Page last updated: 2024-11-11

everolimus

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Cross-References

ID SourceID
PubMed CID6442177
CHEMBL ID1908360
CHEBI ID68478
SCHEMBL ID4378
MeSH IDM000602394

Synonyms (82)

Synonym
rad 001
rapamycin, 42-o-(2-hydroxyethyl)-
(1r,9s,12s,15r,16e,18r,19r,21r,23s,24e,26e,28e,30s,32s,35r)-1,18-dihydroxy-12-((1r)-2-((1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl)-1-methylethyl)-19,30-dimethoxy-15,17,21,23,29,35-hexamethyl-11,36-dioxa-4-azatricyclo(30.3.1.04,9)hexatriaconta-16,2
(1r,9s,12s,15r,16e,18r,19r,21r,23s,24e,26e,28e,30s,32s,35r)-1,18-dihydroxy-12-((1r)-2-((1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl)-1-methylethyl)-19,30-dimethoxy-15,17,21,23,29,35-hexamethyl-11,36-dioxa-4-azatricyclo(30.3.1.0(sup 4,9))hexatriacont
everolimus [usan]
(3s,6r,7e,9r,10r,12r,14s,15e,17e,19e,21s,23s,26r,27r,34as)-9,10,12,13,14,21,22,23,24,25,26,27,32,33,34,34a-hexadecahydro-9,27-dihydroxy-3-((1r)-2-((1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl)-1-methylethyl)-10,21-dimethoxy-6,8,12,14,20,26-hexamethy
HY-10218
votubia
nvp-rad-001
sdz-rad
rad-001c
certican
everolimus ,
afinitor
rad-001
zortress
rad ,
DB01590
42-o-(2-hydroxyethyl)rapamycin
40-o-(2-hydroxyethyl)-rapamycin
rad001
nsc733504
NCGC00167512-01
sdz rad
(3s,6r,7e,9r,10r,12r,14s,15e,17e,19e,21s,23s,26r,27r,34as)-9,27-dihydroxy-3-{(2r)-1-[(1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl]propan-2-yl}-10,21-dimethoxy-6,8,12,14,20,26-hexamethyl-9,10,12,13,14,21,22,23,24,25,26,27,32,33,34,34a-hexadecahydro-3
bdbm50088378
afinitor disperz
chebi:68478 ,
xience v
hsdb 8255
everolimus [usan:inn:ban]
unii-9hw64q8g6g
tox21_112510
dtxsid0040599 ,
cas-159351-69-6
dtxcid8020599
EVEROLIMUS - RAD001
rad 666
CHEMBL1908360
rad-666
9hw64q8g6g ,
everolimus [orange book]
everolimus [who-dd]
everolimus [ep monograph]
everolimus [inn]
everolimus [jan]
everolimus [mart.]
everolimus [vandf]
everolimus [ema epar]
everolimus [usp-rs]
everolimus [mi]
(1r,9s,12s,15r,16e,18r,19r,21r,23s,24e,26e,28e,30s,35r)-1,18-dihydroxy-12-{(2r)-1-[(1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl]propan-2-yl}-19,30-dimethoxy-15,17,21,23,29,35-hexamethyl-11,36-dioxa-4-azatricyclo[30.3.1.0(4,9)]hexatriaconta-16,24,26,
everolimusum
CS-0064
AKOS015850977
40-o-(2-hydroxyethyl)rapamycin
gtpl5889
BRD-K13514097-001-01-2
HKVAMNSJSFKALM-GKUWKFKPSA-N
SCHEMBL4378
Q-101413
rad, sdz
001, rad
everolimus, analytical standard
everolimus (inn)
dihydroxy-[(1r)-2-[(1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxy-cyclohexyl]-1-methyl-ethyl]-dimethoxy-hexamethyl-[?]pentone
Q421052
AS-16971
BRD-K13514097-001-05-3
42-o-(2-hydroxyethyl)-rapamycin
EX-A2057
everolimus; rad001; sdz-rad
(1r,9s,12s,15r,16e,18r,19r,21r,23s,24e,26e,28e,30s,35r)-1,18-dihydroxy-12-((2r)-1-((1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl)propan-2-yl)-19,30-dimethoxy-15,17,21,23,29,35-hexamethyl-11,36-dioxa-4-azatricyclo(30.3.1.0(4,9))hexatriaconta-16,24,26,
everolimus tablets
afinitordisperz
everolimus (usp-rs)
everolimus (mart.)
(3s,6r,7e,9r,10r,12r,14s,15e,17e,19e,21s,23s,26r,27r,34as)-9,27-dihydroxy-3-((2r)-1-((1s,3r,4r)-4-(2-hydroxyethoxy)-3-methoxycyclohexyl)propan-2-yl)-10,21-dimethoxy-6,8,12,14,20,26-hexamethyl-9,10,12,13,14,21,22,23,24,25,26,27,32,33,34,34a-hexadecahydro-3
l04aa18
l01xe10
everolimus (ep monograph)
eve - everolimus

Research Excerpts

Overview

Everolimus is an oral drug that inhibits mTOR with immunosuppressive and antiproliferative characteristics. Everolimus has been increasingly used in high-risk cardiac rhabdomyomas in recent years.

ExcerptReferenceRelevance
"Everolimus is an mTOR pathway inhibitor with synergistic anti-tumor activity when combined with histone deacetylase inhibitors, such as panobinostat, in preclinical lymphoma models."( Phase II Study of Single-Agent and Combination Everolimus and Panobinostat in Relapsed or Refractory Diffuse Large B-Cell Lymphoma.
Beaven, A; de Castro, C; Diehl, L; Islam, P; Li, Z; Lin, C; Moore, J; Morris, T; Rizzieri, D, 2021
)
1.6
"Everolimus is a mammalian target of rapamycin inhibitor/proliferation-signal inhibitor with potent immunosuppressive and anti-proliferative effects."( An overview of the efficacy and safety of everolimus in adult solid organ transplant recipients.
Barten, MJ; Bernhardt, P; De Simone, P; Gawai, A; Gottlieb, J; Pascual, J; Potena, L; Saliba, F; Tedesco-Silva, H, 2022
)
1.71
"Everolimus is an inhibitor of serine/ threonine kinase mTOR. "( Biomarkers of everolimus efficacy in breast cancer therapy.
Voutsadakis, IA, 2022
)
2.52
"Everolimus is a kind of mammalian target of rapamycin (mTOR) inhibitors. "( Exosome-mediated miR-7-5p delivery enhances the anticancer effect of Everolimus via blocking MNK/eIF4E axis in non-small cell lung cancer.
Fan, S; Liu, S; Luo, J; Ma, J; Ning, Y; Peng, J; Wang, H; Wang, W; Wen, Q; Yang, Y; Zang, H; Zhan, Y; Zheng, H, 2022
)
2.4
"Everolimus is an inhibitor of the mammalian target of rapamycin, and its indications include neuroendocrine tumors."( A case of erythema multiforme-like rash induced by everolimus in a patient with a pancreatic neuroendocrine tumor.
Hayama, H; Ito, T; Kubota, Y; Miura, M; Mizuno, Y; Oya, M; Tashiro, A; Tsuneyoshi, M; Ueda, K; Yamashita, A, 2022
)
1.69
"Everolimus is a mTOR inhibitor that has been increasingly used in high-risk cardiac rhabdomyomas in recent years. "( Effect of different dose regimens of everolimus in a series of neonates with giant cardiac rhabdomyomas.
Babaoğlu, K; Başar, EZ; Günlemez, A; Usta, E; Yılmaz, EH, 2023
)
2.63
"Everolimus is a drug with proven efficacy in the treatment of other conditions related to TS, and some evidence suggests that its use benefits the treatment of refractory epilepsy in these patients."( Everolimus as a therapeutic option in refractory epilepsy in children with tuberous sclerosis: a systematic review.
Dutra, LS; Goes, JS; Menezes, CEG; Morais, LAS; Nery, ES; Portela Filho, MB; Santos, DLD; Serpa, ED, 2023
)
3.07
"Everolimus is an oral drug that inhibits mTOR with immunosuppressive and antiproliferative characteristics. "( Evaluating everolimus for the treatment of breast cancer.
Bourbouloux, E; Campone, M; Frenel, JS; Gourmelon, C; Moreau-Bachelard, C; Patsouris, A; Robert, M,
)
1.96
"Everolimus is an inhibitor of mammalian target of rapamycin (mTOR) used in both transplantation and cancer treatment (breast, renal and neuroendocrine). "( First reported double drug-drug interaction in a cancer renal patient under everolimus treatment: therapeutic drug monitoring and review of literature.
Bleda-Perez, C; Del Valle-Celiz, PM; Fontanals-Martínez, S; Fort-Casamartina, E; Gonzalo-Diego, N; Muñoz-Sanchez, C; Otero-Torres, S; Prats-Jimenez, J; Rigo-Bonnin, RF, 2023
)
2.58
"Everolimus is an active agent in the treatment of prolactinomas that warrants further investigation."( The treatment of aggressive prolactinomas with everolimus.
Geer, EB; Lala, N; Lin, AL; Magge, R; Page-Wilson, G; Tabar, V; Young, RJ, 2023
)
2.61
"Everolimus is an alternative second-line therapy, particularly in nonfunctioning NETs and patients with SSTR-negative tumors."( Systemic Therapy for Tumor Control in Metastatic Well-Differentiated Gastroenteropancreatic Neuroendocrine Tumors: ASCO Guideline.
Arshad, J; Basu, S; Bellizzi, AM; Bergsland, EK; Chan, JA; Chauhan, A; Dasari, AN; Del Rivero, J; Gangi, A; Grady, E; Halfdanarson, TR; Howe, JR; Ivanidze, J; Kennedy, EB; Kunz, PL; Lewis, M; Mailman, J; Mittra, ES; Perez, K; Raj, N; Singh, S; Soares, HP; Soulen, MC; Strosberg, JR; Vijayvergia, N; White, SB, 2023
)
1.63
"Everolimus is a Food and Drug Administration-approved inhibitor of TORC1."( Phase 2 clinical trial of TORC1 inhibition with everolimus in men with metastatic castration-resistant prostate cancer.
Anand, M; Armstrong, AJ; George, DJ; Halabi, S; Healy, P; Jonasch, D; Madden, JF; Rasmussen, J; Spritzer, C; Wood, SY, 2020
)
1.54
"Everolimus is an effective immunosuppressant in organ transplantation without impaired renal function. "( Efficacy and safety of everolimus treatment on liver transplant recipients: A meta-analysis.
Chen, KB; Guan, TW; Lin, YJ; Ou, MY, 2019
)
2.27
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor, which is used in immunosuppressive treatment regimens in solid-organ transplant recipients. "( Noncontrast Magnetic Resonance Lymphangiography in a Rare Case of Everolimus-Related Lymphedema.
Boccanera, D; Cellina, M; Gibelli, D; Menozzi, A; Oliva, G; Soresina, M, 2020
)
2.24
"Everolimus is a treatment in NETs that may have antiangiogenic activity."( Everolimus after hepatic arterial embolisation therapy of metastases from gastrointestinal neuroendocrine tumours: The FFCD 1104-EVACEL-GTE phase II study.
Aparicio, T; Barbier, E; Baudin, E; Bouhier-Leporrier, K; Cadiot, G; Caroli-Bosc, FX; Coriat, R; de Baère, T; Di-Fiore, F; Duluc, M; Dupont-Gossart, AC; Gay, F; Ghiringhelli, F; Guimbaud, R; Hentic-Dhome, O; Lecomte, T; Legoux, JL; Lepage, C; Lepere, C; Lombard-Bohas, C; Petorin, C; Smith, D; Walter, T, 2019
)
2.68
"Everolimus is a well-established second-line treatment."( A Phase 4 Study of Everolimus to Evaluate Efficacy and Safety in Patients with Metastatic Renal-Cell Carcinoma after Failure of First-Line Sunitinib or Pazopanib (SUNPAZ).
de Geeter, P; Decker, T; Quiering, C; Resch, A; Schmitz, S; Schostak, M, 2020
)
1.61
"Everolimus is an mTOR inhibitor, approved as a treatment for cancer and as an immunosuppressant agent in solid organ transplantation; it frequently produces toxic metabolic effects, particularly of the most severe kind. "( [Lipid lowering treatment of severe hypertriglyceridemia with acute pancreatitis caused by everolimus in a patient with a neuroendocrine tumor].
Anda Apiñániz, E; Castro Unanua, N; De Carlos Artajo, J; Irigaray Echarri, A; Muruzábal Huarte, E; Vera García, R; Zubiría Gortázar, J, 2020
)
2.22
"Everolimus is a mammalian target of rapamycin inhibitor and is approved as second-line treatment or beyond for renal cell carcinoma. "( Human herpesvirus 6 encephalitis in a patient treated with everolimus for renal cell carcinoma.
Fitsiori, A; Gantzer, J; Kokkali, S; Oddou, I, 2020
)
2.24
"Everolimus is a drug approved for the management of advanced pNETs that can achieve both these goals."( Should everolimus be stopped after radiological progression in metastatic insulinoma? A "cons" point of view.
Amoroso, V; Berruti, A; Consoli, F; Dalla Volta, A; Ferrari, VD; Tovazzi, V, 2020
)
1.73
"Everolimus (EVR) is a derivative of sirolimus with a similar mechanism of action. "( Safety and Efficacy of Everolimus Rescue Treatment After Pediatric Living Donor Liver Transplantation.
Bessho, K; Deguchi, K; Kodama, T; Noguchi, Y; Nomura, M; Okuyama, H; Saka, R; Tazuke, Y; Ueno, T; Watanabe, M,
)
1.88
"Everolimus is a valid therapeutic option for advanced, progressive, well-differentiated NEN, even in patients with positive FDG-PET findings."( [
Annibale, B; Franchi, G; Iannicelli, E; Laghi, A; Magi, L; Mazzuca, F; Panzuto, F; Pilozzi, E; Prosperi, D; Rinzivillo, M; Signore, A, 2021
)
2.06
"Everolimus is a mTOR inhibitor which demonstrates clinical activity in several solid tumors, especially in kidney cancer after first line TKI anti VEGF treatment and in breast cancer in association with exemesthane after failure of aroamatase inhibitors. "( [Tolerablity of everolimus in clinical practice: a retrospective study].
Afani, L; Awada, A; Belbaraka, R, 2020
)
2.35
"Everolimus is an appropriate first-line choice for somatostatin receptor negative tumors and for any patients with progressive disease."( Pulmonary Neuroendocrine Tumors: Adjuvant and Systemic Treatments.
Halfdanarson, TR; Leventakos, K; Molina, JR; Uprety, D, 2020
)
1.28
"Everolimus (Eve) is an immunosuppressive macrolide that is being analyzed in various biological matrices and fluids. "( Development and validation of a simple and versatile method for the quantification of everolimus loaded in H-ferritin nanocages using UHPLC-MS/MS.
Bonizzi, A; Calleri, E; Corsi, F; Cottica, D; Grignani, E; Mazzucchelli, S; Negri, S; Prati, F; Sottani, C, 2020
)
2.22
"Everolimus is a safe and effective therapeutic option for renal angiomyolipoma and various manifestations of TSC, which has been reproduced in real life with six years of follow-up."( Renal angiomyolipoma and tuberous sclerosis complex: long-term safety and efficacy outcomes of Everolimus therapy.
Álvarez-Ossorio Fernández, JL; Ledo Cepero, MJ; Ojeda Claro, AV; Ruiz Guerrero, E; Soto Delgado, M, 2021
)
2.28
"Everolimus is an immunosuppressant agent used in organ transplantation and, more recently, in cancer therapy. "( Complex regional pain syndrome in a patient with neuroendocrine tumour under treatment with everolimus.
Fernández-Rodríguez, T; Oliver-Noguera, A; Pérez de Amezaga-Tomás, L; Rodríguez-Camacho, JM, 2022
)
2.38
"Everolimus is an alternative treatment for several comorbidities, but its early use (< 3 years) requires further study."( [Tuberous sclerosis complex: analysis of areas of involvement, treatment progress and translation to routine clinical practice in a cohort of paediatric patients].
Aparicio-López, C; Bernardino-Cuesta, B; Cantarín-Extremera, V; de Lucas-Collantes, C; Duat-Rodríguez, A; Hernández-Martín, A; Martín-Villaescusa, C; Melero-Llorente, J; Ruiz-Falcó-Rojas, ML; Tamariz Martel-Moreno, A, 2021
)
1.34
"Everolimus is an important drug used for the treatment of NETs."( Effect of everolimus on the expression of Ki-67 and caspase-3 in patients with neuroendocrine tumors.
Cheng, XB; Jin, LD; Li, Y; Yin, HP; Zhang, HY, 2017
)
1.58
"Everolimus is a small molecule that inhibits the mammalian target of rapamycin (mTOR) and is used for treatment of various solid tumours and renal transplant rejection prophylaxis. "( Everolimus-induced nephrotic syndrome precipitated by interaction with voriconazole in a patient with Hodgkin's lymphoma.
Pinter-Brown, LC; Tran, PN, 2017
)
3.34
"Everolimus is a newer generation mammalian target of rapamycin inhibitor approved for immunosuppressive use in a number of advanced medical conditions. "( A Case of Everolimus-Induced Eyelid Edema.
Rodgers, R; Schear, MJ,
)
1.98
"Everolimus is a drug used successfully in a number of different oncology indications, but significant on-target toxicities exist. "( Continuous low plasma concentrations of everolimus provides equivalent efficacy to oral daily dosing in mouse xenograft models of human cancer.
Guthy, D; Laborde, L; McSheehy, P; Oz, F; Ristov, M; Sterker, D, 2017
)
2.17
"Everolimus is a standard treatment option for advanced pancreatic neuroendocrine tumors (pNETs). "( Clinical outcomes of everolimus in patients with advanced, nonfunctioning pancreatic neuroendocrine tumors: a multicenter study in Korea.
Bang, S; Cho, JH; Jeong, S; Kim, YT; Lee, JK; Lee, KH; Lee, KJ; Lee, SH; Lee, TH; Lee, WJ; Paik, WH; Ryu, JK; Song, SY; Woo, SM, 2017
)
2.22
"Everolimus is a promising candidate that combines immunosuppressive properties with anti-neoplastic effects."( Everolimus for pediatric patients with acute graft-versus-host disease after hematopoietic stem cell transplantation: A pilot study.
Chang, YC; Chao, YH; Peng, CT; Weng, TF; Wu, HP; Wu, KH, 2017
)
2.62
"Everolimus is an inhibitor of mammalian target of rapamycin (mTOR), and is available as oral tablets."( Evaluation of the Effect of Everolimus on Retinal Pigment Epithelial Cells and Experimental Proliferative Vitreoretinopathy.
Chen, YH; Ke, MC; Kuo, HK; Kuo, YH; Tseng, YC; Wu, PC, 2018
)
1.5
"Everolimus is an inhibitor of the mammalian target of rapamycin (mTOR) that has been approved by the US Food and Drug Administration for the treatment of subependymal giant cell astrocytoma (SEGA) in patients with tuberous sclerosis complex (TSC). "( Retinal astrocytoma regression in tuberous sclerosis patients treated with everolimus.
Ali, A; Tehrani, NN; Zipori, AB, 2018
)
2.15
"Everolimus is a mammalian target of rapamycin inhibitor, whose immunosuppressant properties are both desired in transplant patients and unwanted in cancer patients, where it is indicated for its antiproliferative efficacy."( The immune system as a chronotoxicity target of the anticancer mTOR inhibitor everolimus.
Cinar, S; Deniz, G; Giacchetti, S; Kaptan, E; Lévi, F; Li, XM; Okyar, A; Ozsoy, N; Ozturk, D; Ozturk, N; Pala-Kara, Z; Sancar-Bas, S, 2018
)
1.43
"Everolimus is a treatment option for hemodialysis patients with metastatic atypical bronchial carcinoid. "( Efficacy and safety of everolimus treatment in a hemodialysis patient with metastatic atypical bronchial carcinoid: case report and literature review.
Brizzi, MP; La Salvia, A; Scagliotti, GV; Sonetto, C; Tampellini, M; Volante, M, 2018
)
2.23
"Everolimus (C53H83NO14) is an efficient anti-cancer drug for breast cancer which targets mammalian target of rapamycin (mTOR)."( Everolimus inhibits breast cancer cell growth through PI3K/AKT/mTOR signaling pathway.
Chen, W; Du, L; Li, X; Mu, L; Song, A; Zhang, Y; Zhen, L, 2018
)
2.64
"Everolimus is a drug from the class of mammalian target of rapamycin inhibitors used for both immunosuppressant and oncological indications. "( A pharmacological rationale for improved everolimus dosing in oncology and transplant patients.
Burger, DM; de Fijter, JW; Guchelaar, HJ; Moes, DJAR; Reinders, MEJ; Ter Heine, R; van Erp, NP; van Herpen, CM, 2018
)
2.19
"Everolimus is an inhibitor of the mammalian target of rapamycin (mTOR) and has been used in combination with calcineurin inhibitors (tacrolimus and cyclosporine) to prevent allograft rejection following organ transplantation. "( Effect of fluconazole on the pharmacokinetics of everolimus and tacrolimus in a heart transplant recipient: Case report.
Matsuda, S; Nakagita, K; Oita, A; Takada, M; Terada, Y; Terakawa, N; Wada, K, 2018
)
2.18
"Everolimus (RAD-001) is an orally active rapamycin analogue shown in preclinical data to produce cytostatic cell inhibition, which may be potentially beneficial in treating melanoma. "( Phase II Study of Everolimus in Metastatic Malignant Melanoma (NCCTG-N0377, Alliance).
Allred, JB; Creagan, ET; Erickson, LA; Kaur, JS; Lowe, VJ; Maples, WJ; Markovic, S; Rao, RD; Suman, VJ; Vera Aguilera, J; Windschitl, HE, 2018
)
2.26
"Everolimus is an allosteric mTOR inhibitor with efficacy in metastatic RCC."( Comprehensive Genomic Profiling of Metastatic Tumors in a Phase 2 Biomarker Study of Everolimus in Advanced Renal Cell Carcinoma.
Bhatt, RS; Catalano, PJ; Choueiri, TK; Gao, X; George, DJ; Gray, C; Jegede, O; Kwiatkowski, DJ; McDermott, DF; McKay, RR; Novak, J; Signoretti, S, 2018
)
1.43
"Everolimus, which is a mammalian target of rapamycin inhibitor, has an anti-fibrosis effect."( Everolimus Rescue Treatment for Chronic Rejection After Pediatric Living Donor Liver Transplantation: 2 Case Reports.
Bessho, K; Hiwatashi, S; Okuyama, H; Saka, R; Takama, Y; Tazuke, Y; Ueno, T; Yamanaka, H, 2018
)
2.64
"Everolimus is an inhibitor of mammalian target of rapamycin (mTOR) and reduces the risk of cytomegalovirus (CMV) infection in transplant recipients. "( Everolimus delayed and suppressed cytomegalovirus DNA synthesis, spread of the infection, and alleviated cytomegalovirus infection.
Sato, N; Shiraki, A; Shiraki, K; Takemura, Y; Tan, L; Yanagita, M; Yoshida, Y, 2019
)
3.4
"Everolimus is a mammalian target of rapamycin (m-TOR) inhibitor that has been approved for the treatment of hormone receptor-positive advanced breast cancer, metastatic renal cancer, and neuroendocrine tumors. "( Therapeutic Drug Monitoring of Everolimus in Oncology: Evidences and Perspectives.
Falkowski, S; Woillard, JB, 2019
)
2.24
"Everolimus is an mTOR (the mammalian target of rapamycin) inhibitor, which is used for the treatment of advanced renal cell carcinoma. "( Severe gastrointestinal hemorrhage related to everolimus: a case report.
Haruki, K; Masubuchi, M; Saito, R; Tsunematsu, M; Watanabe, M; Yanaga, K, 2019
)
2.21
"Everolimus (EVR) is a mammalian target of rapamycin (mTOR) inhibitor commonly used for immunosuppression (IS) after liver transplantation (LT). "( The Effect of Immunosuppression on Coagulation After Liver Transplantation.
Achterfeld, A; Bedreli, S; Herzer, K; Katsounas, A; Saner, F; Straub, K; Wedemeyer, H; Willuweit, K, 2019
)
1.96
"Everolimus is a mammalian target of rapamycin inhibitor used in the treatment of multiple tumor types, and its most common toxicity, stomatitis, can affect patient quality of life. "( A single-arm, phase 2 study of steroid-containing mouthwash for the prevention of everolimus-associated stomatitis in multiple tumor types.
Adachi, Y; Andoh, M; Gondo, N; Hagiwara, S; Hattori, M; Hijioka, S; Iwata, H; Kotani, H; Mizuno, Y; Sawaki, M; Shimizu, J; Tachi, M; Tatematsu, M; Yoshimura, A; Yoshimura, K, 2019
)
2.18
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor that has cytoreductive effects on subependymal giant cell astrocytoma and renal angiomyolipoma in tuberous sclerosis complex (TSC). "( [The Efficacy of Everolimus for Refractory Seizures in Childhood Onset Epilepsy with Tuberous Sclerosis Complex].
Baba, S; Enoki, H; Fujimoto, A; Itamura, S; Nishimura, M; Okanari, K; Okanishi, T, 2019
)
2.3
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved as an immunosuppressant and for second-line therapy of hepatocellular carcinoma (HCC) and renal cell carcinoma (RCC). "( Physiologically based pharmacokinetic models for everolimus and sorafenib in mice.
Fetterly, GJ; Hylander, BH; Jusko, WJ; Ma, WW; Pawaskar, DK; Repasky, EA; Straubinger, RM, 2013
)
2.09
"Everolimus is an immunosuppressant drug used in solid organ transplantation. "( Comparison of a stable isotope-labeled and an analog internal standard for the quantification of everolimus by a liquid chromatography-tandem mass spectrometry method.
Heideloff, C; Payto, D; Wang, S, 2013
)
2.05
"Everolimus is a synthetic, orally available analogue of rapamycin that inhibits the activation of mTOR."( Evaluation of everolimus in renal cell cancer.
Amato, R; Stepankiw, M, 2013
)
1.47
"Everolimus is a potent immunosuppressive agent that has antiproliferative activities. "( Clinical everolimus experience in pediatric renal transplant patients.
Bulut, IK; Dincel, N; Hoşcoşkun, C; Mir, S; Sezer, TÖ, 2013
)
2.25
"Everolimus is a potent, oral inhibitor of the mammalian target of rapamycin (mTOR) that has been investigated in multiple clinical development programs since 1996. "( Development of everolimus, a novel oral mTOR inhibitor, across a spectrum of diseases.
Anak, O; Berg, W; Elmroth, I; Haas, T; Klimovsky, J; Lebwohl, D; Posluszny, J; Sahmoud, T; Saletan, S, 2013
)
2.19
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved for the treatment of advanced renal cell carcinoma, pancreatic neuroendocrine tumors, subependymal giant cell astrocytoma associated with tuberous sclerosis complex, renal angiomyolipoma and tuberous sclerosis complex, and, in combination with exemestane, for hormone receptor-positive HER2-negative advanced breast cancer after failure of treatment with letrozole or anastrozole. "( Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trial.
Peterson, ME, 2013
)
2.05
"Everolimus is an orally administered inhibitor of the mammalian target of rapamycin (mTOR), an intracellular protein kinase downstream of the phosphatidylinositol 3-kinase/AKT pathway involved in key components of tumorigenesis, including cell growth, proliferation, and angiogenesis. "( Practical management of everolimus-related toxicities in patients with advanced solid tumors.
Geberth, M; Grünwald, V; Hörsch, D; Janni, W; Lüftner, D; Pavel, ME; Weber, MM; Weikert, S, 2013
)
2.14
"Everolimus is an immunosuppressant and an antiproliferative macrolide agent which is used for coronary stent coating for local inhibition of in-stent restenosis. "( Everolimus-coated tympanostomy tube on rat tympanic membrane.
Akbulut, S; Altintaş, H; Barisik, NO; Berk, D; Sezen, OS, 2013
)
3.28
"Everolimus is a widely used oral mTOR inhibitor that has the potential for drug interactions that may affect therapeutic outcomes, produce toxicities, or both. "( Drug interactions and the pharmacist: focus on everolimus.
Grabowsky, JA,
)
1.83
"Everolimus is a potent immunosuppressant with several advantages over calcineurin inhibitors, such as good tolerance, preventive effects on cardiovascular morbidity, and mortality and cancer prevention as it inhibits cell proliferation."( Everolimus monotherapy or combined therapy in liver transplantation: indications and results.
Abradelo, M; Alegre, C; Alvaro, E; Calvo, J; Cambra, F; Caso, O; García, M; García-Sesma, A; Jiménez, C; Justo, I; Loinaz, C; Manrique, A; Moreno, E; Sanabria, R, 2013
)
3.28
"Everolimus (EVR) is a semi-synthetic mammalian target of rapamycin inhibitor currently under development for liver transplantation (LTx) in combination with reduced exposure tacrolimus (rTAC). "( mTOR inhibition in liver transplantation: how to dose for effective/safe CNI reduction?
Dumortier, T; Fung, J; Junge, G; Schwende, H, 2013
)
1.83
"Everolimus is an immunosuppressive drug metabolized by enzymes of the CYP family. "( The role of CYP2C8 genotypes in dose requirement and levels of everolimus after heart transplantation.
Kniepeiss, D; Renner, W; Tscheliessnigg, KH; Wagner, D; Wasler, A, 2013
)
2.07
"Everolimus is an mammalian target of rapamycin complex 1 inhibitor with demonstrated benefit in several aspects of TSC."( Everolimus treatment of refractory epilepsy in tuberous sclerosis complex.
Agricola, K; Anderson, AE; Franz, DN; Holland-Bouley, K; Kim, MO; Krueger, DA; Lopez, CM; Mays, M; Tudor, C; Wilfong, AA, 2013
)
2.55
"Everolimus is an orally available inhibitor of the mammalian target of rapamycin (mTOR), which has been approved in combination with exemestane for hormone receptor-positive (HR) breast cancer after failure of treatment with non-steroidal aromatase inhibitors. "( Everolimus: side effect profile and management of toxicities in breast cancer.
O'Regan, R; Paplomata, E; Zelnak, A, 2013
)
3.28
"Everolimus is a mammalian target of rapamycin inhibitor approved for the treatment of metastatic renal cell carcinoma (mRCC). "( Pre-treatment neutrophil-to-lymphocyte ratio may be associated with the outcome in patients treated with everolimus for metastatic renal cell carcinoma.
Amadori, D; Berardi, R; Burattini, L; Cascinu, S; Conti, A; Cortesi, E; De Giorgi, U; Iacovelli, R; Luca Burgio, S; Muzzonigro, G; Rossi, L; Santoni, M, 2013
)
2.05
"Everolimus is a serine-threonine kinase that acts as an inhibitor of mammalian target of rapamycin, which results in reduced growth of cells, angiogenesis, and survival of cells."( Osteonecrosis of the jaw related to everolimus: a case report.
Jung, HD; Jung, YS; Kim, DW; Park, HS, 2013
)
1.39
"Everolimus is a derivative of rapamycin and acts as a signal transduction inhibitor."( The antiproliferative effects of ouabain and everolimus on adrenocortical tumor cells.
Barollo, S; Cicala, MV; Iacobone, M; Mantero, F; Mian, C; Mucignat-Caretta, C; Pezzani, R; Radu, C; Redaelli, M; Rubin, B; Salvà, M; Simioni, P, 2014
)
1.38
"Everolimus is an oral mammalian target of rapamycin inhibitor approved for advanced renal cell carcinoma (RCC)."( A phase 1b clinical trial of the multi-targeted tyrosine kinase inhibitor lenvatinib (E7080) in combination with everolimus for treatment of metastatic renal cell carcinoma (RCC).
Carter, D; Gold, AM; Hutson, TE; Larkin, J; Michaelson, MD; Molina, AM; Motzer, R; Wood, K, 2014
)
1.33
"Everolimus (EVE) is a drug widely used in several renal transplant protocols. "( Everolimus-induced epithelial to mesenchymal transition in immortalized human renal proximal tubular epithelial cells: key role of heparanase.
Gambaro, G; Granata, S; Lupo, A; Masola, V; Onisto, M; Zaza, G, 2013
)
3.28
"Everolimus is an oral inhibitor of mTOR complex 1 (mTORC1) with antitumor activity in a variety of cancers."( Phase II study of everolimus in children and adults with neurofibromatosis type 2 and progressive vestibular schwannomas.
Allen, JC; Derman, A; Filatov, A; Giancotti, FG; Goldberg, JD; Golfinos, JG; Hagiwara, M; Hochman, T; Karajannis, MA; Legault, G; Merkelson, A; Roland, JT; Vega, E; Wisoff, JH, 2014
)
1.46
"Everolimus is an inhibitor of mammalian target of rapamycin, approved in patients with metastatic renal cell carcinoma. "( Immunomodulatory effects of everolimus in a long responsive patient with metastatic renal cell carcinoma.
Adotevi, O; Borg, C; Laheurte, C; Mansi, L; Pivot, X; Royer, B; Thiery-Vuillemin, A, 2014
)
2.14
"Everolimus is an orally administrated mammalian target of rapamycin (mTOR) inhibitor. "( Therapeutic potential and adverse events of everolimus for treatment of hepatocellular carcinoma - systematic review and meta-analysis.
Galli, U; Gao, C; Hatano, E; Houben, P; Lin, S; Petrulionis, M; Richter, S; Schemmer, P; Schultze, D; Winkler, S; Yamanaka, K, 2013
)
2.09
"Everolimus is an oral derivative of rapamycin which acts as a signal transduction inhibitor. "( Everolimus in advanced solid tumors: when to start, early or late?
Procopio, G; Pusceddu, S; Tessari, A; Testa, I,
)
3.02
"Everolimus is an oral raptor mTOR inhibitor and has shown activity in patients with Waldenstrom's macroglobulinemia (WM). "( Long-term results of the phase II trial of the oral mTOR inhibitor everolimus (RAD001) in relapsed or refractory Waldenstrom Macroglobulinemia.
Bergsagel, PL; Camoriano, J; Chuma, S; DeAngelo, D; Gertz, M; Ghobrial, IM; Hayman, S; Lacy, M; LaPlant, B; Treon, SP; Witzig, TE, 2014
)
2.08
"Everolimus is an orally administered mTOR inhibitor. "( Everolimus prolonged survival in transgenic mice with EGFR-driven lung tumors.
Harada, D; Honda, Y; Ichihara, E; Kiura, K; Murakami, T; Ninomiya, T; Ochi, N; Ohashi, K; Takigawa, N; Tanimoto, M; Yasugi, M, 2014
)
3.29
"Everolimus is a targeted agent which was effective in several clinical trials and became registered for treatment of hormone receptor positive breast cancer."( [BOLERO -- another remarkable step in treatment of breast cancer].
Láng, I; Rubovszky, G, 2014
)
1.12
"Everolimus is a mammalian target of rapamycin inhibitor and exhibits synergistic immunosuppressive activity with CNI to permit CNI-reduction."( Everolimus-based calcineurin-inhibitor sparing regimens for kidney transplant recipients: a systematic review and meta-analysis.
Chen, P; Deng, R; Li, H; Su, L; Tam, N; Wu, L, 2014
)
2.57
"Everolimus (ERL) is an immunosuppressive agent with antitumoral properties."( Conversion to everolimus dramatically improves the prognosis of de novo malignancies after liver transplantation for alcoholic liver disease.
Boillot, O; Decullier, E; Dumortier, J; Guillaud, O; Thimonier, E; Vallin, M; Walter, T, 2014
)
1.48
"Everolimus is an orally administered anti-cancer drug that inhibits the mammalian target of rapamycin (mTOR) signal transduction route. "( [Hyperglycaemia during treatment with everolimus].
Beijnen, JH; Huitema, AD; Opdam, FL; Schellens, JH, 2014
)
2.12
"Everolimus is a valid therapeutic option for neuroendocrine tumors (NETs); however, data in a real-world setting outside regulatory trials are sparse. "( Real-world study of everolimus in advanced progressive neuroendocrine tumors.
Ambrosio, MR; Antonuzzo, L; Appetecchia, M; Berruti, A; Brizzi, MP; Campana, D; Cartenì, G; Cascinu, S; Cassano, A; Chiara, S; Colao, A; de Braud, F; Delle Fave, G; Faggiano, A; Falconi, M; Fazio, N; Fontana, A; Funaioli, C; Lugli, F; Luppi, G; Merlano, M; Nobili, E; Nuzzo, C; Panzuto, F; Pastorelli, D; Pusceddu, S; Riccardi, F; Rinzivillo, M; Spada, F; Tomassetti, P; Zaniboni, A; Zatelli, MC, 2014
)
2.17
"Everolimus is a novel proliferation signal inhibitor used in immunosuppressive therapies for the prevention of acute and chronic rejection. "( Latex-enhanced turbidimetric immunoassay for everolimus in whole blood using the Nanopia TDM everolimus assay with the JCA-BM6010 automatic analyzer.
Fukushima, N; Hidaka, Y; Kimura, S; Matsui, M; Mori, K; Suehisa, E, 2014
)
2.1
"Everolimus (EVL) is an immunosuppressant indicated for allograft rejection and cancer therapy, but with narrow therapeutic window."( Oral intake of curcumin markedly activated CYP 3A4: in vivo and ex-vivo studies.
Chao, PD; Hou, YC; Hsieh, YW; Huang, CY; Peng, YH; Yang, SY; Yu, CP, 2014
)
1.12
"Everolimus is an agent frequently associated with specific toxicities. "( Translational studies within the TAMRAD randomized GINECO trial: evidence for mTORC1 activation marker as a predictive factor for everolimus efficacy in advanced breast cancer.
Abadie-Lacourtoisie, S; Arnedos, M; Bachelot, T; Bourcier, AV; Cropet, C; Eymard, JC; Ferrero, JM; Legouffe, E; Levy, C; Lortholary, A; Pujade-Lauraine, E; Spaeth, D; Treilleux, I; Wang, Q, 2015
)
2.06
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor. "( Efficacy and Safety of Sequential Use of Everolimus in Patients With Metastatic Renal Cell Carcinoma Previously Treated With Bevacizumab With or Without Interferon Therapy: Results From the European AVATOR Study.
Emmanouilides, C; Guillot, A; Jacobasch, L; Kelkouli, N; Kim, S; Nguyen Tan Hon, T; Papandreou, C; Schmitz, J; Slimane, K; Theodore, C; Thiery-Vuillemin, A, 2015
)
2.13
"Everolimus is an orally administered inhibitor of the mammalian target of rapamycin (mTOR) recommended for patients with metastatic renal cell carcinoma (mRCC) who progressed on previous vascular endothelial growth factor (VEGF) receptor-tyrosine kinase inhibitor therapy. "( Everolimus in patients with metastatic renal cell carcinoma previously treated with bevacizumab: a prospective multicenter study CRAD001LRU02T.
Gurina, L; Manzuk, L; Safina, S; Snegovoy, A; Tsimafeyeu, I; Varlamov, I; Varlamov, S, 2015
)
3.3
"Everolimus is a mammalian target of rapamycin inhibitor that has been used for immunosuppression, but its effect on recurrence and survival in HCC patients with a high risk of tumor recurrence has not been examined."( Everolimus-based immunosuppression in patients with hepatocellular carcinoma at high risk of recurrence after liver transplantation: a case series.
Diaz, SP; Ferreiro, AO; Gutiérrez, MG; López, FS; Patiño, MJ; Vazquez-Millán, MA, 2014
)
2.57
"Everolimus is an effective and safe treatment option for TSC-related SEGA. "( Factors affecting response to everolimus therapy for subependymal giant cell astrocytomas associated with tuberous sclerosis.
Baranska, D; Dachowska, I; Fendler, W; Jozwiak, S; Kotulska, K; Mlynarski, W; Trelinska, J, 2015
)
2.15
"Everolimus (EVE) is an immunosuppressive drug dosed according to therapeutic drug monitoring in renal transplant recipients. "( Closer to the Site of Action: Everolimus Concentrations in Peripheral Blood Mononuclear Cells Correlate Well With Whole Blood Concentrations.
Åsberg, A; Christensen, H; Falck, P; Midtvedt, K; Robertsen, I; Vethe, NT, 2015
)
2.15
"Everolimus is a promising pharmacological approach to treat clinically significant inoperable cardiac rhabdomyomas or subependymal giant cell astrocytoma associated with tuberous sclerosis complex during the neonatal period."( Use of mTOR inhibitor everolimus in three neonates for treatment of tumors associated with tuberous sclerosis complex.
Dahdah, N; Goyer, I; Major, P, 2015
)
2.17
"Everolimus is an orally-administered inhibitor of the mammalian target of rapamycin."( The ORCHIDEE study: gathering new evidence on the use of everolimus in clinical practice.
Cartenì, G; Floriani, I; Hollander, L; Pappagallo, G; Rizzo, M,
)
1.1
"Everolimus is an immunosuppressant drug that was approved for prophylactic use in the United States to prevent organ rejection in adult kidney and liver transplant recipients. "( Everolimus method comparison between Waters MassTrak™ Immunosuppressants XE (IUO) kit and an in-house laboratory developed LC-MS/MS method in renal transplant patients.
Bradshaw, T; Johnson-Davis, KL; Juenke, JM; Thomas, RL, 2015
)
3.3
"Everolimus is a recently approved option for posttransplant immunosuppression that spares renal function."( Update on the management of the liver transplant patient.
Fix, OK; Kwong, AJ, 2015
)
1.14
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved for the treatment of metastatic renal cell carcinoma (mRCC). "( The changes of lipid metabolism in advanced renal cell carcinoma patients treated with everolimus: a new pharmacodynamic marker?
Ballatore, V; Carteni, G; Cascinu, S; Conti, A; Crucitti, P; Felici, A; Galli, L; Guida, FM; Iacovelli, R; Lugini, A; Milella, M; Modica, D; Ortega, C; Pantano, F; Porta, C; Procopio, G; Rauco, A; Rizzo, M; Russo, A; Santini, D; Santoni, M; Schinzari, G; Silletta, M; Tonini, G; Verzoni, E, 2015
)
2.08
"Everolimus (RAD001) is an orally administered mTOR inhibitor that is well known for its antitumor efficacy and that has been approved for the treatment of several solid tumors, including renal cell carcinoma. "( Successful control of heavily pretreated metastatic gastric cancer with the mTOR inhibitor everolimus (RAD001) in a patient with PIK3CA mutation and pS6 overexpression.
Kang, YK; Na, YS; Park, JH; Park, SR; Park, YS; Rhoo, BY; Ryu, MH, 2015
)
2.08
"Everolimus is an oral agent that targets the mammalian target of rapamycin (mTOR) pathway. "( The mTORC1 inhibitor everolimus has antitumor activity in vitro and produces tumor responses in patients with relapsed T-cell lymphoma.
Ansell, SM; Colgan, JP; Gupta, M; Habermann, TM; Han, JJ; Inwards, DJ; Johnston, PB; LaPlant, B; Macon, W; Markovic, S; Micallef, IN; Nowakowski, GS; Porrata, LF; Reeder, C; Stenson, M; Tun, HW; Witzig, TE, 2015
)
2.18
"Everolimus is a viable option for the treatment of renal dysfunction, and earlier conversion is associated with better recovery of renal function."( Renal function improvement in liver transplant recipients after early everolimus conversion: A clinical practice cohort study in Spain.
Almohalla, C; Bilbao, I; Bustamante, J; Castroagudín, J; Crespo, G; Cuervas-Mons, V; De la Mata, M; Fabregat, J; Gómez, MA; Gonzalez-Dieguez, ML; Guilera, M; Herrero, I; Jimenez, C; Miras, M; Moreno, A; Narvaez, I; Otero, A; Rodrigo, J; Rubín, A; Salcedo, M; Serrano, T, 2015
)
1.37
"Everolimus is an mTOR inhibitor approved for use in a number of indications where mTOR overactivation is implicated, including tuberous sclerosis complex."( Long-Term Everolimus Treatment in Individuals With Tuberous Sclerosis Complex: A Review of the Current Literature.
Tran, LH; Zupanc, ML, 2015
)
1.54
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor used in vascular endothelial growth factor (VEGF)-refractory metastatic renal cell carcinoma (mRCC). "( A Randomised Phase 2 Study of AZD2014 Versus Everolimus in Patients with VEGF-Refractory Metastatic Clear Cell Renal Cancer.
Ahmed, I; Boleti, E; Burke, W; Din, O; Geldart, T; Hussain, S; Powles, T; Ralph, C; Robinson, A; Sarker, SJ; Stockdale, A; Sundar, S; Wheater, M; Wimalasingham, A, 2016
)
2.14
"Everolimus is an oral inhibitor of the mammalian target of rapamycin pathway, which is aberrantly activated in NSCLC."( Toxicity and adverse effects of everolimus in the treatment of advanced nonsmall cell lung cancer pretreated with chemotherapy--Chinese experiences.
Hu, Y; Jiao, S; Ju, Y; Sun, S; Wang, J, 2015
)
1.42
"Everolimus is an mammalian target of rapamycin (mTOR) complex 1 inhibitor that has been successfully used to treat subependymal giant cell astrocytomas and renal angiomyolipomas associated with TSC."( Rapid resolution of cardiac rhabdomyomas following everolimus therapy.
Anwar, S; Choudhry, S; Nguyen, HH, 2015
)
1.39
"Everolimus is an approved agent for use after disease progression with vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) in patients with metastatic renal cell carcinoma. "( Everolimus Versus Temsirolimus in Metastatic Renal Cell Carcinoma After Progression With Previous Systemic Therapies.
Agarwal, AM; Agarwal, N; Gill, DM; Hsu, J; Pal, SK; Patel, SB; Stenehjem, DD; Tantravahi, SK; Vuong, W, 2016
)
3.32
"Everolimus appears to be a safe therapeutic option for patients aged <3 years with TSC-associated SEGA. "( Safety of Everolimus in Patients Younger than 3 Years of Age: Results from EXIST-1, a Randomized, Controlled Clinical Trial.
Berkowitz, N; Brechenmacher, T; Franz, DN; Jóźwiak, S; Kotulska, K, 2016
)
2.28
"Everolimus (E) is a potent inhibitor of mTOR, a pathway frequently activated in HCC."( Sorafenib with or without everolimus in patients with advanced hepatocellular carcinoma (HCC): a randomized multicenter, multinational phase II trial (SAKK 77/08 and SASL 29).
Bodoky, G; Buehlmann, M; Demeter, G; Dufour, JF; Feilchenfeldt, J; Horber, D; Koeberle, D; Lakatos, G; Li, Q; Montemurro, M; Peck-Radosavljevic, M; Rauch, D; Ribi, K; Roth, AD; Saletti, P; Samaras, P; Tschanz, B; Wagner, AD, 2016
)
1.46
"Everolimus is an immunosuppressant agent that has antiproliferative properties and negative effects on wound healing. "( Extending Myringotomy Patency with Topical Everolimus in Rats.
Akbulut, S; Altintas, H; Berk, D; Ozdemir, N,
)
1.84
"Everolimus (EVR) is an orally-administered rapamycin analog that selectively inhibits the mammalian target of rapamycin (mTOR) kinase (mainly mTORC1 and likely mTORC2) and the related signaling pathway. "( Everolimus improves memory and learning while worsening depressive- and anxiety-like behavior in an animal model of depression.
Aiello, R; Chimirri, S; Citraro, R; Constanti, A; Crupi, R; Cuzzocrea, S; De Sarro, G; Leo, A; Lippiello, P; Russo, E; Spiga, R, 2016
)
3.32
"Everolimus (EVE) is a mammalian target of rapamycin inhibitor (mTOR-I) widely used in transplantation that may determine some severe adverse events, including pulmonary fibrosis. "( Everolimus-induced epithelial to mesenchymal transition (EMT) in bronchial/pulmonary cells: when the dosage does matter in transplantation.
Bellin, G; Carratù, P; Chilosi, M; Ficial, M; Gambaro, G; Granata, S; Lupo, A; Masola, V; Onisto, M; Resta, O; Tomei, P; Ventura, VA; Zaza, G, 2016
)
3.32
"Everolimus is a mammalian target of rapamycin inhibitor that has been recently approved for the treatment of patients with advanced progressive pancreatic neuroendocrine tumor. "( Early Response to Everolimus Therapy Detected on 68Ga-DOTATATE PET/CT in a Patient With Pancreatic Neuroendocrine Tumor.
Atmaca, A; Demirag, G; Ozkan, E; Soydal, C; Ucak Semirgin, S; Yapici, O, 2016
)
2.21
"Everolimus is a mTOR inhibitor used for the treatment of different solid malignancies. "( Everolimus pharmacokinetics and its exposure-toxicity relationship in patients with thyroid cancer.
de Wit, D; den Hartigh, J; Gelderblom, H; Guchelaar, HJ; Kapiteijn, E; Links, TP; Moes, DJ; Roozen, CF; Schneider, TC; van der Hoeven, JJ; van Erp, NP, 2016
)
3.32
"Everolimus (EVR) is a potent mammalian target of rapamycin inhibitor that is used after transplantation and to treat advanced malignancies, as we have done in Taiwan after heart transplantation since 2004."( Malignancy After Heart Transplantation Under Everolimus Versus Mycophenolate Mofetil Immunosuppression.
Chen, YS; Chi, NH; Chou, NK; Huang, SC; Shun, CT; Tsai, JT; Tsao, CI; Wang, CH; Wang, SS; Wang, YJ; Wu, IH; Yu, HY, 2016
)
1.42
"Everolimus (EVL) is a novel mTOR-inhibitor similar to sirolimus (SRL) that is used in organ transplant recipients, often in combination with tacrolimus (TAC) or mycophenolate (MPA). "( Immunoregulatory Effects of Everolimus on In Vitro Alloimmune Responses.
Gallon, L; Huang, X; Leventhal, JR; Levitsky, J; Mathew, JM; Miller, J, 2016
)
2.17
"Everolimus is an inhibitor of mTOR, approved for treatment of advanced pancreatic neuroendocrine tumors (NETs). "( The efficacy and safety of everolimus for the treatment of progressive gastroenteropancreatic neuroendocrine tumors: A multi-institution observational study in Taiwan.
Chen, JS; Chen, LT; Chen, MH; Chen, YY; Chou, WC; Ku, FC; Liu, CT; Lu, CH; Shan, YS; Su, YL, 2016
)
2.17
"Everolimus is a licensed medicine in this patient group, but for a different target of effect."( The use of everolimus in the treatment of neurocognitive problems in tuberous sclerosis (TRON): study protocol for a randomised controlled trial.
Angel, L; Cannings-John, R; Davies, DM; de Vries, PJ; Drew, C; Hood, K; Kirby, N; McDermott, E; McNamara, R; Owen-Jones, E; Randell, E; Sampson, JR; Saxena, A; Smalley, M; Stockwell, L, 2016
)
1.55
"Everolimus is an allosteric inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1) widely known for its potent autophagy stimulating properties. "( Continuous administration of the mTORC1 inhibitor everolimus induces tolerance and decreases autophagy in mice.
Apers, S; De Doncker, M; De Meyer, GRY; Kurdi, A; Leloup, A; Lemmens, K; Martinet, W; Neels, H; Timmermans, JP, 2016
)
2.13
"Everolimus seems to be an effective treatment option not only for SEGA and AML, but also for TSC-related epilepsies. "( Efficacy and safety of Everolimus in children with TSC - associated epilepsy - Pilot data from an open single-center prospective study.
Abraham, K; Dressler, A; Feucht, M; Gröppel, G; Kasprian, G; Laccone, F; Mühlebner-Fahrngruber, A; Samueli, S; Scholl, T, 2016
)
2.19
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) in 2016 for treatment of adults with progressive, well-differentiated, non-functional NETs of gastrointestinal (GI) or lung origin that are unresectable, locally advanced, or metastatic."( Budget impact of everolimus for the treatment of progressive, well-differentiated, non-functional neuroendocrine tumors of gastrointestinal or lung origin that are advanced or metastatic.
Cai, B; Neary, MP; Nellesen, D; Rose, DB, 2017
)
1.52
"Everolimus is a mammalian target of rapamycin inhibitor with both antitumor and radiosensitizing effects."( Phase 1 Trial of Everolimus and Radiation Therapy for Salvage Treatment of Biochemical Recurrence in Prostate Cancer Patients Following Prostatectomy.
Bekelman, JE; Christodouleas, JP; Deville, C; Haas, NB; Mick, R; Narayan, V; Rajendran, R; Subramanian, P; Vapiwala, N, 2017
)
1.52
"Everolimus monotherapy is a valid therapeutic option in the relapsed and/or refractory indolent non-Hodgkin lymphoma patients and is well tolerated."( Efficacy of the oral mTORC1 inhibitor everolimus in relapsed or refractory indolent lymphoma.
Ansell, SM; Bennani, NN; Colgan, JP; Ghobrial, IM; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, BR; Macon, WR; Markovic, SN; Micallef, IN; Mikhael, JR; Northfelt, DW; Nowakowski, GS; Porrata, LF; Reeder, CB; Witzig, TE, 2017
)
1.45
"Everolimus is a more recently developed PSI with molecular structure very similar to that of sirolimus."( Everolimus-related pulmonary toxicity in heart transplant recipients.
de Prada, JA; Expósito, V; Fernández-Valls, M; García-Camarero, T; Gómez-Román, JJ; González-Vilchez, F; Llano-Cardenal, M; Martín-Durán, R; Ruano, J, 2008
)
2.51
"Everolimus is an immunosuppressant drug with proven efficacy in transplantation. "( Everolimus: an update on the mechanism of action, pharmacokinetics and recent clinical trials.
Sánchez-Fructuoso, AI, 2008
)
3.23
"Everolimus (RAD001) is an orally administered inhibitor of the mammalian target of rapamycin (mTOR), a therapeutic target for metastatic renal cell carcinoma. "( Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial.
Berg, WJ; Bracarda, S; Escudier, B; Figlin, RA; Grünwald, V; Hollaender, N; Hutson, TE; Kay, A; Lebwohl, D; Motzer, RJ; Oudard, S; Porta, C; Ravaud, A; Thompson, JA; Urbanowitz, G, 2008
)
2.17
"Everolimus is a poorly soluble drug and was, therefore, incorporated in an eye-administrable microemulsion. "( Corneal permeation studies of everolimus microemulsion.
Baspinar, Y; Bertelmann, E; Borchert, HH; Buech, G; Pleyer, U; Siebenbrodt, I, 2008
)
2.08
"Everolimus is a novel proliferation signal inhibitor that has potent immunosuppressive activity. "( Prolongation of corneal allograft survival by topical application of everolimus in experimental keratoplasty.
Bertelmann, E; Buch, G; Li, XQ; Otasevic, L; Pleyer, U; Schlickeiser, S, 2008
)
2.02
"Everolimus is a potential trigger of angioedema."( [67-year-old patient with speech disorder and dysphagia].
Er, F; Erdmann, E; Nia, AM, 2008
)
1.79
"Everolimus is a potent immunosuppressive drug that is as effective as mycophenolate mofetil (MMF) or azathioprine (AZA) in preventing acute transplant rejection when used in combination with cyclosporin A (CyA). "( Everolimus in liver and lung transplantation.
Koch, M, 2009
)
3.24
"Everolimus is an orally administered, targeted therapy indicated for the treatment of advanced renal cell carcinoma. "( Everolimus: in advanced renal cell carcinoma.
Garnock-Jones, KP; Keating, GM, 2009
)
3.24
"Everolimus is a rapamycin-derived macrolide with immunosuppressive and antiproliferative effects."( Severe atopic dermatitis treated with everolimus.
Bruijnzeel-Koomen, CA; Haeck, IM; Van Velsen, SG, 2009
)
1.35
"Everolimus does not seem to be an effective treatment in these two AD patients, either in combination with prednisone or with CsA."( Severe atopic dermatitis treated with everolimus.
Bruijnzeel-Koomen, CA; Haeck, IM; Van Velsen, SG, 2009
)
2.07
"Everolimus is an oral agent targeting raptor mTOR (mTORC1)."( Phase II trial of the oral mammalian target of rapamycin inhibitor everolimus in relapsed or refractory Waldenstrom macroglobulinemia.
Anderson, KC; Ansell, SM; Bergsagel, L; Camoriano, J; Chuma, S; Deangelo, D; Dispenzieri, A; Gertz, M; Ghobrial, IM; Harris, B; Hayman, S; Lacy, M; Laplant, B; Leduc, R; Reeder, C; Richardson, PG; Rourke, M; Treon, SP; Witzig, TE, 2010
)
1.32
"Everolimus (EVL) is a signal proliferation inhibition that reduces graft vascular disease when used de novo."( South American Heart Transplantation Registry of patients receiving everolimus in their immunosuppressive regimens.
Ahualli, L; Amuchástegui, M; Bortman, GV; Burgos, C; Ceruti, B; Colque, R; Diez, F; Muñoz, J; Perrone, SV; Rodriguez, MC; Sgrosso, JL; Vulcano, N,
)
1.09
"Everolimus (RAD001) is an orally administered inhibitor of the mammalian target of rapamycin (mTOR), a therapeutic target for metastatic renal cell carcinoma. "( Acute cholecystitis in a patient with metastatic renal cell carcinoma treated with everolimus.
Baykara, M; Benekli, M; Buyukberber, S; Cetin, B; Coskun, U; Yildiz, R, 2011
)
2.04
"Everolimus is an oral antineoplastic agent that targets the raptor mammalian target of rapamycin (mTORC1). "( A Phase II trial of the oral mTOR inhibitor everolimus in relapsed Hodgkin lymphoma.
Ansell, SM; Colgan, JP; Habermann, TM; Inwards, DJ; Johnston, PB; Kabat, BF; Laplant, BR; Micallef, IN; Porrata, LF; Reeder, CB; Roy, V; Witzig, TE, 2010
)
2.06
"Everolimus is a novel inhibitor of the mammalian target of rapamycin pathway, which is aberrantly activated in nonsmall cell lung cancer (NSCLC). "( Phase 1 and pharmacokinetic study of everolimus, a mammalian target of rapamycin inhibitor, in combination with docetaxel for recurrent/refractory nonsmall cell lung cancer.
Egorin, MJ; Fu, H; Harvey, RD; Kauh, J; Khuri, FR; Owonikoko, TK; Ramalingam, SS; Saba, N; Shin, DM; Strychor, S; Sun, SY; Tighiouart, M, 2010
)
2.08
"Everolimus is an oral rapamycin analog that acts by selectively inhibiting mTOR."( A phase 2 study of the oral mammalian target of rapamycin inhibitor, everolimus, in patients with recurrent endometrial carcinoma.
Broaddus, RR; Burke, TW; Coleman, RL; Gershenson, DM; Johnston, T; Lu, KH; Munsell, M; Ramondetta, LM; Slomovitz, BM; Walker, C; Wolf, J, 2010
)
1.32
"Everolimus is a recently developed immunosuppressive drug for patients following solid organ transplantation. "( Severe stomatitis complicating immune-suppressive switch after cardiac transplantation.
Conraads, V; Rodrigus, IE; Vermeulen, T; Vrints, CJ,
)
1.57
"Everolimus (RAD001) is a novel mammalian target of rapamycin (mTOR) inhibitor, and anti-proliferative activity in various malignancies has been reported. "( Schedule-dependent inhibition of T-cell lymphoma cells by cotreatment with the mTOR inhibitor everolimus and anticancer drugs.
He, XX; Huang, JJ; Huang, Y; Li, ZM; Lin, TY; Tian, Y; Xiao, J, 2012
)
2.04
"Everolimus is an oral agent that targets the raptor mammalian target of rapamycin (mTORC1)."( A phase II trial of the oral mTOR inhibitor everolimus in relapsed aggressive lymphoma.
Ansell, SM; Colgan, JP; Ghobrial, IM; Gupta, M; Habermann, TM; Jacobsen, ED; Johnston, PB; LaPlant, BR; Micallef, IN; Porrata, LF; Reeder, CB; Witzig, TE, 2011
)
1.35
"Everolimus is an oral mammalian target of rapamycin (mTOR) inhibitor approved for the management of patients with advanced renal cell carcinoma who progressed on tyrosine kinase inhibitor therapy. "( Everolimus: a new mammalian target of rapamycin inhibitor for the treatment of advanced renal cell carcinoma.
Grgic, T; Hammond, JM; Mis, L, 2011
)
3.25
"Everolimus is an immunosuppressant requiring routine monitoring in whole blood. "( Evaluation of QMS everolimus assay using Hitachi 917 Analyzer: comparison with liquid chromatography/mass spectrometry.
Chow, L; Dasgupta, A; Davis, B, 2011
)
2.15
"Everolimus is an orally administered inhibitor of the mammalian target of rapamycin that recently received approval from the European Medicines Agency for the treatment of advanced RCC that has progressed on or after treatment with vascular endothelial growth factor (VEGF)-targeted therapy, and from the United States Food and Drug Administration for treatment of advanced RCC after failure of sorafenib or sunitinib."( Management of adverse events associated with the use of everolimus in patients with advanced renal cell carcinoma.
Climent, MA; Creel, P; Dickow, B; Fischer, P; Gornell, SS; Meloni, F; Motzer, RJ; Osanto, S; Porta, C; Ravaud, A; Vaishampayan, U; White, DA, 2011
)
1.34
"Everolimus-induced OU is a frequent, recurrent and sometimes harmful complication. "( Natural history, management and pharmacokinetics of everolimus-induced-oral ulcers: insights into compliance issues.
André, F; Ferté, C; Gomez-Roca, C; Gonzales, DB; Goubar, A; Massard, C; Paci, A; Sahmoud, T; Soria, JC; Zizi, M, 2011
)
2.06
"Everolimus is a novel inhibitor of the mammalian target of rapamycin pathway, which is aberrantly activated in cancer cell. "( Phase I trial of capecitabine plus everolimus (RAD001) in patients with previously treated metastatic gastric cancer.
Ahn, HK; Baek, KK; Han, B; Kang, WK; Kim, KM; Lee, DJ; Lee, HY; Lee, J; Lee, SJ; Lim, HY; Lim, T; Park, JO; Park, SH; Park, YS, 2011
)
2.09
"Everolimus is a proliferation-signal inhibitor which was introduced for heart transplant recipients in 2004. "( Calcineurin inhibitor-free immunosuppression using everolimus (Certican) after heart transplantation: 2 years' follow-up from the University Hospital Münster.
Amler, S; Drees, G; Eckernkemper, S; Engelen, MA; Gunia, S; Rothenburger, M; Rukosujew, A; Sindermann, JR; Stypmann, J; Welp, HA, 2011
)
2.06
"Everolimus is an oral mTOR-inhibitor. "( Phase I and pharmacokinetic study of capecitabine and the oral mTOR inhibitor everolimus in patients with advanced solid malignancies.
Beijnen, JH; Deenen, MJ; Klümpen, HJ; Richel, DJ; Schellens, JH; Sparidans, RW; Weterman, MJ; Wilmink, JW, 2012
)
2.05
"Everolimus is an oral mTOR kinase inhibitor approved for the treatment of patients with metastatic renal cell carcinoma (mRCC) progressing during or after treatment with vascular endothelial growth factor (VEGF)-targeted agents. "( [Metastatic renal cell carcinoma treated with everolimus--data from the RENIS Clinical Registry].
Abrahámová, J; Bortlícek, Z; Büchler, T; Budnáková, D; Dusek, L; Fínek, J; Jarkovský, J; Kandrnal, V; Klimes, D; Poprach, A; Vyzula, R, 2011
)
2.07
"Everolimus is an orally administered inhibitor of the mammalian target of rapamycin (mTOR). "( Everolimus: in patients with subependymal giant cell astrocytoma associated with tuberous sclerosis complex.
Curran, MP, 2012
)
3.26
"Everolimus is a rapamycin analog that inhibits mTOR, but it is more soluble than rapamycin and can be administered orally."( Clinical review: Current scientific rationale for the use of somatostatin analogs and mTOR inhibitors in neuroendocrine tumor therapy.
Billah, SM; Bousquet, C; Caron, P; Chalabi, M; Lasfargues, C; Pyronnet, S; Susini, C; Vezzosi, D, 2012
)
1.1
"Everolimus (RAD001) is an mTOR inhibitor that has been successfully used as an immunosuppressant in solid-organ transplantation. "( Everolimus in combination with cyclosporin a as pre- and posttransplantation immunosuppressive therapy in nonmyeloablative allogeneic hematopoietic stem cell transplantation.
Altmann, S; Drewelow, B; Freund, M; Junghanss, C; Lange, S; Mueller, S; Rathsack, S; Vogel, H; Wacke, R; Weirich, V, 2012
)
3.26
"Everolimus (RAD001) is an oral antineoplastic agent derived from rapamycin, a macrocyclic lactone antibiotic, targeting the mammalian target of rapamycin (mTOR)."( mTOR as a target of everolimus in refractory/relapsed Hodgkin lymphoma.
Gadaleta, CD; Giannoccaro, M; Guarini, A; Iacobazzi, A; Lapietra, A; Minoia, C; Raimondi, A; Rana, A; Ranieri, G; Silvestris, N, 2012
)
1.42
"Everolimus is a mammalian target of rapamycin inhibitor approved for use as an immunosuppressant and antineoplastic agent. "( Fatal acute pulmonary injury associated with everolimus.
Benoit, D; Caes, F; Depuydt, P; Nollet, J; Praet, M, 2012
)
2.08
"Everolimus (EVL) is a mammalian target of rapamycin inhibitor, providing a safe and effective immunosuppressive regime after kidney transplantation."( Everolimus: preventing organ rejection in adult kidney transplant recipients.
Dantal, J, 2012
)
2.54
"Everolimus is a novel macrolide immunosuppressant used in the prevention of acute and chronic rejection of solid organ transplants. "( Population pharmacokinetics and pharmacogenetics of everolimus in renal transplant patients.
de Fijter, JW; den Hartigh, J; Guchelaar, HJ; Moes, DJ; Press, RR; van der Straaten, T, 2012
)
2.07
"Everolimus is an effective agent with several advantages for pediatric solid-organ transplantation. "( Everolimus in pediatric transplantation.
Ahlenstiel, T; Ganschow, R; Pape, L, 2012
)
3.26
"Everolimus is an oral mammalian target of rapamycin (mTOR) inhibitor approved by the FDA in May 2011 for the treatment of progressive, advanced pNET. "( Everolimus: a new treatment option for advanced pancreatic neuroendocrine tumors.
Kim, M; O'Bryant, CL; Thompson, LA; Wenger, SD, 2012
)
3.26
"The everolimus-eluting stent is a second-generation DES that is very effective for de novo coronary lesions."( Repeat drug-eluting stent implantation for in-stent restenosis: first- or second-generation stent.
Araki, H; Ebara, S; Hoshimoto, K; Isomura, N; Obara, C; Ochiai, M; Okabe, T; Saito, S; Yakushiji, T; Yamamoto, MH; Yamashita, K, 2012
)
0.94
"Everolimus is an effective second-line treatment for Taiwanese patients with mRCC. "( Everolimus in metastatic renal cell carcinoma: preliminary experience from Chang Gung Memorial Hospital.
Chang, JW; Chang, YH; Chiang, YJ; Chuang, CK; Hsieh, JJ; Huang, WK; Liaw, CC; Lin, YC; Ou, LY; Pang, ST; Tsai, SI; Wang, HM; Wu, CT; Yang, CH; Yang, CT,
)
3.02
"Everolimus is an immunosuppressant that blocks growth factor-mediated proliferation of hematopoietic cells by targeting the mammalian target of rapamycin (mTOR). "( Interactions of everolimus and sorafenib in whole blood lymphocyte proliferation.
Fetterly, GJ; Jusko, WJ; Ma, WW; Pawaskar, DK; Straubinger, RM, 2013
)
2.18
"Everolimus (RAD001) is an allosteric mTOR inhibitor that has shown marked efficacy in certain cancers but is unable to completely inhibit mTOR activity."( RAD001 enhances the potency of BEZ235 to inhibit mTOR signaling and tumor growth.
Cao, ZA; Chen, Y; Das, R; Finan, PM; Huang, A; Lehár, J; Li, X; Murphy, LO; Nyfeler, B; Pinzon-Ortiz, M; Pradhan, E; Reddy, A; Schlegel, R; Wang, Z, 2012
)
1.1
"Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved for treatment of renal cell carcinoma, subependymal giant cell astrocytoma, breast cancer, and progressive neuroendocrine tumors of pancreatic origin. "( Rash to the mTOR inhibitor everolimus: systematic review and meta-analysis.
Case, EC; Cerci, FB; Lacouture, ME; Ramirez-Fort, MK; Rosen, AC; Wu, S, 2014
)
2.14
"Everolimus is a derivative of sirolimus, and is considered to be free of the latter's pulmonary toxicity. "( Early everolimus-induced pneumonitis in a renal transplant recipient: A case report.
Durlik, M; Gołębiowski, M; Małkowski, P; Miszewska-Szyszkowska, D; Palczewski, P; Sułkowska, K, 2012
)
2.3
"Everolimus is an mTOR inhibitor commonly used to treat metastatic pancreatic neuroendocrine tumors (pNETs) and renal cell carcinoma, and for posttransplant immunosuppression. "( Everolimus causing severe hypertriglyceridemia and acute pancreatitis.
Kunz, PL; Subramaniam, S; Zell, JA, 2013
)
3.28
"Everolimus appears to be a new effective treatment for patients with metastatic insulinoma and refractory hypoglycemia. "( Efficacy of everolimus in patients with metastatic insulinoma and refractory hypoglycemia.
Baudin, E; Bernard, V; Borson-Chazot, F; Caroli-Bosc, FX; Chougnet, CN; Goichot, B; Guimbaud, R; Lombard-Bohas, C; Niccoli, P; Rohmer, V; Ruszniewski, P; Taquet, MC, 2013
)
2.21
"Everolimus is an immunosuppressive macrolide bearing a stable 2-hydroxyethyl chain substitution at position 40 on the sirolimus (rapamycin) structure. "( Clinical pharmacokinetics of everolimus.
Kirchner, GI; Manns, MP; Meier-Wiedenbach, I, 2004
)
2.06
"Everolimus is a derivative of sirolimus, a macrocyclic lactone, originally isolated from Streptomyces hygroscopicus. "( The evolving experience using everolimus in clinical transplantation.
Bia, MJ; Formica, RN; Friedman, AL; Lakkis, F; Lorber, KM; Lorber, MI; Smith, JD, 2004
)
2.05
"Everolimus is a novel macrolide immunosuppressant that acts as a T-lymphocyte proliferation signal inhibitor. "( Experience with everolimus.
Augustine, JJ; Hricik, DE, 2004
)
2.11
"Everolimus is a potent immunosuppressor that inhibits growth factor-stimulated proliferation of hematopoietic and nonhematopoietic cells, including vascular smooth muscle."( Everolimus: a proliferation signal inhibitor targeting primary causes of allograft dysfunction.
Kovarik, JM, 2004
)
2.49
"Everolimus is an immunosuppressant that blocks growth factor-mediated proliferation of haematopoietic and nonhaematopoietic cells. "( Everolimus.
Chapman, TM; Perry, CM, 2004
)
3.21
"Everolimus is a proliferation inhibitor designed to target chronic rejection, including prevention of acute rejection. "( Everolimus (Certican) 12-month safety and efficacy versus mycophenolate mofetil in de novo renal transplant recipients.
Cretin, N; Dantal, J; Jappe, A; Li, Y; Margreiter, R; Viljoen, HG; Vítko, S; Weimar, W, 2004
)
3.21
"Everolimus (Certican) is a novel proliferation signal inhibitor with potent immunosuppressant effects that has been shown to prevent and reverse acute rejection in preclinical models of kidney, heart, and lung transplantation. "( Introducing everolimus (Certican) in organ transplantation: an overview of preclinical and early clinical developments.
Neumayer, HH, 2005
)
2.15
"Everolimus is a novel proliferation signal inhibitor used in immunosuppressive therapies for the prevention of acute and chronic rejection. "( The role of therapeutic monitoring of everolimus in solid organ transplantation.
Ensom, MH; Mabasa, VH, 2005
)
2.04
"Everolimus is a proliferation signal inhibitor with immunosuppressive activity that may reduce the rate of progression of chronic rejection, bronchiolitis obliterans syndrome (BOS), after lung transplantation. "( Everolimus versus azathioprine in maintenance lung transplant recipients: an international, randomized, double-blind clinical trial.
Aris, R; Glanville, AR; Hmissi, A; Loyd, JE; McGiffin, DC; Pirron, U; Roman, A; Snell, GI; Sole, A; Valentine, VG; Vitulo, P, 2006
)
3.22
"Everolimus (Certican) is an orally administered mammalian target of rapamycin inhibitor (proliferation signal inhibitor) derived from sirolimus (rapamycin), which is used as part of immunosuppressant therapy in kidney and heart transplantation. "( Everolimus: a review of its use in renal and cardiac transplantation.
Croom, KF; Dunn, C, 2006
)
3.22
"Everolimus is a macrolide immunosuppressive agent with known consistent absorption. "( Safety, tolerability, and efficacy of everolimus in de novo liver transplant recipients: 12- and 36-month results.
Abecassis, M; Calmus, Y; Fischer, L; Freeman, R; Klempnauer, J; Langnas, A; Levy, G; Mayer, D; Nashan, B; Neuhaus, P; Punch, J; Roberts, J; Rogiers, X; Samuel, D; Schmidli, H; Sloof, M; Tuttle-Newhall, E, 2006
)
2.05
"Everolimus (RAD) is an mTOR inhibitor closely related to rapamycin. "( Everolimus (RAD) inhibits in vivo growth of murine squamous cell carcinoma (SCC VII).
Khariwala, SS; Kjaergaard, J; Lorenz, R; Shu, S; Strome, M; Van Lente, F, 2006
)
3.22
"Everolimus is a novel immunosuppressive agent related to sirolimus. "( Everolimus: an immunosuppressive agent in transplantation.
Kobashigawa, JA; Patel, JK, 2006
)
3.22
"Everolimus is a proliferation signal inhibitor (PSI)/mammalian target of rapamycin inhibitor that is structurally similar to sirolimus, but with a number of important pharmacokinetic differences, including a shorter half-life and time to steady state. "( Everolimus in clinical practice--renal transplantation.
Pascual, J, 2006
)
3.22
"Everolimus is a potent immunosuppressive agent that has anti-proliferative activity. "( Long-term cardiovascular risk in transplantation--insights from the use of everolimus in heart transplantation.
Eisen, H, 2006
)
2.01
"Everolimus is a potent immunosuppressant used in renal transplant therapy, but its effects on renal endothelial cell regeneration after injury are unknown. "( Everolimus inhibits glomerular endothelial cell proliferation and VEGF, but not long-term recovery in experimental thrombotic microangiopathy.
Daniel, C; Endlich, KH; Hugo, C; Johnson, RJ; Keller, K; Kerjaschki, D; Schöcklmann, H, 2006
)
3.22
"Everolimus is an orally active derivative of sirolimus. "( Optimal dosing and duration of oral everolimus to inhibit in-stent neointimal growth in rabbit iliac arteries.
Baffour, R; Hellinga, D; Kolodgie, F; Pakala, R; Seabron, R; Virmani, R; Waksman, R,
)
1.85
"Everolimus (Certican) is a new immunosuppressant derived from sirolimus (Rapamune) with a 2-hydroxyethyl chain at position 40 of the macrolide ring. "( Determination of everolimus in whole blood using the Abbott IMx sirolimus microparticle enzyme immunoassay.
Bouzas, L; Tutor, JC, 2007
)
2.12
"Everolimus is a proliferation signal-inhibitor recently introduced in heart transplant recipients. "( Calcineurin inhibitor-free immunosuppression using everolimus (Certican) in maintenance heart transplant recipients: 6 months' follow-up.
Bara, C; Bruch, C; Hinder, F; Lehmkuhl, H; Rothenburger, M; Schmid, C; Stypmann, J; Suwelack, B; Teerling, E; Wichter, T, 2007
)
2.03
"Everolimus is a novel macrolide immunosuppressant developed for the prophylaxis of allogeneic renal or cardiac transplant rejection. "( Physiologically based pharmacokinetic (PBPK) modeling of everolimus (RAD001) in rats involving non-linear tissue uptake.
Kawai, R; Laplanche, R; Meno-Tetang, GM, 2007
)
2.03
"Everolimus is an immunosuppressive agent that reduces cardiac allograft vasculopathy. "( Prevention of acute rejection and allograft vasculopathy by everolimus in cardiac transplants recipients: a 24-month analysis.
Abeywickrama, K; Benza, R; Boissonnat, P; Cretin, N; Eisen, HJ; Haverich, A; Hill, J; Laufer, G; Love, R; Parameshwar, J; Pulpón, LA; Renlund, D; Starling, RC; Tuzcu, M; Viganò, M, 2007
)
2.02
"Everolimus (RAD001) is an inhibitor of the mammalian target of rapamycin, which is downstream of initial epidermal growth factor receptor signaling."( Rationale for a phase I trial of erlotinib and the mammalian target of rapamycin inhibitor everolimus (RAD001) for patients with relapsed non small cell lung cancer.
Jackman, D; Jänne, PA; Johnson, BE, 2007
)
1.28
"Everolimus is an mTOR inhibitor sirolimus analogue, that has proved, to be highly efficacious to prevent acute myocardial rejection and reduce the severity of cardiac allograft vasculopathy in de novo HTx patients."( Possible role of everolimus in improving renal function in long-term heart transplantation.
Fiocchi, R; Fontana, A; Gamba, A; Gandolfi, L; Iacovoni, A; Sebastiani, R,
)
1.19
"Everolimus is a new immunosuppressant with antitumoral properties and few side effects, but limited use in liver transplantation. "( Use of everolimus as a rescue immunosuppressive therapy in liver transplant patients with neoplasms.
Bañares, R; Clemente, G; Gomez-Camarero, J; Hernando, A; Lo Iacono, O; Rincon, D; Ripoll, C; Salcedo, M; Sanz, C, 2007
)
2.24
"Everolimus is an immunosuppressant used as rejection prophylaxis in patients undergoing transplants. "( Crossreactivity of isolated everolimus metabolites with the Innofluor Certican immunoassay for therapeutic drug monitoring of everolimus.
Arabshahi, L; Boyd, J; Christians, U; Haschke, M; Marbach, P; Roberts, M; Strom, T, 2007
)
2.08
"Everolimus is a selective mammalian target of rapamycin (mTOR) inhibitor with promising anticancer activity. "( Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors.
Baselga, J; Burris, H; Calvo, E; Dimitrijevic, S; Hazell, K; Jones, S; Judson, I; Lane, HA; Lebwohl, D; Macarulla, T; Martinelli, E; Ramon y Cajal, S; Ramos, FJ; Rojo, F; Shand, N; Stumm, M; Tabernero, J; Tang, P; Vidal, L; Zoellner, U, 2008
)
2.01
"Everolimus is a macrolide immunosuppressant intended for acute rejection prophylaxis after kidney transplantation."( Population pharmacokinetics of everolimus in de novo renal transplant patients: impact of ethnicity and comedications.
Berthier, S; Hsu, CH; Kovarik, JM; McMahon, L; Rordorf, C, 2001
)
2.04
"Everolimus is an immunosuppressant intended for use with cyclosporine in acute-rejection prophylaxis following organ transplantation. "( Differential influence of two cyclosporine formulations on everolimus pharmacokinetics: a clinically relevant pharmacokinetic interaction.
Figueiredo, J; Frazier, OL; Kalbag, J; Kovarik, JM; Rordorf, C; Rouilly, M, 2002
)
2

Effects

Everolimus has a high single-agent activity of 73% including MR, with a progression free survival of 21 months, indicating that this agent is active in relapsed/refractory WM. The drug has a significant impact on red blood cell parameters in patients with TSC.

Everolimus (EVR) has been approved for the treatment of various advanced cancers and its indications are increasingly expanding. Everolimus has been shown to be effective for advanced pancreatic neuroendocrine tumours (pNETs)

ExcerptReferenceRelevance
"Everolimus has a significant impact on red blood cell parameters in patients with TSC."( New Insights into Red Blood Cell Microcytosis upon mTOR Inhibitor Administration.
Jakubowska, J; Jóźwiak, S; Kotulska, K; Młynarski, W; Pawlik, B; Stolarska, M; Trelińska, J; Wyka, K, 2021
)
1.34
"Everolimus has a high single-agent activity of 73% including MR, with a progression free survival of 21 months, indicating that this agent is active in relapsed/refractory WM."( Long-term results of the phase II trial of the oral mTOR inhibitor everolimus (RAD001) in relapsed or refractory Waldenstrom Macroglobulinemia.
Bergsagel, PL; Camoriano, J; Chuma, S; DeAngelo, D; Gertz, M; Ghobrial, IM; Hayman, S; Lacy, M; LaPlant, B; Treon, SP; Witzig, TE, 2014
)
1.36
"Everolimus has a favorable pharmacokinetic (PK) profile in early breast cancer studies."( Evaluating the pharmacokinetics and pharmacodynamics of everolimus for treating breast cancer.
Awada, A; Barthélémy, P; Gombos, A, 2015
)
1.38
"Everolimus has a mechanism of action similar to that of sirolimus."( Clinical pharmacokinetics of everolimus.
Kirchner, GI; Manns, MP; Meier-Wiedenbach, I, 2004
)
1.34
"Everolimus has an appropriate rationale for therapeutic use in combination with erlotinib for patients with NSCLC. "( Rationale for a phase I trial of erlotinib and the mammalian target of rapamycin inhibitor everolimus (RAD001) for patients with relapsed non small cell lung cancer.
Jackman, D; Jänne, PA; Johnson, BE, 2007
)
2
"Everolimus has been replaced in the second line setting by these new options."( [Interdisciplinary recommendations for the treatment of advanced renal cell carcinoma].
Bergmann, L; Doehn, C; Grünwald, V; Gschwend, JE; Ivanyi, P; Kuczyk, MA; Miller, K, 2022
)
1.44
"Everolimus (EVR) has been reported to be widely used to prevent the progression of organ fibrosis and graft rejection."( Everolimus Alleviates Renal Allograft Interstitial Fibrosis by Inhibiting Epithelial-to-Mesenchymal Transition Not Only
Chen, H; Fei, S; Gu, M; Gui, Z; Han, Z; Ju, X; Sun, L; Suo, C; Tan, R; Tao, J; Wang, Z; Zhang, H; Zheng, M, 2021
)
2.79
"Everolimus has been continued for 6 years without severe side effects."( A mechanistic target of rapamycin inhibitor, everolimus safely ameliorated lupus nephritis in a patient complicated with tuberous sclerosis.
Katada, Y; Ohshima, S; Okita, Y; Saeki, Y; Yoshimura, M, 2023
)
1.89
"Everolimus has recently been used to prevent graft rejection in liver transplantation and reduces the incidence of kidney dysfunction caused by calcineurin inhibitors. "( Population pharmacokinetics of everolimus in adult liver transplant patients: Comparison to tacrolimus disposition and extrapolation to pediatrics.
Hata, K; Hatano, E; Hira, D; Hirai, M; Imai, S; Ito, T; Itohara, K; Matsubara, K; Nakagawa, S; Nakagawa, T; Sugimoto, M; Terada, T; Yano, I; Yonezawa, A, 2022
)
2.45
"Everolimus (EVE) has been used as a calcineurin inhibitor (CNI) minimization/ elimination agent or to augment immunosuppression in lung transplant recipients (LTR) with CNI-induced nephrotoxicity or neurotoxicity. "( Rescue Everolimus Post Lung Transplantation is Not Associated With an Increased Incidence of CLAD or CLAD-Related Mortality.
Dooley, M; Ivulich, S; Kirkpatrick, C; Paraskeva, M; Paul, E; Snell, G, 2023
)
2.81
"Everolimus has shown positive effects on subependymal giant cell astrocytomas, renal angiomyolipoma and refractory seizures associated with tuberous sclerosis complex."( Everolimus for cardiac rhabdomyomas in children with tuberous sclerosis. The ORACLE study protocol (everOlimus for caRdiac rhAbdomyomas in tuberous sCLErosis): a randomised, multicentre, placebo-controlled, double-blind phase II trial.
Bonanato, L; Camacho-Vilchez, J; Da Silva, MVCP; Danos, P; de Abrantes, FF; de Sousa, CF; Diebold, M; Farina, A; Galvani, MAG; Gresse, S; Gutierrez, A; Idris, OEA; Iqbal, F; Jimenez-Pavón, J; Loss, KL; Mahmoud, RAA; Maki, D; Manterola, C; Morón, EM; Nashwan, MWJ; Ortiz-Lopez, R; Parente, DB; Polania, JP; Raquelo-Menegassio, AF; Stein, A; Stelmaszewski, EV; Thompson, L; Victor, C, 2020
)
2.72
"Everolimus (EVR) has been approved for the treatment of various advanced cancers and its indications are increasingly expanding. "( Designing orodispersible films containing everolimus for enhanced compliance and bioavailability.
Gao, S; Guan, R; Liu, H; Liu, Y; Ma, Y; Song, W; Yang, Y, 2020
)
2.27
"Everolimus has recently been approved by the US Food and Drug Administration and the European Medical Agency for the treatment of refractory seizures associated with TSC starting from the age of 2 years."( Genetic pathogenesis of the epileptogenic lesions in Tuberous Sclerosis Complex: Therapeutic targeting of the mTOR pathway.
Aronica, E; Craiu, D; Curatolo, P; Luinenburg, MJ; Moavero, R; Mühlebner, A, 2022
)
1.44
"Everolimus has demonstrated effectiveness in neuroendocrine neoplasms."( A pilot study of everolimus and radiation for neuroendocrine liver metastases.
Chan, DL; Chen, H; Chung, H; Hallet, J; Hudson, J; Law, C; Milot, L; Myrehaug, S; Patel, C; Rodriguez-Freixinos, V; Singh, S, 2021
)
1.68
"Everolimus has a significant impact on red blood cell parameters in patients with TSC."( New Insights into Red Blood Cell Microcytosis upon mTOR Inhibitor Administration.
Jakubowska, J; Jóźwiak, S; Kotulska, K; Młynarski, W; Pawlik, B; Stolarska, M; Trelińska, J; Wyka, K, 2021
)
1.34
"Everolimus has been shown to be effective for advanced pancreatic neuroendocrine tumours (pNETs), but its positioning in the therapeutic algorithm for pNETs is matter of debate."( Everolimus as first line therapy for pancreatic neuroendocrine tumours: current knowledge and future perspectives.
Colao, A; Faggiano, A; Fanciulli, G; Gallo, M; Malandrino, P; Rota, F, 2017
)
3.34
"Everolimus has single-agent activity in lymphoma."( Adjuvant everolimus in high-risk diffuse large B-cell lymphoma: final results from the PILLAR-2 randomized phase III trial.
Belada, D; Bermudez Silva, CD; Bouabdallah, K; Cavalli, F; Costa, LJ; Fan, J; Fayad, L; Hino, M; Ikeda, T; Kattan, JG; Kim, WS; Kuruvilla, J; Larouche, JF; Louveau, AL; Mayer, J; Ogura, M; Ozcan, M; Rigacci, L; Shi, Y; Tobinai, K; Vanazzi, A; Voi, M; Witzig, TE; Wu, C; Zhu, J, 2018
)
1.62
"Everolimus has proven to be efficacious in size reduction of cardiac rhabdomyomas in cases when surgical resection is not possible."( Giant left ventricular rhabdomyoma treated successfully with everolimus: case report and review of literature.
Aguinaga-Ríos, M; Cardona-Pérez, JA; Cordero-González, G; Coronado-Zarco, A; Escamilla-Sánchez, KI; Martínez-García, A; Michel-Macías, C, 2018
)
2.16
"Everolimus has been shown to overcome endocrine resistance in hormone receptor positive advanced breast cancer patients. "( Mutational Profile of Metastatic Breast Cancer Tissue in Patients Treated with Exemestane Plus Everolimus.
Barbolini, M; Bettelli, S; Caprera, C; Cascinu, S; Conte, PF; Filieri, ME; Guaitoli, G; Kaleci, S; Maiorana, A; Manfredini, S; Moscetti, L; Nasso, C; Omarini, C; Piacentini, F, 2018
)
2.14
"Everolimus has been shown to be effective in overcoming hormonal resistance in Lebanese breast cancer patients with results inferior to those reported in the BOLERO-2 population. "( Efficacy and safety of everolimus in hormone receptor positive breast cancer in a developing country: Real-life single institutional experience.
Assi, T; Chebib, R; El Karak, F; El Rassy, E; Farhat, F; Ghosn, M; Hanna, C; Kattan, J; Moussa, T; Tabchi, S,
)
1.88
"Everolimus has significant clinical benefit and is well tolerated with reversible side effects as second- or third-line therapy for treating RCC. "( Evaluation of everolimus in renal cell cancer.
Amato, R; Stepankiw, M, 2013
)
2.19
"Everolimus conversion has become an excellent choice, offering safety and efficacy with good outcomes."( Clinical everolimus experience in pediatric renal transplant patients.
Bulut, IK; Dincel, N; Hoşcoşkun, C; Mir, S; Sezer, TÖ, 2013
)
2.25
"Everolimus has been well tolerated by all."( Successful everolimus therapy for SEGA in pediatric patients with tuberous sclerosis complex.
Adolfo, F; Cappellano, AM; Cavalheiro, S; Covic, A; Paiva, PM; Pinho, R; Saba, N; Senerchia, AA, 2013
)
1.5
"Everolimus has effective inhibition on aromatase-overexpressing stem cell in vitro and in vivo."( Everolimus in combination with letrozole inhibit human breast cancer MCF-7/Aro stem cells via PI3K/mTOR pathway: an experimental study.
Hou, G; Liu, J; Liu, Y; Zhang, J; Zhang, S; Zhang, X, 2014
)
2.57
"Everolimus has been recently approved as a pharmacotherapy option for TSC patients with subependymal giant-cell astrocytomas (SEGAs) or renal angiomyolipomas (AMLs)."( Is mTOR inhibition a systemic treatment for tuberous sclerosis?
Coniglio, A; Curatolo, P; Garaci, F; Moavero, R, 2013
)
1.11
"Everolimus has shown to stop formation and activity of osteoclasts. "( Everolimus as treatment for breast cancer patients with bone metastases only: results of the phase II RADAR study.
Aktas, B; Barinoff, J; Bauerschlag, D; Bischoff, J; Ettl, J; Harbeck, N; Kümmel, S; Lerchenmüller, C; Loibl, S; Lübbe, K; Lück, HJ; Maass, N; Müller, L; Mundhenke, C; Nekljudova, V; Rösel, S; Schmidt, M; Schwedler, K; von Minckwitz, G; Wagner, S, 2013
)
3.28
"Everolimus has recently been approved by Food and Drug Administration for graft maintenance in liver transplant recipients. "( Analytical performance of QMS everolimus assay on ortho Vitros 5,1 FS fusion analyzer: measuring everolimus trough levels for solid organ transplant recipients.
Bernard, DW; Chandler, WL; Shu, I; Wang, P; Wright, AM, 2014
)
2.13
"Everolimus (EVR) has demonstrated good efficacy after renal transplantation. "( Outcomes in ethnic minority renal transplant recipients receiving everolimus versus mycophenolate: comparative risk assessment results from a pooled analysis.
Delcoro, C; McCague, K; Melancon, K; Mulgaonkar, SP; Shihab, FS; Wiland, A, 2013
)
2.07
"Everolimus has demonstrated efficacy in unresectable and progressive pancreatic neuroendocrine tumors, by doubling the median progression free survival (11 versus 4.6 months), with a median time of exposure to everolimus of nine months."( [Management of metabolic disorders induced by everolimus in patients with differentiated neuroendocrine tumors: expert proposals].
Cariou, B; Coriat, R; François, E; Hammel, P; Hentic, O; Lombard-Bohas, C; N'guyen, T; Niccoli, P; Vergès, B, 2014
)
1.38
"Everolimus has important clinical activity in various malignancies, but its use can be complicated by respiratory adverse events. "( Diagnostic challenges of respiratory adverse events during everolimus treatment.
de Boer, M; De Vos, FY; Jansen, A; Tjan-Heijnen, VC; van Herpen, CM; Willemsen, AE, 2014
)
2.09
"Everolimus has been shown to reduce the severity and incidence of CAV as measured by first year intravascular ultrasound (IVUS)."( Cardiac allograft vasculopathy by intravascular ultrasound in heart transplant patients: substudy from the Everolimus versus mycophenolate mofetil randomized, multicenter trial.
Cantin, B; Dong, G; Eisen, H; Hill, JA; Kobashigawa, JA; Lopez, P; Nicholls, SJ; Pauly, DF; Ross, H; Starling, RC; Wang, SS, 2013
)
1.32
"Everolimus (EVR) has inter-individual pharmacokinetic (PK) variability and a narrow therapeutic index. "( The impact of genetic polymorphisms, diltiazem, and demographic variables on everolimus trough concentrations in lung transplant recipients.
Aquilante, CL; Fish, DN; Kiser, TH; Schoeppler, KE; Zamora, MR, 2014
)
2.07
"Everolimus has demonstrated substantial clinical benefit in randomized, controlled, phase III studies leading to approval for the treatment of advanced renal cell carcinoma, advanced neuroendocrine tumors of pancreatic origin, renal angiomyolipoma and subependymal giant-cell astrocytoma associated with tuberous sclerosis complex, as well as advanced hormone-receptor-positive (HR(+)) and human epidermal growth factor receptor-2-negative advanced breast cancer."( Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer.
Aapro, M; Andre, F; Blackwell, K; Calvo, E; Jahanzeb, M; Papazisis, K; Porta, C; Pritchard, K; Ravaud, A, 2014
)
1.36
"Everolimus has been approved for the treatment of pancreatic neuroendocrine tumors (pNETs), metastatic renal cell carcinoma (mRCC), and breast carcinoma."( Everolimus in advanced solid tumors: when to start, early or late?
Procopio, G; Pusceddu, S; Tessari, A; Testa, I,
)
2.3
"Everolimus has demonstrated its efficacy in metastatic renal cell carcinoma (mRCC). "( Impact of everolimus blood concentration on its anti-cancer activity in patients with metastatic renal cell carcinoma.
Almotlak, H; Foubert, A; Maurina, T; Montange, D; Montcuquet, P; Mouillet, G; Nerich, V; Nguyen Tan Hon, T; Pivot, X; Royer, B; Stein, U; Thiery-Vuillemin, A, 2014
)
2.25
"Everolimus has a high single-agent activity of 73% including MR, with a progression free survival of 21 months, indicating that this agent is active in relapsed/refractory WM."( Long-term results of the phase II trial of the oral mTOR inhibitor everolimus (RAD001) in relapsed or refractory Waldenstrom Macroglobulinemia.
Bergsagel, PL; Camoriano, J; Chuma, S; DeAngelo, D; Gertz, M; Ghobrial, IM; Hayman, S; Lacy, M; LaPlant, B; Treon, SP; Witzig, TE, 2014
)
1.36
"Everolimus has been approved by the FDA and the EMA for the treatment of advanced renal cell carcinoma (RCC), subependymal giant cell astrocytoma (SEGA) associated with tuberous sclerosis (TSC), pancreatic neuroendocrine tumors (PNET), in combination with exemestane in advanced hormone-receptor (HR)-positive, HER2-negative breast cancer."( Everolimus.
Hasskarl, J, 2014
)
2.57
"Both everolimus and axitinib have been considered standard of care after failure of first-line VEGF-TKI; sorafenib has been proposed as an additional option."( Improving outcomes in metastatic clear cell renal cell carcinoma by sequencing therapy.
Schmidinger, M, 2014
)
0.86
"Everolimus has promise as an effective alternative to surgery for subependymal giant cell astrocytomas during early infancy."( Everolimus Treatment for an Early Infantile Subependymal Giant Cell Astrocytoma With Tuberous Sclerosis Complex.
Fukumura, S; Minagawa, K; Takayama, R; Tsutsumi, H; Watanabe, T, 2015
)
2.58
"Everolimus has limited clinical activity in advanced MPM patients. "( SWOG S0722: phase II study of mTOR inhibitor everolimus (RAD001) in advanced malignant pleural mesothelioma (MPM).
Gandara, DR; Garland, LL; Mack, PC; Moon, J; Ou, SH; Testa, JR; Tsao, AS; Wozniak, AJ, 2015
)
2.12
"Everolimus has a favorable pharmacokinetic (PK) profile in early breast cancer studies."( Evaluating the pharmacokinetics and pharmacodynamics of everolimus for treating breast cancer.
Awada, A; Barthélémy, P; Gombos, A, 2015
)
1.38
"Everolimus has also beenrecently studied in HER2 positive locally advanced or metastatic disease in heavily pretreated patients (BOLERO 3 trial)."( [The drug of the month: everolimus (Afinitor) for the treatment of metastatic breast cancer].
Collignon, J; Jerusalem, G; Rorive, A, 2014
)
1.43
"Everolimus has been shown to be an effective HR+/HER2- mBC treatment in both clinical trials and real-world practice. "( Everolimus use and associated factors among post-menopausal women with hormonal receptor positive/human epidermal growth factor receptor 2 negative metastatic breast cancer.
Hao, Y; Li, N; Lin, PL; Signorovitch, JE; Wu, EQ; Xie, J; Zhong, Y; Zhou, Z, 2015
)
3.3
"Everolimus (ERL) has become an alternative to calcineurin inhibitors (CNIs) due to its renal-sparing properties, especially in heart transplant (HTx) recipients with kidney dysfunction. "( Influence of CYP3A5 genetic variation on everolimus maintenance dosing after cardiac transplantation.
Englberger, L; Fiedler, GM; Largiadèr, CR; Lesche, D; Mohacsi, P; Setoud, R; Sigurdardottir, V; Sistonen, J, 2015
)
2.13
"Everolimus has no nephrotoxicity and is used to treat patients with post-liver transplant chronic renal insufficiency. "( Everolimus in de novo liver transplant recipients: a systematic review.
Deng, HJ; Li, QD; Liu, R; Shen, A; Tang, CY; Wei, XF; Wu, YZ; Wu, ZJ, 2015
)
3.3
"Everolimus has been used in patients with hormone receptor-positive breast cancer. "( Treatment with everolimus for a patient with systemic metastatic breast cancer results in severe pulmonary injury: a case report.
Santai, S; Shikai, W; Tao, W; Xiangying, M; Zefei, J, 2017
)
2.25
"Everolimus has been used widely in cancer patients and is associated with the development of hyperglycemia. "( Risk of hyperglycemia attributable to everolimus in cancer patients: A meta-analysis.
Shameem, R; Wu, S; Xu, KY,
)
1.85
"Everolimus (EVR) has been used widely for the purpose of reducing the dosage of calcineurin inhibitor (CNI), leading to decreasing CNI nephrotoxicity. "( Effectiveness of the Combination of Everolimus and Tacrolimus With High Dosage of Mizoribine for Living Donor-Related Kidney Transplantation.
Harada, S; Ito, T; Koshino, K; Nakamura, T; Nakao, T; Nobori, S; Suzuki, T; Ushigome, H; Yoshimura, N, 2016
)
2.15
"Everolimus has activity in relapsed DLBCL."( Everolimus combined with R-CHOP-21 for new, untreated, diffuse large B-cell lymphoma (NCCTG 1085 [Alliance]): safety and efficacy results of a phase 1 and feasibility trial.
Ansell, SM; Colgan, JP; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, B; McPhail, E; Micallef, IN; Nowakowski, GS; Witzig, TE, 2016
)
2.6
"Everolimus has been shown to have a manageable safety profile, with the most common adverse events being stomatitis, rash, diarrhea, fatigue and infections."( Everolimus in the treatment of neuroendocrine tumors of the respiratory and gastroenteropancreatic systems.
Flaum, N; Mansoor, W; McNamara, MG; Valle, JW, 2016
)
2.6
"Everolimus has been used for the treatment of unresectable or metastatic renal cell carcinoma (RCC). "( Population Pharmacokinetics of Everolimus in Relation to Clinical Outcomes in Patients With Advanced Renal Cell Carcinoma.
Fukudo, M; Kamba, T; Masuda, S; Matsubara, K; Ogawa, O; Sato, E; Shinsako, K; Tanaka, A; Yamasaki, T; Yano, I, 2016
)
2.16
"Everolimus has clinically relevant antitumor activity in patients with advanced differentiated thyroid cancer. "( Everolimus in Patients With Advanced Follicular-Derived Thyroid Cancer: Results of a Phase II Clinical Trial.
Bos, M; Corver, WE; de Wit, D; Gelderblom, H; Guchelaar, HJ; Kapiteijn, E; Links, TP; Morreau, H; Roozen, IC; Schneider, TC; Smit, JW; van der Hoeven, JJ; van Erp, NP; van Wezel, T, 2017
)
3.34
"Everolimus has beneficial effects on UF failure, inflammation, and fibrosis."( Effects of everolimus as an antiproliferative agent on regression of encapsulating peritoneal sclerosis in a rat model.
Bozkurt, D; Duman, S; Ertilav, M; Ok, E; Ozkan, S; Sen, S; Sezak, M; Sipahi, S, 2008
)
1.46
"Everolimus has been prescribed both for initial and maintenance therapy after cardiac transplantation. "( Preliminary experience with conversion from calcineurin inhibitors to everolimus in cardiac transplantation maintenance therapy.
Arizón-Muñoz, JM; Benezet-Mazuecos, J; Guisado, A; Jiménez-Díaz, J; Lage-Gallé, E; Martínez, A; Mogollón, MV; Romero-Rodriguez, N; Sánchez-Brotons, JA; Sobrino-Márquez, JM, 2008
)
2.02
"Oral everolimus has activity in metastatic breast cancer that is schedule dependent. "( Randomized phase II study comparing two schedules of everolimus in patients with recurrent/metastatic breast cancer: NCIC Clinical Trials Group IND.163.
Chia, S; Clemons, M; Eisen, A; Ellard, SL; Gelmon, KA; Huntsman, D; Kennecke, H; McIntosh, L; Mishaeli, M; Norris, B; Olivo, M; Pritchard, K; Sauerbrei, E; Seymour, L; Taylor, M; Vandenberg, T; Walsh, W, 2009
)
1.12
"Everolimus has modest antitumor activity against CLL and can mobilize malignant cells from nodal masses into the peripheral circulation in a subset of CLL patients. "( The treatment of recurrent/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) with everolimus results in clinical responses and mobilization of CLL cells into the circulation.
Call, TG; Habermann, TM; Johnston, PB; LaPlant, BR; Micallef, IN; Witzig, TE; Zent, CS, 2010
)
2.02
"Everolimus has shown good results in kidney and heart transplantation, achieving low rates of rejection, of infections, and of tumors compared with calcineurin inhibitors (CNI). "( Everolimus in lung transplantation in Chile.
Alba, A; Parada, MT; Sepúlveda, C,
)
3.02
"Everolimus has single-agent activity in relapsed/refractory HL and provides proof-of-concept that targeting the mTOR pathway in HL is clinically relevant."( A Phase II trial of the oral mTOR inhibitor everolimus in relapsed Hodgkin lymphoma.
Ansell, SM; Colgan, JP; Habermann, TM; Inwards, DJ; Johnston, PB; Kabat, BF; Laplant, BR; Micallef, IN; Porrata, LF; Reeder, CB; Roy, V; Witzig, TE, 2010
)
1.34
"Everolimus has shown in a randomized phase III trial for patients with metastatic renal cell carcinoma having failed under VEGFR tyrosine kinase inhibitor a significant gain in progression-free survival compared to this obtained with placebo VEGFR (1,8 à 4,6 months; HR: 0.33; P < 0.001)."( [mTOR inhibitors: temsirolimus and everolimus in the treatment of renal cell carcinoma].
Bernhard, JC; Digue, L; Ferriere, JM; Gross-Goupil, M; Ravaud, A, 2010
)
1.36
"Everolimus has single-agent activity in relapsed/refractory aggressive NHL and provides proof-of-concept that targeting the mTOR pathway is clinically relevant."( A phase II trial of the oral mTOR inhibitor everolimus in relapsed aggressive lymphoma.
Ansell, SM; Colgan, JP; Ghobrial, IM; Gupta, M; Habermann, TM; Jacobsen, ED; Johnston, PB; LaPlant, BR; Micallef, IN; Porrata, LF; Reeder, CB; Witzig, TE, 2011
)
1.35
"Everolimus has proven efficacy for prevention of rejection in adult de novo renal and cardiac transplant recipient in combination with ciclosporine and corticosteroids. "( [Level of evidence for therapeutic drug monitoring of everolimus].
Goirand, F; Hulin, A; Royer, B; Saint-Marcoux, F,
)
1.82
"Everolimus and sunitinib have been recently introduced as targeted therapies for metastatic pancreatic neuroendocrine tumours."( New therapeutic options for metastatic malignant insulinomas.
de Herder, WW; Feelders, RA; Kwekkeboom, D; van Schaik, E, 2011
)
1.09
"Everolimus has been successfully used in solid organ transplantation, especially of the heart and kidney, but much less often in lung transplantation. "( Long-term use of everolimus in lung transplant patients.
Alba, A; Melo, J; Parada, MT; Sepúlveda, C,
)
1.91
"Everolimus has previously been shown to have some clinical activity when used as a single agent in NSCLC."( Everolimus in combination with pemetrexed in patients with advanced non-small cell lung cancer previously treated with chemotherapy: a phase I study using a novel, adaptive Bayesian dose-escalation model.
Anrys, B; Boyer, M; Di Scala, L; Dimitrijevic, S; Laack, E; Miller, N; Petrovic, K; Pylvaenaeinen, I; Solomon, B; Vansteenkiste, J; Wolf, J, 2011
)
2.53
"Everolimus has potent antifibrotic effects that may potentially affect the clinical course of bronchiolitis obliterans syndrome (BOS) or provide nephroprotective immunosuppressive regimens for lung transplantation."( A retrospective 12-month study of conversion to everolimus in lung transplant recipients.
Borro, JM; Carreño, MC; Roman, A; Santos, F; Solé, A; Ussetti, P; Zurbano, F, 2011
)
2.07
"Everolimus therefore has obvious potential in cancer therapy."( Clinical review: Current scientific rationale for the use of somatostatin analogs and mTOR inhibitors in neuroendocrine tumor therapy.
Billah, SM; Bousquet, C; Caron, P; Chalabi, M; Lasfargues, C; Pyronnet, S; Susini, C; Vezzosi, D, 2012
)
1.1
"Everolimus has shown preliminary evidence of efficacy as a single-agent in heavily pretreated relapsed/refractory HL, with an overall fair safety profile."( mTOR as a target of everolimus in refractory/relapsed Hodgkin lymphoma.
Gadaleta, CD; Giannoccaro, M; Guarini, A; Iacobazzi, A; Lapietra, A; Minoia, C; Raimondi, A; Rana, A; Ranieri, G; Silvestris, N, 2012
)
1.42
"Everolimus has demonstrated antitumor efficacy for various cancers as a result of its inhibition of the mammalian target of rapamycin (mTOR) signaling cascade, which activates cell growth and cell proliferation. "( Preparation and in vivo evaluation of liposomal everolimus for lung carcinoma and thyroid carcinoma.
Iwase, Y; Maitani, Y, 2012
)
2.08
"Everolimus has the potential to become an important agent in de novo and maintenance immunotherapy in kidney transplant recipients."( A drug safety evaluation of everolimus in kidney transplantation.
Åsberg, A; Holdaas, H; Midtvedt, K, 2012
)
1.39
"Everolimus has been approved for second-line treatment of patients with metastatic renal cell carcinoma (mRCC) after failure of sorafenib or sunitinib. "( Everolimus in metastatic renal cell carcinoma: preliminary experience from Chang Gung Memorial Hospital.
Chang, JW; Chang, YH; Chiang, YJ; Chuang, CK; Hsieh, JJ; Huang, WK; Liaw, CC; Lin, YC; Ou, LY; Pang, ST; Tsai, SI; Wang, HM; Wu, CT; Yang, CH; Yang, CT,
)
3.02
"Everolimus, which has greater polarity than sirolimus, was developed in an attempt to improve the pharmacokinetic characteristics of sirolimus, particularly to increase its oral bioavailability."( Clinical pharmacokinetics of everolimus.
Kirchner, GI; Manns, MP; Meier-Wiedenbach, I, 2004
)
1.34
"Everolimus has equivalent efficacy to mycophenolate mofetil in reducing the incidence of acute rejection after renal transplantation and superior efficacy to azathioprine after heart transplantation."( Everolimus: a proliferation signal inhibitor targeting primary causes of allograft dysfunction.
Kovarik, JM, 2004
)
2.49
"Everolimus has been associated with thrombocytopenia, leucopenia and elevated serum lipids and creatinine."( Everolimus.
Chapman, TM; Perry, CM, 2004
)
2.49
"Everolimus has been approved in some European countries and a new drug application has been submitted to the Food and Drug Administration."( Future immunosuppressive agents in solid-organ transplantation.
Cerio, J; Gabardi, S, 2004
)
1.04
"Everolimus has been used in conjunction with a new bioabsorbable polymer and gave promising results in initial clinical studies."( Clinical experiences using everolimus-eluting stents in patients with coronary artery disease.
Grube, E; Haase, J; Hofmann, M; Schwarz, F; Störger, H, 2004
)
1.34
"Everolimus has recently received approval for immunosuppressive therapy in heart transplant recipients in Austria and Germany. "( Immunosuppressive therapy with everolimus can be associated with potentially life-threatening lingual angioedema.
Berthold, HK; Deyerling, KW; Fuchs, U; Koerfer, R; Minami, K; Tenderich, G; Zittermann, A, 2005
)
2.06
"Everolimus has recently shown promise in terms of short- and long-term clinical lung transplant outcomes. "( Everolimus alters the bronchoalveolar lavage and endobronchial biopsy immunologic profile post-human lung transplantation.
Bailey, M; Kotsimbos, TC; Law, L; Levvey, BJ; Orsida, B; Snell, GI; Whitford, HM; Williams, TJ; Zheng, L, 2005
)
3.21
"Everolimus has proven efficacy for prevention of rejection in adult de novo renal and cardiac transplant recipients."( The role of therapeutic monitoring of everolimus in solid organ transplantation.
Ensom, MH; Mabasa, VH, 2005
)
1.32
"Everolimus has shown to reduce acute cellular rejection and may allow CsA dosage reduction."( Everolimus and reduced cyclosporine trough levels in maintenance heart transplant recipients.
Prenner, G; Schwarz, M; Schweiger, M; Stadlbauer, V; Stiegler, P; Tscheliessnigg, K; Wasler, A, 2006
)
2.5
"Everolimus has been shown to be as effective as mycophenolate mofetil in reducing acute rejection in renal transplantation."( Everolimus: an immunosuppressive agent in transplantation.
Kobashigawa, JA; Patel, JK, 2006
)
2.5
"Everolimus has an appropriate rationale for therapeutic use in combination with erlotinib for patients with NSCLC. "( Rationale for a phase I trial of erlotinib and the mammalian target of rapamycin inhibitor everolimus (RAD001) for patients with relapsed non small cell lung cancer.
Jackman, D; Jänne, PA; Johnson, BE, 2007
)
2
"Everolimus (EVL) has shown a potential to reduce nephrotoxicity associated with cyclosporine (CsA) while providing similar protection against rejection. "( Higher incidence of acute rejection in renal transplant recipients with low everolimus exposure.
Castagneto, M; Citterio, F; Favi, E; Renna, R; Romagnoli, J; Salerno, MP; Spagnoletti, G; Tondolo, V,
)
1.8
"Everolimus has recently been introduced into clinical practice with promising perspectives due to its efficacy, lack of nephrotoxicity, and antitumor effects. "( Evaluation of the efficacy and safety of the conversion from a calcineurin inhibitor to an everolimus-based therapy in maintenance renal transplant patients.
Arias, M; Barrientos, A; Calvo, N; Conesa, J; Cotorruelo, J; Gómez-Alamillo, C; Moreno, MA; Rodrigo, E; Ruiz San Millán, JC; Sánchez Fructuoso, A, 2007
)
2
"Everolimus (Eve) has shown good efficacy and safety profiles in clinical trials in combination with low doses of cyclosporine but there is limited experience in other modes, especially with calcineurin inhibitor elimination. "( Use of the new proliferation signal inhibitor everolimus in renal transplant patients in Spain: preliminary results of the EVERODATA registry.
Alarcón, A; Andrés, A; Beneyto, I; Cantarell, C; Díaz, JM; Errasti, P; Fernández, A; Morales, JM; Morey, A; Plaza, JJ; Rengel, M; Romero, R; Ruiz, JC; Sánchez, A; Torregrosa, JV; Zárraga, S, 2007
)
2.04
"Everolimus has not been assessed in patients with severe hepatic impairment."( Influence of hepatic impairment on everolimus pharmacokinetics: implications for dose adjustment.
Dilzer, SC; Figueiredo, J; Kovarik, JM; Lasseter, K; Reynolds, C; Rordorf, C; Sabia, HD; Zimmermann, H, 2001
)
1.31

Actions

Everolimus displays better efficacy in de novo heart transplant patients than azathioprine for prophylaxis of acute rejection episodes of at least ISHLT grade 3A. Everolimus is known to cause ILD, but its pathology is unclear.

ExcerptReferenceRelevance
"Everolimus can enhance the effect of paclitaxel on MDA-MB-231/ADR cells, inhibit cell proliferation, induce apoptosis and arrest cell cycle in the G1 phase mainly by down-regulating the expressions of key proteins in the mTOR signaling pathway."( Inhibitory effects of everolimus in combination with paclitaxel on adriamycin-resistant breast cancer cell line MDA-MB-231.
Li, Q; Liu, P; Shi, C; Wang, X; Xu, T, 2020
)
2.32
"Everolimus could inhibit the growth of both total breast cancer cells and sorted stem cells in vitro and in vivo."( [Drug sensitivity research of mTOR inhibitor on breast cancer stem cells].
Liu, P; Liu, Y; Zhang, J; Zhang, X, 2015
)
1.86
"Everolimus appears to suppress the proliferation of fibroblast and thus may provide an effective treatment strategy in glaucoma filtering surgery."( The Effect of Everolimus on Scar Formation in Glaucoma Filtering Surgery in a Rabbit Model.
Aslan, MŞ; Cinik, R; Karaöz, E; Pirhan, D; Subaşı, C; Yüksel, N, 2016
)
2.24
"Everolimus might also allow postponement of a neurosurgical resection, making it elective with an overall lower risk."( Everolimus Alleviates Obstructive Hydrocephalus due to Subependymal Giant Cell Astrocytomas.
Carai, A; Curatolo, P; Graziola, F; Marciano, S; Mastronuzzi, A; Moavero, R; Vigevano, F, 2017
)
2.62
"Everolimus displays antiproliferative effects on cancer cells, yields antiangiogenic activity in established tumours, and shows synergistic activity with paclitaxel in preclinical models. "( Safety and pharmacokinetics of paclitaxel and the oral mTOR inhibitor everolimus in advanced solid tumours.
Berton Rigaud, D; Bootle, D; Bourbouloux, E; Calvo, F; Campone, M; Dutreix, C; Levy, V; Raymond, E; Shand, N; Zoellner, U, 2009
)
2.03
"Everolimus is known to cause ILD; however, its pathology is unclear."( Granuloma-forming interstitial pneumonia occurring one year after the start of everolimus therapy.
Azuma, A; Fukuda, Y; Gemma, A; Kimura, G; Kondo, Y; Kunugi, S; Minegishi, Y; Miura, Y; Narita, K; Saito, Y; Suzuki, Y, 2013
)
1.34
"Everolimus displays a potent inhibitory effect on PTLD-derived cells in vitro and in vivo in a dose range leading to prevention of allograft rejection and may prove effective in both the prevention and treatment of PTLDs in transplant patients."( Immunosuppressive TOR kinase inhibitor everolimus (RAD) suppresses growth of cells derived from posttransplant lymphoproliferative disorder at allograft-protecting doses.
Fields, L; Joergensen, J; Korecka, M; Kossev, P; Majewski, M; Schuler, W; Shaw, L; Wasik, MA, 2003
)
2.03
"Everolimus patients had lower incidence of efficacy failure compared with azathioprine patients (41.6%, everolimus 1.5 mg; 32.2%, everolimus 3.0 mg; 52.8%, azathioprine). "( Economic evaluation of everolimus vs. azathioprine at one year after de novo heart transplantation.
Cantu, E; Cretin, N; Kaló, Z; Kauf, TL; Radeva, JI; Reed, SD; Schulman, KA, 2005
)
2.08
"Everolimus may allow optimization of immunosuppressant regimens in de novo heart transplant patients so that adequate efficacy can be achieved with reduced CNI exposure, thereby protecting kidney function."( Clinical experience with Certican (everolimus) in de novo heart transplant patients at the Deutsches Herzzentrum Berlin.
Hetzer, R; Lehmkuhl, H, 2005
)
1.33
"Everolimus displays better efficacy in de novo heart transplant patients than azathioprine for prophylaxis of biopsy-proven acute rejection episodes of at least ISHLT grade 3A (P < .001), of allograft vasculopathy (P < .01), and of CMV infections (P < .01)."( Recommendations for use of everolimus after heart transplantation: results from a Latin-American Consensus Meeting.
Ahualli, L; Amuchastegui, M; Bocchi, EA; Boullon, F; Cerutti, B; Colque, R; Fernandez, D; Fiorelli, A; Olaya, P; Perrone, SV; Vulcado, N, 2006
)
1.35
"The everolimus AUC increase with Neoral coadministration was significantly greater than the AUC increase with Sandimmune (p = 0.008)."( Differential influence of two cyclosporine formulations on everolimus pharmacokinetics: a clinically relevant pharmacokinetic interaction.
Figueiredo, J; Frazier, OL; Kalbag, J; Kovarik, JM; Rordorf, C; Rouilly, M, 2002
)
1.04

Treatment

Everolimus showed treatment efficacy and an acceptable safety tolerance with the prevention of side effects in Phase II/III studies. Everolimus as frontline treatment achieves clinical benefits for Chinese patients with mTPBC.

ExcerptReferenceRelevance
"Everolimus targeted treatment showed good efficacy in patients with recurrence/metastasis."( ♣Evaluation of clinicopathological profiles and development of a risk model in renal epithelioid angiomyolipoma patients: a large-scale retrospective cohort study.
Anwaier, A; Ding, T; Qu, YY; Su, JQ; Tian, X; Wang, Y; Xu, WH; Ye, DW; Zhang, HL, 2022
)
1.44
"Everolimus treatment led to a significant and progressive reduction in the cardiac mass volume."( A case of massive fetal cardiac rhabdomyoma: ultrasound features and management.
Balducci, A; Donti, A; Fiorentini, M; Gesuete, V; Montaguti, E; Perolo, A; Pilu, G, 2023
)
1.63
"Everolimus showed treatment efficacy and an acceptable safety tolerance with the prevention of side effects in Phase II/III studies."( Evaluating everolimus for the treatment of breast cancer.
Bourbouloux, E; Campone, M; Frenel, JS; Gourmelon, C; Moreau-Bachelard, C; Patsouris, A; Robert, M,
)
1.24
"Everolimus (EVE)-based treatment is an option for hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC), but a predictive marker has not yet been established. "( Relationship Between Daily Dose of Everolimus and Treatment Effect in Patients With Luminal HER2-negative Metastatic Breast Cancer.
Horimoto, Y; Ishizuka, Y; Murakami, F; Okazaki, M; Ushiyama, Y; Watanabe, J, 2023
)
2.63
"Everolimus as frontline treatment achieves clinical benefits for Chinese patients with mTPBC, which may provide some references for the management of Chinese mTPBC patients."( Efficacy of everolimus-based therapy in advanced triple-positive breast cancer: Experience from three cancer centers in China.
Fan, Y; Jiang, H; Li, Q; Ma, F; Tan, Y; Tian, X; Wang, J; Xu, B; Zhang, P; Zhao, W, 2023
)
2.73
"Everolimus treatment had a statistically significant effect on the renal AML volume reduction during follow-up (P < 0.05), and the mean reduction rate of volume for all cases was 56.47 ± 23.32% over 12 weeks."( Mutational analysis of renal angiomyolipoma associated with tuberous sclerosis complex and the outcome of short-term everolimus therapy.
Hao, C; Liu, F; Liu, P; Ni, J; Qin, W; Wang, T; Wu, G; Yan, F; Yang, B; Yang, X; Yu, L; Yuan, J; Zhang, G, 2019
)
1.44
"Everolimus is a treatment in NETs that may have antiangiogenic activity."( Everolimus after hepatic arterial embolisation therapy of metastases from gastrointestinal neuroendocrine tumours: The FFCD 1104-EVACEL-GTE phase II study.
Aparicio, T; Barbier, E; Baudin, E; Bouhier-Leporrier, K; Cadiot, G; Caroli-Bosc, FX; Coriat, R; de Baère, T; Di-Fiore, F; Duluc, M; Dupont-Gossart, AC; Gay, F; Ghiringhelli, F; Guimbaud, R; Hentic-Dhome, O; Lecomte, T; Legoux, JL; Lepage, C; Lepere, C; Lombard-Bohas, C; Petorin, C; Smith, D; Walter, T, 2019
)
2.68
"Everolimus treatment was continued until disease progression or intolerable toxicity was observed."( A Phase II Trial of Everolimus in Patients with Advanced Pancreatic Neuroendocrine Carcinoma Refractory or Intolerant to Platinum-Containing Chemotherapy (NECTOR Trial).
Asagi, A; Furukawa, M; Furuse, J; Hara, H; Hosokawa, A; Ikeda, M; Kojima, M; Kojima, Y; Mizuno, N; Murohisa, G; Ohkawa, S; Okusaka, T; Okuyama, H; Shioji, K; Yamanaka, T, 2020
)
1.6
"Everolimus treatment also led to decreased PD-1 expression on certain T cell subsets."( Everolimus, an mTORC1/2 inhibitor, in ART-suppressed individuals who received solid organ transplantation: A prospective study.
Aweeka, F; Baker, C; Bakkour, S; Busch, MP; Cameron, C; Chu, S; Deeks, SG; Deitchman, AN; Gibson, EA; Henrich, TJ; Hogan, LE; Liegler, T; Milush, J; Richardson, B; Rogers, R; Rutishauser, RL; Schreiner, C; Stock, PG; Thanh, C; Zarinsefat, A, 2021
)
2.79
"Everolimus treatment resulted in inhibition of mTORC1 activity in peripheral NK cells, whereas mTORC2 activity was preserved."( Cutting Edge: mTORC1 Inhibition in Metastatic Breast Cancer Patients Negatively Affects Peripheral NK Cell Maturation and Number.
Bachelot, T; Besson, L; Charrier, E; Garin, G; Heudel, PE; Marçais, A; Mayet, R; Mery, B; Morelle, M; Parant, F; Perol, D; Ray-Coquard, I; Rocca, Y; Tredan, O; Viel, S; Villard, M; Walzer, T; You, B, 2021
)
1.34
"Everolimus and metformin treated cells were subjected to cytotoxicity and clonogenic assays, western blotting, FACS and metabolic measurements."( Anti-tumor effects of everolimus and metformin are complementary and glucose-dependent in breast cancer cells.
Ariaans, G; Jalving, M; Jong, S; Vries, EG, 2017
)
1.49
"Everolimus treatment resulted in rapid reduction in tumor size, symptomatic improvement, and decrease in cerebrospinal fluid protein."( Acute Management of Symptomatic Subependymal Giant Cell Astrocytoma With Everolimus.
Arroyo, MS; Broomall, E; Franz, DN; Krueger, DA; Stevenson, CB, 2017
)
2.13
"Everolimus treatment remained safe and effective over approximately 4 years. "( Everolimus long-term use in patients with tuberous sclerosis complex: Four-year update of the EXIST-2 study.
Belousova, E; Berkowitz, N; Bissler, JJ; Brakemeier, S; Budde, K; de Vries, PJ; Frost, MD; Kingswood, JC; Peyrard, S; Radzikowska, E; Sauter, M; Voi, M; Zonnenberg, BA, 2017
)
3.34
"Everolimus treatment (1 mg/kg body weight/day) prevented the bone loss observed in OVX mice and concurrently inhibited the metastatic growth of MDA-MB-231 cells by 70% (p < 0.002) while preserving bone mass in an intracardiac model of bone metastasis."( Concurrent antitumor and bone-protective effects of everolimus in osteotropic breast cancer.
Browne, AJ; Chen, D; Göbel, A; Hadji, P; Hofbauer, LC; Kubasch, ML; Rachner, TD; Rauner, M; Stölzel, F, 2017
)
1.43
"Everolimus is a standard treatment option for advanced pancreatic neuroendocrine tumors (pNETs). "( Clinical outcomes of everolimus in patients with advanced, nonfunctioning pancreatic neuroendocrine tumors: a multicenter study in Korea.
Bang, S; Cho, JH; Jeong, S; Kim, YT; Lee, JK; Lee, KH; Lee, KJ; Lee, SH; Lee, TH; Lee, WJ; Paik, WH; Ryu, JK; Song, SY; Woo, SM, 2017
)
2.22
"Everolimus treatment is seriously hampered by its toxicity profile. "( Clinical validation study of dried blood spot for determining everolimus concentration in patients with cancer.
Brüggemann, RJM; Croes, S; de Beer, YM; Knapen, LM; van Erp, NP; van Herpen, CML; Willemsen, AECAB, 2018
)
2.16
"Everolimus is a treatment option for hemodialysis patients with metastatic atypical bronchial carcinoid. "( Efficacy and safety of everolimus treatment in a hemodialysis patient with metastatic atypical bronchial carcinoid: case report and literature review.
Brizzi, MP; La Salvia, A; Scagliotti, GV; Sonetto, C; Tampellini, M; Volante, M, 2018
)
2.23
"Everolimus treatment was performed for AML with a maximum diameter of 4 cm."( Effect of everolimus treatment for regrown renal angiomyolipoma associated with tuberous sclerosis complex after transcatheter arterial embolization.
Egawa, S; Endo, K; Hatano, T; Inaba, H; Matsu-Ura, T; Mori, KI; Tamari, M, 2018
)
1.6
"Everolimus treatment suppressed the conversion of glutamine and glutamate into glutathione both in vitro and in vivo."( Inhibition of mTORC1 in pediatric low-grade glioma depletes glutathione and therapeutically synergizes with carboplatin.
Alt, J; Arnold, A; Eberhart, CG; Poore, B; Price, A; Raabe, EH; Rubens, JA; Slusher, BS; Yuan, M, 2019
)
1.24
"Both everolimus-treated and non-treated mice were sacrificed at several time points, starting 30 min and finishing 7 days after IRI induction."( Effects of everolimus on oxidative stress in kidney model of ischemia/reperfusion injury.
Bajcetic, M; Becker, JU; Kezic, A; Thaiss, F; Tsui, TY, 2013
)
1.23
"Everolimus treatment significantly reduced the number of ICAM-1 positive small vessels (66%; p<0.05) and suppressed the infiltration of leucocytes (CD11a (64%), CD18 (42%); p<0.05) and macrophages (ED1; 22%) in the allografts on POD 30. "( Everolimus's influence on persistent acute rejection after experimental lung transplantation.
Brunner, E; Hirt, SW; Lehle, K; Schmid, C; von Suesskind-Schwendi, M,
)
3.02
"Everolimus treatment was also able to attenuate the activation of primary stellate cells to their activated form."( Everolimus is a potent inhibitor of activated hepatic stellate cell functions in vitro and in vivo, while demonstrating anti-angiogenic activities.
Chatterjee, S; Dufour, JF; Maheshwari, U; Majumder, S; Manjunathan, R; Piguet, AC; Saran, U, 2014
)
2.57
"Everolimus-based salvage treatment of refractory cGVHD results in significant improvement of the NIH Severity Score without impairing control of the malignant disease."( Salvage therapy with everolimus reduces the severity of treatment-refractory chronic GVHD without impairing disease control: a dual center retrospective analysis.
Ammer, J; Ditz, D; Einsele, H; Grigoleit, GU; Holler, E; Kapp, M; Lutz, M; Mielke, S; Schmidhuber, J; Wolff, D, 2014
)
1.44
"Everolimus-treated mice displayed a marked reduction in weight gain from the last day of the treatment period."( Evaluation of the impact of the cancer therapy everolimus on the central nervous system in mice.
Anouar, Y; Castel, H; Dubois, M; Gandolfo, P; Hilber, P; Joly, F; Le Joncour, V; Morin, F; Proust, F; Tonon, MC, 2014
)
1.38
"Everolimus treatment in TSC patients may lead to life-threatening outcomes, including sepsis and death."( Complications of mammalian target of rapamycin inhibitor anticancer treatment among patients with tuberous sclerosis complex are common and occasionally life-threatening.
Dachowska, I; Fendler, W; Jozwiak, S; Kotulska, K; Mlynarski, W; Trelinska, J, 2015
)
1.14
"No everolimus-treated patients experienced renal bleeding."( Everolimus for renal angiomyolipoma in patients with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis: extension of a randomized controlled trial.
Belousova, E; Berkowitz, N; Bissler, JJ; Brakemeier, S; Budde, K; de Vries, PJ; Frost, M; Kingswood, JC; Miao, S; Nonomura, N; Peyrard, S; Radzikowska, E; Sauter, M; Segal, S; Zonnenberg, BA, 2016
)
2.39
"With everolimus treatment, the patient's diarrhea resolved and replacement therapy for hypoproteinemia was less frequent."( Everolimus for Primary Intestinal Lymphangiectasia With Protein-Losing Enteropathy.
Fukao, T; Hori, T; Ibuka, T; Kanda, K; Kawamoto, N; Miyazaki, T; Ozeki, M, 2016
)
2.33
"Everolimus treatment was stopped, and treated with methylprednisolone and empiric antibiotic therapy for 7 days."( Treatment with everolimus for a patient with systemic metastatic breast cancer results in severe pulmonary injury: a case report.
Santai, S; Shikai, W; Tao, W; Xiangying, M; Zefei, J, 2017
)
1.53
"Everolimus treatment improved behavioral deficits in the patient."( Everolimus improves neuropsychiatric symptoms in a patient with tuberous sclerosis carrying a novel TSC2 mutation.
Hwang, SK; Kaang, BK; Lee, JA; Lee, JH; Lee, K; Lee, YS; Lim, CS; Yang, JE, 2016
)
2.6
"Everolimus treatment combined with early CNI elimination is associated with increased densities of Tregs 12-months post-HTx compared to patients receiving CNI based regimen. "( Effect of everolimus initiation and early calcineurin inhibitor withdrawal on myocardial FOXP3+ regulatory T cells in heart transplantation.
Andersen, C; Arora, S; Gullestad, L; Gustafsson, F; Mirza, K, 2016
)
2.28
"Everolimus treatment disrupts the S6K1-IRS-2/PI3K negative feedback loop, leading to BRAF V600E-dependent activation of ERK and Mcl-1 stabilization in colon cancer cells, which in turn blocks the crosstalk from the death receptor to mitochondria."( BRAFV600E-dependent Mcl-1 stabilization leads to everolimus resistance in colon cancer cells.
Chen, D; He, K; Li, X; Ruan, H; Tong, J; Xu, X; Yu, J; Zhang, L, 2016
)
1.41
"Everolimus treatment resulted in significant increases of TCh (from 192.04±40.51mg/dl to 210.74±51.12mg/dl and to 216.69±45.43mg/dl; p=0.0273) and LDL (from 113.21±38.72mg/dl to 133.88±50.71mg/dl and to 141.58±40.67mg/dl; p=0.0006) after three and 12 months respectively."( Everolimus treatment among patients with tuberous sclerosis affects serum lipid profile.
Dachowska, I; Fendler, W; Jóźwiak, S; Kotulska, K; Młynarski, W; Trelińska, J, 2016
)
2.6
"Everolimus treatment had a statistically significant effect on the renal AML volume reduction during follow-up (P < 0.05)."( Effects of everolimus on tuberous sclerosis complex-associated renal angiomyolipoma: A preliminary report.
Fan, HC; Hsu, CC; Sheu, JN; Tsai, JD; Tsai, ML; Tung, MC; Wei, CC; Yang, SH, 2017
)
1.57
"Everolimus based treatment has meaningful activity in heavily pretreated patients with HR-positive MBC but is associated with considerable toxicity and requirement for dose adjustment."( Everolimus in heavily pretreated metastatic breast cancer: Is real world experience different?
Bajpai, J; Ghosh, J; Gulia, S; Gupta, S; Ramaswamy, A,
)
3.02
"Everolimus treatment resulted in reduced neointimal hyperplasia (thickness 3.7+/-1.2 microm) compared to untreated controls (10.1+/-2.5 microm, p=0.008). "( Everolimus attenuates neointimal hyperplasia in cultured human saphenous vein grafts.
Bernecker, OY; Bonatti, JO; Laufer, G; Macfelda, K; Ruttmann-Ulmer, E; Semsroth, S; Stigler, RG; Troppmair, J, 2009
)
3.24
"Everolimus single agent treatment significantly inhibited tumor growth relative to control and cyclophosphamide treatment (T/C 19%, p<0.01). "( mTOR inhibition sensitizes gastric cancer to alkylating chemotherapy in vivo.
Cejka, D; Fuereder, T; Preusser, M; Sieghart, W; Strommer, S; Wacheck, V; Werzowa, J,
)
1.57
"Everolimus-treated heart and lung transplant patients in the lowest tertile for time posttransplant exhibited mean increases of 7.8 mL/min and 4.9 mL/min, respectively."( Everolimus with reduced calcineurin inhibitor in thoracic transplant recipients with renal dysfunction: a multicenter, randomized trial.
Bergh, CH; Bjørtuft, O; Eiskjaer, H; Ekmehag, B; Fagertun, HE; Gude, E; Gullestad, L; Iversen, M; Jansson, K; Mared, L; Mortensen, SA; Riise, GC; Rundqvist, B; Simonsen, S; Solbu, D, 2010
)
2.52
"Everolimus (RAD) treatment was initiated on the day after or 3 days after induction of myocardial infarction (MI) in rats."( Beneficial effects of Mammalian target of rapamycin inhibition on left ventricular remodeling after myocardial infarction.
Bea, F; Bekeredjian, R; Buss, SJ; Hagenmueller, M; Hardt, SE; Izumo, S; Katus, HA; Malekar, P; Muenz, S; Riffel, JH; Schinke-Braun, M; Weiss, CS, 2009
)
1.8
"Everolimus-treated mice had significantly less rejection at all time points compared with non-immunosuppressed mice."( Long-term laryngeal allograft survival using low-dose everolimus.
Dan, O; Lott, DG; Lu, L; Strome, M, 2010
)
1.33
"Everolimus treatment led to 36% of 6-month progression-free survival (PFS) rate and 31% of 3-month PFS rate."( Everolimus in renal cell carcinoma.
Wang, Y, 2010
)
2.52
"The everolimus treatment lasted 20 months (range, 6-60)."( Everolimus in clinical practice in long-term liver transplantation: an observational study.
Argudo, A; Casanovas, T; Peña-Cala, MC,
)
2.05
"Everolimus treatment was inefficient at controlling secretion and tumor growth of one ACTH pituitary carcinoma."( New targeted therapies in pituitary carcinoma resistant to temozolomide.
Borson-Chazot, F; Ducray, F; Favrel, V; Honnorat, J; Jouanneau, E; Raverot, G; Trouillas, J; Wierinckx, A, 2012
)
1.1
"Everolimus treatment resulted in marked tumor regression, significant improvement in patient's ambulation and cessation of seizures."( Effective everolimus treatment of inoperable, life-threatening subependymal giant cell astrocytoma and intractable epilepsy in a patient with tuberous sclerosis complex.
Jóźwiak, S; Jurkiewicz, E; Kotulska, K; Perek, D; Perek-Polnik, M, 2012
)
1.5
"Everolimus treated group (EveG) showed significantly less proteinuria and albuminuria, less glomerular and tubulointerstitial damage and fibrosis, fibroblast activation cell proliferation, when compared with control group (CG), even though the EveG remained with high blood pressure. "( Delayed mTOR inhibition with low dose of everolimus reduces TGFβ expression, attenuates proteinuria and renal damage in the renal mass reduction model.
Arévalo, MA; Boggia, J; Coria, V; Gimenez-Bonafe, P; González-Martínez, F; Grande, MT; Grinyó, J; Herrero-Fresneda, I; Kurdián, M; Lloberas, N; López-Novoa, JM; Malacrida, L; Noboa, O; Torras, J, 2012
)
2.09
"Everolimus treatment reduced VEGF-A levels in the supernatant of all cell lines. "( Measurement of tumor VEGF-A levels with 89Zr-bevacizumab PET as an early biomarker for the antiangiogenic effect of everolimus treatment in an ovarian cancer xenograft model.
de Jong, S; de Vries, EG; Kosterink, JG; Lub-de Hooge, MN; Pot, L; Reyners, AK; Schröder, CP; Terwisscha van Scheltinga, AG; Timmer-Bosscha, H; van der Bilt, AR; van der Zee, AG, 2012
)
2.03
"Pre-everolimus entry treatment options included exemestane, fulvestrant, and tamoxifen."( Budget impact analysis of everolimus for the treatment of hormone receptor positive, human epidermal growth factor receptor-2 negative (HER2-) advanced breast cancer in the United States.
De, G; Diener, M; Namjoshi, M; Wu, EQ; Xie, J; Yang, H, 2013
)
1.17
"Everolimus treatment also induced a significant decrease in Ki67 index and in the phosphorylation levels of the two major effectors of mTOR, p70S6K and 4E-BP1."( Antitumor effect of everolimus in preclinical models of high-grade gastroenteropancreatic neuroendocrine carcinomas.
Benslama, N; Blanc, M; Bollard, J; Couderc, C; Ferraro-Peyret, C; Gouysse, G; Hervieu, V; Lepinasse, F; Poncet, G; Roche, C; Scoazec, JY; Walter, T, 2013
)
1.43
"Both everolimus treatment groups significantly reduced in-stent neointimal growth (46% reduction and 42% reduction in intimal thickness in groups 1 and 2, respectively)."( Oral everolimus inhibits in-stent neointimal growth.
Acampado, E; Farb, A; John, M; Kolodgie, FD; Prescott, MF; Virmani, R, 2002
)
1.28
"In everolimus-treated patients, mean increases in cyclosporine AUC0-12 ranged from 7% to 43%, which were not significantly different across all dosing cohorts including placebo."( Influence of everolimus on steady-state pharmacokinetics of cyclosporine in maintenance renal transplant patients.
Budde, K; Dantal, J; Fauchald, P; Fritsche, L; Lehne, G; Lison, A; Neumayer, HH; Renders, L; Soulillou, JP; Winkler, M, 2005
)
1.21
"Everolimus treatment of established colitis in IL-10-/- mice ameliorated the colitis, probably as a result of decreasing the number of CD4+ T cells in the colonic mucosa and an associated reduction in IFN-gamma production."( Therapeutic effect of a new immunosuppressive agent, everolimus, on interleukin-10 gene-deficient mice with colitis.
Hamanaka, Y; Inoue, M; Ito, T; Kai, Y; Matsuda, C; Matsuda, H; Mizushima, T; Nishida, T; Sawa, Y; Song, J; Tamagawa, H, 2007
)
1.31
"Everolimus treatment markedly reduced the number of 5-bromo-2-deoxyuridine-labeled nuclei in cyst epithelia."( Everolimus retards cyst growth and preserves kidney function in a rodent model for polycystic kidney disease.
Le Hir, M; Serra, AL; Wackerle-Men, Y; Wahl, PR; Wu, M; Wuthrich, RP, 2007
)
2.5
"Everolimus treatment resulted in immediate inhibition (25-55%) of lymphocyte proliferation in renal-allograft recipients; values returning to baseline by 14 days after cessation of everolimus treatment."( Pharmacodynamic effects of everolimus on anti-CD3 antibody-stimulated T-lymphocyte proliferation and interleukin-10 synthesis in stable kidney-transplant patients.
Böhler, T; Budde, K; Kamar, N; Lichter, S; Neumayer, HH; Schumann, B; Waiser, J, 2008
)
1.36
"In everolimus-treated patients for whom rifampin is indicated, alternative agents with less enzyme induction potential than rifampin could be considered. "( Effect of rifampin on apparent clearance of everolimus.
Figueiredo, J; Hartmann, S; Kovarik, JM; Port, A; Rordorf, C; Rouilly, M, 2002
)
1.2
"Treatment with everolimus increased survival of mice infected with C."( Screening the medicine for malaria venture's Pandemic Response Box to identify novel inhibitors of Candida albicans and Candida auris biofilm formation.
Ajetunmobi, OH; Badali, H; Chaturvedi, AK; Lopez-Ribot, JL; Najvar, L; Patterson, TF; Vaccaro, A; Wiederhold, NP; Wormley, FL, 2023
)
1.25
"Treatment with everolimus and exemestane was discontinued and steroid pulse therapy was initiated; however, she required ventilator management because of the severity of the pneumonia."( [A Case of Metastatic Breast Cancer That Recovered from Diffuse Alveolar Damage Associated with Everolims].
Hasegawa, T; Hirakawa, K; Kashiwagi, S; Kinoshita, H; Nakamoto, K; Ohira, M; Teraoka, H, 2020
)
0.9
"Treatment with everolimus reduced allogeneic fibroproliferation, but had no protective effects on the microvasculature; polymerase chain reaction analysis indicated hypoxic stress and inflammation."( Microvasculature in murine tracheal allografts after combined therapy with clopidogrel and everolimus.
Ensminger, SM; Heim, C; Kuckhahn, A; Nicolls, MR; Ramsperger-Gleixner, M; Weyand, M, 2021
)
1.18
"When treated with everolimus, the overall incidence of stomatitis is about two thirds of the patients. "( Stomatitis in mTOR inhibitors treatment and other targeted cancer therapy, possibilities of infl uencing it, and the use of local corticotherapy.
Černá, A; Jánská, V; Kozáková, Š; Liška, J; Vokurka, S, 2020
)
0.89
"Treatment with everolimus was stopped due to adverse events in 8 patients (47.1%). "( Pilot Conversion Study From Mycophenolate Mofetil to Everolimus in Stable ABO-Incompatible Kidney Transplant Recipients: Analysis of 1-Year Follow-Up Data.
Iwai, T; Kabei, K; Kumada, N; Kuwabara, N; Naganuma, T; Nakatani, T; Nishide, S; Takemoto, Y; Uchida, J, 2019
)
1.12
"Treatment with everolimus supports the expansion of immunosuppressive regulatory T cells (Tregs), which exert a negative effect on antitumor immune responses."( Metronomic cyclophosphamide attenuates mTOR-mediated expansion of regulatory T cells, but does not impact clinical outcome in patients with metastatic renal cell cancer treated with everolimus.
de Gruijl, TD; Haanen, JBAG; Hamberg, P; Helgason, HH; Huijts, CM; Los, M; Lougheed, SM; Polee, MB; Tascilar, M; van der Vliet, HJ; Verheul, HM; Werter, IM, 2019
)
1.05
"The treatment with everolimus significantly reduced heme oxygenase-1 expression and increased iNOS mRNA expression when compared to non-treated animals."( Effects of everolimus on oxidative stress in kidney model of ischemia/reperfusion injury.
Bajcetic, M; Becker, JU; Kezic, A; Thaiss, F; Tsui, TY, 2013
)
1.1
"Treatment with everolimus or letrozole resulted in growth inhibition of SCs in a dose-dependent manner."( Everolimus in combination with letrozole inhibit human breast cancer MCF-7/Aro stem cells via PI3K/mTOR pathway: an experimental study.
Hou, G; Liu, J; Liu, Y; Zhang, J; Zhang, S; Zhang, X, 2014
)
2.18
"Treatment with everolimus, an oral mTOR inhibitor, offers patients a noninvasive pharmacotherapeutic treatment option. "( Role of mTOR inhibition in the treatment of patients with renal angiomyolipomas.
Coombs, EJ, 2013
)
0.74
"Treatment with everolimus (10 mg/day) was initiated on POD 32."( [A case of massive pancreatic neuroendocrine carcinoma successfully treated with surgical resection and postoperative everolimus administration].
Egawa, C; Hamanaka, M; Hashimoto, N; Kagawa, Y; Kato, T; Katsura, Y; Kawashima, H; Kusama, H; Matsushita, K; Mukai, Y; Nakahira, S; Okishiro, M; Sakisaka, H; Suzuki, R; Takeda, Y; Takeno, A; Tamura, S; Taniguchi, H, 2013
)
0.94
"Treatment with everolimus significantly enhanced the antitumor activity of trabectedin plus SN-38 or topotecan."( Combination treatment with trabectedin and irinotecan or topotecan has synergistic effects against ovarian clear cell carcinoma cells.
Hamasaki, T; Hisamatsu, T; Kawano, M; Kimura, T; Kishimoto, T; Mabuchi, S; Matsumoto, Y; Sasano, T; Sawada, K; Takahashi, K; Takahashi, R; Takahashi, T, 2014
)
0.74
"Treatment with everolimus, resulted in 56% growth inhibition, and a triple combination of SU11274, everolimus and XAV939, resulted in 95% growth inhibition in RU cells."( Targeting c-Met in melanoma: mechanism of resistance and efficacy of novel combinatorial inhibitor therapy.
Bertram, C; Botting, GM; Crees, Z; Etnyre, D; Fong, JT; Girard, J; Jacobs, RJ; Moravec, DN; Puri, N; Rajanna, S; Shambannagari, MR; Stone, AL; Uppada, SB, 2014
)
0.74
"Treatment with everolimus was started when he was 10 months old."( Everolimus Treatment for an Early Infantile Subependymal Giant Cell Astrocytoma With Tuberous Sclerosis Complex.
Fukumura, S; Minagawa, K; Takayama, R; Tsutsumi, H; Watanabe, T, 2015
)
2.2
"Treatment with everolimus reduces the size of SEGA associated with TSC with an adequate safety profile, and constitutes an alternative to surgery in certain cases."( [Response to everolimus in patients with giant cell astrocytoma associated to tuberous sclerosis complex].
Baena-Gómez, MA; Camino-León, R; Fernández-Ramos, JA; López-Laso, E; Mateos-González, ME; Peña-Rosa, MJ; Vicente-Rueda, J, 2014
)
1.12
"Treatment with everolimus was identified as an independent predictor of longer survival (hazard ratio = 0.34; P = .02)."( Everolimus-based immunosuppression in patients with hepatocellular carcinoma at high risk of recurrence after liver transplantation: a case series.
Diaz, SP; Ferreiro, AO; Gutiérrez, MG; López, FS; Patiño, MJ; Vazquez-Millán, MA, 2014
)
2.18
"The treatment with everolimus significantly inhibited cell growth, and significantly reduced the expression of p-mTOR, downstream signaling proteins, and hypoxic related proteins as well as invasion and migration potentials (P < 0.05)."( Anti-tumor effect of the mammalian target of rapamycin inhibitor everolimus in oral squamous cell carcinoma.
Kawakita, A; Kawasaki, G; Naruse, T; Rokutanda, S; Umeda, M; Yamada, S; Yanamoto, S, 2015
)
0.97
"Treatment with everolimus induced a clear reduction of transaminases within 2 weeks. "( Everolimus treatment for patients with autoimmune hepatitis and poor response to standard therapy and drug alternatives in use.
Larsen, FS; Ytting, H, 2015
)
2.21
"Treatment with everolimus was associated with significant improvement in progression-free survival in patients with progressive lung or gastrointestinal neuroendocrine tumours. "( Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study.
Buzzoni, R; Carnaghi, C; Fave, GD; Fazio, N; Kulke, MH; Lahner, H; Oh, DY; Pacaud, LB; Pavel, ME; Raderer, M; Rouyrre, N; Sachs, C; Shimada, Y; Singh, S; Strosberg, J; Tesselaar, M; Tomasek, J; Valle, JW; Van Cutsem, E; Voi, M; Wolin, E; Yao, JC, 2016
)
2.23
"Treatment with everolimus resulted in growth inhibition of all stem cells in a dose-dependent manner. "( [Drug sensitivity research of mTOR inhibitor on breast cancer stem cells].
Liu, P; Liu, Y; Zhang, J; Zhang, X, 2015
)
0.77
"Treatment with everolimus in patients with extrapancreatic NETs appears to be a promising strategy that is safe and well tolerated. "( Efficacy and Safety of Everolimus in Extrapancreatic Neuroendocrine Tumor: A Comprehensive Review of Literature.
Agrimi, D; Aversano, M; Bassi, V; Colao, A; Faggiano, A; Giordano, EA; Guarnotta, V; Logoluso, FA; Malandrino, P; Messina, E; Modica, R; Nicastro, V; Nuzzo, V; Sciaraffia, M, 2016
)
1.1
"Treatment with everolimus was stopped owing to adverse events in 11 patients (42.3%)."( Clinical Experience of Late Conversion From Antimetabolites With Standard Exposure Calcineurin Inhibitors to Everolimus With Calcineurin Inhibitor Minimization in Stable Kidney Transplant Recipients With Good Renal Function.
Iwai, T; Kabei, K; Kumada, N; Kuwabara, N; Naganuma, T; Nakatanti, T; Nishide, S; Takemoto, Y; Uchida, J; Yamasaki, T, 2016
)
0.99
"Treatment with everolimus markedly inhibited the growth of tumors induced by poorly differentiated HAK-1B and KYN-2 cells and phosphorylation of mTOR Ser2481 in vivo."( Phosphorylation of mTOR Ser2481 is a key target limiting the efficacy of rapalogs for treating hepatocellular carcinoma.
Akiba, J; Kawahara, A; Kuwano, M; Murakami, Y; Nishitani, A; Noda, M; Ono, M; Shibata, T; Watari, K; Yano, H, 2016
)
0.77
"Treatment of everolimus, the specific mTOR inhibitor, significantly attenuated DDX5-mediated cell proliferation."( DDX5 promotes gastric cancer cell proliferation in vitro and in vivo through mTOR signaling pathway.
Chen, L; Du, C; Hou, MX; Li, DQ; Li, N; Li, SS; Xie, MJ; Yang, Y; Zheng, ZD, 2017
)
0.81
"Treatment with everolimus prolongs progression-free survival relative to placebo in patients with metastatic renal cell carcinoma that had progressed on other targeted therapies."( Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial.
Berg, WJ; Bracarda, S; Escudier, B; Figlin, RA; Grünwald, V; Hollaender, N; Hutson, TE; Kay, A; Lebwohl, D; Motzer, RJ; Oudard, S; Porta, C; Ravaud, A; Thompson, JA; Urbanowitz, G, 2008
)
1.08
"Treatment with everolimus was initiated at a mean (SD) of 79.8 (52.7) months (range, 10-163 mo) after transplantation."( Preliminary experience with conversion from calcineurin inhibitors to everolimus in cardiac transplantation maintenance therapy.
Arizón-Muñoz, JM; Benezet-Mazuecos, J; Guisado, A; Jiménez-Díaz, J; Lage-Gallé, E; Martínez, A; Mogollón, MV; Romero-Rodriguez, N; Sánchez-Brotons, JA; Sobrino-Márquez, JM, 2008
)
0.92
"Treatment with everolimus at the minimal effective dose was studied in combination with cyclophosphamide at maximum tolerated dose in a human gastric cancer severe combined immunodeficient (SCID) mouse xenograft model. "( mTOR inhibition sensitizes gastric cancer to alkylating chemotherapy in vivo.
Cejka, D; Fuereder, T; Preusser, M; Sieghart, W; Strommer, S; Wacheck, V; Werzowa, J,
)
0.48
"Treatment with everolimus allowed a primary response."( Maintenance immunosuppressive therapy with everolimus preserves humoral immune responses.
Bemelman, FJ; Idu, MM; Koch, SD; Minnee, RC; Struijk, GH; ten Berge, IJ; van Donselaar-van der Pant, KA; Zwinderman, AH, 2010
)
0.96
"Treatment of everolimus-resistant disease remains largely undefined in metastatic renal cell carcinoma (mRCC). "( Treatment of everolimus-resistant metastatic renal cell carcinoma with VEGF-targeted therapies.
Busch, J; Fenner, M; Ganser, A; Grünwald, V; Seidel, C; Weikert, S, 2011
)
1.11
"Treatment of everolimus-resistant disease was associated with a PFS of 5.5 months. "( Treatment of everolimus-resistant metastatic renal cell carcinoma with VEGF-targeted therapies.
Busch, J; Fenner, M; Ganser, A; Grünwald, V; Seidel, C; Weikert, S, 2011
)
1.11
"Treatment with everolimus also diminished glomerular hypertrophy."( Delayed mTOR inhibition with low dose of everolimus reduces TGFβ expression, attenuates proteinuria and renal damage in the renal mass reduction model.
Arévalo, MA; Boggia, J; Coria, V; Gimenez-Bonafe, P; González-Martínez, F; Grande, MT; Grinyó, J; Herrero-Fresneda, I; Kurdián, M; Lloberas, N; López-Novoa, JM; Malacrida, L; Noboa, O; Torras, J, 2012
)
0.98
"Treatment with everolimus is known to prolong progression-free survival in patients with renal cell carcinoma resistant against tyrosine-kinase inhibitor therapy. "( [Management of side effects of everolimus treatment for metastatic renal cell carcinoma].
Hongo, F; Mikami, K; Miki, T, 2012
)
1.02

Toxicity

Everolimus appears to be safe in mRCC patients with renal impairment or end-stage renal disease requiring dialysis. In pediatric renal transplantation, an everolimus-based regimen with low-dose cyclosporine yields comparable four year results.

ExcerptReferenceRelevance
" There were no significant between-group differences in the composite endpoint of BPAR, graft loss or death, nor any significant between-group differences in adverse events in either study."( Everolimus with optimized cyclosporine dosing in renal transplant recipients: 6-month safety and efficacy results of two randomized studies.
Alves Filho, G; Bernhardt, P; Bourbigot, B; Cambi, V; Campbell, S; Civati, G; Cretin, N; Eris, J; Esmeraldo, R; Garcia, VD; Geissler, J; Haas, T; Jappe, A; Leone, J; Magee, JC; Pascual, J; Rigotti, P; Tedesco, H; Vitko, S; Whelchel, J, 2004
)
1.77
" Adverse events were higher in everolimus-treated patients especially at the 4-mg/day dose, but there was no difference in the incidence of thrombocytopenia or leukopenia between all groups and renal function as determined by serum creatinine, and creatinine clearance remained stable to 36 months in everolimus-treated patients."( Safety, tolerability, and efficacy of everolimus in de novo liver transplant recipients: 12- and 36-month results.
Abecassis, M; Calmus, Y; Fischer, L; Freeman, R; Klempnauer, J; Langnas, A; Levy, G; Mayer, D; Nashan, B; Neuhaus, P; Punch, J; Roberts, J; Rogiers, X; Samuel, D; Schmidli, H; Sloof, M; Tuttle-Newhall, E, 2006
)
0.89
"To date there is a substantial experience with rapamycin conversion in stable renal transplant recipients with respect to the procedure of conversion, initial doses, and target blood levels as well as adverse events, but in the case of Everolimus there is almost no experience with conversion and calcineurin inhibitor (CNI) withdrawal."( Conversion to everolimus in kidney transplant recipients: a safe and simple procedure.
Arias, M; Barrientos, A; Calvo, N; Conesa, J; Cotorruelo, JG; Gómez-Alamillo, C; Rodrigo, E; Ruiz, JC; Sanchez-Fructuoso, A, 2006
)
0.88
" We describe our experience in a series of 78 stable renal transplant patients who were switched to Everolimus with complete and quick elimination of the CNI: the procedure of conversion, pharmacokinetic results after conversion, evolution of renal parameters (renal function, proteinuria, and others), and safety data (acute rejection and adverse events)."( Evaluation of the efficacy and safety of the conversion from a calcineurin inhibitor to an everolimus-based therapy in maintenance renal transplant patients.
Arias, M; Barrientos, A; Calvo, N; Conesa, J; Cotorruelo, J; Gómez-Alamillo, C; Moreno, MA; Rodrigo, E; Ruiz San Millán, JC; Sánchez Fructuoso, A, 2007
)
0.78
"mTOR inhibitors were safe immunosuppressive drugs whose side effects were controlled and easily managed."( Efficacy, tolerance, and safety of mammalian target of rapamycin inhibitors as rescue immunosuppressants in liver transplantation.
Alamo, JM; Barrera, L; Bernal, C; Casado, MD; Cordero, E; Gomez-Bravo, MA; Jimenez, R; Marin, LM; Sanchez-Moreno, L; Sousa, JM; Suarez, G; Suarez-Grau, JM,
)
0.13
", re-introduction of CNI), incidence of adverse events, and mortality up to one year after OLT."( A therapeutic exploratory study to determine the efficacy and safety of calcineurin-inhibitor-free de-novo immunosuppression after liver transplantation: CILT.
Goralczyk, AD; Lorf, T; Obed, A; Ramadori, G; Schnitzbauer, A; Tsui, TY, 2010
)
0.36
" Adverse events led to study drug discontinuation in 17 IE patients (26."( Efficacy and safety of de novo or early everolimus with low cyclosporine in deceased-donor kidney transplant recipients at specified risk of delayed graft function: 12-month results of a randomized, multicenter trial.
Bayle, F; Berthoux, F; Cassuto, E; Charpentier, B; Dantal, J; Di Giambattista, F; Genin, R; Glotz, D; Kossari, N; Lefrançois, N; Legendre, C; Merville, P; Moal, MC; Moulin, B; Quéré, S; Rerolle, JP; Rostaing, L; Toupance, O; Westeel, PF, 2010
)
0.63
" Tolerable dose, need for supportive treatments, and early signals for toxic effect were evaluated in a planned safety analysis of 270 patients."( Integrating bevacizumab, everolimus, and lapatinib into current neoadjuvant chemotherapy regimen for primary breast cancer. Safety results of the GeparQuinto trial.
Blohmer, JU; Costa, SD; Eidtmann, H; Fasching, PA; Fehm, T; Gerber, B; Hanusch, C; Hilfrich, J; Kreienberg, R; Loibl, S; Nekljudova, V; Solbach, C; Strumberg, D; Untch, M; von Minckwitz, G, 2011
)
0.67
"mTORi treatment was prematurely eliminated due to adverse events in 112 patients."( Comparative analysis of adverse events requiring suspension of mTOR inhibitors: everolimus versus sirolimus.
Arias, M; Calvo Romero, N; Gómez Alamillo, C; Pérez-Flores, I; Ruiz, JC; Sánchez-Fructuoso, AI, 2010
)
0.59
"03) and a 43% decrease in major adverse cardiovascular events, cardiac death, myocardial infarction, or ischemic-driven target lesion revascularization (9."( Long-term (three-year) safety and efficacy of everolimus-eluting stents compared to paclitaxel-eluting stents (from the SPIRIT III Trial).
Applegate, RJ; Caputo, R; Cutlip, DE; Doostzadeh, J; Farhat, N; Hermiller, JB; Lansky, AJ; Sood, P; Stone, GW; Sudhir, K; Williams, JE; Yaqub, M; Yu, S, 2011
)
0.63
" The rapid introduction of novel treatment options into clinical practice within a relatively short time frame has created some new challenges pertaining to adverse event (AE) management in patients with advanced RCC."( Treatment-associated adverse event management in the advanced renal cell carcinoma patient treated with targeted therapies.
Ravaud, A, 2011
)
0.37
"In April 2009, an expert group of 11 physicians and clinical nurses met to discuss the management of selected adverse events associated with the use of everolimus for the treatment of metastatic renal cell carcinoma (mRCC)."( Management of adverse events associated with the use of everolimus in patients with advanced renal cell carcinoma.
Climent, MA; Creel, P; Dickow, B; Fischer, P; Gornell, SS; Meloni, F; Motzer, RJ; Osanto, S; Porta, C; Ravaud, A; Vaishampayan, U; White, DA, 2011
)
0.81
" Guidance for management of these adverse events is provided."( Management of adverse events associated with the use of everolimus in patients with advanced renal cell carcinoma.
Climent, MA; Creel, P; Dickow, B; Fischer, P; Gornell, SS; Meloni, F; Motzer, RJ; Osanto, S; Porta, C; Ravaud, A; Vaishampayan, U; White, DA, 2011
)
0.62
" Patients were closely monitored for the development of serious and grades 3/4 adverse events (AEs)."( An international expanded-access programme of everolimus: addressing safety and efficacy in patients with metastatic renal cell carcinoma who progress after initial vascular endothelial growth factor receptor-tyrosine kinase inhibitor therapy.
Anak, O; Bavbek, SE; Blank, CU; Bodrogi, I; Bono, P; Booth, J; Castellano, D; Grünwald, V; Hawkins, R; Karakiewicz, PI; Knox, JJ; Larkin, J; Lee, SH; Machiels, JP; Miller, K; Pirotta, N; Rha, SY; Rosamilia, M, 2012
)
0.64
" The most common severe adverse event probably related to sorafenib was diarrhea (12."( Efficacy and safety of sorafenib in combination with mammalian target of rapamycin inhibitors for recurrent hepatocellular carcinoma after liver transplantation.
Bustamante, J; Castroagudin, JF; Garralda, E; Gomez-Martin, C; Herrero, I; Matilla, A; Salcedo, M; Sangro, B; Testillano, M, 2012
)
0.38
" We sought to determine the causes, incidence, risk factors, and consequences of withdrawal due to adverse events of PSIs in maintenance heart transplantation."( Withdrawal of proliferation signal inhibitors due to adverse events in the maintenance phase of heart transplantation.
Almenar, L; Arizon Del Prado, JM; Brossa, V; Crespo-Leiro, MG; Delgado, JF; Diaz-Molina, B; Gomez-Bueno, M; Gonzalez-Vilchez, F; Lambert, JL; Martínez-Dolz, L; Mirabet, S; Paniagua, MJ; Perez-Villa, F; Roig, E; Ruiz-Cano, MJ; Segovia, J; Vazquez de Prada, JA, 2012
)
0.38
"In this large cohort of maintenance heart transplant recipients taking a PSI, 16% withdrew treatment in the first year, and 25% had stopped PSI due to severe adverse events by the fourth year."( Withdrawal of proliferation signal inhibitors due to adverse events in the maintenance phase of heart transplantation.
Almenar, L; Arizon Del Prado, JM; Brossa, V; Crespo-Leiro, MG; Delgado, JF; Diaz-Molina, B; Gomez-Bueno, M; Gonzalez-Vilchez, F; Lambert, JL; Martínez-Dolz, L; Mirabet, S; Paniagua, MJ; Perez-Villa, F; Roig, E; Ruiz-Cano, MJ; Segovia, J; Vazquez de Prada, JA, 2012
)
0.38
" This has raised challenges in the management of adverse events (AEs) associated with the six targeted agents approved in RCC-sorafenib, sunitinib, pazopanib, bevacizumab (in combination with interferon alpha), temsirolimus, and everolimus."( Targeted therapies for renal cell carcinoma: review of adverse event management strategies.
Eisen, T; Escudier, B; Izzedine, H; Mulders, P; Pyle, L; Robert, C; Sternberg, CN; Zbinden, S, 2012
)
0.56
" Everolimus was generally well tolerated in elderly patients, and most adverse events were grade 1 or 2 in severity."( Efficacy and safety of everolimus in elderly patients with metastatic renal cell carcinoma: an exploratory analysis of the outcomes of elderly patients in the RECORD-1 Trial.
Anak, O; Bracarda, S; Calvo, E; Climent, MA; Escudier, B; Grünwald, V; Hutson, TE; Kim, D; Motzer, RJ; Osanto, S; Panneerselvam, A; Porta, C; Ravaud, A; Vaishampayan, U, 2012
)
1.6
"The XIENCE V stent is safe and effective with low TLR and stent thrombosis rates."( SPIRIT Women, evaluation of the safety and efficacy of the XIENCE V everolimus-eluting stent system in female patients: referral time for coronary intervention and 2-year clinical outcomes.
Grinfeld, L; Li, D; Mauri i Ferré, F; Mikhail, GW; Modena, MG; Morice, MC; Papeleu, P; Rutledge, D; Strasser, RH; Stuteville, M; Sudhir, K; Windecker, S, 2012
)
0.61
" The data of studies and major outcomes include changes in patients' glomerular filtration rate (GFR), urinary protein, total kidney volume (TKV), cyst volume, parenchymal volume, and lipid profile and the frequency of adverse events."( Efficacy and safety of mTOR inhibitor therapy in patients with early-stage autosomal dominant polycystic kidney disease: a meta-analysis of randomized controlled trials.
Chen, J; He, Q; Ji, S; Lin, C, 2012
)
0.38
"Based on the current limited clinical trials, this study suggests that short-duration mTOR inhibitor therapy is relatively safe to slow down the increase in kidney volume in patients with early-stage ADPKD, but it has limited impact on slowing down the decrease in GFR."( Efficacy and safety of mTOR inhibitor therapy in patients with early-stage autosomal dominant polycystic kidney disease: a meta-analysis of randomized controlled trials.
Chen, J; He, Q; Ji, S; Lin, C, 2012
)
0.38
" Most adverse events are present for a short time after the introduction of everolimus, and are manageable."( A drug safety evaluation of everolimus in kidney transplantation.
Åsberg, A; Holdaas, H; Midtvedt, K, 2012
)
0.9
" Correlates of major adverse cardiac events and ST were identified."( Safety and efficacy of everolimus-eluting stents versus sirolimus-eluting stents in women.
Badr, S; Barbash, IM; Dvir, D; Kitabata, H; Loh, JP; Pichard, AD; Sardi, G; Torguson, R; Waksman, R; Xue, Z, 2013
)
0.7
" Dose reduction because of adverse events or intolerance was required in 91% of patients after 26 ± 11 days from the start of treatment."( Adverse events affect sorafenib efficacy in patients with recurrent hepatocellular carcinoma after liver transplantation: experience at a single center and review of the literature.
Airoldi, A; Belli, LS; Cordone, G; Gentiluomo, M; Mancuso, A; Vangeli, M; Viganò, R; Zavaglia, C, 2013
)
0.39
" Adverse events were mostly grade 1 or 2; no patients discontinued treatment because of adverse events."( Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): a multicentre, randomised, placebo-controlled phase 3 trial.
Bebin, EM; Belousova, E; Cauwel, H; Curatolo, P; de Vries, PJ; Flamini, JR; Ford, JP; Franz, DN; Frost, M; Jozwiak, S; Kohrman, MH; Kuperman, R; Lebwohl, D; Sahmoud, T; Shah, G; Sparagana, S; Thiele, EA; Whittemore, VH; Witt, O; Wu, JY, 2013
)
0.7
"In a placebo-controlled trial, the median time to radiologic documentation of cancer worsening or death was extended by about 6 months, although patients experienced many, often serious, adverse effects."( Everolimus 10 mg and pancreatic neuroendocrine tumours: many adverse effects and uncertain benefit.
, 2012
)
1.82
" All serious and grade 3/4 adverse events and grade 1/2 adverse events leading to a change in drug administration were reported."( Safety of everolimus by treatment duration in patients with advanced renal cell cancer in an expanded access program.
Anak, O; Bahl, A; Bavbek, S; Bracarda, S; Grünwald, V; Karakiewicz, P; Kim, D; Ou, YC; Panneerselvam, A; Rha, SY; van den Eertwegh, AJ, 2013
)
0.79
" First occurrence of adverse events presented early in the treatment course for most patients."( Safety of everolimus by treatment duration in patients with advanced renal cell cancer in an expanded access program.
Anak, O; Bahl, A; Bavbek, S; Bracarda, S; Grünwald, V; Karakiewicz, P; Kim, D; Ou, YC; Panneerselvam, A; Rha, SY; van den Eertwegh, AJ, 2013
)
0.79
" All patients reported ≥1 adverse event (AE), mostly grade 1/2 in severity, consistent with that previously reported, and none led to everolimus discontinuation."( Everolimus long-term safety and efficacy in subependymal giant cell astrocytoma.
Agricola, K; Care, MM; Franz, DN; Krueger, DA; Mays, M; Tudor, C, 2013
)
2.04
"Everolimus therapy is safe and effective for longer term (median exposure 34."( Everolimus long-term safety and efficacy in subependymal giant cell astrocytoma.
Agricola, K; Care, MM; Franz, DN; Krueger, DA; Mays, M; Tudor, C, 2013
)
3.28
" The primary endpoint was major adverse cardiac events."( Safety and efficacy of everolimus- versus sirolimus-eluting stents: a systematic review and meta-analysis of 11 randomized trials.
Hahn, S; Hofma, SH; Kandzari, DE; Kang, HJ; Kang, SH; Kim, HS; Koo, BK; Lee, HY; Lim, WH; Oh, BH; Park, KW; Park, YB; Shin, DH; Van Boven, AJ; Velders, MA; Yang, HM, 2013
)
0.7
" Common grade 3/4 adverse events were anemia (20%), hyperglycemia (13%), increased gamma-glutamyltransferase (11%), hyponatremia (8%), dyspnea (8%), hypertriglyceridemia (6%), and lymphopenia (6%)."( Safety and efficacy of everolimus in Chinese patients with metastatic renal cell carcinoma resistant to vascular endothelial growth factor receptor-tyrosine kinase inhibitor therapy: an open-label phase 1b study.
Gogov, S; Guo, J; Huang, Y; Jappe, A; Pirotta, N; Qin, S; Straub, P; Sun, Y; Wang, H; Ye, Z; Zhang, X; Zhou, F, 2013
)
0.7
" Although everolimus is generally well tolerated, as with most therapies administered in an advanced cancer setting, drug-related adverse events (AEs) inevitably occur."( Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trial.
Peterson, ME, 2013
)
1.01
" End points were major adverse cardiovascular events (MACE: defined as the composite of death, myocardial infarction, or target lesion revascularization), target vessel revascularization, and definite stent thrombosis at 1 year."( Safety and efficacy outcomes of overlapping second-generation everolimus-eluting stents versus first-generation drug-eluting stents.
Badr, S; Barbash, IM; Chen, F; Dvir, D; Kent, KM; Kitabata, H; Loh, JP; Minha, S; Pendyala, LK; Pichard, AD; Satler, LF; Suddath, WO; Torguson, R; Waksman, R, 2013
)
0.63
" This review will focus on the adverse events reported with everolimus in breast cancer trials and will provide practical guidelines for the management of these adverse events."( Everolimus: side effect profile and management of toxicities in breast cancer.
O'Regan, R; Paplomata, E; Zelnak, A, 2013
)
2.07
" The most common adverse events (AEs) were hypertriglyceridemia and anemia, similar to Western patients."( Efficacy and safety of everolimus in Korean patients with metastatic renal cell carcinoma.
Ahn, JB; Kim, HS; Kim, KH; Lee, HJ; Rha, SY; Shin, SJ; Yoon, SH, 2013
)
0.7
"Patients who had been treated with everolimus for advanced renal cell carcinoma at 39 Japanese medical centers between January 2010 and November 2011 were retrospectively investigated to assess adverse events and the time to treatment failure."( Adverse event profile and dose modification of everolimus for advanced renal cell carcinoma in real-world Japanese clinical practice.
Azuma, H; Kanayama, HO; Miki, T; Nakatani, T; Nishimura, K; Nonomura, N; Nozawa, M; Tomita, Y; Tsujihata, M; Ueda, T; Uemura, H; Yoshioka, T, 2013
)
0.92
"01) after experiencing adverse events."( Adverse event profile and dose modification of everolimus for advanced renal cell carcinoma in real-world Japanese clinical practice.
Azuma, H; Kanayama, HO; Miki, T; Nakatani, T; Nishimura, K; Nonomura, N; Nozawa, M; Tomita, Y; Tsujihata, M; Ueda, T; Uemura, H; Yoshioka, T, 2013
)
0.65
"The adverse event profile of everolimus may differ between Japanese and Caucasian patients."( Adverse event profile and dose modification of everolimus for advanced renal cell carcinoma in real-world Japanese clinical practice.
Azuma, H; Kanayama, HO; Miki, T; Nakatani, T; Nishimura, K; Nonomura, N; Nozawa, M; Tomita, Y; Tsujihata, M; Ueda, T; Uemura, H; Yoshioka, T, 2013
)
0.94
" Everolimus along with mycophenolate mofetil or azathioprine and prednisolone as a maintenance immunosuppressive therapy was found to be effective and safe in patients with CNIs withdrawal either due to CAN or CNIT."( Safety and efficacy of everolimus in chronic allograft nephropathy.
Dakshinamurty, KV; Das, U, 2013
)
1.61
" Adverse events (AEs) of special interest (all grades) that occurred more frequently with EVE than with PBO included stomatitis, infections, rash, pneumonitis, and hyperglycemia."( Safety and efficacy of everolimus with exemestane vs. exemestane alone in elderly patients with HER2-negative, hormone receptor-positive breast cancer in BOLERO-2.
Baselga, J; Bourgeois, H; Burris, HA; Campone, M; Csöszi, T; Dakhil, S; Gnant, M; Gonzalez Martin, A; Heng, D; Hortobagyi, GN; Ito, Y; Noguchi, S; Osborne, K; Panneerselvam, A; Piccart, M; Pritchard, KI; Puttawibul, P; Rugo, HS; Sahmoud, T; Srimuninnimit, V; Taran, T, 2013
)
0.7
" However, a low rate of tumor reduction and some adverse events have been pointed out."( Therapeutic potential and adverse events of everolimus for treatment of hepatocellular carcinoma - systematic review and meta-analysis.
Galli, U; Gao, C; Hatano, E; Houben, P; Lin, S; Petrulionis, M; Richter, S; Schemmer, P; Schultze, D; Winkler, S; Yamanaka, K, 2013
)
0.65
"Mammalian target of rapamycin (mTOR) inhibitors are associated with dermatological adverse events."( Everolimus-induced human keratinocytes toxicity is mediated by STAT3 inhibition.
Bito, T; Hirai, M; Hirano, T; Kume, M; Makimoto, H; Mukai, A; Nishigori, C; Uda, A; Yamamoto, K; Yamashita, K, 2013
)
1.83
" Adverse events led to temporary SORA discontinuation in 2 patients (28."( Efficacy and safety of combination therapy with everolimus and sorafenib for recurrence of hepatocellular carcinoma after liver transplantation.
Bargellini, I; Bartolozzi, C; Campani, D; Carrai, P; Cioni, D; Crocetti, L; De Simone, P; Della Pina, C; Filipponi, F; Ghinolfi, D; Lencioni, R; Leonardi, G; Pezzati, D; Pollina, L,
)
0.39
"Everolimus has important clinical activity in various malignancies, but its use can be complicated by respiratory adverse events."( Diagnostic challenges of respiratory adverse events during everolimus treatment.
de Boer, M; De Vos, FY; Jansen, A; Tjan-Heijnen, VC; van Herpen, CM; Willemsen, AE, 2014
)
2.09
" We report on the incidence, time course, severity, and resolution of treatment-emergent adverse events (AEs) as well as incidence of dose modifications during the extended follow-up of this study."( Incidence and time course of everolimus-related adverse events in postmenopausal women with hormone receptor-positive advanced breast cancer: insights from BOLERO-2.
Baselga, J; Bauly, H; Burris, HA; Chouinard, E; Csoszi, T; Eakle, J; El-Hashimy, M; Feng, W; Geberth, M; Gnant, M; Hortobagyi, G; Noguchi, S; Perez, A; Piccart, M; Pritchard, KI; Puttawibul, P; Rugo, HS; Srimuninnimit, V; Taran, T, 2014
)
0.69
" End points included probable or definite stent thrombosis and major adverse cardiovascular events (MACE), defined as a composite of all-cause death, Q-wave myocardial infarction, and target lesion revascularization up to 3 years."( Long-term safety and efficacy of second-generation everolimus-eluting stents compared to other limus-eluting stents and bare metal stents in patients with acute coronary syndrome.
Kitabata, H; Loh, JP; Magalhaes, MA; Omar, A; Pendyala, LK; Pichard, AD; Satler, LF; Suddath, WO; Torguson, R; Waksman, R, 2014
)
0.65
"We discuss clinically relevant everolimus-related adverse events from the phase III studies, including stomatitis, noninfectious pneumonitis, rash, selected metabolic abnormalities, and infections, with focus on appropriate clinical management of these events and specific considerations in patients with breast cancer."( Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer.
Aapro, M; Andre, F; Blackwell, K; Calvo, E; Jahanzeb, M; Papazisis, K; Porta, C; Pritchard, K; Ravaud, A, 2014
)
0.92
"The majority of adverse events experienced during everolimus therapy are of mild to moderate severity."( Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer.
Aapro, M; Andre, F; Blackwell, K; Calvo, E; Jahanzeb, M; Papazisis, K; Porta, C; Pritchard, K; Ravaud, A, 2014
)
0.89
"As with all orally administered agents, education of both physicians and patients in the management of adverse events for patients receiving everolimus is critical to achieving optimal exposure and clinical benefit."( Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer.
Aapro, M; Andre, F; Blackwell, K; Calvo, E; Jahanzeb, M; Papazisis, K; Porta, C; Pritchard, K; Ravaud, A, 2014
)
0.84
"Nineteen patients who discontinued treatment with vascular endothelial growth factor receptor-tyrosine kinase inhibitors because of disease progression or adverse events were administered everolimus."( Efficacy of everolimus in patients with advanced renal cell carcinoma refractory or intolerant to VEGFR-TKIs and safety compared with prior VEGFR-TKI treatment.
Fujioka, T; Iwasaki, K; Kato, R; Kato, Y; Matsuura, T; Obara, W, 2014
)
0.97
" Two patients discontinued everolimus treatment because of adverse events."( Efficacy of everolimus in patients with advanced renal cell carcinoma refractory or intolerant to VEGFR-TKIs and safety compared with prior VEGFR-TKI treatment.
Fujioka, T; Iwasaki, K; Kato, R; Kato, Y; Matsuura, T; Obara, W, 2014
)
1.08
" The adverse event profiles of everolimus and vascular endothelial growth factor receptor-tyrosine kinase inhibitors were different."( Efficacy of everolimus in patients with advanced renal cell carcinoma refractory or intolerant to VEGFR-TKIs and safety compared with prior VEGFR-TKI treatment.
Fujioka, T; Iwasaki, K; Kato, R; Kato, Y; Matsuura, T; Obara, W, 2014
)
1.07
" At 2 years, the incidence of major adverse cardiovascular events was 13."( Long-term safety and efficacy of the everolimus-eluting stent compared to first-generation drug-eluting stents in contemporary clinical practice.
Badr, S; Barbash, IM; Ben-Dor, I; Loh, JP; Minha, S; Pichard, AD; Satler, LF; Torguson, R; Waksman, R, 2014
)
0.68
" Two patients required dose reduction due to adverse events."( Activity and safety of RAD001 (everolimus) in patients affected by biliary tract cancer progressing after prior chemotherapy: a phase II ITMO study.
Alabiso, O; Bajetta, E; Bertolini, A; Buzzoni, R; Cantore, M; Ciarlo, A; De Braud, F; Iannacone, C; Mambrini, A; Mazzaferro, V; Platania, M; Pusceddu, S; Turco, C, 2014
)
0.69
"Small vessel diameter and long lesion length are independently associated with increased risk of adverse cardiac events after drug-eluting stent implantation."( Two-year safety and effectiveness of the platinum chromium everolimus-eluting stent for the treatment of small vessels and longer lesions.
Allocco, DJ; Cannon, LA; Dawkins, KD; Dens, J; Dubois, CL; Feldman, RL; Lee, TC; Meredith, IT; Mooney, MR; Pompili, VJ; Rabinowitz, AC; Saito, S; Stone, GW; Teirstein, PS, 2015
)
0.66
" Thus, the adverse effects, described hereby, may be considered as « positive events », predicting efficacy, and thus looked for… Moreover, the sequential approach, with new drugs, emphasizes the need of defining the optimal sequence."( [Revision of therapeutic index for targeted treatment in kidney cancer: What if toxicity could predict efficacy?].
Brugères-Chakiba, C; Chaminade, A; Grellety, T; Gross-Goupil, M; Ravaud, A; Roubaud, G, 2014
)
0.4
"05) in the incidence of discontinuations and serious adverse events between the groups."( Safety and efficacy of the early introduction of everolimus with reduced-exposure cyclosporine a in de novo kidney recipients.
Ha, J; Huh, KH; Kim, YH; Kim, YL; Kim, YS; Oh, CK, 2015
)
0.67
" Because adverse events (AEs) associated with everolimus might differ from AEs that oncologists who treat patients with BC are more familiar with, everolimus AEs and their effective management are reviewed in this article."( Adverse event management of mTOR inhibitors during treatment of hormone receptor-positive advanced breast cancer: considerations for oncologists.
Yardley, DA, 2014
)
0.66
" Discontinuation due to adverse events occurred in three everolimus patients (7."( Efficacy and safety of conversion from cyclosporine to everolimus in living-donor kidney transplant recipients: an analysis from the ZEUS study.
Arns, W; Budde, K; Eisenberger, U; Hauser, IA; Heller, K; Klehr, HU; Lehner, F; May, C; Mühlfeld, A; Nadalin, S; Paulus, EM; Porstner, M; Reinke, P; Sommerer, C; Stahl, R; Stangl, M; Suwelack, B; Witzke, O; Wolters, HH; Wüthrich, RP; Zeier, M, 2014
)
0.9
" However, as other immunosuppressive drugs, mTOR-I may induce the development of several adverse effects (e."( [Extra-renal adverse effects of mTOR inhibitors: know them to optimize their use in renal transplantation].
Caletti, C; Granata, S; Lupo, A; Tomei, P; Zaza, G,
)
0.13
" Adverse clinical events were independently adjudicated."( Safety of second-generation drug-eluting stents three years after randomised use in the TWENTE trial.
de Man, FH; Doggen, CJ; Lam, MK; Linssen, GC; Louwerenburg, JH; Löwik, MM; Sen, H; Stoel, MG; Tandjung, K; van Houwelingen, KG; von Birgelen, C, 2015
)
0.42
" In patients with biliary tract cancer, everolimus is safe but associated with a higher incidence of adverse events."( Safety profile and treatment response of everolimus in different solid tumors: an observational study.
Bajetta, E; Biondani, P; Buzzoni, R; Damato, A; de Braud, F; DeBraud, F; Garanzini, E; Grassi, P; Procopio, G; Pusceddu, S; Testa, I; Verzoni, E, 2014
)
0.94
" This study estimates the costs of managing adverse events (AEs) during EVE + EXE therapy and single-agent chemotherapy in Western Europe."( Cost of adverse events during treatment with everolimus plus exemestane or single-agent chemotherapy in patients with advanced breast cancer in Western Europe.
Campone, M; Faust, E; Gao, H; Kageleiry, A; Signorovitch, JE; Yang, H; Zhang, J, 2014
)
0.66
" The most costly adverse event among all patients treated with EVE + EXE was anemia (on average €152 per patient)."( Cost of adverse events during treatment with everolimus plus exemestane or single-agent chemotherapy in patients with advanced breast cancer in Western Europe.
Campone, M; Faust, E; Gao, H; Kageleiry, A; Signorovitch, JE; Yang, H; Zhang, J, 2014
)
0.66
" The purpose of this study is to compare the adverse event profiles of both agents and to estimate the risk factors for non-infectious pneumonitis in patients with advanced renal cell carcinoma on the basis of our real-world clinical experience."( Differences in adverse event profiles between everolimus and temsirolimus and the risk factors for non-infectious pneumonitis in advanced renal cell carcinoma.
Anai, S; Fujimoto, K; Fukasawa, S; Hongo, F; Miki, T; Nakatani, T; Nozawa, M; Ohzeki, T; Tamada, S; Uemura, H, 2015
)
0.68
" Chi-squared test and univariate and multivariate logistic regression analyses were performed to investigate the differences in adverse event profiles and the risk factors associated with non-infectious pneumonitis, respectively."( Differences in adverse event profiles between everolimus and temsirolimus and the risk factors for non-infectious pneumonitis in advanced renal cell carcinoma.
Anai, S; Fujimoto, K; Fukasawa, S; Hongo, F; Miki, T; Nakatani, T; Nozawa, M; Ohzeki, T; Tamada, S; Uemura, H, 2015
)
0.68
"Our findings suggest that adverse event profiles may differ between everolimus and temsirolimus and that non-infectious pneumonitis may occur more frequently in patients treated with everolimus than temsirolimus."( Differences in adverse event profiles between everolimus and temsirolimus and the risk factors for non-infectious pneumonitis in advanced renal cell carcinoma.
Anai, S; Fujimoto, K; Fukasawa, S; Hongo, F; Miki, T; Nakatani, T; Nozawa, M; Ohzeki, T; Tamada, S; Uemura, H, 2015
)
0.91
" Amenorrhea occurred as a rare adverse event of everolimus."( Amenorrhea as a rare drug-related adverse event associated with everolimus for pancreatic neuroendocrine tumors.
Ito, H; Kawaguchi, Y; Kawashima, Y; Maruno, A; Mine, T; Ogawa, M, 2014
)
0.9
"Biodegradable-polymer drug-eluting stents (BP-DES) were developed to be as effective as second-generation durable-polymer drug-eluting stents (DP-DES) and as safe >1 year as bare-metal stents (BMS)."( Long-term efficacy and safety of biodegradable-polymer biolimus-eluting stents: main results of the Basel Stent Kosten-Effektivitäts Trial-PROspective Validation Examination II (BASKET-PROVE II), a randomized, controlled noninferiority 2-year outcome tria
Alber, H; Buser, P; Conen, D; Eberli, F; Galatius, S; Gilgen, N; Hoffmann, A; Jeger, R; Kaiser, C; Kurz, DJ; Moccetti, T; Müller, C; Naber, C; Pedrazzini, G; Pfisterer, M; Rickenbacher, P; Rickli, H; Skov Jensen, J; Steiner, M; Vogt, DR; Von Felten, S; Vuillomenet, A; Wadt Hansen, K; Wanitschek, M; Weilenmann, D, 2015
)
0.42
" We analyzed several variables associated with this treatment, including patient characteristics, time from liver transplantation to conversion to everolimus, immunosuppression regimens before and after conversion, treatment efficacy, adverse events, and discontinuation after conversion."( Immunosuppression based on everolimus in liver transplant recipients with severe early post-transplantation neurotoxicity.
Bilbao, I; Caralt, M; Castells, L; Charco, R; Dopazo, C; Lazaro, J; Sapisochin, G, 2014
)
0.9
" Patients were followed up to 2 years for major adverse cardiac events (MACE)."( Safety and efficacy of everolimus-eluting stents compared with first-generation drug-eluting stents in patients undergoing primary percutaneous coronary intervention.
Baker, NC; Escárcega, RO; Lipinski, MJ; Magalhaes, MA; Minha, S; Pichard, AD; Satler, LF; Suddath, WO; Torguson, R; Waksman, R,
)
0.44
" All grades of common adverse events (AEs) were consistent with the known safety profile of everolimus."( Efficacy and Safety of Sequential Use of Everolimus in Patients With Metastatic Renal Cell Carcinoma Previously Treated With Bevacizumab With or Without Interferon Therapy: Results From the European AVATOR Study.
Emmanouilides, C; Guillot, A; Jacobasch, L; Kelkouli, N; Kim, S; Nguyen Tan Hon, T; Papandreou, C; Schmitz, J; Slimane, K; Theodore, C; Thiery-Vuillemin, A, 2015
)
0.9
"There is a probable causal relationship between the study medication and the reported serious adverse events."( Phase II study evaluating efficacy and safety of everolimus with letrozole for management of advanced (unresectable or metastatic) non-small cell lung cancer after failure of platinum-based treatment: a preliminary analysis of toxicity.
Brown, MP; Singhal, N; Vatandoust, S, 2015
)
0.67
" Adverse events led to discontinuation of everolimus and CNI in 32."( Renal, efficacy and safety outcomes following late conversion of kidney transplant patients from calcineurin inhibitor therapy to everolimus: the randomized APOLLO study.
Arns, W; Baeumer, D; Budde, K; Haller, H; May, C; Porstner, M; Rath, T; Reinke, P; Sommerer, C; Suwelack, B; Witzke, O, 2015
)
0.89
" The analyzed clinical parameters were the 1-year rates of death, Q-wave myocardial infarction (MI), target lesion revascularization (TLR), target vessel revascularization (TVR), definite stent thrombosis (ST) and major adverse cardiac event (MACE) defined as the composite of death, MI, or TLR at 1-year."( Safety and efficacy of limus-eluting stents and balloon angioplasty for sirolimus-eluting in-stent restenosis.
Chen, F; Kitabata, H; Mahmoudi, M; Ota, H; Pichard, AD; Satler, LF; Suddath, WO; Torguson, R; Waksman, R, 2015
)
0.42
" Other objectives are: to assess the incidence of treated biopsy-proven acute rejection, graft loss, or death; the incidences of components of the composite efficacy endpoint; renal function via estimated glomerular filtration rate using various formulae (MDRD-4, Nankivell, Cockcroft-Gault, chronic kidney disease epidemiology collaboration and Hoek formulae); the incidence of proteinuria; the incidence of adverse events and serious adverse events; the incidence and severity of cytomegalovirus and HCV infections and HCV-related fibrosis."( Evaluating the efficacy, safety and evolution of renal function with early initiation of everolimus-facilitated tacrolimus reduction in de novo liver transplant recipients: Study protocol for a randomized controlled trial.
Braun, F; Dworak, M; Nashan, B; Schemmer, P; Schlitt, H; Wimmer, P, 2015
)
0.64
" Procedural and in-hospital clinical outcomes were examined in addition to the 1-year primary endpoint of death, myocardial infarction, and target lesion revascularization (major adverse cardiac events [MACE])."( Safety and Effectiveness of Everolimus-Eluting Stents in Chronic Total Coronary Occlusion Revascularization: Results From the EXPERT CTO Multicenter Trial (Evaluation of the XIENCE Coronary Stent, Performance, and Technique in Chronic Total Occlusions).
Grantham, JA; Kandzari, DE; Karmpaliotis, D; Kini, AS; Larracas, C; Lembo, NJ; Lombardi, W; Moses, JW; Nicholson, WJ; Orr, C; Popma, JJ; Rutledge, DR; Tummala, PE; Wang, J, 2015
)
0.71
"4%), treatment with EES was associated with significantly lower composite adverse events for both intent-to-treat (18."( Safety and Effectiveness of Everolimus-Eluting Stents in Chronic Total Coronary Occlusion Revascularization: Results From the EXPERT CTO Multicenter Trial (Evaluation of the XIENCE Coronary Stent, Performance, and Technique in Chronic Total Occlusions).
Grantham, JA; Kandzari, DE; Karmpaliotis, D; Kini, AS; Larracas, C; Lembo, NJ; Lombardi, W; Moses, JW; Nicholson, WJ; Orr, C; Popma, JJ; Rutledge, DR; Tummala, PE; Wang, J, 2015
)
0.71
" The analyzed clinical parameters were the 1-year rates of death, Q-wave myocardial infarction (MI), target lesion revascularization (TLR), target vessel revascularization (TVR), definite stent thrombosis (ST) and major adverse cardiac events (MACE) defined as the composite of death, MI, or TLR at 1-year."( Safety and efficacy of everolimus-eluting stents for bare-metal in-stent restenosis.
Mahmoudi, M; Ota, H; Pichard, AD; Satler, LF; Suddath, WO; Torguson, R; Waksman, R,
)
0.44
"The most frequent side-effect of CNIs is nephrotoxicity, which in the long term can contribute, to allograft deterioration."( The safety of calcineurin inhibitors for kidney-transplant patients.
Malvezzi, P; Rostaing, L, 2015
)
0.42
"gov updated to Mar 2014 with safety [major adverse cardiac events (MACE)], all-cause mortality, non-fatal myocardial infarction (MI), stent thrombosis (ST) and efficacy [target vessel revascularization (TVR), target lesion revascularization (TLR), target vessel failure (TVF), target lesion failure (TLF)] endpoints and follow-up of ≥12 months were identified."( Safety and efficacy of everolimus-eluting stent versus zotarolimus-eluting stent: A meta-analysis of randomized controlled clinical trials and observational studies.
Gu, H; Hua, K; Li, W; Wang, Y; Yang, J, 2015
)
0.73
"Sirolimus should not be started within the first month after liver transplantation (LT) because of an increased risk of adverse outcomes."( Everolimus is safe within the first month after liver transplantation.
Barrera, P; Briceño, J; Ciria, R; De la Mata, M; Ferrín, G; González, V; Guerrero-Misas, M; Montero, JL; Pérez-Medrano, I; Poyato, A; Pozo, JC; Rodríguez-Perálvarez, M; Sánchez-Frías, M, 2015
)
1.86
"Everolimus combined with reduced tacrolimus proved to be safe within the first month after LT."( Everolimus is safe within the first month after liver transplantation.
Barrera, P; Briceño, J; Ciria, R; De la Mata, M; Ferrín, G; González, V; Guerrero-Misas, M; Montero, JL; Pérez-Medrano, I; Poyato, A; Pozo, JC; Rodríguez-Perálvarez, M; Sánchez-Frías, M, 2015
)
3.3
"In pediatric renal transplantation, an everolimus-based regimen with low-dose cyclosporine yields comparable four year results as a standard regimen, but with a different side effect profile."( Efficacy and Safety of an Everolimus- vs. a Mycophenolate Mofetil-Based Regimen in Pediatric Renal Transplant Recipients.
Ahlenstiel-Grunow, T; Bald, M; Brunkhorst, LC; Burmeister, G; Fichtner, A; Höcker, B; Krupka, K; Pape, L; Tönshoff, B; Zapf, A, 2015
)
0.99
" Endpoints analyzed included: (1) the composite of all-cause death and non-fatal myocardial infarction (MI); and (2) the composite safety endpoint of major adverse cardiovascular events (MACEs), including death, MI and symptom-driven target lesion revascularization (TLR)."( Evaluation of the Safety of Everolimus-Eluting Bioresorbable Vascular Scaffold (BVS) Implantation in Patients With Chronic Total Coronary Occlusions: Acute Procedural and Short-Term Clinical Results.
Colombo, A; Erdogan, E; Goktekin, O; Latib, A; Panoulas, VF; Sato, K; Shah, I; Tastan, A; Uyarel, H; Yamac, AH, 2015
)
0.71
"Treatment of CTOs with BVS seems to be safe and effective, with a high technical success rate and acceptable MACE at short-term follow-up."( Evaluation of the Safety of Everolimus-Eluting Bioresorbable Vascular Scaffold (BVS) Implantation in Patients With Chronic Total Coronary Occlusions: Acute Procedural and Short-Term Clinical Results.
Colombo, A; Erdogan, E; Goktekin, O; Latib, A; Panoulas, VF; Sato, K; Shah, I; Tastan, A; Uyarel, H; Yamac, AH, 2015
)
0.71
" Theoretically, since vessel scaffolding is temporary and no permanent implant remains in the vessel, BRSs, as opposed to metal stents, once degraded would no longer be potential triggers for stent-related adverse events or side effects."( Evaluation of the short- and long-term safety and therapy outcomes of the everolimus-eluting bioresorbable vascular scaffold system in patients with coronary artery stenosis: Rationale and design of the German-Austrian ABSORB RegIstRy (GABI-R).
Achenbach, S; Biermann, J; Hamm, CW; Kastner, J; Limbourg, T; Mehilli, J; Münzel, T; Naber, C; Nef, H; Neumann, T; Pfannebecker, T; Richardt, G; Schmermund, A; Schneider, S; Wiebe, J; Wöhrle, J; Zahn, R,
)
0.36
" To provide optimal prevention and management strategies, it is crucial that clinicians are aware of the adverse events (AEs) associated with mTOR inhibition."( Dosing and Safety Implications for Oncologists When Administering Everolimus to Patients With Hormone Receptor-Positive Breast Cancer.
Rugo, HS, 2016
)
0.67
" No unexpected adverse events were reported."( Retrospective analysis on safety and efficacy of everolimus in treatment of metastatic renal cancer patients receiving dialysis.
Cosmai, L; del Giovane, C; Di Lorenzo, G; Donati, D; Guida, A; Masini, C; Mighali, P; Milella, M; Porta, C; Prati, V; Sabbatini, R; Santoni, M, 2015
)
0.67
"Everolimus appears to be safe in mRCC patients with renal impairment or end-stage renal disease requiring dialysis."( Retrospective analysis on safety and efficacy of everolimus in treatment of metastatic renal cancer patients receiving dialysis.
Cosmai, L; del Giovane, C; Di Lorenzo, G; Donati, D; Guida, A; Masini, C; Mighali, P; Milella, M; Porta, C; Prati, V; Sabbatini, R; Santoni, M, 2015
)
2.11
" The increased incidence of serious adverse events in early studies halted the enthusiasm as a kidney sparing alternative to calcineurin inhibitors (CNI)."( Safety of mTOR inhibitors in adult solid organ transplantation.
Campistol, JM; Diekmann, F; Ventura-Aguiar, P, 2016
)
0.43
"In the authors' opinion, mTOR inhibitors are a safe alternative to standard immunosuppression therapy with CNI and mycophenolate/azathioprine."( Safety of mTOR inhibitors in adult solid organ transplantation.
Campistol, JM; Diekmann, F; Ventura-Aguiar, P, 2016
)
0.43
"Everolimus-based immunosuppression regimen without corticosteroids and/or induction immediately after liver transplantation seems to be safe and effective when administered with low doses of calcineurin-inhibitor or mycophenolate; although these findings require further investigation, these regimens could avoid adverse effects of standard immunosuppression regimens with higher doses."( The efficacy and safety of mammalian target of rapamycin inhibitors ab initio after liver transplantation without corticosteroids or induction therapy.
Angelico, R; Belardi, C; Cillis, A; Katari, R; Manzia, TM; Mogul, A; Orlando, G; Quaranta, C; Sforza, D; Tariciotti, L; Tisone, G; Toti, L, 2016
)
1.88
"In percutaneous coronary intervention (PCI) patients new-generation drug-eluting stent (DES) has reduced adverse events in comparison to early-generation DES."( Three-year efficacy and safety of new- versus early-generation drug-eluting stents for unprotected left main coronary artery disease insights from the ISAR-LEFT MAIN and ISAR-LEFT MAIN 2 trials.
Byrne, RA; Cassese, S; Fusaro, M; Ibrahim, T; Kastrati, A; Kreutzer, J; Kufner, S; Laugwitz, KL; Mehilli, J; Schunkert, H; Tiroch, K; Tölg, R; Valgimigli, M; Xhepa, E, 2016
)
0.43
" The primary endpoint was the composite of death, myocardial infarction (MI), target lesion revascularization and stroke (MACCE, major adverse cardiac and cerebrovascular event)."( Three-year efficacy and safety of new- versus early-generation drug-eluting stents for unprotected left main coronary artery disease insights from the ISAR-LEFT MAIN and ISAR-LEFT MAIN 2 trials.
Byrne, RA; Cassese, S; Fusaro, M; Ibrahim, T; Kastrati, A; Kreutzer, J; Kufner, S; Laugwitz, KL; Mehilli, J; Schunkert, H; Tiroch, K; Tölg, R; Valgimigli, M; Xhepa, E, 2016
)
0.43
" The novel combination of everolimus and exemestane has been shown to prolong progression-free survival but with increased adverse events compared to exemestane alone."( Occurrence and characterization of everolimus adverse events during first and subsequent cycles in the treatment of metastatic breast cancer.
Berger, MJ; Mrozek, E; Phillips, G; Vargo, CA, 2016
)
1.01
" The most common adverse events leading to dose reduction or interruption was stomatitis (57."( Occurrence and characterization of everolimus adverse events during first and subsequent cycles in the treatment of metastatic breast cancer.
Berger, MJ; Mrozek, E; Phillips, G; Vargo, CA, 2016
)
0.71
" This early onset of adverse events requires thorough patient education and close clinical monitoring during the first 28 days of therapy."( Occurrence and characterization of everolimus adverse events during first and subsequent cycles in the treatment of metastatic breast cancer.
Berger, MJ; Mrozek, E; Phillips, G; Vargo, CA, 2016
)
0.71
"Everolimus appears to be a safe therapeutic option for patients aged <3 years with TSC-associated SEGA."( Safety of Everolimus in Patients Younger than 3 Years of Age: Results from EXIST-1, a Randomized, Controlled Clinical Trial.
Berkowitz, N; Brechenmacher, T; Franz, DN; Jóźwiak, S; Kotulska, K, 2016
)
2.28
" Given that increasing number of patients with TSC receive mTOR inhibitors, the issue of adverse events associated with this therapy becomes practically important."( Management of side effects of mTOR inhibitors in tuberous sclerosis patients.
Jóźwiak, S; Kotulska, K; Sadowski, K, 2016
)
0.43
" Primary endpoint was a composite of major adverse cardiac events (MACE), including cardiac death, myocardial infarction (MI), target vessel revascularization (TVR), and definite stent thrombosis."( Safety and Efficacy of Everolimus- Versus Sirolimus-Eluting Stents: 5-Year Results From SORT OUT IV.
Berencsi, K; Christiansen, EH; Hansen, HS; Hansen, KN; Jensen, LO; Junker, A; Kaltoft, A; Krusell, L; Lassen, JF; Maeng, M; Ravkilde, J; Thayssen, P; Tilsted, HH, 2016
)
0.74
" Conversion to MPS was associated with a higher incidence of adverse events."( Efficacy and Safety of Elective Conversion From Sotrastaurin (STN) to Tacrolimus (TAC) or Mycophenolate (MPS) in Stable Kidney Transplant Recipients.
Aguiar, W; Campos, E; Cristelli, M; Felipe, C; Ferreira, A; Franco, M; Gerbase de Lima, M; Hannun, P; Medina-Pestana, J; Sandes-Freitas, T; Tedesco-Silva, H, 2016
)
0.43
" Most adverse events were either grade 1 or 2 and easy manageable with a dose reduction or temporary interruption and only rarely requiring discontinuation."( Efficacy and Safety of Everolimus in Extrapancreatic Neuroendocrine Tumor: A Comprehensive Review of Literature.
Agrimi, D; Aversano, M; Bassi, V; Colao, A; Faggiano, A; Giordano, EA; Guarnotta, V; Logoluso, FA; Malandrino, P; Messina, E; Modica, R; Nicastro, V; Nuzzo, V; Sciaraffia, M, 2016
)
0.74
"Treatment with everolimus in patients with extrapancreatic NETs appears to be a promising strategy that is safe and well tolerated."( Efficacy and Safety of Everolimus in Extrapancreatic Neuroendocrine Tumor: A Comprehensive Review of Literature.
Agrimi, D; Aversano, M; Bassi, V; Colao, A; Faggiano, A; Giordano, EA; Guarnotta, V; Logoluso, FA; Malandrino, P; Messina, E; Modica, R; Nicastro, V; Nuzzo, V; Sciaraffia, M, 2016
)
1.1
" No severe adverse events were usually reported and discontinuation was rarely required; thus, everolimus should be considered a valid therapeutic option for extrapancreatic NETs."( Efficacy and Safety of Everolimus in Extrapancreatic Neuroendocrine Tumor: A Comprehensive Review of Literature.
Agrimi, D; Aversano, M; Bassi, V; Colao, A; Faggiano, A; Giordano, EA; Guarnotta, V; Logoluso, FA; Malandrino, P; Messina, E; Modica, R; Nicastro, V; Nuzzo, V; Sciaraffia, M, 2016
)
0.96
" The most common adverse events (AEs) were asthenia, hypertension, and diarrhea."( Safety and clinical activity of vascular endothelial growth factor receptor (VEGFR)-tyrosine kinase inhibitors after programmed cell death 1 inhibitor treatment in patients with metastatic clear cell renal cell carcinoma.
Amin, A; Doyle, J; Geynisman, DM; Hammers, HJ; McDermott, DF; Motzer, R; Nadal, R; Plimack, ER; Rini, B; Viudez, A; Voss, MH; Weinstock, M; Zhang, Z, 2016
)
0.43
" Many patients treated with the registered fixed 10 mg dose once daily are in need of dose interruptions, reductions or treatment discontinuation due to severe adverse events."( Everolimus pharmacokinetics and its exposure-toxicity relationship in patients with thyroid cancer.
de Wit, D; den Hartigh, J; Gelderblom, H; Guchelaar, HJ; Kapiteijn, E; Links, TP; Moes, DJ; Roozen, CF; Schneider, TC; van der Hoeven, JJ; van Erp, NP, 2016
)
1.88
" Although the pleural effusion reduced markedly after 5 weeks, stomatitis, diarrhea, melena, and interstitial pneumonia occurred as adverse events."( [An Elderly Patient with Metastatic Breast Cancer Who Developed Severe Adverse Events such as Stomatitis and Interstitial Pneumonia after Everolimus plus Exemestane Treatment].
Goto, Y; Kimura, M; Sakiyama, K; Yoshida, T, 2016
)
0.64
" Adverse events observed were stomatitis (35."( The efficacy and safety of everolimus for the treatment of progressive gastroenteropancreatic neuroendocrine tumors: A multi-institution observational study in Taiwan.
Chen, JS; Chen, LT; Chen, MH; Chen, YY; Chou, WC; Ku, FC; Liu, CT; Lu, CH; Shan, YS; Su, YL, 2016
)
0.73
" The primary objective was safety of EVE plus EXE based on frequency of adverse events (AEs), and serious AEs (SAEs)."( Safety of everolimus plus exemestane in patients with hormone-receptor-positive, HER2-negative locally advanced or metastatic breast cancer progressing on prior non-steroidal aromatase inhibitors: primary results of a phase IIIb, open-label, single-arm, e
Bianchetti, S; Camozzi, M; Ciruelos, EM; Conte, P; Gavila, JG; Generali, D; Jerusalem, G; Lang, I; Lorizzo, K; Mardiak, J; Mariani, G; Martin, M; Michelotti, A; Montemurro, F; Naume, B; Neven, P; Simoncini, E; Tjan-Heijnen, VC, 2016
)
0.84
" The most common grade 3-4 adverse events were haematological; the most common of these was grade 4 neutropenia in 18 (75%) of 24 patients."( Everolimus combined with R-CHOP-21 for new, untreated, diffuse large B-cell lymphoma (NCCTG 1085 [Alliance]): safety and efficacy results of a phase 1 and feasibility trial.
Ansell, SM; Colgan, JP; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, B; McPhail, E; Micallef, IN; Nowakowski, GS; Witzig, TE, 2016
)
1.88
"Due to toxic events, everolimus dosage was reduced to 5 mg in 27% of patients."( Safety analysis, association with response and previous treatments of everolimus and exemestane in 181 metastatic breast cancer patients: A multicenter Italian experience.
Cortesi, E; D'Onofrio, L; Fabbri, MA; Gamucci, T; Giuliani, R; Iezzi, L; Magri, V; Mancini, ML; Marchetti, P; Mauri, M; Mentuccia, L; Moscetti, L; Natoli, C; Pizzuti, L; Ramponi, S; Roma, CL; Ruggeri, EM; Santini, D; Sini, V; Sperduti, I; Vaccaro, A; Vici, P, 2016
)
0.99
"The association of everolimus and exemestane has confirmed to be a safe and effective treatment for endocrine sensitive MBC patients even in routine clinical practice."( Safety analysis, association with response and previous treatments of everolimus and exemestane in 181 metastatic breast cancer patients: A multicenter Italian experience.
Cortesi, E; D'Onofrio, L; Fabbri, MA; Gamucci, T; Giuliani, R; Iezzi, L; Magri, V; Mancini, ML; Marchetti, P; Mauri, M; Mentuccia, L; Moscetti, L; Natoli, C; Pizzuti, L; Ramponi, S; Roma, CL; Ruggeri, EM; Santini, D; Sini, V; Sperduti, I; Vaccaro, A; Vici, P, 2016
)
1
" We summarize the nephrotoxic and diabetogenic mechanisms of mTORi after pancreas transplant, the alternatives to minimize these effects, and report on other adverse events."( mTOR inhibitors in pancreas transplant: adverse effects and drug-drug interactions.
Fernandes-Silva, G; Ivani de Paula, M; Rangel, ÉB, 2017
)
0.46
"To systematically review relevant literature comparing the oncological outcomes and adverse events of different systemic therapies for patients with metastatic non-ccRCC."( A Systematic Review and Meta-analysis Comparing the Effectiveness and Adverse Effects of Different Systemic Treatments for Non-clear Cell Renal Cell Carcinoma.
Albiges, L; Bensalah, K; Bex, A; Canfield, SE; Dabestani, S; Fernández-Pello, S; Giles, RH; Hofmann, F; Hora, M; Kuczyk, MA; Lam, TB; Ljungberg, B; Marconi, L; Merseburger, AS; Powles, T; Staehler, M; Tahbaz, R; Volpe, A, 2017
)
0.46
" Sunitinib was associated with more Grade 3-4 adverse events than everolimus, although this was not statistically significant."( A Systematic Review and Meta-analysis Comparing the Effectiveness and Adverse Effects of Different Systemic Treatments for Non-clear Cell Renal Cell Carcinoma.
Albiges, L; Bensalah, K; Bex, A; Canfield, SE; Dabestani, S; Fernández-Pello, S; Giles, RH; Hofmann, F; Hora, M; Kuczyk, MA; Lam, TB; Ljungberg, B; Marconi, L; Merseburger, AS; Powles, T; Staehler, M; Tahbaz, R; Volpe, A, 2017
)
0.69
"Patients with diabetes and coronary artery disease remain at high risk for adverse cardiovascular events after percutaneous coronary intervention."( Efficacy and safety of everolimus and zotarolimus-eluting stents versus first-generation drug-eluting stents in patients with diabetes: A meta-analysis of randomized trials.
Baber, U; Bavishi, C; Dangas, GD; Kini, AS; Moreno, P; Panwar, S; Pirrotta, S; Sharma, SK; Tamis-Holland, J, 2017
)
0.77
" Efficacy [target vessel revascularization (TVR) and target lesion revascularization (TLR)] and safety [major adverse cardiac events (MACE), all-cause and cardiac mortality, myocardial infarction, stent thrombosis] outcomes were evaluated."( Efficacy and safety of everolimus and zotarolimus-eluting stents versus first-generation drug-eluting stents in patients with diabetes: A meta-analysis of randomized trials.
Baber, U; Bavishi, C; Dangas, GD; Kini, AS; Moreno, P; Panwar, S; Pirrotta, S; Sharma, SK; Tamis-Holland, J, 2017
)
0.77
"In patients with diabetes and coronary artery disease undergoing stenting, EES is the most efficacious and safe DES."( Efficacy and safety of everolimus and zotarolimus-eluting stents versus first-generation drug-eluting stents in patients with diabetes: A meta-analysis of randomized trials.
Baber, U; Bavishi, C; Dangas, GD; Kini, AS; Moreno, P; Panwar, S; Pirrotta, S; Sharma, SK; Tamis-Holland, J, 2017
)
0.77
" There was not a substantial difference in graft loss, mortality, and the occurrence of adverse events (AEs) or serious AEs."( Efficacy and Safety of Everolimus for Maintenance Immunosuppression of Kidney Transplantation: A Meta-Analysis of Randomized Controlled Trials.
Lei, K; Li, J; Liu, D; Liu, J; Xu, Q; You, R; Zhang, C; Zhu, L, 2017
)
0.77
"The aim of this systematic review and meta-analysis was to evaluate the efficacy-related events and adverse events of 2 different doses of everolimus in kidney transplant recipients."( Safety and Efficacy of Two Different Doses of Everolimus in Kidney Transplantation: a Systematic Review and Meta-Analysis.
Arab-Zozani, M; Ghoddoosi Nejad, D; Hasanpoor, E; Kakemam, E; Mahdavi-Mazdeh, M; Sokhanvar, M, 2016
)
0.89
" The relative risk (RR) and 95% confidence interval (CI) for treated efficacy-related events and adverse events were collected to calculate pooled measures."( Safety and Efficacy of Two Different Doses of Everolimus in Kidney Transplantation: a Systematic Review and Meta-Analysis.
Arab-Zozani, M; Ghoddoosi Nejad, D; Hasanpoor, E; Kakemam, E; Mahdavi-Mazdeh, M; Sokhanvar, M, 2016
)
0.69
" Both adverse events were rarely severe and were managed easily and efficiently."( A prospective observational study on the evaluation of everolimus-related adverse events in metastatic renal cell carcinoma after first-line anti-vascular endothelial growth factor therapy: the AFINITE study in France.
Albiges, L; Eymard, JC; Guillot, A; Joly, F; Kelkouli, N; Laguerre, B; Lebret, T; Nguyen, T; Ravaud, A; Rolland, F; Slimane, K; Spaeth, D, 2017
)
0.7
" However, due to the historical predominance of everolimus in the oncology setting, some physicians who treat TSC patients may be unfamiliar with everolimus-associated adverse events (AEs) and appropriate management strategies."( Management of everolimus-associated adverse events in patients with tuberous sclerosis complex: a practical guide.
Davies, M; Kingswood, JC; Saxena, A, 2017
)
1.07
"This meta-analysis explores the relationship between the everolimus minimum (Cmin) and maximum (Cmax) exposure and the risk for pulmonary adverse events (AEs) in Japanese versus non-Japanese patients."( Relationship between Pulmonary Adverse Events and Everolimus Exposure in Japanese and Non-Japanese Patients: A Meta-Analysis of Oncology Trials.
Bouillaud, E; Fan, J; Gallo, J; Ito, T; Jerusalem, G; Noguchi, S; Nonomura, N; Ohno, N; Ohtsu, A; Ravaud, A; Shinohara, N, 2017
)
0.95
" The commonly observed adverse events and laboratory abnormalities were stomatitis (49."( Efficacy and safety of sequential use of everolimus in Japanese patients with advanced renal cell carcinoma after failure of first-line treatment with vascular endothelial growth factor receptor tyrosine kinase inhibitor: a multicenter phase II clinical t
Akaza, H; Fujimoto, K; Hinotsu, S; Kimura, G; Kishida, T; Nozawa, M; Oyama, M; Ozono, S; Shimozuma, K; Sugiyama, T; Tokuda, N, 2017
)
0.72
" In the everolimus arm of the Asian subset, the most common adverse events of any grade were stomatitis (62."( Efficacy and safety of everolimus in combination with trastuzumab and paclitaxel in Asian patients with HER2+ advanced breast cancer in BOLERO-1.
Feng, J; Hurvitz, S; Jiang, Z; Lee, KS; Liu, D; Ng, TY; Noel-Baron, F; Ridolfi, A; Ringeisen, F; Shao, Z; Shen, K; Toi, M; Tseng, LM; Wang, X; Xu, B; Zhang, Q, 2017
)
1.2
"1%), and adverse events (AEs) (16."( Everolimus Plus Exemestane in Advanced Breast Cancer: Safety Results of the BALLET Study on Patients Previously Treated Without and with Chemotherapy in the Metastatic Setting.
Ascione, G; Barone, CA; Bighin, C; Bordonaro, R; Cazzaniga, ME; Cognetti, F; Foglietta, J; Frassoldati, A; Generali, D; Goffredo, F; Ionta, MT; Latini, L; Mafodda, A; Mariani, G; Michelotti, A; Minisini, AM; Molino, A; Montemurro, F; Nolè, F; Nuzzo, F; Piacentini, F; Piovano, P; Portera, G; Riccardi, F; Roila, F; Romito, S; Sartori, D; Schirone, A; Simoncini, EL; Testore, F; Vici, P, 2017
)
1.9
" Thus, it is important to assess even the potential of cumulative and additive toxic effects among the drugs."( Everolimus Plus Exemestane in Advanced Breast Cancer: Safety Results of the BALLET Study on Patients Previously Treated Without and with Chemotherapy in the Metastatic Setting.
Ascione, G; Barone, CA; Bighin, C; Bordonaro, R; Cazzaniga, ME; Cognetti, F; Foglietta, J; Frassoldati, A; Generali, D; Goffredo, F; Ionta, MT; Latini, L; Mafodda, A; Mariani, G; Michelotti, A; Minisini, AM; Molino, A; Montemurro, F; Nolè, F; Nuzzo, F; Piacentini, F; Piovano, P; Portera, G; Riccardi, F; Roila, F; Romito, S; Sartori, D; Schirone, A; Simoncini, EL; Testore, F; Vici, P, 2017
)
1.9
"Women with acute myocardial infarction (MI) undergoing mechanical reperfusion remain at increased risk of adverse cardiac events and mortality compared with their male counterparts."( Long-term Safety and Efficacy of New-Generation Drug-Eluting Stents in Women With Acute Myocardial Infarction: From the Women in Innovation and Drug-Eluting Stents (WIN-DES) Collaboration.
Baber, U; Camenzind, E; Chieffo, A; Dangas, GD; Faggioni, M; Farhan, S; Galatius, S; Giustino, G; Harari, R; Itchhaporia, D; Jeger, RV; Kandzari, D; Kastrati, A; Kim, HS; Kimura, T; Leon, MB; Mehran, R; Mehta, L; Morice, MC; Sartori, S; Serruys, PW; Sharma, M; Smits, PC; Sorrentino, S; Stefanini, GG; Steg, PG; Stone, GW; Valgimigli, M; Von Birgelen, C; Weisz, G; Wijns, W; Windecker, S, 2017
)
0.46
" Grade 3-4 adverse events (AEs) were reported in 37."( Efficacy and safety of Everolimus and Exemestane in hormone-receptor positive (HR+) human-epidermal-growth-factor negative (HER2-) advanced breast cancer patients: New insights beyond clinical trials. The EVA study.
Airoldi, M; Arcangeli, V; Artale, S; Atzori, F; Ballerio, A; Bianchi, GV; Blasi, L; Campidoglio, S; Cazzaniga, ME; Ciccarese, M; Clivio, L; Cursano, MC; Fabi, A; Ferrari, L; Ferzi, A; Ficorella, C; Frassoldati, A; Fumagalli, A; Garrone, O; Gebbia, V; Generali, D; La Verde, N; Maur, M; Michelotti, A; Moretti, G; Musolino, A; Palumbo, R; Piezzo, M; Pistelli, M; Porpiglia, M; Sartori, D; Scavelli, C; Schirone, A; Torri, V; Turletti, A; Valerio, MR; Vici, P; Zambelli, A, 2017
)
0.77
" The combination is safe and the best response could be obtained in patients receiving the full dose of EVE and/or after hormone-therapy as Fulvestrant in ABC."( Efficacy and safety of Everolimus and Exemestane in hormone-receptor positive (HR+) human-epidermal-growth-factor negative (HER2-) advanced breast cancer patients: New insights beyond clinical trials. The EVA study.
Airoldi, M; Arcangeli, V; Artale, S; Atzori, F; Ballerio, A; Bianchi, GV; Blasi, L; Campidoglio, S; Cazzaniga, ME; Ciccarese, M; Clivio, L; Cursano, MC; Fabi, A; Ferrari, L; Ferzi, A; Ficorella, C; Frassoldati, A; Fumagalli, A; Garrone, O; Gebbia, V; Generali, D; La Verde, N; Maur, M; Michelotti, A; Moretti, G; Musolino, A; Palumbo, R; Piezzo, M; Pistelli, M; Porpiglia, M; Sartori, D; Scavelli, C; Schirone, A; Torri, V; Turletti, A; Valerio, MR; Vici, P; Zambelli, A, 2017
)
0.77
"A meta-analysis of randomized controlled trials (RCTs) was performed to examine the risk of everolimus discontinuation due to related and unrelated adverse events (AE) in cancer patients."( Discontinuation of Everolimus Due to Related and Unrelated Adverse Events in Cancer Patients: A Meta-Analysis.
Garcia, CA; Rogers, SC; Wu, S, 2017
)
1
" The most common grade 1-2 adverse events with a suspected association with long-acting pasireotide monotherapy were diarrhoea (15 [37%] of 41), hyperglycaemia (17 [41%]), and weight loss (8 [20%]); those with a suspected association with everolimus monotherapy were stomatitis (26 [62%] of 42) and diarrhoea (16 [38%]); and those suspected to be associated with combination treatment were hyperglycaemia (27 [66%] of 41]), diarrhoea (19 [46%]), and asthenia (8 [20%])."( Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase 2 trial.
Baudin, E; Berruti, A; Brizzi, MP; Buikhuisen, W; Damiano, V; De Castro, J; Do Cao, C; Ferolla, P; Gislimberti, G; Grohé, C; Grønbæk, H; Léna, H; Lombard-Bohas, C; Mansoor, W; Mazieres, J; Meyer, T; Minotti, V; Öberg, K; Reed, N; Singh, N; Stankovic, M; Tiseo, M, 2017
)
0.89
" The objectives of this analysis were to evaluate the safety profile of this combination in a subset of Spanish patients in the BALLET trial and to characterize grade 3 and 4 adverse events (AEs) in routine clinical practice in Spain."( Safety of everolimus plus exemestane in patients with hormone-receptor-positive, HER2-negative locally advanced or metastatic breast cancer: results of phase IIIb BALLET trial in Spain.
Beliera, A; Carabantes, F; Ciruelos, E; Fernández, Y; Fonseca, R; García-Sáenz, JA; Gavilá, J; Martínez de Dueñas, E; Martínez-Jáñez, N; Murillo, L; Vidal, M, 2018
)
0.88
"PtCr-EES demonstrated comparable safety and effectiveness to CoCr-EES through 5 years of follow-up, with low rates of stent thrombosis and other adverse events."( Long-Term Safety and Efficacy of Platinum Chromium Everolimus-Eluting Stents in Coronary Artery Disease: 5-Year Results From the PLATINUM Trial.
Allocco, DJ; Bouchard, A; Carrié, D; Dawkins, KD; Dens, J; Dubois, CL; Farah, B; Feldman, RL; Hagiwara, N; Kelly, CR; Meredith, IT; Pompili, V; Rabinowitz, A; Saito, S; Stone, GW; Teirstein, PS, 2017
)
0.71
" In addition, EVR plus low-dose CNI regimen could reduce the rate of cytomegalovirus and infection, whereas a lower rate of other adverse events were noted in MMF plus standard-dose CNI regimen."( Efficacy and safety of everolimus plus low-dose calcineurin inhibitor vs. mycophenolate mofetil plus standard-dose calcineurin inhibitor in renal transplant recipients: A systematic review and meta-analysis
.
Deng, J; He, L; Jiang, W; Yang, B, 2018
)
0.79
"Lesion length is a major predictor of adverse outcomes after percutaneous coronary intervention."( Treatment with 48-mm everolimus-eluting stents : Procedural safety and 12-month patient outcome.
Arshad, MKM; Ho, HH; Jafary, FH; Loh, JKK; Ong, PJL; Tan, CK; Tin, ZL; Watson, T, 2019
)
0.83
"The Xience 48-mm EES device appears to be safe and efficacious with a low clinical event rate at the 12-month follow-up."( Treatment with 48-mm everolimus-eluting stents : Procedural safety and 12-month patient outcome.
Arshad, MKM; Ho, HH; Jafary, FH; Loh, JKK; Ong, PJL; Tan, CK; Tin, ZL; Watson, T, 2019
)
0.83
" The most common all-grade adverse events(AEs)were stomatitis(82%) and non-infectious lung disease(27%)."( [Efficacy and Safety of Everolimus plus Exemetane in Postmenopausal Endocrine-Responsive Metastatic Breast Cancer Patients].
Kubo, H; Maeda, N; Nagano, H; Nagashima, Y; Sakamoto, K; Sato, Y; Suzuki, N; Takeda, S; Yamamoto, S, 2017
)
0.76
" Adverse events were noted in four patients: infection, stomatitis, and increased triglycerides."( Efficacy and safety of everolimus in patients younger than 12 months with congenital subependymal giant cell astrocytoma.
Ehara, E; Kawawaki, H; Kuki, I; Kunihiro, N; Matsusaka, Y; Okazaki, S; Yoshida, Y, 2018
)
0.79
" This shift has markedly changed adverse event profiles, compared to cytotoxic chemotherapy, affecting a diverse range of organ systems."( Everolimus is a new anti-cancer molecule: Metabolic side effects as lipid disorders and hyperglycemia.
Baldelli, R; Di Giacinto, P; Lenzi, A; Morviducci, L; Nardone, MR; Ramponi, S; Rizza, L; Rota, F; Tubili, C; Zuppi, P, 2018
)
1.92
" The most frequent grade 3/4 adverse events were stomatitis (8."( Efficacy and safety of everolimus plus exemestane in postmenopausal women with hormone receptor-positive, human epidermal growth factor receptor 2-negative locally advanced or metastatic breast cancer: Results of the single-arm, phase IIIB 4EVER trial.
Decker, T; Distelrath, A; Fasching, PA; Hadji, P; Janni, W; Kreuzeder, J; Kurbacher, CM; Lüftner, D; Lux, MP; Marmé, F; Mundhenke, C; Muth, M; Quiering, C; Schneeweiss, A; Stoetzer, O; Taran, FA; Tesch, H, 2019
)
0.82
" Data collection included demographic and clinical information including reason(s) for initiating treatment with mTOR inhibitors, treatment duration, dosing, and corresponding serum trough levels, response to treatment, and adverse events (AE)."( Short-term safety of mTOR inhibitors in infants and very young children with tuberous sclerosis complex (TSC): Multicentre clinical experience.
Bruns, S; Capal, JK; Curatolo, P; de Vries, P; Devinsky, O; Ess, K; Franz, DN; Jansen, AC; Jozwiak, S; Koenig, MK; Krueger, DA; Lawson, J; Mowat, D; Narayanan, V; Ramos, F; Tzadok, M; Wong, M, 2018
)
0.48
" Further studies are necessary to determine if the F-FDG PET/CT could early predict adverse effects of mTOR inhibitors."( Everolimus-induced pulmonary toxicity: Findings on 18F-FDG PET/CT imaging.
Bruna-Muraille, C; Dejust, S; Eymard, JC; Morland, D; Papathanassiou, D; Savoye, AM; Yazbek, G, 2018
)
1.92
" Although lenvatinib is associated with favorable efficacy, it is associated with adverse events (AEs) that the clinician will have to closely monitor for and proactively manage."( Managing the adverse events associated with lenvatinib therapy in radioiodine-refractory differentiated thyroid cancer.
Cabanillas, ME; Takahashi, S, 2019
)
0.51
"Within the safety population (everolimus 1014, MPA 1012), adverse events with a suspected relation to study drug occurred in 62."( Safety of Everolimus With Reduced Calcineurin Inhibitor Exposure in De Novo Kidney Transplants: An Analysis From the Randomized TRANSFORM Study.
Adel Bakr, M; Basic-Jukic, N; Bernhardt, P; Buchler, M; Cassuto, E; Chadban, S; Citterio, F; Cruzado, JM; Garcia, VD; Hernandez Gutierrez, MP; Kim, DY; Kuypers, D; Pascual, J; Russ, G; Sommerer, C; Soo Kim, M; Tedesco-Silva, H; Uyen, HD; Viklicky, O, 2019
)
1.2
"De novo everolimus with reduced-exposure CNI yielded a comparable incidence, though a distinctly different pattern, of adverse events versus current standard of care."( Safety of Everolimus With Reduced Calcineurin Inhibitor Exposure in De Novo Kidney Transplants: An Analysis From the Randomized TRANSFORM Study.
Adel Bakr, M; Basic-Jukic, N; Bernhardt, P; Buchler, M; Cassuto, E; Chadban, S; Citterio, F; Cruzado, JM; Garcia, VD; Hernandez Gutierrez, MP; Kim, DY; Kuypers, D; Pascual, J; Russ, G; Sommerer, C; Soo Kim, M; Tedesco-Silva, H; Uyen, HD; Viklicky, O, 2019
)
1.35
" Supraflex seems a safe and effective alternative drug-eluting stent to other stents in clinical practice."( Safety and efficacy of a sirolimus-eluting coronary stent with ultra-thin strut for treatment of atherosclerotic lesions (TALENT): a prospective multicentre randomised controlled trial.
Cequier, A; Chang, CC; Choudhury, A; Colombo, A; de Winter, RJ; Gil, RJ; Hofma, S; Hoye, A; Ijsselmuiden, S; Iniguez, A; Kaul, U; Kogame, N; Kukreja, N; Mathur, A; Merkely, B; Modolo, R; Moreno, R; Onuma, Y; Petrov, I; Prokopczuk, J; Serra, A; Serruys, PW; Smits, PC; Soliman, O; Spitzer, E; Tonev, G; Tonino, P; Ungi, I; Walsh, S; Zaman, A; Zurakowski, A, 2019
)
0.51
"This retrospective multicenter study demonstrates that mTOR inhibitor treatment with everolimus is safe in TSC patients under the age of 2 years and shows beneficial effects on cardiac manifestations, SEGA size and early epilepsy."( Safety and efficacy of mTOR inhibitor treatment in patients with tuberous sclerosis complex under 2 years of age - a multicenter retrospective study.
Bernhard, MK; Brösse, I; Gorenflo, M; Hahn, A; Hethey, S; Hoffmann, GF; Kaiser, O; Kölker, S; Kreuzaler, P; Merkenschlager, A; Milde, T; Möckel, A; Roser, T; Saffari, A; Schlump, JU; Serfling, A; Spiegler, J; Syrbe, S; van Tilburg, CM; Wagner, R; Wiemer-Kruel, A; Wilken, B; Witt, O; Ziegler, A, 2019
)
0.74
"Patients with diabetes mellitus are prone to increased adverse outcomes after percutaneous coronary intervention, even with contemporary drug-eluting stents."( Efficacy and Safety of Ultrathin, Bioresorbable-Polymer Sirolimus-Eluting Stents Versus Thin, Durable-Polymer Everolimus-Eluting Stents for Coronary Revascularization of Patients With Diabetes Mellitus.
Kandzari, D; Kolm, P; Koolen, JJ; Lipinski, MJ; Saito, S; Shlofmitz, E; Torguson, R; Waksman, R; Windecker, S, 2019
)
0.73
" Through planned 4-year follow-up, the primary composite endpoint of major adverse cardiac events (MACE; death, myocardial infarction [MI] and target lesion revascularization) and rates of individual component endpoints and stent thrombosis were determined."( Late-term safety and effectiveness of everolimus-eluting stents in chronic total coronary occlusion revascularization: Final 4-year results from the evaluation of the XIENCE coronary stent, Performance, and Technique in Chronic Total Occlusions (EXPERT CT
Grantham, JA; Kandzari, DE; Karmpaliotis, D; Kini, AS; Lembo, NJ; Lombardi, W; McGreevy, R; Moses, JW; Nicholson, WJ; Orr, C; Popma, JJ; Tummala, PE; Wang, J; Zhao, W, 2019
)
0.78
"A comprehensive survey of real-world adverse event reporting with conditions for approval under the Pharmaceuticals and Medical Devices Act identified 490 cases of ST with EES."( Patient and lesion characteristics in late/very late stent thrombosis with everolimus-eluting stents from real-world adverse event reporting.
Handa, N; Ho, M; Ishii, K; Koike, K; Konishi, A; Mitsutake, Y; Mochizuki, S, 2020
)
0.79
" Data collection from adverse event reporting could be a helpful strategy for evaluation of this serious but rare complication."( Patient and lesion characteristics in late/very late stent thrombosis with everolimus-eluting stents from real-world adverse event reporting.
Handa, N; Ho, M; Ishii, K; Koike, K; Konishi, A; Mitsutake, Y; Mochizuki, S, 2020
)
0.79
" The outcomes of interest were biopsy-proven acute rejection (BPAR), graft loss, death, renal function and adverse events."( Efficacy and safety of everolimus treatment on liver transplant recipients: A meta-analysis.
Chen, KB; Guan, TW; Lin, YJ; Ou, MY, 2019
)
0.82
" The risk of any adverse event was increased by everolimus combined with reduced CNI therapy (RR = 1."( Efficacy and safety of everolimus treatment on liver transplant recipients: A meta-analysis.
Chen, KB; Guan, TW; Lin, YJ; Ou, MY, 2019
)
1.08
" This regimen might be associated with an increased risk of adverse events, which needs to be elucidated further."( Efficacy and safety of everolimus treatment on liver transplant recipients: A meta-analysis.
Chen, KB; Guan, TW; Lin, YJ; Ou, MY, 2019
)
0.82
" Fifteen patients in the sunitinib group and 13 in the pazopanib group experienced adverse events."( A Phase 4 Study of Everolimus to Evaluate Efficacy and Safety in Patients with Metastatic Renal-Cell Carcinoma after Failure of First-Line Sunitinib or Pazopanib (SUNPAZ).
de Geeter, P; Decker, T; Quiering, C; Resch, A; Schmitz, S; Schostak, M, 2020
)
0.89
" Therefore, we aimed to evaluate long-term adverse events in HBR patients undergoing percutaneous coronary intervention with cobalt-chromium everolimus-eluting stent implantation."( Long-Term Safety and Efficacy of Durable Polymer Cobalt-Chromium Everolimus-Eluting Stents in Patients at High Bleeding Risk: A Patient-Level Stratified Analysis From Four Postapproval Studies.
Angiolillo, DJ; Baber, U; Bangalore, S; Bhatt, DL; Chandiramani, R; Claessen, BE; Ge, J; Guedeney, P; Hermiller, J; Kozuma, K; Krucoff, M; Liu, Y; Makkar, R; Mehran, R; Neumann, FJ; Rau, V; Saito, S; Seth, A; Sorrentino, S; Valgimigli, M; Vogel, B; Wang, J, 2020
)
1
"Everolimus is administered to patients with metastatic renal cell carcinoma in full daily dose of 10 mg or in reduced daily dose of 5 mg in case adverse effect occurred."( Management of metabolic adverse effects of everolimus in patients with renal carcinoma.
Gregorová, J; Holečková, P; Žáková, K, 2019
)
2.22
"The combination of everolimus and buparlisib is safe and well-tolerated at the RP2D of 5 and 60 mg on a continuous daily schedule."( A Phase I Study of Safety, Pharmacokinetics, and Pharmacodynamics of Concurrent Everolimus and Buparlisib Treatment in Advanced Solid Tumors.
Akce, M; Alese, OB; Bilen, MA; Carthon, B; Chen, Z; Collins, H; El-Rayes, BF; Harris, WB; Harvey, RD; Khuri, FR; Kudchagkar, RR; Lawson, DH; Lewis, C; Lonial, S; Owonikoko, TK; Ramalingam, SS; Shaib, WL; Sica, GL; Steuer, CE; Wu, C; Zhang, C, 2020
)
1.11
"2%) adverse events (AEs)."( Efficacy and safety of everolimus plus exemestane in patients with HR+, HER2- advanced breast cancer progressing on/after prior endocrine therapy in routine clinical practice: Primary results from the non-interventional study, STEPAUT.
Bartsch, R; Egle, D; Gnant, M; Greil, R; Haslbauer, F; Helfgott, R; Hennebelle, M; Hock, K; Hubalek, M; Lang, A; Marth, C; Mraz, B; Öhler, L; Petru, E; Pfeiler, G; Redl, A; Steger, GG; Tinchon, C, 2020
)
0.87
" Aims of this study were to evaluate the progression of renal dysfunction after discontinuation of CNIs and to monitor for major adverse events after therapy change."( Long-Term Effects of the Replacement of Calcineurin Inhibitors With Everolimus and Mycophenolate in Patients With Calcineurin Inhibitor-Related Nephrotoxicity.
Acquaro, M; Cattadori, B; D'Armini, AM; Ghio, S; Greco, A; Guida, S; Klersy, C; Pelenghi, S; Pellegrini, C; Raineri, C; Scelsi, L; Turco, A; Visconti, LO, 2020
)
0.79
" Adverse events and changes in lipid panel profile, pulmonary function tests, and VEGF-D were assessed."( A phase II clinical trial of the Safety Of Simvastatin (SOS) in patients with pulmonary lymphangioleiomyomatosis and with tuberous sclerosis complex.
Courtwright, AM; Dorgan, D; Fleck, V; Kotloff, R; Kreider, M; Krymskaya, VP; McCormack, FX, 2020
)
0.56
" The most common adverse events were peripheral edema (30%), cough (30%), and diarrhea (30%)."( A phase II clinical trial of the Safety Of Simvastatin (SOS) in patients with pulmonary lymphangioleiomyomatosis and with tuberous sclerosis complex.
Courtwright, AM; Dorgan, D; Fleck, V; Kotloff, R; Kreider, M; Krymskaya, VP; McCormack, FX, 2020
)
0.56
"The combination of simvastatin with mTORi in LAM patients is safe and well-tolerated from an adverse events perspective."( A phase II clinical trial of the Safety Of Simvastatin (SOS) in patients with pulmonary lymphangioleiomyomatosis and with tuberous sclerosis complex.
Courtwright, AM; Dorgan, D; Fleck, V; Kotloff, R; Kreider, M; Krymskaya, VP; McCormack, FX, 2020
)
0.56
" The objectives of this analysis were to provide additional information on the safety profile of this schedule depending on prior anticancer therapies and to characterize the time course of adverse events (AEs) and serious AEs (SAEs) of clinical interest throughout the study period."( Everolimus plus exemestane in hormone-receptor-positive, HER2-negative locally advanced or metastatic breast cancer: incidence and time course of adverse events in the phase IIIb BALLET population.
Ciruelos, E; Conte, P; Gavila, J; Generali, D; Jerusalem, G; Lang, I; Martin, M; Martínez-Serrano, MJ; Montemurro, F; Neven, P; Perelló, MF; Tjan-Heijnen, VCG, 2020
)
2
" Lipid metabolism disorder was the most common adverse event (69."( Safety and efficacy of sirolimus combined with endocrine therapy in patients with advanced hormone receptor-positive breast cancer and the exploration of biomarkers.
Guan, X; Li, C; Li, H; Li, L; Liu, B; Ma, F; Qian, H; Rong, G; Sun, X; Wang, W; Xu, B; Yi, Z; Zhai, J, 2020
)
0.56
" In 21 patients, EVR dose, trough level, outcomes, and adverse effects were assessed."( Safety and Efficacy of Everolimus Rescue Treatment After Pediatric Living Donor Liver Transplantation.
Bessho, K; Deguchi, K; Kodama, T; Noguchi, Y; Nomura, M; Okuyama, H; Saka, R; Tazuke, Y; Ueno, T; Watanabe, M,
)
0.44
"In this single-center, prospective, randomized study, adverse events (AEs), serious AEs (SAEs), viral infections, laboratory abnormalities, dose reductions, and temporary or permanent discontinuation of the immunosuppressant were compared among patients receiving r-ATG/EVR (n = 85), BAS/EVR (n = 102), and BAS/MPS (n = 101)."( Immunosuppressive Drug-Associated Adverse Event Profiles in De Novo Kidney Transplant Recipients Receiving Everolimus and Reduced Tacrolimus Doses.
Felipe, CR; Medina Pestana, JO; Miranda, TA; Santos, RHN; Tedesco-Silva Junior, H, 2020
)
0.77
" Common (>5%) grade ≥3 adverse events included rash (19%), neutropenia (19%), hypokalaemia (11%) and fatigue (9%)."( Phase 1 safety and pharmacodynamic study of lenalidomide combined with everolimus in patients with advanced solid malignancies with efficacy signal in adenoid cystic carcinoma.
Alese, OB; Bilen, MA; Carthon, BC; Chen, Z; El-Rayes, BF; Harris, WB; Harvey, RD; Hossain, MS; Khuri, FR; Lawson, DH; Lewis, C; Lonial, S; Owonikoko, TK; Ramalingam, SS; Shaib, W; Steuer, CE; Waller, EK; Wu, C; Zhang, C, 2020
)
0.79
"Results of this IPD analysis suggest that the BP-SES with ultrathin struts is as safe as and more efficacious than DP-EES in the overall cohort and especially in small vessels."( Individual patient data analysis of the BIOFLOW study program comparing safety and efficacy of a bioresorbable polymer sirolimus eluting stent to a durable polymer everolimus eluting stent.
Kandzari, DE; Koolen, J; Lefèvre, T; Richardt, G; Saito, S; Slagboom, T; Toelg, R; Waltenberger, J, 2021
)
0.82
" The treatment effects and adverse events were compared between the two groups."( Efficacy and safety of low-dose everolimus treatment for renal angiomyolipoma associated with tuberous sclerosis complex.
Endo, K; Hatano, T; Tamari, M, 2021
)
0.9
"The present study demonstrates that low-dose everolimus treatment is safe and effective for TSC-associated AML."( Efficacy and safety of low-dose everolimus treatment for renal angiomyolipoma associated with tuberous sclerosis complex.
Endo, K; Hatano, T; Tamari, M, 2021
)
1.16
" Primary endpoint was occurrence of any major adverse cardiac event (MACE) up to 12 months' follow-up."( Safety and performance of everolimus-eluting stents comprising of biodegradable polymers with ultrathin stent platforms.
Bolinera, SV; Byrapaneni, S; Chilukuri, M; Gangasani, S; Pamidimukkala, V; Polavarapu, A; Polavarapu, N; Polavarapu, RS; Reddy, SS; Tharaknath, VR, 2020
)
0.86
"Conversion to an EVL-based therapy after pediatric HTX is a safe immunosuppressive regime without increasing risk of acute rejection or CMV-infection."( Conversion to everolimus in pediatric heart transplant recipients is a safe treatment option with an impact on cardiac allograft vasculopathy and renal function.
Dalla Pozza, R; Fernandez Rodriguez, S; Fischer, M; Grimm, K; Haas, NA; Jakob, A; Kozlik-Feldmann, R; Lehner, A; Orban, M; Rosenthal, LL; Ulrich, SM, 2021
)
0.98
"There are limited real-world data available regarding adverse events (AEs) of immunosuppressants."( Mtor inhibitors associated with higher cardiovascular adverse events-A large population database analysis.
Abagyan, R; Nguyen, VN; Tsunoda, SM, 2021
)
0.62
" Absence of complications such as bleeding or glomerular filtration rate decline in the long term, with a favorable safety profile, without interruptions and with mild-moderate, non-cumulative adverse effects, mostly within the first year of treatment."( Renal angiomyolipoma and tuberous sclerosis complex: long-term safety and efficacy outcomes of Everolimus therapy.
Álvarez-Ossorio Fernández, JL; Ledo Cepero, MJ; Ojeda Claro, AV; Ruiz Guerrero, E; Soto Delgado, M, 2021
)
0.84
"Everolimus is a safe and effective therapeutic option for renal angiomyolipoma and various manifestations of TSC, which has been reproduced in real life with six years of follow-up."( Renal angiomyolipoma and tuberous sclerosis complex: long-term safety and efficacy outcomes of Everolimus therapy.
Álvarez-Ossorio Fernández, JL; Ledo Cepero, MJ; Ojeda Claro, AV; Ruiz Guerrero, E; Soto Delgado, M, 2021
)
2.28
" VAN + EV combination is safe and active in refractory solid tumors."( Safety and activity of vandetanib in combination with everolimus in patients with advanced solid tumors: a phase I study.
Amini, B; Bhatt, T; Cascone, T; Conley, AP; Drobnitzky, N; Falchook, GS; Fu, S; Hess, KR; Heymach, JV; Hong, DS; Janku, F; Karp, D; Meric-Bernstam, F; Naing, A; Piha-Paul, SA; Ryan, AJ; Sacks, RL; Sherman, SI; Subbiah, IM; Subbiah, V, 2021
)
0.87
" A landmark progression-free survival (PFS) analysis by central review was performed for patients treated for at least 16 weeks (n = 308) and according to the occurrence of any-grade adverse events (AEs) within this treatment period."( Relationship between metabolic toxicity and efficacy of everolimus in patients with neuroendocrine tumors: A pooled analysis from the randomized, phase 3 RADIANT-3 and RADIANT-4 trials.
Buzzoni, R; Carnaghi, C; Fazio, N; Herbst, F; Kulke, MH; Pavel, ME; Ridolfi, A; Strosberg, J; Valle, JW; Yao, JC, 2021
)
0.87
" Additionally, neutropenia, diarrhoea and fatigue were the most common adverse events caused by these drugs, followed by pain, nausea, decreased appetite, anaemia and vomiting."( Efficacy and safety of targeted drugs in advanced or metastatic gastric and gastroesophageal junction cancer: A network meta-analysis.
Ge, L; Gong, H; Hou, L; Jin, X; Leng, G; Li, C; Li, T; Liu, Y; Ma, L; Niu, S; Su, Y; Zhang, H; Zhang, L; Zhao, L, 2022
)
0.72
"In the TWILIGHT trial, ticagrelor monotherapy after a short course of dual antiplatelet therapy (DAPT) was shown to be a safe bleeding avoidance strategy in high-risk patients undergoing percutaneous coronary intervention (PCI) with drug-eluting stents (DES)."( Safety and efficacy of ticagrelor monotherapy according to drug-eluting stent type: the TWILIGHT-STENT study.
Angiolillo, DJ; Baber, U; Briguori, C; Camaj, A; Cao, D; Cohen, D; Collier, T; Dangas, G; Dudek, D; Escaned, J; Gibson, CM; Gil, R; Giustino, G; Goel, R; Han, YL; Huber, K; Kastrati, A; Kaul, U; Kornowski, R; Krucoff, MW; Kunadian, V; Mehran, R; Mehta, S; Moliterno, DJ; Nicolas, J; Ohman, EM; Oldroyd, KG; Pocock, S; Sardella, G; Sartori, S; Sharma, S; Shlofmitz, R; Steg, PG; Weisz, G; Witzenbichler, B; Zhang, Z, 2022
)
0.72
" Patients in the 14-mg arm were to be uptitrated to lenvatinib 18 mg at cycle 2, day 1, barring intolerable grade 2 or any grade ≥3 treatment-emergent adverse events (TEAEs) requiring dose reduction occurring in the first 28-d cycle."( Assessing the Safety and Efficacy of Two Starting Doses of Lenvatinib Plus Everolimus in Patients with Renal Cell Carcinoma: A Randomized Phase 2 Trial.
Alekseev, B; Binder, TA; Castellano, D; Ciuleanu, T; Glen, H; Heng, DYC; Koralewski, P; Lee, JL; O'Hara, K; Pal, SK; Parnis, F; Peng, L; Puente, J; Rha, SY; Smith, AD; Stroyakovskiy, D; Sunela, K, 2022
)
0.95
"As for adverse events (AEs) caused by everolimus, findings from clinical trials and post-marketing surveillance have reported interstitial lung disease, hyperglycaemia, cardiovascular disease, etc."( Comprehensive analysis of everolimus-induced adverse events using the Japanese real-world database.
Fujiwara, M; Nakano, K; Shimizu, T; Uchida, M; Uesawa, Y, 2022
)
1.29
"Data reported between April 2004 and June 2021 in the Japanese Adverse Drug Event Report database were extracted for use."( Comprehensive analysis of everolimus-induced adverse events using the Japanese real-world database.
Fujiwara, M; Nakano, K; Shimizu, T; Uchida, M; Uesawa, Y, 2022
)
1.02
"Among the top 30 adverse events, 23 adverse event signals were detected and classified into seven categories: lung-related AEs, haematological-related AEs, cancer progression, blood glucose-related AEs, hepatic-related AEs, renal-related AEs and others."( Comprehensive analysis of everolimus-induced adverse events using the Japanese real-world database.
Fujiwara, M; Nakano, K; Shimizu, T; Uchida, M; Uesawa, Y, 2022
)
1.02
"A comprehensive survey of adverse events associated with everolimus administration using the pharmacovigilance database revealed that pulmonary and haematological AEs are frequently reported."( Comprehensive analysis of everolimus-induced adverse events using the Japanese real-world database.
Fujiwara, M; Nakano, K; Shimizu, T; Uchida, M; Uesawa, Y, 2022
)
1.27
" The Absorb everolimus-eluting BVS was safe and showed the favorable clinical outcomes in both patency and TLR, especially in infrapopliteal disease with heavy calcification."( Efficacy and Safety of Absorb Everolimus-Eluting Bioresorbable Vascular Scaffold in Peripheral Artery Disease: A Single-Arm Meta-Analysis.
Fan, W; Feng, B; Li, L; Pei, J; Shi, W; Tan, J; Yu, B; Yuan, G, 2023
)
1.58
"The 48mm-EES is safe and effective to treat long coronary lesions, including CTOs, and provides attractive cost-effectiveness by limiting multiple stenting."( Safety and efficacy of 48 mm Xience Xpedition everolimus-eluting stent for the treatment of long coronary lesions.
Arroyo, D; Benamer, H; Champagne, S; Chevalier, B; Garot, P; Gautier, A; Hovasse, T; Lefèvre, T; Neylon, A; Sanguineti, F; Unterseeh, T, 2022
)
0.98
" The mainstay of treatment is with multiple anti-seizure medications (ASMs); however, the ASMs themselves can be associated with psychobehavioural adverse events, and effects (negative or positive) on cognition and sleep."( Psychobehavioural and Cognitive Adverse Events of Anti-Seizure Medications for the Treatment of Developmental and Epileptic Encephalopathies.
Schubert-Bast, S; Strzelczyk, A, 2022
)
0.72
" For grade ≥ 3 adverse events (AEs), compared with placebo, lenvatinib plus everolimus showed worse safety than all other treatments except for lenvatinib (placebo vs."( Optimizing targeted drug selection in combination therapy for patients with advanced or metastatic renal cell carcinoma: A systematic review and network meta-analysis of safety.
Bi, X; Cao, C; Jiang, W; Shang, B; Shi, H; Shou, J; Wu, J; Xie, R; Zhou, A, 2023
)
1.14
"4%) grade 3 or higher treatment-related adverse events occurred in the combination group, vorolanib group and everolimus group, respectively."( Efficacy and safety of vorolanib plus everolimus in metastatic renal cell carcinoma: A three-arm, randomised, double-blind, multicentre phase III study (CONCEPT).
Chen, K; Chen, P; Ding, L; Fu, C; Guo, H; Guo, J; He, C; He, Z; Hu, Z; Ji, Z; Jiang, K; Li, G; Li, X; Liu, B; Liu, Y; Liu, Z; Luo, H; Qin, S; Sheng, X; Shi, B; Tu, X; Wang, G; Wang, L; Wang, Y; Wang, Z; Wei, Q; Wu, B; Wu, D; Yao, X; Ye, D; Yuan, X; Zhang, D; Zhang, Y; Zhao, Y; Zhou, A; Zhou, F; Zou, Q, 2023
)
1.39
"Literature is limited comparing adverse effects (AEs) of the proliferation signal inhibitors (PSIs) sirolimus (SRL) and everolimus (EVL) in pediatric heart transplant (HTx) recipients."( Assessment of the adverse effects of sirolimus versus everolimus in pediatric heart transplant recipients.
Ahmed, H; Albers, EL; Friedland-Little, JM; Hong, BJ; Kemna, MS; Law, YM; Newland, DM; Rosete, BE; Spencer, KL, 2023
)
1.37
" In patients with PD-L1+ve and -ve ccRCC, niv, atezolizumab, ipi, and pembrolizumab were safe and effective alone and when combined with cabozantinib, tivozanib, axitinib, levantinib, and pegilodecakin."( Efficacy and Safety of Checkpoint Inhibitors in Clear Cell Renal Cell Carcinoma: A Systematic Review of Clinical Trials.
Aiman, W; Ali, MA; Butt, SK; Farrukh, M; Fatima, A; Garg, I; Habib, R; Kashif, T; Khan, H; Naveed, M; Niaz, R; Saeed, M; Zubair, H, 2023
)
0.91
" To determine the safety and efficacy of MTKI therapy in children, adolescents and young adults (AYAs), we conducted a retrospective study on adverse events and treatment outcomes."( Safety and efficacy of multiple tyrosine kinase inhibitors in pediatric/adolescent and young adult patients with relapsed or refractory osteosarcomas: A single-institution retrospective analysis.
Arakawa, A; Iwata, S; Kawai, A; Kobayashi, E; Kumamoto, T; Maniwa, J; Nakajima, M; Ogawa, C; Shirakawa, N; Sugiyama, M; Tanimura, K; Tao, K; Watanabe, Y; Yoneda, A, 2023
)
0.91
" The incidence of treatment-related grade 3 nonhematological adverse events was 14."( Safety and efficacy of multiple tyrosine kinase inhibitors in pediatric/adolescent and young adult patients with relapsed or refractory osteosarcomas: A single-institution retrospective analysis.
Arakawa, A; Iwata, S; Kawai, A; Kobayashi, E; Kumamoto, T; Maniwa, J; Nakajima, M; Ogawa, C; Shirakawa, N; Sugiyama, M; Tanimura, K; Tao, K; Watanabe, Y; Yoneda, A, 2023
)
0.91
" MTKI therapies, particularly regorafenib, against pediatric relapsed osteosarcoma can suppress tumor growth and prolong PFS with tolerable adverse events."( Safety and efficacy of multiple tyrosine kinase inhibitors in pediatric/adolescent and young adult patients with relapsed or refractory osteosarcomas: A single-institution retrospective analysis.
Arakawa, A; Iwata, S; Kawai, A; Kobayashi, E; Kumamoto, T; Maniwa, J; Nakajima, M; Ogawa, C; Shirakawa, N; Sugiyama, M; Tanimura, K; Tao, K; Watanabe, Y; Yoneda, A, 2023
)
0.91
" While this finding is limited by the sample size and warrants prospective verification, initiating therapy at a reduced dose might be practicable and safe in a distinct subset of patients."( Does the dose matter? Antiproliferative efficacy and toxicity of everolimus in patients with neuroendocrine tumors - Experiences from a tertiary referral center.
Haug, A; Kiesewetter, B; Kretschmer-Chott, E; Macheiner, S; Mazal, P; Melhorn, P; Raderer, M; Wolff, L, 2023
)
1.15
"The randomised double-blinded placebo-controlled EXIST-1-3 studies have showed everolimus effective with adverse effects reported as acceptable in treatment of symptoms in patients with tuberous sclerosis complex (TSC), although evidence of outcomes in clinical practice remains limited."( Effectiveness and safety of everolimus treatment in patients with tuberous sclerosis complex in real-world clinical practice.
Christensen, J; Cockerell, I; Grenaa Frederiksen, M; Heimdal, K; Hoei-Hansen, CE; Holst, L; Issa-Epe, AI; Johannessen Landmark, C; Lund, C; Nedregaard, B; Nærland, T; Solhoff, R, 2023
)
1.43
" Adverse effects were reported in 61 of 64 patients (95%) after a median treatment duration of 31 months (range 0-106), with oral ulceration/stomatitis (63%) and upper respiratory tract infections (38%) being the most common."( Effectiveness and safety of everolimus treatment in patients with tuberous sclerosis complex in real-world clinical practice.
Christensen, J; Cockerell, I; Grenaa Frederiksen, M; Heimdal, K; Hoei-Hansen, CE; Holst, L; Issa-Epe, AI; Johannessen Landmark, C; Lund, C; Nedregaard, B; Nærland, T; Solhoff, R, 2023
)
1.2
" Close follow-up is needed for this group, especially for children and patients who may not be able to report adverse effects."( Effectiveness and safety of everolimus treatment in patients with tuberous sclerosis complex in real-world clinical practice.
Christensen, J; Cockerell, I; Grenaa Frederiksen, M; Heimdal, K; Hoei-Hansen, CE; Holst, L; Issa-Epe, AI; Johannessen Landmark, C; Lund, C; Nedregaard, B; Nærland, T; Solhoff, R, 2023
)
1.2

Pharmacokinetics

Everolimus half-life was reduced from an average of 32 hours to 24 hours. Study aimed to evaluate the pharmacokinetic profile and tissue effects of everolimus delivered into arterial wall using biodegradable nanospheres.

ExcerptReferenceRelevance
" On both study days Cmax, and tmax of cyclosporin were not significantly different."( Pharmacokinetics of SDZ RAD and cyclosporin including their metabolites in seven kidney graft patients after the first dose of SDZ RAD.
Blick, S; Franzke, A; Jacobsen, W; Kirchner, GI; Manns, MP; Mueller, L; Sewing, KF; Vidal, C; Wagner, S; Winkler, M, 2000
)
0.31
" The interindividual pharmacokinetic variability for AUC was 85."( Longitudinal assessment of everolimus in de novo renal transplant recipients over the first post-transplant year: pharmacokinetics, exposure-response relationships, and influence on cyclosporine.
Boger, R; Cambon, N; Gerbeau, C; Kahan, BD; Kaplan, B; Kovarik, JM; Lorber, M; Rordorf, C; Rouilly, M; Winkler, M, 2001
)
0.61
" Recipients with bile diversion demonstrated lower peak concentration (Cmax) than those without, but overall drug exposure (AUC) was not altered."( Pharmacokinetics and tolerability of 40-0-[2-hydroxyethyl]rapamycin in de novo liver transplant recipients.
Campestrini, J; Grant, D; Kovarik, JM; Levy, GA; Paradis, K; Smith, T, 2001
)
0.31
" h/ml) and 84% prolonged half-life (79 +/- 42 versus 43 +/- 18 hours) in subjects with hepatic impairment."( Influence of hepatic impairment on everolimus pharmacokinetics: implications for dose adjustment.
Dilzer, SC; Figueiredo, J; Kovarik, JM; Lasseter, K; Reynolds, C; Rordorf, C; Sabia, HD; Zimmermann, H, 2001
)
0.59
"The authors assessed the mutual influence of the immunosuppressant everolimus (Certican) and the HMG-CoA reductase inhibitors atorvastatin and pravastatin when coadministered based on pharmacokinetic and pharmacodynamic measures."( Pharmacokinetic and pharmacodynamic assessments of HMG-CoA reductase inhibitors when coadministered with everolimus.
Berthier, S; Hartmann, S; Hubert, M; Kovarik, JM; Rordorf, C; Rosenkranz, B; Schneider, W, 2002
)
0.76
" Compared with adults from a previous study, apparent clearance (L/h) and distribution volume (L) were lower in pediatric patients, whereas the elimination half-life was similar."( Single-dose pharmacokinetics and tolerability of everolimus in stable pediatric renal transplant patients.
Boger, R; Cambon, N; Ettenger, R; Hoyer, PF; Kovarik, JM; Lemire, J; Mahan, J; McMahon, L; Van Damme-Lombaerts, R; Webb, NA, 2002
)
0.57
" Everolimus half-life was reduced significantly, from an average of 32 hours to 24 hours."( Effect of rifampin on apparent clearance of everolimus.
Figueiredo, J; Hartmann, S; Kovarik, JM; Port, A; Rordorf, C; Rouilly, M, 2002
)
1.49
" Cmax and AUC were evaluated by standard bioequivalence testing to determine relative bioavailability and to quantify the effect of food."( Clinical development of an everolimus pediatric formulation: relative bioavailability, food effect, and steady-state pharmacokinetics.
Berthier, S; Ettenger, R; Hoyer, PF; Kovarik, JM; Langholff, WK; Marion, AS; McMahon, L; Noe, A; Rordorf, C, 2003
)
0.62
" Pharmacokinetic data consisted of 4014 everolimus trough concentrations (Cmin) obtained in all patients and 659 area under the concentration-time curve (AUC) -profiles obtained at months 2, 3, and 6 in a subset of 261 patients."( Pharmacokinetics of an everolimus-cyclosporine immunosuppressive regimen over the first 6 months after kidney transplantation.
Berthier, S; Dantal, J; Hsu, CH; Kahan, BD; Kaplan, B; Kovarik, JM; McMahon, L; Rordorf, C; Silva, HT, 2003
)
0.9
" We used median-effect analysis to characterize exposure-response associations between everolimus average Cmin vs freedom from biopsy-confirmed acute rejection; maximum cholesterol, low density lipoprotein, triglyceride, and creatinine levels; and minimum leukocyte and platelet counts."( Everolimus in de novo cardiac transplantation: pharmacokinetics, therapeutic range, and influence on cyclosporine exposure.
Dorent, R; Eisen, H; Hsu, CH; Kovarik, JM; Mancini, D; Rordorf, C; Rouilly, M; Vigano, M, 2003
)
1.98
" Everolimus Cmin averaged 9-11 ng/mL in the immediate cyclosporine group over the first 3 months."( Influence of delayed initiation of cyclosporine on everolimus pharmacokinetics in de novo renal transplant patients.
Bettoni-Ristic, O; Civati, G; Dantal, J; Kovarik, JM; Rizzo, G; Rordorf, C; Rouilly, M, 2003
)
1.48
" Everolimus, which has greater polarity than sirolimus, was developed in an attempt to improve the pharmacokinetic characteristics of sirolimus, particularly to increase its oral bioavailability."( Clinical pharmacokinetics of everolimus.
Kirchner, GI; Manns, MP; Meier-Wiedenbach, I, 2004
)
1.52
" The secondary objective was to assess the pharmacokinetic profile of two different formulations (capsule and tablet) of everolimus."( Tolerability and steady-state pharmacokinetics of everolimus in maintenance renal transplant patients.
Budde, K; Dantal, J; Fauchald, P; Fritsche, L; Hauser, IA; Lehne, G; Lison, A; Neumayer, HH; Soulillou, JP; Winkler, M, 2004
)
0.78
" Pharmacodynamic assessment showed a relationship between drug exposure and thrombocytopenia."( Tolerability and steady-state pharmacokinetics of everolimus in maintenance renal transplant patients.
Budde, K; Dantal, J; Fauchald, P; Fritsche, L; Hauser, IA; Lehne, G; Lison, A; Neumayer, HH; Soulillou, JP; Winkler, M, 2004
)
0.58
" The role of immunosuppressants in post-transplant outcome is crucial, and associations between exposure-related pharmacokinetic parameters and clinical outcome have been made for several drugs in this class."( Pharmacokinetics of immunosuppressants: a perspective on ethnic differences.
Dirks, NL; Huth, B; Meibohm, B; Yates, CR, 2004
)
0.32
" The objective of this randomized, double-blind, placebo-controlled, dose-escalating Phase 1 study was to evaluate the pharmacokinetic profile of different dosing regimens of everolimus."( Pharmacokinetics of the immunosuppressant everolimus in maintenance renal transplant patients.
Budde, K; Fritsche, L; Glander, P; Neumayer, HH; Slowinski, T; Waiser, J, 2005
)
0.79
" Everolimus half-life was only prolonged to a minor extent (32 +/- 6 vs."( Pharmacokinetic interaction between verapamil and everolimus in healthy subjects.
Allison, MJ; Beyer, D; Bizot, MN; Jiang, Q; Kovarik, JM; Schmouder, RL, 2005
)
1.49
"When CsA pharmacokinetic profiles were considered, the CsA dose requirement was not lower in Chinese patients receiving everolimus than that in patients receiving azathioprine."( Influence of everolimus on cyclosporine Neoral pharmacokinetics in Chinese de novo cardiac transplant recipients.
Chi, NH; Chou, NK; Hsu, RB; Ko, WJ; Tsao, CI; Wang, CH; Wang, SS; Wu, FL, 2006
)
0.91
"2 ng/mL, Cmax 10."( Differential pharmacokinetic interaction of tacrolimus and cyclosporine on everolimus.
Curtis, JJ; Hricik, DE; Kovarik, JM; Pescovitz, MD; Scantlebury, V; Vasquez, A, 2006
)
0.56
" The aim of this work was to characterize the disposition kinetics of everolimus in rats by physiologically based pharmacokinetic (PBPK) modeling."( Physiologically based pharmacokinetic (PBPK) modeling of everolimus (RAD001) in rats involving non-linear tissue uptake.
Kawai, R; Laplanche, R; Meno-Tetang, GM, 2007
)
0.82
"To use preclinical and clinical pharmacokinetic (PK)/pharmacodynamic (PD) modeling to predict optimal clinical regimens of everolimus, a novel oral mammalian target of rapamycin (mTOR) inhibitor, to carry forward to expanded phase I with tumor biopsy studies in cancer patients."( Identifying optimal biologic doses of everolimus (RAD001) in patients with cancer based on the modeling of preclinical and clinical pharmacokinetic and pharmacodynamic data.
Boulay, A; Hattenberger, M; Hazell, K; Judson, I; Kovarik, JM; Lane, HA; O'Reilly, T; Raymond, E; Shand, N; Stephan, C; Tanaka, C; Thomas, G; Zumstein-Mecker, S, 2008
)
0.82
" In order to identify a rationally based dose and schedule for cancer treatment, we have conducted a tumor pharmacodynamic phase I study in patients with advanced solid tumors."( Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors.
Baselga, J; Burris, H; Calvo, E; Dimitrijevic, S; Hazell, K; Jones, S; Judson, I; Lane, HA; Lebwohl, D; Macarulla, T; Martinelli, E; Ramon y Cajal, S; Ramos, FJ; Rojo, F; Shand, N; Stumm, M; Tabernero, J; Tang, P; Vidal, L; Zoellner, U, 2008
)
0.57
"We performed a dose-escalation study in advanced cancer patients administering oral everolimus 5 to 30 mg/wk, with pharmacokinetic (PK) and pharmacodynamic (PD) studies."( Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors.
Brock, C; Burris, HA; Faivre, S; Hazell, K; Jones, S; Judson, I; Kovarik, JM; Lane, HA; O'Donnell, A; Papadimitrakopoulou, V; Raymond, E; Rea, D; Shand, N; Zoellner, U, 2008
)
0.79
" Terminal half-life was 30 hours (range, 26 to 38 hours)."( Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors.
Brock, C; Burris, HA; Faivre, S; Hazell, K; Jones, S; Judson, I; Kovarik, JM; Lane, HA; O'Donnell, A; Papadimitrakopoulou, V; Raymond, E; Rea, D; Shand, N; Zoellner, U, 2008
)
0.56
"This article summarizes this analysis, with special focus on the pharmacokinetic characteristics of everolimus and on the results of its use in renal transplantation."( Everolimus: an update on the mechanism of action, pharmacokinetics and recent clinical trials.
Sánchez-Fructuoso, AI, 2008
)
2
"To describe the management of a pharmacokinetic interaction between azole antifungals (fluconazole and voriconazole) and everolimus in a patient who underwent an orthotopic liver transplant."( Pharmacokinetic interaction between everolimus and antifungal triazoles in a liver transplant patient.
Adani, GL; Baccarani, U; Baraldo, M; Cojutti, P; Franceschi, L; Furlanut, M; Londero, A; Pea, F; Tavio, M; Viale, P, 2008
)
0.83
" This study assessed the safety and the pharmacokinetic interactions of everolimus and paclitaxel in patients with advanced malignancies."( Safety and pharmacokinetics of paclitaxel and the oral mTOR inhibitor everolimus in advanced solid tumours.
Berton Rigaud, D; Bootle, D; Bourbouloux, E; Calvo, F; Campone, M; Dutreix, C; Levy, V; Raymond, E; Shand, N; Zoellner, U, 2009
)
0.82
"Treatment of Japanese cancer patients with RAD001 may be undertaken with the expectation that previously determined pharmacokinetic and safety profiles apply."( Phase I clinical and pharmacokinetic study of RAD001 (everolimus) administered daily to Japanese patients with advanced solid tumors.
Doi, T; Kobayashi, K; Kurei, K; Miyazaki, M; Nakagawa, K; Ohtsu, A; Okamoto, I; Tsuya, A, 2010
)
0.61
"Comparative pharmacokinetic (PK) analysis of the mTOR inhibitor RAD001 (everolimus) in rats and mice."( Comparative pharmacokinetics of RAD001 (everolimus) in normal and tumor-bearing rodents.
Allegrini, PR; Brueggen, J; Bruelisauer, A; Gschwind, HP; Kawai, R; Kretz, O; Lane, HA; McMahon, L; McSheehy, PM; O'Reilly, T, 2010
)
0.86
" Blood concentrations of everolimus were measured up to 240 min, and pharmacokinetic parameters were calculated."( Effect of itraconazole on the pharmacokinetics of everolimus administered by different routes in rats.
Inui, K; Katsura, T; Masuda, S; Sato, E; Yano, I; Yokomasu, A, 2009
)
0.91
" Inhibition of glucose metabolism measured by (18)F-FDG PET was postulated to serve as a pharmacodynamic marker in everolimus-treated non-small cell lung cancer (NSCLC) patients."( Downregulation of 18F-FDG uptake in PET as an early pharmacodynamic effect in treatment of non-small cell lung cancer with the mTOR inhibitor everolimus.
Bangard, C; Boellaard, R; Dietlein, M; Dimitrijevic, S; Eschner, W; Giaccone, G; Gross, SH; Hayes, W; Hoekstra, OS; Hoetjes, N; Kobe, C; Lammertsma, AA; Nogová, L; Pellas, T; Schmidt, K; Thomas, RK; Wolf, J; Zander, T, 2009
)
0.76
" The present pharmacokinetic sub-study from the SPIRIT III Randomized and Controlled Trial (RCT) was to evaluate systemic exposure of patients to everolimus and to further demonstrate safety following implantation of XIENCE Vstents with everolimus doses ranging from 53 to 181 microg."( Pharmacokinetic sub-study in the SPIRIT III Randomized and Controlled Trial of XIENCE V everolimus eluting coronary stent system.
Bol, C; Cannon, L; Gordon, P; Pierson, W; Saucedo, J; Sood, P; Sudhir, K; Wang, Q, 2010
)
0.78
" The authors determined whether a pharmacokinetic interaction occurs between sotrastaurin and everolimus, both of which are substrates and inhibitors of CYP3A4."( Sotrastaurin and everolimus pharmacokinetics after single-dose coadministration.
Antunes, MC; Bartlett, M; Kovarik, JM; Marbach, P; Rordorf, C; van Marle, S; Winter, S, 2010
)
0.92
" Clinical and pharmacokinetic data were collected to Day 5 after each treatment."( Sotrastaurin and everolimus pharmacokinetics after single-dose coadministration.
Antunes, MC; Bartlett, M; Kovarik, JM; Marbach, P; Rordorf, C; van Marle, S; Winter, S, 2010
)
0.7
"A prospective randomized phase II pharmacokinetic study was conducted comparing two fixed EVL dosages (0."( Interaction between everolimus and tacrolimus in renal transplant recipients: a pharmacokinetic controlled trial.
Brunet, M; Cabello, M; del Castillo, D; Fernández, AM; Grinyó, JM; Pallardó, L; Pascual, J, 2010
)
0.68
" The authors conducted a phase 1 and pharmacokinetic study of everolimus and docetaxel for recurrent NSCLC."( Phase 1 and pharmacokinetic study of everolimus, a mammalian target of rapamycin inhibitor, in combination with docetaxel for recurrent/refractory nonsmall cell lung cancer.
Egorin, MJ; Fu, H; Harvey, RD; Kauh, J; Khuri, FR; Owonikoko, TK; Ramalingam, SS; Saba, N; Shin, DM; Strychor, S; Sun, SY; Tighiouart, M, 2010
)
0.87
" Full pharmacokinetic studies of everolimus and CsA were studied at the period of 4-8 weeks after CsA reduction."( Long-term outcome of very early cyclosporine minimization and de novo everolimus therapy in kidney transplant recipients: a pharmacokinetic guided approach.
Chalermsanyakorn, P; Jirasiritham, S; Sumethkul, V; Tankee, P, 2010
)
0.88
" Primary end points were pharmacokinetic parameters and safety and tolerability."( Two-dose-level confirmatory study of the pharmacokinetics and tolerability of everolimus in Chinese patients with advanced solid tumors.
Cherfi, A; Jappe, A; Shen, L; Wang, H; Wu, Y; Xu, B; Ye, D; Yuan, R, 2011
)
0.6
" The observed safety and pharmacokinetic profile of everolimus from this study were consistent with previous studies."( Two-dose-level confirmatory study of the pharmacokinetics and tolerability of everolimus in Chinese patients with advanced solid tumors.
Cherfi, A; Jappe, A; Shen, L; Wang, H; Wu, Y; Xu, B; Ye, D; Yuan, R, 2011
)
0.85
"We retrospectively analyzed data from pharmacokinetic studies of patients implanted with an 18-mm DES: Cypher stent (sirolimus, n = 10), Endeavor stent (zotarolimus, n = 7), Xience V stent (everolimus, n = 6), and Nobori stent (biolimus A9, n = 10), in multicenter trials of Japan."( Comparison of pharmacokinetics of the limus-eluting stents in Japanese patients.
Mitsudo, K; Nakamura, M; Otsuka, Y; Saito, S; Shuto, H, 2011
)
0.56
" The other pharmacokinetic parameters of four DESs differed according to characteristics of the coated drug."( Comparison of pharmacokinetics of the limus-eluting stents in Japanese patients.
Mitsudo, K; Nakamura, M; Otsuka, Y; Saito, S; Shuto, H, 2011
)
0.37
"In Japanese patients, systemic exposure was low, regardless of the type of limus drug eluted from the stents; but specific pharmacokinetic activities were varied according to the drug characteristics, concentration, and DES design."( Comparison of pharmacokinetics of the limus-eluting stents in Japanese patients.
Mitsudo, K; Nakamura, M; Otsuka, Y; Saito, S; Shuto, H, 2011
)
0.37
" There was no pharmacokinetic interaction between everolimus and capecitabine."( Phase I and pharmacokinetic study of capecitabine and the oral mTOR inhibitor everolimus in patients with advanced solid malignancies.
Beijnen, JH; Deenen, MJ; Klümpen, HJ; Richel, DJ; Schellens, JH; Sparidans, RW; Weterman, MJ; Wilmink, JW, 2012
)
0.86
" We evaluated the safety, biologic activity, and pharmacokinetic profile of oral AEE788, a selective inhibitor of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF), plus oral RAD001, a mammalian target of rapamycin inhibitor, in glioblastoma patients."( Pharmacokinetic drug interaction between AEE788 and RAD001 causing thrombocytopenia in patients with glioblastoma.
Alfred Yung, WK; Cloughesy, T; Conrad, C; DiLea, C; Dugan, M; Huang, J; Maes, A; Mietlowski, W; Reardon, DA; Rich, J; Yung, L, 2012
)
0.38
" The purpose of this study was to measure the pharmacokinetic profile of everolimus following stent implantation."( Pharmacokinetic analysis after implantation of everolimus-eluting self-expanding stents in the peripheral vasculature.
Hausegger, KA; Koppensteiner, R; Lammer, J; Menon, RM; Scheinert, D; Schroë, H; Schwartz, LB; Vermassen, F, 2012
)
0.87
" A population pharmacokinetic model was developed and demographic factors and genetic polymorphisms in genes coding for ABCB1, CYP3A5, CYP2C8 and PXR were included as covariates."( Population pharmacokinetics and pharmacogenetics of everolimus in renal transplant patients.
de Fijter, JW; den Hartigh, J; Guchelaar, HJ; Moes, DJ; Press, RR; van der Straaten, T, 2012
)
0.63
" The pharmacokinetic limited sampling model (C(trough) and whole blood drug concentration at 2 hours postdose [C(2)]) resulted in a significantly improved prediction of everolimus exposure compared with the widely used C(trough) monitoring."( Population pharmacokinetics and pharmacogenetics of everolimus in renal transplant patients.
de Fijter, JW; den Hartigh, J; Guchelaar, HJ; Moes, DJ; Press, RR; van der Straaten, T, 2012
)
0.82
"A two-compartment pharmacokinetic model with lag-time describing the concentration-time profile of oral everolimus in renal transplant patients has been developed using pharmacokinetic modelling."( Population pharmacokinetics and pharmacogenetics of everolimus in renal transplant patients.
de Fijter, JW; den Hartigh, J; Guchelaar, HJ; Moes, DJ; Press, RR; van der Straaten, T, 2012
)
0.84
"The aim of this study was, using routine drug monitoring data, to identify patient characteristics that may influence everolimus (EVE) pharmacokinetic parameters and to develop a population pharmacokinetic model to predict EVE whole blood concentrations in cardiac recipients."( Population pharmacokinetics of everolimus in cardiac recipients: comedications, ABCB1, and CYP3A5 polymorphisms.
Antignac, M; Bezian, E; Farinotti, R; Fernandez, C; Goldwirt, L; Lemaitre, F; Urien, S; Varnous, S, 2012
)
0.87
" Population pharmacokinetic modeling was carried out using the nonlinear mixed-effects modeling program."( Population pharmacokinetics of everolimus in cardiac recipients: comedications, ABCB1, and CYP3A5 polymorphisms.
Antignac, M; Bezian, E; Farinotti, R; Fernandez, C; Goldwirt, L; Lemaitre, F; Urien, S; Varnous, S, 2012
)
0.66
"The pharmacokinetic of EVE in cardiac recipients was best described by a 1-compartment model."( Population pharmacokinetics of everolimus in cardiac recipients: comedications, ABCB1, and CYP3A5 polymorphisms.
Antignac, M; Bezian, E; Farinotti, R; Fernandez, C; Goldwirt, L; Lemaitre, F; Urien, S; Varnous, S, 2012
)
0.66
"This study is the first population pharmacokinetic model of EVE in heart transplantation patients."( Population pharmacokinetics of everolimus in cardiac recipients: comedications, ABCB1, and CYP3A5 polymorphisms.
Antignac, M; Bezian, E; Farinotti, R; Fernandez, C; Goldwirt, L; Lemaitre, F; Urien, S; Varnous, S, 2012
)
0.66
" Temsirolimus and everolimus have both been associated with improved outcomes in patients with mRCC but, as reviewed in this paper, the pharmacokinetic characteristics of these agents differ in many respects."( Oral and intravenously administered mTOR inhibitors for metastatic renal cell carcinoma: pharmacokinetic considerations and clinical implications.
Boni, JP; Danesi, R; Ravaud, A, 2013
)
0.72
" Blood samples for pharmacokinetic assessment were collected at predetermined time points up to 168 hours postdosing."( Effects of hepatic impairment on the pharmacokinetics of everolimus: a single-dose, open-label, parallel-group study.
Anak, O; Bouillaud, E; Kobalava, Z; Kunz, T; Paquet, T; Peveling-Oberhag, J; Sellami, D; Urva, S; Yong, WP; Zeuzem, S, 2013
)
0.64
" This study assessed the pharmacokinetics (PK) of everolimus and sorafenib alone and in combination in plasma and tissues, developed physiologically based pharmacokinetic (PBPK) models in mice, and assessed the possibility of PK drug interactions."( Physiologically based pharmacokinetic models for everolimus and sorafenib in mice.
Fetterly, GJ; Hylander, BH; Jusko, WJ; Ma, WW; Pawaskar, DK; Repasky, EA; Straubinger, RM, 2013
)
0.9
" These studies provide the basis for pharmacodynamic evaluation of these drugs in patient-derived primary pancreatic adenocarcinomas explants."( Physiologically based pharmacokinetic models for everolimus and sorafenib in mice.
Fetterly, GJ; Hylander, BH; Jusko, WJ; Ma, WW; Pawaskar, DK; Repasky, EA; Straubinger, RM, 2013
)
0.64
"There is limited data analyzing the pharmacokinetic (PK) and pharmacodynamic (PD) properties of everolimus (EVR) between African Americans and Caucasians."( Racial comparisons of everolimus pharmacokinetics and pharmacodynamics in adult kidney transplant recipients.
Baliga, P; Belk, L; Bratton, C; Chavin, K; Fleming, J; McGillicuddy, J; Meadows, H; Pilch, N; Srinivas, T; Taber, DJ, 2013
)
0.92
" [(18)F]Fluorodeoxyglucose positron emission tomography (FDG-PET) was performed as a pharmacodynamic marker of mTOR inhibition, and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was performed as an indicator of tumor perfusion changes, at 3 time points."( Phase 1 and pharmacodynamic trial of everolimus in combination with cetuximab in patients with advanced cancer.
Avadhani, AN; Ciunci, CA; Divgi, CR; Englander, S; Flaherty, KT; Giantonio, BJ; Harlacker, K; Kang, HC; O'Dwyer, PJ; Perini, RF; Redlinger, M; Rosen, MA; Schnall, M; Song, HK; Sun, W; Troxel, A, 2014
)
0.68
" Pharmacodynamic biomarkers could be useful in distinguishing between (1) disease resistance that occurs even though mTOR is successfully inhibited (suggesting a need for a different treatment strategy) and (2) resistance that might respond to changes in drug dosage or schedule."( Ribosomal S6 kinase and AKT phosphorylation as pharmacodynamic biomarkers in patients with myelodysplastic syndrome treated with RAD001.
Advani, AS; Bates, J; Kalaycio, M; Maciejewski, J; Mahfouz, RZ; Rybicki, L; Saunthararajah, Y; Sekeres, M; Tripp, B, 2014
)
0.4
" Therefore, to evaluate the pharmacodynamic effects of RAD001, S6 and AKT phosphorylation (pS6, pAKT) were measured by peripheral blood flow cytometry."( Ribosomal S6 kinase and AKT phosphorylation as pharmacodynamic biomarkers in patients with myelodysplastic syndrome treated with RAD001.
Advani, AS; Bates, J; Kalaycio, M; Maciejewski, J; Mahfouz, RZ; Rybicki, L; Saunthararajah, Y; Sekeres, M; Tripp, B, 2014
)
0.4
" There were no clinical responses despite the biomarker evidence of intended pharmacodynamic effect."( Ribosomal S6 kinase and AKT phosphorylation as pharmacodynamic biomarkers in patients with myelodysplastic syndrome treated with RAD001.
Advani, AS; Bates, J; Kalaycio, M; Maciejewski, J; Mahfouz, RZ; Rybicki, L; Saunthararajah, Y; Sekeres, M; Tripp, B, 2014
)
0.4
" No difference of EVL Cmax or Tmax was found after atorvastatin coadministration."( Effects of atorvastatin on the pharmacokinetics of everolimus among kidney transplant recipients.
Avihingsanon, Y; Chancharoenthana, W; Praditpornsilpa, K; Somparn, P; Townamchai, N; Vadcharavivad, S; Wanitchanont, A, 2014
)
0.65
" This review focuses on the pharmacokinetic interactions and exposure-response relationships of mTOR inhibitors and tacrolimus (TAC), the most widely used CNI."( Focus on mTOR inhibitors and tacrolimus in renal transplantation: pharmacokinetics, exposure-response relationships, and clinical outcomes.
Christians, U; Kaplan, B; Shihab, F; Smith, L; Wellen, JR, 2014
)
0.4
"2459 ng/mL and mean peak concentration was 13."( Pharmacokinetics of everolimus when combined with cyclosporine in Japanese de novo renal transplant recipients.
Goto, N; Hirano, M; Hoshinaga, K; Kusaka, M; Sasaki, H; Uchida, K; Watarai, Y, 2014
)
0.73
"The pharmacokinetic characteristics of everolimus in Japanese de novo renal transplant patients did not differ from those previously observed in non-Japanese patients, hence the same dosage of everolimus may be acceptable in Japanese patients."( Pharmacokinetics of everolimus when combined with cyclosporine in Japanese de novo renal transplant recipients.
Goto, N; Hirano, M; Hoshinaga, K; Kusaka, M; Sasaki, H; Uchida, K; Watarai, Y, 2014
)
1
"In a pharmacokinetic substudy of the randomized ASSET trial, 46 de novo kidney transplant patients receiving very low (1."( The pharmacokinetics of everolimus in de novo kidney transplant patients receiving tacrolimus: an analysis from the randomized ASSET study.
Christiaans, MH; Kovarik, JM; Pascual, J; Rostaing, L, 2014
)
0.71
"At month 12, mean values for tacrolimus trough concentration (C0), peak concentration (Cmax), and AUC0-12 in the very low tacrolimus group were approximately half that in the low tacrolimus group, but everolimus dose, C0, Cmax, and AUC0-12 were virtually identical in both groups."( The pharmacokinetics of everolimus in de novo kidney transplant patients receiving tacrolimus: an analysis from the randomized ASSET study.
Christiaans, MH; Kovarik, JM; Pascual, J; Rostaing, L, 2014
)
0.9
" Everolimus has a favorable pharmacokinetic (PK) profile in early breast cancer studies."( Evaluating the pharmacokinetics and pharmacodynamics of everolimus for treating breast cancer.
Awada, A; Barthélémy, P; Gombos, A, 2015
)
1.57
"This review article focuses on the biological rationale of using everolimus in breast cancer and on latest advances in the field of everolimus-based combinations with an emphasis on the PK and pharmacodynamic parameters of the drug throughout different studies."( Evaluating the pharmacokinetics and pharmacodynamics of everolimus for treating breast cancer.
Awada, A; Barthélémy, P; Gombos, A, 2015
)
0.9
" We studied the pharmacodynamic effects of everolimus in resectable non-small cell lung cancer (NSCLC) to inform further development of these agents in lung cancer."( A Translational, Pharmacodynamic, and Pharmacokinetic Phase IB Clinical Study of Everolimus in Resectable Non-Small Cell Lung Cancer.
Auffermann, WF; Bechara, R; Behera, M; Chen, Z; Deng, X; Force, SD; Fu, H; Fu, RW; Gal, AA; Harvey, RD; Khuri, FR; Kim, S; Mendel, J; Miller, DL; Owonikoko, TK; Pickens, A; Ramalingam, SS; Rogatko, A; Rossi, MR; Saba, NF; Sica, GL; Sun, SY; Tighiouart, M; Torres, WE, 2015
)
0.91
" Nevertheless, exposures produced by the initial EVR dose, the steady state pharmacokinetic and long-term safety and tolerability have not been explored in detail."( Pharmacokinetics and Long-Term Safety and Tolerability of Everolimus in Renal Transplant Recipients Converted From Cyclosporine.
de Paula, MI; Felipe, CR; Hannun, PG; Medina-Pestana, JO; Oliveira, NI; Tedesco-Silva, H, 2016
)
0.68
" Weekly monitoring of EVR blood concentrations was followed by a full 12 hour pharmacokinetic profile 28 days after conversion."( Pharmacokinetics and Long-Term Safety and Tolerability of Everolimus in Renal Transplant Recipients Converted From Cyclosporine.
de Paula, MI; Felipe, CR; Hannun, PG; Medina-Pestana, JO; Oliveira, NI; Tedesco-Silva, H, 2016
)
0.68
"36 l day(-1) , with an elimination half-life of 11."( Influence of tumour burden on trastuzumab pharmacokinetics in HER2 positive non-metastatic breast cancer.
Arnedos, M; Bachelot, T; Bernadou, G; Campone, M; Diéras, V; Gouilleux-Gruart, V; Jimenez, M; Lokiec, F; Merlin, JL; Paintaud, G; Rezai, K; Ternant, D, 2016
)
0.43
" The elimination half-life of trastuzumab was shorter in the present population of patients than in metastatic breast cancer patients previously studied."( Influence of tumour burden on trastuzumab pharmacokinetics in HER2 positive non-metastatic breast cancer.
Arnedos, M; Bachelot, T; Bernadou, G; Campone, M; Diéras, V; Gouilleux-Gruart, V; Jimenez, M; Lokiec, F; Merlin, JL; Paintaud, G; Rezai, K; Ternant, D, 2016
)
0.43
"A pharmacokinetic interaction between tacrolimus and everolimus was not observed clinically in renal transplant patients."( Influence of everolimus on the pharmacokinetics of tacrolimus in Japanese renal transplant patients.
Akamine, Y; Habuchi, T; Inoue, T; Kagaya, H; Miura, M; Niioka, T; Numakura, K; Saito, M; Satoh, S; Yamamoto, R, 2016
)
1.05
" Serial pharmacokinetic sampling was performed on days 1 and 15."( Everolimus pharmacokinetics and its exposure-toxicity relationship in patients with thyroid cancer.
de Wit, D; den Hartigh, J; Gelderblom, H; Guchelaar, HJ; Kapiteijn, E; Links, TP; Moes, DJ; Roozen, CF; Schneider, TC; van der Hoeven, JJ; van Erp, NP, 2016
)
1.88
"A semi-physiological pharmacokinetic model for everolimus was developed from pharmacokinetic data from 73 patients by non-linear mixed-effects modeling."( A Semi-Physiological Population Model to Quantify the Effect of Hematocrit on Everolimus Pharmacokinetics and Pharmacodynamics in Cancer Patients.
Burger, DM; de Wit, D; Huitema, AD; Kapiteijn, E; Ter Heine, R; van Erp, NP; van Herpen, CM; Willemsen, A, 2016
)
0.92
"A population pharmacokinetic model was developed for everolimus in cancer patients."( A Semi-Physiological Population Model to Quantify the Effect of Hematocrit on Everolimus Pharmacokinetics and Pharmacodynamics in Cancer Patients.
Burger, DM; de Wit, D; Huitema, AD; Kapiteijn, E; Ter Heine, R; van Erp, NP; van Herpen, CM; Willemsen, A, 2016
)
0.91
" The objective of this study was to assess the analytical suitability of a commercial kit-based phosphoflow cytometry protocol using whole blood (WBS) to measure the level of phosphorylated S6RP (p-S6RP) in T-cell subsets to study the pharmacodynamic effects of mTOR inhibitors (mTORi)."( Performance of a phosphoflow assay to determine phosphorylation of S6 ribosomal protein as a pharmacodynamic read out for mTOR inhibition.
Abdel-Kahaar, E; Hartmann, B; Kabakchiev, M; Shipkova, M; Wieland, E, 2016
)
0.43
" Here, we measured blood concentrations of everolimus to obtain the population pharmacokinetic parameters and to examine the relationship between blood concentration and clinical outcomes."( Population Pharmacokinetics of Everolimus in Relation to Clinical Outcomes in Patients With Advanced Renal Cell Carcinoma.
Fukudo, M; Kamba, T; Masuda, S; Matsubara, K; Ogawa, O; Sato, E; Shinsako, K; Tanaka, A; Yamasaki, T; Yano, I, 2016
)
0.98
" Population pharmacokinetic analysis was conducted using the NONMEM software."( Population Pharmacokinetics of Everolimus in Relation to Clinical Outcomes in Patients With Advanced Renal Cell Carcinoma.
Fukudo, M; Kamba, T; Masuda, S; Matsubara, K; Ogawa, O; Sato, E; Shinsako, K; Tanaka, A; Yamasaki, T; Yano, I, 2016
)
0.72
"This study reports the first population pharmacokinetic parameters of everolimus in patients with RCC."( Population Pharmacokinetics of Everolimus in Relation to Clinical Outcomes in Patients With Advanced Renal Cell Carcinoma.
Fukudo, M; Kamba, T; Masuda, S; Matsubara, K; Ogawa, O; Sato, E; Shinsako, K; Tanaka, A; Yamasaki, T; Yano, I, 2016
)
0.95
"AUC0-4h, peak concentration (Cmax), Cmax/minimum concentration, and peak-trough fluctuation in the resting period were significantly lower than those in the active period (p = 0."( Circadian pharmacokinetics and limited sampling strategy of everolimus in heart transplant patients
.
Fukushima, N; Kawabata, A; Kuwahara, T; Matsuda, S; Nakajima, S; Nakatani, T; Sato, T; Seguchi, O; Takada, M; Terada, Y; Wada, K; Watanabe, T; Yanase, M, 2017
)
0.7
"Although the proportion of elderly patients among renal transplant recipients has increased, pharmacokinetic (PK) studies of immunosuppressants rarely include older patients."( Longitudinal Pharmacokinetics of Everolimus When Combined With Low-level of Tacrolimus in Elderly Renal Transplant Recipients.
Agena, F; Coelho, V; David-Neto, E; de Almeida Rezende Ebner, P; Galante, NZ; Lemos, FBC; Ramos, F; Romano, P; Triboni, AHK, 2017
)
0.74
"2 ng/mL), Cmax (19."( Longitudinal Pharmacokinetics of Everolimus When Combined With Low-level of Tacrolimus in Elderly Renal Transplant Recipients.
Agena, F; Coelho, V; David-Neto, E; de Almeida Rezende Ebner, P; Galante, NZ; Lemos, FBC; Ramos, F; Romano, P; Triboni, AHK, 2017
)
0.74
"The objective of the present study was to assess the effect of cyclosporine (CsA) on the pharmacokinetic parameters of mycophenolic acid (MPA), an active mycophenolate mofetil (MMF) metabolite, and to compare with the effect of everolimus (EVR)."( Influence of cyclosporine and everolimus on the main mycophenolate mofetil pharmacokinetic parameters: Cross-sectional study.
Kaduševičius, E; Marquet, P; Noreikaitė, A; Saint-Marcoux, F; Stankevičius, E, 2017
)
0.93
" We aimed to develop pharmacokinetic population models and Bayesian estimators based on a limited sampling strategy for estimation of dose interval exposures of everolimus in whole blood and peripheral blood mononuclear cells (PBMCs) in renal transplant recipients."( A Limited Sampling Strategy to Estimate Exposure of Everolimus in Whole Blood and Peripheral Blood Mononuclear Cells in Renal Transplant Recipients Using Population Pharmacokinetic Modeling and Bayesian Estimators.
Åsberg, A; Debord, J; Marquet, P; Robertsen, I; Woillard, JB, 2018
)
0.93
" The pharmacokinetic model was developed using non-parametric modeling and its performances and those of the derived Bayesian estimator were evaluated in the validation set."( A Limited Sampling Strategy to Estimate Exposure of Everolimus in Whole Blood and Peripheral Blood Mononuclear Cells in Renal Transplant Recipients Using Population Pharmacokinetic Modeling and Bayesian Estimators.
Åsberg, A; Debord, J; Marquet, P; Robertsen, I; Woillard, JB, 2018
)
0.73
"Pharmacodynamic (PD) monitoring may complement routine pharmacokinetic monitoring of mTOR inhibitors (mTORis) in an attempt to better guide individualized sirolimus (SRL) or everolimus (EVR) treatment after organ transplantation."( Pharmacodynamic Monitoring of mTOR Inhibitors.
Brunet, M; Marquet, P; Millán, O; Wieland, E, 2019
)
0.71
" Pharmacokinetic assessment was conducted using blood samples collected on cycle 1, days 8 and 15."( A Phase I Study of Safety, Pharmacokinetics, and Pharmacodynamics of Concurrent Everolimus and Buparlisib Treatment in Advanced Solid Tumors.
Akce, M; Alese, OB; Bilen, MA; Carthon, B; Chen, Z; Collins, H; El-Rayes, BF; Harris, WB; Harvey, RD; Khuri, FR; Kudchagkar, RR; Lawson, DH; Lewis, C; Lonial, S; Owonikoko, TK; Ramalingam, SS; Shaib, WL; Sica, GL; Steuer, CE; Wu, C; Zhang, C, 2020
)
0.79
" Steady-state pharmacokinetic analysis showed dose-normalized maximum concentrations and AUC values for everolimus and buparlisib in combination to be comparable with single-agent pharmacokinetic."( A Phase I Study of Safety, Pharmacokinetics, and Pharmacodynamics of Concurrent Everolimus and Buparlisib Treatment in Advanced Solid Tumors.
Akce, M; Alese, OB; Bilen, MA; Carthon, B; Chen, Z; Collins, H; El-Rayes, BF; Harris, WB; Harvey, RD; Khuri, FR; Kudchagkar, RR; Lawson, DH; Lewis, C; Lonial, S; Owonikoko, TK; Ramalingam, SS; Shaib, WL; Sica, GL; Steuer, CE; Wu, C; Zhang, C, 2020
)
1
" Pharmacokinetic analyses were accompanied by pharmacodynamics assessment of E by FDG-PET."( Sorafenib and everolimus in patients with advanced solid tumors and KRAS-mutated NSCLC: A phase I trial with early pharmacodynamic FDG-PET assessment.
Abdulla, DSY; Backes, H; Behringer, D; Buettner, R; Eichstaedt, M; Fischer, RN; Franklin, J; Frechen, S; Fuhr, U; Gardizi, M; Junge, L; Kaminsky, B; Kinzig, M; Kobe, C; Limburg, M; Mattonet, C; Merkelbach-Bruse, S; Michels, S; Nogova, L; Ouyang, D; Persigehl, T; Rokitta, D; Scheffler, M; Schnell, R; Sörgel, F; Sos, ML; Stelzer, C; Suleiman, AA; Taubert, M; Tian, Y; Wolf, J, 2020
)
0.92
" We conducted a phase 1 study in advanced solid tumour patients to assess safety, efficacy and pharmacodynamic (PD) outcomes."( Phase 1 safety and pharmacodynamic study of lenalidomide combined with everolimus in patients with advanced solid malignancies with efficacy signal in adenoid cystic carcinoma.
Alese, OB; Bilen, MA; Carthon, BC; Chen, Z; El-Rayes, BF; Harris, WB; Harvey, RD; Hossain, MS; Khuri, FR; Lawson, DH; Lewis, C; Lonial, S; Owonikoko, TK; Ramalingam, SS; Shaib, W; Steuer, CE; Waller, EK; Wu, C; Zhang, C, 2020
)
0.79
" Here, we report a patient who showed high sunitinib concentrations, in addition to pharmacokinetic changes in tacrolimus and everolimus after sunitinib therapy."( Model-based assessment of pharmacokinetic changes of sunitinib, tacrolimus, and everolimus in a patient with metastatic renal cell carcinoma after renal transplantation.
Fujisawa, M; Furukawa, J; Harada, K; Ito, T; Ogawa, S; Omura, T; Yamamoto, K; Yano, I, 2020
)
0.99
"This study aimed to evaluate the pharmacokinetic profile and tissue effects of everolimus delivered into arterial wall using biodegradable nanospheres."( Nanospheres encapsulated everolimus delivery into arterial wall-the tissue pharmacokinetics and vascular response experimental study.
Bryła-Wojciechowska, A; Buszman, PE; Buszman, PP; Błachut, A; Gąsior, P; Jelonek, K; Kachel, M; Kasperczyk, J; Kiesz, RS; Krauze, A; Milewski, K; Musiał-Kulik, M; Rousselle, SD; Tellez, A, 2021
)
1.15
"A pharmacokinetic study included 28 porcine coronary arterial segments initially injured with balloon angioplasty followed by the local delivery of everolimus encapsulated in nanospheres (EEN) via injection through a microporous delivery catheter."( Nanospheres encapsulated everolimus delivery into arterial wall-the tissue pharmacokinetics and vascular response experimental study.
Bryła-Wojciechowska, A; Buszman, PE; Buszman, PP; Błachut, A; Gąsior, P; Jelonek, K; Kachel, M; Kasperczyk, J; Kiesz, RS; Krauze, A; Milewski, K; Musiał-Kulik, M; Rousselle, SD; Tellez, A, 2021
)
1.12
" However, the pharmacokinetic and pharmacodynamic information for EVL combined with TAC is limited."( Pharmacodynamic Drug-Drug Interaction on Human Peripheral Blood Mononuclear Cells Between Everolimus and Tacrolimus at the Therapeutic Concentration Range in Renal Transplantation.
Akashi, I; Akiyama, S; Hirano, T; Iwamoto, H; Kihara, Y; Konno, O; Oda, T; Okihara, M; Osato, S; Takeuchi, H; Tanaka, S; Unezaki, S; Yoshinaga, R, 2021
)
0.84
" To this end, we investigated experimentally and assessed computationally the in vitro pharmacodynamic drug-drug interactions of the various dual and triple combinations to assess their subsequent combinatorial effects (synergistic/additive/antagonistic) in a HER2-therapy resistant BC cell line, JIMT-1."( Experimental and computational assessment of the synergistic pharmacodynamic drug-drug interactions of a triple combination therapy in refractory HER2-positive breast cancer cells.
Ait-Oudhia, S; Vaidya, TR, 2022
)
0.72
" In this study, a population pharmacokinetic analysis was conducted to improve the individualization of everolimus therapy."( Population pharmacokinetics of everolimus in adult liver transplant patients: Comparison to tacrolimus disposition and extrapolation to pediatrics.
Hata, K; Hatano, E; Hira, D; Hirai, M; Imai, S; Ito, T; Itohara, K; Matsubara, K; Nakagawa, S; Nakagawa, T; Sugimoto, M; Terada, T; Yano, I; Yonezawa, A, 2022
)
1.22
"When highly purified cannabidiol (CBD; Epidiolex) and the mammalian target of rapamycin inhibitor everolimus are used concomitantly in the treatment of tuberous sclerosis complex, there is evidence of a pharmacokinetic (PK) interaction, leading to increased everolimus systemic exposure."( Pharmacokinetic Drug-Drug Interaction With Coadministration of Cannabidiol and Everolimus in a Phase 1 Healthy Volunteer Trial.
Berwaerts, J; Chen, C; Critchley, D; Hyland, K; Tayo, B; Thai, C; Wray, L, 2023
)
1.35

Compound-Compound Interactions

Inhibition of mTOR by rapalogs, such as rapamycin and everolimus combined with carboplatin chemotherapy may have activity in canine melanoma.

ExcerptReferenceRelevance
"The novel immunosuppressant SDZ RAD, 40-0 (2-hydroxyethyl)rapamycin, is an orally active rapamycin analogue developed for use in combination with cyclosporine (Neoral)."( Suppression of acute rejection in allogeneic rat lung transplantation: a study of the efficacy and pharmacokinetics of rapamycin derivative (SDZ RAD) used alone and in combination with a microemulsion formulation of cyclosporine.
Berry, GJ; Boeke, K; Christians, U; Hausen, B; Morris, RE; Segarra, IT, 1999
)
0.3
" We hypothesized that chloroquine in combination with tacrolimus and the rapamycin derivative SDZ-RAD can synergistically suppress T cell responses and antigen-presenting cell (APC) function in vitro."( Evaluation for synergistic suppression of T cell responses to minor histocompatibility antigens by chloroquine in combination with tacrolimus and a rapamycin derivative, SDZ-RAD.
Bader, S; Forooghian, F; Gilman, A; HayGlass, KT; Hsiao, CC; Rempel, J; Schultz, KR; Su, WN, 2002
)
0.31
"0 mg/day) combined with reduced-exposure CsA (Neoral) in de novo renal transplant patients."( Concentration-controlled everolimus (Certican): combination with reduced dose calcineurin inhibitors.
Pascual, J, 2005
)
0.63
"We evaluated the in vitro capacity of FK778, alone or in combination with other immunosuppressive drugs: Tacrolimus (TRL); Sirolimus (SRL), Everolimus (EVL), to inhibit clonal expansion of T-lymphocytes and expression of lymphocyte-activation surface antigens; secondly, we compared the immunosuppressive potential of FK778 combined with TRL, SRL and EVL with the same combinations using Mycophenolic acid (MPA) as antimetabolite."( Role of FK778 alone or in combination with tacrolimus or mTOR inhibitors as an immunomodulator of immunofunctions: in vitro evaluation of T cell proliferation and the expression of lymphocyte surface antigens.
Barceló, JJ; Brunet, M; Fortuna, V; Jiménez, O; Millán, O,
)
0.33
" In this study we sought to determine whether the oncolytic adenovirus Delta-24-RGD, in combination with everolimus (RAD001), would result in an enhanced anti-glioma effect in vivo."( Delta-24-RGD in combination with RAD001 induces enhanced anti-glioma effect via autophagic cell death.
Alonso, MM; Bekele, NB; Fueyo, J; Gomez-Manzano, C; Jiang, H; Kondo, S; Lang, FF; Xu, J; Yokoyama, T, 2008
)
0.56
" The objective of our study was to evaluate the effects of RAD001 on the growth of CaP in the bone, both alone and in combination with docetaxel and zoledronic acid."( RAD001 (Everolimus) inhibits growth of prostate cancer in the bone and the inhibitory effects are increased by combination with docetaxel and zoledronic acid.
Corey, E; Gross, TS; Morgan, TM; Pitts, TE; Poliachik, SL; Vessella, RL, 2008
)
0.78
"In this study, we examined whether cyclosporine was effective when combined with everolimus in clinical heart transplantation (HT)."( Heart transplantation under cyclosporine or tacrolimus combined with mycophenolate mofetil or everolimus.
Chen, YS; Chi, NH; Chou, NK; Huang, SC; Ko, WJ; Sun, CD; Tsao, CI; Wang, CH; Wang, SS; Wu, IH; Yu, HY, 2008
)
0.79
"Although EVR and FTY720 monotherapy delayed the progression of CAN, their combination with CsA had no beneficial effect."( Treatment of chronic allograft nephropathy at late stages using everolimus or FTY720 in combination with cyclosporine.
Baumann, M; Heemann, U; Liu, S; Lutz, J; Roos, M; Schmaderer, C; Strobl, M, 2008
)
0.58
" 44 (2007) 963-969] and developed a new method, namely the impregnation of sampling paper with a solution of plasma-protein, formic acid and ammonium acetate, in combination with the extraction of the blood spot by filter filtration."( Therapeutic drug monitoring of everolimus using the dried blood spot method in combination with liquid chromatography-mass spectrometry.
Christiaans, M; de Beer, Y; de Jong, GJ; Hoogtanders, K; Neef, C; Stolk, L; van der Heijden, J, 2009
)
0.64
"We report a case of a 54-year-old male renal transplant patient who received antifungal azole treatment in combination with the recently introduced immunosuppressant agent everolimus to prevent post-transplantation aspergillosis reactivation."( Management of metabolic cytochrome P450 3A4 drug-drug interaction between everolimus and azole antifungals in a renal transplant patient.
Abboud, I; Antoine, C; Benammar, M; Berge, M; Billaud, EM; Glotz, D; Lefeuvre, S, 2009
)
0.78
"In phase I, patients received imatinib (600/800 mg/day) combined with weekly (20 mg) or daily (2."( A phase I-II study of everolimus (RAD001) in combination with imatinib in patients with imatinib-resistant gastrointestinal stromal tumors.
Blay, JY; Demetri, GD; Dumez, H; Hollaender, N; Jappe, A; Morgan, JA; Ray-Coquard, I; Reichardt, P; Schöffski, P, 2010
)
0.68
" On the basis of promising preclinical data, the safety and tolerability of therapy with the mTOR inhibitor RAD001 in combination with radiation (RT) and temozolomide (TMZ) was evaluated in this Phase I study."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.61
"All patients received weekly oral RAD001 in combination with standard chemoradiotherapy, followed by RAD001 in combination with standard adjuvant temozolomide."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.61
" On the basis of these results, the recommended Phase II dosage currently being tested is RAD001 70 mg/week in combination with standard chemoradiotherapy."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.61
"RAD001 in combination with RT/TMZ and adjuvant TMZ was reasonably well tolerated."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.61
"The aim was to determine the potential of the allosteric mammalian target of rapamycin inhibitor, everolimus, to act in combination with cytotoxic anticancer compounds in vitro and in vivo."( Evaluation of the mTOR inhibitor, everolimus, in combination with cytotoxic antitumor agents using human tumor models in vitro and in vivo.
Brandt, R; Lane, HA; Lassota, P; McSheehy, PM; O'Reilly, T; Wartmann, M, 2011
)
0.87
"VEGF, mTOR, and EGFR inhibitors have demonstrated anti-tumor and anti-angiogenic effects alone and in combination with each other."( A phase I study of bevacizumab (B) in combination with everolimus (E) and erlotinib (E) in advanced cancer (BEE).
Bendell, JC; Blobe, GC; Broadwater, G; Bullock, KE; Gockerman, JP; Howard, LA; Hurwitz, HI; Lager, JJ; Meadows, KL; Morse, MA; O'Neill, MM; Petros, WP; Truax, R; Uronis, HE; Younis, I; Zafar, SY, 2011
)
0.62
"The present study was designed to investigate the effect of low-dose calcineurin inhibitor (CNI) tacrolimus combined with a mammalian target of rapamycin (mTOR) inhibitor on renal function in transplant recipients without allograft nephropathy."( Low-dose calcineurin inhibitor regimen combined with mammalian target of rapamycin inhibitors preserves kidney functions in renal transplant recipients without allograft nephropathy.
Gurkan, A; Kacar, S; Karaca, C; Tilif, S; Varılsuha, C, 2010
)
0.36
"1 years underwent renal transplantation and were switched to a new second-line treatment of low-dose CNI combined with an mTOR inhibitor, either sirolimus or everolimus."( Low-dose calcineurin inhibitor regimen combined with mammalian target of rapamycin inhibitors preserves kidney functions in renal transplant recipients without allograft nephropathy.
Gurkan, A; Kacar, S; Karaca, C; Tilif, S; Varılsuha, C, 2010
)
0.56
"Low-dose CNI combined with an mTOR inhibitor, as a replacement for mycophenolate mofetil or enteric-coated mycophenolate sodium, seemed to prevent renal dysfunction for at least 6 months among renal transplant patients without allograft nephropathy."( Low-dose calcineurin inhibitor regimen combined with mammalian target of rapamycin inhibitors preserves kidney functions in renal transplant recipients without allograft nephropathy.
Gurkan, A; Kacar, S; Karaca, C; Tilif, S; Varılsuha, C, 2010
)
0.36
"To determine the feasible dose and schedule for everolimus, an oral mTOR inhibitor, combined with vinorelbine and trastuzumab for patients with HER2-overexpressing metastatic breast cancer pretreated with trastuzumab."( Phase I trial of oral mTOR inhibitor everolimus in combination with trastuzumab and vinorelbine in pre-treated patients with HER2-overexpressing metastatic breast cancer.
Bergh, J; Cardoso, F; Dieras, V; Fasolo, A; Gianni, L; Jerusalem, G; Massacesi, C; Sahmoud, T; Vittori, L; Zhang, Y, 2011
)
0.9
" Moreover, trabectedin combined with everolimus may be more efficacious for the management of CCC."( The activity of trabectedin as a single agent or in combination with everolimus for clear cell carcinoma of the ovary.
Hayashi, M; Hisamatsu, T; Kawase, C; Kimura, T; Kurachi, H; Mabuchi, S; Mimura, K; Sawada, K; Takahashi, K; Takahashi, T, 2011
)
0.88
" Patients with HER2-overexpressing MBC who had progressed on trastuzumab-based therapy received trastuzumab every 3 weeks in combination with daily everolimus."( Phase I/II study of trastuzumab in combination with everolimus (RAD001) in patients with HER2-overexpressing metastatic breast cancer who progressed on trastuzumab-based therapy.
Booser, DJ; Coviello, J; Ensor, J; Esteva, FJ; Flores, PR; Hortobagyi, GN; Kindelberger, DW; Krop, IE; Moore, JA; Morrow, PK; Nuñez, R; Sahin, AA; Winer, EP; Wulf, GM; Xiong, Y; Yu, D; Zhang, S, 2011
)
0.82
"In our study, we investigated the efficacy of everolimus in combination with sildenafil on hemodynamic and morphological parameters in rats with monocrotaline-induced pulmonary hypertension (PH)."( Effects of everolimus in combination with sildenafil in monocrotaline-induced pulmonary hypertension in rats.
Atli, O; Burukoglu, D; Ilgin, S; Sirmagul, B, 2012
)
1.03
" Since strong anti-tumor properties became evident with respect to cell growth and adhesion dynamics, the triple drug combination might provide progress in the treatment of advanced prostate cancer."( Molecular targeting of prostate cancer cells by a triple drug combination down-regulates integrin driven adhesion processes, delays cell cycle progression and interferes with the cdk-cyclin axis.
Blaheta, RA; Haferkamp, A; Hudak, L; Juengel, E; Makarević, J; Seibel, JM; Tsaur, I; Waaga-Gasser, A; Wedel, S, 2011
)
0.37
" A Bayesian dose-escalation model was used to determine the feasible doses of daily or weekly everolimus combined with pemetrexed (500 mg/m q3w)."( Everolimus in combination with pemetrexed in patients with advanced non-small cell lung cancer previously treated with chemotherapy: a phase I study using a novel, adaptive Bayesian dose-escalation model.
Anrys, B; Boyer, M; Di Scala, L; Dimitrijevic, S; Laack, E; Miller, N; Petrovic, K; Pylvaenaeinen, I; Solomon, B; Vansteenkiste, J; Wolf, J, 2011
)
2.03
" Primary objective was to determine the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of AEE788 combined with RAD001."( Pharmacokinetic drug interaction between AEE788 and RAD001 causing thrombocytopenia in patients with glioblastoma.
Alfred Yung, WK; Cloughesy, T; Conrad, C; DiLea, C; Dugan, M; Huang, J; Maes, A; Mietlowski, W; Reardon, DA; Rich, J; Yung, L, 2012
)
0.38
"Fifteen patients with metastatic breast cancer were treated with docetaxel (doses of 40-75 mg/m(2) intravenously on day 1 of a 21-day cycle) in combination with everolimus (doses ranging from 20 to 50 mg orally on days 1 and 8 of a 21-day cycle) in a phase 1 trial using the continuous reassessment method to determine maximum tolerated dose."( A phase 1 study of weekly everolimus (RAD001) in combination with docetaxel in patients with metastatic breast cancer.
Booser, D; Brewster, A; Cristofanilli, M; Ensor, J; Giordano, SH; Gladish, G; Gonzalez-Angulo, AM; Hortobagyi, GN; Moore, J; Moulder, S; Murray, JL; Rivera, E; Tran, HT, 2012
)
0.88
"Weekly everolimus in combination with docetaxel every 3 weeks was associated with excessive neutropenia and variable clearance of both drugs, making combination therapy unpredictable, even at low doses of both drugs."( A phase 1 study of weekly everolimus (RAD001) in combination with docetaxel in patients with metastatic breast cancer.
Booser, D; Brewster, A; Cristofanilli, M; Ensor, J; Giordano, SH; Gladish, G; Gonzalez-Angulo, AM; Hortobagyi, GN; Moore, J; Moulder, S; Murray, JL; Rivera, E; Tran, HT, 2012
)
1.13
" In this study, we investigated the therapeutic efficacy of the tumor-VDA ASA404 (DMXAA, 5,6-dimethylxanthenone-4-acetic acid, or vadimezan) in combination with the mTOR inhibitor everolimus (RAD001) against RCC."( Vascular disruption in combination with mTOR inhibition in renal cell carcinoma.
Azabdaftari, G; Ellis, L; Hammers, H; Lehet, K; Pili, R; Seshadri, M; Shah, P; Sotomayor, P, 2012
)
0.57
"25 mg RAD001 twice daily in combination with 15 mg/kg cyclosporin A (CsA) twice daily were identified as appropriate starting doses to achieve the targeted range of RAD001 (3-8 μg/L) when orally administered."( Everolimus in combination with cyclosporin a as pre- and posttransplantation immunosuppressive therapy in nonmyeloablative allogeneic hematopoietic stem cell transplantation.
Altmann, S; Drewelow, B; Freund, M; Junghanss, C; Lange, S; Mueller, S; Rathsack, S; Vogel, H; Wacke, R; Weirich, V, 2012
)
1.82
"Treatment with EVR or SRL alone did not cause severe pancreatic dysfunction and renal injury, but when combined with CsA they aggravated CsA-induced pancreatic and renal injury."( Drug interaction between cyclosporine and mTOR inhibitors in experimental model of chronic cyclosporine nephrotoxicity and pancreatic islet dysfunction.
Bae, SK; Choi, BS; Chung, BH; Lim, SW; Piao, SG; Song, JH; Yang, CW, 2012
)
0.38
" This finding provides better understanding of the difference between EVR and SRL when combined with CsA treatment."( Drug interaction between cyclosporine and mTOR inhibitors in experimental model of chronic cyclosporine nephrotoxicity and pancreatic islet dysfunction.
Bae, SK; Choi, BS; Chung, BH; Lim, SW; Piao, SG; Song, JH; Yang, CW, 2012
)
0.38
"This study evaluated the effects of a mammalian target of mTOR inhibitor everolimus alone or in combination with trastuzumab on stem cells from HER2-overexpressing primary breast cancer cells and the BT474 breast cancer cell line in vitro and in vivo."( Antitumor effect of the mTOR inhibitor everolimus in combination with trastuzumab on human breast cancer stem cells in vitro and in vivo.
Liu, J; Liu, Y; Ma, Y; Zhang, J; Zhang, S; Zhang, X; Zhu, Y, 2012
)
0.88
" This analysis evaluated efficacy and safety of everolimus in combination with tamoxifen in patients with mBC resistant to aromatase inhibitors (AIs)."( Randomized phase II trial of everolimus in combination with tamoxifen in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer with prior exposure to aromatase inhibitors: a GINECO study.
Abadie-Lacourtoisie, S; Allouache, D; Bachelot, T; Bourgier, C; Cropet, C; Debled, M; El Kouri, C; Eymard, JC; Ferrero, JM; Freyer, G; Legouffe, E; Pujade-Lauraine, E; Ray-Coquard, I; Spaëth, D, 2012
)
0.93
"The homogeneous stent strut coverage and the low LLL in the MV reflect proper healing characteristics of the TRYTON Stent in combination with the XIENCE-V™ stent."( Healing responses after bifurcation stenting with the dedicated TRYTON Side-Branch Stent™ in combination with XIENCE-V™ stents: a clinical, angiography, fractional flow reserve, and optical coherence tomography study: the PYTON (Prospective evaluation of
Adriaenssens, T; Bennett, J; Coosemans, M; D'hooge, J; Desmet, W; Dubois, C; Ferdinande, B; Sinnaeve, P; Ughi, G; Wiyono, S, 2013
)
0.39
"BC cell lines expressing aromatase (AROM) and modeling endocrine-sensitive (MCF7-AROM1) and human epidermal growth factor receptor 2 (HER2)-dependent de novo resistant disease (BT474-AROM3) and long-term estrogen-deprived (LTED) MCF7 cells that had acquired resistance associated with HER2 overexpression were treated in vitro and as subcutaneous xenografts with everolimus (RAD001-mTORC1 inhibitor), in combination with tamoxifen or letrozole."( Effectiveness and molecular interactions of the clinically active mTORC1 inhibitor everolimus in combination with tamoxifen or letrozole in vitro and in vivo.
A'Hern, R; Dowsett, M; Evans, DB; Farmer, I; Ghazoui, Z; Guest, S; Johnston, SR; Lane, HA; Martin, LA; Pancholi, S; Ribas, R; Thornhill, AM; Weigel, MT, 2012
)
0.77
" We compared the dose-dependent effects of the structurally related everolimus (EVL) and sirolimus (SRL) alone, and in combination with cyclosporine (CsA), on the rat kidney."( Everolimus and sirolimus in combination with cyclosporine have different effects on renal metabolism in the rat.
Arbelaez, MF; Bohra, R; Brunner, N; Christians, U; Klawitter, J; Lawrence, R; Schmitz, V; Schniedewind, B; Schöning, W; Shokati, T, 2012
)
2.06
" We performed an open label phase II study of everolimus, an inhibitor of mammalian target of rapamycin, in combination with rituximab to examine efficacy and tolerability in patients with relapsed/refractory diffuse large B-cell lymphoma."( Everolimus in combination with rituximab induces complete responses in heavily pretreated diffuse large B-cell lymphoma.
Abramson, JS; Armand, P; Barnes, JA; Feng, Y; Fisher, DC; Freedman, A; Hochberg, EP; Jacobsen, E; Joyce, R; LaCasce, AS; Neuberg, D; Rodig, SJ; Sohani, AR, 2013
)
2.09
" The aim of the study was to characterize the effects in vitro of mTOR inhibitors, used alone and in combination with JAK2 inhibitors, against MPN cells."( mTOR inhibitors alone and in combination with JAK2 inhibitors effectively inhibit cells of myeloproliferative neoplasms.
Bartalucci, N; Bogani, C; Bosi, A; Guglielmelli, P; Martinelli, S; Tozzi, L; Vannucchi, AM, 2013
)
0.39
"We conducted a phase II study of docetaxel in combination with everolimus, a mammalian target of rapamycin (mTOR) inhibitor, for salvage therapy of advanced non-small-cell lung cancer (NSCLC) based on promising preclinical and early-phase clinical data."( Phase II study of docetaxel in combination with everolimus for second- or third-line therapy of advanced non-small-cell lung cancer.
Behera, M; Chen, Z; Fu, H; Gal, AA; Govindan, R; Harvey, RD; Khuri, FR; Klass, CM; Kono, SA; Owonikoko, TK; Ramalingam, SS; Saba, NF; Shin, DM; Sica, G; Subramanian, J; Sun, SY, 2013
)
0.89
" The anti-proliferative activity of CDDP used in combination with RAD001 was statistically significant (P<0."( Everolimus combined with cisplatin has a potential role in treatment of urothelial bladder cancer.
Arantes-Rodrigues, R; Colaço, A; Colaço, B; Costa, C; da Silva, VM; Lopes, C; Oliveira, P; Palmeira, C; Pinto-Leite, R; Santos, L, 2013
)
1.83
"Patients were treated with everolimus 10 mg daily in combination with sorafenib (dose level 1: 200 mg twice daily; dose level 2: 200 mg per morning, 400 mg per evening) using standard phase I dose escalation design."( Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors.
Chan, JA; Jackson, N; Kulke, MH; Malinowski, P; Mayer, RJ; Regan, E, 2013
)
0.94
" Sorafenib 200 mg twice daily in combination with everolimus 10 mg daily was established as the MTD."( Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors.
Chan, JA; Jackson, N; Kulke, MH; Malinowski, P; Mayer, RJ; Regan, E, 2013
)
0.9
"Oral everolimus (Afinitor(®)) in combination with exemestane is indicated for the treatment of hormone receptor-positive, human epidermal growth factor receptor (HER) 2-negative advanced breast cancer in postmenopausal women after failure of treatment with letrozole or anastrozole (in the USA) or after recurrence of progression following a nonsteroidal aromatase inhibitor (AI) in women without symptomatic visceral disease (in the EU)."( Everolimus in combination with exemestane: a review of its use in the treatment of patients with postmenopausal hormone receptor-positive, HER2-negative advanced breast cancer.
Dhillon, S, 2013
)
2.35
"Daily oral everolimus (10 mg) combined with both concurrent radiation and temozolomide followed by adjuvant temozolomide is well tolerated, with an acceptable toxicity profile."( RTOG 0913: a phase 1 study of daily everolimus (RAD001) in combination with radiation therapy and temozolomide in patients with newly diagnosed glioblastoma.
Chinnaiyan, P; Corn, BW; Dipetrillo, TA; Mehta, MP; Rojiani, AM; Wen, PY; Wendland, M; Won, M, 2013
)
1.05
"To evaluate everolimus drug-drug and drug-food interactions, with an emphasis on patients with cancer."( Drug interactions and the pharmacist: focus on everolimus.
Grabowsky, JA,
)
0.77
" Everolimus, an oral mammalian target of rapamycin (mTOR) inhibitor, is indicated for the treatment in adults with progressive neuroendocrine tumors of pancreatic origin that are unresectable, locally advanced, or metastatic; adults with advanced renal cell carcinoma after failure of treatment with sunitinib or sorafenib; and, recently, postmenopausal women with advanced hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer in combination with exemestane after failure of treatment with letrozole or anastrozole."( Drug interactions and the pharmacist: focus on everolimus.
Grabowsky, JA,
)
1.3
" This phase I study was designed to determine the maximum tolerated dose (MTD) of everolimus given with sorafenib 400mg twice daily in patients with advanced HCC of Child-Pugh class A liver function who were naive to systemic therapy."( Phase I study investigating everolimus combined with sorafenib in patients with advanced hepatocellular carcinoma.
Brandt, U; Bruix, J; Chen, LT; Finn, RS; Gomez-Martin, C; Kang, YK; Kim, TY; Klümpen, HJ; Kunz, T; Paquet, T; Poon, RT; Rodriguez-Lope, C; Sellami, D; Yau, T, 2013
)
0.91
"This study evaluated the effects of an mTOR inhibitor everolimus alone or in combination with letrozole on MCF-7/Aro (MCF-7 cells stably transfected with CYP19) in vitro and in vivo."( Everolimus in combination with letrozole inhibit human breast cancer MCF-7/Aro stem cells via PI3K/mTOR pathway: an experimental study.
Hou, G; Liu, J; Liu, Y; Zhang, J; Zhang, S; Zhang, X, 2014
)
2.09
" In this phase 2 multicenter study, adult patients with HER2-positive advanced breast cancer resistant to trastuzumab and pretreated with a taxane received everolimus 10 mg/day in combination with paclitaxel (80 mg/m(2) days 1, 8, and 15 every 4 weeks) and trastuzumab (4 mg/kg loading dose followed by 2 mg/kg weekly), administered in 28-day cycles."( A phase 2 study of everolimus combined with trastuzumab and paclitaxel in patients with HER2-overexpressing advanced breast cancer that progressed during prior trastuzumab and taxane therapy.
André, F; Campone, M; Dalenc, F; El-Hashimy, M; Gligorov, J; Hurvitz, SA; Jhangiani, H; Lincy, J; Llombart, A; Miao, S; Mirshahidi, HR; O'Regan, RM; Sahmoud, T; Soria, JC; Tan-Chiu, E; Taran, T; Tjan-Heijnen, VC, 2013
)
0.92
" The recommended phase 2 dose of oral weekly everolimus is 70 mg in combination with standard cetuximab."( Phase 1 and pharmacodynamic trial of everolimus in combination with cetuximab in patients with advanced cancer.
Avadhani, AN; Ciunci, CA; Divgi, CR; Englander, S; Flaherty, KT; Giantonio, BJ; Harlacker, K; Kang, HC; O'Dwyer, PJ; Perini, RF; Redlinger, M; Rosen, MA; Schnall, M; Song, HK; Sun, W; Troxel, A, 2014
)
0.94
"Lenvatinib 18 mg combined with everolimus 5 mg was associated with manageable toxicity consistent with individual agents and no new safety signals."( A phase 1b clinical trial of the multi-targeted tyrosine kinase inhibitor lenvatinib (E7080) in combination with everolimus for treatment of metastatic renal cell carcinoma (RCC).
Carter, D; Gold, AM; Hutson, TE; Larkin, J; Michaelson, MD; Molina, AM; Motzer, R; Wood, K, 2014
)
0.9
"This phase Ib study aimed to establish the feasible everolimus dose given with standard-dose etoposide plus cisplatin (EP) for extensive-stage small-cell lung cancer (SCLC)."( A phase Ib dose-escalation study of everolimus combined with cisplatin and etoposide as first-line therapy in patients with extensive-stage small-cell lung cancer.
Beck, JT; Besse, B; Camidge, DR; Dimitrijevic, S; Heist, RS; Johnson, BE; Miller, N; Mulatero, C; Papadmitrakopoulou, VA; Petrovic, K; Pylvaenaeinen, I; Schmid, P; Urva, S, 2014
)
0.93
"An adaptive Bayesian dose-escalation model and investigator opinion were used to identify feasible daily or weekly everolimus doses given with EP in adults with treatment-naive extensive-stage SCLC."( A phase Ib dose-escalation study of everolimus combined with cisplatin and etoposide as first-line therapy in patients with extensive-stage small-cell lung cancer.
Beck, JT; Besse, B; Camidge, DR; Dimitrijevic, S; Heist, RS; Johnson, BE; Miller, N; Mulatero, C; Papadmitrakopoulou, VA; Petrovic, K; Pylvaenaeinen, I; Schmid, P; Urva, S, 2014
)
0.89
"5 mg/day plus G-CSF was the only feasible dose given with standard-dose EP in untreated extensive-stage SCLC."( A phase Ib dose-escalation study of everolimus combined with cisplatin and etoposide as first-line therapy in patients with extensive-stage small-cell lung cancer.
Beck, JT; Besse, B; Camidge, DR; Dimitrijevic, S; Heist, RS; Johnson, BE; Miller, N; Mulatero, C; Papadmitrakopoulou, VA; Petrovic, K; Pylvaenaeinen, I; Schmid, P; Urva, S, 2014
)
0.68
" Secondary objectives included evaluation of safety, toxicity and pharmacokinetics of LY2584702 in combination with erlotinib or everolimus."( A phase Ib trial of LY2584702 tosylate, a p70 S6 inhibitor, in combination with erlotinib or everolimus in patients with solid tumours.
Benhadji, KA; Brail, LH; Bumgardner, W; Cohen, EE; Hollebecque, A; Houédé, N; Italiano, A; Massard, C; Miller, J; Soria, JC; Westwood, P, 2014
)
0.83
"Patients with advanced solid tumours were treated with a total daily dose of 50-200mg of LY2584702 in combination with erlotinib 150 mg once daily (Arm A) or everolimus 10mg once daily (Arm B)."( A phase Ib trial of LY2584702 tosylate, a p70 S6 inhibitor, in combination with erlotinib or everolimus in patients with solid tumours.
Benhadji, KA; Brail, LH; Bumgardner, W; Cohen, EE; Hollebecque, A; Houédé, N; Italiano, A; Massard, C; Miller, J; Soria, JC; Westwood, P, 2014
)
0.82
" Exposure of erlotinib increased when administered in combination with LY2584702."( A phase Ib trial of LY2584702 tosylate, a p70 S6 inhibitor, in combination with erlotinib or everolimus in patients with solid tumours.
Benhadji, KA; Brail, LH; Bumgardner, W; Cohen, EE; Hollebecque, A; Houédé, N; Italiano, A; Massard, C; Miller, J; Soria, JC; Westwood, P, 2014
)
0.62
"LY2584702 was not well tolerated when administered with erlotinib, therefore this combination is not feasible."( A phase Ib trial of LY2584702 tosylate, a p70 S6 inhibitor, in combination with erlotinib or everolimus in patients with solid tumours.
Benhadji, KA; Brail, LH; Bumgardner, W; Cohen, EE; Hollebecque, A; Houédé, N; Italiano, A; Massard, C; Miller, J; Soria, JC; Westwood, P, 2014
)
0.62
" We evaluated the optimal schedule of oral topotecan in combination with everolimus in patients with endometrial cancer."( Phase I clinical trial of the mammalian target of rapamycin inhibitor everolimus in combination with oral topotecan for recurrent and advanced endometrial cancer.
Abu-Khalaf, MM; Acevedo-Gadea, C; Azodi, M; Higgins, SA; Ratner, E; Rutherford, T; Santin, AD; Schwartz, PE; Silasi, DA; Urva, S, 2014
)
0.87
"9 mg/m on days 1 to 5 in combination with oral everolimus 5 mg every other day."( Phase I clinical trial of the mammalian target of rapamycin inhibitor everolimus in combination with oral topotecan for recurrent and advanced endometrial cancer.
Abu-Khalaf, MM; Acevedo-Gadea, C; Azodi, M; Higgins, SA; Ratner, E; Rutherford, T; Santin, AD; Schwartz, PE; Silasi, DA; Urva, S, 2014
)
0.89
" The activity and safety of everolimus was assessed in combination with octreotide long-acting repeatable (LAR) in patients with neuroendocrine tumors (NETs) of gastroenteropancreatic and lung origin."( Everolimus in combination with octreotide long-acting repeatable in a first-line setting for patients with neuroendocrine tumors: an ITMO group study.
Aieta, M; Bajetta, E; Bianco, N; Biondani, P; Blanco, G; Catena, L; Fazio, N; Mauri, CM; Pucci, F; Pusceddu, S; Ricci, S; Spada, F; Valente, M, 2014
)
2.14
" Treatment-naive patients with advanced well-differentiated NETs of gastroenteropancreatic tract and lung origin received everolimus 10 mg daily, in combination with octreotide LAR 30 mg every 28 days."( Everolimus in combination with octreotide long-acting repeatable in a first-line setting for patients with neuroendocrine tumors: an ITMO group study.
Aieta, M; Bajetta, E; Bianco, N; Biondani, P; Blanco, G; Catena, L; Fazio, N; Mauri, CM; Pucci, F; Pusceddu, S; Ricci, S; Spada, F; Valente, M, 2014
)
2.05
"This phase I study investigated the safety, dose-limiting toxicity, and efficacy in three cohorts all treated with the mTOR inhibitor everolimus that was delivered (1) in combination with 5-fluorouracil with leucovorin (5-FU/LV), (2) with mFOLFOX6 (5-FU/LV + oxaliplatin), and (3) with mFOLFOX6 + panitumumab in patients with refractory solid tumors."( A phase I trial of everolimus in combination with 5-FU/LV, mFOLFOX6 and mFOLFOX6 plus panitumumab in patients with refractory solid tumors.
Bernard, S; Davies, JM; Dees, EC; Goldberg, RM; Ivanova, A; Keller, K; McRee, AJ; O'Neil, BH; Sanoff, HG, 2014
)
0.93
" Next, a case study is used to illustrate this NMA feasibility assessment process in order to compare everolimus in combination with hormonal therapy to alternative chemotherapies in terms of progression-free survival for women with advanced breast cancer."( A process for assessing the feasibility of a network meta-analysis: a case study of everolimus in combination with hormonal therapy versus chemotherapy for advanced breast cancer.
Cope, S; Jansen, JP; Saletan, S; Schmid, P; Smiechowski, B; Zhang, J, 2014
)
0.84
" Several studies have already used RAD001 in combination with calcineurin inhibitors after hematopoietic stem cell transplantation (HSCT)."( Everolimus in combination with mycophenolate mofetil as pre- and post-transplantation immunosuppression after nonmyeloablative hematopoietic stem cell transplantation in canine littermates.
Junghanss, C; Killian, D; Knuebel, G; Knueppel, A; Lange, S; Lindner, I; Machka, C; Murua Escobar, H; Roolf, C; Vogel, H; Wacke, R; Werner, J, 2014
)
1.85
"Currently, there are no published data on pharmacokinetics (PK) of everolimus in combination with cyclosporine in Japanese renal transplant patients."( Pharmacokinetics of everolimus when combined with cyclosporine in Japanese de novo renal transplant recipients.
Goto, N; Hirano, M; Hoshinaga, K; Kusaka, M; Sasaki, H; Uchida, K; Watarai, Y, 2014
)
0.96
"Platinum-based therapy combined with cetuximab is standard first-line therapy for recurrent or metastatic squamous cell carcinoma of the head and neck (RMSCCHN)."( Phase 1 and pharmacokinetic study of everolimus in combination with cetuximab and carboplatin for recurrent/metastatic squamous cell carcinoma of the head and neck.
Beitler, JJ; Chen, AY; Chen, Z; El-Deiry, M; Harvey, D; Higgins, K; Hurwitz, SJ; Khuri, FR; Kim, S; Kono, SA; Lewis, C; Magliocca, K; Mendel, J; Owonikoko, TK; Ramalingam, SS; Rogerio, J; Saba, NF; Shin, DM; Sun, SY; Wadsworth, T, 2014
)
0.68
" By combination with the mTOR inhibitor RAD001, a synergistic effect on cell proliferation was observed in these cell lines."( The multi-tyrosine kinase inhibitor TKI258, alone or in combination with RAD001, is effective for treatment of human leukemia with BCR-ABL translocation in vitro.
Eucker, J; Habbel, P; Li, X; Liu, H; Neumann, C; Scholz, C; Zang, C; Zhou, Y, 2014
)
0.4
"TKI258 may become a potent therapeutic agent, either alone or in combination with RAD001, for treatment of BCR-ABL(+) leukemia."( The multi-tyrosine kinase inhibitor TKI258, alone or in combination with RAD001, is effective for treatment of human leukemia with BCR-ABL translocation in vitro.
Eucker, J; Habbel, P; Li, X; Liu, H; Neumann, C; Scholz, C; Zang, C; Zhou, Y, 2014
)
0.4
"This phase Ib trial investigated the safety, tolerability, and recommended phase II dose and schedule of the MEK inhibitor trametinib in combination with the mammalian target of rapamycin (mTOR) inhibitor everolimus."( A phase IB trial of the oral MEK inhibitor trametinib (GSK1120212) in combination with everolimus in patients with advanced solid tumors.
Bellew, KM; Bendell, JC; Burris, HA; Cox, DS; Durante, M; Infante, JR; Jones, SF; Le, NT; Papadopoulos, KP; Park, J; Patnaik, A; Rasco, D; Tolcher, AW, 2015
)
0.83
" PK assessment did not suggest drug-drug interactions between these two agents."( A phase IB trial of the oral MEK inhibitor trametinib (GSK1120212) in combination with everolimus in patients with advanced solid tumors.
Bellew, KM; Bendell, JC; Burris, HA; Cox, DS; Durante, M; Infante, JR; Jones, SF; Le, NT; Papadopoulos, KP; Park, J; Patnaik, A; Rasco, D; Tolcher, AW, 2015
)
0.64
"This study was unable to identify a recommended phase II dose and schedule of trametinib in combination with everolimus that provided an acceptable tolerability and adequate drug exposure."( A phase IB trial of the oral MEK inhibitor trametinib (GSK1120212) in combination with everolimus in patients with advanced solid tumors.
Bellew, KM; Bendell, JC; Burris, HA; Cox, DS; Durante, M; Infante, JR; Jones, SF; Le, NT; Papadopoulos, KP; Park, J; Patnaik, A; Rasco, D; Tolcher, AW, 2015
)
0.85
"Azole antifungals, prescribed prophylactically to avoid severe infections in immunosuppressed organ transplant recipients, can interact with drug substrates of CYP3A4."( Worsening pneumonitis due to a pharmacokinetic drug-drug interaction between everolimus and voriconazole in a renal transplant patient.
Amilien, V; Chauvin, B; Eloy, P; Francois, H; Furlan, V; Lecefel, C; Massias, L; Taburet, AM; Wyplosz, B, 2015
)
0.65
" The current study will test the hypothesis that MSC treatment, in combination with the mTOR inhibitor everolimus, facilitates tacrolimus withdrawal, reduces fibrosis and decreases the incidence of opportunistic infections compared to standard tacrolimus dose."( Autologous bone marrow derived mesenchymal stromal cell therapy in combination with everolimus to preserve renal structure and function in renal transplant recipients.
Al Huurman, V; Bank, JR; Claas, FH; de Fijter, JW; Dreyer, GJ; Fibbe, WE; Heidt, S; Lindeman, J; Rabelink, TJ; Reinders, ME; Roelen, DL; Roelofs, H; van Kooten, C, 2014
)
0.84
" Patients in the MSC treated group will receive two doses of autologous bone marrow derived MSCs IV (target 1,5 x 10(6), Range 1-2 x 10(6) million MSCs per/kg body weight), 7 days apart, 6 and 7 weeks transplantation in combination with everolimus and prednisolone."( Autologous bone marrow derived mesenchymal stromal cell therapy in combination with everolimus to preserve renal structure and function in renal transplant recipients.
Al Huurman, V; Bank, JR; Claas, FH; de Fijter, JW; Dreyer, GJ; Fibbe, WE; Heidt, S; Lindeman, J; Rabelink, TJ; Reinders, ME; Roelen, DL; Roelofs, H; van Kooten, C, 2014
)
0.81
"This study will provide information whether MSCs in combination with everolimus can be used for tacrolimus withdrawal, and whether this strategy leads to preservation of renal structure and function in renal recipients."( Autologous bone marrow derived mesenchymal stromal cell therapy in combination with everolimus to preserve renal structure and function in renal transplant recipients.
Al Huurman, V; Bank, JR; Claas, FH; de Fijter, JW; Dreyer, GJ; Fibbe, WE; Heidt, S; Lindeman, J; Rabelink, TJ; Reinders, ME; Roelen, DL; Roelofs, H; van Kooten, C, 2014
)
0.86
" Everolimus, an mTOR inhibitor, has shown antitumor benefit in pNETs alone and in combination with octreotide LAR in RADIANT-1 and RADIANT-3 studies."( Rationale and protocol of the MetNET-1 trial, a prospective, single center, phase II study to evaluate the activity and safety of everolimus in combination with octreotide LAR and metformin in patients with advanced pancreatic neuroendocrine tumors.
Bregant, C; Buzzoni, R; Concas, L; de Braud, F; Formisano, B; Leuzzi, L; Pusceddu, S,
)
1.25
"This phase I study evaluated the safety and efficacy of the oral mTOR inhibitor everolimus in combination with thoracic radiotherapy followed by consolidation chemotherapy in locally advanced or oligometastatic untreated non-small-cell lung cancer (NSCLC)."( Phase I trial of everolimus in combination with thoracic radiotherapy in non-small-cell lung cancer.
Angevin, E; Angokai, M; Bahleda, R; Besse, B; Deandreis, D; Deutsch, E; Faivre, L; Hennequin, C; Le Péchoux, C; Levy, A; Pignon, JP; Rivera, S; Soria, JC; Tao, Y, 2015
)
0.98
"In previously untreated and unselected NSCLC patients, the recommended phase II dose of everolimus in combination with thoracic radiotherapy is 50 mg/week."( Phase I trial of everolimus in combination with thoracic radiotherapy in non-small-cell lung cancer.
Angevin, E; Angokai, M; Bahleda, R; Besse, B; Deandreis, D; Deutsch, E; Faivre, L; Hennequin, C; Le Péchoux, C; Levy, A; Pignon, JP; Rivera, S; Soria, JC; Tao, Y, 2015
)
0.98
" We report the first bench-to-bedside evidence that RET inhibitor combined with an mTOR inhibitor is active against brain-metastatic RET-rearranged lung cancer and the first evidence of blood-brain barrier penetration."( Systemic and CNS activity of the RET inhibitor vandetanib combined with the mTOR inhibitor everolimus in KIF5B-RET re-arranged non-small cell lung cancer with brain metastases.
Ali, SM; Berry, J; Cascone, T; Guha-Thakurta, N; Heymach, JV; McMahon, C; Miller, V; Pai, S; Roxas, M; Subbiah, IM; Subbiah, V; Tang, Z, 2015
)
0.64
"A number of studies have provided information regarding the risks and benefits of mammalian target of rapamycin inhibitors (mTOR-I) combined with calcineurin inhibitors (CNI) versus mycophenolic acid (MPA)."( mTOR inhibitor versus mycophenolic acid as the primary immunosuppression regime combined with calcineurin inhibitor for kidney transplant recipients: a meta-analysis.
Chen, J; Jiang, Y; Lai, X; Shou, Z; Xiang, S; Xie, X, 2015
)
0.42
" Randomized controlled trials comparing mTOR-I to MPA as the primary immunosuppressive regimen in combination with CNI were selected and meta-analyzed."( mTOR inhibitor versus mycophenolic acid as the primary immunosuppression regime combined with calcineurin inhibitor for kidney transplant recipients: a meta-analysis.
Chen, J; Jiang, Y; Lai, X; Shou, Z; Xiang, S; Xie, X, 2015
)
0.42
" Notably, mTOR-I had an increased risk of graft loss when combined with CNI, even when combined with a reduced dose of CNI."( mTOR inhibitor versus mycophenolic acid as the primary immunosuppression regime combined with calcineurin inhibitor for kidney transplant recipients: a meta-analysis.
Chen, J; Jiang, Y; Lai, X; Shou, Z; Xiang, S; Xie, X, 2015
)
0.42
" We conducted a pilot trial to assess the pharmacokinetics (PK), tolerability, and efficacy of JI-101 in combination with everolimus in advanced cancers, and pharmacodynamics (PD), tolerability, and efficacy of JI-101 in ovarian cancer."( A pilot study of JI-101, an inhibitor of VEGFR-2, PDGFR-β, and EphB4 receptors, in combination with everolimus and as a single agent in an ovarian cancer expansion cohort.
Agarwal, N; Boucher, K; Luebke, A; Patel, J; Sharma, S; Wade, ML; Werner, TL, 2015
)
0.84
"To explore the effect of CQ alone or in combination with RAD001, Torin1 or NVP-BEZ235 on autophagy and on NET cell viability, proliferation and apoptosis."( Abrogation of Autophagy by Chloroquine Alone or in Combination with mTOR Inhibitors Induces Apoptosis in Neuroendocrine Tumor Cells.
Avniel-Polak, S; Glaser, B; Gross, DJ; Grozinsky-Glasberg, S; Leibowitz, G; Riahi, Y, 2016
)
0.43
"RAD001 combined with ATRA significantly induced NB4-R1 cells differentiation, but RAD001 or ATRA alone did not enhance NB4-R1 differentiation."( [Everolimus combined with all-trans retinoid acid reverses drug resistance in acute promyelocytic leukemia NB4-R1 cells].
He, Y; Huang, H; Liao, WC; Wang, BS, 2015
)
1.33
"RAD001 combined with ATRA can induce cell differentiation, inhibit cell cycle, resulting the reverse of drug resistance in NB4-R1 cells, which is associated with increase of autophagy level and degradation of PML-RARα."( [Everolimus combined with all-trans retinoid acid reverses drug resistance in acute promyelocytic leukemia NB4-R1 cells].
He, Y; Huang, H; Liao, WC; Wang, BS, 2015
)
1.33
"44 g/day or mycophenolate mofetil at 2 g/day) with tacrolimus; in combination with corticosteroids."( Design and rationale of the ATHENA study--A 12-month, multicentre, prospective study evaluating the outcomes of a de novo everolimus-based regimen in combination with reduced cyclosporine or tacrolimus versus a standard regimen in kidney transplant patien
Dragun, D; Hauser, IA; Nashan, B; Schenker, P; Sommerer, C; Suwelack, B; Thaiss, F, 2016
)
0.64
"The VERITAS (A Phase 1B open-label study to assess the safety and tolerability of everolimus in combination with eribulin in triple-negative breast cancers) trial (EudraCT number: 2014-000135-17) is a phase Ib, open label, multicenter, dose-escalation, safety, pharmacokinetic, and pharmacodynamic study based on the combination of everolimus with eribulin in sequential cohorts of metastatic triple negative breast cancer (TNBC) patients."( A Phase Ib Open-Label Study to Assess the Safety and Tolerability of Everolimus in Combination With Eribulin in Triple-Negative Breast Cancers.
Bottini, A; Cappelletti, MR; Generali, D; Gobbi, A; Milani, M; Roviello, G; Senti, C; Sigala, S; Strina, C; Zanotti, L, 2016
)
0.89
"The primary objective of the study is to identify the recommended dose of everolimus in combination with eribulin."( A Phase Ib Open-Label Study to Assess the Safety and Tolerability of Everolimus in Combination With Eribulin in Triple-Negative Breast Cancers.
Bottini, A; Cappelletti, MR; Generali, D; Gobbi, A; Milani, M; Roviello, G; Senti, C; Sigala, S; Strina, C; Zanotti, L, 2016
)
0.9
"The VERITAS trial is expected to determine the recommended dose of everolimus in combination with eribulin in TBNC."( A Phase Ib Open-Label Study to Assess the Safety and Tolerability of Everolimus in Combination With Eribulin in Triple-Negative Breast Cancers.
Bottini, A; Cappelletti, MR; Generali, D; Gobbi, A; Milani, M; Roviello, G; Senti, C; Sigala, S; Strina, C; Zanotti, L, 2016
)
0.91
" The primary endpoint of the phase 1 portion of this study was to establish the maximum tolerated dose of everolimus that could be combined with R-CHOP-21."( Everolimus combined with R-CHOP-21 for new, untreated, diffuse large B-cell lymphoma (NCCTG 1085 [Alliance]): safety and efficacy results of a phase 1 and feasibility trial.
Ansell, SM; Colgan, JP; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, B; McPhail, E; Micallef, IN; Nowakowski, GS; Witzig, TE, 2016
)
2.09
"The mTORC1 inhibitor everolimus given for 14 days in combination with R-CHOP-21 for patients with DLBCL is safe."( Everolimus combined with R-CHOP-21 for new, untreated, diffuse large B-cell lymphoma (NCCTG 1085 [Alliance]): safety and efficacy results of a phase 1 and feasibility trial.
Ansell, SM; Colgan, JP; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, B; McPhail, E; Micallef, IN; Nowakowski, GS; Witzig, TE, 2016
)
2.2
" We investigated the pharmacologic characteristics of OTX015 as a single agent and combined with targeted therapy or conventional chemotherapies in glioblastoma cell lines."( OTX015 (MK-8628), a novel BET inhibitor, displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma models.
Astorgues-Xerri, L; Bekradda, M; Berenguer-Daizé, C; Cayol, M; Cvitkovic, E; Lokiec, F; MacKenzie, S; Noel, K; Odore, E; Ouafik, L; Rezai, K; Riveiro, ME, 2016
)
0.43
" Here, we report in vivo data for OTX015 in combination with the histone deacetylase inhibitor vorinostat, the Bruton's tyrosine kinase inhibitor ibrutinib, the anti-CD20 monoclonal antibody rituximab, and the mTOR inhibitor everolimus in a diffuse large B cell lymphoma model."( Bromodomain inhibitor OTX015 (MK-8628) combined with targeted agents shows strong in vivo antitumor activity in lymphoma.
Bernasconi, E; Bertoni, F; Cascione, L; Cvitkovic, E; Gaudio, E; Odore, E; Ponzoni, M; Rezai, K; Rinaldi, A; Riveiro, E; Stathis, A; Tarantelli, C; Zucca, E, 2016
)
0.62
" Some clinically relevant drug-drug interactions are highlighted."( mTOR inhibitors in pancreas transplant: adverse effects and drug-drug interactions.
Fernandes-Silva, G; Ivani de Paula, M; Rangel, ÉB, 2017
)
0.46
" In both animal models the anti-lymphoma activity of selinexor is enhanced through combination with DEX or EVER."( Anti-tumor activity of selective inhibitor of nuclear export (SINE) compounds, is enhanced in non-Hodgkin lymphoma through combination with mTOR inhibitor and dexamethasone.
Aboukameel, A; Azmi, AS; Baloglu, E; Bhutani, D; Carlson, R; Elloul, S; Kauffman, M; Mohammad, RM; Muqbil, I; Senapedis, W; Shacham, S; Zonder, J, 2016
)
0.43
"To assess safety and efficacy of mammalian target of rapamycin (mTOR) inhibition in combination with liposomal doxorubicin and bevacizumab in patients with advanced metaplastic TNBC."( Targeting the PI3K/AKT/mTOR Pathway for the Treatment of Mesenchymal Triple-Negative Breast Cancer: Evidence From a Phase 1 Trial of mTOR Inhibition in Combination With Liposomal Doxorubicin and Bevacizumab.
Albarracin, C; Basho, RK; Chavez-MacGregor, M; Gilcrease, M; Helgason, T; Herbrich, SM; Hess, KR; Hong, D; Ibrahim, NK; Janku, F; Karp, DD; Koenig, KB; Kurzrock, R; Litton, JK; Meric-Bernstam, F; Moulder, SL; Murray, JL; Murthy, RK; Subbiah, V; Valero, V, 2017
)
0.46
" The aim of this study was to evaluate the anti-tumor activity of OTX015 as single agent and in combination with everolimus in TNBC models."( The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with everolimus.
Astorgues-Xerri, L; Bekradda, M; Beltrame, L; Bertoni, F; Cvitkovic, E; D'Incalci, M; Erba, E; Frapolli, R; Kwee, I; Odore, E; Panini, N; Rezai, K; Rinaldi, A; Riveiro, ME; Vázquez, R, 2017
)
0.86
" Future trials examining the use of immunotherapy, both as monotherapy and in combination with VEGF targeted therapy, will likely be a dominant influence in the therapeutic landscape of mRCC."( Lenvatinib for use in combination with everolimus for the treatment of patients with advanced renal cell carcinoma following one prior anti-angiogenic therapy.
Larkin, J; O'Reilly, A, 2017
)
0.72
" The changes in steady-state pharmacokinetics of everolimus with BEZ235 highlight potential drug-drug interactions when these two drugs are administered together."( A Phase Ib Study of the Dual PI3K/mTOR Inhibitor Dactolisib (BEZ235) Combined with Everolimus in Patients with Advanced Solid Malignancies.
Beg, MS; Desai, P; Fathallah, H; Karim, NA; Kozma, SC; Mercer, CA; Moorthy, G; Morris, JC; O'Gara, S; Olowokure, O; Rixe, O; Salkeni, MA; Thomas, G; Thomas, HE; Wise-Draper, TM, 2017
)
0.93
" In this study, we aim at determining the antitumor efficacy of everolimus alone or in combination with the SSAs octreotide and pasireotide in primary cultures of pNETs."( Anti-proliferative and anti-secretory effects of everolimus on human pancreatic neuroendocrine tumors primary cultures: is there any benefit from combination with somatostatin analogs?
Albertelli, M; Barbieri, F; Barlier, A; Brue, T; Delpero, JR; Ferone, D; Florio, T; Garcia, S; Gerard, C; Mohamed, A; Moutardier, V; Niccoli, P; Poizat, F; Roche, C; Romano, D; Saveanu, A, 2017
)
0.95
"Palbociclib is the first cyclin-dependent kinase 4/6 inhibitor approved in the United States for HR+/HER2- advanced/metastatic breast cancer, in combination with letrozole as initial endocrine-based therapy in postmenopausal women or with fulvestrant in women with disease progression following endocrine therapy."( Comparison of palbociclib in combination with letrozole or fulvestrant with endocrine therapies for advanced/metastatic breast cancer: network meta-analysis.
Chirila, C; Colosia, A; Iyer, S; Kaye, JA; Ling, C; Mitra, D; Odom, D, 2017
)
0.46
" Patients undergoing hematopoietic stem cell transplantation (HSCT) are exposed to various types of drugs, and understanding how these drugs interact is of the utmost importance."( Development of an assay for cellular efflux of pharmaceutically active agents and its relevance to understanding drug interactions.
Andersson, BS; Hassan, M; Valdez, BC, 2017
)
0.46
" Aim of this study was to evaluate pasireotide alone and in combination with everolimus in patients with MTC."( The antiproliferative effect of pasireotide LAR alone and in combination with everolimus in patients with medullary thyroid cancer: a single-center, open-label, phase II, proof-of-concept study.
Aria, M; Camera, L; Chiofalo, MG; Colao, A; Del Prete, M; Faggiano, A; Ferolla, P; Fonti, R; Modica, R; Pezzullo, L; Severino, R; Vitale, G, 2018
)
0.94
"To assess the tolerability, safety, pharmacokinetics and antitumor activities of lenvatinib, an oral inhibitor of multiple receptor tyrosine kinases, in combination with everolimus, an inhibitor of mammalian target of rapamycin, in Japanese patients with advanced or metastatic renal cell carcinoma after disease progression with vascular endothelial growth factor-targeted therapy."( Lenvatinib in combination with everolimus in patients with advanced or metastatic renal cell carcinoma: A phase 1 study.
Fujiwara, Y; Ikezawa, H; Matsubara, N; Naito, Y; Nakano, K; Namiki, M; Okude, T; Takahashi, S; Yusa, W, 2018
)
0.96
", as a monotherapy or in combination with the known mechanistic target of rapamycin complex 1 (mTORC1) inhibitor, everolimus, against Caki-1 (Von Hippel-Lindau (VHL)+/+) and 786-O (VHL-/-) human RCC cells."( Novel therapeutic roles of MC-4 in combination with everolimus against advanced renal cell carcinoma by dual targeting of Akt/pyruvate kinase muscle isozyme M2 and mechanistic target of rapamycin complex 1 pathways.
Choi, HY; Chung, KH; De, U; Jeon, Y; Kim, HS; Kwak, JH; Lee, BM; Lee, HW; Lee, SJ; Lim, JE; Park, YJ; Rhyu, IJ; Son, JY; Tae, IH; Yoon, S, 2018
)
0.94
" However, there is limited available data on Dmab toxicity in combination with AA therapies in patients with kidney cancer."( Denosumab Toxicity When Combined With Anti-angiogenic Therapies on Patients With Metastatic Renal Cell Carcinoma: A GETUG Study.
Albiges, L; Barthélémy, P; Bouleftour, W; Chevreau, C; Culine, S; Espenel, S; Fizazi, K; Gravis, G; Guillot, A; Houede, N; Joly, C; Mahammedi, H; Meriaux, E; Negrier, S; Oriol, M; Pouessel, D; Roubaud, G; Tartas, S; Tinquaut, F; Vassal, C, 2019
)
0.51
"The incidence of ONJ was high in this real-life population of patients with mRCC treated with AA therapies combined with Dmab."( Denosumab Toxicity When Combined With Anti-angiogenic Therapies on Patients With Metastatic Renal Cell Carcinoma: A GETUG Study.
Albiges, L; Barthélémy, P; Bouleftour, W; Chevreau, C; Culine, S; Espenel, S; Fizazi, K; Gravis, G; Guillot, A; Houede, N; Joly, C; Mahammedi, H; Meriaux, E; Negrier, S; Oriol, M; Pouessel, D; Roubaud, G; Tartas, S; Tinquaut, F; Vassal, C, 2019
)
0.51
" Current studies determined the OATP1B1/1B3-mediated drug-drug interaction (DDI) potential of mammalian target of rapamycin (mTOR) inhibitors, everolimus and sirolimus, using R-value and physiologically based pharmacokinetic models."( Preincubation With Everolimus and Sirolimus Reduces Organic Anion-Transporting Polypeptide (OATP)1B1- and 1B3-Mediated Transport Independently of mTOR Kinase Inhibition: Implication in Assessing OATP1B1- and OATP1B3-Mediated Drug-Drug Interactions.
Alam, K; Crowe, A; Ding, K; Farasyn, T; Hatley, O; Kanyo, J; Lam, TT; Neuhoff, S; Yue, W, 2019
)
1.04
" The purpose of the VicTORia trial was to show superiority of everolimus in combination with vinorelbine versus vinorelbine monotherapy as second-line chemotherapy for patients with advanced HER2 negative breast cancer."( VicTORia: a randomised phase II study to compare vinorelbine in combination with the mTOR inhibitor everolimus versus vinorelbine monotherapy for second-line chemotherapy in advanced HER2-negative breast cancer.
Decker, T; Hagen, V; Jaehnig, P; Lerchenmüller, C; Marschner, N; Muendlein, A; Potthoff, K; Rauh, J; Schröder, H; Welt, A, 2019
)
0.97
" In MPC/MTT spheroids, treatment with clinically relevant doses of BYL719 alone or in combination with everolimus was highly effective, leading to a significant shrinkage or even a complete collapse of the spheroids."( Synergistic Highly Potent Targeted Drug Combinations in Different Pheochromocytoma Models Including Human Tumor Cultures.
Aristizabal Prada, ET; Auernhammer, CJ; Bechmann, N; Beuschlein, F; Bornstein, SR; Eisenhofer, G; Fankhauser, M; Fassnacht, M; Favier, J; Gieldon, L; Goncalves, J; Grossman, AB; Klink, B; Knösel, T; Lauseker, M; Maurer, J; Nölting, S; Orth, M; Pacak, K; Rank, P; Reincke, M; Rubinstein, G; Spitzweg, C; Spöttl, G; William, D; Ziegler, CG, 2019
)
0.73
"Everolimus (EVR) is often administered with cyclosporine A (CsA), according to an established protocol."( Optimal dose of everolimus administered with tacrolimus in living donor kidney transplantation.
Futamura, K; Goto, N; Hiramitsu, T; Ichimori, T; Narumi, S; Okada, M; Tomosugi, T; Watarai, Y, 2019
)
2.3
"The mammalian target of rapamycin (mTOR) inhibitor, everolimus, in combination with reduced-exposure calcineurin inhibitor (CNI), has been demonstrated in clinical trials to have comparable efficacy in low-to-moderate immunological risk kidney transplant recipients to the Standard of Care, mycophenolic acid (MPA) in combination with standard-exposure CNI."( Evidence-based practice: Guidance for using everolimus in combination with low-exposure calcineurin inhibitors as initial immunosuppression in kidney transplant patients.
Berger, SP; Chadban, SJ; Citterio, F; Eisenberger, U; Henry, ML; Hesselink, DA; Kamar, N; Legendre, C; Oppenheimer, F; Pascual, J; Rigotti, P; Russ, GR; Sommerer, C; Srinivas, TR; Tedesco-Silva, H; Vincenti, F; Watarai, Y, 2019
)
1.03
" We completed a phase I, dose-finding trial for the mTOR inhibitor everolimus combined with the HDAC inhibitor panobinostat in advanced clear cell renal cell carcinoma (ccRCC) patients."( Phase I study of the mTOR inhibitor everolimus in combination with the histone deacetylase inhibitor panobinostat in patients with advanced clear cell renal cell carcinoma.
Adra, N; Chintala, S; Damayanti, N; George, S; Pili, R; Wood, A, 2020
)
1.07
" Results The maximum tolerated doses were determined to be vatalanib 1250 mg once daily or 750 mg twice daily in combination with everolimus 10 mg once daily."( A phase I study of the VEGFR kinase inhibitor vatalanib in combination with the mTOR inhibitor, everolimus, in patients with advanced solid tumors.
Adjei, AA; Croghan, GA; Dy, GK; Glockner, J; Hanson, LJ; Molina, JR; Qi, Y; Roos, MM; Tan, AD; Zhu, M, 2020
)
0.98
"We aimed to evaluate the therapeutic effects of paclitaxel in combination with mTOR inhibitor everolimus on adriamycin-resistant breast cancer cell line MDA-MB-231 (MDA-MB-231/ADR)."( Inhibitory effects of everolimus in combination with paclitaxel on adriamycin-resistant breast cancer cell line MDA-MB-231.
Li, Q; Liu, P; Shi, C; Wang, X; Xu, T, 2020
)
1.09
"To explore the efficacy and safety of everolimus combined with endocrine therapy in patients with hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER-2)-negative advanced breast cancer."( Efficacy of everolimus combined with endocrine therapy in HR-positive/HER-2-negativeadvanced breast cancer.
Chen, C; Kong, F; Lu, J; Shan, H; Song, X; Wang, L; Yuan, H,
)
0.78
" Of them, 54 patients were treated with everolimus combined with endocrine drugs (Everolimus group), while the other 54 patients underwent endocrine monotherapy (Control group)."( Efficacy of everolimus combined with endocrine therapy in HR-positive/HER-2-negativeadvanced breast cancer.
Chen, C; Kong, F; Lu, J; Shan, H; Song, X; Wang, L; Yuan, H,
)
0.78
"Everolimus combined with endocrine therapy has significant clinical efficacy in patients with HR-positive/HER-2-negative advanced breast cancer, and can effectively improve the survival of patients with tolerable adverse reactions."( Efficacy of everolimus combined with endocrine therapy in HR-positive/HER-2-negativeadvanced breast cancer.
Chen, C; Kong, F; Lu, J; Shan, H; Song, X; Wang, L; Yuan, H,
)
1.95
" However, the pharmacokinetic and pharmacodynamic information for EVL combined with TAC is limited."( Pharmacodynamic Drug-Drug Interaction on Human Peripheral Blood Mononuclear Cells Between Everolimus and Tacrolimus at the Therapeutic Concentration Range in Renal Transplantation.
Akashi, I; Akiyama, S; Hirano, T; Iwamoto, H; Kihara, Y; Konno, O; Oda, T; Okihara, M; Osato, S; Takeuchi, H; Tanaka, S; Unezaki, S; Yoshinaga, R, 2021
)
0.84
"This case suggests that anterior and posterior spinal fusion combined with everolimus therapy can be a therapeutic option for GSD."( Gorham-Stout Disease Resulting in Spinal Deformity Treated by Fusion Surgery Combined With Everolimus Therapy: A Case Report.
Fujita, N; Matsumoto, M; Nagoshi, N; Nakamura, M; Nori, S; Okada, E; Suzuki, S; Toga, A; Tsuji, O; Watanabe, K; Yagi, M, 2021
)
1.07
" The primary objective was to determine the maximum tolerated dose (MTD)/recommended dose for expansion (RDE) of ALP in combination with EVE and in combination with EVE and exemestane (EXE) and subsequently assess safety, preliminary efficacy and effect of ALP on the pharmacokinetics of EVE and determine the magnitude of the drug-drug interaction."( Alpelisib in combination with everolimus ± exemestane in solid tumours: Phase Ib randomised, open-label, multicentre study.
Ajipa, O; Andre, F; Beck, JT; Blumenstein, L; Curigliano, G; Donnet, V; Fazio, N; Hubner, RA; Jhaveri, K; Lahner, H; Li, Z; Martin, M; Maur, M; Tortora, G, 2021
)
0.91
" There were no clinically relevant drug-drug interactions observed between ALP and EVE."( Alpelisib in combination with everolimus ± exemestane in solid tumours: Phase Ib randomised, open-label, multicentre study.
Ajipa, O; Andre, F; Beck, JT; Blumenstein, L; Curigliano, G; Donnet, V; Fazio, N; Hubner, RA; Jhaveri, K; Lahner, H; Li, Z; Martin, M; Maur, M; Tortora, G, 2021
)
0.91
" Pharmacokinetics of ALP, EVE and EXE was largely unchanged in combination with each other."( Alpelisib in combination with everolimus ± exemestane in solid tumours: Phase Ib randomised, open-label, multicentre study.
Ajipa, O; Andre, F; Beck, JT; Blumenstein, L; Curigliano, G; Donnet, V; Fazio, N; Hubner, RA; Jhaveri, K; Lahner, H; Li, Z; Martin, M; Maur, M; Tortora, G, 2021
)
0.91
" This study aimed to investigate a 1-year clinical follow-up of a prospective multicenter registry of HCR combined with non-saphenous vein graft surgical bypass and PCI using everolimus-eluting metallic stents (the PRIDE-METAL study)."( Prospective multicenter registry of hybrid coronary artery revascularization combined with non-saphenous vein graft surgical bypass and percutaneous coronary intervention using everolimus eluting metallic stents (PRIDE-METAL study).
Doi, H; Fujita, T; Fuku, Y; Fukui, T; Kinoshita, Y; Komiya, T; Kozuma, K; Okawa, Y; Shimokawa, T; Takanashi, S; Tobaru, T; Tsujita, K; Watanabe, Y, 2022
)
1.11
" To this end, we investigated experimentally and assessed computationally the in vitro pharmacodynamic drug-drug interactions of the various dual and triple combinations to assess their subsequent combinatorial effects (synergistic/additive/antagonistic) in a HER2-therapy resistant BC cell line, JIMT-1."( Experimental and computational assessment of the synergistic pharmacodynamic drug-drug interactions of a triple combination therapy in refractory HER2-positive breast cancer cells.
Ait-Oudhia, S; Vaidya, TR, 2022
)
0.72
" Four canine melanoma cell lines were treated in vitro with rapamycin and everolimus as sole treatment or combined with carboplatin."( Investigation of the effects of mTOR inhibitors rapamycin and everolimus in combination with carboplatin on canine malignant melanoma cells.
Bernard, S; Mutsaers, AJ; Poon, AC; Tam, PM, 2021
)
1.09
"When combined with carboplatin chemotherapy, rapamycin or everolimus treatment was overall synergistic in reducing cell viability."( Investigation of the effects of mTOR inhibitors rapamycin and everolimus in combination with carboplatin on canine malignant melanoma cells.
Bernard, S; Mutsaers, AJ; Poon, AC; Tam, PM, 2021
)
1.11
"Inhibition of mTOR by rapalogs, such as rapamycin and everolimus combined with carboplatin chemotherapy may have activity in canine melanoma."( Investigation of the effects of mTOR inhibitors rapamycin and everolimus in combination with carboplatin on canine malignant melanoma cells.
Bernard, S; Mutsaers, AJ; Poon, AC; Tam, PM, 2021
)
1.11
" This case report indicated that remdesivir might interact with cytochrome P450 3A4 substrates, such as tacrolimus and everolimus, and elevate their blood concentrations under high inflammatory conditions."( Drug-drug interaction between remdesivir and immunosuppressant agents in a kidney transplant recipient.
Hirai, T; Inoue, T; Iwamoto, T; Mizuta, A; Nishikawa, K; Sasaki, T, 2022
)
0.93
" 14 mg daily) in combination with everolimus (5 mg daily), following one prior vascular endothelial growth factor-targeted treatment."( Health-Related Quality of Life Outcomes With Two Different Starting Doses of Lenvatinib in Combination With Everolimus for Previously Treated Renal Cell Carcinoma.
Alekseev, B; Bennett, L; Bergerot, C; Castellano, D; Ciuleanu, T; Glen, H; Heng, D; Koralewski, P; Lyun Lee, J; O'Hara, K; Pal, S; Pan, J; Parnis, F; Puente, J; Stroyakovskiy, D; Sunela, K; Wang, J; Young Rha, S, 2023
)
1.4
"The study intended to explore the effects and molecular mechanisms of imatinib combined with everolimus on AtT-20 cells in AtT-20 mouse pituitary tumors."( Effects of Imatinib Combined With Everolimus on Mouse Pituitary Tumor Cell AtT-20.
Lin, Y; Meng, L; Qu, X; Wang, W; Yang, J, 2023
)
1.41
"Imatinib combined with everolimus can affect the AtT-20 cell cycle through the signaling pathway of the phosphatidylin-ositol-3-kinase (PI3K)/Akt/ protein kinase A (PKA) system and can inhibit cell proliferation and induce cell apoptosis."( Effects of Imatinib Combined With Everolimus on Mouse Pituitary Tumor Cell AtT-20.
Lin, Y; Meng, L; Qu, X; Wang, W; Yang, J, 2023
)
1.5
" Three years and six months after surgery, she was administered octreotide LAR 30 mg per month in combination with everolimus."( Everolimus in combination with octreotide LAR in thymic atypical carcinoid.
Hagui, E; Haneda, H; Nakano, T; Okuda, K; Sakane, T, 2023
)
2.56
" A single dose of everolimus 5 mg was well tolerated when administered with multiple doses of CBD."( Pharmacokinetic Drug-Drug Interaction With Coadministration of Cannabidiol and Everolimus in a Phase 1 Healthy Volunteer Trial.
Berwaerts, J; Chen, C; Critchley, D; Hyland, K; Tayo, B; Thai, C; Wray, L, 2023
)
1.47
" Patients received a weekly intravenous infusion of xentuzumab (1000 mg) or placebo in combination with everolimus (10 mg/day orally) and exemestane (25 mg/day orally)."( XENERA-1: a randomised double-blind Phase II trial of xentuzumab in combination with everolimus and exemestane versus everolimus and exemestane in patients with hormone receptor-positive/HER2-negative metastatic breast cancer and non-visceral disease.
Baktash, N; Biyukov, T; Blau, S; Burris, HA; Cortes, J; Díaz-Redondo, T; García-Sáenz, JÁ; Hart, L; Jañez, NM; Joaquim, A; Lemieux, J; Massey, D; Morales, S; Neven, P; Rugo, HS; Schmid, P, 2023
)
1.35
"While this study demonstrated that xentuzumab could be safely combined with everolimus and exemestane in patients with HR-positive/HER2-negative advanced breast cancer with non-visceral disease, there was no PFS benefit with the addition of xentuzumab."( XENERA-1: a randomised double-blind Phase II trial of xentuzumab in combination with everolimus and exemestane versus everolimus and exemestane in patients with hormone receptor-positive/HER2-negative metastatic breast cancer and non-visceral disease.
Baktash, N; Biyukov, T; Blau, S; Burris, HA; Cortes, J; Díaz-Redondo, T; García-Sáenz, JÁ; Hart, L; Jañez, NM; Joaquim, A; Lemieux, J; Massey, D; Morales, S; Neven, P; Rugo, HS; Schmid, P, 2023
)
1.36
" In transplantation, therapeutic drug monitoring (TDM) is recommended due to the potential drug-drug interactions with chronic medications, which can affect everolimus pharmacokinetics."( First reported double drug-drug interaction in a cancer renal patient under everolimus treatment: therapeutic drug monitoring and review of literature.
Bleda-Perez, C; Del Valle-Celiz, PM; Fontanals-Martínez, S; Fort-Casamartina, E; Gonzalo-Diego, N; Muñoz-Sanchez, C; Otero-Torres, S; Prats-Jimenez, J; Rigo-Bonnin, RF, 2023
)
1.34

Bioavailability

RAD001 (everolimus) is a potent, orally bioavailable inhibitor of the mTOR pathway. Low water solubility and low bioavailability of everolimus have prevented its clinical development as an anticancer drug.

ExcerptReferenceRelevance
"The new immunosuppressive agent, SDZ-RAD, and its analog rapamycin were examined for intestinal absorption, metabolism, and bioavailability in Wistar rats."( Absorption and intestinal metabolism of SDZ-RAD and rapamycin in rats.
Bruelisauer, A; Crowe, A; Duerr, L; Guntz, P; Lemaire, M, 1999
)
0.3
" The rate of absorption of everolimus was not altered by hepatic impairment on the basis of the maximum blood concentration (C(max)) and time to reach C(max) (t(max))."( Influence of hepatic impairment on everolimus pharmacokinetics: implications for dose adjustment.
Dilzer, SC; Figueiredo, J; Kovarik, JM; Lasseter, K; Reynolds, C; Rordorf, C; Sabia, HD; Zimmermann, H, 2001
)
0.88
"To quantify the influence of a high-fat meal on the oral bioavailability of the immunosuppressant everolimus in a single-dose study in healthy subjects and to confirm the results in a small food-effect screening assessment in patients with renal transplants who were receiving multiple-dose everolimus."( Effect of food on everolimus absorption: quantification in healthy subjects and a confirmatory screening in patients with renal transplants.
Budde, K; Figueiredo, J; Golor, G; Hartmann, S; Kovarik, JM; Lison, A; Neumayer, HH; Rordorf, C, 2002
)
0.87
" As part of the pediatric clinical development program, the relative bioavailability and food effect for the dispersible tablet were evaluated in healthy adult subjects as a prelude to characterizing the steady-state pharmacokinetics in pediatric kidney allograft recipients."( Clinical development of an everolimus pediatric formulation: relative bioavailability, food effect, and steady-state pharmacokinetics.
Berthier, S; Ettenger, R; Hoyer, PF; Kovarik, JM; Langholff, WK; Marion, AS; McMahon, L; Noe, A; Rordorf, C, 2003
)
0.62
" Black patients may have lower bioavailability and/or higher clearance of everolimus compared with white patients."( Pharmacokinetics of an everolimus-cyclosporine immunosuppressive regimen over the first 6 months after kidney transplantation.
Berthier, S; Dantal, J; Hsu, CH; Kahan, BD; Kaplan, B; Kovarik, JM; McMahon, L; Rordorf, C; Silva, HT, 2003
)
0.86
"The orally bioavailable rapamycin derivative RAD001 (everolimus) targets the mammalian target of rapamycin pathway and possesses potent immunosuppressive and anticancer activities."( Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells.
Beuvink, I; Boulay, A; Haller, R; Heusser, C; Lane, HA; Marti, A; O'Reilly, T; Stephan, C; Stolz, B; Thomas, G; Tobler, S; Zilbermann, F; Zumstein-Mecker, S, 2004
)
0.57
" Everolimus is more hydrophilic, exhibits a shorter elimination half-life (approximately 30 hours), and demonstrates greater relative bioavailability compared to sirolimus."( The evolving experience using everolimus in clinical transplantation.
Bia, MJ; Formica, RN; Friedman, AL; Lakkis, F; Lorber, KM; Lorber, MI; Smith, JD, 2004
)
1.52
" Compared with sirolimus, everolimus has unique pharmacokinetic characteristics including greater bioavailability and a shorter half-life, allowing more rapid achievement of a steady state."( Experience with everolimus.
Augustine, JJ; Hricik, DE, 2004
)
0.97
" Cyclosporine, tacrolimus, sirolimus and everolimus, however, have all been described to exhibit ethnicity-specific differences in bioavailability and/or dose-adjusted systemic exposure, although currently available reports are controversial for some of these drugs."( Pharmacokinetics of immunosuppressants: a perspective on ethnic differences.
Dirks, NL; Huth, B; Meibohm, B; Yates, CR, 2004
)
0.59
" RAD001 (everolimus) is a potent, orally bioavailable inhibitor of the mTOR pathway."( Future directions in the treatment of hormone-sensitive advanced breast cancer: the RAD001 (Everolimus)-letrozole clinical program.
Lane, HA; Lebwohl, D, 2006
)
0.97
" It is a proliferation signal inhibitor with an improved pharmacokinetic profile and bioavailability compared with sirolimus."( Everolimus: an immunosuppressive agent in transplantation.
Kobashigawa, JA; Patel, JK, 2006
)
1.78
" The oral bioavailability value, as estimated with the PBPK, was 12% and was similar to the value obtained by non-compartmental analysis."( Physiologically based pharmacokinetic (PBPK) modeling of everolimus (RAD001) in rats involving non-linear tissue uptake.
Kawai, R; Laplanche, R; Meno-Tetang, GM, 2007
)
0.58
" Closer surveillance for posttransplant infections and alterations in drug bioavailability and dosing as well as break-through immunologic responses are required."( Out with the old, in with the new: immunosuppression minimization in children.
Sarwal, MM, 2008
)
0.35
" The bioavailability after intraportal and intraintestinal administration were 48."( Effect of intestinal and hepatic first-pass extraction on the pharmacokinetics of everolimus in rats.
Inui, K; Katsura, T; Masuda, S; Sato, E; Yano, I; Yokomasu, A, 2008
)
0.57
" NVP-AEW541 is a new, orally bioavailable small molecule inhibitor of the insulin-like growth factor-1 receptor (IGF-1R)."( Myeloma cell growth inhibition is augmented by synchronous inhibition of the insulin-like growth factor-1 receptor by NVP-AEW541 and inhibition of mammalian target of rapamycin by Rad001.
Baumann, P; Franke, D; Hagemeier, H; Mandl-Weber, S; Schmidmaier, R, 2009
)
0.35
" The objective of the study was to evaluate the anti-proliferative efficacy of the mTOR inhibitor, rapamycin, and its orally bioavailable analog RAD001 on the GH-secreting pituitary tumor GH3 and MtT/S cells and in human GH-secreting pituitary adenomas (GH-omas) in primary cell cultures."( Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GH-secreting pituitary tumor cells in vitro.
Cerovac, V; Cohen, ZR; Gorshtein, A; Hadani, M; Kendler, E; Rubinfeld, H; Shimon, I; Stalla, GK; Theodoropoulou, M, 2009
)
0.59
"We conclude that in adult renal transplant recipients, EVL significantly decreases TAC oral bioavailability in a dose-dependent manner."( Interaction between everolimus and tacrolimus in renal transplant recipients: a pharmacokinetic controlled trial.
Brunet, M; Cabello, M; del Castillo, D; Fernández, AM; Grinyó, JM; Pallardó, L; Pascual, J, 2010
)
0.68
" Those drugs have low and variable oral bioavailability that is increased when combined with cyclosporine or tacrolimus (TAC)."( Sirolimus and everolimus intestinal absorption and interaction with calcineurin inhibitors: a differential effect between cyclosporine and tacrolimus.
Boussera, B; Lamoureux, F; Marquet, P; Picard, N; Sauvage, FL, 2012
)
0.74
" Everolimus was more effective than rapamycin in preventing diabetic nephropathy in vivo, which may be contributed to the fact that everolimus has better bioavailability and a higher oral absorption rate."( Everolimus vs. rapamycin for treating diabetic nephropathy in diabetic mouse model.
Chen, J; Cheng, L; Mao, X, 2011
)
2.72
" However, the low water solubility and low bioavailability of everolimus have prevented its clinical development as an anticancer drug."( Preparation and in vivo evaluation of liposomal everolimus for lung carcinoma and thyroid carcinoma.
Iwase, Y; Maitani, Y, 2012
)
0.87
" Temsirolimus has discerning features compared with other currently registered drugs, such as its intravenous administration route-providing predictable bioavailability and adherence to treatment-and potential benefit in nonclear cell histologies."( The clinical potential of temsirolimus in second or later lines of treatment for metastatic renal cell carcinoma.
Greil, R; Grünwald, V; Porta, C; Stenner-Liewen, F, 2013
)
0.39
"Macrocycles are ideal in efforts to tackle "difficult" targets, but our understanding of what makes them cell permeable and orally bioavailable is limited."( Macrocyclic drugs and clinical candidates: what can medicinal chemists learn from their properties?
Giordanetto, F; Kihlberg, J, 2014
)
0.4
"We used wild-type, Abcb1a/1b(-/-), Abcg2(-/-), Abcb1a/1b;Abcg2(-/-), and Cyp3a(-/-) mice to study everolimus oral bioavailability and brain accumulation."( P-glycoprotein, CYP3A, and plasma carboxylesterase determine brain and blood disposition of the mTOR Inhibitor everolimus (Afinitor) in mice.
Beijnen, JH; Cheung, KL; Durmus, S; Fukami, T; Schinkel, AH; Sparidans, RW; Tang, SC; van Vlijmen, BJ; Wagenaar, E; Yokoi, T, 2014
)
0.83
" The physicochemical properties, drug release profile, and bioavailability of SD prepared by SW process were also compared to SD prepared by the conventional co-precipitation method."( Improved oral absorption and chemical stability of everolimus via preparation of solid dispersion using solvent wetting technique.
Jang, SW; Kang, MJ, 2014
)
0.65
" In conclusion, oral intake of curcumin significantly decreased the bioavailability of EVL, a probe substrate of P-gp/CYP 3A4, mainly through marked activation on CYP 3A4."( Oral intake of curcumin markedly activated CYP 3A4: in vivo and ex-vivo studies.
Chao, PD; Hou, YC; Hsieh, YW; Huang, CY; Peng, YH; Yang, SY; Yu, CP, 2014
)
0.4
" Data from large clinical trials have shown that the combination of everolimus , an orally bioavailable mTOR inhibitor with exemestane improves outcome of metastatic breast cancer resistant to non-steroidal aromatase inhibitors."( Evaluating the pharmacokinetics and pharmacodynamics of everolimus for treating breast cancer.
Awada, A; Barthélémy, P; Gombos, A, 2015
)
0.9
" The pharmacokinetics, metabolism and bioavailability of EC0565 were studied in normal rats."( Antiinflammatory Activity of a Novel Folic Acid Targeted Conjugate of the mTOR Inhibitor Everolimus.
Cross, VA; Gehrke, MA; Hahn, SJ; Klein, PJ; Kleindl, PJ; Leamon, CP; Low, PS; Lu, Y; Parker, N; Santhapuram, HK; Stinnette, TW; Vaughn, JF; Vlahov, IR; Wang, K; Westrick, E; Wollak, K; You, F, 2015
)
0.64
"The aims of this study were to improve in vitro dissolution property of poorly water-soluble everolimus (EVR) for enhanced bioavailability without using organic solvents and characterize the effects of microfluidization and freeze-drying on physicochemical properties of EVR nanosuspension and nanoparticle, respectively."( Formulation of Nanoparticle Containing Everolimus Using Microfluidization and Freeze-Drying.
Hwang, KM; Kang, SY; Kim, SH; Park, ES; Seo, JW; Seok, SH, 2016
)
0.92
" Meanwhile, the pharmacokinetics study in vivo confirmed that the delivery of everolimus through the injection of the prodrug can overcome the low bioavailability of oral everolimus."( Synthesis and evaluation of an injectable everolimus prodrug.
Cai, Z; Wang, H; Yu, O; Zheng, S; Zheng, X; Zhu, T, 2017
)
0.95
" BEZ235 systemic exposure increased in a dose-proportional manner while oral bioavailability was quite low, which may be related to gastrointestinal-specific toxicity."( A Phase Ib Study of the Dual PI3K/mTOR Inhibitor Dactolisib (BEZ235) Combined with Everolimus in Patients with Advanced Solid Malignancies.
Beg, MS; Desai, P; Fathallah, H; Karim, NA; Kozma, SC; Mercer, CA; Moorthy, G; Morris, JC; O'Gara, S; Olowokure, O; Rixe, O; Salkeni, MA; Thomas, G; Thomas, HE; Wise-Draper, TM, 2017
)
0.68
"We found that formulation did not influence bioavailability and that use of >20 mg prednisolone daily increased everolimus clearance."( A pharmacological rationale for improved everolimus dosing in oncology and transplant patients.
Burger, DM; de Fijter, JW; Guchelaar, HJ; Moes, DJAR; Reinders, MEJ; Ter Heine, R; van Erp, NP; van Herpen, CM, 2018
)
0.96
" There is growing evidence to suggest that the bioavailability of the potent pro-tumor function of FGFR2 is associated with thrombospondins (TSPs)."( FGFR2 Promotes Gastric Cancer Progression by Inhibiting the Expression of Thrombospondin4 via PI3K-Akt-Mtor Pathway.
Dai, Y; Huang, T; Li, P; Liu, D; Qiu, H; Sun, L; Wang, Y; Xiong, H; Yuan, X; Zou, M, 2018
)
0.48
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
" Being water-insoluble, administration is restricted to the oral route, which results in a low bioavailability of <10%."( Berunda Polypeptides Carrying Rapalogues Inhibit Tumor mTORC1 Better than Oral Everolimus.
MacKay, JA; Peddi, S, 2020
)
0.79
" The aim of this study was to develop a novel sublingual orodispersible film loading everolimus for improving patient compliance and enhancing oral bioavailability of EVR."( Designing orodispersible films containing everolimus for enhanced compliance and bioavailability.
Gao, S; Guan, R; Liu, H; Liu, Y; Ma, Y; Song, W; Yang, Y, 2020
)
1.05
" Pharmacokinetic studies showed that loading into the sublingual orodispersible films significantly increased the oral bioavailability of EVR."( Designing orodispersible films containing everolimus for enhanced compliance and bioavailability.
Gao, S; Guan, R; Liu, H; Liu, Y; Ma, Y; Song, W; Yang, Y, 2020
)
0.82
" Therefore, the aim of this study is to develop a topical nanomicellar formulation of an immunosuppressant drug, everolimus using Soluplus®, a grafted copolymer of polyvinyl caprolactam-polyvinylalcohol-polyethyleneglycol (PVCL-PVA-PEG) for improved permeation through ocular epithelia with minimal or no irritation resulting in enhanced ocular bioavailability at the posterior segments of the eye for the treatment of uveitis."( A grafted copolymer-based nanomicelles for topical ocular delivery of everolimus: Formulation, characterization, ex-vivo permeation, in-vitro ocular toxicity, and stability study.
Aqil, M; Mehra, N; Sultana, Y, 2021
)
1.07
" We found that RCC cells are highly dependent on glutamine for proliferation, and this dependence strongly correlated with sensitivity to telaglenstat (CB-839), an investigational, first-in-class, selective, orally bioavailable GLS inhibitor."( The glutaminase inhibitor telaglenastat enhances the antitumor activity of signal transduction inhibitors everolimus and cabozantinib in models of renal cell carcinoma.
Chen, J; Dang, R; Emberley, E; Gross, M; Huang, T; Li, W; MacKinnon, A; Pan, A; Parlati, F; Singh, D; Sotirovska, N; Steggerda, SM; Wang, T, 2021
)
0.83
" However, due to the low oral bioavailability and narrow response window of oral everolimus, a new delivery system is urgently needed to overcome the above problems."( Tip-concentrated microneedle patch delivering everolimus for therapy of multiple sclerosis.
Liu, H; Ren, Y; Song, W; Wang, N; Yang, Y, 2022
)
1.21
" One phase II trial of the potent orally bioavailable mTOR inhibitor, everolimus, in combination with letrozole demonstrated an unprecedented clinical benefit rate (CBR) of 40% and high objective response rate (RR) of 32% in hormone agnostic endometrial cancers."( Targeting the PI3K Pathway in Gynecologic Malignancies.
Avila, M; Grinsfelder, MO; Pham, M; Westin, SN, 2022
)
0.96
" Analogs of rapamycin were developed to improve its pharmacological properties, such as low oral bioavailability and a long half-life."( Why Is Rapamycin Not a Rapalog?
Kuerec, AH; Maier, AB, 2023
)
0.91

Dosage Studied

Rapamycin inhibitor everolimus was recently approved in the USA and a number of EU countries for use in combination with tacrolimus and corticosteroids for the prophylaxis of organ rejection in adult liver transplant recipients.

ExcerptRelevanceReference
" in the range of the steep part of its dose-response relationship."( The peripheral lymphocyte count predicts graft survival in DA to Lewis heterotopic heart transplantation treated with FTY720 and SDZ RAD.
Baumlin, Y; Hof, A; Hof, RP; Nikolova, Z, 2000
)
0.31
" Potential differences in cyclosporine dosing and pharmacokinetics at different levels of everolimus exposure were assessed in the context of ANOVA."( Longitudinal assessment of everolimus in de novo renal transplant recipients over the first post-transplant year: pharmacokinetics, exposure-response relationships, and influence on cyclosporine.
Boger, R; Cambon, N; Gerbeau, C; Kahan, BD; Kaplan, B; Kovarik, JM; Lorber, M; Rordorf, C; Rouilly, M; Winkler, M, 2001
)
0.83
" For a 4-fold range of everolimus doses there were no differential effects on cyclosporine dosing or pharmacokinetics."( Longitudinal assessment of everolimus in de novo renal transplant recipients over the first post-transplant year: pharmacokinetics, exposure-response relationships, and influence on cyclosporine.
Boger, R; Cambon, N; Gerbeau, C; Kahan, BD; Kaplan, B; Kovarik, JM; Lorber, M; Rordorf, C; Rouilly, M; Winkler, M, 2001
)
0.92
" Drug efficacy and potency was calculated based on dose-response curves of the drug-mediated decrease in CD4(+)/CD8alpha(+)/CD25(+) cells."( Coexpression of CD4 and CD8alpha on rat T-cells in whole blood: a sensitive marker for monitoring T-cell immunosuppressive drugs.
Diaz-Romero, J; Vogt, G; Weckbecker, G, 2001
)
0.31
" There use allows a lower dosage for the calcineurin inhibitor."( [Immunosuppression, ongoing clinical trials].
Rivalan, J, 2001
)
0.31
" This suggested that, although the adult 12-hour dosing interval was appropriate for pediatric patients, they would require reduced dosing based on body size compared with adults."( Safety and efficacy of TOR inhibitors in pediatric renal transplant recipients.
Ettenger, RB; Grimm, EM, 2001
)
0.31
" On two occasions, a pharmacokinetic profile was obtained over the dosing interval after drug administration under fasting conditions and after a high-fat meal in a randomized sequence."( Effect of food on everolimus absorption: quantification in healthy subjects and a confirmatory screening in patients with renal transplants.
Budde, K; Figueiredo, J; Golor, G; Hartmann, S; Kovarik, JM; Lison, A; Neumayer, HH; Rordorf, C, 2002
)
0.65
" This is a lower therapeutic concentration limit when everolimus is used with conventionally dosed cyclosporine."( Exposure-response relationships for everolimus in de novo kidney transplantation: defining a therapeutic range.
Boger, R; Dantal, J; Kahan, BD; Kaplan, B; Kovarik, JM; Rordorf, C; Tedesco Silva, H; Vitko, S, 2002
)
0.84
" Based on this information, pediatric patients will need a dose scaled down for body size, but can probably maintain the same twice-daily dosing schedule used in adults."( Single-dose pharmacokinetics and tolerability of everolimus in stable pediatric renal transplant patients.
Boger, R; Cambon, N; Ettenger, R; Hoyer, PF; Kovarik, JM; Lemire, J; Mahan, J; McMahon, L; Van Damme-Lombaerts, R; Webb, NA, 2002
)
0.57
" Successful dosing was confirmed by measuring splenic weight."( The rapamycin analogue SDZ RAD attenuates bleomycin-induced pulmonary fibrosis in rats.
Chambers, RC; Egan, JJ; Goldsack, NR; Hasleton, PS; Howell, DC; Laurent, GJ; Marshall, RP; Simler, NR, 2002
)
0.31
" The newest drugs to reach clinical evaluation, sirolimus and everolimus, have been studied in the context of concentration-controlled dosing and there is a good rationale for their measurement."( Therapeutic drug monitoring of immunosuppressive drugs in kidney transplantation.
Holt, DW, 2002
)
0.56
" Steady-state concentration-time profiles in pediatric transplant patients receiving the dispersible tablet were comparable to those of adult patients receiving the conventional tablet when both were dosed to yield similar trough concentrations."( Clinical development of an everolimus pediatric formulation: relative bioavailability, food effect, and steady-state pharmacokinetics.
Berthier, S; Ettenger, R; Hoyer, PF; Kovarik, JM; Langholff, WK; Marion, AS; McMahon, L; Noe, A; Rordorf, C, 2003
)
0.62
" Weight-adjusted dosing (mg/kg) does not appear warranted."( Pharmacokinetics of an everolimus-cyclosporine immunosuppressive regimen over the first 6 months after kidney transplantation.
Berthier, S; Dantal, J; Hsu, CH; Kahan, BD; Kaplan, B; Kovarik, JM; McMahon, L; Rordorf, C; Silva, HT, 2003
)
0.63
" There were no trends in C(min) or AUC versus patient age when everolimus was dosed on a mg/m2 basis."( Everolimus in pediatric de nova renal transplant patients.
Ettenger, R; Hoyer, PF; Kovarik, JM; Lemire, J; Loirat, C; Mahan, J; Mayer, H; Mentser, M; Moeller, V; Niaudet, P; Offner, G; VanDamme-Lombaerts, R; Webb, NJ; Wehr, S, 2003
)
2
"These data support the use of body surface area-adjusted dosing for everolimus in pediatric patients."( Everolimus in pediatric de nova renal transplant patients.
Ettenger, R; Hoyer, PF; Kovarik, JM; Lemire, J; Loirat, C; Mahan, J; Mayer, H; Mentser, M; Moeller, V; Niaudet, P; Offner, G; VanDamme-Lombaerts, R; Webb, NJ; Wehr, S, 2003
)
2
" At 8 visits during the first 6 months after transplantation, we obtained 2,328 everolimus trough levels (Cmin) and 129 area-under-the-curve (AUC) profiles over the dosing interval in patients treated with everolimus; we collected 3,258 cyclosporine trough concentrations and 174 profiles in all 3 treatment arms."( Everolimus in de novo cardiac transplantation: pharmacokinetics, therapeutic range, and influence on cyclosporine exposure.
Dorent, R; Eisen, H; Hsu, CH; Kovarik, JM; Mancini, D; Rordorf, C; Rouilly, M; Vigano, M, 2003
)
1.99
"We quantified the influence of delayed initiation of cyclosporine on everolimus pharmacokinetics in order to provide dosing guidance for kidney transplant patients."( Influence of delayed initiation of cyclosporine on everolimus pharmacokinetics in de novo renal transplant patients.
Bettoni-Ristic, O; Civati, G; Dantal, J; Kovarik, JM; Rizzo, G; Rordorf, C; Rouilly, M, 2003
)
0.81
" The synergistic effect allows a dosage reduction that decreases adverse effects."( Clinical pharmacokinetics of everolimus.
Kirchner, GI; Manns, MP; Meier-Wiedenbach, I, 2004
)
0.61
" Everolimus dosing was adjusted to maintain a minimum trough level of 3 ng/mL."( Everolimus with optimized cyclosporine dosing in renal transplant recipients: 6-month safety and efficacy results of two randomized studies.
Alves Filho, G; Bernhardt, P; Bourbigot, B; Cambi, V; Campbell, S; Civati, G; Cretin, N; Eris, J; Esmeraldo, R; Garcia, VD; Geissler, J; Haas, T; Jappe, A; Leone, J; Magee, JC; Pascual, J; Rigotti, P; Tedesco, H; Vitko, S; Whelchel, J, 2004
)
2.68
" However, optimal cyclosporine exposure can now be achieved through monitoring of cyclosporine levels 2 hours after dosing (C(2) monitoring)."( Optimizing the immunosuppressive regimen in heart transplantation.
Eisen, H; Ross, H, 2004
)
0.32
" Cyclosporine dosing began on average at 274 +/- 78 mg bid, was down-titrated in months 2-3 from 181 +/- 80 mg to 81 +/- 33 mg bid, and stabilized at 70 +/- 26 mg bid thereafter."( Everolimus therapeutic concentration range defined from a prospective trial with reduced-exposure cyclosporine in de novo kidney transplantation.
Bizot, MN; Civati, G; Geissler, J; Kovarik, JM; Pascual, J; Schmidli, H; Tedesco, H, 2004
)
1.77
"The aims of the current study were to determine whether therapeutic drug monitoring (TDM) might benefit kidney transplant recipients receiving everolimus, and to establish dosage recommendations when everolimus is used in combination with cyclosporine and corticosteroids."( Therapeutic drug monitoring for everolimus in kidney transplantation using 12-month exposure, efficacy, and safety data.
Kovarik, J; Li, Y; Lorber, MI; Mayer, HW; Ponticelli, C; Schmidli, H; Whelchel, J, 2005
)
0.81
" The objective of this randomized, double-blind, placebo-controlled, dose-escalating Phase 1 study was to evaluate the pharmacokinetic profile of different dosing regimens of everolimus."( Pharmacokinetics of the immunosuppressant everolimus in maintenance renal transplant patients.
Budde, K; Fritsche, L; Glander, P; Neumayer, HH; Slowinski, T; Waiser, J, 2005
)
0.79
" In everolimus-treated patients, mean increases in cyclosporine AUC0-12 ranged from 7% to 43%, which were not significantly different across all dosing cohorts including placebo."( Influence of everolimus on steady-state pharmacokinetics of cyclosporine in maintenance renal transplant patients.
Budde, K; Dantal, J; Fauchald, P; Fritsche, L; Lehne, G; Lison, A; Neumayer, HH; Renders, L; Soulillou, JP; Winkler, M, 2005
)
1.26
"All randomised controlled trials (RCTs) and quasi-RCTs where drug regimens containing TOR-I were compared to alternative drug regimens in the immediate post-transplant period were included, without age restriction, dosage or language of report."( Target of rapamycin inhibitors (TOR-I; sirolimus and everolimus) for primary immunosuppression in kidney transplant recipients.
Chapman, JR; Craig, JC; Lee, VW; Webster, AC, 2006
)
0.58
" Everolimus has shown to reduce acute cellular rejection and may allow CsA dosage reduction."( Everolimus and reduced cyclosporine trough levels in maintenance heart transplant recipients.
Prenner, G; Schwarz, M; Schweiger, M; Stadlbauer, V; Stiegler, P; Tscheliessnigg, K; Wasler, A, 2006
)
2.69
"CsA trough levels and dosage can be significantly reduced in combination with Everolimus without higher rejection rates and with stable kidney function in oHTX patients."( Everolimus and reduced cyclosporine trough levels in maintenance heart transplant recipients.
Prenner, G; Schwarz, M; Schweiger, M; Stadlbauer, V; Stiegler, P; Tscheliessnigg, K; Wasler, A, 2006
)
2
" A reduction in MPA dosage at the time of conversion may be necessary to prevent over-immunosuppression."( Conversion to everolimus in maintenance patients--current clinical strategies.
Pohanka, E, 2006
)
0.69
"9 ng/mL and area under the curve over a dosing interval (AUC) was 132 +/- 56 ng x h/mL."( Differential pharmacokinetic interaction of tacrolimus and cyclosporine on everolimus.
Curtis, JJ; Hricik, DE; Kovarik, JM; Pescovitz, MD; Scantlebury, V; Vasquez, A, 2006
)
0.56
" Basiliximab allows for utilization of lower doses of everolimus with reduced dosing of Neoral)."( 12-month safety and efficacy of everolimus with reduced exposure cyclosporine in de novo renal transplant recipients.
Alves-Filho, G; Bourbigot, B; Cambi, V; Campbell, S; Civati, G; Eris, J; Esmeraldo, R; Garcia, VD; Haas, T; Leone, J; Magee, JC; Pascual, J; Rigotti, P; Tedesco-Silva, H; Vitko, S; Whelchel, J, 2007
)
0.87
" TDM of whole-blood concentrations is mandatory for everolimus (ERL) dosage individualisation."( Comparison of fluorescent polarization immunoassay (FPIA) versus HPLC to measure everolimus blood concentrations in clinical transplantation.
Fruehwirth, F; Halwachs-Baumann, G; Khoschsorur, G; Stettin, M; Zelzer, S, 2007
)
0.82
"Moderate dosage of everolimus inhibits cystogenesis in Han:SPRD rats."( Everolimus retards cyst growth and preserves kidney function in a rodent model for polycystic kidney disease.
Le Hir, M; Serra, AL; Wackerle-Men, Y; Wahl, PR; Wu, M; Wuthrich, RP, 2007
)
2.11
" The results were evaluated by two-way analysis of variance followed by post-hoc tests, and nonlinear regression analysis for dose-response rates."( Everolimus and mycophenolate mofetil sensitize human pancreatic cancer cells to gemcitabine in vitro: a novel adjunct to standard chemotherapy?
Dillmann, S; Henne-Bruns, D; Keller, F; Klug, F; Mayer, JM; Stracke, S; Tuncyurek, P, 2007
)
1.78
"5 mg/m2 in cohorts of three to six patients per dosage level."( Phase I study of everolimus in pediatric patients with refractory solid tumors.
Broniscer, A; Fouladi, M; Furman, WL; Gilbertson, RJ; Houghton, PJ; Laningham, F; Luckett, I; Molina, K; O'Shaughnessy, MA; Spunt, SL; Stewart, CF; Wu, J, 2007
)
0.68
" It requires blood concentration-guided dosing and is extensively metabolized."( Crossreactivity of isolated everolimus metabolites with the Innofluor Certican immunoassay for therapeutic drug monitoring of everolimus.
Arabshahi, L; Boyd, J; Christians, U; Haschke, M; Marbach, P; Roberts, M; Strom, T, 2007
)
0.63
" A growing body of evidence finds that the selection and dosing of immunosuppressive therapies can have great influence on long-term transplantation outcomes."( Proliferation signal inhibitors in transplantation: questions at the cutting edge of everolimus therapy.
Albanell, J; Arns, WA; Campistol, JM; Chapman, JR; Eisen, H; Frigerio, M; Lehmkuhl, H; Marcen, R; Morris, R; Nashan, B; Pascual, J; Pohanka, E; Segovia, J; Valantine, H; Zuckermann, A, 2007
)
0.56
" Plasma trough levels of everolimus were determined on a weekly basis before dosing during the first 4 weeks."( Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors.
Baselga, J; Burris, H; Calvo, E; Dimitrijevic, S; Hazell, K; Jones, S; Judson, I; Lane, HA; Lebwohl, D; Macarulla, T; Martinelli, E; Ramon y Cajal, S; Ramos, FJ; Rojo, F; Shand, N; Stumm, M; Tabernero, J; Tang, P; Vidal, L; Zoellner, U, 2008
)
0.87
" A dosage of 10 mg/d or 50 mg/wk is recommended for further development."( Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors.
Baselga, J; Burris, H; Calvo, E; Dimitrijevic, S; Hazell, K; Jones, S; Judson, I; Lane, HA; Lebwohl, D; Macarulla, T; Martinelli, E; Ramon y Cajal, S; Ramos, FJ; Rojo, F; Shand, N; Stumm, M; Tabernero, J; Tang, P; Vidal, L; Zoellner, U, 2008
)
0.57
"To identify the optimal regimen and dosage of the oral mammalian target of rapamycin inhibitor everolimus (RAD001)."( Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors.
Brock, C; Burris, HA; Faivre, S; Hazell, K; Jones, S; Judson, I; Kovarik, JM; Lane, HA; O'Donnell, A; Papadimitrakopoulou, V; Raymond, E; Rea, D; Shand, N; Zoellner, U, 2008
)
0.78
" PD data prompted investigation of 50 and 70 mg weekly and daily dosing at 5 and 10 mg."( Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors.
Brock, C; Burris, HA; Faivre, S; Hazell, K; Jones, S; Judson, I; Kovarik, JM; Lane, HA; O'Donnell, A; Papadimitrakopoulou, V; Raymond, E; Rea, D; Shand, N; Zoellner, U, 2008
)
0.56
"Metronomic dosing of cytotoxic drugs such as cyclophosphamide has shown anti-angiogenic activity, most likely by inducing hypoxia in tumors."( Everolimus (RAD001) and anti-angiogenic cyclophosphamide show long-term control of gastric cancer growth in vivo.
Cejka, D; Fuereder, T; Preusser, M; Sieghart, W; Strommer, S; Wacheck, V; Werzowa, J; Woehrer, A, 2008
)
1.79
" Especially, clinically relevant immunosuppressive drug interactions require prompt identification, intensified monitoring of drug concentrations and adequate dosing responses."( Influence of interactions between immunosuppressive drugs on therapeutic drug monitoring.
Kuypers, DR, 2008
)
0.35
" Of note, prompt reduction of the everolimus dosage since the first azole coadministration, coupled with intensive therapeutic drug monitoring, represented a useful strategy to prevent drug overexposure."( Pharmacokinetic interaction between everolimus and antifungal triazoles in a liver transplant patient.
Adani, GL; Baccarani, U; Baraldo, M; Cojutti, P; Franceschi, L; Furlanut, M; Londero, A; Pea, F; Tavio, M; Viale, P, 2008
)
0.9
"We studied 56 HT patients who sequentially received PSIs to either withdraw (77%) or reduce the dosage of a calcineurin inhibitor; 42 received everolimus (EVE) and 14 sirolimus (SRL)."( Tolerance profile of the proliferation signal inhibitors everolimus and sirolimus in heart transplantation.
Agüero, J; Almenar, L; Martínez-Dolz, L; Moro, JA; Salvador, A; Sánchez-Lázaro, I, 2008
)
0.79
" Closer surveillance for posttransplant infections and alterations in drug bioavailability and dosing as well as break-through immunologic responses are required."( Out with the old, in with the new: immunosuppression minimization in children.
Sarwal, MM, 2008
)
0.35
" This phase 2 study assessed the efficacy of daily oral dosing with everolimus in patients with RCC."( A phase 2 study with a daily regimen of the oral mTOR inhibitor RAD001 (everolimus) in patients with metastatic clear cell renal cell cancer.
Amato, RJ; Giessinger, S; Jac, J; Saxena, S; Willis, JP, 2009
)
0.82
" These data show that a larger dosage is needed for everolimus than sirolimus to maintain the same trough blood concentration."( Larger dosage required for everolimus than sirolimus to maintain same blood concentration in two pancreatic islet transplant patients with tacrolimus.
Inui, K; Iwanaga, Y; Katsura, T; Masuda, S; Matsumoto, S; Okitsu, T; Sato, E; Shimomura, M; Uemoto, S; Yano, I, 2009
)
0.9
" Tacrolimus was dosed seeking a trough blood level of 8 to 10 ng/mL by month 3 and 5 to 8 ng/mL thereafter."( Prospective clinical trial comparing two immunosuppressive regimens, tacrolimus and mycophenolate mofetil versus everolimus and low-dose cyclosporine, in de novo renal transplant recipients: results at 6 months follow-up.
Castagneto, M; Citterio, F; Delreno, F; Favi, E; Gargiulo, A; Romagnoli, J; Spagnoletti, G, 2009
)
0.56
" However, considering the wide ranges found for the prediction error of sirolimus and everolimus concentrations, the clinical relevance of this dosing model is weak."( Sirolimus and everolimus clearance in maintenance kidney and liver transplant recipients: diagnostic efficiency of the concentration/dose ratio for the prediction of trough steady-state concentrations.
Bouzas, L; Hermida, J; Tutor, JC, 2010
)
0.94
" Cyclosporine or tacrolimus dosage was reduced by 80% (group 1, n = 10) or discontinued (group 2, n = 10)."( Renal function outcomes in kidney transplant recipients after conversion to everolimus-based immunosuppression regimen with CNI reduction or elimination.
Alberú, J; Cataneo-Dávila, A; Correa-Rotter, R; Zúñiga-Varga, J, 2009
)
0.58
" Modelling of the PTK/ZK dose-response curve in this model suggested that any effect of everolimus on the PK of PTK/ZK was unlikely to affect efficacy."( Everolimus and PTK/ZK show synergistic growth inhibition in the orthotopic BL16/BL6 murine melanoma model.
Brueggen, J; Lane, HA; Littlewood-Evans, A; McSheehy, PM; O'Reilly, T; Schnell, C; Wood, JM, 2011
)
2.03
" Single dosing of everolimus is possible and safe and may achieve better patient compliance to multiple-drug immunosuppressive therapy."( Conversion of stable kidney transplant recipients from a twice-daily to once-daily everolimus regimen.
Bernini, M; Carmellini, M; Collini, A; Ruggieri, G, 2010
)
0.92
" Therefore, we investigated whether two commonly used therapeutic drug monitoring methods for everolimus were in agreement and to what extent their differences could lead to differences in dosage advice."( Liquid chromatography-tandem mass spectrometry outperforms fluorescence polarization immunoassay in monitoring everolimus therapy in renal transplantation.
de Fijter, JW; den Hartigh, J; Guchelaar, HJ; Moes, DJ; Press, RR, 2010
)
0.79
" Specifically, the use of FPIA can lead to clinically relevant differences in everolimus dosage advice and higher intrapatient variability."( Liquid chromatography-tandem mass spectrometry outperforms fluorescence polarization immunoassay in monitoring everolimus therapy in renal transplantation.
de Fijter, JW; den Hartigh, J; Guchelaar, HJ; Moes, DJ; Press, RR, 2010
)
0.8
" In these conditions, use of an equivalent dosage is of particular importance."( Relationship between everolimus blood concentration assessed using the Innofluor Certican Fluorescence Polarization Immunoassay and the Architect i System sirolimus chemiluminescent microparticle immunoassay.
Carvalho, C; Coentrão, L; Oliveira, JG; Pestana, MI; Sampaio, S, 2010
)
0.68
" On the basis of these results, the recommended Phase II dosage currently being tested is RAD001 70 mg/week in combination with standard chemoradiotherapy."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.61
"Human T-cell lymphoma cell lines Hut-78 and Jurkat were treated with increasing doses of everolimus, alone or in combination with doxorubicin, etoposide, vincristine, or bortezomib, using different dosing schedules."( Schedule-dependent inhibition of T-cell lymphoma cells by cotreatment with the mTOR inhibitor everolimus and anticancer drugs.
He, XX; Huang, JJ; Huang, Y; Li, ZM; Lin, TY; Tian, Y; Xiao, J, 2012
)
0.82
" With certain dosing schedules, everolimus showed synergism with doxorubicin, etoposide, and bortezomib, but antagonism with vincristine."( Schedule-dependent inhibition of T-cell lymphoma cells by cotreatment with the mTOR inhibitor everolimus and anticancer drugs.
He, XX; Huang, JJ; Huang, Y; Li, ZM; Lin, TY; Tian, Y; Xiao, J, 2012
)
0.88
" CNI dosage was progressively reduced until discontinuation."( Tolerability of everolimus-based immunosuppression in maintenance liver transplant recipients.
Adham, M; Boillot, O; Dumortier, J; Gagnieu, MC; Giostra, E; Guillaud, O; Mentha, G; Morard, I; Morelon, E; Vallin, M,
)
0.48
" Although treatment-associated AEs are common, the majority of AEs reported during clinical trial experiences were grade 1 or 2 in severity and manageable with intervention in the form of supportive measures and/or dosage modification."( Treatment-associated adverse event management in the advanced renal cell carcinoma patient treated with targeted therapies.
Ravaud, A, 2011
)
0.37
" Pulsed immunosuppressive dosing can lessen these risks."( Ten-month laryngeal allograft survival with use of pulsed everolimus and anti-alphabeta T-cell receptor antibody immunosuppression.
Dan, O; Khariwala, SS; Lott, DG; Russell, JO; Strome, M, 2011
)
0.61
" Short-term perioperative therapy followed by pulsed, tapered dosing is a viable alternative to traditional regimens and may decrease associated risks."( Ten-month laryngeal allograft survival with use of pulsed everolimus and anti-alphabeta T-cell receptor antibody immunosuppression.
Dan, O; Khariwala, SS; Lott, DG; Russell, JO; Strome, M, 2011
)
0.61
" The primary end points were determination of a safe dosage of everolimus (phase 1) and progression-free survival (PFS) at 24 weeks (phase 2)."( Phase 1/2 study of everolimus in advanced hepatocellular carcinoma.
Abrams, TA; Blaszkowsky, LS; Borger, DR; Clark, JW; Fuchs, CS; Iafrate, AJ; Jors, KR; Kwak, EL; Meyerhardt, JA; Miksad, R; Muzikansky, A; Ryan, DP; Schrag, D; Zheng, H; Zhu, AX, 2011
)
0.94
" The CSA dosage was reduced by 50%."( Everolimus plus dosage reduction of cyclosporine in cardiac transplant recipients with chronic kidney disease: a two-year follow-up study.
Börgermann, J; Fuchs, U; Gummert, JF; Hakim-Meibodi, K; Schulz, U; Zittermann, A, 2011
)
1.81
" CMV data from 2004 de novo RTx recipients from three-randomized, prospective, EVR studies A2309 (N = 833), B201 (N = 588) and B251 (N = 583) were retrospectively analyzed to identify differences between two EVR dosing groups and MPA."( Cytomegalovirus incidence between everolimus versus mycophenolate in de novo renal transplants: pooled analysis of three clinical trials.
Brennan, DC; Legendre, C; Mange, K; McCague, K; Patel, D; Shihab, FS; Wiland, A, 2011
)
0.65
" Still, there is no consensus considering the recommended dosing and the therapeutic range of MPA."( Monitoring of mycophenolic acid and kidney function during combined immunosuppressive therapy.
Asztalos, L; Ivády, G; Kappelmayer, J; Kovács, AM; Oláh, AV; Varga, E; Varga, J, 2011
)
0.37
" Cell growth and proliferation of PC3(res) was similar to that of PC3(par), and late apoptosis increased in PC3(par) but decreased in PC3(res) following treatment with low dosed everolimus."( The cdk1-cyclin B complex is involved in everolimus triggered resistance in the PC3 prostate cancer cell line.
Bartsch, G; Blaheta, RA; Haferkamp, A; Harder, S; Hudak, L; Juengel, E; Kurosch, M; Makarević, J; Tsaur, I; Wiesner, C, 2011
)
0.83
"The coadministration of AEE788 and RAD001 in glioblastoma patients caused a clinically significant thrombocytopenia and a higher-than-expected RAD001 area under the curve concentration when dosed at 200 and 5 mg/day, respectively."( Pharmacokinetic drug interaction between AEE788 and RAD001 causing thrombocytopenia in patients with glioblastoma.
Alfred Yung, WK; Cloughesy, T; Conrad, C; DiLea, C; Dugan, M; Huang, J; Maes, A; Mietlowski, W; Reardon, DA; Rich, J; Yung, L, 2012
)
0.38
" The first 2 patients developed a dose-limiting toxicity (neutropenic infection), prompting a mandatory dose reduction and PK evaluation of both everolimus and docetaxel for patients enrolled in subsequent dosing cohorts."( A phase 1 study of weekly everolimus (RAD001) in combination with docetaxel in patients with metastatic breast cancer.
Booser, D; Brewster, A; Cristofanilli, M; Ensor, J; Giordano, SH; Gladish, G; Gonzalez-Angulo, AM; Hortobagyi, GN; Moore, J; Moulder, S; Murray, JL; Rivera, E; Tran, HT, 2012
)
0.88
"A dosage of 10 mg everolimus daily with TMZ 150 mg/m(2)/day for five consecutive days every 28 days in patients is the recommended dose for this regimen."( A phase I study of temozolomide and everolimus (RAD001) in patients with newly diagnosed and progressive glioblastoma either receiving or not receiving enzyme-inducing anticonvulsants: an NCIC CTG study.
Easaw, J; Eisenhauer, E; Kavan, P; Lwin, Z; Macdonald, D; Macneil, M; Mason, WP; McIntosh, L; Thiessen, B; Urva, S, 2012
)
0.99
" There have also been no reports on the increased risk of tuberculosis reactivation after an introduction of a PSI, and experience with everolimus dosing during antituberculosis treatment is very limited."( Pulmonary embolism and reactivation of tuberculosis during everolimus therapy in a kidney transplant recipient.
Fijałkowska-Morawska, JB; Jagodzińska, M; Nowicki, M,
)
0.58
"Taxanes remain first line chemotherapy in management of metastatic breast cancer and have a key role in epithelial ovarian cancer, with increasingly common use of weekly paclitaxel dosing regimens."( Paclitaxel resistance is associated with switch from apoptotic to autophagic cell death in MCF-7 breast cancer cells.
Ajabnoor, GM; Coley, HM; Crook, T, 2012
)
0.38
" This dosing regimen has an acceptable safety and tolerability profile and appears to have moderate the clinical activity in refractory tumors."( A phase I study of bevacizumab, everolimus and panitumumab in advanced solid tumors.
Alvarez-Secord, A; Anderson, EL; Bendell, JC; Blobe, GC; Gockerman, J; Hurwitz, HI; Meadows, KL; Morse, MA; Ready, NE; Riedel, RF; Starodub, AN; Uronis, HE; Vlahovic, G; Zafar, SY, 2012
)
0.66
"A reduction in CNI dosage may lead to a decrease in the occurrence of post-transplant malignancy."( Effect of immunosuppressive drugs on spontaneous DNA repair in human peripheral blood mononuclear cells.
Gafter, U; Gafter-Gvili, A; Herman-Edelstein, M; Malachi, T; Ori, Y; Rozen-Zvi, B; Zingerman, B, 2012
)
0.38
" We have developed a model of tumor growth dynamics utilizing serial measurements of the sum of the longest tumor diameters (SLD) from individual RECORD-1 patients to define the dose-response relationship of everolimus."( Dynamic tumor modeling of the dose-response relationship for everolimus in metastatic renal cell carcinoma using data from the phase 3 RECORD-1 trial.
Carter, AA; Chiparus, O; Hollaender, N; Kim, H; Motzer, RJ; Sarr, C; Stein, A; Wang, W, 2012
)
0.81
" This allows evaluation of how an actual dosing history and individual covariates impact on the observed drug effect, and offers the possibility of predicting clinical observations as a function of time."( Dynamic tumor modeling of the dose-response relationship for everolimus in metastatic renal cell carcinoma using data from the phase 3 RECORD-1 trial.
Carter, AA; Chiparus, O; Hollaender, N; Kim, H; Motzer, RJ; Sarr, C; Stein, A; Wang, W, 2012
)
0.62
"In this pharmacodynamic model of tumor response, everolimus more effectively shrinks target lesions in mRCC when dosed 10 mg daily versus 5 mg daily, although a 5-mg dose still shows an antitumor effect."( Dynamic tumor modeling of the dose-response relationship for everolimus in metastatic renal cell carcinoma using data from the phase 3 RECORD-1 trial.
Carter, AA; Chiparus, O; Hollaender, N; Kim, H; Motzer, RJ; Sarr, C; Stein, A; Wang, W, 2012
)
0.87
" Estimated oral clearances of IT decreased on average by 50%, requiring reduced dosing regimens."( Practical management of boceprevir and immunosuppressive therapy in liver transplant recipients with hepatitis C virus recurrence.
Antonini, TM; Barau, C; Bonhomme-Faivre, L; Coilly, A; Duclos-Vallée, JC; Furlan, V; Noël, C; Roche, B; Roque-Afonso, AM; Samuel, D; Taburet, AM, 2012
)
0.38
" However, the newer targeted anticancer therapies have different pharmacokinetic (PK) and dosing characteristics compared with traditional cytotoxic drugs, making it possible to estimate the steady-state drug exposure with a single trough-level measurement."( Evidence for therapeutic drug monitoring of targeted anticancer therapies.
Balakrishnar, B; Clements, A; Gao, B; Gurney, H; Wong, M; Yeap, S, 2012
)
0.38
"Patients with metastatic renal cell carcinoma refractory to vascular endothelial growth factor receptor-tyrosine kinase inhibitor received everolimus (10 mg once daily), with dosing interruption or modifications allowed for toxicity."( Safety of everolimus by treatment duration in patients with advanced renal cell cancer in an expanded access program.
Anak, O; Bahl, A; Bavbek, S; Bracarda, S; Grünwald, V; Karakiewicz, P; Kim, D; Ou, YC; Panneerselvam, A; Rha, SY; van den Eertwegh, AJ, 2013
)
0.99
" The aim of this article is to propose recommendations for the use of everolimus in lung transplant recipients, including indications, dosing schedules and the use of concomitant immunosuppression."( Recommendations on the use of everolimus in lung transplantation.
Borro, JM; Cifrian, JM; de la Torre, M; de Pablo, A; Laporta, R; Monforte, V; Román, A; Santos, F; Solé, A; Ussetti, P; Zurbano, F, 2013
)
0.91
" Everolimus was orally dosed at 10 mg daily."( A phase II study of bevacizumab and everolimus as treatment for refractory metastatic renal cell carcinoma.
Barbeau, S; Harshman, LC; McMillian, A; Srinivas, S, 2013
)
1.58
" In every case, we recommend to start the selected targeted agents at standard dosage and to pursue therapy as long as possible because the control of disease should be the primary endpoint for the management of mRCC."( Management of metastatic renal cell carcinoma progressed after sunitinib or another antiangiogenic treatment.
Cortesi, E; Iacovelli, R; Mezi, S; Naso, G; Palazzo, A; Pellegrino, D; Trenta, P, 2014
)
0.4
"The goal of this study was to assess the pharmacokinetics and safety of everolimus in healthy volunteers with normal hepatic function and patients with mild (Child-Pugh class A), moderate (Child-Pugh class B), and severe (Child-Pugh class C) hepatic impairment in otherwise good health to inform dosing in the clinical setting."( Effects of hepatic impairment on the pharmacokinetics of everolimus: a single-dose, open-label, parallel-group study.
Anak, O; Bouillaud, E; Kobalava, Z; Kunz, T; Paquet, T; Peveling-Oberhag, J; Sellami, D; Urva, S; Yong, WP; Zeuzem, S, 2013
)
0.87
" Pharmacokinetically-directed dosing protocols of everolimus and irinotecan were established and used to assess the in vivo antitumor effects of the agents."( Utilization of quantitative in vivo pharmacology approaches to assess combination effects of everolimus and irinotecan in mouse xenograft models of colorectal cancer.
Bradshaw-Pierce, EL; Eckhardt, SG; Gustafson, DL; Kulikowski, G; Merz, AL; Pitts, TM; Selby, H; Serkova, NJ; Weekes, CD, 2013
)
0.86
" Combination effects of everolimus and irinotecan were determined in CRC xenograft models using clinically-relevant dosing protocols."( Utilization of quantitative in vivo pharmacology approaches to assess combination effects of everolimus and irinotecan in mouse xenograft models of colorectal cancer.
Bradshaw-Pierce, EL; Eckhardt, SG; Gustafson, DL; Kulikowski, G; Merz, AL; Pitts, TM; Selby, H; Serkova, NJ; Weekes, CD, 2013
)
0.92
" Eighty rats were randomly divided into the sham-operated group (n = 20), injury group (n = 20), low dosage of everolimus group (n = 20), and high dosage of everolimus group (n = 20)."( Oral everolimus inhibits intimal proliferation in injured carotid artery in rats.
Feng, RS; Ning, HJ; Shen, DL; Wang, XF; Zhang, JY; Zhao, XY, 2013
)
1.12
" The effect depended on dosage and was associated with the reduction of phosphorylation of P70S6K and the eIF-4E expression level."( Oral everolimus inhibits intimal proliferation in injured carotid artery in rats.
Feng, RS; Ning, HJ; Shen, DL; Wang, XF; Zhang, JY; Zhao, XY, 2013
)
0.9
" Clinical awareness and early identification of such AEs by oncology nurses are essential to dosing (interruptions, reduction, and treatment discontinuation); quality of life; and, ultimately, patient outcomes."( Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trial.
Peterson, ME, 2013
)
0.61
"The mammalian target of rapamycin inhibitor everolimus (Zortress®, Certican®) was recently approved in the USA and a number of EU countries for use in combination with a reduced dosage of tacrolimus and corticosteroids for the prophylaxis of organ rejection in adult liver transplant recipients."( Everolimus: a guide to its use in liver transplantation.
Keating, GM; Lyseng-Williamson, KA, 2013
)
2.09
"In sequential phase II studies assessing two dosing schedules, patients with metastatic colorectal cancers refractory to bevacizumab-, fluoropyrimidine-, oxaliplatin-, and irinotecan-based regimens received everolimus 70 mg/wk (n = 99) or 10 mg/d (n = 100)."( Phase II study of everolimus in patients with metastatic colorectal adenocarcinoma previously treated with bevacizumab-, fluoropyrimidine-, oxaliplatin-, and irinotecan-based regimens.
Arrowsmith, ER; Bajetta, E; Del Prete, SA; Fuchs, CS; Hwang, J; Jin, J; Malek, K; Ng, K; Ryan, DP; Sedova, M; Sharma, S; Tabernero, J, 2013
)
0.91
" Recently, an inhibitor of mammalian target of rapamycin, everolimus (EVL), has emerged as an alternative immunosuppressant drug that may allow CNI dosage reduction and thereby spare renal function."( Everolimus-incorporated immunosuppressant strategy improves renal dysfunction while maintaining low rejection rates after heart transplantation in Japanese patients.
Endo, M; Hatano, M; Imamura, T; Inaba, T; Kagami, Y; Kato, N; Kinugawa, K; Komuro, I; Kyo, S; Maki, H; Minatsuki, S; Muraoka, H; Ono, M; Yao, A, 2013
)
2.08
" Tacrolimus is the basic immunosuppressant for patients after a liver transplant and thanks to its prolonged-release dosage form, which due to its simplicity and reliability of use, replaces tacrolimus twice daily early after the transplant and in the longterm administration, will apparently, for a while, defend its position."( [Immunosuppression after liver transplant, now and in future].
Trunečka, P, 2013
)
0.39
" Specifically, we investigated whether the antitumor effect of an mTOR inhibitor could be improved by changing its dosing schedule in RenCa tumor-bearing mice."( Circadian regulation of mTOR by the ubiquitin pathway in renal cell carcinoma.
Akagawa, Y; Fujioka, T; Koyanagi, S; Kuwano, M; Matsunaga, N; Ohdo, S; Okazaki, F; Okazaki, H; Ono, M; Tsurudome, Y, 2014
)
0.4
" Additionally, our results suggest that clinicians should administer the adequate dosage of EVE in order to increase efficacy and reduce adverse effects."( Everolimus-induced epithelial to mesenchymal transition in immortalized human renal proximal tubular epithelial cells: key role of heparanase.
Gambaro, G; Granata, S; Lupo, A; Masola, V; Onisto, M; Zaza, G, 2013
)
1.83
" The dosage of everolimus had to be increased and refractory status epilepticus followed after 12 days."( Everolimus for the treatment of subependymal giant cell astrocytoma probably causing seizure aggravation in a child with tuberous sclerosis complex: a case report.
Bast, T; Strobl, K; Wiemer-Kruel, A; Woerle, H, 2014
)
2.2
" Pharmacodynamic biomarkers could be useful in distinguishing between (1) disease resistance that occurs even though mTOR is successfully inhibited (suggesting a need for a different treatment strategy) and (2) resistance that might respond to changes in drug dosage or schedule."( Ribosomal S6 kinase and AKT phosphorylation as pharmacodynamic biomarkers in patients with myelodysplastic syndrome treated with RAD001.
Advani, AS; Bates, J; Kalaycio, M; Maciejewski, J; Mahfouz, RZ; Rybicki, L; Saunthararajah, Y; Sekeres, M; Tripp, B, 2014
)
0.4
"In patients with solid tumours, daily everolimus dosing demonstrated dose proportionality and linear pharmacokinetics."( Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology.
Anak, O; Cheung, WK; Grosch, K; Ravaud, A; Sellami, DB; Urva, SR, 2014
)
0.99
"Data on 2004 renal transplant recipients from three EVR studies were pooled to identify the impact of ethnicity on efficacy outcomes across EVR dosing groups and control groups."( Outcomes in ethnic minority renal transplant recipients receiving everolimus versus mycophenolate: comparative risk assessment results from a pooled analysis.
Delcoro, C; McCague, K; Melancon, K; Mulgaonkar, SP; Shihab, FS; Wiland, A, 2013
)
0.63
"Neutropenia after kidney transplant is an adverse event usually treated with a dosage reduction of mycophenolic acid."( Replacement of mycophenolate acid with everolimus in patients who became neutropenic after renal transplant.
Goumenos, DS; Kalliakmani, P; Karavias, D; Kazakopoulos, P; Marangos, M; Papachristou, E; Savvidaki, E; Voliotis, G; Zavvos, V, 2014
)
0.67
" One hundred eighteen episodes of neutropenia were recorded, originally treated by reducing the dosage of mycophenolic acid (765 ± 390 mg/d) and administering granulocyte colony-stimulating factor."( Replacement of mycophenolate acid with everolimus in patients who became neutropenic after renal transplant.
Goumenos, DS; Kalliakmani, P; Karavias, D; Kazakopoulos, P; Marangos, M; Papachristou, E; Savvidaki, E; Voliotis, G; Zavvos, V, 2014
)
0.67
" However, comparable data regarding clinical outcomes in HTx recipients receiving EVL either with dosage reduction of cyclosporine A (CSA) or with dosage reduction of tacrolimus (TAC) is scarce."( Clinical outcome in heart transplant recipients receiving everolimus in combination with dosage reduction of the calcineurin inhibitor cyclosporine A or tacrolimus.
Ensminger, SM; Fuchs, U; Gummert, JF; Schulz, U; Zittermann, A, 2014
)
0.65
"The pharmacokinetic characteristics of everolimus in Japanese de novo renal transplant patients did not differ from those previously observed in non-Japanese patients, hence the same dosage of everolimus may be acceptable in Japanese patients."( Pharmacokinetics of everolimus when combined with cyclosporine in Japanese de novo renal transplant recipients.
Goto, N; Hirano, M; Hoshinaga, K; Kusaka, M; Sasaki, H; Uchida, K; Watarai, Y, 2014
)
1
"From 2007 to 2012, six liver transplant recipients underwent nine major abdominal or thoracic surgical procedures without m-TORi discontinuation or specific dosage adjustment."( Preliminary report of major surgery in liver transplant recipients receiving m-TOR inhibitors without therapeutic discontinuation.
Bernard, D; Calmus, Y; Cauchy, F; Conti, F; Perdigao, F; Scatton, O; Schwarz, L; Sepulveda, A; Soubrane, O, 2014
)
0.4
"Major surgery in liver transplant recipients receiving m-TOR inhibitors appears both feasible and safe without therapeutic discontinuation or specific dosage adjustment."( Preliminary report of major surgery in liver transplant recipients receiving m-TOR inhibitors without therapeutic discontinuation.
Bernard, D; Calmus, Y; Cauchy, F; Conti, F; Perdigao, F; Scatton, O; Schwarz, L; Sepulveda, A; Soubrane, O, 2014
)
0.4
" Early clinical trials of mTOR inhibitors used fixed dosing with no concomitant reduction in calcineurin inhibitor (CNI) exposure, leading to concerns when consistent and marked dyslipidemia was observed."( mTOR inhibitors and dyslipidemia in transplant recipients: a cause for concern?
Holdaas, H; Potena, L; Saliba, F, 2015
)
0.42
" Patients were assigned to one of 10 different cohorts, involving either daily dosing with both agents or daily dosing with trametinib and intermittent everolimus dosing."( A phase IB trial of the oral MEK inhibitor trametinib (GSK1120212) in combination with everolimus in patients with advanced solid tumors.
Bellew, KM; Bendell, JC; Burris, HA; Cox, DS; Durante, M; Infante, JR; Jones, SF; Le, NT; Papadopoulos, KP; Park, J; Patnaik, A; Rasco, D; Tolcher, AW, 2015
)
0.84
" Additionally, a 3% annual discount rate was applied to costs, and the robustness of the model results was tested by conducting sensitivity analyses, including those on dosing scheme and post-progression treatment costs."( Lifetime cost of everolimus vs axitinib in patients with advanced renal cell carcinoma who failed prior sunitinib therapy in the US.
Casciano, R; Chua, A; Culver, K; Garrison, LP; Gorritz, M; Liu, Z; Pal, S; Perrin, A; Sherman, S; Wang, X, 2015
)
0.76
"Everolimus (EVE) is an immunosuppressive drug dosed according to therapeutic drug monitoring in renal transplant recipients."( Closer to the Site of Action: Everolimus Concentrations in Peripheral Blood Mononuclear Cells Correlate Well With Whole Blood Concentrations.
Åsberg, A; Christensen, H; Falck, P; Midtvedt, K; Robertsen, I; Vethe, NT, 2015
)
2.15
"Twelve renal transplant recipients (5 men and 7 women) treated with EVE underwent a pharmacokinetic investigation where EVE concentrations in whole blood and in peripheral blood mononuclear cells (PBMC) were determined within a dosing interval."( Closer to the Site of Action: Everolimus Concentrations in Peripheral Blood Mononuclear Cells Correlate Well With Whole Blood Concentrations.
Åsberg, A; Christensen, H; Falck, P; Midtvedt, K; Robertsen, I; Vethe, NT, 2015
)
0.71
" Phase I studies indicated 10 mg daily to be the best dosing of everolimus in DLBCL."( Everolimus in diffuse large B-cell lymphomas.
Arcaini, L; Ferrario, A; Maffioli, M; Merli, M; Passamonti, F, 2015
)
2.1
" Dosing decisions were based on the probability of experiencing a dose-limiting toxicity (DLT) during the first two 21-day treatment cycles."( Phase I study of everolimus, cetuximab and irinotecan as second-line therapy in metastatic colorectal cancer.
Beck, JT; Brandt, U; Davidson, SJ; Garrett, CR; Hecht, JR; Mackenzie, MJ; Reid, TR; Rizvi, S; Sharma, S, 2015
)
0.76
" Administration of RAD is capable of reducing the dosage of coadministrated calcineurin inhibitors (CNI)."( Histopathologic impacts of everolimus introduction on kidney transplant recipients.
Harada, S; Ito, T; Koshino, K; Nakamura, T; Nakao, T; Nobori, S; Suzuki, T; Takata, T; Ushigome, H; Yoshimura, N, 2015
)
0.71
" We explored a novel dosing schedule to assess safety, toxicity and activity in patients with advanced solid tumors."( Phase I combination of pazopanib and everolimus in PIK3CA mutation positive/PTEN loss patients with advanced solid tumors refractory to standard therapy.
Bellido, J; Falchook, G; Fu, S; Hong, D; Janku, F; Karp, D; Ke, D; Kurzrock, R; Lim, J; Naing, A; Piha-Paul, S; Rodrigues, HV; Stephen, B; Subbiah, V; Tsimberidou, A; Wheler, J; Zinner, R, 2015
)
0.69
" The objective of the model-based work would be the determination of the optimized doses and dosing schedules of everolimus and sorafenib, offering the maximization of the predicted modeled benefit/toxicity ratio in patients with solid tumors."( Multiparameter Phase I trials: a tool for model-based development of targeted agent combinations--example of EVESOR trial.
Barrois, C; Berger, F; Cassier, P; El-Madani, M; Freyer, G; Guitton, J; Hénin, E; Lachuer, J; Lefort, T; Rodriguez-Lafrasse, C; Slimane, K; Tod, M; Valette, PJ; You, B, 2015
)
0.63
" Similarly, SUL was able to reverse the hyper-expression of EMT markers induced by high EVE dosage (100 nM) in cells cultured under both normoxic and hypoxic conditions."( Sulodexide alone or in combination with low doses of everolimus inhibits the hypoxia-mediated epithelial to mesenchymal transition in human renal proximal tubular cells.
Bellin, G; Dalla Gassa, A; Gambaro, G; Granata, S; Lupo, A; Masola, V; Onisto, M; Zaza, G, 2015
)
0.67
"Our data reveal, for the first time, that sulodexide, alone or combined to low doses of everolimus, may hinder EMT in renal cells following hypoxia or minimize fibrotic complications due to high dosage of mammalian target of rapamycin inhibitors."( Sulodexide alone or in combination with low doses of everolimus inhibits the hypoxia-mediated epithelial to mesenchymal transition in human renal proximal tubular cells.
Bellin, G; Dalla Gassa, A; Gambaro, G; Granata, S; Lupo, A; Masola, V; Onisto, M; Zaza, G, 2015
)
0.89
" Coadministration of an mTOR inhibitor could permit lower dosing of chemotherapeutic agents in HCC management, and trials in non-transplant HCC population are exploring combined used with various agents including sorafenib, the vascular endothelial growth factor inhibitor bevacizumab and conventional agents."( mTOR inhibitor therapy: Does it prevent HCC recurrence after liver transplantation?
Duvoux, C; Toso, C, 2015
)
0.42
" Current immunosuppression protocols include a combination of calcineurin inhibitors, such as tacrolimus, and antiproliferative agents (most commonly mycophenolate mofetil), with or without different dosing regimens of corticosteroids."( Current State of Immunosuppression: Past, Present, and Future.
Karam, S; Wali, RK, 2015
)
0.42
" Therefore, the optimal dosage strategies for mTOR-I and CNI need to be further explored."( mTOR inhibitor versus mycophenolic acid as the primary immunosuppression regime combined with calcineurin inhibitor for kidney transplant recipients: a meta-analysis.
Chen, J; Jiang, Y; Lai, X; Shou, Z; Xiang, S; Xie, X, 2015
)
0.42
" The first 3 patients were enrolled onto a 4 + 2 dosing schedule of daily sunitinib 50 mg and weekly everolimus 30 mg."( A Single-Arm Phase 1b Study of Everolimus and Sunitinib in Patients With Advanced Renal Cell Carcinoma.
Armstrong, AJ; George, DJ; Kanesvaran, R; Turnbull, JD; Watt, K; Wolkowiez, MC, 2015
)
0.92
" With limited dosing regimens, the antiarthritic activity of EC0565 was found superior to that of etanercept, everolimus and a nontargeted everolimus analog."( Antiinflammatory Activity of a Novel Folic Acid Targeted Conjugate of the mTOR Inhibitor Everolimus.
Cross, VA; Gehrke, MA; Hahn, SJ; Klein, PJ; Kleindl, PJ; Leamon, CP; Low, PS; Lu, Y; Parker, N; Santhapuram, HK; Stinnette, TW; Vaughn, JF; Vlahov, IR; Wang, K; Westrick, E; Wollak, K; You, F, 2015
)
0.85
" The authors used a different dosage regime compared to literature and documented rapid tumor regression by 3D echocardiography."( Oral Everolimus for Treatment of a Giant Left Ventricular Rhabdomyoma in a Neonate-Rapid Tumor Regression Documented by Real Time 3D Echocardiography.
Daehnert, I; Hornemann, F; Riede, FT; Seki, H; Syrbe, S; Wagner, R; Weidenbach, M, 2015
)
0.93
" Thus, although CNIs remain the major immunosuppressive treatment, their dosage should be minimized by using them with either full-dose MPA or reduced-dose EVR."( The safety of calcineurin inhibitors for kidney-transplant patients.
Malvezzi, P; Rostaing, L, 2015
)
0.42
" However, ERL dosing is challenging due to its narrow therapeutic window combined with high interindividual pharmacokinetic variability."( Influence of CYP3A5 genetic variation on everolimus maintenance dosing after cardiac transplantation.
Englberger, L; Fiedler, GM; Largiadèr, CR; Lesche, D; Mohacsi, P; Setoud, R; Sigurdardottir, V; Sistonen, J, 2015
)
0.68
"To describe dosing patterns and to compare the drug costs per month spent in progression-free survival (PFS) among patients with advanced renal cell carcinoma (aRCC) treated with everolimus or axitinib following a first tyrosine kinase inhibitor (TKI)."( Real-world dosing and drug costs with everolimus or axitinib as second targeted therapies for advanced renal cell carcinoma: a retrospective chart review in the US.
Jonasch, E; Li, N; Liu, Z; Pal, SK; Perez, JR; Reichmann, WM; Signorovitch, JE; Vogelzang, NJ, 2016
)
0.9
" Real-world dosing patterns were summarized."( Real-world dosing and drug costs with everolimus or axitinib as second targeted therapies for advanced renal cell carcinoma: a retrospective chart review in the US.
Jonasch, E; Li, N; Liu, Z; Pal, SK; Perez, JR; Reichmann, WM; Signorovitch, JE; Vogelzang, NJ, 2016
)
0.71
" Close monitoring of the everolimus concentrations and corresponding dosage adjustments are necessary until the target levels are achieved during both periods."( Management of Everolimus and Voriconazole Interaction in Lung Transplant Patients.
Elberdín Pazos, L; Martín Herranz, MI; Outeda Macías, M; Salvador Garrido, P, 2016
)
1.1
" Using total DNA content as a measure of cell number, growth inhibitory dose-response curves of everolimus were determined at the end of resistance induction and over time after everolimus withdrawal."( Long-term acquired everolimus resistance in pancreatic neuroendocrine tumours can be overcome with novel PI3K-AKT-mTOR inhibitors.
Beyens, M; de Beeck, KO; de Herder, W; Dogan, F; Hofland, LJ; Mortier, G; Pauwels, P; Peeters, M; Van Camp, G; van Koetsveld, PM; Vandamme, T; Vangestel, C, 2016
)
0.98
" We analyzed the impact of orally dosed mammalian target of rapamycin inhibitors on abdominal adhesion formation and wound tensile strength in an experimental model."( Effects of mTOR Inhibitors in Prevention of Abdominal Adhesions.
Alwayn, I; Cimen, S; Guler, S; Hart-Matyas, M; Hu, Q; Venkatachalam, AB, 2016
)
0.43
" Dosing patterns and adherence to everolimus were summarized."( Treatment patterns and factors associated with the use of everolimus among post-menopausal women with HR+/HER2- metastatic breast cancer: a retrospective US claims study.
Fang, A; Guérin, A; Hao, Y; Kageleiry, A; Koo, V; Li, N; Peeples, M; Tang, D; Wu, EQ, 2016
)
0.96
" Dosing algorithms are provided, with suggestions for target ranges in specific settings, and treatment strategies for the most common side effects are proposed."( Use of everolimus in liver transplantation: The French experience.
Calmus, Y; Dharancy, S; Dumortier, J; Durand, F; Duvoux, C; Salamé, E; Saliba, F, 2016
)
0.89
" Everolimus was reinitiated in both cases in addition to decreasing the dosage of tacrolimus, and there were no further complications."( Use of Everolimus After Multivisceral Transplantation: A Report of Two Cases.
Jafri, SM; Kazimi, M; Parekh, R; Rao, B; Raoufi, M; Segovia, MC, 2016
)
1.8
" Our findings confirm observations from another study in patients with cancer and show us that everolimus is a good candidate for individualized dosing in patients with cancer."( Everolimus pharmacokinetics and its exposure-toxicity relationship in patients with thyroid cancer.
de Wit, D; den Hartigh, J; Gelderblom, H; Guchelaar, HJ; Kapiteijn, E; Links, TP; Moes, DJ; Roozen, CF; Schneider, TC; van der Hoeven, JJ; van Erp, NP, 2016
)
2.1
"Everolimus (EVR) has been used widely for the purpose of reducing the dosage of calcineurin inhibitor (CNI), leading to decreasing CNI nephrotoxicity."( Effectiveness of the Combination of Everolimus and Tacrolimus With High Dosage of Mizoribine for Living Donor-Related Kidney Transplantation.
Harada, S; Ito, T; Koshino, K; Nakamura, T; Nakao, T; Nobori, S; Suzuki, T; Ushigome, H; Yoshimura, N, 2016
)
2.15
" Further trials addressing the optimal dosage of imatinib and mTORi in kidney transplant recipients are recommended."( Gastrointestinal stromal tumors in kidney transplant recipients: Report of two cases and literature review.
Chan, CK; Chau, KF; Cheng, IK; Cheung, CY; Li, FK; Lo, SH, 2017
)
0.46
"Due to toxic events, everolimus dosage was reduced to 5 mg in 27% of patients."( Safety analysis, association with response and previous treatments of everolimus and exemestane in 181 metastatic breast cancer patients: A multicenter Italian experience.
Cortesi, E; D'Onofrio, L; Fabbri, MA; Gamucci, T; Giuliani, R; Iezzi, L; Magri, V; Mancini, ML; Marchetti, P; Mauri, M; Mentuccia, L; Moscetti, L; Natoli, C; Pizzuti, L; Ramponi, S; Roma, CL; Ruggeri, EM; Santini, D; Sini, V; Sperduti, I; Vaccaro, A; Vici, P, 2016
)
0.99
" Several recent clinical trials have demonstrated their efficacy in combination with reduced cyclosporine dosing in de novo heart transplant recipients, in particular with everolimus."( Recent Advances in Mammalian Target of Rapamycin Inhibitor Use in Heart and Lung Transplantation.
Fine, NM; Kushwaha, SS, 2016
)
0.63
" Second, the question of identifying an optimal dosing strategy for treating with an AI and a cytotoxic agent is addressed."( Microenvironment-Mediated Modeling of Tumor Response to Vascular-Targeting Drugs.
Gevertz, JL, 2016
)
0.43
" In nearly half of orally dosed oncolytics poor dissolution is likely to play a major role in low and unpredictable absorption."( Inventory of oral anticancer agents: Pharmaceutical formulation aspects with focus on the solid dispersion technique.
Beijnen, JH; Nuijen, B; Sawicki, E; Schellens, JH, 2016
)
0.43
"About 26 patients with solid tumors were treated in four different dosing schedules."( EVESOR, a model-based, multiparameter, Phase I trial to optimize the benefit/toxicity ratio of everolimus and sorafenib.
Badary, OA; Barrois, C; Cassier, P; Colomban, O; El-Demerdash, E; El-Madani, M; El-Shenawy, SM; Freyer, G; Gagnieu, MC; Guitton, J; Hommel-Fontaine, J; Ibrahim, BM; Lefort, T; Maillet, D; Peron, J; Rodriguez-Lafrasse, C; Tod, M; Valette, PJ; You, B, 2017
)
0.67
" Calcineurin inhibitor (CNI) dosage was reduced and renal function improved, and mean estimated glomerular filtration rate recovered from 42."( Experience of Quatro-Therapy With Everolimus to Minimize Calcineurin Inhibitor for Kidney Transplant Recipients.
Furuya, R; Irie, S; Matsuoka, H; Matsuzaki, H; Miyajima, S; Miyazaki, T; Nakamura, N; Tanaka, M,
)
0.41
" Key aspects of everolimus dosing and safety profile, drawn up by relevant findings, are included, as well as the role of biomarkers to identify subgroups of mBC patients who may best benefit from everolimus treatment."( Role of everolimus in the treatment of metastatic HER2-negative/HR-positive breast cancer.
Pronzato, P, 2017
)
1.24
" There is tendency not to interrupt the EVE treatment and keep it either in a dosage of 10 or 5mg/day if the oral damage is tolerable."( [Everolimus in Daily Clinical Practice Focusing to Oral Mucosa Damage Issues - Single Oncology Centre Experience within the Course of the Year 2016].
Arnetová, V; Fiala, O; Fínek, J; Kališová, K; Korunková, H; Kulhánková, J; Matějka, VM; Svoboda, T; Vokurka, S; Votavová, M,
)
1.04
" Within five post-operative days, patients (mean age 50±13 years, 27% women) were randomized to EVR or a standard CsA dosage (CsA group)."( Effect of everolimus vs calcineurin inhibitors on quality of life in heart transplant recipients during a 3-year follow-up: Results of a randomized controlled trial (SCHEDULE).
Andreassen, AK; Arora, S; Dellgren, G; Eiskjaer, H; Grov, I; Gude, E; Gullestad, L; Gustafsson, F; Jansson, K; Karason, K; Rådegran, G; Relbo Authen, A; Solbu, D, 2017
)
0.86
" Therefore, with regard to PI3K inhibitors currently in late-stage clinical trials, the identification of appropriate biomarkers of efficacy and the development of optimal combination regimens and dosing schedules are likely to be important for graduation into clinical practice."( Phosphoinositide 3-kinase (PI3K) pathway inhibitors in solid tumors: From laboratory to patients.
Janku, F, 2017
)
0.46
" This beneficial treatment option for patients can minimize the drug dosage and the occurrence of adverse events."( Intermittent everolimus administration for renal angiomyolipoma associated with tuberous sclerosis complex.
Egawa, S; Endo, K; Hatano, T; Inaba, H, 2017
)
0.82
" The CsA dosage was further reduced to keep a lowest value of serum creatinine and a target EVL level."( Ten-Year Follow-up of Pharmacokinetics-Guided Very Early Cyclosporine Minimization Synchronized With Everolimus Initiation in De Novo Kidney Transplantation.
Jirasiritham, S; Sumethkul, V; Tankee, P; Worawichawong, S, 2017
)
0.67
" We created dose-response curves for both compounds."( Preclinical analysis of MTOR complex 1/2 inhibition in diffuse intrinsic pontine glioma.
Amani, V; DeSisto, JA; Donson, A; Flannery, PC; Foreman, N; Green, AL; Hoffman, L; Lemma, RT; Levy, JMM; Moroze, EE; Prince, EW; Venkataraman, S; Vibhakar, R, 2018
)
0.48
" Limits of clinical relevance were set at a difference of ± 25%, as this would lead to a different dosing advice."( Clinical validation study of dried blood spot for determining everolimus concentration in patients with cancer.
Brüggemann, RJM; Croes, S; de Beer, YM; Knapen, LM; van Erp, NP; van Herpen, CML; Willemsen, AECAB, 2018
)
0.72
" However, toxicity with flat dosing limits its usage."( Pharmacokinetically-targeted dosed everolimus maintenance therapy in lymphoma patients.
Ambinder, RF; Asiama, A; Gladstone, DE; Hurtt, J; Jones, RJ; Sawyer, K; Schoch, LK; Shanbhag, S; Swinnen, LJ; Wagner-Johnston, N; Zahurak, M, 2018
)
0.76
"After HDC, everolimus was dosed to serum trough levels (goal 3-15 ng/mL), with quarterly rituximab infusions for 1 year while maintaining < grade II non-hematologic and < grade III hematologic toxicities."( Pharmacokinetically-targeted dosed everolimus maintenance therapy in lymphoma patients.
Ambinder, RF; Asiama, A; Gladstone, DE; Hurtt, J; Jones, RJ; Sawyer, K; Schoch, LK; Shanbhag, S; Swinnen, LJ; Wagner-Johnston, N; Zahurak, M, 2018
)
1.15
"Pharmacokinetically-targeted dosing appears to increase everolimus tolerability."( Pharmacokinetically-targeted dosed everolimus maintenance therapy in lymphoma patients.
Ambinder, RF; Asiama, A; Gladstone, DE; Hurtt, J; Jones, RJ; Sawyer, K; Schoch, LK; Shanbhag, S; Swinnen, LJ; Wagner-Johnston, N; Zahurak, M, 2018
)
1
"Patients with advanced pNET were randomized (1:1) to oral BEZ235 400 mg twice daily or oral everolimus 10 mg once daily on a continuous dosing schedule."( Phase II Study of BEZ235 versus Everolimus in Patients with Mammalian Target of Rapamycin Inhibitor-Naïve Advanced Pancreatic Neuroendocrine Tumors.
Capdevila, J; Custodio, A; Garcia-Carbonero, R; Grande, E; Guimbaud, R; Hendifar, AE; Klümpen, HJ; Kong, O; Libutti, SK; Lombard-Bohas, C; Meyer, T; Reed, N; Ricci, S; Safina, S; Salazar, R; Salomon, H; Tavorath, R; Walenkamp, A; Yao, JC, 2018
)
0.98
" Everolimus toxicity was less severe following dosing at ZT13, as compared to ZT1, as shown with least body weight inhibition in both genders; least reductions in thymus weight both in males (p < 0."( The immune system as a chronotoxicity target of the anticancer mTOR inhibitor everolimus.
Cinar, S; Deniz, G; Giacchetti, S; Kaptan, E; Lévi, F; Li, XM; Okyar, A; Ozsoy, N; Ozturk, D; Ozturk, N; Pala-Kara, Z; Sancar-Bas, S, 2018
)
1.62
" We postulate that there is room for improvement of dosing, as the optimal immunosuppressive dose in calcineurin-free regimens is unknown and since the once daily dosing regimen for oncological indications is often associated with treatment-limiting toxicity."( A pharmacological rationale for improved everolimus dosing in oncology and transplant patients.
Burger, DM; de Fijter, JW; Guchelaar, HJ; Moes, DJAR; Reinders, MEJ; Ter Heine, R; van Erp, NP; van Herpen, CM, 2018
)
0.75
"We developed a mechanistic population pharmacokinetic model for everolimus in cancer and transplant patients and explored alternative dosing regimens."( A pharmacological rationale for improved everolimus dosing in oncology and transplant patients.
Burger, DM; de Fijter, JW; Guchelaar, HJ; Moes, DJAR; Reinders, MEJ; Ter Heine, R; van Erp, NP; van Herpen, CM, 2018
)
0.99
" Waste occurred in 41% of all patients with primary reasons attributed to cancer progression in 25 patients, death in five patients, toxicity in five patients and increase in dosage of targeted therapy in two patients."( Financial impact of oral chemotherapy wastage on society and the patient.
Clamon, G; Meyer, C; Monga, V; Vakiner, B, 2019
)
0.51
" Dosing and administration inputs were consistent with approved prescribing information and the clinical trials used to estimate efficacy and safety inputs."( The Additional Costs per Month of Progression-Free Survival and Overall Survival: An Economic Model Comparing Everolimus with Cabozantinib, Nivolumab, and Axitinib for Second-Line Treatment of Metastatic Renal Cell Carcinoma.
Duchesneau, E; Ghate, S; McDonald, E; Messali, A; Perez, JR; Swallow, E, 2018
)
0.69
"This study included two cohorts; cohort 1 received 30 mg of everolimus by mouth (PO) weekly, and cohort 2 was dosed with 10 mg of everolimus PO daily."( Phase II Study of Everolimus in Metastatic Malignant Melanoma (NCCTG-N0377, Alliance).
Allred, JB; Creagan, ET; Erickson, LA; Kaur, JS; Lowe, VJ; Maples, WJ; Markovic, S; Rao, RD; Suman, VJ; Vera Aguilera, J; Windschitl, HE, 2018
)
1.06
"Key inputs included in the calculation were patient population, dosing information, incidence rates and associated costs of Grade 3/4 AEs, treatment costs (including drug discount programs), and patients' progression-free survival (PFS) as a proxy for length of treatment."( Cost analysis for different sequential treatment regimens for metastatic renal cell carcinoma in China.
Dong, P; Gao, X; Lu, X; Park, SH; Ren, H; Shi, G; Xue, M, 2018
)
0.48
" This study was, therefore, designed to determine the optimal dosage and schedule of CTX when combined with everolimus to prevent this potentially detrimental Treg expansion."( Phase 1 study of everolimus and low-dose oral cyclophosphamide in patients with metastatic renal cell carcinoma.
de Gruijl, TD; Goedegebuure, RS; Haanen, JB; Hamberg, P; Huijts, CM; Lougheed, SM; Tascilar, M; van der Vliet, HJ; van Herpen, CM; Verheul, HM; Werter, IM, 2019
)
1.07
" After dosing was discontinued, TAC-treated mice showed gradual graft rejection, whereas EVL-treated mice sustained long-term robust chimerism."( Evaluation of the impact of conventional immunosuppressant on the establishment of murine transplantation tolerance - an experimental study.
Fukuda, H; Hirai, T; Ikemiyagi, M; Ishii, R; Ishii, Y; Kanzawa, T; Katsumata, H; Miyairi, S; Okumi, M; Omoto, K; Saiga, K; Tanabe, K; Yokoo, T, 2019
)
0.51
"In a multicenter phase 2 study, performed in patients with mRCC not amenable to or progressive on a vascular endothelial growth factor (VEGF)-receptor tyrosine kinase inhibitor (TKI) containing treatment regimen, we assessed whether the addition of this metronomic dosing schedule of cyclophosphamide to therapy with everolimus could result in an improvement of progression-free survival (PFS) after 4 months of treatment."( Metronomic cyclophosphamide attenuates mTOR-mediated expansion of regulatory T cells, but does not impact clinical outcome in patients with metastatic renal cell cancer treated with everolimus.
de Gruijl, TD; Haanen, JBAG; Hamberg, P; Helgason, HH; Huijts, CM; Los, M; Lougheed, SM; Polee, MB; Tascilar, M; van der Vliet, HJ; Verheul, HM; Werter, IM, 2019
)
0.88
" Continuous administration of everolimus at a low dose as opposed to high intermittent dosing schedule has greater antitumor efficacy against thyroid cancer xenografts in vivo."( Evaluation of preclinical efficacy of everolimus and pasireotide in thyroid cancer cell lines and xenograft models.
Chen, Z; Khuri, FR; Lallani, SB; Lonial, S; Marcus, A; Martinson, DE; Owonikoko, TK; Sun, SY; Zhang, G, 2019
)
1.07
" Substituting leflunomide for mycophenolate sodium and increasing dosage of everolimus has been proposed to solve BK nephropathy."( Successful Treatment for BK Virus Nephropathy by Leflunomide in a Kidney Transplant Patient: A Case Report.
Chen, HA; Chen, YW; Hsu, YH; Hung, LY; Lee, CH; Wu, MS; Wu, MY, 2019
)
0.74
" The aim of this study is to characterize the interaction, optimal sequence and dosing of SSA-based and molecularly targeted therapies in PanNET."( Interaction between somatostatin analogues and targeted therapies in neuroendocrine tumor cells.
Egidi, M; Gress, TM; Krug, S; Michl, P; Mordhorst, JP; Moser, F; Rinke, A; Ruffert, C; Theuerkorn, K, 2019
)
0.51
" After 3 months, CNIs were discontinued in patients who tolerated everolimus well, while everolimus dosage was increased (blood trough levels, 6-12 ng/ml)."( Immunosuppressive treatment with everolimus in patients after liver transplant: 4 years of single-center experience.
Grąt, M; Janik, M; Moczydłowska, D; Raszeja-Wyszomirska, J; Wasilewicz, MP; Zieniewicz, K, 2019
)
1.03
" Everolimus 5 mg PO daily and panobinostat 10 mg PO 3 times weekly (weeks 1 and 2) given in 21-day cycles was the recommended phase II dosing based on their maximum tolerated dose."( Phase I study of the mTOR inhibitor everolimus in combination with the histone deacetylase inhibitor panobinostat in patients with advanced clear cell renal cell carcinoma.
Adra, N; Chintala, S; Damayanti, N; George, S; Pili, R; Wood, A, 2020
)
1.74
"All randomised controlled trials (RCTs) and quasi-RCTs in which drug regimens, containing TOR-I commenced within seven days of transplant, were compared to alternative drug regimens, were included without age restriction, dosage or language of report."( Target of rapamycin inhibitors (TOR-I; sirolimus and everolimus) for primary immunosuppression in kidney transplant recipients.
Chapman, JR; Craig, JC; Hahn, D; Hamiwka, LA; Hodson, EM; Lee, VW; Webster, AC, 2019
)
0.76
" Post-HTx patient survival in patients, irrespective of the donor risk factors such as donor age, low LVEF, history of cardiac arrest, was equivalent across cohorts, while longer TIT and higher inotrope dosage prior to procurement surgery were significant risk factors for survival."( Effect of Therapeutic Modification on Outcomes in Heart Transplantation Over the Past Two Decades - A Single-Center Experience in Japan.
Fujita, T; Fukushima, N; Fukushima, S; Ikura, M; Ishibashi-Ueda, H; Iwasaki, K; Kitamura, S; Kobayashi, J; Kuroda, K; Matsuda, S; Mochizuki, H; Nakajima, S; Nakatani, T; Ogawa, H; Seguchi, O; Tadokoro, N; Takenaka, H; Tsujita, K; Watanabe, T; Yanase, M, 2020
)
0.56
" In 21 patients a non-osseous biopsy obtained before dosing was useful for continuous scoring, which demonstrated upregulation of several proteins as compared to readings in corresponding primary tumour tissues."( PI3K pathway protein analyses in metastatic breast cancer patients receiving standard everolimus and exemestane.
Beelen, KJ; Boven, E; de Valk, B; Kruger, DT; Linn, SC; Nieuwenhuis, M; Opdam, M; Sanders, J; van der Noort, V, 2020
)
0.78
" All patients received everolimus at a dosage of 10 mg/day alone or in association with exemesthane for breast cancer."( [Tolerablity of everolimus in clinical practice: a retrospective study].
Afani, L; Awada, A; Belbaraka, R, 2020
)
1.21
"The EVR-loaded sublingual orodispersible films are a promising, economical, and convenient approach for delivering EVR efficiently in a solid dosage form."( Designing orodispersible films containing everolimus for enhanced compliance and bioavailability.
Gao, S; Guan, R; Liu, H; Liu, Y; Ma, Y; Song, W; Yang, Y, 2020
)
0.82
" Discontinuation or dosage reduction of immunosuppressives and other treatment information was documented."( Clinical course of COVID-19 disease in immunosuppressed renal transplant patients
Ateş, ÖF; Çetin, ES; Dheir, H; Fırat, N; Genç, AB; Karabay, O; Köroğlu, M; Muratdağı, G; Özmen, K; Sipahi, S; Tomak, Y; Yaylacı, S, 2021
)
0.62
" Herein, we examined the capacity of single oral dosing with everolimus, an FDA-approved inhibitor of the PI3K/Akt effector mTOR, to reduce incubated cocaine-craving and reverse incubation-associated changes in vmPFC kinase activity and mGlu expression."( Preclinical evidence to support repurposing everolimus for craving reduction during protracted drug withdrawal.
Barger, BD; Chiu, AS; Elias, KN; Fabella, AM; Holder, KN; Huerta Sanchez, LL; Jimenez Chavez, CL; Kang, MC; Kippin, TE; Shin, CB; Szumlinski, KK, 2021
)
1.12
"Saliva and whole blood samples were taken from patients with cancer, who were treated with everolimus in the dosage of either 10 mg once a day or 5 mg twice a day."( Everolimus Concentration in Saliva to Predict Stomatitis: A Feasibility Study in Patients with Cancer.
Beijnen, JH; Dorlo, TPC; Huitema, ADR; Jacobs, BAW; Molenaar-Kuijsten, L; Rosing, H; Steeghs, N; Thijssen, B; Verheijen, RB, 2022
)
2.38
"The study findings support the approved dosing regimen of lenvatinib 18 mg plus everolimus 5 mg daily for patients with advanced RCC."( Assessing the Safety and Efficacy of Two Starting Doses of Lenvatinib Plus Everolimus in Patients with Renal Cell Carcinoma: A Randomized Phase 2 Trial.
Alekseev, B; Binder, TA; Castellano, D; Ciuleanu, T; Glen, H; Heng, DYC; Koralewski, P; Lee, JL; O'Hara, K; Pal, SK; Parnis, F; Peng, L; Puente, J; Rha, SY; Smith, AD; Stroyakovskiy, D; Sunela, K, 2022
)
1.18
" The average dosage of everolimus and sirolimus was 2,65mg (±1,78) and 2,75mg (±0,96) in T patients, respectively, versus 8,75mg (±2,26) for everolimus in K patients."( Pulmonary toxicity of mTOR inhibitors. Comparisons of two populations: Solid organ recipients and cancer patients.
Billaud, EM; Gendarme, S; Gibault, L; Guillemain, R; Israël-Biet, D; Lillo-Lelouet, A; Medioni, J; Oudard, S; Pastré, J,
)
0.44
"The management of immunosuppressors in solid organ transplantation requires pharmacological therapeutic monitoring with regular adaptation of the dosage to the residual level."( Sotorasib associated with tacrolimus and everolimus: A significant drug interaction in lung transplant patients.
Bedouch, P; Briault, A; Degano, B; Falque, L; Liaigre, L; Orhon, P; Perrier, Q; Raymond, CS; Romand, P, 2022
)
0.99
" All drugs were dosed once daily on days 1-21 of each 28-day cycle."( DTRMWXHS-12, a novel Bruton tyrosine kinase inhibitor, in combination with everolimus and pomalidomide in patients with relapsed/refractory lymphomas: An open-label, multicenter, phase 1a/1b study.
Brander, DM; Ding, W; Gui, M; He, W; Huntington, SF; Iqbal, M; Kearney, AS; Koehler, AB; Leis, JF; Mato, AR; McKinlay, TP; Moustafa, MA; Rosenthal, AC; Schuster, SJ; Tun, HW, 2023
)
1.14
" The academic EVESOR trial (NCT01932177) was designed to assess alternative doses and intermittent dosing schedules of EVE and SOR combination therapy to improve the benefit-risk ratio for patients with solid tumors."( Clinical results of the EVESOR trial, a multiparameter phase I trial of everolimus and sorafenib combination in solid tumors.
Augu-Denechere, D; Bonnin, N; Calattini, S; Colomban, O; Fontaine, J; Freyer, G; Guitton, J; Lopez, J; Maillet, D; Payen, L; Peron, J; Puszkiel, A; Schwiertz, V; Tartas, S; Tod, M; Varnier, R; You, B, 2023
)
1.14
"EVESOR is a multiparameter dose-escalation phase I trial investigating different doses and dosing schedules, with the final objective of generating data for modeling and simulation."( Clinical results of the EVESOR trial, a multiparameter phase I trial of everolimus and sorafenib combination in solid tumors.
Augu-Denechere, D; Bonnin, N; Calattini, S; Colomban, O; Fontaine, J; Freyer, G; Guitton, J; Lopez, J; Maillet, D; Payen, L; Peron, J; Puszkiel, A; Schwiertz, V; Tartas, S; Tod, M; Varnier, R; You, B, 2023
)
1.14
"The objective was to develop a pharmacokinetic-pharmacodynamic (PK-PD) model linking everolimus and sorafenib exposure with biomarker dynamics and progression-free survival (PFS) based on data from EVESOR trial in patients with solid tumors treated with everolimus and sorafenib combination therapy and to simulate alternative dosing schedules for sorafenib."( A PK-PD model linking biomarker dynamics to progression-free survival in patients treated with everolimus and sorafenib combination therapy, EVESOR phase I trial.
Augu-Denechere, D; Bonnin, N; Colomban, O; Fontaine, J; Freyer, G; Guitton, J; Lopez, J; Maillet, D; Payen, L; Péron, J; Puszkiel, A; Rousset, P; Tartas, S; Tod, M; Trillet-Lenoir, V; You, B, 2023
)
1.35
"Everolimus (5-10 mg once daily, qd) and sorafenib (200-400 mg twice daily, bid) were administered according to four different dosing schedules in 43 solid tumor patients."( A PK-PD model linking biomarker dynamics to progression-free survival in patients treated with everolimus and sorafenib combination therapy, EVESOR phase I trial.
Augu-Denechere, D; Bonnin, N; Colomban, O; Fontaine, J; Freyer, G; Guitton, J; Lopez, J; Maillet, D; Payen, L; Péron, J; Puszkiel, A; Rousset, P; Tartas, S; Tod, M; Trillet-Lenoir, V; You, B, 2023
)
2.57
" Nonlinear pharmacokinetics (PKs) were observed in clinical studies after intravenous and oral dosing across a wide dose range, including the efficacious doses of 240 and 480 mg."( Developing a mechanistic understanding of the nonlinear pharmacokinetics of letermovir and prospective drug interaction with everolimus using physiological-based pharmacokinetic modeling.
Asari, K; Chen, D; Cho, CR; Hartmann, G; Kuo, Y; McCrea, JB; Menzel, K; Wang, YH, 2023
)
1.12
" Maximum concentration and area under the concentration-time curve (AUC) from time of dosing to the last measurable concentration and extrapolated to infinity, of everolimus in whole blood were estimated using noncompartmental analysis, with geometric mean ratios and 90% confidence intervals for the ratios of everolimus dosed with CBD to everolimus dosed alone."( Pharmacokinetic Drug-Drug Interaction With Coadministration of Cannabidiol and Everolimus in a Phase 1 Healthy Volunteer Trial.
Berwaerts, J; Chen, C; Critchley, D; Hyland, K; Tayo, B; Thai, C; Wray, L, 2023
)
1.33
"Heart transplant recipients experience high rates of skin cancer, likely due to greater length or dosage of immunosuppression."( A review of heart transplant immunosuppressants and nonmelanoma skin cancer.
Camacho, I; Dave, Y; Eckembrecher, DG; Eckembrecher, FJ; Nouri, K; Patel, S; Shah, H, 2023
)
0.91
" Dose-response assays were used to confirm the antifungal activity of initial hits and establish their potency."( Screening the medicine for malaria venture's Pandemic Response Box to identify novel inhibitors of Candida albicans and Candida auris biofilm formation.
Ajetunmobi, OH; Badali, H; Chaturvedi, AK; Lopez-Ribot, JL; Najvar, L; Patterson, TF; Vaccaro, A; Wiederhold, NP; Wormley, FL, 2023
)
0.91
"3) and no difference with regard to dosing was observed (p = ."( Does the dose matter? Antiproliferative efficacy and toxicity of everolimus in patients with neuroendocrine tumors - Experiences from a tertiary referral center.
Haug, A; Kiesewetter, B; Kretschmer-Chott, E; Macheiner, S; Mazal, P; Melhorn, P; Raderer, M; Wolff, L, 2023
)
1.15
" We ascertained whether the risk of drug-induced tremor is influenced by the underlying diagnosis, dosing formulations, drug concentrations, and blood monitoring."( Tremor Induced by Cyclosporine, Tacrolimus, Sirolimus, or Everolimus: A Review of the Literature.
Jha, N; Joyce, M; Khalid, U; Lunny, C; Mahboob, O; Nagaraja, N; Shukla, AM; Wagle Shukla, A, 2023
)
1.15
"The development and analysis of pharmaceutical formulations often involves the determination of multiple active ingredients in a dosage form."( Development and Method Validation of Design of Experiments-Optimized Tablet Formulation for Simultaneous Detection of Exemestane and Everolimus.
Kumar, A; Kurmi, BD; Singh, D,
)
0.34
" Our data might be useful for determining the dosage of EVE in clinical practice."( Relationship Between Daily Dose of Everolimus and Treatment Effect in Patients With Luminal HER2-negative Metastatic Breast Cancer.
Horimoto, Y; Ishizuka, Y; Murakami, F; Okazaki, M; Ushiyama, Y; Watanabe, J, 2023
)
1.19
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (5)

RoleDescription
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
immunosuppressive agentAn agent that suppresses immune function by one of several mechanisms of action. Classical cytotoxic immunosuppressants act by inhibiting DNA synthesis. Others may act through activation of T-cells or by inhibiting the activation of helper cells. In addition, an immunosuppressive agent is a role played by a compound which is exhibited by a capability to diminish the extent and/or voracity of an immune response.
mTOR inhibitorA protein kinase inhibitor of the mammalian target of rapamycin (mTOR), a protein that regulates cell growth, cell proliferation, cell motility, cell survival, protein synthesis and transcription. mTOR inhibitors are used to prevent transplant rejection and in treatment of cancer.
anticoronaviral agentAny antiviral agent which inhibits the activity of coronaviruses.
geroprotectorAny compound that supports healthy aging, slows the biological aging process, or extends lifespan.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (6)

ClassDescription
primary alcoholA primary alcohol is a compound in which a hydroxy group, -OH, is attached to a saturated carbon atom which has either three hydrogen atoms attached to it or only one other carbon atom and two hydrogen atoms attached to it.
secondary alcoholA secondary alcohol is a compound in which a hydroxy group, -OH, is attached to a saturated carbon atom which has two other carbon atoms attached to it.
etherAn organooxygen compound with formula ROR, where R is not hydrogen.
cyclic ketone
cyclic acetalAn acetal in the molecule of which the acetal carbon and one or both oxygen atoms thereon are members of a ring.
macrolide lactamA macrolide in which the macrocyclic lactone ring includes an amide group.
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Protein Targets (317)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
USP1 protein, partialHomo sapiens (human)Potency39.81070.031637.5844354.8130AID504865
TDP1 proteinHomo sapiens (human)Potency0.03440.000811.382244.6684AID686978; AID686979
AR proteinHomo sapiens (human)Potency20.43770.000221.22318,912.5098AID743035; AID743036; AID743042; AID743053; AID743054; AID743063
glucocorticoid receptor [Homo sapiens]Homo sapiens (human)Potency14.43810.000214.376460.0339AID720691; AID720692
pregnane X nuclear receptorHomo sapiens (human)Potency15.84890.005428.02631,258.9301AID1346985
estrogen nuclear receptor alphaHomo sapiens (human)Potency11.84810.000229.305416,493.5996AID743069; AID743075; AID743078; AID743080; AID743091
peroxisome proliferator activated receptor gammaHomo sapiens (human)Potency8.54150.001019.414170.9645AID743094; AID743140
cytochrome P450, family 19, subfamily A, polypeptide 1, isoform CRA_aHomo sapiens (human)Potency0.02660.001723.839378.1014AID743083
vitamin D3 receptor isoform VDRAHomo sapiens (human)Potency39.81070.354828.065989.1251AID504847
thyroid hormone receptor beta isoform 2Rattus norvegicus (Norway rat)Potency0.00290.000323.4451159.6830AID743065; AID743067
nuclear receptor ROR-gamma isoform 1Mus musculus (house mouse)Potency17.74080.00798.23321,122.0200AID2546; AID2551
gemininHomo sapiens (human)Potency26.78520.004611.374133.4983AID624296; AID624297
peripheral myelin protein 22Rattus norvegicus (Norway rat)Potency32.19680.005612.367736.1254AID624032
Cellular tumor antigen p53Homo sapiens (human)Potency23.71010.002319.595674.0614AID651631; AID720552
ATPase family AAA domain-containing protein 5Homo sapiens (human)Potency29.41690.011917.942071.5630AID651632; AID720516
Ataxin-2Homo sapiens (human)Potency26.60320.011912.222168.7989AID651632
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)84.00000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)95.00000.20005.677410.0000AID1473741
Bile salt export pumpHomo sapiens (human)IC50 (µMol)2.00000.11007.190310.0000AID1473738
Potassium voltage-gated channel subfamily E member 1Homo sapiens (human)IC50 (µMol)100.00000.12004.048010.0000AID1207371
Serine/threonine-protein kinase mTORHomo sapiens (human)IC50 (µMol)0.00220.00000.857510.0000AID1662433; AID1876108; AID1916668
Potassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)IC50 (µMol)100.00000.12004.048010.0000AID1207371
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)5,714.80510.00091.901410.0000AID1207465; AID1207496; AID1207525
Sodium channel protein type 5 subunit alphaHomo sapiens (human)IC50 (µMol)415.24150.00033.64849.2000AID1207311; AID1207342
Regulatory-associated protein of mTORHomo sapiens (human)IC50 (µMol)0.00000.00000.11390.6600AID1916668
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)11.30002.41006.343310.0000AID1473739
Target of rapamycin complex subunit LST8Homo sapiens (human)IC50 (µMol)0.00000.00000.09630.6600AID1916668
Potassium voltage-gated channel subfamily D member 3Homo sapiens (human)IC50 (µMol)10,000.00001.40005.35009.3000AID1207431
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Bone morphogenetic protein receptor type-1BHomo sapiens (human)Kd30.00000.00091.14133.7000AID1424922
Cell division cycle 7-related protein kinaseHomo sapiens (human)Kd30.00000.51100.51100.5110AID1424936
Serine/threonine-protein kinase PLK4Homo sapiens (human)Kd30.00000.00081.51449.0000AID1425121
ATP-dependent RNA helicase DDX3XHomo sapiens (human)Kd30.00000.43500.43500.4350AID1424975
Pyridoxal kinaseHomo sapiens (human)Kd30.00000.28605.076516.4040AID1425106
Citron Rho-interacting kinaseHomo sapiens (human)Kd30.00000.03303.064648.8760AID1424954
Serine/threonine-protein kinase Chk1Homo sapiens (human)Kd30.00000.00281.47448.7000AID1424953
Aurora kinase AHomo sapiens (human)Kd30.00000.00010.73429.3000AID1424917
Cyclin-G-associated kinaseHomo sapiens (human)Kd30.00000.00030.908628.6510AID1425009
Ephrin type-B receptor 6Homo sapiens (human)Kd30.00000.00001.07689.0000AID1424995
Peroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)Kd30.00000.02601.31402.6020AID1424896
Receptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)Kd30.00000.00201.621211.4330AID1425155
Mitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)Kd30.00000.09401.39103.5070AID1424926
Dynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)Kd30.00000.01700.36100.7050AID1425097
Eukaryotic translation initiation factor 5BHomo sapiens (human)Kd30.00000.23200.23200.2320AID1424986
Rho-associated protein kinase 2Homo sapiens (human)Kd30.00000.00022.710556.0660AID1425158
Serine/threonine-protein kinase ULK1Homo sapiens (human)Kd30.00000.00081.841023.2730AID1425208
Serine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)Kd30.00000.00572.009512.2010AID1424997
Ribosomal protein S6 kinase alpha-5Homo sapiens (human)Kd30.00000.01701.973729.9570AID1425162
U5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)Kd30.00001.38201.38201.3820AID1425174
Ribosomal protein S6 kinase alpha-4Homo sapiens (human)Kd30.00000.01201.63967.2000AID1425161
Serine/threonine-protein kinase 16Homo sapiens (human)Kd30.00000.00171.24839.9690AID1425179
Serine/threonine-protein kinase 10Homo sapiens (human)Kd30.00000.00002.923457.4530AID1425177
Serine/threonine-protein kinase D3Homo sapiens (human)Kd30.00000.00892.273823.3410AID1425137
Structural maintenance of chromosomes protein 2Homo sapiens (human)Kd30.00000.20900.65751.1060AID1425173
Mitogen-activated protein kinase kinase kinase 6Homo sapiens (human)Kd30.00000.17001.57818.0000AID1425050
Mitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)Kd30.00000.00822.364562.7720AID1425054
Serine/threonine-protein kinase LATS1Homo sapiens (human)Kd30.00000.01401.839310.7330AID1425033
Serine/threonine-protein kinase PAK 4Homo sapiens (human)Kd30.00000.00272.569430.3710AID1425100
Tyrosine-protein kinase ABL1Homo sapiens (human)Kd30.00000.00001.041113.4530AID1424890
Epidermal growth factor receptorHomo sapiens (human)Kd30.00000.00011.351420.8270AID1424983
High affinity nerve growth factor receptorHomo sapiens (human)Kd30.00000.00201.34849.2000AID1425094
Guanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)Kd30.00000.18400.18400.1840AID1425011
ADP/ATP translocase 2Homo sapiens (human)Kd30.00000.45100.45100.4510AID1425169
Protein kinase C beta typeHomo sapiens (human)Kd30.00000.00132.708126.3240AID1425130
Insulin receptorHomo sapiens (human)Kd30.00000.00171.08237.9060AID1425026
Tyrosine-protein kinase LckHomo sapiens (human)Kd30.00000.00021.117424.2210AID1425034
Tyrosine-protein kinase FynHomo sapiens (human)Kd30.00000.00081.42388.4000AID1425008
Cyclin-dependent kinase 1Homo sapiens (human)Kd30.00000.28801.49523.0490AID1424937
Glycogen phosphorylase, liver formHomo sapiens (human)Kd30.00002.12102.12102.1210AID1425146
Tyrosine-protein kinase Fes/FpsHomo sapiens (human)Kd30.00000.00481.09867.4000AID1425003
Adenine phosphoribosyltransferaseHomo sapiens (human)Kd30.00000.02900.02900.0290AID1424914
Tyrosine-protein kinase YesHomo sapiens (human)Kd30.00000.00031.370817.1520AID1425212
Tyrosine-protein kinase LynHomo sapiens (human)Kd30.00000.00061.04855.7000AID1425037
Proto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)Kd30.00000.00070.864227.5420AID1425154
Insulin-like growth factor 1 receptorHomo sapiens (human)Kd30.00000.00101.921119.2170AID1425022
Signal recognition particle receptor subunit alphaHomo sapiens (human)Kd30.00000.00800.00800.0080AID1425176
Cytochrome c1, heme protein, mitochondrialHomo sapiens (human)Kd30.00000.20200.20200.2020AID1424969
Hepatocyte growth factor receptorHomo sapiens (human)Kd30.00000.00021.62978.5000AID1425076
Tyrosine-protein kinase HCKHomo sapiens (human)Kd30.00000.00032.034315.9930AID1425017
Platelet-derived growth factor receptor betaHomo sapiens (human)Kd30.00000.00011.005011.1070AID1425104
Tyrosine-protein kinase FgrHomo sapiens (human)Kd30.00000.00051.07217.8000AID1425005
Serine/threonine-protein kinase A-RafHomo sapiens (human)Kd30.00000.04709.683233.6550AID1424915
Glycogen phosphorylase, brain formHomo sapiens (human)Kd30.00003.56903.56903.5690AID1425145
Breakpoint cluster region proteinHomo sapiens (human)Kd30.00000.00301.219617.3640AID1424919
Serine/threonine-protein kinase pim-1Homo sapiens (human)Kd30.00000.00101.139319.3160AID1425111
Fibroblast growth factor receptor 1Homo sapiens (human)Kd30.00000.00031.55816.2000AID1425004
DNA topoisomerase 2-alphaHomo sapiens (human)Kd30.00000.06400.27500.4860AID1425202
Cyclin-dependent kinase 4Homo sapiens (human)Kd30.00000.00331.60508.6000AID1424946
ADP/ATP translocase 3Homo sapiens (human)Kd30.00000.00600.25050.4950AID1425170
Proto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)Kd30.00000.00021.50779.6000AID1425175
cAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)Kd30.00000.05201.75353.4550AID1425128
Serine/threonine-protein kinase B-rafHomo sapiens (human)Kd30.00000.00021.625826.0180AID1424924
Phosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)Kd30.00000.00012.05699.5000AID1425110
Ribosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)Kd30.00000.00406.755688.9030AID1425093
Tyrosine-protein kinase FerHomo sapiens (human)Kd30.00000.00141.36048.8000AID1425002
Protein kinase C alpha typeHomo sapiens (human)Kd30.00000.00031.792221.3520AID1425129
cAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)Kd30.00000.00392.947923.2450AID1425123
General transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)Kd30.00000.00201.690612.0220AID1424996
Casein kinase II subunit alpha'Homo sapiens (human)Kd30.00000.00102.530928.8720AID1424968
Ras-related protein Rab-6AHomo sapiens (human)Kd30.00000.03300.03300.0330AID1425150
Ephrin type-A receptor 1Homo sapiens (human)Kd30.00000.00411.80009.8000AID1424987
Multifunctional protein ADE2Homo sapiens (human)Kd30.00005.48105.48105.4810AID1425098
cAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)Kd30.00000.00208.557749.2780AID1425125
cAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)Kd30.00000.01300.74084.1000AID1425124
Ferrochelatase, mitochondrialHomo sapiens (human)Kd30.00000.24306.434367.9140AID1425001
Ribosomal protein S6 kinase beta-1Homo sapiens (human)Kd30.00000.00131.18054.8000AID1425164
Tyrosine-protein kinase JAK1Homo sapiens (human)Kd30.00000.00161.21667.8000AID1425030
Cyclin-dependent kinase 2Homo sapiens (human)Kd30.00000.00701.517910.4870AID1424944
Beta-adrenergic receptor kinase 1Homo sapiens (human)Kd30.00000.17005.579122.4940AID1424908
Probable ATP-dependent RNA helicase DDX6Homo sapiens (human)Kd30.00004.10304.10304.1030AID1424977
Mitogen-activated protein kinase 3 Homo sapiens (human)Kd30.00000.43005.27439.8000AID1425061
MAP/microtubule affinity-regulating kinase 3Homo sapiens (human)Kd30.00000.00303.968958.2400AID1425069
Deoxycytidine kinaseHomo sapiens (human)Kd30.00000.01201.08752.1630AID1424970
Mitogen-activated protein kinase 1Homo sapiens (human)Kd30.00000.00012.74417.3000AID1425056
Ephrin type-A receptor 2Homo sapiens (human)Kd30.00000.00091.07528.1980AID1424988
Ephrin type-B receptor 2Homo sapiens (human)Kd30.00000.00043.153653.1980AID1424992
Non-receptor tyrosine-protein kinase TYK2Homo sapiens (human)Kd30.00000.00091.55758.7000AID1425207
UMP-CMP kinase Homo sapiens (human)Kd30.00000.00300.00450.0060AID1424959
Phosphatidylethanolamine-binding protein 1Homo sapiens (human)Kd30.00000.00300.00300.0030AID1425107
Wee1-like protein kinaseHomo sapiens (human)Kd30.00000.00143.538965.1580AID1425210
Heme oxygenase 2Homo sapiens (human)Kd30.00000.11900.11900.1190AID1425018
DnaJ homolog subfamily A member 1Homo sapiens (human)Kd30.00000.96200.96200.9620AID1424980
RAC-alpha serine/threonine-protein kinaseHomo sapiens (human)Kd30.00000.00061.06214.4000AID1424910
RAC-beta serine/threonine-protein kinaseHomo sapiens (human)Kd30.00000.00211.61968.7000AID1424911
Dual specificity protein kinase TTKHomo sapiens (human)Kd30.00000.00651.62698.5000AID1425205
DNA replication licensing factor MCM4Homo sapiens (human)Kd30.00000.62900.62900.6290AID1425072
Myosin-10Homo sapiens (human)Kd30.00000.22900.49350.7580AID1425079
Dual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)Kd30.00000.00391.64299.6000AID1425039
Receptor-type tyrosine-protein kinase FLT3Homo sapiens (human)Kd30.00000.00020.95599.9000AID1425006
Bone morphogenetic protein receptor type-1AHomo sapiens (human)Kd30.00000.06001.50107.0000AID1424921
Activin receptor type-1BHomo sapiens (human)Kd30.00000.00401.511015.2580AID1424901
TGF-beta receptor type-1Homo sapiens (human)Kd30.00000.00502.27859.6000AID1425196
TGF-beta receptor type-2Homo sapiens (human)Kd30.00000.08001.83516.9000AID1425197
Electron transfer flavoprotein subunit betaHomo sapiens (human)Kd30.00000.01200.01200.0120AID1424999
Tyrosine-protein kinase CSKHomo sapiens (human)Kd30.00000.00103.457839.5530AID1424960
Glycine--tRNA ligaseHomo sapiens (human)Kd30.00000.04000.04000.0400AID1425010
Protein kinase C iota typeHomo sapiens (human)Kd30.00000.02609.331651.0180AID1425133
Exosome RNA helicase MTR4Homo sapiens (human)Kd30.00002.60702.60702.6070AID1425168
Serine/threonine-protein kinase mTORHomo sapiens (human)Kd0.00200.00010.59939.2000AID1409403
Tyrosine-protein kinase TecHomo sapiens (human)Kd30.00000.00101.00958.7000AID1425193
Tyrosine-protein kinase ABL2Homo sapiens (human)Kd30.00000.00021.124914.9240AID1424891
Tyrosine-protein kinase FRKHomo sapiens (human)Kd30.00000.00031.242410.8370AID1425007
G protein-coupled receptor kinase 6Homo sapiens (human)Kd30.00001.18901.40201.6150AID1425012
Tyrosine-protein kinase SYKHomo sapiens (human)Kd30.00000.00702.00529.2260AID1425188
26S proteasome regulatory subunit 6BHomo sapiens (human)Kd30.00000.00500.00500.0050AID1425141
Mitogen-activated protein kinase 8Homo sapiens (human)Kd30.00000.01102.096526.0590AID1425063
Mitogen-activated protein kinase 9Homo sapiens (human)Kd30.00000.00201.45968.1000AID1425064
Dual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)Kd30.00000.00502.04626.6000AID1425040
Phosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)Kd30.00000.20803.61257.0170AID1425113
Casein kinase I isoform alphaHomo sapiens (human)Kd30.00000.00102.575619.3520AID1424961
Casein kinase I isoform deltaHomo sapiens (human)Kd30.00000.01502.227018.3960AID1424962
MAP kinase-activated protein kinase 2Homo sapiens (human)Kd30.00000.00032.027414.7420AID1425065
Elongation factor Tu, mitochondrialHomo sapiens (human)Kd30.00000.46400.46400.4640AID1425206
Choline-phosphate cytidylyltransferase AHomo sapiens (human)Kd30.00000.04100.04100.0410AID1425103
Cysteine--tRNA ligase, cytoplasmicHomo sapiens (human)Kd30.00000.01200.33200.6520AID1424932
Casein kinase I isoform epsilonHomo sapiens (human)Kd30.00000.01301.408612.4090AID1424963
Very long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)Kd30.00001.68901.68901.6890AID1424894
Dual specificity protein kinase CLK1Homo sapiens (human)Kd30.00000.00201.879129.8810AID1424955
Dual specificity protein kinase CLK2Homo sapiens (human)Kd30.00000.00701.13846.5000AID1424956
Dual specificity protein kinase CLK3Homo sapiens (human)Kd30.00000.01002.44999.0000AID1424957
Glycogen synthase kinase-3 alphaHomo sapiens (human)Kd30.00000.00602.475422.5430AID1425013
Glycogen synthase kinase-3 betaHomo sapiens (human)Kd30.00000.00701.00576.1680AID1425014
Cyclin-dependent kinase 7Homo sapiens (human)Kd30.00000.00251.67837.7000AID1424949
Cyclin-dependent kinase 9Homo sapiens (human)Kd30.00000.00101.61669.9010AID1424950
Ras-related protein Rab-27AHomo sapiens (human)Kd30.00004.49304.49304.4930AID1425149
Interleukin-1 receptor-associated kinase 1Homo sapiens (human)Kd30.00000.00611.52528.5000AID1425027
Ribosomal protein S6 kinase alpha-3Homo sapiens (human)Kd30.00000.01702.889637.6050AID1425160
Serine/threonine-protein kinase Nek2Homo sapiens (human)Kd30.00000.11001.56496.5000AID1425086
Serine/threonine-protein kinase Nek3Homo sapiens (human)Kd30.00000.17005.936838.0880AID1425087
Dual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)Kd30.00000.00342.39436.5000AID1425043
Serine/threonine-protein kinase PLK1Homo sapiens (human)Kd30.00000.00010.57115.0000AID1425120
LIM domain kinase 1Homo sapiens (human)Kd30.00000.02601.784021.0890AID1425035
LIM domain kinase 2Homo sapiens (human)Kd30.00000.05704.971752.0560AID1425036
Mitogen-activated protein kinase 10Homo sapiens (human)Kd30.00000.00101.63545.9000AID1425057
Tyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)Kd30.00003.31603.31603.3160AID1425211
5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)Kd30.00000.00601.468110.2120AID1425126
Ephrin type-B receptor 3Homo sapiens (human)Kd30.00000.00692.17136.4100AID1424993
Ephrin type-A receptor 5Homo sapiens (human)Kd30.00000.00021.21005.9000AID1424990
Ephrin type-B receptor 4Homo sapiens (human)Kd30.00000.00032.167826.3990AID1424994
Ephrin type-A receptor 4Homo sapiens (human)Kd30.00000.00123.152543.9420AID1424989
Adenylate kinase 2, mitochondrialHomo sapiens (human)Kd30.00001.03601.03601.0360AID1424909
Adenosine kinaseHomo sapiens (human)Kd30.00000.01301.83683.4930AID1424907
Ras-related protein Rab-10Homo sapiens (human)Kd30.00001.34801.34801.3480AID1425148
Actin-related protein 3Homo sapiens (human)Kd30.00000.03602.77355.5110AID1424899
Actin-related protein 2Homo sapiens (human)Kd30.00000.00400.00400.0040AID1424898
GTP-binding nuclear protein RanHomo sapiens (human)Kd30.00000.75900.75900.7590AID1425153
Casein kinase I isoform gamma-2Homo sapiens (human)Kd30.00000.04601.45066.6000AID1424965
Cyclin-dependent kinase 3Homo sapiens (human)Kd30.00000.00803.060263.6140AID1424945
Cyclin-dependent kinase 6Homo sapiens (human)Kd30.00000.03201.20073.3560AID1424948
Cyclin-dependent-like kinase 5 Homo sapiens (human)Kd30.00000.04301.37578.3000AID1424947
Cyclin-dependent kinase 16Homo sapiens (human)Kd30.00000.00111.585510.0000AID1424941
Cyclin-dependent kinase 17Homo sapiens (human)Kd30.00000.00100.82335.6000AID1424942
ATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)Kd30.00000.98300.98300.9830AID1425108
Dual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)Kd30.00000.00021.13868.7730AID1425038
DNA topoisomerase 2-betaHomo sapiens (human)Kd30.00000.14801.22702.5970AID1425203
Protein kinase C theta typeHomo sapiens (human)Kd30.00000.00071.61407.2000AID1425134
Activin receptor type-1Homo sapiens (human)Kd30.00000.00401.485316.1210AID1424900
Macrophage-stimulating protein receptorHomo sapiens (human)Kd30.00000.00302.07188.4000AID1425078
Focal adhesion kinase 1Homo sapiens (human)Kd30.00000.00051.225513.0390AID1425142
Protein kinase C delta typeHomo sapiens (human)Kd30.00000.00021.12619.2060AID1425131
Tyrosine-protein kinase BTKHomo sapiens (human)Kd30.00000.00061.529910.1530AID1424925
Activated CDC42 kinase 1Homo sapiens (human)Kd30.00000.00201.71389.6000AID1425201
Epithelial discoidin domain-containing receptor 1Homo sapiens (human)Kd30.00000.00021.631471.4840AID1424972
Mitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)Kd30.00000.00311.468114.0430AID1425052
Serine/threonine-protein kinase 4Homo sapiens (human)Kd30.00000.00021.712025.9020AID1425185
5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)Kd30.00000.00371.891315.3890AID1425122
Dual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)Kd30.00000.00022.659065.6770AID1425042
Mitogen-activated protein kinase 7Homo sapiens (human)Kd30.00000.04202.00739.9000AID1425062
Serine/threonine-protein kinase PAK 2Homo sapiens (human)Kd30.00000.00312.30456.0000AID1425099
Serine/threonine-protein kinase 3Homo sapiens (human)Kd30.00000.00021.860217.5260AID1425182
Mitogen-activated protein kinase kinase kinase 1Homo sapiens (human)Kd30.00000.09702.599512.4730AID1425044
Integrin-linked protein kinaseHomo sapiens (human)Kd30.00000.02000.46031.3290AID1425024
Rho-associated protein kinase 1Homo sapiens (human)Kd30.00000.00031.755513.4620AID1425157
Non-receptor tyrosine-protein kinase TNK1Homo sapiens (human)Kd30.00000.00181.006411.2690AID1425200
Calcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)Kd30.00000.00051.02097.8000AID1424929
Calcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)Kd30.00000.00031.504420.3010AID1424928
Dual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)Kd30.00000.00012.101640.2910AID1424981
Activin receptor type-2BHomo sapiens (human)Kd30.00000.00762.73289.9000AID1424902
Bone morphogenetic protein receptor type-2Homo sapiens (human)Kd30.00000.01902.591714.3770AID1424923
Protein-tyrosine kinase 6Homo sapiens (human)Kd30.00000.00431.74309.0000AID1425144
cGMP-dependent protein kinase 1 Homo sapiens (human)Kd30.00000.00160.70723.8000AID1425138
Cyclin-dependent kinase 13Homo sapiens (human)Kd30.00000.00091.25714.5180AID1424940
Inhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)Kd30.00000.00511.10938.3000AID1425023
Protein-tyrosine kinase 2-betaHomo sapiens (human)Kd30.00000.00111.945030.4140AID1425143
Maternal embryonic leucine zipper kinaseHomo sapiens (human)Kd30.00000.00492.283529.9330AID1425074
Structural maintenance of chromosomes protein 1AHomo sapiens (human)Kd30.00000.36500.36500.3650AID1425172
Chromodomain-helicase-DNA-binding protein 4Homo sapiens (human)Kd30.00000.00300.00300.0030AID1424952
Peroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)Kd30.00000.01400.14250.2710AID1424895
Ephrin type-A receptor 7Homo sapiens (human)Kd30.00000.00251.44456.5000AID1424991
Delta(24)-sterol reductaseHomo sapiens (human)Kd30.00000.43200.43200.4320AID1424978
Ribosomal protein S6 kinase alpha-1Homo sapiens (human)Kd30.00000.02802.528622.7260AID1425159
Dual specificity testis-specific protein kinase 1Homo sapiens (human)Kd30.00000.03301.75685.6000AID1425194
Myosin light chain kinase, smooth muscleHomo sapiens (human)Kd30.00000.00301.20887.9000AID1425081
Mitogen-activated protein kinase 11Homo sapiens (human)Kd30.00000.00010.46103.7430AID1425058
Serine/threonine-protein kinase STK11Homo sapiens (human)Kd30.00000.00300.99495.9000AID1425178
Serine/threonine-protein kinase N1Homo sapiens (human)Kd30.00000.00133.172949.8130AID1425117
Serine/threonine-protein kinase N2Homo sapiens (human)Kd30.00000.00181.75279.9000AID1425118
Mitogen-activated protein kinase 14Homo sapiens (human)Kd30.00000.00000.50368.5000AID1425059
Calcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)Kd30.00000.03001.92155.4600AID1424930
Mitogen-activated protein kinase kinase kinase 11Homo sapiens (human)Kd30.00000.01101.563917.9840AID1425045
Discoidin domain-containing receptor 2Homo sapiens (human)Kd30.00000.00301.988842.2800AID1424973
AP2-associated protein kinase 1Homo sapiens (human)Kd30.00000.00121.370713.7110AID1424889
Myosin light chain kinase 3Homo sapiens (human)Kd30.00000.00201.618410.4240AID1425082
Putative heat shock protein HSP 90-beta 2Homo sapiens (human)Kd30.00002.56602.56602.5660AID1425019
Serine/threonine-protein kinase MRCK alphaHomo sapiens (human)Kd30.00000.05704.554714.0200AID1424933
Serine/threonine-protein kinase MRCK gammaHomo sapiens (human)Kd30.00000.03701.96259.5000AID1424935
Acyl-CoA dehydrogenase family member 10Homo sapiens (human)Kd30.00000.07801.69973.9570AID1424892
Serine/threonine-protein kinase N3Homo sapiens (human)Kd30.00000.09900.73651.3740AID1425119
Serine/threonine-protein kinase ULK3Homo sapiens (human)Kd30.00000.00121.33509.9000AID1425209
Uncharacterized protein FLJ45252Homo sapiens (human)Kd30.00000.00301.22929.3110AID1425147
Acyl-CoA dehydrogenase family member 11Homo sapiens (human)Kd30.00001.91603.07304.1470AID1424893
Serine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)Kd30.00000.11600.76041.5000AID1424998
Serine/threonine-protein kinase MARK2Homo sapiens (human)Kd30.00000.00011.842511.1030AID1425068
ATP-dependent RNA helicase DHX30Homo sapiens (human)Kd30.00000.00600.00600.0060AID1424979
Serine/threonine-protein kinase TAO1Homo sapiens (human)Kd30.00000.00042.161218.7570AID1425189
STE20-related kinase adapter protein alphaHomo sapiens (human)Kd30.00000.31601.72083.6720AID1425186
AarF domain-containing protein kinase 1Homo sapiens (human)Kd30.00000.02303.113722.7470AID1424904
Mitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)Kd30.00000.00511.641315.4350AID1425053
Eukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)Kd30.00000.00300.00300.0030AID1425015
Atypical kinase COQ8A, mitochondrialHomo sapiens (human)Kd30.00000.09405.167365.3020AID1424905
Phosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)Kd30.00000.00302.75228.8000AID1425115
Mitogen-activated protein kinase 15Homo sapiens (human)Kd30.00000.00490.68804.5000AID1425060
Serine/threonine-protein kinase Nek9Homo sapiens (human)Kd30.00000.01602.742819.6170AID1425089
Serine/threonine-protein kinase Nek7Homo sapiens (human)Kd30.00000.00303.67198.7000AID1425088
ATP-dependent RNA helicase DDX1Homo sapiens (human)Kd30.00000.08600.08600.0860AID1424974
Mitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)Kd30.00000.00100.93785.5000AID1425051
Aurora kinase BHomo sapiens (human)Kd30.00000.00201.061422.8520AID1424918
MAP/microtubule affinity-regulating kinase 4Homo sapiens (human)Kd30.00000.00541.10294.9000AID1425070
Serine/threonine-protein kinase Nek1Homo sapiens (human)Kd30.00000.17002.42948.3000AID1425085
PAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)Kd30.00001.06701.06701.0670AID1425102
Calcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)Kd30.00000.00003.233152.8470AID1424931
EKC/KEOPS complex subunit TP53RKHomo sapiens (human)Kd30.00000.31101.95193.8400AID1425204
Dual specificity testis-specific protein kinase 2Homo sapiens (human)Kd30.00000.00200.00200.0020AID1425195
Membrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)Kd30.00000.04400.92852.9000AID1425116
Mitogen-activated protein kinase kinase kinase 5Homo sapiens (human)Kd30.00000.07006.564750.5360AID1425049
Mitogen-activated protein kinase kinase kinase 3Homo sapiens (human)Kd30.00000.00601.53319.9000AID1425047
Eukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)Kd30.00000.05801.92244.8360AID1424984
Nucleolar GTP-binding protein 1Homo sapiens (human)Kd30.00000.00904.10358.1980AID1425016
Serine/threonine-protein kinase D2Homo sapiens (human)Kd30.00000.00812.372325.0190AID1425136
NUAK family SNF1-like kinase 2Homo sapiens (human)Kd30.00000.00010.67744.6000AID1425095
RNA cytidine acetyltransferaseHomo sapiens (human)Kd30.00001.24001.24001.2400AID1425083
Serine/threonine-protein kinase SIK2Homo sapiens (human)Kd30.00000.00111.816541.7950AID1425166
STE20-like serine/threonine-protein kinase Homo sapiens (human)Kd30.00000.00003.857399.2320AID1425171
Serine/threonine-protein kinase TAO3Homo sapiens (human)Kd30.00000.00022.713114.1960AID1425191
dCTP pyrophosphatase 1Homo sapiens (human)Kd30.00000.57301.74033.0540AID1424971
Dual specificity protein kinase CLK4Homo sapiens (human)Kd30.00000.00201.41228.3000AID1424958
Casein kinase I isoform gamma-1Homo sapiens (human)Kd30.00000.05302.06225.7000AID1424964
Phenylalanine--tRNA ligase beta subunitHomo sapiens (human)Kd30.00000.00300.00450.0060AID1425000
BMP-2-inducible protein kinaseHomo sapiens (human)Kd30.00000.00222.409756.0320AID1424920
Obg-like ATPase 1Homo sapiens (human)Kd30.00000.00300.00500.0070AID1425096
Interleukin-1 receptor-associated kinase 4Homo sapiens (human)Kd30.00000.00173.471934.1450AID1425029
Mitogen-activated protein kinase kinase kinase 20Homo sapiens (human)Kd30.00000.00231.703413.6380AID1425213
Cyclin-dependent kinase 12Homo sapiens (human)Kd30.00000.03201.80325.6350AID1424939
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)Kd30.00003.92003.92003.9200AID1425084
Serine/threonine-protein kinase 26Homo sapiens (human)Kd30.00000.00741.73808.3000AID1425181
Succinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)Kd30.00000.00700.00700.0070AID1425187
Serine/threonine-protein kinase NLKHomo sapiens (human)Kd30.00000.00601.02264.4000AID1425090
5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)Kd30.00000.00501.15819.1280AID1425127
Serine/threonine-protein kinase TBK1Homo sapiens (human)Kd30.00000.00091.767449.6010AID1425192
Septin-9Homo sapiens (human)Kd30.00000.01000.24300.6350AID1425165
Ribosomal protein S6 kinase alpha-6Homo sapiens (human)Kd30.00000.00402.415323.7620AID1425163
TRAF2 and NCK-interacting protein kinaseHomo sapiens (human)Kd30.00000.00471.393510.0000AID1425199
Serine/threonine-protein kinase TAO2Homo sapiens (human)Kd30.00000.01002.017612.9420AID1425190
Long-chain-fatty-acid--CoA ligase 5Homo sapiens (human)Kd30.00000.00800.63531.6900AID1424897
RAC-gamma serine/threonine-protein kinaseHomo sapiens (human)Kd30.00000.00251.76466.2000AID1424912
Serine/threonine-protein kinase SIK3Homo sapiens (human)Kd30.00000.00051.508610.3180AID1425167
Mitogen-activated protein kinase kinase kinase 2Homo sapiens (human)Kd30.00000.00241.32986.9000AID1425046
Thyroid hormone receptor-associated protein 3Homo sapiens (human)Kd30.00002.74602.74602.7460AID1425198
Mitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)Kd30.00000.00051.949450.2140AID1425055
Receptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)Kd30.00000.01101.47976.7000AID1425156
Serine/threonine-protein kinase MRCK betaHomo sapiens (human)Kd30.00000.03403.625250.0050AID1424934
Interleukin-1 receptor-associated kinase 3Homo sapiens (human)Kd30.00000.00701.713725.5810AID1425028
Casein kinase I isoform gamma-3Homo sapiens (human)Kd30.00000.09702.39788.7000AID1424966
Mitogen-activated protein kinase kinase kinase 4Homo sapiens (human)Kd30.00000.03902.39708.4000AID1425048
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (2590)

Processvia Protein(s)Taxonomy
positive regulation of gene expressionBone morphogenetic protein receptor type-1BHomo sapiens (human)
cartilage condensationBone morphogenetic protein receptor type-1BHomo sapiens (human)
ovarian cumulus expansionBone morphogenetic protein receptor type-1BHomo sapiens (human)
osteoblast differentiationBone morphogenetic protein receptor type-1BHomo sapiens (human)
eye developmentBone morphogenetic protein receptor type-1BHomo sapiens (human)
chondrocyte developmentBone morphogenetic protein receptor type-1BHomo sapiens (human)
inflammatory responseBone morphogenetic protein receptor type-1BHomo sapiens (human)
central nervous system neuron differentiationBone morphogenetic protein receptor type-1BHomo sapiens (human)
proteoglycan biosynthetic processBone morphogenetic protein receptor type-1BHomo sapiens (human)
positive regulation of bone mineralizationBone morphogenetic protein receptor type-1BHomo sapiens (human)
BMP signaling pathwayBone morphogenetic protein receptor type-1BHomo sapiens (human)
retinal ganglion cell axon guidanceBone morphogenetic protein receptor type-1BHomo sapiens (human)
positive regulation of chondrocyte differentiationBone morphogenetic protein receptor type-1BHomo sapiens (human)
ovulation cycleBone morphogenetic protein receptor type-1BHomo sapiens (human)
positive regulation of osteoblast differentiationBone morphogenetic protein receptor type-1BHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIBone morphogenetic protein receptor type-1BHomo sapiens (human)
retina development in camera-type eyeBone morphogenetic protein receptor type-1BHomo sapiens (human)
endochondral bone morphogenesisBone morphogenetic protein receptor type-1BHomo sapiens (human)
positive regulation of cartilage developmentBone morphogenetic protein receptor type-1BHomo sapiens (human)
cellular response to BMP stimulusBone morphogenetic protein receptor type-1BHomo sapiens (human)
positive regulation of extrinsic apoptotic signaling pathway via death domain receptorsBone morphogenetic protein receptor type-1BHomo sapiens (human)
negative regulation of chondrocyte proliferationBone morphogenetic protein receptor type-1BHomo sapiens (human)
dorsal/ventral pattern formationBone morphogenetic protein receptor type-1BHomo sapiens (human)
protein phosphorylationBone morphogenetic protein receptor type-1BHomo sapiens (human)
cellular response to growth factor stimulusBone morphogenetic protein receptor type-1BHomo sapiens (human)
G1/S transition of mitotic cell cycleCell division cycle 7-related protein kinaseHomo sapiens (human)
positive regulation of cell population proliferationCell division cycle 7-related protein kinaseHomo sapiens (human)
positive regulation of nuclear cell cycle DNA replicationCell division cycle 7-related protein kinaseHomo sapiens (human)
positive regulation of G2/M transition of mitotic cell cycleCell division cycle 7-related protein kinaseHomo sapiens (human)
cell cycle phase transitionCell division cycle 7-related protein kinaseHomo sapiens (human)
cell divisionCell division cycle 7-related protein kinaseHomo sapiens (human)
peptidyl-serine phosphorylationCell division cycle 7-related protein kinaseHomo sapiens (human)
double-strand break repair via break-induced replicationCell division cycle 7-related protein kinaseHomo sapiens (human)
signal transductionCell division cycle 7-related protein kinaseHomo sapiens (human)
positive regulation of centriole replicationSerine/threonine-protein kinase PLK4Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase PLK4Homo sapiens (human)
centriole replicationSerine/threonine-protein kinase PLK4Homo sapiens (human)
positive regulation of centriole replicationSerine/threonine-protein kinase PLK4Homo sapiens (human)
cilium assemblySerine/threonine-protein kinase PLK4Homo sapiens (human)
trophoblast giant cell differentiationSerine/threonine-protein kinase PLK4Homo sapiens (human)
de novo centriole assembly involved in multi-ciliated epithelial cell differentiationSerine/threonine-protein kinase PLK4Homo sapiens (human)
translational initiationATP-dependent RNA helicase DDX3XHomo sapiens (human)
chromosome segregationATP-dependent RNA helicase DDX3XHomo sapiens (human)
extrinsic apoptotic signaling pathway via death domain receptorsATP-dependent RNA helicase DDX3XHomo sapiens (human)
response to virusATP-dependent RNA helicase DDX3XHomo sapiens (human)
RNA secondary structure unwindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of gene expressionATP-dependent RNA helicase DDX3XHomo sapiens (human)
Wnt signaling pathwayATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of translationATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of cell growthATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of cell growthATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of protein-containing complex assemblyATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of protein autophosphorylationATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of type I interferon productionATP-dependent RNA helicase DDX3XHomo sapiens (human)
DNA duplex unwindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of interferon-alpha productionATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of interferon-beta productionATP-dependent RNA helicase DDX3XHomo sapiens (human)
stress granule assemblyATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of toll-like receptor 7 signaling pathwayATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of toll-like receptor 8 signaling pathwayATP-dependent RNA helicase DDX3XHomo sapiens (human)
intracellular signal transductionATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of translation in response to endoplasmic reticulum stressATP-dependent RNA helicase DDX3XHomo sapiens (human)
cytosolic ribosome assemblyATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of apoptotic processATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of apoptotic processATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of cysteine-type endopeptidase activity involved in apoptotic processATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of viral genome replicationATP-dependent RNA helicase DDX3XHomo sapiens (human)
innate immune responseATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of translationATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of translational initiationATP-dependent RNA helicase DDX3XHomo sapiens (human)
lipid homeostasisATP-dependent RNA helicase DDX3XHomo sapiens (human)
cellular response to arsenic-containing substanceATP-dependent RNA helicase DDX3XHomo sapiens (human)
cellular response to osmotic stressATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of chemokine (C-C motif) ligand 5 productionATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of protein serine/threonine kinase activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of canonical Wnt signaling pathwayATP-dependent RNA helicase DDX3XHomo sapiens (human)
intrinsic apoptotic signaling pathwayATP-dependent RNA helicase DDX3XHomo sapiens (human)
cellular response to virusATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of G1/S transition of mitotic cell cycleATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of NLRP3 inflammasome complex assemblyATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of non-canonical NF-kappaB signal transductionATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of non-canonical NF-kappaB signal transductionATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of protein acetylationATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathway via death domain receptorsATP-dependent RNA helicase DDX3XHomo sapiens (human)
positive regulation of protein K63-linked ubiquitinationATP-dependent RNA helicase DDX3XHomo sapiens (human)
protein localization to cytoplasmic stress granuleATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathwayATP-dependent RNA helicase DDX3XHomo sapiens (human)
negative regulation of gene expressionATP-dependent RNA helicase DDX3XHomo sapiens (human)
gamete generationATP-dependent RNA helicase DDX3XHomo sapiens (human)
cell differentiationATP-dependent RNA helicase DDX3XHomo sapiens (human)
pyridoxal 5'-phosphate salvagePyridoxal kinaseHomo sapiens (human)
pyridoxal metabolic processPyridoxal kinaseHomo sapiens (human)
pyridoxamine metabolic processPyridoxal kinaseHomo sapiens (human)
mitotic cell cycleCitron Rho-interacting kinaseHomo sapiens (human)
mitotic cytokinesisCitron Rho-interacting kinaseHomo sapiens (human)
positive regulation of cytokinesisCitron Rho-interacting kinaseHomo sapiens (human)
negative regulation of hippo signalingCitron Rho-interacting kinaseHomo sapiens (human)
generation of neuronsCitron Rho-interacting kinaseHomo sapiens (human)
neuron apoptotic processCitron Rho-interacting kinaseHomo sapiens (human)
DNA damage checkpoint signalingSerine/threonine-protein kinase Chk1Homo sapiens (human)
G2/M transition of mitotic cell cycleSerine/threonine-protein kinase Chk1Homo sapiens (human)
inner cell mass cell proliferationSerine/threonine-protein kinase Chk1Homo sapiens (human)
DNA replicationSerine/threonine-protein kinase Chk1Homo sapiens (human)
DNA repairSerine/threonine-protein kinase Chk1Homo sapiens (human)
chromatin remodelingSerine/threonine-protein kinase Chk1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase Chk1Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase Chk1Homo sapiens (human)
DNA damage responseSerine/threonine-protein kinase Chk1Homo sapiens (human)
nucleus organizationSerine/threonine-protein kinase Chk1Homo sapiens (human)
mitotic G2 DNA damage checkpoint signalingSerine/threonine-protein kinase Chk1Homo sapiens (human)
regulation of double-strand break repair via homologous recombinationSerine/threonine-protein kinase Chk1Homo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase Chk1Homo sapiens (human)
regulation of cell population proliferationSerine/threonine-protein kinase Chk1Homo sapiens (human)
signal transduction in response to DNA damageSerine/threonine-protein kinase Chk1Homo sapiens (human)
mitotic G2/M transition checkpointSerine/threonine-protein kinase Chk1Homo sapiens (human)
positive regulation of cell cycleSerine/threonine-protein kinase Chk1Homo sapiens (human)
negative regulation of gene expression, epigeneticSerine/threonine-protein kinase Chk1Homo sapiens (human)
negative regulation of mitotic nuclear divisionSerine/threonine-protein kinase Chk1Homo sapiens (human)
regulation of mitotic centrosome separationSerine/threonine-protein kinase Chk1Homo sapiens (human)
negative regulation of G0 to G1 transitionSerine/threonine-protein kinase Chk1Homo sapiens (human)
cellular response to mechanical stimulusSerine/threonine-protein kinase Chk1Homo sapiens (human)
cellular response to caffeineSerine/threonine-protein kinase Chk1Homo sapiens (human)
replicative senescenceSerine/threonine-protein kinase Chk1Homo sapiens (human)
regulation of signal transduction by p53 class mediatorSerine/threonine-protein kinase Chk1Homo sapiens (human)
apoptotic process involved in developmentSerine/threonine-protein kinase Chk1Homo sapiens (human)
negative regulation of DNA biosynthetic processSerine/threonine-protein kinase Chk1Homo sapiens (human)
protein phosphorylationAurora kinase AHomo sapiens (human)
response to woundingAurora kinase AHomo sapiens (human)
liver regenerationAurora kinase AHomo sapiens (human)
G2/M transition of mitotic cell cycleAurora kinase AHomo sapiens (human)
mitotic cell cycleAurora kinase AHomo sapiens (human)
chromatin remodelingAurora kinase AHomo sapiens (human)
protein phosphorylationAurora kinase AHomo sapiens (human)
apoptotic processAurora kinase AHomo sapiens (human)
spindle organizationAurora kinase AHomo sapiens (human)
spindle assembly involved in female meiosis IAurora kinase AHomo sapiens (human)
mitotic centrosome separationAurora kinase AHomo sapiens (human)
anterior/posterior axis specificationAurora kinase AHomo sapiens (human)
regulation of G2/M transition of mitotic cell cycleAurora kinase AHomo sapiens (human)
negative regulation of gene expressionAurora kinase AHomo sapiens (human)
peptidyl-serine phosphorylationAurora kinase AHomo sapiens (human)
regulation of protein stabilityAurora kinase AHomo sapiens (human)
negative regulation of protein bindingAurora kinase AHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processAurora kinase AHomo sapiens (human)
negative regulation of apoptotic processAurora kinase AHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processAurora kinase AHomo sapiens (human)
positive regulation of mitotic nuclear divisionAurora kinase AHomo sapiens (human)
positive regulation of mitotic cell cycleAurora kinase AHomo sapiens (human)
regulation of centrosome cycleAurora kinase AHomo sapiens (human)
protein autophosphorylationAurora kinase AHomo sapiens (human)
cell divisionAurora kinase AHomo sapiens (human)
centrosome localizationAurora kinase AHomo sapiens (human)
cilium disassemblyAurora kinase AHomo sapiens (human)
protein localization to centrosomeAurora kinase AHomo sapiens (human)
positive regulation of mitochondrial fissionAurora kinase AHomo sapiens (human)
positive regulation of oocyte maturationAurora kinase AHomo sapiens (human)
regulation of signal transduction by p53 class mediatorAurora kinase AHomo sapiens (human)
neuron projection extensionAurora kinase AHomo sapiens (human)
mitotic spindle organizationAurora kinase AHomo sapiens (human)
regulation of cytokinesisAurora kinase AHomo sapiens (human)
receptor-mediated endocytosisCyclin-G-associated kinaseHomo sapiens (human)
endoplasmic reticulum organizationCyclin-G-associated kinaseHomo sapiens (human)
Golgi organizationCyclin-G-associated kinaseHomo sapiens (human)
negative regulation of neuron projection developmentCyclin-G-associated kinaseHomo sapiens (human)
synaptic vesicle uncoatingCyclin-G-associated kinaseHomo sapiens (human)
protein localization to Golgi apparatusCyclin-G-associated kinaseHomo sapiens (human)
intracellular transportCyclin-G-associated kinaseHomo sapiens (human)
clathrin coat assemblyCyclin-G-associated kinaseHomo sapiens (human)
chaperone cofactor-dependent protein refoldingCyclin-G-associated kinaseHomo sapiens (human)
clathrin coat disassemblyCyclin-G-associated kinaseHomo sapiens (human)
clathrin-dependent endocytosisCyclin-G-associated kinaseHomo sapiens (human)
protein localization to plasma membraneCyclin-G-associated kinaseHomo sapiens (human)
Golgi to lysosome transportCyclin-G-associated kinaseHomo sapiens (human)
regulation of clathrin coat assemblyCyclin-G-associated kinaseHomo sapiens (human)
protein phosphorylationEphrin type-B receptor 6Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-B receptor 6Homo sapiens (human)
axon guidanceEphrin type-B receptor 6Homo sapiens (human)
fatty acid beta-oxidation using acyl-CoA oxidasePeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
lipid homeostasisPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
prostaglandin secretionMultidrug resistance-associated protein 4Homo sapiens (human)
cilium assemblyMultidrug resistance-associated protein 4Homo sapiens (human)
platelet degranulationMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
bile acid and bile salt transportMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transportMultidrug resistance-associated protein 4Homo sapiens (human)
urate transportMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
cAMP transportMultidrug resistance-associated protein 4Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 4Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 4Homo sapiens (human)
guanine nucleotide transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
toll-like receptor 2 signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of cytokine-mediated signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
adaptive immune responseReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of T-helper 1 type immune responseReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
apoptotic processReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
inflammatory responseReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
signal transductionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
canonical NF-kappaB signal transductionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
JNK cascadeReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cytokine-mediated signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of protein ubiquitinationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
lipopolysaccharide-mediated signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of protein bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of chemokine productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of interferon-alpha productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of interferon-beta productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of type II interferon productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of interleukin-1 beta productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of interleukin-12 productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of interleukin-2 productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of interleukin-6 productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of tumor necrosis factor productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of stress-activated MAPK cascadeReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
immature T cell proliferation in thymusReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of immature T cell proliferation in thymusReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of peptidyl-serine phosphorylationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
toll-like receptor 4 signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
CD4-positive, alpha-beta T cell proliferationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
defense response to bacteriumReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of apoptotic processReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
response to exogenous dsRNAReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
innate immune responseReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of T-helper 1 cell differentiationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of JNK cascadeReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
defense response to Gram-positive bacteriumReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
T cell receptor signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
protein homooligomerizationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
stress-activated MAPK cascadeReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of macrophage cytokine productionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
ERK1 and ERK2 cascadeReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
nucleotide-binding oligomerization domain containing 1 signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
nucleotide-binding oligomerization domain containing 2 signaling pathwayReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
response to interleukin-1Receptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
response to interleukin-12Receptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
response to interleukin-18Receptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cellular response to lipoteichoic acidReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cellular response to peptidoglycanReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cellular response to muramyl dipeptideReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
activation of cysteine-type endopeptidase activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
xenophagyReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of protein K63-linked ubiquitinationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of xenophagyReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of CD4-positive, alpha-beta T cell proliferationReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
chromatin remodelingMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
apoptotic processMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
chromosome segregationMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
regulation of sister chromatid cohesionMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
mitotic spindle assembly checkpoint signalingMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
cell divisionMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
regulation of chromosome segregationMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
positive regulation of maintenance of mitotic sister chromatid cohesion, centromericMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
meiotic sister chromatid cohesion, centromericMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
mitochondrion organizationDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial genome maintenanceDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial fissionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
neural tube closureDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
apoptotic processDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrion organizationDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
inner mitochondrial membrane organizationDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
visual perceptionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial fusionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
axonal transport of mitochondrionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
positive regulation of interleukin-17 productionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
cristae formationDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
negative regulation of apoptotic processDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
GTP metabolic processDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
protein complex oligomerizationDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
membrane fusionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
negative regulation of release of cytochrome c from mitochondriaDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
cellular senescenceDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
membrane tubulationDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
negative regulation of endoplasmic reticulum stress-induced intrinsic apoptotic signaling pathwayDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial inner membrane fusionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
positive regulation of T-helper 17 cell lineage commitmentDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
regulation of translational initiationEukaryotic translation initiation factor 5BHomo sapiens (human)
ribosome assemblyEukaryotic translation initiation factor 5BHomo sapiens (human)
translational initiationEukaryotic translation initiation factor 5BHomo sapiens (human)
epithelial to mesenchymal transitionRho-associated protein kinase 2Homo sapiens (human)
positive regulation of protein phosphorylationRho-associated protein kinase 2Homo sapiens (human)
response to ischemiaRho-associated protein kinase 2Homo sapiens (human)
aortic valve morphogenesisRho-associated protein kinase 2Homo sapiens (human)
protein phosphorylationRho-associated protein kinase 2Homo sapiens (human)
smooth muscle contractionRho-associated protein kinase 2Homo sapiens (human)
canonical NF-kappaB signal transductionRho-associated protein kinase 2Homo sapiens (human)
positive regulation of endothelial cell migrationRho-associated protein kinase 2Homo sapiens (human)
positive regulation of cardiac muscle hypertrophyRho-associated protein kinase 2Homo sapiens (human)
positive regulation of gene expressionRho-associated protein kinase 2Homo sapiens (human)
negative regulation of gene expressionRho-associated protein kinase 2Homo sapiens (human)
positive regulation of centrosome duplicationRho-associated protein kinase 2Homo sapiens (human)
negative regulation of angiogenesisRho-associated protein kinase 2Homo sapiens (human)
actin cytoskeleton organizationRho-associated protein kinase 2Homo sapiens (human)
regulation of cell adhesionRho-associated protein kinase 2Homo sapiens (human)
positive regulation of cell migrationRho-associated protein kinase 2Homo sapiens (human)
cortical actin cytoskeleton organizationRho-associated protein kinase 2Homo sapiens (human)
regulation of nervous system processRho-associated protein kinase 2Homo sapiens (human)
positive regulation of connective tissue growth factor productionRho-associated protein kinase 2Homo sapiens (human)
regulation of actin cytoskeleton organizationRho-associated protein kinase 2Homo sapiens (human)
negative regulation of myosin-light-chain-phosphatase activityRho-associated protein kinase 2Homo sapiens (human)
regulation of circadian rhythmRho-associated protein kinase 2Homo sapiens (human)
positive regulation of MAPK cascadeRho-associated protein kinase 2Homo sapiens (human)
modulation by host of viral processRho-associated protein kinase 2Homo sapiens (human)
negative regulation of nitric oxide biosynthetic processRho-associated protein kinase 2Homo sapiens (human)
regulation of keratinocyte differentiationRho-associated protein kinase 2Homo sapiens (human)
rhythmic processRho-associated protein kinase 2Homo sapiens (human)
centrosome duplicationRho-associated protein kinase 2Homo sapiens (human)
regulation of stress fiber assemblyRho-associated protein kinase 2Homo sapiens (human)
positive regulation of stress fiber assemblyRho-associated protein kinase 2Homo sapiens (human)
regulation of focal adhesion assemblyRho-associated protein kinase 2Homo sapiens (human)
mRNA destabilizationRho-associated protein kinase 2Homo sapiens (human)
negative regulation of biomineral tissue developmentRho-associated protein kinase 2Homo sapiens (human)
cellular response to testosterone stimulusRho-associated protein kinase 2Homo sapiens (human)
response to transforming growth factor betaRho-associated protein kinase 2Homo sapiens (human)
protein localization to plasma membraneRho-associated protein kinase 2Homo sapiens (human)
positive regulation of fibroblast growth factor productionRho-associated protein kinase 2Homo sapiens (human)
blood vessel diameter maintenanceRho-associated protein kinase 2Homo sapiens (human)
regulation of angiotensin-activated signaling pathwayRho-associated protein kinase 2Homo sapiens (human)
negative regulation of protein localization to lysosomeRho-associated protein kinase 2Homo sapiens (human)
regulation of cellular response to hypoxiaRho-associated protein kinase 2Homo sapiens (human)
positive regulation of amyloid-beta formationRho-associated protein kinase 2Homo sapiens (human)
positive regulation of protein localization to early endosomeRho-associated protein kinase 2Homo sapiens (human)
positive regulation of amyloid precursor protein catabolic processRho-associated protein kinase 2Homo sapiens (human)
regulation of establishment of endothelial barrierRho-associated protein kinase 2Homo sapiens (human)
negative regulation of bicellular tight junction assemblyRho-associated protein kinase 2Homo sapiens (human)
cellular response to acetylcholineRho-associated protein kinase 2Homo sapiens (human)
positive regulation of connective tissue replacementRho-associated protein kinase 2Homo sapiens (human)
response to angiotensinRho-associated protein kinase 2Homo sapiens (human)
regulation of establishment of cell polarityRho-associated protein kinase 2Homo sapiens (human)
regulation of cell motilityRho-associated protein kinase 2Homo sapiens (human)
actomyosin structure organizationRho-associated protein kinase 2Homo sapiens (human)
peptidyl-threonine phosphorylationRho-associated protein kinase 2Homo sapiens (human)
mitotic cytokinesisRho-associated protein kinase 2Homo sapiens (human)
embryonic morphogenesisRho-associated protein kinase 2Homo sapiens (human)
regulation of cell junction assemblyRho-associated protein kinase 2Homo sapiens (human)
Rho protein signal transductionRho-associated protein kinase 2Homo sapiens (human)
autophagosome assemblySerine/threonine-protein kinase ULK1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase ULK1Homo sapiens (human)
autophagySerine/threonine-protein kinase ULK1Homo sapiens (human)
signal transductionSerine/threonine-protein kinase ULK1Homo sapiens (human)
protein localizationSerine/threonine-protein kinase ULK1Homo sapiens (human)
negative regulation of cell population proliferationSerine/threonine-protein kinase ULK1Homo sapiens (human)
positive regulation of autophagySerine/threonine-protein kinase ULK1Homo sapiens (human)
regulation of tumor necrosis factor-mediated signaling pathwaySerine/threonine-protein kinase ULK1Homo sapiens (human)
macroautophagySerine/threonine-protein kinase ULK1Homo sapiens (human)
regulation of macroautophagySerine/threonine-protein kinase ULK1Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase ULK1Homo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase ULK1Homo sapiens (human)
neuron projection regenerationSerine/threonine-protein kinase ULK1Homo sapiens (human)
neuron projection developmentSerine/threonine-protein kinase ULK1Homo sapiens (human)
negative regulation of protein-containing complex assemblySerine/threonine-protein kinase ULK1Homo sapiens (human)
cellular response to nutrient levelsSerine/threonine-protein kinase ULK1Homo sapiens (human)
response to starvationSerine/threonine-protein kinase ULK1Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase ULK1Homo sapiens (human)
regulation of protein lipidationSerine/threonine-protein kinase ULK1Homo sapiens (human)
positive regulation of autophagosome assemblySerine/threonine-protein kinase ULK1Homo sapiens (human)
axon extensionSerine/threonine-protein kinase ULK1Homo sapiens (human)
autophagy of mitochondrionSerine/threonine-protein kinase ULK1Homo sapiens (human)
reticulophagySerine/threonine-protein kinase ULK1Homo sapiens (human)
piecemeal microautophagy of the nucleusSerine/threonine-protein kinase ULK1Homo sapiens (human)
negative regulation of collateral sproutingSerine/threonine-protein kinase ULK1Homo sapiens (human)
endothelial cell proliferationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
mRNA catabolic processSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
regulation of macroautophagySerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
positive regulation of RNA splicingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
cellular response to unfolded proteinSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
response to endoplasmic reticulum stressSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
cellular response to vascular endothelial growth factor stimulusSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
peptidyl-serine autophosphorylationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
IRE1-mediated unfolded protein responseSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
positive regulation of JUN kinase activitySerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
mRNA splicing, via endonucleolytic cleavage and ligationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
cellular response to hydrogen peroxideSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
cellular response to glucose stimulusSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
positive regulation of endoplasmic reticulum unfolded protein responseSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
insulin metabolic processSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
peptidyl-serine trans-autophosphorylationSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
negative regulation of cytokine productionRibosomal protein S6 kinase alpha-5Homo sapiens (human)
chromatin remodelingRibosomal protein S6 kinase alpha-5Homo sapiens (human)
regulation of DNA-templated transcriptionRibosomal protein S6 kinase alpha-5Homo sapiens (human)
protein phosphorylationRibosomal protein S6 kinase alpha-5Homo sapiens (human)
inflammatory responseRibosomal protein S6 kinase alpha-5Homo sapiens (human)
axon guidanceRibosomal protein S6 kinase alpha-5Homo sapiens (human)
positive regulation of CREB transcription factor activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
intracellular signal transductionRibosomal protein S6 kinase alpha-5Homo sapiens (human)
post-translational protein modificationRibosomal protein S6 kinase alpha-5Homo sapiens (human)
negative regulation of DNA-templated transcriptionRibosomal protein S6 kinase alpha-5Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIRibosomal protein S6 kinase alpha-5Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
interleukin-1-mediated signaling pathwayRibosomal protein S6 kinase alpha-5Homo sapiens (human)
regulation of postsynapse organizationRibosomal protein S6 kinase alpha-5Homo sapiens (human)
peptidyl-serine phosphorylationRibosomal protein S6 kinase alpha-5Homo sapiens (human)
cis assembly of pre-catalytic spliceosomeU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
spliceosome conformational change to release U4 (or U4atac) and U1 (or U11)U5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
mRNA splicing, via spliceosomeU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
osteoblast differentiationU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
negative regulation of cytokine productionRibosomal protein S6 kinase alpha-4Homo sapiens (human)
chromatin remodelingRibosomal protein S6 kinase alpha-4Homo sapiens (human)
regulation of DNA-templated transcriptionRibosomal protein S6 kinase alpha-4Homo sapiens (human)
protein phosphorylationRibosomal protein S6 kinase alpha-4Homo sapiens (human)
inflammatory responseRibosomal protein S6 kinase alpha-4Homo sapiens (human)
positive regulation of CREB transcription factor activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
intracellular signal transductionRibosomal protein S6 kinase alpha-4Homo sapiens (human)
post-translational protein modificationRibosomal protein S6 kinase alpha-4Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIRibosomal protein S6 kinase alpha-4Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
interleukin-1-mediated signaling pathwayRibosomal protein S6 kinase alpha-4Homo sapiens (human)
peptidyl-serine phosphorylationRibosomal protein S6 kinase alpha-4Homo sapiens (human)
positive regulation of transcription by RNA polymerase IISerine/threonine-protein kinase 16Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase 16Homo sapiens (human)
cellular response to transforming growth factor beta stimulusSerine/threonine-protein kinase 16Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase 10Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase 10Homo sapiens (human)
lymphocyte aggregationSerine/threonine-protein kinase 10Homo sapiens (human)
regulation of lymphocyte migrationSerine/threonine-protein kinase 10Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase D3Homo sapiens (human)
protein kinase C-activating G protein-coupled receptor signaling pathwaySerine/threonine-protein kinase D3Homo sapiens (human)
sphingolipid biosynthetic processSerine/threonine-protein kinase D3Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase D3Homo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwaySerine/threonine-protein kinase D3Homo sapiens (human)
fatty acid metabolic processBile salt export pumpHomo sapiens (human)
bile acid biosynthetic processBile salt export pumpHomo sapiens (human)
xenobiotic metabolic processBile salt export pumpHomo sapiens (human)
xenobiotic transmembrane transportBile salt export pumpHomo sapiens (human)
response to oxidative stressBile salt export pumpHomo sapiens (human)
bile acid metabolic processBile salt export pumpHomo sapiens (human)
response to organic cyclic compoundBile salt export pumpHomo sapiens (human)
bile acid and bile salt transportBile salt export pumpHomo sapiens (human)
canalicular bile acid transportBile salt export pumpHomo sapiens (human)
protein ubiquitinationBile salt export pumpHomo sapiens (human)
regulation of fatty acid beta-oxidationBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transportBile salt export pumpHomo sapiens (human)
bile acid signaling pathwayBile salt export pumpHomo sapiens (human)
cholesterol homeostasisBile salt export pumpHomo sapiens (human)
response to estrogenBile salt export pumpHomo sapiens (human)
response to ethanolBile salt export pumpHomo sapiens (human)
xenobiotic export from cellBile salt export pumpHomo sapiens (human)
lipid homeostasisBile salt export pumpHomo sapiens (human)
phospholipid homeostasisBile salt export pumpHomo sapiens (human)
positive regulation of bile acid secretionBile salt export pumpHomo sapiens (human)
regulation of bile acid metabolic processBile salt export pumpHomo sapiens (human)
transmembrane transportBile salt export pumpHomo sapiens (human)
mitotic chromosome condensationStructural maintenance of chromosomes protein 2Homo sapiens (human)
meiotic chromosome condensationStructural maintenance of chromosomes protein 2Homo sapiens (human)
meiotic chromosome segregationStructural maintenance of chromosomes protein 2Homo sapiens (human)
cell divisionStructural maintenance of chromosomes protein 2Homo sapiens (human)
kinetochore organizationStructural maintenance of chromosomes protein 2Homo sapiens (human)
positive regulation of chromosome segregationStructural maintenance of chromosomes protein 2Homo sapiens (human)
positive regulation of chromosome separationStructural maintenance of chromosomes protein 2Homo sapiens (human)
positive regulation of chromosome condensationStructural maintenance of chromosomes protein 2Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
signal transductionMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
cellular response to stressMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
microvillus assemblyMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
negative regulation of cell-matrix adhesionMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of cell migrationMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of ARF protein signal transductionMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of hippo signalingMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
negative regulation of apoptotic processMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of GTPase activityMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
regulation of JNK cascadeMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of keratinocyte migrationMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of focal adhesion assemblyMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
positive regulation of focal adhesion disassemblyMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
regulation of MAPK cascadeMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
neuron projection morphogenesisMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
G2/M transition of mitotic cell cycleSerine/threonine-protein kinase LATS1Homo sapiens (human)
sister chromatid segregationSerine/threonine-protein kinase LATS1Homo sapiens (human)
inner cell mass cell fate commitmentSerine/threonine-protein kinase LATS1Homo sapiens (human)
inner cell mass cellular morphogenesisSerine/threonine-protein kinase LATS1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase LATS1Homo sapiens (human)
hormone-mediated signaling pathwaySerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of transforming growth factor beta receptor signaling pathwaySerine/threonine-protein kinase LATS1Homo sapiens (human)
keratinocyte differentiationSerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of actin filament polymerizationSerine/threonine-protein kinase LATS1Homo sapiens (human)
positive regulation of peptidyl-serine phosphorylationSerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of intracellular estrogen receptor signaling pathwaySerine/threonine-protein kinase LATS1Homo sapiens (human)
hippo signalingSerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of protein-containing complex assemblySerine/threonine-protein kinase LATS1Homo sapiens (human)
negative regulation of cyclin-dependent protein serine/threonine kinase activitySerine/threonine-protein kinase LATS1Homo sapiens (human)
cytoplasmic sequestering of proteinSerine/threonine-protein kinase LATS1Homo sapiens (human)
cell divisionSerine/threonine-protein kinase LATS1Homo sapiens (human)
mammary gland epithelial cell differentiationSerine/threonine-protein kinase LATS1Homo sapiens (human)
negative regulation of canonical Wnt signaling pathwaySerine/threonine-protein kinase LATS1Homo sapiens (human)
negative regulation of protein localization to nucleusSerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of ubiquitin-dependent protein catabolic processSerine/threonine-protein kinase LATS1Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase LATS1Homo sapiens (human)
G1/S transition of mitotic cell cycleSerine/threonine-protein kinase LATS1Homo sapiens (human)
positive regulation of apoptotic processSerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of organ growthSerine/threonine-protein kinase LATS1Homo sapiens (human)
regulation of cell growthSerine/threonine-protein kinase PAK 4Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cytoskeleton organizationSerine/threonine-protein kinase PAK 4Homo sapiens (human)
signal transductionSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cell migrationSerine/threonine-protein kinase PAK 4Homo sapiens (human)
positive regulation of angiogenesisSerine/threonine-protein kinase PAK 4Homo sapiens (human)
dendritic spine developmentSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cellular response to organic cyclic compoundSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cell-cell adhesionSerine/threonine-protein kinase PAK 4Homo sapiens (human)
negative regulation of endothelial cell apoptotic processSerine/threonine-protein kinase PAK 4Homo sapiens (human)
regulation of MAPK cascadeSerine/threonine-protein kinase PAK 4Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase PAK 4Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase PAK 4Homo sapiens (human)
response to oxidative stressTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of cytosolic calcium ion concentrationTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of ubiquitin-protein transferase activityTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of phospholipase C activityTyrosine-protein kinase ABL1Homo sapiens (human)
mitotic cell cycleTyrosine-protein kinase ABL1Homo sapiens (human)
neural tube closureTyrosine-protein kinase ABL1Homo sapiens (human)
B-1 B cell homeostasisTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of protein phosphorylationTyrosine-protein kinase ABL1Homo sapiens (human)
B cell proliferation involved in immune responseTyrosine-protein kinase ABL1Homo sapiens (human)
transitional one stage B cell differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
mismatch repairTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of DNA-templated transcriptionTyrosine-protein kinase ABL1Homo sapiens (human)
autophagyTyrosine-protein kinase ABL1Homo sapiens (human)
DNA damage responseTyrosine-protein kinase ABL1Homo sapiens (human)
integrin-mediated signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
canonical NF-kappaB signal transductionTyrosine-protein kinase ABL1Homo sapiens (human)
associative learningTyrosine-protein kinase ABL1Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damageTyrosine-protein kinase ABL1Homo sapiens (human)
response to xenobiotic stimulusTyrosine-protein kinase ABL1Homo sapiens (human)
post-embryonic developmentTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of autophagyTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of endothelial cell migrationTyrosine-protein kinase ABL1Homo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase ABL1Homo sapiens (human)
cerebellum morphogenesisTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of cell-cell adhesionTyrosine-protein kinase ABL1Homo sapiens (human)
microspike assemblyTyrosine-protein kinase ABL1Homo sapiens (human)
actin cytoskeleton organizationTyrosine-protein kinase ABL1Homo sapiens (human)
actin filament polymerizationTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of endocytosisTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of cell adhesionTyrosine-protein kinase ABL1Homo sapiens (human)
neuron differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
BMP signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of BMP signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of axon extensionTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of microtubule polymerizationTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of Cdc42 protein signal transductionTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of type II interferon productionTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of interleukin-2 productionTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of actin cytoskeleton organizationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of osteoblast proliferationTyrosine-protein kinase ABL1Homo sapiens (human)
substrate adhesion-dependent cell spreadingTyrosine-protein kinase ABL1Homo sapiens (human)
cellular response to oxidative stressTyrosine-protein kinase ABL1Homo sapiens (human)
response to endoplasmic reticulum stressTyrosine-protein kinase ABL1Homo sapiens (human)
platelet-derived growth factor receptor-beta signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
protein modification processTyrosine-protein kinase ABL1Homo sapiens (human)
peptidyl-tyrosine autophosphorylationTyrosine-protein kinase ABL1Homo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase ABL1Homo sapiens (human)
neuropilin signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
signal transduction in response to DNA damageTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of apoptotic processTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of neuron apoptotic processTyrosine-protein kinase ABL1Homo sapiens (human)
endothelial cell migrationTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of T cell differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of vasoconstrictionTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of mitotic cell cycleTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of mitotic cell cycleTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IITyrosine-protein kinase ABL1Homo sapiens (human)
alpha-beta T cell differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
protein autophosphorylationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of fibroblast proliferationTyrosine-protein kinase ABL1Homo sapiens (human)
spleen developmentTyrosine-protein kinase ABL1Homo sapiens (human)
thymus developmentTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationTyrosine-protein kinase ABL1Homo sapiens (human)
activated T cell proliferationTyrosine-protein kinase ABL1Homo sapiens (human)
T cell receptor signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
B cell receptor signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
neuromuscular process controlling balanceTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of release of sequestered calcium ion into cytosolTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of oxidoreductase activityTyrosine-protein kinase ABL1Homo sapiens (human)
neuron apoptotic processTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of ubiquitin-protein transferase activityTyrosine-protein kinase ABL1Homo sapiens (human)
myoblast proliferationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of stress fiber assemblyTyrosine-protein kinase ABL1Homo sapiens (human)
establishment of localization in cellTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of cell cycleTyrosine-protein kinase ABL1Homo sapiens (human)
mitochondrial depolarizationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of focal adhesion assemblyTyrosine-protein kinase ABL1Homo sapiens (human)
Bergmann glial cell differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
cardiac muscle cell proliferationTyrosine-protein kinase ABL1Homo sapiens (human)
neuroepithelial cell differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
cellular response to hydrogen peroxideTyrosine-protein kinase ABL1Homo sapiens (human)
ERK1 and ERK2 cascadeTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeTyrosine-protein kinase ABL1Homo sapiens (human)
DNA conformation changeTyrosine-protein kinase ABL1Homo sapiens (human)
cellular response to lipopolysaccharideTyrosine-protein kinase ABL1Homo sapiens (human)
cellular response to transforming growth factor beta stimulusTyrosine-protein kinase ABL1Homo sapiens (human)
response to epinephrineTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of protein serine/threonine kinase activityTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of cell migration involved in sprouting angiogenesisTyrosine-protein kinase ABL1Homo sapiens (human)
cellular senescenceTyrosine-protein kinase ABL1Homo sapiens (human)
cell-cell adhesionTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of dendrite developmentTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of substrate adhesion-dependent cell spreadingTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of long-term synaptic potentiationTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of hematopoietic stem cell differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of extracellular matrix organizationTyrosine-protein kinase ABL1Homo sapiens (human)
podocyte apoptotic processTyrosine-protein kinase ABL1Homo sapiens (human)
cellular response to dopamineTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of establishment of T cell polarityTyrosine-protein kinase ABL1Homo sapiens (human)
DN4 thymocyte differentiationTyrosine-protein kinase ABL1Homo sapiens (human)
protein localization to cytoplasmic microtubule plus-endTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of microtubule bindingTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of actin filament bindingTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of modification of synaptic structureTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of blood vessel branchingTyrosine-protein kinase ABL1Homo sapiens (human)
activation of protein kinase C activityTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of double-strand break repair via homologous recombinationTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of Wnt signaling pathway, planar cell polarity pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
regulation of cell motilityTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of endothelial cell apoptotic processTyrosine-protein kinase ABL1Homo sapiens (human)
positive regulation of T cell migrationTyrosine-protein kinase ABL1Homo sapiens (human)
negative regulation of cellular senescenceTyrosine-protein kinase ABL1Homo sapiens (human)
epidermal growth factor receptor signaling pathwayTyrosine-protein kinase ABL1Homo sapiens (human)
protein phosphorylationTyrosine-protein kinase ABL1Homo sapiens (human)
cell surface receptor signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
epidermal growth factor receptor signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
positive regulation of cell population proliferationEpidermal growth factor receptorHomo sapiens (human)
MAPK cascadeEpidermal growth factor receptorHomo sapiens (human)
ossificationEpidermal growth factor receptorHomo sapiens (human)
embryonic placenta developmentEpidermal growth factor receptorHomo sapiens (human)
positive regulation of protein phosphorylationEpidermal growth factor receptorHomo sapiens (human)
hair follicle developmentEpidermal growth factor receptorHomo sapiens (human)
translationEpidermal growth factor receptorHomo sapiens (human)
signal transductionEpidermal growth factor receptorHomo sapiens (human)
epidermal growth factor receptor signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
activation of phospholipase C activityEpidermal growth factor receptorHomo sapiens (human)
salivary gland morphogenesisEpidermal growth factor receptorHomo sapiens (human)
midgut developmentEpidermal growth factor receptorHomo sapiens (human)
learning or memoryEpidermal growth factor receptorHomo sapiens (human)
circadian rhythmEpidermal growth factor receptorHomo sapiens (human)
positive regulation of cell population proliferationEpidermal growth factor receptorHomo sapiens (human)
diterpenoid metabolic processEpidermal growth factor receptorHomo sapiens (human)
peptidyl-tyrosine phosphorylationEpidermal growth factor receptorHomo sapiens (human)
cerebral cortex cell migrationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of cell growthEpidermal growth factor receptorHomo sapiens (human)
lung developmentEpidermal growth factor receptorHomo sapiens (human)
positive regulation of cell migrationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of superoxide anion generationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationEpidermal growth factor receptorHomo sapiens (human)
response to cobalaminEpidermal growth factor receptorHomo sapiens (human)
response to hydroxyisoflavoneEpidermal growth factor receptorHomo sapiens (human)
cellular response to reactive oxygen speciesEpidermal growth factor receptorHomo sapiens (human)
peptidyl-tyrosine autophosphorylationEpidermal growth factor receptorHomo sapiens (human)
ERBB2-EGFR signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
negative regulation of epidermal growth factor receptor signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
negative regulation of protein catabolic processEpidermal growth factor receptorHomo sapiens (human)
vasodilationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of phosphorylationEpidermal growth factor receptorHomo sapiens (human)
ovulation cycleEpidermal growth factor receptorHomo sapiens (human)
hydrogen peroxide metabolic processEpidermal growth factor receptorHomo sapiens (human)
negative regulation of apoptotic processEpidermal growth factor receptorHomo sapiens (human)
positive regulation of MAP kinase activityEpidermal growth factor receptorHomo sapiens (human)
tongue developmentEpidermal growth factor receptorHomo sapiens (human)
positive regulation of cyclin-dependent protein serine/threonine kinase activityEpidermal growth factor receptorHomo sapiens (human)
positive regulation of DNA repairEpidermal growth factor receptorHomo sapiens (human)
positive regulation of DNA replicationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of bone resorptionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of DNA-templated transcriptionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of vasoconstrictionEpidermal growth factor receptorHomo sapiens (human)
negative regulation of mitotic cell cycleEpidermal growth factor receptorHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIEpidermal growth factor receptorHomo sapiens (human)
regulation of JNK cascadeEpidermal growth factor receptorHomo sapiens (human)
symbiont entry into host cellEpidermal growth factor receptorHomo sapiens (human)
protein autophosphorylationEpidermal growth factor receptorHomo sapiens (human)
astrocyte activationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of fibroblast proliferationEpidermal growth factor receptorHomo sapiens (human)
digestive tract morphogenesisEpidermal growth factor receptorHomo sapiens (human)
positive regulation of smooth muscle cell proliferationEpidermal growth factor receptorHomo sapiens (human)
neuron projection morphogenesisEpidermal growth factor receptorHomo sapiens (human)
epithelial cell proliferationEpidermal growth factor receptorHomo sapiens (human)
positive regulation of epithelial cell proliferationEpidermal growth factor receptorHomo sapiens (human)
regulation of peptidyl-tyrosine phosphorylationEpidermal growth factor receptorHomo sapiens (human)
protein insertion into membraneEpidermal growth factor receptorHomo sapiens (human)
response to calcium ionEpidermal growth factor receptorHomo sapiens (human)
regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicEpidermal growth factor receptorHomo sapiens (human)
positive regulation of glial cell proliferationEpidermal growth factor receptorHomo sapiens (human)
morphogenesis of an epithelial foldEpidermal growth factor receptorHomo sapiens (human)
eyelid development in camera-type eyeEpidermal growth factor receptorHomo sapiens (human)
response to UV-AEpidermal growth factor receptorHomo sapiens (human)
positive regulation of mucus secretionEpidermal growth factor receptorHomo sapiens (human)
regulation of ERK1 and ERK2 cascadeEpidermal growth factor receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeEpidermal growth factor receptorHomo sapiens (human)
cellular response to amino acid stimulusEpidermal growth factor receptorHomo sapiens (human)
cellular response to mechanical stimulusEpidermal growth factor receptorHomo sapiens (human)
cellular response to cadmium ionEpidermal growth factor receptorHomo sapiens (human)
cellular response to epidermal growth factor stimulusEpidermal growth factor receptorHomo sapiens (human)
cellular response to estradiol stimulusEpidermal growth factor receptorHomo sapiens (human)
cellular response to xenobiotic stimulusEpidermal growth factor receptorHomo sapiens (human)
cellular response to dexamethasone stimulusEpidermal growth factor receptorHomo sapiens (human)
positive regulation of canonical Wnt signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
liver regenerationEpidermal growth factor receptorHomo sapiens (human)
cell-cell adhesionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of protein kinase C activityEpidermal growth factor receptorHomo sapiens (human)
positive regulation of G1/S transition of mitotic cell cycleEpidermal growth factor receptorHomo sapiens (human)
positive regulation of non-canonical NF-kappaB signal transductionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of prolactin secretionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of miRNA transcriptionEpidermal growth factor receptorHomo sapiens (human)
positive regulation of protein localization to plasma membraneEpidermal growth factor receptorHomo sapiens (human)
negative regulation of cardiocyte differentiationEpidermal growth factor receptorHomo sapiens (human)
neurogenesisEpidermal growth factor receptorHomo sapiens (human)
multicellular organism developmentEpidermal growth factor receptorHomo sapiens (human)
positive regulation of kinase activityEpidermal growth factor receptorHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayEpidermal growth factor receptorHomo sapiens (human)
cellular response to amyloid-betaHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of protein phosphorylationHigh affinity nerve growth factor receptorHomo sapiens (human)
protein phosphorylationHigh affinity nerve growth factor receptorHomo sapiens (human)
axon guidanceHigh affinity nerve growth factor receptorHomo sapiens (human)
learning or memoryHigh affinity nerve growth factor receptorHomo sapiens (human)
circadian rhythmHigh affinity nerve growth factor receptorHomo sapiens (human)
negative regulation of cell population proliferationHigh affinity nerve growth factor receptorHomo sapiens (human)
response to xenobiotic stimulusHigh affinity nerve growth factor receptorHomo sapiens (human)
programmed cell death involved in cell developmentHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of neuron projection developmentHigh affinity nerve growth factor receptorHomo sapiens (human)
peptidyl-tyrosine phosphorylationHigh affinity nerve growth factor receptorHomo sapiens (human)
olfactory nerve developmentHigh affinity nerve growth factor receptorHomo sapiens (human)
B cell differentiationHigh affinity nerve growth factor receptorHomo sapiens (human)
response to nutrient levelsHigh affinity nerve growth factor receptorHomo sapiens (human)
peptidyl-tyrosine autophosphorylationHigh affinity nerve growth factor receptorHomo sapiens (human)
nerve growth factor signaling pathwayHigh affinity nerve growth factor receptorHomo sapiens (human)
mechanoreceptor differentiationHigh affinity nerve growth factor receptorHomo sapiens (human)
negative regulation of apoptotic processHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of programmed cell deathHigh affinity nerve growth factor receptorHomo sapiens (human)
negative regulation of neuron apoptotic processHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of GTPase activityHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of Ras protein signal transductionHigh affinity nerve growth factor receptorHomo sapiens (human)
protein autophosphorylationHigh affinity nerve growth factor receptorHomo sapiens (human)
neurotrophin TRK receptor signaling pathwayHigh affinity nerve growth factor receptorHomo sapiens (human)
ephrin receptor signaling pathwayHigh affinity nerve growth factor receptorHomo sapiens (human)
sympathetic nervous system developmentHigh affinity nerve growth factor receptorHomo sapiens (human)
response to axon injuryHigh affinity nerve growth factor receptorHomo sapiens (human)
detection of temperature stimulus involved in sensory perception of painHigh affinity nerve growth factor receptorHomo sapiens (human)
detection of mechanical stimulus involved in sensory perception of painHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of NF-kappaB transcription factor activityHigh affinity nerve growth factor receptorHomo sapiens (human)
neuron apoptotic processHigh affinity nerve growth factor receptorHomo sapiens (human)
response to hydrostatic pressureHigh affinity nerve growth factor receptorHomo sapiens (human)
response to electrical stimulusHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of synapse assemblyHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicHigh affinity nerve growth factor receptorHomo sapiens (human)
Sertoli cell developmentHigh affinity nerve growth factor receptorHomo sapiens (human)
axonogenesis involved in innervationHigh affinity nerve growth factor receptorHomo sapiens (human)
behavioral response to formalin induced painHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeHigh affinity nerve growth factor receptorHomo sapiens (human)
cellular response to nicotineHigh affinity nerve growth factor receptorHomo sapiens (human)
cellular response to nerve growth factor stimulusHigh affinity nerve growth factor receptorHomo sapiens (human)
multicellular organism developmentHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of kinase activityHigh affinity nerve growth factor receptorHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeHigh affinity nerve growth factor receptorHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionHigh affinity nerve growth factor receptorHomo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycle G2/M phase transitionCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
ER overload responseCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
mitophagyCellular tumor antigen p53Homo sapiens (human)
in utero embryonic developmentCellular tumor antigen p53Homo sapiens (human)
somitogenesisCellular tumor antigen p53Homo sapiens (human)
release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
hematopoietic progenitor cell differentiationCellular tumor antigen p53Homo sapiens (human)
T cell proliferation involved in immune responseCellular tumor antigen p53Homo sapiens (human)
B cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
T cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
response to ischemiaCellular tumor antigen p53Homo sapiens (human)
nucleotide-excision repairCellular tumor antigen p53Homo sapiens (human)
double-strand break repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
protein import into nucleusCellular tumor antigen p53Homo sapiens (human)
autophagyCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorCellular tumor antigen p53Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
Ras protein signal transductionCellular tumor antigen p53Homo sapiens (human)
gastrulationCellular tumor antigen p53Homo sapiens (human)
neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
protein localizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA replicationCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
determination of adult lifespanCellular tumor antigen p53Homo sapiens (human)
mRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
rRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
response to salt stressCellular tumor antigen p53Homo sapiens (human)
response to inorganic substanceCellular tumor antigen p53Homo sapiens (human)
response to X-rayCellular tumor antigen p53Homo sapiens (human)
response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
positive regulation of gene expressionCellular tumor antigen p53Homo sapiens (human)
cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
viral processCellular tumor antigen p53Homo sapiens (human)
glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
cerebellum developmentCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell growthCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCellular tumor antigen p53Homo sapiens (human)
negative regulation of telomere maintenance via telomeraseCellular tumor antigen p53Homo sapiens (human)
T cell differentiation in thymusCellular tumor antigen p53Homo sapiens (human)
tumor necrosis factor-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
regulation of tissue remodelingCellular tumor antigen p53Homo sapiens (human)
cellular response to UVCellular tumor antigen p53Homo sapiens (human)
multicellular organism growthCellular tumor antigen p53Homo sapiens (human)
positive regulation of mitochondrial membrane permeabilityCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
entrainment of circadian clock by photoperiodCellular tumor antigen p53Homo sapiens (human)
mitochondrial DNA repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
transcription initiation-coupled chromatin remodelingCellular tumor antigen p53Homo sapiens (human)
negative regulation of proteolysisCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of RNA polymerase II transcription preinitiation complex assemblyCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
response to antibioticCellular tumor antigen p53Homo sapiens (human)
fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
circadian behaviorCellular tumor antigen p53Homo sapiens (human)
bone marrow developmentCellular tumor antigen p53Homo sapiens (human)
embryonic organ developmentCellular tumor antigen p53Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationCellular tumor antigen p53Homo sapiens (human)
protein stabilizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of helicase activityCellular tumor antigen p53Homo sapiens (human)
protein tetramerizationCellular tumor antigen p53Homo sapiens (human)
chromosome organizationCellular tumor antigen p53Homo sapiens (human)
neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
hematopoietic stem cell differentiationCellular tumor antigen p53Homo sapiens (human)
negative regulation of glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
type II interferon-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
cardiac septum morphogenesisCellular tumor antigen p53Homo sapiens (human)
positive regulation of programmed necrotic cell deathCellular tumor antigen p53Homo sapiens (human)
protein-containing complex assemblyCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressCellular tumor antigen p53Homo sapiens (human)
thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
necroptotic processCellular tumor antigen p53Homo sapiens (human)
cellular response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
cellular response to xenobiotic stimulusCellular tumor antigen p53Homo sapiens (human)
cellular response to ionizing radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to UV-CCellular tumor antigen p53Homo sapiens (human)
stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
cellular response to actinomycin DCellular tumor antigen p53Homo sapiens (human)
positive regulation of release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
cellular senescenceCellular tumor antigen p53Homo sapiens (human)
replicative senescenceCellular tumor antigen p53Homo sapiens (human)
oxidative stress-induced premature senescenceCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
oligodendrocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of execution phase of apoptosisCellular tumor antigen p53Homo sapiens (human)
negative regulation of mitophagyCellular tumor antigen p53Homo sapiens (human)
regulation of mitochondrial membrane permeability involved in apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of G1 to G0 transitionCellular tumor antigen p53Homo sapiens (human)
negative regulation of miRNA processingCellular tumor antigen p53Homo sapiens (human)
negative regulation of glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
negative regulation of pentose-phosphate shuntCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
regulation of fibroblast apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
positive regulation of cellular senescenceCellular tumor antigen p53Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
signal transductionGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
G protein-coupled receptor signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
negative regulation of adenylate cyclase activityGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
G protein-coupled acetylcholine receptor signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
response to nutrientGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
cell population proliferationGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of cell population proliferationGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of cell migrationGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of superoxide anion generationGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of urine volumeGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
negative regulation of calcium ion-dependent exocytosisGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of insulin receptor signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
negative regulation of synaptic transmissionGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
cell divisionGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
regulation of calcium ion transportGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
negative regulation of adenylate cyclase-activating adrenergic receptor signaling pathway involved in heart processGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
positive regulation of neural precursor cell proliferationGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
negative regulation of apoptotic signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
G protein-coupled adenosine receptor signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
chromosome segregationADP/ATP translocase 2Homo sapiens (human)
positive regulation of cell population proliferationADP/ATP translocase 2Homo sapiens (human)
adenine transportADP/ATP translocase 2Homo sapiens (human)
B cell differentiationADP/ATP translocase 2Homo sapiens (human)
erythrocyte differentiationADP/ATP translocase 2Homo sapiens (human)
regulation of mitochondrial membrane permeabilityADP/ATP translocase 2Homo sapiens (human)
adenine nucleotide transportADP/ATP translocase 2Homo sapiens (human)
mitochondrial ADP transmembrane transportADP/ATP translocase 2Homo sapiens (human)
negative regulation of mitochondrial outer membrane permeabilization involved in apoptotic signaling pathwayADP/ATP translocase 2Homo sapiens (human)
positive regulation of mitophagyADP/ATP translocase 2Homo sapiens (human)
proton transmembrane transportADP/ATP translocase 2Homo sapiens (human)
mitochondrial ATP transmembrane transportADP/ATP translocase 2Homo sapiens (human)
cellular response to leukemia inhibitory factorADP/ATP translocase 2Homo sapiens (human)
adaptive thermogenesisADP/ATP translocase 2Homo sapiens (human)
adaptive immune responseProtein kinase C beta typeHomo sapiens (human)
chromatin remodelingProtein kinase C beta typeHomo sapiens (human)
regulation of transcription by RNA polymerase IIProtein kinase C beta typeHomo sapiens (human)
protein phosphorylationProtein kinase C beta typeHomo sapiens (human)
calcium ion transportProtein kinase C beta typeHomo sapiens (human)
intracellular calcium ion homeostasisProtein kinase C beta typeHomo sapiens (human)
apoptotic processProtein kinase C beta typeHomo sapiens (human)
mitotic nuclear membrane disassemblyProtein kinase C beta typeHomo sapiens (human)
signal transductionProtein kinase C beta typeHomo sapiens (human)
phospholipase C-activating G protein-coupled acetylcholine receptor signaling pathwayProtein kinase C beta typeHomo sapiens (human)
response to xenobiotic stimulusProtein kinase C beta typeHomo sapiens (human)
response to glucoseProtein kinase C beta typeHomo sapiens (human)
regulation of glucose transmembrane transportProtein kinase C beta typeHomo sapiens (human)
negative regulation of glucose transmembrane transportProtein kinase C beta typeHomo sapiens (human)
regulation of dopamine secretionProtein kinase C beta typeHomo sapiens (human)
dibenzo-p-dioxin metabolic processProtein kinase C beta typeHomo sapiens (human)
positive regulation of vascular endothelial growth factor receptor signaling pathwayProtein kinase C beta typeHomo sapiens (human)
positive regulation of insulin secretionProtein kinase C beta typeHomo sapiens (human)
response to vitamin DProtein kinase C beta typeHomo sapiens (human)
regulation of growthProtein kinase C beta typeHomo sapiens (human)
B cell activationProtein kinase C beta typeHomo sapiens (human)
positive regulation of odontogenesis of dentin-containing toothProtein kinase C beta typeHomo sapiens (human)
lipoprotein transportProtein kinase C beta typeHomo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionProtein kinase C beta typeHomo sapiens (human)
post-translational protein modificationProtein kinase C beta typeHomo sapiens (human)
response to ethanolProtein kinase C beta typeHomo sapiens (human)
positive regulation of angiogenesisProtein kinase C beta typeHomo sapiens (human)
positive regulation of DNA-templated transcriptionProtein kinase C beta typeHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayProtein kinase C beta typeHomo sapiens (human)
B cell receptor signaling pathwayProtein kinase C beta typeHomo sapiens (human)
positive regulation of B cell receptor signaling pathwayProtein kinase C beta typeHomo sapiens (human)
cellular response to carbohydrate stimulusProtein kinase C beta typeHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionProtein kinase C beta typeHomo sapiens (human)
regulation of synaptic vesicle exocytosisProtein kinase C beta typeHomo sapiens (human)
peptidyl-serine phosphorylationProtein kinase C beta typeHomo sapiens (human)
intracellular signal transductionProtein kinase C beta typeHomo sapiens (human)
positive regulation of MAP kinase activityInsulin receptorHomo sapiens (human)
positive regulation of protein phosphorylationInsulin receptorHomo sapiens (human)
positive regulation of receptor internalizationInsulin receptorHomo sapiens (human)
heart morphogenesisInsulin receptorHomo sapiens (human)
regulation of DNA-templated transcriptionInsulin receptorHomo sapiens (human)
protein phosphorylationInsulin receptorHomo sapiens (human)
receptor-mediated endocytosisInsulin receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayInsulin receptorHomo sapiens (human)
learningInsulin receptorHomo sapiens (human)
memoryInsulin receptorHomo sapiens (human)
positive regulation of cell population proliferationInsulin receptorHomo sapiens (human)
insulin receptor signaling pathwayInsulin receptorHomo sapiens (human)
epidermis developmentInsulin receptorHomo sapiens (human)
male gonad developmentInsulin receptorHomo sapiens (human)
peptidyl-tyrosine phosphorylationInsulin receptorHomo sapiens (human)
male sex determinationInsulin receptorHomo sapiens (human)
adrenal gland developmentInsulin receptorHomo sapiens (human)
positive regulation of cell migrationInsulin receptorHomo sapiens (human)
exocrine pancreas developmentInsulin receptorHomo sapiens (human)
receptor internalizationInsulin receptorHomo sapiens (human)
activation of protein kinase activityInsulin receptorHomo sapiens (human)
activation of protein kinase B activityInsulin receptorHomo sapiens (human)
cellular response to insulin stimulusInsulin receptorHomo sapiens (human)
glucose homeostasisInsulin receptorHomo sapiens (human)
positive regulation of protein-containing complex disassemblyInsulin receptorHomo sapiens (human)
positive regulation of MAPK cascadeInsulin receptorHomo sapiens (human)
positive regulation of nitric oxide biosynthetic processInsulin receptorHomo sapiens (human)
positive regulation of glycogen biosynthetic processInsulin receptorHomo sapiens (human)
positive regulation of glycolytic processInsulin receptorHomo sapiens (human)
positive regulation of mitotic nuclear divisionInsulin receptorHomo sapiens (human)
positive regulation of DNA-templated transcriptionInsulin receptorHomo sapiens (human)
regulation of embryonic developmentInsulin receptorHomo sapiens (human)
positive regulation of glucose importInsulin receptorHomo sapiens (human)
symbiont entry into host cellInsulin receptorHomo sapiens (human)
protein autophosphorylationInsulin receptorHomo sapiens (human)
positive regulation of developmental growthInsulin receptorHomo sapiens (human)
positive regulation of meiotic cell cycleInsulin receptorHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionInsulin receptorHomo sapiens (human)
positive regulation of respiratory burstInsulin receptorHomo sapiens (human)
cellular response to growth factor stimulusInsulin receptorHomo sapiens (human)
dendritic spine maintenanceInsulin receptorHomo sapiens (human)
amyloid-beta clearanceInsulin receptorHomo sapiens (human)
transport across blood-brain barrierInsulin receptorHomo sapiens (human)
neuron projection maintenanceInsulin receptorHomo sapiens (human)
regulation of female gonad developmentInsulin receptorHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayInsulin receptorHomo sapiens (human)
multicellular organism developmentInsulin receptorHomo sapiens (human)
positive regulation of kinase activityInsulin receptorHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase LckHomo sapiens (human)
intracellular zinc ion homeostasisTyrosine-protein kinase LckHomo sapiens (human)
activation of cysteine-type endopeptidase activity involved in apoptotic processTyrosine-protein kinase LckHomo sapiens (human)
response to xenobiotic stimulusTyrosine-protein kinase LckHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase LckHomo sapiens (human)
hemopoiesisTyrosine-protein kinase LckHomo sapiens (human)
platelet activationTyrosine-protein kinase LckHomo sapiens (human)
T cell differentiationTyrosine-protein kinase LckHomo sapiens (human)
T cell costimulationTyrosine-protein kinase LckHomo sapiens (human)
positive regulation of heterotypic cell-cell adhesionTyrosine-protein kinase LckHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase LckHomo sapiens (human)
peptidyl-tyrosine autophosphorylationTyrosine-protein kinase LckHomo sapiens (human)
Fc-gamma receptor signaling pathwayTyrosine-protein kinase LckHomo sapiens (human)
T cell receptor signaling pathwayTyrosine-protein kinase LckHomo sapiens (human)
positive regulation of T cell receptor signaling pathwayTyrosine-protein kinase LckHomo sapiens (human)
positive regulation of T cell activationTyrosine-protein kinase LckHomo sapiens (human)
leukocyte migrationTyrosine-protein kinase LckHomo sapiens (human)
release of sequestered calcium ion into cytosolTyrosine-protein kinase LckHomo sapiens (human)
regulation of lymphocyte activationTyrosine-protein kinase LckHomo sapiens (human)
positive regulation of leukocyte cell-cell adhesionTyrosine-protein kinase LckHomo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayTyrosine-protein kinase LckHomo sapiens (human)
innate immune responseTyrosine-protein kinase LckHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase LckHomo sapiens (human)
B cell receptor signaling pathwayTyrosine-protein kinase LckHomo sapiens (human)
response to singlet oxygenTyrosine-protein kinase FynHomo sapiens (human)
neuron migrationTyrosine-protein kinase FynHomo sapiens (human)
stimulatory C-type lectin receptor signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
adaptive immune responseTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusTyrosine-protein kinase FynHomo sapiens (human)
heart processTyrosine-protein kinase FynHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase FynHomo sapiens (human)
calcium ion transportTyrosine-protein kinase FynHomo sapiens (human)
G protein-coupled glutamate receptor signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
axon guidanceTyrosine-protein kinase FynHomo sapiens (human)
learningTyrosine-protein kinase FynHomo sapiens (human)
feeding behaviorTyrosine-protein kinase FynHomo sapiens (human)
regulation of cell shapeTyrosine-protein kinase FynHomo sapiens (human)
gene expressionTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of gene expressionTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of hydrogen peroxide biosynthetic processTyrosine-protein kinase FynHomo sapiens (human)
positive regulation of neuron projection developmentTyrosine-protein kinase FynHomo sapiens (human)
protein ubiquitinationTyrosine-protein kinase FynHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase FynHomo sapiens (human)
protein catabolic processTyrosine-protein kinase FynHomo sapiens (human)
forebrain developmentTyrosine-protein kinase FynHomo sapiens (human)
T cell costimulationTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of protein ubiquitinationTyrosine-protein kinase FynHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase FynHomo sapiens (human)
cellular response to platelet-derived growth factor stimulusTyrosine-protein kinase FynHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of protein catabolic processTyrosine-protein kinase FynHomo sapiens (human)
positive regulation of tyrosine phosphorylation of STAT proteinTyrosine-protein kinase FynHomo sapiens (human)
response to ethanolTyrosine-protein kinase FynHomo sapiens (human)
vascular endothelial growth factor receptor signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
ephrin receptor signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
dendrite morphogenesisTyrosine-protein kinase FynHomo sapiens (human)
regulation of peptidyl-tyrosine phosphorylationTyrosine-protein kinase FynHomo sapiens (human)
activated T cell proliferationTyrosine-protein kinase FynHomo sapiens (human)
modulation of chemical synaptic transmissionTyrosine-protein kinase FynHomo sapiens (human)
T cell receptor signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
leukocyte migrationTyrosine-protein kinase FynHomo sapiens (human)
detection of mechanical stimulus involved in sensory perception of painTyrosine-protein kinase FynHomo sapiens (human)
cellular response to hydrogen peroxideTyrosine-protein kinase FynHomo sapiens (human)
cellular response to transforming growth factor beta stimulusTyrosine-protein kinase FynHomo sapiens (human)
positive regulation of protein targeting to membraneTyrosine-protein kinase FynHomo sapiens (human)
dendritic spine maintenanceTyrosine-protein kinase FynHomo sapiens (human)
positive regulation of protein localization to nucleusTyrosine-protein kinase FynHomo sapiens (human)
regulation of glutamate receptor signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of oxidative stress-induced intrinsic apoptotic signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
negative regulation of dendritic spine maintenanceTyrosine-protein kinase FynHomo sapiens (human)
response to amyloid-betaTyrosine-protein kinase FynHomo sapiens (human)
cellular response to amyloid-betaTyrosine-protein kinase FynHomo sapiens (human)
cellular response to L-glutamateTyrosine-protein kinase FynHomo sapiens (human)
cellular response to glycineTyrosine-protein kinase FynHomo sapiens (human)
positive regulation of protein localization to membraneTyrosine-protein kinase FynHomo sapiens (human)
regulation of calcium ion import across plasma membraneTyrosine-protein kinase FynHomo sapiens (human)
positive regulation of cysteine-type endopeptidase activityTyrosine-protein kinase FynHomo sapiens (human)
innate immune responseTyrosine-protein kinase FynHomo sapiens (human)
cell differentiationTyrosine-protein kinase FynHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase FynHomo sapiens (human)
G1/S transition of mitotic cell cycleCyclin-dependent kinase 1Homo sapiens (human)
G2/M transition of mitotic cell cycleCyclin-dependent kinase 1Homo sapiens (human)
microtubule cytoskeleton organizationCyclin-dependent kinase 1Homo sapiens (human)
DNA replicationCyclin-dependent kinase 1Homo sapiens (human)
DNA repairCyclin-dependent kinase 1Homo sapiens (human)
chromatin remodelingCyclin-dependent kinase 1Homo sapiens (human)
regulation of transcription by RNA polymerase IICyclin-dependent kinase 1Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 1Homo sapiens (human)
apoptotic processCyclin-dependent kinase 1Homo sapiens (human)
DNA damage responseCyclin-dependent kinase 1Homo sapiens (human)
mitotic nuclear membrane disassemblyCyclin-dependent kinase 1Homo sapiens (human)
centrosome cycleCyclin-dependent kinase 1Homo sapiens (human)
pronuclear fusionCyclin-dependent kinase 1Homo sapiens (human)
response to xenobiotic stimulusCyclin-dependent kinase 1Homo sapiens (human)
response to toxic substanceCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of gene expressionCyclin-dependent kinase 1Homo sapiens (human)
negative regulation of gene expressionCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of G2/M transition of mitotic cell cycleCyclin-dependent kinase 1Homo sapiens (human)
regulation of Schwann cell differentiationCyclin-dependent kinase 1Homo sapiens (human)
response to amineCyclin-dependent kinase 1Homo sapiens (human)
response to activityCyclin-dependent kinase 1Homo sapiens (human)
cell migrationCyclin-dependent kinase 1Homo sapiens (human)
peptidyl-serine phosphorylationCyclin-dependent kinase 1Homo sapiens (human)
peptidyl-threonine phosphorylationCyclin-dependent kinase 1Homo sapiens (human)
chromosome condensationCyclin-dependent kinase 1Homo sapiens (human)
epithelial cell differentiationCyclin-dependent kinase 1Homo sapiens (human)
animal organ regenerationCyclin-dependent kinase 1Homo sapiens (human)
protein localization to kinetochoreCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of protein import into nucleusCyclin-dependent kinase 1Homo sapiens (human)
regulation of circadian rhythmCyclin-dependent kinase 1Homo sapiens (human)
negative regulation of apoptotic processCyclin-dependent kinase 1Homo sapiens (human)
response to ethanolCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of DNA replicationCyclin-dependent kinase 1Homo sapiens (human)
regulation of embryonic developmentCyclin-dependent kinase 1Homo sapiens (human)
response to cadmium ionCyclin-dependent kinase 1Homo sapiens (human)
response to copper ionCyclin-dependent kinase 1Homo sapiens (human)
symbiont entry into host cellCyclin-dependent kinase 1Homo sapiens (human)
fibroblast proliferationCyclin-dependent kinase 1Homo sapiens (human)
rhythmic processCyclin-dependent kinase 1Homo sapiens (human)
response to axon injuryCyclin-dependent kinase 1Homo sapiens (human)
cell divisionCyclin-dependent kinase 1Homo sapiens (human)
ventricular cardiac muscle cell developmentCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of cardiac muscle cell proliferationCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of mitotic sister chromatid segregationCyclin-dependent kinase 1Homo sapiens (human)
protein-containing complex assemblyCyclin-dependent kinase 1Homo sapiens (human)
cellular response to hydrogen peroxideCyclin-dependent kinase 1Homo sapiens (human)
ERK1 and ERK2 cascadeCyclin-dependent kinase 1Homo sapiens (human)
cellular response to organic cyclic compoundCyclin-dependent kinase 1Homo sapiens (human)
Golgi disassemblyCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of protein localization to nucleusCyclin-dependent kinase 1Homo sapiens (human)
regulation of attachment of mitotic spindle microtubules to kinetochoreCyclin-dependent kinase 1Homo sapiens (human)
microtubule cytoskeleton organization involved in mitosisCyclin-dependent kinase 1Homo sapiens (human)
positive regulation of mitochondrial ATP synthesis coupled electron transportCyclin-dependent kinase 1Homo sapiens (human)
mitotic G2 DNA damage checkpoint signalingCyclin-dependent kinase 1Homo sapiens (human)
protein deubiquitinationCyclin-dependent kinase 1Homo sapiens (human)
glycogen metabolic processGlycogen phosphorylase, liver formHomo sapiens (human)
5-phosphoribose 1-diphosphate biosynthetic processGlycogen phosphorylase, liver formHomo sapiens (human)
response to bacteriumGlycogen phosphorylase, liver formHomo sapiens (human)
glucose homeostasisGlycogen phosphorylase, liver formHomo sapiens (human)
necroptotic processGlycogen phosphorylase, liver formHomo sapiens (human)
glycogen catabolic processGlycogen phosphorylase, liver formHomo sapiens (human)
microtubule bundle formationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
centrosome cycleTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of cell shapeTyrosine-protein kinase Fes/FpsHomo sapiens (human)
positive regulation of neuron projection developmentTyrosine-protein kinase Fes/FpsHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of cell adhesionTyrosine-protein kinase Fes/FpsHomo sapiens (human)
positive regulation of microtubule polymerizationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of cell population proliferationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of mast cell degranulationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of cell differentiationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
positive regulation of myeloid cell differentiationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
positive regulation of monocyte differentiationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
protein autophosphorylationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
myoblast proliferationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
cardiac muscle cell proliferationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of vesicle-mediated transportTyrosine-protein kinase Fes/FpsHomo sapiens (human)
cellular response to vitamin DTyrosine-protein kinase Fes/FpsHomo sapiens (human)
regulation of cell motilityTyrosine-protein kinase Fes/FpsHomo sapiens (human)
chemotaxisTyrosine-protein kinase Fes/FpsHomo sapiens (human)
cell adhesionTyrosine-protein kinase Fes/FpsHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase Fes/FpsHomo sapiens (human)
purine ribonucleoside salvageAdenine phosphoribosyltransferaseHomo sapiens (human)
grooming behaviorAdenine phosphoribosyltransferaseHomo sapiens (human)
GMP salvageAdenine phosphoribosyltransferaseHomo sapiens (human)
IMP salvageAdenine phosphoribosyltransferaseHomo sapiens (human)
AMP salvageAdenine phosphoribosyltransferaseHomo sapiens (human)
adenine salvageAdenine phosphoribosyltransferaseHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusTyrosine-protein kinase YesHomo sapiens (human)
regulation of glucose transmembrane transportTyrosine-protein kinase YesHomo sapiens (human)
T cell costimulationTyrosine-protein kinase YesHomo sapiens (human)
cellular response to platelet-derived growth factor stimulusTyrosine-protein kinase YesHomo sapiens (human)
protein modification processTyrosine-protein kinase YesHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase YesHomo sapiens (human)
regulation of vascular permeabilityTyrosine-protein kinase YesHomo sapiens (human)
positive regulation of transcription by RNA polymerase IITyrosine-protein kinase YesHomo sapiens (human)
ephrin receptor signaling pathwayTyrosine-protein kinase YesHomo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationTyrosine-protein kinase YesHomo sapiens (human)
leukocyte migrationTyrosine-protein kinase YesHomo sapiens (human)
cellular response to retinoic acidTyrosine-protein kinase YesHomo sapiens (human)
cellular response to transforming growth factor beta stimulusTyrosine-protein kinase YesHomo sapiens (human)
innate immune responseTyrosine-protein kinase YesHomo sapiens (human)
cell differentiationTyrosine-protein kinase YesHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase YesHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase YesHomo sapiens (human)
DNA damage checkpoint signalingTyrosine-protein kinase LynHomo sapiens (human)
B cell homeostasisTyrosine-protein kinase LynHomo sapiens (human)
regulation of cytokine productionTyrosine-protein kinase LynHomo sapiens (human)
regulation of protein phosphorylationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of protein phosphorylationTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of protein phosphorylationTyrosine-protein kinase LynHomo sapiens (human)
stimulatory C-type lectin receptor signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
hematopoietic progenitor cell differentiationTyrosine-protein kinase LynHomo sapiens (human)
adaptive immune responseTyrosine-protein kinase LynHomo sapiens (human)
Fc receptor mediated stimulatory signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
tolerance induction to self antigenTyrosine-protein kinase LynHomo sapiens (human)
histamine secretion by mast cellTyrosine-protein kinase LynHomo sapiens (human)
platelet degranulationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of myeloid leukocyte differentiationTyrosine-protein kinase LynHomo sapiens (human)
immune response-regulating cell surface receptor signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
Fc receptor mediated inhibitory signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusTyrosine-protein kinase LynHomo sapiens (human)
regulation of B cell apoptotic processTyrosine-protein kinase LynHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase LynHomo sapiens (human)
DNA damage responseTyrosine-protein kinase LynHomo sapiens (human)
response to sterol depletionTyrosine-protein kinase LynHomo sapiens (human)
signal transductionTyrosine-protein kinase LynHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of cell population proliferationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of cell population proliferationTyrosine-protein kinase LynHomo sapiens (human)
response to xenobiotic stimulusTyrosine-protein kinase LynHomo sapiens (human)
response to toxic substanceTyrosine-protein kinase LynHomo sapiens (human)
response to hormoneTyrosine-protein kinase LynHomo sapiens (human)
response to carbohydrateTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of neuron projection developmentTyrosine-protein kinase LynHomo sapiens (human)
oligodendrocyte developmentTyrosine-protein kinase LynHomo sapiens (human)
response to organic cyclic compoundTyrosine-protein kinase LynHomo sapiens (human)
fatty acid transportTyrosine-protein kinase LynHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase LynHomo sapiens (human)
erythrocyte differentiationTyrosine-protein kinase LynHomo sapiens (human)
eosinophil differentiationTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of cell migrationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of B cell proliferationTyrosine-protein kinase LynHomo sapiens (human)
T cell costimulationTyrosine-protein kinase LynHomo sapiens (human)
lipopolysaccharide-mediated signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
response to insulinTyrosine-protein kinase LynHomo sapiens (human)
regulation of mast cell activationTyrosine-protein kinase LynHomo sapiens (human)
regulation of cell adhesion mediated by integrinTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of toll-like receptor 2 signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
toll-like receptor 4 signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of toll-like receptor 4 signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
cellular response to heatTyrosine-protein kinase LynHomo sapiens (human)
interleukin-5-mediated signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
Fc-epsilon receptor signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase LynHomo sapiens (human)
C-X-C chemokine receptor CXCR4 signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of tyrosine phosphorylation of STAT proteinTyrosine-protein kinase LynHomo sapiens (human)
response to amino acidTyrosine-protein kinase LynHomo sapiens (human)
regulation of mast cell degranulationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of MAP kinase activityTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of MAPK cascadeTyrosine-protein kinase LynHomo sapiens (human)
regulation of erythrocyte differentiationTyrosine-protein kinase LynHomo sapiens (human)
protein autophosphorylationTyrosine-protein kinase LynHomo sapiens (human)
ephrin receptor signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
response to axon injuryTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of immune responseTyrosine-protein kinase LynHomo sapiens (human)
B cell receptor signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
regulation of B cell receptor signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
leukocyte migrationTyrosine-protein kinase LynHomo sapiens (human)
regulation of release of sequestered calcium ion into cytosolTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of glial cell proliferationTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of Fc receptor mediated stimulatory signaling pathwayTyrosine-protein kinase LynHomo sapiens (human)
growth hormone receptor signaling pathway via JAK-STATTyrosine-protein kinase LynHomo sapiens (human)
regulation of ERK1 and ERK2 cascadeTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of oligodendrocyte progenitor proliferationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of mast cell proliferationTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of mast cell proliferationTyrosine-protein kinase LynHomo sapiens (human)
cellular response to retinoic acidTyrosine-protein kinase LynHomo sapiens (human)
regulation of monocyte chemotaxisTyrosine-protein kinase LynHomo sapiens (human)
regulation of platelet aggregationTyrosine-protein kinase LynHomo sapiens (human)
dendritic cell differentiationTyrosine-protein kinase LynHomo sapiens (human)
negative regulation of intracellular signal transductionTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of aspartic-type endopeptidase activity involved in amyloid precursor protein catabolic processTyrosine-protein kinase LynHomo sapiens (human)
positive regulation of dendritic cell apoptotic processTyrosine-protein kinase LynHomo sapiens (human)
neuron projection developmentTyrosine-protein kinase LynHomo sapiens (human)
innate immune responseTyrosine-protein kinase LynHomo sapiens (human)
MAPK cascadeProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
ureteric bud developmentProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
neural crest cell migrationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
embryonic epithelial tube formationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
protein phosphorylationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
homophilic cell adhesion via plasma membrane adhesion moleculesProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
neuron cell-cell adhesionProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
signal transductionProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
axon guidanceProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
posterior midgut developmentProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
response to xenobiotic stimulusProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of gene expressionProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of neuron projection developmentProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of neuron maturationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
regulation of cell adhesionProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of cell migrationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylation of STAT proteinProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
membrane protein proteolysisProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of cell adhesion mediated by integrinProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
ureter maturationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
glial cell-derived neurotrophic factor receptor signaling pathwayProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
neuron maturationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of MAPK cascadeProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of cell sizeProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of DNA-templated transcriptionProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
response to painProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
enteric nervous system developmentProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
regulation of axonogenesisProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
retina development in camera-type eyeProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
innervationProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
Peyer's patch morphogenesisProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
cellular response to retinoic acidProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of metanephric glomerulus developmentProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
lymphocyte migration into lymphoid organsProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
GDF15-GFRAL signaling pathwayProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of extrinsic apoptotic signaling pathway in absence of ligandProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
positive regulation of kinase activityProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
multicellular organism developmentProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
peptidyl-tyrosine autophosphorylationInsulin-like growth factor 1 receptorHomo sapiens (human)
cardiac atrium developmentInsulin-like growth factor 1 receptorHomo sapiens (human)
immune responseInsulin-like growth factor 1 receptorHomo sapiens (human)
signal transductionInsulin-like growth factor 1 receptorHomo sapiens (human)
axonogenesisInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of cell population proliferationInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin receptor signaling pathwayInsulin-like growth factor 1 receptorHomo sapiens (human)
negative regulation of muscle cell apoptotic processInsulin-like growth factor 1 receptorHomo sapiens (human)
cerebellum developmentInsulin-like growth factor 1 receptorHomo sapiens (human)
hippocampus developmentInsulin-like growth factor 1 receptorHomo sapiens (human)
establishment of cell polarityInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of cell migrationInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of cytokinesisInsulin-like growth factor 1 receptorHomo sapiens (human)
response to vitamin EInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of osteoblast proliferationInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to zinc ion starvationInsulin-like growth factor 1 receptorHomo sapiens (human)
response to nicotineInsulin-like growth factor 1 receptorHomo sapiens (human)
negative regulation of apoptotic processInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of protein-containing complex disassemblyInsulin-like growth factor 1 receptorHomo sapiens (human)
response to alkaloidInsulin-like growth factor 1 receptorHomo sapiens (human)
negative regulation of MAPK cascadeInsulin-like growth factor 1 receptorHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionInsulin-like growth factor 1 receptorHomo sapiens (human)
estrous cycleInsulin-like growth factor 1 receptorHomo sapiens (human)
transcytosisInsulin-like growth factor 1 receptorHomo sapiens (human)
response to ethanolInsulin-like growth factor 1 receptorHomo sapiens (human)
regulation of JNK cascadeInsulin-like growth factor 1 receptorHomo sapiens (human)
protein autophosphorylationInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin-like growth factor receptor signaling pathwayInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of smooth muscle cell proliferationInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of axon regenerationInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of DNA metabolic processInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to mechanical stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to estradiol stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to progesterone stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to testosterone stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to dexamethasone stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to transforming growth factor beta stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of steroid hormone biosynthetic processInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular senescenceInsulin-like growth factor 1 receptorHomo sapiens (human)
dendritic spine maintenanceInsulin-like growth factor 1 receptorHomo sapiens (human)
amyloid-beta clearanceInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of cold-induced thermogenesisInsulin-like growth factor 1 receptorHomo sapiens (human)
response to L-glutamateInsulin-like growth factor 1 receptorHomo sapiens (human)
negative regulation of hepatocyte apoptotic processInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to aldosteroneInsulin-like growth factor 1 receptorHomo sapiens (human)
negative regulation of cholangiocyte apoptotic processInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to angiotensinInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to amyloid-betaInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to insulin-like growth factor stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
multicellular organism developmentInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of kinase activityInsulin-like growth factor 1 receptorHomo sapiens (human)
cellular response to glucose stimulusInsulin-like growth factor 1 receptorHomo sapiens (human)
positive regulation of MAPK cascadeInsulin-like growth factor 1 receptorHomo sapiens (human)
cotranslational protein targeting to membraneSignal recognition particle receptor subunit alphaHomo sapiens (human)
SRP-dependent cotranslational protein targeting to membrane, signal sequence recognitionSignal recognition particle receptor subunit alphaHomo sapiens (human)
intracellular protein transportSignal recognition particle receptor subunit alphaHomo sapiens (human)
protein targeting to ERSignal recognition particle receptor subunit alphaHomo sapiens (human)
mitochondrial electron transport, ubiquinol to cytochrome cCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
response to glucagonCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
cellular respirationCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
proton transmembrane transportCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
endothelial cell morphogenesisHepatocyte growth factor receptorHomo sapiens (human)
signal transductionHepatocyte growth factor receptorHomo sapiens (human)
cell surface receptor signaling pathwayHepatocyte growth factor receptorHomo sapiens (human)
negative regulation of autophagyHepatocyte growth factor receptorHomo sapiens (human)
positive regulation of microtubule polymerizationHepatocyte growth factor receptorHomo sapiens (human)
negative regulation of Rho protein signal transductionHepatocyte growth factor receptorHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIHepatocyte growth factor receptorHomo sapiens (human)
hepatocyte growth factor receptor signaling pathwayHepatocyte growth factor receptorHomo sapiens (human)
branching morphogenesis of an epithelial tubeHepatocyte growth factor receptorHomo sapiens (human)
positive chemotaxisHepatocyte growth factor receptorHomo sapiens (human)
negative regulation of stress fiber assemblyHepatocyte growth factor receptorHomo sapiens (human)
excitatory postsynaptic potentialHepatocyte growth factor receptorHomo sapiens (human)
establishment of skin barrierHepatocyte growth factor receptorHomo sapiens (human)
negative regulation of thrombin-activated receptor signaling pathwayHepatocyte growth factor receptorHomo sapiens (human)
semaphorin-plexin signaling pathwayHepatocyte growth factor receptorHomo sapiens (human)
negative regulation of hydrogen peroxide-mediated programmed cell deathHepatocyte growth factor receptorHomo sapiens (human)
negative regulation of guanyl-nucleotide exchange factor activityHepatocyte growth factor receptorHomo sapiens (human)
positive regulation of endothelial cell chemotaxisHepatocyte growth factor receptorHomo sapiens (human)
liver developmentHepatocyte growth factor receptorHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayHepatocyte growth factor receptorHomo sapiens (human)
phagocytosisHepatocyte growth factor receptorHomo sapiens (human)
multicellular organism developmentHepatocyte growth factor receptorHomo sapiens (human)
neuron differentiationHepatocyte growth factor receptorHomo sapiens (human)
positive regulation of kinase activityHepatocyte growth factor receptorHomo sapiens (human)
cell migrationHepatocyte growth factor receptorHomo sapiens (human)
pancreas developmentHepatocyte growth factor receptorHomo sapiens (human)
nervous system developmentHepatocyte growth factor receptorHomo sapiens (human)
leukocyte migration involved in immune responseTyrosine-protein kinase HCKHomo sapiens (human)
innate immune response-activating signaling pathwayTyrosine-protein kinase HCKHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusTyrosine-protein kinase HCKHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase HCKHomo sapiens (human)
inflammatory responseTyrosine-protein kinase HCKHomo sapiens (human)
cell adhesionTyrosine-protein kinase HCKHomo sapiens (human)
integrin-mediated signaling pathwayTyrosine-protein kinase HCKHomo sapiens (human)
mesoderm developmentTyrosine-protein kinase HCKHomo sapiens (human)
positive regulation of cell population proliferationTyrosine-protein kinase HCKHomo sapiens (human)
regulation of cell shapeTyrosine-protein kinase HCKHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase HCKHomo sapiens (human)
cytokine-mediated signaling pathwayTyrosine-protein kinase HCKHomo sapiens (human)
positive regulation of actin filament polymerizationTyrosine-protein kinase HCKHomo sapiens (human)
lipopolysaccharide-mediated signaling pathwayTyrosine-protein kinase HCKHomo sapiens (human)
regulation of actin cytoskeleton organizationTyrosine-protein kinase HCKHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase HCKHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase HCKHomo sapiens (human)
negative regulation of apoptotic processTyrosine-protein kinase HCKHomo sapiens (human)
leukocyte degranulationTyrosine-protein kinase HCKHomo sapiens (human)
respiratory burst after phagocytosisTyrosine-protein kinase HCKHomo sapiens (human)
protein autophosphorylationTyrosine-protein kinase HCKHomo sapiens (human)
regulation of inflammatory responseTyrosine-protein kinase HCKHomo sapiens (human)
regulation of phagocytosisTyrosine-protein kinase HCKHomo sapiens (human)
regulation of DNA-binding transcription factor activityTyrosine-protein kinase HCKHomo sapiens (human)
type II interferon-mediated signaling pathwayTyrosine-protein kinase HCKHomo sapiens (human)
regulation of podosome assemblyTyrosine-protein kinase HCKHomo sapiens (human)
cell differentiationTyrosine-protein kinase HCKHomo sapiens (human)
innate immune responseTyrosine-protein kinase HCKHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase HCKHomo sapiens (human)
signal transductionPlatelet-derived growth factor receptor betaHomo sapiens (human)
G protein-coupled receptor signaling pathwayPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of cell population proliferationPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of phospholipase C activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of smooth muscle cell migrationPlatelet-derived growth factor receptor betaHomo sapiens (human)
peptidyl-tyrosine phosphorylationPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of cell migrationPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of phosphoprotein phosphatase activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
regulation of actin cytoskeleton organizationPlatelet-derived growth factor receptor betaHomo sapiens (human)
cell migration involved in vasculogenesisPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet-derived growth factor receptor-beta signaling pathwayPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of metanephric mesenchymal cell migration by platelet-derived growth factor receptor-beta signaling pathwayPlatelet-derived growth factor receptor betaHomo sapiens (human)
aorta morphogenesisPlatelet-derived growth factor receptor betaHomo sapiens (human)
cellular response to platelet-derived growth factor stimulusPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of cell proliferation by VEGF-activated platelet derived growth factor receptor signaling pathwayPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of MAP kinase activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of mitotic nuclear divisionPlatelet-derived growth factor receptor betaHomo sapiens (human)
phosphatidylinositol metabolic processPlatelet-derived growth factor receptor betaHomo sapiens (human)
protein autophosphorylationPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet-derived growth factor receptor signaling pathwayPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of smooth muscle cell proliferationPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of calcium-mediated signalingPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of chemotaxisPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionPlatelet-derived growth factor receptor betaHomo sapiens (human)
cardiac myofibril assemblyPlatelet-derived growth factor receptor betaHomo sapiens (human)
cell chemotaxisPlatelet-derived growth factor receptor betaHomo sapiens (human)
cell migration involved in coronary angiogenesisPlatelet-derived growth factor receptor betaHomo sapiens (human)
retina vasculature development in camera-type eyePlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadePlatelet-derived growth factor receptor betaHomo sapiens (human)
smooth muscle cell chemotaxisPlatelet-derived growth factor receptor betaHomo sapiens (human)
metanephric glomerular mesangial cell proliferation involved in metanephros developmentPlatelet-derived growth factor receptor betaHomo sapiens (human)
metanephric glomerular capillary formationPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of calcium ion importPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of reactive oxygen species metabolic processPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of DNA biosynthetic processPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of kinase activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
angiogenesisPlatelet-derived growth factor receptor betaHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayPlatelet-derived growth factor receptor betaHomo sapiens (human)
multicellular organism developmentPlatelet-derived growth factor receptor betaHomo sapiens (human)
positive regulation of cytokine productionTyrosine-protein kinase FgrHomo sapiens (human)
immune response-regulating cell surface receptor signaling pathwayTyrosine-protein kinase FgrHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusTyrosine-protein kinase FgrHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase FgrHomo sapiens (human)
integrin-mediated signaling pathwayTyrosine-protein kinase FgrHomo sapiens (human)
regulation of cell shapeTyrosine-protein kinase FgrHomo sapiens (human)
response to virusTyrosine-protein kinase FgrHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase FgrHomo sapiens (human)
bone mineralizationTyrosine-protein kinase FgrHomo sapiens (human)
positive regulation of cell migrationTyrosine-protein kinase FgrHomo sapiens (human)
negative regulation of natural killer cell activationTyrosine-protein kinase FgrHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase FgrHomo sapiens (human)
positive regulation of mast cell degranulationTyrosine-protein kinase FgrHomo sapiens (human)
regulation of innate immune responseTyrosine-protein kinase FgrHomo sapiens (human)
regulation of protein kinase activityTyrosine-protein kinase FgrHomo sapiens (human)
protein autophosphorylationTyrosine-protein kinase FgrHomo sapiens (human)
skeletal system morphogenesisTyrosine-protein kinase FgrHomo sapiens (human)
regulation of phagocytosisTyrosine-protein kinase FgrHomo sapiens (human)
defense response to Gram-positive bacteriumTyrosine-protein kinase FgrHomo sapiens (human)
myoblast proliferationTyrosine-protein kinase FgrHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionTyrosine-protein kinase FgrHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase FgrHomo sapiens (human)
cell differentiationTyrosine-protein kinase FgrHomo sapiens (human)
innate immune responseTyrosine-protein kinase FgrHomo sapiens (human)
regulation of TOR signalingSerine/threonine-protein kinase A-RafHomo sapiens (human)
regulation of proteasomal ubiquitin-dependent protein catabolic processSerine/threonine-protein kinase A-RafHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationSerine/threonine-protein kinase A-RafHomo sapiens (human)
protein modification processSerine/threonine-protein kinase A-RafHomo sapiens (human)
negative regulation of apoptotic processSerine/threonine-protein kinase A-RafHomo sapiens (human)
MAPK cascadeSerine/threonine-protein kinase A-RafHomo sapiens (human)
glycogen catabolic processGlycogen phosphorylase, brain formHomo sapiens (human)
negative regulation of cellular extravasationBreakpoint cluster region proteinHomo sapiens (human)
renal system processBreakpoint cluster region proteinHomo sapiens (human)
protein phosphorylationBreakpoint cluster region proteinHomo sapiens (human)
phagocytosisBreakpoint cluster region proteinHomo sapiens (human)
signal transductionBreakpoint cluster region proteinHomo sapiens (human)
small GTPase-mediated signal transductionBreakpoint cluster region proteinHomo sapiens (human)
brain developmentBreakpoint cluster region proteinHomo sapiens (human)
actin cytoskeleton organizationBreakpoint cluster region proteinHomo sapiens (human)
keratinocyte differentiationBreakpoint cluster region proteinHomo sapiens (human)
regulation of Rho protein signal transductionBreakpoint cluster region proteinHomo sapiens (human)
inner ear morphogenesisBreakpoint cluster region proteinHomo sapiens (human)
regulation of vascular permeabilityBreakpoint cluster region proteinHomo sapiens (human)
neutrophil degranulationBreakpoint cluster region proteinHomo sapiens (human)
negative regulation of neutrophil degranulationBreakpoint cluster region proteinHomo sapiens (human)
focal adhesion assemblyBreakpoint cluster region proteinHomo sapiens (human)
homeostasis of number of cellsBreakpoint cluster region proteinHomo sapiens (human)
negative regulation of inflammatory responseBreakpoint cluster region proteinHomo sapiens (human)
positive regulation of phagocytosisBreakpoint cluster region proteinHomo sapiens (human)
modulation of chemical synaptic transmissionBreakpoint cluster region proteinHomo sapiens (human)
neuromuscular process controlling balanceBreakpoint cluster region proteinHomo sapiens (human)
regulation of small GTPase mediated signal transductionBreakpoint cluster region proteinHomo sapiens (human)
regulation of cell cycleBreakpoint cluster region proteinHomo sapiens (human)
definitive hemopoiesisBreakpoint cluster region proteinHomo sapiens (human)
negative regulation of respiratory burstBreakpoint cluster region proteinHomo sapiens (human)
negative regulation of blood vessel remodelingBreakpoint cluster region proteinHomo sapiens (human)
intracellular protein transmembrane transportBreakpoint cluster region proteinHomo sapiens (human)
cellular response to lipopolysaccharideBreakpoint cluster region proteinHomo sapiens (human)
activation of GTPase activityBreakpoint cluster region proteinHomo sapiens (human)
macrophage migrationBreakpoint cluster region proteinHomo sapiens (human)
negative regulation of macrophage migrationBreakpoint cluster region proteinHomo sapiens (human)
negative regulation of reactive oxygen species metabolic processBreakpoint cluster region proteinHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase pim-1Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase pim-1Homo sapiens (human)
regulation of transmembrane transporter activitySerine/threonine-protein kinase pim-1Homo sapiens (human)
negative regulation of apoptotic processSerine/threonine-protein kinase pim-1Homo sapiens (human)
negative regulation of DNA-binding transcription factor activitySerine/threonine-protein kinase pim-1Homo sapiens (human)
negative regulation of innate immune responseSerine/threonine-protein kinase pim-1Homo sapiens (human)
positive regulation of DNA-templated transcriptionSerine/threonine-protein kinase pim-1Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase pim-1Homo sapiens (human)
protein stabilizationSerine/threonine-protein kinase pim-1Homo sapiens (human)
positive regulation of cardiac muscle cell proliferationSerine/threonine-protein kinase pim-1Homo sapiens (human)
vitamin D receptor signaling pathwaySerine/threonine-protein kinase pim-1Homo sapiens (human)
cellular response to type II interferonSerine/threonine-protein kinase pim-1Homo sapiens (human)
positive regulation of brown fat cell differentiationSerine/threonine-protein kinase pim-1Homo sapiens (human)
regulation of hematopoietic stem cell proliferationSerine/threonine-protein kinase pim-1Homo sapiens (human)
positive regulation of TORC1 signalingSerine/threonine-protein kinase pim-1Homo sapiens (human)
positive regulation of cardioblast proliferationSerine/threonine-protein kinase pim-1Homo sapiens (human)
cellular detoxificationSerine/threonine-protein kinase pim-1Homo sapiens (human)
positive regulation of cell population proliferationFibroblast growth factor receptor 1Homo sapiens (human)
fibroblast growth factor receptor signaling pathwayFibroblast growth factor receptor 1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIFibroblast growth factor receptor 1Homo sapiens (human)
MAPK cascadeFibroblast growth factor receptor 1Homo sapiens (human)
skeletal system developmentFibroblast growth factor receptor 1Homo sapiens (human)
angiogenesisFibroblast growth factor receptor 1Homo sapiens (human)
ureteric bud developmentFibroblast growth factor receptor 1Homo sapiens (human)
in utero embryonic developmentFibroblast growth factor receptor 1Homo sapiens (human)
organ inductionFibroblast growth factor receptor 1Homo sapiens (human)
neuron migrationFibroblast growth factor receptor 1Homo sapiens (human)
epithelial to mesenchymal transitionFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of mesenchymal cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
chondrocyte differentiationFibroblast growth factor receptor 1Homo sapiens (human)
protein phosphorylationFibroblast growth factor receptor 1Homo sapiens (human)
sensory perception of soundFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of cell population proliferationFibroblast growth factor receptor 1Homo sapiens (human)
fibroblast growth factor receptor signaling pathwayFibroblast growth factor receptor 1Homo sapiens (human)
mesenchymal cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
gene expressionFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of phospholipase activityFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of phospholipase C activityFibroblast growth factor receptor 1Homo sapiens (human)
regulation of phosphate transportFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of neuron projection developmentFibroblast growth factor receptor 1Homo sapiens (human)
cell migrationFibroblast growth factor receptor 1Homo sapiens (human)
peptidyl-tyrosine phosphorylationFibroblast growth factor receptor 1Homo sapiens (human)
ventricular zone neuroblast divisionFibroblast growth factor receptor 1Homo sapiens (human)
cell projection assemblyFibroblast growth factor receptor 1Homo sapiens (human)
embryonic limb morphogenesisFibroblast growth factor receptor 1Homo sapiens (human)
midbrain developmentFibroblast growth factor receptor 1Homo sapiens (human)
neuron projection developmentFibroblast growth factor receptor 1Homo sapiens (human)
fibroblast growth factor receptor signaling pathway involved in orbitofrontal cortex developmentFibroblast growth factor receptor 1Homo sapiens (human)
inner ear morphogenesisFibroblast growth factor receptor 1Homo sapiens (human)
outer ear morphogenesisFibroblast growth factor receptor 1Homo sapiens (human)
middle ear morphogenesisFibroblast growth factor receptor 1Homo sapiens (human)
chordate embryonic developmentFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of MAP kinase activityFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of MAPK cascadeFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of blood vessel endothelial cell migrationFibroblast growth factor receptor 1Homo sapiens (human)
cellular response to fibroblast growth factor stimulusFibroblast growth factor receptor 1Homo sapiens (human)
regulation of cell differentiationFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of neuron differentiationFibroblast growth factor receptor 1Homo sapiens (human)
protein autophosphorylationFibroblast growth factor receptor 1Homo sapiens (human)
phosphatidylinositol-mediated signalingFibroblast growth factor receptor 1Homo sapiens (human)
paraxial mesoderm developmentFibroblast growth factor receptor 1Homo sapiens (human)
regulation of lateral mesodermal cell fate specificationFibroblast growth factor receptor 1Homo sapiens (human)
cell maturationFibroblast growth factor receptor 1Homo sapiens (human)
skeletal system morphogenesisFibroblast growth factor receptor 1Homo sapiens (human)
stem cell differentiationFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionFibroblast growth factor receptor 1Homo sapiens (human)
calcium ion homeostasisFibroblast growth factor receptor 1Homo sapiens (human)
cardiac muscle cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of cardiac muscle cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
auditory receptor cell developmentFibroblast growth factor receptor 1Homo sapiens (human)
branching involved in salivary gland morphogenesisFibroblast growth factor receptor 1Homo sapiens (human)
lung-associated mesenchyme developmentFibroblast growth factor receptor 1Homo sapiens (human)
regulation of branching involved in salivary gland morphogenesis by mesenchymal-epithelial signalingFibroblast growth factor receptor 1Homo sapiens (human)
vitamin D3 metabolic processFibroblast growth factor receptor 1Homo sapiens (human)
diphosphate metabolic processFibroblast growth factor receptor 1Homo sapiens (human)
cementum mineralizationFibroblast growth factor receptor 1Homo sapiens (human)
stem cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of MAPKKK cascade by fibroblast growth factor receptor signaling pathwayFibroblast growth factor receptor 1Homo sapiens (human)
negative regulation of fibroblast growth factor productionFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of mitotic cell cycle DNA replicationFibroblast growth factor receptor 1Homo sapiens (human)
response to sodium phosphateFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of vascular endothelial cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of stem cell proliferationFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of parathyroid hormone secretionFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of endothelial cell chemotaxisFibroblast growth factor receptor 1Homo sapiens (human)
regulation of extrinsic apoptotic signaling pathway in absence of ligandFibroblast growth factor receptor 1Homo sapiens (human)
multicellular organism developmentFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of cell differentiationFibroblast growth factor receptor 1Homo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayFibroblast growth factor receptor 1Homo sapiens (human)
positive regulation of kinase activityFibroblast growth factor receptor 1Homo sapiens (human)
hematopoietic progenitor cell differentiationDNA topoisomerase 2-alphaHomo sapiens (human)
DNA topological changeDNA topoisomerase 2-alphaHomo sapiens (human)
DNA ligationDNA topoisomerase 2-alphaHomo sapiens (human)
DNA damage responseDNA topoisomerase 2-alphaHomo sapiens (human)
chromosome segregationDNA topoisomerase 2-alphaHomo sapiens (human)
female meiotic nuclear divisionDNA topoisomerase 2-alphaHomo sapiens (human)
apoptotic chromosome condensationDNA topoisomerase 2-alphaHomo sapiens (human)
embryonic cleavageDNA topoisomerase 2-alphaHomo sapiens (human)
regulation of circadian rhythmDNA topoisomerase 2-alphaHomo sapiens (human)
positive regulation of apoptotic processDNA topoisomerase 2-alphaHomo sapiens (human)
positive regulation of single stranded viral RNA replication via double stranded DNA intermediateDNA topoisomerase 2-alphaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIDNA topoisomerase 2-alphaHomo sapiens (human)
rhythmic processDNA topoisomerase 2-alphaHomo sapiens (human)
negative regulation of DNA duplex unwindingDNA topoisomerase 2-alphaHomo sapiens (human)
resolution of meiotic recombination intermediatesDNA topoisomerase 2-alphaHomo sapiens (human)
sister chromatid segregationDNA topoisomerase 2-alphaHomo sapiens (human)
G1/S transition of mitotic cell cycleCyclin-dependent kinase 4Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 4Homo sapiens (human)
positive regulation of cell population proliferationCyclin-dependent kinase 4Homo sapiens (human)
response to xenobiotic stimulusCyclin-dependent kinase 4Homo sapiens (human)
regulation of gene expressionCyclin-dependent kinase 4Homo sapiens (human)
positive regulation of G2/M transition of mitotic cell cycleCyclin-dependent kinase 4Homo sapiens (human)
positive regulation of fibroblast proliferationCyclin-dependent kinase 4Homo sapiens (human)
cell divisionCyclin-dependent kinase 4Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 4Homo sapiens (human)
regulation of transcription initiation by RNA polymerase IICyclin-dependent kinase 4Homo sapiens (human)
regulation of type B pancreatic cell proliferationCyclin-dependent kinase 4Homo sapiens (human)
cellular response to lipopolysaccharideCyclin-dependent kinase 4Homo sapiens (human)
cellular response to interleukin-4Cyclin-dependent kinase 4Homo sapiens (human)
cellular response to phorbol 13-acetate 12-myristateCyclin-dependent kinase 4Homo sapiens (human)
cellular response to ionomycinCyclin-dependent kinase 4Homo sapiens (human)
response to organic substanceCyclin-dependent kinase 4Homo sapiens (human)
regulation of G2/M transition of mitotic cell cycleCyclin-dependent kinase 4Homo sapiens (human)
signal transductionCyclin-dependent kinase 4Homo sapiens (human)
apoptotic processADP/ATP translocase 3Homo sapiens (human)
mitochondrial ADP transmembrane transportADP/ATP translocase 3Homo sapiens (human)
mitochondrial ATP transmembrane transportADP/ATP translocase 3Homo sapiens (human)
negative regulation of mitochondrial outer membrane permeabilization involved in apoptotic signaling pathwayADP/ATP translocase 3Homo sapiens (human)
GMP biosynthetic processInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
GTP biosynthetic processInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
circadian rhythmInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
lymphocyte proliferationInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
cellular response to interleukin-4Inosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
'de novo' XMP biosynthetic processInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
peptidyl-tyrosine phosphorylationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
primary ovarian follicle growthProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of cytokine productionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
stimulatory C-type lectin receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
signal transductionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
signal complex assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
epidermal growth factor receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
transforming growth factor beta receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
integrin-mediated signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
spermatogenesisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
learning or memoryProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to xenobiotic stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to mechanical stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to acidic pHProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of gene expressionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of epithelial cell migrationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of epithelial cell migrationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of glucose metabolic processProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of protein processingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
skeletal muscle cell proliferationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of smooth muscle cell migrationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
macroautophagyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
peptidyl-tyrosine phosphorylationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of cell-cell adhesionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
platelet activationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
forebrain developmentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
T cell costimulationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of protein-containing complex assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
protein destabilizationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to nutrient levelsProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of telomere maintenance via telomeraseProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to insulin stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of intracellular estrogen receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of integrin activationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of toll-like receptor 3 signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
adherens junction organizationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
substrate adhesion-dependent cell spreadingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of dephosphorylationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of hippo signalingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
intracellular signal transductionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
entry of bacterium into host cellProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
osteoclast developmentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to platelet-derived growth factor stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
ERBB2 signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
angiotensin-activated signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
odontogenesisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of apoptotic processProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of apoptotic processProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of vascular permeabilityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
stress fiber assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
transcytosisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of bone resorptionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
bone resorptionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of Notch signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of bone resorptionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of Ras protein signal transductionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of insulin receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
protein autophosphorylationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
platelet-derived growth factor receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
vascular endothelial growth factor receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
neurotrophin TRK receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
ephrin receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
focal adhesion assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
oogenesisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
progesterone receptor signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
leukocyte migrationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of small GTPase mediated signal transductionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of protein transportProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to mineralocorticoidProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
myoblast proliferationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to electrical stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of focal adhesion assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of mitochondrial depolarizationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of telomerase activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
uterus developmentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
branching involved in mammary gland duct morphogenesisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of cell projection assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
intestinal epithelial cell developmentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
interleukin-6-mediated signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to hydrogen peroxideProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
response to interleukin-1Proto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to lipopolysaccharideProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to peptide hormone stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to progesterone stimulusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to fatty acidProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to hypoxiaProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to fluid shear stressProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of podosome assemblyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
DNA biosynthetic processProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of protein serine/threonine kinase activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of heart rate by cardiac conductionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of canonical Wnt signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cell-cell adhesionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of protein localization to nucleusProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of non-membrane spanning protein tyrosine kinase activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of TORC1 signalingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cellular response to prolactinProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of male germ cell proliferationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of ovarian follicle developmentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of lamellipodium morphogenesisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
positive regulation of platelet-derived growth factor receptor-beta signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of early endosome to late endosome transportProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of anoikisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathwayProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of caveolin-mediated endocytosisProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cell differentiationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cell adhesionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
innate immune responseProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
protein phosphorylationProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
symbiont entry into host cellProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
regulation of protein phosphorylationcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
intracellular signal transductioncAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
negative regulation of cAMP/PKA signal transductioncAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
negative regulation of cAMP-dependent protein kinase activitycAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
MAPK cascadeSerine/threonine-protein kinase B-rafHomo sapiens (human)
myeloid progenitor cell differentiationSerine/threonine-protein kinase B-rafHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase B-rafHomo sapiens (human)
epidermal growth factor receptor signaling pathwaySerine/threonine-protein kinase B-rafHomo sapiens (human)
visual learningSerine/threonine-protein kinase B-rafHomo sapiens (human)
animal organ morphogenesisSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of gene expressionSerine/threonine-protein kinase B-rafHomo sapiens (human)
negative regulation of fibroblast migrationSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of glucose transmembrane transportSerine/threonine-protein kinase B-rafHomo sapiens (human)
synaptic vesicle exocytosisSerine/threonine-protein kinase B-rafHomo sapiens (human)
thyroid gland developmentSerine/threonine-protein kinase B-rafHomo sapiens (human)
T cell differentiation in thymusSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationSerine/threonine-protein kinase B-rafHomo sapiens (human)
substrate adhesion-dependent cell spreadingSerine/threonine-protein kinase B-rafHomo sapiens (human)
somatic stem cell population maintenanceSerine/threonine-protein kinase B-rafHomo sapiens (human)
regulation of cell population proliferationSerine/threonine-protein kinase B-rafHomo sapiens (human)
negative regulation of apoptotic processSerine/threonine-protein kinase B-rafHomo sapiens (human)
stress fiber assemblySerine/threonine-protein kinase B-rafHomo sapiens (human)
CD4-positive, alpha-beta T cell differentiationSerine/threonine-protein kinase B-rafHomo sapiens (human)
CD4-positive or CD8-positive, alpha-beta T cell lineage commitmentSerine/threonine-protein kinase B-rafHomo sapiens (human)
response to peptide hormoneSerine/threonine-protein kinase B-rafHomo sapiens (human)
negative regulation of neuron apoptotic processSerine/threonine-protein kinase B-rafHomo sapiens (human)
regulation of T cell differentiationSerine/threonine-protein kinase B-rafHomo sapiens (human)
thymus developmentSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of axon regenerationSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of axonogenesisSerine/threonine-protein kinase B-rafHomo sapiens (human)
T cell receptor signaling pathwaySerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of stress fiber assemblySerine/threonine-protein kinase B-rafHomo sapiens (human)
response to cAMPSerine/threonine-protein kinase B-rafHomo sapiens (human)
long-term synaptic potentiationSerine/threonine-protein kinase B-rafHomo sapiens (human)
head morphogenesisSerine/threonine-protein kinase B-rafHomo sapiens (human)
face developmentSerine/threonine-protein kinase B-rafHomo sapiens (human)
ERK1 and ERK2 cascadeSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeSerine/threonine-protein kinase B-rafHomo sapiens (human)
cellular response to calcium ionSerine/threonine-protein kinase B-rafHomo sapiens (human)
cellular response to xenobiotic stimulusSerine/threonine-protein kinase B-rafHomo sapiens (human)
endothelial cell apoptotic processSerine/threonine-protein kinase B-rafHomo sapiens (human)
establishment of protein localization to membraneSerine/threonine-protein kinase B-rafHomo sapiens (human)
positive regulation of substrate adhesion-dependent cell spreadingSerine/threonine-protein kinase B-rafHomo sapiens (human)
cellular response to nerve growth factor stimulusSerine/threonine-protein kinase B-rafHomo sapiens (human)
negative regulation of synaptic vesicle exocytosisSerine/threonine-protein kinase B-rafHomo sapiens (human)
negative regulation of endothelial cell apoptotic processSerine/threonine-protein kinase B-rafHomo sapiens (human)
epithelial cell maturationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
male gonad developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
vestibular nucleus developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
secretory granule organizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to acidic pHPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to light stimulusPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell action potential involved in contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of protein targeting to membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
glycogen metabolic processPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
glycogen biosynthetic processPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
glycogen catabolic processPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
generation of precursor metabolites and energyPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
protein phosphorylationPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
positive regulation of glycogen catabolic processPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
quinone catabolic processRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
microtubule cytoskeleton organizationTyrosine-protein kinase FerHomo sapiens (human)
regulation of protein phosphorylationTyrosine-protein kinase FerHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase FerHomo sapiens (human)
tyrosine phosphorylation of STAT proteinTyrosine-protein kinase FerHomo sapiens (human)
germ cell developmentTyrosine-protein kinase FerHomo sapiens (human)
positive regulation of cell population proliferationTyrosine-protein kinase FerHomo sapiens (human)
insulin receptor signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
regulation of lamellipodium assemblyTyrosine-protein kinase FerHomo sapiens (human)
regulation of fibroblast migrationTyrosine-protein kinase FerHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase FerHomo sapiens (human)
cytokine-mediated signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
actin cytoskeleton organizationTyrosine-protein kinase FerHomo sapiens (human)
positive regulation of cell migrationTyrosine-protein kinase FerHomo sapiens (human)
positive regulation of actin filament polymerizationTyrosine-protein kinase FerHomo sapiens (human)
response to lipopolysaccharideTyrosine-protein kinase FerHomo sapiens (human)
negative regulation of mast cell activation involved in immune responseTyrosine-protein kinase FerHomo sapiens (human)
adherens junction assemblyTyrosine-protein kinase FerHomo sapiens (human)
substrate adhesion-dependent cell spreadingTyrosine-protein kinase FerHomo sapiens (human)
cellular response to reactive oxygen speciesTyrosine-protein kinase FerHomo sapiens (human)
extracellular matrix-cell signalingTyrosine-protein kinase FerHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase FerHomo sapiens (human)
cellular response to macrophage colony-stimulating factor stimulusTyrosine-protein kinase FerHomo sapiens (human)
response to platelet-derived growth factorTyrosine-protein kinase FerHomo sapiens (human)
Fc-epsilon receptor signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
Kit signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
regulation of epidermal growth factor receptor signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
cell-cell adhesion mediated by cadherinTyrosine-protein kinase FerHomo sapiens (human)
protein autophosphorylationTyrosine-protein kinase FerHomo sapiens (human)
platelet-derived growth factor receptor signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
diapedesisTyrosine-protein kinase FerHomo sapiens (human)
positive regulation of NF-kappaB transcription factor activityTyrosine-protein kinase FerHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionTyrosine-protein kinase FerHomo sapiens (human)
Sertoli cell developmentTyrosine-protein kinase FerHomo sapiens (human)
interleukin-6-mediated signaling pathwayTyrosine-protein kinase FerHomo sapiens (human)
seminiferous tubule developmentTyrosine-protein kinase FerHomo sapiens (human)
adherens junction disassemblyTyrosine-protein kinase FerHomo sapiens (human)
cell adhesionTyrosine-protein kinase FerHomo sapiens (human)
chemotaxisTyrosine-protein kinase FerHomo sapiens (human)
angiogenesisProtein kinase C alpha typeHomo sapiens (human)
positive regulation of endothelial cell proliferationProtein kinase C alpha typeHomo sapiens (human)
desmosome assemblyProtein kinase C alpha typeHomo sapiens (human)
chromatin remodelingProtein kinase C alpha typeHomo sapiens (human)
protein phosphorylationProtein kinase C alpha typeHomo sapiens (human)
mitotic nuclear membrane disassemblyProtein kinase C alpha typeHomo sapiens (human)
cell adhesionProtein kinase C alpha typeHomo sapiens (human)
positive regulation of endothelial cell migrationProtein kinase C alpha typeHomo sapiens (human)
positive regulation of cardiac muscle hypertrophyProtein kinase C alpha typeHomo sapiens (human)
peptidyl-serine phosphorylationProtein kinase C alpha typeHomo sapiens (human)
peptidyl-threonine phosphorylationProtein kinase C alpha typeHomo sapiens (human)
positive regulation of cell migrationProtein kinase C alpha typeHomo sapiens (human)
positive regulation of lipopolysaccharide-mediated signaling pathwayProtein kinase C alpha typeHomo sapiens (human)
negative regulation of glial cell apoptotic processProtein kinase C alpha typeHomo sapiens (human)
regulation of mRNA stabilityProtein kinase C alpha typeHomo sapiens (human)
positive regulation of blood vessel endothelial cell migrationProtein kinase C alpha typeHomo sapiens (human)
post-translational protein modificationProtein kinase C alpha typeHomo sapiens (human)
positive regulation of macrophage differentiationProtein kinase C alpha typeHomo sapiens (human)
positive regulation of angiogenesisProtein kinase C alpha typeHomo sapiens (human)
positive regulation of bone resorptionProtein kinase C alpha typeHomo sapiens (human)
positive regulation of cell adhesionProtein kinase C alpha typeHomo sapiens (human)
positive regulation of mitotic cell cycleProtein kinase C alpha typeHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeProtein kinase C alpha typeHomo sapiens (human)
response to interleukin-1Protein kinase C alpha typeHomo sapiens (human)
regulation of platelet aggregationProtein kinase C alpha typeHomo sapiens (human)
apoptotic signaling pathwayProtein kinase C alpha typeHomo sapiens (human)
positive regulation of adenylate cyclase-activating G protein-coupled receptor signaling pathwayProtein kinase C alpha typeHomo sapiens (human)
positive regulation of angiotensin-activated signaling pathwayProtein kinase C alpha typeHomo sapiens (human)
positive regulation of dense core granule biogenesisProtein kinase C alpha typeHomo sapiens (human)
intracellular signal transductionProtein kinase C alpha typeHomo sapiens (human)
positive regulation of insulin secretionProtein kinase C alpha typeHomo sapiens (human)
mesoderm formationcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
neural tube closurecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of heart ratecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
renal water homeostasiscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
mRNA processingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein phosphorylationcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein export from nucleuscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwaycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of cardiac muscle contraction by regulation of the release of sequestered calcium ioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of macroautophagycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
peptidyl-serine phosphorylationcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cytokine-mediated signaling pathwaycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
positive regulation of insulin secretioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
negative regulation of interleukin-2 productioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
high-density lipoprotein particle assemblycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cellular response to heatcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
mitochondrial protein catabolic processcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of osteoblast differentiationcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
positive regulation of gluconeogenesiscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
negative regulation of smoothened signaling pathwaycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
positive regulation of protein export from nucleuscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
sperm capacitationcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
positive regulation of calcium-mediated signalingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of cell cyclecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of cardiac muscle contractioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of proteasomal protein catabolic processcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cellular response to coldcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of protein processingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cellular response to glucose stimuluscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cellular response to parathyroid hormone stimuluscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cellular response to glucagon stimuluscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cellular response to epinephrine stimuluscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cell communication by electrical coupling involved in cardiac conductioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
postsynaptic modulation of chemical synaptic transmissioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of cardiac conductioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
negative regulation of TORC1 signalingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
negative regulation of glycolytic process through fructose-6-phosphatecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein localization to lipid dropletcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
regulation of bicellular tight junction assemblycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein kinase A signalingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
maturation of SSU-rRNA from tricistronic rRNA transcript (SSU-rRNA, 5.8S rRNA, LSU-rRNA)General transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
response to hypoxiaGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
in utero embryonic developmentGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription-coupled nucleotide-excision repairGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
nucleotide-excision repairGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
regulation of transcription by RNA polymerase IIGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription elongation by RNA polymerase IGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription by RNA polymerase IIGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription initiation at RNA polymerase II promoterGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
apoptotic processGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
response to oxidative stressGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
chromosome segregationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
determination of adult lifespanGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
UV protectionGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
post-embryonic developmentGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
spinal cord developmentGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
extracellular matrix organizationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
bone mineralizationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
central nervous system myelin formationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
DNA duplex unwindingGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
multicellular organism growthGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
hair cell differentiationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
embryonic cleavageGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
erythrocyte maturationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
insulin-like growth factor receptor signaling pathwayGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
embryonic organ developmentGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
hair follicle maturationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
hematopoietic stem cell differentiationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
hematopoietic stem cell proliferationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
regulation of mitotic cell cycle phase transitionGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
positive regulation of mitotic recombinationGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
double-strand break repairCasein kinase II subunit alpha'Homo sapiens (human)
apoptotic processCasein kinase II subunit alpha'Homo sapiens (human)
spermatogenesisCasein kinase II subunit alpha'Homo sapiens (human)
Wnt signaling pathwayCasein kinase II subunit alpha'Homo sapiens (human)
cerebral cortex developmentCasein kinase II subunit alpha'Homo sapiens (human)
negative regulation of proteasomal ubiquitin-dependent protein catabolic processCasein kinase II subunit alpha'Homo sapiens (human)
liver regenerationCasein kinase II subunit alpha'Homo sapiens (human)
regulation of mitophagyCasein kinase II subunit alpha'Homo sapiens (human)
positive regulation of protein targeting to mitochondrionCasein kinase II subunit alpha'Homo sapiens (human)
regulation of chromosome separationCasein kinase II subunit alpha'Homo sapiens (human)
negative regulation of apoptotic signaling pathwayCasein kinase II subunit alpha'Homo sapiens (human)
peptidyl-threonine phosphorylationCasein kinase II subunit alpha'Homo sapiens (human)
peptidyl-serine phosphorylationCasein kinase II subunit alpha'Homo sapiens (human)
peptidyl-cysteine methylationRas-related protein Rab-6AHomo sapiens (human)
retrograde vesicle-mediated transport, Golgi to endoplasmic reticulumRas-related protein Rab-6AHomo sapiens (human)
antigen processing and presentationRas-related protein Rab-6AHomo sapiens (human)
neuron projection developmentRas-related protein Rab-6AHomo sapiens (human)
protein localization to Golgi apparatusRas-related protein Rab-6AHomo sapiens (human)
early endosome to Golgi transportRas-related protein Rab-6AHomo sapiens (human)
minus-end-directed organelle transport along microtubuleRas-related protein Rab-6AHomo sapiens (human)
protein localization to Golgi membraneRas-related protein Rab-6AHomo sapiens (human)
intracellular protein transportRas-related protein Rab-6AHomo sapiens (human)
intra-Golgi vesicle-mediated transportRas-related protein Rab-6AHomo sapiens (human)
retrograde transport, endosome to GolgiRas-related protein Rab-6AHomo sapiens (human)
positive regulation of cell-matrix adhesionEphrin type-A receptor 1Homo sapiens (human)
negative regulation of protein kinase activityEphrin type-A receptor 1Homo sapiens (human)
cell surface receptor signaling pathwayEphrin type-A receptor 1Homo sapiens (human)
positive regulation of cell population proliferationEphrin type-A receptor 1Homo sapiens (human)
peptidyl-tyrosine phosphorylationEphrin type-A receptor 1Homo sapiens (human)
positive regulation of cell migrationEphrin type-A receptor 1Homo sapiens (human)
negative regulation of cell migrationEphrin type-A receptor 1Homo sapiens (human)
substrate adhesion-dependent cell spreadingEphrin type-A receptor 1Homo sapiens (human)
regulation of GTPase activityEphrin type-A receptor 1Homo sapiens (human)
positive regulation of angiogenesisEphrin type-A receptor 1Homo sapiens (human)
protein autophosphorylationEphrin type-A receptor 1Homo sapiens (human)
positive regulation of stress fiber assemblyEphrin type-A receptor 1Homo sapiens (human)
activation of GTPase activityEphrin type-A receptor 1Homo sapiens (human)
positive regulation of kinase activityEphrin type-A receptor 1Homo sapiens (human)
multicellular organism developmentEphrin type-A receptor 1Homo sapiens (human)
angiogenesisEphrin type-A receptor 1Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-A receptor 1Homo sapiens (human)
GMP biosynthetic processMultifunctional protein ADE2Homo sapiens (human)
'de novo' IMP biosynthetic processMultifunctional protein ADE2Homo sapiens (human)
purine nucleobase biosynthetic processMultifunctional protein ADE2Homo sapiens (human)
'de novo' AMP biosynthetic processMultifunctional protein ADE2Homo sapiens (human)
'de novo' XMP biosynthetic processMultifunctional protein ADE2Homo sapiens (human)
renal water homeostasiscAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
spermatogenesiscAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
male gonad developmentcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
high-density lipoprotein particle assemblycAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
protein kinase A signalingcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
neural tube closurecAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
renal water homeostasiscAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
protein phosphorylationcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
signal transductioncAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
adenylate cyclase-modulating G protein-coupled receptor signaling pathwaycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
high-density lipoprotein particle assemblycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
negative regulation of smoothened signaling pathwaycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
regulation of protein processingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
negative regulation of TORC1 signalingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
protein kinase A signalingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
generation of precursor metabolites and energyFerrochelatase, mitochondrialHomo sapiens (human)
heme biosynthetic processFerrochelatase, mitochondrialHomo sapiens (human)
heme A biosynthetic processFerrochelatase, mitochondrialHomo sapiens (human)
heme B biosynthetic processFerrochelatase, mitochondrialHomo sapiens (human)
cholesterol metabolic processFerrochelatase, mitochondrialHomo sapiens (human)
response to xenobiotic stimulusFerrochelatase, mitochondrialHomo sapiens (human)
response to light stimulusFerrochelatase, mitochondrialHomo sapiens (human)
detection of UVFerrochelatase, mitochondrialHomo sapiens (human)
response to lead ionFerrochelatase, mitochondrialHomo sapiens (human)
regulation of eIF2 alpha phosphorylation by hemeFerrochelatase, mitochondrialHomo sapiens (human)
response to insecticideFerrochelatase, mitochondrialHomo sapiens (human)
erythrocyte differentiationFerrochelatase, mitochondrialHomo sapiens (human)
very-low-density lipoprotein particle assemblyFerrochelatase, mitochondrialHomo sapiens (human)
response to ethanolFerrochelatase, mitochondrialHomo sapiens (human)
protoporphyrinogen IX metabolic processFerrochelatase, mitochondrialHomo sapiens (human)
response to arsenic-containing substanceFerrochelatase, mitochondrialHomo sapiens (human)
regulation of hemoglobin biosynthetic processFerrochelatase, mitochondrialHomo sapiens (human)
heme O biosynthetic processFerrochelatase, mitochondrialHomo sapiens (human)
response to methylmercuryFerrochelatase, mitochondrialHomo sapiens (human)
multicellular organismal-level iron ion homeostasisFerrochelatase, mitochondrialHomo sapiens (human)
response to platinum ionFerrochelatase, mitochondrialHomo sapiens (human)
cellular response to dexamethasone stimulusFerrochelatase, mitochondrialHomo sapiens (human)
G1/S transition of mitotic cell cycleRibosomal protein S6 kinase beta-1Homo sapiens (human)
behavioral fear responseRibosomal protein S6 kinase beta-1Homo sapiens (human)
skeletal muscle contractionRibosomal protein S6 kinase beta-1Homo sapiens (human)
apoptotic processRibosomal protein S6 kinase beta-1Homo sapiens (human)
signal transductionRibosomal protein S6 kinase beta-1Homo sapiens (human)
germ cell developmentRibosomal protein S6 kinase beta-1Homo sapiens (human)
long-term memoryRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to xenobiotic stimulusRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to mechanical stimulusRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to toxic substanceRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to glucoseRibosomal protein S6 kinase beta-1Homo sapiens (human)
skeletal muscle atrophyRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to electrical stimulus involved in regulation of muscle adaptationRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of smooth muscle cell migrationRibosomal protein S6 kinase beta-1Homo sapiens (human)
cell migrationRibosomal protein S6 kinase beta-1Homo sapiens (human)
peptidyl-serine phosphorylationRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to nutrient levelsRibosomal protein S6 kinase beta-1Homo sapiens (human)
cellular response to nutrientRibosomal protein S6 kinase beta-1Homo sapiens (human)
TOR signalingRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to lipopolysaccharideRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to testosteroneRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to glucagonRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to tumor necrosis factorRibosomal protein S6 kinase beta-1Homo sapiens (human)
negative regulation of apoptotic processRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to L-leucineRibosomal protein S6 kinase beta-1Homo sapiens (human)
long-chain fatty acid import into cellRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to ethanolRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of translationRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of mitotic cell cycleRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of translational initiationRibosomal protein S6 kinase beta-1Homo sapiens (human)
regulation of glucose importRibosomal protein S6 kinase beta-1Homo sapiens (human)
negative regulation of insulin receptor signaling pathwayRibosomal protein S6 kinase beta-1Homo sapiens (human)
phosphatidylinositol-mediated signalingRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of skeletal muscle tissue growthRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of smooth muscle cell proliferationRibosomal protein S6 kinase beta-1Homo sapiens (human)
modulation of chemical synaptic transmissionRibosomal protein S6 kinase beta-1Homo sapiens (human)
cellular response to type II interferonRibosomal protein S6 kinase beta-1Homo sapiens (human)
cellular response to growth factor stimulusRibosomal protein S6 kinase beta-1Homo sapiens (human)
cellular response to dexamethasone stimulusRibosomal protein S6 kinase beta-1Homo sapiens (human)
positive regulation of TORC1 signalingRibosomal protein S6 kinase beta-1Homo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathwayRibosomal protein S6 kinase beta-1Homo sapiens (human)
cellular response to insulin stimulusRibosomal protein S6 kinase beta-1Homo sapiens (human)
response to antibioticTyrosine-protein kinase JAK1Homo sapiens (human)
protein phosphorylationTyrosine-protein kinase JAK1Homo sapiens (human)
cell surface receptor signaling pathway via JAK-STATTyrosine-protein kinase JAK1Homo sapiens (human)
cytokine-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
positive regulation of homotypic cell-cell adhesionTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-15-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-4-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-2-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-9-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-11-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
type III interferon-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
type II interferon-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
type I interferon-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-6-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
T-helper 17 cell lineage commitmentTyrosine-protein kinase JAK1Homo sapiens (human)
cellular response to virusTyrosine-protein kinase JAK1Homo sapiens (human)
interleukin-10-mediated signaling pathwayTyrosine-protein kinase JAK1Homo sapiens (human)
protein localization to cell-cell junctionTyrosine-protein kinase JAK1Homo sapiens (human)
positive regulation of protein localization to nucleusTyrosine-protein kinase JAK1Homo sapiens (human)
positive regulation of sprouting angiogenesisTyrosine-protein kinase JAK1Homo sapiens (human)
intracellular signal transductionTyrosine-protein kinase JAK1Homo sapiens (human)
tyrosine phosphorylation of STAT proteinTyrosine-protein kinase JAK1Homo sapiens (human)
cell differentiationTyrosine-protein kinase JAK1Homo sapiens (human)
growth hormone receptor signaling pathway via JAK-STATTyrosine-protein kinase JAK1Homo sapiens (human)
G1/S transition of mitotic cell cycleCyclin-dependent kinase 2Homo sapiens (human)
G2/M transition of mitotic cell cycleCyclin-dependent kinase 2Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICyclin-dependent kinase 2Homo sapiens (human)
DNA replicationCyclin-dependent kinase 2Homo sapiens (human)
DNA repairCyclin-dependent kinase 2Homo sapiens (human)
chromatin remodelingCyclin-dependent kinase 2Homo sapiens (human)
DNA-templated transcriptionCyclin-dependent kinase 2Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 2Homo sapiens (human)
potassium ion transportCyclin-dependent kinase 2Homo sapiens (human)
centriole replicationCyclin-dependent kinase 2Homo sapiens (human)
Ras protein signal transductionCyclin-dependent kinase 2Homo sapiens (human)
regulation of mitotic cell cycleCyclin-dependent kinase 2Homo sapiens (human)
positive regulation of cell population proliferationCyclin-dependent kinase 2Homo sapiens (human)
peptidyl-serine phosphorylationCyclin-dependent kinase 2Homo sapiens (human)
positive regulation of heterochromatin formationCyclin-dependent kinase 2Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCyclin-dependent kinase 2Homo sapiens (human)
positive regulation of DNA-templated DNA replication initiationCyclin-dependent kinase 2Homo sapiens (human)
telomere maintenance in response to DNA damageCyclin-dependent kinase 2Homo sapiens (human)
post-translational protein modificationCyclin-dependent kinase 2Homo sapiens (human)
positive regulation of DNA replicationCyclin-dependent kinase 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionCyclin-dependent kinase 2Homo sapiens (human)
centrosome duplicationCyclin-dependent kinase 2Homo sapiens (human)
cell divisionCyclin-dependent kinase 2Homo sapiens (human)
meiotic cell cycleCyclin-dependent kinase 2Homo sapiens (human)
cellular response to nitric oxideCyclin-dependent kinase 2Homo sapiens (human)
cellular senescenceCyclin-dependent kinase 2Homo sapiens (human)
regulation of anaphase-promoting complex-dependent catabolic processCyclin-dependent kinase 2Homo sapiens (human)
regulation of G2/M transition of mitotic cell cycleCyclin-dependent kinase 2Homo sapiens (human)
signal transductionCyclin-dependent kinase 2Homo sapiens (human)
regulation of gene expressionCyclin-dependent kinase 2Homo sapiens (human)
response to organic substanceCyclin-dependent kinase 2Homo sapiens (human)
desensitization of G protein-coupled receptor signaling pathwayBeta-adrenergic receptor kinase 1Homo sapiens (human)
negative regulation of the force of heart contraction by chemical signalBeta-adrenergic receptor kinase 1Homo sapiens (human)
G protein-coupled receptor signaling pathwayBeta-adrenergic receptor kinase 1Homo sapiens (human)
G protein-coupled acetylcholine receptor signaling pathwayBeta-adrenergic receptor kinase 1Homo sapiens (human)
tachykinin receptor signaling pathwayBeta-adrenergic receptor kinase 1Homo sapiens (human)
heart developmentBeta-adrenergic receptor kinase 1Homo sapiens (human)
peptidyl-serine phosphorylationBeta-adrenergic receptor kinase 1Homo sapiens (human)
viral genome replicationBeta-adrenergic receptor kinase 1Homo sapiens (human)
receptor internalizationBeta-adrenergic receptor kinase 1Homo sapiens (human)
positive regulation of catecholamine secretionBeta-adrenergic receptor kinase 1Homo sapiens (human)
negative regulation of striated muscle contractionBeta-adrenergic receptor kinase 1Homo sapiens (human)
symbiont entry into host cellBeta-adrenergic receptor kinase 1Homo sapiens (human)
cardiac muscle contractionBeta-adrenergic receptor kinase 1Homo sapiens (human)
negative regulation of relaxation of smooth muscleBeta-adrenergic receptor kinase 1Homo sapiens (human)
regulation of the force of heart contractionBeta-adrenergic receptor kinase 1Homo sapiens (human)
protein phosphorylationBeta-adrenergic receptor kinase 1Homo sapiens (human)
P-body assemblyProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
miRNA-mediated gene silencing by inhibition of translationProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
negative regulation of translationProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
viral RNA genome packagingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
stem cell population maintenanceProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
neuron differentiationProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
P-body assemblyProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
negative regulation of neuron differentiationProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
stress granule assemblyProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
positive regulation of macrophage chemotaxisMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of macrophage proliferationMitogen-activated protein kinase 3 Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase 3 Homo sapiens (human)
DNA-templated transcriptionMitogen-activated protein kinase 3 Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase 3 Homo sapiens (human)
apoptotic processMitogen-activated protein kinase 3 Homo sapiens (human)
insulin receptor signaling pathwayMitogen-activated protein kinase 3 Homo sapiens (human)
Schwann cell developmentMitogen-activated protein kinase 3 Homo sapiens (human)
phosphorylationMitogen-activated protein kinase 3 Homo sapiens (human)
sensory perception of painMitogen-activated protein kinase 3 Homo sapiens (human)
regulation of ossificationMitogen-activated protein kinase 3 Homo sapiens (human)
BMP signaling pathwayMitogen-activated protein kinase 3 Homo sapiens (human)
regulation of cellular pHMitogen-activated protein kinase 3 Homo sapiens (human)
thyroid gland developmentMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of cyclase activityMitogen-activated protein kinase 3 Homo sapiens (human)
lipopolysaccharide-mediated signaling pathwayMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of telomere maintenance via telomeraseMitogen-activated protein kinase 3 Homo sapiens (human)
regulation of stress-activated MAPK cascadeMitogen-activated protein kinase 3 Homo sapiens (human)
cellular response to amino acid starvationMitogen-activated protein kinase 3 Homo sapiens (human)
cellular response to reactive oxygen speciesMitogen-activated protein kinase 3 Homo sapiens (human)
peptidyl-tyrosine autophosphorylationMitogen-activated protein kinase 3 Homo sapiens (human)
ERBB2-ERBB3 signaling pathwayMitogen-activated protein kinase 3 Homo sapiens (human)
outer ear morphogenesisMitogen-activated protein kinase 3 Homo sapiens (human)
myelinationMitogen-activated protein kinase 3 Homo sapiens (human)
signal transduction in response to DNA damageMitogen-activated protein kinase 3 Homo sapiens (human)
response to exogenous dsRNAMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIMitogen-activated protein kinase 3 Homo sapiens (human)
insulin-like growth factor receptor signaling pathwayMitogen-activated protein kinase 3 Homo sapiens (human)
thymus developmentMitogen-activated protein kinase 3 Homo sapiens (human)
modulation of chemical synaptic transmissionMitogen-activated protein kinase 3 Homo sapiens (human)
cartilage developmentMitogen-activated protein kinase 3 Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase 3 Homo sapiens (human)
regulation of cytoskeleton organizationMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of telomerase activityMitogen-activated protein kinase 3 Homo sapiens (human)
Bergmann glial cell differentiationMitogen-activated protein kinase 3 Homo sapiens (human)
face developmentMitogen-activated protein kinase 3 Homo sapiens (human)
lung morphogenesisMitogen-activated protein kinase 3 Homo sapiens (human)
trachea formationMitogen-activated protein kinase 3 Homo sapiens (human)
cardiac neural crest cell development involved in heart developmentMitogen-activated protein kinase 3 Homo sapiens (human)
ERK1 and ERK2 cascadeMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeMitogen-activated protein kinase 3 Homo sapiens (human)
interleukin-1-mediated signaling pathwayMitogen-activated protein kinase 3 Homo sapiens (human)
response to epidermal growth factorMitogen-activated protein kinase 3 Homo sapiens (human)
cellular response to mechanical stimulusMitogen-activated protein kinase 3 Homo sapiens (human)
cellular response to cadmium ionMitogen-activated protein kinase 3 Homo sapiens (human)
cellular response to tumor necrosis factorMitogen-activated protein kinase 3 Homo sapiens (human)
caveolin-mediated endocytosisMitogen-activated protein kinase 3 Homo sapiens (human)
regulation of Golgi inheritanceMitogen-activated protein kinase 3 Homo sapiens (human)
xenophagyMitogen-activated protein kinase 3 Homo sapiens (human)
negative regulation of TORC1 signalingMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of telomere cappingMitogen-activated protein kinase 3 Homo sapiens (human)
positive regulation of xenophagyMitogen-activated protein kinase 3 Homo sapiens (human)
regulation of early endosome to late endosome transportMitogen-activated protein kinase 3 Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase 3 Homo sapiens (human)
protein phosphorylationMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
peptidyl-serine phosphorylationMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
positive regulation of protein bindingMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
negative regulation of hippo signalingMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
peptidyl-serine autophosphorylationMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
negative regulation of protein localization to nucleusMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
intracellular signal transductionMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
pyrimidine nucleotide metabolic processDeoxycytidine kinaseHomo sapiens (human)
CMP biosynthetic processDeoxycytidine kinaseHomo sapiens (human)
dAMP salvageDeoxycytidine kinaseHomo sapiens (human)
nucleoside phosphate biosynthetic processDeoxycytidine kinaseHomo sapiens (human)
positive regulation of macrophage chemotaxisMitogen-activated protein kinase 1Homo sapiens (human)
positive regulation of macrophage proliferationMitogen-activated protein kinase 1Homo sapiens (human)
regulation of transcription by RNA polymerase IIMitogen-activated protein kinase 1Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase 1Homo sapiens (human)
apoptotic processMitogen-activated protein kinase 1Homo sapiens (human)
chemotaxisMitogen-activated protein kinase 1Homo sapiens (human)
DNA damage responseMitogen-activated protein kinase 1Homo sapiens (human)
signal transductionMitogen-activated protein kinase 1Homo sapiens (human)
chemical synaptic transmissionMitogen-activated protein kinase 1Homo sapiens (human)
learning or memoryMitogen-activated protein kinase 1Homo sapiens (human)
insulin receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationMitogen-activated protein kinase 1Homo sapiens (human)
Schwann cell developmentMitogen-activated protein kinase 1Homo sapiens (human)
peptidyl-serine phosphorylationMitogen-activated protein kinase 1Homo sapiens (human)
peptidyl-threonine phosphorylationMitogen-activated protein kinase 1Homo sapiens (human)
cytosine metabolic processMitogen-activated protein kinase 1Homo sapiens (human)
regulation of ossificationMitogen-activated protein kinase 1Homo sapiens (human)
androgen receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
regulation of cellular pHMitogen-activated protein kinase 1Homo sapiens (human)
thyroid gland developmentMitogen-activated protein kinase 1Homo sapiens (human)
regulation of protein stabilityMitogen-activated protein kinase 1Homo sapiens (human)
lipopolysaccharide-mediated signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
positive regulation of telomere maintenance via telomeraseMitogen-activated protein kinase 1Homo sapiens (human)
regulation of stress-activated MAPK cascadeMitogen-activated protein kinase 1Homo sapiens (human)
mammary gland epithelial cell proliferationMitogen-activated protein kinase 1Homo sapiens (human)
cellular response to amino acid starvationMitogen-activated protein kinase 1Homo sapiens (human)
cellular response to reactive oxygen speciesMitogen-activated protein kinase 1Homo sapiens (human)
response to nicotineMitogen-activated protein kinase 1Homo sapiens (human)
ERBB signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
ERBB2-ERBB3 signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
outer ear morphogenesisMitogen-activated protein kinase 1Homo sapiens (human)
myelinationMitogen-activated protein kinase 1Homo sapiens (human)
response to exogenous dsRNAMitogen-activated protein kinase 1Homo sapiens (human)
steroid hormone mediated signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
negative regulation of cell differentiationMitogen-activated protein kinase 1Homo sapiens (human)
insulin-like growth factor receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
thymus developmentMitogen-activated protein kinase 1Homo sapiens (human)
progesterone receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
T cell receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
B cell receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase 1Homo sapiens (human)
regulation of cytoskeleton organizationMitogen-activated protein kinase 1Homo sapiens (human)
positive regulation of telomerase activityMitogen-activated protein kinase 1Homo sapiens (human)
Bergmann glial cell differentiationMitogen-activated protein kinase 1Homo sapiens (human)
long-term synaptic potentiationMitogen-activated protein kinase 1Homo sapiens (human)
face developmentMitogen-activated protein kinase 1Homo sapiens (human)
lung morphogenesisMitogen-activated protein kinase 1Homo sapiens (human)
trachea formationMitogen-activated protein kinase 1Homo sapiens (human)
labyrinthine layer blood vessel developmentMitogen-activated protein kinase 1Homo sapiens (human)
cardiac neural crest cell development involved in heart developmentMitogen-activated protein kinase 1Homo sapiens (human)
ERK1 and ERK2 cascadeMitogen-activated protein kinase 1Homo sapiens (human)
response to epidermal growth factorMitogen-activated protein kinase 1Homo sapiens (human)
cellular response to cadmium ionMitogen-activated protein kinase 1Homo sapiens (human)
cellular response to tumor necrosis factorMitogen-activated protein kinase 1Homo sapiens (human)
caveolin-mediated endocytosisMitogen-activated protein kinase 1Homo sapiens (human)
regulation of Golgi inheritanceMitogen-activated protein kinase 1Homo sapiens (human)
positive regulation of telomere cappingMitogen-activated protein kinase 1Homo sapiens (human)
regulation of early endosome to late endosome transportMitogen-activated protein kinase 1Homo sapiens (human)
cell surface receptor signaling pathwayMitogen-activated protein kinase 1Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase 1Homo sapiens (human)
skeletal system developmentEphrin type-A receptor 2Homo sapiens (human)
vasculogenesisEphrin type-A receptor 2Homo sapiens (human)
osteoblast differentiationEphrin type-A receptor 2Homo sapiens (human)
blood vessel endothelial cell proliferation involved in sprouting angiogenesisEphrin type-A receptor 2Homo sapiens (human)
inflammatory responseEphrin type-A receptor 2Homo sapiens (human)
cell adhesionEphrin type-A receptor 2Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damageEphrin type-A receptor 2Homo sapiens (human)
regulation of lamellipodium assemblyEphrin type-A receptor 2Homo sapiens (human)
notochord formationEphrin type-A receptor 2Homo sapiens (human)
cell migrationEphrin type-A receptor 2Homo sapiens (human)
negative regulation of angiogenesisEphrin type-A receptor 2Homo sapiens (human)
neural tube developmentEphrin type-A receptor 2Homo sapiens (human)
neuron differentiationEphrin type-A receptor 2Homo sapiens (human)
keratinocyte differentiationEphrin type-A receptor 2Homo sapiens (human)
osteoclast differentiationEphrin type-A receptor 2Homo sapiens (human)
positive regulation of cell migrationEphrin type-A receptor 2Homo sapiens (human)
negative regulation of chemokine productionEphrin type-A receptor 2Homo sapiens (human)
mammary gland epithelial cell proliferationEphrin type-A receptor 2Homo sapiens (human)
regulation of cell adhesion mediated by integrinEphrin type-A receptor 2Homo sapiens (human)
post-anal tail morphogenesisEphrin type-A receptor 2Homo sapiens (human)
regulation of blood vessel endothelial cell migrationEphrin type-A receptor 2Homo sapiens (human)
regulation of angiogenesisEphrin type-A receptor 2Homo sapiens (human)
cAMP metabolic processEphrin type-A receptor 2Homo sapiens (human)
symbiont entry into host cellEphrin type-A receptor 2Homo sapiens (human)
bone remodelingEphrin type-A receptor 2Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-A receptor 2Homo sapiens (human)
axial mesoderm formationEphrin type-A receptor 2Homo sapiens (human)
cell motilityEphrin type-A receptor 2Homo sapiens (human)
defense response to Gram-positive bacteriumEphrin type-A receptor 2Homo sapiens (human)
notochord cell developmentEphrin type-A receptor 2Homo sapiens (human)
cell chemotaxisEphrin type-A receptor 2Homo sapiens (human)
branching involved in mammary gland duct morphogenesisEphrin type-A receptor 2Homo sapiens (human)
lens fiber cell morphogenesisEphrin type-A receptor 2Homo sapiens (human)
regulation of ERK1 and ERK2 cascadeEphrin type-A receptor 2Homo sapiens (human)
response to growth factorEphrin type-A receptor 2Homo sapiens (human)
protein localization to plasma membraneEphrin type-A receptor 2Homo sapiens (human)
activation of GTPase activityEphrin type-A receptor 2Homo sapiens (human)
negative regulation of lymphangiogenesisEphrin type-A receptor 2Homo sapiens (human)
positive regulation of protein localization to plasma membraneEphrin type-A receptor 2Homo sapiens (human)
positive regulation of bicellular tight junction assemblyEphrin type-A receptor 2Homo sapiens (human)
pericyte cell differentiationEphrin type-A receptor 2Homo sapiens (human)
positive regulation of kinase activityEphrin type-A receptor 2Homo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayEphrin type-A receptor 2Homo sapiens (human)
multicellular organism developmentEphrin type-A receptor 2Homo sapiens (human)
negative regulation of protein kinase activityEphrin type-B receptor 2Homo sapiens (human)
regulation of autophagosome assemblyEphrin type-B receptor 2Homo sapiens (human)
angiogenesisEphrin type-B receptor 2Homo sapiens (human)
urogenital system developmentEphrin type-B receptor 2Homo sapiens (human)
negative regulation of protein phosphorylationEphrin type-B receptor 2Homo sapiens (human)
positive regulation of immunoglobulin productionEphrin type-B receptor 2Homo sapiens (human)
negative regulation of cell adhesionEphrin type-B receptor 2Homo sapiens (human)
nervous system developmentEphrin type-B receptor 2Homo sapiens (human)
axon guidanceEphrin type-B receptor 2Homo sapiens (human)
axonal fasciculationEphrin type-B receptor 2Homo sapiens (human)
learning or memoryEphrin type-B receptor 2Homo sapiens (human)
learningEphrin type-B receptor 2Homo sapiens (human)
positive regulation of gene expressionEphrin type-B receptor 2Homo sapiens (human)
phosphorylationEphrin type-B receptor 2Homo sapiens (human)
peptidyl-tyrosine phosphorylationEphrin type-B receptor 2Homo sapiens (human)
optic nerve morphogenesisEphrin type-B receptor 2Homo sapiens (human)
hindbrain tangential cell migrationEphrin type-B receptor 2Homo sapiens (human)
central nervous system projection neuron axonogenesisEphrin type-B receptor 2Homo sapiens (human)
corpus callosum developmentEphrin type-B receptor 2Homo sapiens (human)
regulation of blood coagulationEphrin type-B receptor 2Homo sapiens (human)
positive regulation of cell migrationEphrin type-B receptor 2Homo sapiens (human)
positive regulation of B cell proliferationEphrin type-B receptor 2Homo sapiens (human)
retinal ganglion cell axon guidanceEphrin type-B receptor 2Homo sapiens (human)
positive regulation of synaptic plasticityEphrin type-B receptor 2Homo sapiens (human)
positive regulation of tumor necrosis factor productionEphrin type-B receptor 2Homo sapiens (human)
B cell activationEphrin type-B receptor 2Homo sapiens (human)
inner ear morphogenesisEphrin type-B receptor 2Homo sapiens (human)
regulation of receptor signaling pathway via JAK-STATEphrin type-B receptor 2Homo sapiens (human)
negative regulation of Ras protein signal transductionEphrin type-B receptor 2Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-B receptor 2Homo sapiens (human)
regulation of neuronal synaptic plasticityEphrin type-B receptor 2Homo sapiens (human)
positive regulation of long-term neuronal synaptic plasticityEphrin type-B receptor 2Homo sapiens (human)
camera-type eye morphogenesisEphrin type-B receptor 2Homo sapiens (human)
negative regulation of axonogenesisEphrin type-B receptor 2Homo sapiens (human)
regulation of body fluid levelsEphrin type-B receptor 2Homo sapiens (human)
regulation of filopodium assemblyEphrin type-B receptor 2Homo sapiens (human)
positive regulation of synapse assemblyEphrin type-B receptor 2Homo sapiens (human)
roof of mouth developmentEphrin type-B receptor 2Homo sapiens (human)
dendritic spine developmentEphrin type-B receptor 2Homo sapiens (human)
dendritic spine morphogenesisEphrin type-B receptor 2Homo sapiens (human)
positive regulation of dendritic spine morphogenesisEphrin type-B receptor 2Homo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeEphrin type-B receptor 2Homo sapiens (human)
cellular response to lipopolysaccharideEphrin type-B receptor 2Homo sapiens (human)
commissural neuron axon guidanceEphrin type-B receptor 2Homo sapiens (human)
postsynaptic membrane assemblyEphrin type-B receptor 2Homo sapiens (human)
trans-synaptic signaling by trans-synaptic complex, modulating synaptic transmissionEphrin type-B receptor 2Homo sapiens (human)
neuron projection retractionEphrin type-B receptor 2Homo sapiens (human)
vesicle-mediated intercellular transportEphrin type-B receptor 2Homo sapiens (human)
tight junction assemblyEphrin type-B receptor 2Homo sapiens (human)
negative regulation of cytokine production involved in inflammatory responseEphrin type-B receptor 2Homo sapiens (human)
positive regulation of long-term synaptic potentiationEphrin type-B receptor 2Homo sapiens (human)
positive regulation of protein localization to plasma membraneEphrin type-B receptor 2Homo sapiens (human)
cellular response to amyloid-betaEphrin type-B receptor 2Homo sapiens (human)
negative regulation of NMDA glutamate receptor activityEphrin type-B receptor 2Homo sapiens (human)
positive regulation of NMDA glutamate receptor activityEphrin type-B receptor 2Homo sapiens (human)
positive regulation of protein localization to cell surfaceEphrin type-B receptor 2Homo sapiens (human)
regulation of T-helper 17 type immune responseEphrin type-B receptor 2Homo sapiens (human)
regulation of behavioral fear responseEphrin type-B receptor 2Homo sapiens (human)
protein phosphorylationEphrin type-B receptor 2Homo sapiens (human)
protein phosphorylationNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
immune responseNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cell surface receptor signaling pathway via JAK-STATNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cytokine-mediated signaling pathwayNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of type II interferon productionNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of interleukin-17 productionNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of natural killer cell proliferationNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
interleukin-12-mediated signaling pathwayNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
type III interferon-mediated signaling pathwayNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of T cell proliferationNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of receptor signaling pathway via JAK-STATNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of NK T cell proliferationNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
type II interferon-mediated signaling pathwayNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
type I interferon-mediated signaling pathwayNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cellular response to virusNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
interleukin-10-mediated signaling pathwayNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of protein localization to nucleusNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
positive regulation of T-helper 17 type immune responseNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
intracellular signal transductionNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cell differentiationNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
growth hormone receptor signaling pathway via JAK-STATNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
'de novo' pyrimidine nucleobase biosynthetic processUMP-CMP kinase Homo sapiens (human)
UMP biosynthetic processUMP-CMP kinase Homo sapiens (human)
UDP biosynthetic processUMP-CMP kinase Homo sapiens (human)
pyrimidine ribonucleotide biosynthetic processUMP-CMP kinase Homo sapiens (human)
nucleobase-containing small molecule interconversionUMP-CMP kinase Homo sapiens (human)
nucleoside monophosphate phosphorylationUMP-CMP kinase Homo sapiens (human)
CDP biosynthetic processUMP-CMP kinase Homo sapiens (human)
negative regulation of MAPK cascadePhosphatidylethanolamine-binding protein 1Homo sapiens (human)
G2/M transition of mitotic cell cycleWee1-like protein kinaseHomo sapiens (human)
microtubule cytoskeleton organizationWee1-like protein kinaseHomo sapiens (human)
negative regulation of G2/M transition of mitotic cell cycleWee1-like protein kinaseHomo sapiens (human)
establishment of cell polarityWee1-like protein kinaseHomo sapiens (human)
positive regulation of DNA replicationWee1-like protein kinaseHomo sapiens (human)
neuron projection morphogenesisWee1-like protein kinaseHomo sapiens (human)
cell divisionWee1-like protein kinaseHomo sapiens (human)
negative regulation of G1/S transition of mitotic cell cycleWee1-like protein kinaseHomo sapiens (human)
protein phosphorylationWee1-like protein kinaseHomo sapiens (human)
response to hypoxiaHeme oxygenase 2Homo sapiens (human)
response to oxidative stressHeme oxygenase 2Homo sapiens (human)
heme catabolic processHeme oxygenase 2Homo sapiens (human)
heme oxidationHeme oxygenase 2Homo sapiens (human)
S-adenosylmethionine biosynthetic processS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
one-carbon metabolic processS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
protein hexamerizationS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
protein heterooligomerizationS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
cellular response to methionineS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
positive regulation of TORC1 signalingS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
cellular response to leukemia inhibitory factorS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
protein foldingDnaJ homolog subfamily A member 1Homo sapiens (human)
response to unfolded proteinDnaJ homolog subfamily A member 1Homo sapiens (human)
spermatogenesisDnaJ homolog subfamily A member 1Homo sapiens (human)
response to heatDnaJ homolog subfamily A member 1Homo sapiens (human)
flagellated sperm motilityDnaJ homolog subfamily A member 1Homo sapiens (human)
androgen receptor signaling pathwayDnaJ homolog subfamily A member 1Homo sapiens (human)
negative regulation of protein ubiquitinationDnaJ homolog subfamily A member 1Homo sapiens (human)
positive regulation of apoptotic processDnaJ homolog subfamily A member 1Homo sapiens (human)
negative regulation of apoptotic processDnaJ homolog subfamily A member 1Homo sapiens (human)
negative regulation of JUN kinase activityDnaJ homolog subfamily A member 1Homo sapiens (human)
regulation of protein transportDnaJ homolog subfamily A member 1Homo sapiens (human)
protein localization to mitochondrionDnaJ homolog subfamily A member 1Homo sapiens (human)
negative regulation of establishment of protein localization to mitochondrionDnaJ homolog subfamily A member 1Homo sapiens (human)
negative regulation of nitrosative stress-induced intrinsic apoptotic signaling pathwayDnaJ homolog subfamily A member 1Homo sapiens (human)
protein refoldingDnaJ homolog subfamily A member 1Homo sapiens (human)
protein phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
activation-induced cell death of T cellsRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
intracellular signal transductionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
osteoblast differentiationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
maternal placenta developmentRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of endothelial cell proliferationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cell migration involved in sprouting angiogenesisRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
sphingosine-1-phosphate receptor signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
glycogen biosynthetic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of glycogen biosynthetic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
glucose metabolic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of translationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of protein kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein import into nucleusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
nitric oxide biosynthetic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
inflammatory responseRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to oxidative stressRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
signal transductionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
epidermal growth factor receptor signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
G protein-coupled receptor signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
canonical NF-kappaB signal transductionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cell population proliferationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
insulin receptor signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
apoptotic mitochondrial changesRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to heatRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
gene expressionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of autophagyRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of endothelial cell migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of gene expressionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of gene expressionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of long-chain fatty acid import across plasma membraneRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
fibroblast migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of fibroblast migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of sodium ion transportRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of glucose metabolic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of endopeptidase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of neuron projection developmentRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of macroautophagyRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein ubiquitinationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
peptidyl-serine phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
peptidyl-threonine phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
virus-mediated perturbation of host defense responseRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cytokine-mediated signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
mammalian oogenesis stageRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cell differentiationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cell growthRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of cell migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cell migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
T cell costimulationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of protein ubiquitinationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of myelinationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
lipopolysaccharide-mediated signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
TOR signalingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of fatty acid beta-oxidationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of endodeoxyribonuclease activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of protein bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to foodRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
peripheral nervous system myelin maintenanceRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to insulin stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to fluid shear stressRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to reactive oxygen speciesRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
interleukin-18-mediated signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to vascular endothelial growth factor stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to decreased oxygen levelsRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
non-canonical NF-kappaB signal transductionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
glucose homeostasisRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of apoptotic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of apoptotic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
anoikisRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of mRNA stabilityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of blood vessel endothelial cell migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of nitric oxide biosynthetic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of fat cell differentiationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of glycogen biosynthetic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cyclin-dependent protein serine/threonine kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of Notch signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of proteolysisRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of DNA-templated transcriptionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of glucose importRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of organ growthRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein autophosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of lipid biosynthetic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
insulin-like growth factor receptor signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
behavioral response to painRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of smooth muscle cell proliferationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of nitric-oxide synthase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of DNA-binding transcription factor activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
striated muscle cell differentiationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein metabolic processRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
excitatory postsynaptic potentialRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to growth hormoneRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
mammary gland epithelial cell differentiationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
labyrinthine layer blood vessel developmentRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to UV-ARAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to growth factorRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to cadmium ionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to tumor necrosis factorRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to epidermal growth factor stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to prostaglandin E stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of protein serine/threonine kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
establishment of protein localization to mitochondrionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
maintenance of protein location in mitochondrionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of release of cytochrome c from mitochondriaRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to granulocyte macrophage colony-stimulating factor stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
execution phase of apoptosisRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of postsynapse organizationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of tRNA methylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to oxidised low-density lipoprotein particle stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of protein localization to lysosomeRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of cGAS/STING signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of G1/S transition of mitotic cell cycleRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein localization to nucleusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to peptideRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of signal transduction by p53 class mediatorRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of cilium assemblyRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of oxidative stress-induced intrinsic apoptotic signaling pathwayRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of leukocyte cell-cell adhesionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein localization to plasma membraneRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of I-kappaB phosphorylationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of TORC1 signalingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein localization to endoplasmic reticulumRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cellular response to nerve growth factor stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
response to insulin-like growth factor stimulusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein localization to cell surfaceRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
regulation of type B pancreatic cell developmentRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of lymphocyte migrationRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathway in absence of ligandRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
intracellular signal transductionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
glycogen biosynthetic processRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
glucose metabolic processRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
regulation of translationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
signal transductionRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
insulin receptor signaling pathwayRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of long-chain fatty acid import across plasma membraneRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of glucose metabolic processRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
regulation of cell migrationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cell migrationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of fatty acid beta-oxidationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
peripheral nervous system myelin maintenanceRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
cellular response to insulin stimulusRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
protein modification processRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
fat cell differentiationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of glycogen biosynthetic processRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of glucose importRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
regulation of cell cycleRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
mammary gland epithelial cell differentiationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
cellular response to high light intensityRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
organic substance transportRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
protein localization to plasma membraneRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of protein targeting to membraneRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
retinal rod cell apoptotic processRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cell motilityRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
intracellular signal transductionRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
peptidyl-serine phosphorylationRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
spindle organizationDual specificity protein kinase TTKHomo sapiens (human)
mitotic spindle organizationDual specificity protein kinase TTKHomo sapiens (human)
positive regulation of cell population proliferationDual specificity protein kinase TTKHomo sapiens (human)
female meiosis chromosome segregationDual specificity protein kinase TTKHomo sapiens (human)
protein localization to meiotic spindle midzoneDual specificity protein kinase TTKHomo sapiens (human)
chromosome segregationDual specificity protein kinase TTKHomo sapiens (human)
peptidyl-serine phosphorylationDual specificity protein kinase TTKHomo sapiens (human)
protein localization to kinetochoreDual specificity protein kinase TTKHomo sapiens (human)
mitotic spindle assembly checkpoint signalingDual specificity protein kinase TTKHomo sapiens (human)
meiotic spindle assembly checkpoint signalingDual specificity protein kinase TTKHomo sapiens (human)
DNA replicationDNA replication licensing factor MCM4Homo sapiens (human)
DNA unwinding involved in DNA replicationDNA replication licensing factor MCM4Homo sapiens (human)
regulation of DNA-templated DNA replication initiationDNA replication licensing factor MCM4Homo sapiens (human)
double-strand break repair via break-induced replicationDNA replication licensing factor MCM4Homo sapiens (human)
DNA strand elongation involved in DNA replicationDNA replication licensing factor MCM4Homo sapiens (human)
mitotic DNA replication initiationDNA replication licensing factor MCM4Homo sapiens (human)
mitotic cytokinesisMyosin-10Homo sapiens (human)
actin filament-based movementMyosin-10Homo sapiens (human)
cell adhesionMyosin-10Homo sapiens (human)
actomyosin structure organizationMyosin-10Homo sapiens (human)
positive regulation of protein secretionMyosin-10Homo sapiens (human)
mitotic cytokinesisMyosin-10Homo sapiens (human)
regulation of cell shapeMyosin-10Homo sapiens (human)
heart developmentDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
positive regulation of gene expressionDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
Schwann cell developmentDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
thyroid gland developmentDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
regulation of stress-activated MAPK cascadeDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
peptidyl-serine autophosphorylationDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
ERBB2-ERBB3 signaling pathwayDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
myelinationDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
insulin-like growth factor receptor signaling pathwayDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
thymus developmentDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
regulation of axon regenerationDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
positive regulation of axonogenesisDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
face developmentDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
trachea formationDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
epithelial cell proliferation involved in lung morphogenesisDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
ERK1 and ERK2 cascadeDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
positive regulation of protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
regulation of Golgi inheritanceDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
positive regulation of cell motilityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
regulation of early endosome to late endosome transportDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
MAPK cascadeDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
hemopoiesisReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
leukocyte homeostasisReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
myeloid progenitor cell differentiationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
pro-B cell differentiationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
positive regulation of cell population proliferationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
response to organonitrogen compoundReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
peptidyl-tyrosine phosphorylationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
cytokine-mediated signaling pathwayReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
B cell differentiationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
animal organ regenerationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
common myeloid progenitor cell proliferationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
vascular endothelial growth factor signaling pathwayReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
positive regulation of tyrosine phosphorylation of STAT proteinReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
regulation of apoptotic processReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
positive regulation of MAP kinase activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
positive regulation of MAPK cascadeReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
lymphocyte proliferationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
protein autophosphorylationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
cellular response to cytokine stimulusReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
cellular response to glucocorticoid stimulusReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
dendritic cell differentiationReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
positive regulation of kinase activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
multicellular organism developmentReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
regulation of cardiac muscle cell apoptotic processBone morphogenetic protein receptor type-1AHomo sapiens (human)
regulation of neural crest cell differentiationBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of gene expressionBone morphogenetic protein receptor type-1AHomo sapiens (human)
negative regulation of gene expressionBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of transforming growth factor beta2 productionBone morphogenetic protein receptor type-1AHomo sapiens (human)
angiogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
osteoblast differentiationBone morphogenetic protein receptor type-1AHomo sapiens (human)
in utero embryonic developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
mesoderm formationBone morphogenetic protein receptor type-1AHomo sapiens (human)
somitogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
Mullerian duct regressionBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of mesenchymal cell proliferationBone morphogenetic protein receptor type-1AHomo sapiens (human)
chondrocyte differentiationBone morphogenetic protein receptor type-1AHomo sapiens (human)
outflow tract septum morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
outflow tract morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
cardiac conduction system developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
atrioventricular valve developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
mitral valve morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
tricuspid valve morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
endocardial cushion morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
cardiac right ventricle morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
ventricular trabecula myocardium morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
ventricular compact myocardium morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
endocardial cushion formationBone morphogenetic protein receptor type-1AHomo sapiens (human)
immune responseBone morphogenetic protein receptor type-1AHomo sapiens (human)
transforming growth factor beta receptor signaling pathwayBone morphogenetic protein receptor type-1AHomo sapiens (human)
ectoderm developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
dorsal/ventral axis specificationBone morphogenetic protein receptor type-1AHomo sapiens (human)
neural crest cell developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
negative regulation of smooth muscle cell migrationBone morphogenetic protein receptor type-1AHomo sapiens (human)
central nervous system neuron differentiationBone morphogenetic protein receptor type-1AHomo sapiens (human)
pituitary gland developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
neural plate mediolateral regionalizationBone morphogenetic protein receptor type-1AHomo sapiens (human)
lung developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of bone mineralizationBone morphogenetic protein receptor type-1AHomo sapiens (human)
BMP signaling pathwayBone morphogenetic protein receptor type-1AHomo sapiens (human)
somatic stem cell population maintenanceBone morphogenetic protein receptor type-1AHomo sapiens (human)
hindlimb morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
dorsal aorta morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
odontogenesis of dentin-containing toothBone morphogenetic protein receptor type-1AHomo sapiens (human)
embryonic digit morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of osteoblast differentiationBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIBone morphogenetic protein receptor type-1AHomo sapiens (human)
paraxial mesoderm structural organizationBone morphogenetic protein receptor type-1AHomo sapiens (human)
lateral mesoderm developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
regulation of lateral mesodermal cell fate specificationBone morphogenetic protein receptor type-1AHomo sapiens (human)
mesendoderm developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
embryonic organ developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
developmental growthBone morphogenetic protein receptor type-1AHomo sapiens (human)
epithelial cell proliferationBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of epithelial cell proliferationBone morphogenetic protein receptor type-1AHomo sapiens (human)
negative regulation of neurogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
negative regulation of muscle cell differentiationBone morphogenetic protein receptor type-1AHomo sapiens (human)
roof of mouth developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
regulation of cardiac muscle cell proliferationBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of cardiac muscle cell proliferationBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of SMAD protein signal transductionBone morphogenetic protein receptor type-1AHomo sapiens (human)
ventricular septum morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
heart formationBone morphogenetic protein receptor type-1AHomo sapiens (human)
atrioventricular node cell developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
pharyngeal arch artery morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
cellular response to BMP stimulusBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of miRNA transcriptionBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of cardiac ventricle developmentBone morphogenetic protein receptor type-1AHomo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationBone morphogenetic protein receptor type-1AHomo sapiens (human)
fibrous ring of heart morphogenesisBone morphogenetic protein receptor type-1AHomo sapiens (human)
regulation of cellular senescenceBone morphogenetic protein receptor type-1AHomo sapiens (human)
protein phosphorylationBone morphogenetic protein receptor type-1AHomo sapiens (human)
dorsal/ventral pattern formationBone morphogenetic protein receptor type-1AHomo sapiens (human)
cellular response to growth factor stimulusBone morphogenetic protein receptor type-1AHomo sapiens (human)
G1/S transition of mitotic cell cycleActivin receptor type-1BHomo sapiens (human)
in utero embryonic developmentActivin receptor type-1BHomo sapiens (human)
hair follicle developmentActivin receptor type-1BHomo sapiens (human)
regulation of DNA-templated transcriptionActivin receptor type-1BHomo sapiens (human)
signal transductionActivin receptor type-1BHomo sapiens (human)
cell surface receptor protein serine/threonine kinase signaling pathwayActivin receptor type-1BHomo sapiens (human)
positive regulation of gene expressionActivin receptor type-1BHomo sapiens (human)
negative regulation of gene expressionActivin receptor type-1BHomo sapiens (human)
peptidyl-threonine phosphorylationActivin receptor type-1BHomo sapiens (human)
negative regulation of cell growthActivin receptor type-1BHomo sapiens (human)
activin receptor signaling pathwayActivin receptor type-1BHomo sapiens (human)
positive regulation of activin receptor signaling pathwayActivin receptor type-1BHomo sapiens (human)
nodal signaling pathwayActivin receptor type-1BHomo sapiens (human)
positive regulation of erythrocyte differentiationActivin receptor type-1BHomo sapiens (human)
protein autophosphorylationActivin receptor type-1BHomo sapiens (human)
extrinsic apoptotic signaling pathwayActivin receptor type-1BHomo sapiens (human)
positive regulation of trophoblast cell migrationActivin receptor type-1BHomo sapiens (human)
cellular response to growth factor stimulusActivin receptor type-1BHomo sapiens (human)
protein phosphorylationActivin receptor type-1BHomo sapiens (human)
nervous system developmentActivin receptor type-1BHomo sapiens (human)
proepicardium developmentTGF-beta receptor type-1Homo sapiens (human)
negative regulation of cell migrationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of extracellular matrix assemblyTGF-beta receptor type-1Homo sapiens (human)
skeletal system developmentTGF-beta receptor type-1Homo sapiens (human)
in utero embryonic developmentTGF-beta receptor type-1Homo sapiens (human)
kidney developmentTGF-beta receptor type-1Homo sapiens (human)
blastocyst developmentTGF-beta receptor type-1Homo sapiens (human)
epithelial to mesenchymal transitionTGF-beta receptor type-1Homo sapiens (human)
endothelial cell proliferationTGF-beta receptor type-1Homo sapiens (human)
negative regulation of endothelial cell proliferationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of endothelial cell proliferationTGF-beta receptor type-1Homo sapiens (human)
lens development in camera-type eyeTGF-beta receptor type-1Homo sapiens (human)
ventricular trabecula myocardium morphogenesisTGF-beta receptor type-1Homo sapiens (human)
ventricular compact myocardium morphogenesisTGF-beta receptor type-1Homo sapiens (human)
regulation of DNA-templated transcriptionTGF-beta receptor type-1Homo sapiens (human)
apoptotic processTGF-beta receptor type-1Homo sapiens (human)
signal transductionTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayTGF-beta receptor type-1Homo sapiens (human)
heart developmentTGF-beta receptor type-1Homo sapiens (human)
positive regulation of cell population proliferationTGF-beta receptor type-1Homo sapiens (human)
germ cell migrationTGF-beta receptor type-1Homo sapiens (human)
male gonad developmentTGF-beta receptor type-1Homo sapiens (human)
post-embryonic developmentTGF-beta receptor type-1Homo sapiens (human)
anterior/posterior pattern specificationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of gene expressionTGF-beta receptor type-1Homo sapiens (human)
regulation of epithelial to mesenchymal transitionTGF-beta receptor type-1Homo sapiens (human)
positive regulation of epithelial to mesenchymal transitionTGF-beta receptor type-1Homo sapiens (human)
peptidyl-serine phosphorylationTGF-beta receptor type-1Homo sapiens (human)
collagen fibril organizationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of cell growthTGF-beta receptor type-1Homo sapiens (human)
positive regulation of cell migrationTGF-beta receptor type-1Homo sapiens (human)
regulation of protein ubiquitinationTGF-beta receptor type-1Homo sapiens (human)
negative regulation of chondrocyte differentiationTGF-beta receptor type-1Homo sapiens (human)
activin receptor signaling pathwayTGF-beta receptor type-1Homo sapiens (human)
intracellular signal transductionTGF-beta receptor type-1Homo sapiens (human)
myofibroblast differentiationTGF-beta receptor type-1Homo sapiens (human)
wound healingTGF-beta receptor type-1Homo sapiens (human)
endothelial cell activationTGF-beta receptor type-1Homo sapiens (human)
extracellular structure organizationTGF-beta receptor type-1Homo sapiens (human)
endothelial cell migrationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of DNA-templated transcriptionTGF-beta receptor type-1Homo sapiens (human)
filopodium assemblyTGF-beta receptor type-1Homo sapiens (human)
thymus developmentTGF-beta receptor type-1Homo sapiens (human)
neuron fate commitmentTGF-beta receptor type-1Homo sapiens (human)
embryonic cranial skeleton morphogenesisTGF-beta receptor type-1Homo sapiens (human)
skeletal system morphogenesisTGF-beta receptor type-1Homo sapiens (human)
mesenchymal cell differentiationTGF-beta receptor type-1Homo sapiens (human)
artery morphogenesisTGF-beta receptor type-1Homo sapiens (human)
cell motilityTGF-beta receptor type-1Homo sapiens (human)
positive regulation of filopodium assemblyTGF-beta receptor type-1Homo sapiens (human)
positive regulation of stress fiber assemblyTGF-beta receptor type-1Homo sapiens (human)
regulation of cell cycleTGF-beta receptor type-1Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionTGF-beta receptor type-1Homo sapiens (human)
parathyroid gland developmentTGF-beta receptor type-1Homo sapiens (human)
roof of mouth developmentTGF-beta receptor type-1Homo sapiens (human)
pharyngeal system developmentTGF-beta receptor type-1Homo sapiens (human)
regulation of cardiac muscle cell proliferationTGF-beta receptor type-1Homo sapiens (human)
cardiac epithelial to mesenchymal transitionTGF-beta receptor type-1Homo sapiens (human)
positive regulation of SMAD protein signal transductionTGF-beta receptor type-1Homo sapiens (human)
ventricular septum morphogenesisTGF-beta receptor type-1Homo sapiens (human)
angiogenesis involved in coronary vascular morphogenesisTGF-beta receptor type-1Homo sapiens (human)
coronary artery morphogenesisTGF-beta receptor type-1Homo sapiens (human)
response to cholesterolTGF-beta receptor type-1Homo sapiens (human)
cellular response to transforming growth factor beta stimulusTGF-beta receptor type-1Homo sapiens (human)
positive regulation of mesenchymal stem cell proliferationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of vasculature developmentTGF-beta receptor type-1Homo sapiens (human)
positive regulation of epithelial to mesenchymal transition involved in endocardial cushion formationTGF-beta receptor type-1Homo sapiens (human)
positive regulation of tight junction disassemblyTGF-beta receptor type-1Homo sapiens (human)
epicardium morphogenesisTGF-beta receptor type-1Homo sapiens (human)
positive regulation of apoptotic signaling pathwayTGF-beta receptor type-1Homo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathwayTGF-beta receptor type-1Homo sapiens (human)
protein phosphorylationTGF-beta receptor type-1Homo sapiens (human)
cellular response to growth factor stimulusTGF-beta receptor type-1Homo sapiens (human)
nervous system developmentTGF-beta receptor type-1Homo sapiens (human)
cell proliferation involved in endocardial cushion morphogenesisTGF-beta receptor type-2Homo sapiens (human)
superior endocardial cushion morphogenesisTGF-beta receptor type-2Homo sapiens (human)
blood vessel developmentTGF-beta receptor type-2Homo sapiens (human)
branching involved in blood vessel morphogenesisTGF-beta receptor type-2Homo sapiens (human)
vasculogenesisTGF-beta receptor type-2Homo sapiens (human)
in utero embryonic developmentTGF-beta receptor type-2Homo sapiens (human)
epithelial to mesenchymal transitionTGF-beta receptor type-2Homo sapiens (human)
heart loopingTGF-beta receptor type-2Homo sapiens (human)
positive regulation of mesenchymal cell proliferationTGF-beta receptor type-2Homo sapiens (human)
lens development in camera-type eyeTGF-beta receptor type-2Homo sapiens (human)
positive regulation of tolerance induction to self antigenTGF-beta receptor type-2Homo sapiens (human)
positive regulation of B cell tolerance inductionTGF-beta receptor type-2Homo sapiens (human)
positive regulation of T cell tolerance inductionTGF-beta receptor type-2Homo sapiens (human)
outflow tract septum morphogenesisTGF-beta receptor type-2Homo sapiens (human)
membranous septum morphogenesisTGF-beta receptor type-2Homo sapiens (human)
outflow tract morphogenesisTGF-beta receptor type-2Homo sapiens (human)
aortic valve morphogenesisTGF-beta receptor type-2Homo sapiens (human)
atrioventricular valve morphogenesisTGF-beta receptor type-2Homo sapiens (human)
tricuspid valve morphogenesisTGF-beta receptor type-2Homo sapiens (human)
cardiac left ventricle morphogenesisTGF-beta receptor type-2Homo sapiens (human)
endocardial cushion fusionTGF-beta receptor type-2Homo sapiens (human)
growth plate cartilage chondrocyte growthTGF-beta receptor type-2Homo sapiens (human)
apoptotic processTGF-beta receptor type-2Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayTGF-beta receptor type-2Homo sapiens (human)
Notch signaling pathwayTGF-beta receptor type-2Homo sapiens (human)
smoothened signaling pathwayTGF-beta receptor type-2Homo sapiens (human)
gastrulationTGF-beta receptor type-2Homo sapiens (human)
brain developmentTGF-beta receptor type-2Homo sapiens (human)
heart developmentTGF-beta receptor type-2Homo sapiens (human)
positive regulation of cell population proliferationTGF-beta receptor type-2Homo sapiens (human)
response to xenobiotic stimulusTGF-beta receptor type-2Homo sapiens (human)
regulation of gene expressionTGF-beta receptor type-2Homo sapiens (human)
positive regulation of epithelial cell migrationTGF-beta receptor type-2Homo sapiens (human)
positive regulation of epithelial to mesenchymal transitionTGF-beta receptor type-2Homo sapiens (human)
activation of protein kinase activityTGF-beta receptor type-2Homo sapiens (human)
activin receptor signaling pathwayTGF-beta receptor type-2Homo sapiens (human)
embryonic hemopoiesisTGF-beta receptor type-2Homo sapiens (human)
aorta morphogenesisTGF-beta receptor type-2Homo sapiens (human)
regulation of cell population proliferationTGF-beta receptor type-2Homo sapiens (human)
myeloid dendritic cell differentiationTGF-beta receptor type-2Homo sapiens (human)
positive regulation of angiogenesisTGF-beta receptor type-2Homo sapiens (human)
embryonic cranial skeleton morphogenesisTGF-beta receptor type-2Homo sapiens (human)
artery morphogenesisTGF-beta receptor type-2Homo sapiens (human)
positive regulation of NK T cell differentiationTGF-beta receptor type-2Homo sapiens (human)
roof of mouth developmentTGF-beta receptor type-2Homo sapiens (human)
positive regulation of SMAD protein signal transductionTGF-beta receptor type-2Homo sapiens (human)
SMAD protein signal transductionTGF-beta receptor type-2Homo sapiens (human)
ventricular septum morphogenesisTGF-beta receptor type-2Homo sapiens (human)
bronchus morphogenesisTGF-beta receptor type-2Homo sapiens (human)
trachea formationTGF-beta receptor type-2Homo sapiens (human)
mammary gland morphogenesisTGF-beta receptor type-2Homo sapiens (human)
lung lobe morphogenesisTGF-beta receptor type-2Homo sapiens (human)
Langerhans cell differentiationTGF-beta receptor type-2Homo sapiens (human)
secondary palate developmentTGF-beta receptor type-2Homo sapiens (human)
response to cholesterolTGF-beta receptor type-2Homo sapiens (human)
regulation of stem cell proliferationTGF-beta receptor type-2Homo sapiens (human)
positive regulation of epithelial to mesenchymal transition involved in endocardial cushion formationTGF-beta receptor type-2Homo sapiens (human)
inferior endocardial cushion morphogenesisTGF-beta receptor type-2Homo sapiens (human)
lens fiber cell apoptotic processTGF-beta receptor type-2Homo sapiens (human)
miRNA transportTGF-beta receptor type-2Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processTGF-beta receptor type-2Homo sapiens (human)
positive regulation of CD4-positive, alpha-beta T cell proliferationTGF-beta receptor type-2Homo sapiens (human)
regulation of stem cell differentiationTGF-beta receptor type-2Homo sapiens (human)
cellular response to growth factor stimulusTGF-beta receptor type-2Homo sapiens (human)
protein phosphorylationTGF-beta receptor type-2Homo sapiens (human)
amino acid catabolic processElectron transfer flavoprotein subunit betaHomo sapiens (human)
respiratory electron transport chainElectron transfer flavoprotein subunit betaHomo sapiens (human)
fatty acid beta-oxidation using acyl-CoA dehydrogenaseElectron transfer flavoprotein subunit betaHomo sapiens (human)
adaptive immune responseTyrosine-protein kinase CSKHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of cell population proliferationTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of low-density lipoprotein particle clearanceTyrosine-protein kinase CSKHomo sapiens (human)
T cell costimulationTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of interleukin-6 productionTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of Golgi to plasma membrane protein transportTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of bone resorptionTyrosine-protein kinase CSKHomo sapiens (human)
oligodendrocyte differentiationTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of phagocytosisTyrosine-protein kinase CSKHomo sapiens (human)
T cell receptor signaling pathwayTyrosine-protein kinase CSKHomo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeTyrosine-protein kinase CSKHomo sapiens (human)
cellular response to peptide hormone stimulusTyrosine-protein kinase CSKHomo sapiens (human)
regulation of Fc receptor mediated stimulatory signaling pathwayTyrosine-protein kinase CSKHomo sapiens (human)
adherens junction organizationTyrosine-protein kinase CSKHomo sapiens (human)
tRNA aminoacylation for protein translationGlycine--tRNA ligaseHomo sapiens (human)
diadenosine tetraphosphate biosynthetic processGlycine--tRNA ligaseHomo sapiens (human)
mitochondrial glycyl-tRNA aminoacylationGlycine--tRNA ligaseHomo sapiens (human)
protein phosphorylationProtein kinase C iota typeHomo sapiens (human)
protein targeting to membraneProtein kinase C iota typeHomo sapiens (human)
cytoskeleton organizationProtein kinase C iota typeHomo sapiens (human)
actin filament organizationProtein kinase C iota typeHomo sapiens (human)
positive regulation of neuron projection developmentProtein kinase C iota typeHomo sapiens (human)
vesicle-mediated transportProtein kinase C iota typeHomo sapiens (human)
cell migrationProtein kinase C iota typeHomo sapiens (human)
cellular response to insulin stimulusProtein kinase C iota typeHomo sapiens (human)
negative regulation of glial cell apoptotic processProtein kinase C iota typeHomo sapiens (human)
establishment of apical/basal cell polarityProtein kinase C iota typeHomo sapiens (human)
eye photoreceptor cell developmentProtein kinase C iota typeHomo sapiens (human)
negative regulation of apoptotic processProtein kinase C iota typeHomo sapiens (human)
negative regulation of neuron apoptotic processProtein kinase C iota typeHomo sapiens (human)
establishment or maintenance of epithelial cell apical/basal polarityProtein kinase C iota typeHomo sapiens (human)
cell-cell junction organizationProtein kinase C iota typeHomo sapiens (human)
positive regulation of Notch signaling pathwayProtein kinase C iota typeHomo sapiens (human)
positive regulation of glucose importProtein kinase C iota typeHomo sapiens (human)
secretionProtein kinase C iota typeHomo sapiens (human)
Golgi vesicle buddingProtein kinase C iota typeHomo sapiens (human)
positive regulation of NF-kappaB transcription factor activityProtein kinase C iota typeHomo sapiens (human)
positive regulation of glial cell proliferationProtein kinase C iota typeHomo sapiens (human)
membrane organizationProtein kinase C iota typeHomo sapiens (human)
cellular response to chemical stressProtein kinase C iota typeHomo sapiens (human)
response to interleukin-1Protein kinase C iota typeHomo sapiens (human)
regulation of postsynaptic membrane neurotransmitter receptor levelsProtein kinase C iota typeHomo sapiens (human)
positive regulation of protein localization to plasma membraneProtein kinase C iota typeHomo sapiens (human)
positive regulation of endothelial cell apoptotic processProtein kinase C iota typeHomo sapiens (human)
intracellular signal transductionProtein kinase C iota typeHomo sapiens (human)
peptidyl-serine phosphorylationProtein kinase C iota typeHomo sapiens (human)
mRNA splicing, via spliceosomeExosome RNA helicase MTR4Homo sapiens (human)
maturation of 5.8S rRNAExosome RNA helicase MTR4Homo sapiens (human)
rRNA processingExosome RNA helicase MTR4Homo sapiens (human)
RNA catabolic processExosome RNA helicase MTR4Homo sapiens (human)
DNA damage responseExosome RNA helicase MTR4Homo sapiens (human)
snRNA catabolic processExosome RNA helicase MTR4Homo sapiens (human)
protein destabilizationSerine/threonine-protein kinase mTORHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of macroautophagySerine/threonine-protein kinase mTORHomo sapiens (human)
phosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of cell growthSerine/threonine-protein kinase mTORHomo sapiens (human)
T-helper 1 cell lineage commitmentSerine/threonine-protein kinase mTORHomo sapiens (human)
heart morphogenesisSerine/threonine-protein kinase mTORHomo sapiens (human)
heart valve morphogenesisSerine/threonine-protein kinase mTORHomo sapiens (human)
energy reserve metabolic processSerine/threonine-protein kinase mTORHomo sapiens (human)
'de novo' pyrimidine nucleobase biosynthetic processSerine/threonine-protein kinase mTORHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
inflammatory responseSerine/threonine-protein kinase mTORHomo sapiens (human)
DNA damage responseSerine/threonine-protein kinase mTORHomo sapiens (human)
cytoskeleton organizationSerine/threonine-protein kinase mTORHomo sapiens (human)
lysosome organizationSerine/threonine-protein kinase mTORHomo sapiens (human)
germ cell developmentSerine/threonine-protein kinase mTORHomo sapiens (human)
response to nutrientSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of cell sizeSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to starvationSerine/threonine-protein kinase mTORHomo sapiens (human)
response to heatSerine/threonine-protein kinase mTORHomo sapiens (human)
post-embryonic developmentSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of autophagySerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of lamellipodium assemblySerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of gene expressionSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of epithelial to mesenchymal transitionSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of myotube differentiationSerine/threonine-protein kinase mTORHomo sapiens (human)
macroautophagySerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of macroautophagySerine/threonine-protein kinase mTORHomo sapiens (human)
phosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
neuronal action potentialSerine/threonine-protein kinase mTORHomo sapiens (human)
protein catabolic processSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of cell growthSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of actin filament polymerizationSerine/threonine-protein kinase mTORHomo sapiens (human)
T cell costimulationSerine/threonine-protein kinase mTORHomo sapiens (human)
ruffle organizationSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of myelinationSerine/threonine-protein kinase mTORHomo sapiens (human)
response to nutrient levelsSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to nutrient levelsSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to nutrientSerine/threonine-protein kinase mTORHomo sapiens (human)
TOR signalingSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of phosphoprotein phosphatase activitySerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to insulin stimulusSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of actin cytoskeleton organizationSerine/threonine-protein kinase mTORHomo sapiens (human)
calcineurin-NFAT signaling cascadeSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to amino acid starvationSerine/threonine-protein kinase mTORHomo sapiens (human)
multicellular organism growthSerine/threonine-protein kinase mTORHomo sapiens (human)
TORC1 signalingSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of circadian rhythmSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of apoptotic processSerine/threonine-protein kinase mTORHomo sapiens (human)
response to amino acidSerine/threonine-protein kinase mTORHomo sapiens (human)
anoikisSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of osteoclast differentiationSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of translationSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of cell sizeSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of glycolytic processSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIISerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of translational initiationSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of lipid biosynthetic processSerine/threonine-protein kinase mTORHomo sapiens (human)
behavioral response to painSerine/threonine-protein kinase mTORHomo sapiens (human)
rhythmic processSerine/threonine-protein kinase mTORHomo sapiens (human)
oligodendrocyte differentiationSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of oligodendrocyte differentiationSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationSerine/threonine-protein kinase mTORHomo sapiens (human)
voluntary musculoskeletal movementSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of stress fiber assemblySerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of keratinocyte migrationSerine/threonine-protein kinase mTORHomo sapiens (human)
nucleus localizationSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionSerine/threonine-protein kinase mTORHomo sapiens (human)
cardiac muscle cell developmentSerine/threonine-protein kinase mTORHomo sapiens (human)
cardiac muscle contractionSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to methionineSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of calcineurin-NFAT signaling cascadeSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to amino acid stimulusSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to L-leucineSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to hypoxiaSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to osmotic stressSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of membrane permeabilitySerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of cellular response to heatSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of protein localization to nucleusSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of signal transduction by p53 class mediatorSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of transcription of nucleolar large rRNA by RNA polymerase ISerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of wound healing, spreading of epidermal cellsSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of locomotor rhythmSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of cytoplasmic translational initiationSerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of lysosome organizationSerine/threonine-protein kinase mTORHomo sapiens (human)
positive regulation of pentose-phosphate shuntSerine/threonine-protein kinase mTORHomo sapiens (human)
cellular response to leucine starvationSerine/threonine-protein kinase mTORHomo sapiens (human)
regulation of autophagosome assemblySerine/threonine-protein kinase mTORHomo sapiens (human)
negative regulation of macroautophagySerine/threonine-protein kinase mTORHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase TecHomo sapiens (human)
integrin-mediated signaling pathwayTyrosine-protein kinase TecHomo sapiens (human)
regulation of platelet activationTyrosine-protein kinase TecHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase TecHomo sapiens (human)
tissue regenerationTyrosine-protein kinase TecHomo sapiens (human)
B cell receptor signaling pathwayTyrosine-protein kinase TecHomo sapiens (human)
adaptive immune responseTyrosine-protein kinase TecHomo sapiens (human)
T cell receptor signaling pathwayTyrosine-protein kinase TecHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of phospholipase C activityTyrosine-protein kinase ABL2Homo sapiens (human)
negative regulation of Rho protein signal transductionTyrosine-protein kinase ABL2Homo sapiens (human)
exploration behaviorTyrosine-protein kinase ABL2Homo sapiens (human)
cell adhesionTyrosine-protein kinase ABL2Homo sapiens (human)
signal transductionTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of autophagyTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of neuron projection developmentTyrosine-protein kinase ABL2Homo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of endocytosisTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of cell adhesionTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of actin cytoskeleton organizationTyrosine-protein kinase ABL2Homo sapiens (human)
protein modification processTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of oxidoreductase activityTyrosine-protein kinase ABL2Homo sapiens (human)
cellular response to retinoic acidTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of establishment of T cell polarityTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of cell motilityTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of T cell migrationTyrosine-protein kinase ABL2Homo sapiens (human)
epidermal growth factor receptor signaling pathwayTyrosine-protein kinase ABL2Homo sapiens (human)
protein phosphorylationTyrosine-protein kinase ABL2Homo sapiens (human)
negative regulation of transcription by RNA polymerase IITyrosine-protein kinase FRKHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase FRKHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase FRKHomo sapiens (human)
cell differentiationTyrosine-protein kinase FRKHomo sapiens (human)
innate immune responseTyrosine-protein kinase FRKHomo sapiens (human)
G protein-coupled receptor signaling pathwayG protein-coupled receptor kinase 6Homo sapiens (human)
regulation of G protein-coupled receptor signaling pathwayG protein-coupled receptor kinase 6Homo sapiens (human)
Wnt signaling pathwayG protein-coupled receptor kinase 6Homo sapiens (human)
regulation of signal transductionG protein-coupled receptor kinase 6Homo sapiens (human)
protein phosphorylationG protein-coupled receptor kinase 6Homo sapiens (human)
protein import into nucleusTyrosine-protein kinase SYKHomo sapiens (human)
regulation of DNA-binding transcription factor activityTyrosine-protein kinase SYKHomo sapiens (human)
angiogenesisTyrosine-protein kinase SYKHomo sapiens (human)
cell activationTyrosine-protein kinase SYKHomo sapiens (human)
lymph vessel developmentTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of receptor internalizationTyrosine-protein kinase SYKHomo sapiens (human)
stimulatory C-type lectin receptor signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
adaptive immune responseTyrosine-protein kinase SYKHomo sapiens (human)
macrophage activation involved in immune responseTyrosine-protein kinase SYKHomo sapiens (human)
neutrophil activation involved in immune responseTyrosine-protein kinase SYKHomo sapiens (human)
leukocyte activation involved in immune responseTyrosine-protein kinase SYKHomo sapiens (human)
serotonin secretion by plateletTyrosine-protein kinase SYKHomo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusTyrosine-protein kinase SYKHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase SYKHomo sapiens (human)
leukocyte cell-cell adhesionTyrosine-protein kinase SYKHomo sapiens (human)
integrin-mediated signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
animal organ morphogenesisTyrosine-protein kinase SYKHomo sapiens (human)
regulation of platelet activationTyrosine-protein kinase SYKHomo sapiens (human)
regulation of tumor necrosis factor-mediated signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase SYKHomo sapiens (human)
leukotriene biosynthetic processTyrosine-protein kinase SYKHomo sapiens (human)
calcium-mediated signalingTyrosine-protein kinase SYKHomo sapiens (human)
platelet activationTyrosine-protein kinase SYKHomo sapiens (human)
B cell differentiationTyrosine-protein kinase SYKHomo sapiens (human)
neutrophil chemotaxisTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of protein-containing complex assemblyTyrosine-protein kinase SYKHomo sapiens (human)
receptor internalizationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of type I interferon productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of granulocyte macrophage colony-stimulating factor productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of interleukin-10 productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of interleukin-12 productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of interleukin-3 productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of interleukin-4 productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of interleukin-6 productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of interleukin-8 productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of tumor necrosis factor productionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of mast cell cytokine productionTyrosine-protein kinase SYKHomo sapiens (human)
regulation of superoxide anion generationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of superoxide anion generationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of cell adhesion mediated by integrinTyrosine-protein kinase SYKHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase SYKHomo sapiens (human)
collagen-activated tyrosine kinase receptor signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
Fc-epsilon receptor signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisTyrosine-protein kinase SYKHomo sapiens (human)
interleukin-3-mediated signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
gamma-delta T cell differentiationTyrosine-protein kinase SYKHomo sapiens (human)
defense response to bacteriumTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of cysteine-type endopeptidase activity involved in apoptotic processTyrosine-protein kinase SYKHomo sapiens (human)
mast cell degranulationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of mast cell degranulationTyrosine-protein kinase SYKHomo sapiens (human)
regulation of neutrophil degranulationTyrosine-protein kinase SYKHomo sapiens (human)
beta selectionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of MAPK cascadeTyrosine-protein kinase SYKHomo sapiens (human)
innate immune responseTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of B cell differentiationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of gamma-delta T cell differentiationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of bone resorptionTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of alpha-beta T cell differentiationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of alpha-beta T cell proliferationTyrosine-protein kinase SYKHomo sapiens (human)
blood vessel morphogenesisTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationTyrosine-protein kinase SYKHomo sapiens (human)
regulation of phagocytosisTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of calcium-mediated signalingTyrosine-protein kinase SYKHomo sapiens (human)
B cell receptor signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of killing of cells of another organismTyrosine-protein kinase SYKHomo sapiens (human)
regulation of ERK1 and ERK2 cascadeTyrosine-protein kinase SYKHomo sapiens (human)
cellular response to molecule of fungal originTyrosine-protein kinase SYKHomo sapiens (human)
cellular response to lipidTyrosine-protein kinase SYKHomo sapiens (human)
cellular response to low-density lipoprotein particle stimulusTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of monocyte chemotactic protein-1 productionTyrosine-protein kinase SYKHomo sapiens (human)
regulation of arachidonic acid secretionTyrosine-protein kinase SYKHomo sapiens (human)
regulation of platelet aggregationTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of cold-induced thermogenesisTyrosine-protein kinase SYKHomo sapiens (human)
positive regulation of TORC1 signalingTyrosine-protein kinase SYKHomo sapiens (human)
cellular response to lectinTyrosine-protein kinase SYKHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayTyrosine-protein kinase SYKHomo sapiens (human)
cell differentiationTyrosine-protein kinase SYKHomo sapiens (human)
blastocyst development26S proteasome regulatory subunit 6BHomo sapiens (human)
proteolysis26S proteasome regulatory subunit 6BHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic process26S proteasome regulatory subunit 6BHomo sapiens (human)
positive regulation of proteasomal protein catabolic process26S proteasome regulatory subunit 6BHomo sapiens (human)
JUN phosphorylationMitogen-activated protein kinase 8Homo sapiens (human)
response to UVMitogen-activated protein kinase 8Homo sapiens (human)
negative regulation of apoptotic processMitogen-activated protein kinase 8Homo sapiens (human)
cellular response to lipopolysaccharideMitogen-activated protein kinase 8Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase 8Homo sapiens (human)
response to oxidative stressMitogen-activated protein kinase 8Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase 8Homo sapiens (human)
JUN phosphorylationMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of gene expressionMitogen-activated protein kinase 8Homo sapiens (human)
regulation of macroautophagyMitogen-activated protein kinase 8Homo sapiens (human)
peptidyl-serine phosphorylationMitogen-activated protein kinase 8Homo sapiens (human)
peptidyl-threonine phosphorylationMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of cyclase activityMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of cell killingMitogen-activated protein kinase 8Homo sapiens (human)
negative regulation of protein bindingMitogen-activated protein kinase 8Homo sapiens (human)
regulation of protein localizationMitogen-activated protein kinase 8Homo sapiens (human)
cellular response to amino acid starvationMitogen-activated protein kinase 8Homo sapiens (human)
cellular response to oxidative stressMitogen-activated protein kinase 8Homo sapiens (human)
cellular response to reactive oxygen speciesMitogen-activated protein kinase 8Homo sapiens (human)
Fc-epsilon receptor signaling pathwayMitogen-activated protein kinase 8Homo sapiens (human)
regulation of circadian rhythmMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of apoptotic processMitogen-activated protein kinase 8Homo sapiens (human)
negative regulation of apoptotic processMitogen-activated protein kinase 8Homo sapiens (human)
rhythmic processMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of protein metabolic processMitogen-activated protein kinase 8Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase 8Homo sapiens (human)
cellular response to mechanical stimulusMitogen-activated protein kinase 8Homo sapiens (human)
cellular response to cadmium ionMitogen-activated protein kinase 8Homo sapiens (human)
cellular senescenceMitogen-activated protein kinase 8Homo sapiens (human)
energy homeostasisMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of NLRP3 inflammasome complex assemblyMitogen-activated protein kinase 8Homo sapiens (human)
response to mechanical stimulusMitogen-activated protein kinase 8Homo sapiens (human)
positive regulation of establishment of protein localization to mitochondrionMitogen-activated protein kinase 8Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase 9Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of gene expressionMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of macrophage derived foam cell differentiationMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of protein ubiquitinationMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processMitogen-activated protein kinase 9Homo sapiens (human)
cellular response to reactive oxygen speciesMitogen-activated protein kinase 9Homo sapiens (human)
Fc-epsilon receptor signaling pathwayMitogen-activated protein kinase 9Homo sapiens (human)
regulation of circadian rhythmMitogen-activated protein kinase 9Homo sapiens (human)
rhythmic processMitogen-activated protein kinase 9Homo sapiens (human)
modulation of chemical synaptic transmissionMitogen-activated protein kinase 9Homo sapiens (human)
protein localization to tricellular tight junctionMitogen-activated protein kinase 9Homo sapiens (human)
cellular response to cadmium ionMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of podosome assemblyMitogen-activated protein kinase 9Homo sapiens (human)
cellular senescenceMitogen-activated protein kinase 9Homo sapiens (human)
inflammatory response to woundingMitogen-activated protein kinase 9Homo sapiens (human)
apoptotic signaling pathwayMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of cytokine production involved in inflammatory responseMitogen-activated protein kinase 9Homo sapiens (human)
positive regulation of apoptotic signaling pathwayMitogen-activated protein kinase 9Homo sapiens (human)
regulation of cytokine productionDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
response to ischemiaDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
inflammatory responseDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
signal transductionDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
heart developmentDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
stress-activated protein kinase signaling cascadeDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
negative regulation of hippo signalingDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
cellular response to vascular endothelial growth factor stimulusDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
p38MAPK cascadeDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
positive regulation of MAPK cascadeDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
positive regulation of blood vessel endothelial cell migrationDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
positive regulation of protein kinase activityDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
positive regulation of DNA-templated transcriptionDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
positive regulation of nitric-oxide synthase biosynthetic processDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
cardiac muscle contractionDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
cellular response to lipopolysaccharideDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
cellular response to sorbitolDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
cellular senescenceDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
MAPK cascadeDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
regulation of autophagyPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
megakaryocyte developmentPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
autophagosome-lysosome fusionPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
vesicle-mediated cholesterol transportPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate biosynthetic processPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
positive regulation of autophagosome assemblyPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
phosphatidylinositol phosphate biosynthetic processPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
protein phosphorylationCasein kinase I isoform alphaHomo sapiens (human)
Golgi organizationCasein kinase I isoform alphaHomo sapiens (human)
cell surface receptor signaling pathwayCasein kinase I isoform alphaHomo sapiens (human)
Wnt signaling pathwayCasein kinase I isoform alphaHomo sapiens (human)
peptidyl-serine phosphorylationCasein kinase I isoform alphaHomo sapiens (human)
viral protein processingCasein kinase I isoform alphaHomo sapiens (human)
cellular response to nutrientCasein kinase I isoform alphaHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processCasein kinase I isoform alphaHomo sapiens (human)
positive regulation of Rho protein signal transductionCasein kinase I isoform alphaHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processCasein kinase I isoform alphaHomo sapiens (human)
intermediate filament cytoskeleton organizationCasein kinase I isoform alphaHomo sapiens (human)
cell divisionCasein kinase I isoform alphaHomo sapiens (human)
negative regulation of canonical Wnt signaling pathwayCasein kinase I isoform alphaHomo sapiens (human)
negative regulation of NLRP3 inflammasome complex assemblyCasein kinase I isoform alphaHomo sapiens (human)
positive regulation of TORC1 signalingCasein kinase I isoform alphaHomo sapiens (human)
signal transductionCasein kinase I isoform alphaHomo sapiens (human)
microtubule nucleationCasein kinase I isoform deltaHomo sapiens (human)
Golgi organizationCasein kinase I isoform deltaHomo sapiens (human)
protein localization to Golgi apparatusCasein kinase I isoform deltaHomo sapiens (human)
protein localization to ciliumCasein kinase I isoform deltaHomo sapiens (human)
protein localization to centrosomeCasein kinase I isoform deltaHomo sapiens (human)
non-motile cilium assemblyCasein kinase I isoform deltaHomo sapiens (human)
positive regulation of protein phosphorylationCasein kinase I isoform deltaHomo sapiens (human)
protein phosphorylationCasein kinase I isoform deltaHomo sapiens (human)
Wnt signaling pathwayCasein kinase I isoform deltaHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processCasein kinase I isoform deltaHomo sapiens (human)
circadian regulation of gene expressionCasein kinase I isoform deltaHomo sapiens (human)
regulation of circadian rhythmCasein kinase I isoform deltaHomo sapiens (human)
COPII vesicle coatingCasein kinase I isoform deltaHomo sapiens (human)
spindle assemblyCasein kinase I isoform deltaHomo sapiens (human)
positive regulation of canonical Wnt signaling pathwayCasein kinase I isoform deltaHomo sapiens (human)
midbrain dopaminergic neuron differentiationCasein kinase I isoform deltaHomo sapiens (human)
cellular response to nerve growth factor stimulusCasein kinase I isoform deltaHomo sapiens (human)
positive regulation of non-canonical Wnt signaling pathwayCasein kinase I isoform deltaHomo sapiens (human)
peptidyl-serine phosphorylationCasein kinase I isoform deltaHomo sapiens (human)
signal transductionCasein kinase I isoform deltaHomo sapiens (human)
non-motile cilium assemblyCasein kinase I isoform deltaHomo sapiens (human)
endocytosisCasein kinase I isoform deltaHomo sapiens (human)
MAPK cascadeMAP kinase-activated protein kinase 2Homo sapiens (human)
toll-like receptor signaling pathwayMAP kinase-activated protein kinase 2Homo sapiens (human)
protein phosphorylationMAP kinase-activated protein kinase 2Homo sapiens (human)
leukotriene metabolic processMAP kinase-activated protein kinase 2Homo sapiens (human)
inflammatory responseMAP kinase-activated protein kinase 2Homo sapiens (human)
DNA damage responseMAP kinase-activated protein kinase 2Homo sapiens (human)
regulation of tumor necrosis factor-mediated signaling pathwayMAP kinase-activated protein kinase 2Homo sapiens (human)
peptidyl-serine phosphorylationMAP kinase-activated protein kinase 2Homo sapiens (human)
response to lipopolysaccharideMAP kinase-activated protein kinase 2Homo sapiens (human)
regulation of interleukin-6 productionMAP kinase-activated protein kinase 2Homo sapiens (human)
regulation of tumor necrosis factor productionMAP kinase-activated protein kinase 2Homo sapiens (human)
positive regulation of tumor necrosis factor productionMAP kinase-activated protein kinase 2Homo sapiens (human)
response to cytokineMAP kinase-activated protein kinase 2Homo sapiens (human)
cellular response to vascular endothelial growth factor stimulusMAP kinase-activated protein kinase 2Homo sapiens (human)
p38MAPK cascadeMAP kinase-activated protein kinase 2Homo sapiens (human)
regulation of mRNA stabilityMAP kinase-activated protein kinase 2Homo sapiens (human)
macropinocytosisMAP kinase-activated protein kinase 2Homo sapiens (human)
vascular endothelial growth factor receptor signaling pathwayMAP kinase-activated protein kinase 2Homo sapiens (human)
inner ear developmentMAP kinase-activated protein kinase 2Homo sapiens (human)
positive regulation of macrophage cytokine productionMAP kinase-activated protein kinase 2Homo sapiens (human)
3'-UTR-mediated mRNA stabilizationMAP kinase-activated protein kinase 2Homo sapiens (human)
regulation of cellular response to heatMAP kinase-activated protein kinase 2Homo sapiens (human)
protein autophosphorylationMAP kinase-activated protein kinase 2Homo sapiens (human)
intracellular signal transductionMAP kinase-activated protein kinase 2Homo sapiens (human)
translational elongationElongation factor Tu, mitochondrialHomo sapiens (human)
response to ethanolElongation factor Tu, mitochondrialHomo sapiens (human)
mitochondrial translational elongationElongation factor Tu, mitochondrialHomo sapiens (human)
phosphatidylcholine biosynthetic processCholine-phosphate cytidylyltransferase AHomo sapiens (human)
CDP-choline pathwayCholine-phosphate cytidylyltransferase AHomo sapiens (human)
cysteinyl-tRNA aminoacylationCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
DNA repairCasein kinase I isoform epsilonHomo sapiens (human)
protein phosphorylationCasein kinase I isoform epsilonHomo sapiens (human)
protein localizationCasein kinase I isoform epsilonHomo sapiens (human)
negative regulation of Wnt signaling pathwayCasein kinase I isoform epsilonHomo sapiens (human)
negative regulation of protein bindingCasein kinase I isoform epsilonHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processCasein kinase I isoform epsilonHomo sapiens (human)
regulation of protein localizationCasein kinase I isoform epsilonHomo sapiens (human)
circadian regulation of gene expressionCasein kinase I isoform epsilonHomo sapiens (human)
regulation of circadian rhythmCasein kinase I isoform epsilonHomo sapiens (human)
circadian behaviorCasein kinase I isoform epsilonHomo sapiens (human)
canonical Wnt signaling pathwayCasein kinase I isoform epsilonHomo sapiens (human)
positive regulation of amyloid-beta formationCasein kinase I isoform epsilonHomo sapiens (human)
cellular response to nerve growth factor stimulusCasein kinase I isoform epsilonHomo sapiens (human)
positive regulation of non-canonical Wnt signaling pathwayCasein kinase I isoform epsilonHomo sapiens (human)
peptidyl-serine phosphorylationCasein kinase I isoform epsilonHomo sapiens (human)
endocytosisCasein kinase I isoform epsilonHomo sapiens (human)
positive regulation of canonical Wnt signaling pathwayCasein kinase I isoform epsilonHomo sapiens (human)
signal transductionCasein kinase I isoform epsilonHomo sapiens (human)
temperature homeostasisVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
response to coldVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
energy derivation by oxidation of organic compoundsVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
epithelial cell differentiationVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
fatty acid beta-oxidation using acyl-CoA dehydrogenaseVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
negative regulation of fatty acid biosynthetic processVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
negative regulation of fatty acid oxidationVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
regulation of cholesterol metabolic processVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
regulation of RNA splicingDual specificity protein kinase CLK1Homo sapiens (human)
peptidyl-tyrosine phosphorylationDual specificity protein kinase CLK1Homo sapiens (human)
protein phosphorylationDual specificity protein kinase CLK2Homo sapiens (human)
response to ionizing radiationDual specificity protein kinase CLK2Homo sapiens (human)
regulation of RNA splicingDual specificity protein kinase CLK2Homo sapiens (human)
negative regulation of gluconeogenesisDual specificity protein kinase CLK2Homo sapiens (human)
protein autophosphorylationDual specificity protein kinase CLK2Homo sapiens (human)
peptidyl-tyrosine phosphorylationDual specificity protein kinase CLK2Homo sapiens (human)
protein phosphorylationDual specificity protein kinase CLK3Homo sapiens (human)
regulation of RNA splicingDual specificity protein kinase CLK3Homo sapiens (human)
regulation of systemic arterial blood pressureGlycogen synthase kinase-3 alphaHomo sapiens (human)
cardiac left ventricle morphogenesisGlycogen synthase kinase-3 alphaHomo sapiens (human)
glycogen metabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
dopamine receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
nervous system developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
insulin receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of autophagyGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of gene expressionGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of UDP-glucose catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
Wnt signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
cell migrationGlycogen synthase kinase-3 alphaHomo sapiens (human)
peptidyl-threonine phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
viral protein processingGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of protein ubiquitinationGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of TOR signalingGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to insulin stimulusGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to interleukin-3Glycogen synthase kinase-3 alphaHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glycogen biosynthetic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of protein catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of heart contractionGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glucose importGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
excitatory postsynaptic potentialGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of cell growth involved in cardiac muscle cell developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to lithium ionGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to glucocorticoid stimulusGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of adenylate cyclase-activating adrenergic receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of canonical Wnt signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
extrinsic apoptotic signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
extrinsic apoptotic signaling pathway in absence of ligandGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of adenylate cyclase-activating G protein-coupled receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
autosome genomic imprintingGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of mitochondrial outer membrane permeabilization involved in apoptotic signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
regulation of mitophagyGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of amyloid-beta formationGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of protein targeting to mitochondrionGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glycogen synthase activity, transferring glucose-1-phosphateGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of type B pancreatic cell developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glycogen (starch) synthase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of glycogen (starch) synthase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
cell differentiationGlycogen synthase kinase-3 alphaHomo sapiens (human)
regulation of microtubule cytoskeleton organizationGlycogen synthase kinase-3 alphaHomo sapiens (human)
regulation of neuron projection developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of gene expressionGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of gene expressionGlycogen synthase kinase-3 betaHomo sapiens (human)
ER overload responseGlycogen synthase kinase-3 betaHomo sapiens (human)
peptidyl-serine phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
intracellular signal transductionGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of apoptotic processGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein export from nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
epithelial to mesenchymal transitionGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of cell-matrix adhesionGlycogen synthase kinase-3 betaHomo sapiens (human)
glycogen metabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
protein phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
mitochondrion organizationGlycogen synthase kinase-3 betaHomo sapiens (human)
dopamine receptor signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
circadian rhythmGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of autophagyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of gene expressionGlycogen synthase kinase-3 betaHomo sapiens (human)
peptidyl-serine phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
peptidyl-threonine phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
viral protein processingGlycogen synthase kinase-3 betaHomo sapiens (human)
hippocampus developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
establishment of cell polarityGlycogen synthase kinase-3 betaHomo sapiens (human)
maintenance of cell polarityGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of cell migrationGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of axon extensionGlycogen synthase kinase-3 betaHomo sapiens (human)
neuron projection developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of protein-containing complex assemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein-containing complex assemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein ubiquitinationGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of phosphoprotein phosphatase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of microtubule-based processGlycogen synthase kinase-3 betaHomo sapiens (human)
intracellular signal transductionGlycogen synthase kinase-3 betaHomo sapiens (human)
cellular response to interleukin-3Glycogen synthase kinase-3 betaHomo sapiens (human)
regulation of circadian rhythmGlycogen synthase kinase-3 betaHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of GTPase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of cell differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of osteoblast differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of glycogen biosynthetic processGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of cilium assemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein catabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
protein autophosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of protein export from nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of dendrite morphogenesisGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of axonogenesisGlycogen synthase kinase-3 betaHomo sapiens (human)
canonical Wnt signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
excitatory postsynaptic potentialGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of microtubule cytoskeleton organizationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of calcineurin-NFAT signaling cascadeGlycogen synthase kinase-3 betaHomo sapiens (human)
superior temporal gyrus developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
cellular response to retinoic acidGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of canonical Wnt signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
extrinsic apoptotic signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
extrinsic apoptotic signaling pathway in absence of ligandGlycogen synthase kinase-3 betaHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionGlycogen synthase kinase-3 betaHomo sapiens (human)
neuron projection organizationGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of microtubule anchoring at centrosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of cellular response to heatGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of protein localization to nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of long-term synaptic potentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of mitochondrial outer membrane permeabilization involved in apoptotic signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of protein acetylationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathway via death domain receptorsGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein localization to ciliumGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of dopaminergic neuron differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
cellular response to amyloid-betaGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein localization to centrosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
beta-catenin destruction complex disassemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of type B pancreatic cell developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of glycogen (starch) synthase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of mesenchymal stem cell differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of TOR signalingGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of neuron projection developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
cell differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
insulin receptor signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
DNA repairCyclin-dependent kinase 7Homo sapiens (human)
transcription by RNA polymerase IICyclin-dependent kinase 7Homo sapiens (human)
transcription initiation at RNA polymerase II promoterCyclin-dependent kinase 7Homo sapiens (human)
snRNA transcription by RNA polymerase IICyclin-dependent kinase 7Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICyclin-dependent kinase 7Homo sapiens (human)
protein stabilizationCyclin-dependent kinase 7Homo sapiens (human)
cell divisionCyclin-dependent kinase 7Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 7Homo sapiens (human)
regulation of G1/S transition of mitotic cell cycleCyclin-dependent kinase 7Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 7Homo sapiens (human)
regulation of mitotic cell cycleCyclin-dependent kinase 9Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 9Homo sapiens (human)
DNA repairCyclin-dependent kinase 9Homo sapiens (human)
regulation of DNA repairCyclin-dependent kinase 9Homo sapiens (human)
transcription by RNA polymerase IICyclin-dependent kinase 9Homo sapiens (human)
transcription initiation at RNA polymerase II promoterCyclin-dependent kinase 9Homo sapiens (human)
transcription elongation by RNA polymerase IICyclin-dependent kinase 9Homo sapiens (human)
cell population proliferationCyclin-dependent kinase 9Homo sapiens (human)
replication fork processingCyclin-dependent kinase 9Homo sapiens (human)
regulation of mRNA 3'-end processingCyclin-dependent kinase 9Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IICyclin-dependent kinase 9Homo sapiens (human)
positive regulation by host of viral transcriptionCyclin-dependent kinase 9Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICyclin-dependent kinase 9Homo sapiens (human)
regulation of muscle cell differentiationCyclin-dependent kinase 9Homo sapiens (human)
nucleus localizationCyclin-dependent kinase 9Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 9Homo sapiens (human)
cellular response to cytokine stimulusCyclin-dependent kinase 9Homo sapiens (human)
negative regulation of protein localization to chromatinCyclin-dependent kinase 9Homo sapiens (human)
positive regulation of protein localization to chromatinCyclin-dependent kinase 9Homo sapiens (human)
transcription elongation-coupled chromatin remodelingCyclin-dependent kinase 9Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 9Homo sapiens (human)
exocytosisRas-related protein Rab-27AHomo sapiens (human)
blood coagulationRas-related protein Rab-27AHomo sapiens (human)
protein secretionRas-related protein Rab-27AHomo sapiens (human)
positive regulation of gene expressionRas-related protein Rab-27AHomo sapiens (human)
antigen processing and presentationRas-related protein Rab-27AHomo sapiens (human)
melanocyte differentiationRas-related protein Rab-27AHomo sapiens (human)
melanosome localizationRas-related protein Rab-27AHomo sapiens (human)
melanosome transportRas-related protein Rab-27AHomo sapiens (human)
multivesicular body organizationRas-related protein Rab-27AHomo sapiens (human)
cytotoxic T cell degranulationRas-related protein Rab-27AHomo sapiens (human)
natural killer cell degranulationRas-related protein Rab-27AHomo sapiens (human)
positive regulation of exocytosisRas-related protein Rab-27AHomo sapiens (human)
synaptic vesicle transportRas-related protein Rab-27AHomo sapiens (human)
positive regulation of phagocytosisRas-related protein Rab-27AHomo sapiens (human)
multivesicular body sorting pathwayRas-related protein Rab-27AHomo sapiens (human)
complement-dependent cytotoxicityRas-related protein Rab-27AHomo sapiens (human)
positive regulation of regulated secretory pathwayRas-related protein Rab-27AHomo sapiens (human)
positive regulation of reactive oxygen species biosynthetic processRas-related protein Rab-27AHomo sapiens (human)
positive regulation of constitutive secretory pathwayRas-related protein Rab-27AHomo sapiens (human)
exosomal secretionRas-related protein Rab-27AHomo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein autophosphorylationInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
regulation of cytokine-mediated signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
toll-like receptor signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
MyD88-dependent toll-like receptor signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
canonical NF-kappaB signal transductionInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
JNK cascadeInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
lipopolysaccharide-mediated signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
negative regulation of NF-kappaB transcription factor activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
positive regulation of type I interferon productionInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
response to lipopolysaccharideInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
toll-like receptor 2 signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
toll-like receptor 9 signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
cellular response to heatInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
interleukin-33-mediated signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein autophosphorylationInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
positive regulation of smooth muscle cell proliferationInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
type I interferon-mediated signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
interleukin-1-mediated signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
response to interleukin-1Interleukin-1 receptor-associated kinase 1Homo sapiens (human)
cellular response to hypoxiaInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
positive regulation of leukocyte adhesion to vascular endothelial cellInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
toll-like receptor 4 signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
positive regulation of MAP kinase activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
cellular response to lipopolysaccharideInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
intracellular signal transductionInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
Toll signaling pathwayInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
innate immune responseInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
gastrin-induced gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
glucose metabolic processPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
heart developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
rhythmic behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of blood pressurePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of heart ratePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
iodide transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
erythrocyte differentiationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intracellular chloride ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
response to insulinPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
social behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
corticosterone secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear morphogenesisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intestinal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
detection of mechanical stimulus involved in sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
auditory receptor cell developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
stomach developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal sodium ion absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to epinephrine stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
adrenergic receptor signaling pathwayPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cochlea developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
non-motile cilium assemblyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
skeletal system developmentRibosomal protein S6 kinase alpha-3Homo sapiens (human)
toll-like receptor signaling pathwayRibosomal protein S6 kinase alpha-3Homo sapiens (human)
signal transductionRibosomal protein S6 kinase alpha-3Homo sapiens (human)
chemical synaptic transmissionRibosomal protein S6 kinase alpha-3Homo sapiens (human)
central nervous system developmentRibosomal protein S6 kinase alpha-3Homo sapiens (human)
peptidyl-serine phosphorylationRibosomal protein S6 kinase alpha-3Homo sapiens (human)
positive regulation of cell growthRibosomal protein S6 kinase alpha-3Homo sapiens (human)
response to lipopolysaccharideRibosomal protein S6 kinase alpha-3Homo sapiens (human)
intracellular signal transductionRibosomal protein S6 kinase alpha-3Homo sapiens (human)
negative regulation of apoptotic processRibosomal protein S6 kinase alpha-3Homo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processRibosomal protein S6 kinase alpha-3Homo sapiens (human)
regulation of translation in response to stressRibosomal protein S6 kinase alpha-3Homo sapiens (human)
positive regulation of cell differentiationRibosomal protein S6 kinase alpha-3Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIRibosomal protein S6 kinase alpha-3Homo sapiens (human)
mitotic cell cycleSerine/threonine-protein kinase Nek2Homo sapiens (human)
blastocyst developmentSerine/threonine-protein kinase Nek2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase Nek2Homo sapiens (human)
chromosome segregationSerine/threonine-protein kinase Nek2Homo sapiens (human)
regulation of mitotic nuclear divisionSerine/threonine-protein kinase Nek2Homo sapiens (human)
positive regulation of telomere maintenance via telomeraseSerine/threonine-protein kinase Nek2Homo sapiens (human)
regulation of mitotic centrosome separationSerine/threonine-protein kinase Nek2Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase Nek2Homo sapiens (human)
spindle assemblySerine/threonine-protein kinase Nek2Homo sapiens (human)
centrosome separationSerine/threonine-protein kinase Nek2Homo sapiens (human)
cell divisionSerine/threonine-protein kinase Nek2Homo sapiens (human)
meiotic cell cycleSerine/threonine-protein kinase Nek2Homo sapiens (human)
positive regulation of telomerase activitySerine/threonine-protein kinase Nek2Homo sapiens (human)
regulation of attachment of spindle microtubules to kinetochoreSerine/threonine-protein kinase Nek2Homo sapiens (human)
mitotic spindle assemblySerine/threonine-protein kinase Nek2Homo sapiens (human)
negative regulation of centriole-centriole cohesionSerine/threonine-protein kinase Nek2Homo sapiens (human)
positive regulation of telomere cappingSerine/threonine-protein kinase Nek2Homo sapiens (human)
mitotic cell cycleSerine/threonine-protein kinase Nek3Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase Nek3Homo sapiens (human)
establishment of cell polaritySerine/threonine-protein kinase Nek3Homo sapiens (human)
neuron projection morphogenesisSerine/threonine-protein kinase Nek3Homo sapiens (human)
cell divisionSerine/threonine-protein kinase Nek3Homo sapiens (human)
regulation of tubulin deacetylationSerine/threonine-protein kinase Nek3Homo sapiens (human)
MAPK cascadeDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
osteoblast differentiationDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
positive regulation of protein phosphorylationDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
response to ischemiaDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
apoptotic processDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
signal transductionDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
response to xenobiotic stimulusDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
ovulation cycle processDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
stress-activated protein kinase signaling cascadeDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
positive regulation of prostaglandin secretionDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
nucleotide-binding domain, leucine rich repeat containing receptor signaling pathwayDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
p38MAPK cascadeDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
signal transduction in response to DNA damageDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
positive regulation of apoptotic processDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
positive regulation of MAPK cascadeDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
stress-activated MAPK cascadeDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
regulation of cell cycleDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
positive regulation of nitric-oxide synthase biosynthetic processDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
cardiac muscle contractionDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
bone developmentDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
cellular response to sorbitolDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
cellular senescenceDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
negative regulation of cold-induced thermogenesisDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
regulation of signal transduction by p53 class mediatorDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
establishment of protein localizationSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic sister chromatid segregationSerine/threonine-protein kinase PLK1Homo sapiens (human)
G2/M transition of mitotic cell cycleSerine/threonine-protein kinase PLK1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IISerine/threonine-protein kinase PLK1Homo sapiens (human)
establishment of mitotic spindle orientationSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic cell cycleSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic cytokinesisSerine/threonine-protein kinase PLK1Homo sapiens (human)
microtubule bundle formationSerine/threonine-protein kinase PLK1Homo sapiens (human)
double-strand break repairSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic spindle organizationSerine/threonine-protein kinase PLK1Homo sapiens (human)
sister chromatid cohesionSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic chromosome condensationSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic nuclear membrane disassemblySerine/threonine-protein kinase PLK1Homo sapiens (human)
metaphase/anaphase transition of mitotic cell cycleSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic spindle assembly checkpoint signalingSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic G2 DNA damage checkpoint signalingSerine/threonine-protein kinase PLK1Homo sapiens (human)
centrosome cycleSerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of mitotic cell cycleSerine/threonine-protein kinase PLK1Homo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationSerine/threonine-protein kinase PLK1Homo sapiens (human)
female meiosis chromosome segregationSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein ubiquitinationSerine/threonine-protein kinase PLK1Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of mitotic metaphase/anaphase transitionSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein destabilizationSerine/threonine-protein kinase PLK1Homo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processSerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of cytokinesisSerine/threonine-protein kinase PLK1Homo sapiens (human)
negative regulation of apoptotic processSerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of protein bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
homologous chromosome segregationSerine/threonine-protein kinase PLK1Homo sapiens (human)
negative regulation of cyclin-dependent protein serine/threonine kinase activitySerine/threonine-protein kinase PLK1Homo sapiens (human)
positive regulation of proteolysisSerine/threonine-protein kinase PLK1Homo sapiens (human)
Golgi inheritanceSerine/threonine-protein kinase PLK1Homo sapiens (human)
nuclear membrane disassemblySerine/threonine-protein kinase PLK1Homo sapiens (human)
positive regulation of ubiquitin-protein transferase activitySerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of cell cycleSerine/threonine-protein kinase PLK1Homo sapiens (human)
synaptonemal complex disassemblySerine/threonine-protein kinase PLK1Homo sapiens (human)
protein localization to chromatinSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein localization to nuclear envelopeSerine/threonine-protein kinase PLK1Homo sapiens (human)
double-strand break repair via alternative nonhomologous end joiningSerine/threonine-protein kinase PLK1Homo sapiens (human)
positive regulation of protein localization to nucleusSerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of mitotic spindle assemblySerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of mitotic cell cycle phase transitionSerine/threonine-protein kinase PLK1Homo sapiens (human)
positive regulation of ubiquitin protein ligase activitySerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of protein localization to cell cortexSerine/threonine-protein kinase PLK1Homo sapiens (human)
regulation of anaphase-promoting complex-dependent catabolic processSerine/threonine-protein kinase PLK1Homo sapiens (human)
negative regulation of double-strand break repair via homologous recombinationSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein phosphorylationLIM domain kinase 1Homo sapiens (human)
signal transductionLIM domain kinase 1Homo sapiens (human)
Rho protein signal transductionLIM domain kinase 1Homo sapiens (human)
nervous system developmentLIM domain kinase 1Homo sapiens (human)
positive regulation of actin filament bundle assemblyLIM domain kinase 1Homo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisLIM domain kinase 1Homo sapiens (human)
stress fiber assemblyLIM domain kinase 1Homo sapiens (human)
positive regulation of axon extensionLIM domain kinase 1Homo sapiens (human)
axon extensionLIM domain kinase 1Homo sapiens (human)
negative regulation of ubiquitin-protein transferase activityLIM domain kinase 1Homo sapiens (human)
positive regulation of stress fiber assemblyLIM domain kinase 1Homo sapiens (human)
actin cytoskeleton organizationLIM domain kinase 1Homo sapiens (human)
positive regulation of protein phosphorylationLIM domain kinase 2Homo sapiens (human)
protein phosphorylationLIM domain kinase 2Homo sapiens (human)
spermatogenesisLIM domain kinase 2Homo sapiens (human)
phosphorylationLIM domain kinase 2Homo sapiens (human)
astral microtubule organizationLIM domain kinase 2Homo sapiens (human)
establishment of vesicle localizationLIM domain kinase 2Homo sapiens (human)
head developmentLIM domain kinase 2Homo sapiens (human)
cornea development in camera-type eyeLIM domain kinase 2Homo sapiens (human)
positive regulation of protein localization to nucleusLIM domain kinase 2Homo sapiens (human)
negative regulation of cilium assemblyLIM domain kinase 2Homo sapiens (human)
actin cytoskeleton organizationLIM domain kinase 2Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase 10Homo sapiens (human)
signal transductionMitogen-activated protein kinase 10Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase 10Homo sapiens (human)
response to light stimulusMitogen-activated protein kinase 10Homo sapiens (human)
Fc-epsilon receptor signaling pathwayMitogen-activated protein kinase 10Homo sapiens (human)
regulation of circadian rhythmMitogen-activated protein kinase 10Homo sapiens (human)
rhythmic processMitogen-activated protein kinase 10Homo sapiens (human)
cellular senescenceMitogen-activated protein kinase 10Homo sapiens (human)
tyrosyl-tRNA aminoacylationTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
apoptotic processTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
response to starvationTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
regulation of glycolytic process5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
protein phosphorylation5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
fatty acid biosynthetic process5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
signal transduction5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
spermatogenesis5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
positive regulation of gene expression5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
cellular response to nutrient levels5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
positive regulation of protein kinase activity5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
import into nucleus5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
regulation of catalytic activity5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
angiogenesisEphrin type-B receptor 3Homo sapiens (human)
urogenital system developmentEphrin type-B receptor 3Homo sapiens (human)
axon guidanceEphrin type-B receptor 3Homo sapiens (human)
axonal fasciculationEphrin type-B receptor 3Homo sapiens (human)
cell migrationEphrin type-B receptor 3Homo sapiens (human)
central nervous system projection neuron axonogenesisEphrin type-B receptor 3Homo sapiens (human)
corpus callosum developmentEphrin type-B receptor 3Homo sapiens (human)
regulation of cell-cell adhesionEphrin type-B receptor 3Homo sapiens (human)
retinal ganglion cell axon guidanceEphrin type-B receptor 3Homo sapiens (human)
substrate adhesion-dependent cell spreadingEphrin type-B receptor 3Homo sapiens (human)
regulation of GTPase activityEphrin type-B receptor 3Homo sapiens (human)
protein autophosphorylationEphrin type-B receptor 3Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-B receptor 3Homo sapiens (human)
thymus developmentEphrin type-B receptor 3Homo sapiens (human)
digestive tract morphogenesisEphrin type-B receptor 3Homo sapiens (human)
regulation of axonogenesisEphrin type-B receptor 3Homo sapiens (human)
positive regulation of synapse assemblyEphrin type-B receptor 3Homo sapiens (human)
roof of mouth developmentEphrin type-B receptor 3Homo sapiens (human)
dendritic spine developmentEphrin type-B receptor 3Homo sapiens (human)
dendritic spine morphogenesisEphrin type-B receptor 3Homo sapiens (human)
protein phosphorylationEphrin type-B receptor 3Homo sapiens (human)
axon guidanceEphrin type-A receptor 5Homo sapiens (human)
cAMP-mediated signalingEphrin type-A receptor 5Homo sapiens (human)
hippocampus developmentEphrin type-A receptor 5Homo sapiens (human)
positive regulation of CREB transcription factor activityEphrin type-A receptor 5Homo sapiens (human)
regulation of actin cytoskeleton organizationEphrin type-A receptor 5Homo sapiens (human)
regulation of GTPase activityEphrin type-A receptor 5Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-A receptor 5Homo sapiens (human)
neuron developmentEphrin type-A receptor 5Homo sapiens (human)
regulation of insulin secretion involved in cellular response to glucose stimulusEphrin type-A receptor 5Homo sapiens (human)
protein phosphorylationEphrin type-A receptor 5Homo sapiens (human)
angiogenesisEphrin type-B receptor 4Homo sapiens (human)
cell migration involved in sprouting angiogenesisEphrin type-B receptor 4Homo sapiens (human)
heart morphogenesisEphrin type-B receptor 4Homo sapiens (human)
cell adhesionEphrin type-B receptor 4Homo sapiens (human)
protein autophosphorylationEphrin type-B receptor 4Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-B receptor 4Homo sapiens (human)
multicellular organism developmentEphrin type-B receptor 4Homo sapiens (human)
positive regulation of kinase activityEphrin type-B receptor 4Homo sapiens (human)
negative regulation of cellular response to hypoxiaEphrin type-A receptor 4Homo sapiens (human)
cell adhesionEphrin type-A receptor 4Homo sapiens (human)
negative regulation of cell adhesionEphrin type-A receptor 4Homo sapiens (human)
adult walking behaviorEphrin type-A receptor 4Homo sapiens (human)
motor neuron axon guidanceEphrin type-A receptor 4Homo sapiens (human)
positive regulation of cell population proliferationEphrin type-A receptor 4Homo sapiens (human)
glial cell migrationEphrin type-A receptor 4Homo sapiens (human)
negative regulation of epithelial to mesenchymal transitionEphrin type-A receptor 4Homo sapiens (human)
negative regulation of neuron projection developmentEphrin type-A receptor 4Homo sapiens (human)
negative regulation of translationEphrin type-A receptor 4Homo sapiens (human)
peptidyl-tyrosine phosphorylationEphrin type-A receptor 4Homo sapiens (human)
corticospinal tract morphogenesisEphrin type-A receptor 4Homo sapiens (human)
positive regulation of cell migrationEphrin type-A receptor 4Homo sapiens (human)
negative regulation of cell migrationEphrin type-A receptor 4Homo sapiens (human)
adherens junction organizationEphrin type-A receptor 4Homo sapiens (human)
regulation of GTPase activityEphrin type-A receptor 4Homo sapiens (human)
positive regulation of cell adhesionEphrin type-A receptor 4Homo sapiens (human)
protein autophosphorylationEphrin type-A receptor 4Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-A receptor 4Homo sapiens (human)
negative regulation of axon regenerationEphrin type-A receptor 4Homo sapiens (human)
regulation of astrocyte differentiationEphrin type-A receptor 4Homo sapiens (human)
regulation of axonogenesisEphrin type-A receptor 4Homo sapiens (human)
positive regulation of dendrite morphogenesisEphrin type-A receptor 4Homo sapiens (human)
protein stabilizationEphrin type-A receptor 4Homo sapiens (human)
regulation of dendritic spine morphogenesisEphrin type-A receptor 4Homo sapiens (human)
positive regulation of protein tyrosine kinase activityEphrin type-A receptor 4Homo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeEphrin type-A receptor 4Homo sapiens (human)
nephric duct morphogenesisEphrin type-A receptor 4Homo sapiens (human)
cochlea developmentEphrin type-A receptor 4Homo sapiens (human)
fasciculation of sensory neuron axonEphrin type-A receptor 4Homo sapiens (human)
fasciculation of motor neuron axonEphrin type-A receptor 4Homo sapiens (human)
neuron projection guidanceEphrin type-A receptor 4Homo sapiens (human)
synapse pruningEphrin type-A receptor 4Homo sapiens (human)
neuron projection fasciculationEphrin type-A receptor 4Homo sapiens (human)
negative regulation of long-term synaptic potentiationEphrin type-A receptor 4Homo sapiens (human)
positive regulation of amyloid-beta formationEphrin type-A receptor 4Homo sapiens (human)
positive regulation of aspartic-type endopeptidase activity involved in amyloid precursor protein catabolic processEphrin type-A receptor 4Homo sapiens (human)
negative regulation of proteolysis involved in protein catabolic processEphrin type-A receptor 4Homo sapiens (human)
cellular response to amyloid-betaEphrin type-A receptor 4Homo sapiens (human)
regulation of modification of synaptic structureEphrin type-A receptor 4Homo sapiens (human)
regulation of synapse pruningEphrin type-A receptor 4Homo sapiens (human)
positive regulation of Rho guanyl-nucleotide exchange factor activityEphrin type-A receptor 4Homo sapiens (human)
protein phosphorylationEphrin type-A receptor 4Homo sapiens (human)
axon guidanceEphrin type-A receptor 4Homo sapiens (human)
ADP biosynthetic processAdenylate kinase 2, mitochondrialHomo sapiens (human)
nucleobase-containing small molecule interconversionAdenylate kinase 2, mitochondrialHomo sapiens (human)
AMP metabolic processAdenylate kinase 2, mitochondrialHomo sapiens (human)
ATP metabolic processAdenylate kinase 2, mitochondrialHomo sapiens (human)
nucleoside monophosphate phosphorylationAdenylate kinase 2, mitochondrialHomo sapiens (human)
purine ribonucleoside salvageAdenosine kinaseHomo sapiens (human)
dATP biosynthetic processAdenosine kinaseHomo sapiens (human)
ribonucleoside monophosphate biosynthetic processAdenosine kinaseHomo sapiens (human)
GMP salvageAdenosine kinaseHomo sapiens (human)
AMP salvageAdenosine kinaseHomo sapiens (human)
dAMP salvageAdenosine kinaseHomo sapiens (human)
purine nucleobase metabolic processAdenosine kinaseHomo sapiens (human)
Golgi to plasma membrane transportRas-related protein Rab-10Homo sapiens (human)
axonogenesisRas-related protein Rab-10Homo sapiens (human)
vesicle-mediated transportRas-related protein Rab-10Homo sapiens (human)
endosomal transportRas-related protein Rab-10Homo sapiens (human)
antigen processing and presentationRas-related protein Rab-10Homo sapiens (human)
polarized epithelial cell differentiationRas-related protein Rab-10Homo sapiens (human)
cellular response to insulin stimulusRas-related protein Rab-10Homo sapiens (human)
Golgi to plasma membrane protein transportRas-related protein Rab-10Homo sapiens (human)
regulated exocytosisRas-related protein Rab-10Homo sapiens (human)
establishment of neuroblast polarityRas-related protein Rab-10Homo sapiens (human)
endoplasmic reticulum tubular network organizationRas-related protein Rab-10Homo sapiens (human)
protein localization to plasma membraneRas-related protein Rab-10Homo sapiens (human)
establishment of protein localization to membraneRas-related protein Rab-10Homo sapiens (human)
establishment of protein localization to endoplasmic reticulum membraneRas-related protein Rab-10Homo sapiens (human)
cell-cell adhesionRas-related protein Rab-10Homo sapiens (human)
protein localization to basolateral plasma membraneRas-related protein Rab-10Homo sapiens (human)
exocytosisRas-related protein Rab-10Homo sapiens (human)
protein secretionRas-related protein Rab-10Homo sapiens (human)
establishment or maintenance of cell polarityActin-related protein 3Homo sapiens (human)
asymmetric cell divisionActin-related protein 3Homo sapiens (human)
positive regulation of lamellipodium assemblyActin-related protein 3Homo sapiens (human)
meiotic chromosome movement towards spindle poleActin-related protein 3Homo sapiens (human)
meiotic cytokinesisActin-related protein 3Homo sapiens (human)
Arp2/3 complex-mediated actin nucleationActin-related protein 3Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIActin-related protein 3Homo sapiens (human)
spindle localizationActin-related protein 3Homo sapiens (human)
cilium assemblyActin-related protein 3Homo sapiens (human)
actin polymerization-dependent cell motilityActin-related protein 3Homo sapiens (human)
cellular response to type II interferonActin-related protein 3Homo sapiens (human)
regulation of double-strand break repair via nonhomologous end joiningActin-related protein 2Homo sapiens (human)
cilium assemblyActin-related protein 2Homo sapiens (human)
establishment or maintenance of cell polarityActin-related protein 2Homo sapiens (human)
asymmetric cell divisionActin-related protein 2Homo sapiens (human)
positive regulation of lamellipodium assemblyActin-related protein 2Homo sapiens (human)
meiotic chromosome movement towards spindle poleActin-related protein 2Homo sapiens (human)
cytosolic transportActin-related protein 2Homo sapiens (human)
meiotic cytokinesisActin-related protein 2Homo sapiens (human)
Arp2/3 complex-mediated actin nucleationActin-related protein 2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIActin-related protein 2Homo sapiens (human)
spindle localizationActin-related protein 2Homo sapiens (human)
cellular response to type II interferonActin-related protein 2Homo sapiens (human)
positive regulation of double-strand break repair via homologous recombinationActin-related protein 2Homo sapiens (human)
ribosomal large subunit export from nucleusGTP-binding nuclear protein RanHomo sapiens (human)
ribosomal small subunit export from nucleusGTP-binding nuclear protein RanHomo sapiens (human)
mitotic sister chromatid segregationGTP-binding nuclear protein RanHomo sapiens (human)
mitotic cell cycleGTP-binding nuclear protein RanHomo sapiens (human)
DNA metabolic processGTP-binding nuclear protein RanHomo sapiens (human)
protein import into nucleusGTP-binding nuclear protein RanHomo sapiens (human)
protein export from nucleusGTP-binding nuclear protein RanHomo sapiens (human)
mitotic spindle organizationGTP-binding nuclear protein RanHomo sapiens (human)
spermatid developmentGTP-binding nuclear protein RanHomo sapiens (human)
viral processGTP-binding nuclear protein RanHomo sapiens (human)
hippocampus developmentGTP-binding nuclear protein RanHomo sapiens (human)
actin cytoskeleton organizationGTP-binding nuclear protein RanHomo sapiens (human)
positive regulation of protein bindingGTP-binding nuclear protein RanHomo sapiens (human)
pre-miRNA export from nucleusGTP-binding nuclear protein RanHomo sapiens (human)
positive regulation of protein import into nucleusGTP-binding nuclear protein RanHomo sapiens (human)
GTP metabolic processGTP-binding nuclear protein RanHomo sapiens (human)
cell divisionGTP-binding nuclear protein RanHomo sapiens (human)
snRNA import into nucleusGTP-binding nuclear protein RanHomo sapiens (human)
cellular response to mineralocorticoid stimulusGTP-binding nuclear protein RanHomo sapiens (human)
protein localization to nucleolusGTP-binding nuclear protein RanHomo sapiens (human)
ribosomal subunit export from nucleusGTP-binding nuclear protein RanHomo sapiens (human)
protein phosphorylationCasein kinase I isoform gamma-2Homo sapiens (human)
Wnt signaling pathwayCasein kinase I isoform gamma-2Homo sapiens (human)
sphingolipid biosynthetic processCasein kinase I isoform gamma-2Homo sapiens (human)
signal transductionCasein kinase I isoform gamma-2Homo sapiens (human)
peptidyl-serine phosphorylationCasein kinase I isoform gamma-2Homo sapiens (human)
endocytosisCasein kinase I isoform gamma-2Homo sapiens (human)
positive regulation of canonical Wnt signaling pathwayCasein kinase I isoform gamma-2Homo sapiens (human)
DNA damage responseCyclin-dependent kinase 3Homo sapiens (human)
G1/S transition of mitotic cell cycleCyclin-dependent kinase 3Homo sapiens (human)
cell population proliferationCyclin-dependent kinase 3Homo sapiens (human)
G0 to G1 transitionCyclin-dependent kinase 3Homo sapiens (human)
negative regulation of Notch signaling pathwayCyclin-dependent kinase 3Homo sapiens (human)
cell divisionCyclin-dependent kinase 3Homo sapiens (human)
regulation of G2/M transition of mitotic cell cycleCyclin-dependent kinase 3Homo sapiens (human)
response to organic substanceCyclin-dependent kinase 3Homo sapiens (human)
signal transductionCyclin-dependent kinase 3Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 3Homo sapiens (human)
regulation of gene expressionCyclin-dependent kinase 3Homo sapiens (human)
G1/S transition of mitotic cell cycleCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICyclin-dependent kinase 6Homo sapiens (human)
positive regulation of cell-matrix adhesionCyclin-dependent kinase 6Homo sapiens (human)
type B pancreatic cell developmentCyclin-dependent kinase 6Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 6Homo sapiens (human)
Notch signaling pathwayCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of cell population proliferationCyclin-dependent kinase 6Homo sapiens (human)
response to virusCyclin-dependent kinase 6Homo sapiens (human)
regulation of gene expressionCyclin-dependent kinase 6Homo sapiens (human)
positive regulation of gene expressionCyclin-dependent kinase 6Homo sapiens (human)
astrocyte developmentCyclin-dependent kinase 6Homo sapiens (human)
dentate gyrus developmentCyclin-dependent kinase 6Homo sapiens (human)
lateral ventricle developmentCyclin-dependent kinase 6Homo sapiens (human)
T cell differentiation in thymusCyclin-dependent kinase 6Homo sapiens (human)
gliogenesisCyclin-dependent kinase 6Homo sapiens (human)
cell dedifferentiationCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of cell differentiationCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of myeloid cell differentiationCyclin-dependent kinase 6Homo sapiens (human)
regulation of erythrocyte differentiationCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of monocyte differentiationCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of osteoblast differentiationCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of cell cycleCyclin-dependent kinase 6Homo sapiens (human)
positive regulation of fibroblast proliferationCyclin-dependent kinase 6Homo sapiens (human)
generation of neuronsCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of epithelial cell proliferationCyclin-dependent kinase 6Homo sapiens (human)
cell divisionCyclin-dependent kinase 6Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 6Homo sapiens (human)
hematopoietic stem cell differentiationCyclin-dependent kinase 6Homo sapiens (human)
regulation of hematopoietic stem cell differentiationCyclin-dependent kinase 6Homo sapiens (human)
regulation of cell motilityCyclin-dependent kinase 6Homo sapiens (human)
negative regulation of cellular senescenceCyclin-dependent kinase 6Homo sapiens (human)
regulation of G2/M transition of mitotic cell cycleCyclin-dependent kinase 6Homo sapiens (human)
response to organic substanceCyclin-dependent kinase 6Homo sapiens (human)
signal transductionCyclin-dependent kinase 6Homo sapiens (human)
microtubule cytoskeleton organizationCyclin-dependent-like kinase 5 Homo sapiens (human)
neuron migrationCyclin-dependent-like kinase 5 Homo sapiens (human)
synaptic transmission, dopaminergicCyclin-dependent-like kinase 5 Homo sapiens (human)
protein phosphorylationCyclin-dependent-like kinase 5 Homo sapiens (human)
intracellular protein transportCyclin-dependent-like kinase 5 Homo sapiens (human)
cell-matrix adhesionCyclin-dependent-like kinase 5 Homo sapiens (human)
chemical synaptic transmissionCyclin-dependent-like kinase 5 Homo sapiens (human)
synapse assemblyCyclin-dependent-like kinase 5 Homo sapiens (human)
skeletal muscle tissue developmentCyclin-dependent-like kinase 5 Homo sapiens (human)
motor neuron axon guidanceCyclin-dependent-like kinase 5 Homo sapiens (human)
visual learningCyclin-dependent-like kinase 5 Homo sapiens (human)
Schwann cell developmentCyclin-dependent-like kinase 5 Homo sapiens (human)
synaptic vesicle exocytosisCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of macroautophagyCyclin-dependent-like kinase 5 Homo sapiens (human)
phosphorylationCyclin-dependent-like kinase 5 Homo sapiens (human)
peptidyl-serine phosphorylationCyclin-dependent-like kinase 5 Homo sapiens (human)
peptidyl-threonine phosphorylationCyclin-dependent-like kinase 5 Homo sapiens (human)
sensory perception of painCyclin-dependent-like kinase 5 Homo sapiens (human)
cerebellar cortex formationCyclin-dependent-like kinase 5 Homo sapiens (human)
hippocampus developmentCyclin-dependent-like kinase 5 Homo sapiens (human)
layer formation in cerebral cortexCyclin-dependent-like kinase 5 Homo sapiens (human)
central nervous system neuron developmentCyclin-dependent-like kinase 5 Homo sapiens (human)
corpus callosum developmentCyclin-dependent-like kinase 5 Homo sapiens (human)
neuron differentiationCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of cell migrationCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of axon extensionCyclin-dependent-like kinase 5 Homo sapiens (human)
neuron projection developmentCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of protein ubiquitinationCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of synaptic plasticityCyclin-dependent-like kinase 5 Homo sapiens (human)
receptor catabolic processCyclin-dependent-like kinase 5 Homo sapiens (human)
synaptic transmission, glutamatergicCyclin-dependent-like kinase 5 Homo sapiens (human)
protein localization to synapseCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of apoptotic processCyclin-dependent-like kinase 5 Homo sapiens (human)
receptor clusteringCyclin-dependent-like kinase 5 Homo sapiens (human)
positive regulation of neuron apoptotic processCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of cell cycleCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of proteolysisCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of DNA-templated transcriptionCyclin-dependent-like kinase 5 Homo sapiens (human)
positive regulation of calcium ion-dependent exocytosisCyclin-dependent-like kinase 5 Homo sapiens (human)
negative regulation of protein export from nucleusCyclin-dependent-like kinase 5 Homo sapiens (human)
behavioral response to cocaineCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of synaptic plasticityCyclin-dependent-like kinase 5 Homo sapiens (human)
synaptic vesicle endocytosisCyclin-dependent-like kinase 5 Homo sapiens (human)
rhythmic processCyclin-dependent-like kinase 5 Homo sapiens (human)
axon extensionCyclin-dependent-like kinase 5 Homo sapiens (human)
oligodendrocyte differentiationCyclin-dependent-like kinase 5 Homo sapiens (human)
dendrite morphogenesisCyclin-dependent-like kinase 5 Homo sapiens (human)
cell divisionCyclin-dependent-like kinase 5 Homo sapiens (human)
neuron apoptotic processCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of cell cycleCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of synaptic transmission, glutamatergicCyclin-dependent-like kinase 5 Homo sapiens (human)
excitatory postsynaptic potentialCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of dendritic spine morphogenesisCyclin-dependent-like kinase 5 Homo sapiens (human)
calcium ion importCyclin-dependent-like kinase 5 Homo sapiens (human)
positive regulation of protein targeting to membraneCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of protein localization to plasma membraneCyclin-dependent-like kinase 5 Homo sapiens (human)
regulation of synaptic vesicle recyclingCyclin-dependent-like kinase 5 Homo sapiens (human)
cellular response to amyloid-betaCyclin-dependent-like kinase 5 Homo sapiens (human)
axonogenesisCyclin-dependent-like kinase 5 Homo sapiens (human)
synaptic vesicle transportCyclin-dependent-like kinase 5 Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 16Homo sapiens (human)
exocytosisCyclin-dependent kinase 16Homo sapiens (human)
spermatogenesisCyclin-dependent kinase 16Homo sapiens (human)
positive regulation of autophagyCyclin-dependent kinase 16Homo sapiens (human)
growth hormone secretionCyclin-dependent kinase 16Homo sapiens (human)
neuron projection developmentCyclin-dependent kinase 16Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 16Homo sapiens (human)
regulation of insulin secretion involved in cellular response to glucose stimulusCyclin-dependent kinase 16Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 17Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 17Homo sapiens (human)
cellular response to leukemia inhibitory factorATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
canonical glycolysisATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
fructose 1,6-bisphosphate metabolic processATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
fructose 6-phosphate metabolic processATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
chemotaxisDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
signal transductionDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
heart developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
negative regulation of cell population proliferationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
positive regulation of gene expressionDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
Schwann cell developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
cerebellar cortex formationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
keratinocyte differentiationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
thyroid gland developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
regulation of stress-activated MAPK cascadeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
endodermal cell differentiationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
ERBB2-ERBB3 signaling pathwayDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
myelinationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
type B pancreatic cell proliferationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
positive regulation of DNA-templated transcriptionDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
insulin-like growth factor receptor signaling pathwayDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
thymus developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
regulation of axon regenerationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
cell motilityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
positive regulation of axonogenesisDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
Bergmann glial cell differentiationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
face developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
trachea formationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
epithelial cell proliferation involved in lung morphogenesisDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
placenta blood vessel developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
labyrinthine layer developmentDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
ERK1 and ERK2 cascadeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
positive regulation of protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
regulation of Golgi inheritanceDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
cellular senescenceDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
positive regulation of endodermal cell differentiationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
regulation of early endosome to late endosome transportDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
neuron differentiationDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
MAPK cascadeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
neuron migrationDNA topoisomerase 2-betaHomo sapiens (human)
DNA topological changeDNA topoisomerase 2-betaHomo sapiens (human)
axonogenesisDNA topoisomerase 2-betaHomo sapiens (human)
B cell differentiationDNA topoisomerase 2-betaHomo sapiens (human)
forebrain developmentDNA topoisomerase 2-betaHomo sapiens (human)
positive regulation of single stranded viral RNA replication via double stranded DNA intermediateDNA topoisomerase 2-betaHomo sapiens (human)
cellular response to hydrogen peroxideDNA topoisomerase 2-betaHomo sapiens (human)
cellular response to ATPDNA topoisomerase 2-betaHomo sapiens (human)
cellular senescenceDNA topoisomerase 2-betaHomo sapiens (human)
positive regulation of double-strand break repair via nonhomologous end joiningDNA topoisomerase 2-betaHomo sapiens (human)
sister chromatid segregationDNA topoisomerase 2-betaHomo sapiens (human)
resolution of meiotic recombination intermediatesDNA topoisomerase 2-betaHomo sapiens (human)
regulation of cell growthProtein kinase C theta typeHomo sapiens (human)
regulation of DNA-templated transcriptionProtein kinase C theta typeHomo sapiens (human)
protein phosphorylationProtein kinase C theta typeHomo sapiens (human)
membrane protein ectodomain proteolysisProtein kinase C theta typeHomo sapiens (human)
inflammatory responseProtein kinase C theta typeHomo sapiens (human)
axon guidanceProtein kinase C theta typeHomo sapiens (human)
positive regulation of telomere maintenance via telomeraseProtein kinase C theta typeHomo sapiens (human)
positive regulation of interleukin-17 productionProtein kinase C theta typeHomo sapiens (human)
positive regulation of interleukin-2 productionProtein kinase C theta typeHomo sapiens (human)
positive regulation of interleukin-4 productionProtein kinase C theta typeHomo sapiens (human)
intracellular signal transductionProtein kinase C theta typeHomo sapiens (human)
CD4-positive, alpha-beta T cell proliferationProtein kinase C theta typeHomo sapiens (human)
Fc-epsilon receptor signaling pathwayProtein kinase C theta typeHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayProtein kinase C theta typeHomo sapiens (human)
positive regulation of T cell activationProtein kinase C theta typeHomo sapiens (human)
positive regulation of NF-kappaB transcription factor activityProtein kinase C theta typeHomo sapiens (human)
positive regulation of telomerase activityProtein kinase C theta typeHomo sapiens (human)
cell chemotaxisProtein kinase C theta typeHomo sapiens (human)
negative regulation of T cell apoptotic processProtein kinase C theta typeHomo sapiens (human)
regulation of platelet aggregationProtein kinase C theta typeHomo sapiens (human)
positive regulation of telomere cappingProtein kinase C theta typeHomo sapiens (human)
positive regulation of T-helper 17 type immune responseProtein kinase C theta typeHomo sapiens (human)
positive regulation of CD4-positive, alpha-beta T cell proliferationProtein kinase C theta typeHomo sapiens (human)
positive regulation of T-helper 2 cell activationProtein kinase C theta typeHomo sapiens (human)
peptidyl-serine phosphorylationProtein kinase C theta typeHomo sapiens (human)
outflow tract septum morphogenesisActivin receptor type-1Homo sapiens (human)
branching involved in blood vessel morphogenesisActivin receptor type-1Homo sapiens (human)
in utero embryonic developmentActivin receptor type-1Homo sapiens (human)
gastrulation with mouth forming secondActivin receptor type-1Homo sapiens (human)
mesoderm formationActivin receptor type-1Homo sapiens (human)
neural crest cell migrationActivin receptor type-1Homo sapiens (human)
acute inflammatory responseActivin receptor type-1Homo sapiens (human)
embryonic heart tube morphogenesisActivin receptor type-1Homo sapiens (human)
atrioventricular valve morphogenesisActivin receptor type-1Homo sapiens (human)
mitral valve morphogenesisActivin receptor type-1Homo sapiens (human)
endocardial cushion formationActivin receptor type-1Homo sapiens (human)
endocardial cushion fusionActivin receptor type-1Homo sapiens (human)
atrial septum primum morphogenesisActivin receptor type-1Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayActivin receptor type-1Homo sapiens (human)
germ cell developmentActivin receptor type-1Homo sapiens (human)
determination of left/right symmetryActivin receptor type-1Homo sapiens (human)
negative regulation of signal transductionActivin receptor type-1Homo sapiens (human)
regulation of ossificationActivin receptor type-1Homo sapiens (human)
positive regulation of cell migrationActivin receptor type-1Homo sapiens (human)
positive regulation of bone mineralizationActivin receptor type-1Homo sapiens (human)
BMP signaling pathwayActivin receptor type-1Homo sapiens (human)
activin receptor signaling pathwayActivin receptor type-1Homo sapiens (human)
negative regulation of activin receptor signaling pathwayActivin receptor type-1Homo sapiens (human)
positive regulation of osteoblast differentiationActivin receptor type-1Homo sapiens (human)
positive regulation of DNA-templated transcriptionActivin receptor type-1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIActivin receptor type-1Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationActivin receptor type-1Homo sapiens (human)
smooth muscle cell differentiationActivin receptor type-1Homo sapiens (human)
pharyngeal system developmentActivin receptor type-1Homo sapiens (human)
positive regulation of SMAD protein signal transductionActivin receptor type-1Homo sapiens (human)
ventricular septum morphogenesisActivin receptor type-1Homo sapiens (human)
cardiac muscle cell fate commitmentActivin receptor type-1Homo sapiens (human)
endocardial cushion cell fate commitmentActivin receptor type-1Homo sapiens (human)
positive regulation of cardiac epithelial to mesenchymal transitionActivin receptor type-1Homo sapiens (human)
cellular response to BMP stimulusActivin receptor type-1Homo sapiens (human)
positive regulation of determination of dorsal identityActivin receptor type-1Homo sapiens (human)
negative regulation of G1/S transition of mitotic cell cycleActivin receptor type-1Homo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathwayActivin receptor type-1Homo sapiens (human)
dorsal/ventral pattern formationActivin receptor type-1Homo sapiens (human)
heart developmentActivin receptor type-1Homo sapiens (human)
protein phosphorylationActivin receptor type-1Homo sapiens (human)
cellular response to growth factor stimulusActivin receptor type-1Homo sapiens (human)
defense responseMacrophage-stimulating protein receptorHomo sapiens (human)
signal transductionMacrophage-stimulating protein receptorHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayMacrophage-stimulating protein receptorHomo sapiens (human)
single fertilizationMacrophage-stimulating protein receptorHomo sapiens (human)
positive regulation of cell population proliferationMacrophage-stimulating protein receptorHomo sapiens (human)
response to virusMacrophage-stimulating protein receptorHomo sapiens (human)
macrophage colony-stimulating factor signaling pathwayMacrophage-stimulating protein receptorHomo sapiens (human)
positive regulation of MAP kinase activityMacrophage-stimulating protein receptorHomo sapiens (human)
innate immune responseMacrophage-stimulating protein receptorHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionMacrophage-stimulating protein receptorHomo sapiens (human)
nervous system developmentMacrophage-stimulating protein receptorHomo sapiens (human)
cell migrationMacrophage-stimulating protein receptorHomo sapiens (human)
phagocytosisMacrophage-stimulating protein receptorHomo sapiens (human)
positive regulation of kinase activityMacrophage-stimulating protein receptorHomo sapiens (human)
multicellular organism developmentMacrophage-stimulating protein receptorHomo sapiens (human)
positive regulation of macrophage chemotaxisFocal adhesion kinase 1Homo sapiens (human)
positive regulation of macrophage proliferationFocal adhesion kinase 1Homo sapiens (human)
angiogenesisFocal adhesion kinase 1Homo sapiens (human)
placenta developmentFocal adhesion kinase 1Homo sapiens (human)
regulation of protein phosphorylationFocal adhesion kinase 1Homo sapiens (human)
positive regulation of protein phosphorylationFocal adhesion kinase 1Homo sapiens (human)
heart morphogenesisFocal adhesion kinase 1Homo sapiens (human)
signal complex assemblyFocal adhesion kinase 1Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
integrin-mediated signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
axon guidanceFocal adhesion kinase 1Homo sapiens (human)
positive regulation of cell population proliferationFocal adhesion kinase 1Homo sapiens (human)
regulation of cell shapeFocal adhesion kinase 1Homo sapiens (human)
regulation of endothelial cell migrationFocal adhesion kinase 1Homo sapiens (human)
regulation of epithelial cell migrationFocal adhesion kinase 1Homo sapiens (human)
positive regulation of epithelial cell migrationFocal adhesion kinase 1Homo sapiens (human)
positive regulation of epithelial to mesenchymal transitionFocal adhesion kinase 1Homo sapiens (human)
positive regulation of fibroblast migrationFocal adhesion kinase 1Homo sapiens (human)
cell migrationFocal adhesion kinase 1Homo sapiens (human)
peptidyl-tyrosine phosphorylationFocal adhesion kinase 1Homo sapiens (human)
negative regulation of cell-cell adhesionFocal adhesion kinase 1Homo sapiens (human)
establishment of cell polarityFocal adhesion kinase 1Homo sapiens (human)
positive regulation of cell migrationFocal adhesion kinase 1Homo sapiens (human)
regulation of cell adhesion mediated by integrinFocal adhesion kinase 1Homo sapiens (human)
detection of muscle stretchFocal adhesion kinase 1Homo sapiens (human)
netrin-activated signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisFocal adhesion kinase 1Homo sapiens (human)
regulation of cell population proliferationFocal adhesion kinase 1Homo sapiens (human)
negative regulation of apoptotic processFocal adhesion kinase 1Homo sapiens (human)
regulation of GTPase activityFocal adhesion kinase 1Homo sapiens (human)
regulation of osteoblast differentiationFocal adhesion kinase 1Homo sapiens (human)
positive regulation of protein kinase activityFocal adhesion kinase 1Homo sapiens (human)
protein autophosphorylationFocal adhesion kinase 1Homo sapiens (human)
vascular endothelial growth factor receptor signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
ephrin receptor signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
cell motilityFocal adhesion kinase 1Homo sapiens (human)
regulation of cytoskeleton organizationFocal adhesion kinase 1Homo sapiens (human)
regulation of focal adhesion assemblyFocal adhesion kinase 1Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionFocal adhesion kinase 1Homo sapiens (human)
growth hormone receptor signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
positive regulation of wound healingFocal adhesion kinase 1Homo sapiens (human)
regulation of substrate adhesion-dependent cell spreadingFocal adhesion kinase 1Homo sapiens (human)
positive regulation of ubiquitin-dependent protein catabolic processFocal adhesion kinase 1Homo sapiens (human)
negative regulation of anoikisFocal adhesion kinase 1Homo sapiens (human)
protein phosphorylationFocal adhesion kinase 1Homo sapiens (human)
epidermal growth factor receptor signaling pathwayFocal adhesion kinase 1Homo sapiens (human)
regulation of cell adhesionFocal adhesion kinase 1Homo sapiens (human)
protein phosphorylationProtein kinase C delta typeHomo sapiens (human)
apoptotic processProtein kinase C delta typeHomo sapiens (human)
DNA damage responseProtein kinase C delta typeHomo sapiens (human)
signal transductionProtein kinase C delta typeHomo sapiens (human)
intrinsic apoptotic signaling pathway in response to oxidative stressProtein kinase C delta typeHomo sapiens (human)
regulation of signaling receptor activityProtein kinase C delta typeHomo sapiens (human)
immunoglobulin mediated immune responseProtein kinase C delta typeHomo sapiens (human)
peptidyl-serine phosphorylationProtein kinase C delta typeHomo sapiens (human)
peptidyl-threonine phosphorylationProtein kinase C delta typeHomo sapiens (human)
termination of signal transductionProtein kinase C delta typeHomo sapiens (human)
negative regulation of actin filament polymerizationProtein kinase C delta typeHomo sapiens (human)
positive regulation of endodeoxyribonuclease activityProtein kinase C delta typeHomo sapiens (human)
negative regulation of protein bindingProtein kinase C delta typeHomo sapiens (human)
activation of protein kinase activityProtein kinase C delta typeHomo sapiens (human)
positive regulation of superoxide anion generationProtein kinase C delta typeHomo sapiens (human)
regulation of actin cytoskeleton organizationProtein kinase C delta typeHomo sapiens (human)
negative regulation of glial cell apoptotic processProtein kinase C delta typeHomo sapiens (human)
cellular response to UVProtein kinase C delta typeHomo sapiens (human)
positive regulation of protein dephosphorylationProtein kinase C delta typeHomo sapiens (human)
Fc-gamma receptor signaling pathway involved in phagocytosisProtein kinase C delta typeHomo sapiens (human)
B cell proliferationProtein kinase C delta typeHomo sapiens (human)
neutrophil activationProtein kinase C delta typeHomo sapiens (human)
positive regulation of protein import into nucleusProtein kinase C delta typeHomo sapiens (human)
defense response to bacteriumProtein kinase C delta typeHomo sapiens (human)
negative regulation of MAP kinase activityProtein kinase C delta typeHomo sapiens (human)
regulation of mRNA stabilityProtein kinase C delta typeHomo sapiens (human)
post-translational protein modificationProtein kinase C delta typeHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayProtein kinase C delta typeHomo sapiens (human)
negative regulation of inflammatory responseProtein kinase C delta typeHomo sapiens (human)
negative regulation of peptidyl-tyrosine phosphorylationProtein kinase C delta typeHomo sapiens (human)
protein stabilizationProtein kinase C delta typeHomo sapiens (human)
negative regulation of filopodium assemblyProtein kinase C delta typeHomo sapiens (human)
cell chemotaxisProtein kinase C delta typeHomo sapiens (human)
cellular response to hydrogen peroxideProtein kinase C delta typeHomo sapiens (human)
cellular response to hydroperoxideProtein kinase C delta typeHomo sapiens (human)
negative regulation of platelet aggregationProtein kinase C delta typeHomo sapiens (human)
cellular senescenceProtein kinase C delta typeHomo sapiens (human)
positive regulation of phospholipid scramblase activityProtein kinase C delta typeHomo sapiens (human)
cellular response to angiotensinProtein kinase C delta typeHomo sapiens (human)
regulation of ceramide biosynthetic processProtein kinase C delta typeHomo sapiens (human)
positive regulation of ceramide biosynthetic processProtein kinase C delta typeHomo sapiens (human)
positive regulation of glucosylceramide catabolic processProtein kinase C delta typeHomo sapiens (human)
positive regulation of sphingomyelin catabolic processProtein kinase C delta typeHomo sapiens (human)
positive regulation of apoptotic signaling pathwayProtein kinase C delta typeHomo sapiens (human)
intracellular signal transductionProtein kinase C delta typeHomo sapiens (human)
neutrophil homeostasisTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of type III hypersensitivityTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of type I hypersensitivityTyrosine-protein kinase BTKHomo sapiens (human)
adaptive immune responseTyrosine-protein kinase BTKHomo sapiens (human)
B cell affinity maturationTyrosine-protein kinase BTKHomo sapiens (human)
histamine secretion by mast cellTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of immunoglobulin productionTyrosine-protein kinase BTKHomo sapiens (human)
regulation of B cell cytokine productionTyrosine-protein kinase BTKHomo sapiens (human)
MyD88-dependent toll-like receptor signaling pathwayTyrosine-protein kinase BTKHomo sapiens (human)
regulation of B cell apoptotic processTyrosine-protein kinase BTKHomo sapiens (human)
protein phosphorylationTyrosine-protein kinase BTKHomo sapiens (human)
mesoderm developmentTyrosine-protein kinase BTKHomo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase BTKHomo sapiens (human)
calcium-mediated signalingTyrosine-protein kinase BTKHomo sapiens (human)
proteoglycan catabolic processTyrosine-protein kinase BTKHomo sapiens (human)
negative regulation of B cell proliferationTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of B cell proliferationTyrosine-protein kinase BTKHomo sapiens (human)
response to lipopolysaccharideTyrosine-protein kinase BTKHomo sapiens (human)
negative regulation of interleukin-10 productionTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of interleukin-6 productionTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of tumor necrosis factor productionTyrosine-protein kinase BTKHomo sapiens (human)
cellular response to reactive oxygen speciesTyrosine-protein kinase BTKHomo sapiens (human)
intracellular signal transductionTyrosine-protein kinase BTKHomo sapiens (human)
Fc-epsilon receptor signaling pathwayTyrosine-protein kinase BTKHomo sapiens (human)
B cell activationTyrosine-protein kinase BTKHomo sapiens (human)
innate immune responseTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of B cell differentiationTyrosine-protein kinase BTKHomo sapiens (human)
cell maturationTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of phagocytosisTyrosine-protein kinase BTKHomo sapiens (human)
B cell receptor signaling pathwayTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of NF-kappaB transcription factor activityTyrosine-protein kinase BTKHomo sapiens (human)
monocyte proliferationTyrosine-protein kinase BTKHomo sapiens (human)
cellular response to molecule of fungal originTyrosine-protein kinase BTKHomo sapiens (human)
apoptotic signaling pathwayTyrosine-protein kinase BTKHomo sapiens (human)
cellular response to interleukin-7Tyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of interleukin-17A productionTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of NLRP3 inflammasome complex assemblyTyrosine-protein kinase BTKHomo sapiens (human)
positive regulation of synoviocyte proliferationTyrosine-protein kinase BTKHomo sapiens (human)
eosinophil homeostasisTyrosine-protein kinase BTKHomo sapiens (human)
T cell receptor signaling pathwayTyrosine-protein kinase BTKHomo sapiens (human)
endocytosisActivated CDC42 kinase 1Homo sapiens (human)
cell surface receptor signaling pathwayActivated CDC42 kinase 1Homo sapiens (human)
small GTPase-mediated signal transductionActivated CDC42 kinase 1Homo sapiens (human)
phosphorylationActivated CDC42 kinase 1Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationActivated CDC42 kinase 1Homo sapiens (human)
regulation of clathrin-dependent endocytosisActivated CDC42 kinase 1Homo sapiens (human)
protein phosphorylationActivated CDC42 kinase 1Homo sapiens (human)
regulation of cell growthEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
regulation of cell-matrix adhesionEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
cell adhesionEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
embryo implantationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
lactationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
cell population proliferationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
negative regulation of cell population proliferationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
regulation of extracellular matrix disassemblyEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
smooth muscle cell migrationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
collagen-activated tyrosine kinase receptor signaling pathwayEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
peptidyl-tyrosine autophosphorylationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
ear developmentEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
wound healing, spreading of cellsEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
protein autophosphorylationEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
branching involved in mammary gland duct morphogenesisEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
mammary gland alveolus developmentEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
smooth muscle cell-matrix adhesionEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
axon developmentEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
neuron projection extensionEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
multicellular organism developmentEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
positive regulation of kinase activityEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
positive regulation of neuron projection developmentEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
vesicle targetingMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
immune responseMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
positive regulation of JUN kinase activityMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
innate immune responseMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
positive regulation of JNK cascadeMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
cell morphogenesisSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of protein phosphorylationSerine/threonine-protein kinase 4Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of protein bindingSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of peptidyl-serine phosphorylationSerine/threonine-protein kinase 4Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase 4Homo sapiens (human)
protein stabilizationSerine/threonine-protein kinase 4Homo sapiens (human)
branching involved in blood vessel morphogenesisSerine/threonine-protein kinase 4Homo sapiens (human)
neural tube formationSerine/threonine-protein kinase 4Homo sapiens (human)
endocardium developmentSerine/threonine-protein kinase 4Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase 4Homo sapiens (human)
protein import into nucleusSerine/threonine-protein kinase 4Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase 4Homo sapiens (human)
signal transductionSerine/threonine-protein kinase 4Homo sapiens (human)
central nervous system developmentSerine/threonine-protein kinase 4Homo sapiens (human)
extrinsic apoptotic signaling pathway via death domain receptorsSerine/threonine-protein kinase 4Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase 4Homo sapiens (human)
keratinocyte differentiationSerine/threonine-protein kinase 4Homo sapiens (human)
organ growthSerine/threonine-protein kinase 4Homo sapiens (human)
hippo signalingSerine/threonine-protein kinase 4Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of apoptotic processSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of fat cell differentiationSerine/threonine-protein kinase 4Homo sapiens (human)
negative regulation of organ growthSerine/threonine-protein kinase 4Homo sapiens (human)
epithelial cell proliferationSerine/threonine-protein kinase 4Homo sapiens (human)
negative regulation of epithelial cell proliferationSerine/threonine-protein kinase 4Homo sapiens (human)
protein tetramerizationSerine/threonine-protein kinase 4Homo sapiens (human)
canonical Wnt signaling pathwaySerine/threonine-protein kinase 4Homo sapiens (human)
primitive hemopoiesisSerine/threonine-protein kinase 4Homo sapiens (human)
cell differentiation involved in embryonic placenta developmentSerine/threonine-protein kinase 4Homo sapiens (human)
regulation of cell differentiation involved in embryonic placenta developmentSerine/threonine-protein kinase 4Homo sapiens (human)
negative regulation of canonical Wnt signaling pathwaySerine/threonine-protein kinase 4Homo sapiens (human)
hepatocyte apoptotic processSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of extrinsic apoptotic signaling pathway via death domain receptorsSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of hepatocyte apoptotic processSerine/threonine-protein kinase 4Homo sapiens (human)
positive regulation of substrate-dependent cell migration, cell attachment to substrateSerine/threonine-protein kinase 4Homo sapiens (human)
regulation of MAPK cascadeSerine/threonine-protein kinase 4Homo sapiens (human)
lipid droplet disassembly5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
response to hypoxia5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
glucose metabolic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
chromatin remodeling5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein phosphorylation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
fatty acid biosynthetic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cholesterol biosynthetic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
autophagy5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
signal transduction5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of cell population proliferation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
lipid biosynthetic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
response to UV5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cold acclimation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
response to gamma radiation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of autophagy5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of gene expression5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of gene expression5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
response to activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
bile acid and bile salt transport5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
Wnt signaling pathway5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
fatty acid oxidation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
response to caffeine5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to nutrient levels5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of TOR signaling5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
regulation of peptidyl-serine phosphorylation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to oxidative stress5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
bile acid signaling pathway5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to glucose starvation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
glucose homeostasis5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
regulation of circadian rhythm5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of apoptotic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
response to estrogen5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of cholesterol biosynthetic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of glycolytic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of DNA-templated transcription5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of glucosylceramide biosynthetic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of insulin receptor signaling pathway5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
rhythmic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of skeletal muscle tissue development5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of lipid catabolic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
fatty acid homeostasis5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
regulation of vesicle-mediated transport5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
motor behavior5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
CAMKK-AMPK signaling cascade5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
regulation of stress granule assembly5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
neuron cellular homeostasis5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to hydrogen peroxide5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
regulation of microtubule cytoskeleton organization5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to calcium ion5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to glucose stimulus5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to ethanol5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to prostaglandin E stimulus5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to organonitrogen compound5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to hypoxia5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cellular response to xenobiotic stimulus5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
energy homeostasis5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
regulation of bile acid secretion5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of mitochondrial transcription5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of protein localization5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of hepatocyte apoptotic process5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of protein targeting to mitochondrion5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of adipose tissue development5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of TORC1 signaling5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of tubulin deacetylation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein localization to lipid droplet5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
positive regulation of peptidyl-lysine acetylation5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
MAPK cascadeDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
signal transductionDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
heart developmentDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
positive regulation of cell growthDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of NF-kappaB transcription factor activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of interleukin-8 productionDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of heterotypic cell-cell adhesionDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of smooth muscle cell apoptotic processDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
positive regulation of MAP kinase activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
insulin-like growth factor receptor signaling pathwayDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
positive regulation of epithelial cell proliferationDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
positive regulation of protein metabolic processDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of response to cytokine stimulusDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
ERK5 cascadeDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
cellular response to growth factor stimulusDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
cellular response to laminar fluid shear stressDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of cell migration involved in sprouting angiogenesisDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of chemokine (C-X-C motif) ligand 2 productionDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathway in absence of ligandDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase 7Homo sapiens (human)
signal transductionMitogen-activated protein kinase 7Homo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwayMitogen-activated protein kinase 7Homo sapiens (human)
cell differentiationMitogen-activated protein kinase 7Homo sapiens (human)
calcineurin-NFAT signaling cascadeMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of heterotypic cell-cell adhesionMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of smooth muscle cell apoptotic processMitogen-activated protein kinase 7Homo sapiens (human)
regulation of angiogenesisMitogen-activated protein kinase 7Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of inflammatory responseMitogen-activated protein kinase 7Homo sapiens (human)
positive regulation of protein metabolic processMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of response to cytokine stimulusMitogen-activated protein kinase 7Homo sapiens (human)
cellular response to hydrogen peroxideMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of calcineurin-NFAT signaling cascadeMitogen-activated protein kinase 7Homo sapiens (human)
cellular response to growth factor stimulusMitogen-activated protein kinase 7Homo sapiens (human)
cellular response to laminar fluid shear stressMitogen-activated protein kinase 7Homo sapiens (human)
cellular response to transforming growth factor beta stimulusMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of oxidative stress-induced intrinsic apoptotic signaling pathwayMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of endothelial cell apoptotic processMitogen-activated protein kinase 7Homo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathway in absence of ligandMitogen-activated protein kinase 7Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase 7Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
stimulatory C-type lectin receptor signaling pathwaySerine/threonine-protein kinase PAK 2Homo sapiens (human)
cardiac muscle hypertrophySerine/threonine-protein kinase PAK 2Homo sapiens (human)
negative regulation of protein kinase activitySerine/threonine-protein kinase PAK 2Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase PAK 2Homo sapiens (human)
signal transductionSerine/threonine-protein kinase PAK 2Homo sapiens (human)
phosphorylationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
adherens junction assemblySerine/threonine-protein kinase PAK 2Homo sapiens (human)
negative regulation of apoptotic processSerine/threonine-protein kinase PAK 2Homo sapiens (human)
vascular endothelial growth factor receptor signaling pathwaySerine/threonine-protein kinase PAK 2Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
regulation of cytoskeleton organizationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
negative regulation of stress fiber assemblySerine/threonine-protein kinase PAK 2Homo sapiens (human)
dendritic spine developmentSerine/threonine-protein kinase PAK 2Homo sapiens (human)
bicellular tight junction assemblySerine/threonine-protein kinase PAK 2Homo sapiens (human)
cellular response to organic cyclic compoundSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cellular response to transforming growth factor beta stimulusSerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein localization to cell-cell junctionSerine/threonine-protein kinase PAK 2Homo sapiens (human)
positive regulation of extrinsic apoptotic signaling pathwaySerine/threonine-protein kinase PAK 2Homo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in execution phase of apoptosisSerine/threonine-protein kinase PAK 2Homo sapiens (human)
regulation of axonogenesisSerine/threonine-protein kinase PAK 2Homo sapiens (human)
regulation of MAPK cascadeSerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase PAK 2Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase PAK 2Homo sapiens (human)
positive regulation of protein bindingSerine/threonine-protein kinase 3Homo sapiens (human)
protein stabilizationSerine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of DNA-binding transcription factor activitySerine/threonine-protein kinase 3Homo sapiens (human)
neural tube formationSerine/threonine-protein kinase 3Homo sapiens (human)
endocardium developmentSerine/threonine-protein kinase 3Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase 3Homo sapiens (human)
protein import into nucleusSerine/threonine-protein kinase 3Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase 3Homo sapiens (human)
JNK cascadeSerine/threonine-protein kinase 3Homo sapiens (human)
central nervous system developmentSerine/threonine-protein kinase 3Homo sapiens (human)
extrinsic apoptotic signaling pathway via death domain receptorsSerine/threonine-protein kinase 3Homo sapiens (human)
organ growthSerine/threonine-protein kinase 3Homo sapiens (human)
hippo signalingSerine/threonine-protein kinase 3Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of apoptotic processSerine/threonine-protein kinase 3Homo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionSerine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of fat cell differentiationSerine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of JNK cascadeSerine/threonine-protein kinase 3Homo sapiens (human)
negative regulation of organ growthSerine/threonine-protein kinase 3Homo sapiens (human)
epithelial cell proliferationSerine/threonine-protein kinase 3Homo sapiens (human)
negative regulation of epithelial cell proliferationSerine/threonine-protein kinase 3Homo sapiens (human)
protein tetramerizationSerine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionSerine/threonine-protein kinase 3Homo sapiens (human)
canonical Wnt signaling pathwaySerine/threonine-protein kinase 3Homo sapiens (human)
primitive hemopoiesisSerine/threonine-protein kinase 3Homo sapiens (human)
cell differentiation involved in embryonic placenta developmentSerine/threonine-protein kinase 3Homo sapiens (human)
regulation of cell differentiation involved in embryonic placenta developmentSerine/threonine-protein kinase 3Homo sapiens (human)
protein localization to centrosomeSerine/threonine-protein kinase 3Homo sapiens (human)
negative regulation of canonical Wnt signaling pathwaySerine/threonine-protein kinase 3Homo sapiens (human)
hepatocyte apoptotic processSerine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of extrinsic apoptotic signaling pathway via death domain receptorsSerine/threonine-protein kinase 3Homo sapiens (human)
regulation of MAPK cascadeSerine/threonine-protein kinase 3Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
Fc-epsilon receptor signaling pathwayMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
cellular response to mechanical stimulusMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
cell morphogenesisIntegrin-linked protein kinaseHomo sapiens (human)
integrin-mediated signaling pathwayIntegrin-linked protein kinaseHomo sapiens (human)
branching involved in ureteric bud morphogenesisIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of protein phosphorylationIntegrin-linked protein kinaseHomo sapiens (human)
outflow tract morphogenesisIntegrin-linked protein kinaseHomo sapiens (human)
protein phosphorylationIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of cell population proliferationIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of signal transductionIntegrin-linked protein kinaseHomo sapiens (human)
fibroblast migrationIntegrin-linked protein kinaseHomo sapiens (human)
nerve developmentIntegrin-linked protein kinaseHomo sapiens (human)
myelination in peripheral nervous systemIntegrin-linked protein kinaseHomo sapiens (human)
cell projection organizationIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of BMP signaling pathwayIntegrin-linked protein kinaseHomo sapiens (human)
tumor necrosis factor-mediated signaling pathwayIntegrin-linked protein kinaseHomo sapiens (human)
substrate adhesion-dependent cell spreadingIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of phosphorylationIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionIntegrin-linked protein kinaseHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionIntegrin-linked protein kinaseHomo sapiens (human)
establishment or maintenance of epithelial cell apical/basal polarityIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of osteoblast differentiationIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of DNA-templated transcriptionIntegrin-linked protein kinaseHomo sapiens (human)
neural precursor cell proliferationIntegrin-linked protein kinaseHomo sapiens (human)
platelet aggregationIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of canonical Wnt signaling pathwayIntegrin-linked protein kinaseHomo sapiens (human)
positive regulation of substrate adhesion-dependent cell spreadingIntegrin-linked protein kinaseHomo sapiens (human)
negative regulation of neural precursor cell proliferationIntegrin-linked protein kinaseHomo sapiens (human)
cell-matrix adhesionIntegrin-linked protein kinaseHomo sapiens (human)
integrin-mediated signaling pathwayIntegrin-linked protein kinaseHomo sapiens (human)
epithelial to mesenchymal transitionRho-associated protein kinase 1Homo sapiens (human)
aortic valve morphogenesisRho-associated protein kinase 1Homo sapiens (human)
apical constrictionRho-associated protein kinase 1Homo sapiens (human)
protein phosphorylationRho-associated protein kinase 1Homo sapiens (human)
smooth muscle contractionRho-associated protein kinase 1Homo sapiens (human)
leukocyte cell-cell adhesionRho-associated protein kinase 1Homo sapiens (human)
signal transductionRho-associated protein kinase 1Homo sapiens (human)
canonical NF-kappaB signal transductionRho-associated protein kinase 1Homo sapiens (human)
Rho protein signal transductionRho-associated protein kinase 1Homo sapiens (human)
positive regulation of autophagyRho-associated protein kinase 1Homo sapiens (human)
positive regulation of cardiac muscle hypertrophyRho-associated protein kinase 1Homo sapiens (human)
positive regulation of gene expressionRho-associated protein kinase 1Homo sapiens (human)
positive regulation of phosphatase activityRho-associated protein kinase 1Homo sapiens (human)
negative regulation of angiogenesisRho-associated protein kinase 1Homo sapiens (human)
peptidyl-serine phosphorylationRho-associated protein kinase 1Homo sapiens (human)
membrane to membrane dockingRho-associated protein kinase 1Homo sapiens (human)
actin cytoskeleton organizationRho-associated protein kinase 1Homo sapiens (human)
regulation of cell adhesionRho-associated protein kinase 1Homo sapiens (human)
regulation of cell migrationRho-associated protein kinase 1Homo sapiens (human)
cortical actin cytoskeleton organizationRho-associated protein kinase 1Homo sapiens (human)
neuron projection developmentRho-associated protein kinase 1Homo sapiens (human)
bleb assemblyRho-associated protein kinase 1Homo sapiens (human)
negative regulation of protein bindingRho-associated protein kinase 1Homo sapiens (human)
regulation of actin cytoskeleton organizationRho-associated protein kinase 1Homo sapiens (human)
positive regulation of dephosphorylationRho-associated protein kinase 1Homo sapiens (human)
negative regulation of myosin-light-chain-phosphatase activityRho-associated protein kinase 1Homo sapiens (human)
negative regulation of phosphorylationRho-associated protein kinase 1Homo sapiens (human)
positive regulation of MAPK cascadeRho-associated protein kinase 1Homo sapiens (human)
regulation of keratinocyte differentiationRho-associated protein kinase 1Homo sapiens (human)
regulation of neuron differentiationRho-associated protein kinase 1Homo sapiens (human)
leukocyte migrationRho-associated protein kinase 1Homo sapiens (human)
leukocyte tethering or rollingRho-associated protein kinase 1Homo sapiens (human)
negative regulation of membrane protein ectodomain proteolysisRho-associated protein kinase 1Homo sapiens (human)
myoblast migrationRho-associated protein kinase 1Homo sapiens (human)
regulation of stress fiber assemblyRho-associated protein kinase 1Homo sapiens (human)
regulation of focal adhesion assemblyRho-associated protein kinase 1Homo sapiens (human)
positive regulation of focal adhesion assemblyRho-associated protein kinase 1Homo sapiens (human)
mRNA destabilizationRho-associated protein kinase 1Homo sapiens (human)
negative regulation of biomineral tissue developmentRho-associated protein kinase 1Homo sapiens (human)
regulation of microtubule cytoskeleton organizationRho-associated protein kinase 1Homo sapiens (human)
response to transforming growth factor betaRho-associated protein kinase 1Homo sapiens (human)
protein localization to plasma membraneRho-associated protein kinase 1Homo sapiens (human)
regulation of synapse maturationRho-associated protein kinase 1Homo sapiens (human)
podocyte cell migrationRho-associated protein kinase 1Homo sapiens (human)
motor neuron apoptotic processRho-associated protein kinase 1Homo sapiens (human)
blood vessel diameter maintenanceRho-associated protein kinase 1Homo sapiens (human)
regulation of angiotensin-activated signaling pathwayRho-associated protein kinase 1Homo sapiens (human)
neuron projection arborizationRho-associated protein kinase 1Homo sapiens (human)
positive regulation of amyloid-beta clearanceRho-associated protein kinase 1Homo sapiens (human)
regulation of synaptic vesicle endocytosisRho-associated protein kinase 1Homo sapiens (human)
negative regulation of amyloid-beta formationRho-associated protein kinase 1Homo sapiens (human)
negative regulation of amyloid precursor protein catabolic processRho-associated protein kinase 1Homo sapiens (human)
regulation of establishment of endothelial barrierRho-associated protein kinase 1Homo sapiens (human)
negative regulation of bicellular tight junction assemblyRho-associated protein kinase 1Homo sapiens (human)
positive regulation of connective tissue replacementRho-associated protein kinase 1Homo sapiens (human)
response to angiotensinRho-associated protein kinase 1Homo sapiens (human)
regulation of establishment of cell polarityRho-associated protein kinase 1Homo sapiens (human)
regulation of cell motilityRho-associated protein kinase 1Homo sapiens (human)
negative regulation of motor neuron apoptotic processRho-associated protein kinase 1Homo sapiens (human)
regulation of cell junction assemblyRho-associated protein kinase 1Homo sapiens (human)
mitotic cytokinesisRho-associated protein kinase 1Homo sapiens (human)
embryonic morphogenesisRho-associated protein kinase 1Homo sapiens (human)
peptidyl-threonine phosphorylationRho-associated protein kinase 1Homo sapiens (human)
actomyosin structure organizationRho-associated protein kinase 1Homo sapiens (human)
protein phosphorylationNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
protein autophosphorylationNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
nervous system developmentCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
regulation of neuron projection developmentCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
regulation of skeletal muscle adaptationCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
insulin secretionCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
cell differentiationCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
regulation of calcium ion transportCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
regulation of cell growthCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of the force of heart contractionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of membrane depolarizationCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of transcription by RNA polymerase IICalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
protein phosphorylationCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of heart contractionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
positive regulation of cardiac muscle hypertrophyCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of cell communication by electrical couplingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of release of sequestered calcium ion into cytosol by sarcoplasmic reticulumCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of cardiac muscle contraction by regulation of the release of sequestered calcium ionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
peptidyl-serine phosphorylationCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
peptidyl-threonine phosphorylationCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
endoplasmic reticulum calcium ion homeostasisCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
protein autophosphorylationCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
relaxation of cardiac muscleCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of ryanodine-sensitive calcium-release channel activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of cellular localizationCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
cellular response to calcium ionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
cardiac muscle cell contractionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of heart rate by cardiac conductionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of cardiac muscle cell action potentialCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of cardiac muscle cell action potential involved in regulation of contractionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of cell communication by electrical coupling involved in cardiac conductionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of relaxation of cardiac muscleCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
negative regulation of sodium ion transmembrane transportCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
regulation of calcium ion transmembrane transport via high voltage-gated calcium channelCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
negative regulation of sodium ion transmembrane transporter activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
peptidyl-tyrosine phosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
chromatin remodelingDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
regulation of transcription by RNA polymerase IIDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein phosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
nervous system developmentDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
circadian rhythmDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
peptidyl-serine phosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
peptidyl-threonine phosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
peptidyl-tyrosine phosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
negative regulation of microtubule polymerizationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
positive regulation of RNA splicingDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
amyloid-beta formationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
peptidyl-serine autophosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
peptidyl-tyrosine autophosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
negative regulation of DNA damage response, signal transduction by p53 class mediatorDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein autophosphorylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
negative regulation of mRNA splicing, via spliceosomeDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
negative regulation of DNA methylation-dependent heterochromatin formationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
positive regulation of protein deacetylationDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
positive regulation of DNA-templated transcriptionDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIActivin receptor type-2BHomo sapiens (human)
gastrulation with mouth forming secondActivin receptor type-2BHomo sapiens (human)
kidney developmentActivin receptor type-2BHomo sapiens (human)
lymphangiogenesisActivin receptor type-2BHomo sapiens (human)
blood vessel remodelingActivin receptor type-2BHomo sapiens (human)
regulation of DNA-templated transcriptionActivin receptor type-2BHomo sapiens (human)
signal transductionActivin receptor type-2BHomo sapiens (human)
cell surface receptor protein serine/threonine kinase signaling pathwayActivin receptor type-2BHomo sapiens (human)
determination of left/right symmetryActivin receptor type-2BHomo sapiens (human)
mesoderm developmentActivin receptor type-2BHomo sapiens (human)
heart developmentActivin receptor type-2BHomo sapiens (human)
response to glucoseActivin receptor type-2BHomo sapiens (human)
post-embryonic developmentActivin receptor type-2BHomo sapiens (human)
anterior/posterior pattern specificationActivin receptor type-2BHomo sapiens (human)
insulin secretionActivin receptor type-2BHomo sapiens (human)
lung developmentActivin receptor type-2BHomo sapiens (human)
positive regulation of bone mineralizationActivin receptor type-2BHomo sapiens (human)
BMP signaling pathwayActivin receptor type-2BHomo sapiens (human)
pancreas developmentActivin receptor type-2BHomo sapiens (human)
activin receptor signaling pathwayActivin receptor type-2BHomo sapiens (human)
positive regulation of activin receptor signaling pathwayActivin receptor type-2BHomo sapiens (human)
organ growthActivin receptor type-2BHomo sapiens (human)
odontogenesis of dentin-containing toothActivin receptor type-2BHomo sapiens (human)
positive regulation of osteoblast differentiationActivin receptor type-2BHomo sapiens (human)
embryonic foregut morphogenesisActivin receptor type-2BHomo sapiens (human)
skeletal system morphogenesisActivin receptor type-2BHomo sapiens (human)
roof of mouth developmentActivin receptor type-2BHomo sapiens (human)
lymphatic endothelial cell differentiationActivin receptor type-2BHomo sapiens (human)
artery developmentActivin receptor type-2BHomo sapiens (human)
venous blood vessel developmentActivin receptor type-2BHomo sapiens (human)
retina vasculature development in camera-type eyeActivin receptor type-2BHomo sapiens (human)
negative regulation of cold-induced thermogenesisActivin receptor type-2BHomo sapiens (human)
cellular response to growth factor stimulusActivin receptor type-2BHomo sapiens (human)
protein phosphorylationActivin receptor type-2BHomo sapiens (human)
outflow tract septum morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
atrioventricular valve morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
cardiac muscle tissue developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
pharyngeal arch artery morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of gene expressionBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of SMAD protein signal transductionBone morphogenetic protein receptor type-2Homo sapiens (human)
osteoblast differentiationBone morphogenetic protein receptor type-2Homo sapiens (human)
mesoderm formationBone morphogenetic protein receptor type-2Homo sapiens (human)
maternal placenta developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
endothelial cell proliferationBone morphogenetic protein receptor type-2Homo sapiens (human)
lymphangiogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
blood vessel remodelingBone morphogenetic protein receptor type-2Homo sapiens (human)
chondrocyte developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of systemic arterial blood pressureBone morphogenetic protein receptor type-2Homo sapiens (human)
outflow tract morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
aortic valve developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
pulmonary valve developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
mitral valve morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
tricuspid valve morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
endocardial cushion developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of cell proliferation involved in heart valve morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
cell surface receptor protein serine/threonine kinase signaling pathwayBone morphogenetic protein receptor type-2Homo sapiens (human)
cellular response to starvationBone morphogenetic protein receptor type-2Homo sapiens (human)
anterior/posterior pattern specificationBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of epithelial cell migrationBone morphogenetic protein receptor type-2Homo sapiens (human)
regulation of lung blood pressureBone morphogenetic protein receptor type-2Homo sapiens (human)
proteoglycan biosynthetic processBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of cell growthBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of bone mineralizationBone morphogenetic protein receptor type-2Homo sapiens (human)
BMP signaling pathwayBone morphogenetic protein receptor type-2Homo sapiens (human)
activin receptor signaling pathwayBone morphogenetic protein receptor type-2Homo sapiens (human)
regulation of cell population proliferationBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of osteoblast differentiationBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of ossificationBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of vasoconstrictionBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIBone morphogenetic protein receptor type-2Homo sapiens (human)
lung alveolus developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of smooth muscle cell proliferationBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of axon extension involved in axon guidanceBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of muscle cell differentiationBone morphogenetic protein receptor type-2Homo sapiens (human)
limb developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
endochondral bone morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of SMAD protein signal transductionBone morphogenetic protein receptor type-2Homo sapiens (human)
ventricular septum morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
atrial septum morphogenesisBone morphogenetic protein receptor type-2Homo sapiens (human)
lung vasculature developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
lymphatic endothelial cell differentiationBone morphogenetic protein receptor type-2Homo sapiens (human)
artery developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
venous blood vessel developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
positive regulation of cartilage developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
retina vasculature development in camera-type eyeBone morphogenetic protein receptor type-2Homo sapiens (human)
cellular response to BMP stimulusBone morphogenetic protein receptor type-2Homo sapiens (human)
endothelial cell apoptotic processBone morphogenetic protein receptor type-2Homo sapiens (human)
negative regulation of chondrocyte proliferationBone morphogenetic protein receptor type-2Homo sapiens (human)
semi-lunar valve developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
cellular response to growth factor stimulusBone morphogenetic protein receptor type-2Homo sapiens (human)
blood vessel developmentBone morphogenetic protein receptor type-2Homo sapiens (human)
protein phosphorylationBone morphogenetic protein receptor type-2Homo sapiens (human)
protein phosphorylationProtein-tyrosine kinase 6Homo sapiens (human)
tyrosine phosphorylation of STAT proteinProtein-tyrosine kinase 6Homo sapiens (human)
positive regulation of neuron projection developmentProtein-tyrosine kinase 6Homo sapiens (human)
cell migrationProtein-tyrosine kinase 6Homo sapiens (human)
ERBB2 signaling pathwayProtein-tyrosine kinase 6Homo sapiens (human)
positive regulation of tyrosine phosphorylation of STAT proteinProtein-tyrosine kinase 6Homo sapiens (human)
positive regulation of DNA replicationProtein-tyrosine kinase 6Homo sapiens (human)
positive regulation of epidermal growth factor receptor signaling pathwayProtein-tyrosine kinase 6Homo sapiens (human)
positive regulation of cell cycleProtein-tyrosine kinase 6Homo sapiens (human)
negative regulation of growthProtein-tyrosine kinase 6Homo sapiens (human)
protein autophosphorylationProtein-tyrosine kinase 6Homo sapiens (human)
intestinal epithelial cell differentiationProtein-tyrosine kinase 6Homo sapiens (human)
negative regulation of protein tyrosine kinase activityProtein-tyrosine kinase 6Homo sapiens (human)
cellular response to retinoic acidProtein-tyrosine kinase 6Homo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayProtein-tyrosine kinase 6Homo sapiens (human)
innate immune responseProtein-tyrosine kinase 6Homo sapiens (human)
cell differentiationProtein-tyrosine kinase 6Homo sapiens (human)
positive regulation of receptor signaling pathway via JAK-STATProtein-tyrosine kinase 6Homo sapiens (human)
protein phosphorylationcGMP-dependent protein kinase 1 Homo sapiens (human)
neuron migrationcGMP-dependent protein kinase 1 Homo sapiens (human)
signal transductioncGMP-dependent protein kinase 1 Homo sapiens (human)
positive regulation of cytosolic calcium ion concentrationcGMP-dependent protein kinase 1 Homo sapiens (human)
spermatid developmentcGMP-dependent protein kinase 1 Homo sapiens (human)
negative regulation of inositol phosphate biosynthetic processcGMP-dependent protein kinase 1 Homo sapiens (human)
negative regulation of glutamate secretioncGMP-dependent protein kinase 1 Homo sapiens (human)
dendrite developmentcGMP-dependent protein kinase 1 Homo sapiens (human)
cGMP-mediated signalingcGMP-dependent protein kinase 1 Homo sapiens (human)
cerebellum developmentcGMP-dependent protein kinase 1 Homo sapiens (human)
actin cytoskeleton organizationcGMP-dependent protein kinase 1 Homo sapiens (human)
forebrain developmentcGMP-dependent protein kinase 1 Homo sapiens (human)
positive regulation of circadian rhythmcGMP-dependent protein kinase 1 Homo sapiens (human)
regulation of GTPase activitycGMP-dependent protein kinase 1 Homo sapiens (human)
collateral sproutingcGMP-dependent protein kinase 1 Homo sapiens (human)
relaxation of vascular associated smooth musclecGMP-dependent protein kinase 1 Homo sapiens (human)
cell growth involved in cardiac muscle cell developmentcGMP-dependent protein kinase 1 Homo sapiens (human)
negative regulation of platelet aggregationcGMP-dependent protein kinase 1 Homo sapiens (human)
negative regulation of vascular associated smooth muscle cell proliferationcGMP-dependent protein kinase 1 Homo sapiens (human)
negative regulation of vascular associated smooth muscle cell migrationcGMP-dependent protein kinase 1 Homo sapiens (human)
regulation of testosterone biosynthetic processcGMP-dependent protein kinase 1 Homo sapiens (human)
protein kinase A signalingcGMP-dependent protein kinase 1 Homo sapiens (human)
alternative mRNA splicing, via spliceosomeCyclin-dependent kinase 13Homo sapiens (human)
regulation of signal transductionCyclin-dependent kinase 13Homo sapiens (human)
hemopoiesisCyclin-dependent kinase 13Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IICyclin-dependent kinase 13Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICyclin-dependent kinase 13Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 13Homo sapiens (human)
negative regulation of stem cell differentiationCyclin-dependent kinase 13Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 13Homo sapiens (human)
immune responseInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damageInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
gene expressionInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
positive regulation of lipid storageInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
positive regulation of type I interferon productionInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
response to interferon-betaInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
regulation of protein-containing complex assemblyInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
mRNA stabilizationInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
positive regulation of DNA-binding transcription factor activityInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
defense response to virusInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
type I interferon-mediated signaling pathwayInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
positive regulation of type I interferon-mediated signaling pathwayInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
interleukin-17-mediated signaling pathwayInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
cellular response to virusInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
peptidyl-serine phosphorylationInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionProtein-tyrosine kinase 2-betaHomo sapiens (human)
MAPK cascadeProtein-tyrosine kinase 2-betaHomo sapiens (human)
oocyte maturationProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to hypoxiaProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of cell-matrix adhesionProtein-tyrosine kinase 2-betaHomo sapiens (human)
sprouting angiogenesisProtein-tyrosine kinase 2-betaHomo sapiens (human)
adaptive immune responseProtein-tyrosine kinase 2-betaHomo sapiens (human)
marginal zone B cell differentiationProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to ischemiaProtein-tyrosine kinase 2-betaHomo sapiens (human)
protein phosphorylationProtein-tyrosine kinase 2-betaHomo sapiens (human)
apoptotic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
cellular defense responseProtein-tyrosine kinase 2-betaHomo sapiens (human)
signal transductionProtein-tyrosine kinase 2-betaHomo sapiens (human)
cell surface receptor signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
signal complex assemblyProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationProtein-tyrosine kinase 2-betaHomo sapiens (human)
integrin-mediated signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of cell population proliferationProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of cell population proliferationProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of cell shapeProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to xenobiotic stimulusProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to mechanical stimulusProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to hormoneProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to glucoseProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of endothelial cell migrationProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of muscle cell apoptotic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of macrophage chemotaxisProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of neuron projection developmentProtein-tyrosine kinase 2-betaHomo sapiens (human)
glial cell proliferationProtein-tyrosine kinase 2-betaHomo sapiens (human)
peptidyl-tyrosine phosphorylationProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of cell adhesionProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of cell growthProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of cell migrationProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of bone mineralizationProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of actin filament polymerizationProtein-tyrosine kinase 2-betaHomo sapiens (human)
cortical cytoskeleton organizationProtein-tyrosine kinase 2-betaHomo sapiens (human)
neuron projection developmentProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of actin cytoskeleton organizationProtein-tyrosine kinase 2-betaHomo sapiens (human)
tumor necrosis factor-mediated signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to immobilization stressProtein-tyrosine kinase 2-betaHomo sapiens (human)
peptidyl-tyrosine autophosphorylationProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to cocaineProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to hydrogen peroxideProtein-tyrosine kinase 2-betaHomo sapiens (human)
activation of Janus kinase activityProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of apoptotic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
stress fiber assemblyProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to cation stressProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of potassium ion transportProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of neuron apoptotic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
blood vessel endothelial cell migrationProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of nitric oxide biosynthetic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
bone resorptionProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to ethanolProtein-tyrosine kinase 2-betaHomo sapiens (human)
negative regulation of myeloid cell differentiationProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of translationProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of angiogenesisProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of protein kinase activityProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of JNK cascadeProtein-tyrosine kinase 2-betaHomo sapiens (human)
protein autophosphorylationProtein-tyrosine kinase 2-betaHomo sapiens (human)
vascular endothelial growth factor receptor signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
focal adhesion assemblyProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of synaptic plasticityProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of release of sequestered calcium ion into cytosolProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to cAMPProtein-tyrosine kinase 2-betaHomo sapiens (human)
response to calcium ionProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicProtein-tyrosine kinase 2-betaHomo sapiens (human)
long-term synaptic potentiationProtein-tyrosine kinase 2-betaHomo sapiens (human)
long-term synaptic depressionProtein-tyrosine kinase 2-betaHomo sapiens (human)
protein-containing complex assemblyProtein-tyrosine kinase 2-betaHomo sapiens (human)
chemokine-mediated signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeProtein-tyrosine kinase 2-betaHomo sapiens (human)
cellular response to retinoic acidProtein-tyrosine kinase 2-betaHomo sapiens (human)
cellular response to fluid shear stressProtein-tyrosine kinase 2-betaHomo sapiens (human)
endothelin receptor signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of postsynaptic density assemblyProtein-tyrosine kinase 2-betaHomo sapiens (human)
postsynaptic modulation of chemical synaptic transmissionProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of ubiquitin-dependent protein catabolic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of ubiquitin-dependent protein catabolic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of reactive oxygen species metabolic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of B cell chemotaxisProtein-tyrosine kinase 2-betaHomo sapiens (human)
positive regulation of DNA biosynthetic processProtein-tyrosine kinase 2-betaHomo sapiens (human)
epidermal growth factor receptor signaling pathwayProtein-tyrosine kinase 2-betaHomo sapiens (human)
regulation of heart rateSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac conduction system developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac ventricle developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
brainstem developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
response to denervation involved in regulation of muscle adaptationSodium channel protein type 5 subunit alphaHomo sapiens (human)
telencephalon developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cerebellum developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
odontogenesis of dentin-containing toothSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of epithelial cell proliferationSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cellular response to calcium ionSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle cell action potential involved in contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of cardiac muscle cell contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
ventricular cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during Purkinje myocyte cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell to bundle of His cell communicationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of heart rate by cardiac conductionSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
G2/M transition of mitotic cell cycleMaternal embryonic leucine zipper kinaseHomo sapiens (human)
apoptotic processMaternal embryonic leucine zipper kinaseHomo sapiens (human)
cell population proliferationMaternal embryonic leucine zipper kinaseHomo sapiens (human)
intrinsic apoptotic signaling pathway in response to oxidative stressMaternal embryonic leucine zipper kinaseHomo sapiens (human)
hemopoiesisMaternal embryonic leucine zipper kinaseHomo sapiens (human)
positive regulation of apoptotic processMaternal embryonic leucine zipper kinaseHomo sapiens (human)
protein autophosphorylationMaternal embryonic leucine zipper kinaseHomo sapiens (human)
neural precursor cell proliferationMaternal embryonic leucine zipper kinaseHomo sapiens (human)
protein phosphorylationMaternal embryonic leucine zipper kinaseHomo sapiens (human)
mitotic sister chromatid segregationStructural maintenance of chromosomes protein 1AHomo sapiens (human)
DNA repairStructural maintenance of chromosomes protein 1AHomo sapiens (human)
sister chromatid cohesionStructural maintenance of chromosomes protein 1AHomo sapiens (human)
mitotic sister chromatid cohesionStructural maintenance of chromosomes protein 1AHomo sapiens (human)
response to radiationStructural maintenance of chromosomes protein 1AHomo sapiens (human)
establishment of mitotic sister chromatid cohesionStructural maintenance of chromosomes protein 1AHomo sapiens (human)
establishment of meiotic sister chromatid cohesionStructural maintenance of chromosomes protein 1AHomo sapiens (human)
somatic stem cell population maintenanceStructural maintenance of chromosomes protein 1AHomo sapiens (human)
cell divisionStructural maintenance of chromosomes protein 1AHomo sapiens (human)
meiotic cell cycleStructural maintenance of chromosomes protein 1AHomo sapiens (human)
response to DNA damage checkpoint signalingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
mitotic spindle assemblyStructural maintenance of chromosomes protein 1AHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
double-strand break repair via homologous recombinationChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
chromatin remodelingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
negative regulation of gene expressionChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
regulation of cell fate specificationChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
negative regulation of DNA-templated transcriptionChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
positive regulation of DNA-templated transcriptionChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
regulation of synapse assemblyChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
terminal button organizationChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
regulation of stem cell differentiationChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
very long-chain fatty acid metabolic processPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
generation of precursor metabolites and energyPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
lipid metabolic processPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
prostaglandin metabolic processPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
spermatogenesisPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
fatty acid catabolic processPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
fatty acid oxidationPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
fatty acid beta-oxidation using acyl-CoA oxidasePeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
hydrogen peroxide biosynthetic processPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
very long-chain fatty acid beta-oxidationPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
lipid homeostasisPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
positive regulation of protein phosphorylationEphrin type-A receptor 7Homo sapiens (human)
brain developmentEphrin type-A receptor 7Homo sapiens (human)
phosphorylationEphrin type-A receptor 7Homo sapiens (human)
regulation of cell-cell adhesionEphrin type-A receptor 7Homo sapiens (human)
retinal ganglion cell axon guidanceEphrin type-A receptor 7Homo sapiens (human)
regulation of protein autophosphorylationEphrin type-A receptor 7Homo sapiens (human)
regulation of cysteine-type endopeptidase activity involved in apoptotic processEphrin type-A receptor 7Homo sapiens (human)
positive regulation of neuron apoptotic processEphrin type-A receptor 7Homo sapiens (human)
ephrin receptor signaling pathwayEphrin type-A receptor 7Homo sapiens (human)
negative regulation of collateral sproutingEphrin type-A receptor 7Homo sapiens (human)
branching morphogenesis of a nerveEphrin type-A receptor 7Homo sapiens (human)
regulation of peptidyl-tyrosine phosphorylationEphrin type-A receptor 7Homo sapiens (human)
modulation of chemical synaptic transmissionEphrin type-A receptor 7Homo sapiens (human)
negative chemotaxisEphrin type-A receptor 7Homo sapiens (human)
neuron apoptotic processEphrin type-A receptor 7Homo sapiens (human)
negative regulation of synapse assemblyEphrin type-A receptor 7Homo sapiens (human)
regulation of ERK1 and ERK2 cascadeEphrin type-A receptor 7Homo sapiens (human)
nephric duct morphogenesisEphrin type-A receptor 7Homo sapiens (human)
regulation of postsynapse organizationEphrin type-A receptor 7Homo sapiens (human)
axon guidanceEphrin type-A receptor 7Homo sapiens (human)
protein phosphorylationEphrin type-A receptor 7Homo sapiens (human)
cholesterol biosynthetic processDelta(24)-sterol reductaseHomo sapiens (human)
cholesterol biosynthetic processDelta(24)-sterol reductaseHomo sapiens (human)
Ras protein signal transductionDelta(24)-sterol reductaseHomo sapiens (human)
protein localizationDelta(24)-sterol reductaseHomo sapiens (human)
negative regulation of cell population proliferationDelta(24)-sterol reductaseHomo sapiens (human)
response to hormoneDelta(24)-sterol reductaseHomo sapiens (human)
tissue developmentDelta(24)-sterol reductaseHomo sapiens (human)
male genitalia developmentDelta(24)-sterol reductaseHomo sapiens (human)
plasminogen activationDelta(24)-sterol reductaseHomo sapiens (human)
cholesterol biosynthetic process via desmosterolDelta(24)-sterol reductaseHomo sapiens (human)
cholesterol biosynthetic process via lathosterolDelta(24)-sterol reductaseHomo sapiens (human)
amyloid precursor protein catabolic processDelta(24)-sterol reductaseHomo sapiens (human)
skin developmentDelta(24)-sterol reductaseHomo sapiens (human)
membrane organizationDelta(24)-sterol reductaseHomo sapiens (human)
steroid metabolic processDelta(24)-sterol reductaseHomo sapiens (human)
protein phosphorylationRibosomal protein S6 kinase alpha-1Homo sapiens (human)
signal transductionRibosomal protein S6 kinase alpha-1Homo sapiens (human)
chemical synaptic transmissionRibosomal protein S6 kinase alpha-1Homo sapiens (human)
positive regulation of cell growthRibosomal protein S6 kinase alpha-1Homo sapiens (human)
negative regulation of TOR signalingRibosomal protein S6 kinase alpha-1Homo sapiens (human)
intracellular signal transductionRibosomal protein S6 kinase alpha-1Homo sapiens (human)
negative regulation of apoptotic processRibosomal protein S6 kinase alpha-1Homo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processRibosomal protein S6 kinase alpha-1Homo sapiens (human)
regulation of translation in response to stressRibosomal protein S6 kinase alpha-1Homo sapiens (human)
positive regulation of cell differentiationRibosomal protein S6 kinase alpha-1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIRibosomal protein S6 kinase alpha-1Homo sapiens (human)
hepatocyte proliferationRibosomal protein S6 kinase alpha-1Homo sapiens (human)
positive regulation of hepatic stellate cell activationRibosomal protein S6 kinase alpha-1Homo sapiens (human)
positive regulation of DNA-templated transcriptionRibosomal protein S6 kinase alpha-1Homo sapiens (human)
peptidyl-serine phosphorylationRibosomal protein S6 kinase alpha-1Homo sapiens (human)
positive regulation of protein phosphorylationDual specificity testis-specific protein kinase 1Homo sapiens (human)
spermatogenesisDual specificity testis-specific protein kinase 1Homo sapiens (human)
negative regulation of protein autophosphorylationDual specificity testis-specific protein kinase 1Homo sapiens (human)
regulation of protein localizationDual specificity testis-specific protein kinase 1Homo sapiens (human)
regulation of actin cytoskeleton organizationDual specificity testis-specific protein kinase 1Homo sapiens (human)
negative regulation of phosphorylationDual specificity testis-specific protein kinase 1Homo sapiens (human)
positive regulation of stress fiber assemblyDual specificity testis-specific protein kinase 1Homo sapiens (human)
establishment of vesicle localizationDual specificity testis-specific protein kinase 1Homo sapiens (human)
negative regulation of protein serine/threonine kinase activityDual specificity testis-specific protein kinase 1Homo sapiens (human)
podocyte cell migrationDual specificity testis-specific protein kinase 1Homo sapiens (human)
positive regulation of substrate adhesion-dependent cell spreadingDual specificity testis-specific protein kinase 1Homo sapiens (human)
positive regulation of protein localization to nucleusDual specificity testis-specific protein kinase 1Homo sapiens (human)
negative regulation of cilium assemblyDual specificity testis-specific protein kinase 1Homo sapiens (human)
actin cytoskeleton organizationDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein phosphorylationMyosin light chain kinase, smooth muscleHomo sapiens (human)
smooth muscle contractionMyosin light chain kinase, smooth muscleHomo sapiens (human)
tonic smooth muscle contractionMyosin light chain kinase, smooth muscleHomo sapiens (human)
positive regulation of cell migrationMyosin light chain kinase, smooth muscleHomo sapiens (human)
bleb assemblyMyosin light chain kinase, smooth muscleHomo sapiens (human)
positive regulation of calcium ion transportMyosin light chain kinase, smooth muscleHomo sapiens (human)
aorta smooth muscle tissue morphogenesisMyosin light chain kinase, smooth muscleHomo sapiens (human)
cellular hypotonic responseMyosin light chain kinase, smooth muscleHomo sapiens (human)
positive regulation of wound healingMyosin light chain kinase, smooth muscleHomo sapiens (human)
positive regulation of erythrocyte differentiationMitogen-activated protein kinase 11Homo sapiens (human)
osteoblast differentiationMitogen-activated protein kinase 11Homo sapiens (human)
positive regulation of gene expressionMitogen-activated protein kinase 11Homo sapiens (human)
stress-activated protein kinase signaling cascadeMitogen-activated protein kinase 11Homo sapiens (human)
positive regulation of interleukin-12 productionMitogen-activated protein kinase 11Homo sapiens (human)
p38MAPK cascadeMitogen-activated protein kinase 11Homo sapiens (human)
positive regulation of muscle cell differentiationMitogen-activated protein kinase 11Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase 11Homo sapiens (human)
cardiac muscle cell proliferationMitogen-activated protein kinase 11Homo sapiens (human)
negative regulation of cardiac muscle cell proliferationMitogen-activated protein kinase 11Homo sapiens (human)
bone developmentMitogen-activated protein kinase 11Homo sapiens (human)
cellular response to interleukin-1Mitogen-activated protein kinase 11Homo sapiens (human)
cellular response to UV-BMitogen-activated protein kinase 11Homo sapiens (human)
cellular senescenceMitogen-activated protein kinase 11Homo sapiens (human)
cellular response to virusMitogen-activated protein kinase 11Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase 11Homo sapiens (human)
G1 to G0 transitionSerine/threonine-protein kinase STK11Homo sapiens (human)
regulation of cell growthSerine/threonine-protein kinase STK11Homo sapiens (human)
tissue homeostasisSerine/threonine-protein kinase STK11Homo sapiens (human)
vasculature developmentSerine/threonine-protein kinase STK11Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase STK11Homo sapiens (human)
protein dephosphorylationSerine/threonine-protein kinase STK11Homo sapiens (human)
autophagySerine/threonine-protein kinase STK11Homo sapiens (human)
DNA damage responseSerine/threonine-protein kinase STK11Homo sapiens (human)
spermatogenesisSerine/threonine-protein kinase STK11Homo sapiens (human)
axonogenesisSerine/threonine-protein kinase STK11Homo sapiens (human)
negative regulation of cell population proliferationSerine/threonine-protein kinase STK11Homo sapiens (human)
response to ionizing radiationSerine/threonine-protein kinase STK11Homo sapiens (human)
positive regulation of autophagySerine/threonine-protein kinase STK11Homo sapiens (human)
response to activitySerine/threonine-protein kinase STK11Homo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase STK11Homo sapiens (human)
establishment of cell polaritySerine/threonine-protein kinase STK11Homo sapiens (human)
negative regulation of cell growthSerine/threonine-protein kinase STK11Homo sapiens (human)
positive regulation of transforming growth factor beta receptor signaling pathwaySerine/threonine-protein kinase STK11Homo sapiens (human)
activation of protein kinase activitySerine/threonine-protein kinase STK11Homo sapiens (human)
response to glucagonSerine/threonine-protein kinase STK11Homo sapiens (human)
response to lipidSerine/threonine-protein kinase STK11Homo sapiens (human)
protein localization to nucleusSerine/threonine-protein kinase STK11Homo sapiens (human)
glucose homeostasisSerine/threonine-protein kinase STK11Homo sapiens (human)
anoikisSerine/threonine-protein kinase STK11Homo sapiens (human)
positive thymic T cell selectionSerine/threonine-protein kinase STK11Homo sapiens (human)
positive regulation of gluconeogenesisSerine/threonine-protein kinase STK11Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase STK11Homo sapiens (human)
regulation of dendrite morphogenesisSerine/threonine-protein kinase STK11Homo sapiens (human)
positive regulation of axonogenesisSerine/threonine-protein kinase STK11Homo sapiens (human)
T cell receptor signaling pathwaySerine/threonine-protein kinase STK11Homo sapiens (human)
Golgi localizationSerine/threonine-protein kinase STK11Homo sapiens (human)
regulation of cell cycleSerine/threonine-protein kinase STK11Homo sapiens (human)
regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionSerine/threonine-protein kinase STK11Homo sapiens (human)
epithelial cell proliferation involved in prostate gland developmentSerine/threonine-protein kinase STK11Homo sapiens (human)
negative regulation of epithelial cell proliferation involved in prostate gland developmentSerine/threonine-protein kinase STK11Homo sapiens (human)
cellular response to UV-BSerine/threonine-protein kinase STK11Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorSerine/threonine-protein kinase STK11Homo sapiens (human)
negative regulation of canonical Wnt signaling pathwaySerine/threonine-protein kinase STK11Homo sapiens (human)
response to thyroid hormoneSerine/threonine-protein kinase STK11Homo sapiens (human)
dendrite extensionSerine/threonine-protein kinase STK11Homo sapiens (human)
negative regulation of cold-induced thermogenesisSerine/threonine-protein kinase STK11Homo sapiens (human)
positive regulation of protein localization to nucleusSerine/threonine-protein kinase STK11Homo sapiens (human)
positive regulation of vesicle transport along microtubuleSerine/threonine-protein kinase STK11Homo sapiens (human)
regulation of signal transduction by p53 class mediatorSerine/threonine-protein kinase STK11Homo sapiens (human)
negative regulation of TORC1 signalingSerine/threonine-protein kinase STK11Homo sapiens (human)
signal transductionSerine/threonine-protein kinase STK11Homo sapiens (human)
regulation of Wnt signaling pathwaySerine/threonine-protein kinase STK11Homo sapiens (human)
B cell homeostasisSerine/threonine-protein kinase N1Homo sapiens (human)
B cell apoptotic processSerine/threonine-protein kinase N1Homo sapiens (human)
negative regulation of protein phosphorylationSerine/threonine-protein kinase N1Homo sapiens (human)
regulation of germinal center formationSerine/threonine-protein kinase N1Homo sapiens (human)
regulation of immunoglobulin productionSerine/threonine-protein kinase N1Homo sapiens (human)
renal system processSerine/threonine-protein kinase N1Homo sapiens (human)
chromatin remodelingSerine/threonine-protein kinase N1Homo sapiens (human)
regulation of transcription by RNA polymerase IISerine/threonine-protein kinase N1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase N1Homo sapiens (human)
hyperosmotic responseSerine/threonine-protein kinase N1Homo sapiens (human)
signal transductionSerine/threonine-protein kinase N1Homo sapiens (human)
epithelial cell migrationSerine/threonine-protein kinase N1Homo sapiens (human)
negative regulation of B cell proliferationSerine/threonine-protein kinase N1Homo sapiens (human)
post-translational protein modificationSerine/threonine-protein kinase N1Homo sapiens (human)
positive regulation of DNA-templated transcriptionSerine/threonine-protein kinase N1Homo sapiens (human)
spleen developmentSerine/threonine-protein kinase N1Homo sapiens (human)
regulation of androgen receptor signaling pathwaySerine/threonine-protein kinase N1Homo sapiens (human)
regulation of cell motilitySerine/threonine-protein kinase N1Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase N1Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase N1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase N2Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase N2Homo sapiens (human)
cell adhesionSerine/threonine-protein kinase N2Homo sapiens (human)
signal transductionSerine/threonine-protein kinase N2Homo sapiens (human)
epithelial cell migrationSerine/threonine-protein kinase N2Homo sapiens (human)
cell projection organizationSerine/threonine-protein kinase N2Homo sapiens (human)
positive regulation of cytokinesisSerine/threonine-protein kinase N2Homo sapiens (human)
apical junction assemblySerine/threonine-protein kinase N2Homo sapiens (human)
positive regulation of viral genome replicationSerine/threonine-protein kinase N2Homo sapiens (human)
positive regulation of mitotic cell cycleSerine/threonine-protein kinase N2Homo sapiens (human)
cell divisionSerine/threonine-protein kinase N2Homo sapiens (human)
regulation of cell motilitySerine/threonine-protein kinase N2Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase N2Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase N2Homo sapiens (human)
positive regulation of blood vessel endothelial cell migrationMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to lipopolysaccharideMitogen-activated protein kinase 14Homo sapiens (human)
DNA damage checkpoint signalingMitogen-activated protein kinase 14Homo sapiens (human)
cell morphogenesisMitogen-activated protein kinase 14Homo sapiens (human)
cartilage condensationMitogen-activated protein kinase 14Homo sapiens (human)
angiogenesisMitogen-activated protein kinase 14Homo sapiens (human)
osteoblast differentiationMitogen-activated protein kinase 14Homo sapiens (human)
placenta developmentMitogen-activated protein kinase 14Homo sapiens (human)
response to dietary excessMitogen-activated protein kinase 14Homo sapiens (human)
chondrocyte differentiationMitogen-activated protein kinase 14Homo sapiens (human)
negative regulation of inflammatory response to antigenic stimulusMitogen-activated protein kinase 14Homo sapiens (human)
glucose metabolic processMitogen-activated protein kinase 14Homo sapiens (human)
regulation of transcription by RNA polymerase IIMitogen-activated protein kinase 14Homo sapiens (human)
transcription by RNA polymerase IIMitogen-activated protein kinase 14Homo sapiens (human)
apoptotic processMitogen-activated protein kinase 14Homo sapiens (human)
chemotaxisMitogen-activated protein kinase 14Homo sapiens (human)
signal transductionMitogen-activated protein kinase 14Homo sapiens (human)
cell surface receptor signaling pathwayMitogen-activated protein kinase 14Homo sapiens (human)
cell surface receptor protein serine/threonine kinase signaling pathwayMitogen-activated protein kinase 14Homo sapiens (human)
skeletal muscle tissue developmentMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of gene expressionMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of myotube differentiationMitogen-activated protein kinase 14Homo sapiens (human)
peptidyl-serine phosphorylationMitogen-activated protein kinase 14Homo sapiens (human)
fatty acid oxidationMitogen-activated protein kinase 14Homo sapiens (human)
platelet activationMitogen-activated protein kinase 14Homo sapiens (human)
regulation of ossificationMitogen-activated protein kinase 14Homo sapiens (human)
osteoclast differentiationMitogen-activated protein kinase 14Homo sapiens (human)
stress-activated protein kinase signaling cascadeMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of cyclase activityMitogen-activated protein kinase 14Homo sapiens (human)
lipopolysaccharide-mediated signaling pathwayMitogen-activated protein kinase 14Homo sapiens (human)
response to muramyl dipeptideMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of interleukin-12 productionMitogen-activated protein kinase 14Homo sapiens (human)
response to insulinMitogen-activated protein kinase 14Homo sapiens (human)
negative regulation of hippo signalingMitogen-activated protein kinase 14Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to vascular endothelial growth factor stimulusMitogen-activated protein kinase 14Homo sapiens (human)
response to muscle stretchMitogen-activated protein kinase 14Homo sapiens (human)
p38MAPK cascadeMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of protein import into nucleusMitogen-activated protein kinase 14Homo sapiens (human)
signal transduction in response to DNA damageMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of erythrocyte differentiationMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of myoblast differentiationMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIMitogen-activated protein kinase 14Homo sapiens (human)
glucose importMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of glucose importMitogen-activated protein kinase 14Homo sapiens (human)
vascular endothelial growth factor receptor signaling pathwayMitogen-activated protein kinase 14Homo sapiens (human)
stem cell differentiationMitogen-activated protein kinase 14Homo sapiens (human)
striated muscle cell differentiationMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of muscle cell differentiationMitogen-activated protein kinase 14Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of cardiac muscle cell proliferationMitogen-activated protein kinase 14Homo sapiens (human)
bone developmentMitogen-activated protein kinase 14Homo sapiens (human)
3'-UTR-mediated mRNA stabilizationMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to lipoteichoic acidMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to tumor necrosis factorMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to ionizing radiationMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to UV-BMitogen-activated protein kinase 14Homo sapiens (human)
negative regulation of canonical Wnt signaling pathwayMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of brown fat cell differentiationMitogen-activated protein kinase 14Homo sapiens (human)
cellular senescenceMitogen-activated protein kinase 14Homo sapiens (human)
stress-induced premature senescenceMitogen-activated protein kinase 14Homo sapiens (human)
cellular response to virusMitogen-activated protein kinase 14Homo sapiens (human)
regulation of synaptic membrane adhesionMitogen-activated protein kinase 14Homo sapiens (human)
regulation of cytokine production involved in inflammatory responseMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of myoblast fusionMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processMitogen-activated protein kinase 14Homo sapiens (human)
positive regulation of myeloid dendritic cell cytokine productionCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
adaptive immune responseCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
protein phosphorylationCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
inflammatory responseCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
signal transductionCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
long-term memoryCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
regulation of T cell differentiation in thymusCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
myeloid dendritic cell differentiationCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
regulation of osteoclast differentiationCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
positive regulation of DNA-templated transcriptionCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
protein autophosphorylationCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
peptidyl-serine phosphorylationCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
intracellular signal transductionCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
microtubule-based processMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
positive regulation of JUN kinase activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
positive regulation of neuron apoptotic processMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
cell cycle G1/S phase transitionMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
positive regulation of JNK cascadeMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
protein autophosphorylationMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
positive regulation of apoptotic processMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
ossificationDiscoidin domain-containing receptor 2Homo sapiens (human)
endochondral bone growthDiscoidin domain-containing receptor 2Homo sapiens (human)
cell adhesionDiscoidin domain-containing receptor 2Homo sapiens (human)
signal transductionDiscoidin domain-containing receptor 2Homo sapiens (human)
regulation of extracellular matrix disassemblyDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of fibroblast migrationDiscoidin domain-containing receptor 2Homo sapiens (human)
peptidyl-tyrosine phosphorylationDiscoidin domain-containing receptor 2Homo sapiens (human)
collagen fibril organizationDiscoidin domain-containing receptor 2Homo sapiens (human)
regulation of bone mineralizationDiscoidin domain-containing receptor 2Homo sapiens (human)
biomineral tissue developmentDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of collagen biosynthetic processDiscoidin domain-containing receptor 2Homo sapiens (human)
regulation of tissue remodelingDiscoidin domain-containing receptor 2Homo sapiens (human)
chondrocyte proliferationDiscoidin domain-containing receptor 2Homo sapiens (human)
response to muscle stretchDiscoidin domain-containing receptor 2Homo sapiens (human)
collagen-activated tyrosine kinase receptor signaling pathwayDiscoidin domain-containing receptor 2Homo sapiens (human)
negative regulation of apoptotic processDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of osteoblast differentiationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of protein kinase activityDiscoidin domain-containing receptor 2Homo sapiens (human)
protein autophosphorylationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of fibroblast proliferationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of DNA-binding transcription factor activityDiscoidin domain-containing receptor 2Homo sapiens (human)
cellular response to hypoxiaDiscoidin domain-containing receptor 2Homo sapiens (human)
cellular response to transforming growth factor beta stimulusDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of extracellular matrix disassemblyDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of wound healingDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of G1/S transition of mitotic cell cycleDiscoidin domain-containing receptor 2Homo sapiens (human)
negative regulation of hydrogen peroxide-mediated programmed cell deathDiscoidin domain-containing receptor 2Homo sapiens (human)
cellular response to angiotensinDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell migrationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of hepatic stellate cell proliferationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of hepatic stellate cell activationDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of neuron projection developmentDiscoidin domain-containing receptor 2Homo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionDiscoidin domain-containing receptor 2Homo sapiens (human)
multicellular organism developmentDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of kinase activityDiscoidin domain-containing receptor 2Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeDiscoidin domain-containing receptor 2Homo sapiens (human)
protein phosphorylationAP2-associated protein kinase 1Homo sapiens (human)
regulation of protein localizationAP2-associated protein kinase 1Homo sapiens (human)
positive regulation of Notch signaling pathwayAP2-associated protein kinase 1Homo sapiens (human)
protein stabilizationAP2-associated protein kinase 1Homo sapiens (human)
membrane organizationAP2-associated protein kinase 1Homo sapiens (human)
presynaptic endocytosisAP2-associated protein kinase 1Homo sapiens (human)
regulation of clathrin-dependent endocytosisAP2-associated protein kinase 1Homo sapiens (human)
regulation of vascular permeability involved in acute inflammatory responseMyosin light chain kinase 3Homo sapiens (human)
protein phosphorylationMyosin light chain kinase 3Homo sapiens (human)
sarcomere organizationMyosin light chain kinase 3Homo sapiens (human)
sarcomerogenesisMyosin light chain kinase 3Homo sapiens (human)
cardiac myofibril assemblyMyosin light chain kinase 3Homo sapiens (human)
positive regulation of sarcomere organizationMyosin light chain kinase 3Homo sapiens (human)
cellular response to interleukin-1Myosin light chain kinase 3Homo sapiens (human)
biological_processPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
cellular response to heatPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
protein stabilizationPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
protein foldingPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
cell migrationSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
actin cytoskeleton organizationSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
actomyosin structure organizationSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
actin cytoskeleton organizationSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
actomyosin structure organizationSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
fatty acid beta-oxidationAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
fatty acid beta-oxidation using acyl-CoA dehydrogenaseAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase N3Homo sapiens (human)
signal transductionSerine/threonine-protein kinase N3Homo sapiens (human)
epithelial cell migrationSerine/threonine-protein kinase N3Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase N3Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase N3Homo sapiens (human)
autophagySerine/threonine-protein kinase ULK3Homo sapiens (human)
smoothened signaling pathwaySerine/threonine-protein kinase ULK3Homo sapiens (human)
negative regulation of smoothened signaling pathwaySerine/threonine-protein kinase ULK3Homo sapiens (human)
positive regulation of smoothened signaling pathwaySerine/threonine-protein kinase ULK3Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase ULK3Homo sapiens (human)
fibroblast activationSerine/threonine-protein kinase ULK3Homo sapiens (human)
cellular senescenceSerine/threonine-protein kinase ULK3Homo sapiens (human)
reticulophagySerine/threonine-protein kinase ULK3Homo sapiens (human)
piecemeal microautophagy of the nucleusSerine/threonine-protein kinase ULK3Homo sapiens (human)
response to starvationSerine/threonine-protein kinase ULK3Homo sapiens (human)
autophagosome assemblySerine/threonine-protein kinase ULK3Homo sapiens (human)
autophagy of mitochondrionSerine/threonine-protein kinase ULK3Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase ULK3Homo sapiens (human)
regulation of autophagySerine/threonine-protein kinase ULK3Homo sapiens (human)
fatty acid beta-oxidationAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
fatty acid beta-oxidation using acyl-CoA dehydrogenaseAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
mRNA processingSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
rRNA catabolic processSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
apoptotic chromosome condensationSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
response to endoplasmic reticulum stressSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
negative regulation of DNA-templated transcriptionSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
IRE1-mediated unfolded protein responseSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
autophagy of mitochondrionSerine/threonine-protein kinase MARK2Homo sapiens (human)
neuron migrationSerine/threonine-protein kinase MARK2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase MARK2Homo sapiens (human)
positive regulation of neuron projection developmentSerine/threonine-protein kinase MARK2Homo sapiens (human)
Wnt signaling pathwaySerine/threonine-protein kinase MARK2Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase MARK2Homo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase MARK2Homo sapiens (human)
establishment of cell polaritySerine/threonine-protein kinase MARK2Homo sapiens (human)
activation of protein kinase activitySerine/threonine-protein kinase MARK2Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase MARK2Homo sapiens (human)
establishment or maintenance of epithelial cell apical/basal polaritySerine/threonine-protein kinase MARK2Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase MARK2Homo sapiens (human)
regulation of axonogenesisSerine/threonine-protein kinase MARK2Homo sapiens (human)
regulation of cytoskeleton organizationSerine/threonine-protein kinase MARK2Homo sapiens (human)
mitochondrion localizationSerine/threonine-protein kinase MARK2Homo sapiens (human)
axon developmentSerine/threonine-protein kinase MARK2Homo sapiens (human)
regulation of microtubule cytoskeleton organizationSerine/threonine-protein kinase MARK2Homo sapiens (human)
establishment or maintenance of cell polarity regulating cell shapeSerine/threonine-protein kinase MARK2Homo sapiens (human)
regulation of microtubule bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
microtubule cytoskeleton organizationSerine/threonine-protein kinase MARK2Homo sapiens (human)
central nervous system developmentATP-dependent RNA helicase DHX30Homo sapiens (human)
DNA duplex unwindingATP-dependent RNA helicase DHX30Homo sapiens (human)
mitochondrial large ribosomal subunit assemblyATP-dependent RNA helicase DHX30Homo sapiens (human)
microtubule cytoskeleton organizationSerine/threonine-protein kinase TAO1Homo sapiens (human)
DNA repairSerine/threonine-protein kinase TAO1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase TAO1Homo sapiens (human)
DNA damage responseSerine/threonine-protein kinase TAO1Homo sapiens (human)
negative regulation of microtubule depolymerizationSerine/threonine-protein kinase TAO1Homo sapiens (human)
mitotic G2 DNA damage checkpoint signalingSerine/threonine-protein kinase TAO1Homo sapiens (human)
phosphorylationSerine/threonine-protein kinase TAO1Homo sapiens (human)
central nervous system neuron developmentSerine/threonine-protein kinase TAO1Homo sapiens (human)
positive regulation of stress-activated MAPK cascadeSerine/threonine-protein kinase TAO1Homo sapiens (human)
regulation of actin cytoskeleton organizationSerine/threonine-protein kinase TAO1Homo sapiens (human)
positive regulation of JNK cascadeSerine/threonine-protein kinase TAO1Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase TAO1Homo sapiens (human)
regulation of cytoskeleton organizationSerine/threonine-protein kinase TAO1Homo sapiens (human)
neuron cellular homeostasisSerine/threonine-protein kinase TAO1Homo sapiens (human)
regulation of microtubule cytoskeleton organizationSerine/threonine-protein kinase TAO1Homo sapiens (human)
execution phase of apoptosisSerine/threonine-protein kinase TAO1Homo sapiens (human)
positive regulation of protein acetylationSerine/threonine-protein kinase TAO1Homo sapiens (human)
protein phosphorylationSTE20-related kinase adapter protein alphaHomo sapiens (human)
G1 to G0 transitionSTE20-related kinase adapter protein alphaHomo sapiens (human)
protein export from nucleusSTE20-related kinase adapter protein alphaHomo sapiens (human)
activation of protein kinase activitySTE20-related kinase adapter protein alphaHomo sapiens (human)
skeletal muscle contractionMyosin-14Homo sapiens (human)
mitochondrion organizationMyosin-14Homo sapiens (human)
skeletal muscle tissue developmentMyosin-14Homo sapiens (human)
sensory perception of soundMyosin-14Homo sapiens (human)
regulation of cell shapeMyosin-14Homo sapiens (human)
neuronal action potentialMyosin-14Homo sapiens (human)
actin filament-based movementMyosin-14Homo sapiens (human)
actomyosin structure organizationMyosin-14Homo sapiens (human)
vocalization behaviorMyosin-14Homo sapiens (human)
negative regulation of mitochondrial fusionAarF domain-containing protein kinase 1Homo sapiens (human)
positive regulation of cristae formationAarF domain-containing protein kinase 1Homo sapiens (human)
mitochondrion organizationAarF domain-containing protein kinase 1Homo sapiens (human)
lipid homeostasisAarF domain-containing protein kinase 1Homo sapiens (human)
protein localizationATP-dependent RNA helicase DDX42Homo sapiens (human)
regulation of apoptotic processATP-dependent RNA helicase DDX42Homo sapiens (human)
U2-type prespliceosome assemblyATP-dependent RNA helicase DDX42Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
response to UVMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
response to tumor necrosis factorMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
nuclear-transcribed mRNA catabolic process, nonsense-mediated decayEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
translational terminationEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
translationEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
regulation of cell growthRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of endothelial cell proliferationRegulatory-associated protein of mTORHomo sapiens (human)
DNA damage responseRegulatory-associated protein of mTORHomo sapiens (human)
regulation of cell sizeRegulatory-associated protein of mTORHomo sapiens (human)
response to xenobiotic stimulusRegulatory-associated protein of mTORHomo sapiens (human)
negative regulation of autophagyRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of cell growthRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to nutrient levelsRegulatory-associated protein of mTORHomo sapiens (human)
TOR signalingRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of TOR signalingRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationRegulatory-associated protein of mTORHomo sapiens (human)
social behaviorRegulatory-associated protein of mTORHomo sapiens (human)
TORC1 signalingRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of osteoclast differentiationRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of glycolytic processRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIIRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of lipid biosynthetic processRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to amino acid stimulusRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to L-leucineRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to glucose stimulusRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to hypoxiaRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to osmotic stressRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of G1/S transition of mitotic cell cycleRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of odontoblast differentiationRegulatory-associated protein of mTORHomo sapiens (human)
positive regulation of pentose-phosphate shuntRegulatory-associated protein of mTORHomo sapiens (human)
regulation of autophagyRegulatory-associated protein of mTORHomo sapiens (human)
cellular response to starvationRegulatory-associated protein of mTORHomo sapiens (human)
protein phosphorylationAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
ubiquinone biosynthetic processAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
phosphorylationAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
regulation of autophagyPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate biosynthetic processPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
positive regulation of autophagosome assemblyPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
phosphatidylinositol phosphate biosynthetic processPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
MAPK cascadeMitogen-activated protein kinase 15Homo sapiens (human)
regulation of COPII vesicle coatingMitogen-activated protein kinase 15Homo sapiens (human)
DNA damage responseMitogen-activated protein kinase 15Homo sapiens (human)
endoplasmic reticulum organizationMitogen-activated protein kinase 15Homo sapiens (human)
positive regulation of cell population proliferationMitogen-activated protein kinase 15Homo sapiens (human)
regulation of autophagyMitogen-activated protein kinase 15Homo sapiens (human)
negative regulation of cell migrationMitogen-activated protein kinase 15Homo sapiens (human)
positive regulation of telomere maintenance via telomeraseMitogen-activated protein kinase 15Homo sapiens (human)
protein autophosphorylationMitogen-activated protein kinase 15Homo sapiens (human)
positive regulation of telomerase activityMitogen-activated protein kinase 15Homo sapiens (human)
dopamine uptakeMitogen-activated protein kinase 15Homo sapiens (human)
regulation of cilium assemblyMitogen-activated protein kinase 15Homo sapiens (human)
positive regulation of telomere cappingMitogen-activated protein kinase 15Homo sapiens (human)
protein localization to ciliary transition zoneMitogen-activated protein kinase 15Homo sapiens (human)
positive regulation of metaphase/anaphase transition of meiosis IMitogen-activated protein kinase 15Homo sapiens (human)
positive regulation of spindle assemblyMitogen-activated protein kinase 15Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase 15Homo sapiens (human)
mitotic cell cycleSerine/threonine-protein kinase Nek9Homo sapiens (human)
regulation of mitotic cell cycleSerine/threonine-protein kinase Nek9Homo sapiens (human)
cell divisionSerine/threonine-protein kinase Nek9Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase Nek7Homo sapiens (human)
regulation of mitotic cell cycleSerine/threonine-protein kinase Nek7Homo sapiens (human)
positive regulation of telomere maintenance via telomeraseSerine/threonine-protein kinase Nek7Homo sapiens (human)
cellular response to potassium ionSerine/threonine-protein kinase Nek7Homo sapiens (human)
spindle assemblySerine/threonine-protein kinase Nek7Homo sapiens (human)
positive regulation of telomerase activitySerine/threonine-protein kinase Nek7Homo sapiens (human)
positive regulation of NLRP3 inflammasome complex assemblySerine/threonine-protein kinase Nek7Homo sapiens (human)
positive regulation of telomere cappingSerine/threonine-protein kinase Nek7Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase Nek7Homo sapiens (human)
spliceosomal complex assemblyATP-dependent RNA helicase DDX1Homo sapiens (human)
positive regulation of myeloid dendritic cell cytokine productionATP-dependent RNA helicase DDX1Homo sapiens (human)
double-strand break repairATP-dependent RNA helicase DDX1Homo sapiens (human)
tRNA splicing, via endonucleolytic cleavage and ligationATP-dependent RNA helicase DDX1Homo sapiens (human)
regulation of translational initiationATP-dependent RNA helicase DDX1Homo sapiens (human)
DNA duplex unwindingATP-dependent RNA helicase DDX1Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionATP-dependent RNA helicase DDX1Homo sapiens (human)
response to exogenous dsRNAATP-dependent RNA helicase DDX1Homo sapiens (human)
innate immune responseATP-dependent RNA helicase DDX1Homo sapiens (human)
defense response to virusATP-dependent RNA helicase DDX1Homo sapiens (human)
nucleic acid metabolic processATP-dependent RNA helicase DDX1Homo sapiens (human)
protein localization to cytoplasmic stress granuleATP-dependent RNA helicase DDX1Homo sapiens (human)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
cell population proliferationMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
peptidyl-serine phosphorylationMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
positive regulation of MAPK cascadeMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
protein autophosphorylationMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
cellular response to phorbol 13-acetate 12-myristateMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIAurora kinase BHomo sapiens (human)
mitotic cell cycleAurora kinase BHomo sapiens (human)
mitotic cytokinesisAurora kinase BHomo sapiens (human)
negative regulation of B cell apoptotic processAurora kinase BHomo sapiens (human)
protein phosphorylationAurora kinase BHomo sapiens (human)
spindle organizationAurora kinase BHomo sapiens (human)
attachment of spindle microtubules to kinetochoreAurora kinase BHomo sapiens (human)
abscissionAurora kinase BHomo sapiens (human)
negative regulation of protein bindingAurora kinase BHomo sapiens (human)
positive regulation of telomere maintenance via telomeraseAurora kinase BHomo sapiens (human)
negative regulation of cytokinesisAurora kinase BHomo sapiens (human)
positive regulation of cytokinesisAurora kinase BHomo sapiens (human)
protein localization to kinetochoreAurora kinase BHomo sapiens (human)
cellular response to UVAurora kinase BHomo sapiens (human)
cleavage furrow formationAurora kinase BHomo sapiens (human)
post-translational protein modificationAurora kinase BHomo sapiens (human)
cell cycle G2/M phase transitionAurora kinase BHomo sapiens (human)
mitotic cytokinesis checkpoint signalingAurora kinase BHomo sapiens (human)
negative regulation of innate immune responseAurora kinase BHomo sapiens (human)
protein autophosphorylationAurora kinase BHomo sapiens (human)
mitotic spindle midzone assemblyAurora kinase BHomo sapiens (human)
positive regulation of telomerase activityAurora kinase BHomo sapiens (human)
regulation of chromosome segregationAurora kinase BHomo sapiens (human)
positive regulation of mitotic sister chromatid segregationAurora kinase BHomo sapiens (human)
positive regulation of mitotic cell cycle spindle assembly checkpointAurora kinase BHomo sapiens (human)
mitotic spindle assemblyAurora kinase BHomo sapiens (human)
negative regulation of cGAS/STING signaling pathwayAurora kinase BHomo sapiens (human)
regulation of signal transduction by p53 class mediatorAurora kinase BHomo sapiens (human)
positive regulation of mitotic sister chromatid separationAurora kinase BHomo sapiens (human)
positive regulation of attachment of mitotic spindle microtubules to kinetochoreAurora kinase BHomo sapiens (human)
positive regulation of mitotic cytokinesisAurora kinase BHomo sapiens (human)
positive regulation of telomere cappingAurora kinase BHomo sapiens (human)
positive regulation of lateral attachment of mitotic spindle microtubules to kinetochoreAurora kinase BHomo sapiens (human)
mitotic spindle organizationAurora kinase BHomo sapiens (human)
regulation of cytokinesisAurora kinase BHomo sapiens (human)
microtubule cytoskeleton organizationMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
positive regulation of cell cycleMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
microtubule cytoskeleton organizationMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
microtubule bundle formationMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
protein phosphorylationMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
nervous system developmentMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
positive regulation of programmed cell deathMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
cilium organizationMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
positive regulation of cilium assemblyMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
regulation of centrosome cycleMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
cell divisionMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
positive regulation of NLRP3 inflammasome complex assemblyMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
positive regulation of protein localization to centrosomeMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
intracellular signal transductionMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase Nek1Homo sapiens (human)
cell divisionSerine/threonine-protein kinase Nek1Homo sapiens (human)
cilium assemblySerine/threonine-protein kinase Nek1Homo sapiens (human)
cell population proliferationATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of B cell proliferationATPase family AAA domain-containing protein 5Homo sapiens (human)
nuclear DNA replicationATPase family AAA domain-containing protein 5Homo sapiens (human)
signal transduction in response to DNA damageATPase family AAA domain-containing protein 5Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorATPase family AAA domain-containing protein 5Homo sapiens (human)
isotype switchingATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of DNA replicationATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of isotype switching to IgG isotypesATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA clamp unloadingATPase family AAA domain-containing protein 5Homo sapiens (human)
regulation of mitotic cell cycle phase transitionATPase family AAA domain-containing protein 5Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorATPase family AAA domain-containing protein 5Homo sapiens (human)
positive regulation of cell cycle G2/M phase transitionATPase family AAA domain-containing protein 5Homo sapiens (human)
protein phosphorylationPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
regulation of respiratory gaseous exchangePAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of glycogen biosynthetic processPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of translationPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
protein autophosphorylationPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
regulation of glucagon secretionPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
energy homeostasisPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
intracellular signal transductionPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
MAPK cascadeCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
positive regulation of protein phosphorylationCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
protein phosphorylationCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
calcium-mediated signalingCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
cellular response to reactive oxygen speciesCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
regulation of protein kinase activityCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
protein autophosphorylationCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
CAMKK-AMPK signaling cascadeCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
positive regulation of autophagy of mitochondrionCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
protein phosphorylationEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
tRNA processingEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
regulation of signal transduction by p53 class mediatorEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
tRNA threonylcarbamoyladenosine metabolic processEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
protein phosphorylationDual specificity testis-specific protein kinase 2Homo sapiens (human)
spermatogenesisDual specificity testis-specific protein kinase 2Homo sapiens (human)
actin cytoskeleton organizationDual specificity testis-specific protein kinase 2Homo sapiens (human)
focal adhesion assemblyDual specificity testis-specific protein kinase 2Homo sapiens (human)
regulation of cyclin-dependent protein serine/threonine kinase activityMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
G2/M transition of mitotic cell cycleMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
mitotic cell cycleMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
regulation of mitotic nuclear divisionMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
negative regulation of G2/M transition of mitotic cell cycleMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
protein phosphorylationMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
negative regulation of G2/MI transition of meiotic cell cycleMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
meiotic cell cycleMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
response to ischemiaMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to oxidative stressMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cellular response to amino acid starvationMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
response to endoplasmic reticulum stressMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
neuron intrinsic apoptotic signaling pathway in response to oxidative stressMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
p38MAPK cascadeMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of apoptotic processMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of cysteine-type endopeptidase activity involved in apoptotic processMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of JUN kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
innate immune responseMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of myoblast differentiationMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of protein kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of DNA-templated transcriptionMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of JNK cascadeMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
neuron apoptotic processMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cellular response to hydrogen peroxideMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cellular response to tumor necrosis factorMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
endothelial cell apoptotic processMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cellular senescenceMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
apoptotic signaling pathwayMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
programmed necrotic cell deathMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of p38MAPK cascadeMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cellular response to reactive nitrogen speciesMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cellular response to stressMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
negative regulation of receptor internalizationAtaxin-2Homo sapiens (human)
regulation of translationAtaxin-2Homo sapiens (human)
RNA metabolic processAtaxin-2Homo sapiens (human)
P-body assemblyAtaxin-2Homo sapiens (human)
stress granule assemblyAtaxin-2Homo sapiens (human)
RNA transportAtaxin-2Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
protein autophosphorylationMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
blood vessel developmentMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
positive regulation of cell proliferation in bone marrowMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
positive regulation of cell migration involved in sprouting angiogenesisMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
positive regulation of p38MAPK cascadeMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
negative regulation of cellular senescenceMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
acute inflammatory responseEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
phagocytosisEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
negative regulation of cell population proliferationEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
regulation of eIF2 alpha phosphorylation by hemeEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
macrophage differentiationEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
negative regulation of translational initiation by ironEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
protoporphyrinogen IX metabolic processEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
protein autophosphorylationEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
negative regulation of hemoglobin biosynthetic processEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
establishment of localization in cellEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
multicellular organismal-level iron ion homeostasisEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
integrated stress response signalingEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
HRI-mediated signalingEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
positive regulation of mitophagyEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
response to iron ion starvationEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
regulation of translational initiation by eIF2 alpha phosphorylationEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
DNA damage responseTarget of rapamycin complex subunit LST8Homo sapiens (human)
cytoskeleton organizationTarget of rapamycin complex subunit LST8Homo sapiens (human)
negative regulation of autophagyTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of cell growthTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of actin filament polymerizationTarget of rapamycin complex subunit LST8Homo sapiens (human)
cellular response to nutrient levelsTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of TOR signalingTarget of rapamycin complex subunit LST8Homo sapiens (human)
regulation of actin cytoskeleton organizationTarget of rapamycin complex subunit LST8Homo sapiens (human)
TORC1 signalingTarget of rapamycin complex subunit LST8Homo sapiens (human)
negative regulation of apoptotic processTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of glycolytic processTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of lipid biosynthetic processTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationTarget of rapamycin complex subunit LST8Homo sapiens (human)
cellular response to hypoxiaTarget of rapamycin complex subunit LST8Homo sapiens (human)
cellular response to osmotic stressTarget of rapamycin complex subunit LST8Homo sapiens (human)
positive regulation of pentose-phosphate shuntTarget of rapamycin complex subunit LST8Homo sapiens (human)
TOR signalingTarget of rapamycin complex subunit LST8Homo sapiens (human)
regulation of cyclin-dependent protein serine/threonine kinase activityNucleolar GTP-binding protein 1Homo sapiens (human)
maturation of LSU-rRNA from tricistronic rRNA transcript (SSU-rRNA, 5.8S rRNA, LSU-rRNA)Nucleolar GTP-binding protein 1Homo sapiens (human)
osteoblast differentiationNucleolar GTP-binding protein 1Homo sapiens (human)
negative regulation of DNA replicationNucleolar GTP-binding protein 1Homo sapiens (human)
negative regulation of cell population proliferationNucleolar GTP-binding protein 1Homo sapiens (human)
negative regulation of cell-cell adhesionNucleolar GTP-binding protein 1Homo sapiens (human)
negative regulation of cell migrationNucleolar GTP-binding protein 1Homo sapiens (human)
negative regulation of protein ubiquitinationNucleolar GTP-binding protein 1Homo sapiens (human)
negative regulation of collagen bindingNucleolar GTP-binding protein 1Homo sapiens (human)
ribosomal large subunit biogenesisNucleolar GTP-binding protein 1Homo sapiens (human)
protein stabilizationNucleolar GTP-binding protein 1Homo sapiens (human)
angiogenesisSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of endothelial cell proliferationSerine/threonine-protein kinase D2Homo sapiens (human)
adaptive immune responseSerine/threonine-protein kinase D2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase D2Homo sapiens (human)
cell adhesionSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of endothelial cell migrationSerine/threonine-protein kinase D2Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase D2Homo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase D2Homo sapiens (human)
sphingolipid biosynthetic processSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of vascular endothelial growth factor receptor signaling pathwaySerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of interleukin-2 productionSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of interleukin-8 productionSerine/threonine-protein kinase D2Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase D2Homo sapiens (human)
cellular response to vascular endothelial growth factor stimulusSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of blood vessel endothelial cell migrationSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of fibroblast growth factor receptor signaling pathwaySerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of angiogenesisSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of cell adhesionSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IISerine/threonine-protein kinase D2Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase D2Homo sapiens (human)
vascular endothelial growth factor receptor signaling pathwaySerine/threonine-protein kinase D2Homo sapiens (human)
T cell receptor signaling pathwaySerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of T cell receptor signaling pathwaySerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of DNA-binding transcription factor activitySerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activitySerine/threonine-protein kinase D2Homo sapiens (human)
endothelial tube morphogenesisSerine/threonine-protein kinase D2Homo sapiens (human)
regulation of T cell apoptotic processSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of DNA biosynthetic processSerine/threonine-protein kinase D2Homo sapiens (human)
positive regulation of endothelial cell chemotaxisSerine/threonine-protein kinase D2Homo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwaySerine/threonine-protein kinase D2Homo sapiens (human)
protein phosphorylationNUAK family SNF1-like kinase 2Homo sapiens (human)
apoptotic processNUAK family SNF1-like kinase 2Homo sapiens (human)
actin cytoskeleton organizationNUAK family SNF1-like kinase 2Homo sapiens (human)
protein localization to nucleusNUAK family SNF1-like kinase 2Homo sapiens (human)
regulation of hippo signalingNUAK family SNF1-like kinase 2Homo sapiens (human)
cellular response to glucose starvationNUAK family SNF1-like kinase 2Homo sapiens (human)
negative regulation of apoptotic processNUAK family SNF1-like kinase 2Homo sapiens (human)
rRNA modificationRNA cytidine acetyltransferaseHomo sapiens (human)
regulation of translationRNA cytidine acetyltransferaseHomo sapiens (human)
protein acetylationRNA cytidine acetyltransferaseHomo sapiens (human)
regulation of centrosome duplicationRNA cytidine acetyltransferaseHomo sapiens (human)
negative regulation of telomere maintenance via telomeraseRNA cytidine acetyltransferaseHomo sapiens (human)
ribosomal small subunit biogenesisRNA cytidine acetyltransferaseHomo sapiens (human)
positive regulation of translationRNA cytidine acetyltransferaseHomo sapiens (human)
tRNA acetylationRNA cytidine acetyltransferaseHomo sapiens (human)
rRNA acetylation involved in maturation of SSU-rRNARNA cytidine acetyltransferaseHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase SIK2Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase SIK2Homo sapiens (human)
regulation of insulin receptor signaling pathwaySerine/threonine-protein kinase SIK2Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase SIK2Homo sapiens (human)
apoptotic processSTE20-like serine/threonine-protein kinase Homo sapiens (human)
regulation of cell migrationSTE20-like serine/threonine-protein kinase Homo sapiens (human)
cytoplasmic microtubule organizationSTE20-like serine/threonine-protein kinase Homo sapiens (human)
regulation of apoptotic processSTE20-like serine/threonine-protein kinase Homo sapiens (human)
protein autophosphorylationSTE20-like serine/threonine-protein kinase Homo sapiens (human)
regulation of focal adhesion assemblySTE20-like serine/threonine-protein kinase Homo sapiens (human)
protein phosphorylationSTE20-like serine/threonine-protein kinase Homo sapiens (human)
MAPK cascadeSerine/threonine-protein kinase TAO3Homo sapiens (human)
DNA repairSerine/threonine-protein kinase TAO3Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase TAO3Homo sapiens (human)
DNA damage responseSerine/threonine-protein kinase TAO3Homo sapiens (human)
mitotic G2 DNA damage checkpoint signalingSerine/threonine-protein kinase TAO3Homo sapiens (human)
positive regulation of stress-activated MAPK cascadeSerine/threonine-protein kinase TAO3Homo sapiens (human)
positive regulation of JUN kinase activitySerine/threonine-protein kinase TAO3Homo sapiens (human)
negative regulation of JNK cascadeSerine/threonine-protein kinase TAO3Homo sapiens (human)
positive regulation of JNK cascadeSerine/threonine-protein kinase TAO3Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase TAO3Homo sapiens (human)
regulation of MAPK cascadeSerine/threonine-protein kinase TAO3Homo sapiens (human)
neuron projection morphogenesisSerine/threonine-protein kinase TAO3Homo sapiens (human)
dTTP catabolic processdCTP pyrophosphatase 1Homo sapiens (human)
dCTP catabolic processdCTP pyrophosphatase 1Homo sapiens (human)
nucleoside triphosphate catabolic processdCTP pyrophosphatase 1Homo sapiens (human)
DNA protectiondCTP pyrophosphatase 1Homo sapiens (human)
regulation of RNA splicingDual specificity protein kinase CLK4Homo sapiens (human)
peptidyl-tyrosine phosphorylationDual specificity protein kinase CLK4Homo sapiens (human)
Wnt signaling pathwayCasein kinase I isoform gamma-1Homo sapiens (human)
positive regulation of canonical Wnt signaling pathwayCasein kinase I isoform gamma-1Homo sapiens (human)
signal transductionCasein kinase I isoform gamma-1Homo sapiens (human)
endocytosisCasein kinase I isoform gamma-1Homo sapiens (human)
peptidyl-serine phosphorylationCasein kinase I isoform gamma-1Homo sapiens (human)
translationPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
phenylalanyl-tRNA aminoacylationPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
protein heterotetramerizationPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
positive regulation of Notch signaling pathwayBMP-2-inducible protein kinaseHomo sapiens (human)
regulation of clathrin-dependent endocytosisBMP-2-inducible protein kinaseHomo sapiens (human)
regulation of bone mineralizationBMP-2-inducible protein kinaseHomo sapiens (human)
ATP metabolic processObg-like ATPase 1Homo sapiens (human)
ribosomal large subunit assemblyMidasinHomo sapiens (human)
ribosomal large subunit export from nucleusMidasinHomo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
toll-like receptor signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
neutrophil mediated immunityInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
MyD88-dependent toll-like receptor signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
JNK cascadeInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
toll-like receptor 4 signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
toll-like receptor 9 signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
interleukin-33-mediated signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
innate immune responseInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
positive regulation of smooth muscle cell proliferationInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
interleukin-1-mediated signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
neutrophil migrationInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
cytokine-mediated signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
Toll signaling pathwayInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
cellular response to lipopolysaccharideInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
intracellular signal transductionInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
positive regulation of programmed cell deathMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
GCN2-mediated signalingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
pyroptosisMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
DNA damage checkpoint signalingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
inflammatory responseMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cytoskeleton organizationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
JNK cascadeMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cell deathMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cell differentiationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
stress-activated protein kinase signaling cascadeMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
p38MAPK cascadeMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
embryonic digit morphogenesisMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
positive regulation of apoptotic processMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein autophosphorylationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
stress-activated MAPK cascadeMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
limb developmentMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cellular response to gamma radiationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cellular response to UV-BMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
positive regulation of mitotic DNA damage checkpointMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
peptidyl-serine phosphorylationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
regulation of mitotic metaphase/anaphase transitionMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
chromosome segregationMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
transcription by RNA polymerase IICyclin-dependent kinase 12Homo sapiens (human)
mRNA processingCyclin-dependent kinase 12Homo sapiens (human)
RNA splicingCyclin-dependent kinase 12Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IICyclin-dependent kinase 12Homo sapiens (human)
regulation of MAP kinase activityCyclin-dependent kinase 12Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICyclin-dependent kinase 12Homo sapiens (human)
protein autophosphorylationCyclin-dependent kinase 12Homo sapiens (human)
regulation of cell cycleCyclin-dependent kinase 12Homo sapiens (human)
negative regulation of stem cell differentiationCyclin-dependent kinase 12Homo sapiens (human)
protein phosphorylationCyclin-dependent kinase 12Homo sapiens (human)
aerobic respirationNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
negative regulation of cell growthNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
mitochondrial respiratory chain complex I assemblyNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
cellular response to interferon-betaNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
proton motive force-driven mitochondrial ATP synthesisNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
protein insertion into mitochondrial inner membraneNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
positive regulation of protein catabolic processNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
negative regulation of DNA-templated transcriptionNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
cellular response to retinoic acidNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
reactive oxygen species metabolic processNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
apoptotic signaling pathwayNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
extrinsic apoptotic signaling pathwayNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
positive regulation of execution phase of apoptosisNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathwayNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase 26Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase 26Homo sapiens (human)
cellular response to starvationSerine/threonine-protein kinase 26Homo sapiens (human)
microvillus assemblySerine/threonine-protein kinase 26Homo sapiens (human)
negative regulation of cell migrationSerine/threonine-protein kinase 26Homo sapiens (human)
cellular response to oxidative stressSerine/threonine-protein kinase 26Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase 26Homo sapiens (human)
regulation of apoptotic processSerine/threonine-protein kinase 26Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase 26Homo sapiens (human)
tricarboxylic acid cycleSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinate metabolic processSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinyl-CoA pathwaySuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinyl-CoA catabolic processSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinyl-CoA metabolic processSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
MAPK cascadeSerine/threonine-protein kinase NLKHomo sapiens (human)
regulation of DNA-templated transcriptionSerine/threonine-protein kinase NLKHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase NLKHomo sapiens (human)
transforming growth factor beta receptor signaling pathwaySerine/threonine-protein kinase NLKHomo sapiens (human)
Wnt signaling pathway, calcium modulating pathwaySerine/threonine-protein kinase NLKHomo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase NLKHomo sapiens (human)
negative regulation of Wnt signaling pathwaySerine/threonine-protein kinase NLKHomo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase NLKHomo sapiens (human)
protein stabilizationSerine/threonine-protein kinase NLKHomo sapiens (human)
cellular response to osmotic stressSerine/threonine-protein kinase NLKHomo sapiens (human)
negative regulation of TORC1 signalingSerine/threonine-protein kinase NLKHomo sapiens (human)
positive regulation of receptor signaling pathway via STATSerine/threonine-protein kinase NLKHomo sapiens (human)
glycogen metabolic process5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
regulation of glycolytic process5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
negative regulation of protein kinase activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
fatty acid biosynthetic process5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
ATP biosynthetic process5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
positive regulation of peptidyl-threonine phosphorylation5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
sterol biosynthetic process5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
regulation of fatty acid metabolic process5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
cellular response to nutrient levels5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
intracellular signal transduction5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
positive regulation of protein kinase activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
regulation of fatty acid oxidation5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
regulation of glucose import5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
regulation of catalytic activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
protein phosphorylation5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
activation of innate immune responseSerine/threonine-protein kinase TBK1Homo sapiens (human)
cytoplasmic pattern recognition receptor signaling pathwaySerine/threonine-protein kinase TBK1Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase TBK1Homo sapiens (human)
inflammatory responseSerine/threonine-protein kinase TBK1Homo sapiens (human)
canonical NF-kappaB signal transductionSerine/threonine-protein kinase TBK1Homo sapiens (human)
response to virusSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of autophagySerine/threonine-protein kinase TBK1Homo sapiens (human)
negative regulation of gene expressionSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of macroautophagySerine/threonine-protein kinase TBK1Homo sapiens (human)
peptidyl-serine phosphorylationSerine/threonine-protein kinase TBK1Homo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase TBK1Homo sapiens (human)
regulation of type I interferon productionSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of type I interferon productionSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of interferon-alpha productionSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of interferon-beta productionSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of peptidyl-serine phosphorylationSerine/threonine-protein kinase TBK1Homo sapiens (human)
toll-like receptor 4 signaling pathwaySerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionSerine/threonine-protein kinase TBK1Homo sapiens (human)
dendritic cell proliferationSerine/threonine-protein kinase TBK1Homo sapiens (human)
innate immune responseSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IISerine/threonine-protein kinase TBK1Homo sapiens (human)
defense response to Gram-positive bacteriumSerine/threonine-protein kinase TBK1Homo sapiens (human)
defense response to virusSerine/threonine-protein kinase TBK1Homo sapiens (human)
type I interferon-mediated signaling pathwaySerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of type I interferon-mediated signaling pathwaySerine/threonine-protein kinase TBK1Homo sapiens (human)
antiviral innate immune responseSerine/threonine-protein kinase TBK1Homo sapiens (human)
cGAS/STING signaling pathwaySerine/threonine-protein kinase TBK1Homo sapiens (human)
negative regulation of TORC1 signalingSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of TORC1 signalingSerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of xenophagySerine/threonine-protein kinase TBK1Homo sapiens (human)
macroautophagySerine/threonine-protein kinase TBK1Homo sapiens (human)
positive regulation of non-motile cilium assemblySeptin-9Homo sapiens (human)
protein localizationSeptin-9Homo sapiens (human)
cytoskeleton-dependent cytokinesisSeptin-9Homo sapiens (human)
potassium ion transportPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein homooligomerizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorRibosomal protein S6 kinase alpha-6Homo sapiens (human)
signal transductionRibosomal protein S6 kinase alpha-6Homo sapiens (human)
central nervous system developmentRibosomal protein S6 kinase alpha-6Homo sapiens (human)
negative regulation of embryonic developmentRibosomal protein S6 kinase alpha-6Homo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeRibosomal protein S6 kinase alpha-6Homo sapiens (human)
negative regulation of mesoderm developmentRibosomal protein S6 kinase alpha-6Homo sapiens (human)
peptidyl-serine phosphorylationRibosomal protein S6 kinase alpha-6Homo sapiens (human)
positive regulation of protein phosphorylationTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein phosphorylationTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
cytoskeleton organizationTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
response to organonitrogen compoundTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
Wnt signaling pathwayTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
microvillus assemblyTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
actin cytoskeleton organizationTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
intracellular signal transductionTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
positive regulation of JNK cascadeTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein autophosphorylationTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
regulation of dendrite morphogenesisTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein localization to plasma membraneTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
neuron projection morphogenesisTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
regulation of MAPK cascadeTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
MAPK cascadeTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
regulation of cell growthSerine/threonine-protein kinase TAO2Homo sapiens (human)
protein targeting to membraneSerine/threonine-protein kinase TAO2Homo sapiens (human)
apoptotic processSerine/threonine-protein kinase TAO2Homo sapiens (human)
DNA damage responseSerine/threonine-protein kinase TAO2Homo sapiens (human)
mitotic G2 DNA damage checkpoint signalingSerine/threonine-protein kinase TAO2Homo sapiens (human)
axonogenesisSerine/threonine-protein kinase TAO2Homo sapiens (human)
regulation of cell shapeSerine/threonine-protein kinase TAO2Homo sapiens (human)
cell migrationSerine/threonine-protein kinase TAO2Homo sapiens (human)
actin cytoskeleton organizationSerine/threonine-protein kinase TAO2Homo sapiens (human)
positive regulation of protein autophosphorylationSerine/threonine-protein kinase TAO2Homo sapiens (human)
activation of protein kinase activitySerine/threonine-protein kinase TAO2Homo sapiens (human)
positive regulation of stress-activated MAPK cascadeSerine/threonine-protein kinase TAO2Homo sapiens (human)
regulation of actin cytoskeleton organizationSerine/threonine-protein kinase TAO2Homo sapiens (human)
positive regulation of MAPK cascadeSerine/threonine-protein kinase TAO2Homo sapiens (human)
positive regulation of JNK cascadeSerine/threonine-protein kinase TAO2Homo sapiens (human)
protein autophosphorylationSerine/threonine-protein kinase TAO2Homo sapiens (human)
focal adhesion assemblySerine/threonine-protein kinase TAO2Homo sapiens (human)
stress-activated MAPK cascadeSerine/threonine-protein kinase TAO2Homo sapiens (human)
basal dendrite morphogenesisSerine/threonine-protein kinase TAO2Homo sapiens (human)
basal dendrite arborizationSerine/threonine-protein kinase TAO2Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase TAO2Homo sapiens (human)
long-chain fatty acid metabolic processLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
long-chain fatty-acyl-CoA biosynthetic processLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
positive regulation of long-chain fatty acid import across plasma membraneLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
mitochondrial genome maintenanceRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of endothelial cell proliferationRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
protein phosphorylationRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
signal transductionRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of TOR signalingRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of blood vessel endothelial cell migrationRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of angiogenesisRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cell sizeRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
brain morphogenesisRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
homeostasis of number of cells within a tissueRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of cell migration involved in sprouting angiogenesisRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of vascular endothelial cell proliferationRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
positive regulation of artery morphogenesisRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
negative regulation of cellular senescenceRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
intracellular signal transductionRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
peptidyl-serine phosphorylationRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase SIK3Homo sapiens (human)
positive regulation of TORC1 signalingSerine/threonine-protein kinase SIK3Homo sapiens (human)
positive regulation of TORC2 signalingSerine/threonine-protein kinase SIK3Homo sapiens (human)
microtubule cytoskeleton organizationSerine/threonine-protein kinase SIK3Homo sapiens (human)
intracellular signal transductionSerine/threonine-protein kinase SIK3Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
cellular response to mechanical stimulusMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIThyroid hormone receptor-associated protein 3Homo sapiens (human)
regulation of alternative mRNA splicing, via spliceosomeThyroid hormone receptor-associated protein 3Homo sapiens (human)
nuclear-transcribed mRNA catabolic processThyroid hormone receptor-associated protein 3Homo sapiens (human)
mRNA processingThyroid hormone receptor-associated protein 3Homo sapiens (human)
circadian rhythmThyroid hormone receptor-associated protein 3Homo sapiens (human)
RNA splicingThyroid hormone receptor-associated protein 3Homo sapiens (human)
positive regulation of circadian rhythmThyroid hormone receptor-associated protein 3Homo sapiens (human)
positive regulation of DNA-templated transcriptionThyroid hormone receptor-associated protein 3Homo sapiens (human)
positive regulation of mRNA splicing, via spliceosomeThyroid hormone receptor-associated protein 3Homo sapiens (human)
mRNA stabilizationThyroid hormone receptor-associated protein 3Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIThyroid hormone receptor-associated protein 3Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
protein phosphorylationMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
regulation of T cell mediated cytotoxicityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
regulation of adaptive immune responseReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of phosphatase activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
activation of protein kinase activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
regulation of type II interferon productionReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
T cell differentiation in thymusReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein modification processReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
non-canonical NF-kappaB signal transductionReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
regulation of apoptotic processReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
T cell homeostasisReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of DNA-templated transcriptionReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
regulation of activated T cell proliferationReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein autophosphorylationReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
lymph node developmentReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
spleen developmentReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
thymus developmentReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
defense response to virusReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of necroptotic processReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
regulation of activation-induced cell death of T cellsReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
necroptotic processReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
cellular response to hydrogen peroxideReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
reactive oxygen species metabolic processReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
apoptotic signaling pathwayReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
programmed necrotic cell deathReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
necroptotic signaling pathwayReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
execution phase of necroptosisReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
amyloid fibril formationReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
regulation of CD8-positive, alpha-beta cytotoxic T cell extravasationReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
signal transductionReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein phosphorylationSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
cytoskeleton organizationSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
establishment or maintenance of cell polaritySerine/threonine-protein kinase MRCK betaHomo sapiens (human)
signal transductionSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
cell migrationSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
actin cytoskeleton organizationSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
actomyosin structure organizationSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
peptidyl-threonine phosphorylationSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
positive regulation of cytokine productionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of cytokine-mediated signaling pathwayInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
MyD88-dependent toll-like receptor signaling pathwayInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein phosphorylationInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
response to virusInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
positive regulation of macrophage tolerance inductionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of macrophage cytokine productionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
cytokine-mediated signaling pathwayInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of NF-kappaB transcription factor activityInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
response to peptidoglycanInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
response to lipopolysaccharideInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of interleukin-12 productionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of interleukin-6 productionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of tumor necrosis factor productionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of toll-like receptor signaling pathwayInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of protein catabolic processInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of protein-containing complex disassemblyInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
regulation of protein-containing complex disassemblyInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
response to exogenous dsRNAInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of MAP kinase activityInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
negative regulation of innate immune responseInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activityInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
interleukin-1-mediated signaling pathwayInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
response to interleukin-1Interleukin-1 receptor-associated kinase 3Homo sapiens (human)
Toll signaling pathwayInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
cellular response to lipopolysaccharideInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
intracellular signal transductionInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
Wnt signaling pathwayCasein kinase I isoform gamma-3Homo sapiens (human)
protein modification processCasein kinase I isoform gamma-3Homo sapiens (human)
peptidyl-serine phosphorylationCasein kinase I isoform gamma-3Homo sapiens (human)
signal transductionCasein kinase I isoform gamma-3Homo sapiens (human)
positive regulation of canonical Wnt signaling pathwayCasein kinase I isoform gamma-3Homo sapiens (human)
endocytosisCasein kinase I isoform gamma-3Homo sapiens (human)
MAPK cascadeMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
placenta developmentMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
response to UV-CMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
regulation of gene expressionMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
male germ-line sex determinationMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
positive regulation of telomere maintenance via telomeraseMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
intracellular signal transductionMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
positive regulation of JUN kinase activityMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
positive regulation of telomerase activityMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
chorionic trophoblast cell differentiationMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
positive regulation of p38MAPK cascadeMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
positive regulation of telomere cappingMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (534)

Processvia Protein(s)Taxonomy
protein serine/threonine kinase activityBone morphogenetic protein receptor type-1BHomo sapiens (human)
transmembrane receptor protein serine/threonine kinase activityBone morphogenetic protein receptor type-1BHomo sapiens (human)
transmembrane signaling receptor activityBone morphogenetic protein receptor type-1BHomo sapiens (human)
protein bindingBone morphogenetic protein receptor type-1BHomo sapiens (human)
ATP bindingBone morphogenetic protein receptor type-1BHomo sapiens (human)
BMP bindingBone morphogenetic protein receptor type-1BHomo sapiens (human)
SMAD bindingBone morphogenetic protein receptor type-1BHomo sapiens (human)
metal ion bindingBone morphogenetic protein receptor type-1BHomo sapiens (human)
BMP receptor activityBone morphogenetic protein receptor type-1BHomo sapiens (human)
transforming growth factor beta receptor activity, type IBone morphogenetic protein receptor type-1BHomo sapiens (human)
protein kinase activityCell division cycle 7-related protein kinaseHomo sapiens (human)
protein bindingCell division cycle 7-related protein kinaseHomo sapiens (human)
ATP bindingCell division cycle 7-related protein kinaseHomo sapiens (human)
kinase activityCell division cycle 7-related protein kinaseHomo sapiens (human)
metal ion bindingCell division cycle 7-related protein kinaseHomo sapiens (human)
protein serine kinase activityCell division cycle 7-related protein kinaseHomo sapiens (human)
protein serine/threonine kinase activityCell division cycle 7-related protein kinaseHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase PLK4Homo sapiens (human)
protein bindingSerine/threonine-protein kinase PLK4Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase PLK4Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase PLK4Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase PLK4Homo sapiens (human)
DNA bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
DNA helicase activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
RNA bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
RNA helicase activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
mRNA bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
GTPase activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
protein bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
ATP bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
transcription factor bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
poly(A) bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
eukaryotic initiation factor 4E bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
ATP hydrolysis activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
ribonucleoside triphosphate phosphatase activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
translation initiation factor bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
RNA strand annealing activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
signaling adaptor activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
RNA stem-loop bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
gamma-tubulin bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
ribosomal small subunit bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
CTPase activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
protein serine/threonine kinase activator activityATP-dependent RNA helicase DDX3XHomo sapiens (human)
cadherin bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
mRNA 5'-UTR bindingATP-dependent RNA helicase DDX3XHomo sapiens (human)
magnesium ion bindingPyridoxal kinaseHomo sapiens (human)
ATP bindingPyridoxal kinaseHomo sapiens (human)
zinc ion bindingPyridoxal kinaseHomo sapiens (human)
pyridoxal kinase activityPyridoxal kinaseHomo sapiens (human)
pyridoxal phosphate bindingPyridoxal kinaseHomo sapiens (human)
potassium ion bindingPyridoxal kinaseHomo sapiens (human)
sodium ion bindingPyridoxal kinaseHomo sapiens (human)
lithium ion bindingPyridoxal kinaseHomo sapiens (human)
protein homodimerization activityPyridoxal kinaseHomo sapiens (human)
transcription coactivator bindingCitron Rho-interacting kinaseHomo sapiens (human)
protein serine/threonine kinase activityCitron Rho-interacting kinaseHomo sapiens (human)
protein bindingCitron Rho-interacting kinaseHomo sapiens (human)
ATP bindingCitron Rho-interacting kinaseHomo sapiens (human)
SH3 domain bindingCitron Rho-interacting kinaseHomo sapiens (human)
protein kinase bindingCitron Rho-interacting kinaseHomo sapiens (human)
PDZ domain bindingCitron Rho-interacting kinaseHomo sapiens (human)
protein serine/threonine kinase inhibitor activityCitron Rho-interacting kinaseHomo sapiens (human)
metal ion bindingCitron Rho-interacting kinaseHomo sapiens (human)
scaffold protein bindingCitron Rho-interacting kinaseHomo sapiens (human)
protein serine kinase activityCitron Rho-interacting kinaseHomo sapiens (human)
protein kinase activitySerine/threonine-protein kinase Chk1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase Chk1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase Chk1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase Chk1Homo sapiens (human)
protein domain specific bindingSerine/threonine-protein kinase Chk1Homo sapiens (human)
histone H3T11 kinase activitySerine/threonine-protein kinase Chk1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase Chk1Homo sapiens (human)
protein kinase activityAurora kinase AHomo sapiens (human)
protein serine/threonine kinase activityAurora kinase AHomo sapiens (human)
protein serine/threonine/tyrosine kinase activityAurora kinase AHomo sapiens (human)
protein bindingAurora kinase AHomo sapiens (human)
ATP bindingAurora kinase AHomo sapiens (human)
protein kinase bindingAurora kinase AHomo sapiens (human)
ubiquitin protein ligase bindingAurora kinase AHomo sapiens (human)
histone H3S10 kinase activityAurora kinase AHomo sapiens (human)
protein heterodimerization activityAurora kinase AHomo sapiens (human)
protein serine kinase activityAurora kinase AHomo sapiens (human)
molecular function activator activityAurora kinase AHomo sapiens (human)
protein serine/threonine kinase activityCyclin-G-associated kinaseHomo sapiens (human)
protein bindingCyclin-G-associated kinaseHomo sapiens (human)
ATP bindingCyclin-G-associated kinaseHomo sapiens (human)
cyclin bindingCyclin-G-associated kinaseHomo sapiens (human)
protein-folding chaperone bindingCyclin-G-associated kinaseHomo sapiens (human)
protein serine kinase activityCyclin-G-associated kinaseHomo sapiens (human)
clathrin bindingCyclin-G-associated kinaseHomo sapiens (human)
ephrin receptor activityEphrin type-B receptor 6Homo sapiens (human)
protein bindingEphrin type-B receptor 6Homo sapiens (human)
ATP bindingEphrin type-B receptor 6Homo sapiens (human)
signaling receptor activityEphrin type-B receptor 6Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-B receptor 6Homo sapiens (human)
FAD bindingPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
flavin adenine dinucleotide bindingPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
pristanoyl-CoA oxidase activityPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
fatty acid bindingPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
guanine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
urate transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
purine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type bile acid transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
15-hydroxyprostaglandin dehydrogenase (NAD+) activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein serine/threonine kinase activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
signaling receptor bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
protein bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
ATP bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
LIM domain bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
signaling adaptor activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
identical protein bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
protein homodimerization activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
CARD domain bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
caspase bindingReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
protein serine kinase activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
JUN kinase kinase kinase activityReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
protein kinase activityMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
protein serine/threonine kinase activityMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
protein bindingMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
ATP bindingMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
protein serine kinase activityMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
histone H2A kinase activityMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
magnesium ion bindingDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
GTPase activityDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
protein bindingDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
GTP bindingDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
phosphatidic acid bindingDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
GTPase-dependent fusogenic activityDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
membrane bending activityDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
cardiolipin bindingDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
microtubule bindingDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
tRNA bindingEukaryotic translation initiation factor 5BHomo sapiens (human)
RNA bindingEukaryotic translation initiation factor 5BHomo sapiens (human)
translation initiation factor activityEukaryotic translation initiation factor 5BHomo sapiens (human)
GTPase activityEukaryotic translation initiation factor 5BHomo sapiens (human)
protein bindingEukaryotic translation initiation factor 5BHomo sapiens (human)
GTP bindingEukaryotic translation initiation factor 5BHomo sapiens (human)
metal ion bindingEukaryotic translation initiation factor 5BHomo sapiens (human)
protease bindingRho-associated protein kinase 2Homo sapiens (human)
RNA bindingRho-associated protein kinase 2Homo sapiens (human)
protein serine/threonine kinase activityRho-associated protein kinase 2Homo sapiens (human)
structural molecule activityRho-associated protein kinase 2Homo sapiens (human)
protein bindingRho-associated protein kinase 2Homo sapiens (human)
ATP bindingRho-associated protein kinase 2Homo sapiens (human)
small GTPase bindingRho-associated protein kinase 2Homo sapiens (human)
metal ion bindingRho-associated protein kinase 2Homo sapiens (human)
tau protein bindingRho-associated protein kinase 2Homo sapiens (human)
tau-protein kinase activityRho-associated protein kinase 2Homo sapiens (human)
endopeptidase activator activityRho-associated protein kinase 2Homo sapiens (human)
Rho-dependent protein serine/threonine kinase activityRho-associated protein kinase 2Homo sapiens (human)
protein serine kinase activityRho-associated protein kinase 2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase ULK1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase ULK1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase ULK1Homo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase ULK1Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase ULK1Homo sapiens (human)
protein-containing complex bindingSerine/threonine-protein kinase ULK1Homo sapiens (human)
GTPase bindingSerine/threonine-protein kinase ULK1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase ULK1Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
RNA endonuclease activitySerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
platelet-derived growth factor receptor bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
enzyme bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
Hsp70 protein bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
protein homodimerization activitySerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
ADP bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
Hsp90 protein bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
unfolded protein bindingSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
magnesium ion bindingRibosomal protein S6 kinase alpha-5Homo sapiens (human)
protein serine/threonine kinase activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
protein tyrosine kinase activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
protein bindingRibosomal protein S6 kinase alpha-5Homo sapiens (human)
ATP bindingRibosomal protein S6 kinase alpha-5Homo sapiens (human)
histone H3S10 kinase activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
histone H3S28 kinase activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
histone H2AS1 kinase activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
protein serine kinase activityRibosomal protein S6 kinase alpha-5Homo sapiens (human)
RNA bindingU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
RNA helicase activityU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
helicase activityU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
protein bindingU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
ATP bindingU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
ATP hydrolysis activityU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
identical protein bindingU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
magnesium ion bindingRibosomal protein S6 kinase alpha-4Homo sapiens (human)
protein serine/threonine kinase activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
ribosomal protein S6 kinase activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
protein bindingRibosomal protein S6 kinase alpha-4Homo sapiens (human)
ATP bindingRibosomal protein S6 kinase alpha-4Homo sapiens (human)
histone H3S10 kinase activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
histone H3S28 kinase activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
protein serine kinase activityRibosomal protein S6 kinase alpha-4Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingSerine/threonine-protein kinase 16Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activitySerine/threonine-protein kinase 16Homo sapiens (human)
protein bindingSerine/threonine-protein kinase 16Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase 16Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase 16Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase 16Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase 10Homo sapiens (human)
protein bindingSerine/threonine-protein kinase 10Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase 10Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase 10Homo sapiens (human)
protein homodimerization activitySerine/threonine-protein kinase 10Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase 10Homo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activitySerine/threonine-protein kinase D3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase D3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase D3Homo sapiens (human)
kinase activitySerine/threonine-protein kinase D3Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase D3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase D3Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase D3Homo sapiens (human)
protein bindingBile salt export pumpHomo sapiens (human)
ATP bindingBile salt export pumpHomo sapiens (human)
ABC-type xenobiotic transporter activityBile salt export pumpHomo sapiens (human)
bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
canalicular bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transporter activityBile salt export pumpHomo sapiens (human)
ABC-type bile acid transporter activityBile salt export pumpHomo sapiens (human)
ATP hydrolysis activityBile salt export pumpHomo sapiens (human)
single-stranded DNA bindingStructural maintenance of chromosomes protein 2Homo sapiens (human)
protein bindingStructural maintenance of chromosomes protein 2Homo sapiens (human)
ATP bindingStructural maintenance of chromosomes protein 2Homo sapiens (human)
ATP hydrolysis activityStructural maintenance of chromosomes protein 2Homo sapiens (human)
chromatin bindingStructural maintenance of chromosomes protein 2Homo sapiens (human)
magnesium ion bindingMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 6Homo sapiens (human)
creatine kinase activityMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
microtubule bindingMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
MAP kinase kinase kinase kinase activityMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase LATS1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase LATS1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase LATS1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase LATS1Homo sapiens (human)
protein kinase bindingSerine/threonine-protein kinase LATS1Homo sapiens (human)
nuclear estrogen receptor bindingSerine/threonine-protein kinase LATS1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase LATS1Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase PAK 4Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase PAK 4Homo sapiens (human)
protein bindingSerine/threonine-protein kinase PAK 4Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cadherin binding involved in cell-cell adhesionSerine/threonine-protein kinase PAK 4Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase PAK 4Homo sapiens (human)
supercoiled DNA bindingTyrosine-protein kinase ABL1Homo sapiens (human)
magnesium ion bindingTyrosine-protein kinase ABL1Homo sapiens (human)
four-way junction DNA bindingTyrosine-protein kinase ABL1Homo sapiens (human)
bubble DNA bindingTyrosine-protein kinase ABL1Homo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase ABL1Homo sapiens (human)
DNA bindingTyrosine-protein kinase ABL1Homo sapiens (human)
transcription coactivator activityTyrosine-protein kinase ABL1Homo sapiens (human)
actin monomer bindingTyrosine-protein kinase ABL1Homo sapiens (human)
nicotinate-nucleotide adenylyltransferase activityTyrosine-protein kinase ABL1Homo sapiens (human)
protein kinase activityTyrosine-protein kinase ABL1Homo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase ABL1Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase ABL1Homo sapiens (human)
protein kinase C bindingTyrosine-protein kinase ABL1Homo sapiens (human)
protein bindingTyrosine-protein kinase ABL1Homo sapiens (human)
ATP bindingTyrosine-protein kinase ABL1Homo sapiens (human)
kinase activityTyrosine-protein kinase ABL1Homo sapiens (human)
SH3 domain bindingTyrosine-protein kinase ABL1Homo sapiens (human)
syntaxin bindingTyrosine-protein kinase ABL1Homo sapiens (human)
manganese ion bindingTyrosine-protein kinase ABL1Homo sapiens (human)
neuropilin bindingTyrosine-protein kinase ABL1Homo sapiens (human)
SH2 domain bindingTyrosine-protein kinase ABL1Homo sapiens (human)
ephrin receptor bindingTyrosine-protein kinase ABL1Homo sapiens (human)
actin filament bindingTyrosine-protein kinase ABL1Homo sapiens (human)
mitogen-activated protein kinase bindingTyrosine-protein kinase ABL1Homo sapiens (human)
proline-rich region bindingTyrosine-protein kinase ABL1Homo sapiens (human)
delta-catenin bindingTyrosine-protein kinase ABL1Homo sapiens (human)
sequence-specific double-stranded DNA bindingTyrosine-protein kinase ABL1Homo sapiens (human)
epidermal growth factor receptor activityEpidermal growth factor receptorHomo sapiens (human)
virus receptor activityEpidermal growth factor receptorHomo sapiens (human)
chromatin bindingEpidermal growth factor receptorHomo sapiens (human)
double-stranded DNA bindingEpidermal growth factor receptorHomo sapiens (human)
MAP kinase kinase kinase activityEpidermal growth factor receptorHomo sapiens (human)
protein tyrosine kinase activityEpidermal growth factor receptorHomo sapiens (human)
transmembrane receptor protein tyrosine kinase activityEpidermal growth factor receptorHomo sapiens (human)
transmembrane signaling receptor activityEpidermal growth factor receptorHomo sapiens (human)
epidermal growth factor receptor activityEpidermal growth factor receptorHomo sapiens (human)
integrin bindingEpidermal growth factor receptorHomo sapiens (human)
protein bindingEpidermal growth factor receptorHomo sapiens (human)
calmodulin bindingEpidermal growth factor receptorHomo sapiens (human)
ATP bindingEpidermal growth factor receptorHomo sapiens (human)
enzyme bindingEpidermal growth factor receptorHomo sapiens (human)
kinase bindingEpidermal growth factor receptorHomo sapiens (human)
protein kinase bindingEpidermal growth factor receptorHomo sapiens (human)
protein phosphatase bindingEpidermal growth factor receptorHomo sapiens (human)
protein tyrosine kinase activator activityEpidermal growth factor receptorHomo sapiens (human)
transmembrane receptor protein tyrosine kinase activator activityEpidermal growth factor receptorHomo sapiens (human)
ubiquitin protein ligase bindingEpidermal growth factor receptorHomo sapiens (human)
identical protein bindingEpidermal growth factor receptorHomo sapiens (human)
cadherin bindingEpidermal growth factor receptorHomo sapiens (human)
actin filament bindingEpidermal growth factor receptorHomo sapiens (human)
ATPase bindingEpidermal growth factor receptorHomo sapiens (human)
epidermal growth factor bindingEpidermal growth factor receptorHomo sapiens (human)
protein tyrosine kinase activityHigh affinity nerve growth factor receptorHomo sapiens (human)
transmembrane receptor protein tyrosine kinase activityHigh affinity nerve growth factor receptorHomo sapiens (human)
GPI-linked ephrin receptor activityHigh affinity nerve growth factor receptorHomo sapiens (human)
neurotrophin p75 receptor bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
protein bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
ATP bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
nerve growth factor receptor activityHigh affinity nerve growth factor receptorHomo sapiens (human)
kinase bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
identical protein bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
protein homodimerization activityHigh affinity nerve growth factor receptorHomo sapiens (human)
nerve growth factor bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
neurotrophin bindingHigh affinity nerve growth factor receptorHomo sapiens (human)
neurotrophin receptor activityHigh affinity nerve growth factor receptorHomo sapiens (human)
transcription cis-regulatory region bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
core promoter sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
TFIID-class transcription factor complex bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
protease bindingCellular tumor antigen p53Homo sapiens (human)
p53 bindingCellular tumor antigen p53Homo sapiens (human)
DNA bindingCellular tumor antigen p53Homo sapiens (human)
chromatin bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activityCellular tumor antigen p53Homo sapiens (human)
mRNA 3'-UTR bindingCellular tumor antigen p53Homo sapiens (human)
copper ion bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCellular tumor antigen p53Homo sapiens (human)
zinc ion bindingCellular tumor antigen p53Homo sapiens (human)
enzyme bindingCellular tumor antigen p53Homo sapiens (human)
receptor tyrosine kinase bindingCellular tumor antigen p53Homo sapiens (human)
ubiquitin protein ligase bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase regulator activityCellular tumor antigen p53Homo sapiens (human)
ATP-dependent DNA/DNA annealing activityCellular tumor antigen p53Homo sapiens (human)
identical protein bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase bindingCellular tumor antigen p53Homo sapiens (human)
protein heterodimerization activityCellular tumor antigen p53Homo sapiens (human)
protein-folding chaperone bindingCellular tumor antigen p53Homo sapiens (human)
protein phosphatase 2A bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingCellular tumor antigen p53Homo sapiens (human)
14-3-3 protein bindingCellular tumor antigen p53Homo sapiens (human)
MDM2/MDM4 family protein bindingCellular tumor antigen p53Homo sapiens (human)
disordered domain specific bindingCellular tumor antigen p53Homo sapiens (human)
general transcription initiation factor bindingCellular tumor antigen p53Homo sapiens (human)
molecular function activator activityCellular tumor antigen p53Homo sapiens (human)
promoter-specific chromatin bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
GTP bindingGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
metal ion bindingGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
G-protein beta/gamma-subunit complex bindingGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
G protein-coupled receptor bindingGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
GTPase activityGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
adenine nucleotide transmembrane transporter activityADP/ATP translocase 2Homo sapiens (human)
RNA bindingADP/ATP translocase 2Homo sapiens (human)
ATP:ADP antiporter activityADP/ATP translocase 2Homo sapiens (human)
protein bindingADP/ATP translocase 2Homo sapiens (human)
proton transmembrane transporter activityADP/ATP translocase 2Homo sapiens (human)
adenine transmembrane transporter activityADP/ATP translocase 2Homo sapiens (human)
oxidative phosphorylation uncoupler activityADP/ATP translocase 2Homo sapiens (human)
ubiquitin protein ligase bindingADP/ATP translocase 2Homo sapiens (human)
chromatin bindingProtein kinase C beta typeHomo sapiens (human)
protein serine/threonine kinase activityProtein kinase C beta typeHomo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activityProtein kinase C beta typeHomo sapiens (human)
protein kinase C bindingProtein kinase C beta typeHomo sapiens (human)
calcium channel regulator activityProtein kinase C beta typeHomo sapiens (human)
protein bindingProtein kinase C beta typeHomo sapiens (human)
ATP bindingProtein kinase C beta typeHomo sapiens (human)
zinc ion bindingProtein kinase C beta typeHomo sapiens (human)
nuclear receptor coactivator activityProtein kinase C beta typeHomo sapiens (human)
histone H3T6 kinase activityProtein kinase C beta typeHomo sapiens (human)
histone bindingProtein kinase C beta typeHomo sapiens (human)
nuclear androgen receptor bindingProtein kinase C beta typeHomo sapiens (human)
protein serine kinase activityProtein kinase C beta typeHomo sapiens (human)
amyloid-beta bindingInsulin receptorHomo sapiens (human)
protein tyrosine kinase activityInsulin receptorHomo sapiens (human)
insulin receptor activityInsulin receptorHomo sapiens (human)
insulin-like growth factor receptor bindingInsulin receptorHomo sapiens (human)
protein bindingInsulin receptorHomo sapiens (human)
ATP bindingInsulin receptorHomo sapiens (human)
GTP bindingInsulin receptorHomo sapiens (human)
protein domain specific bindingInsulin receptorHomo sapiens (human)
insulin-like growth factor I bindingInsulin receptorHomo sapiens (human)
insulin-like growth factor II bindingInsulin receptorHomo sapiens (human)
cargo receptor activityInsulin receptorHomo sapiens (human)
phosphatidylinositol 3-kinase bindingInsulin receptorHomo sapiens (human)
insulin bindingInsulin receptorHomo sapiens (human)
insulin receptor substrate bindingInsulin receptorHomo sapiens (human)
protein-containing complex bindingInsulin receptorHomo sapiens (human)
PTB domain bindingInsulin receptorHomo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase LckHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase LckHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase LckHomo sapiens (human)
protein serine/threonine phosphatase activityTyrosine-protein kinase LckHomo sapiens (human)
protein bindingTyrosine-protein kinase LckHomo sapiens (human)
ATP bindingTyrosine-protein kinase LckHomo sapiens (human)
phospholipase activator activityTyrosine-protein kinase LckHomo sapiens (human)
protein kinase bindingTyrosine-protein kinase LckHomo sapiens (human)
protein phosphatase bindingTyrosine-protein kinase LckHomo sapiens (human)
SH2 domain bindingTyrosine-protein kinase LckHomo sapiens (human)
T cell receptor bindingTyrosine-protein kinase LckHomo sapiens (human)
CD4 receptor bindingTyrosine-protein kinase LckHomo sapiens (human)
CD8 receptor bindingTyrosine-protein kinase LckHomo sapiens (human)
identical protein bindingTyrosine-protein kinase LckHomo sapiens (human)
phospholipase bindingTyrosine-protein kinase LckHomo sapiens (human)
phosphatidylinositol 3-kinase bindingTyrosine-protein kinase LckHomo sapiens (human)
ATPase bindingTyrosine-protein kinase LckHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase LckHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase FynHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase FynHomo sapiens (human)
protein bindingTyrosine-protein kinase FynHomo sapiens (human)
ATP bindingTyrosine-protein kinase FynHomo sapiens (human)
phospholipase activator activityTyrosine-protein kinase FynHomo sapiens (human)
enzyme bindingTyrosine-protein kinase FynHomo sapiens (human)
type 5 metabotropic glutamate receptor bindingTyrosine-protein kinase FynHomo sapiens (human)
identical protein bindingTyrosine-protein kinase FynHomo sapiens (human)
alpha-tubulin bindingTyrosine-protein kinase FynHomo sapiens (human)
phospholipase bindingTyrosine-protein kinase FynHomo sapiens (human)
transmembrane transporter bindingTyrosine-protein kinase FynHomo sapiens (human)
metal ion bindingTyrosine-protein kinase FynHomo sapiens (human)
ephrin receptor bindingTyrosine-protein kinase FynHomo sapiens (human)
tau protein bindingTyrosine-protein kinase FynHomo sapiens (human)
tau-protein kinase activityTyrosine-protein kinase FynHomo sapiens (human)
growth factor receptor bindingTyrosine-protein kinase FynHomo sapiens (human)
scaffold protein bindingTyrosine-protein kinase FynHomo sapiens (human)
disordered domain specific bindingTyrosine-protein kinase FynHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase FynHomo sapiens (human)
virus receptor activityCyclin-dependent kinase 1Homo sapiens (human)
chromatin bindingCyclin-dependent kinase 1Homo sapiens (human)
protein kinase activityCyclin-dependent kinase 1Homo sapiens (human)
protein serine/threonine kinase activityCyclin-dependent kinase 1Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 1Homo sapiens (human)
protein bindingCyclin-dependent kinase 1Homo sapiens (human)
ATP bindingCyclin-dependent kinase 1Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityCyclin-dependent kinase 1Homo sapiens (human)
kinase activityCyclin-dependent kinase 1Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 1Homo sapiens (human)
Hsp70 protein bindingCyclin-dependent kinase 1Homo sapiens (human)
histone kinase activityCyclin-dependent kinase 1Homo sapiens (human)
cyclin-dependent protein kinase activityCyclin-dependent kinase 1Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 1Homo sapiens (human)
purine nucleobase bindingGlycogen phosphorylase, liver formHomo sapiens (human)
protein bindingGlycogen phosphorylase, liver formHomo sapiens (human)
ATP bindingGlycogen phosphorylase, liver formHomo sapiens (human)
glucose bindingGlycogen phosphorylase, liver formHomo sapiens (human)
glycogen phosphorylase activityGlycogen phosphorylase, liver formHomo sapiens (human)
AMP bindingGlycogen phosphorylase, liver formHomo sapiens (human)
vitamin bindingGlycogen phosphorylase, liver formHomo sapiens (human)
bile acid bindingGlycogen phosphorylase, liver formHomo sapiens (human)
identical protein bindingGlycogen phosphorylase, liver formHomo sapiens (human)
linear malto-oligosaccharide phosphorylase activityGlycogen phosphorylase, liver formHomo sapiens (human)
SHG alpha-glucan phosphorylase activityGlycogen phosphorylase, liver formHomo sapiens (human)
pyridoxal phosphate bindingGlycogen phosphorylase, liver formHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase Fes/FpsHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase Fes/FpsHomo sapiens (human)
protein bindingTyrosine-protein kinase Fes/FpsHomo sapiens (human)
ATP bindingTyrosine-protein kinase Fes/FpsHomo sapiens (human)
microtubule bindingTyrosine-protein kinase Fes/FpsHomo sapiens (human)
immunoglobulin receptor bindingTyrosine-protein kinase Fes/FpsHomo sapiens (human)
phosphatidylinositol bindingTyrosine-protein kinase Fes/FpsHomo sapiens (human)
adenine phosphoribosyltransferase activityAdenine phosphoribosyltransferaseHomo sapiens (human)
protein bindingAdenine phosphoribosyltransferaseHomo sapiens (human)
AMP bindingAdenine phosphoribosyltransferaseHomo sapiens (human)
adenine bindingAdenine phosphoribosyltransferaseHomo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase YesHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase YesHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase YesHomo sapiens (human)
protein bindingTyrosine-protein kinase YesHomo sapiens (human)
ATP bindingTyrosine-protein kinase YesHomo sapiens (human)
enzyme bindingTyrosine-protein kinase YesHomo sapiens (human)
transmembrane transporter bindingTyrosine-protein kinase YesHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase YesHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase LynHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase LynHomo sapiens (human)
platelet-derived growth factor receptor bindingTyrosine-protein kinase LynHomo sapiens (human)
integrin bindingTyrosine-protein kinase LynHomo sapiens (human)
protein bindingTyrosine-protein kinase LynHomo sapiens (human)
ATP bindingTyrosine-protein kinase LynHomo sapiens (human)
kinase activityTyrosine-protein kinase LynHomo sapiens (human)
SH3 domain bindingTyrosine-protein kinase LynHomo sapiens (human)
ubiquitin protein ligase bindingTyrosine-protein kinase LynHomo sapiens (human)
gamma-tubulin bindingTyrosine-protein kinase LynHomo sapiens (human)
glycosphingolipid bindingTyrosine-protein kinase LynHomo sapiens (human)
transmembrane transporter bindingTyrosine-protein kinase LynHomo sapiens (human)
ephrin receptor bindingTyrosine-protein kinase LynHomo sapiens (human)
phosphoprotein bindingTyrosine-protein kinase LynHomo sapiens (human)
scaffold protein bindingTyrosine-protein kinase LynHomo sapiens (human)
phosphorylation-dependent protein bindingTyrosine-protein kinase LynHomo sapiens (human)
phosphatidylinositol 3-kinase activator activityTyrosine-protein kinase LynHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase LynHomo sapiens (human)
protein tyrosine kinase activityProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
transmembrane receptor protein tyrosine kinase activityProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
calcium ion bindingProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
protein bindingProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
ATP bindingProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
signaling receptor activityProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
G-protein alpha-subunit bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
protein tyrosine kinase activityInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin-like growth factor receptor activityInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin receptor bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
protein bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin-like growth factor bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
ATP bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin-like growth factor I bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
identical protein bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
phosphatidylinositol 3-kinase bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin receptor substrate bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
protein-containing complex bindingInsulin-like growth factor 1 receptorHomo sapiens (human)
protein transporter activityInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin receptor activityInsulin-like growth factor 1 receptorHomo sapiens (human)
RNA bindingSignal recognition particle receptor subunit alphaHomo sapiens (human)
GTP bindingSignal recognition particle receptor subunit alphaHomo sapiens (human)
ATP hydrolysis activitySignal recognition particle receptor subunit alphaHomo sapiens (human)
signal recognition particle bindingSignal recognition particle receptor subunit alphaHomo sapiens (human)
GTPase activitySignal recognition particle receptor subunit alphaHomo sapiens (human)
protein bindingCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
ubiquinol-cytochrome-c reductase activityCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
heme bindingCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
metal ion bindingCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
protein tyrosine kinase activityHepatocyte growth factor receptorHomo sapiens (human)
protein bindingHepatocyte growth factor receptorHomo sapiens (human)
ATP bindingHepatocyte growth factor receptorHomo sapiens (human)
semaphorin receptor activityHepatocyte growth factor receptorHomo sapiens (human)
protein phosphatase bindingHepatocyte growth factor receptorHomo sapiens (human)
identical protein bindingHepatocyte growth factor receptorHomo sapiens (human)
molecular function activator activityHepatocyte growth factor receptorHomo sapiens (human)
hepatocyte growth factor receptor activityHepatocyte growth factor receptorHomo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase HCKHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase HCKHomo sapiens (human)
protein bindingTyrosine-protein kinase HCKHomo sapiens (human)
ATP bindingTyrosine-protein kinase HCKHomo sapiens (human)
lipid bindingTyrosine-protein kinase HCKHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase HCKHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase HCKHomo sapiens (human)
protein kinase activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
protein tyrosine kinase activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet activating factor receptor activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet-derived growth factor receptor activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet-derived growth factor beta-receptor activityPlatelet-derived growth factor receptor betaHomo sapiens (human)
signaling receptor bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet-derived growth factor receptor bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
protein bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
ATP bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
enzyme bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
protein kinase bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
vascular endothelial growth factor bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
platelet-derived growth factor bindingPlatelet-derived growth factor receptor betaHomo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase FgrHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase FgrHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase FgrHomo sapiens (human)
protein bindingTyrosine-protein kinase FgrHomo sapiens (human)
ATP bindingTyrosine-protein kinase FgrHomo sapiens (human)
protein kinase bindingTyrosine-protein kinase FgrHomo sapiens (human)
immunoglobulin receptor bindingTyrosine-protein kinase FgrHomo sapiens (human)
Fc-gamma receptor I complex bindingTyrosine-protein kinase FgrHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase FgrHomo sapiens (human)
protein kinase activitySerine/threonine-protein kinase A-RafHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase A-RafHomo sapiens (human)
protein bindingSerine/threonine-protein kinase A-RafHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase A-RafHomo sapiens (human)
metal ion bindingSerine/threonine-protein kinase A-RafHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase A-RafHomo sapiens (human)
MAP kinase kinase kinase activitySerine/threonine-protein kinase A-RafHomo sapiens (human)
protein bindingGlycogen phosphorylase, brain formHomo sapiens (human)
glycogen phosphorylase activityGlycogen phosphorylase, brain formHomo sapiens (human)
linear malto-oligosaccharide phosphorylase activityGlycogen phosphorylase, brain formHomo sapiens (human)
SHG alpha-glucan phosphorylase activityGlycogen phosphorylase, brain formHomo sapiens (human)
pyridoxal phosphate bindingGlycogen phosphorylase, brain formHomo sapiens (human)
protein serine/threonine kinase activityBreakpoint cluster region proteinHomo sapiens (human)
protein tyrosine kinase activityBreakpoint cluster region proteinHomo sapiens (human)
guanyl-nucleotide exchange factor activityBreakpoint cluster region proteinHomo sapiens (human)
GTPase activator activityBreakpoint cluster region proteinHomo sapiens (human)
protein bindingBreakpoint cluster region proteinHomo sapiens (human)
ATP bindingBreakpoint cluster region proteinHomo sapiens (human)
protein serine kinase activityBreakpoint cluster region proteinHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase pim-1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase pim-1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase pim-1Homo sapiens (human)
transcription factor bindingSerine/threonine-protein kinase pim-1Homo sapiens (human)
manganese ion bindingSerine/threonine-protein kinase pim-1Homo sapiens (human)
ribosomal small subunit bindingSerine/threonine-protein kinase pim-1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase pim-1Homo sapiens (human)
protein tyrosine kinase activityFibroblast growth factor receptor 1Homo sapiens (human)
fibroblast growth factor receptor activityFibroblast growth factor receptor 1Homo sapiens (human)
protein bindingFibroblast growth factor receptor 1Homo sapiens (human)
ATP bindingFibroblast growth factor receptor 1Homo sapiens (human)
heparin bindingFibroblast growth factor receptor 1Homo sapiens (human)
fibroblast growth factor bindingFibroblast growth factor receptor 1Homo sapiens (human)
SH2 domain bindingFibroblast growth factor receptor 1Homo sapiens (human)
identical protein bindingFibroblast growth factor receptor 1Homo sapiens (human)
protein homodimerization activityFibroblast growth factor receptor 1Homo sapiens (human)
receptor-receptor interactionFibroblast growth factor receptor 1Homo sapiens (human)
magnesium ion bindingDNA topoisomerase 2-alphaHomo sapiens (human)
DNA bindingDNA topoisomerase 2-alphaHomo sapiens (human)
chromatin bindingDNA topoisomerase 2-alphaHomo sapiens (human)
RNA bindingDNA topoisomerase 2-alphaHomo sapiens (human)
DNA topoisomerase type II (double strand cut, ATP-hydrolyzing) activityDNA topoisomerase 2-alphaHomo sapiens (human)
protein kinase C bindingDNA topoisomerase 2-alphaHomo sapiens (human)
protein bindingDNA topoisomerase 2-alphaHomo sapiens (human)
ATP bindingDNA topoisomerase 2-alphaHomo sapiens (human)
ATP-dependent activity, acting on DNADNA topoisomerase 2-alphaHomo sapiens (human)
DNA binding, bendingDNA topoisomerase 2-alphaHomo sapiens (human)
protein homodimerization activityDNA topoisomerase 2-alphaHomo sapiens (human)
ubiquitin bindingDNA topoisomerase 2-alphaHomo sapiens (human)
protein heterodimerization activityDNA topoisomerase 2-alphaHomo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 4Homo sapiens (human)
protein bindingCyclin-dependent kinase 4Homo sapiens (human)
ATP bindingCyclin-dependent kinase 4Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase regulator activityCyclin-dependent kinase 4Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 4Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 4Homo sapiens (human)
ATP:ADP antiporter activityADP/ATP translocase 3Homo sapiens (human)
protein bindingADP/ATP translocase 3Homo sapiens (human)
nucleotide bindingInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
DNA bindingInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
RNA bindingInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
IMP dehydrogenase activityInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
protein bindingInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
metal ion bindingInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
protein kinase activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
protein tyrosine kinase activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
protein kinase C bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
signaling receptor bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
insulin receptor bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
integrin bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
protein bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
ATP bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
phospholipase activator activityProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
enzyme bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
heme bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
nuclear estrogen receptor bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
SH2 domain bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
phospholipase bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
transmembrane transporter bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cadherin bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
ephrin receptor bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
ATPase bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
phosphoprotein bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
BMP receptor bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
connexin bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
scaffold protein bindingProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cAMP-dependent protein kinase inhibitor activitycAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
protein bindingcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
cAMP-dependent protein kinase regulator activitycAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
protein domain specific bindingcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
ubiquitin protein ligase bindingcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
protein kinase A catalytic subunit bindingcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
cAMP bindingcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
protein kinase activitySerine/threonine-protein kinase B-rafHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase B-rafHomo sapiens (human)
MAP kinase kinase activitySerine/threonine-protein kinase B-rafHomo sapiens (human)
calcium ion bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
protein bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
mitogen-activated protein kinase kinase bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
identical protein bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
protein-containing complex bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
scaffold protein bindingSerine/threonine-protein kinase B-rafHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase B-rafHomo sapiens (human)
MAP kinase kinase kinase activitySerine/threonine-protein kinase B-rafHomo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium channel regulator activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
telethonin bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein-containing complex bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein serine/threonine kinase activityPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
phosphorylase kinase activityPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
protein bindingPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
calmodulin bindingPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
ATP bindingPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
enzyme bindingPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
tau-protein kinase activityPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
dihydronicotinamide riboside quinone reductase activityRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
protein bindingRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
zinc ion bindingRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
electron transfer activityRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
oxidoreductase activityRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
oxidoreductase activity, acting on other nitrogenous compounds as donorsRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
chloride ion bindingRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
protein homodimerization activityRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
FAD bindingRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
melatonin bindingRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
resveratrol bindingRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
NAD(P)H dehydrogenase (quinone) activityRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase FerHomo sapiens (human)
epidermal growth factor receptor bindingTyrosine-protein kinase FerHomo sapiens (human)
protein bindingTyrosine-protein kinase FerHomo sapiens (human)
ATP bindingTyrosine-protein kinase FerHomo sapiens (human)
protein phosphatase 1 bindingTyrosine-protein kinase FerHomo sapiens (human)
lipid bindingTyrosine-protein kinase FerHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase FerHomo sapiens (human)
protein kinase activityProtein kinase C alpha typeHomo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activityProtein kinase C alpha typeHomo sapiens (human)
calcium,diacylglycerol-dependent serine/threonine kinase activityProtein kinase C alpha typeHomo sapiens (human)
integrin bindingProtein kinase C alpha typeHomo sapiens (human)
protein bindingProtein kinase C alpha typeHomo sapiens (human)
ATP bindingProtein kinase C alpha typeHomo sapiens (human)
zinc ion bindingProtein kinase C alpha typeHomo sapiens (human)
enzyme bindingProtein kinase C alpha typeHomo sapiens (human)
histone H3T6 kinase activityProtein kinase C alpha typeHomo sapiens (human)
protein serine kinase activityProtein kinase C alpha typeHomo sapiens (human)
protein serine/threonine kinase activityProtein kinase C alpha typeHomo sapiens (human)
diacylglycerol bindingProtein kinase C alpha typeHomo sapiens (human)
magnesium ion bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein kinase activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein serine/threonine kinase activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
AMP-activated protein kinase activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cAMP-dependent protein kinase activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein serine/threonine/tyrosine kinase activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
ATP bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein kinase bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein domain specific bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
manganese ion bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
ubiquitin protein ligase bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein kinase A regulatory subunit bindingcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
channel activator activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein serine kinase activitycAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
protein bindingGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
ATP bindingGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
ATP hydrolysis activityGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
protein-macromolecule adaptor activityGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
5'-3' DNA helicase activityGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
metal ion bindingGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
4 iron, 4 sulfur cluster bindingGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
DNA helicase activityGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
damaged DNA bindingGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
protein serine/threonine kinase activityCasein kinase II subunit alpha'Homo sapiens (human)
protein bindingCasein kinase II subunit alpha'Homo sapiens (human)
ATP bindingCasein kinase II subunit alpha'Homo sapiens (human)
protein serine kinase activityCasein kinase II subunit alpha'Homo sapiens (human)
GTPase activityRas-related protein Rab-6AHomo sapiens (human)
protein bindingRas-related protein Rab-6AHomo sapiens (human)
GTP bindingRas-related protein Rab-6AHomo sapiens (human)
protein domain specific bindingRas-related protein Rab-6AHomo sapiens (human)
myosin V bindingRas-related protein Rab-6AHomo sapiens (human)
fibronectin bindingEphrin type-A receptor 1Homo sapiens (human)
protein kinase activityEphrin type-A receptor 1Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-A receptor 1Homo sapiens (human)
ATP bindingEphrin type-A receptor 1Homo sapiens (human)
protein kinase bindingEphrin type-A receptor 1Homo sapiens (human)
transmembrane receptor protein tyrosine kinase activityEphrin type-A receptor 1Homo sapiens (human)
phosphoribosylaminoimidazole carboxylase activityMultifunctional protein ADE2Homo sapiens (human)
phosphoribosylaminoimidazolesuccinocarboxamide synthase activityMultifunctional protein ADE2Homo sapiens (human)
protein bindingMultifunctional protein ADE2Homo sapiens (human)
ATP bindingMultifunctional protein ADE2Homo sapiens (human)
identical protein bindingMultifunctional protein ADE2Homo sapiens (human)
5-amino-4-imidazole carboxylate lyase activityMultifunctional protein ADE2Homo sapiens (human)
cadherin bindingMultifunctional protein ADE2Homo sapiens (human)
protein serine/threonine kinase activitycAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
AMP-activated protein kinase activitycAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
cAMP-dependent protein kinase activitycAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
protein bindingcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
ATP bindingcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
protein serine kinase activitycAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
protein kinase A regulatory subunit bindingcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
magnesium ion bindingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
protein serine/threonine kinase activitycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
AMP-activated protein kinase activitycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
cAMP-dependent protein kinase activitycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
protein bindingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
ATP bindingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
ubiquitin protein ligase bindingcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
protein serine kinase activitycAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
ferrochelatase activityFerrochelatase, mitochondrialHomo sapiens (human)
protein bindingFerrochelatase, mitochondrialHomo sapiens (human)
ferrous iron bindingFerrochelatase, mitochondrialHomo sapiens (human)
heme bindingFerrochelatase, mitochondrialHomo sapiens (human)
iron-responsive element bindingFerrochelatase, mitochondrialHomo sapiens (human)
identical protein bindingFerrochelatase, mitochondrialHomo sapiens (human)
protein homodimerization activityFerrochelatase, mitochondrialHomo sapiens (human)
2 iron, 2 sulfur cluster bindingFerrochelatase, mitochondrialHomo sapiens (human)
protein kinase activityRibosomal protein S6 kinase beta-1Homo sapiens (human)
protein serine/threonine kinase activityRibosomal protein S6 kinase beta-1Homo sapiens (human)
ribosomal protein S6 kinase activityRibosomal protein S6 kinase beta-1Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityRibosomal protein S6 kinase beta-1Homo sapiens (human)
protein bindingRibosomal protein S6 kinase beta-1Homo sapiens (human)
ATP bindingRibosomal protein S6 kinase beta-1Homo sapiens (human)
PDZ domain bindingRibosomal protein S6 kinase beta-1Homo sapiens (human)
peptide bindingRibosomal protein S6 kinase beta-1Homo sapiens (human)
identical protein bindingRibosomal protein S6 kinase beta-1Homo sapiens (human)
protein phosphatase 2A bindingRibosomal protein S6 kinase beta-1Homo sapiens (human)
protein serine kinase activityRibosomal protein S6 kinase beta-1Homo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase JAK1Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase JAK1Homo sapiens (human)
growth hormone receptor bindingTyrosine-protein kinase JAK1Homo sapiens (human)
protein bindingTyrosine-protein kinase JAK1Homo sapiens (human)
ATP bindingTyrosine-protein kinase JAK1Homo sapiens (human)
protein phosphatase bindingTyrosine-protein kinase JAK1Homo sapiens (human)
ubiquitin protein ligase bindingTyrosine-protein kinase JAK1Homo sapiens (human)
CCR5 chemokine receptor bindingTyrosine-protein kinase JAK1Homo sapiens (human)
metal ion bindingTyrosine-protein kinase JAK1Homo sapiens (human)
histone kinase activityCyclin-dependent kinase 2Homo sapiens (human)
magnesium ion bindingCyclin-dependent kinase 2Homo sapiens (human)
protein serine/threonine kinase activityCyclin-dependent kinase 2Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 2Homo sapiens (human)
protein bindingCyclin-dependent kinase 2Homo sapiens (human)
ATP bindingCyclin-dependent kinase 2Homo sapiens (human)
protein domain specific bindingCyclin-dependent kinase 2Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 2Homo sapiens (human)
cyclin-dependent protein kinase activityCyclin-dependent kinase 2Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 2Homo sapiens (human)
protein kinase activityBeta-adrenergic receptor kinase 1Homo sapiens (human)
G protein-coupled receptor kinase activityBeta-adrenergic receptor kinase 1Homo sapiens (human)
protein bindingBeta-adrenergic receptor kinase 1Homo sapiens (human)
ATP bindingBeta-adrenergic receptor kinase 1Homo sapiens (human)
alpha-2A adrenergic receptor bindingBeta-adrenergic receptor kinase 1Homo sapiens (human)
Edg-2 lysophosphatidic acid receptor bindingBeta-adrenergic receptor kinase 1Homo sapiens (human)
beta-adrenergic receptor kinase activityBeta-adrenergic receptor kinase 1Homo sapiens (human)
G protein-coupled receptor bindingBeta-adrenergic receptor kinase 1Homo sapiens (human)
RNA bindingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
RNA helicase activityProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
helicase activityProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
protein bindingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
ATP bindingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
ATP hydrolysis activityProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
protein domain specific bindingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
cadherin bindingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
mRNA bindingProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
phosphotyrosine residue bindingMitogen-activated protein kinase 3 Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 3 Homo sapiens (human)
MAP kinase activityMitogen-activated protein kinase 3 Homo sapiens (human)
protein bindingMitogen-activated protein kinase 3 Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 3 Homo sapiens (human)
phosphatase bindingMitogen-activated protein kinase 3 Homo sapiens (human)
identical protein bindingMitogen-activated protein kinase 3 Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 3 Homo sapiens (human)
DNA-binding transcription factor bindingMitogen-activated protein kinase 3 Homo sapiens (human)
protein serine/threonine kinase activityMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
protein bindingMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
ATP bindingMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
tau protein bindingMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
tau-protein kinase activityMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
protein serine kinase activityMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
deoxyadenosine kinase activityDeoxycytidine kinaseHomo sapiens (human)
deoxycytidine kinase activityDeoxycytidine kinaseHomo sapiens (human)
deoxyguanosine kinase activityDeoxycytidine kinaseHomo sapiens (human)
ATP bindingDeoxycytidine kinaseHomo sapiens (human)
protein homodimerization activityDeoxycytidine kinaseHomo sapiens (human)
cytidine kinase activityDeoxycytidine kinaseHomo sapiens (human)
phosphotyrosine residue bindingMitogen-activated protein kinase 1Homo sapiens (human)
DNA bindingMitogen-activated protein kinase 1Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 1Homo sapiens (human)
MAP kinase activityMitogen-activated protein kinase 1Homo sapiens (human)
protein bindingMitogen-activated protein kinase 1Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 1Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityMitogen-activated protein kinase 1Homo sapiens (human)
phosphatase bindingMitogen-activated protein kinase 1Homo sapiens (human)
identical protein bindingMitogen-activated protein kinase 1Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 1Homo sapiens (human)
virus receptor activityEphrin type-A receptor 2Homo sapiens (human)
transmembrane receptor protein tyrosine kinase activityEphrin type-A receptor 2Homo sapiens (human)
ephrin receptor activityEphrin type-A receptor 2Homo sapiens (human)
protein bindingEphrin type-A receptor 2Homo sapiens (human)
ATP bindingEphrin type-A receptor 2Homo sapiens (human)
growth factor bindingEphrin type-A receptor 2Homo sapiens (human)
cadherin bindingEphrin type-A receptor 2Homo sapiens (human)
molecular function activator activityEphrin type-A receptor 2Homo sapiens (human)
amyloid-beta bindingEphrin type-B receptor 2Homo sapiens (human)
protein tyrosine kinase activityEphrin type-B receptor 2Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-B receptor 2Homo sapiens (human)
signaling receptor bindingEphrin type-B receptor 2Homo sapiens (human)
protein bindingEphrin type-B receptor 2Homo sapiens (human)
ATP bindingEphrin type-B receptor 2Homo sapiens (human)
axon guidance receptor activityEphrin type-B receptor 2Homo sapiens (human)
identical protein bindingEphrin type-B receptor 2Homo sapiens (human)
protein-containing complex bindingEphrin type-B receptor 2Homo sapiens (human)
protein tyrosine kinase activityNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
growth hormone receptor bindingNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
protein bindingNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
ATP bindingNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
type 1 angiotensin receptor bindingNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
nucleoside diphosphate kinase activityUMP-CMP kinase Homo sapiens (human)
uridine kinase activityUMP-CMP kinase Homo sapiens (human)
ATP bindingUMP-CMP kinase Homo sapiens (human)
UMP kinase activityUMP-CMP kinase Homo sapiens (human)
CMP kinase activityUMP-CMP kinase Homo sapiens (human)
dCMP kinase activityUMP-CMP kinase Homo sapiens (human)
nucleoside monophosphate kinase activityUMP-CMP kinase Homo sapiens (human)
cytidylate kinase activityUMP-CMP kinase Homo sapiens (human)
RNA bindingPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
serine-type endopeptidase inhibitor activityPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
protein bindingPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
ATP bindingPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
phosphatidylethanolamine bindingPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
enzyme bindingPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
protein kinase bindingPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
magnesium ion bindingWee1-like protein kinaseHomo sapiens (human)
protein tyrosine kinase activityWee1-like protein kinaseHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityWee1-like protein kinaseHomo sapiens (human)
protein bindingWee1-like protein kinaseHomo sapiens (human)
ATP bindingWee1-like protein kinaseHomo sapiens (human)
heme oxygenase (decyclizing) activityHeme oxygenase 2Homo sapiens (human)
protein bindingHeme oxygenase 2Homo sapiens (human)
metal ion bindingHeme oxygenase 2Homo sapiens (human)
heme bindingHeme oxygenase 2Homo sapiens (human)
methionine adenosyltransferase activityS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
protein bindingS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
ATP bindingS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
small molecule bindingS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
identical protein bindingS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
metal ion bindingS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
G protein-coupled receptor bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
ATPase activator activityDnaJ homolog subfamily A member 1Homo sapiens (human)
protein bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
ATP bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
Hsp70 protein bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
Tat protein bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
ubiquitin protein ligase bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
metal ion bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
low-density lipoprotein particle receptor bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
unfolded protein bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
protein-folding chaperone bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
C3HC4-type RING finger domain bindingDnaJ homolog subfamily A member 1Homo sapiens (human)
protein kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein serine/threonine kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein serine/threonine/tyrosine kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
calmodulin bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
ATP bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
phosphatidylinositol-3,4,5-trisphosphate bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
enzyme bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein kinase bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
nitric-oxide synthase regulator activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein serine/threonine kinase inhibitor activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
identical protein bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein homodimerization activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
phosphatidylinositol-3,4-bisphosphate bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
14-3-3 protein bindingRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
potassium channel activator activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein serine kinase activityRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein serine/threonine kinase activityRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
protein bindingRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
ATP bindingRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
metal ion bindingRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
protein serine kinase activityRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
molecular function activator activityRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
protein tyrosine kinase activityDual specificity protein kinase TTKHomo sapiens (human)
protein bindingDual specificity protein kinase TTKHomo sapiens (human)
ATP bindingDual specificity protein kinase TTKHomo sapiens (human)
identical protein bindingDual specificity protein kinase TTKHomo sapiens (human)
kinetochore bindingDual specificity protein kinase TTKHomo sapiens (human)
protein serine kinase activityDual specificity protein kinase TTKHomo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity protein kinase TTKHomo sapiens (human)
protein serine/threonine kinase activityDual specificity protein kinase TTKHomo sapiens (human)
DNA helicase activityDNA replication licensing factor MCM4Homo sapiens (human)
single-stranded DNA bindingDNA replication licensing factor MCM4Homo sapiens (human)
protein bindingDNA replication licensing factor MCM4Homo sapiens (human)
ATP bindingDNA replication licensing factor MCM4Homo sapiens (human)
ATP hydrolysis activityDNA replication licensing factor MCM4Homo sapiens (human)
single-stranded DNA helicase activityDNA replication licensing factor MCM4Homo sapiens (human)
microfilament motor activityMyosin-10Homo sapiens (human)
actin filament bindingMyosin-10Homo sapiens (human)
microfilament motor activityMyosin-10Homo sapiens (human)
actin bindingMyosin-10Homo sapiens (human)
protein bindingMyosin-10Homo sapiens (human)
calmodulin bindingMyosin-10Homo sapiens (human)
ATP bindingMyosin-10Homo sapiens (human)
RNA stem-loop bindingMyosin-10Homo sapiens (human)
ADP bindingMyosin-10Homo sapiens (human)
mRNA 5'-UTR bindingMyosin-10Homo sapiens (human)
actin filament bindingMyosin-10Homo sapiens (human)
protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
MAP kinase kinase activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
protein tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
MAP-kinase scaffold activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
protein bindingDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
ATP bindingDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
PDZ domain bindingDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
protein serine/threonine kinase activator activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
metal ion bindingDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
scaffold protein bindingDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
protein serine kinase activityDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
protein tyrosine kinase activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
transmembrane receptor protein tyrosine kinase activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
cytokine receptor activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
vascular endothelial growth factor receptor activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
protein bindingReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
ATP bindingReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
nuclear glucocorticoid receptor bindingReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
protein-containing complex bindingReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
phosphatidylinositol 3-kinase activator activityReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
growth factor bindingReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
protein serine/threonine kinase activityBone morphogenetic protein receptor type-1AHomo sapiens (human)
transmembrane receptor protein serine/threonine kinase activityBone morphogenetic protein receptor type-1AHomo sapiens (human)
transforming growth factor beta receptor activity, type IBone morphogenetic protein receptor type-1AHomo sapiens (human)
protein bindingBone morphogenetic protein receptor type-1AHomo sapiens (human)
ATP bindingBone morphogenetic protein receptor type-1AHomo sapiens (human)
BMP bindingBone morphogenetic protein receptor type-1AHomo sapiens (human)
protein homodimerization activityBone morphogenetic protein receptor type-1AHomo sapiens (human)
SMAD bindingBone morphogenetic protein receptor type-1AHomo sapiens (human)
metal ion bindingBone morphogenetic protein receptor type-1AHomo sapiens (human)
BMP receptor activityBone morphogenetic protein receptor type-1AHomo sapiens (human)
activin receptor activityActivin receptor type-1BHomo sapiens (human)
growth factor bindingActivin receptor type-1BHomo sapiens (human)
activin bindingActivin receptor type-1BHomo sapiens (human)
protein serine/threonine kinase activityActivin receptor type-1BHomo sapiens (human)
transmembrane receptor protein serine/threonine kinase activityActivin receptor type-1BHomo sapiens (human)
protein bindingActivin receptor type-1BHomo sapiens (human)
ATP bindingActivin receptor type-1BHomo sapiens (human)
activin receptor activity, type IActivin receptor type-1BHomo sapiens (human)
activin receptor activityActivin receptor type-1BHomo sapiens (human)
ubiquitin protein ligase bindingActivin receptor type-1BHomo sapiens (human)
inhibin bindingActivin receptor type-1BHomo sapiens (human)
SMAD bindingActivin receptor type-1BHomo sapiens (human)
metal ion bindingActivin receptor type-1BHomo sapiens (human)
I-SMAD bindingActivin receptor type-1BHomo sapiens (human)
transforming growth factor beta receptor activityTGF-beta receptor type-1Homo sapiens (human)
growth factor bindingTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta bindingTGF-beta receptor type-1Homo sapiens (human)
protein kinase activityTGF-beta receptor type-1Homo sapiens (human)
protein serine/threonine kinase activityTGF-beta receptor type-1Homo sapiens (human)
transmembrane receptor protein serine/threonine kinase activityTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta receptor activityTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta receptor activity, type ITGF-beta receptor type-1Homo sapiens (human)
type II transforming growth factor beta receptor bindingTGF-beta receptor type-1Homo sapiens (human)
protein bindingTGF-beta receptor type-1Homo sapiens (human)
ATP bindingTGF-beta receptor type-1Homo sapiens (human)
ubiquitin protein ligase bindingTGF-beta receptor type-1Homo sapiens (human)
SMAD bindingTGF-beta receptor type-1Homo sapiens (human)
metal ion bindingTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta bindingTGF-beta receptor type-1Homo sapiens (human)
I-SMAD bindingTGF-beta receptor type-1Homo sapiens (human)
activin receptor activity, type ITGF-beta receptor type-1Homo sapiens (human)
activin bindingTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta bindingTGF-beta receptor type-2Homo sapiens (human)
transmembrane receptor protein serine/threonine kinase activityTGF-beta receptor type-2Homo sapiens (human)
transforming growth factor beta receptor activityTGF-beta receptor type-2Homo sapiens (human)
transforming growth factor beta receptor activity, type IITGF-beta receptor type-2Homo sapiens (human)
protein bindingTGF-beta receptor type-2Homo sapiens (human)
ATP bindingTGF-beta receptor type-2Homo sapiens (human)
glycosaminoglycan bindingTGF-beta receptor type-2Homo sapiens (human)
kinase activator activityTGF-beta receptor type-2Homo sapiens (human)
type I transforming growth factor beta receptor bindingTGF-beta receptor type-2Homo sapiens (human)
SMAD bindingTGF-beta receptor type-2Homo sapiens (human)
metal ion bindingTGF-beta receptor type-2Homo sapiens (human)
transforming growth factor beta bindingTGF-beta receptor type-2Homo sapiens (human)
molecular adaptor activityTGF-beta receptor type-2Homo sapiens (human)
activin receptor activityTGF-beta receptor type-2Homo sapiens (human)
activin bindingTGF-beta receptor type-2Homo sapiens (human)
protein serine/threonine kinase activityTGF-beta receptor type-2Homo sapiens (human)
protein bindingElectron transfer flavoprotein subunit betaHomo sapiens (human)
electron transfer activityElectron transfer flavoprotein subunit betaHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase CSKHomo sapiens (human)
protein bindingTyrosine-protein kinase CSKHomo sapiens (human)
ATP bindingTyrosine-protein kinase CSKHomo sapiens (human)
protein phosphatase bindingTyrosine-protein kinase CSKHomo sapiens (human)
protein kinase A catalytic subunit bindingTyrosine-protein kinase CSKHomo sapiens (human)
identical protein bindingTyrosine-protein kinase CSKHomo sapiens (human)
metal ion bindingTyrosine-protein kinase CSKHomo sapiens (human)
proline-rich region bindingTyrosine-protein kinase CSKHomo sapiens (human)
protein tyrosine kinase bindingTyrosine-protein kinase CSKHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase CSKHomo sapiens (human)
bis(5'-nucleosyl)-tetraphosphatase (asymmetrical) activityGlycine--tRNA ligaseHomo sapiens (human)
glycine-tRNA ligase activityGlycine--tRNA ligaseHomo sapiens (human)
protein bindingGlycine--tRNA ligaseHomo sapiens (human)
ATP bindingGlycine--tRNA ligaseHomo sapiens (human)
transferase activityGlycine--tRNA ligaseHomo sapiens (human)
identical protein bindingGlycine--tRNA ligaseHomo sapiens (human)
protein dimerization activityGlycine--tRNA ligaseHomo sapiens (human)
protein kinase activityProtein kinase C iota typeHomo sapiens (human)
protein serine/threonine kinase activityProtein kinase C iota typeHomo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activityProtein kinase C iota typeHomo sapiens (human)
protein bindingProtein kinase C iota typeHomo sapiens (human)
ATP bindingProtein kinase C iota typeHomo sapiens (human)
phospholipid bindingProtein kinase C iota typeHomo sapiens (human)
metal ion bindingProtein kinase C iota typeHomo sapiens (human)
protein serine kinase activityProtein kinase C iota typeHomo sapiens (human)
RNA bindingExosome RNA helicase MTR4Homo sapiens (human)
RNA helicase activityExosome RNA helicase MTR4Homo sapiens (human)
protein bindingExosome RNA helicase MTR4Homo sapiens (human)
ATP bindingExosome RNA helicase MTR4Homo sapiens (human)
ATP hydrolysis activityExosome RNA helicase MTR4Homo sapiens (human)
RNA polymerase III type 1 promoter sequence-specific DNA bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
RNA polymerase III type 2 promoter sequence-specific DNA bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
RNA polymerase III type 3 promoter sequence-specific DNA bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
TFIIIC-class transcription factor complex bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
protein kinase activitySerine/threonine-protein kinase mTORHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase mTORHomo sapiens (human)
protein bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
kinase activitySerine/threonine-protein kinase mTORHomo sapiens (human)
identical protein bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
ribosome bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
phosphoprotein bindingSerine/threonine-protein kinase mTORHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase mTORHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase TecHomo sapiens (human)
protein bindingTyrosine-protein kinase TecHomo sapiens (human)
ATP bindingTyrosine-protein kinase TecHomo sapiens (human)
phospholipid bindingTyrosine-protein kinase TecHomo sapiens (human)
metal ion bindingTyrosine-protein kinase TecHomo sapiens (human)
magnesium ion bindingTyrosine-protein kinase ABL2Homo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase ABL2Homo sapiens (human)
actin monomer bindingTyrosine-protein kinase ABL2Homo sapiens (human)
protein kinase activityTyrosine-protein kinase ABL2Homo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase ABL2Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase ABL2Homo sapiens (human)
protein bindingTyrosine-protein kinase ABL2Homo sapiens (human)
ATP bindingTyrosine-protein kinase ABL2Homo sapiens (human)
manganese ion bindingTyrosine-protein kinase ABL2Homo sapiens (human)
actin filament bindingTyrosine-protein kinase ABL2Homo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase FRKHomo sapiens (human)
protein bindingTyrosine-protein kinase FRKHomo sapiens (human)
ATP bindingTyrosine-protein kinase FRKHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase FRKHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase FRKHomo sapiens (human)
protein bindingG protein-coupled receptor kinase 6Homo sapiens (human)
ATP bindingG protein-coupled receptor kinase 6Homo sapiens (human)
beta-adrenergic receptor kinase activityG protein-coupled receptor kinase 6Homo sapiens (human)
G protein-coupled receptor kinase activityG protein-coupled receptor kinase 6Homo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase SYKHomo sapiens (human)
protein kinase activityTyrosine-protein kinase SYKHomo sapiens (human)
protein serine/threonine kinase activityTyrosine-protein kinase SYKHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase SYKHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase SYKHomo sapiens (human)
signaling receptor bindingTyrosine-protein kinase SYKHomo sapiens (human)
integrin bindingTyrosine-protein kinase SYKHomo sapiens (human)
protein bindingTyrosine-protein kinase SYKHomo sapiens (human)
ATP bindingTyrosine-protein kinase SYKHomo sapiens (human)
interleukin-15 receptor bindingTyrosine-protein kinase SYKHomo sapiens (human)
kinase activityTyrosine-protein kinase SYKHomo sapiens (human)
protein kinase bindingTyrosine-protein kinase SYKHomo sapiens (human)
phosphatase bindingTyrosine-protein kinase SYKHomo sapiens (human)
Toll-like receptor bindingTyrosine-protein kinase SYKHomo sapiens (human)
SH2 domain bindingTyrosine-protein kinase SYKHomo sapiens (human)
phospholipase bindingTyrosine-protein kinase SYKHomo sapiens (human)
scaffold protein bindingTyrosine-protein kinase SYKHomo sapiens (human)
protein binding26S proteasome regulatory subunit 6BHomo sapiens (human)
ATP binding26S proteasome regulatory subunit 6BHomo sapiens (human)
ATP hydrolysis activity26S proteasome regulatory subunit 6BHomo sapiens (human)
proteasome-activating activity26S proteasome regulatory subunit 6BHomo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 8Homo sapiens (human)
JUN kinase activityMitogen-activated protein kinase 8Homo sapiens (human)
protein bindingMitogen-activated protein kinase 8Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 8Homo sapiens (human)
enzyme bindingMitogen-activated protein kinase 8Homo sapiens (human)
protein phosphatase bindingMitogen-activated protein kinase 8Homo sapiens (human)
histone deacetylase regulator activityMitogen-activated protein kinase 8Homo sapiens (human)
histone deacetylase bindingMitogen-activated protein kinase 8Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 8Homo sapiens (human)
protein serine/threonine kinase bindingMitogen-activated protein kinase 8Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 9Homo sapiens (human)
JUN kinase activityMitogen-activated protein kinase 9Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityMitogen-activated protein kinase 9Homo sapiens (human)
protein bindingMitogen-activated protein kinase 9Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 9Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 9Homo sapiens (human)
protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
MAP kinase kinase activityDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
protein tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
protein bindingDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
ATP bindingDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
protein kinase bindingDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
protein serine kinase activityDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
protein bindingPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
ATP bindingPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
1-phosphatidylinositol-4-phosphate 5-kinase activityPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
1-phosphatidylinositol-5-phosphate 4-kinase activityPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
protein homodimerization activityPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
protein kinase activityCasein kinase I isoform alphaHomo sapiens (human)
protein serine/threonine kinase activityCasein kinase I isoform alphaHomo sapiens (human)
protein bindingCasein kinase I isoform alphaHomo sapiens (human)
ATP bindingCasein kinase I isoform alphaHomo sapiens (human)
protein serine kinase activityCasein kinase I isoform alphaHomo sapiens (human)
protein kinase activityCasein kinase I isoform deltaHomo sapiens (human)
protein serine/threonine kinase activityCasein kinase I isoform deltaHomo sapiens (human)
protein bindingCasein kinase I isoform deltaHomo sapiens (human)
ATP bindingCasein kinase I isoform deltaHomo sapiens (human)
cadherin bindingCasein kinase I isoform deltaHomo sapiens (human)
tau-protein kinase activityCasein kinase I isoform deltaHomo sapiens (human)
protein serine kinase activityCasein kinase I isoform deltaHomo sapiens (human)
protein kinase activityMAP kinase-activated protein kinase 2Homo sapiens (human)
protein serine/threonine kinase activityMAP kinase-activated protein kinase 2Homo sapiens (human)
protein bindingMAP kinase-activated protein kinase 2Homo sapiens (human)
ATP bindingMAP kinase-activated protein kinase 2Homo sapiens (human)
protein serine kinase activityMAP kinase-activated protein kinase 2Homo sapiens (human)
calcium-dependent protein serine/threonine kinase activityMAP kinase-activated protein kinase 2Homo sapiens (human)
calmodulin bindingMAP kinase-activated protein kinase 2Homo sapiens (human)
calmodulin-dependent protein kinase activityMAP kinase-activated protein kinase 2Homo sapiens (human)
mitogen-activated protein kinase bindingMAP kinase-activated protein kinase 2Homo sapiens (human)
RNA bindingElongation factor Tu, mitochondrialHomo sapiens (human)
translation elongation factor activityElongation factor Tu, mitochondrialHomo sapiens (human)
GTPase activityElongation factor Tu, mitochondrialHomo sapiens (human)
protein bindingElongation factor Tu, mitochondrialHomo sapiens (human)
GTP bindingElongation factor Tu, mitochondrialHomo sapiens (human)
choline-phosphate cytidylyltransferase activityCholine-phosphate cytidylyltransferase AHomo sapiens (human)
protein bindingCholine-phosphate cytidylyltransferase AHomo sapiens (human)
calmodulin bindingCholine-phosphate cytidylyltransferase AHomo sapiens (human)
identical protein bindingCholine-phosphate cytidylyltransferase AHomo sapiens (human)
protein homodimerization activityCholine-phosphate cytidylyltransferase AHomo sapiens (human)
molecular function inhibitor activityCholine-phosphate cytidylyltransferase AHomo sapiens (human)
phosphatidylcholine bindingCholine-phosphate cytidylyltransferase AHomo sapiens (human)
tRNA bindingCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
cysteine-tRNA ligase activityCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
protein bindingCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
ATP bindingCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
identical protein bindingCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
metal ion bindingCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
RNA bindingCasein kinase I isoform epsilonHomo sapiens (human)
protein kinase activityCasein kinase I isoform epsilonHomo sapiens (human)
protein serine/threonine kinase activityCasein kinase I isoform epsilonHomo sapiens (human)
protein bindingCasein kinase I isoform epsilonHomo sapiens (human)
ATP bindingCasein kinase I isoform epsilonHomo sapiens (human)
protein serine kinase activityCasein kinase I isoform epsilonHomo sapiens (human)
acyl-CoA dehydrogenase activityVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
long-chain fatty acyl-CoA dehydrogenase activityVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
protein bindingVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
very-long-chain fatty acyl-CoA dehydrogenase activityVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
identical protein bindingVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
flavin adenine dinucleotide bindingVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
fatty-acyl-CoA bindingVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
protein serine/threonine kinase activityDual specificity protein kinase CLK1Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity protein kinase CLK1Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityDual specificity protein kinase CLK1Homo sapiens (human)
protein bindingDual specificity protein kinase CLK1Homo sapiens (human)
ATP bindingDual specificity protein kinase CLK1Homo sapiens (human)
protein serine kinase activityDual specificity protein kinase CLK1Homo sapiens (human)
protein tyrosine kinase activityDual specificity protein kinase CLK1Homo sapiens (human)
protein serine/threonine kinase activityDual specificity protein kinase CLK2Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity protein kinase CLK2Homo sapiens (human)
protein bindingDual specificity protein kinase CLK2Homo sapiens (human)
ATP bindingDual specificity protein kinase CLK2Homo sapiens (human)
identical protein bindingDual specificity protein kinase CLK2Homo sapiens (human)
protein serine kinase activityDual specificity protein kinase CLK2Homo sapiens (human)
protein tyrosine kinase activityDual specificity protein kinase CLK2Homo sapiens (human)
RNA bindingDual specificity protein kinase CLK3Homo sapiens (human)
protein serine/threonine kinase activityDual specificity protein kinase CLK3Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity protein kinase CLK3Homo sapiens (human)
protein tyrosine kinase activityDual specificity protein kinase CLK3Homo sapiens (human)
protein bindingDual specificity protein kinase CLK3Homo sapiens (human)
ATP bindingDual specificity protein kinase CLK3Homo sapiens (human)
identical protein bindingDual specificity protein kinase CLK3Homo sapiens (human)
protein serine kinase activityDual specificity protein kinase CLK3Homo sapiens (human)
protein serine/threonine kinase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
signaling receptor bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
ATP bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein kinase A catalytic subunit bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
tau protein bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
tau-protein kinase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein serine kinase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
protease bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
p53 bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein serine/threonine kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
ATP bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
beta-catenin bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
ubiquitin protein ligase bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase A catalytic subunit bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
dynactin bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
tau protein bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
tau-protein kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
NF-kappaB bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
protein serine kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase activityCyclin-dependent kinase 7Homo sapiens (human)
protein serine/threonine kinase activityCyclin-dependent kinase 7Homo sapiens (human)
protein bindingCyclin-dependent kinase 7Homo sapiens (human)
ATP bindingCyclin-dependent kinase 7Homo sapiens (human)
ATP-dependent activity, acting on DNACyclin-dependent kinase 7Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityCyclin-dependent kinase 7Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 7Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 7Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCyclin-dependent kinase 9Homo sapiens (human)
transcription coactivator bindingCyclin-dependent kinase 9Homo sapiens (human)
DNA bindingCyclin-dependent kinase 9Homo sapiens (human)
chromatin bindingCyclin-dependent kinase 9Homo sapiens (human)
transcription elongation factor activityCyclin-dependent kinase 9Homo sapiens (human)
protein kinase activityCyclin-dependent kinase 9Homo sapiens (human)
protein serine/threonine kinase activityCyclin-dependent kinase 9Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 9Homo sapiens (human)
protein bindingCyclin-dependent kinase 9Homo sapiens (human)
ATP bindingCyclin-dependent kinase 9Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityCyclin-dependent kinase 9Homo sapiens (human)
kinase activityCyclin-dependent kinase 9Homo sapiens (human)
protein kinase bindingCyclin-dependent kinase 9Homo sapiens (human)
7SK snRNA bindingCyclin-dependent kinase 9Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 9Homo sapiens (human)
GTPase activityRas-related protein Rab-27AHomo sapiens (human)
G protein activityRas-related protein Rab-27AHomo sapiens (human)
protein bindingRas-related protein Rab-27AHomo sapiens (human)
GTP bindingRas-related protein Rab-27AHomo sapiens (human)
GDP bindingRas-related protein Rab-27AHomo sapiens (human)
protein domain specific bindingRas-related protein Rab-27AHomo sapiens (human)
myosin V bindingRas-related protein Rab-27AHomo sapiens (human)
protein kinase activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein serine/threonine kinase activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein bindingInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
ATP bindingInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
kinase activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein kinase bindingInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
heat shock protein bindingInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
identical protein bindingInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein homodimerization activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein heterodimerization activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein serine kinase activityInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
calmodulin bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
phosphatidylinositol-4,5-bisphosphate bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein phosphatase 1 bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
outward rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A catalytic subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A regulatory subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in atrial cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
magnesium ion bindingRibosomal protein S6 kinase alpha-3Homo sapiens (human)
protein kinase activityRibosomal protein S6 kinase alpha-3Homo sapiens (human)
protein serine/threonine kinase activityRibosomal protein S6 kinase alpha-3Homo sapiens (human)
protein bindingRibosomal protein S6 kinase alpha-3Homo sapiens (human)
ATP bindingRibosomal protein S6 kinase alpha-3Homo sapiens (human)
protein kinase bindingRibosomal protein S6 kinase alpha-3Homo sapiens (human)
cysteine-type endopeptidase inhibitor activity involved in apoptotic processRibosomal protein S6 kinase alpha-3Homo sapiens (human)
protein serine kinase activityRibosomal protein S6 kinase alpha-3Homo sapiens (human)
ribosomal protein S6 kinase activityRibosomal protein S6 kinase alpha-3Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase Nek2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase Nek2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase Nek2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase Nek2Homo sapiens (human)
protein phosphatase bindingSerine/threonine-protein kinase Nek2Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase Nek2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase Nek2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase Nek3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase Nek3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase Nek3Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase Nek3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase Nek3Homo sapiens (human)
protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
MAP kinase kinase activityDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
protein tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
protein bindingDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
ATP bindingDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
protein kinase bindingDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
protein serine kinase activityDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase PLK1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase PLK1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
microtubule bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
anaphase-promoting complex bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein kinase bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase PLK1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase PLK1Homo sapiens (human)
protein kinase activityLIM domain kinase 1Homo sapiens (human)
protein serine/threonine kinase activityLIM domain kinase 1Homo sapiens (human)
protein bindingLIM domain kinase 1Homo sapiens (human)
ATP bindingLIM domain kinase 1Homo sapiens (human)
heat shock protein bindingLIM domain kinase 1Homo sapiens (human)
metal ion bindingLIM domain kinase 1Homo sapiens (human)
protein serine kinase activityLIM domain kinase 1Homo sapiens (human)
protein serine/threonine kinase activityLIM domain kinase 2Homo sapiens (human)
protein bindingLIM domain kinase 2Homo sapiens (human)
ATP bindingLIM domain kinase 2Homo sapiens (human)
metal ion bindingLIM domain kinase 2Homo sapiens (human)
protein serine kinase activityLIM domain kinase 2Homo sapiens (human)
JUN kinase activityMitogen-activated protein kinase 10Homo sapiens (human)
MAP kinase kinase activityMitogen-activated protein kinase 10Homo sapiens (human)
protein bindingMitogen-activated protein kinase 10Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 10Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 10Homo sapiens (human)
tRNA bindingTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
RNA bindingTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
tyrosine-tRNA ligase activityTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
interleukin-8 receptor bindingTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
protein bindingTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
ATP bindingTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
small molecule bindingTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
protein kinase activity5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
cAMP-dependent protein kinase activity5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
protein binding5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
ATP binding5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
cAMP-dependent protein kinase regulator activity5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
AMP binding5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
protein kinase regulator activity5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
protein kinase binding5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
ADP binding5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
ephrin receptor activityEphrin type-B receptor 3Homo sapiens (human)
protein bindingEphrin type-B receptor 3Homo sapiens (human)
ATP bindingEphrin type-B receptor 3Homo sapiens (human)
axon guidance receptor activityEphrin type-B receptor 3Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-B receptor 3Homo sapiens (human)
ephrin receptor activityEphrin type-A receptor 5Homo sapiens (human)
GPI-linked ephrin receptor activityEphrin type-A receptor 5Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-A receptor 5Homo sapiens (human)
protein bindingEphrin type-A receptor 5Homo sapiens (human)
ATP bindingEphrin type-A receptor 5Homo sapiens (human)
transmembrane receptor protein tyrosine kinase activityEphrin type-B receptor 4Homo sapiens (human)
ephrin receptor activityEphrin type-B receptor 4Homo sapiens (human)
protein bindingEphrin type-B receptor 4Homo sapiens (human)
ATP bindingEphrin type-B receptor 4Homo sapiens (human)
amyloid-beta bindingEphrin type-A receptor 4Homo sapiens (human)
protein kinase activityEphrin type-A receptor 4Homo sapiens (human)
protein tyrosine kinase activityEphrin type-A receptor 4Homo sapiens (human)
GPI-linked ephrin receptor activityEphrin type-A receptor 4Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-A receptor 4Homo sapiens (human)
protein bindingEphrin type-A receptor 4Homo sapiens (human)
ATP bindingEphrin type-A receptor 4Homo sapiens (human)
kinase activityEphrin type-A receptor 4Homo sapiens (human)
PH domain bindingEphrin type-A receptor 4Homo sapiens (human)
identical protein bindingEphrin type-A receptor 4Homo sapiens (human)
ephrin receptor bindingEphrin type-A receptor 4Homo sapiens (human)
DH domain bindingEphrin type-A receptor 4Homo sapiens (human)
protein tyrosine kinase bindingEphrin type-A receptor 4Homo sapiens (human)
adenylate kinase activityAdenylate kinase 2, mitochondrialHomo sapiens (human)
protein bindingAdenylate kinase 2, mitochondrialHomo sapiens (human)
ATP bindingAdenylate kinase 2, mitochondrialHomo sapiens (human)
RNA bindingAdenosine kinaseHomo sapiens (human)
deoxyadenosine kinase activityAdenosine kinaseHomo sapiens (human)
ATP bindingAdenosine kinaseHomo sapiens (human)
metal ion bindingAdenosine kinaseHomo sapiens (human)
adenosine kinase activityAdenosine kinaseHomo sapiens (human)
G protein activityRas-related protein Rab-10Homo sapiens (human)
protein bindingRas-related protein Rab-10Homo sapiens (human)
GTP bindingRas-related protein Rab-10Homo sapiens (human)
GDP bindingRas-related protein Rab-10Homo sapiens (human)
myosin V bindingRas-related protein Rab-10Homo sapiens (human)
cadherin binding involved in cell-cell adhesionRas-related protein Rab-10Homo sapiens (human)
actin filament bindingActin-related protein 3Homo sapiens (human)
structural constituent of cytoskeletonActin-related protein 3Homo sapiens (human)
protein bindingActin-related protein 3Homo sapiens (human)
ATP bindingActin-related protein 3Homo sapiens (human)
actin filament bindingActin-related protein 2Homo sapiens (human)
structural constituent of cytoskeletonActin-related protein 2Homo sapiens (human)
protein bindingActin-related protein 2Homo sapiens (human)
ATP bindingActin-related protein 2Homo sapiens (human)
nuclear export signal receptor activityGTP-binding nuclear protein RanHomo sapiens (human)
pre-miRNA bindingGTP-binding nuclear protein RanHomo sapiens (human)
magnesium ion bindingGTP-binding nuclear protein RanHomo sapiens (human)
chromatin bindingGTP-binding nuclear protein RanHomo sapiens (human)
RNA bindingGTP-binding nuclear protein RanHomo sapiens (human)
GTPase activityGTP-binding nuclear protein RanHomo sapiens (human)
G protein activityGTP-binding nuclear protein RanHomo sapiens (human)
protein bindingGTP-binding nuclear protein RanHomo sapiens (human)
GTP bindingGTP-binding nuclear protein RanHomo sapiens (human)
GDP bindingGTP-binding nuclear protein RanHomo sapiens (human)
protein domain specific bindingGTP-binding nuclear protein RanHomo sapiens (human)
cadherin bindingGTP-binding nuclear protein RanHomo sapiens (human)
dynein intermediate chain bindingGTP-binding nuclear protein RanHomo sapiens (human)
protein heterodimerization activityGTP-binding nuclear protein RanHomo sapiens (human)
importin-alpha family protein bindingGTP-binding nuclear protein RanHomo sapiens (human)
protein serine/threonine kinase activityCasein kinase I isoform gamma-2Homo sapiens (human)
protein bindingCasein kinase I isoform gamma-2Homo sapiens (human)
ATP bindingCasein kinase I isoform gamma-2Homo sapiens (human)
protein serine kinase activityCasein kinase I isoform gamma-2Homo sapiens (human)
protein bindingCyclin-dependent kinase 3Homo sapiens (human)
ATP bindingCyclin-dependent kinase 3Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 3Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 3Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 3Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 6Homo sapiens (human)
protein bindingCyclin-dependent kinase 6Homo sapiens (human)
ATP bindingCyclin-dependent kinase 6Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 6Homo sapiens (human)
FBXO family protein bindingCyclin-dependent kinase 6Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 6Homo sapiens (human)
microtubule bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
p53 bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
protein kinase activityCyclin-dependent-like kinase 5 Homo sapiens (human)
protein serine/threonine kinase activityCyclin-dependent-like kinase 5 Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent-like kinase 5 Homo sapiens (human)
ErbB-2 class receptor bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
protein bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
ATP bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
kinase activityCyclin-dependent-like kinase 5 Homo sapiens (human)
acetylcholine receptor activator activityCyclin-dependent-like kinase 5 Homo sapiens (human)
ErbB-3 class receptor bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
tau protein bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
tau-protein kinase activityCyclin-dependent-like kinase 5 Homo sapiens (human)
Hsp90 protein bindingCyclin-dependent-like kinase 5 Homo sapiens (human)
protein serine kinase activityCyclin-dependent-like kinase 5 Homo sapiens (human)
protein serine/threonine kinase activityCyclin-dependent kinase 16Homo sapiens (human)
protein bindingCyclin-dependent kinase 16Homo sapiens (human)
ATP bindingCyclin-dependent kinase 16Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 16Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 16Homo sapiens (human)
protein kinase activityCyclin-dependent kinase 17Homo sapiens (human)
protein bindingCyclin-dependent kinase 17Homo sapiens (human)
ATP bindingCyclin-dependent kinase 17Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 17Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 17Homo sapiens (human)
6-phosphofructokinase activityATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
protein bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
protein-containing complex bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
cadherin bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
metal ion bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
ATP bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
monosaccharide bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
AMP bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
identical protein bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
fructose-6-phosphate bindingATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
protein kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
MAP kinase kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
MAP-kinase scaffold activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein bindingDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
ATP bindingDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein kinase activator activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein serine/threonine kinase activator activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
scaffold protein bindingDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
protein serine kinase activityDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
DNA bindingDNA topoisomerase 2-betaHomo sapiens (human)
chromatin bindingDNA topoisomerase 2-betaHomo sapiens (human)
DNA topoisomerase type II (double strand cut, ATP-hydrolyzing) activityDNA topoisomerase 2-betaHomo sapiens (human)
protein bindingDNA topoisomerase 2-betaHomo sapiens (human)
ATP bindingDNA topoisomerase 2-betaHomo sapiens (human)
ribonucleoprotein complex bindingDNA topoisomerase 2-betaHomo sapiens (human)
metal ion bindingDNA topoisomerase 2-betaHomo sapiens (human)
protein kinase activityProtein kinase C theta typeHomo sapiens (human)
protein serine/threonine kinase activityProtein kinase C theta typeHomo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activityProtein kinase C theta typeHomo sapiens (human)
protein bindingProtein kinase C theta typeHomo sapiens (human)
ATP bindingProtein kinase C theta typeHomo sapiens (human)
metal ion bindingProtein kinase C theta typeHomo sapiens (human)
protein serine kinase activityProtein kinase C theta typeHomo sapiens (human)
activin receptor activity, type IActivin receptor type-1Homo sapiens (human)
protein kinase activityActivin receptor type-1Homo sapiens (human)
protein serine/threonine kinase activityActivin receptor type-1Homo sapiens (human)
transmembrane receptor protein serine/threonine kinase activityActivin receptor type-1Homo sapiens (human)
protein bindingActivin receptor type-1Homo sapiens (human)
ATP bindingActivin receptor type-1Homo sapiens (human)
peptide hormone bindingActivin receptor type-1Homo sapiens (human)
protein homodimerization activityActivin receptor type-1Homo sapiens (human)
cadherin bindingActivin receptor type-1Homo sapiens (human)
SMAD bindingActivin receptor type-1Homo sapiens (human)
metal ion bindingActivin receptor type-1Homo sapiens (human)
activin bindingActivin receptor type-1Homo sapiens (human)
transforming growth factor beta bindingActivin receptor type-1Homo sapiens (human)
BMP receptor activityActivin receptor type-1Homo sapiens (human)
protein tyrosine kinase bindingActivin receptor type-1Homo sapiens (human)
transforming growth factor beta receptor activity, type IActivin receptor type-1Homo sapiens (human)
macrophage colony-stimulating factor receptor activityMacrophage-stimulating protein receptorHomo sapiens (human)
protein bindingMacrophage-stimulating protein receptorHomo sapiens (human)
ATP bindingMacrophage-stimulating protein receptorHomo sapiens (human)
enzyme bindingMacrophage-stimulating protein receptorHomo sapiens (human)
transmembrane receptor protein tyrosine kinase activityMacrophage-stimulating protein receptorHomo sapiens (human)
actin bindingFocal adhesion kinase 1Homo sapiens (human)
protein tyrosine kinase activityFocal adhesion kinase 1Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityFocal adhesion kinase 1Homo sapiens (human)
protein tyrosine phosphatase activityFocal adhesion kinase 1Homo sapiens (human)
integrin bindingFocal adhesion kinase 1Homo sapiens (human)
protein bindingFocal adhesion kinase 1Homo sapiens (human)
ATP bindingFocal adhesion kinase 1Homo sapiens (human)
JUN kinase bindingFocal adhesion kinase 1Homo sapiens (human)
protein kinase bindingFocal adhesion kinase 1Homo sapiens (human)
protein phosphatase bindingFocal adhesion kinase 1Homo sapiens (human)
SH2 domain bindingFocal adhesion kinase 1Homo sapiens (human)
molecular function activator activityFocal adhesion kinase 1Homo sapiens (human)
protein kinase activityProtein kinase C delta typeHomo sapiens (human)
protein serine/threonine kinase activityProtein kinase C delta typeHomo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activityProtein kinase C delta typeHomo sapiens (human)
diacylglycerol-dependent, calcium-independent serine/threonine kinase activityProtein kinase C delta typeHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityProtein kinase C delta typeHomo sapiens (human)
protein bindingProtein kinase C delta typeHomo sapiens (human)
ATP bindingProtein kinase C delta typeHomo sapiens (human)
enzyme activator activityProtein kinase C delta typeHomo sapiens (human)
enzyme bindingProtein kinase C delta typeHomo sapiens (human)
protein kinase bindingProtein kinase C delta typeHomo sapiens (human)
insulin receptor substrate bindingProtein kinase C delta typeHomo sapiens (human)
metal ion bindingProtein kinase C delta typeHomo sapiens (human)
protein serine kinase activityProtein kinase C delta typeHomo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase BTKHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase BTKHomo sapiens (human)
protein bindingTyrosine-protein kinase BTKHomo sapiens (human)
ATP bindingTyrosine-protein kinase BTKHomo sapiens (human)
phosphatidylinositol-3,4,5-trisphosphate bindingTyrosine-protein kinase BTKHomo sapiens (human)
phospholipase activator activityTyrosine-protein kinase BTKHomo sapiens (human)
identical protein bindingTyrosine-protein kinase BTKHomo sapiens (human)
phospholipase bindingTyrosine-protein kinase BTKHomo sapiens (human)
metal ion bindingTyrosine-protein kinase BTKHomo sapiens (human)
protein serine/threonine kinase activityActivated CDC42 kinase 1Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityActivated CDC42 kinase 1Homo sapiens (human)
protein tyrosine kinase activityActivated CDC42 kinase 1Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityActivated CDC42 kinase 1Homo sapiens (human)
GTPase inhibitor activityActivated CDC42 kinase 1Homo sapiens (human)
epidermal growth factor receptor bindingActivated CDC42 kinase 1Homo sapiens (human)
protein bindingActivated CDC42 kinase 1Homo sapiens (human)
ATP bindingActivated CDC42 kinase 1Homo sapiens (human)
ubiquitin protein ligase bindingActivated CDC42 kinase 1Homo sapiens (human)
identical protein bindingActivated CDC42 kinase 1Homo sapiens (human)
metal ion bindingActivated CDC42 kinase 1Homo sapiens (human)
WW domain bindingActivated CDC42 kinase 1Homo sapiens (human)
protein serine kinase activityActivated CDC42 kinase 1Homo sapiens (human)
transmembrane receptor protein tyrosine kinase activityEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
protein bindingEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
collagen bindingEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
ATP bindingEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
protein tyrosine kinase collagen receptor activityEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
metal ion bindingEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
mitogen-activated protein kinase kinase kinase bindingMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
MAP kinase kinase kinase kinase activityMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase 4Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase 4Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase 4Homo sapiens (human)
protein bindingSerine/threonine-protein kinase 4Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase 4Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase 4Homo sapiens (human)
protein homodimerization activitySerine/threonine-protein kinase 4Homo sapiens (human)
protein serine/threonine kinase activator activitySerine/threonine-protein kinase 4Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingSerine/threonine-protein kinase 4Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase 4Homo sapiens (human)
chromatin binding5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein serine/threonine kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
AMP-activated protein kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cAMP-dependent protein kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein binding5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
ATP binding5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
metal ion binding5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
[hydroxymethylglutaryl-CoA reductase (NADPH)] kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
tau protein binding5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
tau-protein kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein serine kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
histone H2BS36 kinase activity5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
protein kinase activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
protein serine/threonine kinase activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
protein tyrosine kinase activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
protein bindingDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
ATP bindingDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
metal ion bindingDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
protein serine kinase activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
MAP kinase kinase activityDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
MAP kinase activityMitogen-activated protein kinase 7Homo sapiens (human)
enzyme inhibitor activityMitogen-activated protein kinase 7Homo sapiens (human)
protein bindingMitogen-activated protein kinase 7Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 7Homo sapiens (human)
mitogen-activated protein kinase bindingMitogen-activated protein kinase 7Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 7Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 7Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase PAK 2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein kinase bindingSerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein tyrosine kinase activator activitySerine/threonine-protein kinase PAK 2Homo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase PAK 2Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cadherin bindingSerine/threonine-protein kinase PAK 2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase PAK 2Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase 3Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase 3Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase 3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase 3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase 3Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase 3Homo sapiens (human)
protein serine/threonine kinase activator activitySerine/threonine-protein kinase 3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase 3Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
zinc ion bindingMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
protein kinase bindingMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
protein serine/threonine kinase activityIntegrin-linked protein kinaseHomo sapiens (human)
protein bindingIntegrin-linked protein kinaseHomo sapiens (human)
ATP bindingIntegrin-linked protein kinaseHomo sapiens (human)
protein kinase bindingIntegrin-linked protein kinaseHomo sapiens (human)
protein serine kinase activityIntegrin-linked protein kinaseHomo sapiens (human)
protein kinase activityRho-associated protein kinase 1Homo sapiens (human)
protein serine/threonine kinase activityRho-associated protein kinase 1Homo sapiens (human)
protein bindingRho-associated protein kinase 1Homo sapiens (human)
ATP bindingRho-associated protein kinase 1Homo sapiens (human)
small GTPase bindingRho-associated protein kinase 1Homo sapiens (human)
metal ion bindingRho-associated protein kinase 1Homo sapiens (human)
tau protein bindingRho-associated protein kinase 1Homo sapiens (human)
tau-protein kinase activityRho-associated protein kinase 1Homo sapiens (human)
Rho-dependent protein serine/threonine kinase activityRho-associated protein kinase 1Homo sapiens (human)
protein serine kinase activityRho-associated protein kinase 1Homo sapiens (human)
protein tyrosine kinase activityNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
protein bindingNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
ATP bindingNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
calcium-dependent protein serine/threonine phosphatase activityCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
protein bindingCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
calmodulin bindingCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
ATP bindingCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
identical protein bindingCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
protein homodimerization activityCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
protein serine kinase activityCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
calmodulin-dependent protein kinase activityCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
protein serine/threonine kinase activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
calmodulin-dependent protein kinase activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
protein bindingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
calmodulin bindingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
ATP bindingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
sodium channel inhibitor activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
titin bindingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
identical protein bindingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
protein homodimerization activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
transmembrane transporter bindingCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
protein serine kinase activityCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
protein kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein serine/threonine kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein tyrosine kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein bindingDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
ATP bindingDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
identical protein bindingDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
tau protein bindingDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
tau-protein kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
protein serine kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
histone H3T45 kinase activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
transcription coactivator activityDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
activin receptor activity, type IIActivin receptor type-2BHomo sapiens (human)
protein serine/threonine kinase activityActivin receptor type-2BHomo sapiens (human)
protein serine/threonine/tyrosine kinase activityActivin receptor type-2BHomo sapiens (human)
protein bindingActivin receptor type-2BHomo sapiens (human)
ATP bindingActivin receptor type-2BHomo sapiens (human)
activin receptor activity, type IIActivin receptor type-2BHomo sapiens (human)
kinase activator activityActivin receptor type-2BHomo sapiens (human)
growth factor bindingActivin receptor type-2BHomo sapiens (human)
metal ion bindingActivin receptor type-2BHomo sapiens (human)
activin bindingActivin receptor type-2BHomo sapiens (human)
activin receptor activityActivin receptor type-2BHomo sapiens (human)
protein bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
ATP bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
activin receptor activity, type IIBone morphogenetic protein receptor type-2Homo sapiens (human)
growth factor bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
BMP bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
cadherin bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
metal ion bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
BMP receptor activityBone morphogenetic protein receptor type-2Homo sapiens (human)
protein tyrosine kinase bindingBone morphogenetic protein receptor type-2Homo sapiens (human)
transforming growth factor beta receptor activityBone morphogenetic protein receptor type-2Homo sapiens (human)
protein tyrosine kinase activityProtein-tyrosine kinase 6Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityProtein-tyrosine kinase 6Homo sapiens (human)
protein bindingProtein-tyrosine kinase 6Homo sapiens (human)
ATP bindingProtein-tyrosine kinase 6Homo sapiens (human)
identical protein bindingProtein-tyrosine kinase 6Homo sapiens (human)
signaling receptor bindingProtein-tyrosine kinase 6Homo sapiens (human)
protein kinase activitycGMP-dependent protein kinase 1 Homo sapiens (human)
cGMP-dependent protein kinase activitycGMP-dependent protein kinase 1 Homo sapiens (human)
calcium channel regulator activitycGMP-dependent protein kinase 1 Homo sapiens (human)
protein bindingcGMP-dependent protein kinase 1 Homo sapiens (human)
ATP bindingcGMP-dependent protein kinase 1 Homo sapiens (human)
cGMP bindingcGMP-dependent protein kinase 1 Homo sapiens (human)
identical protein bindingcGMP-dependent protein kinase 1 Homo sapiens (human)
mitogen-activated protein kinase p38 bindingcGMP-dependent protein kinase 1 Homo sapiens (human)
protein serine kinase activitycGMP-dependent protein kinase 1 Homo sapiens (human)
RNA bindingCyclin-dependent kinase 13Homo sapiens (human)
protein kinase activityCyclin-dependent kinase 13Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 13Homo sapiens (human)
protein bindingCyclin-dependent kinase 13Homo sapiens (human)
ATP bindingCyclin-dependent kinase 13Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityCyclin-dependent kinase 13Homo sapiens (human)
protein kinase bindingCyclin-dependent kinase 13Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 13Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 13Homo sapiens (human)
K63-linked polyubiquitin modification-dependent protein bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
protein serine/threonine kinase activityInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
protein bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
ATP bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
IkappaB kinase activityInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
protein phosphatase bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
ubiquitin protein ligase bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
K48-linked polyubiquitin modification-dependent protein bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
identical protein bindingInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
calmodulin-dependent protein kinase activityProtein-tyrosine kinase 2-betaHomo sapiens (human)
protein tyrosine kinase activityProtein-tyrosine kinase 2-betaHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityProtein-tyrosine kinase 2-betaHomo sapiens (human)
protein bindingProtein-tyrosine kinase 2-betaHomo sapiens (human)
ATP bindingProtein-tyrosine kinase 2-betaHomo sapiens (human)
ubiquitin protein ligase bindingProtein-tyrosine kinase 2-betaHomo sapiens (human)
glutamate receptor bindingProtein-tyrosine kinase 2-betaHomo sapiens (human)
3-phosphoinositide-dependent protein kinase bindingProtein-tyrosine kinase 2-betaHomo sapiens (human)
protein-containing complex bindingProtein-tyrosine kinase 2-betaHomo sapiens (human)
neurotransmitter receptor regulator activityProtein-tyrosine kinase 2-betaHomo sapiens (human)
voltage-gated sodium channel activitySodium channel protein type 5 subunit alphaHomo sapiens (human)
protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
calmodulin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
fibroblast growth factor bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
enzyme bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein kinase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein domain specific bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ankyrin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ubiquitin protein ligase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
transmembrane transporter bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
nitric-oxide synthase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in Purkinje myocyte action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
scaffold protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein serine/threonine kinase activityMaternal embryonic leucine zipper kinaseHomo sapiens (human)
non-membrane spanning protein tyrosine kinase activityMaternal embryonic leucine zipper kinaseHomo sapiens (human)
calcium ion bindingMaternal embryonic leucine zipper kinaseHomo sapiens (human)
protein bindingMaternal embryonic leucine zipper kinaseHomo sapiens (human)
ATP bindingMaternal embryonic leucine zipper kinaseHomo sapiens (human)
lipid bindingMaternal embryonic leucine zipper kinaseHomo sapiens (human)
protein serine kinase activityMaternal embryonic leucine zipper kinaseHomo sapiens (human)
chromatin bindingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
RNA bindingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
protein bindingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
ATP bindingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
ATP hydrolysis activityStructural maintenance of chromosomes protein 1AHomo sapiens (human)
mediator complex bindingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
protein heterodimerization activityStructural maintenance of chromosomes protein 1AHomo sapiens (human)
DNA bindingStructural maintenance of chromosomes protein 1AHomo sapiens (human)
nucleosomal DNA bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
transcription coregulator bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
transcription corepressor activityChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
helicase activityChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
protein bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
ATP bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
zinc ion bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
ATP hydrolysis activityChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
histone deacetylase bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
ATP-dependent chromatin remodeler activityChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
chromatin bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
DNA bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
histone bindingChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
acyl-CoA oxidase activityPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
protein bindingPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
PDZ domain bindingPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
protein homodimerization activityPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
FAD bindingPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
fatty acid bindingPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
flavin adenine dinucleotide bindingPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
palmitoyl-CoA oxidase activityPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
protein tyrosine kinase activityEphrin type-A receptor 7Homo sapiens (human)
GPI-linked ephrin receptor activityEphrin type-A receptor 7Homo sapiens (human)
protein bindingEphrin type-A receptor 7Homo sapiens (human)
ATP bindingEphrin type-A receptor 7Homo sapiens (human)
axon guidance receptor activityEphrin type-A receptor 7Homo sapiens (human)
growth factor bindingEphrin type-A receptor 7Homo sapiens (human)
chemorepellent activityEphrin type-A receptor 7Homo sapiens (human)
transmembrane-ephrin receptor activityEphrin type-A receptor 7Homo sapiens (human)
delta24(24-1) sterol reductase activityDelta(24)-sterol reductaseHomo sapiens (human)
protein bindingDelta(24)-sterol reductaseHomo sapiens (human)
oxidoreductase activity, acting on the CH-CH group of donors, NAD or NADP as acceptorDelta(24)-sterol reductaseHomo sapiens (human)
enzyme bindingDelta(24)-sterol reductaseHomo sapiens (human)
peptide antigen bindingDelta(24)-sterol reductaseHomo sapiens (human)
delta24-sterol reductase activityDelta(24)-sterol reductaseHomo sapiens (human)
FAD bindingDelta(24)-sterol reductaseHomo sapiens (human)
magnesium ion bindingRibosomal protein S6 kinase alpha-1Homo sapiens (human)
protein serine/threonine kinase activityRibosomal protein S6 kinase alpha-1Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityRibosomal protein S6 kinase alpha-1Homo sapiens (human)
protein bindingRibosomal protein S6 kinase alpha-1Homo sapiens (human)
ATP bindingRibosomal protein S6 kinase alpha-1Homo sapiens (human)
cysteine-type endopeptidase inhibitor activity involved in apoptotic processRibosomal protein S6 kinase alpha-1Homo sapiens (human)
protein serine kinase activityRibosomal protein S6 kinase alpha-1Homo sapiens (human)
ribosomal protein S6 kinase activityRibosomal protein S6 kinase alpha-1Homo sapiens (human)
protein kinase activityDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein tyrosine kinase activityDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein bindingDual specificity testis-specific protein kinase 1Homo sapiens (human)
ATP bindingDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein kinase bindingDual specificity testis-specific protein kinase 1Homo sapiens (human)
metal ion bindingDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein serine kinase activityDual specificity testis-specific protein kinase 1Homo sapiens (human)
protein serine/threonine kinase activityDual specificity testis-specific protein kinase 1Homo sapiens (human)
actin bindingMyosin light chain kinase, smooth muscleHomo sapiens (human)
myosin light chain kinase activityMyosin light chain kinase, smooth muscleHomo sapiens (human)
protein bindingMyosin light chain kinase, smooth muscleHomo sapiens (human)
calmodulin bindingMyosin light chain kinase, smooth muscleHomo sapiens (human)
ATP bindingMyosin light chain kinase, smooth muscleHomo sapiens (human)
metal ion bindingMyosin light chain kinase, smooth muscleHomo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 11Homo sapiens (human)
MAP kinase activityMitogen-activated protein kinase 11Homo sapiens (human)
protein bindingMitogen-activated protein kinase 11Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 11Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 11Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase STK11Homo sapiens (human)
p53 bindingSerine/threonine-protein kinase STK11Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase STK11Homo sapiens (human)
protein bindingSerine/threonine-protein kinase STK11Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase STK11Homo sapiens (human)
LRR domain bindingSerine/threonine-protein kinase STK11Homo sapiens (human)
protein kinase activator activitySerine/threonine-protein kinase STK11Homo sapiens (human)
protein-containing complex bindingSerine/threonine-protein kinase STK11Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase STK11Homo sapiens (human)
chromatin bindingSerine/threonine-protein kinase N1Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase N1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase N1Homo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activitySerine/threonine-protein kinase N1Homo sapiens (human)
protein kinase C bindingSerine/threonine-protein kinase N1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase N1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase N1Homo sapiens (human)
nuclear receptor coactivator activitySerine/threonine-protein kinase N1Homo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase N1Homo sapiens (human)
histone H3T11 kinase activitySerine/threonine-protein kinase N1Homo sapiens (human)
histone bindingSerine/threonine-protein kinase N1Homo sapiens (human)
histone deacetylase bindingSerine/threonine-protein kinase N1Homo sapiens (human)
nuclear androgen receptor bindingSerine/threonine-protein kinase N1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase N1Homo sapiens (human)
RNA bindingSerine/threonine-protein kinase N2Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase N2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase N2Homo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activitySerine/threonine-protein kinase N2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase N2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase N2Homo sapiens (human)
kinase activitySerine/threonine-protein kinase N2Homo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase N2Homo sapiens (human)
histone deacetylase bindingSerine/threonine-protein kinase N2Homo sapiens (human)
cadherin bindingSerine/threonine-protein kinase N2Homo sapiens (human)
RNA polymerase bindingSerine/threonine-protein kinase N2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase N2Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 14Homo sapiens (human)
MAP kinase activityMitogen-activated protein kinase 14Homo sapiens (human)
MAP kinase kinase activityMitogen-activated protein kinase 14Homo sapiens (human)
protein bindingMitogen-activated protein kinase 14Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 14Homo sapiens (human)
enzyme bindingMitogen-activated protein kinase 14Homo sapiens (human)
protein phosphatase bindingMitogen-activated protein kinase 14Homo sapiens (human)
mitogen-activated protein kinase p38 bindingMitogen-activated protein kinase 14Homo sapiens (human)
NFAT protein bindingMitogen-activated protein kinase 14Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 14Homo sapiens (human)
calmodulin-dependent protein kinase activityCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
ATP bindingCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
protein serine kinase activityCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
calmodulin bindingCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
calcium-dependent protein serine/threonine kinase activityCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
JUN kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
small GTPase bindingMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
mitogen-activated protein kinase kinase bindingMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
mitogen-activated protein kinase kinase kinase bindingMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
identical protein bindingMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
protein homodimerization activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
transmembrane receptor protein tyrosine kinase activityDiscoidin domain-containing receptor 2Homo sapiens (human)
protein bindingDiscoidin domain-containing receptor 2Homo sapiens (human)
collagen bindingDiscoidin domain-containing receptor 2Homo sapiens (human)
ATP bindingDiscoidin domain-containing receptor 2Homo sapiens (human)
protein tyrosine kinase collagen receptor activityDiscoidin domain-containing receptor 2Homo sapiens (human)
protein serine/threonine kinase activityAP2-associated protein kinase 1Homo sapiens (human)
Notch bindingAP2-associated protein kinase 1Homo sapiens (human)
protein bindingAP2-associated protein kinase 1Homo sapiens (human)
ATP bindingAP2-associated protein kinase 1Homo sapiens (human)
AP-2 adaptor complex bindingAP2-associated protein kinase 1Homo sapiens (human)
protein serine kinase activityAP2-associated protein kinase 1Homo sapiens (human)
calmodulin-dependent protein kinase activityMyosin light chain kinase 3Homo sapiens (human)
myosin light chain kinase activityMyosin light chain kinase 3Homo sapiens (human)
protein bindingMyosin light chain kinase 3Homo sapiens (human)
ATP bindingMyosin light chain kinase 3Homo sapiens (human)
molecular_functionPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
ATP hydrolysis activityPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
ATP-dependent protein folding chaperonePutative heat shock protein HSP 90-beta 2Homo sapiens (human)
disordered domain specific bindingPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
ATP bindingPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
unfolded protein bindingPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
protein bindingSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
identical protein bindingSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
protein bindingSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
acyl-CoA dehydrogenase activityAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
flavin adenine dinucleotide bindingAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase N3Homo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activitySerine/threonine-protein kinase N3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase N3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase N3Homo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase N3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase N3Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase N3Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase ULK3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase ULK3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase ULK3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase ULK3Homo sapiens (human)
acyl-CoA dehydrogenase activityAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
long-chain fatty acyl-CoA dehydrogenase activityAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
protein bindingAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
very-long-chain fatty acyl-CoA dehydrogenase activityAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
flavin adenine dinucleotide bindingAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
medium-chain fatty acyl-CoA dehydrogenase activityAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
endonuclease activitySerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
RNA endonuclease activitySerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
unfolded protein bindingSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
RNA bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase MARK2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
lipid bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
protein kinase activator activitySerine/threonine-protein kinase MARK2Homo sapiens (human)
cadherin bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
tau protein bindingSerine/threonine-protein kinase MARK2Homo sapiens (human)
tau-protein kinase activitySerine/threonine-protein kinase MARK2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase MARK2Homo sapiens (human)
chromatin bindingATP-dependent RNA helicase DHX30Homo sapiens (human)
RNA bindingATP-dependent RNA helicase DHX30Homo sapiens (human)
RNA helicase activityATP-dependent RNA helicase DHX30Homo sapiens (human)
double-stranded RNA bindingATP-dependent RNA helicase DHX30Homo sapiens (human)
protein bindingATP-dependent RNA helicase DHX30Homo sapiens (human)
ATP bindingATP-dependent RNA helicase DHX30Homo sapiens (human)
ATP hydrolysis activityATP-dependent RNA helicase DHX30Homo sapiens (human)
G-quadruplex RNA bindingATP-dependent RNA helicase DHX30Homo sapiens (human)
DNA helicase activityATP-dependent RNA helicase DHX30Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase TAO1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase TAO1Homo sapiens (human)
kinase activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
transferase activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
alpha-tubulin bindingSerine/threonine-protein kinase TAO1Homo sapiens (human)
protein serine/threonine kinase activator activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
tau protein bindingSerine/threonine-protein kinase TAO1Homo sapiens (human)
beta-tubulin bindingSerine/threonine-protein kinase TAO1Homo sapiens (human)
tau-protein kinase activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase TAO1Homo sapiens (human)
protein kinase activitySTE20-related kinase adapter protein alphaHomo sapiens (human)
protein bindingSTE20-related kinase adapter protein alphaHomo sapiens (human)
ATP bindingSTE20-related kinase adapter protein alphaHomo sapiens (human)
kinase bindingSTE20-related kinase adapter protein alphaHomo sapiens (human)
protein kinase activator activitySTE20-related kinase adapter protein alphaHomo sapiens (human)
protein serine/threonine kinase activator activitySTE20-related kinase adapter protein alphaHomo sapiens (human)
microfilament motor activityMyosin-14Homo sapiens (human)
actin filament bindingMyosin-14Homo sapiens (human)
calmodulin bindingMyosin-14Homo sapiens (human)
ATP bindingMyosin-14Homo sapiens (human)
protein serine/threonine kinase activityAarF domain-containing protein kinase 1Homo sapiens (human)
ATP bindingAarF domain-containing protein kinase 1Homo sapiens (human)
RNA bindingATP-dependent RNA helicase DDX42Homo sapiens (human)
RNA helicase activityATP-dependent RNA helicase DDX42Homo sapiens (human)
protein bindingATP-dependent RNA helicase DDX42Homo sapiens (human)
ATP bindingATP-dependent RNA helicase DDX42Homo sapiens (human)
ATP hydrolysis activityATP-dependent RNA helicase DDX42Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
MAP kinase kinase kinase kinase activityMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
RNA bindingEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
translation release factor activityEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
GTPase activityEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
protein bindingEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
GTP bindingEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
protein bindingRegulatory-associated protein of mTORHomo sapiens (human)
protein kinase bindingRegulatory-associated protein of mTORHomo sapiens (human)
protein serine/threonine kinase inhibitor activityRegulatory-associated protein of mTORHomo sapiens (human)
protein kinase activator activityRegulatory-associated protein of mTORHomo sapiens (human)
protein-macromolecule adaptor activityRegulatory-associated protein of mTORHomo sapiens (human)
small GTPase bindingRegulatory-associated protein of mTORHomo sapiens (human)
protein-containing complex bindingRegulatory-associated protein of mTORHomo sapiens (human)
14-3-3 protein bindingRegulatory-associated protein of mTORHomo sapiens (human)
enzyme-substrate adaptor activityRegulatory-associated protein of mTORHomo sapiens (human)
protein kinase activityAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
protein bindingAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
ATP bindingAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
kinase activityAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
ADP bindingAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
protein bindingPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
ATP bindingPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
1-phosphatidylinositol-4-phosphate 5-kinase activityPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
identical protein bindingPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
1-phosphatidylinositol-5-phosphate 4-kinase activityPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
chromatin bindingMitogen-activated protein kinase 15Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase 15Homo sapiens (human)
MAP kinase activityMitogen-activated protein kinase 15Homo sapiens (human)
protein bindingMitogen-activated protein kinase 15Homo sapiens (human)
ATP bindingMitogen-activated protein kinase 15Homo sapiens (human)
kinase activityMitogen-activated protein kinase 15Homo sapiens (human)
SH3 domain bindingMitogen-activated protein kinase 15Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase 15Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase 15Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase Nek9Homo sapiens (human)
protein bindingSerine/threonine-protein kinase Nek9Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase Nek9Homo sapiens (human)
protein kinase bindingSerine/threonine-protein kinase Nek9Homo sapiens (human)
protein kinase activator activitySerine/threonine-protein kinase Nek9Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase Nek9Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase Nek9Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase Nek7Homo sapiens (human)
protein bindingSerine/threonine-protein kinase Nek7Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase Nek7Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase Nek7Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase Nek7Homo sapiens (human)
molecular function activator activitySerine/threonine-protein kinase Nek7Homo sapiens (human)
DNA bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
chromatin bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
transcription coregulator activityATP-dependent RNA helicase DDX1Homo sapiens (human)
RNA bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
RNA helicase activityATP-dependent RNA helicase DDX1Homo sapiens (human)
double-stranded RNA bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
nuclease activityATP-dependent RNA helicase DDX1Homo sapiens (human)
exonuclease activityATP-dependent RNA helicase DDX1Homo sapiens (human)
protein bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
ATP bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
poly(A) bindingATP-dependent RNA helicase DDX1Homo sapiens (human)
ATP hydrolysis activityATP-dependent RNA helicase DDX1Homo sapiens (human)
DNA/RNA helicase activityATP-dependent RNA helicase DDX1Homo sapiens (human)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
MAP kinase kinase kinase kinase activityMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
protein serine/threonine kinase activityAurora kinase BHomo sapiens (human)
protein serine/threonine kinase activityAurora kinase BHomo sapiens (human)
protein serine/threonine/tyrosine kinase activityAurora kinase BHomo sapiens (human)
protein bindingAurora kinase BHomo sapiens (human)
ATP bindingAurora kinase BHomo sapiens (human)
kinase bindingAurora kinase BHomo sapiens (human)
protein serine kinase activityAurora kinase BHomo sapiens (human)
protein serine/threonine kinase activityMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
protein bindingMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
ATP bindingMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
microtubule bindingMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
cytoskeletal anchor activityMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
gamma-tubulin bindingMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
ubiquitin bindingMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
tau protein bindingMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
tau-protein kinase activityMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
protein serine kinase activityMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase Nek1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase Nek1Homo sapiens (human)
protein tyrosine kinase activitySerine/threonine-protein kinase Nek1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase Nek1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase Nek1Homo sapiens (human)
kinase activitySerine/threonine-protein kinase Nek1Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase Nek1Homo sapiens (human)
14-3-3 protein bindingSerine/threonine-protein kinase Nek1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase Nek1Homo sapiens (human)
protein bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
ATP bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
ATP hydrolysis activityATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA clamp unloader activityATPase family AAA domain-containing protein 5Homo sapiens (human)
DNA bindingATPase family AAA domain-containing protein 5Homo sapiens (human)
protein serine/threonine kinase activityPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
protein bindingPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
ATP bindingPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
phosphatidylinositol bindingPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
protein serine kinase activityPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
calmodulin-dependent protein kinase activityCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
protein tyrosine kinase activityCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
calcium ion bindingCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
calmodulin bindingCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
ATP bindingCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
protein serine kinase activityCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
protein serine/threonine kinase activityCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
p53 bindingEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
protein serine/threonine kinase activityEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
protein bindingEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
ATP bindingEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
hydrolase activityEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
protein serine kinase activityEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
protein kinase activityDual specificity testis-specific protein kinase 2Homo sapiens (human)
protein serine/threonine kinase activityDual specificity testis-specific protein kinase 2Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity testis-specific protein kinase 2Homo sapiens (human)
protein tyrosine kinase activityDual specificity testis-specific protein kinase 2Homo sapiens (human)
protein bindingDual specificity testis-specific protein kinase 2Homo sapiens (human)
ATP bindingDual specificity testis-specific protein kinase 2Homo sapiens (human)
metal ion bindingDual specificity testis-specific protein kinase 2Homo sapiens (human)
protein serine kinase activityDual specificity testis-specific protein kinase 2Homo sapiens (human)
protein serine/threonine kinase activityMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
protein bindingMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
ATP bindingMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
kinase activityMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
metal ion bindingMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
protein serine kinase activityMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
protein kinase activityMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
magnesium ion bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
JUN kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein kinase bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein phosphatase bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein domain specific bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
identical protein bindingMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein homodimerization activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
RNA bindingAtaxin-2Homo sapiens (human)
epidermal growth factor receptor bindingAtaxin-2Homo sapiens (human)
protein bindingAtaxin-2Homo sapiens (human)
mRNA bindingAtaxin-2Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
metal ion bindingMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
eukaryotic translation initiation factor 2alpha kinase activityEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
protein bindingEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
ATP bindingEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
heme bindingEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
protein homodimerization activityEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
protein serine kinase activityEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
protein bindingTarget of rapamycin complex subunit LST8Homo sapiens (human)
protein serine/threonine kinase activator activityTarget of rapamycin complex subunit LST8Homo sapiens (human)
RNA bindingNucleolar GTP-binding protein 1Homo sapiens (human)
GTPase activityNucleolar GTP-binding protein 1Homo sapiens (human)
protein bindingNucleolar GTP-binding protein 1Homo sapiens (human)
GTP bindingNucleolar GTP-binding protein 1Homo sapiens (human)
preribosome bindingNucleolar GTP-binding protein 1Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase D2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase D2Homo sapiens (human)
diacylglycerol-dependent serine/threonine kinase activitySerine/threonine-protein kinase D2Homo sapiens (human)
protein kinase C bindingSerine/threonine-protein kinase D2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase D2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase D2Homo sapiens (human)
metal ion bindingSerine/threonine-protein kinase D2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase D2Homo sapiens (human)
magnesium ion bindingNUAK family SNF1-like kinase 2Homo sapiens (human)
protein serine/threonine kinase activityNUAK family SNF1-like kinase 2Homo sapiens (human)
protein bindingNUAK family SNF1-like kinase 2Homo sapiens (human)
ATP bindingNUAK family SNF1-like kinase 2Homo sapiens (human)
protein serine kinase activityNUAK family SNF1-like kinase 2Homo sapiens (human)
RNA bindingRNA cytidine acetyltransferaseHomo sapiens (human)
protein bindingRNA cytidine acetyltransferaseHomo sapiens (human)
ATP bindingRNA cytidine acetyltransferaseHomo sapiens (human)
N-acetyltransferase activityRNA cytidine acetyltransferaseHomo sapiens (human)
tRNA N-acetyltransferase activityRNA cytidine acetyltransferaseHomo sapiens (human)
DNA polymerase bindingRNA cytidine acetyltransferaseHomo sapiens (human)
mRNA N-acetyltransferase activityRNA cytidine acetyltransferaseHomo sapiens (human)
tRNA bindingRNA cytidine acetyltransferaseHomo sapiens (human)
rRNA cytidine N-acetyltransferase activityRNA cytidine acetyltransferaseHomo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase SIK2Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase SIK2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase SIK2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase SIK2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase SIK2Homo sapiens (human)
protein serine/threonine kinase activitySTE20-like serine/threonine-protein kinase Homo sapiens (human)
protein bindingSTE20-like serine/threonine-protein kinase Homo sapiens (human)
ATP bindingSTE20-like serine/threonine-protein kinase Homo sapiens (human)
identical protein bindingSTE20-like serine/threonine-protein kinase Homo sapiens (human)
protein homodimerization activitySTE20-like serine/threonine-protein kinase Homo sapiens (human)
cadherin bindingSTE20-like serine/threonine-protein kinase Homo sapiens (human)
protein serine kinase activitySTE20-like serine/threonine-protein kinase Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase TAO3Homo sapiens (human)
protein kinase inhibitor activitySerine/threonine-protein kinase TAO3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase TAO3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase TAO3Homo sapiens (human)
transferase activitySerine/threonine-protein kinase TAO3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase TAO3Homo sapiens (human)
magnesium ion bindingdCTP pyrophosphatase 1Homo sapiens (human)
protein bindingdCTP pyrophosphatase 1Homo sapiens (human)
pyrimidine deoxyribonucleotide bindingdCTP pyrophosphatase 1Homo sapiens (human)
identical protein bindingdCTP pyrophosphatase 1Homo sapiens (human)
nucleoside triphosphate diphosphatase activitydCTP pyrophosphatase 1Homo sapiens (human)
dCTP diphosphatase activitydCTP pyrophosphatase 1Homo sapiens (human)
protein serine/threonine kinase activityDual specificity protein kinase CLK4Homo sapiens (human)
protein serine/threonine/tyrosine kinase activityDual specificity protein kinase CLK4Homo sapiens (human)
protein bindingDual specificity protein kinase CLK4Homo sapiens (human)
ATP bindingDual specificity protein kinase CLK4Homo sapiens (human)
protein serine kinase activityDual specificity protein kinase CLK4Homo sapiens (human)
protein tyrosine kinase activityDual specificity protein kinase CLK4Homo sapiens (human)
protein serine/threonine kinase activityCasein kinase I isoform gamma-1Homo sapiens (human)
protein bindingCasein kinase I isoform gamma-1Homo sapiens (human)
ATP bindingCasein kinase I isoform gamma-1Homo sapiens (human)
protein serine kinase activityCasein kinase I isoform gamma-1Homo sapiens (human)
magnesium ion bindingPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
RNA bindingPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
phenylalanine-tRNA ligase activityPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
protein bindingPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
ATP bindingPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
protein bindingBMP-2-inducible protein kinaseHomo sapiens (human)
ATP bindingBMP-2-inducible protein kinaseHomo sapiens (human)
protein serine kinase activityBMP-2-inducible protein kinaseHomo sapiens (human)
phosphatase regulator activityBMP-2-inducible protein kinaseHomo sapiens (human)
AP-2 adaptor complex bindingBMP-2-inducible protein kinaseHomo sapiens (human)
protein serine/threonine kinase activityBMP-2-inducible protein kinaseHomo sapiens (human)
protein bindingObg-like ATPase 1Homo sapiens (human)
ATP bindingObg-like ATPase 1Homo sapiens (human)
GTP bindingObg-like ATPase 1Homo sapiens (human)
ATP hydrolysis activityObg-like ATPase 1Homo sapiens (human)
ribosomal large subunit bindingObg-like ATPase 1Homo sapiens (human)
cadherin bindingObg-like ATPase 1Homo sapiens (human)
metal ion bindingObg-like ATPase 1Homo sapiens (human)
protein bindingMidasinHomo sapiens (human)
ATP bindingMidasinHomo sapiens (human)
ATP hydrolysis activityMidasinHomo sapiens (human)
magnesium ion bindingInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
protein serine/threonine kinase activityInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
interleukin-1 receptor bindingInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
protein bindingInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
ATP bindingInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
kinase activityInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
protein kinase bindingInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
protein serine kinase activityInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
magnesium ion bindingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
RNA bindingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
JUN kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein kinase activator activityMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
ribosome bindingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
small ribosomal subunit rRNA bindingMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
protein kinase activityCyclin-dependent kinase 12Homo sapiens (human)
cyclin-dependent protein serine/threonine kinase activityCyclin-dependent kinase 12Homo sapiens (human)
protein bindingCyclin-dependent kinase 12Homo sapiens (human)
ATP bindingCyclin-dependent kinase 12Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityCyclin-dependent kinase 12Homo sapiens (human)
protein kinase bindingCyclin-dependent kinase 12Homo sapiens (human)
cyclin bindingCyclin-dependent kinase 12Homo sapiens (human)
protein serine kinase activityCyclin-dependent kinase 12Homo sapiens (human)
protein bindingNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
ATP bindingNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
endopeptidase activator activityNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase 26Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase 26Homo sapiens (human)
protein bindingSerine/threonine-protein kinase 26Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase 26Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase 26Homo sapiens (human)
protein homodimerization activitySerine/threonine-protein kinase 26Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase 26Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase 26Homo sapiens (human)
magnesium ion bindingSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinate-CoA ligase (ADP-forming) activitySuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
protein bindingSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
ATP bindingSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase NLKHomo sapiens (human)
protein kinase activitySerine/threonine-protein kinase NLKHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase NLKHomo sapiens (human)
MAP kinase activitySerine/threonine-protein kinase NLKHomo sapiens (human)
protein bindingSerine/threonine-protein kinase NLKHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase NLKHomo sapiens (human)
ubiquitin protein ligase bindingSerine/threonine-protein kinase NLKHomo sapiens (human)
SH2 domain bindingSerine/threonine-protein kinase NLKHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase NLKHomo sapiens (human)
DNA-binding transcription factor bindingSerine/threonine-protein kinase NLKHomo sapiens (human)
AMP-activated protein kinase activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
cAMP-dependent protein kinase inhibitor activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
protein binding5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
ATP binding5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
cAMP-dependent protein kinase regulator activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
phosphorylase kinase regulator activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
protein kinase regulator activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
protein kinase binding5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
protein kinase activator activity5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
ADP binding5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
AMP binding5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
nucleic acid bindingSerine/threonine-protein kinase TBK1Homo sapiens (human)
protein kinase activitySerine/threonine-protein kinase TBK1Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase TBK1Homo sapiens (human)
protein bindingSerine/threonine-protein kinase TBK1Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase TBK1Homo sapiens (human)
protein phosphatase bindingSerine/threonine-protein kinase TBK1Homo sapiens (human)
identical protein bindingSerine/threonine-protein kinase TBK1Homo sapiens (human)
phosphoprotein bindingSerine/threonine-protein kinase TBK1Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase TBK1Homo sapiens (human)
protein bindingSeptin-9Homo sapiens (human)
GTP bindingSeptin-9Homo sapiens (human)
cadherin bindingSeptin-9Homo sapiens (human)
GTPase activitySeptin-9Homo sapiens (human)
molecular adaptor activitySeptin-9Homo sapiens (human)
A-type (transient outward) potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
metal ion bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
magnesium ion bindingRibosomal protein S6 kinase alpha-6Homo sapiens (human)
protein kinase activityRibosomal protein S6 kinase alpha-6Homo sapiens (human)
protein bindingRibosomal protein S6 kinase alpha-6Homo sapiens (human)
ATP bindingRibosomal protein S6 kinase alpha-6Homo sapiens (human)
protein serine kinase activityRibosomal protein S6 kinase alpha-6Homo sapiens (human)
ribosomal protein S6 kinase activityRibosomal protein S6 kinase alpha-6Homo sapiens (human)
protein kinase activityTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein serine/threonine kinase activityTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein bindingTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
ATP bindingTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein serine kinase activityTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase TAO2Homo sapiens (human)
MAP kinase kinase kinase activitySerine/threonine-protein kinase TAO2Homo sapiens (human)
protein bindingSerine/threonine-protein kinase TAO2Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase TAO2Homo sapiens (human)
mitogen-activated protein kinase kinase bindingSerine/threonine-protein kinase TAO2Homo sapiens (human)
neuropilin bindingSerine/threonine-protein kinase TAO2Homo sapiens (human)
protein serine/threonine kinase activator activitySerine/threonine-protein kinase TAO2Homo sapiens (human)
tau protein bindingSerine/threonine-protein kinase TAO2Homo sapiens (human)
tau-protein kinase activitySerine/threonine-protein kinase TAO2Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase TAO2Homo sapiens (human)
long-chain fatty acid-CoA ligase activityLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
protein bindingLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
ATP bindingLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
arachidonate-CoA ligase activityLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
oleoyl-CoA ligase activityLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
protein kinase activityRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
protein serine/threonine kinase activityRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
protein bindingRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
ATP bindingRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
protein serine kinase activityRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase SIK3Homo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase SIK3Homo sapiens (human)
protein bindingSerine/threonine-protein kinase SIK3Homo sapiens (human)
ATP bindingSerine/threonine-protein kinase SIK3Homo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase SIK3Homo sapiens (human)
tau-protein kinase activitySerine/threonine-protein kinase SIK3Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
protein kinase bindingMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
metal ion bindingMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
transcription coregulator activityThyroid hormone receptor-associated protein 3Homo sapiens (human)
transcription coactivator activityThyroid hormone receptor-associated protein 3Homo sapiens (human)
RNA bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
protein bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
ATP bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
nuclear receptor coactivator activityThyroid hormone receptor-associated protein 3Homo sapiens (human)
nuclear vitamin D receptor bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
nuclear thyroid hormone receptor bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
phosphoprotein bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
DNA bindingThyroid hormone receptor-associated protein 3Homo sapiens (human)
protein kinase activityMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
protein serine/threonine kinase activityMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
MAP kinase kinase kinase kinase activityMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
transcription coactivator activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein kinase activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein serine/threonine kinase activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein bindingReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
ATP bindingReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
identical protein bindingReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein-containing complex bindingReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein serine kinase activityReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
magnesium ion bindingSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
protein kinase activitySerine/threonine-protein kinase MRCK betaHomo sapiens (human)
protein serine/threonine kinase activitySerine/threonine-protein kinase MRCK betaHomo sapiens (human)
ATP bindingSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
small GTPase bindingSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
protein-containing complex bindingSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
protein serine kinase activitySerine/threonine-protein kinase MRCK betaHomo sapiens (human)
magnesium ion bindingInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein serine/threonine kinase activityInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein bindingInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
ATP bindingInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein kinase bindingInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein homodimerization activityInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein heterodimerization activityInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
protein kinase activityCasein kinase I isoform gamma-3Homo sapiens (human)
protein serine/threonine kinase activityCasein kinase I isoform gamma-3Homo sapiens (human)
ATP bindingCasein kinase I isoform gamma-3Homo sapiens (human)
protein serine kinase activityCasein kinase I isoform gamma-3Homo sapiens (human)
MAP kinase kinase kinase activityMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
protein bindingMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
ATP bindingMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
metal ion bindingMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
protein serine kinase activityMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (410)

Processvia Protein(s)Taxonomy
plasma membraneBone morphogenetic protein receptor type-1BHomo sapiens (human)
dendriteBone morphogenetic protein receptor type-1BHomo sapiens (human)
neuronal cell bodyBone morphogenetic protein receptor type-1BHomo sapiens (human)
receptor complexBone morphogenetic protein receptor type-1BHomo sapiens (human)
HFE-transferrin receptor complexBone morphogenetic protein receptor type-1BHomo sapiens (human)
plasma membraneBone morphogenetic protein receptor type-1BHomo sapiens (human)
nucleusCell division cycle 7-related protein kinaseHomo sapiens (human)
nucleoplasmCell division cycle 7-related protein kinaseHomo sapiens (human)
cytoplasmCell division cycle 7-related protein kinaseHomo sapiens (human)
intercellular bridgeCell division cycle 7-related protein kinaseHomo sapiens (human)
mitotic spindleCell division cycle 7-related protein kinaseHomo sapiens (human)
nucleusCell division cycle 7-related protein kinaseHomo sapiens (human)
cytoplasmCell division cycle 7-related protein kinaseHomo sapiens (human)
XY bodySerine/threonine-protein kinase PLK4Homo sapiens (human)
nucleolusSerine/threonine-protein kinase PLK4Homo sapiens (human)
centrosomeSerine/threonine-protein kinase PLK4Homo sapiens (human)
centrioleSerine/threonine-protein kinase PLK4Homo sapiens (human)
cytosolSerine/threonine-protein kinase PLK4Homo sapiens (human)
cleavage furrowSerine/threonine-protein kinase PLK4Homo sapiens (human)
deuterosomeSerine/threonine-protein kinase PLK4Homo sapiens (human)
procentrioleSerine/threonine-protein kinase PLK4Homo sapiens (human)
procentriole replication complexSerine/threonine-protein kinase PLK4Homo sapiens (human)
nucleusSerine/threonine-protein kinase PLK4Homo sapiens (human)
eukaryotic translation initiation factor 3 complexATP-dependent RNA helicase DDX3XHomo sapiens (human)
cytosolic small ribosomal subunitATP-dependent RNA helicase DDX3XHomo sapiens (human)
cytoplasmATP-dependent RNA helicase DDX3XHomo sapiens (human)
extracellular regionATP-dependent RNA helicase DDX3XHomo sapiens (human)
nucleusATP-dependent RNA helicase DDX3XHomo sapiens (human)
nucleoplasmATP-dependent RNA helicase DDX3XHomo sapiens (human)
cytoplasmATP-dependent RNA helicase DDX3XHomo sapiens (human)
centrosomeATP-dependent RNA helicase DDX3XHomo sapiens (human)
cytosolATP-dependent RNA helicase DDX3XHomo sapiens (human)
plasma membraneATP-dependent RNA helicase DDX3XHomo sapiens (human)
cytoplasmic stress granuleATP-dependent RNA helicase DDX3XHomo sapiens (human)
lamellipodiumATP-dependent RNA helicase DDX3XHomo sapiens (human)
cell leading edgeATP-dependent RNA helicase DDX3XHomo sapiens (human)
secretory granule lumenATP-dependent RNA helicase DDX3XHomo sapiens (human)
extracellular exosomeATP-dependent RNA helicase DDX3XHomo sapiens (human)
ficolin-1-rich granule lumenATP-dependent RNA helicase DDX3XHomo sapiens (human)
NLRP3 inflammasome complexATP-dependent RNA helicase DDX3XHomo sapiens (human)
nucleusATP-dependent RNA helicase DDX3XHomo sapiens (human)
P granuleATP-dependent RNA helicase DDX3XHomo sapiens (human)
extracellular regionPyridoxal kinaseHomo sapiens (human)
nucleusPyridoxal kinaseHomo sapiens (human)
nucleoplasmPyridoxal kinaseHomo sapiens (human)
cytosolPyridoxal kinaseHomo sapiens (human)
secretory granule lumenPyridoxal kinaseHomo sapiens (human)
specific granule lumenPyridoxal kinaseHomo sapiens (human)
extracellular exosomePyridoxal kinaseHomo sapiens (human)
cytosolPyridoxal kinaseHomo sapiens (human)
cytosolCitron Rho-interacting kinaseHomo sapiens (human)
membraneCitron Rho-interacting kinaseHomo sapiens (human)
chromosome, telomeric regionSerine/threonine-protein kinase Chk1Homo sapiens (human)
condensed nuclear chromosomeSerine/threonine-protein kinase Chk1Homo sapiens (human)
extracellular spaceSerine/threonine-protein kinase Chk1Homo sapiens (human)
nucleusSerine/threonine-protein kinase Chk1Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase Chk1Homo sapiens (human)
replication forkSerine/threonine-protein kinase Chk1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase Chk1Homo sapiens (human)
centrosomeSerine/threonine-protein kinase Chk1Homo sapiens (human)
cytosolSerine/threonine-protein kinase Chk1Homo sapiens (human)
intracellular membrane-bounded organelleSerine/threonine-protein kinase Chk1Homo sapiens (human)
chromatinSerine/threonine-protein kinase Chk1Homo sapiens (human)
protein-containing complexSerine/threonine-protein kinase Chk1Homo sapiens (human)
nucleusSerine/threonine-protein kinase Chk1Homo sapiens (human)
spindle microtubuleAurora kinase AHomo sapiens (human)
nucleusAurora kinase AHomo sapiens (human)
nucleoplasmAurora kinase AHomo sapiens (human)
centrosomeAurora kinase AHomo sapiens (human)
centrioleAurora kinase AHomo sapiens (human)
spindleAurora kinase AHomo sapiens (human)
cytosolAurora kinase AHomo sapiens (human)
postsynaptic densityAurora kinase AHomo sapiens (human)
microtubule cytoskeletonAurora kinase AHomo sapiens (human)
basolateral plasma membraneAurora kinase AHomo sapiens (human)
midbodyAurora kinase AHomo sapiens (human)
spindle pole centrosomeAurora kinase AHomo sapiens (human)
ciliary basal bodyAurora kinase AHomo sapiens (human)
germinal vesicleAurora kinase AHomo sapiens (human)
axon hillockAurora kinase AHomo sapiens (human)
pronucleusAurora kinase AHomo sapiens (human)
perinuclear region of cytoplasmAurora kinase AHomo sapiens (human)
mitotic spindleAurora kinase AHomo sapiens (human)
meiotic spindleAurora kinase AHomo sapiens (human)
mitotic spindle poleAurora kinase AHomo sapiens (human)
glutamatergic synapseAurora kinase AHomo sapiens (human)
spindle pole centrosomeAurora kinase AHomo sapiens (human)
chromosome passenger complexAurora kinase AHomo sapiens (human)
spindle midzoneAurora kinase AHomo sapiens (human)
kinetochoreAurora kinase AHomo sapiens (human)
Golgi apparatusCyclin-G-associated kinaseHomo sapiens (human)
cytosolCyclin-G-associated kinaseHomo sapiens (human)
focal adhesionCyclin-G-associated kinaseHomo sapiens (human)
membraneCyclin-G-associated kinaseHomo sapiens (human)
clathrin-coated vesicleCyclin-G-associated kinaseHomo sapiens (human)
vesicleCyclin-G-associated kinaseHomo sapiens (human)
intracellular membrane-bounded organelleCyclin-G-associated kinaseHomo sapiens (human)
perinuclear region of cytoplasmCyclin-G-associated kinaseHomo sapiens (human)
presynapseCyclin-G-associated kinaseHomo sapiens (human)
vesicleCyclin-G-associated kinaseHomo sapiens (human)
cytoplasmCyclin-G-associated kinaseHomo sapiens (human)
intracellular membrane-bounded organelleCyclin-G-associated kinaseHomo sapiens (human)
extracellular regionEphrin type-B receptor 6Homo sapiens (human)
cytosolEphrin type-B receptor 6Homo sapiens (human)
plasma membraneEphrin type-B receptor 6Homo sapiens (human)
plasma membraneEphrin type-B receptor 6Homo sapiens (human)
dendriteEphrin type-B receptor 6Homo sapiens (human)
peroxisomePeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
peroxisomal matrixPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
cytosolPeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
membranePeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
peroxisomePeroxisomal acyl-coenzyme A oxidase 3Homo sapiens (human)
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
nucleolusMultidrug resistance-associated protein 4Homo sapiens (human)
Golgi apparatusMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
platelet dense granule membraneMultidrug resistance-associated protein 4Homo sapiens (human)
external side of apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cytoplasmReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
endoplasmic reticulumReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cytosolReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
cytoskeletonReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
vesicleReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
protein-containing complexReceptor-interacting serine/threonine-protein kinase 2Homo sapiens (human)
kinetochoreMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
nucleoplasmMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
cytosolMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
membraneMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
intracellular membrane-bounded organelleMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
outer kinetochoreMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
kinetochoreMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
nucleusMitotic checkpoint serine/threonine-protein kinase BUB1Homo sapiens (human)
nucleoplasmDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrionDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial outer membraneDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial inner membraneDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial intermembrane spaceDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
cytosolDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
membraneDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial cristaDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
dendriteDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
axon cytoplasmDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial intermembrane spaceDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
cytoplasmDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
microtubuleDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
mitochondrial membraneDynamin-like 120 kDa protein, mitochondrialHomo sapiens (human)
nucleusEukaryotic translation initiation factor 5BHomo sapiens (human)
cytoplasmEukaryotic translation initiation factor 5BHomo sapiens (human)
cytosolEukaryotic translation initiation factor 5BHomo sapiens (human)
synapseEukaryotic translation initiation factor 5BHomo sapiens (human)
cytoplasmEukaryotic translation initiation factor 5BHomo sapiens (human)
nucleusRho-associated protein kinase 2Homo sapiens (human)
centrosomeRho-associated protein kinase 2Homo sapiens (human)
cytosolRho-associated protein kinase 2Homo sapiens (human)
plasma membraneRho-associated protein kinase 2Homo sapiens (human)
cytoplasmic ribonucleoprotein granuleRho-associated protein kinase 2Homo sapiens (human)
centrosomeRho-associated protein kinase 2Homo sapiens (human)
cytoskeletonRho-associated protein kinase 2Homo sapiens (human)
cytoplasmRho-associated protein kinase 2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase ULK1Homo sapiens (human)
phagophore assembly siteSerine/threonine-protein kinase ULK1Homo sapiens (human)
autophagosome membraneSerine/threonine-protein kinase ULK1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase ULK1Homo sapiens (human)
mitochondrial outer membraneSerine/threonine-protein kinase ULK1Homo sapiens (human)
autophagosomeSerine/threonine-protein kinase ULK1Homo sapiens (human)
endoplasmic reticulum membraneSerine/threonine-protein kinase ULK1Homo sapiens (human)
cytosolSerine/threonine-protein kinase ULK1Homo sapiens (human)
axonSerine/threonine-protein kinase ULK1Homo sapiens (human)
phagophore assembly site membraneSerine/threonine-protein kinase ULK1Homo sapiens (human)
recycling endosomeSerine/threonine-protein kinase ULK1Homo sapiens (human)
omegasome membraneSerine/threonine-protein kinase ULK1Homo sapiens (human)
Atg1/ULK1 kinase complexSerine/threonine-protein kinase ULK1Homo sapiens (human)
cytosolSerine/threonine-protein kinase ULK1Homo sapiens (human)
phagophore assembly siteSerine/threonine-protein kinase ULK1Homo sapiens (human)
autophagosomeSerine/threonine-protein kinase ULK1Homo sapiens (human)
phagophore assembly site membraneSerine/threonine-protein kinase ULK1Homo sapiens (human)
nuclear inner membraneSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
mitochondrionSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
endoplasmic reticulumSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
endoplasmic reticulum membraneSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
Ire1 complexSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
AIP1-IRE1 complexSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
IRE1-TRAF2-ASK1 complexSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
IRE1-RACK1-PP2A complexSerine/threonine-protein kinase/endoribonuclease IRE1Homo sapiens (human)
nucleusRibosomal protein S6 kinase alpha-5Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-5Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase alpha-5Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-5Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase alpha-5Homo sapiens (human)
nucleusU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
nucleoplasmU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
membraneU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
U4/U6 x U5 tri-snRNP complexU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
spliceosomal complexU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
U5 snRNPU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
U2-type precatalytic spliceosomeU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
U2-type catalytic step 1 spliceosomeU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
catalytic step 2 spliceosomeU5 small nuclear ribonucleoprotein 200 kDa helicaseHomo sapiens (human)
nucleusRibosomal protein S6 kinase alpha-4Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-4Homo sapiens (human)
cytosolRibosomal protein S6 kinase alpha-4Homo sapiens (human)
synapseRibosomal protein S6 kinase alpha-4Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase alpha-4Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-4Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase 16Homo sapiens (human)
Golgi-associated vesicleSerine/threonine-protein kinase 16Homo sapiens (human)
cytosolSerine/threonine-protein kinase 16Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase 16Homo sapiens (human)
perinuclear region of cytoplasmSerine/threonine-protein kinase 16Homo sapiens (human)
Golgi apparatusSerine/threonine-protein kinase 16Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 16Homo sapiens (human)
cytosolSerine/threonine-protein kinase 10Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase 10Homo sapiens (human)
specific granule membraneSerine/threonine-protein kinase 10Homo sapiens (human)
extracellular exosomeSerine/threonine-protein kinase 10Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 10Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase D3Homo sapiens (human)
cytosolSerine/threonine-protein kinase D3Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase D3Homo sapiens (human)
cytosolSerine/threonine-protein kinase D3Homo sapiens (human)
basolateral plasma membraneBile salt export pumpHomo sapiens (human)
Golgi membraneBile salt export pumpHomo sapiens (human)
endosomeBile salt export pumpHomo sapiens (human)
plasma membraneBile salt export pumpHomo sapiens (human)
cell surfaceBile salt export pumpHomo sapiens (human)
apical plasma membraneBile salt export pumpHomo sapiens (human)
intercellular canaliculusBile salt export pumpHomo sapiens (human)
intracellular canaliculusBile salt export pumpHomo sapiens (human)
recycling endosomeBile salt export pumpHomo sapiens (human)
recycling endosome membraneBile salt export pumpHomo sapiens (human)
extracellular exosomeBile salt export pumpHomo sapiens (human)
membraneBile salt export pumpHomo sapiens (human)
nuclear chromosomeStructural maintenance of chromosomes protein 2Homo sapiens (human)
condensed chromosomeStructural maintenance of chromosomes protein 2Homo sapiens (human)
condensed nuclear chromosomeStructural maintenance of chromosomes protein 2Homo sapiens (human)
condensin complexStructural maintenance of chromosomes protein 2Homo sapiens (human)
nucleusStructural maintenance of chromosomes protein 2Homo sapiens (human)
nucleoplasmStructural maintenance of chromosomes protein 2Homo sapiens (human)
nucleolusStructural maintenance of chromosomes protein 2Homo sapiens (human)
cytoplasmStructural maintenance of chromosomes protein 2Homo sapiens (human)
cytosolStructural maintenance of chromosomes protein 2Homo sapiens (human)
extracellular exosomeStructural maintenance of chromosomes protein 2Homo sapiens (human)
condensed chromosomeStructural maintenance of chromosomes protein 2Homo sapiens (human)
chromatinStructural maintenance of chromosomes protein 2Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
focal adhesionMitogen-activated protein kinase kinase kinase kinase 4Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase LATS1Homo sapiens (human)
spindle poleSerine/threonine-protein kinase LATS1Homo sapiens (human)
nucleusSerine/threonine-protein kinase LATS1Homo sapiens (human)
centrosomeSerine/threonine-protein kinase LATS1Homo sapiens (human)
cytosolSerine/threonine-protein kinase LATS1Homo sapiens (human)
midbodySerine/threonine-protein kinase LATS1Homo sapiens (human)
spindle poleSerine/threonine-protein kinase LATS1Homo sapiens (human)
Golgi apparatusSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cytosolSerine/threonine-protein kinase PAK 4Homo sapiens (human)
adherens junctionSerine/threonine-protein kinase PAK 4Homo sapiens (human)
focal adhesionSerine/threonine-protein kinase PAK 4Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase PAK 4Homo sapiens (human)
ruffleTyrosine-protein kinase ABL1Homo sapiens (human)
nucleusTyrosine-protein kinase ABL1Homo sapiens (human)
nucleoplasmTyrosine-protein kinase ABL1Homo sapiens (human)
nucleolusTyrosine-protein kinase ABL1Homo sapiens (human)
cytoplasmTyrosine-protein kinase ABL1Homo sapiens (human)
mitochondrionTyrosine-protein kinase ABL1Homo sapiens (human)
cytosolTyrosine-protein kinase ABL1Homo sapiens (human)
actin cytoskeletonTyrosine-protein kinase ABL1Homo sapiens (human)
nuclear bodyTyrosine-protein kinase ABL1Homo sapiens (human)
dendriteTyrosine-protein kinase ABL1Homo sapiens (human)
growth coneTyrosine-protein kinase ABL1Homo sapiens (human)
nuclear membraneTyrosine-protein kinase ABL1Homo sapiens (human)
neuronal cell bodyTyrosine-protein kinase ABL1Homo sapiens (human)
perinuclear region of cytoplasmTyrosine-protein kinase ABL1Homo sapiens (human)
postsynapseTyrosine-protein kinase ABL1Homo sapiens (human)
protein-containing complexTyrosine-protein kinase ABL1Homo sapiens (human)
plasma membraneTyrosine-protein kinase ABL1Homo sapiens (human)
endosomeEpidermal growth factor receptorHomo sapiens (human)
plasma membraneEpidermal growth factor receptorHomo sapiens (human)
ruffle membraneEpidermal growth factor receptorHomo sapiens (human)
Golgi membraneEpidermal growth factor receptorHomo sapiens (human)
extracellular spaceEpidermal growth factor receptorHomo sapiens (human)
nucleusEpidermal growth factor receptorHomo sapiens (human)
cytoplasmEpidermal growth factor receptorHomo sapiens (human)
endosomeEpidermal growth factor receptorHomo sapiens (human)
endoplasmic reticulum membraneEpidermal growth factor receptorHomo sapiens (human)
plasma membraneEpidermal growth factor receptorHomo sapiens (human)
focal adhesionEpidermal growth factor receptorHomo sapiens (human)
cell surfaceEpidermal growth factor receptorHomo sapiens (human)
endosome membraneEpidermal growth factor receptorHomo sapiens (human)
membraneEpidermal growth factor receptorHomo sapiens (human)
basolateral plasma membraneEpidermal growth factor receptorHomo sapiens (human)
apical plasma membraneEpidermal growth factor receptorHomo sapiens (human)
cell junctionEpidermal growth factor receptorHomo sapiens (human)
clathrin-coated endocytic vesicle membraneEpidermal growth factor receptorHomo sapiens (human)
early endosome membraneEpidermal growth factor receptorHomo sapiens (human)
nuclear membraneEpidermal growth factor receptorHomo sapiens (human)
membrane raftEpidermal growth factor receptorHomo sapiens (human)
perinuclear region of cytoplasmEpidermal growth factor receptorHomo sapiens (human)
multivesicular body, internal vesicle lumenEpidermal growth factor receptorHomo sapiens (human)
intracellular vesicleEpidermal growth factor receptorHomo sapiens (human)
protein-containing complexEpidermal growth factor receptorHomo sapiens (human)
receptor complexEpidermal growth factor receptorHomo sapiens (human)
Shc-EGFR complexEpidermal growth factor receptorHomo sapiens (human)
basal plasma membraneEpidermal growth factor receptorHomo sapiens (human)
early endosomeHigh affinity nerve growth factor receptorHomo sapiens (human)
late endosomeHigh affinity nerve growth factor receptorHomo sapiens (human)
plasma membraneHigh affinity nerve growth factor receptorHomo sapiens (human)
cell surfaceHigh affinity nerve growth factor receptorHomo sapiens (human)
endosome membraneHigh affinity nerve growth factor receptorHomo sapiens (human)
dendriteHigh affinity nerve growth factor receptorHomo sapiens (human)
early endosome membraneHigh affinity nerve growth factor receptorHomo sapiens (human)
late endosome membraneHigh affinity nerve growth factor receptorHomo sapiens (human)
neuronal cell bodyHigh affinity nerve growth factor receptorHomo sapiens (human)
recycling endosome membraneHigh affinity nerve growth factor receptorHomo sapiens (human)
protein-containing complexHigh affinity nerve growth factor receptorHomo sapiens (human)
receptor complexHigh affinity nerve growth factor receptorHomo sapiens (human)
axonHigh affinity nerve growth factor receptorHomo sapiens (human)
plasma membraneHigh affinity nerve growth factor receptorHomo sapiens (human)
nuclear bodyCellular tumor antigen p53Homo sapiens (human)
nucleusCellular tumor antigen p53Homo sapiens (human)
nucleoplasmCellular tumor antigen p53Homo sapiens (human)
replication forkCellular tumor antigen p53Homo sapiens (human)
nucleolusCellular tumor antigen p53Homo sapiens (human)
cytoplasmCellular tumor antigen p53Homo sapiens (human)
mitochondrionCellular tumor antigen p53Homo sapiens (human)
mitochondrial matrixCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulumCellular tumor antigen p53Homo sapiens (human)
centrosomeCellular tumor antigen p53Homo sapiens (human)
cytosolCellular tumor antigen p53Homo sapiens (human)
nuclear matrixCellular tumor antigen p53Homo sapiens (human)
PML bodyCellular tumor antigen p53Homo sapiens (human)
transcription repressor complexCellular tumor antigen p53Homo sapiens (human)
site of double-strand breakCellular tumor antigen p53Homo sapiens (human)
germ cell nucleusCellular tumor antigen p53Homo sapiens (human)
chromatinCellular tumor antigen p53Homo sapiens (human)
transcription regulator complexCellular tumor antigen p53Homo sapiens (human)
protein-containing complexCellular tumor antigen p53Homo sapiens (human)
nucleoplasmGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
cytoplasmGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
centrosomeGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
cytosolGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
plasma membraneGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
membraneGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
dendriteGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
midbodyGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
cell bodyGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
synapseGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
extracellular exosomeGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
neuronal dense core vesicleGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
extracellular vesicleGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
heterotrimeric G-protein complexGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
cytoplasmGuanine nucleotide-binding protein G(i) subunit alpha-2Homo sapiens (human)
nucleusADP/ATP translocase 2Homo sapiens (human)
mitochondrionADP/ATP translocase 2Homo sapiens (human)
mitochondrial inner membraneADP/ATP translocase 2Homo sapiens (human)
plasma membraneADP/ATP translocase 2Homo sapiens (human)
membraneADP/ATP translocase 2Homo sapiens (human)
mitochondrial nucleoidADP/ATP translocase 2Homo sapiens (human)
mitochondrial permeability transition pore complexADP/ATP translocase 2Homo sapiens (human)
MMXD complexADP/ATP translocase 2Homo sapiens (human)
nucleusProtein kinase C beta typeHomo sapiens (human)
nucleoplasmProtein kinase C beta typeHomo sapiens (human)
cytoplasmProtein kinase C beta typeHomo sapiens (human)
centrosomeProtein kinase C beta typeHomo sapiens (human)
cytosolProtein kinase C beta typeHomo sapiens (human)
plasma membraneProtein kinase C beta typeHomo sapiens (human)
brush border membraneProtein kinase C beta typeHomo sapiens (human)
calyx of HeldProtein kinase C beta typeHomo sapiens (human)
extracellular exosomeProtein kinase C beta typeHomo sapiens (human)
presynaptic cytosolProtein kinase C beta typeHomo sapiens (human)
spectrinProtein kinase C beta typeHomo sapiens (human)
nuclear envelopeInsulin receptorHomo sapiens (human)
nuclear lumenInsulin receptorHomo sapiens (human)
lysosomeInsulin receptorHomo sapiens (human)
late endosomeInsulin receptorHomo sapiens (human)
plasma membraneInsulin receptorHomo sapiens (human)
caveolaInsulin receptorHomo sapiens (human)
external side of plasma membraneInsulin receptorHomo sapiens (human)
endosome membraneInsulin receptorHomo sapiens (human)
membraneInsulin receptorHomo sapiens (human)
dendrite membraneInsulin receptorHomo sapiens (human)
neuronal cell body membraneInsulin receptorHomo sapiens (human)
extracellular exosomeInsulin receptorHomo sapiens (human)
insulin receptor complexInsulin receptorHomo sapiens (human)
receptor complexInsulin receptorHomo sapiens (human)
plasma membraneInsulin receptorHomo sapiens (human)
axonInsulin receptorHomo sapiens (human)
pericentriolar materialTyrosine-protein kinase LckHomo sapiens (human)
immunological synapseTyrosine-protein kinase LckHomo sapiens (human)
cytosolTyrosine-protein kinase LckHomo sapiens (human)
plasma membraneTyrosine-protein kinase LckHomo sapiens (human)
membrane raftTyrosine-protein kinase LckHomo sapiens (human)
extracellular exosomeTyrosine-protein kinase LckHomo sapiens (human)
plasma membraneTyrosine-protein kinase LckHomo sapiens (human)
membrane raftTyrosine-protein kinase FynHomo sapiens (human)
dendriteTyrosine-protein kinase FynHomo sapiens (human)
nucleusTyrosine-protein kinase FynHomo sapiens (human)
mitochondrionTyrosine-protein kinase FynHomo sapiens (human)
endosomeTyrosine-protein kinase FynHomo sapiens (human)
cytosolTyrosine-protein kinase FynHomo sapiens (human)
actin filamentTyrosine-protein kinase FynHomo sapiens (human)
plasma membraneTyrosine-protein kinase FynHomo sapiens (human)
postsynaptic densityTyrosine-protein kinase FynHomo sapiens (human)
dendriteTyrosine-protein kinase FynHomo sapiens (human)
perikaryonTyrosine-protein kinase FynHomo sapiens (human)
cell bodyTyrosine-protein kinase FynHomo sapiens (human)
membrane raftTyrosine-protein kinase FynHomo sapiens (human)
perinuclear region of cytoplasmTyrosine-protein kinase FynHomo sapiens (human)
perinuclear endoplasmic reticulumTyrosine-protein kinase FynHomo sapiens (human)
glial cell projectionTyrosine-protein kinase FynHomo sapiens (human)
Schaffer collateral - CA1 synapseTyrosine-protein kinase FynHomo sapiens (human)
plasma membraneTyrosine-protein kinase FynHomo sapiens (human)
mitochondrial matrixCyclin-dependent kinase 1Homo sapiens (human)
chromosome, telomeric regionCyclin-dependent kinase 1Homo sapiens (human)
nucleusCyclin-dependent kinase 1Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 1Homo sapiens (human)
mitochondrionCyclin-dependent kinase 1Homo sapiens (human)
endoplasmic reticulum membraneCyclin-dependent kinase 1Homo sapiens (human)
centrosomeCyclin-dependent kinase 1Homo sapiens (human)
cytosolCyclin-dependent kinase 1Homo sapiens (human)
spindle microtubuleCyclin-dependent kinase 1Homo sapiens (human)
membraneCyclin-dependent kinase 1Homo sapiens (human)
midbodyCyclin-dependent kinase 1Homo sapiens (human)
extracellular exosomeCyclin-dependent kinase 1Homo sapiens (human)
mitotic spindleCyclin-dependent kinase 1Homo sapiens (human)
cyclin A1-CDK1 complexCyclin-dependent kinase 1Homo sapiens (human)
cyclin A2-CDK1 complexCyclin-dependent kinase 1Homo sapiens (human)
cyclin B1-CDK1 complexCyclin-dependent kinase 1Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 1Homo sapiens (human)
cytoplasmCyclin-dependent kinase 1Homo sapiens (human)
nucleusCyclin-dependent kinase 1Homo sapiens (human)
extracellular regionGlycogen phosphorylase, liver formHomo sapiens (human)
cytosolGlycogen phosphorylase, liver formHomo sapiens (human)
secretory granule lumenGlycogen phosphorylase, liver formHomo sapiens (human)
extracellular exosomeGlycogen phosphorylase, liver formHomo sapiens (human)
ficolin-1-rich granule lumenGlycogen phosphorylase, liver formHomo sapiens (human)
cytoplasmGlycogen phosphorylase, liver formHomo sapiens (human)
cytoplasmic vesicleTyrosine-protein kinase Fes/FpsHomo sapiens (human)
cytoplasmTyrosine-protein kinase Fes/FpsHomo sapiens (human)
Golgi apparatusTyrosine-protein kinase Fes/FpsHomo sapiens (human)
cytosolTyrosine-protein kinase Fes/FpsHomo sapiens (human)
focal adhesionTyrosine-protein kinase Fes/FpsHomo sapiens (human)
cytoplasmic side of plasma membraneTyrosine-protein kinase Fes/FpsHomo sapiens (human)
microtubule cytoskeletonTyrosine-protein kinase Fes/FpsHomo sapiens (human)
plasma membraneTyrosine-protein kinase Fes/FpsHomo sapiens (human)
extracellular regionAdenine phosphoribosyltransferaseHomo sapiens (human)
nucleoplasmAdenine phosphoribosyltransferaseHomo sapiens (human)
cytoplasmAdenine phosphoribosyltransferaseHomo sapiens (human)
cytosolAdenine phosphoribosyltransferaseHomo sapiens (human)
secretory granule lumenAdenine phosphoribosyltransferaseHomo sapiens (human)
extracellular exosomeAdenine phosphoribosyltransferaseHomo sapiens (human)
cytoplasmAdenine phosphoribosyltransferaseHomo sapiens (human)
Golgi apparatusTyrosine-protein kinase YesHomo sapiens (human)
centrosomeTyrosine-protein kinase YesHomo sapiens (human)
cytosolTyrosine-protein kinase YesHomo sapiens (human)
actin filamentTyrosine-protein kinase YesHomo sapiens (human)
plasma membraneTyrosine-protein kinase YesHomo sapiens (human)
focal adhesionTyrosine-protein kinase YesHomo sapiens (human)
extracellular exosomeTyrosine-protein kinase YesHomo sapiens (human)
plasma membraneTyrosine-protein kinase YesHomo sapiens (human)
plasma membraneTyrosine-protein kinase LynHomo sapiens (human)
cytoplasmic side of plasma membraneTyrosine-protein kinase LynHomo sapiens (human)
nucleusTyrosine-protein kinase LynHomo sapiens (human)
cytoplasmTyrosine-protein kinase LynHomo sapiens (human)
lysosomal membraneTyrosine-protein kinase LynHomo sapiens (human)
Golgi apparatusTyrosine-protein kinase LynHomo sapiens (human)
cytosolTyrosine-protein kinase LynHomo sapiens (human)
plasma membraneTyrosine-protein kinase LynHomo sapiens (human)
adherens junctionTyrosine-protein kinase LynHomo sapiens (human)
mitochondrial cristaTyrosine-protein kinase LynHomo sapiens (human)
endocytic vesicle membraneTyrosine-protein kinase LynHomo sapiens (human)
intracellular membrane-bounded organelleTyrosine-protein kinase LynHomo sapiens (human)
membrane raftTyrosine-protein kinase LynHomo sapiens (human)
perinuclear region of cytoplasmTyrosine-protein kinase LynHomo sapiens (human)
extracellular exosomeTyrosine-protein kinase LynHomo sapiens (human)
glutamatergic synapseTyrosine-protein kinase LynHomo sapiens (human)
postsynaptic specialization, intracellular componentTyrosine-protein kinase LynHomo sapiens (human)
integrin alpha2-beta1 complexTyrosine-protein kinase LynHomo sapiens (human)
plasma membraneProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
early endosomeProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
plasma membraneProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
endosome membraneProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
dendriteProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
neuronal cell bodyProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
receptor complexProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
plasma membrane protein complexProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
axonProto-oncogene tyrosine-protein kinase receptor RetHomo sapiens (human)
plasma membraneInsulin-like growth factor 1 receptorHomo sapiens (human)
caveolaInsulin-like growth factor 1 receptorHomo sapiens (human)
membraneInsulin-like growth factor 1 receptorHomo sapiens (human)
T-tubuleInsulin-like growth factor 1 receptorHomo sapiens (human)
neuronal cell bodyInsulin-like growth factor 1 receptorHomo sapiens (human)
intracellular membrane-bounded organelleInsulin-like growth factor 1 receptorHomo sapiens (human)
alphav-beta3 integrin-IGF-1-IGF1R complexInsulin-like growth factor 1 receptorHomo sapiens (human)
receptor complexInsulin-like growth factor 1 receptorHomo sapiens (human)
protein kinase complexInsulin-like growth factor 1 receptorHomo sapiens (human)
axonInsulin-like growth factor 1 receptorHomo sapiens (human)
plasma membraneInsulin-like growth factor 1 receptorHomo sapiens (human)
insulin receptor complexInsulin-like growth factor 1 receptorHomo sapiens (human)
signal recognition particle receptor complexSignal recognition particle receptor subunit alphaHomo sapiens (human)
endoplasmic reticulum membraneSignal recognition particle receptor subunit alphaHomo sapiens (human)
membraneSignal recognition particle receptor subunit alphaHomo sapiens (human)
extracellular exosomeSignal recognition particle receptor subunit alphaHomo sapiens (human)
endoplasmic reticulum membraneSignal recognition particle receptor subunit alphaHomo sapiens (human)
nucleusCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
mitochondrionCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
mitochondrial inner membraneCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
mitochondrial respiratory chain complex IIICytochrome c1, heme protein, mitochondrialHomo sapiens (human)
membraneCytochrome c1, heme protein, mitochondrialHomo sapiens (human)
extracellular regionHepatocyte growth factor receptorHomo sapiens (human)
plasma membraneHepatocyte growth factor receptorHomo sapiens (human)
basal plasma membraneHepatocyte growth factor receptorHomo sapiens (human)
cell surfaceHepatocyte growth factor receptorHomo sapiens (human)
membraneHepatocyte growth factor receptorHomo sapiens (human)
postsynapseHepatocyte growth factor receptorHomo sapiens (human)
basal plasma membraneHepatocyte growth factor receptorHomo sapiens (human)
plasma membraneHepatocyte growth factor receptorHomo sapiens (human)
receptor complexHepatocyte growth factor receptorHomo sapiens (human)
actin filamentTyrosine-protein kinase HCKHomo sapiens (human)
nucleusTyrosine-protein kinase HCKHomo sapiens (human)
lysosomeTyrosine-protein kinase HCKHomo sapiens (human)
Golgi apparatusTyrosine-protein kinase HCKHomo sapiens (human)
cytosolTyrosine-protein kinase HCKHomo sapiens (human)
plasma membraneTyrosine-protein kinase HCKHomo sapiens (human)
caveolaTyrosine-protein kinase HCKHomo sapiens (human)
focal adhesionTyrosine-protein kinase HCKHomo sapiens (human)
cytoplasmic side of plasma membraneTyrosine-protein kinase HCKHomo sapiens (human)
transport vesicleTyrosine-protein kinase HCKHomo sapiens (human)
cell projectionTyrosine-protein kinase HCKHomo sapiens (human)
intracellular membrane-bounded organelleTyrosine-protein kinase HCKHomo sapiens (human)
plasma membraneTyrosine-protein kinase HCKHomo sapiens (human)
nucleusPlatelet-derived growth factor receptor betaHomo sapiens (human)
cytoplasmPlatelet-derived growth factor receptor betaHomo sapiens (human)
Golgi apparatusPlatelet-derived growth factor receptor betaHomo sapiens (human)
plasma membranePlatelet-derived growth factor receptor betaHomo sapiens (human)
focal adhesionPlatelet-derived growth factor receptor betaHomo sapiens (human)
membranePlatelet-derived growth factor receptor betaHomo sapiens (human)
apical plasma membranePlatelet-derived growth factor receptor betaHomo sapiens (human)
cytoplasmic vesiclePlatelet-derived growth factor receptor betaHomo sapiens (human)
lysosomal lumenPlatelet-derived growth factor receptor betaHomo sapiens (human)
intracellular membrane-bounded organellePlatelet-derived growth factor receptor betaHomo sapiens (human)
plasma membranePlatelet-derived growth factor receptor betaHomo sapiens (human)
receptor complexPlatelet-derived growth factor receptor betaHomo sapiens (human)
cytoskeletonTyrosine-protein kinase FgrHomo sapiens (human)
actin cytoskeletonTyrosine-protein kinase FgrHomo sapiens (human)
ruffle membraneTyrosine-protein kinase FgrHomo sapiens (human)
extracellular regionTyrosine-protein kinase FgrHomo sapiens (human)
mitochondrial inner membraneTyrosine-protein kinase FgrHomo sapiens (human)
mitochondrial intermembrane spaceTyrosine-protein kinase FgrHomo sapiens (human)
cytosolTyrosine-protein kinase FgrHomo sapiens (human)
plasma membraneTyrosine-protein kinase FgrHomo sapiens (human)
aggresomeTyrosine-protein kinase FgrHomo sapiens (human)
secretory granule lumenTyrosine-protein kinase FgrHomo sapiens (human)
extracellular exosomeTyrosine-protein kinase FgrHomo sapiens (human)
plasma membraneTyrosine-protein kinase FgrHomo sapiens (human)
cellular_componentSerine/threonine-protein kinase A-RafHomo sapiens (human)
cytosolSerine/threonine-protein kinase A-RafHomo sapiens (human)
cytosolSerine/threonine-protein kinase A-RafHomo sapiens (human)
mitochondrionSerine/threonine-protein kinase A-RafHomo sapiens (human)
extracellular regionGlycogen phosphorylase, brain formHomo sapiens (human)
cytoplasmGlycogen phosphorylase, brain formHomo sapiens (human)
membraneGlycogen phosphorylase, brain formHomo sapiens (human)
azurophil granule lumenGlycogen phosphorylase, brain formHomo sapiens (human)
extracellular exosomeGlycogen phosphorylase, brain formHomo sapiens (human)
cytoplasmGlycogen phosphorylase, brain formHomo sapiens (human)
cytosolBreakpoint cluster region proteinHomo sapiens (human)
plasma membraneBreakpoint cluster region proteinHomo sapiens (human)
postsynaptic densityBreakpoint cluster region proteinHomo sapiens (human)
membraneBreakpoint cluster region proteinHomo sapiens (human)
axonBreakpoint cluster region proteinHomo sapiens (human)
dendritic spineBreakpoint cluster region proteinHomo sapiens (human)
extracellular exosomeBreakpoint cluster region proteinHomo sapiens (human)
protein-containing complexBreakpoint cluster region proteinHomo sapiens (human)
Schaffer collateral - CA1 synapseBreakpoint cluster region proteinHomo sapiens (human)
glutamatergic synapseBreakpoint cluster region proteinHomo sapiens (human)
membraneBreakpoint cluster region proteinHomo sapiens (human)
nucleusSerine/threonine-protein kinase pim-1Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase pim-1Homo sapiens (human)
nucleolusSerine/threonine-protein kinase pim-1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase pim-1Homo sapiens (human)
cytosolSerine/threonine-protein kinase pim-1Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase pim-1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase pim-1Homo sapiens (human)
extracellular regionFibroblast growth factor receptor 1Homo sapiens (human)
nucleusFibroblast growth factor receptor 1Homo sapiens (human)
cytosolFibroblast growth factor receptor 1Homo sapiens (human)
plasma membraneFibroblast growth factor receptor 1Homo sapiens (human)
membraneFibroblast growth factor receptor 1Homo sapiens (human)
cytoplasmic vesicleFibroblast growth factor receptor 1Homo sapiens (human)
receptor complexFibroblast growth factor receptor 1Homo sapiens (human)
plasma membraneFibroblast growth factor receptor 1Homo sapiens (human)
nucleolusDNA topoisomerase 2-alphaHomo sapiens (human)
nuclear chromosomeDNA topoisomerase 2-alphaHomo sapiens (human)
centrioleDNA topoisomerase 2-alphaHomo sapiens (human)
chromosome, centromeric regionDNA topoisomerase 2-alphaHomo sapiens (human)
condensed chromosomeDNA topoisomerase 2-alphaHomo sapiens (human)
male germ cell nucleusDNA topoisomerase 2-alphaHomo sapiens (human)
nucleusDNA topoisomerase 2-alphaHomo sapiens (human)
nucleoplasmDNA topoisomerase 2-alphaHomo sapiens (human)
nucleolusDNA topoisomerase 2-alphaHomo sapiens (human)
cytoplasmDNA topoisomerase 2-alphaHomo sapiens (human)
DNA topoisomerase type II (double strand cut, ATP-hydrolyzing) complexDNA topoisomerase 2-alphaHomo sapiens (human)
protein-containing complexDNA topoisomerase 2-alphaHomo sapiens (human)
ribonucleoprotein complexDNA topoisomerase 2-alphaHomo sapiens (human)
nucleusDNA topoisomerase 2-alphaHomo sapiens (human)
nucleusCyclin-dependent kinase 4Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 4Homo sapiens (human)
nucleolusCyclin-dependent kinase 4Homo sapiens (human)
cytosolCyclin-dependent kinase 4Homo sapiens (human)
bicellular tight junctionCyclin-dependent kinase 4Homo sapiens (human)
nuclear membraneCyclin-dependent kinase 4Homo sapiens (human)
cyclin D1-CDK4 complexCyclin-dependent kinase 4Homo sapiens (human)
cyclin D2-CDK4 complexCyclin-dependent kinase 4Homo sapiens (human)
cyclin D3-CDK4 complexCyclin-dependent kinase 4Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 4Homo sapiens (human)
chromatinCyclin-dependent kinase 4Homo sapiens (human)
transcription regulator complexCyclin-dependent kinase 4Homo sapiens (human)
nucleusCyclin-dependent kinase 4Homo sapiens (human)
cytoplasmCyclin-dependent kinase 4Homo sapiens (human)
nucleusADP/ATP translocase 3Homo sapiens (human)
mitochondrionADP/ATP translocase 3Homo sapiens (human)
mitochondrial inner membraneADP/ATP translocase 3Homo sapiens (human)
membraneADP/ATP translocase 3Homo sapiens (human)
TIM23 mitochondrial import inner membrane translocase complexADP/ATP translocase 3Homo sapiens (human)
extracellular regionInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
nucleusInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
cytoplasmInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
peroxisomal membraneInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
cytosolInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
membraneInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
secretory granule lumenInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
extracellular exosomeInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
ficolin-1-rich granule lumenInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
cytoplasmInosine-5'-monophosphate dehydrogenase 2Homo sapiens (human)
podosomeProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
nucleoplasmProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cytoplasmProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
mitochondrionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
mitochondrial inner membraneProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
lysosomeProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
late endosomeProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cytosolProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
actin filamentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
plasma membraneProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
caveolaProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
focal adhesionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
cell junctionProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
ruffle membraneProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
neuronal cell bodyProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
dendritic growth coneProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
membrane raftProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
perinuclear region of cytoplasmProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
extracellular exosomeProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
synaptic membraneProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
glutamatergic synapseProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
postsynaptic specialization, intracellular componentProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
dendritic filopodiumProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
plasma membraneProto-oncogene tyrosine-protein kinase SrcHomo sapiens (human)
axonemecAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
cytoplasmcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
centrosomecAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
cytosolcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
plasma membranecAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
focal adhesioncAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
membranecAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
plasma membrane raftcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
extracellular exosomecAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
ciliary basecAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
cAMP-dependent protein kinase complexcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
nucleotide-activated protein kinase complexcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
protein-containing complexcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
cytosolcAMP-dependent protein kinase type II-alpha regulatory subunitHomo sapiens (human)
nucleusSerine/threonine-protein kinase B-rafHomo sapiens (human)
cytosolSerine/threonine-protein kinase B-rafHomo sapiens (human)
plasma membraneSerine/threonine-protein kinase B-rafHomo sapiens (human)
neuron projectionSerine/threonine-protein kinase B-rafHomo sapiens (human)
intracellular membrane-bounded organelleSerine/threonine-protein kinase B-rafHomo sapiens (human)
cell bodySerine/threonine-protein kinase B-rafHomo sapiens (human)
presynapseSerine/threonine-protein kinase B-rafHomo sapiens (human)
cytosolSerine/threonine-protein kinase B-rafHomo sapiens (human)
mitochondrionSerine/threonine-protein kinase B-rafHomo sapiens (human)
plasma membraneSerine/threonine-protein kinase B-rafHomo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
Z discPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cytosolPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
phosphorylase kinase complexPhosphorylase b kinase gamma catalytic chain, liver/testis isoformHomo sapiens (human)
nucleoplasmRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
cytosolRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
extracellular exosomeRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
cytosolRibosyldihydronicotinamide dehydrogenase [quinone]Homo sapiens (human)
actin cytoskeletonTyrosine-protein kinase FerHomo sapiens (human)
microtubule cytoskeletonTyrosine-protein kinase FerHomo sapiens (human)
lamellipodiumTyrosine-protein kinase FerHomo sapiens (human)
cell junctionTyrosine-protein kinase FerHomo sapiens (human)
nucleusTyrosine-protein kinase FerHomo sapiens (human)
cytoplasmTyrosine-protein kinase FerHomo sapiens (human)
cytosolTyrosine-protein kinase FerHomo sapiens (human)
adherens junctionTyrosine-protein kinase FerHomo sapiens (human)
cell cortexTyrosine-protein kinase FerHomo sapiens (human)
cytoplasmic side of plasma membraneTyrosine-protein kinase FerHomo sapiens (human)
chromatinTyrosine-protein kinase FerHomo sapiens (human)
plasma membraneTyrosine-protein kinase FerHomo sapiens (human)
ciliary basal bodyProtein kinase C alpha typeHomo sapiens (human)
nucleoplasmProtein kinase C alpha typeHomo sapiens (human)
cytoplasmProtein kinase C alpha typeHomo sapiens (human)
mitochondrionProtein kinase C alpha typeHomo sapiens (human)
endoplasmic reticulumProtein kinase C alpha typeHomo sapiens (human)
cytosolProtein kinase C alpha typeHomo sapiens (human)
plasma membraneProtein kinase C alpha typeHomo sapiens (human)
mitochondrial membraneProtein kinase C alpha typeHomo sapiens (human)
perinuclear region of cytoplasmProtein kinase C alpha typeHomo sapiens (human)
extracellular exosomeProtein kinase C alpha typeHomo sapiens (human)
alphav-beta3 integrin-PKCalpha complexProtein kinase C alpha typeHomo sapiens (human)
axonemecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cytoplasmcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
acrosomal vesiclecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
nucleuscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
nucleoplasmcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cytoplasmcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
mitochondrial matrixcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
centrosomecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cytosolcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
plasma membranecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
nuclear speckcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
neuromuscular junctioncAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
sperm flagellumcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
dendritic spinecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
plasma membrane raftcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
perinuclear region of cytoplasmcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
extracellular exosomecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
ciliary basecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
glutamatergic synapsecAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cAMP-dependent protein kinase complexcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
calcium channel complexcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
cytosolcAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
nucleuscAMP-dependent protein kinase catalytic subunit alphaHomo sapiens (human)
nucleusGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
nucleoplasmGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
cytoplasmGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
spindleGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
cytosolGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription factor TFIIH core complexGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription factor TFIID complexGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
transcription factor TFIIH holo complexGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
CAK-ERCC2 complexGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
MMXD complexGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
nucleusGeneral transcription and DNA repair factor IIH helicase subunit XPDHomo sapiens (human)
PcG protein complexCasein kinase II subunit alpha'Homo sapiens (human)
acrosomal vesicleCasein kinase II subunit alpha'Homo sapiens (human)
nucleusCasein kinase II subunit alpha'Homo sapiens (human)
nucleoplasmCasein kinase II subunit alpha'Homo sapiens (human)
cytosolCasein kinase II subunit alpha'Homo sapiens (human)
protein kinase CK2 complexCasein kinase II subunit alpha'Homo sapiens (human)
chromatinCasein kinase II subunit alpha'Homo sapiens (human)
cytosolCasein kinase II subunit alpha'Homo sapiens (human)
nucleusCasein kinase II subunit alpha'Homo sapiens (human)
Golgi membraneRas-related protein Rab-6AHomo sapiens (human)
acrosomal membraneRas-related protein Rab-6AHomo sapiens (human)
endoplasmic reticulum membraneRas-related protein Rab-6AHomo sapiens (human)
Golgi apparatusRas-related protein Rab-6AHomo sapiens (human)
trans-Golgi networkRas-related protein Rab-6AHomo sapiens (human)
cytosolRas-related protein Rab-6AHomo sapiens (human)
plasma membraneRas-related protein Rab-6AHomo sapiens (human)
membraneRas-related protein Rab-6AHomo sapiens (human)
secretory granule membraneRas-related protein Rab-6AHomo sapiens (human)
cytoplasmic vesicleRas-related protein Rab-6AHomo sapiens (human)
trans-Golgi network membraneRas-related protein Rab-6AHomo sapiens (human)
extracellular exosomeRas-related protein Rab-6AHomo sapiens (human)
endosome to plasma membrane transport vesicleRas-related protein Rab-6AHomo sapiens (human)
Golgi apparatusRas-related protein Rab-6AHomo sapiens (human)
endomembrane systemRas-related protein Rab-6AHomo sapiens (human)
plasma membraneEphrin type-A receptor 1Homo sapiens (human)
receptor complexEphrin type-A receptor 1Homo sapiens (human)
plasma membraneEphrin type-A receptor 1Homo sapiens (human)
cytoplasmMultifunctional protein ADE2Homo sapiens (human)
cytosolMultifunctional protein ADE2Homo sapiens (human)
membraneMultifunctional protein ADE2Homo sapiens (human)
extracellular exosomeMultifunctional protein ADE2Homo sapiens (human)
nucleoplasmcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
cytosolcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
ciliary basecAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
cytosolcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
nucleuscAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
cAMP-dependent protein kinase complexcAMP-dependent protein kinase catalytic subunit gammaHomo sapiens (human)
nucleoplasmcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
centrosomecAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
cytosolcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
plasma membranecAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
extracellular exosomecAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
ciliary basecAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
cAMP-dependent protein kinase complexcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
cytosolcAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
nucleuscAMP-dependent protein kinase catalytic subunit betaHomo sapiens (human)
mitochondrial inner membraneFerrochelatase, mitochondrialHomo sapiens (human)
mitochondrial matrixFerrochelatase, mitochondrialHomo sapiens (human)
mitochondrionFerrochelatase, mitochondrialHomo sapiens (human)
nucleoplasmRibosomal protein S6 kinase beta-1Homo sapiens (human)
mitochondrionRibosomal protein S6 kinase beta-1Homo sapiens (human)
mitochondrial outer membraneRibosomal protein S6 kinase beta-1Homo sapiens (human)
cytosolRibosomal protein S6 kinase beta-1Homo sapiens (human)
cell surfaceRibosomal protein S6 kinase beta-1Homo sapiens (human)
neuron projectionRibosomal protein S6 kinase beta-1Homo sapiens (human)
perinuclear region of cytoplasmRibosomal protein S6 kinase beta-1Homo sapiens (human)
postsynapseRibosomal protein S6 kinase beta-1Homo sapiens (human)
glutamatergic synapseRibosomal protein S6 kinase beta-1Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase beta-1Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase beta-1Homo sapiens (human)
cytoplasmTyrosine-protein kinase JAK1Homo sapiens (human)
plasma membraneTyrosine-protein kinase JAK1Homo sapiens (human)
cytoplasmic side of plasma membraneTyrosine-protein kinase JAK1Homo sapiens (human)
extrinsic component of cytoplasmic side of plasma membraneTyrosine-protein kinase JAK1Homo sapiens (human)
nucleusTyrosine-protein kinase JAK1Homo sapiens (human)
cytoplasmTyrosine-protein kinase JAK1Homo sapiens (human)
endosomeTyrosine-protein kinase JAK1Homo sapiens (human)
cytosolTyrosine-protein kinase JAK1Homo sapiens (human)
cytoskeletonTyrosine-protein kinase JAK1Homo sapiens (human)
focal adhesionTyrosine-protein kinase JAK1Homo sapiens (human)
cytosolTyrosine-protein kinase JAK1Homo sapiens (human)
chromosome, telomeric regionCyclin-dependent kinase 2Homo sapiens (human)
condensed chromosomeCyclin-dependent kinase 2Homo sapiens (human)
X chromosomeCyclin-dependent kinase 2Homo sapiens (human)
Y chromosomeCyclin-dependent kinase 2Homo sapiens (human)
male germ cell nucleusCyclin-dependent kinase 2Homo sapiens (human)
nucleusCyclin-dependent kinase 2Homo sapiens (human)
nuclear envelopeCyclin-dependent kinase 2Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 2Homo sapiens (human)
cytoplasmCyclin-dependent kinase 2Homo sapiens (human)
endosomeCyclin-dependent kinase 2Homo sapiens (human)
centrosomeCyclin-dependent kinase 2Homo sapiens (human)
cytosolCyclin-dependent kinase 2Homo sapiens (human)
Cajal bodyCyclin-dependent kinase 2Homo sapiens (human)
cyclin A1-CDK2 complexCyclin-dependent kinase 2Homo sapiens (human)
cyclin A2-CDK2 complexCyclin-dependent kinase 2Homo sapiens (human)
cyclin E1-CDK2 complexCyclin-dependent kinase 2Homo sapiens (human)
cyclin E2-CDK2 complexCyclin-dependent kinase 2Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 2Homo sapiens (human)
transcription regulator complexCyclin-dependent kinase 2Homo sapiens (human)
cytoplasmCyclin-dependent kinase 2Homo sapiens (human)
nucleusCyclin-dependent kinase 2Homo sapiens (human)
cytoplasmBeta-adrenergic receptor kinase 1Homo sapiens (human)
cytosolBeta-adrenergic receptor kinase 1Homo sapiens (human)
plasma membraneBeta-adrenergic receptor kinase 1Homo sapiens (human)
ciliumBeta-adrenergic receptor kinase 1Homo sapiens (human)
membraneBeta-adrenergic receptor kinase 1Homo sapiens (human)
presynapseBeta-adrenergic receptor kinase 1Homo sapiens (human)
postsynapseBeta-adrenergic receptor kinase 1Homo sapiens (human)
P-bodyProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
nucleusProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
cytoplasmProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
cytosolProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
cytoplasmic stress granuleProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
membraneProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
cytoplasmic ribonucleoprotein granuleProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
RISC complexProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
cytoplasmic stress granuleProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
P-bodyProbable ATP-dependent RNA helicase DDX6Homo sapiens (human)
nucleusMitogen-activated protein kinase 3 Homo sapiens (human)
nuclear envelopeMitogen-activated protein kinase 3 Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 3 Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 3 Homo sapiens (human)
mitochondrionMitogen-activated protein kinase 3 Homo sapiens (human)
early endosomeMitogen-activated protein kinase 3 Homo sapiens (human)
late endosomeMitogen-activated protein kinase 3 Homo sapiens (human)
endoplasmic reticulum lumenMitogen-activated protein kinase 3 Homo sapiens (human)
Golgi apparatusMitogen-activated protein kinase 3 Homo sapiens (human)
cytosolMitogen-activated protein kinase 3 Homo sapiens (human)
cytoskeletonMitogen-activated protein kinase 3 Homo sapiens (human)
plasma membraneMitogen-activated protein kinase 3 Homo sapiens (human)
caveolaMitogen-activated protein kinase 3 Homo sapiens (human)
focal adhesionMitogen-activated protein kinase 3 Homo sapiens (human)
pseudopodiumMitogen-activated protein kinase 3 Homo sapiens (human)
glutamatergic synapseMitogen-activated protein kinase 3 Homo sapiens (human)
nucleusMitogen-activated protein kinase 3 Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 3 Homo sapiens (human)
cytoplasmMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
cytosolMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
plasma membraneMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
dendriteMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
extracellular exosomeMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
plasma membraneMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
cytoplasmMAP/microtubule affinity-regulating kinase 3Homo sapiens (human)
nucleoplasmDeoxycytidine kinaseHomo sapiens (human)
cytosolDeoxycytidine kinaseHomo sapiens (human)
mitochondrionDeoxycytidine kinaseHomo sapiens (human)
cytoplasmDeoxycytidine kinaseHomo sapiens (human)
extracellular regionMitogen-activated protein kinase 1Homo sapiens (human)
nucleusMitogen-activated protein kinase 1Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 1Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 1Homo sapiens (human)
mitochondrionMitogen-activated protein kinase 1Homo sapiens (human)
early endosomeMitogen-activated protein kinase 1Homo sapiens (human)
late endosomeMitogen-activated protein kinase 1Homo sapiens (human)
endoplasmic reticulum lumenMitogen-activated protein kinase 1Homo sapiens (human)
Golgi apparatusMitogen-activated protein kinase 1Homo sapiens (human)
centrosomeMitogen-activated protein kinase 1Homo sapiens (human)
cytosolMitogen-activated protein kinase 1Homo sapiens (human)
cytoskeletonMitogen-activated protein kinase 1Homo sapiens (human)
plasma membraneMitogen-activated protein kinase 1Homo sapiens (human)
caveolaMitogen-activated protein kinase 1Homo sapiens (human)
focal adhesionMitogen-activated protein kinase 1Homo sapiens (human)
pseudopodiumMitogen-activated protein kinase 1Homo sapiens (human)
azurophil granule lumenMitogen-activated protein kinase 1Homo sapiens (human)
synapseMitogen-activated protein kinase 1Homo sapiens (human)
mitotic spindleMitogen-activated protein kinase 1Homo sapiens (human)
ficolin-1-rich granule lumenMitogen-activated protein kinase 1Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 1Homo sapiens (human)
nucleusMitogen-activated protein kinase 1Homo sapiens (human)
plasma membraneEphrin type-A receptor 2Homo sapiens (human)
focal adhesionEphrin type-A receptor 2Homo sapiens (human)
cell surfaceEphrin type-A receptor 2Homo sapiens (human)
lamellipodiumEphrin type-A receptor 2Homo sapiens (human)
leading edge membraneEphrin type-A receptor 2Homo sapiens (human)
lamellipodium membraneEphrin type-A receptor 2Homo sapiens (human)
ruffle membraneEphrin type-A receptor 2Homo sapiens (human)
tight junctionEphrin type-A receptor 2Homo sapiens (human)
receptor complexEphrin type-A receptor 2Homo sapiens (human)
plasma membraneEphrin type-A receptor 2Homo sapiens (human)
extracellular regionEphrin type-B receptor 2Homo sapiens (human)
nucleoplasmEphrin type-B receptor 2Homo sapiens (human)
cytosolEphrin type-B receptor 2Homo sapiens (human)
plasma membraneEphrin type-B receptor 2Homo sapiens (human)
cell surfaceEphrin type-B receptor 2Homo sapiens (human)
axonEphrin type-B receptor 2Homo sapiens (human)
dendriteEphrin type-B receptor 2Homo sapiens (human)
presynaptic membraneEphrin type-B receptor 2Homo sapiens (human)
neuronal cell bodyEphrin type-B receptor 2Homo sapiens (human)
dendritic spineEphrin type-B receptor 2Homo sapiens (human)
postsynaptic membraneEphrin type-B receptor 2Homo sapiens (human)
hippocampal mossy fiber to CA3 synapseEphrin type-B receptor 2Homo sapiens (human)
postsynapseEphrin type-B receptor 2Homo sapiens (human)
glutamatergic synapseEphrin type-B receptor 2Homo sapiens (human)
plasma membraneEphrin type-B receptor 2Homo sapiens (human)
dendriteEphrin type-B receptor 2Homo sapiens (human)
plasma membraneNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cytoplasmic side of plasma membraneNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
extrinsic component of plasma membraneNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
extrinsic component of cytoplasmic side of plasma membraneNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
nucleusNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cytoplasmNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cytosolNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cytoskeletonNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
plasma membraneNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
interleukin-12 receptor complexNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
extracellular exosomeNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
interleukin-23 receptor complexNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
cytosolNon-receptor tyrosine-protein kinase TYK2Homo sapiens (human)
nucleoplasmUMP-CMP kinase Homo sapiens (human)
nucleolusUMP-CMP kinase Homo sapiens (human)
cytosolUMP-CMP kinase Homo sapiens (human)
extracellular exosomeUMP-CMP kinase Homo sapiens (human)
cytoplasmUMP-CMP kinase Homo sapiens (human)
nucleusUMP-CMP kinase Homo sapiens (human)
nucleusPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
cytosolPhosphatidylethanolamine-binding protein 1Homo sapiens (human)
extracellular exosomePhosphatidylethanolamine-binding protein 1Homo sapiens (human)
nucleusWee1-like protein kinaseHomo sapiens (human)
nucleoplasmWee1-like protein kinaseHomo sapiens (human)
nucleolusWee1-like protein kinaseHomo sapiens (human)
cytoplasmWee1-like protein kinaseHomo sapiens (human)
endoplasmic reticulum membraneHeme oxygenase 2Homo sapiens (human)
plasma membraneHeme oxygenase 2Homo sapiens (human)
membraneHeme oxygenase 2Homo sapiens (human)
specific granule membraneHeme oxygenase 2Homo sapiens (human)
cytosolS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
methionine adenosyltransferase complexS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
cytosolS-adenosylmethionine synthase isoform type-2Homo sapiens (human)
nucleusDnaJ homolog subfamily A member 1Homo sapiens (human)
mitochondrionDnaJ homolog subfamily A member 1Homo sapiens (human)
cytosolDnaJ homolog subfamily A member 1Homo sapiens (human)
microtubule cytoskeletonDnaJ homolog subfamily A member 1Homo sapiens (human)
membraneDnaJ homolog subfamily A member 1Homo sapiens (human)
perinuclear region of cytoplasmDnaJ homolog subfamily A member 1Homo sapiens (human)
extracellular exosomeDnaJ homolog subfamily A member 1Homo sapiens (human)
cytoplasmic side of endoplasmic reticulum membraneDnaJ homolog subfamily A member 1Homo sapiens (human)
cytoplasmDnaJ homolog subfamily A member 1Homo sapiens (human)
cytosolDnaJ homolog subfamily A member 1Homo sapiens (human)
cytoplasmRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
mitochondrial intermembrane spaceRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
membraneRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
nucleusRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
nucleoplasmRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cytoplasmRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
spindleRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cytosolRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
plasma membraneRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cell-cell junctionRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
cell cortexRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
microtubule cytoskeletonRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
lamellipodiumRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
vesicleRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
ciliary basal bodyRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
postsynapseRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
glutamatergic synapseRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
protein-containing complexRAC-alpha serine/threonine-protein kinaseHomo sapiens (human)
nucleusRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
nucleoplasmRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
early endosomeRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
cytosolRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
plasma membraneRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
cell cortexRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
ruffle membraneRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
intracellular membrane-bounded organelleRAC-beta serine/threonine-protein kinaseHomo sapiens (human)
cytoplasmDual specificity protein kinase TTKHomo sapiens (human)
spindleDual specificity protein kinase TTKHomo sapiens (human)
membraneDual specificity protein kinase TTKHomo sapiens (human)
kinetochoreDual specificity protein kinase TTKHomo sapiens (human)
nucleusDual specificity protein kinase TTKHomo sapiens (human)
chromosome, telomeric regionDNA replication licensing factor MCM4Homo sapiens (human)
nucleusDNA replication licensing factor MCM4Homo sapiens (human)
nucleoplasmDNA replication licensing factor MCM4Homo sapiens (human)
membraneDNA replication licensing factor MCM4Homo sapiens (human)
MCM complexDNA replication licensing factor MCM4Homo sapiens (human)
CMG complexDNA replication licensing factor MCM4Homo sapiens (human)
nucleusDNA replication licensing factor MCM4Homo sapiens (human)
postsynaptic actin cytoskeletonMyosin-10Homo sapiens (human)
stress fiberMyosin-10Homo sapiens (human)
nucleusMyosin-10Homo sapiens (human)
cytoplasmMyosin-10Homo sapiens (human)
cytosolMyosin-10Homo sapiens (human)
cell cortexMyosin-10Homo sapiens (human)
lamellipodiumMyosin-10Homo sapiens (human)
midbodyMyosin-10Homo sapiens (human)
cleavage furrowMyosin-10Homo sapiens (human)
actomyosinMyosin-10Homo sapiens (human)
extracellular exosomeMyosin-10Homo sapiens (human)
myosin II filamentMyosin-10Homo sapiens (human)
myosin complexMyosin-10Homo sapiens (human)
myosin II complexMyosin-10Homo sapiens (human)
myosin filamentMyosin-10Homo sapiens (human)
cytoplasmMyosin-10Homo sapiens (human)
extracellular regionDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
nucleusDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
mitochondrionDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
early endosomeDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
late endosomeDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
peroxisomal membraneDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
endoplasmic reticulumDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
Golgi apparatusDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
cytosolDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
microtubuleDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
cell-cell junctionDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
focal adhesionDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
cytoplasmic side of plasma membraneDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
perinuclear region of cytoplasmDual specificity mitogen-activated protein kinase kinase 2Homo sapiens (human)
endoplasmic reticulumReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
endoplasmic reticulum lumenReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
plasma membraneReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
endosome membraneReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
receptor complexReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
plasma membraneReceptor-type tyrosine-protein kinase FLT3Homo sapiens (human)
caveolaBone morphogenetic protein receptor type-1AHomo sapiens (human)
plasma membraneBone morphogenetic protein receptor type-1AHomo sapiens (human)
external side of plasma membraneBone morphogenetic protein receptor type-1AHomo sapiens (human)
membraneBone morphogenetic protein receptor type-1AHomo sapiens (human)
dendriteBone morphogenetic protein receptor type-1AHomo sapiens (human)
neuronal cell bodyBone morphogenetic protein receptor type-1AHomo sapiens (human)
HFE-transferrin receptor complexBone morphogenetic protein receptor type-1AHomo sapiens (human)
plasma membraneBone morphogenetic protein receptor type-1AHomo sapiens (human)
receptor complexBone morphogenetic protein receptor type-1AHomo sapiens (human)
cytosolActivin receptor type-1BHomo sapiens (human)
plasma membraneActivin receptor type-1BHomo sapiens (human)
cell surfaceActivin receptor type-1BHomo sapiens (human)
receptor complexActivin receptor type-1BHomo sapiens (human)
activin receptor complexActivin receptor type-1BHomo sapiens (human)
plasma membraneActivin receptor type-1BHomo sapiens (human)
nucleusTGF-beta receptor type-1Homo sapiens (human)
endosomeTGF-beta receptor type-1Homo sapiens (human)
plasma membraneTGF-beta receptor type-1Homo sapiens (human)
bicellular tight junctionTGF-beta receptor type-1Homo sapiens (human)
cell surfaceTGF-beta receptor type-1Homo sapiens (human)
membrane raftTGF-beta receptor type-1Homo sapiens (human)
transforming growth factor beta ligand-receptor complexTGF-beta receptor type-1Homo sapiens (human)
receptor complexTGF-beta receptor type-1Homo sapiens (human)
plasma membraneTGF-beta receptor type-1Homo sapiens (human)
activin receptor complexTGF-beta receptor type-1Homo sapiens (human)
extracellular regionTGF-beta receptor type-2Homo sapiens (human)
cytosolTGF-beta receptor type-2Homo sapiens (human)
plasma membraneTGF-beta receptor type-2Homo sapiens (human)
caveolaTGF-beta receptor type-2Homo sapiens (human)
external side of plasma membraneTGF-beta receptor type-2Homo sapiens (human)
membraneTGF-beta receptor type-2Homo sapiens (human)
membrane raftTGF-beta receptor type-2Homo sapiens (human)
transforming growth factor beta ligand-receptor complexTGF-beta receptor type-2Homo sapiens (human)
receptor complexTGF-beta receptor type-2Homo sapiens (human)
plasma membraneTGF-beta receptor type-2Homo sapiens (human)
mitochondrionElectron transfer flavoprotein subunit betaHomo sapiens (human)
mitochondrial matrixElectron transfer flavoprotein subunit betaHomo sapiens (human)
electron transfer flavoprotein complexElectron transfer flavoprotein subunit betaHomo sapiens (human)
mitochondrionElectron transfer flavoprotein subunit betaHomo sapiens (human)
cytoplasmTyrosine-protein kinase CSKHomo sapiens (human)
cytosolTyrosine-protein kinase CSKHomo sapiens (human)
plasma membraneTyrosine-protein kinase CSKHomo sapiens (human)
cell-cell junctionTyrosine-protein kinase CSKHomo sapiens (human)
extracellular exosomeTyrosine-protein kinase CSKHomo sapiens (human)
plasma membraneTyrosine-protein kinase CSKHomo sapiens (human)
mitochondrial matrixGlycine--tRNA ligaseHomo sapiens (human)
cytosolGlycine--tRNA ligaseHomo sapiens (human)
secretory granuleGlycine--tRNA ligaseHomo sapiens (human)
axonGlycine--tRNA ligaseHomo sapiens (human)
extracellular exosomeGlycine--tRNA ligaseHomo sapiens (human)
cytoplasmGlycine--tRNA ligaseHomo sapiens (human)
mitochondrionGlycine--tRNA ligaseHomo sapiens (human)
Golgi membraneProtein kinase C iota typeHomo sapiens (human)
nucleusProtein kinase C iota typeHomo sapiens (human)
nucleoplasmProtein kinase C iota typeHomo sapiens (human)
endosomeProtein kinase C iota typeHomo sapiens (human)
cytosolProtein kinase C iota typeHomo sapiens (human)
plasma membraneProtein kinase C iota typeHomo sapiens (human)
brush borderProtein kinase C iota typeHomo sapiens (human)
bicellular tight junctionProtein kinase C iota typeHomo sapiens (human)
microtubule cytoskeletonProtein kinase C iota typeHomo sapiens (human)
apical plasma membraneProtein kinase C iota typeHomo sapiens (human)
cell leading edgeProtein kinase C iota typeHomo sapiens (human)
Schmidt-Lanterman incisureProtein kinase C iota typeHomo sapiens (human)
intercellular bridgeProtein kinase C iota typeHomo sapiens (human)
extracellular exosomeProtein kinase C iota typeHomo sapiens (human)
tight junctionProtein kinase C iota typeHomo sapiens (human)
Schaffer collateral - CA1 synapseProtein kinase C iota typeHomo sapiens (human)
glutamatergic synapseProtein kinase C iota typeHomo sapiens (human)
PAR polarity complexProtein kinase C iota typeHomo sapiens (human)
nuclear exosome (RNase complex)Exosome RNA helicase MTR4Homo sapiens (human)
exosome (RNase complex)Exosome RNA helicase MTR4Homo sapiens (human)
nucleusExosome RNA helicase MTR4Homo sapiens (human)
nucleoplasmExosome RNA helicase MTR4Homo sapiens (human)
nucleolusExosome RNA helicase MTR4Homo sapiens (human)
nuclear speckExosome RNA helicase MTR4Homo sapiens (human)
TRAMP complexExosome RNA helicase MTR4Homo sapiens (human)
catalytic step 2 spliceosomeExosome RNA helicase MTR4Homo sapiens (human)
nucleusExosome RNA helicase MTR4Homo sapiens (human)
PML bodySerine/threonine-protein kinase mTORHomo sapiens (human)
lysosomal membraneSerine/threonine-protein kinase mTORHomo sapiens (human)
cytosolSerine/threonine-protein kinase mTORHomo sapiens (human)
Golgi membraneSerine/threonine-protein kinase mTORHomo sapiens (human)
nucleoplasmSerine/threonine-protein kinase mTORHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase mTORHomo sapiens (human)
mitochondrial outer membraneSerine/threonine-protein kinase mTORHomo sapiens (human)
lysosomeSerine/threonine-protein kinase mTORHomo sapiens (human)
lysosomal membraneSerine/threonine-protein kinase mTORHomo sapiens (human)
endoplasmic reticulum membraneSerine/threonine-protein kinase mTORHomo sapiens (human)
cytosolSerine/threonine-protein kinase mTORHomo sapiens (human)
endomembrane systemSerine/threonine-protein kinase mTORHomo sapiens (human)
membraneSerine/threonine-protein kinase mTORHomo sapiens (human)
dendriteSerine/threonine-protein kinase mTORHomo sapiens (human)
TORC1 complexSerine/threonine-protein kinase mTORHomo sapiens (human)
TORC2 complexSerine/threonine-protein kinase mTORHomo sapiens (human)
phagocytic vesicleSerine/threonine-protein kinase mTORHomo sapiens (human)
nuclear envelopeSerine/threonine-protein kinase mTORHomo sapiens (human)
nucleusSerine/threonine-protein kinase mTORHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase mTORHomo sapiens (human)
cytosolTyrosine-protein kinase TecHomo sapiens (human)
cytoskeletonTyrosine-protein kinase TecHomo sapiens (human)
plasma membraneTyrosine-protein kinase TecHomo sapiens (human)
plasma membraneTyrosine-protein kinase TecHomo sapiens (human)
cytosolTyrosine-protein kinase ABL2Homo sapiens (human)
actin cytoskeletonTyrosine-protein kinase ABL2Homo sapiens (human)
plasma membraneTyrosine-protein kinase ABL2Homo sapiens (human)
extracellular regionTyrosine-protein kinase FRKHomo sapiens (human)
nucleusTyrosine-protein kinase FRKHomo sapiens (human)
nucleoplasmTyrosine-protein kinase FRKHomo sapiens (human)
cytosolTyrosine-protein kinase FRKHomo sapiens (human)
azurophil granule lumenTyrosine-protein kinase FRKHomo sapiens (human)
specific granule lumenTyrosine-protein kinase FRKHomo sapiens (human)
extracellular exosomeTyrosine-protein kinase FRKHomo sapiens (human)
plasma membraneTyrosine-protein kinase FRKHomo sapiens (human)
plasma membraneG protein-coupled receptor kinase 6Homo sapiens (human)
membraneG protein-coupled receptor kinase 6Homo sapiens (human)
cytoplasmG protein-coupled receptor kinase 6Homo sapiens (human)
cytoplasmTyrosine-protein kinase SYKHomo sapiens (human)
nucleusTyrosine-protein kinase SYKHomo sapiens (human)
cytoplasmTyrosine-protein kinase SYKHomo sapiens (human)
cytosolTyrosine-protein kinase SYKHomo sapiens (human)
plasma membraneTyrosine-protein kinase SYKHomo sapiens (human)
early phagosomeTyrosine-protein kinase SYKHomo sapiens (human)
B cell receptor complexTyrosine-protein kinase SYKHomo sapiens (human)
protein-containing complexTyrosine-protein kinase SYKHomo sapiens (human)
T cell receptor complexTyrosine-protein kinase SYKHomo sapiens (human)
plasma membraneTyrosine-protein kinase SYKHomo sapiens (human)
proteasome complex26S proteasome regulatory subunit 6BHomo sapiens (human)
nucleus26S proteasome regulatory subunit 6BHomo sapiens (human)
nucleoplasm26S proteasome regulatory subunit 6BHomo sapiens (human)
cytosol26S proteasome regulatory subunit 6BHomo sapiens (human)
membrane26S proteasome regulatory subunit 6BHomo sapiens (human)
inclusion body26S proteasome regulatory subunit 6BHomo sapiens (human)
synapse26S proteasome regulatory subunit 6BHomo sapiens (human)
proteasome accessory complex26S proteasome regulatory subunit 6BHomo sapiens (human)
cytosolic proteasome complex26S proteasome regulatory subunit 6BHomo sapiens (human)
proteasome regulatory particle, base subcomplex26S proteasome regulatory subunit 6BHomo sapiens (human)
cytoplasmMitogen-activated protein kinase 8Homo sapiens (human)
nucleusMitogen-activated protein kinase 8Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 8Homo sapiens (human)
cytosolMitogen-activated protein kinase 8Homo sapiens (human)
axonMitogen-activated protein kinase 8Homo sapiens (human)
synapseMitogen-activated protein kinase 8Homo sapiens (human)
basal dendriteMitogen-activated protein kinase 8Homo sapiens (human)
nucleusMitogen-activated protein kinase 8Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 9Homo sapiens (human)
mitochondrionMitogen-activated protein kinase 9Homo sapiens (human)
cytosolMitogen-activated protein kinase 9Homo sapiens (human)
plasma membraneMitogen-activated protein kinase 9Homo sapiens (human)
nuclear speckMitogen-activated protein kinase 9Homo sapiens (human)
Schaffer collateral - CA1 synapseMitogen-activated protein kinase 9Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 9Homo sapiens (human)
nucleusMitogen-activated protein kinase 9Homo sapiens (human)
cytoplasmDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
nucleoplasmDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
cytosolDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
membraneDual specificity mitogen-activated protein kinase kinase 3Homo sapiens (human)
photoreceptor outer segmentPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
photoreceptor inner segmentPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
nucleoplasmPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
lysosomePhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
autophagosomePhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
cytosolPhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
plasma membranePhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
plasma membranePhosphatidylinositol 5-phosphate 4-kinase type-2 alphaHomo sapiens (human)
mRNA cleavage and polyadenylation specificity factor complexCasein kinase I isoform alphaHomo sapiens (human)
keratin filamentCasein kinase I isoform alphaHomo sapiens (human)
kinetochoreCasein kinase I isoform alphaHomo sapiens (human)
centrosomeCasein kinase I isoform alphaHomo sapiens (human)
spindleCasein kinase I isoform alphaHomo sapiens (human)
cytosolCasein kinase I isoform alphaHomo sapiens (human)
ciliumCasein kinase I isoform alphaHomo sapiens (human)
membraneCasein kinase I isoform alphaHomo sapiens (human)
nuclear speckCasein kinase I isoform alphaHomo sapiens (human)
beta-catenin destruction complexCasein kinase I isoform alphaHomo sapiens (human)
ciliary basal bodyCasein kinase I isoform alphaHomo sapiens (human)
cytoplasmCasein kinase I isoform alphaHomo sapiens (human)
nucleusCasein kinase I isoform alphaHomo sapiens (human)
nucleusCasein kinase I isoform deltaHomo sapiens (human)
nucleoplasmCasein kinase I isoform deltaHomo sapiens (human)
Golgi apparatusCasein kinase I isoform deltaHomo sapiens (human)
centrosomeCasein kinase I isoform deltaHomo sapiens (human)
spindleCasein kinase I isoform deltaHomo sapiens (human)
cytosolCasein kinase I isoform deltaHomo sapiens (human)
spindle microtubuleCasein kinase I isoform deltaHomo sapiens (human)
plasma membraneCasein kinase I isoform deltaHomo sapiens (human)
endoplasmic reticulum-Golgi intermediate compartment membraneCasein kinase I isoform deltaHomo sapiens (human)
ciliary basal bodyCasein kinase I isoform deltaHomo sapiens (human)
perinuclear region of cytoplasmCasein kinase I isoform deltaHomo sapiens (human)
nucleusCasein kinase I isoform deltaHomo sapiens (human)
cytoplasmCasein kinase I isoform deltaHomo sapiens (human)
spindle microtubuleCasein kinase I isoform deltaHomo sapiens (human)
nucleusMAP kinase-activated protein kinase 2Homo sapiens (human)
nucleoplasmMAP kinase-activated protein kinase 2Homo sapiens (human)
cytoplasmMAP kinase-activated protein kinase 2Homo sapiens (human)
centrosomeMAP kinase-activated protein kinase 2Homo sapiens (human)
cytosolMAP kinase-activated protein kinase 2Homo sapiens (human)
extracellular exosomeMAP kinase-activated protein kinase 2Homo sapiens (human)
nucleusMAP kinase-activated protein kinase 2Homo sapiens (human)
cytoplasmMAP kinase-activated protein kinase 2Homo sapiens (human)
mitochondrionElongation factor Tu, mitochondrialHomo sapiens (human)
mitochondrial outer membraneElongation factor Tu, mitochondrialHomo sapiens (human)
membraneElongation factor Tu, mitochondrialHomo sapiens (human)
mitochondrial nucleoidElongation factor Tu, mitochondrialHomo sapiens (human)
synapseElongation factor Tu, mitochondrialHomo sapiens (human)
extracellular exosomeElongation factor Tu, mitochondrialHomo sapiens (human)
mitochondrionElongation factor Tu, mitochondrialHomo sapiens (human)
nucleusCholine-phosphate cytidylyltransferase AHomo sapiens (human)
nuclear envelopeCholine-phosphate cytidylyltransferase AHomo sapiens (human)
endoplasmic reticulumCholine-phosphate cytidylyltransferase AHomo sapiens (human)
endoplasmic reticulum membraneCholine-phosphate cytidylyltransferase AHomo sapiens (human)
cytosolCholine-phosphate cytidylyltransferase AHomo sapiens (human)
glycogen granuleCholine-phosphate cytidylyltransferase AHomo sapiens (human)
endoplasmic reticulumCholine-phosphate cytidylyltransferase AHomo sapiens (human)
cytoplasmCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
cytosolCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
cytoplasmCysteine--tRNA ligase, cytoplasmicHomo sapiens (human)
nucleusCasein kinase I isoform epsilonHomo sapiens (human)
nucleoplasmCasein kinase I isoform epsilonHomo sapiens (human)
cytoplasmCasein kinase I isoform epsilonHomo sapiens (human)
cytosolCasein kinase I isoform epsilonHomo sapiens (human)
growth coneCasein kinase I isoform epsilonHomo sapiens (human)
neuronal cell bodyCasein kinase I isoform epsilonHomo sapiens (human)
ribonucleoprotein complexCasein kinase I isoform epsilonHomo sapiens (human)
cytoplasmCasein kinase I isoform epsilonHomo sapiens (human)
nucleusCasein kinase I isoform epsilonHomo sapiens (human)
nucleoplasmVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
nucleolusVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
mitochondrionVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
mitochondrial inner membraneVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
mitochondrial matrixVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
mitochondrial membraneVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
mitochondrial nucleoidVery long-chain specific acyl-CoA dehydrogenase, mitochondrialHomo sapiens (human)
nucleusDual specificity protein kinase CLK1Homo sapiens (human)
nucleusDual specificity protein kinase CLK2Homo sapiens (human)
nucleoplasmDual specificity protein kinase CLK2Homo sapiens (human)
nuclear bodyDual specificity protein kinase CLK2Homo sapiens (human)
nuclear speckDual specificity protein kinase CLK2Homo sapiens (human)
nucleusDual specificity protein kinase CLK2Homo sapiens (human)
acrosomal vesicleDual specificity protein kinase CLK3Homo sapiens (human)
nucleusDual specificity protein kinase CLK3Homo sapiens (human)
nucleoplasmDual specificity protein kinase CLK3Homo sapiens (human)
membraneDual specificity protein kinase CLK3Homo sapiens (human)
nuclear speckDual specificity protein kinase CLK3Homo sapiens (human)
intermediate filament cytoskeletonDual specificity protein kinase CLK3Homo sapiens (human)
mitochondrionGlycogen synthase kinase-3 alphaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 alphaHomo sapiens (human)
beta-catenin destruction complexGlycogen synthase kinase-3 alphaHomo sapiens (human)
neuronal cell bodyGlycogen synthase kinase-3 alphaHomo sapiens (human)
apical dendriteGlycogen synthase kinase-3 alphaHomo sapiens (human)
postsynapseGlycogen synthase kinase-3 alphaHomo sapiens (human)
proximal dendriteGlycogen synthase kinase-3 alphaHomo sapiens (human)
cytoplasmGlycogen synthase kinase-3 alphaHomo sapiens (human)
nucleusGlycogen synthase kinase-3 alphaHomo sapiens (human)
axonGlycogen synthase kinase-3 alphaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 alphaHomo sapiens (human)
glutamatergic synapseGlycogen synthase kinase-3 betaHomo sapiens (human)
nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
nucleoplasmGlycogen synthase kinase-3 betaHomo sapiens (human)
cytoplasmGlycogen synthase kinase-3 betaHomo sapiens (human)
mitochondrionGlycogen synthase kinase-3 betaHomo sapiens (human)
centrosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 betaHomo sapiens (human)
plasma membraneGlycogen synthase kinase-3 betaHomo sapiens (human)
axonGlycogen synthase kinase-3 betaHomo sapiens (human)
dendriteGlycogen synthase kinase-3 betaHomo sapiens (human)
beta-catenin destruction complexGlycogen synthase kinase-3 betaHomo sapiens (human)
presynapseGlycogen synthase kinase-3 betaHomo sapiens (human)
postsynapseGlycogen synthase kinase-3 betaHomo sapiens (human)
Wnt signalosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 betaHomo sapiens (human)
axonGlycogen synthase kinase-3 betaHomo sapiens (human)
nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
cytoplasmGlycogen synthase kinase-3 betaHomo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 7Homo sapiens (human)
fibrillar centerCyclin-dependent kinase 7Homo sapiens (human)
male germ cell nucleusCyclin-dependent kinase 7Homo sapiens (human)
nucleusCyclin-dependent kinase 7Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 7Homo sapiens (human)
cytosolCyclin-dependent kinase 7Homo sapiens (human)
plasma membraneCyclin-dependent kinase 7Homo sapiens (human)
perinuclear region of cytoplasmCyclin-dependent kinase 7Homo sapiens (human)
transcription factor TFIIH core complexCyclin-dependent kinase 7Homo sapiens (human)
transcription factor TFIIH holo complexCyclin-dependent kinase 7Homo sapiens (human)
CAK-ERCC2 complexCyclin-dependent kinase 7Homo sapiens (human)
transcription factor TFIIK complexCyclin-dependent kinase 7Homo sapiens (human)
cytoplasmCyclin-dependent kinase 7Homo sapiens (human)
nucleusCyclin-dependent kinase 7Homo sapiens (human)
nucleusCyclin-dependent kinase 9Homo sapiens (human)
nucleusCyclin-dependent kinase 9Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 9Homo sapiens (human)
cyclin/CDK positive transcription elongation factor complexCyclin-dependent kinase 9Homo sapiens (human)
membraneCyclin-dependent kinase 9Homo sapiens (human)
PML bodyCyclin-dependent kinase 9Homo sapiens (human)
cytoplasmic ribonucleoprotein granuleCyclin-dependent kinase 9Homo sapiens (human)
transcription elongation factor complexCyclin-dependent kinase 9Homo sapiens (human)
P-TEFb complexCyclin-dependent kinase 9Homo sapiens (human)
photoreceptor outer segmentRas-related protein Rab-27AHomo sapiens (human)
extracellular regionRas-related protein Rab-27AHomo sapiens (human)
lysosomeRas-related protein Rab-27AHomo sapiens (human)
late endosomeRas-related protein Rab-27AHomo sapiens (human)
cytosolRas-related protein Rab-27AHomo sapiens (human)
dendriteRas-related protein Rab-27AHomo sapiens (human)
multivesicular body membraneRas-related protein Rab-27AHomo sapiens (human)
Weibel-Palade bodyRas-related protein Rab-27AHomo sapiens (human)
melanosome membraneRas-related protein Rab-27AHomo sapiens (human)
specific granule lumenRas-related protein Rab-27AHomo sapiens (human)
melanosomeRas-related protein Rab-27AHomo sapiens (human)
extracellular exosomeRas-related protein Rab-27AHomo sapiens (human)
exocytic vesicleRas-related protein Rab-27AHomo sapiens (human)
exocytic vesicleRas-related protein Rab-27AHomo sapiens (human)
apical plasma membraneRas-related protein Rab-27AHomo sapiens (human)
Golgi apparatusRas-related protein Rab-27AHomo sapiens (human)
secretory granuleRas-related protein Rab-27AHomo sapiens (human)
melanosomeRas-related protein Rab-27AHomo sapiens (human)
cytoplasmInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
cell surfaceInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
nucleoplasmInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
lipid dropletInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
cytosolInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
plasma membraneInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
endosome membraneInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
protein-containing complexInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
nucleusInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
plasma membraneInterleukin-1 receptor-associated kinase 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
late endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transport vesiclePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmic vesicle membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuron projectionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ciliary basePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lumenal side of membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral part of cellPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
monoatomic ion channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-3Homo sapiens (human)
nucleolusRibosomal protein S6 kinase alpha-3Homo sapiens (human)
cytosolRibosomal protein S6 kinase alpha-3Homo sapiens (human)
synapseRibosomal protein S6 kinase alpha-3Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase alpha-3Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-3Homo sapiens (human)
kinetochoreSerine/threonine-protein kinase Nek2Homo sapiens (human)
kinetochoreSerine/threonine-protein kinase Nek2Homo sapiens (human)
condensed nuclear chromosomeSerine/threonine-protein kinase Nek2Homo sapiens (human)
spindle poleSerine/threonine-protein kinase Nek2Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase Nek2Homo sapiens (human)
nucleolusSerine/threonine-protein kinase Nek2Homo sapiens (human)
centrosomeSerine/threonine-protein kinase Nek2Homo sapiens (human)
cytosolSerine/threonine-protein kinase Nek2Homo sapiens (human)
microtubuleSerine/threonine-protein kinase Nek2Homo sapiens (human)
midbodySerine/threonine-protein kinase Nek2Homo sapiens (human)
protein-containing complexSerine/threonine-protein kinase Nek2Homo sapiens (human)
centrosomeSerine/threonine-protein kinase Nek2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase Nek2Homo sapiens (human)
nucleusSerine/threonine-protein kinase Nek2Homo sapiens (human)
nucleusSerine/threonine-protein kinase Nek3Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase Nek3Homo sapiens (human)
axonSerine/threonine-protein kinase Nek3Homo sapiens (human)
cytoplasmDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
cytosolDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
nucleoplasmDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
cytosolDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
cytoskeletonDual specificity mitogen-activated protein kinase kinase 6Homo sapiens (human)
spindle microtubuleSerine/threonine-protein kinase PLK1Homo sapiens (human)
kinetochoreSerine/threonine-protein kinase PLK1Homo sapiens (human)
synaptonemal complexSerine/threonine-protein kinase PLK1Homo sapiens (human)
spindle poleSerine/threonine-protein kinase PLK1Homo sapiens (human)
nucleusSerine/threonine-protein kinase PLK1Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase PLK1Homo sapiens (human)
centrosomeSerine/threonine-protein kinase PLK1Homo sapiens (human)
centrioleSerine/threonine-protein kinase PLK1Homo sapiens (human)
spindleSerine/threonine-protein kinase PLK1Homo sapiens (human)
cytosolSerine/threonine-protein kinase PLK1Homo sapiens (human)
microtubule cytoskeletonSerine/threonine-protein kinase PLK1Homo sapiens (human)
midbodySerine/threonine-protein kinase PLK1Homo sapiens (human)
centriolar satelliteSerine/threonine-protein kinase PLK1Homo sapiens (human)
spindle midzoneSerine/threonine-protein kinase PLK1Homo sapiens (human)
mitotic spindle poleSerine/threonine-protein kinase PLK1Homo sapiens (human)
chromatinSerine/threonine-protein kinase PLK1Homo sapiens (human)
outer kinetochoreSerine/threonine-protein kinase PLK1Homo sapiens (human)
nucleusSerine/threonine-protein kinase PLK1Homo sapiens (human)
centrosomeSerine/threonine-protein kinase PLK1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase PLK1Homo sapiens (human)
spindle poleSerine/threonine-protein kinase PLK1Homo sapiens (human)
kinetochoreSerine/threonine-protein kinase PLK1Homo sapiens (human)
postsynapseLIM domain kinase 1Homo sapiens (human)
glutamatergic synapseLIM domain kinase 1Homo sapiens (human)
male germ cell nucleusLIM domain kinase 1Homo sapiens (human)
cytoplasmLIM domain kinase 1Homo sapiens (human)
cytosolLIM domain kinase 1Homo sapiens (human)
cytoskeletonLIM domain kinase 1Homo sapiens (human)
focal adhesionLIM domain kinase 1Homo sapiens (human)
membraneLIM domain kinase 1Homo sapiens (human)
nuclear speckLIM domain kinase 1Homo sapiens (human)
lamellipodiumLIM domain kinase 1Homo sapiens (human)
neuron projectionLIM domain kinase 1Homo sapiens (human)
nucleusLIM domain kinase 1Homo sapiens (human)
neuron projectionLIM domain kinase 1Homo sapiens (human)
cytoplasmLIM domain kinase 1Homo sapiens (human)
nucleusLIM domain kinase 2Homo sapiens (human)
cytoplasmLIM domain kinase 2Homo sapiens (human)
cis-Golgi networkLIM domain kinase 2Homo sapiens (human)
centrosomeLIM domain kinase 2Homo sapiens (human)
perinuclear region of cytoplasmLIM domain kinase 2Homo sapiens (human)
mitotic spindleLIM domain kinase 2Homo sapiens (human)
nucleusLIM domain kinase 2Homo sapiens (human)
cytoplasmLIM domain kinase 2Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 10Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 10Homo sapiens (human)
mitochondrionMitogen-activated protein kinase 10Homo sapiens (human)
cytosolMitogen-activated protein kinase 10Homo sapiens (human)
plasma membraneMitogen-activated protein kinase 10Homo sapiens (human)
nucleusMitogen-activated protein kinase 10Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 10Homo sapiens (human)
nucleusTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
cytoplasmTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
cytosolTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
extracellular spaceTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
cytosolTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
nuclear bodyTyrosine--tRNA ligase, cytoplasmicHomo sapiens (human)
nucleus5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
nucleoplasm5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
cytosol5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
membrane5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
nucleotide-activated protein kinase complex5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
nucleus5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
cytoplasm5'-AMP-activated protein kinase subunit gamma-1Homo sapiens (human)
extracellular regionEphrin type-B receptor 3Homo sapiens (human)
cytosolEphrin type-B receptor 3Homo sapiens (human)
plasma membraneEphrin type-B receptor 3Homo sapiens (human)
dendriteEphrin type-B receptor 3Homo sapiens (human)
plasma membraneEphrin type-B receptor 3Homo sapiens (human)
rough endoplasmic reticulumEphrin type-A receptor 5Homo sapiens (human)
plasma membraneEphrin type-A receptor 5Homo sapiens (human)
external side of plasma membraneEphrin type-A receptor 5Homo sapiens (human)
axonEphrin type-A receptor 5Homo sapiens (human)
dendriteEphrin type-A receptor 5Homo sapiens (human)
neuronal cell bodyEphrin type-A receptor 5Homo sapiens (human)
perinuclear region of cytoplasmEphrin type-A receptor 5Homo sapiens (human)
plasma membraneEphrin type-A receptor 5Homo sapiens (human)
dendriteEphrin type-A receptor 5Homo sapiens (human)
extracellular regionEphrin type-B receptor 4Homo sapiens (human)
cytosolEphrin type-B receptor 4Homo sapiens (human)
plasma membraneEphrin type-B receptor 4Homo sapiens (human)
extracellular exosomeEphrin type-B receptor 4Homo sapiens (human)
receptor complexEphrin type-B receptor 4Homo sapiens (human)
plasma membraneEphrin type-B receptor 4Homo sapiens (human)
cytoplasmEphrin type-A receptor 4Homo sapiens (human)
mitochondrial outer membraneEphrin type-A receptor 4Homo sapiens (human)
plasma membraneEphrin type-A receptor 4Homo sapiens (human)
adherens junctionEphrin type-A receptor 4Homo sapiens (human)
cell surfaceEphrin type-A receptor 4Homo sapiens (human)
filopodiumEphrin type-A receptor 4Homo sapiens (human)
axonEphrin type-A receptor 4Homo sapiens (human)
dendriteEphrin type-A receptor 4Homo sapiens (human)
neuromuscular junctionEphrin type-A receptor 4Homo sapiens (human)
early endosome membraneEphrin type-A receptor 4Homo sapiens (human)
presynaptic membraneEphrin type-A receptor 4Homo sapiens (human)
dendritic spineEphrin type-A receptor 4Homo sapiens (human)
dendritic shaftEphrin type-A receptor 4Homo sapiens (human)
perikaryonEphrin type-A receptor 4Homo sapiens (human)
axon terminusEphrin type-A receptor 4Homo sapiens (human)
axonal growth coneEphrin type-A receptor 4Homo sapiens (human)
Schaffer collateral - CA1 synapseEphrin type-A receptor 4Homo sapiens (human)
postsynaptic density membraneEphrin type-A receptor 4Homo sapiens (human)
glutamatergic synapseEphrin type-A receptor 4Homo sapiens (human)
plasma membraneEphrin type-A receptor 4Homo sapiens (human)
dendriteEphrin type-A receptor 4Homo sapiens (human)
mitochondrial intermembrane spaceAdenylate kinase 2, mitochondrialHomo sapiens (human)
extracellular exosomeAdenylate kinase 2, mitochondrialHomo sapiens (human)
sperm mitochondrial sheathAdenylate kinase 2, mitochondrialHomo sapiens (human)
cytoplasmAdenylate kinase 2, mitochondrialHomo sapiens (human)
mitochondrionAdenylate kinase 2, mitochondrialHomo sapiens (human)
nucleoplasmAdenosine kinaseHomo sapiens (human)
cytosolAdenosine kinaseHomo sapiens (human)
plasma membraneAdenosine kinaseHomo sapiens (human)
nucleusAdenosine kinaseHomo sapiens (human)
cytosolAdenosine kinaseHomo sapiens (human)
exocystRas-related protein Rab-10Homo sapiens (human)
plasma membraneRas-related protein Rab-10Homo sapiens (human)
Golgi membraneRas-related protein Rab-10Homo sapiens (human)
endosomeRas-related protein Rab-10Homo sapiens (human)
endoplasmic reticulum membraneRas-related protein Rab-10Homo sapiens (human)
Golgi apparatusRas-related protein Rab-10Homo sapiens (human)
trans-Golgi networkRas-related protein Rab-10Homo sapiens (human)
cytosolRas-related protein Rab-10Homo sapiens (human)
cytoskeletonRas-related protein Rab-10Homo sapiens (human)
plasma membraneRas-related protein Rab-10Homo sapiens (human)
adherens junctionRas-related protein Rab-10Homo sapiens (human)
focal adhesionRas-related protein Rab-10Homo sapiens (human)
ciliumRas-related protein Rab-10Homo sapiens (human)
endosome membraneRas-related protein Rab-10Homo sapiens (human)
cytoplasmic vesicle membraneRas-related protein Rab-10Homo sapiens (human)
secretory granule membraneRas-related protein Rab-10Homo sapiens (human)
phagocytic vesicle membraneRas-related protein Rab-10Homo sapiens (human)
insulin-responsive compartmentRas-related protein Rab-10Homo sapiens (human)
perinuclear region of cytoplasmRas-related protein Rab-10Homo sapiens (human)
recycling endosomeRas-related protein Rab-10Homo sapiens (human)
recycling endosome membraneRas-related protein Rab-10Homo sapiens (human)
extracellular exosomeRas-related protein Rab-10Homo sapiens (human)
exocytic vesicleRas-related protein Rab-10Homo sapiens (human)
endoplasmic reticulum tubular networkRas-related protein Rab-10Homo sapiens (human)
recycling endosomeRas-related protein Rab-10Homo sapiens (human)
secretory vesicleRas-related protein Rab-10Homo sapiens (human)
membraneRas-related protein Rab-10Homo sapiens (human)
Golgi apparatusRas-related protein Rab-10Homo sapiens (human)
nucleusActin-related protein 3Homo sapiens (human)
cytoplasmActin-related protein 3Homo sapiens (human)
cytosolActin-related protein 3Homo sapiens (human)
brush borderActin-related protein 3Homo sapiens (human)
cell-cell junctionActin-related protein 3Homo sapiens (human)
focal adhesionActin-related protein 3Homo sapiens (human)
actin cytoskeletonActin-related protein 3Homo sapiens (human)
membraneActin-related protein 3Homo sapiens (human)
lamellipodiumActin-related protein 3Homo sapiens (human)
site of double-strand breakActin-related protein 3Homo sapiens (human)
extracellular exosomeActin-related protein 3Homo sapiens (human)
Arp2/3 protein complexActin-related protein 3Homo sapiens (human)
extracellular regionActin-related protein 2Homo sapiens (human)
nucleusActin-related protein 2Homo sapiens (human)
cytoplasmActin-related protein 2Homo sapiens (human)
cytosolActin-related protein 2Homo sapiens (human)
focal adhesionActin-related protein 2Homo sapiens (human)
actin cytoskeletonActin-related protein 2Homo sapiens (human)
membraneActin-related protein 2Homo sapiens (human)
actin capActin-related protein 2Homo sapiens (human)
azurophil granule lumenActin-related protein 2Homo sapiens (human)
site of double-strand breakActin-related protein 2Homo sapiens (human)
cell projectionActin-related protein 2Homo sapiens (human)
extracellular exosomeActin-related protein 2Homo sapiens (human)
ficolin-1-rich granule lumenActin-related protein 2Homo sapiens (human)
Arp2/3 protein complexActin-related protein 2Homo sapiens (human)
cell cortexActin-related protein 2Homo sapiens (human)
Flemming bodyGTP-binding nuclear protein RanHomo sapiens (human)
male germ cell nucleusGTP-binding nuclear protein RanHomo sapiens (human)
manchetteGTP-binding nuclear protein RanHomo sapiens (human)
nucleusGTP-binding nuclear protein RanHomo sapiens (human)
nuclear envelopeGTP-binding nuclear protein RanHomo sapiens (human)
nucleoplasmGTP-binding nuclear protein RanHomo sapiens (human)
nucleolusGTP-binding nuclear protein RanHomo sapiens (human)
cytoplasmGTP-binding nuclear protein RanHomo sapiens (human)
centrioleGTP-binding nuclear protein RanHomo sapiens (human)
cytosolGTP-binding nuclear protein RanHomo sapiens (human)
membraneGTP-binding nuclear protein RanHomo sapiens (human)
midbodyGTP-binding nuclear protein RanHomo sapiens (human)
sperm flagellumGTP-binding nuclear protein RanHomo sapiens (human)
melanosomeGTP-binding nuclear protein RanHomo sapiens (human)
recycling endosomeGTP-binding nuclear protein RanHomo sapiens (human)
extracellular exosomeGTP-binding nuclear protein RanHomo sapiens (human)
chromatinGTP-binding nuclear protein RanHomo sapiens (human)
nuclear poreGTP-binding nuclear protein RanHomo sapiens (human)
protein-containing complexGTP-binding nuclear protein RanHomo sapiens (human)
RNA nuclear export complexGTP-binding nuclear protein RanHomo sapiens (human)
nucleusGTP-binding nuclear protein RanHomo sapiens (human)
cytoplasmGTP-binding nuclear protein RanHomo sapiens (human)
cytosolCasein kinase I isoform gamma-2Homo sapiens (human)
cell cortexCasein kinase I isoform gamma-2Homo sapiens (human)
membraneCasein kinase I isoform gamma-2Homo sapiens (human)
cytoplasmCasein kinase I isoform gamma-2Homo sapiens (human)
plasma membraneCasein kinase I isoform gamma-2Homo sapiens (human)
nucleusCasein kinase I isoform gamma-2Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 3Homo sapiens (human)
nucleusCyclin-dependent kinase 3Homo sapiens (human)
cytoplasmCyclin-dependent kinase 3Homo sapiens (human)
ruffleCyclin-dependent kinase 6Homo sapiens (human)
nucleusCyclin-dependent kinase 6Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 6Homo sapiens (human)
cytoplasmCyclin-dependent kinase 6Homo sapiens (human)
centrosomeCyclin-dependent kinase 6Homo sapiens (human)
cytosolCyclin-dependent kinase 6Homo sapiens (human)
cyclin D1-CDK6 complexCyclin-dependent kinase 6Homo sapiens (human)
cyclin D3-CDK6 complexCyclin-dependent kinase 6Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 6Homo sapiens (human)
cyclin D2-CDK6 complexCyclin-dependent kinase 6Homo sapiens (human)
cytoplasmCyclin-dependent kinase 6Homo sapiens (human)
nucleusCyclin-dependent kinase 6Homo sapiens (human)
microtubuleCyclin-dependent-like kinase 5 Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent-like kinase 5 Homo sapiens (human)
nucleusCyclin-dependent-like kinase 5 Homo sapiens (human)
nucleoplasmCyclin-dependent-like kinase 5 Homo sapiens (human)
cytoplasmCyclin-dependent-like kinase 5 Homo sapiens (human)
cytosolCyclin-dependent-like kinase 5 Homo sapiens (human)
plasma membraneCyclin-dependent-like kinase 5 Homo sapiens (human)
postsynaptic densityCyclin-dependent-like kinase 5 Homo sapiens (human)
membraneCyclin-dependent-like kinase 5 Homo sapiens (human)
protein kinase 5 complexCyclin-dependent-like kinase 5 Homo sapiens (human)
lamellipodiumCyclin-dependent-like kinase 5 Homo sapiens (human)
cell junctionCyclin-dependent-like kinase 5 Homo sapiens (human)
filopodiumCyclin-dependent-like kinase 5 Homo sapiens (human)
axonCyclin-dependent-like kinase 5 Homo sapiens (human)
dendriteCyclin-dependent-like kinase 5 Homo sapiens (human)
growth coneCyclin-dependent-like kinase 5 Homo sapiens (human)
neuromuscular junctionCyclin-dependent-like kinase 5 Homo sapiens (human)
neuron projectionCyclin-dependent-like kinase 5 Homo sapiens (human)
neuronal cell bodyCyclin-dependent-like kinase 5 Homo sapiens (human)
perikaryonCyclin-dependent-like kinase 5 Homo sapiens (human)
presynapseCyclin-dependent-like kinase 5 Homo sapiens (human)
nucleusCyclin-dependent-like kinase 5 Homo sapiens (human)
cytoplasmCyclin-dependent-like kinase 5 Homo sapiens (human)
synaptic vesicleCyclin-dependent kinase 16Homo sapiens (human)
cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 16Homo sapiens (human)
cytoplasmCyclin-dependent kinase 16Homo sapiens (human)
cytosolCyclin-dependent kinase 16Homo sapiens (human)
plasma membraneCyclin-dependent kinase 16Homo sapiens (human)
cytoplasmic side of plasma membraneCyclin-dependent kinase 16Homo sapiens (human)
microtubule cytoskeletonCyclin-dependent kinase 16Homo sapiens (human)
neuron projectionCyclin-dependent kinase 16Homo sapiens (human)
cytoplasmCyclin-dependent kinase 16Homo sapiens (human)
nucleusCyclin-dependent kinase 16Homo sapiens (human)
cytoplasmCyclin-dependent kinase 17Homo sapiens (human)
nucleusCyclin-dependent kinase 17Homo sapiens (human)
nucleusATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
cytoplasmATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
cytosolATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
membraneATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
extracellular exosomeATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
membraneATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
6-phosphofructokinase complexATP-dependent 6-phosphofructokinase, platelet typeHomo sapiens (human)
nucleusDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
mitochondrionDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
early endosomeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
late endosomeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
endoplasmic reticulumDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
Golgi apparatusDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
centrosomeDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
cytosolDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
plasma membraneDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
focal adhesionDual specificity mitogen-activated protein kinase kinase 1Homo sapiens (human)
nucleolusDNA topoisomerase 2-betaHomo sapiens (human)
heterochromatinDNA topoisomerase 2-betaHomo sapiens (human)
nucleusDNA topoisomerase 2-betaHomo sapiens (human)
nucleoplasmDNA topoisomerase 2-betaHomo sapiens (human)
nucleolusDNA topoisomerase 2-betaHomo sapiens (human)
cytosolDNA topoisomerase 2-betaHomo sapiens (human)
ribonucleoprotein complexDNA topoisomerase 2-betaHomo sapiens (human)
nucleusDNA topoisomerase 2-betaHomo sapiens (human)
immunological synapseProtein kinase C theta typeHomo sapiens (human)
cytosolProtein kinase C theta typeHomo sapiens (human)
plasma membraneProtein kinase C theta typeHomo sapiens (human)
aggresomeProtein kinase C theta typeHomo sapiens (human)
centriolar satelliteProtein kinase C theta typeHomo sapiens (human)
plasma membraneActivin receptor type-1Homo sapiens (human)
apical part of cellActivin receptor type-1Homo sapiens (human)
activin receptor complexActivin receptor type-1Homo sapiens (human)
BMP receptor complexActivin receptor type-1Homo sapiens (human)
plasma membraneActivin receptor type-1Homo sapiens (human)
stress fiberMacrophage-stimulating protein receptorHomo sapiens (human)
vacuoleMacrophage-stimulating protein receptorHomo sapiens (human)
plasma membraneMacrophage-stimulating protein receptorHomo sapiens (human)
cell surfaceMacrophage-stimulating protein receptorHomo sapiens (human)
receptor complexMacrophage-stimulating protein receptorHomo sapiens (human)
plasma membraneMacrophage-stimulating protein receptorHomo sapiens (human)
stress fiberFocal adhesion kinase 1Homo sapiens (human)
nucleusFocal adhesion kinase 1Homo sapiens (human)
cytoplasmFocal adhesion kinase 1Homo sapiens (human)
centrosomeFocal adhesion kinase 1Homo sapiens (human)
cytosolFocal adhesion kinase 1Homo sapiens (human)
cytoskeletonFocal adhesion kinase 1Homo sapiens (human)
plasma membraneFocal adhesion kinase 1Homo sapiens (human)
focal adhesionFocal adhesion kinase 1Homo sapiens (human)
cell cortexFocal adhesion kinase 1Homo sapiens (human)
ciliary basal bodyFocal adhesion kinase 1Homo sapiens (human)
intracellular membrane-bounded organelleFocal adhesion kinase 1Homo sapiens (human)
perinuclear region of cytoplasmFocal adhesion kinase 1Homo sapiens (human)
plasma membraneFocal adhesion kinase 1Homo sapiens (human)
focal adhesionFocal adhesion kinase 1Homo sapiens (human)
dendritic spineFocal adhesion kinase 1Homo sapiens (human)
extracellular regionProtein kinase C delta typeHomo sapiens (human)
nucleusProtein kinase C delta typeHomo sapiens (human)
nucleoplasmProtein kinase C delta typeHomo sapiens (human)
cytoplasmProtein kinase C delta typeHomo sapiens (human)
mitochondrionProtein kinase C delta typeHomo sapiens (human)
endoplasmic reticulumProtein kinase C delta typeHomo sapiens (human)
cytosolProtein kinase C delta typeHomo sapiens (human)
plasma membraneProtein kinase C delta typeHomo sapiens (human)
cell-cell junctionProtein kinase C delta typeHomo sapiens (human)
nuclear matrixProtein kinase C delta typeHomo sapiens (human)
azurophil granule lumenProtein kinase C delta typeHomo sapiens (human)
endolysosomeProtein kinase C delta typeHomo sapiens (human)
perinuclear region of cytoplasmProtein kinase C delta typeHomo sapiens (human)
extracellular exosomeProtein kinase C delta typeHomo sapiens (human)
nucleusTyrosine-protein kinase BTKHomo sapiens (human)
cytoplasmTyrosine-protein kinase BTKHomo sapiens (human)
cytosolTyrosine-protein kinase BTKHomo sapiens (human)
plasma membraneTyrosine-protein kinase BTKHomo sapiens (human)
cytoplasmic vesicleTyrosine-protein kinase BTKHomo sapiens (human)
membrane raftTyrosine-protein kinase BTKHomo sapiens (human)
perinuclear region of cytoplasmTyrosine-protein kinase BTKHomo sapiens (human)
plasma membraneTyrosine-protein kinase BTKHomo sapiens (human)
nucleusActivated CDC42 kinase 1Homo sapiens (human)
cytoplasmActivated CDC42 kinase 1Homo sapiens (human)
endosomeActivated CDC42 kinase 1Homo sapiens (human)
cytosolActivated CDC42 kinase 1Homo sapiens (human)
plasma membraneActivated CDC42 kinase 1Homo sapiens (human)
clathrin-coated pitActivated CDC42 kinase 1Homo sapiens (human)
adherens junctionActivated CDC42 kinase 1Homo sapiens (human)
membraneActivated CDC42 kinase 1Homo sapiens (human)
clathrin-coated vesicleActivated CDC42 kinase 1Homo sapiens (human)
cytoplasmic vesicle membraneActivated CDC42 kinase 1Homo sapiens (human)
intracellular membrane-bounded organelleActivated CDC42 kinase 1Homo sapiens (human)
perinuclear region of cytoplasmActivated CDC42 kinase 1Homo sapiens (human)
cytoophidiumActivated CDC42 kinase 1Homo sapiens (human)
Grb2-EGFR complexActivated CDC42 kinase 1Homo sapiens (human)
plasma membraneActivated CDC42 kinase 1Homo sapiens (human)
extracellular spaceEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
plasma membraneEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
extracellular exosomeEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
receptor complexEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
plasma membraneEpithelial discoidin domain-containing receptor 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
Golgi membraneMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
basolateral plasma membraneMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 4Homo sapiens (human)
nucleusSerine/threonine-protein kinase 4Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase 4Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 4Homo sapiens (human)
cytosolSerine/threonine-protein kinase 4Homo sapiens (human)
nuclear bodySerine/threonine-protein kinase 4Homo sapiens (human)
protein-containing complexSerine/threonine-protein kinase 4Homo sapiens (human)
cytoplasm5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
nucleus5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
nucleoplasm5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cytoplasm5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
cytosol5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
apical plasma membrane5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
nuclear speck5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
axon5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
dendrite5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
nucleotide-activated protein kinase complex5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
neuronal cell body5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
chromatin5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
nucleus5'-AMP-activated protein kinase catalytic subunit alpha-1Homo sapiens (human)
spindleDual specificity mitogen-activated protein kinase kinase 5Homo sapiens (human)
nucleusMitogen-activated protein kinase 7Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 7Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 7Homo sapiens (human)
cytosolMitogen-activated protein kinase 7Homo sapiens (human)
PML bodyMitogen-activated protein kinase 7Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 7Homo sapiens (human)
nucleusMitogen-activated protein kinase 7Homo sapiens (human)
nucleusSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cytosolSerine/threonine-protein kinase PAK 2Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cell-cell junctionSerine/threonine-protein kinase PAK 2Homo sapiens (human)
postsynaptic densitySerine/threonine-protein kinase PAK 2Homo sapiens (human)
secretory granuleSerine/threonine-protein kinase PAK 2Homo sapiens (human)
perinuclear region of cytoplasmSerine/threonine-protein kinase PAK 2Homo sapiens (human)
glutamatergic synapseSerine/threonine-protein kinase PAK 2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 3Homo sapiens (human)
centrosomeSerine/threonine-protein kinase 3Homo sapiens (human)
nucleusSerine/threonine-protein kinase 3Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 3Homo sapiens (human)
cytosolSerine/threonine-protein kinase 3Homo sapiens (human)
protein-containing complexSerine/threonine-protein kinase 3Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase 1Homo sapiens (human)
cytosolIntegrin-linked protein kinaseHomo sapiens (human)
plasma membraneIntegrin-linked protein kinaseHomo sapiens (human)
focal adhesionIntegrin-linked protein kinaseHomo sapiens (human)
membraneIntegrin-linked protein kinaseHomo sapiens (human)
sarcomereIntegrin-linked protein kinaseHomo sapiens (human)
lamellipodiumIntegrin-linked protein kinaseHomo sapiens (human)
focal adhesionIntegrin-linked protein kinaseHomo sapiens (human)
stress fiberIntegrin-linked protein kinaseHomo sapiens (human)
Golgi membraneRho-associated protein kinase 1Homo sapiens (human)
ruffleRho-associated protein kinase 1Homo sapiens (human)
extracellular regionRho-associated protein kinase 1Homo sapiens (human)
centrioleRho-associated protein kinase 1Homo sapiens (human)
cytosolRho-associated protein kinase 1Homo sapiens (human)
cytoskeletonRho-associated protein kinase 1Homo sapiens (human)
plasma membraneRho-associated protein kinase 1Homo sapiens (human)
cytoplasmic stress granuleRho-associated protein kinase 1Homo sapiens (human)
lamellipodiumRho-associated protein kinase 1Homo sapiens (human)
blebRho-associated protein kinase 1Homo sapiens (human)
secretory granule lumenRho-associated protein kinase 1Homo sapiens (human)
Schaffer collateral - CA1 synapseRho-associated protein kinase 1Homo sapiens (human)
cytoskeletonRho-associated protein kinase 1Homo sapiens (human)
cytoplasmRho-associated protein kinase 1Homo sapiens (human)
cytoplasmic stress granuleRho-associated protein kinase 1Homo sapiens (human)
cytoplasmNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
membraneNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
plasma membraneNon-receptor tyrosine-protein kinase TNK1Homo sapiens (human)
nucleoplasmCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
cytosolCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
membraneCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
endocytic vesicle membraneCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
sarcoplasmic reticulum membraneCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
calcium- and calmodulin-dependent protein kinase complexCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
neuron projectionCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
cytoplasmCalcium/calmodulin-dependent protein kinase type II subunit gammaHomo sapiens (human)
nucleusCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
nucleoplasmCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
cytoplasmCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
cytosolCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
membraneCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
endocytic vesicle membraneCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
sarcoplasmic reticulum membraneCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
sarcolemmaCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
calcium- and calmodulin-dependent protein kinase complexCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
cytoplasmCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
neuron projectionCalcium/calmodulin-dependent protein kinase type II subunit deltaHomo sapiens (human)
cytoskeletonDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
nucleusDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
nucleusDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
nucleoplasmDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
cytoplasmDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
nuclear speckDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
axonDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
dendriteDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
ribonucleoprotein complexDual specificity tyrosine-phosphorylation-regulated kinase 1AHomo sapiens (human)
cytoplasmActivin receptor type-2BHomo sapiens (human)
plasma membraneActivin receptor type-2BHomo sapiens (human)
protein-containing complexActivin receptor type-2BHomo sapiens (human)
receptor complexActivin receptor type-2BHomo sapiens (human)
activin receptor complexActivin receptor type-2BHomo sapiens (human)
plasma membraneActivin receptor type-2BHomo sapiens (human)
caveolaBone morphogenetic protein receptor type-2Homo sapiens (human)
extracellular spaceBone morphogenetic protein receptor type-2Homo sapiens (human)
nucleoplasmBone morphogenetic protein receptor type-2Homo sapiens (human)
plasma membraneBone morphogenetic protein receptor type-2Homo sapiens (human)
clathrin-coated pitBone morphogenetic protein receptor type-2Homo sapiens (human)
adherens junctionBone morphogenetic protein receptor type-2Homo sapiens (human)
basal plasma membraneBone morphogenetic protein receptor type-2Homo sapiens (human)
cell surfaceBone morphogenetic protein receptor type-2Homo sapiens (human)
postsynaptic densityBone morphogenetic protein receptor type-2Homo sapiens (human)
apical plasma membraneBone morphogenetic protein receptor type-2Homo sapiens (human)
axonBone morphogenetic protein receptor type-2Homo sapiens (human)
dendriteBone morphogenetic protein receptor type-2Homo sapiens (human)
neuronal cell bodyBone morphogenetic protein receptor type-2Homo sapiens (human)
plasma membraneBone morphogenetic protein receptor type-2Homo sapiens (human)
receptor complexBone morphogenetic protein receptor type-2Homo sapiens (human)
ruffleProtein-tyrosine kinase 6Homo sapiens (human)
nucleusProtein-tyrosine kinase 6Homo sapiens (human)
nucleoplasmProtein-tyrosine kinase 6Homo sapiens (human)
cytoplasmProtein-tyrosine kinase 6Homo sapiens (human)
cytosolProtein-tyrosine kinase 6Homo sapiens (human)
plasma membraneProtein-tyrosine kinase 6Homo sapiens (human)
nuclear bodyProtein-tyrosine kinase 6Homo sapiens (human)
plasma membraneProtein-tyrosine kinase 6Homo sapiens (human)
acrosomal vesiclecGMP-dependent protein kinase 1 Homo sapiens (human)
nucleoplasmcGMP-dependent protein kinase 1 Homo sapiens (human)
cytoplasmcGMP-dependent protein kinase 1 Homo sapiens (human)
Golgi apparatuscGMP-dependent protein kinase 1 Homo sapiens (human)
cytosolcGMP-dependent protein kinase 1 Homo sapiens (human)
plasma membranecGMP-dependent protein kinase 1 Homo sapiens (human)
sarcolemmacGMP-dependent protein kinase 1 Homo sapiens (human)
cyclin K-CDK13 complexCyclin-dependent kinase 13Homo sapiens (human)
extracellular regionCyclin-dependent kinase 13Homo sapiens (human)
extracellular spaceCyclin-dependent kinase 13Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 13Homo sapiens (human)
Golgi apparatusCyclin-dependent kinase 13Homo sapiens (human)
cytosolCyclin-dependent kinase 13Homo sapiens (human)
nuclear speckCyclin-dependent kinase 13Homo sapiens (human)
ficolin-1-rich granule lumenCyclin-dependent kinase 13Homo sapiens (human)
nuclear cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 13Homo sapiens (human)
nucleusCyclin-dependent kinase 13Homo sapiens (human)
cyclin/CDK positive transcription elongation factor complexCyclin-dependent kinase 13Homo sapiens (human)
cytoplasmInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
nucleusInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
nucleoplasmInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
cytoplasmInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
cytosolInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
PML bodyInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
mitochondrial membraneInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
serine/threonine protein kinase complexInhibitor of nuclear factor kappa-B kinase subunit epsilonHomo sapiens (human)
NMDA selective glutamate receptor complexProtein-tyrosine kinase 2-betaHomo sapiens (human)
nucleusProtein-tyrosine kinase 2-betaHomo sapiens (human)
cytoplasmProtein-tyrosine kinase 2-betaHomo sapiens (human)
cytosolProtein-tyrosine kinase 2-betaHomo sapiens (human)
cytoskeletonProtein-tyrosine kinase 2-betaHomo sapiens (human)
focal adhesionProtein-tyrosine kinase 2-betaHomo sapiens (human)
cell cortexProtein-tyrosine kinase 2-betaHomo sapiens (human)
postsynaptic densityProtein-tyrosine kinase 2-betaHomo sapiens (human)
lamellipodiumProtein-tyrosine kinase 2-betaHomo sapiens (human)
dendriteProtein-tyrosine kinase 2-betaHomo sapiens (human)
growth coneProtein-tyrosine kinase 2-betaHomo sapiens (human)
neuronal cell bodyProtein-tyrosine kinase 2-betaHomo sapiens (human)
cell bodyProtein-tyrosine kinase 2-betaHomo sapiens (human)
perinuclear region of cytoplasmProtein-tyrosine kinase 2-betaHomo sapiens (human)
apical dendriteProtein-tyrosine kinase 2-betaHomo sapiens (human)
Schaffer collateral - CA1 synapseProtein-tyrosine kinase 2-betaHomo sapiens (human)
presynapseProtein-tyrosine kinase 2-betaHomo sapiens (human)
glutamatergic synapseProtein-tyrosine kinase 2-betaHomo sapiens (human)
postsynaptic density, intracellular componentProtein-tyrosine kinase 2-betaHomo sapiens (human)
dendritic spineProtein-tyrosine kinase 2-betaHomo sapiens (human)
focal adhesionProtein-tyrosine kinase 2-betaHomo sapiens (human)
plasma membraneProtein-tyrosine kinase 2-betaHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleolusSodium channel protein type 5 subunit alphaHomo sapiens (human)
endoplasmic reticulumSodium channel protein type 5 subunit alphaHomo sapiens (human)
plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
cell surfaceSodium channel protein type 5 subunit alphaHomo sapiens (human)
intercalated discSodium channel protein type 5 subunit alphaHomo sapiens (human)
membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
lateral plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
Z discSodium channel protein type 5 subunit alphaHomo sapiens (human)
T-tubuleSodium channel protein type 5 subunit alphaHomo sapiens (human)
sarcolemmaSodium channel protein type 5 subunit alphaHomo sapiens (human)
perinuclear region of cytoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel complexSodium channel protein type 5 subunit alphaHomo sapiens (human)
plasma membraneMaternal embryonic leucine zipper kinaseHomo sapiens (human)
cell cortexMaternal embryonic leucine zipper kinaseHomo sapiens (human)
membraneMaternal embryonic leucine zipper kinaseHomo sapiens (human)
cytoplasmMaternal embryonic leucine zipper kinaseHomo sapiens (human)
chromosome, centromeric regionStructural maintenance of chromosomes protein 1AHomo sapiens (human)
kinetochoreStructural maintenance of chromosomes protein 1AHomo sapiens (human)
condensed nuclear chromosomeStructural maintenance of chromosomes protein 1AHomo sapiens (human)
nucleusStructural maintenance of chromosomes protein 1AHomo sapiens (human)
nucleoplasmStructural maintenance of chromosomes protein 1AHomo sapiens (human)
chromosomeStructural maintenance of chromosomes protein 1AHomo sapiens (human)
cytosolStructural maintenance of chromosomes protein 1AHomo sapiens (human)
nuclear matrixStructural maintenance of chromosomes protein 1AHomo sapiens (human)
mitotic cohesin complexStructural maintenance of chromosomes protein 1AHomo sapiens (human)
meiotic cohesin complexStructural maintenance of chromosomes protein 1AHomo sapiens (human)
mitotic spindle poleStructural maintenance of chromosomes protein 1AHomo sapiens (human)
cohesin complexStructural maintenance of chromosomes protein 1AHomo sapiens (human)
nucleusStructural maintenance of chromosomes protein 1AHomo sapiens (human)
chromosome, telomeric regionChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
nucleusChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
nucleoplasmChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
cytoplasmChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
centrosomeChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
membraneChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
NuRD complexChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
site of DNA damageChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
cerebellar granule cell to Purkinje cell synapseChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
chromatinChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
protein-containing complexChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
RNA polymerase II transcription regulator complexChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
nucleusChromodomain-helicase-DNA-binding protein 4Homo sapiens (human)
peroxisomePeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
peroxisomePeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
peroxisomal membranePeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
peroxisomal matrixPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
cytosolPeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
membranePeroxisomal acyl-coenzyme A oxidase 1Homo sapiens (human)
plasma membraneEphrin type-A receptor 7Homo sapiens (human)
glutamatergic synapseEphrin type-A receptor 7Homo sapiens (human)
plasma membraneEphrin type-A receptor 7Homo sapiens (human)
dendriteEphrin type-A receptor 7Homo sapiens (human)
Golgi membraneDelta(24)-sterol reductaseHomo sapiens (human)
nucleusDelta(24)-sterol reductaseHomo sapiens (human)
endoplasmic reticulumDelta(24)-sterol reductaseHomo sapiens (human)
endoplasmic reticulum membraneDelta(24)-sterol reductaseHomo sapiens (human)
membraneDelta(24)-sterol reductaseHomo sapiens (human)
cytoplasmDelta(24)-sterol reductaseHomo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-1Homo sapiens (human)
cytosolRibosomal protein S6 kinase alpha-1Homo sapiens (human)
synapseRibosomal protein S6 kinase alpha-1Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-1Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase alpha-1Homo sapiens (human)
cytoplasmic vesicleDual specificity testis-specific protein kinase 1Homo sapiens (human)
cytoplasmDual specificity testis-specific protein kinase 1Homo sapiens (human)
centrosomeDual specificity testis-specific protein kinase 1Homo sapiens (human)
cytosolDual specificity testis-specific protein kinase 1Homo sapiens (human)
lamellipodiumDual specificity testis-specific protein kinase 1Homo sapiens (human)
perinuclear region of cytoplasmDual specificity testis-specific protein kinase 1Homo sapiens (human)
cytoplasmDual specificity testis-specific protein kinase 1Homo sapiens (human)
nucleusDual specificity testis-specific protein kinase 1Homo sapiens (human)
stress fiberMyosin light chain kinase, smooth muscleHomo sapiens (human)
cytoplasmMyosin light chain kinase, smooth muscleHomo sapiens (human)
cytosolMyosin light chain kinase, smooth muscleHomo sapiens (human)
plasma membraneMyosin light chain kinase, smooth muscleHomo sapiens (human)
actin cytoskeletonMyosin light chain kinase, smooth muscleHomo sapiens (human)
lamellipodiumMyosin light chain kinase, smooth muscleHomo sapiens (human)
cleavage furrowMyosin light chain kinase, smooth muscleHomo sapiens (human)
cleavage furrowMyosin light chain kinase, smooth muscleHomo sapiens (human)
stress fiberMyosin light chain kinase, smooth muscleHomo sapiens (human)
lamellipodiumMyosin light chain kinase, smooth muscleHomo sapiens (human)
cytoplasmMyosin light chain kinase, smooth muscleHomo sapiens (human)
nucleoplasmMitogen-activated protein kinase 11Homo sapiens (human)
cytosolMitogen-activated protein kinase 11Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 11Homo sapiens (human)
nucleusMitogen-activated protein kinase 11Homo sapiens (human)
nucleusSerine/threonine-protein kinase STK11Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase STK11Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase STK11Homo sapiens (human)
mitochondrionSerine/threonine-protein kinase STK11Homo sapiens (human)
cytosolSerine/threonine-protein kinase STK11Homo sapiens (human)
membraneSerine/threonine-protein kinase STK11Homo sapiens (human)
Z discSerine/threonine-protein kinase STK11Homo sapiens (human)
extracellular exosomeSerine/threonine-protein kinase STK11Homo sapiens (human)
serine/threonine protein kinase complexSerine/threonine-protein kinase STK11Homo sapiens (human)
intracellular protein-containing complexSerine/threonine-protein kinase STK11Homo sapiens (human)
nucleusSerine/threonine-protein kinase STK11Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase STK11Homo sapiens (human)
nucleusSerine/threonine-protein kinase N1Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase N1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase N1Homo sapiens (human)
endosomeSerine/threonine-protein kinase N1Homo sapiens (human)
cytosolSerine/threonine-protein kinase N1Homo sapiens (human)
midbodySerine/threonine-protein kinase N1Homo sapiens (human)
cleavage furrowSerine/threonine-protein kinase N1Homo sapiens (human)
protein-containing complexSerine/threonine-protein kinase N1Homo sapiens (human)
nucleusSerine/threonine-protein kinase N2Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase N2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase N2Homo sapiens (human)
centrosomeSerine/threonine-protein kinase N2Homo sapiens (human)
cytosolSerine/threonine-protein kinase N2Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase N2Homo sapiens (human)
nuclear bodySerine/threonine-protein kinase N2Homo sapiens (human)
lamellipodiumSerine/threonine-protein kinase N2Homo sapiens (human)
midbodySerine/threonine-protein kinase N2Homo sapiens (human)
cleavage furrowSerine/threonine-protein kinase N2Homo sapiens (human)
apical junction complexSerine/threonine-protein kinase N2Homo sapiens (human)
intermediate filament cytoskeletonSerine/threonine-protein kinase N2Homo sapiens (human)
perinuclear region of cytoplasmSerine/threonine-protein kinase N2Homo sapiens (human)
protein-containing complexSerine/threonine-protein kinase N2Homo sapiens (human)
cytosolMitogen-activated protein kinase 14Homo sapiens (human)
spindle poleMitogen-activated protein kinase 14Homo sapiens (human)
extracellular regionMitogen-activated protein kinase 14Homo sapiens (human)
nucleusMitogen-activated protein kinase 14Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase 14Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 14Homo sapiens (human)
mitochondrionMitogen-activated protein kinase 14Homo sapiens (human)
cytosolMitogen-activated protein kinase 14Homo sapiens (human)
nuclear speckMitogen-activated protein kinase 14Homo sapiens (human)
secretory granule lumenMitogen-activated protein kinase 14Homo sapiens (human)
glutamatergic synapseMitogen-activated protein kinase 14Homo sapiens (human)
ficolin-1-rich granule lumenMitogen-activated protein kinase 14Homo sapiens (human)
nucleusMitogen-activated protein kinase 14Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 14Homo sapiens (human)
fibrillar centerCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
nucleoplasmCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
extracellular exosomeCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
cytoplasmCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
nucleusCalcium/calmodulin-dependent protein kinase type IVHomo sapiens (human)
centrosomeMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
microtubuleMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
membraneMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
centrosomeMitogen-activated protein kinase kinase kinase 11Homo sapiens (human)
plasma membraneDiscoidin domain-containing receptor 2Homo sapiens (human)
focal adhesionDiscoidin domain-containing receptor 2Homo sapiens (human)
actin cytoskeletonDiscoidin domain-containing receptor 2Homo sapiens (human)
apical plasma membraneDiscoidin domain-containing receptor 2Homo sapiens (human)
receptor complexDiscoidin domain-containing receptor 2Homo sapiens (human)
plasma membraneDiscoidin domain-containing receptor 2Homo sapiens (human)
cytosolAP2-associated protein kinase 1Homo sapiens (human)
plasma membraneAP2-associated protein kinase 1Homo sapiens (human)
clathrin-coated pitAP2-associated protein kinase 1Homo sapiens (human)
clathrin-coated vesicleAP2-associated protein kinase 1Homo sapiens (human)
cell leading edgeAP2-associated protein kinase 1Homo sapiens (human)
terminal boutonAP2-associated protein kinase 1Homo sapiens (human)
intracellular membrane-bounded organelleAP2-associated protein kinase 1Homo sapiens (human)
presynapseAP2-associated protein kinase 1Homo sapiens (human)
cytoplasmMyosin light chain kinase 3Homo sapiens (human)
cytosolMyosin light chain kinase 3Homo sapiens (human)
cytoplasmMyosin light chain kinase 3Homo sapiens (human)
actin cytoskeletonMyosin light chain kinase 3Homo sapiens (human)
extracellular exosomePutative heat shock protein HSP 90-beta 2Homo sapiens (human)
perinuclear region of cytoplasmPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
protein-containing complexPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
plasma membranePutative heat shock protein HSP 90-beta 2Homo sapiens (human)
cytosolPutative heat shock protein HSP 90-beta 2Homo sapiens (human)
cytosolSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
cell-cell junctionSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
lamellipodiumSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
cell leading edgeSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
actomyosinSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
extracellular exosomeSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
actomyosinSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
cytoskeletonSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase MRCK alphaHomo sapiens (human)
cytosolSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
cell leading edgeSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
centriolar satelliteSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
cytoskeletonSerine/threonine-protein kinase MRCK gammaHomo sapiens (human)
mitochondrionAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
mitochondrial matrixAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
cytoplasmAcyl-CoA dehydrogenase family member 10Homo sapiens (human)
nucleusSerine/threonine-protein kinase N3Homo sapiens (human)
Golgi apparatusSerine/threonine-protein kinase N3Homo sapiens (human)
cytosolSerine/threonine-protein kinase N3Homo sapiens (human)
perinuclear region of cytoplasmSerine/threonine-protein kinase N3Homo sapiens (human)
phagophore assembly siteSerine/threonine-protein kinase ULK3Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase ULK3Homo sapiens (human)
ciliary tipSerine/threonine-protein kinase ULK3Homo sapiens (human)
phagophore assembly site membraneSerine/threonine-protein kinase ULK3Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase ULK3Homo sapiens (human)
cytosolSerine/threonine-protein kinase ULK3Homo sapiens (human)
autophagosomeSerine/threonine-protein kinase ULK3Homo sapiens (human)
phagophore assembly siteSerine/threonine-protein kinase ULK3Homo sapiens (human)
nucleusAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
mitochondrial inner membraneAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
peroxisomeAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
mitochondrial membraneAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
mitochondrionAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
cytoplasmAcyl-CoA dehydrogenase family member 11Homo sapiens (human)
endoplasmic reticulumSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
cytosolSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
endoplasmic reticulum quality control compartmentSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
IRE1-TRAF2-ASK1 complexSerine/threonine-protein kinase/endoribonuclease IRE2Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase MARK2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase MARK2Homo sapiens (human)
mitochondrionSerine/threonine-protein kinase MARK2Homo sapiens (human)
actin filamentSerine/threonine-protein kinase MARK2Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase MARK2Homo sapiens (human)
membraneSerine/threonine-protein kinase MARK2Homo sapiens (human)
lateral plasma membraneSerine/threonine-protein kinase MARK2Homo sapiens (human)
dendriteSerine/threonine-protein kinase MARK2Homo sapiens (human)
microtubule bundleSerine/threonine-protein kinase MARK2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase MARK2Homo sapiens (human)
cytoplasmATP-dependent RNA helicase DHX30Homo sapiens (human)
mitochondrionATP-dependent RNA helicase DHX30Homo sapiens (human)
cytosolATP-dependent RNA helicase DHX30Homo sapiens (human)
ribonucleoprotein granuleATP-dependent RNA helicase DHX30Homo sapiens (human)
mitochondrial nucleoidATP-dependent RNA helicase DHX30Homo sapiens (human)
cytoplasmATP-dependent RNA helicase DHX30Homo sapiens (human)
nucleusATP-dependent RNA helicase DHX30Homo sapiens (human)
intracellular anatomical structureATP-dependent RNA helicase DHX30Homo sapiens (human)
cytosolSerine/threonine-protein kinase TAO1Homo sapiens (human)
microtubule cytoskeletonSerine/threonine-protein kinase TAO1Homo sapiens (human)
perinuclear region of cytoplasmSerine/threonine-protein kinase TAO1Homo sapiens (human)
extracellular exosomeSerine/threonine-protein kinase TAO1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase TAO1Homo sapiens (human)
nucleusSTE20-related kinase adapter protein alphaHomo sapiens (human)
nucleoplasmSTE20-related kinase adapter protein alphaHomo sapiens (human)
cytoplasmSTE20-related kinase adapter protein alphaHomo sapiens (human)
cytosolSTE20-related kinase adapter protein alphaHomo sapiens (human)
serine/threonine protein kinase complexSTE20-related kinase adapter protein alphaHomo sapiens (human)
intracellular protein-containing complexSTE20-related kinase adapter protein alphaHomo sapiens (human)
stress fiberMyosin-14Homo sapiens (human)
cytosolMyosin-14Homo sapiens (human)
brush borderMyosin-14Homo sapiens (human)
membraneMyosin-14Homo sapiens (human)
growth coneMyosin-14Homo sapiens (human)
actomyosinMyosin-14Homo sapiens (human)
extracellular exosomeMyosin-14Homo sapiens (human)
myosin II filamentMyosin-14Homo sapiens (human)
myosin II complexMyosin-14Homo sapiens (human)
cytoplasmMyosin-14Homo sapiens (human)
myosin filamentMyosin-14Homo sapiens (human)
mitochondrionAarF domain-containing protein kinase 1Homo sapiens (human)
mitochondrial inner membraneAarF domain-containing protein kinase 1Homo sapiens (human)
nucleusATP-dependent RNA helicase DDX42Homo sapiens (human)
nucleoplasmATP-dependent RNA helicase DDX42Homo sapiens (human)
cytoplasmATP-dependent RNA helicase DDX42Homo sapiens (human)
cytosolATP-dependent RNA helicase DDX42Homo sapiens (human)
Cajal bodyATP-dependent RNA helicase DDX42Homo sapiens (human)
membraneATP-dependent RNA helicase DDX42Homo sapiens (human)
nuclear speckATP-dependent RNA helicase DDX42Homo sapiens (human)
U2-type prespliceosomeATP-dependent RNA helicase DDX42Homo sapiens (human)
nucleusATP-dependent RNA helicase DDX42Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 3Homo sapiens (human)
cytosolEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
translation release factor complexEukaryotic peptide chain release factor GTP-binding subunit ERF3BHomo sapiens (human)
lysosomal membraneRegulatory-associated protein of mTORHomo sapiens (human)
nucleoplasmRegulatory-associated protein of mTORHomo sapiens (human)
cytoplasmRegulatory-associated protein of mTORHomo sapiens (human)
lysosomeRegulatory-associated protein of mTORHomo sapiens (human)
lysosomal membraneRegulatory-associated protein of mTORHomo sapiens (human)
cytosolRegulatory-associated protein of mTORHomo sapiens (human)
cytoplasmic stress granuleRegulatory-associated protein of mTORHomo sapiens (human)
dendriteRegulatory-associated protein of mTORHomo sapiens (human)
TORC1 complexRegulatory-associated protein of mTORHomo sapiens (human)
neuronal cell bodyRegulatory-associated protein of mTORHomo sapiens (human)
cytoplasmRegulatory-associated protein of mTORHomo sapiens (human)
mitochondrionAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
membraneAtypical kinase COQ8A, mitochondrialHomo sapiens (human)
nucleoplasmPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
autophagosomePhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
endoplasmic reticulumPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
cytosolPhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
plasma membranePhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
intracellular organellePhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
extracellular exosomePhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
plasma membranePhosphatidylinositol 5-phosphate 4-kinase type-2 gammaHomo sapiens (human)
axonemeMitogen-activated protein kinase 15Homo sapiens (human)
extracellular regionMitogen-activated protein kinase 15Homo sapiens (human)
nucleusMitogen-activated protein kinase 15Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 15Homo sapiens (human)
autophagosomeMitogen-activated protein kinase 15Homo sapiens (human)
Golgi apparatusMitogen-activated protein kinase 15Homo sapiens (human)
centrioleMitogen-activated protein kinase 15Homo sapiens (human)
cell-cell junctionMitogen-activated protein kinase 15Homo sapiens (human)
bicellular tight junctionMitogen-activated protein kinase 15Homo sapiens (human)
cytoplasmic vesicleMitogen-activated protein kinase 15Homo sapiens (human)
ciliary basal bodyMitogen-activated protein kinase 15Homo sapiens (human)
meiotic spindleMitogen-activated protein kinase 15Homo sapiens (human)
cytoplasmMitogen-activated protein kinase 15Homo sapiens (human)
nucleusMitogen-activated protein kinase 15Homo sapiens (human)
centrosomeSerine/threonine-protein kinase Nek9Homo sapiens (human)
nucleusSerine/threonine-protein kinase Nek9Homo sapiens (human)
cytosolSerine/threonine-protein kinase Nek9Homo sapiens (human)
microtubule organizing centerSerine/threonine-protein kinase Nek7Homo sapiens (human)
spindle poleSerine/threonine-protein kinase Nek7Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase Nek7Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase Nek7Homo sapiens (human)
centrosomeSerine/threonine-protein kinase Nek7Homo sapiens (human)
microtubuleSerine/threonine-protein kinase Nek7Homo sapiens (human)
nucleusSerine/threonine-protein kinase Nek7Homo sapiens (human)
nucleusATP-dependent RNA helicase DDX1Homo sapiens (human)
nucleoplasmATP-dependent RNA helicase DDX1Homo sapiens (human)
cytoplasmATP-dependent RNA helicase DDX1Homo sapiens (human)
mitochondrionATP-dependent RNA helicase DDX1Homo sapiens (human)
cytosolATP-dependent RNA helicase DDX1Homo sapiens (human)
cytoplasmic stress granuleATP-dependent RNA helicase DDX1Homo sapiens (human)
membraneATP-dependent RNA helicase DDX1Homo sapiens (human)
cleavage bodyATP-dependent RNA helicase DDX1Homo sapiens (human)
tRNA-splicing ligase complexATP-dependent RNA helicase DDX1Homo sapiens (human)
ribonucleoprotein complexATP-dependent RNA helicase DDX1Homo sapiens (human)
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
membraneMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 1Homo sapiens (human)
kinetochoreAurora kinase BHomo sapiens (human)
condensed chromosome, centromeric regionAurora kinase BHomo sapiens (human)
nucleusAurora kinase BHomo sapiens (human)
nucleoplasmAurora kinase BHomo sapiens (human)
spindleAurora kinase BHomo sapiens (human)
cytosolAurora kinase BHomo sapiens (human)
chromocenterAurora kinase BHomo sapiens (human)
microtubule cytoskeletonAurora kinase BHomo sapiens (human)
midbodyAurora kinase BHomo sapiens (human)
chromosome passenger complexAurora kinase BHomo sapiens (human)
mitotic spindle poleAurora kinase BHomo sapiens (human)
mitotic spindle midzoneAurora kinase BHomo sapiens (human)
kinetochoreAurora kinase BHomo sapiens (human)
spindle pole centrosomeAurora kinase BHomo sapiens (human)
spindle microtubuleAurora kinase BHomo sapiens (human)
spindle midzoneAurora kinase BHomo sapiens (human)
microtubule organizing centerMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
cytoplasmMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
centrosomeMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
microtubule organizing centerMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
cytosolMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
microtubule cytoskeletonMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
dendriteMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
midbodyMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
neuron projectionMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
gamma-tubulin complexMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
ciliary basal bodyMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
cytoplasmMAP/microtubule affinity-regulating kinase 4Homo sapiens (human)
pericentriolar materialSerine/threonine-protein kinase Nek1Homo sapiens (human)
nucleusSerine/threonine-protein kinase Nek1Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase Nek1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase Nek1Homo sapiens (human)
centrosomeSerine/threonine-protein kinase Nek1Homo sapiens (human)
cytosolSerine/threonine-protein kinase Nek1Homo sapiens (human)
centriolar satelliteSerine/threonine-protein kinase Nek1Homo sapiens (human)
Elg1 RFC-like complexATPase family AAA domain-containing protein 5Homo sapiens (human)
nucleusATPase family AAA domain-containing protein 5Homo sapiens (human)
nucleusPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
cytoplasmPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
cytosolPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
cytosolPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
cytoplasmPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
nucleusPAS domain-containing serine/threonine-protein kinaseHomo sapiens (human)
nucleoplasmCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
cytosolCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
neuron projectionCalcium/calmodulin-dependent protein kinase kinase 2Homo sapiens (human)
nucleusEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
nucleoplasmEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
cytoplasmEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
EKC/KEOPS complexEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
cytosolEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
nucleusEKC/KEOPS complex subunit TP53RKHomo sapiens (human)
nucleusDual specificity testis-specific protein kinase 2Homo sapiens (human)
nucleoplasmDual specificity testis-specific protein kinase 2Homo sapiens (human)
nuclear bodyDual specificity testis-specific protein kinase 2Homo sapiens (human)
nucleusDual specificity testis-specific protein kinase 2Homo sapiens (human)
cytoplasmDual specificity testis-specific protein kinase 2Homo sapiens (human)
Golgi membraneMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
nucleoplasmMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
nucleolusMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
endoplasmic reticulumMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
endoplasmic reticulum membraneMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
Golgi apparatusMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
cytosolMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
membraneMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
cytoplasmMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
nucleusMembrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinaseHomo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
external side of plasma membraneMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein-containing complexMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
protein kinase complexMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
IRE1-TRAF2-ASK1 complexMitogen-activated protein kinase kinase kinase 5Homo sapiens (human)
cytoplasmAtaxin-2Homo sapiens (human)
Golgi apparatusAtaxin-2Homo sapiens (human)
trans-Golgi networkAtaxin-2Homo sapiens (human)
cytosolAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
membraneAtaxin-2Homo sapiens (human)
perinuclear region of cytoplasmAtaxin-2Homo sapiens (human)
ribonucleoprotein complexAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase 3Homo sapiens (human)
cytosolEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
cytosolEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
nucleusEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
cytoplasmEukaryotic translation initiation factor 2-alpha kinase 1Homo sapiens (human)
nucleoplasmTarget of rapamycin complex subunit LST8Homo sapiens (human)
cytoplasmTarget of rapamycin complex subunit LST8Homo sapiens (human)
lysosomal membraneTarget of rapamycin complex subunit LST8Homo sapiens (human)
cytosolTarget of rapamycin complex subunit LST8Homo sapiens (human)
TORC1 complexTarget of rapamycin complex subunit LST8Homo sapiens (human)
TORC2 complexTarget of rapamycin complex subunit LST8Homo sapiens (human)
nucleusNucleolar GTP-binding protein 1Homo sapiens (human)
nucleoplasmNucleolar GTP-binding protein 1Homo sapiens (human)
nucleolusNucleolar GTP-binding protein 1Homo sapiens (human)
cytoplasmNucleolar GTP-binding protein 1Homo sapiens (human)
cytosolNucleolar GTP-binding protein 1Homo sapiens (human)
membraneNucleolar GTP-binding protein 1Homo sapiens (human)
nuclear membraneNucleolar GTP-binding protein 1Homo sapiens (human)
perinuclear region of cytoplasmNucleolar GTP-binding protein 1Homo sapiens (human)
nucleolusNucleolar GTP-binding protein 1Homo sapiens (human)
nucleusSerine/threonine-protein kinase D2Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase D2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase D2Homo sapiens (human)
Golgi apparatusSerine/threonine-protein kinase D2Homo sapiens (human)
cytosolSerine/threonine-protein kinase D2Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase D2Homo sapiens (human)
cytosolSerine/threonine-protein kinase D2Homo sapiens (human)
chromosome, telomeric regionRNA cytidine acetyltransferaseHomo sapiens (human)
nucleusRNA cytidine acetyltransferaseHomo sapiens (human)
nucleoplasmRNA cytidine acetyltransferaseHomo sapiens (human)
nucleolusRNA cytidine acetyltransferaseHomo sapiens (human)
membraneRNA cytidine acetyltransferaseHomo sapiens (human)
midbodyRNA cytidine acetyltransferaseHomo sapiens (human)
telomerase holoenzyme complexRNA cytidine acetyltransferaseHomo sapiens (human)
small-subunit processomeRNA cytidine acetyltransferaseHomo sapiens (human)
nucleolusRNA cytidine acetyltransferaseHomo sapiens (human)
endoplasmic reticulum membraneSerine/threonine-protein kinase SIK2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase SIK2Homo sapiens (human)
nucleusSerine/threonine-protein kinase SIK2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase SIK2Homo sapiens (human)
cytoplasmSTE20-like serine/threonine-protein kinase Homo sapiens (human)
cytosolSTE20-like serine/threonine-protein kinase Homo sapiens (human)
cell leading edgeSTE20-like serine/threonine-protein kinase Homo sapiens (human)
perinuclear region of cytoplasmSTE20-like serine/threonine-protein kinase Homo sapiens (human)
extracellular exosomeSTE20-like serine/threonine-protein kinase Homo sapiens (human)
cytoplasmSTE20-like serine/threonine-protein kinase Homo sapiens (human)
plasma membraneSerine/threonine-protein kinase TAO3Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase TAO3Homo sapiens (human)
nucleusdCTP pyrophosphatase 1Homo sapiens (human)
nucleoplasmdCTP pyrophosphatase 1Homo sapiens (human)
mitochondriondCTP pyrophosphatase 1Homo sapiens (human)
cytosoldCTP pyrophosphatase 1Homo sapiens (human)
cytosoldCTP pyrophosphatase 1Homo sapiens (human)
nucleusDual specificity protein kinase CLK4Homo sapiens (human)
cytosolCasein kinase I isoform gamma-1Homo sapiens (human)
nucleusCasein kinase I isoform gamma-1Homo sapiens (human)
plasma membraneCasein kinase I isoform gamma-1Homo sapiens (human)
cytoplasmCasein kinase I isoform gamma-1Homo sapiens (human)
cytoplasmPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
cytosolPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
phenylalanine-tRNA ligase complexPhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
membranePhenylalanine--tRNA ligase beta subunitHomo sapiens (human)
nuclear speckBMP-2-inducible protein kinaseHomo sapiens (human)
cytoplasmBMP-2-inducible protein kinaseHomo sapiens (human)
nucleusBMP-2-inducible protein kinaseHomo sapiens (human)
extracellular regionObg-like ATPase 1Homo sapiens (human)
nucleolusObg-like ATPase 1Homo sapiens (human)
cytoplasmObg-like ATPase 1Homo sapiens (human)
centrosomeObg-like ATPase 1Homo sapiens (human)
cytosolObg-like ATPase 1Homo sapiens (human)
membraneObg-like ATPase 1Homo sapiens (human)
platelet alpha granule lumenObg-like ATPase 1Homo sapiens (human)
extracellular exosomeObg-like ATPase 1Homo sapiens (human)
cytoplasmObg-like ATPase 1Homo sapiens (human)
nucleusMidasinHomo sapiens (human)
nucleoplasmMidasinHomo sapiens (human)
nucleolusMidasinHomo sapiens (human)
cytosolMidasinHomo sapiens (human)
membraneMidasinHomo sapiens (human)
intermediate filament cytoskeletonMidasinHomo sapiens (human)
nucleusMidasinHomo sapiens (human)
preribosome, large subunit precursorMidasinHomo sapiens (human)
cytoplasmInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
cell surfaceInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
extrinsic component of plasma membraneInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
extracellular spaceInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
cytosolInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
plasma membraneInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
endosome membraneInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
nucleusInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
plasma membraneInterleukin-1 receptor-associated kinase 4Homo sapiens (human)
nucleusMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 20Homo sapiens (human)
cyclin K-CDK12 complexCyclin-dependent kinase 12Homo sapiens (human)
nucleoplasmCyclin-dependent kinase 12Homo sapiens (human)
nuclear speckCyclin-dependent kinase 12Homo sapiens (human)
nuclear cyclin-dependent protein kinase holoenzyme complexCyclin-dependent kinase 12Homo sapiens (human)
nucleusCyclin-dependent kinase 12Homo sapiens (human)
cyclin/CDK positive transcription elongation factor complexCyclin-dependent kinase 12Homo sapiens (human)
nucleoplasmNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
cytoplasmNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
mitochondrionNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
mitochondrial inner membraneNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
mitochondrial respirasomeNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
mitochondrial respiratory chain complex INADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
mitochondrial membraneNADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 26Homo sapiens (human)
Golgi apparatusSerine/threonine-protein kinase 26Homo sapiens (human)
Golgi-associated vesicleSerine/threonine-protein kinase 26Homo sapiens (human)
cytosolSerine/threonine-protein kinase 26Homo sapiens (human)
vesicle membraneSerine/threonine-protein kinase 26Homo sapiens (human)
membraneSerine/threonine-protein kinase 26Homo sapiens (human)
apical plasma membraneSerine/threonine-protein kinase 26Homo sapiens (human)
perinuclear region of cytoplasmSerine/threonine-protein kinase 26Homo sapiens (human)
extracellular exosomeSerine/threonine-protein kinase 26Homo sapiens (human)
cell peripherySerine/threonine-protein kinase 26Homo sapiens (human)
FAR/SIN/STRIPAK complexSerine/threonine-protein kinase 26Homo sapiens (human)
Golgi apparatusSerine/threonine-protein kinase 26Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase 26Homo sapiens (human)
mitochondrionSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
mitochondrial matrixSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinate-CoA ligase complex (ADP-forming)Succinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
extracellular exosomeSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
mitochondrionSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
succinate-CoA ligase complexSuccinate--CoA ligase [ADP-forming] subunit beta, mitochondrialHomo sapiens (human)
nucleusSerine/threonine-protein kinase NLKHomo sapiens (human)
nucleoplasmSerine/threonine-protein kinase NLKHomo sapiens (human)
cytosolSerine/threonine-protein kinase NLKHomo sapiens (human)
nucleusSerine/threonine-protein kinase NLKHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase NLKHomo sapiens (human)
extracellular space5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
nucleoplasm5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
cytosol5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
nucleotide-activated protein kinase complex5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
cytoplasm5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
nucleus5'-AMP-activated protein kinase subunit gamma-2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase TBK1Homo sapiens (human)
cytosolSerine/threonine-protein kinase TBK1Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase TBK1Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase TBK1Homo sapiens (human)
cytosolSerine/threonine-protein kinase TBK1Homo sapiens (human)
intracellular membrane-bounded organelleSerine/threonine-protein kinase TBK1Homo sapiens (human)
serine/threonine protein kinase complexSerine/threonine-protein kinase TBK1Homo sapiens (human)
stress fiberSeptin-9Homo sapiens (human)
cytoplasmSeptin-9Homo sapiens (human)
microtubuleSeptin-9Homo sapiens (human)
axonemeSeptin-9Homo sapiens (human)
actin cytoskeletonSeptin-9Homo sapiens (human)
perinuclear region of cytoplasmSeptin-9Homo sapiens (human)
non-motile ciliumSeptin-9Homo sapiens (human)
septin complexSeptin-9Homo sapiens (human)
septin ringSeptin-9Homo sapiens (human)
microtubule cytoskeletonSeptin-9Homo sapiens (human)
cell division siteSeptin-9Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
sarcolemmaPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
GABA-ergic synapsePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic specialization membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
dendritic spinePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
fibrillar centerRibosomal protein S6 kinase alpha-6Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-6Homo sapiens (human)
nucleolusRibosomal protein S6 kinase alpha-6Homo sapiens (human)
mitochondrionRibosomal protein S6 kinase alpha-6Homo sapiens (human)
cytosolRibosomal protein S6 kinase alpha-6Homo sapiens (human)
cytoplasmRibosomal protein S6 kinase alpha-6Homo sapiens (human)
nucleoplasmRibosomal protein S6 kinase alpha-6Homo sapiens (human)
nucleusTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
nucleoplasmTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
cytoplasmTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
cytosolTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
cytoskeletonTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
apical plasma membraneTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
recycling endosomeTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
extracellular exosomeTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
presynapseTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
glutamatergic synapseTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
postsynaptic density, intracellular componentTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
cytoplasmTRAF2 and NCK-interacting protein kinaseHomo sapiens (human)
actin cytoskeletonSerine/threonine-protein kinase TAO2Homo sapiens (human)
nucleoplasmSerine/threonine-protein kinase TAO2Homo sapiens (human)
nucleolusSerine/threonine-protein kinase TAO2Homo sapiens (human)
cytosolSerine/threonine-protein kinase TAO2Homo sapiens (human)
axonSerine/threonine-protein kinase TAO2Homo sapiens (human)
cytoplasmic vesicle membraneSerine/threonine-protein kinase TAO2Homo sapiens (human)
cytoplasmic vesicleSerine/threonine-protein kinase TAO2Homo sapiens (human)
neuron projectionSerine/threonine-protein kinase TAO2Homo sapiens (human)
dendritic growth coneSerine/threonine-protein kinase TAO2Homo sapiens (human)
axonal growth coneSerine/threonine-protein kinase TAO2Homo sapiens (human)
receptor complexSerine/threonine-protein kinase TAO2Homo sapiens (human)
cytoplasmSerine/threonine-protein kinase TAO2Homo sapiens (human)
nucleoplasmLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
nucleolusLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
mitochondrionLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
mitochondrial outer membraneLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
endoplasmic reticulumLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
endoplasmic reticulum membraneLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
plasma membraneLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
membraneLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
endoplasmic reticulumLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
membraneLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
mitochondrionLong-chain-fatty-acid--CoA ligase 5Homo sapiens (human)
nucleusRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
cytoplasmRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
membraneRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
nucleoplasmRAC-gamma serine/threonine-protein kinaseHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase SIK3Homo sapiens (human)
nucleoplasmMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase 2Homo sapiens (human)
exon-exon junction complexThyroid hormone receptor-associated protein 3Homo sapiens (human)
nucleusThyroid hormone receptor-associated protein 3Homo sapiens (human)
nucleoplasmThyroid hormone receptor-associated protein 3Homo sapiens (human)
nuclear speckThyroid hormone receptor-associated protein 3Homo sapiens (human)
extracellular exosomeThyroid hormone receptor-associated protein 3Homo sapiens (human)
mediator complexThyroid hormone receptor-associated protein 3Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
cytosolMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
plasma membraneMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase kinase 5Homo sapiens (human)
nucleusReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
nucleusReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
cytosolReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
protein-containing complexReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
cytoplasmReceptor-interacting serine/threonine-protein kinase 3Homo sapiens (human)
cytosolSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
plasma membraneSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
cell-cell junctionSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
lamellipodiumSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
cell leading edgeSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
actomyosinSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
extracellular exosomeSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
cytoskeletonSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
cytoplasmSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
actomyosinSerine/threonine-protein kinase MRCK betaHomo sapiens (human)
nucleusInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
cytoplasmInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
nucleusInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
plasma membraneInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
cytoplasmInterleukin-1 receptor-associated kinase 3Homo sapiens (human)
cytoplasmCasein kinase I isoform gamma-3Homo sapiens (human)
plasma membraneCasein kinase I isoform gamma-3Homo sapiens (human)
cytoplasmCasein kinase I isoform gamma-3Homo sapiens (human)
nucleusCasein kinase I isoform gamma-3Homo sapiens (human)
cytoplasmMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
perinuclear region of cytoplasmMitogen-activated protein kinase kinase kinase 4Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (391)

Assay IDTitleYearJournalArticle
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1876329Antiviral activity against MERS-CoV infected in 1 hr pretreated human Huh-7 cells assessed as inhibition of viral growth at 10 uM measured after 48 hrs by plaque reduction assay relative to control2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Kinases as Potential Therapeutic Targets for Anti-coronaviral Therapy.
AID1425005Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425174Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718215Elimination half life in C57BL/6J mouse plasma at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1424992Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425117Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425203Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718226AUC (0 to 8 hrs) in C57BL/6J mouse brain at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1425000Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425060Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424925Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153720Cytotoxicity against human COLO205 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1424929Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425147Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424505Antitumor activity in renal cell carcinoma patient without previous VEGFR TKI treatment assessed as progression free survival (Rvb = 1.9 months)2017European journal of medicinal chemistry, Dec-15, Volume: 142Recent developments in small molecule therapies for renal cell carcinoma.
AID1425122Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1409403Binding affinity to mTOR FKBP12 site (unknown origin)2018Journal of medicinal chemistry, 11-21, Volume: 61, Issue:22
Discovery and Preclinical Characterization of 5-[4,6-Bis({3-oxa-8-azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoromethyl)pyridin-2-amine (PQR620), a Highly Potent and Selective mTORC1/2 Inhibitor for Cancer and Neurological Disorders.
AID1662433Inhibition of mTOR in HEK293 cells2020Journal of medicinal chemistry, 09-24, Volume: 63, Issue:18
p62/SQSTM1, a Central but Unexploited Target: Advances in Its Physiological/Pathogenic Functions and Small Molecular Modulators.
AID1424937Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425010Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425170Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425164Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424960Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1207342Inhibition of fast sodium current (INa) in HEK293 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID1425188Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1876111Cytotoxicity against human Huh-7 cells2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1425024Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424948Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424922Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425159Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718223Cmax in C57BL/6J mouse brain at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1425115Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424955Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425161Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425107Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424949Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153724Inhibition of mTORC1 in human COLO205 cells assessed as reduction of S6 phosphorylation at 0.1 to 8 uM after 24 hrs by Western blotting2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1425104Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425202Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071501Cytotoxicity against human Caki1 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1424915Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718218Cmax in C57BL/6J mouse plasma at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1424947Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425128Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424897Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424975Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425072Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425133Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425046Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1065394Oral bioavailability in human2014Journal of medicinal chemistry, Jan-23, Volume: 57, Issue:2
Macrocyclic drugs and clinical candidates: what can medicinal chemists learn from their properties?
AID1424959Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424506Antitumor activity in renal cell carcinoma patient with 2 previous VEGFR TKI treatment assessed as progression free survival (Rvb = 1.8 months)2017European journal of medicinal chemistry, Dec-15, Volume: 142Recent developments in small molecule therapies for renal cell carcinoma.
AID1424956Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1207402Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells transfected with KCNQ1 / Kv1.7 / KvLQT1 and KCNE1/minK measured using IonWorks automated patch clamp platform
AID1425050Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425088Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425026Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153721Inhibition of mTORC1 in human SK-HEP1 cells assessed as reduction of S6 phosphorylation at 0.1 to 8 uM after 24 hrs by Western blotting2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1425137Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1207282Inhibition of long-lasting type calcium current (ICaL) in HEK293 cells (alpha1C/beta2a/alpha2delta1) cells measured using IonWorks Barracuda automated patch clamp platform
AID1236017Induction of autophagic punctation in human A549 cells assessed as increase of puncta with p62 at 5 uM after 6 hrs by immunofluorescence staining-based microscopic analysis relative to control2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1207311Inhibition of fast sodium current (INa) in Chinese Hamster Ovary (CHO) K1 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID1425173Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425012Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424928Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425068Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425197Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424970Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425078Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425087Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424969Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425127Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424988Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424991Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718230Elimination half life in C57BL/6J mouse brain at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1207371Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells expressing hKvLQT1/hminK measured using IonWorks Quattro automated patch clamp platform
AID1425019Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425204Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425053Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425083Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424912Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425210Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425001Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424944Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425064Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425195Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425044Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424923Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425193Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718225Elimination half life in C57BL/6J mouse brain at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1425136Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425168Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424983Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425029Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425150Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473740Inhibition of human MRP3 overexpressed in Sf9 insect cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 10 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1425080Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1236023Inhibition of AKT in human A549 cells assessed as downregulation of PRAS40 phosphorylation at thr246 at 5 uM after 3 to 12 hrs by Western blot analysis2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1425062Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473910Ratio of drug concentration at steady state in human at 0.25 to 20 mg, po after 24 hrs to IC50 for human BSEP overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1424950Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425014Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424926Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425102Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071500Cytotoxicity against human HT-29 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1424918Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424965Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425004Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473912Ratio of drug concentration at steady state in human at 0.25 to 20 mg, po after 24 hrs to IC50 for human MRP3 overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1424909Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718232Clearance in C57BL/6J mouse brain at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1424908Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425171Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425097Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424940Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473739Inhibition of human MRP2 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1718222Clearance in C57BL/6J mouse plasma at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1425106Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425063Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1236019Inhibition of mTOR phosphorylation at ser2448 in human A549 cells at 5 uM after 3 to 24 hrs by Western blot analysis2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1424979Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424905Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425153Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425038Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425018Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425074Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425054Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1236012Induction of autophagy in human A549 cells assessed as increase in conversion of LC3-1 to LC3-2 at 5 uM after 6 hrs by Western blot analysis2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1425201Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153726Inhibition of mTORC1 in human COLO205 cells assessed as AKT phosphorylation at Ser473 at 0.1 to 8 uM after 24 hrs by Western blotting2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1425166Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424958Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473908AUC in human at 0.25 to 20 mg, po QD after 24 hrs2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1153719Cytotoxicity against human HOP62 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1425144Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424935Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1207431Inhibition of transient outward potassium current (Ito) current in Chinese Hamster Ovary (CHO) K1 cells expressing human Kv4.3 measured using IonWorks Quattro automated patch clamp platform
AID1071505Cytotoxicity against human COLO205 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1424896Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425059Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1876110Cytotoxicity against human HaCaT cells2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1425175Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425198Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424892Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID706794Increase in allograft survival in stringent Dark Agouti to Lewis rat heterotopic vascular heart allotransplantation model assessed as allograft mean survival time at 0.3 mg/kg, po qd (Rvb = 6 days)2012Journal of medicinal chemistry, Nov-26, Volume: 55, Issue:22
An oral sphingosine 1-phosphate receptor 1 (S1P(1)) antagonist prodrug with efficacy in vivo: discovery, synthesis, and evaluation.
AID1425149Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425108Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425206Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153725Inhibition of mTORC1 in human COLO205 cells assessed as reduction of 4-EBP1 phosphorylation at 0.1 to 8 uM after 24 hrs by Western blotting2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1425042Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1875901Antiviral activity against MERS-CoV infected in 1 hr pretreated human Huh-7 cells at 10 uM relative to control2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1424917Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424984Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424978Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425051Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1876108Inhibition of mTOR (unknown origin)2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1583277Protection against C57BL/6J mouse model of neuronal mTOR hyperactivity assessed as survival rate at 6 mg/kg, ip administered on postnatal day 22 daily and measured on postnatal day 52 (Rvb = 48 %)2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery of a Brain-Penetrant ATP-Competitive Inhibitor of the Mechanistic Target of Rapamycin (mTOR) for CNS Disorders.
AID1425176Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425095Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425103Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718267Toxicity in Tsc1-GFAP knockout Balb/c mouse assessed as change in body weight at 10 mg/kg, po
AID1424933Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424914Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424890Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425002Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425035Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425189Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425140Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424977Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424993Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424987Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718213Cmax in C57BL/6J mouse plasma at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1424994Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473741Inhibition of human MRP4 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1718231AUC (0 to 8 hrs) in C57BL/6J mouse brain at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1425185Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425118Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425027Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425090Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424920Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718262Protection against Tsc1-GFAP knockout induced mortality in Balb/c mouse assessed as increase in survival rate at 10 mg/kg, po administered as single dose
AID1424911Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718214Tmax in C57BL/6J mouse plasma at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1424971Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424973Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425116Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1425073Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425213Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425003Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1876090Antiviral activity against MERS-CoV at 10 uM relative to control2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1424968Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1916668Inhibition of mTORC1 (unknown origin)2022European journal of medicinal chemistry, Aug-05, Volume: 238Contemporary mTOR inhibitor scaffolds to diseases breakdown: A patent review (2015-2021).
AID1425138Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425043Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071502Cytotoxicity against human ColoR cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1425179Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425007Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425154Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425022Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424921Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425208Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425061Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424967Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425045Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424931Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1236021Inhibition of mTOR in human A549 cells assessed as downregulation of p70S6K phosphorylation at thr389 at 5 uM after 3 to 24 hrs by Western blot analysis2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1425156Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425132Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424899Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424900Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425030Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424972Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424898Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425119Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718229Tmax in C57BL/6J mouse brain at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1718220Elimination half life in C57BL/6J mouse plasma at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1425099Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425191Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425056Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425049Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425016Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425039Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718217Clearance in C57BL/6J mouse plasma at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1425036Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425181Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425211Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425177Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425143Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425160Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425113Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071503Cytotoxicity against human HCT116 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1207465Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) K1 cells stably expressing hERG measured using IonWorks Quattro automated patch clamp platform
AID1424942Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424919Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424910Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1207559Inhibition of long-lasting type calcium current (hICa) in Chinese Hamster Ovary (CHO) cells expressing hCav1.2 measured using IonWorks Quattro automated patch clamp platform
AID1425098Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424907Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473909Drug concentration at steady state in human at 0.25 to 20 mg, po QD after 24 hrs2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1425192Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425196Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718219Tmax in C57BL/6J mouse plasma at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1876109Cytotoxicity against human HeLa cells2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1425142Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425081Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425100Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425089Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425124Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425126Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153722Inhibition of mTORC1 in human SK-HEP1 cells assessed as reduction of 4-EBP1 phosphorylation at 0.1 to 8 uM after 24 hrs by Western blotting2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1425121Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424954Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425172Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424997Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424985Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425028Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425190Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424503Antitumor activity in patient with metastatic renal cell carcinoma assessed as progression free survival2017European journal of medicinal chemistry, Dec-15, Volume: 142Recent developments in small molecule therapies for renal cell carcinoma.
AID1424893Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425163Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425082Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425017Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153723Inhibition of mTORC1 in human SK-HEP1 cells assessed as AKT phosphorylation at Ser473 at 0.1 to 8 uM after 24 hrs by Western blotting2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1424962Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718243In vivo inhibition of PI3Kalpha in C57BL/6J mouse assessed as increase in plasma glucose concentration at 10 mg/kg, po measured after 8 hrs by glucoseoxidase-peroxide coupled colorimetric assay
AID1424889Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071499Cytotoxicity against human OVCAR3 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1425199Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425182Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424986Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425169Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425048Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425131Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425162Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425096Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424936Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424946Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425057Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425047Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425008Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424934Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424930Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425055Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425187Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424963Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425079Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718247In vivo inhibition of mTORC1 signalling in C57BL/6J mouse assessed as reduction in RPS6 phosphorylation at S235/236 residue in cortex at 10 mg/kg, po incubated for 30 mins by Western blot analysis
AID1424974Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425155Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425037Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425058Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424894Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425040Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425141Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425013Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424952Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424999Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425157Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425178Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425186Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425212Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425129Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071495Cytotoxicity against human HOP62 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1425023Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473913Ratio of drug concentration at steady state in human at 0.25 to 20 mg, po after 24 hrs to IC50 for human MRP4 overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1425200Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425093Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424902Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425086Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424485Antitumor activity in patient with metastatic renal cell carcinoma assessed as overall survival2017European journal of medicinal chemistry, Dec-15, Volume: 142Recent developments in small molecule therapies for renal cell carcinoma.
AID1424996Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425069Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425209Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425167Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1236011Prevention of autophagosome degradation in human A549 cells assessed as upregulation of p62 expression at 5 uM after 6 hrs by Western blot analysis2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1424981Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424957Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718245In vivo inhibition of PI3Kalpha C57BL/6J mouse assessed as increase in plasma insulin concentration at 10 mg/kg, po measured after 8 hrs by ELISA
AID1207496Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) cells stable expressing hERG measured using IonWorks Barracuda automated patch clamp platform
AID1071498Cytotoxicity against human DU145 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1425125Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718224Tmax in C57BL/6J mouse brain at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1425070Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424895Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424924Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425034Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425205Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1153718Cytotoxicity against human SQ20B cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Jun-23, Volume: 81Benzofuran derivatives as anticancer inhibitors of mTOR signaling.
AID1718216AUC (0 to 8 hrs) in C57BL/6J mouse plasma at 10 mg/kg, po administered as single dose by mass spectrometry based UHPLC analysis
AID1207525Inhibition of rapid delayed inward rectifying potassium current (IKr) measured using manual patch clamp assay
AID1425111Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425148Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425123Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1236016Induction of autophagic punctation in human A549 cells assessed as increase of puncta with LC3 at 5 uM after 6 hrs by immunofluorescence staining-based microscopic analysis relative to control2015Journal of natural products, Jul-24, Volume: 78, Issue:7
Bisleuconothine A Induces Autophagosome Formation by Interfering with AKT-mTOR Signaling Pathway.
AID1424998Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425207Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425025Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425134Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424891Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424939Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425194Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425130Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473738Inhibition of human BSEP overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-taurocholate in presence of ATP measured after 15 to 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1425110Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425011Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424945Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425105Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424904Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718221AUC (0 to 8 hrs) in C57BL/6J mouse plasma at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1424989Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1425076Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425094Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1718228Cmax in C57BL/6J mouse brain at 10 mg/kg, po administered once a day for 4 consecutive days by mass spectrometry based UHPLC analysis
AID1425006Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424976Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425145Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424932Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1583253Inhibition of oversizing of mouse primary cortical neuron harboring TSC1 deletion mutant at 20 nM incubated for 7 days by immunofluorescence assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery of a Brain-Penetrant ATP-Competitive Inhibitor of the Mechanistic Target of Rapamycin (mTOR) for CNS Disorders.
AID1425158Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425165Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1473911Ratio of drug concentration at steady state in human at 0.25 to 20 mg, po after 24 hrs to IC50 for human MRP2 overexpressed in Sf9 insect cells2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1425015Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1875938Antiviral activity against MERS-CoV infected in human Huh-7 cells at 10 uM relative to control2022Journal of medicinal chemistry, 01-27, Volume: 65, Issue:2
Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections.
AID1425084Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425033Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425146Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425120Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424953Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425085Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424941Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071497Cytotoxicity against human SKHEP1 cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1425052Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425071Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1071513Cytotoxicity against human SQ20B cells after 72 hrs by MTT assay2014European journal of medicinal chemistry, Mar-03, Volume: 74Benzofuran derivatives as a novel class of inhibitors of mTOR signaling.
AID1424966Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424901Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424990Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424964Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424995Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424980Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1425065Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1424961Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID1583252Inhibition of overgrowth of mouse primary cortical neuron harboring TSC1 deletion mutant at 20 nM incubated for 7 days by immunofluorescence assay2020Journal of medicinal chemistry, 02-13, Volume: 63, Issue:3
Discovery of a Brain-Penetrant ATP-Competitive Inhibitor of the Mechanistic Target of Rapamycin (mTOR) for CNS Disorders.
AID1425009Kinobeads (epsilon), multiple immobilized ATP-competitive broad spectrum kinase inhibitors, used to assess residual binding of ~300 proteins simultaneously from cell lysate in the presence of a compound. Quantitative readout performed by mass spectrometry2017Science (New York, N.Y.), 12-01, Volume: 358, Issue:6367
The target landscape of clinical kinase drugs.
AID504749qHTS profiling for inhibitors of Plasmodium falciparum proliferation2011Science (New York, N.Y.), Aug-05, Volume: 333, Issue:6043
Chemical genomic profiling for antimalarial therapies, response signatures, and molecular targets.
AID1345726Human mechanistic target of rapamycin kinase (FRAP subfamily)1998Transplantation proceedings, Aug, Volume: 30, Issue:5
Chemical modification of rapamycin: the discovery of SDZ RAD.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (5,432)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's20 (0.37)18.2507
2000's596 (10.97)29.6817
2010's3917 (72.11)24.3611
2020's899 (16.55)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 82.19

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index82.19 (24.57)
Research Supply Index8.84 (2.92)
Research Growth Index6.25 (4.65)
Search Engine Demand Index148.64 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (82.19)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials1,289 (22.89%)5.53%
Reviews767 (13.62%)6.00%
Case Studies596 (10.58%)4.05%
Observational147 (2.61%)0.25%
Other2,832 (50.29%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (895)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Outcome-related Factors in Patients With Metastatic Renal Cell Carcinoma Treated With Everolimus After Failure of a First-line Treatment With VEGF Inhibitor [NCT02338570]Phase 431 participants (Actual)Interventional2015-07-31Terminated(stopped due to Stopped on November 16th 2016, because of recruitment failure.)
A Phase 1b Trial of ARV-471 in Combination With Everolimus in Patients With ER+, HER2- Advanced or Metastatic Breast Cancer [NCT05501769]Phase 132 participants (Anticipated)Interventional2022-09-08Recruiting
An Open-label, Phase II, Single-arm Study of Everolimus in Combination With Letrozole in the Treatment of Postmenopausal Women With Estrogen Receptor Positive HER2 Negative Metastatic or Locally Advanced Breast Cancer [NCT01698918]Phase 2202 participants (Actual)Interventional2013-03-07Completed
Phase I Study of the Combination of Bevacizumab, Everolimus and LBH589 (BEL) for the Treatment of Advanced Solid Tumors [NCT01055795]Phase 114 participants (Actual)Interventional2010-03-31Completed
Phase II Trial on Efficacy of mTOR Inhibitor RAD001 as Maintenance Therapy for Patients Above 60 Years in Mantle Cell Lymphoma After First and Second Line Chemotherapy [NCT00727207]Phase 235 participants (Anticipated)Interventional2008-05-31Terminated(stopped due to Lack of participations (8 of 25))
Phase II Study of Everolimus in Children and Adolescents With Refractory or Relapsed Osteosarcoma [NCT01216826]Phase 220 participants (Anticipated)Interventional2011-03-31Recruiting
A Phase IB/II Study of Second Line Therapy With Panitumumab, Irinotecan and Everolimus (PIE) in Metastatic Colorectal Cancer With KRAS WT [NCT01139138]Phase 1/Phase 249 participants (Actual)Interventional2010-06-30Completed
Randomized Phase II Study of Sorafenib With or Without Everolimus in Patients With Radioactive Iodine Refractory Hurthle Cell Thyroid Cancer [NCT02143726]Phase 235 participants (Actual)Interventional2014-10-01Active, not recruiting
A Twelve-month, Multicenter, Open-label, Randomized Study of the Safety, Tolerability and Efficacy of Everolimus With IL-2 Receptor Antagonist, Corticosteroids and Two Different Exposure Levels of Tacrolimus in de Novo Renal Transplant Recipients [NCT00369161]Phase 4228 participants (Actual)Interventional2006-06-30Completed
Everolimus Added to Long Acting Octreotide as a Volume Reducing Treatment of Polycystic Livers [NCT01157858]Phase 244 participants (Actual)Interventional2010-06-30Completed
[NCT01167530]Phase 136 participants (Anticipated)Interventional2008-03-31Recruiting
An Open Label Multi-Center Phase II Study of RAD001 in Advanced Thyroid Cancer [NCT01164176]Phase 240 participants (Actual)Interventional2010-03-31Completed
A Phase Ib, Open-label Study to Evaluate RAD001 as Monotherapy Treatment in Chinese Patients With Advanced Pulmonary Neuroendocrine Tumor [NCT01175096]Phase 1/Phase 220 participants (Anticipated)Interventional2010-07-31Active, not recruiting
Hormone Receptor Positive Disease Across Solid Tumor Types: A Phase I Study of Single-Agent Hormone Blockade and Combination Approaches With Targeted Agents to Provide Synergy and Overcome Resistance [NCT01197170]Phase 1277 participants (Actual)Interventional2010-09-07Completed
A Phase II Study of the Mammalian Target of Rapamycin (mTOR) Inhibitor RAD001 in Combination With Imatinib Mesylate in Patients With Previously Treated Advanced Renal Carcinoma [NCT00331409]Phase 223 participants (Actual)Interventional2006-01-31Completed
A Randomized Three Arm Phase II Study of (1) Everolimus, (2) Estrogen Deprivation Therapy (EDT) With Leuprolide + Letrozole and (3) Everolimus + EDT in Patients With Unresectable Fibrolamellar Hepatocellular Carcinoma (FLL-HCC) [NCT01642186]Phase 228 participants (Actual)Interventional2012-07-12Completed
A Clinical Evaluation of the Everolimus Eluting Bioresorbable Vascular Scaffold System (BVS) for the Treatment of Subjects With Critical Limb Ischemia (CLI) From Occlusive Vascular Disease of the Tibial Arteries [NCT01341340]2 participants (Actual)Interventional2011-11-30Terminated(stopped due to Discontinued due to poor enrollment. Insufficient number of patients enrolled to permit a statistically rigorous assessment of safety and efficacy.)
2-phased Randomized Comparison Between PromusTMElementTM Versus Xience PRIME® Stent [NCT01581515]Phase 4100 participants (Anticipated)Interventional2012-01-31Recruiting
NOBORI Biolimus-Eluting Versus XIENCE/PROMUS Everolimus-eluting Stent Trial [NCT01303640]Phase 43,235 participants (Actual)Interventional2011-05-31Completed
BARIS - BIBF1120 and RAD001 in Solid Tumors. A Phase I Trial to Evaluate the Safety and Tolerability of Combined BIBF 1120 and RAD001 in Solid Tumors and to Determine the Maximum Tolerated Dose (MTD) of the Combination [NCT01349296]Phase 118 participants (Actual)Interventional2012-07-31Completed
A Phase II, Randomized Two-Arm Study of Everolimus and Letrozole, +/- Ribociclib (LEE011) in Patients With Advanced or Recurrent Endometrial Carcinoma [NCT03008408]Phase 299 participants (Anticipated)Interventional2017-08-18Recruiting
Assessment of Neointimal Stent Strut Coverage and Endothelial Function After Second Generation Drug-Eluting Stent Systems [NCT01073111]0 participants (Actual)Interventional2010-04-30Withdrawn
Multicenter International Durability and Safety of Sirolimus in LAM Trial (MIDAS) [NCT02432560]600 participants (Anticipated)Observational2015-03-31Recruiting
Phase II Study of the IGF-1 Inhibitor Pasireotide Lar in Combination With the m-TOR Inhibitor Everolimus in Patients With Relapsed/Refractory Multiple Myeloma [NCT01234974]Phase 20 participants (Actual)Interventional2010-12-31Withdrawn(stopped due to Study never undertaken)
Dose-finding Study of CAELYXTM and RAD001 in Patients With Advanced Solid Tumors [NCT01148628]Phase 154 participants (Actual)Interventional2007-10-31Active, not recruiting
A Phase II/III Clinical Study to Evaluate the Efficacy and Safety of Fruquintinib in Combination With Sintilimab Versus Axitinib or Everolimus as Second-line Treatment for Locally Advanced or Metastatic Renal Cell Carcinoma [NCT05522231]Phase 2/Phase 3264 participants (Anticipated)Interventional2022-10-27Recruiting
Phase II Study of RAD001monotherapy in Patients With Unresectable Adenoid Cystic Carcinoma [NCT01152840]Phase 233 participants (Actual)Interventional2008-07-31Completed
Comparison of the Efficacy and Safety of New Platform Everolimus-eluting Coronary Stent System (Promus Element) With Zotarolimus-eluting Coronary Stent System (Endeavor Resolute) and Triple Anti-platelet Therapy With Double-dose Clopidogrel Anti-platelet [NCT01267734]Phase 43,750 participants (Anticipated)Interventional2010-06-30Recruiting
A 6-Month Open Pilot Study to Investigate the Safety and Tolerability of Immediate Conversion From Calcineurin Inhibitor Tacrolimus to Everolimus in Stable Maintenance Renal Transplant Recipients [NCT01269684]Phase 40 participants (Actual)Interventional2008-08-31Withdrawn(stopped due to No patient has never been included in the trial)
Phase I Trial of Gemcitabine and Split-Dose Cisplatin Plus Everolimus (RAD001) in Patients With Advanced Solid Tumor Malignancies [NCT01182168]Phase 112 participants (Actual)Interventional2010-08-31Completed
An Observational Safety Evaluation of Patients Treated With the NEVO™ Sirolimus-eluting Coronary Stent. [NCT01202058]Phase 3156 participants (Actual)Interventional2010-08-31Terminated(stopped due to The NEVO™ stent will not be commercialized. Cordis decided to close the study after 1 years. This decision took the absence of safety signals into account.)
Phase I Study of Anti-IGF-1R Monoclonal Antibody, IMC-A12, and mTOR Inhibitor, Everolimus, in Advanced Low to Intermediate Grade Neuroendocrine Carcinoma [NCT01204476]Phase 127 participants (Actual)Interventional2010-10-31Completed
Phase II Study of Everolimus (RAD001, Afinitor®) for Children With Recurrent or Progressive Ependymoma [NCT02155920]Phase 211 participants (Actual)Interventional2015-02-28Completed
Effect of the Inhibition of the Mammalian Target of Rapamycin on Metabolism and Exercise. [NCT01561404]Phase 43 participants (Actual)Interventional2011-09-30Terminated(stopped due to lack of potential patients)
A Phase II Trial of Everolimus and Bevacizumab in Advanced Non-Clear Cell Renal Cell Carcinoma (RCC) [NCT01399918]Phase 257 participants (Actual)Interventional2011-07-31Completed
A Phase II Study on Everolimus, an mTOR Inhibitor (Oral Formulation), With Octreotide LAR® in Adult Patients With Advanced, Non-functioning, Well-differentiated Gastrointestinal Neuroendocrine Tumours (GI NET) [NCT01567488]Phase 243 participants (Actual)Interventional2011-06-08Completed
A Prospective, Active Control, Open-label, Multinational, Randomized Clinical Trial Comparing Single Long BioMimeTM Morph Coronary Stent System vs. Two Overlapping Xience Family Coronary Stent Systems to Evaluate Safety and Performance in Patients With Lo [NCT03721614]200 participants (Anticipated)Interventional2019-09-15Not yet recruiting
A Phase I/II Trial of RAD001/Capecitabine in Refractory Gastric Cancer [NCT01099527]Phase 1/Phase 259 participants (Actual)Interventional2009-10-31Completed
A Phase I Study of Oral Administration of mTOR Inhibitor Everolimus (RAD001) in Association With Cisplatin and Radiotherapy for the Treatment of Locally Advanced Cervix Cancer [NCT01217177]Phase 114 participants (Actual)Interventional2011-12-31Completed
Phase II Study of Everolimus in Children and Adolescents With Refractory or Relapsed Rhabdomyosarcoma and Other Soft Tissue Sarcomas [NCT01216839]Phase 220 participants (Anticipated)Interventional2011-03-31Recruiting
RANDOMIZED TRIAL OF ZOTAROLIMUS- AND EVEROLIMUS-ELUTING STENTS REGARDING STENT COVERAGE ASSESSED BY OCT [NCT01230723]Phase 430 participants (Anticipated)Interventional2010-10-31Active, not recruiting
Comparison of the Everolimus Eluting (XIENCE-V®, XIENCE-Prime® or PROMUS® Stent) With the Biolimus A9 Eluting NOBORI® Stent in All-comers: a Randomized Open Label Study [NCT01233453]Phase 42,700 participants (Anticipated)Interventional2009-01-31Active, not recruiting
A New Strategy Regarding Discontinuation of Dual Antiplatelet; Real Safety and Efficacy of a 3-month Dual Antiplatelet Therapy Following Zotarolimus-eluting Stents Implantation (RESET Trial) [NCT01145079]Phase 4982 participants (Actual)Interventional2009-05-31Completed
Comparison of the Efficacy of Everolimus-Eluting Versus Zotarolimus-Eluting Stent for Coronary Lesions in Acute Myocardial Infarction [NCT01347554]Phase 4461 participants (Actual)Interventional2009-01-31Completed
A Phase I and Surgical Study of Ribociclib and Everolimus (RAD001) in Children With Recurrent or Refractory Malignant Brain Tumors [NCT03387020]Phase 122 participants (Actual)Interventional2018-01-13Completed
Clinical Outcome of de Novo Everolimus-based Immunosuppressive Therapy for Renal Transplantation Using Rituximab Induction [NCT01312064]Phase 42 participants (Actual)Interventional2011-04-30Terminated(stopped due to Difficulty collecting)
A Phase Ib, Multi-center, Open-label Study to Evaluate the Safety of RAD001 in Chinese Patients With Metastatic Renal Cell Cancer Who Are Intolerant of or Who Have Progressed Despite Treatment With VEGF-targeted Therapies [NCT01152801]Phase 164 participants (Actual)Interventional2010-05-31Completed
A Phase II Study of Everolimus and Sorafenib in Patients With Metastatic Differentiated Thyroid Cancer Who Have Progressed on Sorafenib Alone [NCT01263951]Phase 235 participants (Actual)Interventional2010-11-30Active, not recruiting
An Open Label, Single Arm Trial to Characterize Patients With Metastatic Renal Cell Carcinoma Treated With Everolimus After Failure of the First VEGF-targeted Therapy [NCT01266837]Phase 464 participants (Actual)Interventional2011-03-31Completed
A Multicenter Phase II Study to Evaluate the Clinical Activity and the Safety Profile of Everolimus (RAD001) in Marginal Zone B-cell Lymphomas (MZL) EudraCT Number 2009-011725-14 [NCT01164267]Phase 230 participants (Actual)Interventional2010-03-31Completed
Evaluation of Late Clinical Events After Drug-eluting Versus Bare-metal Stents in Patients at Risk: BAsel Stent Kosten Effektivitäts Trial - PROspective Validation Examination Part II (BASKET-PROVE II) [NCT01166685]Phase 42,291 participants (Actual)Interventional2010-04-30Completed
Pilot Study of Everolimus in the Treatment of Neoplasms in Patients With Peutz-Jeghers Syndrome [NCT01178151]Phase 20 participants (Actual)Interventional2010-10-31Withdrawn(stopped due to No patients)
Polyomavirus BK Nephropathy After Renal Transplantation: Randomized Clinical Trial to Demonstrate That Switching to mTOR Inhibitor is More Effective Than a Reduction of Immunosuppressive Therapy [NCT01289301]Phase 4124 participants (Anticipated)Interventional2011-10-31Not yet recruiting
Targeted Therapy Selection Based on Tumor Tissue Kinase Activity Profiles for Patients With Advanced Solid Malignancies, an Exploratory Study [NCT01190241]45 participants (Actual)Interventional2010-08-31Terminated
Z-SEA-SIDE: Sirolimus Versus Everolimus Versus Zotarolimus-eluting Stent Assessment in Bifurcated Lesions and Clinical SIgnificance of Residual siDE-branch Stenosis [NCT01200693]Phase 480 participants (Actual)Interventional2008-11-30Completed
A Phase I/II Study of RAD001 Combined With CHOP in Newly Diagnosed Peripheral T-cell Lymphomas [NCT01198665]Phase 1/Phase 246 participants (Actual)Interventional2010-07-31Completed
PI3K/mTOR Pathway Activation Selected Phase II Study of Everolimus (RAD001) With and Without Temozolomide in the Treatment of Adult Patients With Supratentorial Low-Grade Glioma [NCT02023905]Phase 227 participants (Actual)Interventional2014-03-19Terminated(stopped due to Sponsor decision)
Phase I Study of the Combination of Axitinib (AX) Plus Everolimus (EV) in Patients With Malignant Advanced Solid Tumors [NCT01334073]Phase 119 participants (Actual)Interventional2011-03-31Completed
A Phase I Dose Escalation Study of the mTOR Inhibitor Everolimus (RAD001) and Erlotinib Concurrently With Radiation Therapy in the Re-Irradiation Setting for Head and Neck Cancer [NCT01332279]Phase 10 participants (Actual)Interventional2011-04-30Withdrawn(stopped due to Pharmaceutical co. withdrew support. Study was never activated and did not accrue any patients.)
CRAD001X2401: Treatment Plan for an Individual Patient With Recurrent Uterine Papillary Serous Carcinoma (UPSC) With PIK3CA Gene Mutation [NCT03285802]Phase 2/Phase 31 participants (Actual)Interventional2017-09-07Completed
A Six-month, Prospective, Single-center, Pilot Study Determining the Pharmacokinetics and Effectiveness of Twice-daily Tacolimus and Everolimus Regimen Convert to Once-daily Tacrolimus and Everolimus Regimen in Liver Transplant Patients [NCT03256864]Phase 410 participants (Actual)Interventional2016-01-31Completed
mTor-inhibitor (EVERolimus) Based Immunosuppressive Strategies for CNI Minimisation in OLD for Old Renal Transplantation [NCT01028092]Phase 3327 participants (Actual)Interventional2009-03-31Completed
Phase I/II Trial of Everolimus in Combination With Lonafarnib in Progeria [NCT02579044]Phase 1/Phase 280 participants (Anticipated)Interventional2015-12-31Enrolling by invitation
Everolimus-eluting Bioresorbable Vascular Scaffold System in the Treatment of Cardiac Allograft Vasculopathy in Heart Transplant Recipients: A Prospective Multicenter Pilot Study [NCT02377648]Phase 434 participants (Actual)Interventional2015-01-31Completed
An Open-Label Phase I/II Safety and Efficacy Study of Itacitinib In Combination With Everolimus In Subjects With Relapsed/Refractory Classical Hodgkin Lymphoma [NCT03697408]Phase 1/Phase 223 participants (Actual)Interventional2019-02-11Active, not recruiting
Phase I Trial of Ribociclib in Combination With Everolimus and Dexamethasone in Children and Young Adults With Relapsed Acute Lymphoblastic Leukemia [NCT03740334]Phase 145 participants (Anticipated)Interventional2019-01-30Active, not recruiting
A Neoadjuvant Phase II Trial of Aromatase Inhibitors in Combination With Everolimus in Postmenopausal Women With Hormone Receptor Positive/HER2 Negative Breast Cancers With Low and Intermediate Risk (< 25) Oncotype Dx Recurrence Scores [NCT02236572]Phase 217 participants (Actual)Interventional2014-11-29Terminated(stopped due to Phase 2 of recruitment was contingent on 5 of 15 patients responding, which did not occur.)
DUrable Polymer-based STent CHallenge of Promus Element Versus ReSolute Integrity (DUTCH PEERS): Randomized Multicenter Trial in All Comers Population Treated Within Eastern NeThErlands-2 (TWENTE-2) [NCT01331707]Phase 41,811 participants (Actual)Interventional2010-11-30Completed
Biomarkers Predicting Successful Tacrolimus Withdrawal and Everolimus (Zortress) Monotherapy Early After Liver Transplantation [NCT02736227]28 participants (Actual)Interventional2016-03-31Completed
A Randomized, Double-Blind, Placebo-Controlled Phase 2 Study Comparing CB-839 in Combination With Everolimus (CBE) vs. Placebo With Everolimus (PboE) in Patients With Advanced or Metastatic Renal Cell Carcinoma (RCC) [NCT03163667]Phase 269 participants (Actual)Interventional2017-09-06Completed
Treatment of Neuroendocrine Tumors (NETs) With Combination of Everolimus and Radiolabeled Somatostatin Analogue [NCT03629847]Phase 1/Phase 230 participants (Anticipated)Interventional2012-12-31Recruiting
Clinical Evaluation of Everolimus (a Rapamycin Analog) in Restoring Salivary Gland Function to Patients Treated With Radiotherapy for Head and Neck Cancer [NCT03578432]Early Phase 12 participants (Actual)Interventional2018-05-04Terminated(stopped due to UACC-PHX is no longer able to support the study and Novartis is unable to provide drug beyond December 2019.)
To Evaluate Prevention Effect Of Everolimus and Low-dose Tacrolimus in Comparison With Standard-dose Tacrolimus Therapy With Mycophenolic Acid on the New Onset Diabetes Mellitus After Transplantation in the Renal Allograft Recipients [NCT02036554]Phase 4234 participants (Anticipated)Interventional2013-03-31Recruiting
Study of ABSORB Stent in Acute Coronary Syndrome [NCT02071342]300 participants (Anticipated)Observational2013-09-30Recruiting
Everolimus in Association With Very Low-dose Tacrolimus Versus Enteric-coated Mycophenolate Sodium With Low-dose Tacrolimus in de Novo Renal Transplant Recipients - A Prospective Single Center Study [NCT02084446]Phase 4120 participants (Actual)Interventional2012-12-31Completed
DORA: A Phase I and Randomized Phase II Study of Docetaxel and RAD001 (Everolimus) in Advanced/Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck [NCT01313390]Phase 1/Phase 24 participants (Actual)Interventional2009-06-30Terminated(stopped due to Lack of recruitment)
SPIRIT III: A Clinical Evaluation of the Investigational Device XIENCE V® Everolimus Eluting Coronary Stent System (EECSS) in the Treatment of Subjects With de Novo Native Coronary Artery Lesions [NCT00180479]Phase 31,002 participants (Actual)Interventional2005-06-30Completed
Enzalutamide Plus Everolimus in Men With Metastatic Castrate-Resistant Prostate Cancer: A Phase I Study With a Maximum Tolerated Dose Expansion Cohort [NCT02125084]Phase 138 participants (Actual)Interventional2014-10-31Completed
Prospective, Randomized Trial of Belatacept Switch in Renal Transplant Recipients With Delayed Graft Function [NCT02134288]Early Phase 18 participants (Actual)Interventional2014-04-30Terminated(stopped due to Drug unavailability due to a manufacturing transition.)
Phase I/II Study of Induction Chemotherapy With Weekly RAD001, Carboplatin and Paclitaxel in Unresectable or Inoperable Locally Advanced Head and Neck Squamous Cell Carcinoma (HNSCC) [NCT01333085]Phase 1/Phase 249 participants (Actual)Interventional2009-10-31Completed
Differentiating Sirolimus and Everolimus in Combination With Calcineurin Inhibitors in Long-term Maintenance of Kidney Transplant Patients - The Effects on Vascular Endothelial and Kidney Function. The DESIRE Study. [NCT02062892]Phase 40 participants (Actual)Interventional2013-12-31Withdrawn(stopped due to Patients meeting the inclusion/ exclusion criteria did not agree to participate. It became obvious that recruitment goals could not be met.)
The Effect of Everolimus on the Pharmacokinetics of Tacrolimus in Renal Transplant Patients, and the Effect of ABCB1、CYP3A4、CYP3A5、PORGenetic Polymorphism on the Two Drugs [NCT02077556]Phase 414 participants (Actual)Interventional2014-04-30Completed
Phase I Study of Everolimus (RAD001) in Combination With Lenalidomide in Patients With Advanced Solid Malignancies Enriched for Renal Cell Carcinoma [NCT01218555]Phase 144 participants (Actual)Interventional2010-09-09Completed
Efficacy and Safety of Induction Strategies Combined With Low Tacrolimus Exposure in Kidney Transplant Recipients Receiving Everolimus or Sodium Mycophenolate [NCT01354301]Phase 4300 participants (Actual)Interventional2011-05-31Completed
To Compare the Safety and Performance of MeRes100 Sirolimus Eluting BioResorbable Vascular Scaffold System Versus Contemporary DES Platforms in Patients With de Novo Coronary Artery Lesions [NCT05417893]1,872 participants (Anticipated)Interventional2022-10-15Not yet recruiting
Assessment of Vessel Healing After DES Implantation With STEMI, NSTEMI and Stable/Unstable Angina Patients: a Randomized Study Between Everolimus and Biolimus A9-eluting Stents: an Optical Coherence Tomography (OCT) and Intravascular Ultrasound Tissue Cha [NCT01065519]64 participants (Actual)Interventional2009-06-30Completed
A Phase 2 Study of Panobinostat in Combination With Everolimus for Children and Young Adults With Gliomas Harboring H3.3 or H3.1 K27M Mutation [NCT03632317]Phase 20 participants (Actual)Interventional2019-10-31Withdrawn(stopped due to low accrual)
A Phase Ib/II Randomized Study of BI 836845 in Combination With Exemestane and Everolimus Versus Exemestane and Everolimus Alone in Women With Locally Advanced or Metastatic Breast Cancer [NCT02123823]Phase 1164 participants (Actual)Interventional2014-05-15Completed
RIBS IV (Restenosis Intra-stent of Drug-eluting Stents: Paclitaxel-eluting Balloon vs Everolimus-eluting Stent). A Prospective, Multicenter and Randomized Clinical Trial [NCT01239940]Phase 4310 participants (Anticipated)Interventional2010-01-31Recruiting
Phase I Study of Carboplatin, Pegylated Liposomal Doxorubicin (PLD) and Everolimus in Patients With Platinum-Sensitive Epithelial Ovarian, Fallopian Tube or Primary Peritoneal Cancer in First Relapse [NCT01281514]Phase 121 participants (Actual)Interventional2010-12-14Completed
Percutaneous Treatment of LONG Native Coronary Lesions With Drug-Eluting Stent-V:Everolimus-eluting(PROMUS-ELEMENT) vs. Biolimus A9-Eluting(NOBORI) Stents [NCT01186120]Phase 4500 participants (Actual)Interventional2010-08-31Completed
Percutaneous Treatment of LONG Native Coronary Lesions With Drug-Eluting Stent-III: Sirolimus vs. Everolimus-eluting Stent [NCT01078038]Phase 4451 participants (Actual)Interventional2008-06-30Completed
ABSORB PHYSIOLOGY Clinical Investigation: Clinical Evaluation of the Short and Long-Term Effects of the Abbott Vascular Everolimus-Eluting Bioresorbable Vascular Scaffold on Coronary Artery Blood Flow and Physiological Responsiveness [NCT01308346]1 participants (Actual)Interventional2011-11-30Terminated(stopped due to Due to a challenging protocol inclusion/ exclusion criteria, only one subject was enrolled since the trial was initiated in June 2011.)
A Single Arm, Multicenter Phase II Study of Everolimus in Patients With Metastatic Colorectal Adenocarcinoma Whose Cancer Has Progressed Despite Prior Therapy With an Anti-EGFR Antibody (if Appropriate), Bevacizumab, Fluoropyrimidine, Oxaliplatin, and Iri [NCT00419159]Phase 2199 participants (Actual)Interventional2006-12-31Completed
A Randomized, Double-blind Placebo-controlled, Multicenter Phase III Study in Patients With Advanced Carcinoid Tumor Receiving Octreotide Depot and Everolimus 10 mg/Day or Octreotide Depot and Placebo [NCT00412061]Phase 3429 participants (Actual)Interventional2006-12-31Completed
A Phase I/II Trial of Fixed Doses of Daily Gefitinib With Escalating Doses of Daily RAD001 in Advanced Non-Small Cell Lung Cancer [NCT00096486]Phase 1/Phase 274 participants (Actual)Interventional2004-05-31Completed
A Phase I/ II, Non-randomized, Feasibility/ Safety and Efficacy Study of the Combination of Everolimus, Cetuximab and Capecitabine in Patients With Metastatic Pancreatic Cancer [NCT01077986]Phase 1/Phase 235 participants (Actual)Interventional2009-08-31Completed
A Phase I/II, Non-randomized, Multi-center, Dose-escalating, Two-stage Efficacy and Feasibility Study of the Combination of Everolimus and Capecitabine in Patients With Advanced Malignancies [NCT01079702]Phase 1/Phase 235 participants (Anticipated)Interventional2008-04-30Recruiting
Impact and Efficacy of Everolimus Rescue Immunosuppression in the Treatment of Chronic Allograft Dysfunction in Renal Transplant Recipients [NCT01046045]Phase 417 participants (Actual)Interventional2008-04-30Completed
A Phase I Study of Combination Anticancer Therapy of Paclitaxel and Everolimus for Relapsed or Refractory Small Cell Lung Cancer [NCT01079481]Phase 121 participants (Actual)Interventional2009-12-31Completed
Drug Eluting Balloon Angioplasty in Tunisian Population Versus Everolimus Platinum Chrome Stent [NCT05516446]290 participants (Anticipated)Interventional2021-08-25Recruiting
Targeting of Renal Cell Cancer With Specific Inhibitors: A Model for Selective Adaptive Medicine Based on Molecular Alterations [NCT02560012]Phase 24 participants (Actual)Interventional2016-01-04Terminated(stopped due to Loss of laboratory performing molecular analysis)
A Randomized, Open-Label (Formerly Double-Blind), Phase 2 Trial to Assess Safety and Efficacy of Lenvatinib at Two Different Starting Doses (18 mg vs. 14 mg QD) in Combination With Everolimus (5 mg QD) in Renal Cell Carcinoma Following One Prior VEGF-Targ [NCT03173560]Phase 2343 participants (Actual)Interventional2017-08-17Active, not recruiting
Phase II Study of Oral PRednisone 5 mg Bid Plus EVerolimus in Patients With Metastatic Renal Cell Cancer After Failure of Vascular Endothelial Growth Factor Receptor-tyrosine Kinase Inhibitors [NCT02479490]Phase 28 participants (Actual)Interventional2015-09-22Terminated
A Phase 2, Open-label Study to Evaluate the Efficacy and Safety of Single-Agent MLN0128 and the Combination of MLN0128+MLN1117 Compared With Everolimus in the Treatment of Adult Patients With Advanced or Metastatic Clear-Cell Renal Cell Carcinoma That Has [NCT02724020]Phase 296 participants (Actual)Interventional2016-06-30Completed
P2 Efficacy Evaluation of Sabizabulin Monotherapy Versus Active Control for Treatment of ER+HER2- MBC in Patients Who Have Shown Previous Disease Progression on a Nonsteroidal Aromatase Inhibitor, Fulvestrant and CDK 4/6 Inhibitor [NCT05079360]Phase 20 participants (Actual)Interventional2023-03-15Withdrawn(stopped due to Decided to halt and will potentially reopen in the future.)
Autophagy Inhibition to Augment Mammilian Target of Rapamycin (mTOR) Inhibition: A Phase I/II Trial of RAD001 and Hydroxychloroquine (HCQ) in Patients With Previously Treated Renal Cell Carcinoma [NCT01510119]Phase 1/Phase 240 participants (Actual)Interventional2011-09-30Completed
A Randomized, Controlled, Open Label, Two Arms, Exploratory Study to Evaluate the Effect of Everolimus on Histologically Assessed Fibrosis Progression (Ishak-Knodell) in Liver Transplant Recipients With Recurrent Hepatitis C Viral Infection as Compared to [NCT00582738]Phase 243 participants (Actual)Interventional2007-12-31Terminated
[NCT02449538]Phase 210 participants (Actual)Interventional2015-02-28Completed
Clinical Evaluation of a Bioresorbable Sirolimus-eluting Coronary Scaffold in the Treatment of Patients With Denovo Coronary Artery Lesion (NeoVas): Randomized Controlled Trial [NCT02305485]560 participants (Anticipated)Interventional2014-11-30Active, not recruiting
A Multicenter, Randomized Phase ll Study of Letrozole Versus Letrozole Plus Everolimus for Hormone Receptor-PositivePremenopausal Women With Recurrent or Metastatic Breast Cancer on Goserelin Treatment After Progression on Tamoxifen [NCT02313051]Phase 2200 participants (Anticipated)Interventional2014-12-31Not yet recruiting
A Phase II Randomized, Double-Blinded Evaluation of Oral Everolimus (RAD001) Plus Bevacizumab vs. Oral Placebo Plus Bevacizumab in the Treatment of Recurrent or Persistent Epithelial Ovarian, Fallopian Tube or Primary Peritoneal Cancer [NCT00886691]Phase 2150 participants (Actual)Interventional2010-12-27Completed
A Phase I Study of Everolimus (mTOR Inhibitor) and OSI-906 (Dual IGFR and IR Tyrosine Kinase Inhibitor) for the Treatment of Patients With Refractory Metastatic Colorectal Cancer [NCT01154335]Phase 118 participants (Actual)Interventional2010-07-31Completed
A Phase 1b/2a Study Of Palbociclib In Combination With Everolimus And Exemestane In Postmenopausal Women With Estrogen Receptor Positive and HER2 Negative Metastatic Breast Cancer [NCT02871791]Phase 1/Phase 241 participants (Actual)Interventional2016-08-24Completed
Ovarian Suppression Plus Letrozole Plus Everolimus for Hormone Receptor-Positive, Tamoxifen and Ovarian Suppression Pretreated, Premenopausal Women With Recurrent or Metastatic Breast Cancer[LEO] [NCT02344550]Phase 2137 participants (Actual)Interventional2014-01-31Completed
Prospective Multicenter Randomized Openlabel Study to Evaluate the Benefit on Renal Function at 12months Post-transplantation of Immunosuppressive Treatment With Withdrawal of Calcineurin Inhibitor at 3months and Combining Mycophenolate Sodium-Everolimus [NCT02334488]Phase 3329 participants (Actual)Interventional2014-12-11Completed
Phase I/II Study of Weekly Abraxane and RAD001 in Women With Locally Advanced or Metastatic Breast Cancer. A Study of the Cancer Institute of New Jersey Oncology Group (CINJOG) [NCT00934895]Phase 1/Phase 227 participants (Actual)Interventional2009-07-15Terminated(stopped due to Closed early due to slow accrual)
Phase II Single Arm Trial With Combination of Everolimus and Letrozole in Treatment of Platinum Resistant Relapse or Refractory or Persistent Ovarian Cancer/Endometrial Cancer (CRAD001CUS242T) [NCT02188550]Phase 220 participants (Anticipated)Interventional2014-06-30Recruiting
Molecular Determinants for Therapy Response on Renal Cell Carcinoma [NCT02208128]4 participants (Anticipated)Interventional2014-05-31Recruiting
Drug-Eluting Balloon Versus Drug-eluting Stent On de Novo coRonary Artery Disease in Patients With High Bleeding Risk [NCT04885816]5 participants (Actual)Interventional2021-04-20Terminated(stopped due to Slow recruitment)
CERTICOEUR a Secondary Prevention Study of Skin Cancers in Heart Transplant Patients. An Open Labelled Randomized Everolimus vs Calcineurin Inhibitors Multicenter Trial [NCT00799188]Phase 3175 participants (Actual)Interventional2008-10-31Active, not recruiting
A Phase II, Open-Label, Randomized Study of GDC-0980 Versus Everolimus in Patients With Metastatic Renal Cell Carcinoma Who Have Progressed on or Following VEGF-Targeted Therapy [NCT01442090]Phase 285 participants (Actual)Interventional2011-10-31Completed
Phase I Study of Pasireotide (SOM230) in Combination With RAD001 in Patients With Advanced Neuroendocrine Tumors [NCT00804336]Phase 122 participants (Actual)Interventional2008-10-31Completed
Safety and Tolerability of Everolimus as Second-line Treatment in Poorly Differentiated Neuroendocrine Carcinoma / Neuroendocrine Carcinoma G3 (WHO 2010) and Neuroendocrine Tumor G3 - an Investigator Initiated Phase II Study [NCT02113800]Phase 240 participants (Actual)Interventional2015-08-31Completed
Phase I/II Trial of Concurrent RAD001 (Everolimus) With Temozolomide/Radiation Followed by Adjuvant RAD001/Temozolomide in Newly Diagnosed Glioblastoma [NCT01062399]Phase 1/Phase 2279 participants (Actual)Interventional2010-12-31Completed
Pet Imaging as a Biomarker of Everolimus Added Value in Hormone Refractory postmenopausaL Women [NCT02028364]Phase 255 participants (Actual)Interventional2014-01-12Completed
A Single Arm, Phase Ib Study of RAD001 and Sunitinib in Patients With Advanced Renal Cell Carcinoma [NCT00788060]Phase 15 participants (Actual)Interventional2008-10-31Completed
Activity and Safety of Everolimus in Combination With Octreotide LAR and Metformin in Patients With Advanced Pancreatic Well-differentiated Neuroendocrine Tumors (pWDNETs): a Phase II, Open, Monocentric, Prospective Study [NCT02294006]Phase 226 participants (Actual)Interventional2014-06-30Active, not recruiting
Phase I Combination of Pazopanib and Everolimus in PI3KCA Mutation Positive/PTEN Loss Patients With Advanced Solid Tumors Refractory to Standard Therapy [NCT01430572]Phase 162 participants (Actual)Interventional2011-10-07Completed
Intracoronary Analysis of Cardiac Allograft Vasculopathy in Comparison to Coronary Artery Disease by Means of Optical Coherence Tomography [NCT02254668]Phase 4278 participants (Anticipated)Interventional2013-12-31Recruiting
N-of-1 Trial of Actionable Target Identification in Metastatic Cancer for Palliative Systemic Therapy [NCT02142036]Phase 250 participants (Actual)Interventional2014-05-31Completed
Treatment of Canadian Men and Pre/Postmenopausal Women With ER+ Advanced Breast Cancer in the Real-World Setting With Hormone Therapy ± Targeted Therapy [NCT02753686]440 participants (Actual)Observational [Patient Registry]2016-03-15Completed
BIOTRONIK-Safety and Clinical Performance of the Drug Eluting Resorbable Coronary MAGnesium Scaffold System (DREAMS 3G) in the Treatment of Subjects With de Novo Lesions in Native Coronary Arteries: BIOMAG-II: A Randomized Controlled Trial [NCT05540223]1,859 participants (Anticipated)Interventional2024-04-30Not yet recruiting
Biomarker and Tumor Cell Culture-Driven Pilot Trial for Treatment of Recurrent Glioblastoma [NCT05432518]Early Phase 110 participants (Anticipated)Interventional2023-06-27Recruiting
Aflac LL1602 ENCERT: A Phase 1 Trial Using Everolimus in Combination With Nelarabine, Cyclophosphamide and Etoposide in Relapsed T Cell Lymphoblastic Leukemia/Lymphoma [NCT03328104]Phase 18 participants (Actual)Interventional2018-07-24Completed
Everolimus (Afinitor) in Patients With Metastatic Renal Cell Carcinoma Following Progression on Prior Bevacizumab Treatment [NCT02056587]Phase 437 participants (Actual)Interventional2011-11-30Completed
Multi-center, Single Arm, Open-label, Phase 4 STudy to Evaluate the Efficacy and Safety of CertiroBELL® Tablet Plus TAcrolimus in Primary Living Donor Liver Transplant Recipients [NCT04867720]Phase 4112 participants (Anticipated)Interventional2021-03-16Recruiting
A Randomized, Open-Label, Phase 3 Study of Nivolumab (BMS-936558) vs. Everolimus in Subjects With Advanced or Metastatic Clear-Cell Renal Cell Carcinoma Who Have Received Prior Anti-Angiogenic Therapy [NCT01668784]Phase 3821 participants (Actual)Interventional2012-10-09Completed
PhaseII Study of Ribociclib and Everolimus Following Radiotherapy in Pediatric and Young Adult Patients Newly Diagnosed With HGG Including DIPG, Which Harbor Alterations of the Cell Cycle and/or PI3K/mTOR Pathways [NCT05843253]Phase 2100 participants (Anticipated)Interventional2024-01-15Not yet recruiting
Serial Measurements of Molecular and Architectural Responses to Therapy (SMMART) Trial: PRIME [NCT03878524]Phase 12 participants (Actual)Interventional2020-04-01Active, not recruiting
A 12 Month, Multicenter, Randomized, Parallel, Open-label Study, to Evaluate Renal Function and Efficacy of Everolimus With Basiliximab and Cyclosporine Microemulsion Discontinuation at 3 Month Post-transplant Versus Minimization, in de Novo Kidney Transp [NCT00154284]Phase 3114 participants (Actual)Interventional2005-07-31Completed
A Phase I-II Trial of Everolimus and Sorafenib in Patients With Recurrent High-Grade Gliomas [NCT01434602]Phase 1/Phase 286 participants (Actual)Interventional2012-10-02Completed
A Prospective, Randomized, Double-blind, Placebo-Controlled Phase Ⅱ Study to Evaluate the Efficacy of Adjunctive Everolimus Treatment in Patients With Refractory Epilepsy [NCT05613166]Phase 2108 participants (Anticipated)Interventional2022-11-30Recruiting
A Prospective, Active Control Open Label, Multicentre Randomized Clinical Trial for Comparison Between BioMime Sirolimus Eluting Stent of Meril Life Sciences and Xience Everolimus Eluting Stent of Abbott Vascular Inc. to Evaluate Efficacy and Safety in Co [NCT02112981]256 participants (Actual)Interventional2014-11-05Active, not recruiting
Evaluation of Acute Rejection Rates in de Novo Renal Transplant Recipients Following Thymoglobulin Induction, CNI-free, Nulojix (Belatacept)-Based Immunosuppression [NCT02137239]Phase 258 participants (Actual)Interventional2015-12-31Completed
An Open-labelled Multicenter Randomized Study on the Efficacy of Everolimus in Reducing Total Native Kidney Volume in Kidney Transplanted Patients With Autosomal Dominant Polycystic Kidney Disease [NCT02134899]Phase 33 participants (Actual)Interventional2014-10-14Completed
Phase 1 Study of Everolimus in Combination With Brentuximab Vedotin in Patients With Relapsed or Refractory Hodgkin Lymphoma [NCT02254239]Phase 110 participants (Actual)Interventional2016-02-04Terminated(stopped due to funding withdrawal)
Phase I/II Trial With Sorafenib in Combination With RAD001 Administered Orally in Patients With Advanced Solid Tumors, Selected on the Base of Molecular Targets [NCT01226056]Phase 1/Phase 245 participants (Anticipated)Interventional2009-03-31Suspended(stopped due to toxicity (protocol amendment under approval))
Multi-center, Open-label, Prospective, Randomized, Parallel Group Study Investigating a CNI-free Regimen With Enteric-Coated Mycophenolate Sodium (EC-MPS) and Everolimus in Comparison to Standard Therapy With Enteric-Coated Mycophenolate Sodium (EC-MPS) a [NCT00154310]Phase 4300 participants (Actual)Interventional2005-06-30Completed
A Single Arm, Multicenter, Phase II Trial of RAD001 as Monotherapy in the Palliative Treatment of Patients With Locally Advanced or Metastatic Transitional Cell Carcinoma After Failure of Platinum-based Chemotherapy [NCT00714025]Phase 240 participants (Actual)Interventional2008-11-30Completed
A Phase I Study of TS-1, Cisplatin & RAD001 (Everolimus) [NCT01096199]Phase 135 participants InterventionalTerminated
[NCT02616848]Phase 11 participants (Anticipated)Interventional2015-11-30Recruiting
A Prospective Multicenter Post Market Trial to Assess the Safety and Effectiveness of the Firehawk™ Rapamycin Target Eluting Cobalt Chromium Coronary Stent System (Firehawk™ Stent System) for the Treatment of Atherosclerotic Lesion(s) [NCT02520180]1,653 participants (Actual)Interventional2015-12-31Completed
A Phase II Study to Investigate the Efficacy of RAD001 (Afinitor®, Everolimus) in Patients With Irresectable Recurrent or Metastatic Differentiated, Undifferentiated (Anaplastic) and Medullary Thyroid Carcinoma [NCT01118065]Phase 242 participants (Anticipated)Interventional2010-05-31Recruiting
Phase I Study of mTOR Inhibitor RAD001 in Combination With IGF-1R Inhibitor AMG479 for Patients With Advanced Solid Tumors [NCT01122199]Phase 127 participants (Actual)Interventional2010-05-14Completed
A Phase II Study of Lenvatinib in Combination With Everolimus in Patients With Advanced Carcinoid Tumors [NCT03950609]Phase 236 participants (Actual)Interventional2019-07-30Active, not recruiting
EMBER: A Phase 1a/1b Study of LY3484356 Administered as Monotherapy and in Combination With Anticancer Therapies for Patients With ER+ Locally Advanced or Metastatic Breast Cancer and Other Select Non-Breast Cancers [NCT04188548]Phase 1500 participants (Anticipated)Interventional2019-12-10Active, not recruiting
Optical Coherence Tomography Assessment of Intimal Tissue and Malapposition: A Randomized Comparison of the Biolimus A9-eluting and Everolimus-eluting Coronary Stents [NCT01137019]Phase 380 participants (Anticipated)Interventional2010-10-31Active, not recruiting
An Angiogenic Study in Patients With Well/Moderately Differentiated Metastatic Pancreatic Neuroendocrine Tumors Treated With Everolimus [NCT02305810]Phase 254 participants (Actual)Interventional2013-09-30Completed
A 12 Month, Single-center, Open-label, Randomized-controlled Trial to Investigate Efficacy, Safety and Tolerability of Everolimus in Combination With Cyclosporine A and Corticosteroid in de Novo Transplant Recipients of Expanded Criteria Donor Kidneys or [NCT02314312]Phase 348 participants (Anticipated)Interventional2012-01-31Active, not recruiting
Managed Access Program (MAP) Cohort Treatment Plan CRAD001Y2002M to Provide Access to Afinitor for Patients With Advanced Hormone Receptor Positive (HR+) Breast Cancer [NCT05108740]0 participants Expanded AccessNo longer available
Phase IV, Single Arm Study of Safety and Efficacy of Everolimus in Chinese Adults With Tuberous Sclerosis Complex Who Have Renal Angiomyolipoma Not Requiring Immediate Surgery [NCT03525834]Phase 440 participants (Actual)Interventional2018-11-09Completed
Clinical Trial Program of a Medical Instrument Product [NCT01157455]Phase 41,900 participants (Anticipated)Interventional2010-05-31Recruiting
Comparison of Biolimus-eluting Biodegradable Polymer, Everolimus-eluting and Sirolimus-eluting Coronary Stents [NCT01268371]Phase 41,462 participants (Anticipated)Interventional2010-12-31Active, not recruiting
An Open Label, Time-To-Event Continuous Reassessment Method, Phase I/II Study of the Mammalian Target of Rapamycin (mTOR) Inhibitor RAD001 in Combination With Imatinib (Gleevec) in Patients With Chronic Phase Chronic Myeloid Leukemia (CML) With Persistent [NCT01188889]Phase 1/Phase 20 participants (Actual)Interventional2013-09-30Withdrawn(stopped due to Unable to obtain sufficient funding.)
A Pilot Study of Neo-Adjuvant Everolimus in Patients With Advanced Renal Cell Carcinoma Undergoing Definitive Therapy With Radical Nephrectomy, or Cytoreductive Nephrectomy - Analysis of Serum and Tissue Biomarkers [NCT01107509]20 participants (Actual)Interventional2010-10-01Completed
Precision Platform Study of HR+/ HER2-advanced Breast Cancer Based on SNF Typing (A Prospective, Open-label, Multi-center, Phase II Platform Study) [NCT05594095]Phase 2140 participants (Anticipated)Interventional2022-12-30Recruiting
A Single Arm, Multicenter Single Stage Phase II Trial of RAD001 as Monotherapy in the Treatment of Metastatic Non Syndromic Neuro-endocrine Tumors [NCT00688623]Phase 273 participants (Actual)Interventional2009-06-24Completed
A Single-center,Randomized,Open-label,12 Months Study,2 Parallel Group to Compare the Efficacy of Everolimus Combination + Tacrolimus in Regression of Left Ventricular Hypertrophy vs Tacrolimus + MMF in Renal Transplant Patients [NCT03415750]Phase 420 participants (Actual)Interventional2016-11-30Completed
RIBS V (Restenosis Intra-stent of Bare Metal Stents: Paclitaxel-eluting Balloon vs Everolimus-eluting Stent). A Prospective, Multicenter and Randomized Clinical Trial [NCT01239953]Phase 4190 participants (Actual)Interventional2010-01-31Active, not recruiting
Alterations in Cognitive Function and Cerebral Blood Flow After Conversion From Calcineurin Inhibitors to Everolimus [NCT03413722]30 participants (Anticipated)Observational2018-02-01Recruiting
Phase II Study of RAD001monotherapy in Patients With Unresectable Pheochromocytoma or Extra-adrenal Paraganglioma or Non-functioning Carcinoid [NCT01152827]Phase 233 participants (Actual)Interventional2008-07-31Completed
An Open-label Phase I Study Evaluating the Safety and Tolerability of Pasireotide LAR in Combination With Everolimus in Advanced Metastatic NETs - The COOPERATE-1 Study [NCT01263353]Phase 136 participants (Actual)Interventional2010-11-30Completed
Phase II Trial of Lapatinib and RAD-001 for HER2 Positive Metastatic Breast Cancer [NCT01283789]Phase 223 participants (Actual)Interventional2011-02-28Active, not recruiting
A Phase II Study With Tumor Molecular Pharmacodynamic (MPD) Evaluation of Oral mTOR-inhibitor Everolimus (RAD001) 10 mg Daily in Patients Suffering From Classic or Endemic Kaposi's Sarcoma [NCT01412515]Phase 211 participants (Actual)Interventional2008-06-30Terminated(stopped due to results from interim analysis conducted to study interruption)
An Open, Single Centre, Randomised, Parallel Group Study to Investigate Three Different Immunosuppressive Regimens for de Novo Renal Transplant Recipients: A Comparison of a Sirolimus / EC-MPS (Myfortic) / Tacrolimus Regimen, an Everolimus / EC-MPS / Tacr [NCT01183247]Phase 463 participants (Actual)Interventional2008-07-31Completed
A Phase IIIb, Multi-center, Open-label Study of RAD001 in Combination With EXemestane in Post-menopausal Women With EStrogen Receptor Positive, Human Epidermal Growth Factor Receptor 2 Negative Locally Advanced or Metastatic Breast Cancer [NCT03176238]Phase 3235 participants (Actual)Interventional2013-03-29Completed
Safety and Feasibility of Bioabsorbable Everolimus-Eluting Stents for Patients With Internal Pudendal Artery-Related Arteriogenic Erectile Dysfunction (PERFECT-ABSORB) [NCT02492386]Phase 2/Phase 315 participants (Anticipated)Interventional2015-07-31Not yet recruiting
A Phase I and Feasibility Study of Everolimus (RAD001) Plus R-CHOP for New Untreated Diffuse Large B-Cell Lymphoma (DLBCL) [NCT01334502]Phase 126 participants (Actual)Interventional2012-03-31Completed
A Phase I/II Study Investigating the Combination of RAD001 and Rituximab in Patients With Non-Hodgkin's Lymphomas [NCT01567475]Phase 121 participants (Actual)Interventional2011-12-31Completed
89Zr-bevacizumab PET Imaging as Predictive Biomarker for Everolimus Efficacy in Patients With Neuroendocrine Tumors [NCT01338090]14 participants (Actual)Observational2010-04-30Completed
A Safety and Tolerability Study of RAD001 (mTOR Inhibitor) in Combination With Two Dosing Schedules of LBH589B (Histone Deacetylase Inhibitor) in Solid Tumors/ Lymphomas With Enrichment for EBV-Driven Tumors [NCT01341834]Phase 149 participants (Actual)Interventional2010-03-31Active, not recruiting
A Phase 1 Dose Escalation and Expansion Study of AZD9833 Alone or in Combination With Palbociclib or Everolimus in Chinese Patients With Oestrogen Receptor Positive (ER+), Human Epidermal Growth Factor Receptor 2 Negative (HER2-) Metastatic Breast Cancer [NCT04818632]Phase 128 participants (Actual)Interventional2021-11-12Completed
The Rome Trial From Histology to Target: the Road to Personalize Target Therapy and Immunotherapy [NCT04591431]Phase 2400 participants (Actual)Interventional2020-10-07Active, not recruiting
An Open-Label, Single-Arm, Phase II Study of RAD001 in Patients With Mantle Cell Lymphoma Who Are Refractory or Intolerant to Velcade® (Bortezomib) [NCT00702052]Phase 258 participants (Actual)Interventional2008-08-22Completed
Prospective Randomized Trial of Everolimus- and Zotarolimus-eluting Stents for Treatment of Unprotected Left Main Coronary Artery Disease: ISAR-LEFT-MAIN-2 [NCT00598637]Phase 4650 participants (Actual)Interventional2007-12-31Completed
A Randomized Phase III, Double-blind, Placebo-controlled, Multi-center Study to Evaluate the Efficacy and Safety of Everolimus (RAD001) in Adult Patients With Advanced Hepatocellular Carcinoma After Failure of Sorafenib Treatment - The EVOLVE-1 Study [NCT01035229]Phase 3546 participants (Actual)Interventional2010-04-30Completed
A Phase II Study of RAD001 (Everolimus) and Pasireotide (SOM230) LAR in Patients With Advanced Uveal Melanoma [NCT01252251]Phase 214 participants (Actual)Interventional2010-11-30Completed
A Phase I/IIB Study of Combination Weekly Carboplatin, Cetuximab and Dose Escalation of RAD001 in Recurrent Metastatic Squamous Cell Carcinoma of the Head and Neck (SCCHN) [NCT01283334]Phase 1/Phase 220 participants (Actual)Interventional2011-01-31Completed
A Phase Ib/II Study of Cisplatin, Paclitaxel, and RAD001 in Patients With Metastatic Breast Cancer [NCT01031446]Phase 1/Phase 255 participants (Actual)Interventional2009-10-31Completed
NET RETREAT: A Phase II Study of 177 Lutetium-DOTATATE Retreatment vs. Everolimus in Metastatic/Unresectable Midgut NET [NCT05773274]Phase 2100 participants (Anticipated)Interventional2024-03-01Recruiting
Phase II Open Label Study of RAD001 (Everolimus) in Combination With Letrozole in the Treatment of Postmenopausal Women With Locally Advanced or Metastatic, Estrogen Receptor Positive Breast Cancer, After Failure of Tamoxifen and/or Anastrozole and/or Let [NCT01231659]Phase 272 participants (Actual)Interventional2011-08-09Completed
A Randomized Phase III, Double-blind, Placebo-controlled Multicenter Trial of Daily Everolimus in Combination With Trastuzumab and Vinorelbine, in Pretreated Women With HER2/Neu Over-expressing Locally Advanced or Metastatic Breast Cancer. [NCT01007942]Phase 3569 participants (Actual)Interventional2009-10-31Completed
Early vs. Delayed EVERolimus in de Novo HEART Transplant Recipients: Optimization of the Safety/Efficacy Profile (EVERHEART Study) [NCT01017029]Phase 4182 participants (Actual)Interventional2009-09-30Completed
MEGALiT - a Multicenter, Basket and Umbrella Explorative Trial on the Efficacy and Safety of Molecular Profile Selected Commercially Available Targeted Anti-cancer Drugs in Patients With Advanced Cancers Progressive on Standard Therapy [NCT04185831]Phase 2154 participants (Anticipated)Interventional2020-10-20Recruiting
Once-a-day Regimen or Steroid Withdrawal in de Novo Kidney Transplant Recipients Treated With Everolimus, Cyclosporine and Steroids: a 12-month, Prospective, Randomized, Multicenter, Open-label Study. The EVIDENCE Study (EVerolImus Once-a-Day rEgimen With [NCT01023815]Phase 3330 participants (Actual)Interventional2009-04-30Completed
A Single Arm, Multicenter Phase II Trial of RAD001 as Monotherapy in the Treatment of Advanced Papillary Renal Cell Cancer [NCT00688753]Phase 292 participants (Actual)Interventional2009-07-31Completed
A Dose-Escalation Study to Evaluate the Safety and Tolerability of SCH 717454 in Combination With Different Treatment Regimens in Subjects With Advanced Solid Tumors (Phase 1B/2; Protocol No. P04722) [NCT00954512]Phase 1/Phase 215 participants (Actual)Interventional2009-09-25Terminated(stopped due to This study was terminated for business reasons.)
Phase II Study of RAD001 and Bevacizumab in Recurrent Ovarian, Peritoneal, and Fallopian Tube Cancer An Investigator-initiated, Single-institution Trial at Magee-Womens Hospital [NCT01031381]Phase 250 participants (Actual)Interventional2010-09-30Completed
Phase II Clinical Trial of the Combination of RAD001 and Erlotinib in Patients With Recurrent Squamous Cell Carcinoma of the Head and Neck [NCT00942734]Phase 249 participants (Actual)Interventional2009-07-31Completed
A Phase II Study of Everolimus (RAD001) for Children With Chemotherapy and/or Radiation-Refractory Progressive or Recurrent Low-Grade Gliomas [NCT00782626]Phase 223 participants (Actual)Interventional2009-06-30Completed
Efficiency of Everolimus for the Treatment of Kidney Transplanted Patients Presenting a Missing Self-induced Natural Killer Cells Mediated (NK-mediated) Rejection [NCT03955172]20 participants (Anticipated)Interventional2020-12-03Recruiting
Phase I/II Study of RAD001 in Patients With Relapsed or Refractory Acute Myeloid Leukemia, Acute Lymphocytic Leukemia, Chronic Myeloid Leukemia in Blastic-Phase, Agnogenic Myeloid Metaplasia, Chronic Lymphocytic Leukemia, T-Cell Leukemia, or Mantle Cell L [NCT00081874]Phase 1/Phase 229 participants (Actual)Interventional2004-04-30Completed
A Randomized Controlled, Open Labeled, Two Arm, Study of Addition of Everolimus to Standard of Care in Carcinoma Gallbladder [NCT05833815]Phase 2/Phase 356 participants (Anticipated)Interventional2022-11-01Recruiting
A Randomized Double-Blind, Placebo-Controlled Study of Everolimus in Combination With Exemestane in the Treatment of Postmenopausal Women With Estrogen Receptor Positive Locally Advanced or Metastatic Breast Cancer Who Are Refractory to Letrozole or Anast [NCT00863655]Phase 3724 participants (Actual)Interventional2009-06-03Completed
Comparison of the Efficacy of Everolimus-Eluting Versus Sirolimus-Eluting Stent for Coronary Lesions [NCT00698607]Phase 41,466 participants (Anticipated)Interventional2008-06-30Active, not recruiting
Phase 1b Study of ARN 509 Plus Everolimus in Men With Progressive Metastatic Castration-Resistant Prostate Cancer After Treatment With Abiraterone Acetate [NCT02106507]Phase 19 participants (Actual)Interventional2014-04-30Completed
A Single-armed, Open-labeled and Single-centered Phase II Trial of Everolimus in Selected Patients With Metastatic Melanoma: Efficacy and Safety Study [NCT01960829]Phase 260 participants (Anticipated)Interventional2013-09-30Recruiting
Dried Blood SPOT Analysis of Everolimus in Cancer Patients [NCT02809404]75 participants (Actual)Observational2015-06-30Completed
Phase II Multicenter Study Evaluating the Tolerability and Efficacy of RAD001 (Everolimus) in Patients With Relapsed or Metastatic Endometrial Cancer [NCT00870337]Phase 244 participants (Actual)Interventional2008-03-31Completed
A Randomized, Open-label Phase II Trial Comparing Durable Overall Response Rate at 56 Days in Patients With Steroid-resistant Severe Acute GvHD After Allogeneic HSCT Treated With Decidual Stromal Cells or Best Available Treatment [NCT05132166]Phase 250 participants (Anticipated)Interventional2021-12-22Active, not recruiting
Randomized Phase II Trial of Lutetium Lu 177 Dotatate Versus Everolimus in Somatostatin Receptor Positive Bronchial Neuroendocrine Tumors [NCT04665739]Phase 2108 participants (Anticipated)Interventional2023-02-03Recruiting
A Phase I Study of Temozolomide and RAD001C in Patients With Malignant Glioblastoma Multiforme [NCT00387400]Phase 132 participants (Actual)Interventional2007-03-20Completed
A Randomized Phase II Study of Two Different Schedules of RAD001C in Patients With Recurrent/Metastatic Breast Cancer [NCT00255788]Phase 249 participants (Actual)Interventional2005-05-10Completed
Bioresorbable Vascular Scaffold in Patient With ST Elevation Myocardial Infarction: a Randomized Comparison With Everolimus Eluting Stent [NCT02151929]Phase 4100 participants (Actual)Interventional2013-05-31Completed
A National Multicentre Randomized Study Comparing the Early Versus Delayed Administration of Everolimus in de Novo Kidney Transplant Recipients at Risk of Delayed Graft Function [NCT00154297]Phase 4139 participants (Actual)Interventional2005-06-30Completed
Combination Treatment With Everolimus, Letrozole and Trastuzumab in Hormone Receptor and HER2/Neu-Positive Patients With Advanced Metastatic Breast Cancer and Other Solid Tumors: Evaluating Synergy and Overcoming Resistance [NCT02152943]Phase 137 participants (Actual)Interventional2014-07-17Completed
Phase II Study of Everolimus (RAD001) in Metastatic Transitional Cell Carcinoma of the Urothelium [NCT00805129]Phase 246 participants (Actual)Interventional2008-12-05Completed
Comprehensive Imaging and Interventional Therapy Studies for Arteriogenic Erectile Dysfunction and Lower Urinary Tract Symptoms: A Multi-modality, Multi-Specialty Collaborative Study (PERFECT Program) [NCT02178761]300 participants (Anticipated)Interventional2012-10-31Recruiting
Multi-center, Open-label, Randomized Controlled Phase 4 Study to Evaluate the Efficacy and Safety of CertiroBell® Compared With Mycophenolate Mofetil in Primary Living Donor Liver Transplant Recipients. [NCT04471441]Phase 4150 participants (Anticipated)Interventional2020-06-30Recruiting
An Open-label, Randomized Phase 3 Study of MK-6482 Versus Everolimus in Participants With Advanced Renal Cell Carcinoma That Has Progressed After Prior PD-1/L1 and VEGF-Targeted Therapies [NCT04195750]Phase 3736 participants (Anticipated)Interventional2020-02-27Active, not recruiting
MAINtenance Afinitor: A Randomized Trial Comparing Maintenance Aromatase Inhibitors (AIs) + Everolimus (Afinitor) vs AIs in Hormone Receptor Positive (HR+) Metastatic Breast Cancer Patients With Disease Control After First Line Chemotherapy [NCT02511639]Phase 3110 participants (Actual)Interventional2014-07-30Completed
Effects of Everolimus in Left Ventricular Hypertrophy After Conversion From Azathioprine: A Pilot Study [NCT02493465]Phase 450 participants (Anticipated)Interventional2015-07-31Recruiting
6-month, Open-label, Randomized, Multicenter, Prospective, Controlled Study to Evaluate the Efficacy, Safety and Tolerability of Everolimus in de Novo Renal Transplant Recipients Participating in the Eurotransplant Senior Program [NCT00956293]Phase 4207 participants (Actual)Interventional2009-07-31Terminated(stopped due to The study was terminated because the required sample size of 240-260 de novo senior renal transplant patients was not achieved within a reasonable time.)
Protection of Renal Function After Liver Transplant Using Everolimus Monotherapy as the Immunosuppression Regimen [NCT04063865]Phase 314 participants (Actual)Interventional2019-05-09Terminated(stopped due to Study was larger than expected and became a burden to faculty and staff resources.)
Comparison of Titanium-Nitride-Oxide Coated Bio-Active-Stent (Optimax™) to the Drug (Everolimus) -Eluting Stent (Synergy™) in Acute Coronary Syndrome [NCT02049229]Phase 41,800 participants (Anticipated)Interventional2014-01-31Recruiting
Sequential Two-agent Assessment in Renal Cell Carcinoma Therapy: The START Trial [NCT01217931]Phase 2180 participants (Actual)Interventional2011-01-19Active, not recruiting
A Phase Ib Dose-finding Study of BYL719 Plus Everolimus and BYL719 Plus Everolimus Plus Exemestane in Patients With Advanced Solid Tumors, With Dose-expansion Cohorts in Renal Cell Cancer (RCC), Pancreatic Neuroendocrine Tumors (pNETs), and Advanced Breas [NCT02077933]Phase 179 participants (Actual)Interventional2014-05-14Completed
Combination of Everolimus and 177Lu-DOTATATE in the Treatment of Grades 2 and 3 Refractory Meningioma: a Phase IIb Clinical Trial [NCT06126588]Phase 228 participants (Anticipated)Interventional2024-03-01Not yet recruiting
AN OBSERVATIONAL REAL-WORLD STUDY OF THE SYSTEMIC TREATMENT OF WELL-DIFFERENTIATED, UNRESECTABLE OR METASTATIC, PROGRESSIVE PANCREATIC NEUROENDOCRINE TUMOURS (PNET): A STUDY OF MORBIDITY AND MORTALITY AT 2 YEARS [NCT02264665]144 participants (Actual)Observational2015-05-12Completed
XIENCE V® Everolimus Eluting Coronary Stent System USA Post- Approval Study (XIENCE V® USA DAPT Cohort) (XVU-AV DAPT) [NCT01106534]Phase 4870 participants (Actual)Interventional2009-08-31Completed
MicroRNA Expression in Everolimus-based Versus Tacrolimus-based Maintenance Immunosuppressive Regimens in Kidney Transplantation [NCT02091973]50 participants (Anticipated)Interventional2010-06-30Recruiting
Randomized Clinical Comparison of Everolimus-Eluting SYNERGY® and Biolimus-Eluting BioMatrix NeoFlex® Coronary Stents in Non-Selected Patients With Ischemic Heart Disease [NCT02093845]2,800 participants (Actual)Interventional2014-02-10Active, not recruiting
Phosphatidylinositol 3-kinase (PI3K) Pathway Analysis in Tumor Tissue and Circulating DNA to Obtain Further Insight in the Efficacy of Everolimus When Combined With Exemestane: A Side-study Protocol Attached to Standard Treatment With Everolimus and Exeme [NCT02109913]175 participants (Anticipated)Interventional2014-03-31Active, not recruiting
A Phase I Study of LDE225 in Combination With Everolimus in Patients With Advanced Gastroesophageal Adenocarcinoma [NCT02138929]Phase 125 participants (Actual)Interventional2014-11-10Completed
Phase I Study of the Pan-ERBB Inhibitor Neratinib Given in Combination With Everolimus, Palbociclib, or Trametinib in Advanced Cancer Subjects With EGFR Mutation/Amplification, HER2 Mutation/Amplification, or HER3/4 Mutation or KRAS Mutation [NCT03065387]Phase 193 participants (Actual)Interventional2017-10-31Active, not recruiting
Phase 1 Study of Lenvatinib in Combination With Everolimus in Subjects With Unresectable Advanced or Metastatic RCC [NCT02454478]Phase 17 participants (Actual)Interventional2015-07-01Completed
Etiology of the Platelet-Cancer Metastatic Pathway - A Study of Inflammatory Markers, Platelet Characteristics and Metastatic Surrogates in Cancer Patients and Controls [NCT02450175]50 participants (Anticipated)Interventional2014-06-30Recruiting
Comparison of Safety and Efficacy of de Novo Everolimus Plus Low Dose of Cyclosporine With Standard Dose of Cyclosporine Plus Cellcept on CMV and BK Virus Infections Prevention in Renal Transplant Patients [NCT04906304]35 participants (Actual)Interventional2020-01-01Completed
Prospective, Multicenter, Randomized, Open-label, Phase 2, Lasting 12 Weeks, Evaluating the Pharmacodynamics, Efficacy and Safety of Basiliximab in de Novo Adult Renal Transplant Patients at Low Risk Receiving Either a Cumulative Dose of Basiliximab of 40 [NCT01596062]Phase 216 participants (Actual)Interventional2012-03-31Completed
Treg Adoptive Therapy in Subclinical Inflammation in Kidney Transplantation (CTOT-21) [NCT02711826]Phase 1/Phase 214 participants (Anticipated)Interventional2016-09-20Completed
Phase II Study of RAD001 Plus Octreotide Depot in Patients With Metastatic or Unresectable Low Grade Neuroendocrine Carcinoma (Carcinoid, Islet Cell) [NCT00113360]Phase 267 participants (Actual)Interventional2005-01-31Completed
A Phase II Study of RAD001 in Patients With Recurrent Endometrial Cancer [NCT00087685]Phase 235 participants (Actual)Interventional2004-06-30Completed
A Prospective, Open Label, Multi-center Trial of Firehawk™ Coronary Stent System in the Treatment of Coronary Chronic Total Occlusion Lesion(s) by Optical Coherent Tomography (OCT) and Coronary Angiography [NCT03040934]196 participants (Actual)Interventional2017-11-10Active, not recruiting
Precision Treatment of Luminal Advanced Breast Cancer Based on Molecular Subtyping [NCT04355858]Phase 2319 participants (Anticipated)Interventional2020-05-01Recruiting
Open-label Study on Treatment of Primary Aldosteronism With Everolimus [NCT03174171]Phase 212 participants (Actual)Interventional2017-05-22Completed
Proof of Concept for Lenvatinib and Everolimus Prior to Nephrectomy in Eligible Patients With Local and Metastatic Renal Cell Carcinoma (RCC) [NCT03324373]Phase 111 participants (Actual)Interventional2019-03-20Active, not recruiting
Phase II Study of Erlotinib and RAD001 (Everolimus) in Patients With Previously Treated Advanced Pancreatic Cancer [NCT00640978]Phase 216 participants (Actual)Interventional2008-03-31Terminated(stopped due to Significant Adverse Effects - Futility)
Evaluation of RAD001 as Therapy for Patients With Systemic Mastocytosis [NCT00449748]Phase 210 participants (Actual)Interventional2007-04-30Completed
A Phase I Trial to Evaluate Acute and Late Toxicities of Concurrent Treatment With Everolimus (RAD001) and Radio-Hormonotherapy in High-risk Prostate Cancer.(RHOMUS) [NCT00943956]Phase 130 participants (Actual)Interventional2009-01-31Completed
A Phase II Study of Temozolomide and Everolimus (RAD001) Therapy for Metastatic Melanoma [NCT00521001]Phase 249 participants (Actual)Interventional2008-01-31Completed
A Phase II, Single-Arm Study of RAD001 (Everolimus), Letrozole, and Metformin in Patients With Advanced or Recurrent Endometrial Carcinoma [NCT01797523]Phase 262 participants (Actual)Interventional2013-10-07Active, not recruiting
Optical Coherence Tomography Assessment of Gender diVersity In Primary Angioplasty: The OCTAVIA Trial [NCT02577965]Phase 4140 participants (Actual)Interventional2011-03-31Completed
ComparisiOn of Neointimal coVerage betwEen zotaRolimus Eluting Stent and Everolimus Eluting Stent Using Optical Coherence Tomography at 9 Months (COVER OCT) [NCT00894062]Phase 440 participants (Actual)Interventional2008-12-31Completed
COREV : A Multi-center, Randomized, Open-label Study Evaluating the Efficacy on Renal Function of Everolimus in Heart Transplant Recipients With Established Chronic Renal Failure [NCT00716573]Phase 4120 participants (Actual)Interventional2008-09-16Completed
AngiographiC Evaluation of the Everolimus-Eluting Stent in Chronic Total Occlusions - the ACE-CTO Study [NCT01012869]Phase 4100 participants (Anticipated)Interventional2009-11-30Completed
Impact of Cyclosporine or Steroid Withdrawal at 3 Months Post Transplantation on Graft Function, Patient Survival and Cardiovascular Surrogate Markers the First 5 Years After Renal Transplantation. [NCT00903188]Phase 4152 participants (Anticipated)Interventional2008-10-31Recruiting
Phase 1b/3 Global, Randomized, Controlled, Open-label Trial Comparing Treatment With RYZ101 to Standard of Care Therapy in Subjects With Inoperable, Advanced, SSTR+, Well-differentiated GEP-NETs That Have Progressed Following Prior 177Lu-SSA Therapy [NCT05477576]Phase 3288 participants (Anticipated)Interventional2022-03-24Recruiting
A Phase 1b/2 Study of Ibrutinib Combination Therapy in Selected Advanced Gastrointestinal and Genitourinary Tumors [NCT02599324]Phase 1/Phase 2263 participants (Actual)Interventional2015-12-01Completed
A Single Arm, Multi-center Phase II Trial to Evaluate Paclitaxel Plus RAD001 in Urothelial Carcinoma After Failure of Prior Platin-based Chemotherapy [NCT00933374]Phase 228 participants (Actual)Interventional2009-07-31Terminated(stopped due to delayed recruitment)
A Phase II Trial of LEE011 in Combination With Everolimus in the Treatment of Advanced Well Differentiated Neuroendocrine Tumors of Foregut Origin [NCT03070301]Phase 221 participants (Actual)Interventional2017-02-27Active, not recruiting
Phase II Trial of Everolimus (RAD001) in Relapsed/Refractory Lymphoma [NCT00436618]Phase 2277 participants (Actual)Interventional2005-08-31Completed
A Phase Ib/II Study Investigating the Combination of Everolimus With Trastuzumab and Paclitaxel in Patients With HER2-overexpressing Metastatic Breast Cancer [NCT00426556]Phase 188 participants (Actual)Interventional2007-07-31Completed
Phase I Everolimus Dose Finding Study for the Treatment of Stage IV or Recurrent, Non-resectable, Cervical Cancer With Standard Whole Pelvic Radiation Therapy in Combination With Weekly Cisplatin and Daily Everolimus [NCT00967928]Phase 10 participants (Actual)Interventional2009-12-31Withdrawn(stopped due to Lack of enrollment)
A Prospective Multicenter Open-label Randomized Study to Assess Efficacy and Safety of Everolimus in Combination With Cyclosporine Microemulsion Versus Everolimus Without Calcineurine Inhibitor in Combination With Enteric-coated Mycophenolate Sodium (EC-M [NCT00425308]Phase 330 participants (Actual)Interventional2006-10-31Completed
Multiple Ascending Dose (MAD) Phase Ib/II Study of the mTOR Inhibitor (RAD001) in Combination With the IGF-1R Antagonist (R1507) for the Treatment of Patients With Advanced Solid Tumors [NCT00985374]Phase 111 participants (Actual)Interventional2009-11-30Terminated
A Phase I/Ib Dose Escalation and Biomarker Study of Ceritinib (LDK378) in Combination With Everolimus in Patients With Locally Advanced or Metastatic Solid Tumors With an Expansion in NSCLC Characterized by Abnormalities in Anaplastic Lymphoma Kinase (ALK [NCT02321501]Phase 137 participants (Actual)Interventional2016-06-22Active, not recruiting
Efficacy and Safety of Anti-cytomegalovirus Prophylaxis Versus Pre-emptive Approaches With Valganciclovir in Heart Transplant Recipients Treated With Everolimus or Mycophenolate. A Randomized Open-label Study for Prevention of Cardiaca Allograft Vasculopa [NCT00966836]Phase 3100 participants (Anticipated)Interventional2009-04-30Recruiting
A Randomized Study of mTOR Inhibition by RAD001 (Everolimus) in Invasive Breast Cancer Patients After Pre-operative Use of Anthracycline and/or Taxane-based Chemotherapy [NCT01088893]Phase 250 participants (Anticipated)Interventional2009-11-30Recruiting
Evaluation of the Early Conversion From a Calcineurin Inhibitor-based Immunosuppressive Regimen to Everolimus in de Novo Renal Transplant Recipients, a Multicenter Experience [NCT01609673]1 participants (Actual)Interventional2013-03-31Terminated(stopped due to The inclusion of proposed sample turned to be infeasible due to Ethical Non-approval of 2 research sites and another site no transplant activity since 2012.)
Antiangiogenic Treatment of Advanced or Metastatic Hepatocellular Cancer (HCC) - An Open Label, Stratified, Single-arm Phase II Study of Bevacizumab and RAD001 [NCT00775073]Phase 233 participants (Actual)Interventional2008-10-31Completed
A Prospective, Randomized, Single Center Pilot Study Comparing Patient and Graft Survival, Adverse Events and Tolerability of Zortress® (Everolimus) Versus Rapamune® (Sirolimus) in Combination With Low Dose Neoral® (Cyclosporine) Dosed by C2 Monitoring, i [NCT01976390]60 participants (Actual)Interventional2013-10-01Completed
A Phase Ib, Open-label Study to Evaluate RAD001 as Monotherapy Treatment in Chinese Patients With Advanced Pulmonary Neuroendocrine Tumor [NCT01324492]Phase 116 participants (Actual)Interventional2010-12-31Completed
Phase II Trial of RAD001 (Everolimus) in Previously Treated Small Cell Lung Cancer [NCT00374140]Phase 240 participants (Actual)Interventional2006-10-31Completed
Phase I Trial of Carboplatin and Etoposide in Combination With Everolimus (RAD001) in Advanced Solid Tumors, With Emphasis on Small Cell Lung Cancer (SCLC) [NCT00807755]Phase 15 participants (Actual)Interventional2009-03-31Terminated(stopped due to Number of known toxicities observed despite a treatment-naïve population)
A Phase I Dose Finding Study of RAD001 in Combination With Capecitabine and Oxaliplatin (XELOX) in Patients With Advanced Gastric Cancer [NCT01049620]Phase 140 participants (Actual)Interventional2010-02-28Completed
A Two Parts, Biomarker Study to Identify Genetic Aberrations Predictive for Response on Everolimus in Solid Tumors Without Regular Treatment Options (CPCT-03) [NCT01566279]73 participants (Actual)Interventional2012-08-31Completed
Nordic Everolimus (Certican) Trial in Heart and Lung Transplantation: Results at 24 Months [NCT00377962]Phase 4282 participants (Actual)Interventional2005-12-31Completed
Multicentric Study of GOELAMS Phase I Evaluation of RAD001 in Association With Aracytine and Daunorubicine in AML Treatment in Patients Less Than 65 Years in Relapse More Than One Year After First Complete Remission [NCT01074086]Phase 131 participants (Actual)Interventional2008-02-29Completed
A Phase I/II Study of RAD001 and Radiation Therapy in Patients With Brain Metastasis From Non-small Cell Lung Cancer (NSCLC) [NCT00892801]Phase 15 participants (Actual)Interventional2009-05-31Terminated(stopped due to Terminated due to low accrual. Study was closed to accrual prematurely and did not continue on to Phase II.)
Safety and Efficacy of Cemiplimab (PD-1 Blockade) in Selected Organ Transplant Recipients With Advanced Cutaneous Squamous Cell Carcinoma (CONTRAC) [NCT04339062]Phase 112 participants (Anticipated)Interventional2020-07-15Active, not recruiting
A Multi-Centered Randomized Phase II Study Comparison of Single-Agent Carboplatin vs the Combination of Carboplatin and Everolimus for the Treatment of Advanced Triple-Negative Breast Cancer [NCT02531932]Phase 262 participants (Anticipated)Interventional2015-12-16Active, not recruiting
A Single-centre Study of Certican (Everolimus) as Prophylaxis for Graft-versus-Host Disease Following Post-Transplantation Cyclophosphamide After Allogeneic Stem Cell Transplantation [NCT02812940]Phase 219 participants (Actual)Interventional2016-04-30Completed
Comparison of the Vasomotor Function and Myocardial Flow in Patients Treated [NCT02738658]Phase 470 participants (Actual)Interventional2015-03-01Completed
Everolimus (RAD001) in Combination With Intravenous Carboplatin in Taxane- and Anthracycline-pretreated Patients With Progressive Metastatic Breast Cancer [NCT00930475]Phase 1/Phase 254 participants (Anticipated)Interventional2009-02-28Recruiting
Pharmacokinetics of Everolimus and Enteric-Coated Mycophenolatesodium Before and After Withdrawal of Cyclosporine in Stable Renal Transplant Patients [NCT00443937]Phase 415 participants (Anticipated)Interventional2004-01-31Completed
Spirit Small Vessel Registry (SPIRIT SV) [NCT00783796]150 participants (Actual)Interventional2008-10-31Terminated(stopped due to Study stopped at 3 years. Funding for trial withdrawn by sponsor.)
Everolimus-Eluting Stent in the Treatment of Bifurcation Lesions: Comparison of Main Vessel Stent to Main Vessel and Side Branch Stent [NCT00916695]Phase 4332 participants (Anticipated)Interventional2009-06-30Recruiting
An Open-Label Controlled Study of Adjunctive Everolimus (RAD 001) Therapy for Epilepsy in Children With Sturge-Weber Syndrome [NCT01997255]Phase 20 participants (Actual)Interventional2014-04-30Withdrawn(stopped due to Site did not want to pursue study)
PROlonging Dual Antiplatelet Treatment In Patients With Coronary Artery Disease After Graded Stent-induced Intimal Hyperplasia studY [NCT00611286]Phase 41,700 participants (Anticipated)Interventional2006-12-31Completed
Precise Targeted Therapy for Refractory HER2 Positive Advanced Breast Cancer Based on Genome Signature and Drug Sensitivity of PDO Model [NCT05429684]Phase 3120 participants (Anticipated)Interventional2021-01-01Recruiting
A Multicenter, Randomized, Double-blind, Phase II Study to Evaluate the Tolerability of an Induction Dose Escalation of Everolimus in Patients With Metastatic Breast Cancer [NCT02387099]Phase 2156 participants (Actual)Interventional2015-05-31Completed
A Randomized, Double-blind, Multi-center Phase III Study Comparing Everolimus (RAD001) Plus Best Supportive Care Versus Placebo Plus Best Supportive Care in Patients With Advanced Gastric Cancer After Progression on 1 or 2 Prior Systemic Chemotherapy [NCT00879333]Phase 3656 participants (Actual)Interventional2009-07-31Completed
The Landscape of ESR1 Mutations in Asian Estrogen Receptor-positive Metastatic Breast Cancer Patients Detected by Liquid Biopsy and Impact on Hormonal Therapy-based Treatments [NCT04212702]123 participants (Actual)Observational2017-05-22Active, not recruiting
A Multicentre, Open-label, Randomised, Controlled Study of Molecularly Precision Target Therapy Based on Tumor Molecular Profiling With GEMOX in Advanced or Recurrent Extrahepatic Cholangiocarcinoma and Gallbladder Carcinoma [NCT02836847]Phase 2152 participants (Anticipated)Interventional2016-07-31Recruiting
Genomics-Based Target Therapy for Children With Relapsed or Refractory Malignancy [NCT02638428]Phase 290 participants (Anticipated)Interventional2015-12-31Recruiting
Phase IV Study of the Choice of Optimal Strategy for Bifurcation Lesions With Normal Side Branch [NCT00694005]Phase 4504 participants (Actual)Interventional2008-01-31Completed
A Phase 1 Trial of the Combination of Everolimus (RAD001) and Bortezomib (VELCADE) for Relapsed or Refractory Lymphoma [NCT00671112]Phase 130 participants (Actual)Interventional2008-06-30Terminated(stopped due to Treatment ineffective)
Certican® (Everolimus) Against Cytomegalovirus Disease in Renal Transplant Patients [NCT00828503]Phase 240 participants (Anticipated)Interventional2008-12-31Recruiting
Everolimus and Mycophenolate Sodium as GvHD Prophylaxis in Allogeneic Stem Cell Transplantation [NCT00856505]Phase 1/Phase 238 participants (Anticipated)Interventional2008-03-31Recruiting
A Phase 1 Dose Escalation and Expansion Study of AZD9833 Alone or in Combination in Women With ER-positive, HER2-negative Advanced Breast Cancer (SERENA-1) [NCT03616587]Phase 1386 participants (Anticipated)Interventional2018-10-11Recruiting
A Multicenter, Open-label, Phase I Clinical Study to Evaluate the Safety, Pharmacokinetics, and Antitumor Efficacy of SIM0270 Alone or in Combination in Subjects With ER-positive, HER-2 Negative Locally Advanced or Metastatic Breast Cancer [NCT05293964]Phase 1210 participants (Anticipated)Interventional2022-05-18Recruiting
A Randomized Phase 2 Study of MK-2206 in Comparison With Everolimus in Refractory Renal Cell Carcinoma [NCT01239342]Phase 243 participants (Actual)Interventional2011-01-27Terminated
XENERA™1: A Multi-centre, Double-blind, Placebo-controlled, Randomised Phase II Trial to Compare Efficacy of Xentuzumab in Combination With Everolimus and Exemestane Versus Everolimus and Exemestane in Women With HR+ / HER2- Metastatic Breast Cancer and N [NCT03659136]Phase 2103 participants (Actual)Interventional2018-11-28Completed
European Proof-of-Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory Tumors [NCT02813135]Phase 1/Phase 2460 participants (Anticipated)Interventional2016-08-03Recruiting
Phase II Randomized Multicenter Study of Everolimus as Maintenance Therapy for Metastatic Neuroendocrine Carcinoma With Pulmonary or Gastroenteropancreatic Origin [NCT02687958]Phase 230 participants (Actual)Interventional2015-05-31Active, not recruiting
An Exploratory Study Evaluating FDG-PET as a Predictive Marker for mTOR Directed Therapy With RAD001 in Metastatic Renal Cell Cancer [NCT00529802]Phase 260 participants (Actual)Interventional2007-09-30Completed
A Phase I/Ib, Open-label, Multi-center Study of DFF332 as a Single Agent and in Combination With Everolimus or IO Agents in Patients With Advanced/Relapsed ccRCC and Other Malignancies With HIF2α Stabilizing Mutations [NCT04895748]Phase 140 participants (Actual)Interventional2021-11-30Active, not recruiting
An Umbrella Trial Based on Molecular Pathway for Patients With Unresectable Locally Advanced or Metastatic Triple Negative Breast Cancer (FUTURE SUPER) [NCT04395989]Phase 2139 participants (Actual)Interventional2020-07-28Active, not recruiting
A 36 Month Multi-center, Open Label, Randomized, Comparator Study to Evaluate the Efficacy and Safety of Everolimus Immunosuppression Treatment in Liver Transplantation for Hepatocellular Carcinoma Exceeding Milan Criteria [NCT02081755]Phase 4336 participants (Anticipated)Interventional2014-03-31Active, not recruiting
Phase I, Open Label, Dose Escalation Study of the Safety, Tolerability, and Pharmacokinetics of the Combination RAD001 Plus Docetaxel in Patients With Metastatic Breast Cancer [NCT00253318]Phase 115 participants (Actual)Interventional2005-11-01Terminated(stopped due to Toxicity and Lack of Efficacy)
An Open-label, Multicenter Phase II Study to Compare the Efficacy and Safety of RAD001 as First-line Followed by Second-line Sunitinib Versus Sunitinib as First-line Followed by Second-line RAD001 in the Treatment of Patients With Metastatic Renal Cell Ca [NCT00903175]Phase 2471 participants (Actual)Interventional2009-10-31Completed
Multi-center, Open-label, Prospective, Randomized, Parallel Group, Long-term Study Investigating a Standard Regimen in de Novo Kidney Transplant Patients Versus a CNI-free Regimen and a CNI-low Dose Regimen [NCT00514514]Phase 3802 participants (Actual)Interventional2007-07-31Completed
A Phase I Trial of the IGF-1R Antibody AMG 479 in Combination With Everolimus (RAD001) and Panitumumab in Patients With Advanced Cancer (The RAP Trial) [NCT01061788]Phase 143 participants (Actual)Interventional2010-04-30Completed
A Randomized Double-blind Phase III Study of RAD001 10 mg/d Plus Best Supportive Care Versus Placebo Plus Best Supportive Care in the Treatment of Patients With Advanced Pancreatic Neuroendocrine Tumor (NET) [NCT00510068]Phase 3410 participants (Actual)Interventional2007-07-31Completed
Phase I Trial of Everolimus, Gemcitabine, and Cisplatin for Patients With Solid Tumors Refractory to Standard Therapy [NCT00949949]Phase 138 participants (Actual)Interventional2009-09-30Completed
A 24-month, Multi-center, Randomized, Open-label, Non-inferiority Study of Efficacy and Safety Comparing Two Exposures of Concentration-controlled Everolimus With Reduced Cyclosporine Versus 3.0 g Mycophenolate Mofetil With Standard Dose Cyclosporine in d [NCT00300274]Phase 3721 participants (Actual)Interventional2006-01-31Completed
Phase III Placebo-Controlled Trial to Evaluate Dexamethasone Use for Everolimus-Induced Oral Stomatitis: Prevention Versus Early Treatment Approaches: MIST (My Individualized Stomatitis Treatment) [NCT03839940]Phase 339 participants (Actual)Interventional2019-02-15Terminated(stopped due to Severe lack of accrual)
A Phase I/II Study of RAD001 With Docetaxel in the Treatment of Metastatic, Androgen Independent Prostate Cancer [NCT00459186]Phase 1/Phase 219 participants (Actual)Interventional2005-11-30Completed
A Phase I Study Evaluating the Combination of the Deacetylase Inhibitor, LBH589 Plus the mTOR Inhibitor RAD001, in Relapsed and Refractory Adult Patients With Lymphoma [NCT00962507]Phase 111 participants (Actual)Interventional2009-07-31Completed
An Open-label, Single-dose Study to Assess the Pharmacokinetics of Oral Everolimus (Afinitor®) in Subjects With Impaired Hepatic Function [NCT00968591]Phase 134 participants (Actual)Interventional2009-11-30Completed
An Open Label, Stratified, Single-arm Phase II Study of Everolimus in Patients With Advanced Pancreatic Neuroendocrine Tumor (NET) After Failure of Cytotoxic Chemotherapy [NCT00363051]Phase 2160 participants (Actual)Interventional2006-06-30Completed
Post-Approval Study of the PROMUS PREMIERTM Everolimus-Eluting Platinum Chromium Coronary Stent System in China [NCT03273023]2,059 participants (Actual)Observational [Patient Registry]2018-01-11Active, not recruiting
A Randomized, Double-blind, Placebo-controlled Study of Everolimus in the Treatment of Patients With Subependymal Giant Cell Astrocytomas (SEGA) Associated With Tuberous Sclerosis Complex (TSC) [NCT00789828]Phase 3117 participants (Actual)Interventional2009-08-31Completed
A Randomized Controlled Trial of Everolimus-eluting Stents and Paclitaxel-eluting Stents for Coronary Revascularization in Daily Practice: The COMPARE Trial [NCT01016041]Phase 31,800 participants (Actual)Interventional2007-02-28Completed
Open Label Randomized Clinical Trial of Standard Neoadjuvant Chemotherapy (Paclitaxel Followed by FEC) Versus the Combination of Paclitaxel and RAD001 Followed by FEC in Women With Triple Receptor-Negative Breast Cancer (CRAD001C24101) [NCT00499603]Phase 262 participants (Actual)Interventional2007-07-31Active, not recruiting
A Phase I Study of BKM120 and Everolimus in Advanced Solid Malignancies [NCT01470209]Phase 143 participants (Actual)Interventional2012-01-31Completed
A Clinical Evaluation of the Bioabsorbable Everolimus Eluting Coronary Stent System (BVS EECSS) in the Treatment of Patients With de Novo Native Coronary Artery Lesions. [NCT00856856]101 participants (Actual)Interventional2009-03-31Completed
A Phase II Trial of RAD001 in Triple Negative Metastatic Breast Cancer [NCT00827567]Phase 26 participants (Actual)Interventional2009-04-30Terminated(stopped due to Slow accrual)
A Phase I Trial of the mTOR Inhibitor RAD001 in Combination With VEGF Receptor Tyrosine Kinase Inhibitor PTK787/ZK 222584 in Patients With Advanced Solid Tumors [NCT00655655]Phase 196 participants (Anticipated)Interventional2004-12-31Completed
A Phase Ib Study Investigating the Combination of RAD001 With Cisplatin and Etoposide in Patients With Extensive-stage Small-cell Lung Cancer Not Previously Treated With Chemotherapy [NCT00466466]Phase 120 participants (Actual)Interventional2007-04-30Completed
A Randomized Comparison of the Supralimus® Stent With the Xience V™ Stent in the Treatment of Patients With de Novo Native Coronary Artery Lesions [NCT00917163]Phase 413 participants (Actual)Interventional2009-07-31Terminated(stopped due to due to unavoidable circumstances relating to logistic issues and regulatory processes in various countries causing unacceptable delays.)
A Two-step Phase 1 Study Investigating the Combination of RAD001 With Carboplatin, Paclitaxel and Bevacizumab in Non-small-cell Lung Cancer (NSCLC) Patients Not Treated Previously With Systemic Therapy [NCT00457119]Phase 169 participants (Actual)Interventional2007-02-28Completed
CLEVER Pilot Trial: A Phase II Pilot Trial of HydroxyChLoroquine, EVErolimus or the Combination for Prevention of Recurrent Breast Cancer [NCT03032406]Phase 254 participants (Actual)Interventional2017-01-23Active, not recruiting
A Phase I Trial of RAD001 (Everolimus) and Avastin(R) (Bevacizumab) in Children With Recurrent Solid Tumors [NCT00756340]Phase 116 participants (Actual)Interventional2008-07-31Completed
A Randomized, Double-blind, Placebo-controlled Study of RAD0001 in the Treatment of Angiomyolipoma in Patients With Either Tuberous Sclerosis Complex (TSC) or Sporadic Lymphangioleiomyomatosis (LAM) [NCT00790400]Phase 3118 participants (Actual)Interventional2009-04-30Completed
A Randomized Phase III, Double-Blind, Placebo-Controlled Multicenter Trial of Everolimus in Combination With Trastuzumab and Paclitaxel, as First Line Therapy in Women With HER2 Positive Locally Advanced or Metastatic Breast Cancer [NCT00876395]Phase 3719 participants (Actual)Interventional2009-09-10Completed
Phase IV Study of Optimal Stenting Strategy For True Bifurcation Lesions [NCT00693251]Phase 4420 participants (Actual)Interventional2008-01-31Completed
Everolimus in Combination With Rituximab for Relapsed/Refractory Diffuse Large B Cell Lymphoma [NCT00869999]Phase 226 participants (Actual)Interventional2009-05-31Completed
Long Term Follow Up for RAD001 Therapy of Angiomyolipomata in Patients With Tuberous Sclerosis Complex and Sporadic Lymphangioleiomyomatosis [NCT00792766]Phase 1/Phase 220 participants (Actual)Interventional2008-12-31Completed
Phase II Trial of mTOR Inhibitor, Everolimus (RAD001), in Malignant Pleural Mesothelioma (MPM) [NCT00770120]Phase 261 participants (Actual)Interventional2008-12-31Completed
A Phase II Trial of RAD001 Plus Bevacizumab in the Treatment of Patients With Metastatic Melanoma [NCT00591734]Phase 257 participants (Actual)Interventional2008-01-31Completed
Open Label Phase II Study of Everolimus (RAD001) in Patients With Segmental Overgrowth Syndrome [NCT02569125]Phase 20 participants (Actual)Interventional2016-01-31Withdrawn(stopped due to Change in supply of study medication)
A Multi-center, Randomized, Open-label, Parallel Group Study Investigating the Renal Tolerability, Efficacy and Safety of a CNI-free Regimen (Everolimus and MPA) Versus a CNI-regimen With Everolimus in Heart Transplant Recipients [NCT00862979]Phase 4162 participants (Actual)Interventional2009-02-24Completed
A Randomized, Open-label, Multi-center Phase II Study to Compare Bevacizumab Plus RAD001 Versus Interferon Alfa-2a Plus Bevacizumab for the First-line Treatment of Patients With Metastatic Clear Cell Carcinoma of the Kidney [NCT00719264]Phase 2365 participants (Actual)Interventional2008-11-12Completed
Multicenter, Triple-arm, Single-stage, Phase II Trial to Determine the Preliminary Efficacy and Safety of RAD001 in Patients With Histological Evidence of Progressive or Metastatic Bone or Soft Tissue Sarcomas [NCT00767819]Phase 271 participants (Actual)Interventional2008-03-31Completed
A Phase II Study of Combined Fulvestrant (Faslodex) and Everolimus in Advanced/Metastatic Breast Cancer After Aromatase Inhibitor Failure [NCT00570921]Phase 233 participants (Actual)Interventional2008-04-30Completed
Randomized Comparison Between Sirolimus-eluting and Everolimus-eluting Coronary Stents in Chronic Coronary Occlusions [NCT00793221]Phase 3207 participants (Actual)Interventional2008-11-30Completed
Phase 1 Study of the Combination of CM082 With Everolimus in Patients With Metastatic Renal Cell Carcinoma (RCC): Safety, Tolerability, Pharmacokinetics, and Preliminary Efficacy [NCT02577458]Phase 118 participants (Anticipated)Interventional2015-09-30Recruiting
Phase I/II Study of Hyper-CVAD Plus RAD001 (Everolimus) for Patients With Relapsed or Refractory Acute Lymphocytic Leukemia (ALL) [NCT00968253]Phase 1/Phase 224 participants (Actual)Interventional2009-11-30Completed
Phase II Trial of EVEROLIMUS ± Trastuzumab in Hormone-Refractory Metastatic Breast Cancer [NCT00912340]Phase 270 participants (Actual)Interventional2009-05-31Completed
A Phase 2b Randomized Trial of Autologous Dendritic Cell Immunotherapy (CMN-001) Plus Standard Treatment of Advanced Renal Cell Carcinoma [NCT04203901]Phase 216 participants (Actual)Interventional2020-07-22Terminated(stopped due to Strategic corporate decision)
A Phase 0/II Study of Ribociclib (LEE011) in Combination With Everolimus in Preoperative Rb-Intact Recurrent High-Grade Glioma Patients Scheduled for Resection to Evaluate Central Nervous System (CNS) Penetration [NCT03834740]Early Phase 127 participants (Actual)Interventional2018-12-21Completed
A Phase I Study of Capecitabine, Oxaliplatin, Bevacizumab, and RAD001 (XELOX-A-Ev) for Subjects With Advanced Solid Tumors [NCT00849550]Phase 132 participants (Actual)Interventional2009-07-31Completed
A Phase I Study of Ribociclib and Everolimus Following Radiation Therapy in Children With Newly Diagnosed Non-biopsied Diffuse Pontine Gliomas (DIPG) and RB+ Biopsied DIPG and High Grade Gliomas (HGG) [NCT03355794]Phase 119 participants (Actual)Interventional2017-11-14Completed
A Phase II Study Evaluating The Efficacy And Tolerability Of Everolimus (RAD001) In Combination With Trastuzumab And Vinorelbine In The Treatment Of Progressive HER2-Positive Breast Cancer Brain Metastases [NCT01305941]Phase 232 participants (Actual)Interventional2011-09-30Completed
An Open-Label Phase I Study of the Safety of and Efficacy of RAD001 in Combination With Lenalidomide in the Treatment of Subjects With Relapsed and Relapsed/Refractory Multiple Myeloma [NCT00729638]Phase 128 participants (Actual)Interventional2008-06-30Completed
Harmonizing Optimal Strategy for Treatment of Coronary Atherosclerotic Lesions- Registry-based Study on the Effect and Safety of Xience Xpedition™/Alpine™, Everolimus-eluting Stent for Coronary Atherosclerotic Lesions: HOST-Alpine Registry [NCT02845804]1,533 participants (Anticipated)Observational [Patient Registry]2015-07-31Recruiting
An Open-label, Multi-center Phase 2 Study to Evaluate Everolimus as Monotherapy Treatment for Patients With Metastatic Recurrent and/or Unresectable Renal Cell Carcinoma (EVERMORE). [NCT01206764]Phase 4143 participants (Actual)Interventional2009-11-11Completed
An Extension to a 12-month, Open-label, Randomised, Multicenter, Sequential Cohort, Dose Finding Study to Evaluate the Efficacy, Safety and Tolerability of Oral AEB071 Versus Cyclosporine in Combination With Everolimus, Basiliximab and Corticosteroids in [NCT00820911]Phase 2175 participants (Actual)Interventional2008-09-30Completed
Extension Study to the Multicenter, Open-label, Randomized, Controlled Study CRAD001H2304 to Evaluate the Long-term Efficacy and Safety of Concentration-controlled Everolimus in Liver Transplant Recipient [NCT01150097]Phase 3284 participants (Actual)Interventional2010-03-31Completed
A 24 Month, Multicenter, Open-label, Randomized, Controlled Study to Evaluate the Efficacy and Safety of Concentration-controlled Everolimus to Eliminate or to Reduce Tacrolimus Compared to Tacrolimus in de Novo Liver Transplant Recipients [NCT00622869]Phase 3719 participants (Actual)Interventional2008-01-31Completed
A Clinical Evaluation of Everolimus Eluting Coronary Stents in the Treatment of Patients With ST-segment Elevation Myocardial Infarction.EXAMINATION Study [NCT00828087]1,504 participants (Actual)Interventional2008-12-31Completed
A Phase I Study of the Weekly Oral RAD001 in Combination With Oral Topotecan in Patients With Advanced or Recurrent Endometrial Cancers [NCT00703807]Phase 110 participants (Actual)Interventional2008-12-31Completed
Phase I/II Study of Combination Everolimus (RAD001), and Rituximab (Rituxan), OR Everolimus, Bortezomib (Velcade, PS-341), and Rituximab in Patients With Relapsed and/or Relapsed/Refractory Waldenstrom's Macroglobulinemia [NCT01125293]Phase 1/Phase 246 participants (Actual)Interventional2010-04-30Terminated(stopped due to Per protocol, Phase II is to be terminated early if a less than 5% of the Phase II participants observe a Very Good Partial Response or Better in the first 23 participants enrolled to Phase II.)
Phase I Dose Escalation of Gleevec in Combination With RAD001 Plus Hydroxyurea for Patients With Recurrent Malignant Glioma [NCT00613132]Phase 178 participants (Actual)Interventional2005-05-31Completed
A 24-month, Multi-center, Open-label, Randomized, Controlled Trial to Investigate Efficacy, Safety and Evolution of Cardiovascular Parameters in de Novo Renal Transplant Recipients After Early Calcineurin Inhibitor to Everolimus Conversion [NCT01114529]Phase 3828 participants (Actual)Interventional2010-08-09Completed
A Phase I/II Study of the Histone Deacetylase (HDAC) Inhibitor LBH589 (Panobinostat) in Combination With mTOR Inhibitor RAD001 (Everolimus) in Patients With Relapsed Multiple Myeloma or Lymphoma [NCT00918333]Phase 1/Phase 2124 participants (Actual)Interventional2009-06-30Completed
A Phase I Dose Escalation Study of RAD001 Administered in Patients With Relapsed or Refractory Non-Hodgkin's Lymphoma [NCT00622258]Phase 113 participants (Actual)Interventional2008-03-31Completed
Phase I Study of Sorafenib in Combination With RAD001 in Patients With Advanced Neuroendocrine Tumors [NCT00942682]Phase 121 participants (Actual)Interventional2009-07-31Completed
An Open-Label, Single Arm, Pilot Study of the Renal Safety of Everolimus in Addition to Neoral® in Cardiac Transplant Recipients With Established Allograft Vasculopathy [NCT00097968]Phase 30 participants Interventional2004-08-31Completed
A Phase I Study of Radiation Therapy (IMRT) + RAD001 (Everolimus) + Cisplatin for Patients With Head and Neck Cancer [NCT00858663]Phase 113 participants (Actual)Interventional2009-03-31Completed
A Randomized, Phase 2 Study to Assess the Safety and Efficacy of CRLX101 in Combination With Bevacizumab in Patients With Metastatic Renal Cell Carcinoma (RCC) Versus Standard of Care (SOC) (Investigator's Choice) [NCT02187302]Phase 2115 participants (Actual)Interventional2014-07-31Completed
Observational Study Evaluating Efficacy, Tolerability and Treatment Algorithm of Advanced Renal Cell Cancer Patients Under Afinitor Treatment [NCT01390519]30 participants (Actual)Observational2011-06-30Completed
EndothelIal progeNitor Cell Capture steNt With 1-mOnth Dual Antiplatelet Therapy Versus eVerolimus-eluting Stent With stAndard 12-month Dual anTIplatelet Therapy in Elderly (≥ 70 Year) With Stable corONary Artery Disease - INNOVATION Trial [NCT01394848]Phase 41 participants (Actual)Interventional2011-10-31Terminated(stopped due to Previous other study including EPC capture stent raised the issue of safety (significant high incidence of instent restenosis))
A Multicenter, Open-labeled, Randomized Controlled Trial Comparing Three 2nd Generation Drug-Eluting Stents in Real-World Practice [NCT01397175]1,960 participants (Actual)Interventional2013-01-16Terminated(stopped due to Slow enrollment)
The DESTINY Trial: a Prospective Randomized Multicenter Trial Comparing the Implant of a Drug Eluting Stent (XIENCE V, Abbott Vascular) vs. a Bare Metal Stent (MULTILINK VISION, Abbott Vascular) in the Critically Ischemic Lower Leg [NCT00510393]Phase 2140 participants (Actual)Interventional2008-03-31Completed
Efficacy and Safety of Certican® in Combination With Myfortic® in Adult Renal Allograft Recipients Following Calcineurin Inhibitor Withdrawal at Week 16 Compared to Patients Who Are Maintained on Tacrolimus and Myfortic® [NCT01399242]Phase 440 participants (Anticipated)Interventional2011-08-31Not yet recruiting
A Randomized Phase II Study to Explore the Efficacy and Feasibility of Upfront Bi-monthly Rotations Between Everolimus and Pazopanib in Patients With Advanced or Metastatic Clear Cell Renal Cancer [NCT01408004]Phase 2101 participants (Actual)Interventional2011-11-30Completed
A Therapeutic Exploratory Study to Determine the Efficacy and Safety of Calcineurin-Inhibitor-Free De-novo Immunosuppression After Liver Transplantation [NCT00890253]Phase 229 participants (Anticipated)Interventional2010-01-31Recruiting
Randomized Comparison of Biolimus-Eluting and Everolimus-Eluting Stents With Optical Coherence Tomography Guided Stent Implantation in ST Elevation Myocardial Infarction.A 9-Month Angiographic and Optical Coherence Tomography Follow-up. [NCT00888758]Phase 4400 participants (Anticipated)Interventional2009-05-31Not yet recruiting
Multi-center, Open-label, Prospective, Randomized, Parallel Group Study Investigating a CNI-free Regimen With Enteric-coated Mycophenolate Sodium and Everolimus in Comparison to Standard Therapy With Enteric-coated Mycophenolate Sodium and Ciclosporin Mic [NCT00332839]Phase 493 participants (Actual)Interventional2005-11-30Terminated(stopped due to The trial was terminated early due to slow enrollment. It was determined that the planned sample size of 300 could not be achieved.)
Randomized Phase I/II of Rapamycin Analog, RAD001, in Advanced Hepatocellular Carcinoma - With a Pharmacokinetic Study of RAD001 [NCT00390195]Phase 1/Phase 2134 participants (Anticipated)Interventional2006-10-31Recruiting
Prospective and Randomized Study to Evaluate the Effect of Everolimus in the Clinical and Intra-Cardiac Ecography Progression of Heart Graft Vascular Illness. [NCT00695344]Phase 452 participants (Actual)Interventional2006-01-31Active, not recruiting
A Phase II Trial Using RAD001 for Patients With Radioiodine Refractory Thyroid Cancer [NCT00936858]Phase 250 participants (Actual)Interventional2009-07-31Completed
A Phase II Study of Carboplatin (CBDCA), Paclitaxel (TAXOL), and Everolimus (RAD001) in Previously Untreated Patients With Measurable Disease With Cancer of Unknown Primary (CUP) [NCT00936702]Phase 246 participants (Actual)Interventional2009-09-30Completed
A Phase II Neo-Adjuvant Study of Cisplatin, Paclitaxel With or Without RAD001 in Patients With Triple-negative Locally Advanced Breast Cancer. [NCT00930930]Phase 2145 participants (Actual)Interventional2009-06-30Completed
SEA-SIDE: Sirolimus Versus Everolimus-eluting Stent Randomized Assessment in Bifurcated Lesions and Clinical SIgnificance of Residual siDE-branch Stenosis [NCT00697372]Phase 4150 participants (Actual)Interventional2007-09-30Completed
Everolimus-Eluting Bioresorbable Scaffolds Versus Everolimus-Eluting Metallic Stents for Diffuse Long Coronary Artery Disease [NCT02831205]Phase 4800 participants (Anticipated)Interventional2016-07-31Terminated(stopped due to Due to current BVS safety issue)
SORAVE-Sorafenib and Everolimus in Solid Tumors. A Phase I Clinical Trial to Evaluate the Safety of Combined Sorafenib and Everolimus Treatment in Patients With Relapsed Solid Tumors [NCT00933777]Phase 136 participants (Actual)Interventional2009-07-31Completed
An Open-label, Single-arm Phase II Study of RAD001 in Patients With Relapsed/Refractory Classical Hodgkin Lymphoma [NCT01022996]Phase 257 participants (Actual)Interventional2009-12-31Completed
XIENCE V® Everolimus Eluting Coronary Stent System (EECSS) USA Post-Approval Study [NCT00676520]8,053 participants (Actual)Observational2008-07-31Completed
An Open-label Phase I/II (Proof of Concept) Trial of an Combination of Nilotinib (AMN 107) and RAD001 in Patients With Acute Myeloid Leukemia [NCT00762632]Phase 1/Phase 240 participants (Anticipated)Interventional2007-12-31Completed
AERO: Adjuvant EveRolimus Outcomes in Laryngotracheal Stenosis [NCT05153668]Early Phase 120 participants (Anticipated)Interventional2022-09-30Active, not recruiting
A 12-month, Multicenter, Randomized, Open-label Study to Investigate Efficacy and Safety of Concentration Controlled Everolimus With Reduced Dose Cyclosporine A Versus Mycophenolate Mofetil With Standard Dose Cyclosporine A in de Novo Renal Transplant Adu [NCT00658320]Phase 3122 participants (Actual)Interventional2008-02-29Completed
A Randomized, Double-blind, Placebo-controlled, Multi-center Phase III Study of RAD001 Adjuvant Therapy in Poor Risk Patients With Diffuse Large B-Cell Lymphoma (DLBCL) of RAD001 Versus Matching Placebo After Patients Have Achieved Complete Response With [NCT00790036]Phase 3742 participants (Actual)Interventional2009-07-24Completed
A Phase Ib Trial of LEE011 in Combination With Everolimus (RAD001) and Exemestane in the Treatment of Postmenopausal Women With Hormone Receptor Positive, HER2 Negative Locally Advanced or Metastatic Breast Cancer [NCT01857193]Phase 1132 participants (Actual)Interventional2013-09-06Completed
Randomized Comparison of Coronary Artery Bypass Surgery and Everolimus-Eluting Stent Implantation in the Treatment of Patients With Multivessel Coronary Artery Disease (BEST) [NCT00997828]888 participants (Actual)Interventional2008-07-28Terminated(stopped due to Problem with recruitment of subjects)
A Phase 1 Open Label/ Phase 2 Randomized, Double-blind, Multicenter Study Investigating the Combination of RAD001 and Sorafenib (Nexavar®) in Patients With Advanced Hepatocellular Carcinoma [NCT00828594]Phase 1130 participants (Actual)Interventional2008-12-31Terminated
Neoadjuvant Everolimus (RAD001)for Advanced RCC Before Cytoreductive Nephrectomy, With Correlative Tumor Studies (Protocol #: 06-08-20-01) [NCT00831480]Phase 215 participants (Actual)Interventional2011-04-30Terminated(stopped due to Difficulty in accrual and by the order of the cancer center.)
An Open-Label, Dose-Escalation, Phase IB II Study to Investigate the Safety, Pharmacokinetics, Pharmacodynamics and Clinical Activity of the MEK Inhibitor GSK1120212 in Combination With Oral Everolimus in Subjects With Solid Tumors [NCT00955773]Phase 164 participants (Actual)Interventional2009-08-17Completed
A Phase II Trial of Short-Term Everolimus (RAD001) to Predict Response in Women With Operable Breast Cancer [NCT00855114]Phase 20 participants (Actual)Interventional2008-07-31Withdrawn(stopped due to Withdrawn due to no accrual)
Phase II Study of RAD001 in Advanced Cholangiocarcinoma [NCT00973713]Phase 227 participants (Anticipated)Interventional2009-09-30Recruiting
Rapalogues for Autism Phenotype in TSC: A Feasibility Study [NCT01929642]Phase 23 participants (Actual)Interventional2013-07-31Completed
Everolimus First-line Therapy in Non-rapidly Progressive Castration Resistant Prostate Cancer (CRPC). A Multicenter Phase II Trial. [NCT00976755]Phase 237 participants (Actual)Interventional2009-09-14Completed
Everolimus for the Treatment of Uveitis Unresponsive to Cyclosporine A [NCT00803816]Phase 212 participants (Actual)Interventional2007-11-30Completed
Master Protocol for Mantle Cell Lymphoma A Multicenter Phase II Trial Testing Everolimus (RAD001) for the Treatment of Patients With Relapsed or Therapy Resistant Mantle Cell Lymphoma [NCT00516412]Phase 235 participants (Actual)Interventional2007-08-31Completed
A Single Arm, Prospective, Open-label, Pilot Study to Assess Effects of the Switch From Sirolimus to Everolimus in Stable Maintenance Renal Transplant Patients Receiving a Calcineurin Inhibitor Free Regimen [NCT00170820]Phase 420 participants Interventional2005-02-28Completed
A One-year Multicenter, Randomized, Open-label Study of the Safety and Efficacy of Everolimus Versus Mycophenolate Mofetil in Combination With Reduced Dose Cyclosporine Microemulsion in Maintenance Heart Transplant Recipients [NCT00170859]Phase 40 participants Interventional2004-08-31Completed
ABILITY Diabetes Global [NCT04236609]3,050 participants (Actual)Interventional2020-06-15Active, not recruiting
A Facilitated Access Program to Provide Everolimus (RAD) Maintenance for Patients Completing Therapy in RAD Trials in Solid Organ Transplantation [NCT00149981]0 participants Expanded AccessNo longer available
Phase I Clinical Trial of RAD001 in Combination With CP-751,871 in Patients With Advanced Sarcomas and Other Malignant Neoplasms [NCT00927966]Phase 121 participants (Actual)Interventional2009-07-31Completed
Phase I Study Evaluating the Chemosensitizing Effect of Everolimus Administered With Cytarabine and Daunorubicin in Patients With Acute Myeloid Leukemia in Relapse [NCT00544999]Phase 121 participants (Anticipated)Interventional2007-09-30Recruiting
EXecutive Randomized Controlled Trial (RCT): XIENCE V® Everolimus Eluting Coronary Stent System (XIENCE V® EECSS) in the Treatment of the Specific Setting of Patients With Multi-vessel Coronary Artery Disease. [NCT00531011]Phase 4200 participants (Actual)Interventional2007-09-30Completed
A 3-Armed Prospective Randomized Controlled, Open-Labeled Phase II Trial to Evaluate Late Introduction of Cyclosporine or Everolimus Versus a 5-day Delay of Cyclosporine in Combination With MMF in Liver Transplant Recipients With MELD-Scores≥25 [NCT01023542]Phase 2/Phase 345 participants (Anticipated)Interventional2011-06-30Recruiting
Randomized Evaluation of Sirolimus-eluting Versus Everolimus-eluting Stent Trial [NCT01035450]Phase 43,206 participants (Actual)Interventional2010-02-28Completed
A Randomized, Double-blind, Placebo-controlled, Multicenter Phase III Study to Compare the Safety and Efficacy of RAD001 Plus Best Supportive Care (BSC) Versus BSC Plus Placebo in Patients With Metastatic Carcinoma of the Kidney Which Has Progressed on VE [NCT00410124]Phase 3416 participants (Actual)Interventional2006-11-30Completed
A Multicenter, Randomized, Placebo-controlled, Double-blind Study on the Efficacy, Safety and Tolerability of Everolimus in Preventing End-stage Renal Disease (ESRD) in Patients With Autosomal Dominant Polycystic Kidney Disease (ADPKD) [NCT00414440]Phase 4431 participants (Actual)Interventional2006-12-31Completed
SPIRIT IV Clinical Trial: Clinical Evaluation of the XIENCE V® Everolimus Eluting Coronary Stent System in the Treatment of Subjects With de Novo Native Coronary Artery Lesions [NCT00307047]Phase 33,687 participants (Actual)Interventional2006-08-31Completed
Phase I Study of Daily RAD001 Administered Orally in Combination With Mitomycin C, Administered Every Three Weeks to Patients With Advanced Gastric Cancer or Cancer of the Esophagogastric Junction [NCT01042782]Phase 116 participants (Actual)Interventional2008-01-31Completed
A 6-month, Multicenter, Randomized, Open-label Study of Safety and Efficacy of Everolimus-based Regimen Versus Calcineurin Inhibitor (CNI)-Based Regimen in Maintenance Liver Transplant Recipients [NCT00267189]Phase 3145 participants (Actual)Interventional2005-11-30Completed
A Prospective, Open-label, Controlled Multicenter Trial to Assess the Efficacy and Safety of an Induction Regimen of Cyclosporine Micro Emulsion, Enteric-coated Mycophenolate Sodium (EC-MPS) and Corticosteroids, Followed by Administration of Everolimus an [NCT00371826]Phase 4126 participants (Actual)Interventional2006-03-31Completed
A Randomised Fase I/II Trial With Irinotecan, Cetuximab and Everolimus (ICE)Compared to Capecitabine and Oxaliplatin (CapOx) for Patients With Gemcitabin Resistant Pancreatic Cancer [NCT01042028]Phase 1/Phase 239 participants (Actual)Interventional2010-01-31Terminated(stopped due to Emergence of FOLFIRINOX and slow recruitment)
AN INTERVENTIONAL, OPEN-LABEL, RANDOMIZED, MULTICENTER PHASE 3 STUDY OF PF-07220060 PLUS FULVESTRANT COMPARED TO INVESTIGATOR'S CHOICE OF THERAPY IN PARTICIPANTS OVER 18 YEARS OF AGE WITH HORMONE RECEPTOR-POSITIVE, HER2-NEGATIVE ADVANCED/METASTATIC BREAST [NCT06105632]Phase 3510 participants (Anticipated)Interventional2023-12-15Not yet recruiting
Presentation of Renal Function in Liver Transplant Recipients With Certican Therapy: PROTECT Study A Twelve-month, Multicenter, Randomized, Open-label Study of Safety, Tolerability and Efficacy of Certican-based Regimen Versus Calcineurin Inhibitor-based [NCT00378014]Phase 3276 participants (Actual)Interventional2006-08-31Completed
A Phase I Trial Using RAD001 With Weekly Cisplatin and Radiation Therapy in Patients With Locally Advanced Head and Neck Cancer [NCT01058408]Phase 13 participants (Actual)Interventional2010-02-28Terminated
The Real-World Endeavor Resolute Versus XIENCE V Drug-Eluting SteNt Study: Head-to-head Comparison of Clinical Outcome After Implantation of Second Generation Drug-eluting Stents in a Real World Scenario [NCT01066650]Phase 41,391 participants (Actual)Interventional2008-06-30Completed
Efficacy and Safety Study Comparing Concentration-controlled Everolimus in Two Doses (1.5 and 3.0 mg/Day Starting Doses) With Reduced Cyclosporine Versus 1.44 g Mycophenolic Acid (as Sodium Salt) With Standard Dose Cyclosporine in de Novo Renal Transplant [NCT00251004]Phase 3833 participants (Actual)Interventional2005-10-31Completed
A Phase I Dose Escalation Study of RAD001 in Combination With Chemotherapy and Radiation in Patients With NSCLC [NCT01063478]Phase 13 participants (Actual)Interventional2010-02-28Terminated(stopped due to Poor accrual)
Exploring the Activity of RAD001 in Vestibular Schwannomas and Meningiomas [NCT01880749]Early Phase 15 participants (Actual)Interventional2013-06-30Completed
Effect of Rotational Atherectomy on Balloon-resistant Calcified Coronary Lesion the During a Long-term Follow-up Study [NCT01887704]240 participants (Actual)Interventional2010-01-31Completed
Phase II Study Assessing Everolimus as Fist Line Treatment in Patients With Metastatic Kidney Cancer of Bad Prognosis [NCT01888042]Phase 261 participants (Actual)Interventional2011-07-31Terminated
[NCT01895049]Phase 4171 participants (Actual)Interventional2013-08-31Completed
EVESOR: a Phase 1 Trial of Everolimus and Sorafenib to Assess the Impact of Doses and Administration Sequences on Pharmacokinetic and Pharmacodynamic Effects of the Combination [NCT01932177]Phase 160 participants (Anticipated)Interventional2013-04-30Recruiting
Regulatory T Cell Modulation in Kidney Transplantation With Biologic Blockade of Dual Effector Pathways, CD28 and IL-6 (CTOT-24) [NCT04066114]Phase 1/Phase 210 participants (Actual)Interventional2019-12-11Completed
A Phase Ib Trial of Gemcitabine and Cisplatin With RAD001 in Patients With Metastatic Triple Negative Breast Cancer Proceeding to an Open Label Randomized Phase II Trial Comparing Gemcitabine/Cisplatin With or Without RAD001. [NCT01939418]Phase 1/Phase 223 participants (Actual)Interventional2013-08-31Terminated(stopped due to slow recruitment)
Intracoronary Scaffold Assessment a Randomised Evaluation of Absorb in Myocardial Infarction (ISAR-Absorb MI) A Prospective, Randomized Trial of BVS Veruss EES in Patients Undergoing Coronary Stenting for Myocardial Infarction [NCT01942070]262 participants (Actual)Interventional2013-09-30Active, not recruiting
Influence of Exceptional Patient Characteristics on Everolimus Exposure [NCT01948960]Phase 456 participants (Actual)Interventional2013-08-31Completed
Combination of Everolimus and Octreotide LAR in Aggressive Recurrent Meningiomas. [NCT02333565]Phase 220 participants (Actual)Interventional2015-01-22Completed
TRON: A Randomised, Double Blind, Placebo-controlled Study of RAD001 (Everolimus) in the Treatment of Neurocognitive Problems in Tuberous Sclerosis [NCT01954693]Phase 248 participants (Anticipated)Interventional2012-06-30Active, not recruiting
Treatment of Drug-Eluting Stent REstenosis Using Drug-Eluting STents vs. Drug-COated Balloon for Preventing REcurrent In-Stent Restenosis [NCT01967199]Phase 4175 participants (Actual)Interventional2013-04-18Terminated
A Phase Ib/II, Open-Label, Multicenter, Randomized Umbrella Study Evaluating the Efficacy and Safety of Multiple Treatment Combinations in Patients With Breast Cancer (MORPHEUS- BREAST CANCER) [NCT04802759]Phase 1/Phase 2510 participants (Anticipated)Interventional2021-06-20Recruiting
A Phase I Trial of Sirolimus or Everolimus or Temsirolimus (mTOR Inhibitor) and Vorinostat (Histone Deacetylase Inhibitor) in Patients With Advanced Cancer [NCT01087554]Phase 1249 participants (Anticipated)Interventional2010-03-31Active, not recruiting
A Single Center, Open-label, Randomized, Controlled Pilot Trial to Evaluate the Efficacy and Safety of Everolimus Conversion Versus Standard Immunosuppression in Liver Transplant Recipients [NCT01998789]Phase 250 participants (Anticipated)Interventional2013-10-31Recruiting
A Phase I/II Trial of an Oral MTOR Protein Kinase Inhibitor (Everolimus, RAD001) in Combination With an Oral EGFR Tyrosine Kinase Inhibitor (Erlotinib, Tarceva™) In Patients With Metastatic Breast Cancer [NCT00574366]Phase 114 participants (Actual)Interventional2005-12-31Completed
Strut Coverage With SYNERGY Stents and Bioresorbable Vascular Scaffold in Acute Myocardial Infarction: An Intracoronary Optical Coherence Tomography Randomized Study [NCT02890589]22 participants (Actual)Interventional2016-07-31Completed
A Randomized Phase II Trial of Carboplatin, Paclitaxel, Bevacizumab, With or Without Everolimus for Therapy of Metastatic Malignant Melanoma [NCT00976573]Phase 2149 participants (Actual)Interventional2010-04-30Completed
Phase IV, Open-label, Multi-center, Single-arm Study of the Safety and Efficacy of Everolimus (Afinitor) in Adult Patients With Local Advanced or Metastatic, Well Differentiated Progressive Pancreatic Neuroendocrine Tumors (pNET) in China. [NCT02842749]Phase 461 participants (Actual)Interventional2016-03-14Active, not recruiting
A Randomized Prospective, Multicentric, Open Label, Phase II Study Aiming to Evaluate the Efficacity and Safety of EVEROLIMUS as Neo-adjuvant Therapy in Patients With Primary or Relapsed Chondrosarcomas [NCT02008019]Phase 257 participants (Anticipated)Interventional2014-08-14Suspended(stopped due to The study was suspended since july 27th 2016 because of unavailability of Everolimus)
A Pilot Study of Genomic Sequencing Guided Individualized Therapy in Gastrointestinal Cancers [NCT02013089]50 participants (Anticipated)Interventional2013-12-31Recruiting
Multicenter Comparison of Early and Late Vascular Responses to Everolimus-eluting Cobalt-CHromium Stent and Platelet AggregatioN studIeS for TreatMent of Acute Myocardial Infarction: MECHANISM-AMI [NCT02014753]100 participants (Anticipated)Observational2014-04-30Completed
The Feasibility of Selecting Patient-Specific Biologically Targeted Therapy With Sorafenib, Everolimus, Erlotinib or Dasatinib for Pediatric Patients With Refractory Or Recurrent Brain Tumors [NCT02015728]20 participants (Anticipated)Interventional2013-12-31Active, not recruiting
Incidence and Predictors for Late Acquired Stent Malaposition of Drug-Eluting-Stents With Second-Generation Permanent and Biodegradable Polymer-Coatings - a Prospective, Randomized Comparison Using Optical Coherence Tomography [NCT02018991]Phase 469 participants (Actual)Interventional2010-10-31Completed
Observational Study of Everolimus in Combination With Exemestane in Postmenopausal Patients With Hormone Receptor-positive, HER 2-negative Advanced Breast Cancer [NCT02023359]7 participants (Actual)Observational2013-12-31Completed
A Phase II Open-label Pilot Study Evaluating the Maintenance Therapy With Exemestane Plus Everolimus After Induction Chemotherapy in Patients With Hormone-receptor Positive Metastatic Breast Cancer [NCT02025712]Phase 235 participants (Anticipated)InterventionalNot yet recruiting
A Multicenter, Open-label, Randomized, Phase 3 Trial to Compare the Efficacy and Safety of Lenvatinib in Combination With Everolimus or Pembrolizumab Versus Sunitinib Alone in First-Line Treatment of Subjects With Advanced Renal Cell Carcinoma (CLEAR) [NCT02811861]Phase 31,069 participants (Actual)Interventional2016-10-13Active, not recruiting
Phase Ib, Open-label, Multi-center Study to Characterize the Safety, Tolerability and Pharmacodynamics (PD) of PDR001 in Combination With LCL161, Everolimus (RAD001) or Panobinostat (LBH589) [NCT02890069]Phase 1298 participants (Actual)Interventional2016-10-14Completed
A Phase I/II Study of Preoperative (Neoadjuvant) Combination of Letrozole (Femara), Everolimus (Afinitor), and TRC105 in Postmenopausal Women With Newly Diagnosed Local or Locally Advanced Potentially Resectable Hormone-Receptor Positive and Her2 Negative [NCT02520063]Phase 1/Phase 215 participants (Actual)Interventional2016-02-01Completed
Phase II Study of Everolimus (RAD001) for the Treatment of Malignant Pleural Mesothelioma With Merlin/NF2 Loss as a Biomarker to Predict Sensitivity [NCT01024946]Phase 211 participants (Actual)Interventional2009-12-31Completed
Combination of Antiangiogenic Therapy Using the mTOR-inhibitor RAD001 and Low Dose Chemotherapy for Locally Advanced and/or Metastatic Pancreatic Cancer - a Dose Finding Study [NCT00560963]Phase 1/Phase 221 participants (Actual)Interventional2007-10-31Completed
A Phase III Trials Program Exploring the Integration of Bevacizumab, Everolimus (RAD001), and Lapatinib Into Current Neoadjuvant Chemotherapy Regimes for Primary Breast Cancer [NCT00567554]Phase 32,600 participants (Actual)Interventional2007-10-31Completed
Phase Ib/II Evaluation of RAD001 With Docetaxel and Bevacizumab in Patients With Metastatic Androgen Independent Prostate Cancer [NCT00574769]Phase 1/Phase 227 participants (Actual)Interventional2010-02-17Completed
A Phase I Study of Bevacizumab, Everolimus, and Panitumumab for Patients With Advanced Solid Tumors [NCT00586443]Phase 156 participants (Actual)Interventional2007-11-30Completed
Assessment of Everolimus in Addition to Calcineurin Inhibitors Reduction in Maintenance Renal Transplant Recipients [NCT00170846]Phase 4394 participants (Actual)Interventional2005-02-28Completed
A Phase I/II Study of the Raf Kinase/VEGFR Inhibitor Sorafenib in Combination With the mTOR Inhibitor RAD001 (Everolimus) in Patients With Relapsed Non-Hodgkin Lymphoma, Hodgkin Lymphoma, or Multiple Myeloma [NCT00474929]Phase 1/Phase 2103 participants (Actual)Interventional2007-08-29Completed
A Phase I Study to Investigate the Safety and Clinical Activity of Idelalisib in Combination With Chemotherapeutic Agents, Immunomodulatory Agents and Anti-CD20 mAb in Subjects With Relapsed or Refractory Indolent B-cell Non-Hodgkin Lymphoma, Mantle Cell [NCT01088048]Phase 1241 participants (Actual)Interventional2010-03-25Completed
Everolimus for Treatment of Disfiguring Cutaneous Lesions in Neurofibromatosis1- CRAD001CUS232T [NCT02332902]Phase 224 participants (Actual)Interventional2015-02-28Completed
Expanded Access to Everolimus, for an Individual Patient With GIST (Gastrointestinal Stromal Tumors)(CTMS#18-0019) [NCT03493152]0 participants Expanded AccessTemporarily not available
A Randomized Phase II Study of Fulvestrant in Combination With the Dual mTOR Inhibitor AZD2014 or Everolimus or Fulvestrant Alone in Estrogen Receptor-positive Advanced or Metastatic Breast Cancer [NCT02216786]Phase 2333 participants (Actual)Interventional2014-01-31Active, not recruiting
PNOC021: A Phase I Trial Evaluating the Combination of Trametinib and Everolimus in Pediatric and Young Adult Patients With Recurrent Low-Grade Gliomas and High Grade Gliomas [NCT04485559]Phase 150 participants (Anticipated)Interventional2020-12-09Recruiting
A Phase 1b Study of Abemaciclib in Combination With Therapies for Patients With Metastatic Breast Cancer [NCT02057133]Phase 1198 participants (Anticipated)Interventional2014-03-10Active, not recruiting
A Feasibility Trial of Everolimus (RAD001),an mTOR Inhibitor, Given in Combination With Multiagent Re-Induction Chemotherapy in Pediatric Patients With Relapsed Acute Lymphoblastic Leukemia (ALL) [NCT01523977]Phase 122 participants (Actual)Interventional2011-11-30Completed
A Phase II Study of RAD001 in the Treatment of Patients With Plexiform Neurofibromas (PN) Associated With Neurofibromatosis Type 1 (NF1) [NCT01365468]Phase 29 participants (Actual)Interventional2012-04-30Terminated(stopped due to Poor patients' accrual)
Multicenter, Open-label, Randomized, 24 Months Follow-up, Two Arm Study to Compare the Efficacy of Everolimus in Improving the Cardiovascular Profile in a Regimen With Mycophenolic Acid vs. a Regimen of CNI+MPA in Maintenance Renal Transplant Recipients. [NCT01169701]Phase 471 participants (Actual)Interventional2010-08-31Completed
Intracoronary Stenting and Angiographic Results: Test Efficacy of 3 Limus-Eluting STents (ISAR-TEST 4): Prospective, Randomized Trial of 3-limus Agent-eluting Stents With Different Polymer Coatings [NCT00598676]Phase 42,600 participants (Actual)Interventional2007-09-30Completed
A Multi-centric, Open-label, Phase II Study Investigating the Combination of Afinitor With Paclitaxel and Carboplatin in First Line Treatment of Patients With Advanced (Stage IV) Large Cell Lung Cancer With Neuroendocrine Differentiation (LC-NEC) [NCT01317615]Phase 449 participants (Actual)Interventional2011-04-30Completed
An Open Label Randomized Phase II Study of SOM230 and Everolimus in Castrate-Resistant, Chemotherapy-Naïve Prostate Cancer Patients [NCT01313559]Phase 26 participants (Actual)Interventional2011-06-30Terminated(stopped due to Slow accrual)
A Phase I/II Clinical Trial of the mTor Inhibitor RAD001 (Everolimus) in Combination With Lenalidomide (Revlimid) for Patients With Relapsed or Refractory Lymphoid Malignancy [NCT01075321]Phase 1/Phase 258 participants (Actual)Interventional2011-01-10Completed
Phase II Trial of Everolimus or Everolimus Plus Paclitaxel as First-line Therapy in Cisplatin-ineligible Patients With Advanced Urothelial Carcinoma: Hoosier Cancer Research Network GU10-147 [NCT01215136]Phase 236 participants (Actual)Interventional2010-12-31Terminated(stopped due to Lack of Accrual)
A Phase I Study of RAD001 (Everolimus) + Docetaxel + Cisplatin as Induction Chemotherapy in Patients With Local-Regional Advanced Head and Neck Cancer [NCT00935961]Phase 118 participants (Actual)Interventional2009-07-31Completed
Multicentre, Open Study for the Setting up of Population Pharmacokinetic Models and Bayesian Estimators for Individual Dose Adjustment of Immunosuppressive Drugs (Cyclosporine, Tacrolimus, Mycophenolate Mofetil, Everolimus) During the First Year Post-graf [NCT00812786]Phase 442 participants (Actual)Interventional2007-07-31Completed
A Randomized, Double-masked, Parallel Group Study to Assess the Efficacy of Oral Everolimus, Either Alone or Added to Lucentis, in Patients With Neovascular Age-related Macular Degeneration [NCT00857259]Phase 216 participants (Actual)Interventional2009-02-28Terminated
Sorafenib Alone or in Combination With Everolimus in Patients With Unresectable Hepatocellular Carcinoma. A Randomized Multicenter Phase II Trial. [NCT01005199]Phase 2106 participants (Actual)Interventional2009-11-30Completed
Expanded Cohort of Patients With Refractory Metastatic Colorectal Cancer (MCRC) Treated With Bevacizumab and Everolimus [NCT00597506]Phase 250 participants (Actual)Interventional2007-10-31Completed
A Phase I Open Label/Phase II Randomized, Double-Blind, Multicenter Trial Investigating the Combination of Everolimus and TransArterial ChemoEmbolization (TACE) With Doxorubicin in Patients With Hepatocellular Carcinoma [NCT01009801]Phase 1/Phase 227 participants (Actual)Interventional2010-02-28Terminated(stopped due to due to slow patient recruitement.)
Everolimus on CKD (Chronic Kidney Disease) Progression in ADPKD Patients [NCT01009957]Phase 2/Phase 371 participants (Actual)Interventional2008-06-30Terminated(stopped due to After primary completition date, experimental drug was no longer available)
A Phase II, Randomized, Multi-center Study, Assessing Value of Adding Everolimus (RAD001) to Trastuzumab as Preoperative Therapy of HER-2 Positive Primary Breast Cancer Amenable to Surgery. [NCT00674414]Phase 282 participants (Actual)Interventional2008-04-30Terminated(stopped due to IDMC decision due to accrual issue (82 pts accrued / 120 expected))
A Phase III, Randomized, Open-Label, Multicenter Study Evaluating the Efficacy and Safety of Giredestrant Plus Everolimus Compared With The Physician's Choice of Endocrine Therapy Plus Everolimus in Patients With Estrogen Receptor-Positive, HER2-Negative, [NCT05306340]Phase 3320 participants (Anticipated)Interventional2022-08-03Recruiting
LAte Stent Strut APPosition and COverage After Drug-Eluting Stent ImplantaTIOn by Optical Coherence Tomography in PatieNts With Acute Myocardial Infarction II(APPOSITION-AMI II) [NCT02770651]69 participants (Anticipated)Observational2016-05-31Enrolling by invitation
Everolimus Versus Sunitinib Therapy in Patients With Advanced Non-clear Cell Renal Cell Carcinoma [NCT01185366]Phase 273 participants (Actual)Interventional2010-08-31Terminated
XIENCE V® Everolimus Eluting Coronary Stent System India Post-marketing Single-Arm Study [NCT00631228]1,000 participants (Actual)Observational2008-06-30Completed
Phase I/II Study of BNC105P in Combination With Everolimus or Following Everolimus For Progressive Metastatic Clear Cell Renal Cell Carcinoma Following Prior Tyrosine Kinase Inhibitors [NCT01034631]Phase 1/Phase 2154 participants (Actual)Interventional2010-01-31Completed
Phase II Study of Cisplatinum and Everolimus in Patients With Metastatic or Unresectable Neuroendocrine Carcinomas (NEC) of Extrapulmonary Origin [NCT02695459]Phase 239 participants (Actual)Interventional2016-03-30Active, not recruiting
Everolimus-eluting Bioresorbable Vascular Scaffolds Versus Everolimus-eluting Stents in Patients With Chronic Total Occlusion [NCT02739685]17 participants (Actual)Interventional2016-09-30Terminated(stopped due to Slow enrollment, access to devices)
A 12 Month, Multi-center, Randomized, Open-label Non-inferiority Study Comparing Safety and Efficacy of Concentration-controlled Everolimus With Low Dose Tacrolimus to Mycophenolate Mofetil With Standard Dose Tacrolimus in de Novo Renal Transplant Recipie [NCT01025817]Phase 3613 participants (Actual)Interventional2010-01-31Completed
A Sequential Phase I Study Of The Combination Of Everolimus (Rad001) With 5-Fu/Lv (De Gramont), Folfox6, And Folfox6/Panitumumab In Patients With Refractory Solid Malignancies [NCT00610948]Phase 174 participants (Actual)Interventional2008-03-31Completed
Everolimus (RAD001) Therapy for Epilepsy in Patients With Tuberous Sclerosis Complex [NCT01070316]Phase 1/Phase 220 participants (Actual)Interventional2010-01-31Completed
A Phase I Dose Finding Study of Everolimus (RAD001) in Elderly Patients With Acute Myeloid Leukemia (AML) Unfit for Intensive Induction Chemotherapy [NCT00636922]Phase 140 participants (Anticipated)Interventional2010-02-28Active, not recruiting
A Phase II Biomarker Trial of Everolimus in Patients With Advanced Renal Cell Carcinoma [NCT00827359]Phase 225 participants (Actual)Interventional2009-03-31Completed
A Phase 2 Study to Evaluate the Efficacy of Epigenetic Modulation in Relapsed/Refractory Diffuse Large B-Cell Lymphoma (DLBCL) [NCT00978432]Phase 250 participants (Actual)Interventional2012-02-29Terminated(stopped due to The toxicity seemed to outweigh the benefit.)
Selection of Chemoradiotherapy Based on Response to Induction Chemotherapy - a Phase II Study in Locally Advanced Squamous Cell Carcinoma of Head and Neck [NCT01133678]Phase 250 participants (Actual)Interventional2010-05-04Terminated(stopped due to Primary endpoint reached futility boundary)
An Open-Label, Multi-Center, Expanded Access Study of RAD001 in Patients With Metastatic Carcinoma of the Kidney Who Are Intolerant of or Have Progressed Despite Any AvailableVascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy [NCT00655252]0 participants Expanded AccessNo longer available
Phase II Study of RAD001 in a Neoadjuvant Setting in Men With Intermediate or High Risk Prostate Cancer [NCT00657982]Phase 215 participants (Anticipated)Interventional2008-04-30Recruiting
Efficacy and Safety of Certican® (Everolimus) in Combination With Myfortic® (EC-MPS, Enteric-coated Mycophenolate Sodium) After Early CNI Elimination Versus Myfortic® in Combination With Prograf® in Renal Transplant Recipients [NCT00965094]Phase 436 participants (Actual)Interventional2009-12-31Completed
Topical Everolimus Versus Placebo for the Treatment of Facial Angiofibromas in Patients With Tuberous Sclerosis Complex. A Phase II/III, Multicentre, Randomized, Double-blind, Placebo-controlled Study of 3 Doses of Topical Everolimus. [NCT02860494]Phase 2/Phase 30 participants (Actual)Interventional2020-12-31Withdrawn(stopped due to Pharmaceutical and financial difficulties)
A Phase I Study of Cisplatin, Paclitaxel, and RAD001 Patients With Metastatic Breast Cancer [NCT00680758]Phase 118 participants (Actual)Interventional2008-05-31Completed
An Expanded Phase I Study of Pazopanib and Everolimus in Patients With Advanced Solid Tumors and Previously Treated Advanced Urothelial Cancer [NCT01184326]Phase 123 participants (Actual)Interventional2011-01-31Completed
A Randomized, Double-blind, Placebo Controlled, Phase II Study of Everolimus in Combination With Exemestane in the Treatment of Chinese Postmenopausal Women With Estrogen Receptor Positive, HER-2 Negative, Locally Advanced, Recurrent, or Metastatic Breast [NCT03312738]Phase 2159 participants (Actual)Interventional2017-09-15Completed
[NCT02796157]950 participants (Anticipated)Interventional2016-06-30Recruiting
Phase II Examination of Irinotecan, Cetuximab and Everolimus to Chemotherapy Resistent Patients With Metastatic Colorectal Cancer and KRAS Mutations or After Progression With KRAS Wildtype on Irinotecan and Cetuximab - Effect and Biological Markers [NCT01387880]Phase 2100 participants (Actual)Interventional2010-01-31Completed
A Single Arm, Multicenter Phase II a Trial of RAD001 as Monotherapy in the Treatment of Neurofibromatosis 1 Related Internal Plexiform Neurofibromas That Cannot be Removed by Surgery [NCT01412892]Phase 230 participants (Actual)Interventional2011-04-30Completed
Terapia Con Everolimus Nel Trapianto de Novo di Fegato: Uno Studio Multicentrico Randomizzato [NCT01423708]Phase 2117 participants (Anticipated)Interventional2010-02-28Recruiting
A Phase 1b/2 Study of Retaspimycin HCl (IPI-504) in Combination With Everolimus in Patients With KRAS Mutant NSCLC [NCT01427946]Phase 1/Phase 247 participants (Actual)Interventional2011-07-31Completed
Plasmonic Photothermal and Stem Cell Therapy of Atherosclerosis With The Use of Gold Nanoparticles With Iron Oxide-Silica Shells Versus Stenting [NCT01436123]Phase 162 participants (Actual)Interventional2010-12-31Terminated(stopped due to The study was terminated under the political pressure of the Federal Security Service of the Russian Federation (FSB) and the Russian Society of Cardiology)
P3 Efficacy Evaluation of Enobosarm in Combo With Abemaciclib Compared to Estrogen Blocking Agent for 2nd Line Treatment of ER+HER2- MBC in Patients Who Have Shown Previous Disease Progression on an Estrogen Blocking Agent Plus Palbociclib [NCT05065411]Phase 3186 participants (Anticipated)Interventional2022-04-11Active, not recruiting
A Phase I Study of Weekly Low-Dose Cisplatin Plus Escalating Doses of Oral RAD001(Everolimus) for Patients With Advanced Solid Tumors [NCT00423865]Phase 130 participants (Actual)Interventional2006-11-30Completed
Multicenter, Randomized, Open-label Trial to Evaluate the Safety, Tolerability and Efficacy of Two Regimens of Everolimus Plus Cyclosporine Microemulsion, Given According to Different Blood Target Levels, in de Novo Renal Transplant Recipients [NCT00170885]Phase 40 participants Interventional2005-05-31Completed
A Phase II Trial of RAD001 in Patients With Recurrent Glioblastoma Multiforme [NCT00515086]Phase 241 participants (Actual)Interventional2007-08-31Terminated(stopped due to Early termination due to slow enrollment and protocol-defined stopping rule.)
RESOLUTE-III All-comers Trial: A Randomized Comparison of a Zotarolimus-Eluting Stent With an Everolimus-Eluting Stent for Percutaneous Coronary Intervention [NCT00617084]Phase 42,292 participants (Actual)Interventional2008-04-30Completed
Phase II Study of Low-dose RAD001(Everolimus) Plus Cisplatin and HDFL (Weekly 24-Hour Infusion of High-dose 5-Fluorouracil and Leucovorin) Chemotherapy for First-line Treatment of Unresectable, Recurrent or Metastatic Gastric Cancer [NCT00632268]Phase 240 participants (Actual)Interventional2008-02-29Completed
A Randomised Pilot Study of Neoadjuvant Everolimus Plus Letrozole Compared With FEC in Postmenopausal Patients With ER-positive, HER2-negative Breast Cancer [NCT02742051]Phase 240 participants (Actual)Interventional2016-06-30Completed
A Phase 1 Study of RAD001 in Combination w/ Cetuximab and Cisplatin as First-line Therapy in Recurrent & Metastatic Squamous Cell Cancer of the Head & Neck [NCT01009346]Phase 1/Phase 29 participants (Actual)Interventional2009-10-31Terminated(stopped due to Toxicity)
Phase II Trial of RAD001 in Relapsed/Refractory Multiple Myeloma [NCT00618345]Phase 235 participants (Anticipated)Interventional2005-03-31Completed
A Phase I Pilot Study of the Oral mTOR Inhibitor RAD001 in Combination With Capecitabine for Metastatic Breast Cancer [NCT00473005]Phase 118 participants (Actual)Interventional2007-08-31Terminated(stopped due to Principal Investigator (Dr. Guardino) left Stanford)
A Pilot, Rapid Sequencing of First Line Cabozantinib, Ipilimumab and Nivolumab, and Lenvatinib and Everolimus in Patients With Metastatic or Unresectable Clear Cell Renal Cell Carcinoma [NCT05188118]Early Phase 120 participants (Anticipated)Interventional2023-02-24Recruiting
An Open-Label, Single-arm, Phase Ib/II Study of AEB071 (a Protein Kinase C Inhibitor) and Everolimus (mTOR Inhibitor) in Patients With CD79-mutant or ABC Subtype Diffuse Large B-Cell Lymphoma [NCT01854606]Phase 131 participants (Actual)Interventional2013-12-05Completed
[NCT01856374]Phase 460 participants (Actual)Interventional2011-08-31Completed
Phase II Study of the Levonorgestrel Intrauterine Device Alone or in Combination With the mTORC1 Inhibitor, Everolimus, for the Treatment of Complex Atypical Hyperplasia and Stage Ia Grade 1 Endometrial Cancer [NCT02397083]Phase 2270 participants (Anticipated)Interventional2015-09-23Recruiting
[NCT01857843]Phase 4160 participants (Actual)Interventional2009-11-30Completed
Amsterdam Investigator-initiateD Absorb Strategy All-comers Trial (AIDA Trial): A Clinical Evaluation Comparing the Efficacy and Performance of ABSORB™ Everolimus Eluting Bioresorbable Vascular Scaffold Strategy Versus the XIENCE Family (XIENCE PRIME™ or [NCT01858077]1,845 participants (Actual)Interventional2013-08-31Active, not recruiting
Open Label, Non-randomized, Phase 2 Study Investigating the Effect of RAD001 Monotherapy in Patients With Advanced NSCLC Previously Treated With Either Chemotherapy Only or With Chemotherapy and EGFR Inhibitor(s) [NCT00124280]Phase 285 participants (Actual)Interventional2005-07-31Completed
Phase I Study of TheraSphere and Everolimus Among Patients With Neuroendocrine Tumors and Liver Only or Liver Dominant Disease [NCT01864070]Phase 10 participants (Actual)Interventional2014-05-31Withdrawn
Everolimus (RAD001)Therapy of Giant Cell Astrocytomas in Patients With Tuberous Sclerosis Complex [NCT00411619]Phase 1/Phase 228 participants (Actual)Interventional2007-01-31Completed
SCHEDULE - Scandinavian Heart Transplant Everolimus de Novo Study With Early CNI Avoidance [NCT01266148]Phase 4115 participants (Actual)Interventional2009-11-30Completed
Phase III Multicenter Open-label Randomized Clinical Trial Comparing Everolimus and Low Dose Tacrolimus to Tacrolimus and Mycophenolate Mofetil at 6 mo Post-Transplant to Prevent Long-term Complications After Pediatric Heart Transplantation [NCT03386539]Phase 3211 participants (Actual)Interventional2018-01-29Active, not recruiting
A Pilot Study Comparing the Safety and Efficacy of Zortress (Everolimus) With Low Dose Tacrolimus to Early Conversion to Calcineulin Inhibitor-Free Regimen and Mycophenolic Acid With Standard Dose Tacrolimus in Recipients of ECD/DCD Kidneys [NCT01878786]Phase 2/Phase 325 participants (Actual)Interventional2013-06-30Terminated(stopped due to Interim results suggested a concern for patient outcomes and safety)
Pilot Study of mTOR Inhibitor Therapy for Treatment of Intestinal Polyps in Peutz-Jeghers Syndrome [NCT00811590]Phase 23 participants (Actual)Interventional2008-11-30Terminated(stopped due to The study was ended early due to low enrollment.)
Phase IB/II Study of Pasireotide, Everolimus and Selective Internal Radioembolization Therapy (SIRT) for Unresectable Neuroendocrine Hepatic Metastases [NCT01469572]Phase 113 participants (Actual)Interventional2011-12-31Completed
Randomized Comparison of the Effects of PLatinum Chromium Everolimus-eluting Stent vs. cobAlT Chromium Everolimus-eluting Stent on inFlammatOry maRkers and Endothelial daMage - The PLATFORM Trial [NCT01489202]Phase 4100 participants (Anticipated)Interventional2014-09-30Not yet recruiting
A Single Arm, Single Center, Phase II Trial of RAD001 as Monotherapy in the Treatment of Neurofibromatosis Type 2 - Related Vestibular Schwannoma [NCT01490476]Phase 210 participants (Actual)Interventional2012-01-31Completed
Phase I/IIB Study of Induction Chemotherapy With XELOX, Followed by Radiation Therapy and Dose Escalation of RAD001 in Patients With Esophageal Cancer [NCT01490749]Phase 117 participants (Actual)Interventional2012-02-29Completed
A Phase 1/2 Study of Lenvatinib in Combination With Everolimus in Recurrent and Refractory Pediatric Solid Tumors, Including CNS Tumors [NCT03245151]Phase 1/Phase 264 participants (Actual)Interventional2017-11-16Completed
PhaseII,Open-label,Pilot Study Evaluating the Safety+Efficacy of Certican ® in the Prevention of Chronic Graft-versus-host Disease+Late Pulmonary Complications After Allogeneic Hematopoietic Cell Transplantation Blood [NCT01509560]Phase 221 participants (Actual)Interventional2011-11-01Completed
Phase I/II Trial of RAD001 Plus Nexavar® For Patients With Metastatic Renal Cell Carcinoma [NCT00448149]Phase 1/Phase 255 participants (Actual)Interventional2006-12-31Completed
A Combined Phase 1 and 2 Study Investigating the Combination of RAD001 and Erlotinib in Patients With Advanced NSCLC Previously Treated Only With Chemotherapy [NCT00456833]Phase 1248 participants (Actual)Interventional2005-06-30Completed
Reduction or Discontinuation of Calcineurine Inhibitors With Conversion to Everolimus-Based Immunosuppresion to Alleviate Chronic Allograft Nephropathy (CAN) in Kidney Transplant Recipients: A Prospective Randomized Study [NCT00443508]Phase 460 participants Interventional2007-02-28Recruiting
RAD001 Therapy of Angiomyolipomata in Patients With Tuberous Sclerosis Complex and Sporadic Lymphangioleiomyomatosis [NCT00457964]Phase 1/Phase 236 participants (Actual)Interventional2005-08-31Completed
A Phase II Randomized, Double-blinded, Multicenter Asian Study Investigating the Combination of Transcatheter Arterial Chemoembolization (TACE) and Oral Everolimus (RAD001, Afinitor®) in Localised Unresectable Hepatocellular Carcinoma (HCC) - The TRACER S [NCT01379521]Phase 265 participants (Actual)Interventional2011-06-30Terminated(stopped due to The study was terminated due to low enrollment. This resulted in the study being underpowered and inconclusive.)
[NCT01605721]Phase 42,000 participants (Anticipated)Interventional2011-05-31Recruiting
An Open-label, Multicenter Phase 1 Study Investigating the Combination of RAD001, Cetuximab and Irinotecan as Second-line Therapy After FOLFOX (or XELOX) Plus Bevacizumab (if Given as Part of Local Standard Practice) in Patient With Metastatic Colorectal [NCT00478634]Phase 119 participants (Actual)Interventional2007-05-31Completed
Comparison Between Drug-Eluting Stents for the Treatment of the Diffuse Type In-Stent Restenosis After Drug-Eluting Stents Implantation: Sirolimus-Eluting vs. Paclitaxel-Eluting Stents [NCT00485030]Phase 460 participants (Actual)Interventional2007-03-31Completed
A 12 Month Open-label, Randomized, Multicenter, Sequential Cohort-group, Dose Finding Study to Evaluate the Efficacy, Safety and Tolerability of Oral AEB071 Versus Cyclosporine in Combination With Everolimus, Basiliximab and Corticosteroids in de Novo Adu [NCT00504543]Phase 2311 participants (Actual)Interventional2007-07-31Completed
A 2-year Extension to a 1-year Multicenter, Randomized, Open Label, Parallel Group Study of the Safety, Tolerability and Efficacy of Two Doses (1.5 and 3 mg/Day) of Everolimus With Steroids and Optimized Administration of Cyclosporine in de Novo Renal Tra [NCT00531063]Phase 3237 participants (Anticipated)Interventional2001-11-30Completed
Phase I / Randomized Phase II Study of Second Line Therapy With Irinotecan and Cetuximab With or Without RAD001, an Oral mTOR Inhibitor for Patients With Metastatic Colorectal Cancer: Hoosier Oncology Group GI05-102 [NCT00522665]Phase 1/Phase 241 participants (Actual)Interventional2007-08-31Completed
A 2 Year Extension to a 1 Year Multicenter, Randomized, Open Label, Parallel Group Study of the Safety, Tolerability and Efficacy of Two Doses (1.5 and 3 mg/Day) of Everolimus (RAD001) With Basiliximab, Corticosteroids and Optimized Administration of Cycl [NCT00531440]Phase 3256 participants (Actual)Interventional2001-11-30Completed
A Prospective, Multi-Center, Single-Blinded, Randomized Trial of the Sirolimus-Eluting Iron Bioresorbable Coronary Scaffold System in Patients With Coronary Artery Disease: IRONMAN-II [NCT05206084]518 participants (Actual)Interventional2022-03-10Active, not recruiting
Phase 2 Study Assessing the Tolerance and Efficacy of Tamoxifen Alone Versus the Association Tamoxifen-RAD001 (Everolimus) in Patients With Anti-aromatase Resistant Breast Metastatic Cancer [NCT01298713]Phase 2111 participants (Actual)Interventional2008-03-31Completed
A Phase 1b/2 Study of Imatinib in Combination With Everolimus in Synovial Sarcoma [NCT01281865]Phase 1/Phase 214 participants (Actual)Interventional2011-01-31Completed
Comparison of Everolimus- and Biolimus-Eluting Stents With Everolimus-Eluting Bioresorbable Vascular Scaffold Stents [NCT01711931]Phase 4240 participants (Actual)Interventional2012-10-31Completed
Ph1 Study of the Safety, PK, and PDn of Escalating Oral Doses of the Glutaminase Inhibitor CB-839, as a Single Agent and in Combination With Standard Chemotherapy in Patients With Advanced and/or Treatment-Refractory Solid Tumors [NCT02071862]Phase 1210 participants (Actual)Interventional2014-02-28Completed
Phase IV: Effect of Everolimus and CNI Minimalization on Renal Function. [NCT00596557]Phase 420 participants (Actual)Interventional2008-02-29Completed
Phase I-II Study of Trastuzumab in Combination With RAD001 in Patients With HER-2 Overexpressing, PTEN-deficient Metastatic Breast Cancer Progressing on Trastuzumab-Based Therapy [NCT00317720]Phase 1/Phase 240 participants (Actual)Interventional2006-04-30Completed
An Exploratory, Open-label, Non-randomized, Within-patient Multiple Dose-escalation Safety, Tolerability, PK and Efficacy Trial of RAD001 (Everolimus) in Patients With Lymphangioleiomyomatosis [NCT01059318]Phase 224 participants (Actual)Interventional2010-01-31Completed
A Multi Center Randomized Control Study of MeRes100 Sirolimus Eluting BioResorbable Vascular Scaffold System in Treatment of Coronary Artery Disease Patients: MeRes - China. [NCT03454724]484 participants (Anticipated)Interventional2020-01-01Not yet recruiting
Cytokines Evaluation in Early Calcineurin Inhibitors Withdrawn on Renal Transplant [NCT01239472]Phase 430 participants (Actual)Interventional2011-01-31Completed
A Single-arm, Open-label and Multi-center Phase I Study on Safety, Tolerance and Pharmacokinetics of HRS-8080 Tablets Monotherapy and Combined With Other Anti-cancer Therapy in Patients With Metastatic or Local Advanced Breast Cancer [NCT05189717]Phase 1156 participants (Anticipated)Interventional2022-02-17Enrolling by invitation
Phase I Trial of Everolimus, Pomalidomide and Dexamethasone in Patients With Relapsed/Refractory Multiple Myeloma [NCT01889420]Phase 11 participants (Actual)Interventional2014-07-31Terminated(stopped due to Low accrual)
Zortress (Everolimus) to Prevent Alloantibody Formation in Patients With Late Stage Renal Allograft Failure: The Everolimus-Transplant Exit Strategy Trial (E-TEST) [NCT01636466]Phase 31 participants (Actual)Interventional2013-06-30Terminated(stopped due to Feasibility)
ABSORB II RANDOMIZED CONTROLLED TRIAL A Clinical Evaluation to Compare the Safety, Efficacy and Performance of ABSORB Everolimus Eluting Bioresorbable Vascular Scaffold System Against XIENCE Everolimus Eluting Coronary Stent System in the Treatment of Sub [NCT01425281]501 participants (Actual)Interventional2011-11-30Completed
A Phase II Study to Evaluate the Safety and Efficacy of RAD001 Plus Erlotinib in Patients With Well- to Moderately-Differentiated Neuroendocrine Tumors [NCT00843531]Phase 217 participants (Actual)Interventional2009-06-25Terminated(stopped due to Low Accrual)
Randomized Open Label Study to Compare the Efficacy and Safety of Everolimus Followed by Chemotherapy With Streptozotocin- Fluorouracilo (STZ-5FU) Upon Progression or the Reverse Sequence, in Advanced Progressive Pancreatic NETs (pNETs) [NCT02246127]Phase 3141 participants (Actual)Interventional2014-10-27Completed
A Phase 3, Randomized, Controlled Study of Cabozantinib (XL184) vs Everolimus in Subjects With Metastatic Renal Cell Carcinoma That Has Progressed After Prior VEGFR Tyrosine Kinase Inhibitor Therapy [NCT01865747]Phase 3658 participants (Actual)Interventional2013-06-30Completed
A Randomized, Double-Blind, Placebo-Controlled Phase II Trial of Weekly Paclitaxel/Bevacizumab +/- Everolimus as First-Line Chemotherapy for Patients With HER2-Negative Metastatic Breast Cancer (MBC) [NCT00915603]Phase 2113 participants (Actual)Interventional2009-07-31Completed
The Efficacy and Safety of CM082 Combined With Everolimus in Chinese Patients With Metastatic Renal Cell Carcinoma: a Randomized, Double-blind, Double Dummy, Multicenter Study [NCT03095040]Phase 3390 participants (Anticipated)Interventional2016-12-16Active, not recruiting
A Phase I/II Study of LEE011 Plus Everolimus in Patients With Metastatic Pancreatic Adenocarcinoma Refractory to Chemotherapy [NCT02985125]Phase 1/Phase 244 participants (Anticipated)Interventional2017-05-04Active, not recruiting
A Phase II Study of Everolimus in Combination With Paclitaxel and Carboplatin in Patients With Metastatic Melanoma [NCT01014351]Phase 270 participants (Actual)Interventional2010-02-28Completed
5, 6 or 7 Year Follow-up Control After the SCHEDULE Study (SCANDINAVIAN HEART TRANSPLANT EVEROLIMUS DE NOVO STUDY WITH EARLY CNI AVOIDANCE) [NCT02864706]Phase 495 participants (Actual)Interventional2016-01-18Completed
Conversion From MPA to Zortress (Everolimus) for GI Toxicity Post-renal Transplantation [NCT02974686]Phase 41 participants (Actual)Interventional2016-11-30Terminated(stopped due to low recruitment)
A Phase I-II, Open-label Study of RAD001 in Combination With Glivec®/Gleevec™ (Imatinib) in Patients With Glivec/Gleevec-refractory/Resistant Gastrointestinal Stromal Tumors. [NCT01275222]Phase 1/Phase 2117 participants (Actual)Interventional2002-11-13Completed
Randomized ph. II Study to Explore Efficacy and Feasibility of Upfront Rotations Between SUNitinib and Everolimus vs Sequential Treatment of 1st lIne Sunitinib & 2nd Line EverolimuS Until Progression in Pats Met. Clear Cell Renal Cancer [NCT01784978]Phase 241 participants (Actual)Interventional2013-02-12Terminated(stopped due to Lack of recruitment)
A Prospective, Single-center, Open-label, Pilot Study to Investigate the Effect of Switching to Certican® in Viremia of Hepatitis C Virus in Adult Renal Allograft Recipients. [NCT01469884]Phase 430 participants (Actual)Interventional2011-11-30Completed
A Phase II Trial of RAD001 (Everolimus) for 2nd Line Treatment After Failure of Fluoropyrimidine Plus Platinum Chemotherapy in Patients With Metastatic or Recurrent Gastric Cancer With pS6 Ser 240/4 Expression [NCT01482299]Phase 245 participants (Actual)Interventional2011-11-30Completed
Six-month, Multicenter, Randomized, Open-label Study of the Safety, Tolerability and Efficacy of Two Neoral Doses in Addition to Certican and Steroids in de Novo Heart Transplant Recipients [NCT00098007]Phase 3199 participants (Actual)Interventional2004-08-09Completed
Multicenter Trial of the Safety & Efficacy of Certican in Pediatric de Novo Renal Transplant Patients [NCT00098241]Phase 345 participants (Anticipated)Interventional2000-06-30Completed
Phase II Trial Of RAD-001 In Metastatic Malignant Melanoma [NCT00098553]Phase 253 participants (Actual)Interventional2005-04-30Completed
Randomized 2x2 Factorial Trial Comparing the Cre8 Amphilimus-sirolimus Eluting Stent vs. the Synergy Everolimus-eluting Stent and a Personalized vs. Standard Duration of Dual Antiplatelet Therapy in All-comers Patients Undergoing Percutaneous Coronary Int [NCT04135989]Phase 42,106 participants (Anticipated)Interventional2020-01-01Active, not recruiting
Pilot Study on the Determination of Intratumoral Concentrations of Kinase Inhibitors in Patients With Advanced Solid Malignancies. [NCT01636908]43 participants (Actual)Interventional2011-08-31Completed
A Phase 2, Double-blind, Randomized, Placebo-controlled, Multi-center Study Assessing the Value of Adding Everolimus to Letrozole as Preoperative Therapy of Primary Breast Cancer in Postmenopausal Women [NCT00107016]Phase 2267 participants (Actual)Interventional2005-03-31Completed
A One-Year, Single-Center, Prospective, Open-Label Study of the Safety, Tolerability, and Preliminary Efficacy of Anti-Thymocyte Globulin, Cyclosporine, and RAD in Type 1 Diabetic Islet Transplant Recipients [NCT00286624]Phase 1/Phase 26 participants (Actual)Interventional2003-03-31Completed
Levels of Circulating Tumor DNA as a Predictive Marker for Early Switch in Treatment for Patients With Metastatic (Stage IV) Breast Cancer: A Phase 2 Randomized, Open-Label Study [NCT05826964]Phase 2500 participants (Anticipated)Interventional2023-06-12Recruiting
XIENCE Skypoint Large Vessel Post Approval Study [NCT05423379]100 participants (Anticipated)Observational2022-09-14Recruiting
Multicenter Randomized Two-arms Study Evaluating the BK Viral Clearance in Kidney Transplant Recipients With BK Viremia After Reduction of Immunosuppression Alone vs. Reduction of Immunosuppression and Replacement of Mycophenolate Mofetil by Everolimus [NCT03216967]Phase 4130 participants (Actual)Interventional2018-01-15Active, not recruiting
A Phase 2 Study of Dasatinib in Combination With Everolimus for Children With Gliomas Harboring PDGFR Alterations [NCT03352427]Phase 23 participants (Actual)Interventional2017-12-06Terminated(stopped due to Low accrual)
A Phase I-II, Study of RAD001 in Combination With Imatinib (Glivec®/Gleevec™) in Patients With Chronic Myelogenous Leukemia (CML) in Chronic Phase Who Are Not In Complete Cytogenetic Response to Imatinib-Alone at Study Entry [NCT00093639]Phase 1/Phase 20 participants Interventional2004-08-31Completed
A Phase I Study Investigating Everolimus and Dovitinib in Metastatic Clear Cell Renal Cancer [NCT01714765]Phase 117 participants (Actual)Interventional2011-04-30Completed
Phase II Study Evaluating the Combination of Everolimus and Sorafenib in the Treatment of Thyroid Cancer [NCT01141309]Phase 241 participants (Actual)Interventional2010-06-30Active, not recruiting
Reservoir-Based Polymer-Free Amphilimus-Eluting Stent Versus Polymer-Based Everolimus-Eluting Stent in Diabetic Patients (RESERVOIR) Trial [NCT01710748]Phase 3112 participants (Anticipated)Interventional2012-10-31Active, not recruiting
A Phase IB/II Multicenter, Two-Arm, Dose-Escalation Study of Oral AEE788 Administered in Combination With Oral RAD001 on a Continuous Once Daily Dosing Schedule in Adult Patients With First or Second Recurrent or Relapsing Glioblastoma Multiforme [NCT00107237]Phase 1/Phase 216 participants (Actual)Interventional2003-10-31Completed
RADAR: A Randomized Discontinuation Phase II Study to Determine the Efficacy of RAD001 in Breast Cancer Patients With Bone Metastases [NCT00466102]Phase 2130 participants (Anticipated)Interventional2006-12-31Active, not recruiting
Monocenter, Double Blinded, Prospective, Randomized Placebo Controlled Study Investigating Prevention of Major Adverse Cardiac Events (MACEs) Within 6 Months by Systemic Treatment With Everolimus After Coronary Intervention With Bare Metal Stents in Patie [NCT00426049]Phase 3484 participants Interventional2006-10-31Recruiting
Phase II Study of RAD001 (Everolimus) in Patient's With Metastatic Renal Cell Cancer [NCT00446368]Phase 266 participants (Actual)Interventional2005-05-31Completed
Prophylaxis of the Graft-Versus-Host-Disease in Patients After Allogeneic Stem Cell Transplantation With a Combination of Tacrolimus and Everolimus [NCT00117702]Phase 2/Phase 324 participants (Actual)Interventional2005-10-31Terminated(stopped due to safety reasons)
A Single Arm, Open-label Multicenter Phase II Trial of Everolimus in Patients With Relapsed/Refractory Germ Cell Cancer [NCT01242631]Phase 225 participants (Actual)Interventional2010-11-30Completed
SPIRIT V: A Clinical Evaluation of the XIENCE V® Everolimus Eluting Coronary Stent System in the Treatment of Patients With de Novo Coronary Artery Lesions [NCT00402272]Phase 42,700 participants (Actual)Interventional2006-11-30Completed
Phase I Study of Patupilone and RAD001 in Patients With Refractory Solid Tumor Malignancy [NCT00496600]Phase 142 participants (Actual)Interventional2007-07-31Completed
A Phase II Study of Everolimus in Combination With Imatinib in Metastatic Melanoma [NCT00402662]Phase 23 participants (Actual)Interventional2006-02-28Terminated(stopped due to unexpected level of toxicity)
A Randomized, Double-blind, Multi-center Phase III Study Evaluating Paclitaxel With and Without RAD001 in Patients With Gastric Carcinoma Who Have Progressed After Therapy With a Fluoropyrimidine-containing Regimen [NCT01248403]Phase 3300 participants (Actual)Interventional2011-10-31Completed
A Phase I Study Investigating the Combination of RAD001 With Standard Induction and Consolidation Therapy in Older Patients With Newly Diagnosed Acute Myeloid Leukemia (AML) [NCT01154439]Phase 111 participants (Actual)Interventional2010-10-31Completed
Efficacy and Safety of Everolimus to SAve REnal Function (SAREFU) in Long Term Heart Transplanted Patients [NCT00505102]Phase 4200 participants (Anticipated)Interventional2007-01-31Active, not recruiting
PNOC 001: Phase II Study of Everolimus for Recurrent or Progressive Low-grade Gliomas in Children [NCT01734512]Phase 265 participants (Actual)Interventional2012-12-13Active, not recruiting
Trial of RAD001 in Patients With Operable Non-Small Cell Lung Cancer (NSCLC) [NCT00401778]Phase 133 participants (Actual)Interventional2006-11-30Completed
A Phase 1b Trial of LY2584702 in Combination With Erlotinib or Everolimus in Patients With Solid Tumors [NCT01115803]Phase 129 participants (Actual)Interventional2010-03-31Terminated(stopped due to Primary objective has been met; safety and pharmacokinetics have been characterized.)
Circulating Tumor Cells and Tumor DNA in HCC and NET - Patient-specific Biomarkers for Clinical Decision Support and Tailored Relapse Diagnostics [NCT02973204]167 participants (Actual)Observational [Patient Registry]2016-11-30Completed
Phase II Trial of Bicalutamide and RAD001 in Patients With Hormone-Independent Prostatic Adenocarcinoma (HIPC) After the First-Line Androgen Deprivation Therapy [NCT00814788]Phase 224 participants (Actual)Interventional2008-12-31Completed
Phase I/II of Panobinostat (LBH589) Plus Everolimus (RAD001) in Patients With Relapsed and Refractory Lymphoma [NCT00967044]Phase 1/Phase 231 participants (Actual)Interventional2009-11-30Completed
Phase II Study of Everolimus (RAD001) in Primary Therapy of Waldenstrom's Macroglobulinemia [NCT00976248]Phase 233 participants (Actual)Interventional2009-11-30Completed
[NCT01093300]Phase 4264 participants (Anticipated)InterventionalNot yet recruiting
Randomized Phase III Trial Comparing Everolimus Versus Everolimus Plus Bevacizumab for Advanced Renal Cell Carcinoma Progressing After Treatment With Tyrosine Kinase Inhibitors [NCT01198158]Phase 377 participants (Actual)Interventional2010-09-15Terminated
Efficacy, Safety and Patient-reported Outcomes of Peptide Receptor Radionuclide Therapy With 177Lu-edotreotide Compared to Everolimus in Somatostatin Receptor Positive Neuroendocrine Tumors of the Lung and Thymus. [NCT05918302]Phase 3120 participants (Anticipated)Interventional2023-10-27Recruiting
Clinical Evaluation of mTORC1 Inhibition for Geroprotection [NCT05835999]Phase 286 participants (Anticipated)Interventional2023-03-24Recruiting
A Phase I Dose Escalation Study With Everolimus in Combination With Cyclosporine A and Prednisolone for the Treatment of Acute GVHD After Allogeneic Hematopoietic Stem Cell Transplantation [NCT00373815]Phase 110 participants (Anticipated)Interventional2006-09-30Terminated(stopped due to high incidence of TTP, poor recrual)
Phase III Randomized, Placebo-Controlled Clinical Trial Evaluating the Use of Adjuvant Endocrine Therapy +/- One Year of Everolimus in Patients With High-Risk, Hormone Receptor-Positive and HER2/Neu Negative Breast Cancer [NCT01674140]Phase 31,939 participants (Actual)Interventional2013-09-12Active, not recruiting
Prospective Randomised Study Comparing Efficacy of Treatment Coronary In-stent Restenosis Using Drug Eluting Paclitaxel-coated Balloon and Drug Eluting Stent With Everolimus. [NCT01735825]199 participants (Actual)Interventional2012-01-31Completed
An Open-Label, Multicenter, Phase 1b/2 Study of E7080 Alone, and in Combination With Everolimus in Subjects With Unresectable Advanced or Metastatic Renal Cell Carcinoma Following One Prior VEGF-Targeted Treatment [NCT01136733]Phase 1/Phase 2173 participants (Actual)Interventional2010-08-05Completed
Phase II Trial of RAD001 Plus Carboplatin in Patients With Triple-Negative Metastatic Breast Cancer [NCT01127763]Phase 225 participants (Actual)Interventional2010-06-30Completed
A Phase Ib Study Investigating the Combination of Everolimus With Trastuzumab and Vinorelbine in Patients With HER2-overexpressing Metastatic Breast Cancer [NCT00426530]Phase 150 participants (Actual)Interventional2007-02-28Completed
A Phase I Study Investigating the Combination of Everolimus With Pemetrexed in Patients With Advanced Non Small Cell Lung Cancer (NSCLC) Previously Treated With Chemotherapy [NCT00434174]Phase 148 participants (Actual)Interventional2006-12-31Completed
A Study to Evaluate the Safety and Performance of the Dynalink®-E, Everolimus Eluting Peripheral Stent System for Treating Atherosclerotic de Novo or Restenotic Native Superficial Femoral and Proximal Popliteal Artery Lesions [NCT00475566]104 participants (Actual)Interventional2007-05-31Completed
A Pilot Study to Evaluate Feasibility and Safety of Early Switch to Everolimus From Cyclosporine in de Novo Renal Transplant [NCT00464399]Phase 320 participants Interventional2006-09-30Completed
Immune Monitoring to Facilitate Belatacept Monotherapy [NCT04177095]Phase 417 participants (Actual)Interventional2020-02-11Completed
A Single Arm, Multi-center Phase II Study of RAD001 in Patients With Advanced Gastric Carcinoma Whose Cancer Has Progressed Despite Prior Treatment [NCT00519324]Phase 254 participants (Actual)Interventional2007-08-31Completed
Phase I/II Study of Paclitaxel / Carboplatin / RAD001 as First Line Therapy for Advanced Adenocarcinoma of the Stomach [NCT01514110]Phase 1/Phase 235 participants (Actual)Interventional2008-01-23Completed
Hybrid Sirolimus-eluting Stent With Bioresorbable Polymer Versus Everolimus-eluting Stent With Durable Polymer for Total Coronary Occlusions in Native Coronary Arteries (PRISON-IV) [NCT01516723]Phase 3330 participants (Anticipated)Interventional2012-02-29Active, not recruiting
Randomized Phase II Study to Compare Vinorelbine In Combination With the mTOR Inhibitor Everolimus vs. Vinorelbin Monotherapy for Second-line Treatment in Advanced Breast Cancer [NCT01520103]Phase 2139 participants (Actual)Interventional2012-01-31Completed
Impact of Two Prednisone-free Maintenance Immunosuppressive Regimens With Reduced Dose FK506+Everolimus vs. Standard Dose Tacrolimus (FK506)+ Mycophenolate Mofetil (MMF) on Subpopulation of T and B Cells, Renal Allograft Function and Gene Expression Profi [NCT01653847]88 participants (Actual)Interventional2013-02-28Completed
A Phase II Single Arm Study to Evaluate the Safety and Efficacy of RAD001 as Monotherapy in Treatment naïve Advanced Cholangiocarcinoma [NCT01525719]Phase 240 participants (Anticipated)Interventional2012-01-31Active, not recruiting
Phase I-II Randomized Prospective Double-blind Multi-center Trial on the Effects of a Short Course of Oral Everolimus on Infarct Size, Left Ventricular Remodeling and Inflammation in Patients With Acute ST-Elevation Myocardial Infarction [NCT01529554]Phase 1/Phase 2150 participants (Actual)Interventional2015-01-08Completed
Phase 1 Trial of the Mammalian Target of Rapamycin (mTOR) Inhibitor Everolimus Plus Radiation Therapy (RT) for Salvage Treatment of Biochemical Recurrence in Prostate Cancer Patients Following Prostatectomy [NCT01548807]Phase 119 participants (Actual)Interventional2010-09-30Completed
A Single-arm, Open Label Phase II Study of RAD001 in Soft Tissue Extremity and/or Retroperitoneal Sarcomas [NCT01048723]Phase 22 participants (Actual)Interventional2010-01-31Terminated(stopped due to Novartis terminated funding)
Trametinib Combined With Everolimus and Lenvatinib in the Treatment of Recurrent/Refractory Advanced Solid Tumors: a Phase II Clinical Trial [NCT04803318]Phase 2100 participants (Anticipated)Interventional2021-01-01Recruiting
A Trial of Maintenance Rituximab With mTor Inhibition After High-dose Consolidative Therapy in CD20+, B-cell Lymphomas, Gray Zone Lymphoma, and Hodgkin's Lymphoma [NCT01665768]Phase 256 participants (Actual)Interventional2012-09-30Completed
Phase II Study of Everolimus in Patients With Advanced Solid Malignancies With TSC1, TSC2, NF1, NF2, or STK11 Mutations [NCT02352844]Phase 212 participants (Actual)Interventional2015-10-07Completed
A Ph2 Randomized Trial to Evaluate the Safety Preliminary Efficacy and Biomarker Response of Host Directed Therapies Added to Rifabutin-modified Standard Therapy in Adults With Drug-Sensitive Smear-Positive Pulmonary TB [NCT02968927]Phase 2200 participants (Actual)Interventional2016-11-30Active, not recruiting
Phase II Expansion Cohorts Studies of a Novel Triple Combination Therapy, DTRM-555, in Patients With Relapsed/Refractory Chronic Lymphocytic Leukemia or Relapsed/Refractory Non-Hodgkin's Lymphomas [NCT04305444]Phase 2120 participants (Anticipated)Interventional2020-04-24Recruiting
Phase II Study of Everolimus in Refractory Testicular Germ Cell Cancer. [NCT01466231]Phase 215 participants (Actual)Interventional2011-11-30Completed
Phase I Study of Bevacizumab in Combination With Everolimus and Erlotinib in Advanced Cancer [NCT00276575]Phase 165 participants (Actual)Interventional2005-03-31Completed
Pilot Trial of an Implantable Microdevice for In Vivo Drug Sensitivity Testing in Patients With Sarcomas [NCT04199026]Early Phase 120 participants (Anticipated)Interventional2024-01-31Not yet recruiting
Envarsus® Tablets Administered Once Daily in Combination With Everolimus in Elderly De-novo Kidney Transplant Recipients: Open-label, Multicentre, Single-arm, Pharmacokinetic and Clinical Study [NCT02970630]Phase 228 participants (Actual)Interventional2017-01-31Completed
A Single Arm, Monocenter Phase II Trial of RAD001 as Monotherapy in the Treatment of Neurofibromatosis Type 2 - Related Vestibular Schwannoma [NCT01345136]Phase 24 participants (Actual)Interventional2015-07-01Active, not recruiting
Immunosuppressive Therapy With Certican (Everolimus) After Lung Transplantation [NCT00402532]Phase 3190 participants (Actual)Interventional2005-03-31Completed
An Open Label, Randomised Phase II Study, Comparing AZD2014 Versus Everolimus With Advanced Metastatic Renal Cancer and Progression on VEGF Targeted Therapy [NCT01793636]Phase 249 participants (Actual)Interventional2013-02-28Terminated(stopped due to Inferior efficacy of study drug in renal indication.)
A 12-month, Multicenter, Randomized, Open-label Non-inferiority Study of Renal Function and Efficacy Comparing Concentration-controlled Certican (1.5 mg/Day Starting Dose) With Reduced Neoral Dose Versus MMF With Standard Neoral Dose in de Novo Heart Tran [NCT00150046]Phase 3176 participants (Anticipated)Interventional2004-12-31Completed
Prospective Randomized Study to Characterize Risk Factors of Polyomavirus-related Transplant Nephropathy and the Impact of Three Immunosuppressive Regimens on Nephropathy Incidence [NCT00160966]Phase 4108 participants (Actual)Interventional2004-09-30Completed
A One Year, Multicenter, Open-Label, Single Arm, Pilot Study of the Renal Safety of Everolimus in Addition to Cyclosporine Microemulsion in Cardiac Transplant Recipients. [NCT00170794]Phase 30 participants Interventional2004-09-30Completed
A Prospective, Multicenter, Open Label, Randomized Study of the Safety, Tolerability and Efficacy of Everolimus (RAD001) With Basiliximab, Corticosteroids and Lower Levels Versus Higher Levels of Tacrolimus in de Novo Renal Transplant Recipients [NCT00170833]Phase 380 participants Interventional2003-11-30Completed
A Clinical Evaluation of the XIENCE V® Everolimus Eluting Coronary Stent System in the Treatment of Patients With de Novo Native Coronary Artery Lesions [NCT00180310]Phase 3300 participants (Actual)Interventional2005-07-31Completed
Phase I Trial of RAD001C (Everolimus) in Pediatric Patients With Recurrent Refractory Solid Tumors or Brain Tumors [NCT00187174]Phase 141 participants (Actual)Interventional2004-10-31Completed
Polymorphism of the Cytochrome P450-system and the MDR-system in Renal Transplants Receiving the Immunosuppressive Drugs Tacrolimus, Sirolimus, Everolimus or Cyclosporine A [NCT00223054]200 participants (Actual)Observational2005-03-31Completed
Effects of Zortress® + Tacrolimus vs. Standard Immunosuppression on Progression of Coronary Artery Calcifications and Bone Disease in de Novo Renal Transplant Recipients [NCT01612299]0 participants (Actual)Interventional2012-05-31Withdrawn(stopped due to Funding was withdrawn for this study by the sponsor.)
Efficacy and Safety of Precision Therapy in Refractory Tumor (Long March Pathway) [NCT03239015]Phase 2300 participants (Anticipated)Interventional2017-01-01Recruiting
A Brief Dose Escalation Followed by a Phase 2 Study of RAD001 in Combination With Trastuzumab in HER2-Positive Metastatic Breast Cancer [NCT00458237]Phase 1/Phase 211 participants (Actual)Interventional2007-04-30Completed
Single Arm, Single Center, Prospective, Phase II Clinical Study of Anlotinib Hydrochloride Capsule Combined With Everolimus in the First-line Treatment of Advanced Non Clear Cell Renal Cell Carcinoma [NCT05124431]Phase 230 participants (Anticipated)Interventional2021-12-31Not yet recruiting
Randomized Phase II Study of Two Different Doses of RAD-001 (Everolimus) as Neo-Adjuvant Therapy in Patients With Localized Prostate Cancer [NCT00526591]Phase 217 participants (Actual)Interventional2007-09-30Terminated(stopped due to Slow accrual)
Study Examining the PROMUS Element Everolimus-eluting Stent in Multi-center Coronary Intervention of Complex Arterial Lesion Subsets [NCT01670318]800 participants (Anticipated)Interventional2012-08-31Recruiting
A Phase II Randomized, Double-blind, Placebo-controlled Trial of Radium-223 Dichloride in Combination With Exemestane and Everolimus Versus Placebo in Combination With Exemestane and Everolimus When Administered to Metastatic HER2 Negative Hormone Recepto [NCT02258451]Phase 2283 participants (Actual)Interventional2015-06-04Completed
Everolimus as Treatment After Embolization or Chemoembolization for Liver Metastases From Digestive Endocrine Tumors [NCT01678664]Phase 274 participants (Actual)Interventional2012-10-31Completed
A 3-Arm Randomized Phase II Trial Evaluating Single Agent and Combined Efficacy of Pasireotide and Everolimus in Adult Patients With Radioiodine-Refractory Differentiated and Medullary Thyroid Cancer [NCT01270321]Phase 242 participants (Actual)Interventional2010-11-30Completed
A Single Arm, Two Center, Phase II Study of RAD001 in Patients With Metastatic, Hormone-Refractory Prostate Cancer [NCT00629525]Phase 235 participants (Actual)Interventional2005-08-31Completed
An 18 Month Extension to the Multicenter, Randomized, Open-label Trial (NCT00170885) to Evaluate the Safety, Tolerability, and Efficacy of Two Regimens of Everolimus Plus Cyclosporine Microemulsion, Given According to Different Blood Target Levels, in de [NCT01276457]Phase 3223 participants (Actual)Interventional2006-05-31Completed
A Phase 2 Trial of RAD001(Everolimus) in Low and Intermediate-1 Risk Myelodysplastic Syndrome [NCT00809185]Phase 27 participants (Actual)Interventional2005-11-30Terminated(stopped due to slow accrual)
Phase II Trial of RAD001 in Patients With Recurrent Low Grade Glioma [NCT00823459]Phase 258 participants (Actual)Interventional2009-01-23Completed
Comparison Between Abluminal Biodegradable Polymer Ultrathin Sirolimus-eluting Stent and Durable-polymer Everolimus-eluting Stent (GENOSS Randomized Clinical Trial) [NCT05444452]850 participants (Anticipated)Interventional2022-04-24Recruiting
Study to Investigate Outcome of Individualized Treatment Based on Pharmacogenomic Profiling & Ex Vivo Drug Sensitivity Testing of Patient-derived Organoids in Patients With Metastatic Colorectal Cancer [NCT05725200]Phase 240 participants (Anticipated)Interventional2022-09-27Recruiting
A Multicenter, Two Arms, Randomized, Open Label Clinical Phase IV Study Investigating the Proportion of CMV Seropositive Kidney Transplant Recipients Who Will Develop a CMV Infection Within the First 6 Months Post-transplantation When Treated With an Immu [NCT02328963]Phase 4186 participants (Actual)Interventional2014-05-02Completed
Neoadjuvant Phase II Study Of Everolimus Plus Cisplatin In Triple Negative Breast Cancer Patients With Residual Disease After Standard Chemotherapy [NCT01931163]Phase 224 participants (Actual)Interventional2013-07-31Completed
A Phase I Trial of Escalating Dose of RAD001 in Combination With PKC412 in Patients With Relapsed, Refractory or Poor Prognosis AML or MDS [NCT00819546]Phase 129 participants (Anticipated)Interventional2009-01-31Active, not recruiting
A Study in Adults on Pre LT Dialysis With Basiliximab, Delayed Tacrolimus (TAC), Mycophenolate (MMF), Steroids (Grp 1) vs. Basiliximab, Delayed TAC, MMF, Steroids, With Everolimus 30d Post LT(Grp 2), vs. TAC, MMF, Steroids (Grp 3). [NCT04104438]Phase 490 participants (Anticipated)Interventional2021-01-15Recruiting
AXITINIB IN ADVANCED / METASTATIC RENAL CELL CARCINOMA - A NON-INTERVENTIONAL STUDY OF REAL WORLD TREATMENT OUTCOMES IN PATIENTS RECEIVING 2ND LINE AXITINIB AFTER 1ST LINE SUNITINIB (ADONIS) [NCT02184416]573 participants (Actual)Observational2014-10-31Completed
A Phase II Multicenter Trial of RAD001 in Patients With Metastatic or Recurrent Sarcomas [NCT01830153]Phase 241 participants (Actual)Interventional2010-04-30Completed
An Open-label Randomized Controlled Clinical Trial to Assess the Efficacy and Safety of the Conversion to Everolimus 3 Months After Kidney Transplantation. [NCT01608412]Phase 4120 participants (Anticipated)Interventional2012-02-29Recruiting
Placebo-Controlled, Randomized, Prospective and Multicenter Trial of Everolimus in Castrated Resistant Prostate Cancer Patients With PI3K-AKT-mTOR Signaling Pathway Deficiency [NCT03580239]Phase 3120 participants (Anticipated)Interventional2019-01-01Not yet recruiting
Phase IV, Prospective Single Arm Study of Safety and Efficacy of Votubia (Everolimus) in Taiwanese Adults With Tuberous Sclerosis Complex Who Have Renal Angiomyolipoma [NCT05252585]Phase 410 participants (Anticipated)Interventional2023-05-01Recruiting
Phase II Trial of Carboplatin and Everolimus (RAD001) in Metastatic Castrate Resistant Prostate Cancer (CRPC) Pretreated With Docetaxel Chemotherapy. [NCT01051570]Phase 226 participants (Actual)Interventional2010-02-28Completed
PTK787/ZK222584 and RAD001 for Patients With Advanced Solid Tumors [NCT00303732]Phase 137 participants (Actual)Interventional2004-12-31Completed
Bioabsorbable Vascular Solutions First in Man Clinical Investigation: A Clinical Evaluation of the Bioabsorbable Vascular Solutions Everolimus Eluting Coronary Stent System (BVS EECSS) in the Treatment of Patients With Single de Novo Native Coronary Arter [NCT00300131]30 participants (Actual)Observational2006-03-31Completed
An Open-label, Multi-center Phase I Dose-finding Study of RAD001 (Everolimus, Afinitor®) in Combination With BEZ235 in Patients With Advanced Solid Tumors [NCT01482156]Phase 146 participants (Actual)Interventional2012-01-31Completed
Safety and Efficacy of Low-dose Cyclosporine in Association With Everolimus to Minimize Renal Dysfunction in Heart Transplant Recipients [NCT00420537]Phase 434 participants (Actual)Interventional2006-09-30Terminated(stopped due to A cluster of adverse events in everolimus arm was noted.)
Phase II Trial of RAD001 in Patients With Refractory Colorectal Cancer [NCT00337545]Phase 222 participants (Actual)Interventional2006-05-31Completed
A Phase 1b/2, Open-Label Umbrella Study To Evaluate Safety And Efficacy Of Elacestrant In Various Combination In Patients With Metastatic Breast Cancer [NCT05563220]Phase 1/Phase 2322 participants (Anticipated)Interventional2023-01-24Recruiting
Open Label Multicenter Randomized Trial Comparing Standard Immunosuppression With Tacrolimus and Mycophenolate Mofetil With a Low Exposure Tacrolimus Regimen in Combination With Everolimus in de Novo Renal Transplantation in Elderly Patient [NCT03797196]Phase 4374 participants (Anticipated)Interventional2019-07-29Active, not recruiting
PREVENT: a Prospective, Multi-center, Monitored Trial Investigating the Implant of the Promus Everolimus-Eluting Stent System in Critically Ischemic Lesions BTK [NCT01500070]Phase 270 participants (Anticipated)Interventional2012-08-31Completed
An Open, Randomized Phase III Study of Everolimus With Investigator's Choice of Chemotherapy Versus Chemotherapy in The First-Line Treatment of Luminal Androgen Receptor (LAR) Subtype With PI3K/AKT/mTOR Pathway Mutation of Locally Recurrent Inoperable or [NCT05954442]Phase 3203 participants (Anticipated)Interventional2023-09-13Recruiting
A Dose Escalation, Single Arm, Phase 1b-2 Combination Study of BEZ235 With Everolimus to Determine the Safety, Pharmacodynamics and Pharmacokinetics in Subjects With Advanced Solid Malignancies [NCT01508104]Phase 119 participants (Actual)Interventional2012-01-31Terminated(stopped due to funding)
The Beneficial Role of Percutaneous Coronary Intervention Over Optimal Medical Therapy in Elderly Patients (Age > 75 Years Old) With Coronary Artery Disease: a Randomized Controlled Study [NCT01508663]Phase 41,600 participants (Anticipated)Interventional2010-08-31Recruiting
TITANIC-XV Trial: Prospective, Multicenter and Randomized Trial (Bioactive Bare Metal Titanium Stent Versus Everolimus Drug Eluting Stent) [NCT01510509]Phase 40 participants Interventional2009-01-31Active, not recruiting
A Phase I Trial of Anakinra (IL-1 Receptor Antagonist) or Denosumab (Anti-RANKL Monoclonal Antibody) in Combination With Everolimus (mTOR Inhibitor) in Patients With Advanced Malignancies [NCT01624766]Phase 157 participants (Actual)Interventional2012-06-19Completed
Mono Centre, Open Label Proof of Concept Study SOM230 in Progressive Medullary Thyroid Cancer Patients and the Combination With RAD001 Upon Progression [NCT01625520]Phase 219 participants (Actual)Interventional2012-02-29Completed
A Phase IIIB, Multi-Center, Open Label Study For Postmenopausal Women With Estrogen Receptor Positive Locally Advanced or Metastatic Breast Cancer Treated With Everolimus (RAD001) in Combination With Exemestane: 4EVER - Efficacy, Safety, Health Economics, [NCT01626222]Phase 3301 participants (Actual)Interventional2012-06-30Completed
Immunosuppression in Renal Transplantation in The Elderly: Time to Rethink. - nEverOld Study [NCT01631058]Phase 490 participants (Anticipated)Interventional2012-07-31Recruiting
A Phase I Evaluation of Cetuximab and RAD001 in Patients With Solid Tumors [NCT01637194]Phase 112 participants (Actual)Interventional2008-11-30Completed
Effect of Different Therapeutic Strategies on Regulatory T Cells in Kidney Transplantation: a Randomized Study [NCT01640743]58 participants (Actual)Interventional2010-03-31Completed
Phase II Multicenter Single-arm Study Evaluating the Safety and Efficacy of Everolimus as a First-line Treatment in Newly-diagnosed Patients With Advanced GI Neuroendocrine Tumors. [NCT01648465]Phase 225 participants (Actual)Interventional2012-08-06Terminated
Certican (Everolimus) for Recipients of Kidney From HLA-identical Living Donors [NCT01653041]Phase 447 participants (Actual)Interventional2012-08-31Completed
Phase II Single Arm Study of Everolimus and Pasireotide (SOM230) in Patients With Advanced or Metastatic Hepatocellular Carcinoma (HCC) [NCT01488487]Phase 224 participants (Actual)Interventional2011-12-31Completed
"Effects of the Use of de Novo Everolimus for the Expression of Cytokines in Kidneys From Extended Criteria Donors and With Delayed Graft Function" [NCT01663805]Phase 480 participants (Anticipated)Interventional2012-01-31Recruiting
Randomized Comparison of Everolimus Eluting Stents and Sirolimus Eluting Stent in Patients With ST Elevation Myocardial Infarction (STEMI) (RaCES-MI Trial) [NCT01684982]Phase 4500 participants (Actual)Interventional2007-04-30Completed
Phase I/II Study of RAD001 and Intravesical Gemcitabine in BCG-Refractory Primary or Secondary Carcinoma In Situ of the Bladder [NCT01259063]Phase 1/Phase 233 participants (Actual)Interventional2010-12-31Completed
An Open Label, Single Arm Trial to Evaluate Patients With Metastatic Renal Cell Carcinoma Treated With Everolimus After Failure of First Line Therapy With Sunitinib or Pazopanib [NCT01514448]Phase 429 participants (Actual)Interventional2012-05-21Completed
Phase I Dose Escalation Study of Everolimus, Pemetrexed, Carboplatin, and Bevacizumab in Stage IV Non-Squamous Non-Small Cell Lung Cancer [NCT01700400]Phase 113 participants (Actual)Interventional2012-09-30Completed
[NCT01706471]Phase 460 participants (Actual)Interventional2009-06-30Completed
An Unicenter, Prospective, Randomized, Pilot Study Comparing the Effect of Everolimus-containing Versus mTOR Inhibitor Free Immunosuppression in the Evolution of Hepatitis C Fibrosis After Liver Transplantation. [NCT01707849]Phase 318 participants (Actual)Interventional2012-10-31Completed
Phase 2 Study of Everolimus Therapy in Patients With Birt-Hogg-Dube Syndrome (BHD)-Associated Kidney Cancer or Sporadic Chromophobe Renal Cancer [NCT02504892]Phase 23 participants (Actual)Interventional2015-07-21Terminated(stopped due to Study was terminated due to slow, insufficient accrual.)
A Comparison of Blood and Tissue Biomarkers Before and After Nephrectomy in the First-line Setting With Everolimus in Patients With Locally Advanced or Metastatic Renal Cell Carcinoma [NCT01715935]Phase 225 participants (Actual)Interventional2012-06-30Completed
Efficacy of RAD001/Everolimus in Autism and NeuroPsychological Deficits in Children With Tuberous Sclerosis Complex [NCT01730209]Phase 2/Phase 360 participants (Anticipated)Interventional2012-11-30Recruiting
A Prospective, Open-label, Multicenter, Randomized Phase II Trial: Sequential Therapy With BEvacizumab, RAd001 (Everolimus) and Tyrosinekinase Inhibitors (TKI) in Metastatic Renal Cell Carcinoma (mRCC) (BERAT Study) [NCT01731158]Phase 222 participants (Actual)Interventional2012-10-31Completed
A Randomized Proof-of-concept Phase II Trial Comparing Therapy Based on Tumor Molecular Profiling Versus Conventional Therapy in Patients With Refractory Cancer. [NCT01771458]Phase 2742 participants (Actual)Interventional2012-10-31Active, not recruiting
A Multicenter Randomized, Double Blind, Placebo- Controlled, Phase II Study to Compare Endocrine Treatment Alone Versus Endocrine Treatment With Everolimus in Patients With HR+/HER2- Metastatic Breast Cancer and Progression After Previous Treatment With E [NCT01773460]Phase 33 participants (Actual)Interventional2013-07-31Terminated(stopped due to Study was stopped terminated early due to low recruitment.)
Apposition Assessed Using Optical Coherence Tomography of Chromium Stents Eluting Everolimus From Cobalt Versus Platinum Alloy Platforms. [NCT01776567]Phase 460 participants (Anticipated)Interventional2012-07-31Recruiting
Drug Interaction and Pharmacokinetic Assessment of Everolimus When Coadministered With Atorvastatin in Renal Transplantation Recipient [NCT01780948]18 participants (Actual)Interventional2012-09-30Completed
Phase 1b/2 Single-arm Trial Evaluating the Combination of Lapatinib, Everolimus and Capecitabine for the Treatment of Patients With HER2-positive Metastatic Breast Cancer With CNS Progression After Trastuzumab [NCT01783756]Phase 1/Phase 29 participants (Actual)Interventional2013-06-26Completed
A Multicentre Phase III Trial to Determine the Efficacy of RAD 001 (Everolimus, Afinitor) as Second Line Therapy in Patients With Transitional Cell Carcinoma TCC of the Urothelium Which Failed or Progressed After First Line Chemotherapy [NCT01801137]Phase 254 participants (Actual)Interventional2011-06-30Completed
A Phase II, Open Label, Non-randomized Study of Second or Third Line Treatment With the Combination of Sorafenib and Everolimus in Patients Affected by Relapsed and Non-resectable High-grade Osteosarcoma [NCT01804374]Phase 238 participants (Actual)Interventional2011-06-30Completed
Prospective Evaluation of the Efficacy and Safety of Zortress (Everolimus)/Myfortic (Enteric Coated Mycophenolate Sodium) Conversion in High MELD Liver Transplantation [NCT01807767]0 participants (Actual)Interventional2013-03-31Withdrawn(stopped due to funding was withdrawn)
Drug- Drug Interaction Study of JI-101 & Everolimus in Advanced Solid Tumors, Expansion Pharmacodynamic Study of JI-101 in Advanced Low Grade Endocrine Tumors, Ovarian Cancers or K-RAS Mutant Colon Cancers [NCT01149434]Phase 1/Phase 219 participants (Actual)Interventional2010-09-30Terminated(stopped due to Lack of drug efficacy)
Sirolimus-coated Balloon Versus Drug-eluting Stent in Native Coronary Vessels [NCT04893291]1,820 participants (Anticipated)Interventional2021-11-16Recruiting
Randomized Double-Blind Phase 2 Trial Of RAD001 For Neurocognition In Individuals With Tuberous Sclerosis Complex [NCT01289912]Phase 252 participants (Actual)Interventional2011-01-31Completed
PHASE II TRIAL OF THE mTOR INHIBITOR EVEROLIMUS IN RELAPSED OR REFRACTORY CUTANEOUS T-CELL LYMPHOMA (CTCL) [NCT01637090]Phase 23 participants (Actual)Interventional2012-06-30Terminated(stopped due to Poor enrollment)
A Phase II Study of Lenvatinib Plus Everolimus Versus Cabozantinib in Patients With Metastatic Renal Cell Carcinoma That Progressed on A PD-1/PD-L1 Checkpoint Inhibitor [NCT05012371]Phase 290 participants (Anticipated)Interventional2022-02-16Recruiting
Phase II Trial of Lapatinib in Combination With Everolimus in Triple Negative Metastatic or Locally Advanced Breast Cancer [NCT01272141]Phase 25 participants (Actual)Interventional2010-12-31Terminated(stopped due to Slow accrual, Funding stopped)
A Phase 1b/2, Protocol Evaluating the Safety, Tolerability, Pharmacokinetics, and Efficacy of Sotorasib Monotherapy and in Combination With Other Anti-cancer Therapies in Subjects With Advanced Solid Tumors With KRAS p.G12C Mutation (CodeBreak 101) [NCT04185883]Phase 1/Phase 21,143 participants (Anticipated)Interventional2019-12-17Recruiting
A Randomized Comparison of a Sirolimus-eluting Stent With Biodegradable Polymer Versus an Everolimus-eluting Stent With a Durable Polymer for Percutaneous Coronary Revascularization [NCT01443104]2,119 participants (Actual)Interventional2012-02-29Completed
Phase 1 Pilot Study Using Autologous CD4+CD25+FoxP3+ T Regulatory Cells and Campath-1H to Induce Renal Transplant Tolerance [NCT01446484]Phase 1/Phase 230 participants (Anticipated)Interventional2011-10-31Recruiting
Women's Triple-Negative First-Line Study: A Phase II Trial of Liposomal Doxorubicin, Bevacizumab and Everolimus (DAE) in Patients With Localized Triple-Negative Breast Cancer (TNBC) With Tumors Predicted Insensitive to Standard Neoadjuvant Chemotherapy [NCT02456857]Phase 217 participants (Actual)Interventional2016-01-12Completed
Phase 1-2 Study of Everolimus and Low-dose Cyclophosphamide in Patients With Metastatic Renal Cell Cancer. [NCT01462214]Phase 1/Phase 296 participants (Anticipated)Interventional2011-10-31Completed
Phase II Trial of Bevacizumab(Avastin) and RAD001(Everolimus)in the Treatment of Patients With Advanced Clear Cell Renal Carcinoma [NCT00323739]Phase 280 participants (Actual)Interventional2006-05-31Completed
A Phase II Trial of RAD001 and Bicalutamide for Androgen Independent Prostate Cancer [NCT00630344]Phase 236 participants (Actual)Interventional2008-02-29Completed
Prospective, Multicenter, Randomized Open Study to Evaluate the Progression of Renal Graft Fibrosis According to the Epithelial-mesenchymal Transition (EMT) in de Novo Renal Transplant Recipients Treated Either by a CNI Free Immunosuppressive Regimen With [NCT01079143]Phase 3194 participants (Actual)Interventional2009-09-30Completed
German-Austrian Register to Evaluate the Short and Long-term Safety and Therapy Outcomes of the ABSORB Everolimus-eluting Bioresorbable Vascular Scaffold System in Patients With Coronary Artery Stenosis [NCT02066623]3,330 participants (Actual)Observational [Patient Registry]2013-11-08Completed
A Phase II Trial of the Combination of Bevacizumab and Everolimus in Patients With Refractory, Progressive Intracranial Meningioma [NCT00972335]Phase 218 participants (Actual)Interventional2010-01-31Terminated(stopped due to Study terminated early due to slow accrual)
A Phase 2 Trial of Letrozole and Everolimus in Relapsed Hormone Receptor Positive Ovarian, Fallopian Tube or Primary Peritoneal Carcinomas [NCT02283658]Phase 220 participants (Actual)Interventional2014-11-14Completed
BIOTRONIK - A Prospective Randomized Multicenter Study to Assess the SaFety and Effectiveness of the Orsiro SiroLimus Eluting Coronary Stent System in the Treatment Of Subjects With up to Three De Novo or Restenotic Coronary Artery Lesions - V [NCT02389946]1,334 participants (Actual)Interventional2015-05-31Completed
Phase Ib Trial of mFOLFOX6 and Everolimus (NSC-733504) in Patients With Metastatic Gastroesophageal Adenocarcinoma [NCT01231399]Phase 1/Phase 26 participants (Actual)Interventional2012-02-29Completed
A 12-month, Multi-center, Open-label, Randomized, Controlled Study to Evaluate Efficacy/Safety and Evolution of Renal Function of Everolimus in Co-exposure With Tacrolimus in de Novo Liver Transplant Recipients [NCT01551212]Phase 4339 participants (Actual)Interventional2012-05-24Completed
BrUOG-H&N-229-RAD001, Cisplatin and Concurrent Radiation for Locally Advanced, Inoperable Head and Neck Cancer: A PHASE I STUDY- Novartis CRAD001CUS134T [NCT01057277]Phase 13 participants (Actual)Interventional2009-12-31Terminated(stopped due to Pharmaceutical co- re aligned their specialties- no longer will fund H&N ca)
XIENCE V Everolimus Eluting Coronary Stent System (EECSS) China: Post-Approval, Single-Arm Study [NCT01249027]2,605 participants (Actual)Observational2010-11-30Completed
A Phase I/II Study of the HDAC Inhibitor LBH-589 in Combination With the mTOR Inhibitor Everolimus (RAD001) in Metastatic Renal Cell Carcinoma [NCT01582009]Phase 1/Phase 226 participants (Actual)Interventional2010-03-31Terminated(stopped due to pts off study, PI left institute)
A Phase 1 Study of Fosbretabulin in Combination With Everolimus in Neuroendocrine Tumors (Grades 1-3) That Have Progressed After at Least One Prior Regimen for Metastatic Disease [NCT03014297]Phase 117 participants (Actual)Interventional2017-03-06Terminated(stopped due to Part I completed. Part 2 will not be done due to sponsor decision.)
An Open-label, Multi-center Long-term Safety Roll-over Study in Patients With Tuberous Sclerosis Complex (TSC) and Refractory Seizures Who Are Judged by the Investigator to Benefit From Continued Treatment With Everolimus After Completion of Study CRAD001 [NCT02962414]Phase 3206 participants (Actual)Interventional2017-06-08Active, not recruiting
A Phase II Study of RAD001 (Everolimus) for Children With NeurF1 and Chemotherapy-Refractory Radiographic Progressive Low Grade Gliomas [NCT01158651]Phase 223 participants (Actual)Interventional2010-07-10Completed
Randomized Phase II Study of Everolimus Alone Versus Everolimus Plus Bevacizumab in Patients With Locally Advanced or Metastatic Pancreatic Neuroendocrine Tumors [NCT01229943]Phase 2150 participants (Actual)Interventional2010-10-15Completed
A Single-arm, Multicenter, Phase 2 Trial to Evaluate Efficacy and Safety of Lenvatinib in Combination With Everolimus in Subjects With Unresectable Advanced or Metastatic Non Clear Cell Renal Cell Carcinoma (nccRCC) Who Have Not Received Any Chemotherapy [NCT02915783]Phase 241 participants (Actual)Interventional2017-02-20Completed
Circulating FGF21 Levels and Efficacy of Exemestane, Everolimus and Metformin in Postmenopausal Women With Hormone Receptor Positive Metastatic Breast Cancer and BMI >/= 25 [NCT01627067]Phase 223 participants (Actual)Interventional2012-09-30Terminated(stopped due to Komen Foundation Funding Terminated)
A Prospective, Open-label, Controlled, Randomized Study to Evaluate the Safety and Efficacy of Switching Calcineurin Inhibitor to Everolimus After 90 to 150 Days After Kidney Transplantation in Adults, Maintaining Corticosteroid and Mycophenolate Sodium C [NCT01455649]Phase 430 participants (Anticipated)Interventional2011-11-30Not yet recruiting
A 12-month, Multicenter, Open Label, Randomized, Controlled Study to Evaluate the Efficacy, Tolerability and Safety of Early Introduction of Everolimus, Reduced CNI, and Early Steroid Elimination Compared to Standard CNI, Mycophenolate Mofetil and Steroid [NCT01544491]Phase 3106 participants (Actual)Interventional2012-08-17Completed
Preservation of Renal Function After Liver Transplant for Patients With Pre-existing Chronic Kidney Disease or Peri-operative Acute Kidney Injury Using Everolimus Plus Mycophenolate Mofetil Immunosuppression Regimen [NCT04258423]Phase 34 participants (Actual)Interventional2019-12-19Terminated(stopped due to Study was larger than expected and became a burden to faculty and staff resources.)
Phase II Study of Everolimus (RAD001) in Children and Adults With Neurofibromatosis Type 2 [NCT01419639]Phase 210 participants (Actual)Interventional2011-10-31Completed
A Prospective, Randomized, Double-blind, Placebo-controlled Cross Over Study Investigating the Anti-epileptic Efficacy of Afinitor (Everolimus) in Patients With Refractory Seizures Who Have Focal Cortical Dysplasia Type II (FCD II) [NCT03198949]Phase 223 participants (Actual)Interventional2018-05-24Completed
An Open Label, Single-Centre, Pilot Study for Radiosensitization of Everolimus With External Beam Radiotherapy for the Treatment of Metastatic Neuroendocrine Liver Metastasis [NCT02205515]Phase 1/Phase 217 participants (Actual)Interventional2016-03-31Completed
Phase II Study of Everolimus in Patients With Thymoma and Thymic Carcinoma Previously Treated With Chemotherapy [NCT02049047]Phase 241 participants (Actual)Interventional2011-02-28Completed
Safety and Efficacy of Everolimus Transition in Minimizing Progressive Graft Dysfunction and Interstitial Fibrosis in Adult Kidney Transplant Recipients [NCT02096107]Phase 460 participants (Actual)Interventional2014-02-28Completed
Observationnal Multicenter Study on a Prospective Cohort of Kidney Transplanted Patients Receiving a Year After Transplant an Extended Releasing Tacrolimus-Everolimus Association [NCT03228576]16 participants (Actual)Observational2017-04-14Terminated(stopped due to No recruitment)
A Randomized Double-Blind Controlled Trial of Everolimus in Individuals With PTEN Mutations (RAD001XUS257T) [NCT02991807]Phase 1/Phase 246 participants (Actual)Interventional2017-06-12Completed
Phase 2 Study of the mTOR Inhibitor Everolimus in Combination With Bevacizumab in Patients With Sporadic and Neurofibromatosis Type 1 (NF1) Related Refractory Malignant Peripheral Nerve Sheath Tumors [NCT01661283]Phase 225 participants (Actual)Interventional2012-09-30Completed
Safer mTOR Inhibition for Human Geroprotection [NCT05949658]Phase 172 participants (Anticipated)Interventional2024-01-31Not yet recruiting
Study of Efficacy of Everolimus Combined With First-line Endocrine Therapy for HR+/HER2- (Open, Randomized, Phase II ) [NCT05949541]Phase 2265 participants (Anticipated)Interventional2023-07-31Not yet recruiting
A Phase III Randomized Open-label Multi-center Study of Ruxolitinib vs. Best Available Therapy in Patients With Corticosteroid-refractory Chronic Graft vs Host Disease After Allogeneic Stem Cell Transplantation (REACH3) [NCT03112603]Phase 3330 participants (Actual)Interventional2017-06-29Completed
A Phase 1/2 Study for the Safety, Efficacy, Pharmacokinetic and Pharmacodynamics Evaluation of Amcenestrant (SAR439859), Administered Orally as Monotherapy, Then in Combination With Other Anti-cancer Therapies in Postmenopausal Women With Estrogen Recepto [NCT03284957]Phase 1/Phase 2136 participants (Actual)Interventional2017-09-20Active, not recruiting
The Efficacy of Everolimus With Reduced-dose Tacrolimus Versus Reduced-dose Tacrolimus in Treatment of BK Virus Infection in Kidney Transplantation Recipient [NCT04542733]50 participants (Anticipated)Interventional2021-02-10Recruiting
Phase I Study of Everolimus + Bendamustine in Patients With Relapsed/Refractory Hematological Malignancies [NCT02240719]Phase 120 participants (Actual)Interventional2014-10-31Completed
A Phase I/II Study of RAD001, FOLFOX and Bevacizumab in Treatment of Colorectal Carcinoma [NCT01047293]Phase 1/Phase 247 participants (Actual)Interventional2010-05-31Completed
A Phase I Trial of VS-6766 (RO5126766) (a Dual RAF/MEK Inhibitor) Exploring Intermittent, Oral Dosing Regimens in Patients With Solid Tumours or Multiple Myeloma, With an Expansion to Explore Intermittent Dosing in Combination With Everolimus [NCT02407509]Phase 1104 participants (Anticipated)Interventional2013-06-17Recruiting
A Phase 1 Trial of Vandetanib (a Multi-Kinase Inhibitor of EGFR, VEGFR, and RET Inhibitor) in Combination With Everolimus (an mTOR Inhibitor) in Advanced Cancer [NCT01582191]Phase 1153 participants (Actual)Interventional2012-05-14Active, not recruiting
EVEREST: EVErolimus for Renal Cancer Ensuing Surgical Therapy, A Phase III Study [NCT01120249]Phase 31,545 participants (Actual)Interventional2011-04-01Active, not recruiting
"Extension Study to the Open-label Phase I Study Evaluating the Safety and Tolerability of Pasireotide LAR in Combination With Everolimus in Advanced Metastatic NETs - The COOPERATE-1 Study" [NCT01590199]Phase 118 participants (Actual)Interventional2012-05-18Completed
A Prospective, Open-label Study to Assess Safety of Certican in Kidney Transplant Patients [NCT01594268]Phase 454 participants (Actual)Interventional2012-03-31Completed
An Open-label, Multi-center, Expanded Access Study of Everolimus in Participants With Advanced Neuroendocrine Tumors (NETs) (Core Study) and an Extension Study to the Open-label, Multi-center, Expanded Access Study of Everolimus in Patients With Advanced [NCT01595009]Phase 4246 participants (Actual)Interventional2011-04-27Completed
Randomized, Multicenter, Open-label, Comparative Study of Efficacy and Safety of Treatment With a Calcineurin Inhibitor (CNI), Associating Myfortic ® and Neoral ® Compared to a CNI-free Treatment, Combining Myfortic ® and Certican ® , in Adult Patients Wi [NCT01595984]Phase 390 participants (Actual)Interventional2012-05-03Active, not recruiting
A Phase I Dose-Escalation Study of the BRAF Inhibitor Vemurafenib (Zelboraf®) in Combination With an mTOR Inhibitor, Everolimus (Afinitor®) or Temsirolimus (Torisel®), in Subjects With Advanced Cancer [NCT01596140]Phase 127 participants (Actual)Interventional2012-12-18Completed
Biological Medicine for Diffuse Intrinsic Pontine Glioma (DIPG) Eradication [NCT05476939]Phase 3368 participants (Anticipated)Interventional2022-09-29Recruiting
A Randomized Non-Comparative Multicenter Phase II Study of Everolimus/Sorafenib or Sunitinib in Patients With Metastatic Renal Cell Carcinoma (RCC) [NCT01616186]Phase 20 participants (Actual)Interventional2012-07-31Withdrawn
A Controlled Randomized Open-label Multicenter Study Evaluating if Early Conversion to Everolimus (Certican) From Cyclosporine (Neoral) in de Novo Renal Transplant Recipients Can Improve Long-term Renal Function and Slow Down the Progression of Chronic Al [NCT00634920]Phase 4204 participants (Actual)Interventional2008-03-31Completed
A Phase I/II Study of Sorafenib and Everolimus in Patients With Advanced Solid Tumors and Gemcitabine-Refractory Metastatic Pancreatic Cancer [NCT00981162]Phase 112 participants (Actual)Interventional2009-08-31Completed
A Multicenter, Open-label, Randomized, Phase I/II Clinical Trial Comparing Safety and Durable Overall Response Day 56 in Patients With Steroid Resistent Acute GvHD After Allogeneic Hematopoietic Stem Cell Transplant Treated With DSC or BAT [NCT04118556]Phase 1/Phase 250 participants (Anticipated)Interventional2021-12-01Recruiting
XIENCE PRIME SV Everolimus Eluting Coronary Stent Japan Post Marketing Surveillance [NCT02513719]312 participants (Actual)Observational2013-05-13Completed
Phase I Study Evaluating the Combination of Lapatinib (GW572016; NSC-727989) and Everolimus (RAD001) in Patients With Advanced Solid Tumors [NCT00352443]Phase 166 participants (Actual)Interventional2006-09-30Completed
Randomized Comparison of Dual Drug-Eluting Cilotax Stent and Everolimus -Eluting Stent Implantation for DE Novo Coronary Artery DisEase in Patients With DIABETES Mellitus [NCT01515228]Phase 4291 participants (Actual)Interventional2012-01-31Completed
A Pilot Study to Investigate Pharmacokinetic Characteristics of Everolimus in Patients Treated With Tacrolimus-Based Immunosuppression in De Novo Kidney Transplantation [NCT00325325]Phase 240 participants Interventional2006-01-31Recruiting
A Phase I/II Study of RAD001 and AV-951 in Patients With Refractory, Metastatic Colorectal Cancer [NCT01058655]Phase 1/Phase 256 participants (Actual)Interventional2010-02-28Completed
Phase II Study of RAD001 for Treatment of Refractory, Recurrent, Locally Advanced Squamous Cell Carcinoma of the Head and Neck [NCT01051791]Phase 213 participants (Actual)Interventional2010-01-31Terminated
Expanded Access to Everolimus, for an Individual Patient With Uterine Sarcoma (CTMS#18-0020) [NCT03493165]0 participants Expanded AccessAvailable
A Phase II Trial of Everolimus for Cancer Patients With Inactivating Mutations in TSC1 or TSC2 or Activating MTOR Mutations [NCT02201212]Phase 230 participants (Actual)Interventional2014-09-30Completed
A Prospective, Single Blind, Multi-center, Randomized Trial to Compare the TAXUS Element™ Coronary Stent System Against the XIENCE Prime™ Coronary Stent System in the Treatment of a Diabetic Patient PopulatiOn in India [NCT03125772]1,830 participants (Actual)Interventional2011-06-30Completed
Phase II Open Label Study of Everolimus in Combination With Anti-estrogen Therapy in Hormone Receptor-Positive HER2-Negative Advanced Breast Cancer [NCT02291913]Phase 248 participants (Actual)Interventional2014-12-18Completed
A Phase II Trial of Mutation-Targeted Therapy With Sunitinib or Everolimus in Patients With Advanced Low-or Intermediate Grade Neuroendocrine Tumors of the Gastrointestinal Tract and Pancreas With or Without Cytoreductive Surgery [NCT02315625]Phase 216 participants (Actual)Interventional2015-04-08Terminated(stopped due to Study closed due to poor accrual.)
A Phase II Study of Letrozole and RAD001 (Everolimus) in Patients With Advanced or Recurrent Endometrial Cancer [NCT01068249]Phase 242 participants (Actual)Interventional2010-04-30Active, not recruiting
An Open Label, Multi-center Everolimus Roll-over Protocol for Patients Who Have Completed a Previous Novartis-sponsored Everolimus Study and Are Judged by the Investigator to Benefit From Continued Everolimus Treatment. [NCT02017860]Phase 24 participants (Actual)Interventional2014-01-16Completed
12 Month, Multi-center, Open-label, Prospective, Randomized, Parallel Group Study Investigating a Standard Regimen in de Novo Kidney Transplant Patients Versus a Certican® Based Regimen Either in Combination With Cyclosporin A or Tacrolimus [NCT01843348]Phase 3612 participants (Actual)Interventional2012-12-27Completed
An Open-label, Multi-center Everolimus Roll-over Protocol for Patients Who Have Completed a Previous Novartis-sponsored Everolimus Study and Are Judged by the Investigator to Benefit From Continued Everolimus Treatment [NCT01789281]Phase 434 participants (Actual)Interventional2013-05-14Completed
De Novo Everolimus Versus Tacrolimus in Combination With Mofetil Mycophenolate and Low Dose Corticosteroids to Reduce Tacrolimus Induced Nephrotoxicity in Liver Transplantation: a Prospective, Multicentric, Randomised Study [NCT02909335]Phase 30 participants (Actual)Interventional2016-11-30Withdrawn
A Randomized Prospective Trial of Conversion to Everolimus Therapy From Calcineurin Inhibitor Based Maintenance Immunosuppression in Association With Mycophenolic Acid in Liver Transplantation: Examination of Impact on Long Term Renal Function. [NCT01936519]24 participants (Actual)Interventional2013-12-16Completed
The Safety of Simvastatin (SOS) in Patients With Pulmonary Lymphangioleiomyomatosis (LAM) and With Tuberous Sclerosis Complex (TSC) [NCT02061397]Phase 1/Phase 210 participants (Actual)Interventional2014-03-31Completed
Phase II Trial of Ribociclib in Combination With Everolimus in Advanced Dedifferentiated Liposarcoma (DDL) and Leiomyosarcoma (LMS) [NCT03114527]Phase 248 participants (Actual)Interventional2017-08-08Active, not recruiting
Randomized, Double Blind, Multicentric Phase III Trial Evaluating the Safety and Benefit of Adding Everolimus to Adjuvant Hormone Therapy in Women With High Risk of Relapse, ER+ and HER2- Primary Breast Cancer Who Remain Free of Disease After Receiving at [NCT01805271]Phase 31,278 participants (Actual)Interventional2013-03-31Active, not recruiting
A Randomized, Double-Blinded, Placebo-Controlled Phase II Study of Adjuvant Everolimus Following the Resection of Metastatic Pancreatic Neuroendocrine Tumors to the Liver [NCT02031536]Phase 22 participants (Actual)Interventional2014-04-10Terminated(stopped due to Slow accrual)
A Phase I Study of Sorafenib and RAD001 in Patients With Metastatic Renal Cell Carcinoma [NCT00384969]Phase 121 participants (Actual)Interventional2006-10-31Completed
Impact of Everolimus on HIV Persistence Post Kidney (and Kidney/Pancreas) or Liver Transplant [NCT02429869]Phase 410 participants (Actual)Interventional2016-02-24Completed
Once-daily Regimen With Envarsus® to Optimize Immunosuppression Management and Outcomes in Kidney Transplant Recipients [NCT02954198]40 participants (Actual)Interventional2016-12-01Completed
Phase I/II Trial of Sorafenib (Nexavar) and RAD001 (Everolimus)in the Treatment of Patients With Advanced Clear Cell Renal Cell Carcinoma [NCT00392821]Phase 1/Phase 278 participants (Actual)Interventional2006-12-31Completed
Genetically-informed Therapy for ER+ Breast Cancer in a Post-CDK4/6 Inhibitor Setting: a Phase II Umbrella Study (GERTRUDE) [NCT05933395]Phase 2135 participants (Anticipated)Interventional2023-10-10Recruiting
Continuous Access to Advanced and Metastatic Renal Cell Carcinoma Therapy With Everolimus Post Pazopanib Treatment [NCT01545817]Phase 274 participants (Actual)Interventional2012-04-19Terminated(stopped due to As the therapeutic landscape in renal cell carcinoma is changing, it has become apparent that information gained so far by CATChEz study is sufficient.)
Effect of the Introduction of mTor Inhibitors in the Activation of the Cytomegalovirus (CMV) -Specific Cellular Immune Response to Control Viral Replication in Kidney Transplant Patients [NCT04936971]Phase 446 participants (Anticipated)Interventional2021-09-30Not yet recruiting
A Randomized, Multicenter Study of Belatacept, Everolimus, rATG and Early Steroid Withdrawal Versus Belatacept, Mycophenolate, rATG and Chronic Steroids in Renal Transplantation [NCT04849533]Phase 4120 participants (Anticipated)Interventional2021-04-09Recruiting
Evaluation of WSS and Neointimal Healing Following Percutaneous Coronary Intervention of Angulated Vessels With Resolute® Integrity Zotarolimus Eluting Coronary Stent Compared to XIENCE Xpedition® Everolimus Eluting Coronary Stent [NCT02098876]86 participants (Actual)Interventional2014-05-31Completed
Fulvestrant Followed by Everolimus Plus Exemestane vs Examestane and Everolimus Followed by Fulvestrant in Postmenopausal Women With HR+ and HER2- Locally Advanced (LABC) or Metastatic Breast Cancer (MBC) Previously Treated With NSAI [NCT02404051]Phase 3745 participants (Anticipated)Interventional2015-12-31Recruiting
A Randomized, Open-label Phase II Multicenter Study Evaluating the Efficacy of Oral Everolimus Alone or in Combination With Pasireotide LAR i.m. in Advanced Progressive Pancreatic Neuroendocrine Tumors (PNET) - The COOPERATE-2 Study [NCT01374451]Phase 2160 participants (Actual)Interventional2011-06-30Terminated(stopped due to The study was stopped for not meeting the primary endpoint for PFS.)
Evaluation of Oral Care to Prevent Oral Mucositis in Estrogen Receptor Positive Metastatic Breast Cancer Patients Treated With Everolimus: Phase III Randomized Control Trial [NCT02376985]Phase 3174 participants (Actual)Interventional2015-03-26Completed
GCC 0901- A Phase II Study of Letrozole in Combination With Lapatinib Followed by an Addition of Everolimus in Postmenopausal Women With Advanced Endocrine Resistant Breast Cancer [NCT01499160]Phase 27 participants (Actual)Interventional2012-05-31Terminated(stopped due to low accrual)
Mechanisms and Treatment of Chronic Allograft Injury (CAI) Due to Calcineurin Inhibitor (CNI) Toxicity [NCT01473732]2 participants (Actual)Interventional2012-03-31Terminated(stopped due to terminated after 2 patients due to difficulty in enrollment)
An Open-label, Multicenter Phase II Study to Examine the Efficacy and Safety of Everolimus as Second-line Therapy in the Treatment of Patients With Metastatic Renal Cell Carcinoma [NCT01491672]Phase 2134 participants (Actual)Interventional2011-11-30Completed
UPCC 36315 A Phase II Study Of Everolimus (RAD001) And Lenvatinib (E7080) In Patients With Metastatic Differentiated Thyroid Cancer Who Have Progressed on Lenvatinib Alone [NCT03139747]Phase 25 participants (Actual)Interventional2017-04-03Suspended(stopped due to lack of accrual reevaluating feasibility)
A Phase I/II Study of RAD001 in Advanced Hepatocellular Carcinoma [NCT00516165]Phase 1/Phase 228 participants (Actual)Interventional2007-08-31Completed
Exploratory Study of Avastin (Bevacizumab) and RAD001 (Everolimus) in Advanced Low or Intermediate Grade Neuroendocrine Carcinoma (AVF3961s) (CRAD001C2481) [NCT00607113]Phase 241 participants (Actual)Interventional2008-01-31Completed
A 2-arm, Prospective, Randomized, Controlled, Open-label, 12 Month Phase III Trial to Evaluate the Efficacy Regarding Renal Function of Everolimus in Combination With a Centre Specific Standard Immunosuppressive Regimen Consisting of CNI, Purinantagonists [NCT01404325]Phase 3130 participants (Actual)Interventional2012-02-01Completed
A Phase II Study to Evaluate the Efficacy of RAD001 in Metastatic Non-clear Cell Renal Cell Carcinoma [NCT00830895]Phase 249 participants (Actual)Interventional2009-01-31Completed
A Randomized Phase II Study of Afinitor (RAD001) vs. Sutent (Sunitinib) in Patients With Metastatic Non-Clear Cell Renal Cell Carcinoma (ASPEN) [NCT01108445]Phase 2131 participants (Actual)Interventional2010-09-30Completed
An Open Label, randomIzed Controlled Prospective Multicenter Two Arm Phase IV Trial to Determine Patient Preference for Everolimus in Combination With Exemestane or Capecitabine in Combination With Bevacizumab for Advanced (Inoperable or Metastatic) HER2- [NCT02248571]Phase 485 participants (Actual)Interventional2014-08-31Completed
A Pilot Study To Evaluate The Effects of Everolimus on Brain mTOR Activity and Cortical Hyperexcitability in TSC and FCD [NCT02451696]Phase 215 participants (Actual)Interventional2014-01-31Completed
A Randomized Phase II Trial of Everolimus and Letrozole or Hormonal Therapy (Tamoxifen/Medroxyprogesterone Acetate) in Women With Advanced, Recurrent, or Persistent Endometria Carcinoma [NCT02228681]Phase 274 participants (Actual)Interventional2015-02-28Active, not recruiting
Treatment of Relapsed/Refractory Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL) With Everolimus (RAD001) and Alemtuzumab: A Phase I/II Study [NCT00935792]Phase 1/Phase 228 participants (Actual)Interventional2009-07-31Completed
Randomized Phase II Trial of Everolimus Versus Placebo as Adjuvant Therapy in Patients With Locally Advanced Squamous Cell Cancer of the Head and Neck (SCCHN) [NCT01111058]Phase 252 participants (Actual)Interventional2010-04-30Terminated(stopped due to Slow accrual)
Healing Responses After Treatment of Bare Metal Stent Restenosis With Implantation of an Everolimus-eluting Xience V Stent Versus Use of a Paclitaxel-eluting Balloon: Optical Coherence Tomography Study [NCT01065532]50 participants (Anticipated)Interventional2009-05-31Completed
A Phase I/II Drug Withdrawal Study of Alloantigen-Specific Tregs in Liver Transplantation [NCT03654040]Phase 1/Phase 230 participants (Actual)Interventional2021-04-22Terminated(stopped due to The study was terminated by the Sponsor prior to any participants receiving the investigational product.)
Long-term Follow-up Study to Monitor the Growth and Development of Pediatric Patients Previously Treated With Everolimus in Study CRAD001M2301 [NCT02338609]Phase 415 participants (Actual)Interventional2014-12-17Active, not recruiting
A Phase II Study of the mTOR Inhibitor RAD001 in Previously Treated Patients With Unresectable or Metastatic Adenocarcinoma of the Esophagus and Stomach [NCT00985192]Phase 249 participants (Actual)Interventional2009-09-30Completed
An Exploratory Evaluation of Early Use of Everolimus (EVE) on Tacrolimus (TAC)-Based Immunosuppressive Regiment vs. Mycophenolate Sodium (MPS) on Cytomegalovirus (CMV) Infection in Renal Transplant Recipients. [NCT01927588]Phase 450 participants (Anticipated)Interventional2013-08-31Not yet recruiting
A Prospective, Randomized, Placebo-controlled, Multicenter, International Phase I/II Trial of RAD001 (Everolimus) in Combination With DHAP as Induction Therapy in Patients With Relapsed or Refractory Hodgkin Lymphoma (HL) [NCT01453504]Phase 1/Phase 273 participants (Actual)Interventional2012-08-31Completed
A Randomized, Double-blind, Multicenter, Phase III Study of Everolimus (RAD001) Plus Best Supportive Care Versus Placebo Plus Best Supportive Care in the Treatment of Patients With Advanced NET of GI or Lung Origin [NCT01524783]Phase 3302 participants (Actual)Interventional2012-03-30Completed
Signal TrAnsduction Pathway Activity Analysis in OVarian cancER [NCT03458221]Phase 2/Phase 3148 participants (Anticipated)Interventional2023-01-31Recruiting
Phase II Study of Single Agent RAD001 in Patients With Colon Cancer and Activating Mutations in the PI3KCA Gene [NCT00390364]Phase 21 participants (Actual)Interventional2006-10-31Terminated(stopped due to Withdrawn due to low accrual)
Phase I/II Trial of RAD001 Plus Docetaxel in Patients With Metastatic or Recurrent Non-Small Cell Lung Cancer [NCT00406276]Phase 1/Phase 228 participants (Actual)Interventional2006-11-30Terminated(stopped due to data analysis showed insufficient drug efficacy)
Molecular Profiling-Based Assignment of Cancer Therapy for Patients With Advanced Solid Tumors [NCT01827384]Phase 2208 participants (Actual)Interventional2014-01-07Completed
A Phase II Study of Sporadic Angiomyolipomas (AMLs) Growth Kinetics While on Everolimus Therapy [NCT02539459]Phase 220 participants (Actual)Interventional2015-09-23Terminated(stopped due to Extreme toxicity, met toxicity stopping rules)
Randomised Comparison of Two Different Immunosuppressive Regimens on Progression of Arteriosclerosis in Renal Transplant Patients. [NCT01276834]Phase 413 participants (Actual)Interventional2010-09-30Terminated(stopped due to insufficient patients enrolled)
A Phase 1 Evaluation of the Safety and Tolerability of Niraparib in Combination With Everolimus in Advanced Gynecologic Malignancies and Breast Cancer [NCT03154281]Phase 124 participants (Anticipated)Interventional2017-07-17Active, not recruiting
A Phase IV Multicentre, Open Label Study of Postmenopausal Women With Oestrogen Receptor Positive Locally Advanced or Metastatic Breast Cancer Treated With Everolimus (RAD001) in Combination With Exemestane, With Exploratory Epigenetic Marker Analysis [NCT01743560]Phase 452 participants (Actual)Interventional2013-01-31Completed
A Phase 1b, Open-label, Dose-escalation Study to Evaluate the Safety and Tolerability of SGN-75 in Combination With Everolimus in Patients With CD70-positive Metastatic Renal Cell Carcinoma [NCT01677390]Phase 14 participants (Actual)Interventional2012-08-31Terminated
Multicenter, Prospective, Open-label, Controlled, Randomized, Parallel Groups Study to Evaluate the Renal Function of Adult Liver Transplant Recipients Treated With Two Everolimus-based Immunosuppressive Regimens (Tacrolimus Withdrawal vs. Minimization) U [NCT02115113]Phase 378 participants (Actual)Interventional2014-03-28Completed
A Phase I/II Trial to Assess the Tolerability of RAD 001 With Gefitinib in Patients With Glioblastoma Multiforme and Prostate Cancer and Efficacy in Patients With Castrate Metastatic Prostate Cancer [NCT00085566]Phase 1/Phase 261 participants (Actual)Interventional2004-03-31Completed
A Prospective, Randomised, Controlled, Open-label, Multicentre Study to Evaluate Efficacy, Safety and Patient-Reported Outcomes of Peptide Receptor Radionuclide Therapy (PRRT) With 177Lu-Edotreotide Compared to Best Standard of Care in Patients With Well- [NCT04919226]Phase 3202 participants (Anticipated)Interventional2021-12-21Recruiting
A Phase I/II Drug Withdrawal Study of Alloantigen-Specific Tregs in Liver Transplantation (ITN073ST) [NCT03577431]Phase 1/Phase 29 participants (Anticipated)Interventional2019-03-29Active, not recruiting
Ten-year Follow-up of the Clinical Evaluation of Everolimus-Eluting Coronary Stents in the Treatment of Patients With ST-segment Elevation Myocardial Infarction: EXAMINATION-EXTEND Trial [NCT04462315]1,498 participants (Actual)Observational2015-05-01Completed
A Prospective, Pilot Trial to Evaluate Safety and Tolerability of Everolimus for the Prevention of BK and CMV Viremia in HLA Sensitized Kidney Transplant Recipients [NCT01911546]Phase 220 participants (Actual)Interventional2013-06-30Completed
Evaluate the Continued Safety and Effectiveness of the XIENCE PRIME EECSS in a Cohort of Real-world Patients Receiving the XIENCE PRIME EECSS During Commercial Use. [NCT01894152]2,002 participants (Actual)Observational2013-07-31Completed
Phase II Study of RAD001 in Previously Treated Patients With Metastatic Pancreatic Cancer [NCT00409292]Phase 233 participants (Actual)Interventional2007-01-31Completed
PLATINUM: A Prospective, Randomized, Multicenter Trial to Assess an Everolimus-Eluting Coronary Stent System (PROMUS Element™) for the Treatment of up to Two De Novo Coronary Artery Lesions - Pharmacokinetics Sub-trial [NCT01510327]Phase 322 participants (Actual)Interventional2009-10-31Completed
Phase I/II Study of RAD001 in Combination With Temozolomide in Patients With Advanced Pancreatic Neuroendocrine Tumors [NCT00576680]Phase 1/Phase 243 participants (Actual)Interventional2008-05-31Completed
Treatment of Refractory Metastatic Renal Cell Carcinoma With Bevacizumab and RAD001 (Everolimus) [NCT00651482]Phase 210 participants (Actual)Interventional2008-08-31Terminated(stopped due to slow accrual)
Phase I/IB Trial of Eribulin and Everolimus in Patients With Triple Negative Metastatic Breast Cancer [NCT02120469]Phase 127 participants (Actual)Interventional2014-10-01Completed
A Phase II Study of RAD001 (Everolimus) in Patients With Fluoropyrimidine and Platinum-refractory Advanced Gastric Cancer [NCT00729482]Phase 254 participants (Actual)Interventional2008-07-31Completed
A 24-month, Single Center, Pilot, Open Label, Controlled Trial to Evaluate the Efficacy and Safety of Calcineurin-inhibitor Reduction With Conversion at 2 Months to Reduced Dose Tacrolimus/Everolimus in Adult Renal Transplant Recipients Following Campath® [NCT01935128]Phase 455 participants (Actual)Interventional2013-07-03Completed
A 3-month, Multicenter, Randomized, Open Label Study to Evaluate the Impact of Early Versus Delayed Introduction of Everolimus on Wound Healing in de Novo Kidney Transplant Recipients With a Follow-up Evaluation at 12 Month After Transplant (NEVERWOUND St [NCT01410448]Phase 3383 participants (Actual)Interventional2011-11-30Completed
Everolimus and Temozolomide as 1-line Treatment in Advanced Gastroenteropancreatic Neuroendocrine Carcinoma (G3) With a Ki67 of 20-55% [NCT02248012]Phase 238 participants (Actual)Interventional2014-12-31Completed
Safety and Efficacy of Mycophenolic Acid Withdrawal With Conversion to Zortress (Everolimus) in Renal Transplant Recipients With BK Virus Infection [NCT01624948]Phase 440 participants (Actual)Interventional2012-09-30Completed
MEK114375: A Rollover Study to Provide Continued Treatment With GSK1120212 to Subjects With Solid Tumors or Leukemia [NCT01376310]Phase 2159 participants (Actual)Interventional2010-11-02Terminated(stopped due to Company Decision)
Randomized Comparison of the Biolimus-eluting Biomatrix Neoflex™ og Everolimus-eluting SYNERGY™ Stents in All-comer Patients With Ischemic Heart Disease - The OCT SORT-OUT VIII [NCT02253108]160 participants (Anticipated)Interventional2014-05-31Active, not recruiting
A Phase I/II Study of X-82, an Oral Anti-VEGFR Tyrosine Kinase Inhibitor, With Everolimus for Patients With Pancreatic Neuroendocrine Tumors [NCT01784861]Phase 1/Phase 223 participants (Actual)Interventional2013-05-03Terminated(stopped due to Pharmaceutical company decision)
Phase I/II Evaluation of Everolimus (RAD001), Radiation and Temozolomide (TMZ) Followed by Adjuvant Temozolomide and Everolimus in Newly Diagnosed Glioblastoma [NCT00553150]Phase 1/Phase 2122 participants (Actual)Interventional2009-03-31Completed
Comparison of the Efficacy and Safety of Sirolimus, Everolimus or Mycophenolate in Renal Transplant Recipients Receiving Induction With Anti-thymocyte Globulin, Tacrolimus and Prednisone [NCT03468478]Phase 41,209 participants (Actual)Interventional2017-06-18Completed
Randomized, Double-Blind, Placebo-Controlled Phase II Trial of Fulvestrant (Faslodex) Plus Everolimus in Post-Menopausal Patients With Hormone-Receptor Positive Metastatic Breast Cancer Resistant to Aromatase Inhibitor Therapy [NCT01797120]Phase 2131 participants (Actual)Interventional2013-05-31Completed
Multicenter 3-arm Trial to Evaluate the Efficacy and Safety of Pasireotide LAR or Everolimus Alone or in Combination in Patients With Well Differentiated Neuroendocrine Carcinoma of the Lung and Thymus - LUNA Trial [NCT01563354]Phase 2124 participants (Actual)Interventional2013-08-16Completed
A 24-month Multi-center, Open-label, Randomized, Controlled Study to Evaluate the Evolution of Renal Function in Maintenance Liver Transplant Recipients Receiving Either RAD001 (Everolimus) Plus Reduced TAC or RAD001 (Everolimus) Plus Mycophenolate Mofeti [NCT03596970]Phase 30 participants (Actual)Interventional2015-09-30Withdrawn
Biological Medicine for Diffuse Intrinsic Pontine Glioma (DIPG) Eradication [NCT02233049]Phase 2250 participants (Anticipated)Interventional2014-10-31Recruiting
Phase II Evaluation of Real-Time, Pharmacokinetically Guided Everolimus in Patients With Hormone Receptor Positive Breast Cancer, Pancreatic Neuroendocrine Tumors (PNET), and Renal Cell Carcinoma [NCT02273752]Phase 22 participants (Actual)Interventional2014-11-30Terminated(stopped due to Slow accrual)
A Prospective Cohort, Open, Phase II Clinical Study of Chidamide/Everolimus Combined With Endocrine Therapy for PIK3CA Wild-type/Mutant Hormone Receptor Positive/Human Epidermal Growth Factor Receptor 2 Negative Advanced Breast Cancer [NCT05983107]Phase 2102 participants (Anticipated)Interventional2023-07-20Recruiting
A Multi-institutional Phase I and Biomarker Study of Everolimus Added to Combined Hormonal and Radiation Therapy for High Risk Prostate Cancer [NCT01642732]Phase 11 participants (Actual)Interventional2012-10-31Terminated(stopped due to Lack of accrual and funding expires in June, 2014.)
Randomized, Open-Label Trial of Tacrolimus/Everolimus vs. Tacrolimus/Enteric-Coated Mycophenolate Sodium to Prevent Biopsy-Proven Acute Rejection and Chronic Allograft Injury in Adult, Primary Kidney Transplantation [NCT01680861]Phase 332 participants (Actual)Interventional2012-11-30Completed
A Phase II Study of Concurrent Radiation Therapy, Temozolomide, and Bevacizumab Followed by Bevacizumab/Everolimus in the First-line of Treatment of Patients With Glioblastoma Multiforme [NCT00805961]Phase 268 participants (Actual)Interventional2009-01-31Completed
A Phase I/II, Single Arm, Open-label Study of Ribociclib in Combination With Everolimus + Exemestane in the Treatment of Men and Postmenopausal Women With HR+, HER2- Locally Advanced or Metastatic Breast Cancer Following Progression on a CDK 4/6 Inhibitor [NCT02732119]Phase 1/Phase 2104 participants (Actual)Interventional2016-06-14Completed
A Multicenter, Dose-finding Study to Determine if Oral BEZ235 Alone or in Combination With RAD001 Decreases the Incidence of Respiratory Tract Infections in Elderly Subjects [NCT03373903]Phase 2652 participants (Actual)Interventional2017-11-15Active, not recruiting
Precision Treatment of Refractory Triple Negative Breast Cancer Based on Molecular Subtyping --FUSCC-TNBC- Umbrella Trial [NCT03805399]Phase 1/Phase 2140 participants (Anticipated)Interventional2018-10-18Recruiting
Fulvestrant Alone Versus Fulvestrant and Everolimus Versus Fulvestrant, Everolimus and Anastrozole: A Phase III Randomized Placebo-Controlled Trial in Postmenopausal Patients [NCT02137837]Phase 337 participants (Actual)Interventional2014-05-31Terminated(stopped due to lack of accrual)
Japan-USA Harmonized Assessment by Randomized, Multi-Center Study of OrbusNEich's Combo StEnt (Japan-USA HARMONEE): Assessment of a Novel DES Platform For Percutaneous Coronary Revascularization in Patients With Ischemic Coronary Disease and NSTEMI Acute [NCT02073565]572 participants (Actual)Interventional2014-02-28Completed
A National, Multi-center, Randomized, Open Label Study to Evaluate the Efficacy and Safety of Everolimus Combined With Enteric-coated Mycophenolate Sodium Compared to the Standard Treatment Combining Tacrolimus and Enteric-coated Mycophenolate Sodium in d [NCT01625377]Phase 3188 participants (Actual)Interventional2012-12-31Completed
A 24-month, Multi-center, Single Arm, Prospective Study to Evaluate Renal Function, Efficacy, Safety and Tolerability of Everolimus in Combination With Reduced Exposure Cyclosporine or Tacrolimus in Paediatric Liver Transplant Recipients. [NCT01598987]Phase 356 participants (Actual)Interventional2012-10-31Completed
A Phase II, Single Arm Study of the Use of Steroid-based Mouthwash to Prevent Stomatitis in Postmenopausal Women With Advanced or Metastatic Hormone Receptor Positive Breast Cancer Being Treated With Everolimus Plus Exemestane [NCT02069093]Phase 292 participants (Actual)Interventional2014-05-31Completed
A Prospective, Randomised, Controlled, Open-label, Multicentre Phase III Study to Evaluate Efficacy and Safety of Peptide Receptor Radionuclide Therapy (PRRT) With 177Lu-Edotreotide Compared to Targeted Molecular Therapy With Everolimus in Patients With I [NCT03049189]Phase 3309 participants (Actual)Interventional2017-02-02Active, not recruiting
A Two-period, Multicenter, Randomized, Open-label, Phase II Study Evaluating the Clinical Benefit of a Maintenance Treatment Targeting Tumor Molecular Alterations in Patients With Progressive Locally-advanced or Metastatic Solid Tumors [NCT02029001]Phase 2560 participants (Anticipated)Interventional2014-03-31Recruiting
A Phase I Study of RAD001 and Sunitinib in Metastatic Renal Cell Carcinoma Patients [NCT00422344]Phase 120 participants (Actual)Interventional2006-10-31Completed
A Randomized, Open-label, Single-dose, Two-way Crossover Clinical Trial to Investigate the Pharmacokinetic and Safety After Oral Administration of SVG101(Dispersible Tablet of Everolimus) 5mg and Afinitor 5mg in Healthy Adults [NCT05490095]Phase 126 participants (Actual)Interventional2021-07-23Completed
A 24 Month, Randomized, Controlled, Study to Evaluate the Efficacy and Safety of Concentration-controlled Everolimus Plus Reduced Tacrolimus Compared to Standard Tacrolimus in Recipients of Living Donor Liver Transplants and Long Term Extension to Evaluat [NCT01888432]Phase 3285 participants (Actual)Interventional2013-09-25Completed
Donor-Alloantigen-Reactive Regulatory T Cell (darTreg) Therapy in Liver Transplantation (RTB-002) [NCT02188719]Phase 115 participants (Actual)Interventional2014-12-17Terminated(stopped due to The trial could not be completed within the grant timeline.)
Randomized Phase II Study of BEZ235 or Everolimus in Advanced Pancreatic Neuroendocrine Tumors [NCT01628913]Phase 262 participants (Actual)Interventional2012-10-31Terminated(stopped due to This trial was terminated based on an interim analysis as BEZ235 did not demonstrate a progression free survival advantage to everolimus treatment.)
A Three-arm, Randomized, Open Label, Phase II Study of Everolimus in Combination With Exemestane Versus Everolimus Alone Versus Capecitabine in the Treatment of Postmenopausal Women With Estrogen Receptor Positive, Locally Advanced, Recurrent, or Metastat [NCT01783444]Phase 2309 participants (Actual)Interventional2013-02-26Completed
Phase II Study of Everolimus Beyond Progression in Postmenopausal Women With Advanced, Hormone Receptor Positive Breast Cancer [NCT02269670]Phase 23 participants (Actual)Interventional2014-11-25Terminated(stopped due to Slow to accrual)
A Three-arm, Randomized, Double-blind, Placebo-controlled Study of the Efficacy and Safety of Two Trough-ranges of Everolimus as Adjunctive Therapy in Patients With Tuberous Sclerosis Complex (TSC) Who Have Refractory Partial-onset Seizures [NCT01713946]Phase 3366 participants (Actual)Interventional2013-04-29Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00085566 (1) [back to overview]Overall Objective Response
NCT00087685 (2) [back to overview]Clinical Benefit Rate
NCT00087685 (2) [back to overview]Number of Participants With Objective Response Plus Stable Disease Rate (CR + PR + SD)
NCT00096486 (1) [back to overview]Overall Objective Response
NCT00113360 (1) [back to overview]Progression Free Survival (PFS)
NCT00154284 (4) [back to overview]Calculated Creatinine Clearance at 6 Month and 12 Month
NCT00154284 (4) [back to overview]Number of Participants With Biopsy-proven Acute Rejection (BPAR) Episodes, Graft Loss, Death or Loss to Follow-up
NCT00154284 (4) [back to overview]Renal Function Measured by Calculated Glomerular Filtration Rate (GFR Calculated According to the Nankivell Formula)
NCT00154284 (4) [back to overview]Serum Creatinine at Month 6 and 12
NCT00154297 (6) [back to overview]Number of Participants With Any Wound Healing Disorder During the 12-month Treatment Period
NCT00154297 (6) [back to overview]Number of Participants Considered in Failure for the Primary Failure Endpoint at 6 Months Post-transplantation.
NCT00154297 (6) [back to overview]Number of Participants Considered in Failure for the Primary Failure Endpoint at 3 Months
NCT00154297 (6) [back to overview]Number of Participants Considered in Failure for the Primary Failure Endpoint at 12 Months Post-transplantation.
NCT00154297 (6) [back to overview]Number of Participants Who Underwent Any Dialysis Within the 12-month Treatment Period
NCT00154297 (6) [back to overview]Duration of Dialysis
NCT00154310 (5) [back to overview]Number of Participants With Occurrence of Treatment Failures
NCT00154310 (5) [back to overview]Number of Participants With Occurrence of Biopsy Proven Acute Rejection (BPAR), Graft Loss or Death
NCT00154310 (5) [back to overview]Number of Participants Who Experienced an Adverse Event or Serious Adverse Event
NCT00154310 (5) [back to overview]Changes in Cardiovascular Risk From Month 4.5 to Final Assessment at Month 12
NCT00154310 (5) [back to overview]Renal Function (Nankivell Formula) at Month 12 Post Transplantation.
NCT00170846 (3) [back to overview]Number of Participants With Safety Parameters
NCT00170846 (3) [back to overview]Change in mGFR by Baseline Calculated Creatinine Clearance (Cockcroft-Gault Formula)
NCT00170846 (3) [back to overview]Renal Function Assessed by Measured GFR (mGFR)
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00180479 (44) [back to overview]% Volume Obstruction (% VO)
NCT00180479 (44) [back to overview]Acute Success: Clinical Device
NCT00180479 (44) [back to overview]Acute Success: Clinical Procedure
NCT00180479 (44) [back to overview]Distal Late Loss
NCT00180479 (44) [back to overview]In-segment % Angiographic Binary Restenosis (% ABR) Rate
NCT00180479 (44) [back to overview]In-segment % Diameter Stenosis (% DS)
NCT00180479 (44) [back to overview]In-stent % Angiographic Binary Restenosis (% ABR) Rate
NCT00180479 (44) [back to overview]In-stent % Diameter Stenosis (% DS)
NCT00180479 (44) [back to overview]In-stent Late Loss
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event(MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Major Adverse Cardiac Event (MACE)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00180479 (44) [back to overview]Major Secondary Endpoint: Ischemia Driven Target Vessel Failure (ID-TVF)
NCT00180479 (44) [back to overview]Persisting Incomplete Stent Apposition, Late-acquired Incomplete Stent Apposition, Aneurysm, Thrombosis, and Persisting Dissection
NCT00180479 (44) [back to overview]Primary Endpoint: In-segment Late Loss (LL)
NCT00180479 (44) [back to overview]Proximal Late Loss
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00180479 (44) [back to overview]Target Vessel Failure (TVF)
NCT00251004 (4) [back to overview]Number of Participants With Composite Efficacy Endpoints - 12 Month Analysis
NCT00251004 (4) [back to overview]Percentage of Participants With the Composite Incidence of Graft Loss, Death or Loss to Follow up at 12 Months Post-transplantation
NCT00251004 (4) [back to overview]Non-inferiority Analysis of Renal Function, Calculated by Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula
NCT00251004 (4) [back to overview]Non-inferiority Analysis on Percentage of Participants With Composite Efficacy Endpoints
NCT00267189 (3) [back to overview]Percentage of Patients With Efficacy Failure (Biopsy Proven Acute Rejection [BPAR], Graft Loss or Death)
NCT00267189 (3) [back to overview]Mean Change From Baseline in Cockcroft-Gault Calculated Creatinine Clearance (CrCl)
NCT00267189 (3) [back to overview]Number of Patients With Discontinuation of Study Medication
NCT00300274 (10) [back to overview]Percentage of Participants With Biopsy-proven Acute Rejection (BPAR of ISHLT Grade ≥ 3A), Acute Rejection (AR) Associated With Hemodynamic Compromise (HDC), Graft Loss/Re-transplant and Death at Month 24
NCT00300274 (10) [back to overview]Renal Function Measured by Glomerular Filtration Rate (GFR) at 12 Months
NCT00300274 (10) [back to overview]Renal Function Calculated by Glomerular Filtration Rate (GFR) at 24 Months
NCT00300274 (10) [back to overview]Percentage of Participants With Graft Loss/Re-transplant, Death or Loss to Follow-up at 24 Months
NCT00300274 (10) [back to overview]Percentage of Participants With Graft Loss/Re-transplant, Death or Loss to Follow-up at 12 Months
NCT00300274 (10) [back to overview]Percentage of Participants With Composite Efficacy Failure at 24 Months
NCT00300274 (10) [back to overview]Percentage of Participants With Composite Efficacy Failure at 12 Months
NCT00300274 (10) [back to overview]Percentage of Participants With Cardiac Allograft Vasculopathy (CAV) at Month 12
NCT00300274 (10) [back to overview]Percentage of Participants With Biopsy-proven Acute Rejection (BPAR of ISHLT Grade ≥ 3A), Acute Rejection Associated With Hemodynamic Compromise (HDC), Graft Loss/Re-transplant and Death at Month 12
NCT00300274 (10) [back to overview]Change From Baseline in the Average Maximum Intimal Thickness at Month 12
NCT00307047 (66) [back to overview]Ischemia Driven Major Adverse Cardiac Events (MACE)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Failure (TLF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Failure (TLF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Failure (TLF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Failure (TLF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Failure (TLF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Revascularization (TLR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Revascularization (TLR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Revascularization (TLR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Revascularization (TLR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Revascularization (TLR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Revascularization (TLR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Failure (TVF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Failure (TVF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Failure (TVF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Failure (TVF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Failure (TVF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Failure (TVF)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Revascularization (TVR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Revascularization (TVR)
NCT00307047 (66) [back to overview]All Cause Mortality
NCT00307047 (66) [back to overview]All Cause Mortality
NCT00307047 (66) [back to overview]All Cause Mortality
NCT00307047 (66) [back to overview]All Cause Mortality
NCT00307047 (66) [back to overview]All Cause Mortality
NCT00307047 (66) [back to overview]Acute Success (Clinical Procedure)
NCT00307047 (66) [back to overview]Acute Success (Clinical Device)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Revascularization (TVR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Revascularization (TVR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Revascularization (TVR)
NCT00307047 (66) [back to overview]Ischemia Driven Target Vessel Revascularization (TVR)
NCT00307047 (66) [back to overview]Ischemia Driven Major Adverse Cardiac Events (MACE)
NCT00307047 (66) [back to overview]Protocol Defined Stent Thrombosis Rate
NCT00307047 (66) [back to overview]Protocol Defined Stent Thrombosis Rate
NCT00307047 (66) [back to overview]Protocol Defined Stent Thrombosis Rate
NCT00307047 (66) [back to overview]Protocol Defined Stent Thrombosis Rate
NCT00307047 (66) [back to overview]All Myocardial Infarction (MI)
NCT00307047 (66) [back to overview]Ischemia Driven Target Lesion Failure (TLF)
NCT00307047 (66) [back to overview]Protocol Defined Stent Thrombosis Rate
NCT00307047 (66) [back to overview]Ischemia Driven Major Adverse Cardiac Events (MACE)
NCT00307047 (66) [back to overview]Ischemia Driven Major Adverse Cardiac Events (MACE)
NCT00307047 (66) [back to overview]Ischemia Driven Major Adverse Cardiac Events (MACE)
NCT00307047 (66) [back to overview]Ischemia Driven Major Adverse Cardiac Events (MACE)
NCT00307047 (66) [back to overview]Definite + Probable Stent Thrombosis Rate Based on ARC Definition
NCT00307047 (66) [back to overview]Definite + Probable Stent Thrombosis Rate Based on ARC Definition
NCT00307047 (66) [back to overview]Definite + Probable Stent Thrombosis Rate Based on ARC Definition
NCT00307047 (66) [back to overview]Definite + Probable Stent Thrombosis Rate Based on ARC Definition
NCT00307047 (66) [back to overview]Definite + Probable Stent Thrombosis Rate Based on Academic Research Consortium (ARC) Definition
NCT00307047 (66) [back to overview]Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)
NCT00307047 (66) [back to overview]Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)
NCT00307047 (66) [back to overview]Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)
NCT00307047 (66) [back to overview]Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)
NCT00307047 (66) [back to overview]Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)
NCT00307047 (66) [back to overview]Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)
NCT00307047 (66) [back to overview]Cardiac Death or Target Vessel MI Rate
NCT00307047 (66) [back to overview]Cardiac Death or Target Vessel MI Rate
NCT00307047 (66) [back to overview]Cardiac Death or Target Vessel MI Rate
NCT00307047 (66) [back to overview]Cardiac Death or Target Vessel MI Rate
NCT00307047 (66) [back to overview]Cardiac Death or Target Vessel MI Rate
NCT00307047 (66) [back to overview]Cardiac Death or Target Vessel MI Rate
NCT00307047 (66) [back to overview]All MI
NCT00307047 (66) [back to overview]All MI
NCT00307047 (66) [back to overview]All MI
NCT00307047 (66) [back to overview]All MI
NCT00307047 (66) [back to overview]All MI
NCT00307047 (66) [back to overview]All Cause Mortality
NCT00317720 (2) [back to overview]Clinical Benefit Response Rate (CBR)
NCT00317720 (2) [back to overview]Optimal Dose of RAD001 in Combination With Trastuzumab (Phase I)
NCT00323739 (2) [back to overview]Progression Free Survival (PFS), the Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Worsening of Their Disease
NCT00323739 (2) [back to overview]Overall Survival (OS), the Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Death
NCT00331409 (5) [back to overview]Median Time to Progression
NCT00331409 (5) [back to overview]Number of Participants With Adverse Events
NCT00331409 (5) [back to overview]Number of Subjects That Demonstrated a Reduction in Tumor Measurements.
NCT00331409 (5) [back to overview]Overall Number of Participants Who Achieve a Response Rate (Complete Response, Partial Response, and Stable Disease) at 3 Months
NCT00331409 (5) [back to overview]Progression-free Survival at 3 Months
NCT00332839 (1) [back to overview]Number of Participants Who Experienced Adverse Events and Death
NCT00363051 (11) [back to overview]Everolimus Trough Level Determination by Pharmacokinetics Parameter in Both Strata (Stratum 1 and 2)
NCT00363051 (11) [back to overview]Objective Response Rate: Percentage of Participants With Best Over All Response of Complete Response or Partial Response by Central Radiology Review (Stratum 1) Based on Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00363051 (11) [back to overview]Objective Response Rate: Percentage of Participants With Best Over All Response of Complete Response or Partial Response by Central Radiology Review (Stratum 2) Based on Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00363051 (11) [back to overview]Time to Overall Survival (OS) (Stratum 2)
NCT00363051 (11) [back to overview]Time to Progression Free Survival (PFS) Per Central Radiology Review (Stratum 2)
NCT00363051 (11) [back to overview]Time to Overall Survival (OS)(Stratum 1)
NCT00363051 (11) [back to overview]Time to Progression Free Survival (PFS) Per Central Radiology Review (Stratum 1)
NCT00363051 (11) [back to overview]Effect of Octreotide Depot on the Trough Concentrations of Everolimus
NCT00363051 (11) [back to overview]Number of Participants With Adverse Events (AEs), Death, Serious Adverse Events (SAEs) [Stratum 2]
NCT00363051 (11) [back to overview]Number of Participants With Adverse Events (AEs), Death, Serious Adverse Events (SAEs)[Stratum 1]
NCT00363051 (11) [back to overview]Duration of Overall Response (Stratum 1) Based on Response Evaluation Criteria in Solid Tumors (RECIST)- Central Radiology Review
NCT00369161 (3) [back to overview]Number of Participants With Incidence of Biopsy-proven Acute Rejection (BPAR)
NCT00369161 (3) [back to overview]Renal Function Assessed by Calculated Glomerular Filtration Rate (cGFR)
NCT00369161 (3) [back to overview]Percentage of Participants With Efficacy Failure
NCT00371826 (39) [back to overview]Number of Participants With Wound Problems(12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Sub Clinical Acute Rejection (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Sub Clinical Acute Rejection (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Post Transplant Diabetes Mellitus (PTDM) and Impaired Fasting Glucose (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Notable Abnormalities in Total Cholesterol and Triglycerides as Measurement of Effect of Treatment on Cardiovascular Health (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Notable Abnormalities in Total Cholesterol and Triglycerides as Measurement of Effect of Treatment on Cardiovascular Health (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Notable Abnormal Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP) as Measurement of Effect of Treatment on Cardiovascular Health (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Notable Abnormal Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP) as Measurement of Effect of Treatment on Cardiovascular Health (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With New Onset Diabetes Mellitus After Transplantation (NODAT) and Impaired Fasting Glucose (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Histological Evidence Chronic Allograft Nephropathy (CAN) (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Histological Evidence Chronic Allograft Nephropathy (CAN) (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Employment Status (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Employment Status (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Composite Endpoint of Treatment Failure (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Biopsy Proven Acute Rejection (BPAR) Per Treatment Group (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Biopsy Proven Acute Rejection (BPAR) Influenced by Demographic Characteristics and Morbidities (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Biopsy Proven Acute Rejection (BPAR) Influenced by Demographic Characteristics and Morbidities (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Any Wound Problems (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Antibody-mediated Rejection Per Treatment Group (36 Months Analysis)
NCT00371826 (39) [back to overview]Mean Urine Albumin/Creatinine Ratio [ACR] as Measurement of Proteinuria (36 Months Analysis)
NCT00371826 (39) [back to overview]Mean Short-form 36 Health Survey (SF-36) Score as a Measure of Quality of Life Assessment (36 Months Analysis)
NCT00371826 (39) [back to overview]Mean Short-form 36 Health Survey (SF-36) Score as a Measure of Quality of Life Assessment (12 Months Analysis)
NCT00371826 (39) [back to overview]Mean Serum Creatinine (36 Months Analysis)
NCT00371826 (39) [back to overview]Creatinine Clearance Calculated by the Cockcroft-Gault Formula (36 Months Analysis)
NCT00371826 (39) [back to overview]Change in Bone Mineral Density Between Week 2 and Month 24 (36 Months Analysis)
NCT00371826 (39) [back to overview]Calculated Glomerular Filtration Rate (cGFR) After Kidney Transplant to Evaluate Kidney Function (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Erythropoietin Usage (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Erythropoietin Usage (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Composite Endpoint of Treatment Failure (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Biopsy Proven Acute Rejection (BPAR) Per Treatment Group (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants With Antibody-mediated Rejection Per Treatment Group (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants Hospitalized for Reasons Other Than Primary Transplantation (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Participants Hospitalized for Reasons Other Than Primary Transplantation (12 Months Analysis)
NCT00371826 (39) [back to overview]Mean Urine Albumin/Creatinine Ratio (ACR) as Measurement of Proteinuria (12 Months Analysis)
NCT00371826 (39) [back to overview]Mean Serum Creatinine (12 Months Analysis)
NCT00371826 (39) [back to overview]Creatinine Clearance (CrCl) Calculated by the Cockcroft-Gault Formula (12 Months Analysis)
NCT00371826 (39) [back to overview]Calculated Glomerular Filtration Rate (cGFR) After Kidney Transplant to Evaluate Kidney Function (12 Months Analysis)
NCT00371826 (39) [back to overview]Number of Patient Survival and Graft Survival (36 Months Analysis)
NCT00371826 (39) [back to overview]Number of Patient Survival and Graft Survival (12 Months Analysis)
NCT00374140 (4) [back to overview]Progression-free Survival
NCT00374140 (4) [back to overview]Overall Survival
NCT00374140 (4) [back to overview]Objective Response Rate
NCT00374140 (4) [back to overview]Determine the Proportion of Previously Treated Small Cell Lung Cancer (SCLC) Patients Whose Disease Has Not Progressed Following 6-weeks (2 Cycles) of Treatment With RAD001.
NCT00377962 (12) [back to overview]Number of Patients With Biopsy-proven Acute Rejection From Month 12 to End of Study (Month 24)
NCT00377962 (12) [back to overview]Number of Patients in Need of Dialysis From Month 12 to End of Study (Month 24)
NCT00377962 (12) [back to overview]Mean Days of Hospitalization From Baseline to End of Study (Month 24)
NCT00377962 (12) [back to overview]Change in Forced Vital Capacity (FVC) From Baseline to End of Study (Month 24) in the Lung Transplant Subgroup
NCT00377962 (12) [back to overview]Change in Forced Expiratory Volume in 1 Second (FEV1) From Baseline to End of Study (Month 24) in the Lung Transplant Subgroup
NCT00377962 (12) [back to overview]Number of Patients Who Died and Number of Patients With Graft Loss From Month 12 to End of Study (Month 24)
NCT00377962 (12) [back to overview]Change in Measured Glomerular Filtration Rate (mGFR) From Baseline to Month 12
NCT00377962 (12) [back to overview]Change in Left Ventricular Function (Diameter and Thickness Parameters) From Baseline to End of Study (Month 24) in the Heart Transplant Subgroup
NCT00377962 (12) [back to overview]Change in Left Ventricular Function (Filling and Ejection Fraction Parameters) From Baseline to End of Study (Month 24) in the Heart Transplant Subgroup
NCT00377962 (12) [back to overview]Change in Measured Glomerular Filtration Rate (mGFR) From Baseline to End of Study (Month 24)
NCT00377962 (12) [back to overview]Number of Patients Discontinued From the Study Due to Adverse Events From Month 12 to End of Study (Month 24)
NCT00377962 (12) [back to overview]Change in Serum Creatinine From Baseline to End of Study (Month 24)
NCT00378014 (9) [back to overview]Number of Patients Who Experienced Adverse Events, Serious Adverse Events and Death
NCT00378014 (9) [back to overview]Incidence of Efficacy Failure
NCT00378014 (9) [back to overview]Incidence of the Need for a Change in the Immunosuppressive Regimen
NCT00378014 (9) [back to overview]Patient and Graft Survival
NCT00378014 (9) [back to overview]Renal Function (cGFR)
NCT00378014 (9) [back to overview]Hepatitis C Virus (HCV) Replication in HCV-positive Patients
NCT00378014 (9) [back to overview]Number of Patients Who Experienced Adverse Events, Serious Adverse Events and Death
NCT00378014 (9) [back to overview]Incidence of Treated BPAR
NCT00378014 (9) [back to overview]Calculated Glomerular Filtration Rate (cGFR)
NCT00390364 (1) [back to overview]Response Rate: The Total Number of Participants With Progression of Disease
NCT00392821 (2) [back to overview]Progression-Free Survival for Patients Treated at MTD/Phase II Dose Level
NCT00392821 (2) [back to overview]Overall Response Rate (ORR) of Patients Treated at MTD/Phase II Dose Level
NCT00401778 (3) [back to overview]Clinical Response as Assessed Metabolically by Changes in Positron Emission Tomography (PET) Scan Between Baseline and Immediately Prior to Surgery.
NCT00401778 (3) [back to overview]Effects of RAD001 on the Regulation of Key Proteins Involved With the Mammalian Target of Rapamycin (mTOR) Axis in Tumor Specimens and Buccal Mucosa in Patients With Operable Non-small Cell Lung Cancer (NSCLC).
NCT00401778 (3) [back to overview]Duration of Hospital Stay Following Surgery.
NCT00406276 (2) [back to overview]Time to Progression:Time Period (in Months) From Study Entry Until Disease Progression, Death, or Last Date of Contact.
NCT00406276 (2) [back to overview]Number of Subjects Showing Partial Response and Stable Disease With the Combination of RAD001 and Docetaxel.
NCT00409292 (4) [back to overview]To Assess the Safety of RAD001 in Patients With Metastatic Pancreatic Cancer
NCT00409292 (4) [back to overview]to Assess Overall Survival Associated With RAD001 in This Patient Population.
NCT00409292 (4) [back to overview]To Assess Progression-free Survival of RAD001 at Two Months in Patients With Metastatic Pancreatic Cancer Whose Disease Has Progressed on Gemcitabine Chemotherapy.
NCT00409292 (4) [back to overview]to Assess Response Rate Associated With RAD001 in This Patient Population.
NCT00410124 (13) [back to overview]Time to Definitive Deterioration of the FKS-DRS Risk Score by at Least 2 Score Units Using Kaplan-Meier Method, by Treatment.
NCT00410124 (13) [back to overview]Progressive Free Survival (PFS) in Patients Who Receive RAD001 Plus Best Supportive Care(BSC) Versus Patients Who Receive Matching Placebo Plus BSC
NCT00410124 (13) [back to overview]Overall Survival (OS) Assessed by the Monthly Overall Survival Assessments
NCT00410124 (13) [back to overview]Pharmacokinetics of RAD001: Time of the Last Quantifiable Concentration in a Dosing Interval - (Tlast)
NCT00410124 (13) [back to overview]Pharmacokinetics of RAD001: Time at Which C-Max Occurs (t-Max)
NCT00410124 (13) [back to overview]Pharmacokinetics of RAD001: Normalized to Body Surface Area (CL/F)
NCT00410124 (13) [back to overview]Pharmacokinetics of RAD001: Area Under Curve (AUC) in a Dosing Interval From Time-zero to Time of the Last Quantifiable Concentration. (AUC 0-tlast)
NCT00410124 (13) [back to overview]Duration of Response in Patients Who Receive RAD001 Plus BSC Versus Placebo Plus BSC
NCT00410124 (13) [back to overview]Best Overall Response Rate in Patients Who Receive RAD001 Plus BSC Versus Matching Placebo Plus BSC
NCT00410124 (13) [back to overview]Analysis of Time to Definitive Deterioration of the Global Health Status/QoL Scale(QL) Scores of the EORTC QLQ-30 Questionnaire by at Least 10 Percent Using Kaplan Meier Method, by Treatment.
NCT00410124 (13) [back to overview]Pharmacokinetics of RAD001: Apparent Systemic Clearance From Blood Following Extravascular Administration (CL/F)
NCT00410124 (13) [back to overview]Pharmacokinetics of RAD001:Peak Concentration in a Dosing Interval (C-max); Pre-dose Concentration at 24-h Time Point in Dosing Interval (C-min) and Average Concentration in a Dosing Interval =(C-avg)
NCT00410124 (13) [back to overview]Time to Definitive Deterioration of the Physical Functioning Scale (PF)Score of the EORTC QLQ-C30 Questionnaire by at Least 10 Percent Using Kaplan_Meier Method, by Treatment.
NCT00411619 (2) [back to overview]Overall Reduction in SEGA Tumor Volume.
NCT00411619 (2) [back to overview]Number With Observed Adverse Side Effects
NCT00412061 (7) [back to overview]Overall Survival Using Kaplan-Meier Methodology
NCT00412061 (7) [back to overview]Progression Free Survival (PFS) as Per Adjudicated Central Review by Baseline 5-hydroxyindoleacetic Acid (5-HIAA) Level
NCT00412061 (7) [back to overview]Number of Patients With Adverse Events (AEs), Clinically Notable AE, Death, Serious Adverse Events (SAEs) (Double-Blind Phase)
NCT00412061 (7) [back to overview]Progression Free Survival (PFS) as Per Adjudicated Central Radiology Review
NCT00412061 (7) [back to overview]Best Overall Response Rate as Per Adjudicated Central Radiology Review Based on Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00412061 (7) [back to overview]Progression Free Survival (PFS) as Per Adjudicated Central Review by Baseline Chromogranin A (CgA)
NCT00412061 (7) [back to overview]Number of Patients With Adverse Events (AEs), Clinically Notable AE, Death, Serious Adverse Events (SAEs) (Open Label Phase)
NCT00414440 (5) [back to overview]Course of Calculated GFR (mL/Min/1.73 m^2) From Month 24 to Month 60
NCT00414440 (5) [back to overview]Changes in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
NCT00414440 (5) [back to overview]Calculated GFR, Change From Baseline at Month 60 by Baseline cGFR
NCT00414440 (5) [back to overview]Primary Efficacy Analysis of Total Kidney Volume (mITT Set, Multiple Imputation)
NCT00414440 (5) [back to overview]Calculated GFR (mL/Min/1.73 m^2), Change From Baseline by Visit
NCT00419159 (13) [back to overview]Biomarkers Predictive of Clinical Benefit (DCR by PTEN ) on Everolimus (RAD001) 10mg/Day
NCT00419159 (13) [back to overview]Biomarker Predictive of Clinical Benefit (DCR by KRAS) on Everolimus (RAD001) 70 mg/Week
NCT00419159 (13) [back to overview]Biomarkers Predictive of Clinical Benefit (Median PFS and OS by PTEN ) on Everolimus (RAD001) 10mg/Day
NCT00419159 (13) [back to overview]Biomarker Predictive of Clinical Benefit (DCR by KRAS) on Everolimus (RAD001) 10 mg/Day
NCT00419159 (13) [back to overview]Number of Patients Who Died, Had an Serious Adverse Event (SAE), Had Grade 3 to 4 Adverse Event (AE), Discontinued Due to an AE, or Had a Clinical Notable AE by Treatment (tr).
NCT00419159 (13) [back to overview]Disease Control Rate (DCR) and Objective Response Rate (ORR) According to the Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00419159 (13) [back to overview]Biomarkers Predictive of Clinical Benefit (Median PFS and OS by PTEN ) on Everolimus (RAD001) 70mg/Week
NCT00419159 (13) [back to overview]The Number of Participants With Best Overall Response According to Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00419159 (13) [back to overview]Biomarkers Predictive of Clinical Benefit (Median PFS and OS by KRAS) on Everolimus (RAD001) 10mg/Day
NCT00419159 (13) [back to overview]Progression-free Survival (PFS)
NCT00419159 (13) [back to overview]Biomarkers Predictive of Clinical Benefit (Median PFS and OS by KRAS ) on Everolimus (RAD001) 70mg/Week
NCT00419159 (13) [back to overview]Overall Survival (OS)
NCT00419159 (13) [back to overview]Biomarkers Predictive of Clinical Benefit (DCR by PTEN ) on Everolimus (RAD001) 70mg/Week
NCT00425308 (12) [back to overview]Number of Participants With Biopsy-proven Acute Rejection (BPAR) at Month 6 and Month 12.
NCT00425308 (12) [back to overview]Number of Participants With Treatment Failures Assessed by Biopsy-proven Acute Rejection (BPAR), Graft Loss/Re-transplantation, Death or Lost to Follow-up at Month 12.
NCT00425308 (12) [back to overview]Change in the Glomerular Filtration Rate Estimated by Iohexol Plasma Clearance 12 Months After Randomization Between the 2 Groups of Patients Who Completed Trial
NCT00425308 (12) [back to overview]Change in Glomerular Filtration Rate Estimated by Iohexol Plasma Clearance 12 Months After Randomization Between the 2 Groups of Patients.
NCT00425308 (12) [back to overview]Assessing Cardiovascular Risk Factors Based on Fasting C-reactive Protein (CRP).
NCT00425308 (12) [back to overview]Assessing Cardiovascular Risk Factors Based on Fasting Triglycerides.
NCT00425308 (12) [back to overview]Assessing Cardiovascular Risk Factors Based on Fasting Total Cholesterol.
NCT00425308 (12) [back to overview]Assessing Cardiovascular Risk Factors Based on Fasting High-density Lipoprotein (HDL) Cholesterol, Low-density Lipoprotein (LDL) Cholesterol.
NCT00425308 (12) [back to overview]Change in Renal Function Assessed by Creatinine Clearance at Month 3, Month 6 and Month 12
NCT00425308 (12) [back to overview]Change in Renal Function Assessed by Proteinuria at Month 3, Month 6 and Month 12
NCT00425308 (12) [back to overview]Change in Renal Function Assessed by Serum Creatinine at Month 3, Month 6 and Month 12
NCT00425308 (12) [back to overview]Assessing Cardiovascular Risk Factors Based on Fasting Glucose.
NCT00426556 (4) [back to overview]Phase I: Best Overall Response (BOR)
NCT00426556 (4) [back to overview]Phase II: Progression Free Survival (PFS)
NCT00426556 (4) [back to overview]Phase II: Overall Survival (OS)
NCT00426556 (4) [back to overview]Phase II: Overall Response Rate
NCT00436618 (4) [back to overview]Progression-free Survival
NCT00436618 (4) [back to overview]Tumor Response, Defined by Disease: Chronic Lymphocytic Leukemia(CLL): Clinical Complete or Complete or Nodular Partial or Partial Remission, Waldenstrom: Complete or Partial Response, All Others: Complete or Complete Unconfirmed or Partial Response.
NCT00436618 (4) [back to overview]Time to Progression
NCT00436618 (4) [back to overview]Overall Survival
NCT00449748 (1) [back to overview]Number of Participants With Objective Response
NCT00458237 (2) [back to overview]Clinical Response Rate
NCT00458237 (2) [back to overview]Maximum Tolerated Dose (MTD)
NCT00459186 (2) [back to overview]Number of Patients Free of Dose Limiting Toxicity
NCT00459186 (2) [back to overview]Response Based on PET Scan
NCT00474929 (4) [back to overview]Survival Time
NCT00474929 (4) [back to overview]Progression Free Survival
NCT00474929 (4) [back to overview]Number of Participants Reporting a Dose Limiting Toxicity (DLT)
NCT00474929 (4) [back to overview]Proportion of Confirmed Tumor Responses
NCT00499603 (3) [back to overview]Participant Responses Per Treatment Arm at 12 Weeks
NCT00499603 (3) [back to overview]Number Participants With Inhibition of PI3K/PTEN/AKT Pathway at 48 Hours
NCT00499603 (3) [back to overview]Participant Responses Per Treatment Arm at 24 Weeks
NCT00510068 (18) [back to overview]Progression Free Survival According to Neuron Specific Enolase Tumor Marker (NSE) Baseline Level and According to NSE Early Response
NCT00510068 (18) [back to overview]Progression Free Survival According to Ki-67 Levels Categorized as: Less Than or Equal to 2%, > 2% to Less Than or Equal to 5% and > 5%
NCT00510068 (18) [back to overview]Progression Free Survival According to Chromogramin A Tumor Marker (CgA) Baseline Level and According to CgA Early Response
NCT00510068 (18) [back to overview]Plasma Angiogenesis Marker: Vascular Endothelial Growth Factor (VEGF)
NCT00510068 (18) [back to overview]Plasma Angiogenesis Marker: Soluble Vascular Endothelial Growth Factor Receptor 1 (sVEGFR1)
NCT00510068 (18) [back to overview]Plasma Angiogenesis Marker: Placental Growth Factor (PLGF)
NCT00510068 (18) [back to overview]Evaluation of Pharmacokinetics (PK) Parameter: CL/F
NCT00510068 (18) [back to overview]Plasma Angiogenesis Marker: Basic Fibroblast Growth Factor (bFGF)
NCT00510068 (18) [back to overview]Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) (Open-label Period)
NCT00510068 (18) [back to overview]Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs)
NCT00510068 (18) [back to overview]Evaluation of Pharmacokinetics (PK) Parameters: Cmax, Cmin
NCT00510068 (18) [back to overview]Evaluation of Pharmacokinetics (PK) Parameter: Tmax -Time to Maximum (Peak) Drug Concentration
NCT00510068 (18) [back to overview]Overall Survival
NCT00510068 (18) [back to overview]Percentage of Participants With Objective Response Rate ( CR {Complete Response} OR PR {Partial Response})
NCT00510068 (18) [back to overview]Time to Progression Free Survival (PFS) Based as Per Investigator Using Kaplan-Meier Methodology
NCT00510068 (18) [back to overview]Analysis of Time to Definitive Deterioration of WHO Performance Status Using Kaplan-Meier
NCT00510068 (18) [back to overview]Evaluation of Pharmacokinetics (PK) Parameter: AUC0-t Last
NCT00510068 (18) [back to overview]Plasma Angiogenesis Marker: Soluble Vascular Endothelial Growth Factor Receptor 2 (sVEGFR2)
NCT00514514 (13) [back to overview]Efficacy Event Data After Month 12 to Month 60
NCT00514514 (13) [back to overview]Efficacy Event Data Baseline 2 (Month 3) to Month 12
NCT00514514 (13) [back to overview]Efficacy Event Data From Baseline 2 (Month 3) to Month 6
NCT00514514 (13) [back to overview]Change From BL2 (Month 3) to Month 12 in Cardiovascular Risk (Framingham Score; 10-year Cardiovascular Risk)
NCT00514514 (13) [back to overview]GFR at Month 60 Utilizing Cockcroft-Gault Formula
NCT00514514 (13) [back to overview]GFR at Month 12 Utilizing Cockcroft-Gault Formula
NCT00514514 (13) [back to overview]GFR at Month 12 Utilizing Modification of Diet in Renal Disease (MDRD) Method
NCT00514514 (13) [back to overview]Mean Change in Serum Creatinine From Month 3 to Month 60
NCT00514514 (13) [back to overview]GFR at Month 60 Utilizing Modification of Diet in Renal Disease (MDRD) Method
NCT00514514 (13) [back to overview]GFR Calculated Via Nankivell Formula at Month 60
NCT00514514 (13) [back to overview]GFR Via Nankivell Formula at Month 12 - All Regimens
NCT00514514 (13) [back to overview]GFR Via Nankivell Method at Month 12 - CNI-Free vs Standard Regimen
NCT00514514 (13) [back to overview]Mean Change in Serum Creatinine From Month 3 to Month 12
NCT00515086 (4) [back to overview]Surgery Group: Progression-free Survival
NCT00515086 (4) [back to overview]No Surgery Group: Progression Free Survival
NCT00515086 (4) [back to overview]Surgery Group: Number of Participants With Adverse Events
NCT00515086 (4) [back to overview]No Surgery Group: Best Overall Tumor Response
NCT00516165 (6) [back to overview]Time to Progression
NCT00516165 (6) [back to overview]Progression-free Survival Rate at 24 Weeks
NCT00516165 (6) [back to overview]Overall Survival
NCT00516165 (6) [back to overview]Number of Patients With Adverse Events Who Were Treated With RAD001 for Advanced HCC
NCT00516165 (6) [back to overview]Maximum Tolerated Dose of RAD001 in Patients With Advanced Hepatocellular Carcinoma (HCC).
NCT00516165 (6) [back to overview]Overall Response Rate
NCT00521001 (4) [back to overview]Time to Disease Progression
NCT00521001 (4) [back to overview]9-week Progression-free Survival Rate
NCT00521001 (4) [back to overview]Confirmed Response Rate (Complete Response and Partial Response)
NCT00521001 (4) [back to overview]Survival Time
NCT00526591 (3) [back to overview]Change in PSA
NCT00526591 (3) [back to overview]Proportion of Patients Who Are P0 (i.e., no Clinically Detectable Tumor in the Pathologic Specimen) at Surgery
NCT00526591 (3) [back to overview]Toxicity Profile of Each Dose (Number of Patients With Worst Grade Toxicity)
NCT00529802 (2) [back to overview]Percent Change in FDG-PETUptake Following 2 Weeks of Therapy
NCT00529802 (2) [back to overview]Relative Tumor Size Change Following 8 Weeks of Therapy.
NCT00531011 (19) [back to overview]Revascularizations
NCT00531011 (19) [back to overview]Distal Minimum Lumen Diameter (MLD).
NCT00531011 (19) [back to overview]In-stent Late Loss (LL)
NCT00531011 (19) [back to overview]In-stent Minimum Lumen Diameter (MLD).
NCT00531011 (19) [back to overview]Composite Rate of Cardiac Death, Myocardial Infarction (MI, Both Q-wave and Non Q-wave), and Ischemia-driven Target Lesion Revascularization (TLR) .
NCT00531011 (19) [back to overview]Composite Rate of All Death, MI (Q-wave and Non Q-wave), and Target Vessel Revascularization (TVR).
NCT00531011 (19) [back to overview]Device Success
NCT00531011 (19) [back to overview]Composite Endpoint of All Death, MI (Q-wave and Non Q-wave), and TVR.
NCT00531011 (19) [back to overview]Procedural Success
NCT00531011 (19) [back to overview]Adjudicated Stent Thrombosis.
NCT00531011 (19) [back to overview]Adjudicated Stent Thrombosis.
NCT00531011 (19) [back to overview]Composite Endpoint of Cardiac Death, MI (Q-wave and Non Q-wave), and Ischemia-driven TLR .
NCT00531011 (19) [back to overview]Lesion Success
NCT00531011 (19) [back to overview]In-segment Binary Restenosis Rate
NCT00531011 (19) [back to overview]In-segment Late Loss (LL)
NCT00531011 (19) [back to overview]In-segment Minimum Lumen Diameter (MLD).
NCT00531011 (19) [back to overview]In-stent Binary Restenosis Rate
NCT00531011 (19) [back to overview]Proximal Minimum Lumen Diameter (MLD).
NCT00531011 (19) [back to overview]Revascularizations
NCT00553150 (6) [back to overview]Overall Survival Time
NCT00553150 (6) [back to overview]Maximum Tolerated Dose (MTD) of Everolimus (RAD001) in Combination With Temozolomide (TMZ) and 3D-conformal Radiotherapy (RT) or Intensity-modulated Radiotherapy (IMRT) Followed by Adjuvant TMZ With or Without RAD001 (Phase I)
NCT00553150 (6) [back to overview]Progression-free-survival at 6 Months (Phase II)
NCT00553150 (6) [back to overview]Response Rate, as Measured in Patients Receiving FLT-PET Imaging (Phase II)
NCT00553150 (6) [back to overview]Overall Survival at 12 Months (Phase II)
NCT00553150 (6) [back to overview]Time to Progression (Phase II)
NCT00570921 (3) [back to overview]Time to Progression
NCT00570921 (3) [back to overview]Objective Response Rates
NCT00570921 (3) [back to overview]Clinical Benefit Rate
NCT00576680 (3) [back to overview]Response Rate
NCT00576680 (3) [back to overview]Progression-free Survival
NCT00576680 (3) [back to overview]To Determine the Safety and Tolerability of This Drug Combination.
NCT00582738 (9) [back to overview]Percentage of Patients in Each Study Arm With Increase of ≥1 Point in the Ishak-Knodell Staging Score in Fibrosis
NCT00582738 (9) [back to overview]Change From Baseline in Fibrosis Staging Score (Measured by the Ishak-Knodell Staging Score) Between Baseline and 24 Months Post-transplant.
NCT00582738 (9) [back to overview]Comparison of Renal Function (Glomerular Filtration Rate [GFR] Calculated Using the Modification of Diet in Renal Disease Study Group [MDRD] Formula) Between Study Groups
NCT00582738 (9) [back to overview]Percentage of Patients With Death, Graft Loss and Biopsy Proven Acute Rejection (BPAR) Between Study Groups
NCT00582738 (9) [back to overview]Number of Patients With Events (Progression to Cirrhosis, Retransplantation, HCV Related Death, First BPAR, Graft Loss)at 12 and 24 Months
NCT00582738 (9) [back to overview]Comparison of the Effect of Both Regimens on the Inflammatory (Acti-test) and Fibrosis (Fibro-test) Components of Fibrosure, and on Fibrosis Area Assessed by Histomorphometry
NCT00582738 (9) [back to overview]Comparison of the Effect of Both Regimens in the Necroinflammatory Grading Score (Ishak-Knodell) (Portal Inflammation)
NCT00582738 (9) [back to overview]Change From Baseline in Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Viral Load at 12 and 24 Months Post Randomization
NCT00582738 (9) [back to overview]Change From Baseline in Fibrosis Metavir Scoring at 12 and 24 Months Post Randomization
NCT00591734 (3) [back to overview]Objective Response Rate (ORR)
NCT00591734 (3) [back to overview]Overall Survival Rate
NCT00591734 (3) [back to overview]Progression-free Survival
NCT00597506 (2) [back to overview]Progression Free Survival (PFS)
NCT00597506 (2) [back to overview]Overall Response
NCT00607113 (1) [back to overview]Net Change Relative to Baseline in Tumor Blood Flow
NCT00617084 (2) [back to overview]In-Stent Percent Diameter Stenosis
NCT00617084 (2) [back to overview]Target Lesion Failure
NCT00622869 (4) [back to overview]Incidence Rate of Composite Efficacy Failure From Randomization to Month 24
NCT00622869 (4) [back to overview]Incidence Rate of Composite Efficacy Failure From Randomization to Month 12
NCT00622869 (4) [back to overview]Incidence Rate of Treated Biopsy Proven Acute Rejection (tBPAR) at Months 12 and 24
NCT00622869 (4) [back to overview]Change in Renal Function From Randomization to Months 12 and 24
NCT00629525 (5) [back to overview]Biochemical Response Rate
NCT00629525 (5) [back to overview]Clinical Response
NCT00629525 (5) [back to overview]Molecular Response
NCT00629525 (5) [back to overview]Progression Free Survival
NCT00629525 (5) [back to overview]Pathologic Response
NCT00630344 (6) [back to overview]Incidence of Grade 4 Treatment-Related Toxicity
NCT00630344 (6) [back to overview]Incidence of Grade 1-3 Treatment-Related Fatigue Toxicity
NCT00630344 (6) [back to overview]Time to Progression (TTP)
NCT00630344 (6) [back to overview]Incidence of Grade 1-3 Treatment-Related Mucositis Toxicity
NCT00630344 (6) [back to overview]Incidence of Grade 1-3 Treatment-Related Rash Toxicity
NCT00630344 (6) [back to overview]Overall Response Rate
NCT00634920 (19) [back to overview]Progression of Measured Glomerular Filtration Rate
NCT00634920 (19) [back to overview]Percentage of Participants With Treatment Failures
NCT00634920 (19) [back to overview]Percentage of Participants With Graft Loss or Death
NCT00634920 (19) [back to overview]Percentage of Participants With Biopsy Proven Acute Rejection (BPAR)
NCT00634920 (19) [back to overview]Percentage of Participants on Lipid-lowering Drugs
NCT00634920 (19) [back to overview]Percentage of Participants Who Had Donor Specific Antibodies (DSA)
NCT00634920 (19) [back to overview]Percentage of Participants Who Developed CAN (Chronic Allograft Nephropathy)
NCT00634920 (19) [back to overview]Time to First Malignancy
NCT00634920 (19) [back to overview]Measured Glomerular Filtration Rate
NCT00634920 (19) [back to overview]Calculated Glomerular Filtration Rate
NCT00634920 (19) [back to overview]Health-related Quality of Life (QoL) as Measured by EuroQoL EQ-5D
NCT00634920 (19) [back to overview]Lipid Profile for Apolipoprotein
NCT00634920 (19) [back to overview]Lipid Profile for HDL-C, LDL-C,Total Cholesterol, and Triglycerides
NCT00634920 (19) [back to overview]Number of Lipid-lowering Drugs Taken
NCT00634920 (19) [back to overview]Percentage of Participants on Antihypertensive Drugs
NCT00634920 (19) [back to overview]Time to Treatment Failure
NCT00634920 (19) [back to overview]Measured Glomerular Filtration Rate
NCT00634920 (19) [back to overview]Proteinuria (Measured as Urine Albumin/Creatinine Ratio (mg/mmol))
NCT00634920 (19) [back to overview]Number of Antihypertensive Drugs Taken
NCT00640978 (1) [back to overview]Number of Participants Surviving at 6 Months
NCT00651482 (9) [back to overview]Progression-free Survival (PFS)
NCT00651482 (9) [back to overview]Overall Survival (OS)
NCT00651482 (9) [back to overview]Number of Subjects With Drug-related SAEs
NCT00651482 (9) [back to overview]Objective Response (OR)
NCT00651482 (9) [back to overview]Objective Response (OR) Duration
NCT00651482 (9) [back to overview]Time-to-Treatment Failure (TTF)
NCT00651482 (9) [back to overview]Total Number of Drug-related SAEs
NCT00651482 (9) [back to overview]Treatment Discontinuation Due to Disease Progression
NCT00651482 (9) [back to overview]Treatment Discontinuation Due to Toxicity
NCT00658320 (10) [back to overview]Core Study: Number of Patients With Composite Efficacy Endpoint
NCT00658320 (10) [back to overview]Extension Study: Renal Function Measured by Calculated Glomerular Filtration Rate (cGFR) Using the Modification of Diet in Renal Disease (MDRD) Formula
NCT00658320 (10) [back to overview]Core Study: Renal Function Measured by Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula
NCT00658320 (10) [back to overview]Core Study: Number Participants With Combined Graft Loss, Death or Loss to Follow-up
NCT00658320 (10) [back to overview]Extension Study: Renal Function Measured by Calculated Glomerular Filtration Rate (cGFR) Using the Modification of Diet in Renal Disease (MDRD) Formula
NCT00658320 (10) [back to overview]Extension Study: Renal Function Measured by Calculated Glomerular Filtration Rate (GFR) Using the Nankivell Formula
NCT00658320 (10) [back to overview]Extension Study: Number of Participants With Combined Efficacy Endpoint: Graft Loss, Death, Loss to Follow-up and/or Treated Biopsy Proven Acute Rejection (BPAR)
NCT00658320 (10) [back to overview]Extension Study: Number of Participants With Adverse Events and Serious Adverse Events
NCT00658320 (10) [back to overview]Extension Study: Everolimus Trough Levels
NCT00658320 (10) [back to overview]Extension Study: Cyclosporine Trough Levels
NCT00676520 (42) [back to overview]Death (Cardiac Death, Vascular Death, and Non-cardiovascular Death)
NCT00676520 (42) [back to overview]Death (Cardiac Death, Vascular Death, and Non-cardiovascular Death)
NCT00676520 (42) [back to overview]Death (Cardiac Death, Vascular Death, and Non-cardiovascular Death)
NCT00676520 (42) [back to overview]Dual Antiplatelet Medication Usage
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death, Any MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Target Lesion Revascularization (TLR) (PCI and CABG)
NCT00676520 (42) [back to overview]Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Dual Antiplatelet Medication Usage
NCT00676520 (42) [back to overview]Dual Antiplatelet Therapy Non-compliance Through 1 Year
NCT00676520 (42) [back to overview]Major Bleeding Complications
NCT00676520 (42) [back to overview]Major Bleeding Complications
NCT00676520 (42) [back to overview]Major Bleeding Complications
NCT00676520 (42) [back to overview]Major Bleeding Complications
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death, Any MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Target Lesion Revascularization (TLR) (PCI and CABG)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death and MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Clinically-indicated Target Lesion Revascularization (CI-TLR) (PCI and CABG) (This Composite Endpoint is Also Denoted as TLF)
NCT00676520 (42) [back to overview]Patient Health Status, Physical Limitations Assessed Using the SAQ (Seattle Angina Questionaire)
NCT00676520 (42) [back to overview]Patient Health Status, Physical Limitations Assessed Using the SAQ (Seattle Angina Questionaire)
NCT00676520 (42) [back to overview]SAQ (Seattle Angina Questionaire)
NCT00676520 (42) [back to overview]SAQ (Seattle Angina Questionaire)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death and MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Clinically-indicated Target Lesion Revascularization (CI-TLR) (PCI and CABG) (This Composite Endpoint is Also Denoted as TLF)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death and MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Clinically-indicated Target Lesion Revascularization (CI-TLR) (PCI and CABG) (This Composite Endpoint is Also Denoted as TLF)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death and Any Myocardial Infarction (MI)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death and Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Composite Rate of All Death, Any MI (Q-wave and Non Q-wave) and Any Repeat Revascularization (Percutaneous Coronary Intervention [PCI] and Coronary Artery Bypass Graft [CABG])
NCT00676520 (42) [back to overview]SAQ (Seattle Angina Questionaire)
NCT00676520 (42) [back to overview]Composite Rate of All Death, Any MI (Q-wave and Non Q-wave) and Any Repeat Revascularization (Percutaneous Coronary Intervention [PCI] and Coronary Artery Bypass Graft [CABG])
NCT00676520 (42) [back to overview]Composite Rate of All Death, Any MI (Q-wave and Non Q-wave) and Any Repeat Revascularization (Percutaneous Coronary Intervention [PCI] and Coronary Artery Bypass Graft [CABG])
NCT00676520 (42) [back to overview]Patient Health Status, Physical Limitations Assessed Using the SAQ (Seattle Angina Questionaire)
NCT00676520 (42) [back to overview]Composite Rate of All Death and Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Composite Rate of All Death and Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death, Any MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Target Lesion Revascularization (TLR) (PCI and CABG)
NCT00676520 (42) [back to overview]Procedural Success
NCT00676520 (42) [back to overview]Revascularization (Target Lesion, Target Vessel [TVR], and Non-target Vessel) (PCI and CABG)
NCT00676520 (42) [back to overview]Revascularization (Target Lesion, Target Vessel [TVR], and Non-target Vessel) (PCI and CABG)
NCT00676520 (42) [back to overview]Composite Rate of All Death and Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Composite Rate of Cardiac Death and Any MI (Q-wave and Non Q-wave)
NCT00676520 (42) [back to overview]Clinical Device Success
NCT00676520 (42) [back to overview]Dual Antiplatelet Medication Usage
NCT00676520 (42) [back to overview]Stent Thrombosis (Definite and Probable) Rate as Defined by ARC (Academic Research Constortium).
NCT00676520 (42) [back to overview]Dual Antiplatelet Medication Usage
NCT00676520 (42) [back to overview]Revascularization (Target Lesion, Target Vessel [TVR], and Non-target Vessel) (PCI and CABG)
NCT00688623 (7) [back to overview]Overall Survival (OS) for Per Protocol (PP) and ITT Sets
NCT00688623 (7) [back to overview]Percentage of Participants With a Overall Response Rate With a Complete Response (CR) or Partial Response (PR) at 12 Months ITT Set
NCT00688623 (7) [back to overview]Percentage of Participants With Disease Control Rate (DCR) at 12 Months for Per Protocol (PP) and ITT Sets
NCT00688623 (7) [back to overview]Percentage of Participants' Best Overall Response Rate at 12 Months - Per Protocol Set (PP)
NCT00688623 (7) [back to overview]Duration of Progression Free Survival (PFS) for Per Protocol (PP) and ITT Sets
NCT00688623 (7) [back to overview]Percentage of Participants With Objective Response Rate at 12 Months - Per Protocol Set (PP)
NCT00688623 (7) [back to overview]Percentage of Participants With Objective Response Rate at 12 Months ITT Set
NCT00688753 (6) [back to overview]Objective Response Rate
NCT00688753 (6) [back to overview]To Evaluate Efficacy of RAD001 as Monotherapy for the Treatment of Papillary Renal Cancer. Efficacy is Defined as the Percentage of Patients Progression-free at 6 Months.
NCT00688753 (6) [back to overview]Duration of Response
NCT00688753 (6) [back to overview]Median Progression Free Survival
NCT00688753 (6) [back to overview]Disease Control Rate (SD + PR + CR)
NCT00688753 (6) [back to overview]Incidence of Adverse Events, Serious Adverse Events, and Death.
NCT00702052 (5) [back to overview]Disease Control Rate (DCR)
NCT00702052 (5) [back to overview]Progression Free Survival (PFS)
NCT00702052 (5) [back to overview]Overall Survival
NCT00702052 (5) [back to overview]Overall Response Rate (ORR)
NCT00702052 (5) [back to overview]Number of Participants With At Least One Adverse Event (AE) and Serious Adverse Event (SAE)
NCT00719264 (8) [back to overview]Best Overall Response in Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab
NCT00719264 (8) [back to overview]Time to Definitive Deterioration of the Functional Assessment of Cancer Therapy Kidney Symptom Index, Disease Related Symptoms (FKSI-DRS) Risk Score by at Least 2 Score Units
NCT00719264 (8) [back to overview]Response Duration Differences in Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab
NCT00719264 (8) [back to overview]Overall Survival (OS) Treatment Effect in Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab
NCT00719264 (8) [back to overview]Duration of Exposure of RAD001 in Participants Randomized to the Treatment Combination of RAD001 and Bevacizumab
NCT00719264 (8) [back to overview]Progression-free Survival (PFS) of Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab
NCT00719264 (8) [back to overview]Time to Definitive Deterioration of the Global Health Status and the Physical Functioning (PF) Subscale Scores of the European Organization for the Research and Treatment of Cancer (EORTC)-Core Quality of Life Questionnaire (QLQ-C30) by at Least 10%
NCT00719264 (8) [back to overview]Number of Participants Who Experienced Adverse Events (AEs), Serious Adverse Events and Deaths
NCT00729482 (4) [back to overview]Number of Participants With Adverse Events
NCT00729482 (4) [back to overview]Overall Survival
NCT00729482 (4) [back to overview]Progression-free Survival Rate at 4-month (16 Weeks)
NCT00729482 (4) [back to overview]Response Rate
NCT00767819 (6) [back to overview]Objective Tumor Response Rates (Complete Response and Partial Response) at Week 16 (ITT)
NCT00767819 (6) [back to overview]Best Overall Response Rates by Week 16 (ITT)
NCT00767819 (6) [back to overview]Time to Progression (TTP) (ITT)
NCT00767819 (6) [back to overview]Percentage of Participants With Progression-free Survival (PFS) at 16 Weeks
NCT00767819 (6) [back to overview]Percentage of Participants With Overall Survival (OS) at Week 16 (ITT)
NCT00767819 (6) [back to overview]Percentage of Participants With Duration of Response (CR, PR, SD) at 16 Weeks.
NCT00770120 (4) [back to overview]Frequency and Severity of Toxicities
NCT00770120 (4) [back to overview]Response
NCT00770120 (4) [back to overview]Progression-Free Survival
NCT00770120 (4) [back to overview]Overall Survival
NCT00782626 (1) [back to overview]Overall Response
NCT00783796 (99) [back to overview]Cardiac Death/MI
NCT00783796 (99) [back to overview]Cardiac Death/ All MI /CI-TLR
NCT00783796 (99) [back to overview]Cardiac Death/ All MI /CI-TLR
NCT00783796 (99) [back to overview]Cardiac Death/ All MI /CI-TLR
NCT00783796 (99) [back to overview]Cardiac Death/ All MI /CI-TLR
NCT00783796 (99) [back to overview]Cardiac Death/ All MI /CI-TLR
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)
NCT00783796 (99) [back to overview]All TVR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TVR (CI and Non-CI)
NCT00783796 (99) [back to overview]Clinically Indicated Target Lesion Revascularization (CI-TLR)
NCT00783796 (99) [back to overview]Clinically Indicated Target Vessel Revascularization
NCT00783796 (99) [back to overview]Clinically Indicated Target Vessel Revascularization
NCT00783796 (99) [back to overview]Clinically Indicated Target Vessel Revascularization
NCT00783796 (99) [back to overview]Clinically Indicated Target Vessel Revascularization
NCT00783796 (99) [back to overview]All TVR (CI and Non-CI)
NCT00783796 (99) [back to overview]Clinically Indicated Target Vessel Revascularization (TVR)
NCT00783796 (99) [back to overview]Composite Rate of Cardiac Death, Target Vessel Myocardial Infarction (MI) (Per Protocol Definition) & Clinically Indicated Target Lesion Revascularization (CI-TLR).
NCT00783796 (99) [back to overview]All TVR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TVR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TLR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TLR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TLR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TLR (CI and Non-CI)
NCT00783796 (99) [back to overview]All TLR (CI and Non-CI)
NCT00783796 (99) [back to overview]All Death/ All MI/All Coronary Revascularization
NCT00783796 (99) [back to overview]All Death/ All MI/All Coronary Revascularization
NCT00783796 (99) [back to overview]All Death/ All MI/All Coronary Revascularization
NCT00783796 (99) [back to overview]All Death/ All MI/All Coronary Revascularization
NCT00783796 (99) [back to overview]All Death/ All MI/All Coronary Revascularization
NCT00783796 (99) [back to overview]All Death (Cardiac, Vascular, Non-cardiovascular)
NCT00783796 (99) [back to overview]All Death (Cardiac, Vascular, Non-cardiovascular)
NCT00783796 (99) [back to overview]All Death (Cardiac, Vascular, Non-cardiovascular)
NCT00783796 (99) [back to overview]All Death (Cardiac, Vascular, Non-cardiovascular)
NCT00783796 (99) [back to overview]All Death (Cardiac, Vascular, Non-cardiovascular)
NCT00783796 (99) [back to overview]All Coronary Revascularization (TVR and Non-TVR)
NCT00783796 (99) [back to overview]All Coronary Revascularization (TVR and Non-TVR)
NCT00783796 (99) [back to overview]All Coronary Revascularization (TVR and Non-TVR)
NCT00783796 (99) [back to overview]All Coronary Revascularization (TVR and Non-TVR)
NCT00783796 (99) [back to overview]All Coronary Revascularization (TVR and Non-TVR)
NCT00783796 (99) [back to overview]Clinically Indicated Target Lesion Revascularization (CI-TLR)
NCT00783796 (99) [back to overview]Composite Rate of Cardiac Death, Target Vessel Myocardial Infarction (MI) (Per Protocol Definition) & Clinically Indicated Target Lesion Revascularization (CI-TLR).
NCT00783796 (99) [back to overview]Device Success (Per Lesion Basis, for Target Lesions Treated by 2.25 mm XIENCE V EECS With or Without Planned Overlap)
NCT00783796 (99) [back to overview]Distal % Diameter Stenosis
NCT00783796 (99) [back to overview]Distal Angiographic Binary Restenosis (ABR) Rate
NCT00783796 (99) [back to overview]Distal Late Loss
NCT00783796 (99) [back to overview]In-segment % Diameter Stenosis
NCT00783796 (99) [back to overview]In-segment Angiographic Binary Restenosis (ABR) Rate
NCT00783796 (99) [back to overview]In-segment Late Loss (LL)
NCT00783796 (99) [back to overview]In-stent % Diameter Stenosis
NCT00783796 (99) [back to overview]In-stent Angiographic Binary Restenosis (ABR) Rate
NCT00783796 (99) [back to overview]In-Stent Late Loss
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)
NCT00783796 (99) [back to overview]Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)
NCT00783796 (99) [back to overview]Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)
NCT00783796 (99) [back to overview]Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)
NCT00783796 (99) [back to overview]Clinically Indicated Target Lesion Revascularization (CI-TLR)
NCT00783796 (99) [back to overview]Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)
NCT00783796 (99) [back to overview]Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)
NCT00783796 (99) [back to overview]Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Clinically Indicated Target Lesion Revascularization (CI-TLR)
NCT00783796 (99) [back to overview]Clinically Indicated Target Lesion Revascularization (CI-TLR)
NCT00783796 (99) [back to overview]Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Procedural Success (Per Subject Basis, for ALL Target and Non-target Lesions)
NCT00783796 (99) [back to overview]Proximal % Diameter Stenosis
NCT00783796 (99) [back to overview]Proximal Angiographic Binary Restenosis (ABR) Rate
NCT00783796 (99) [back to overview]Proximal Late Loss
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (ARC Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Stent Thrombosis (Protocol Defined)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Cardiac Death/MI
NCT00783796 (99) [back to overview]Cardiac Death/MI
NCT00783796 (99) [back to overview]Cardiac Death/MI
NCT00783796 (99) [back to overview]Cardiac Death/MI
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per ARC)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)
NCT00783796 (99) [back to overview]Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)
NCT00783796 (99) [back to overview]Composite Rate of Cardiac Death, Target Vessel Myocardial Infarction (MI) (Per Protocol Definition) & Clinically Indicated Target Lesion Revascularization (CI-TLR).
NCT00789828 (11) [back to overview]Everolimus Blood Concentration (C2h) at 2 Hours Post Dose
NCT00789828 (11) [back to overview]Everolimus Trough Concentrations (Cmin) at 24 Hours After Last Dose
NCT00789828 (11) [back to overview]Percentage of Participants With Renal Impairment During Core Period
NCT00789828 (11) [back to overview]Change From Baseline in Frequency of Total Seizure Events Per 24 Hours at Week 24 in Both Core and Extension Period
NCT00789828 (11) [back to overview]Duration of SEGA Response
NCT00789828 (11) [back to overview]Duration of Skin Lesion Response in Everolimus Treated Participants
NCT00789828 (11) [back to overview]Percentage of Participants With Best Overall Subependymal Giant Cell Astrocytomas (SEGA) Response
NCT00789828 (11) [back to overview]Percentage of Participants With Skin Lesions Assessed Using Physician's Global Assessement Overall Score
NCT00789828 (11) [back to overview]Time to SEGA Progression
NCT00789828 (11) [back to overview]Time to SEGA Response
NCT00789828 (11) [back to overview]Time to SEGA Worsening
NCT00790036 (3) [back to overview]Disease-free Survival (DFS)
NCT00790036 (3) [back to overview]Lymphoma-specific Survival (LSS)
NCT00790036 (3) [back to overview]Overall Survival (OS)
NCT00790400 (10) [back to overview]Angiomyolipoma Response Rate as Per Central Radiology Review
NCT00790400 (10) [back to overview]Duration of Angiomyolipoma Response - Only Everolimus Patients With Angiomyolipoma Response
NCT00790400 (10) [back to overview]Duration of Skin Lesion Response - Only Everolimus Patients With Best Overall Skin Lesion Response of Complete Clinical Response (CCR) or Partial Response (PR)
NCT00790400 (10) [back to overview]Skin Lesion Response Rate as Per Investigator (Only Patients With at Least One Skin Lesion at Baseline)
NCT00790400 (10) [back to overview]Time to Angiomyolipoma Progression as Per Central Radiology Review
NCT00790400 (10) [back to overview]Time to Angiomyolipoma Response - Only Everolimus Patients With Angiomyolipoma Response
NCT00790400 (10) [back to overview]Change From Baseline in Plasma Angiogenic Molecules - Vascular Endothelial Growth Factor (VEGF) Marker
NCT00790400 (10) [back to overview]Everolimus Blood Concentrations (C2h) at 2 Hours Post-dose
NCT00790400 (10) [back to overview]Everolimus Trough Concentrations (Cmin)
NCT00790400 (10) [back to overview]Percentage of Participants With Renal Impairment
NCT00805129 (2) [back to overview]Proportion of Pretreatment Primary Tumor Samples With mTOR Pathway Markers
NCT00805129 (2) [back to overview]Number of Participants Evaluated for Toxicity
NCT00805961 (4) [back to overview]Overall Survival of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen
NCT00805961 (4) [back to overview]Progression-free Survival (PFS)
NCT00805961 (4) [back to overview]Number of Participants Experiencing Toxicity After This Novel Multimodality Regimen
NCT00805961 (4) [back to overview]Objective Response Rate of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen
NCT00809185 (3) [back to overview]Number of Participants With Bone Marrow Morphology and Cytogenetics Pre- and Post-therapy
NCT00809185 (3) [back to overview]Number of Patients With Either a Major or Minor Erythroid Response (Hemoglobin Change From Baseline Measure)
NCT00809185 (3) [back to overview]Number of Dose- and Non-dose-limiting Toxicities
NCT00814788 (3) [back to overview]Progression-free Survival
NCT00814788 (3) [back to overview]Overall Survival
NCT00814788 (3) [back to overview]PSA Response Rate
NCT00823459 (5) [back to overview]Progression-free Survival at 6 Months.
NCT00823459 (5) [back to overview]To Assess the Correlation of Activation of the PI3K/mTOR Pathway With Survival
NCT00823459 (5) [back to overview]Objective Response Rate (ORR) in Patients Treated With RAD001.
NCT00823459 (5) [back to overview]Overall Survival (OS) in Patients Treated With RAD001.
NCT00823459 (5) [back to overview]RAD001 Safety Profile in Patients With Recurrent LLG
NCT00827359 (3) [back to overview]Best Overall Response by PI3K-AKT-MTOR Mutation
NCT00827359 (3) [back to overview]Median Progression Free Survival
NCT00827359 (3) [back to overview]Best Overall Response Rate
NCT00831480 (1) [back to overview]Disease Progression Diagnosed by Biopsy
NCT00843531 (3) [back to overview]Objective Response Rate (ORR)
NCT00843531 (3) [back to overview]Number of Patients With Dose-limiting Toxicity (DLT)
NCT00843531 (3) [back to overview]Duration of Objective Response
NCT00856856 (194) [back to overview]In-scaffold Percent Diameter Stenosis (%DS)
NCT00856856 (194) [back to overview]In-scaffold Percent Diameter Stenosis (%DS)
NCT00856856 (194) [back to overview]In-scaffold Percent Diameter Stenosis (%DS)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Ischemia Driven Target Vessel Revascularization (ID-TVR)
NCT00856856 (194) [back to overview]Late Incomplete Apposition
NCT00856856 (194) [back to overview]Late Incomplete Apposition
NCT00856856 (194) [back to overview]Late Incomplete Apposition
NCT00856856 (194) [back to overview]Late Incomplete Apposition
NCT00856856 (194) [back to overview]Luminal Volume
NCT00856856 (194) [back to overview]Luminal Volume
NCT00856856 (194) [back to overview]Luminal Volume
NCT00856856 (194) [back to overview]Luminal Volume
NCT00856856 (194) [back to overview]Mean Flow Area
NCT00856856 (194) [back to overview]Mean Flow Area
NCT00856856 (194) [back to overview]Mean Flow Area
NCT00856856 (194) [back to overview]Mean Flow Area
NCT00856856 (194) [back to overview]Mean Luminal Area
NCT00856856 (194) [back to overview]Mean Luminal Area
NCT00856856 (194) [back to overview]Mean Luminal Area
NCT00856856 (194) [back to overview]Mean Luminal Area
NCT00856856 (194) [back to overview]Mean Luminal Diameter
NCT00856856 (194) [back to overview]Mean Luminal Diameter
NCT00856856 (194) [back to overview]Mean Luminal Diameter
NCT00856856 (194) [back to overview]Mean Luminal Diameter
NCT00856856 (194) [back to overview]Mean Reference Area
NCT00856856 (194) [back to overview]Mean Reference Area
NCT00856856 (194) [back to overview]Mean Reference Area
NCT00856856 (194) [back to overview]Mean Reference Area
NCT00856856 (194) [back to overview]Mean Scaffold Area
NCT00856856 (194) [back to overview]Mean Scaffold Area
NCT00856856 (194) [back to overview]Mean Scaffold Diameter
NCT00856856 (194) [back to overview]Mean Scaffold Diameter
NCT00856856 (194) [back to overview]Mean Stent Area
NCT00856856 (194) [back to overview]Mean Stent Diameter
NCT00856856 (194) [back to overview]Mean Strut Core Area
NCT00856856 (194) [back to overview]Mean Strut Core Area
NCT00856856 (194) [back to overview]Mean Strut Core Area
NCT00856856 (194) [back to overview]Minimum Flow Area
NCT00856856 (194) [back to overview]Minimum Flow Area
NCT00856856 (194) [back to overview]Minimum Flow Area
NCT00856856 (194) [back to overview]Minimum Flow Area
NCT00856856 (194) [back to overview]Minimum Luminal Area
NCT00856856 (194) [back to overview]Minimum Luminal Area
NCT00856856 (194) [back to overview]Minimum Luminal Area
NCT00856856 (194) [back to overview]Minimum Luminal Area
NCT00856856 (194) [back to overview]Minimum Luminal Diameter
NCT00856856 (194) [back to overview]Minimum Luminal Diameter
NCT00856856 (194) [back to overview]Minimum Luminal Diameter
NCT00856856 (194) [back to overview]Minimum Luminal Diameter (MLD)
NCT00856856 (194) [back to overview]Minimum Scaffold Area
NCT00856856 (194) [back to overview]Minimum Scaffold Area
NCT00856856 (194) [back to overview]Minimum Scaffold Diameter
NCT00856856 (194) [back to overview]Minimum Scaffold Diameter
NCT00856856 (194) [back to overview]Minimum Stent Area
NCT00856856 (194) [back to overview]Minimum Stent Diameter
NCT00856856 (194) [back to overview]Myocardial Infarction
NCT00856856 (194) [back to overview]Myocardial Infarction
NCT00856856 (194) [back to overview]Myocardial Infarction
NCT00856856 (194) [back to overview]Myocardial Infarction
NCT00856856 (194) [back to overview]Myocardial Infarction
NCT00856856 (194) [back to overview]Myocardial Infarction
NCT00856856 (194) [back to overview]Number of Struts in Side Branch
NCT00856856 (194) [back to overview]Number of Struts in Side Branch
NCT00856856 (194) [back to overview]Number of Struts in Side Branch
NCT00856856 (194) [back to overview]Number of Struts in Side Branch
NCT00856856 (194) [back to overview]Number of Struts Per BVS
NCT00856856 (194) [back to overview]Number of Struts Per BVS
NCT00856856 (194) [back to overview]Number of Struts Per BVS
NCT00856856 (194) [back to overview]Number of Struts Per BVS
NCT00856856 (194) [back to overview]Percent (%) Lumen Area Stenosis
NCT00856856 (194) [back to overview]Percent (%) Lumen Area Stenosis
NCT00856856 (194) [back to overview]Percent (%) Lumen Area Stenosis
NCT00856856 (194) [back to overview]Percent (%) Lumen Area Stenosis
NCT00856856 (194) [back to overview]Persisting Dissection
NCT00856856 (194) [back to overview]Persisting Dissection
NCT00856856 (194) [back to overview]Persisting Dissection
NCT00856856 (194) [back to overview]Persisting Dissection
NCT00856856 (194) [back to overview]Persisting Dissection
NCT00856856 (194) [back to overview]Persisting Incomplete Apposition
NCT00856856 (194) [back to overview]Persisting Incomplete Apposition
NCT00856856 (194) [back to overview]Persisting Incomplete Apposition
NCT00856856 (194) [back to overview]Persisting Incomplete Apposition
NCT00856856 (194) [back to overview]Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 1 Year
NCT00856856 (194) [back to overview]Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 180 Days
NCT00856856 (194) [back to overview]Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 2 Years
NCT00856856 (194) [back to overview]Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 3 Years
NCT00856856 (194) [back to overview]Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 5 Years
NCT00856856 (194) [back to overview]Scaffold Thrombosis
NCT00856856 (194) [back to overview]Scaffold Thrombosis
NCT00856856 (194) [back to overview]Scaffold Thrombosis
NCT00856856 (194) [back to overview]Scaffold Thrombosis
NCT00856856 (194) [back to overview]Scaffold Thrombosis
NCT00856856 (194) [back to overview]Scaffold Thrombosis
NCT00856856 (194) [back to overview]Scaffold Volume
NCT00856856 (194) [back to overview]Scaffold Volume
NCT00856856 (194) [back to overview]Stent Volume
NCT00856856 (194) [back to overview]Strut Volume
NCT00856856 (194) [back to overview]Strut Volume
NCT00856856 (194) [back to overview]Strut Volume
NCT00856856 (194) [back to overview]Thrombus
NCT00856856 (194) [back to overview]Thrombus
NCT00856856 (194) [back to overview]Thrombus
NCT00856856 (194) [back to overview]Thrombus
NCT00856856 (194) [back to overview]Thrombus
NCT00856856 (194) [back to overview]Tissue Coverage Area BVS (Neointimal Area)
NCT00856856 (194) [back to overview]Tissue Coverage Area BVS (Neointimal Area)
NCT00856856 (194) [back to overview]Tissue Coverage Area BVS (Neointimal Area)
NCT00856856 (194) [back to overview]Tissue Coverage Area Classical
NCT00856856 (194) [back to overview]Tissue Coverage Area Classical
NCT00856856 (194) [back to overview]Tissue Coverage Area Classical
NCT00856856 (194) [back to overview]Tissue Coverage Obstruction Volume BVS
NCT00856856 (194) [back to overview]Tissue Coverage Obstruction Volume BVS
NCT00856856 (194) [back to overview]Tissue Coverage Obstruction Volume BVS
NCT00856856 (194) [back to overview]Ischemia Driven Target Lesion Revascularization (ID-TLR)
NCT00856856 (194) [back to overview]Tissue Coverage Obstruction Volume Classical
NCT00856856 (194) [back to overview]Tissue Coverage Obstruction Volume Classical
NCT00856856 (194) [back to overview]Tissue Coverage Volume BVS
NCT00856856 (194) [back to overview]Tissue Coverage Volume BVS
NCT00856856 (194) [back to overview]Tissue Coverage Volume BVS
NCT00856856 (194) [back to overview]Tissue Coverage Volume Classical
NCT00856856 (194) [back to overview]Tissue Coverage Volume Classical
NCT00856856 (194) [back to overview]Tissue Coverage Volume Classical
NCT00856856 (194) [back to overview]Volume Obstruction (VO)
NCT00856856 (194) [back to overview]Volume Obstruction (VO)
NCT00856856 (194) [back to overview]Volume Obstruction (VO)
NCT00856856 (194) [back to overview]Volume Obstruction (VO)
NCT00856856 (194) [back to overview]Vasomotion Analysis: In-scaffold Mean Luminal Diameter
NCT00856856 (194) [back to overview]Cardiac Death
NCT00856856 (194) [back to overview]Clinical Device Success (Per Lesion)
NCT00856856 (194) [back to overview]Clinical Procedure Success (Per Patient)
NCT00856856 (194) [back to overview]Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 1 Year
NCT00856856 (194) [back to overview]Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 180 Days
NCT00856856 (194) [back to overview]Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 2 Years
NCT00856856 (194) [back to overview]Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 3 Years
NCT00856856 (194) [back to overview]Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 5 Years
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Major Adverse Cardiac Event (MACE)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]Hierarchical Target Vessel Failure (TVF)
NCT00856856 (194) [back to overview]In-scaffold Angiographic Binary Restenosis (ABR)
NCT00856856 (194) [back to overview]In-scaffold Angiographic Binary Restenosis (ABR)
NCT00856856 (194) [back to overview]In-scaffold Angiographic Binary Restenosis (ABR)
NCT00856856 (194) [back to overview]In-scaffold Angiographic Binary Restenosis (ABR)
NCT00856856 (194) [back to overview]In-scaffold Angiographic Binary Restenosis (ABR)
NCT00856856 (194) [back to overview]In-scaffold Late Loss (LL): In-scaffold MLD Post-procedure - In-scaffold MLD at 2 Years
NCT00856856 (194) [back to overview]In-scaffold Late Loss (LL): In-scaffold MLD Post-procedure - In-scaffold MLD at 3 Years
NCT00856856 (194) [back to overview]In-scaffold Late Loss (LL): In-scaffold MLD Post-procedure - In-scaffold MLD at 5 Years
NCT00856856 (194) [back to overview]In-scaffold Late Loss: In-scaffold MLD Post-procedure - In-scaffold MLD at 1 Year
NCT00856856 (194) [back to overview]In-scaffold Late Loss: In-scaffold MLD Post-procedure - In-scaffold MLD at 180 Days
NCT00856856 (194) [back to overview]Tissue Coverage Obstruction Volume Classical
NCT00856856 (194) [back to overview]% of Acutely Covered Struts
NCT00856856 (194) [back to overview]% of Acutely Covered Struts
NCT00856856 (194) [back to overview]% of Covered Struts (150 µm)
NCT00856856 (194) [back to overview]% of Uncovered Struts (150 µm)
NCT00856856 (194) [back to overview]% of Uncovered Struts (150 µm)
NCT00856856 (194) [back to overview]% of Uncovered Struts (150 µm)
NCT00856856 (194) [back to overview]Aneurysm
NCT00856856 (194) [back to overview]Aneurysm
NCT00856856 (194) [back to overview]Aneurysm
NCT00856856 (194) [back to overview]Aneurysm
NCT00856856 (194) [back to overview]Aneurysm
NCT00856856 (194) [back to overview]Cardiac Death
NCT00856856 (194) [back to overview]Cardiac Death
NCT00856856 (194) [back to overview]Cardiac Death
NCT00856856 (194) [back to overview]Cardiac Death
NCT00856856 (194) [back to overview]Cardiac Death
NCT00856856 (194) [back to overview]In-scaffold Percent Diameter Stenosis (%DS)
NCT00856856 (194) [back to overview]In-scaffold Percent Diameter Stenosis (%DS)
NCT00857259 (2) [back to overview]Change in Visual Acuity From Baseline to Week 4 in Patients Treated With Everolimus
NCT00857259 (2) [back to overview]Change in Central Retinal Thickness From Baseline to Week 4, as Measured by Optical Coherence Tomography (OCT)
NCT00862979 (6) [back to overview]Occurrence of Treatment Failures From Month 6 to 9 and Month 9 to 18
NCT00862979 (6) [back to overview]Calculated Glomerular Filtration Rate (cGFR) According to Cockcroft-Gault at Month 12 and 18
NCT00862979 (6) [back to overview]Serum Creatinine at Month 6, 8, 9, 10 12 and 18
NCT00862979 (6) [back to overview]Reciprocal Creatinine Slope Between Month 6 and Month 18
NCT00862979 (6) [back to overview]Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula at Month 18
NCT00862979 (6) [back to overview]Occurrence of Major Cardiac Events (MACE) From Month 6 to 18
NCT00863655 (12) [back to overview]Overall Response Rate (ORR)
NCT00863655 (12) [back to overview]Estradiol Plasma Concentrations
NCT00863655 (12) [back to overview]Clinical Benefit Rate (CBR)
NCT00863655 (12) [back to overview]Duration of Response (Among Participants With Best Overall Response of CR or PR) Estimated Per Kaplan-Meier
NCT00863655 (12) [back to overview]Overall Survival (OS) by Number of Deaths
NCT00863655 (12) [back to overview]Progression-free Survival (PFS) Based on Local Radiology Review of Tumor Assessments.
NCT00863655 (12) [back to overview]Overall Survival (OS) by Median
NCT00863655 (12) [back to overview]Proportion of Patients With no Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG PS) Using Kaplan-Meier
NCT00863655 (12) [back to overview]Proportion of Patients With Having no Overall Response Based on Investigator Assessment
NCT00863655 (12) [back to overview]Patient-reported Outcomes (PROs): Time to Deterioration of PRO Scores Using Kaplan Meier - EORTC QLQ-C30
NCT00863655 (12) [back to overview]Exemestane Concentrations at Week 4
NCT00863655 (12) [back to overview]Everolimus Concentrations at Week 4
NCT00869999 (3) [back to overview]Progression-free Survival
NCT00869999 (3) [back to overview]Duration of Overall Response
NCT00869999 (3) [back to overview]Overall Response Rate
NCT00876395 (17) [back to overview]Clinical Benefit Rate (CBR) Equal to or Greater Than 24 Weeks - Full Population
NCT00876395 (17) [back to overview]Clinical Benefit Rate (CBR) Equal to or Greater Than 24 Weeks - HR-negative Population
NCT00876395 (17) [back to overview]Overall Response Rate (ORR) - HR-negative Population
NCT00876395 (17) [back to overview]Overall Survival (OS) - Full Population
NCT00876395 (17) [back to overview]Time to Overall Response Based on Investigator - HR-negative Population
NCT00876395 (17) [back to overview]Time to Overall Response Based on Investigator - Full Population
NCT00876395 (17) [back to overview]Everolimus Blood Level Concentrations at Steady States for Everolimus
NCT00876395 (17) [back to overview]Overall Response (OR) - Full Population
NCT00876395 (17) [back to overview]Overall Response (OR) - HR-negative Population
NCT00876395 (17) [back to overview]Paclitaxel Plasma Concentrations
NCT00876395 (17) [back to overview]Time to Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG-PS) Score - HR-negative Population
NCT00876395 (17) [back to overview]Time to Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG-PS) Score - Full Population
NCT00876395 (17) [back to overview]Progression-free Survival (PFS) Per Investigators' Assessment Based on Local Radiology Review - Full Population
NCT00876395 (17) [back to overview]Trastuzumab Serum Concentrations
NCT00876395 (17) [back to overview]Overall Response Rate (ORR) - Full Population
NCT00876395 (17) [back to overview]Progression-free Survival (PFS) Per Investigators' Assessment Based on Local Radiology Review - (Hormone Receptor (HR)-Negative Population
NCT00876395 (17) [back to overview]Overall Survival (OS) - HR-negative Population
NCT00879333 (7) [back to overview]Patient Reported Outcome (PRO): Time to Definitive Deterioration of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire 30 (EORTC QLQ-C30) Scores
NCT00879333 (7) [back to overview]Overall Response Rate (ORR)
NCT00879333 (7) [back to overview]Progression Free Survival (PFS)
NCT00879333 (7) [back to overview]Overall Survival (OS)
NCT00879333 (7) [back to overview]Time to Definitive Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG PS) Score
NCT00879333 (7) [back to overview]Everolimus Steady State Concentraions at Predose (Cmin) and Cmax at Week 5
NCT00879333 (7) [back to overview]Everolimus Steady State Concentraions at Predose (Cmin) and Cmax by Region Asia vs. Rest of the World (ROW) at Week 5
NCT00886691 (5) [back to overview]Progression-free Survival
NCT00886691 (5) [back to overview]The Proportion of Patients With Measurable Disease Who Have Objective Tumor Responses by Treatment.
NCT00886691 (5) [back to overview]Percentage of Participants With at Least One Cancer Antigen 125 (CA-125) Response
NCT00886691 (5) [back to overview]Characterize and Compare Progression-free Survival and Overall Survival in Patients With Measurable Disease (RECIST Criteria) and Patients With Detectable (Non-measurable) Disease
NCT00886691 (5) [back to overview]Incidence of Adverse Events as Assessed by Common Terminology Criteria for Adverse Events Version 4.0 (CTCAE v4.0)
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the Global Health Status/QoL Scores of the EORTC QLQ-C30 by First-Line Drug
NCT00903175 (13) [back to overview]Overall Response Rate (ORR) - First -Line (1-L)
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the Physical Functioning Scale of the EORTC QLQ-C30 - by First and Second-Line Drugs Combined
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the Global Health Status/QoL Scores of the EORTC QLQ-C30 by First and Second-Line Drugs Combined
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the FKSI-DRS Risk Score by at Least 3 Score Units by First-line Drug
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the FKSI-DRS Risk Score by at Least 3 Score Units by First and Second-line Drugs Combined
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the Fatigue Scale of the EORTC QLQ-C30 by First-Line Drug
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the Fatigue Scale of the EORTC QLQ-C30 by First and Second-Line Drugs Combined
NCT00903175 (13) [back to overview]Progression-free Survival Combined (PFS-C)
NCT00903175 (13) [back to overview]Progression Free Survival First-Line (PFS 1-L)
NCT00903175 (13) [back to overview]Overall Survival (OS)
NCT00903175 (13) [back to overview]Duration of Response (DoR) - First-Line (1-L)
NCT00903175 (13) [back to overview]Time to Definitive Deterioration of the Physical Functioning (PF) Scale of the EORTC QLQ-C30 - by First-Line (1L) Drug
NCT00912340 (2) [back to overview]Progression-free Survival (PFS) Until First Progression
NCT00912340 (2) [back to overview]Progression-free Survival (PFS) in Patients Who Crossed Over
NCT00915603 (5) [back to overview]Number of Patients With Treatment-related Adverse Events (AEs) as a Measure of Safety and Tolerability
NCT00915603 (5) [back to overview]Duration of Response (DOR)
NCT00915603 (5) [back to overview]Overall Response Rate (ORR)
NCT00915603 (5) [back to overview]Overall Survival (OS)
NCT00915603 (5) [back to overview]Progression-Free Survival (PFS)
NCT00918333 (5) [back to overview]Number of Phase I Participants With Dose-Limiting Toxicity Events (Phase I)
NCT00918333 (5) [back to overview]Overall Response Rate (Phase II)
NCT00918333 (5) [back to overview]Duration of Response (Phase II)
NCT00918333 (5) [back to overview]Progression-free Survival (Phase II)
NCT00918333 (5) [back to overview]Overall Survival Time (Phase II)
NCT00930930 (4) [back to overview]Number of Patients With Pathological Complete Response
NCT00930930 (4) [back to overview]Clinical Tumor Response to Neoadjuvant Therapy as Measured by Ultrasound Immediately Before Surgery
NCT00930930 (4) [back to overview]Number of Patients With Each Worst-grade Toxicity Response
NCT00930930 (4) [back to overview]Number of Patients That Underwent Breast Conservation Surgery
NCT00934895 (1) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of RAD001 in Combination of Weekly Abraxane and Determine the Phase II Dose of RAD001.
NCT00935792 (7) [back to overview]Progression-free Survival
NCT00935792 (7) [back to overview]Clinical Response (Complete or Partial Remission)
NCT00935792 (7) [back to overview]Survival Time
NCT00935792 (7) [back to overview]Time to Subsequent Therapy
NCT00935792 (7) [back to overview]Duration of Response
NCT00935792 (7) [back to overview]Test the Safety and Tolerability of the Combination of Everolimus and Alemtuzumab.
NCT00935792 (7) [back to overview]Number of Participants With Dose-Limiting Toxicities
NCT00936702 (5) [back to overview]Duration of Response
NCT00936702 (5) [back to overview]Overall Survival
NCT00936702 (5) [back to overview]Percentage of Participants With Confirmed Tumor Responses
NCT00936702 (5) [back to overview]Progression-free Survival
NCT00936702 (5) [back to overview]Time to Treatment Failure
NCT00936858 (4) [back to overview]Median Overall Survival
NCT00936858 (4) [back to overview]Median Progression Free Survival
NCT00936858 (4) [back to overview]Objective Response Rate
NCT00936858 (4) [back to overview]Mean Change in Quality of Life [Medullary Thyroid Cancer Population Only]
NCT00942734 (1) [back to overview]12-Week Progression-Free Survival (PFS)
NCT00954512 (2) [back to overview]Part 1: Number of Participants Who Experienced One or More Adverse Events (AEs)
NCT00954512 (2) [back to overview]Part 2: Number of Participants With Each Type of Response Evaluation Criteria in Solid Tumors (RECIST)-Determined Overall Best Response
NCT00956293 (1) [back to overview]Number of Participants Who Experienced Adverse Events, Serious Adverse Events and Death
NCT00965094 (5) [back to overview]Change in Renal Function (Creatinine Slope)
NCT00965094 (5) [back to overview]Participants Who Had Occurrence of Biopsy Proven Acute Rejection, Graft Loss or Death.
NCT00965094 (5) [back to overview]Participants Who Had Occurrence of Treatment Failure.
NCT00965094 (5) [back to overview]Assessment of GFR by the Cockcroft-Gault Method (LOCF)
NCT00965094 (5) [back to overview]Renal Function Assessed as Glomerula Filtration Rate (GFR) - Nankivell Method - 9 Months After Renal Transplantation (LOCF)
NCT00967044 (1) [back to overview]Maximum Tolerated Dose (MTD) of Everolimus With Panobinostat
NCT00968253 (3) [back to overview]Overall Response Rate (OR) Where OR = CR + CRp + CRi
NCT00968253 (3) [back to overview]Participant Responses by Daily Dose Level Assignment (RAD001 5 mg, 10 mg and MTD 5 mg)
NCT00968253 (3) [back to overview]Maximum Tolerated Dose [MTD] Determination by Number of Participants With Dose Limiting Toxicity (DLT)
NCT00972335 (2) [back to overview]Progression-free Survival (PFS), in the Treatment of Patients With Refractory Meningioma.
NCT00972335 (2) [back to overview]To Evaluate the Toxicity of Bevacizumab/Everolimus in Patients With Recurrent Meningioma.
NCT00976248 (3) [back to overview]Time to Next Therapy With Single Agent RAD001 Therapy in Previously Untreated WM
NCT00976248 (3) [back to overview]Overall Response Rate of RAD001 in Patients With Previously Untreated WM
NCT00976248 (3) [back to overview]Time to Progression With Single Agent RAD001 Therapy in Previously Untreated WM.
NCT00976573 (4) [back to overview]Confirmed Tumor Response Rate (Complete Response [CR] or Partial Response [PR]) According to Response Evaluation Criteria in Solid Tumors (RECIST) Criteria
NCT00976573 (4) [back to overview]Progression-free Survival
NCT00976573 (4) [back to overview]Overall Survival Time
NCT00976573 (4) [back to overview]Toxicity
NCT00978432 (2) [back to overview]Overall Response Rate
NCT00978432 (2) [back to overview]Summary of Adverse Events (AEs)
NCT00985192 (6) [back to overview]Biomarker Correlations: Time to Progression
NCT00985192 (6) [back to overview]Efficacy in Terms of Progression Free Response
NCT00985192 (6) [back to overview]Overall Disease-control Rate in Patients With Previously Treated Unresectable or Metastatic Adenocarcinoma of the Upper Gastrointestinal Tract Treated With Everolimus.
NCT00985192 (6) [back to overview]Overall Survival
NCT00985192 (6) [back to overview]Biomarker Correlations: Progression Free Survival
NCT00985192 (6) [back to overview]Observed Biomarkers
NCT01007942 (9) [back to overview]Vinorelbine Blood Concentrations by Leading Dose and Time Point
NCT01007942 (9) [back to overview]Trastuzumab Blood Concentrations by Leading Dose and Time Point
NCT01007942 (9) [back to overview]Progressive-free Survival (PFS) Per Investigator Assessment
NCT01007942 (9) [back to overview]Overall Survival (OS)
NCT01007942 (9) [back to overview]Overall Response Rate (ORR)
NCT01007942 (9) [back to overview]Median Time to Deterioration of the ECOG Performance Status Score
NCT01007942 (9) [back to overview]Clinical Benefit Rate (CBR)
NCT01007942 (9) [back to overview]Everolimus Blood Concentrations by Leading Dose and Time Point
NCT01007942 (9) [back to overview]PRO: Time to Deterioration in Global Health Status/QoL Domain Score of the European Organization for the Research and Treatment of Cancer (EORTC)-Core Quality of Life Questionnaire (QLQ-C30) (by at Least 10%)
NCT01009346 (2) [back to overview]Progression Free Survival (PFS) of RAD001 in Combination With Weekly Cetuximab and Cisplatin.
NCT01009346 (2) [back to overview]Maximum Tolerated Dose (MTD) of RAD001 in Combination With Cetuximab and Cisplatin.
NCT01014351 (3) [back to overview]Overall Survival (OS)
NCT01014351 (3) [back to overview]Objective Response Rate (ORR)
NCT01014351 (3) [back to overview]Progression-free Survival (PFS)
NCT01017029 (6) [back to overview]Absolute and Percent Frequencies of Patients With LDL ≥ 100 mg/mL at 1, 3 and 6 Months, by Treatment Group
NCT01017029 (6) [back to overview]Participants With at Least One Occurrence of Safety Composite Endpoint After 6 Months by Treatment Group
NCT01017029 (6) [back to overview]Participants With at Least One Occurrence of Composite Treatment Failure Events
NCT01017029 (6) [back to overview]Hazard Cox's Model Analysis of Pericardial/Pleural Effusions
NCT01017029 (6) [back to overview]Partcipants With at Least One Occurrence of Each Safety Composite Endpoint Event After 6 Months by Treatment Group
NCT01017029 (6) [back to overview]Participants With CMV Infection and CMV Syndrome/Disease After 6 Months by Treatment Group
NCT01022996 (6) [back to overview]Overall Response Rate (ORR) Based on the Assessments by Investigator
NCT01022996 (6) [back to overview]Disease Control Rate (DCR)
NCT01022996 (6) [back to overview]Duration of Disease Control
NCT01022996 (6) [back to overview]Progression Free Survival (PFS) by Kaplan-Meier Estimate
NCT01022996 (6) [back to overview]Time to Overall Response (TTR) Per Kaplan-Meier Estimate
NCT01022996 (6) [back to overview]Duration of Overall Response (DoR)
NCT01023815 (6) [back to overview]Treatment Failure Rate
NCT01023815 (6) [back to overview]Number of Participants With Graft and Patient Survival After Randomization
NCT01023815 (6) [back to overview]Changes in the Estimated Glomerular Filtration Rate (eGFR) Between Randomization (Month 3) and Month 12
NCT01023815 (6) [back to overview]Biopsy Proven Acute Rejection (BPAR) Rate Between Randomization and Month 12
NCT01023815 (6) [back to overview]Change in Serum Creatinine
NCT01023815 (6) [back to overview]Change in Estimated Creatine Clearance
NCT01024946 (1) [back to overview]To Determine the Rate of Clinical Benefit (i.e. Rate of Complete or Partial Response Plus Stable Disease) at 16 Weeks for Patients With Malignant Mesothelioma Treated With Everolimus as Second or Third Line Therapy.
NCT01025817 (7) [back to overview]Number of Participants With Incidence of Proteinuria Events
NCT01025817 (7) [back to overview]Number of Participants With Incidence Rates of BKV Viremia, BKV Viruria, or BKV Nephropathy
NCT01025817 (7) [back to overview]Number of Participants With Incidence of Adverse Events, Serious Adverse Events, and Tacrolimus-associated Adverse Events
NCT01025817 (7) [back to overview]Number of Participants With Incidence of CMV (Viremia, Syndrome and Disease)
NCT01025817 (7) [back to overview]Estimated Glomerular Filtration Rate (eGFR)
NCT01025817 (7) [back to overview]Number of Participants With Incidence of Composite Efficacy Failure
NCT01025817 (7) [back to overview]Number of Participants With Incidence of New Onset of Diabetes Mellitus
NCT01031381 (2) [back to overview]Total Number of Participants Experienced a Response (Complete Response+Partial Response+Stable Disease)
NCT01031381 (2) [back to overview]Progression-free Survival (PFS) at 6-months
NCT01031446 (6) [back to overview]Patients With Overall Response
NCT01031446 (6) [back to overview]Maximum Feasible Dose in Milligrams Per Meter Squared of Body Surface Area (mg/m2) of Cisplatin and Paclitaxel for Women With Metastatic Breast Cancer
NCT01031446 (6) [back to overview]Time to Progression in Patients With Metastatic Basal-like Breast Cancer.
NCT01031446 (6) [back to overview]Time to Progression
NCT01031446 (6) [back to overview]Patients With Progression-free Survival
NCT01031446 (6) [back to overview]Maximum Feasible Dose in mg of RAD001 (Everolimus)for Women With Metastatic Breast Cancer
NCT01034631 (10) [back to overview]Exploratory Objective: Correlation of PFS With Biomarkers
NCT01034631 (10) [back to overview]Phase II: Response Rate With Combination Therapy Compared to Everolimus Alone
NCT01034631 (10) [back to overview]Phase I: Toxicities of BNC105P in Combination With Everolimus.
NCT01034631 (10) [back to overview]Phase II: Adverse Events of Everolimus and BNC105P When Administered as a Combination or Sequential Regimen.
NCT01034631 (10) [back to overview]Phase I: Response Rate of BNC105P in Combination With Everolimus.
NCT01034631 (10) [back to overview]Geometric Mean Half-life of BNC105 and BNC105P in Combination With Everolimus.
NCT01034631 (10) [back to overview]Phase I: Maximum Tolerated Dose of BNC105P in Combination With Everolimus.
NCT01034631 (10) [back to overview]Phase II: 6-month Progression Free Survival (PFS) With the Addition of BNC105P to Everolimus.
NCT01034631 (10) [back to overview]Phase II: Overall Survival
NCT01034631 (10) [back to overview]Phase II: Progression Free Survival (PFS) With BNC105P Alone in Patients After Progressing on Everolimus.
NCT01035229 (7) [back to overview]Time to Tumor Progression (TTP)
NCT01035229 (7) [back to overview]Time to Definitive Deterioration of EORTC QLQ-C30 Scores
NCT01035229 (7) [back to overview]Time to Definitive Deterioration of ECOG Performance Score (PS) Score
NCT01035229 (7) [back to overview]Pharmacokinetics Assessments - Cmin
NCT01035229 (7) [back to overview]Pharmacokinetics Assessments - Cmax
NCT01035229 (7) [back to overview]Percentage of Participants With Disease Control Rate (DCR)
NCT01035229 (7) [back to overview]Overall Survival (OS)
NCT01047293 (2) [back to overview]Evaluate Safety of the Combination at a Daily Dosing of 2.5mg RAD001, 5 mg RAD001 or 10 mg RAD001 (Phase 1 Part)
NCT01047293 (2) [back to overview]Progression Free Survival at Six Months
NCT01051570 (6) [back to overview]PSA Response Rate
NCT01051570 (6) [back to overview]Association of PSA Response Rate With Correlative Markers (Phospho mTOR, pAKT, and p70S6)
NCT01051570 (6) [back to overview]Overall Survival
NCT01051570 (6) [back to overview]Pharmacokinetics: Observed Carboplatin AUC Was Estimated Based on the Concentration in the 2.75-h Sample.
NCT01051570 (6) [back to overview]Time to Progression (TTP)
NCT01051570 (6) [back to overview]Number of Participants With Toxicity as Measured by NCI CTCAE v3.0 Criteria
NCT01051791 (4) [back to overview]Overall Survival (OS)
NCT01051791 (4) [back to overview]Progression Free Survival (PFS)
NCT01051791 (4) [back to overview]Objective Response Rate (ORR)
NCT01051791 (4) [back to overview]Clinical Benefit Rate (CBR)
NCT01057277 (1) [back to overview]Response
NCT01058655 (6) [back to overview]Overall Survival (OS) [Phase II]
NCT01058655 (6) [back to overview]Everolimus Maximum Tolerated Dose (MTD) [Phase I]
NCT01058655 (6) [back to overview]Dose Limiting Toxicity (DLT) [Phase I]
NCT01058655 (6) [back to overview]Disease Control Rate (DCR) [Phase II]
NCT01058655 (6) [back to overview]Tivozanib Maximum Tolerated Dose (MTD) [Phase I]
NCT01058655 (6) [back to overview]Progression-Free Survival (PFS) [Phase II]
NCT01059318 (8) [back to overview]Change From Baseline in Forced Vital Capacity (FVC)
NCT01059318 (8) [back to overview]Change From Baseline in Forced Expiratory Volume in 1 Second (FEV1)
NCT01059318 (8) [back to overview]Change From Baseline in Extended Pulmonary Function Testing
NCT01059318 (8) [back to overview]Change From Baseline in 6-minute Walk Test Score to Measure Exercise Capacity
NCT01059318 (8) [back to overview]Change From Baseline in Carbon Monoxide Diffusing Capacity (DLCO)
NCT01059318 (8) [back to overview]Mean Trough (C0,ss) and Peak (C2,ss) Drug Concentration at Steady State
NCT01059318 (8) [back to overview]Change From Baseline in Vascular Endothelial Growth Factor-D (VEGF-D) Concentrations
NCT01059318 (8) [back to overview]Change From Baseline in Oxygen Saturation
NCT01062399 (5) [back to overview]Phase II: Distribution of Worst Adverse Event Grade
NCT01062399 (5) [back to overview]Phase I: Number of Patients With Dose-limiting Toxicity (DLT)
NCT01062399 (5) [back to overview]Phase II: Overall Survival (OS)
NCT01062399 (5) [back to overview]Phase II: Progression-free Survival (PFS)
NCT01062399 (5) [back to overview]Phase I: Distribution of Worst Adverse Event Grade
NCT01068249 (5) [back to overview]Number of Participants With Disease Progression
NCT01068249 (5) [back to overview]Number of Participants With Objective Response Rate
NCT01068249 (5) [back to overview]Number of Participants With Adverse Events (All Grades)
NCT01068249 (5) [back to overview]Median Overall Survival (OS)
NCT01068249 (5) [back to overview]Median Progression Free Survival (PFS)
NCT01070316 (2) [back to overview]Reduction in Seizure Frequency
NCT01070316 (2) [back to overview]Number of Participants Continuing Study Medication Over Time
NCT01075321 (5) [back to overview]Number of Patients Reporting Dose-Limiting Toxicity (DLT) (Phase I)
NCT01075321 (5) [back to overview]Duration of Response for All Eligible Patients
NCT01075321 (5) [back to overview]Overall Survival for All Eligible Patients
NCT01075321 (5) [back to overview]Time to Treatment Failure for All Eligible Patients
NCT01075321 (5) [back to overview]Progression-Free Survival For All Eligible Patients
NCT01079143 (18) [back to overview]Change in Interstitial Fibrosis/Tabular Atrophy (IF/TA) Grade
NCT01079143 (18) [back to overview]Number of Participants With Progression of Renal Graft Fibrosis (Primary Comparison - ITT Population
NCT01079143 (18) [back to overview]Number of Participants With Progression of Renal Fibrosis Using Numerical Quantification
NCT01079143 (18) [back to overview]Number of Participants With Epithelial-mesenchymal Transition (EMT) Status
NCT01079143 (18) [back to overview]Number of Participants With Epithelial-mesenchymal Transition (EMT) Score
NCT01079143 (18) [back to overview]Interstitial Fibrosis/Tabular Atrophy (IF/TA)
NCT01079143 (18) [back to overview]Incidence (Number) of BPAR
NCT01079143 (18) [back to overview]Change in EMT Score
NCT01079143 (18) [back to overview]Incidence (Number) of Subclinical Rejections and Borderline Lesions
NCT01079143 (18) [back to overview]Change in Urine Protein/Creatinine Ratio (Without Imputation)
NCT01079143 (18) [back to overview]Change From Baseline (M3) in Estimated Glomerular Filtration Rate (eGFR)
NCT01079143 (18) [back to overview]Change in Estimated Glomerular Filtration Rate (eGFR) at M12 From Baseline (M3) - ANCOVA Model
NCT01079143 (18) [back to overview]Incidence (Number) of Participants With Graft Losses
NCT01079143 (18) [back to overview]Type of Biopsy Proven Acute Rejection (BPAR)
NCT01079143 (18) [back to overview]Treatment Failures
NCT01079143 (18) [back to overview]Change in Percentage of Interstitial Fibrosis (IF) by Numerical Quantification
NCT01079143 (18) [back to overview]Severity of BPAR
NCT01079143 (18) [back to overview]Risk Factors of IF/TA Progression
NCT01088048 (15) [back to overview]Plasma Concentration of Lenalidomide
NCT01088048 (15) [back to overview]Plasma Concentration of Everolimus
NCT01088048 (15) [back to overview]Plasma Concentration of Bendamustine
NCT01088048 (15) [back to overview]Toxicity of Administration of IDELA
NCT01088048 (15) [back to overview]Time to Response
NCT01088048 (15) [back to overview]Progression-free Survival
NCT01088048 (15) [back to overview]Overall Survival
NCT01088048 (15) [back to overview]Overall Response Rate
NCT01088048 (15) [back to overview]Duration of Response
NCT01088048 (15) [back to overview]Duration of Exposure to IDELA
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 6)
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 7)
NCT01088048 (15) [back to overview]Sub-study: Plasma Concentration of IDELA (Cohorts 1-4)
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 4)
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 1, Cohorts 2 and 3, Cohort 5)
NCT01108445 (15) [back to overview]Percentage of Participants With Stable Disease (SD)
NCT01108445 (15) [back to overview]Percentage of Participants With Adverse Events
NCT01108445 (15) [back to overview]Overall Response Rate
NCT01108445 (15) [back to overview]Median OS
NCT01108445 (15) [back to overview]12 Week Clinical Benefit Rate as Percentage
NCT01108445 (15) [back to overview]Median Duration of Response (CR, PR, and SD)
NCT01108445 (15) [back to overview]Change in Quality-of-life
NCT01108445 (15) [back to overview]Change in Quality-of-life
NCT01108445 (15) [back to overview]Progression Free Survival Rates
NCT01108445 (15) [back to overview]PFS Expressed in Months
NCT01108445 (15) [back to overview]Best Tumor Shrinkage as a Percentile in Each Arm
NCT01108445 (15) [back to overview]Time-to-new Metastatic Disease in Each Treatment Arm
NCT01108445 (15) [back to overview]Anti-tumor Activity as Measured by Median Progression Free Survival Time
NCT01108445 (15) [back to overview]Overall Survival Rates
NCT01108445 (15) [back to overview]Change in Quality-of-life
NCT01111058 (5) [back to overview]Site of Progression: Distant
NCT01111058 (5) [back to overview]Site of Progression: Local-regional
NCT01111058 (5) [back to overview]Site of Progression: Unknown
NCT01111058 (5) [back to overview]2 Year Progression Free Survival Rate
NCT01111058 (5) [back to overview]Number of Participants With Toxicity
NCT01114529 (4) [back to overview]Estimated Glomerular Filtration Rate (eGFR)
NCT01114529 (4) [back to overview]Incidence of Composite Efficacy Endpoint for Each Arm at Month 12 and Month 24
NCT01114529 (4) [back to overview]Comparison of Incidence Rates of Efficacy Endpoints Between Treatment Arms (Full Analysis Set - 24 Month Analysis)
NCT01114529 (4) [back to overview]Change in Left Ventricular Mass Index (LVMi) From Randomization to Month 12 and Month 24
NCT01115803 (7) [back to overview]Percentage of Participants Achieving Complete Response (CR) or Partial Response (PR) [Response Rate (RR)]
NCT01115803 (7) [back to overview]Number of Participants With Complete Response (CR), Partial Response (PR) or Stable Disease (SD) [Best Overall Response (BOR) (CR+PR+SD)]
NCT01115803 (7) [back to overview]Number of Participants Who Died Due to Progressive Disease Within 30 Days of Study Drug Discontinuation
NCT01115803 (7) [back to overview]Pharmacokinetics, Maximum Observed Plasma Concentration (Cmax) of LY2584702
NCT01115803 (7) [back to overview]Pharmacokinetics, Area Under the Concentration Time Curve (AUC)
NCT01115803 (7) [back to overview]Clinically Significant Effects (Number of Participants With Adverse Events)
NCT01115803 (7) [back to overview]Recommended Dose for Phase 2 Studies
NCT01125293 (10) [back to overview]PTEN Mutation Rate [Phase II]
NCT01125293 (10) [back to overview]Very-good-partial-response-or-better Rate [Phase II]
NCT01125293 (10) [back to overview]Treatment-Emergent Sensory Neuropathy Rate [Phase I]
NCT01125293 (10) [back to overview]Phase II Duration of Response (DoR)
NCT01125293 (10) [back to overview]Phase II Overall Response Rate
NCT01125293 (10) [back to overview]2 Year Progression-free-survival [Phase II]
NCT01125293 (10) [back to overview]2-year Time-to-progression Probability (TTP) [Phase II]
NCT01125293 (10) [back to overview]Everolimus Dose Limiting Toxicity (DLT) [Phase I]
NCT01125293 (10) [back to overview]Everolimus Maximum Tolerated Dose (MTD) Stage A [Phase I]
NCT01125293 (10) [back to overview]Everolimus Maximum Tolerated Dose (MTD) Stage B [Phase I]
NCT01127763 (4) [back to overview]Median Progression-free Survival Time
NCT01127763 (4) [back to overview]Toxicity Profile-Non Hematological
NCT01127763 (4) [back to overview]Toxicity Profile-Hematological
NCT01127763 (4) [back to overview]Clinical Benefit Rate (Complete Response, Partial Response, and Stable Disease That Lasts More Than 6 Months)
NCT01133678 (1) [back to overview]Tumor Responses
NCT01136733 (16) [back to overview]Summary of Plasma Concentrations of Lenvatinib for Sparse Pharmacokinetic (PK) Sampling for Phase 1b and Phase 2
NCT01136733 (16) [back to overview]Maximum Concentration of Everolimus (Cmax) in Blood When Administered Alone or in Combination With Lenvatinib
NCT01136733 (16) [back to overview]Phase 2: Objective Response Rate (ORR)
NCT01136733 (16) [back to overview]Phase 2: Overall Survival (OS)
NCT01136733 (16) [back to overview]Phase 1b: Number of Participants With Dose-limiting Toxicity (DLT)
NCT01136733 (16) [back to overview]Area Under the Blood Concentration-Time Curve From 0 to 24 Hours for Everolimus When Administered Alone or in Combination With Lenvatinib
NCT01136733 (16) [back to overview]Phase 1b: Maximum Tolerated Dose (MTD) and Recommended Phase 2 (RP2) Dose
NCT01136733 (16) [back to overview]Area Under the Plasma Concentration-Time Curve From 0 to 24 Hours (AUC(0-24)) for Lenvatinib When Administered Alone or in Combination With Everolimus
NCT01136733 (16) [back to overview]Phase 2: Progression-Free Survival (PFS)
NCT01136733 (16) [back to overview]Summary of Blood Concentrations of Everolimus for Sparse PK Sampling for Phase 1b and Phase 2
NCT01136733 (16) [back to overview]Clinical Benefit Rate (CBR)
NCT01136733 (16) [back to overview]Disease Control Rate (DCR)
NCT01136733 (16) [back to overview]Time to Cmax (Tmax) for Everolimus When Administered Alone or in Combination With Lenvatinib
NCT01136733 (16) [back to overview]Durable Stable Disease (SD) Rate
NCT01136733 (16) [back to overview]Maximum Concentration (Cmax) of Lenvatinib in Plasma When Administered Alone or in Combination With Everolimus
NCT01136733 (16) [back to overview]Time to Cmax (Tmax) for Lenvatinib When Administered Alone or in Combination With Everolimus
NCT01149434 (6) [back to overview]Effect of RAD001 on Pharmacokinetics AUC(0-inf) of JI-101
NCT01149434 (6) [back to overview]Progression Free-Survival in the Ovarian Cancer Cohort
NCT01149434 (6) [back to overview]Safety and Tolerability of JI-101
NCT01149434 (6) [back to overview]Effect of JI 101 on Pharmacokinetics Area Under Curve (AUC) (0-inf) of RAD001
NCT01149434 (6) [back to overview]Tumor Response in the Ovarian Cancer Cohort
NCT01149434 (6) [back to overview]Tumor Response
NCT01150097 (6) [back to overview]Incidence Rate of Composite Efficacy Failure Defined as Treated Biopsy Proven Acute Rejection (tBPAR ), Graft Loss or Death
NCT01150097 (6) [back to overview]Incidence Rate of Composite Efficacy Failure Defined as Graft Loss or Death
NCT01150097 (6) [back to overview]Change in Renal Function
NCT01150097 (6) [back to overview]Incidence Rate of Composite Efficacy Failure Defined as Graft Loss or Death
NCT01150097 (6) [back to overview]Incidence Rate of tBPAR
NCT01150097 (6) [back to overview]Incidence Rate of Composite Efficacy Failure Defined as Treated Biopsy Proven Acute Rejection (tBPAR ), Graft Loss or Death
NCT01154335 (4) [back to overview]Progression-Free Survival (PFS)
NCT01154335 (4) [back to overview]Response Rate
NCT01154335 (4) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of the Combination of OSI-906 and Everolimus for the Treatment of Patients With Refractory Metastatic Colorectal Cancer.
NCT01154335 (4) [back to overview]Overall Survival (OS)
NCT01158651 (2) [back to overview]Common Terminology Criteria for Adverse Events (CTCAE) Events
NCT01158651 (2) [back to overview]RAD001 Response Rate Based on 2D MRI Change From Baseline
NCT01169701 (14) [back to overview]Change From Baseline in Cardiovascular Biomarkers, C-reactive Protein (CRP)
NCT01169701 (14) [back to overview]Percentage of Participants With Biopsy-proven Acute Rejection (BPAR), Graft Loss, Death and Lost to Follow up
NCT01169701 (14) [back to overview]Change From Baseline in the Cardiovascular Biomarker, Type 1 Procollagen Amino-terminal-propeptide (PINP)
NCT01169701 (14) [back to overview]Renal Function as Measured by Estimated Glomerular Filtration Rate (eGFR)
NCT01169701 (14) [back to overview]Renal Function Measured by Serum Creatinine
NCT01169701 (14) [back to overview]Change From Baseline in the Cardiovascular Biomarker, N-terminal Pro-brain Natriuretic Peptide Fraction (NT-proBNP)
NCT01169701 (14) [back to overview]Change From Baseline in the Cardiovascular Biomarker, Glycosylated Hemoglobin (HbA1c)
NCT01169701 (14) [back to overview]Change From Baseline in Cardiovascular Biomarkers: Troponin I and Collagen Type 1 C-telopeptide (ICTP)
NCT01169701 (14) [back to overview]Change From Baseline in the Cardiovascular Biomarker, Myeloperoxidase (MPO)
NCT01169701 (14) [back to overview]Renal Function as Measured by Creatinine Clearance
NCT01169701 (14) [back to overview]Pulse Wave Velocity (PWV)
NCT01169701 (14) [back to overview]Change From Baseline in Left Ventricular Mass Index (LVMI)
NCT01169701 (14) [back to overview]Percentage of Participants With Major Cardiovascular Events (MACE)
NCT01169701 (14) [back to overview]Change From Baseline in Mean 24 Hour Systolic and Diastolic Blood Pressure
NCT01184326 (7) [back to overview]Grade 4 Treatment-Related Toxicity Rate [Dose Finding]
NCT01184326 (7) [back to overview]Dose Limiting Toxicity (DLT) [Phase I Dose Finding]
NCT01184326 (7) [back to overview]Objective Response Rate [Expansion]
NCT01184326 (7) [back to overview]Median Progression-Free Survival [Expansion]
NCT01184326 (7) [back to overview]Mean Duration of Response [Expansion]
NCT01184326 (7) [back to overview]Grade 4 Treatment-Related Toxicity Rate [Expansion]
NCT01184326 (7) [back to overview]Maximum Tolerated Dose (MTD) [Phase I Dose Finding]
NCT01185366 (5) [back to overview]Progression Free Survival (PFS) for First Line Medication
NCT01185366 (5) [back to overview]Number of Participants Who Experienced Either a Grade 3 or 4 Adverse Event
NCT01185366 (5) [back to overview]Number of Participants With Best Overall Response for First Line Medication
NCT01185366 (5) [back to overview]Overall Survival of the First Line Therapy
NCT01185366 (5) [back to overview]Progression Free Survival (PFS) for Crossover Medication
NCT01198158 (4) [back to overview]Toxicities, Graded According to the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0
NCT01198158 (4) [back to overview]Progression-free Survival (PFS)
NCT01198158 (4) [back to overview]Objective Response Rate (CR + PR)
NCT01198158 (4) [back to overview]Overall Survival (OS)
NCT01206764 (5) [back to overview]Disease Control Rate (Stable Disease [SD] + Partial Response [PR] + Complete Response [CR]);
NCT01206764 (5) [back to overview]PFS (Progression-Free Survival)
NCT01206764 (5) [back to overview]Overall Survival
NCT01206764 (5) [back to overview]Objective Response Rate (ORR; Where ORR = CR + PR)
NCT01206764 (5) [back to overview]Duration of Response (DOR)
NCT01215136 (4) [back to overview]Number of Adverse Events as a Measure of Safety and Tolerability
NCT01215136 (4) [back to overview]Response Rate of Single-Agent Everolimus and Everolimus + Paclitaxel
NCT01215136 (4) [back to overview]Progression Free Survival
NCT01215136 (4) [back to overview]Overall Survival
NCT01229943 (3) [back to overview]Progression Free Survival
NCT01229943 (3) [back to overview]Overall Survival (OS)
NCT01229943 (3) [back to overview]Overall Response Rate
NCT01231399 (4) [back to overview]Progression-free Survival
NCT01231399 (4) [back to overview]Overall Survival
NCT01231399 (4) [back to overview]Number of Subject With Overall Response
NCT01231399 (4) [back to overview]Maximum Tolerated Dose (MTD) of Everolimus
NCT01231659 (6) [back to overview]Disease Control Rate (DCR)
NCT01231659 (6) [back to overview]Median Time to Overall Survival (OS)
NCT01231659 (6) [back to overview]Median Time to Progression-Free Survival (PFS)
NCT01231659 (6) [back to overview]Percentage of Participants With Overall Response Rate (ORR)
NCT01231659 (6) [back to overview]All Collected Deaths
NCT01231659 (6) [back to overview]Long-term Safety and Tolerability
NCT01239342 (4) [back to overview]Median Overall Survival (OS) in Months
NCT01239342 (4) [back to overview]Summary of Selected Toxicities Grade 3 or Greater Toxicity Based on the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0
NCT01239342 (4) [back to overview]Median Progression Free Survival (PFS) in Months
NCT01239342 (4) [back to overview]Overall Response Rate (ORR) Defined as Complete Response (CR) + Partial Response (PR)
NCT01239472 (2) [back to overview]Evaluation of Renal Function
NCT01239472 (2) [back to overview]Cytokines Evaluation
NCT01249027 (66) [back to overview]Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)
NCT01249027 (66) [back to overview]Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)
NCT01249027 (66) [back to overview]Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)
NCT01249027 (66) [back to overview]Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)
NCT01249027 (66) [back to overview]Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)
NCT01249027 (66) [back to overview]Number of Participants Experiencing Death
NCT01249027 (66) [back to overview]Number of Participants Experiencing Death
NCT01249027 (66) [back to overview]Number of Participants Experiencing Death
NCT01249027 (66) [back to overview]Number of Participants Experiencing Death
NCT01249027 (66) [back to overview]Number of Participants With All Target Lesion Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Lesion Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Lesion Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Lesions Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Lesions Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Vessel Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Vessel Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Vessel Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Vessel Revascularization
NCT01249027 (66) [back to overview]Number of Participants With All Target Vessel Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Any MI
NCT01249027 (66) [back to overview]Number of Participants With Any MI
NCT01249027 (66) [back to overview]Number of Participants With Any MI
NCT01249027 (66) [back to overview]Number of Participants With Any MI
NCT01249027 (66) [back to overview]Number of Participants With Any MI
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death and Any MI
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death and Any MI
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death and Any MI
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death and Any MI
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death and Any MI
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)
NCT01249027 (66) [back to overview]Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)
NCT01249027 (66) [back to overview]Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01249027 (66) [back to overview]Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01249027 (66) [back to overview]Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01249027 (66) [back to overview]Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01249027 (66) [back to overview]Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01249027 (66) [back to overview]Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))
NCT01249027 (66) [back to overview]Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))
NCT01249027 (66) [back to overview]Number of Participants Experiencing Death
NCT01249027 (66) [back to overview]Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))
NCT01249027 (66) [back to overview]Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))
NCT01249027 (66) [back to overview]Number of Participants With Major Bleeding Complications (According to GUSTO Classification)
NCT01249027 (66) [back to overview]Number of Participants With Major Bleeding Complications (According to GUSTO Classification)
NCT01249027 (66) [back to overview]Number of Participants With Major Bleeding Complications (According to GUSTO Classification)
NCT01249027 (66) [back to overview]Number of Participants With Major Bleeding Complications (According to GUSTO Classification)
NCT01249027 (66) [back to overview]Number of Participants With Major Bleeding Complications (According to GUSTO Classification)
NCT01249027 (66) [back to overview]Number of Participants With Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Revascularization
NCT01249027 (66) [back to overview]Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)
NCT01249027 (66) [back to overview]Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)
NCT01249027 (66) [back to overview]Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)
NCT01249027 (66) [back to overview]Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)
NCT01249027 (66) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)
NCT01249027 (66) [back to overview]Number of Participants With Stent Thrombosis
NCT01249027 (66) [back to overview]Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))
NCT01252251 (3) [back to overview]Number of Participants With Stable Disease (SD)
NCT01252251 (3) [back to overview]Median Progression Free Survival(PFS)
NCT01252251 (3) [back to overview]Median Overall Survival (OS)
NCT01259063 (5) [back to overview]Survival of Patients Treated
NCT01259063 (5) [back to overview]Complete Response (CR) Rate
NCT01259063 (5) [back to overview]Phase I - Dose-limiting Toxicity (DLT)
NCT01259063 (5) [back to overview]Phase II - Patients Who Are Free of Disease at 1 Year
NCT01259063 (5) [back to overview]Phase I - Maximum Tolerated Dose (MTD)
NCT01266148 (11) [back to overview]Number of Rejections Leading to Hemodynamic Compromise
NCT01266148 (11) [back to overview]Measured Glomerular Filtration Rate (mGFR), 12 Months After Heart Transplantation
NCT01266148 (11) [back to overview]Average Level of Protenuria at Week 52
NCT01266148 (11) [back to overview]Change in Quality of Life - Euro Quality of Life 5D (EQ-5D)
NCT01266148 (11) [back to overview]Change in Calculated Glomerular Filtration Rate From Pre-transplantation to Week 52
NCT01266148 (11) [back to overview]Progression of Chronic Allograft Vasculopathy (CAV) Based on Maximal Intimal Thickness (MIT) From Baseline to Week 52
NCT01266148 (11) [back to overview]Progression of Chronic Allograft Vasculopathy (CAV) Based on Incidence of Chronic Allograft Vasculopathy (CAV) From Baseline to Week 52
NCT01266148 (11) [back to overview]Occurrence of Treatment Failures up to 12 Months After Transplant
NCT01266148 (11) [back to overview]Calculated Glomerular Filtration Rate From Pre-Transplantation to Week 52
NCT01266148 (11) [back to overview]Lipid Profile at 12 Months
NCT01266148 (11) [back to overview]Change in Quality of Life Assessed by SF-36 (Minnesota Living With Heart Failure Questionnaire ([MLHF)]) From Pre-transplant to Week 52 of Treatment
NCT01270321 (2) [back to overview]Number of Participants With Progression-free Survival
NCT01270321 (2) [back to overview]Number of Participants With Response Per Responsive Evaluation Criteria in Solid Tumors Criteria (RECIST v1.0)
NCT01275222 (8) [back to overview]All Collected Deaths
NCT01275222 (8) [back to overview]4-Month Progression-free Survival (PFS) Rate - Phase II
NCT01275222 (8) [back to overview]Best Overall Response (BOR) as Assessed by Overall Response (Complete Response (CR) & Partial Response (PR)) - Phase I & II
NCT01275222 (8) [back to overview]Number of Participants With Adverse Events (AEs): Phase I & II
NCT01275222 (8) [back to overview]Overall Survival (OS) - Phase I & II
NCT01275222 (8) [back to overview]Progression-free Survival (PFS) - Phase II
NCT01275222 (8) [back to overview]Trough Concentrations for RAD001 and for Imatinib - Phase II
NCT01275222 (8) [back to overview]Trough Concentrations for RAD001 and for Imatinib - Phase II
NCT01276457 (4) [back to overview]Renal Function Assessed by Creatinine Clearance
NCT01276457 (4) [back to overview]Number of Participants Who Died, Number of Participants Who Lost Their Graft, and Number of Participants Who Died or Lost Their Graft
NCT01276457 (4) [back to overview]Number of Participants With Adverse Events (AE), Serious Adverse Events (SAE) or Deaths
NCT01276457 (4) [back to overview]Number of Participants With Biopsy-proven Acute Rejection
NCT01281865 (1) [back to overview]Response Rate (CR + PR) Assessed by RECIST 1.1 (Phase II)
NCT01283334 (2) [back to overview]Progression-free Survival (PFS)
NCT01283334 (2) [back to overview]To Measure the Safety and Clinical Effectiveness of the Combination of Carboplatin, Cetuximab and RAD001 in Patients With Advanced (Recurrent or Metastatic) Head and Neck Cancer
NCT01289912 (6) [back to overview]Evaluation of the Efficacy of RAD001 on Neurocognition in Patients With TSC Compared With Placebo.
NCT01289912 (6) [back to overview]Evaluation of the Safety of RAD001 on Neurocognition in Patients With TSC Compared With Placebo in Patients With TSC.
NCT01289912 (6) [back to overview]Comparison of Autism Spectrum Disorders Features Between Patients Taking RAD001 vs. Placebo
NCT01289912 (6) [back to overview]Comparison of Behavioral Problems Between Patients Taking RAD001 vs Placebo
NCT01289912 (6) [back to overview]Evaluation of the Efficacy of RAD001 on Neurocognition (Cambridge Neuropsychological Test Automated Battery) in Patients With TSC Compared With Placebo.
NCT01289912 (6) [back to overview]Comparison of Academic Skills Between Patients Taking RAD001 vs. Placebo
NCT01305941 (9) [back to overview]Extracranial Response
NCT01305941 (9) [back to overview]Time to Intracranial Progression.
NCT01305941 (9) [back to overview]Toxicity
NCT01305941 (9) [back to overview]Overall Survival
NCT01305941 (9) [back to overview]Intracranial Response Rate- MacDonald Criteria
NCT01305941 (9) [back to overview]Functional Assessment of Cancer Therapy- Brain (FACT-Br) Change From Baseline to Assess Impact of Everolimus in Combination With Trastuzumab and Vinorelbine on Quality of Life
NCT01305941 (9) [back to overview]Intracranial Objective Response Rate- Modified RECIST Criteria
NCT01305941 (9) [back to overview]Functional Assessment Cancer Therapy- Breast (FACT-B) From Baseline to 9 Weeks of Treatment to Assess Impact of Everolimus in Combination With Trastuzumab and Vinorelbine on Quality of Life
NCT01305941 (9) [back to overview]Extracranial Time to Progression
NCT01313559 (4) [back to overview]Number of Participants Without New Bone Lesions After 12 Weeks of Treatment
NCT01313559 (4) [back to overview]Number of Participants With > 50% Decline From Baseline PSA Level
NCT01313559 (4) [back to overview]Number of Participants With Progression Free Survival (PFS) Based on RECIST 1.1 Criteria
NCT01313559 (4) [back to overview]Number of Participants Alive and Progression Free After 12 Weeks of Treatment
NCT01317615 (6) [back to overview]Percentage of Participants Progression-free
NCT01317615 (6) [back to overview]Overall Survival (OS)
NCT01317615 (6) [back to overview]Percentage of Participants With Disease Control Rate (DCR)
NCT01317615 (6) [back to overview]Progression Free Survival (PFS)
NCT01317615 (6) [back to overview]Percentage of Participants With Overall Response Rate (ORR)
NCT01317615 (6) [back to overview]Percentage of Participants Progression-free
NCT01347554 (4) [back to overview]Any Bleeding
NCT01347554 (4) [back to overview]Device-oriented Composite Outcome
NCT01347554 (4) [back to overview]Device-oriented Composite Outcome
NCT01347554 (4) [back to overview]Stent Thrombosis
NCT01365468 (3) [back to overview]Number of Patients With Objective Radiographic Responses Based on Volumetric MRI Measurements (In Stratum 2 Only)
NCT01365468 (3) [back to overview]Number of Patients With Adverse Events Assessed by Common Toxicity Criteria for Adverse Events (CTCAE) V.04
NCT01365468 (3) [back to overview]Time to Disease Progression (TTP) Based on Change in Volumetric MRI Measurements in Children and Adults (In Stratum I Only)
NCT01374451 (10) [back to overview]Overall Survival (OS) Using Kaplan Meier Method
NCT01374451 (10) [back to overview]Progression-free Survival (PFS) Per Local Radiological Review
NCT01374451 (10) [back to overview]Summary of Pharmacokinetics (PK) for Everolimus for AUClast
NCT01374451 (10) [back to overview]Summary of Pharmacokinetics (PK) for Everolimus for CL/F
NCT01374451 (10) [back to overview]Summary of Pharmacokinetics (PK) for Everolimus for Cmax and Cmin
NCT01374451 (10) [back to overview]Summary of Pharmacokinetics (PK) for Everolimus for Tmax
NCT01374451 (10) [back to overview]Safety and Tolerability Profile of Everolimus Alone or in Combination With Pasireotide LAR
NCT01374451 (10) [back to overview]Summary of Pasireotide Concentrations Following Intramuscular Injection of Pasireotide LAR 60mg
NCT01374451 (10) [back to overview]Objective Response Rate (ORR) as Per Radiology Review
NCT01374451 (10) [back to overview]Disease Control Rate (DCR) as Per Radiology Review
NCT01376310 (1) [back to overview]Number of Participants With Adverse Events
NCT01379521 (3) [back to overview]Time to Progression (TTP) Based on the Modified RECIST Criteria
NCT01379521 (3) [back to overview]Percentage of Participants With a Decrease in the Sum of the of Longest Diameters (SLD) of Target Lesions From Baseline to 30 Months
NCT01379521 (3) [back to overview]Overall Survival (OS)
NCT01399918 (3) [back to overview]Participants Evaluated for Toxicity
NCT01399918 (3) [back to overview]Secondary Endpoint Will be the Overall Response Rate (ORR)
NCT01399918 (3) [back to overview]To Evaluate the Efficacy of Combining Everolimus and Bevacizumab in Patients With Advanced RCC of Non-clear Cell Histology
NCT01404325 (26) [back to overview]Incidence of Bacterial, Viral, and Fungal Infections at Month 12
NCT01404325 (26) [back to overview]Incidence of Acute Rejection Episodes at Month 6 and Month 12
NCT01404325 (26) [back to overview]High-Density Lipoprotein (HDL)Cholesterol Levels at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Exercise Capacity 6-Minute Walk Test(6MWT) at Month 6 and Month 12
NCT01404325 (26) [back to overview]Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Calculated Glomerular Filtration Rate (cGFR) According to Cystatin C-based Hoek's Formula at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Calculated Glomerular Filtration Rate (cGFR) According to Chronic Kidney Disease Epidemiology Collaboration (CKD EPI) at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Adherence to Target Ranges of Tacrolimus at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Adherence to Target Ranges of Everolimus at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Adherence to Target Ranges of Cyclosporine A (CsA) at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Calculated Glomerular Filtration Rate (cGFR) According to Chronic Kidney Disease Epidemiology Collaboration (CKD EPI) at 12 Months
NCT01404325 (26) [back to overview]Calculated Glomerular Filtration Rate (cGFR) According to Cockcroft-Gault at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Trough Levels of Everolimus at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Trough Levels of Tacrolimus at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Incidence of Patients Experiencing a Decline in GFR of < 10, 10-15, 15-20, 20-25 and > 25 mL/Min From Baseline to Month 6 and 12.
NCT01404325 (26) [back to overview]Trough Levels of Cyclosporine A (CsA) at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Triglyceride Levels at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Total Cholesterol Levels at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Quality of Life (QoL, SF36) at Month 6 and Month 12
NCT01404325 (26) [back to overview]Incidence of Treated Arterial Hypertension up to Month 12
NCT01404325 (26) [back to overview]Incidence of Renal Replacement Therapy at Month 6 and Month 12
NCT01404325 (26) [back to overview]Incidence of Graft Loss/Re-transplantation at Month 6 and Month 12
NCT01404325 (26) [back to overview]Low-Density Lipoprotein (LDL)Cholesterol Levels at Month 1, 3, 6, 9, 12
NCT01404325 (26) [back to overview]Incidence of Diabetes Mellitus up to Month 12
NCT01404325 (26) [back to overview]Incidence of Death at Month 6 and Month 12
NCT01404325 (26) [back to overview]Incidence of Bronchiolitis Obliterans Syndrome (BOS) at Month 6 and Month 12
NCT01410448 (17) [back to overview]Change From Baseline in Serum Creatinine - ITT
NCT01410448 (17) [back to overview]Change From Baseline in Serum Creatinine - Modified ITT
NCT01410448 (17) [back to overview]Percentage of Participants With Proteinuria
NCT01410448 (17) [back to overview]Percentage of Participants Who Experienced Treatment Failure - Worst-case Scenario
NCT01410448 (17) [back to overview]Percentage of Participants With a New Onset of Diabetes
NCT01410448 (17) [back to overview]Percentage of Participants With a New Onset of Malignancy
NCT01410448 (17) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate (eGFR) (Calculated With Modified Diet in Renal Disease (MDRD)-4 Formula - ITT
NCT01410448 (17) [back to overview]Percentage of Participants With Acute Rejection (AR)
NCT01410448 (17) [back to overview]Participant/Graft Survival Rate: Percentage of Participants With Failure Events of Death or Graft Loss - Worst-case Scenario
NCT01410448 (17) [back to overview]Percentage of Participants Without Wound Healing Complications - Worst-case Scenario
NCT01410448 (17) [back to overview]Duration of DGF
NCT01410448 (17) [back to overview]Patient Survival Rate: Percentage of Deaths - Worst-case Scenario
NCT01410448 (17) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate (eGFR) (Calculated With Modified Diet in Renal Disease (MDRD)-4 Formula - Modified ITT
NCT01410448 (17) [back to overview]Percentage of Participants Without Wound Healing Complications - Worst-case Scenario
NCT01410448 (17) [back to overview]Graft Survival Rate: Percentage of Participants With Graft Loss - Worst-case Scenario
NCT01410448 (17) [back to overview]Percentage of Participants With BPAR - Worst-case Scenario
NCT01410448 (17) [back to overview]Percentage of Participants With Delayed Graft Function (DGF) -
NCT01419639 (3) [back to overview]Audiologic Response
NCT01419639 (3) [back to overview]Change in Tumor Size From Baseline
NCT01419639 (3) [back to overview]Radiographic Response
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With DMR (All Death, All MI, All Revascularization)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Non-Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Procedural Success
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, Target Vessel Myocardial Infarction (TV-MI), ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Failure (TLF) (Cardiac Death,(Target Vessel Myocardial Infarction(TV-MI), ID-TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Lesion Revascularization (TLR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Target Vessel Revascularization (TVR)
NCT01425281 (106) [back to overview]Number of Participants With Acute Stent/Scaffold Thrombosis
NCT01425281 (106) [back to overview]Number of Participants With Acute/Subacute Stent/Scaffold Thrombosis
NCT01425281 (106) [back to overview]Number of Participants With Cumulative Stent/Scaffold Thrombosis
NCT01425281 (106) [back to overview]Number of Participants With Late Stent/Scaffold Thrombosis
NCT01425281 (106) [back to overview]Number of Participants With Subacute Stent/Scaffold Thrombosis
NCT01425281 (106) [back to overview]Number of Participants With Very Late Stent/Scaffold Thrombosis
NCT01425281 (106) [back to overview]Number of Participants With Non Target Vessel Revascularization (Non-TVR)
NCT01425281 (106) [back to overview]Absolute Difference (3 Years Post Nitrate- 3 Years Pre Nitrate) In-Scaffold Mean Lumen Diameter (MLD)
NCT01425281 (106) [back to overview]Absolute Difference (3 Years Post-nitrate - Post Procedure Post-nitrate) In-Scaffold Minimum Lumen Diameter
NCT01425281 (106) [back to overview]Device Success
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing All Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants Experiencing Cardiac Death/All MI
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Myocardial Infarction (Per Protocol Definition)
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01425281 (106) [back to overview]Number of Participants With All Revascularization
NCT01434602 (16) [back to overview]Maximum Tolerated Dose of Sorafenib for Participants With Recurrent Malignant Gliomas
NCT01434602 (16) [back to overview]Median Amount of Time a Participant Survives After Therapy for Participants With Recurrent Malignant Glioma (Group A, Group B, and Group C) Treated With Everolimus and Sorafenib Measured From the Time of Study Enrollment
NCT01434602 (16) [back to overview]Median Amount of Time Participants Survives Without Disease Progression for Groups A, B, and C Treated With Everolimus and Sorafenib
NCT01434602 (16) [back to overview]Number of Participants With a Dose-limiting Toxicity (DLT)
NCT01434602 (16) [back to overview]Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0)
NCT01434602 (16) [back to overview]Percentage of Participants Who Survived Without Disease Progression at 3 Months After Treatment for Glioblastoma Participants With Prior Bevacizumab Exposure
NCT01434602 (16) [back to overview]Rate of Participants Symptom Interference With Function
NCT01434602 (16) [back to overview]Percentage of Participants Who Survived Without Disease Progression at 6 Months After Treatment for Anaplastic Glioma (AG) Participants With no Prior Bevacizumab Exposure
NCT01434602 (16) [back to overview]Percentage of Participants Who Survived Without Disease Progression at 6 Months After Treatment for Glioblastoma Participants With no Prior Bevacizumab Exposure
NCT01434602 (16) [back to overview]Objective Response for Participants With Recurrent Malignant Gliomas (Group A, Group B, and Group C) Treated With Everolimus and Sorafenib
NCT01434602 (16) [back to overview]Rate of Participants Symptom Severity
NCT01434602 (16) [back to overview]Rate of Participants Symptom Severity
NCT01434602 (16) [back to overview]Rate of Participants Symptom Severity
NCT01434602 (16) [back to overview]Rate of Participants Symptom Interference With Function
NCT01434602 (16) [back to overview]Rate of Participants Symptom Interference With Function
NCT01434602 (16) [back to overview]Maximum Tolerated Dose of Everolimus for Participants With Recurrent Malignant Gliomas
NCT01488487 (4) [back to overview]Number of Individuals Experiencing Toxicity
NCT01488487 (4) [back to overview]Objective Response Rate (ORR)
NCT01488487 (4) [back to overview]Time to Progression (TTP)
NCT01488487 (4) [back to overview]Overall Survival (OS)
NCT01491672 (6) [back to overview]Duration of Response (DoR)
NCT01491672 (6) [back to overview]Clinical Benefit Rate (CBR)
NCT01491672 (6) [back to overview]Duration of PFS for Each First-line Treatment Cohort
NCT01491672 (6) [back to overview]Progression-free Survival (PFS) - All Participants
NCT01491672 (6) [back to overview]Overall Survival (OS)
NCT01491672 (6) [back to overview]Objective Response Rate (ORR)
NCT01499160 (2) [back to overview]Clinical Benefit Rate of Patients Treated With the Combination of Letrozole and Lapatinib and Then After Progression, Treated With Everolimus, Letrozole and Lapatinib.
NCT01499160 (2) [back to overview]PROGRESSION FREE SURVIVAL TUMOR ASSESSMENT
NCT01510327 (14) [back to overview]Myocardial Infarction (MI) Related to the Target Vessel
NCT01510327 (14) [back to overview]Maximum Observed Everolimus Blood Concentration (Cmax)
NCT01510327 (14) [back to overview]Definite + Probable Stent Thrombosis Rate Based on Academic Research Consortium (ARC) Definition
NCT01510327 (14) [back to overview]Definite + Probable Stent Thrombosis Rate Based on Academic Research Consortium (ARC) Definition
NCT01510327 (14) [back to overview]Definite + Probable Stent Thrombosis (ST) Rate Based on Academic Research Consortium (ARC) Definition
NCT01510327 (14) [back to overview]Area Under the Concentration Versus Time Curve (AUC 0-t) Everolimus
NCT01510327 (14) [back to overview]Area Under the Concentration Versus Time Curve (AUC 0-infinity) Everolimus
NCT01510327 (14) [back to overview]Area Under the Concentration Versus Time Curve (AUC 0-24), Everolimus
NCT01510327 (14) [back to overview]All Death
NCT01510327 (14) [back to overview]Time of Occurrence of Maximum Everolimus Concentration (Tmax)
NCT01510327 (14) [back to overview]Total Blood Clearance - Everolimus (CL)
NCT01510327 (14) [back to overview]Terminal Phase Half-life (t1/2) Everolimus
NCT01510327 (14) [back to overview]Target Vessel Revascularization (TVR)
NCT01510327 (14) [back to overview]Target Lesion Revascularization (TLR)
NCT01514448 (4) [back to overview]Overall Survival (OS) of Patients Treated With Everolimus After Failure of First-line Sunitinib or Pazopanib Therapy up to 48 Months
NCT01514448 (4) [back to overview]Percentage of Patients With Overall Response Rate (ORR) Treated With Everolimus After Failure of First-line Sunitinib or Pazopanib Therapy at Month 6
NCT01514448 (4) [back to overview]Percentage of Progression-free Patients by Month 6
NCT01514448 (4) [back to overview]Progression-Free Survival (PFS) as the Time Interval Between First Intake of Everolimus and First Documented Disease Progression or Death Due to Any Cause at 24 Months
NCT01524783 (10) [back to overview]Time to Definitive Deterioration in Functional Assessment of Cancer Therapy - General (FACT-G) Questionnaire Total Score
NCT01524783 (10) [back to overview]Overall Survival (OS)
NCT01524783 (10) [back to overview]Disease Control Rate (DCR) Based on Modified RECIST 1.0 and as Per Central Radiology Assessment
NCT01524783 (10) [back to overview]Pharmacokinetics (PK): Predose Concentration (Cmin) of Everolimus at Day 29
NCT01524783 (10) [back to overview]Overall Response Rate (ORR) as Per Modified RECIST 1.0 According to Central Evaluation
NCT01524783 (10) [back to overview]Time to Definitive Deterioration in World Health Organization (WHO) Performance Status (PS) Change
NCT01524783 (10) [back to overview]Probability of Participants Remaining Event-Free in Progression-Free Survival (PFS) Based on Central Radiology Assessment
NCT01524783 (10) [back to overview]Change From Baseline in Neuron Specific Enolase (NSE) Levels
NCT01524783 (10) [back to overview]Change From Baseline in Chromogranin A (CgA) Levels
NCT01524783 (10) [back to overview]All Collected Deaths
NCT01544491 (11) [back to overview]Incidence of Biopsy Proven Antibody Mediated Rejection.
NCT01544491 (11) [back to overview]Number of Participants Having Reached the Composite Efficacy Endpoint of Biopsy-proven Acute Rejection
NCT01544491 (11) [back to overview]To Evaluate Renal Function, Assessed by Glomerular Filtration Rate (eGFR) and Estimated by the Schwartz Formula (Extended), at Month 12
NCT01544491 (11) [back to overview]Chronic Allograft Nephropathy / Interstitial Fibrosis and Tubular Atrophy
NCT01544491 (11) [back to overview]Composite Efficacy Endpoint
NCT01544491 (11) [back to overview]Evaluation of Evolution of Renal Allograft Function Over Time
NCT01544491 (11) [back to overview]Growth/Development : Weight, Height, BMI : Change From Baseline
NCT01544491 (11) [back to overview]To Evaluate Renal Function, Assessed by Glomerular Filtration Rate (eGFR) and Estimated by the Schwartz Formula (Abbreviated), at Month 12 and 36
NCT01544491 (11) [back to overview]To Evaluate the Severity of BPAR (Acute T-cell Mediated Rejection Only) (Banff 2009)
NCT01544491 (11) [back to overview]To Evaluate the Time to Event of BPAR
NCT01544491 (11) [back to overview]Proteinuria (Urinary Protein/Creatinine Ratio)
NCT01545817 (7) [back to overview]Overall Survival From the Start (OSS) of Study Treatment
NCT01545817 (7) [back to overview]Overall Survival of Everolimus (OSE)
NCT01545817 (7) [back to overview]Progression Free Survival (PFS) Rates
NCT01545817 (7) [back to overview]Objective Response Rate (ORR) for the Pazopanib Treatment Period Using RECIST
NCT01545817 (7) [back to overview]Objective Response Rate (ORR) for the Everolimus Treatment Period Using RECIST
NCT01545817 (7) [back to overview]Progression Free Survival (PFS) for the Everolimus Treatment Period Using RECIST
NCT01545817 (7) [back to overview]PFS for the Pazopanib Treatment Period Using RECIST
NCT01551212 (6) [back to overview]Incidence of HCV Related Fibrosis
NCT01551212 (6) [back to overview]Estimated GFR - PP Set
NCT01551212 (6) [back to overview]Estimated Glomerular Filtration Rate (GFR)
NCT01551212 (6) [back to overview]Incidence of de Novo HCC Malignancies
NCT01551212 (6) [back to overview]Number of Participants With HCV
NCT01551212 (6) [back to overview]Percentage of Participants With Treated Biopsy Proven Acute Rejection (BPAR), Graft Loss or Death
NCT01563354 (12) [back to overview]Biochemical Response Rate (BRR) for 5HIAA Levels
NCT01563354 (12) [back to overview]Duration of Biochemical Response (DBR), by Treatment (Full Analysis Set)
NCT01563354 (12) [back to overview]Summary of Duration of Response (Months)
NCT01563354 (12) [back to overview]Percentage of Participants Progression-free at 9 Months Based on Response Evaluation Criteria In Solid Tumors v1.1 (RECIST v1.1)
NCT01563354 (12) [back to overview]12-month Disease Control Rate (DCR) and Objective Response Rate (ORR)
NCT01563354 (12) [back to overview]Summary of Progression-free Survival (PFS) Based on RECIST v1.1
NCT01563354 (12) [back to overview]Biochemical Response Rate (BRR) for Chromogranin A (CgA) Levels
NCT01563354 (12) [back to overview]Kaplan-Meier Estimates of Progression-free Survival (PFS)
NCT01563354 (12) [back to overview]Summary of Time to Response (Months)
NCT01563354 (12) [back to overview]Kaplan-Meier Event-free Probability Estimate for Biochemical Progression-free Survival Based on CgA Levels
NCT01563354 (12) [back to overview]Kaplan-Meier Event-free Probability Estimate Based on CgA Levels
NCT01563354 (12) [back to overview]Summary of Biochemical Progression-free Survival Based on CgA Levels by Treatment
NCT01582009 (5) [back to overview]Number of Participants With an Adverse Event.
NCT01582009 (5) [back to overview]Number of Participants With Clinical Response
NCT01582009 (5) [back to overview]Progression-free Survival (PFS)
NCT01582009 (5) [back to overview]Median Progression Free Survival
NCT01582009 (5) [back to overview]6-month Overall Survival Rate
NCT01595009 (16) [back to overview]Mean EORTC QLQ-G.I. NET21 Score (Core)
NCT01595009 (16) [back to overview]Change in EORTC QLQ-G.I. NET21 Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)
NCT01595009 (16) [back to overview]Number and Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) and Deaths During the Extension Phase (E1)
NCT01595009 (16) [back to overview]Investigator-assessed Best Overall Response During the Extension Phase (E1)
NCT01595009 (16) [back to overview]Number and Percentage of Participants With Ratings of 'no Problem, 'Some Problem' and 'Extreme Problem' in the EuroQol Five Dimensions Questionnaire (EQ-5D) (Core)
NCT01595009 (16) [back to overview]Number and Percentage of Participants With Ratings of 'no Problem, 'Some Problem' and 'Extreme Problem' in the EuroQol Five Dimensions Questionnaire (EQ-5D) (Core)
NCT01595009 (16) [back to overview]Change in EORTC QLQ-G.I. NET21 Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)
NCT01595009 (16) [back to overview]Investigator-assessed Progression Free Survival (PFS) (E1)
NCT01595009 (16) [back to overview]Investigator-assessed Progression Free Survival (PFS) (Core)
NCT01595009 (16) [back to overview]Change in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-C30 (EORTC QLQ-C30) Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)
NCT01595009 (16) [back to overview]Mean European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-C30 (EORTC QLQ-C30) Score (Core)
NCT01595009 (16) [back to overview]Mean EQ-5D Visual Analogue Scale (VAS) Score (Core)
NCT01595009 (16) [back to overview]Mean EORTC QLQ-G.I. NET21 Score (Core)
NCT01595009 (16) [back to overview]Number and Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) and Deaths (Core)
NCT01595009 (16) [back to overview]Change in EuroQol Five Dimensions Questionnaire (EQ-5D) Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)
NCT01595009 (16) [back to overview]Investigator-assessed Best Overall Response (Core)
NCT01596062 (9) [back to overview]Estimated Glomerular Filtration Rate (eGFR) at Day 8 and Week 24
NCT01596062 (9) [back to overview]AUC of Basiliximab Binding to CD25 Receptors From Day 0 to Day 84
NCT01596062 (9) [back to overview]Area Under the Curve (AUC) of CD25 Saturation by Basiliximab From Day 0 to Day 84
NCT01596062 (9) [back to overview]Saturation Rate of CD25 Antigen Saturation by Basiliximab
NCT01596062 (9) [back to overview]Percentage of Participants With Biopsy Proven Acute Rejection (BPAR) According to Type and Severity
NCT01596062 (9) [back to overview]Percentage of Participants With of Biopsy Proven Acute Rejection (BPAR)
NCT01596062 (9) [back to overview]Percentage of T-cells That Bind Basiliximab to CD25 Receptors
NCT01596062 (9) [back to overview]Proportion of CD3+, CD4+, CD8+, CD19+ and CD56+ T Cells
NCT01596062 (9) [back to overview]Percentage of Participants With of Treatment Failures
NCT01598987 (7) [back to overview]Growth Development - Height at Baseline and Month 24
NCT01598987 (7) [back to overview]Growth Development - Height at Baseline and Month 12
NCT01598987 (7) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate - Month 12
NCT01598987 (7) [back to overview]Change From Baseline in Estimated Glomerular Filtration Rate - Month 24
NCT01598987 (7) [back to overview]Kaplan-Meier Estimates for Failure Rates of Efficacy Endpoints
NCT01598987 (7) [back to overview]Growth Development - Weight at Baseline and Month 24
NCT01598987 (7) [back to overview]Growth Development - Weight at Baseline and Month 12
NCT01624948 (6) [back to overview]Evidence of Reduction of BK Viruria and/or Clearance of BK Viremia
NCT01624948 (6) [back to overview]Proteinuria
NCT01624948 (6) [back to overview]p70S6 Kinase Phosphorylation
NCT01624948 (6) [back to overview]Cholesterol
NCT01624948 (6) [back to overview]Evaluation for the Development of BK Virus Nephropathy or Doubling of BK Viremia Levels
NCT01624948 (6) [back to overview]Median Between the Calculated Mean Residual Expression of NFAT-regulated Genes
NCT01625377 (13) [back to overview]Change From Baseline (Randomization) in Serum Creatinine
NCT01625377 (13) [back to overview]Number of Patients With Treated or Not Treated Biopsy Proven Acute Rejection (BPAR)
NCT01625377 (13) [back to overview]Change From Baseline (Randomization) in Creatinine Clearance Estimated Using the Adjusted Cockcroft-Gault Formula
NCT01625377 (13) [back to overview]Change From Baseline (Randomization) in Glomerular Filtration Rate Estimated by CKD-EPI Formula
NCT01625377 (13) [back to overview]Change From Baseline (Randomization) in Renal Function
NCT01625377 (13) [back to overview]Change From Baseline (Randomization) in Urine Protein/Creatinine Ratio
NCT01625377 (13) [back to overview]Change From Baseline (Randomization) in Glomerular Filtration Rate Estimated by Abbreviated Modification of Diet in Renal Disease (MDRD) Formula
NCT01625377 (13) [back to overview]Number of Patients in Different Stages of Chronic Kidney Diseases According to the K/DOQI Classification System
NCT01625377 (13) [back to overview]Number of Patients Reported With Different Categories of Severity of BPAR According to Banff Classification
NCT01625377 (13) [back to overview]Number of Patients With Any Adverse Events, Serious Adverse Events, Death and Premature Discontinuation
NCT01625377 (13) [back to overview]Number of Patients With Death or Graft Loss
NCT01625377 (13) [back to overview]Number of Patients With Treated or Untreated BPAR With RAI Score Greater Than 3
NCT01625377 (13) [back to overview]Number of Patients With Treatment Failures
NCT01627067 (5) [back to overview]Overall Survival (OS)
NCT01627067 (5) [back to overview]Progression-Free Survival (PFS)
NCT01627067 (5) [back to overview]Compare Overall Survival (OS) Between the Number of Obese and Overweight Participants
NCT01627067 (5) [back to overview]Compare Progression Free Survival (PFS) Between the Number of Obese and Overweight Participants
NCT01627067 (5) [back to overview]Number of Participants With Response
NCT01628913 (1) [back to overview]Progression Free Survival (PFS)
NCT01637090 (5) [back to overview]Efficacy of Treatment
NCT01637090 (5) [back to overview]Progression-free Survival
NCT01637090 (5) [back to overview]Effect of mTOR on Tumors
NCT01637090 (5) [back to overview]Time to Response
NCT01637090 (5) [back to overview]Number of Participants With Adverse Events as a Measure of Safety and Tolerability
NCT01642186 (5) [back to overview]Median PFS
NCT01642186 (5) [back to overview]Number of Participants With One or More Adverse Events/Toxicity
NCT01642186 (5) [back to overview]Number of Participants With Tissue Biomarkers Collected
NCT01642186 (5) [back to overview]Percentage of Participants With Stable Disease
NCT01642186 (5) [back to overview]Median Overall Survival (OS)
NCT01653847 (5) [back to overview]Change in T Cell & B Cell Generation
NCT01653847 (5) [back to overview]Renal Allograft Survival
NCT01653847 (5) [back to overview]Patient Survival
NCT01653847 (5) [back to overview]Acute Rejection
NCT01653847 (5) [back to overview]Change in Glomerular Filtration Rate (GFR)
NCT01661283 (3) [back to overview]Vascular Endothelial Growth Factor (VEGF) and Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) Levels at Baseline and Pre-Cycles 3 and 5
NCT01661283 (3) [back to overview]Clinical Benefit Rate (Complete Response, Partial Response, and Stable Disease at ≥ 4 Months Using World Health Organization (WHO) Criteria) of Everolimus in Combination With Bevacizumab
NCT01661283 (3) [back to overview]Number of Participants With Response Stratified by Individuals With Sporadic or NF1 Associated MPNST
NCT01665768 (2) [back to overview]Safety as Assessed by Avoidance of Grade 3-4 Adverse Events
NCT01665768 (2) [back to overview]Event Free Survival (EFS)
NCT01668784 (11) [back to overview]Number of Participants With Abnormal Hematology and Serum Chemistry Laboratory Parameters by Worse CTC Grade - SI Units
NCT01668784 (11) [back to overview]Number of Participants Meeting Marked Laboratory Abnormality Criteria in Specific Liver and Thyroid Tests
NCT01668784 (11) [back to overview]Overall Survival (OS) at Primary Endpoint
NCT01668784 (11) [back to overview]Investigator-assessed Objective Response Rate (ORR)
NCT01668784 (11) [back to overview]Extended Collection to Post Hoc Overall Survival (OS)
NCT01668784 (11) [back to overview]Investigator-assessed Duration of Objective Response
NCT01668784 (11) [back to overview]Number of Participants With Serious Adverse Events, Death, Discontinuation Due to Adverse Events
NCT01668784 (11) [back to overview]Investigator-assessed Time of Progression-free Survival (PFS)
NCT01668784 (11) [back to overview]Percentage of Participants With Disease-related Symptom Progression (DRSP)
NCT01668784 (11) [back to overview]Overall Survival (OS) by Programmed Death-Ligand 1 (PD-L1) Expression Level
NCT01668784 (11) [back to overview]Investigator-assessed Time to Objective Response
NCT01674140 (4) [back to overview]Distant Recurrence-Free Survival (DRFS)
NCT01674140 (4) [back to overview]Invasive Disease-Free Survival (IDFS)
NCT01674140 (4) [back to overview]Overall Survival (OS)
NCT01674140 (4) [back to overview]Toxicity Based on Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0, Assessed up to 10 Years.
NCT01680861 (7) [back to overview]Incidence of Chronic Allograft Nephropathy (CAI) at 12 Months Post-transplant
NCT01680861 (7) [back to overview]BPAR (Biopsy-proven Acute Rejection) Incidence During the First 12 Months Post-transplant
NCT01680861 (7) [back to overview]Discontinuance of Any Study Medication (Tacrolimus, Everolimus, or EC-MPS)
NCT01680861 (7) [back to overview]eGFR (Calculated Glomerular Filtration Rate), i.e., Renal Function, at 1 Month Post-transplant.
NCT01680861 (7) [back to overview]eGFR (Renal Function) at 6 Months Post-transplant
NCT01680861 (7) [back to overview]eGFR (Renal Function) at Month 3 Post-transplant
NCT01680861 (7) [back to overview]Graft Loss (Return to Permanent Dialysis or Death)
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Overall Health at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment: Progression-free Survival (PFS)
NCT01698918 (23) [back to overview]First-line Treatment: Overall Response Rate (ORR)
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness Limiting Swallowing, Drinking, Eating, Talking and Sleeping at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity Affecting Daily Activities at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment: Duration of First Stomatitis Based on OSDQ
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Duration of First Stomatitis Based on OSDQ
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness Limiting Swallowing, Drinking, Eating, Talking and Sleeping at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment: Clinical Benefit Rate (CBR)
NCT01698918 (23) [back to overview]All Collected Deaths
NCT01698918 (23) [back to overview]Second-line Treatment: Progression-free Survival (PFS)
NCT01698918 (23) [back to overview]First-line Treatment (Stomatitis Sub-study): Time to First Stomatitis Episode as Assessed by the OSDQ
NCT01698918 (23) [back to overview]First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment: Time to First Stomatitis Episode as Assessed by the Oral Stomatitis Daily Questionnaire (OSDQ)
NCT01698918 (23) [back to overview]First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity Affecting Daily Activities at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]Second-line Treatment: Overall Response Rate (ORR)
NCT01698918 (23) [back to overview]First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Overall Health at the End of the First Stomatitis Episode
NCT01698918 (23) [back to overview]Second-line Treatment: Clinical Benefit Rate (CBR)
NCT01698918 (23) [back to overview]Number of Participants With Clinical Benfit During Extension Phase
NCT01698918 (23) [back to overview]Overall Survival (OS)
NCT01713946 (22) [back to overview]Seizure Free Rates by Time Window
NCT01713946 (22) [back to overview]Long Term Evaluation: Relationship Between Seizure Frequency and Time-normalized Everolimus Concentration at Trough (Cmin,TN) - Repeated Measures Analysis
NCT01713946 (22) [back to overview]Core Phase: Median Percentage Change From Baseline in Seizure Frequency by Time Normalized Minimum Concentration
NCT01713946 (22) [back to overview]Core Phase: Food & Drug Administration (FDA): Percentage Change From Baseline in Partial Onset-seizure Frequency
NCT01713946 (22) [back to overview]Core Phase: Percentage of Patients With at Least a 25% Reduction in Seizure Frequency
NCT01713946 (22) [back to overview]Core Phase: Impact of Everolimus on Anti-epileptic Drugs (AEDs) Concentrations
NCT01713946 (22) [back to overview]Percentage of Seizure-free Patients During the Maintenance Period of the Core Phase
NCT01713946 (22) [back to overview]Core Phase: Change From Baseline in the Overall Vineland-II Adaptive Behavior Composite (ABC) Score
NCT01713946 (22) [back to overview]Long Term Evaluation: Incidence of Suicide Attempt, Suicidal Ideation or Behavior During Core Phase Per Columbia Suicide Severity Rating Scale (C-SSRS) Outcomes
NCT01713946 (22) [back to overview]Core Phase: Response Rate in Seizure Frequency by Time Normalized Minimum Concentration
NCT01713946 (22) [back to overview]Core Phase: European Medicine Agency (EMA): Seizure Frequency Response Rate
NCT01713946 (22) [back to overview]Core Phase: Changes From Baseline in Number of Seizure-free Days
NCT01713946 (22) [back to overview]Core Phase: Change From Baseline in the QOLIE-31-P Overall Quality-of-life Score for Patients Aged >=18 Years
NCT01713946 (22) [back to overview]Long Term Evaluation: Effect of Everolimus Over Time in the Overall Wechsler Nonverbal Composite Score
NCT01713946 (22) [back to overview]Long Term Evaluation: Effect of Everolimus Over Time in the Overall Vineland-II Adaptive Behavior Composite (ABC) Score
NCT01713946 (22) [back to overview]Core Phase: Probability That a Patient Remains On-treatment up to a Specified Time Point
NCT01713946 (22) [back to overview]Core Phase: Incidence of Suicide Attempt, Suicidal Ideation or Behavior During Core Phase Per Columbia Suicide Severity Rating Scale (C-SSRS) Outcomes
NCT01713946 (22) [back to overview]Core Phase: Distribution of Reduction From Baseline in Seizure Frequency
NCT01713946 (22) [back to overview]Core Phase: Change From Baseline in the QOLIE-AD-48 Overall Quality-of-life Score for Patients >=11 to 18 Years
NCT01713946 (22) [back to overview]Core Phase: Change From Baseline in Wechsler Nonverbal Composite Score
NCT01713946 (22) [back to overview]Long Term Evaluation: Percentage Change From Start of Everolimus in Seizure Frequency by Time Window
NCT01713946 (22) [back to overview]Core Phase: Change From Baseline in the QOLCE Overall Quality-of-life Score for Patients <11 Years
NCT01734512 (1) [back to overview]Percentage of Participants With Progression Free Survival at 6 Months
NCT01743560 (10) [back to overview]Overall Survival (OS) Events (Number of Deaths) - FAS
NCT01743560 (10) [back to overview]Overall Survival (OS) - % Event-free Probability Estimate - FAS
NCT01743560 (10) [back to overview]Change From Baseline EORTC Quality of Life Questionnaire of Cancer Patients QLQ-C30 at Each Time Point
NCT01743560 (10) [back to overview]Progression-free Survival (PFS) - % Event-free Probability Estimate - FAS
NCT01743560 (10) [back to overview]Progression-free Survival (PFS) Events as Per Investigators - FAS
NCT01743560 (10) [back to overview]Change From Baseline in EuroQoL 5-dimension Visual Analogue Scores - FAS
NCT01743560 (10) [back to overview]Overall Response Rate of Everolimus and Exemestane Treatment in Postmenopausal Women With Hormone Receptor Positive Locally Advanced or Metastatic Breast Cancer
NCT01743560 (10) [back to overview]Progression-free Survival (PFS) by Median Time in Weeks as Per Investigators - FAS
NCT01743560 (10) [back to overview]Percentage of Patient Responses in EuroQoL 5-dimension Questionnaire - FAS
NCT01743560 (10) [back to overview]Best Overall Response of Everolimus and Exemestane Treatment in Postmenopausal Women With Hormone Receptor Positive Locally Advanced or Metastatic Breast Cancer
NCT01783444 (9) [back to overview]Mean Change in Treatment Satisfaction Questionnaire for Medication (TSQM) Between Week 3 and 12
NCT01783444 (9) [back to overview]Time to Eastern Cooperative Oncology Group (ECOG) Performance Deterioration
NCT01783444 (9) [back to overview]Time to 10% Definitive Deterioration in the Global Health Status / Quality of Life
NCT01783444 (9) [back to overview]Progression Free Survival (PFS) - Everolimus Plus Exemestane Versus Capecitabine Alone
NCT01783444 (9) [back to overview]Progression Free Survival (PFS) - Everolimus Plus Exemestane Versus Everolimus Alone
NCT01783444 (9) [back to overview]Clinical Benefit Rate (CBR)
NCT01783444 (9) [back to overview]Overall Response Rate (ORR)
NCT01783444 (9) [back to overview]All Collected Deaths
NCT01783444 (9) [back to overview]Overall Survival (OS)
NCT01784861 (8) [back to overview]Disease Stabilization Rate - Phase II
NCT01784861 (8) [back to overview]Progression Free Survival (PFS) - Phase II
NCT01784861 (8) [back to overview]Recommended Phase II Dose of X-82
NCT01784861 (8) [back to overview]Number of Participants With Toxicity - Phase II
NCT01784861 (8) [back to overview]Overall Toxicities - Phase I
NCT01784861 (8) [back to overview]Number of Participants With Dose Limiting Toxicities - Phase I
NCT01784861 (8) [back to overview]Objective Response Rate (Complete Response + Partial Response) - Phase II
NCT01784861 (8) [back to overview]Overall Survival - Phase II
NCT01789281 (2) [back to overview]Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT01789281 (2) [back to overview]Percentage of Patients With Clinical Benefit
NCT01797120 (4) [back to overview]Clinical Benefit Rate
NCT01797120 (4) [back to overview]Objective Response Rate
NCT01797120 (4) [back to overview]Overall Survival
NCT01797120 (4) [back to overview]Progression-free Survival
NCT01827384 (3) [back to overview]Number of Participants With an Objective Response
NCT01827384 (3) [back to overview]Proportion of Participants With 4 Month Progression-free Survival (PFS)
NCT01827384 (3) [back to overview]Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v5.0)
NCT01843348 (11) [back to overview]Percent of Participants With Delayed Graft Function by Day
NCT01843348 (11) [back to overview]Glomular Filtration Rate (GFR) mL/Min Via Cockcroft- Gault Method at Month 12 Post Transplant
NCT01843348 (11) [back to overview]Glomular Filtration Rate (GFR) Via Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) Method at Month 12 Post Transplant
NCT01843348 (11) [back to overview]Glomular Filtration Rate (GFR) Via Modification of Diet in Renal Disease (MDRD) Method at Month 12 Post Transplant
NCT01843348 (11) [back to overview]Duration of Wound Healing
NCT01843348 (11) [back to overview]Glomular Filtration Rate (GFR) mL/Min Via Nankivell Method at Month 12 - Standard Regimen vs Certican Regimens
NCT01843348 (11) [back to overview]Percent of Participants With Delayed Graft Function and Slow Graft Function
NCT01843348 (11) [back to overview]Percent of Participants With Viral Infections
NCT01843348 (11) [back to overview]Percent of Participants With Wound Healing Complications During Study
NCT01843348 (11) [back to overview]Percentage of Participants With Composite Treatment Failure Endpoints - Difference Between Groups at Month 12
NCT01843348 (11) [back to overview]Percentage of Participants With Treatment Failure Endpoints at Month 12
NCT01865747 (3) [back to overview]Objective Response Rate (ORR)
NCT01865747 (3) [back to overview]Overall Survival (OS)
NCT01865747 (3) [back to overview]Progression-free Survival (PFS)
NCT01888432 (16) [back to overview]Composite Efficacy Failure of Treated Biopsy in Everolimus With Reduced Tacrolimus Group Compared to Standard Tacrolimus in Patients From Japan Only
NCT01888432 (16) [back to overview]Number of Participants With Composite of tBPAR, Graft Loss, and Death
NCT01888432 (16) [back to overview]Number of Participants With Time to Recurrence of HCC in Subjects With a Diagnosis of HCC at the Time of Liver Transplantation
NCT01888432 (16) [back to overview]Compare Renal Function Over Time Assessed by the Change by eGFR, Post-randomization
NCT01888432 (16) [back to overview]Renal Function by Estimated Glomerular Filtration Rate (eGFR) From Randomization
NCT01888432 (16) [back to overview]Number of Subjects Experiencing Adverse Events/Infections by SOC
NCT01888432 (16) [back to overview]Number of Participants With Composite Efficacy Failure of Treated Biopsy Proven Acute Rejection, Graft Loss or Death in Everolimus With Reduced Tacrolimus Group Compared to Standard Tacrolimus
NCT01888432 (16) [back to overview]Compare Incidence of tAR
NCT01888432 (16) [back to overview]Renal Function by Estimated Glomerular Filtration Rate (All Extension Patients)
NCT01888432 (16) [back to overview]Compare Incidence of a Composite of Death or Graft Loss
NCT01888432 (16) [back to overview]Compare Incidence of AR
NCT01888432 (16) [back to overview]Compare Incidence of BPAR
NCT01888432 (16) [back to overview]Compare Incidence of Death
NCT01888432 (16) [back to overview]Compare Incidence of Graft Loss
NCT01888432 (16) [back to overview]Compare Incidence of Notable Safety Events (SAEs, Infections and Serious Infections Leading to Premature Discontinuation)
NCT01888432 (16) [back to overview]Compare Incidence of tBPAR
NCT01894152 (38) [back to overview]Number of All Death (Cardiac, Vascular, and Non-cardiovascular)
NCT01894152 (38) [back to overview]Number of All Deaths, Myocardial Infarction, Any Repetitive Revascularization Composite Endpoints
NCT01894152 (38) [back to overview]Number of Participants With All Target Vessel Revascularization (TVR)
NCT01894152 (38) [back to overview]Number of Participants With All TLR
NCT01894152 (38) [back to overview]Number of Participants With All TLR
NCT01894152 (38) [back to overview]Number of Participants With ID-TVR (TLR and TVR, Non-target Lesion)
NCT01894152 (38) [back to overview]Number of Participants With Cardiac Death and All Myocardial Infarction (MI) (Q-wave and Non-Q Wave) Composite Endpoint
NCT01894152 (38) [back to overview]Number of Participants With All TVR (TLR and TVR, Non-target Lesion)
NCT01894152 (38) [back to overview]Number of Participants With ID-TVR (TLR and TVR, Non-target Lesion)
NCT01894152 (38) [back to overview]Number of Participants With Target Lesion Failure (TLF)
NCT01894152 (38) [back to overview]Number of Participants With Sub-acute Stent Thrombosis
NCT01894152 (38) [back to overview]Number of Participants With ID-TVR, Non-target Lesion
NCT01894152 (38) [back to overview]Number of Participants With ID-TVR, Non-target Lesion
NCT01894152 (38) [back to overview]Number of Participants With Late Stent Thrombosis
NCT01894152 (38) [back to overview]Number of Participants With Overall Stent Thrombosis
NCT01894152 (38) [back to overview]Number of All Death (Cardiac, Vascular, and Non-cardiovascular)
NCT01894152 (38) [back to overview]Number of Participants With Early Stent Thrombosis
NCT01894152 (38) [back to overview]Number of All Deaths, Myocardial Infarction, Any Repetitive Revascularization Composite Endpoints
NCT01894152 (38) [back to overview]Number of Participants With All TVR (TLR and TVR, Non-target Lesion)
NCT01894152 (38) [back to overview]Number of Participants With Target Lesion Failure (TLF)
NCT01894152 (38) [back to overview]Number of Participants With Very Late Stent Thrombosis
NCT01894152 (38) [back to overview]Number of Participants With Acute Stent Thrombosis
NCT01894152 (38) [back to overview]Number of Participants With Target Vessel Failure (ID-TVF) (Cardiac Death, All Myocardial Infarctions and Ischemia-driven Target Vessel Revascularization)
NCT01894152 (38) [back to overview]Number of Participants With Target Vessel ARC MI
NCT01894152 (38) [back to overview]Number of Participants With Target Vessel ARC MI
NCT01894152 (38) [back to overview]Number of Participants With Composite Rate of All Deaths and Myocardial Infarctions (MI)
NCT01894152 (38) [back to overview]Number of Participants With ID-TLR
NCT01894152 (38) [back to overview]Number of Participants With ID-TLR
NCT01894152 (38) [back to overview]Number of Participants With Target Vessel Failure (ID-TVF) (Cardiac Death, All Myocardial Infarctions and Ischemia-driven Target Vessel Revascularization)
NCT01894152 (38) [back to overview]Number of Participants With Cardiogenic Death, Target Vessel Blood Flow Myocardial Infarction, Target Lesion Revascularization Composite Endpoint
NCT01894152 (38) [back to overview]Number of Participants With Cardiogenic Death, Target Vessel Blood Flow Myocardial Infarction, Target Lesion Revascularization Composite Endpoint
NCT01894152 (38) [back to overview]Number of Participants With Composite Rate of All Deaths and Myocardial Infarctions (MI)
NCT01894152 (38) [back to overview]Number of Participants With All Target Vessel Revascularization (TVR)
NCT01894152 (38) [back to overview]Number of Participants With All Revascularization (Target Lesion, Target Vessel, and Non-target Vessel) (PCI and Coronary Artery Bypass Graft [CABG])
NCT01894152 (38) [back to overview]Number of Participants With All Revascularization (Target Lesion, Target Vessel, and Non-target Vessel) (PCI and Coronary Artery Bypass Graft [CABG])
NCT01894152 (38) [back to overview]Number of Participants With All Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01894152 (38) [back to overview]Number of Participants With All Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)
NCT01894152 (38) [back to overview]Number of Participants With Cardiac Death and All Myocardial Infarction (MI) (Q-wave and Non-Q Wave) Composite Endpoint
NCT01911546 (4) [back to overview]The Number of Polyoma BK Viremia Patients
NCT01911546 (4) [back to overview]The Number of CMV Viremia
NCT01911546 (4) [back to overview]Incidence of Cell Mediated Rejection (CMR)
NCT01911546 (4) [back to overview]Incidence of Antibody Mediated Rejection (ABMR)
NCT01929642 (7) [back to overview]Frequency of Seizures Assessed by Total Number of Seizures
NCT01929642 (7) [back to overview]Feasibility Measurements of Parental Stress
NCT01929642 (7) [back to overview]Cognitive Function as Assessed by the Capute Scale
NCT01929642 (7) [back to overview]Caregiver Burden
NCT01929642 (7) [back to overview]Number of Participants With Compliance to the Treatment Protocol.
NCT01929642 (7) [back to overview]Total Number of Aggressions or Self-injuries
NCT01929642 (7) [back to overview]Repetitive Behavior
NCT01931163 (1) [back to overview]Tumor Response
NCT01935128 (20) [back to overview]Leukopenia
NCT01935128 (20) [back to overview]Infection Requiring Hospitalization
NCT01935128 (20) [back to overview]Impaired Glucose Tolerance
NCT01935128 (20) [back to overview]Graft Survival
NCT01935128 (20) [back to overview]Gastrointestinal Complaints
NCT01935128 (20) [back to overview]Development of Donor Specific Antibody
NCT01935128 (20) [back to overview]Cytomegalovirus
NCT01935128 (20) [back to overview]BK Nephropathy
NCT01935128 (20) [back to overview]BK Infection
NCT01935128 (20) [back to overview]Biopsy Proven Acute Rejection
NCT01935128 (20) [back to overview]Cardiovascular Complications
NCT01935128 (20) [back to overview]Lipid Levels
NCT01935128 (20) [back to overview]Thrombocytopenia
NCT01935128 (20) [back to overview]Renal Function
NCT01935128 (20) [back to overview]Proteinuria
NCT01935128 (20) [back to overview]Pneumonitis
NCT01935128 (20) [back to overview]Patient Survival
NCT01935128 (20) [back to overview]Neurotoxicity
NCT01935128 (20) [back to overview]Mouth Ulcers
NCT01935128 (20) [back to overview]Malignancies
NCT01936519 (6) [back to overview]Recipient and Donor Genotyping for Selected Variants of CYP3A5, ABCB1 (MDR1), and CYP4A Genes
NCT01936519 (6) [back to overview]Renal Function as Measured by 24 Hour Urine Creatinine Clearance
NCT01936519 (6) [back to overview]Renal Function as Measured by Cockcroft Gault Creatinine Clearance
NCT01936519 (6) [back to overview]Renal Function as Measured by Iothalamate Clearance
NCT01936519 (6) [back to overview]Renal Function as Measured by Modification of Diet in Renal Disease (MDRD) Estimated Glomerular Filtration Rate (eGFR)
NCT01936519 (6) [back to overview]Renal Function as Measured by Serum Creatinine Level
NCT01976390 (1) [back to overview]Number of Participants With Composite Endpoint of Graft Survival (Non-death Censored) and Biopsy Proven Acute Rejection at 1 Year
NCT02017860 (1) [back to overview]Number of Pariticipants With Adverse Events as a Measure of Safety and Tolerability
NCT02023905 (6) [back to overview]Rate of Reduction in Seizures
NCT02023905 (6) [back to overview]Progression-Free Survival Rate (PFS) (Arms 1 and 2)
NCT02023905 (6) [back to overview]Progression-Free Survival Rate (PFS) (Arm 3)
NCT02023905 (6) [back to overview]Overall Survival Rate (OS)
NCT02023905 (6) [back to overview]Median Progression Free Survival (PFS)
NCT02023905 (6) [back to overview]Objective Response Rate (ORR)
NCT02031536 (2) [back to overview]Overall Survival (OS)
NCT02031536 (2) [back to overview]Disease Free Survival (DFS)
NCT02061397 (2) [back to overview]Safety of Simvastatin in the Treatment of LAM-S and LAM-TS Patients
NCT02061397 (2) [back to overview]Percent Predicted FEV1
NCT02069093 (6) [back to overview]Time to Resolution of Stomatitis From Grade 2 or Greater to Grade 1 or Less
NCT02069093 (6) [back to overview]Blood Concentration of Everolimus and Exemestane
NCT02069093 (6) [back to overview]Dose Intensity of Everolimus and Exemestane
NCT02069093 (6) [back to overview]Number of Participants With All Grades of Stomatitis
NCT02069093 (6) [back to overview]Number of Participants With Stomatitis Grade ≥ 2
NCT02069093 (6) [back to overview]Median Number of Mouthwashes Per Day
NCT02073565 (4) [back to overview]Percentage of Healthy Tissue Coverage That Was Greater Than 40 Micrometers
NCT02073565 (4) [back to overview]Number of Patients Exhibiting Human Antimurine Antibody (HAMA) Reaction
NCT02073565 (4) [back to overview]Number of Participants With Target Vessel Failure (TVF)
NCT02073565 (4) [back to overview]Number of Patients With Clinically and Functionally Ischemia-Driven Target Lesion Revascularization (TLR)
NCT02096107 (6) [back to overview]Adverse Drug Reactions
NCT02096107 (6) [back to overview]Percentage of Participants Discontinuing or Modifying Immunosuppressant Use
NCT02096107 (6) [back to overview]Kidney Allograft Survival
NCT02096107 (6) [back to overview]Infection
NCT02096107 (6) [back to overview]Renal Function Measured by Estimated Glomerular Filtration Rate (eGFR)
NCT02096107 (6) [back to overview]Kidney Allograft Fibrosis Assessment
NCT02098876 (8) [back to overview]In Stent: Mean Thickness of Strut Coverage at Follow up
NCT02098876 (8) [back to overview]In Stent Mean Cross-sectional Area of Neo-intimal Tissue Coverage
NCT02098876 (8) [back to overview]Stent Edge -Change in Plaque Area (Efficacy Endpoint) at 5 mm Proximal and Distal to Stent.
NCT02098876 (8) [back to overview]In Stent: Plaque Prolapse Post-PCI (In-Stent Mechanistic Endpoint)
NCT02098876 (8) [back to overview]Stent Edge: Percent Area With Low Wall Shear Stress (WSS) at Stent Edges Post-PCI (Mechanistic Endpoint)
NCT02098876 (8) [back to overview]In-Stent: Degree of Vascular Straightening Post-percutaneous Coronary Intervention (PCI) (In-Stent Mechanistic Endpoint)
NCT02098876 (8) [back to overview]In-Stent: Percent Area of Low Wall Shear Stress (WSS)-(In-Stent Mechanistic Endpoint)
NCT02098876 (8) [back to overview]Stent Edge: Degree of Vascular Straightening Post-percutaneous Coronary Intervention (PCI) at the Stent Edges (Stent Edge Mechanistic Endpoint)
NCT02115113 (3) [back to overview]Renal Function Assessed by Estimated Glomerular Filtartion Rate (eGFR)
NCT02115113 (3) [back to overview]Percentage of Participants With Treated Biopsy Proven Acute Rejection (tBPAR) Acute Rejection (AR), Graft Loss (GL) or Death (D)
NCT02115113 (3) [back to overview]Change From Baseline (Randomization) in Serum Creatinine at 12 Months Post-transplant
NCT02120469 (5) [back to overview]Number of Subjects With Disease Progression Using the Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (Phase IB)
NCT02120469 (5) [back to overview]Number of Participants With Grade 3 or Higher Toxicities (Phase IB)
NCT02120469 (5) [back to overview]Number of Participants Experiencing a Dose Limiting Toxicity (DLT) (Phase I) and the Recommended Phase IB Dose (RP2D)
NCT02120469 (5) [back to overview]Median Progression Free Survival (Phase IB)
NCT02120469 (5) [back to overview]Median Overall Survival (Phase IB)
NCT02137239 (28) [back to overview]Absolute Values of Blood Pressure: Mean
NCT02137239 (28) [back to overview]Absolute Calculated Glomerular Filtration Rate (cGFR): Mean
NCT02137239 (28) [back to overview]Time to Clinically-suspected Biopsy-proven Acute Rejection (CSBPAR).
NCT02137239 (28) [back to overview]Percentage of Participants With Serious Adverse Events (SAEs)
NCT02137239 (28) [back to overview]Percentage of Participants With Adverse Events (AEs)
NCT02137239 (28) [back to overview]Percentage of Particpants With Laboratory Test Abnormalities (LTAs)
NCT02137239 (28) [back to overview]Urine Protein Creatinine Ratio (UPr/Cr)
NCT02137239 (28) [back to overview]Treatment Differences in Therapeutic Modalities
NCT02137239 (28) [back to overview]Time to Event: Graft Loss and Death
NCT02137239 (28) [back to overview]Percentage of Participants With New Onset Diabetes After Transplant
NCT02137239 (28) [back to overview]Percentage of Participants With Events of Special Interest (ESIs)
NCT02137239 (28) [back to overview]Percentage of Participants With Donor Specific Anti-HLA Antibodies (DSA)
NCT02137239 (28) [back to overview]Percentage of Participants With De Novo Donor Specific Anti-HLA Antibodies (DSA)
NCT02137239 (28) [back to overview]Percentage of Participants With BANFF Grade by Severity Grades. BANFF Type (Grade) for Acute/Active Rejection
NCT02137239 (28) [back to overview]Percentage of Clinically-suspected Biopsy-proven Acute Rejection (CSBPAR) at 6 Months
NCT02137239 (28) [back to overview]Number of Participants Who Survive With a Functioning Graft
NCT02137239 (28) [back to overview]Number of Participants Who Experience Graft Loss Post Transplant
NCT02137239 (28) [back to overview]Number of Participants Deaths Post Transplant
NCT02137239 (28) [back to overview]Median Calculated Glomerular Filtration Rate (cGFR)
NCT02137239 (28) [back to overview]Mean Changes From Baseline Values of Lipid Values
NCT02137239 (28) [back to overview]Mean Changes From Baseline Values for Blood Pressure
NCT02137239 (28) [back to overview]Mean Change From Month 3 in cGFR
NCT02137239 (28) [back to overview]Mean and Mean Change From Baseline in Whole Blood HbA1c
NCT02137239 (28) [back to overview]Mean and Mean Change From Baseline in Blood Glucose
NCT02137239 (28) [back to overview]Clinically-suspected Biopsy-proven Acute Rejection (CSBPAR) at 6, 12 and 24 Months
NCT02137239 (28) [back to overview]Absolute Values of Fasting Lipid Values: Median
NCT02137239 (28) [back to overview]Absolute Values of Fasting Lipid Values: Mean
NCT02137239 (28) [back to overview]Absolute Values of Blood Pressure: Median
NCT02137837 (8) [back to overview]Progression-free Survival (Fulvestrant Versus Fulvestrant + Everolimus + Anastrozole)
NCT02137837 (8) [back to overview]Progression Free Survival (Fulvestrant + Everolimus vs Fulvestrant + Everolimus + Anastrozole)
NCT02137837 (8) [back to overview]Overall Survival
NCT02137837 (8) [back to overview]Molecular Determinants of Response in Circulating Tumor Cells: CTC-ETI
NCT02137837 (8) [back to overview]Clinical Benefit Rate
NCT02137837 (8) [back to overview]Number of Patients With Gr 3 Through 5 Adverse Events That Are Related to Study Drugs
NCT02137837 (8) [back to overview]Response Rate
NCT02137837 (8) [back to overview]Progression-Free Survival (Fulvestrant vs Fulvestrant + Everolimus )
NCT02143726 (4) [back to overview]Progression Free Survival
NCT02143726 (4) [back to overview]Overall Survival
NCT02143726 (4) [back to overview]Confirmed Response Rate
NCT02143726 (4) [back to overview]Number of Participants With Grade 3 or Higher Adverse Events
NCT02188719 (5) [back to overview]Percent of Participants With Grade 3 or Higher Infectious Adverse Event(s)
NCT02188719 (5) [back to overview]Percent of Participants With Adverse Events (AEs) Attributable to the Donor Alloantigen Reactive Tregs (darTregs) Infusion
NCT02188719 (5) [back to overview]Percent of Participants With Grade 2 or Higher Hematologic Adverse Events (AEs) of Anemia, Neutropenia, and/or Thrombocytopenia
NCT02188719 (5) [back to overview]Percent of Participants With Biopsy-Proven Acute and/or Chronic Rejection
NCT02188719 (5) [back to overview]Percent of Participants With Grade 3 or Higher Wound Complication(s) Adverse Event(s)
NCT02201212 (5) [back to overview]Progression-free Survival
NCT02201212 (5) [back to overview]Duration of Response
NCT02201212 (5) [back to overview]Toxicity Rate
NCT02201212 (5) [back to overview]Objective Response Rate
NCT02201212 (5) [back to overview]Overall Survival
NCT02228681 (4) [back to overview]Median Survival
NCT02228681 (4) [back to overview]Frequency of Response
NCT02228681 (4) [back to overview]Frequency and Severity of CTCAE (Common Toxicity Criteria for Adverse Events) Version 4
NCT02228681 (4) [back to overview]Median Progression-free Survival
NCT02236572 (2) [back to overview]PEPI Score
NCT02236572 (2) [back to overview]Participant Ability to Tolerate Study Treatment With Minimal Side Effects
NCT02258451 (15) [back to overview]Symptomatic Skeletal Event-free Survival (SSE-FS)
NCT02258451 (15) [back to overview]Number of Participants With Hematological Toxicities: Worst Grade Under Treatment (From First Dosing Till Primary Analysis)
NCT02258451 (15) [back to overview]Number of Participants With Hematological Toxicities: Worst Grade Under Treatment
NCT02258451 (15) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs) (From First Dosing Till Primary Analysis)
NCT02258451 (15) [back to overview]Radiological Progression-free Survival (rPFS)
NCT02258451 (15) [back to overview]Number of Participants With Post-treatment Chemotherapy Related Adverse Events (From First Dosing Till Primary Analysis)
NCT02258451 (15) [back to overview]Number of Participants With Post-treatment Chemotherapy Related Adverse Events
NCT02258451 (15) [back to overview]Number of Participants With New Primary Malignancies
NCT02258451 (15) [back to overview]Number of Participants With New Primary Malignancies During Study Treatment Till Primary Analysis
NCT02258451 (15) [back to overview]Time to Cytotoxic Chemotherapy
NCT02258451 (15) [back to overview]Time to Opiate Use for Cancer Pain
NCT02258451 (15) [back to overview]Overall Survival
NCT02258451 (15) [back to overview]Percentage of Participants With Pain Improvement
NCT02258451 (15) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs)
NCT02258451 (15) [back to overview]Time to Pain Progression
NCT02264665 (13) [back to overview]Mean of Number of Tumor Assessment Visits - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Number of Participants With Adverse Events Leading to Death - Based on Treatment Received At-least Once During the Study
NCT02264665 (13) [back to overview]OS Rate at 2 Years- Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Number of Participants Who Had a Change in Their Treatment - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Overall Survival (OS) Rate at 2 Years - Based on Type of Treatment at Inclusion
NCT02264665 (13) [back to overview]PFS at 2 Years Assessed by Investigator Per RECIST v1.1 - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), Treatment Related AEs, SAEs and SAEs With Common Terminology Criteria For Adverse Events (CTCAE) 3, 4 and 5, v4.0-Based on Treatment Received At-least Once During Study
NCT02264665 (13) [back to overview]Number of Participants With Adverse Events Leading to Discontinuation of Treatment - Based on Treatment Received At-least Once During the Study
NCT02264665 (13) [back to overview]Number of Participants With Different Types of Investigation Used for Tumor Assessment - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Number of Participants With Types of Main Lines of Treatment Received During the Follow-up - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Number of Main Lines of Treatment Received During the Study - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Number of Combined Main Lines of Treatments Received - Based on Targeted Therapy Group and Other Treatments Group at Inclusion
NCT02264665 (13) [back to overview]Progression-Free Survival (PFS) at 2 Years Assessed by Investigator Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1 - Based on Type of Treatment at Inclusion
NCT02283658 (6) [back to overview]Progression Free Survival (PFS)
NCT02283658 (6) [back to overview]Percentage of Participants With CA-125 Response
NCT02283658 (6) [back to overview]Percentage of Patients Alive and Progression Free Survival at 12 Weeks
NCT02283658 (6) [back to overview]Confirmed Response Rate, Estimated Using RECIST 1.1 Criteria
NCT02283658 (6) [back to overview]Overall Suravival(OS)
NCT02283658 (6) [back to overview]Number of Participants Experiencing Adverse Events
NCT02291913 (6) [back to overview]Median Time From First Occurrence of CR or PR to Disease Progression or Death Also Called Duration of Response (DOR)
NCT02291913 (6) [back to overview]Median Progression Free Survival (PFS)
NCT02291913 (6) [back to overview]Number of Patients With Adverse Events (AEs) as a Measure of Safety and Tolerability
NCT02291913 (6) [back to overview]Number of Patients With an Objective Response (CR or PR) Also Called the Overall Response Rate (ORR).
NCT02291913 (6) [back to overview]Number of Participants With CR, PR, or 6 Months of SD Also Called Clinical Benefit Rate (CBR)
NCT02291913 (6) [back to overview]Median Overall Survival (OS)
NCT02315625 (5) [back to overview]Overall Survival
NCT02315625 (5) [back to overview]Number of Participants With Serious and Non-Serious Adverse Events
NCT02315625 (5) [back to overview]Number of Participants With an Overall Response
NCT02315625 (5) [back to overview]Median Survival Time (MST)
NCT02315625 (5) [back to overview]Median Amount of Time Subject Survives Without Disease Progression After Treatment
NCT02332902 (2) [back to overview]3D Photographic Measurement of Surface Volume of Cutaneous Neurofibroma Lesion
NCT02332902 (2) [back to overview]Number of Participants With Grade 3-4 Adverse Events
NCT02352844 (3) [back to overview]Mutations Associated With Therapeutic Response
NCT02352844 (3) [back to overview]Response Rate (RR)
NCT02352844 (3) [back to overview]Genetic Changes Associated With Disease Progression
NCT02389946 (10) [back to overview]Number of Lesions With Lesion Success
NCT02389946 (10) [back to overview]Number of Participants With Procedure Success
NCT02389946 (10) [back to overview]Number of Participants With TLF and Individual TLF Components
NCT02389946 (10) [back to overview]Percentage of Participants With Target Lesion Failure (TLF) at 12 Months Post-Index Procedure by Bayesian Estimation
NCT02389946 (10) [back to overview]Number of Participants With MACE and Individual MACE Components
NCT02389946 (10) [back to overview]Number of Participants With Myocardial Infarction
NCT02389946 (10) [back to overview]Number of Participants With Myocardial Infarction or Cardiac Death
NCT02389946 (10) [back to overview]Number of Participants With Stent Thrombosis
NCT02389946 (10) [back to overview]Number of Participants With Target Vessel Failure (TVF) and Individual TVF Components
NCT02389946 (10) [back to overview]Number of Lesions With Device Success
NCT02429869 (3) [back to overview]Plasma HIV RNA
NCT02429869 (3) [back to overview]Cell-associated Total HIV RNA
NCT02429869 (3) [back to overview]Cell-associated HIV DNA
NCT02451696 (5) [back to overview]Blood Total VEGF Levels (Not Only VEGF-D)
NCT02451696 (5) [back to overview]mTOR Brain Tissue-S6 Phosphate by Western Blot
NCT02451696 (5) [back to overview]Number of Patients With Adverse Events
NCT02451696 (5) [back to overview]HMGB1 Expression in Brain Tissue
NCT02451696 (5) [back to overview]Blood Everolimus Levels
NCT02454478 (11) [back to overview]Objective Response Rate (ORR)
NCT02454478 (11) [back to overview]Tss,Max: Time to Reach the Maximum Plasma Concentration (Cmax) at Steady State for Levatinib and Everolimus
NCT02454478 (11) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT02454478 (11) [back to overview]Number of Participants With the Minimum Percent Change From Baseline in the Sum of Diameters of Target Lesions
NCT02454478 (11) [back to overview]Disease Control Rate (DCR)
NCT02454478 (11) [back to overview]Number of Participants With Best Overall Response (BOR)
NCT02454478 (11) [back to overview]Css,Max: Maximum Observed Plasma Concentration at Steady State for Levatinib and Everolimus
NCT02454478 (11) [back to overview]Cmax: Maximum Observed Plasma Concentration for Levatinib and Everolimus
NCT02454478 (11) [back to overview]Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Levatinib and Everolimus
NCT02454478 (11) [back to overview]Number of Participants Who Experienced Any Dose Limiting Toxicity (DLT)
NCT02454478 (11) [back to overview]AUC 0-t: Area Under the Concentration-time Curve From Zero (Pre-dose) to Time of Last Quantifiable Concentration for Levatinib and Everolimus
NCT02504892 (4) [back to overview]Count of Participants With Non-serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0)
NCT02504892 (4) [back to overview]Progression-free Survival (PFS)
NCT02504892 (4) [back to overview]Overall Response Rate With Everolimus Treatment.
NCT02504892 (4) [back to overview]Overall Survival (OS)
NCT02513719 (97) [back to overview]Number of Participants With All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Myocardial Infarction
NCT02513719 (97) [back to overview]Number of Participants With Myocardial Infarction
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or Target Vessel MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or Target Vessel MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or Target Vessel MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or Target Vessel MI
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Failure
NCT02513719 (97) [back to overview]Number of Participants With Myocardial Infarction
NCT02513719 (97) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE)
NCT02513719 (97) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE)
NCT02513719 (97) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE)
NCT02513719 (97) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE)
NCT02513719 (97) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE)
NCT02513719 (97) [back to overview]Number of Participants With Major Adverse Cardiac Events (MACE)
NCT02513719 (97) [back to overview]Number of Participants With Hemorrhage
NCT02513719 (97) [back to overview]Number of Participants With Hemorrhage
NCT02513719 (97) [back to overview]Number of Participants With Hemorrhage
NCT02513719 (97) [back to overview]Number of Participants With Hemorrhage
NCT02513719 (97) [back to overview]Number of Participants With Hemorrhage
NCT02513719 (97) [back to overview]Number of Participants With Hemorrhage
NCT02513719 (97) [back to overview]Number of Participants With Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Death or MI
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or Target-Vessel MI
NCT02513719 (97) [back to overview]Net Gain: In-stent, In-segment
NCT02513719 (97) [back to overview]Late Loss(LL): In-stent,In-segment,Proximal, and Distal
NCT02513719 (97) [back to overview]Acute Gain: In-stent,In-segment
NCT02513719 (97) [back to overview]Success Rate: XIENCE PRIME Implant Success by Patient
NCT02513719 (97) [back to overview]Success Rate: Percentage of Lesions With Procedural Success
NCT02513719 (97) [back to overview]Success Rate: Percentage of Devices With Implant Success
NCT02513719 (97) [back to overview]Percent Diameter Stenosis (%DS)
NCT02513719 (97) [back to overview]Percent Diameter Stenosis (%DS)
NCT02513719 (97) [back to overview]Percent Diameter Stenosis (%DS)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Vessel Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Failure
NCT02513719 (97) [back to overview]Number of Participants With Cardiac Death or Target Vessel-MI
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Failure
NCT02513719 (97) [back to overview]Number of Participants With Target Lesion Failure
NCT02513719 (97) [back to overview]Number of Participants With Stent Thrombosis: Very Late
NCT02513719 (97) [back to overview]Number of Death
NCT02513719 (97) [back to overview]Number of Death
NCT02513719 (97) [back to overview]Number of Death
NCT02513719 (97) [back to overview]Number of Death
NCT02513719 (97) [back to overview]Number of Death
NCT02513719 (97) [back to overview]Number of Death
NCT02513719 (97) [back to overview]Number of Participants With All Death/All MI/All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Death/All MI/All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Death/All MI/All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Death/All MI/All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With All Death/All MI/All Revascularization
NCT02513719 (97) [back to overview]Number of Participants With Stent Thrombosis: Subacute
NCT02513719 (97) [back to overview]Number of Participants With Stent Thrombosis: Late
NCT02513719 (97) [back to overview]Number of Participants With Stent Thrombosis: Acute
NCT02513719 (97) [back to overview]Number of Participants With Non-target Vessel Revascularization (Non-TVR)
NCT02513719 (97) [back to overview]Number of Participants With Non-target Vessel Revascularization (Non-TVR)
NCT02513719 (97) [back to overview]Number of Participants With Non-target Vessel Revascularization (Non-TVR)
NCT02513719 (97) [back to overview]Number of Participants With Non-target Vessel Revascularization (Non-TVR)
NCT02513719 (97) [back to overview]Number of Participants With Non-target Vessel Revascularization (Non-TVR)
NCT02513719 (97) [back to overview]Number of Participants With Non-target Vessel Revascularization (Non-TVR)
NCT02513719 (97) [back to overview]Number of Participants With Myocardial Infarction
NCT02513719 (97) [back to overview]Number of Participants With Myocardial Infarction
NCT02513719 (97) [back to overview]Number of Participants With Myocardial Infarction
NCT02513719 (97) [back to overview]Number of Participants With All Death/All MI/All Revascularization
NCT02520063 (1) [back to overview]Number of Participants Who Experienced Dose-limiting Toxicities
NCT02539459 (2) [back to overview]Number of Patients With Tumor Volume Reduction Greater Than 25%
NCT02539459 (2) [back to overview]Safety and Tolerability of Everolimus in Patients With Sporadic AML
NCT02560012 (1) [back to overview]Number of Participants Who Progressed
NCT02599324 (19) [back to overview]Phase 1b: Terminal Elimination Rate Constant (λz) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Area Under the Concentration-Time Curve to Last Observed Time Point (AUClast) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Apparent Total Clearance at Steady-State (CLss/F) for Ibrutinib in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Terminal Elimination Rate Constant (λz) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Terminal Elimination Half-Life (t1/2term) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Observed Maximum Concentration (Cmax) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Disease Control Rate (DCR) in Cohorts 1 to 6
NCT02599324 (19) [back to overview]Phase 1b: Number of Participants With Dose-Limiting Toxicities (DLTs) in Cohorts 1 to 6
NCT02599324 (19) [back to overview]Phase 1b: ORR in Cohorts 1 to 6
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: DCR in Cohorts 1 to 6
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: Duration of Response (DOR) in Cohorts 1 to 6
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: ORR in Cohorts 1 and 2
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: Overall Response Rate (ORR) as Assessed by Investigator in Cohorts 3 to 6
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: Overall Survival (OS) in Cohorts 1 to 6
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: PFS in Cohorts 3 to 6
NCT02599324 (19) [back to overview]Phase 1b/2 RP2D: Progression-Free Survival (PFS) as Assessed by Investigator in Cohorts 1 and 2
NCT02599324 (19) [back to overview]Phase 1b: Area Under the Concentration-Time Curve From Time 0 to Hour 24 (AUC0-24h) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Time of Last Observed Concentration (Tlast) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02599324 (19) [back to overview]Phase 1b: Time to Cmax (Tmax) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4
NCT02724020 (7) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs)
NCT02724020 (7) [back to overview]Objective Response Rate (ORR)
NCT02724020 (7) [back to overview]Overall Survival (OS)
NCT02724020 (7) [back to overview]Progression-Free Survival (PFS)
NCT02724020 (7) [back to overview]Time-to-progression (TTP)
NCT02724020 (7) [back to overview]Clinical Benefit Rate (CBR)
NCT02724020 (7) [back to overview]CBR With SD Duration of at Least 16 Weeks
NCT02732119 (12) [back to overview]Duration of Overall Response (DOR) by Group - Phase II
NCT02732119 (12) [back to overview]Participants With Dose Limiting Toxicities by Preferred Term in Cycle 1 (28 Days) - in Phase I
NCT02732119 (12) [back to overview]Everolimus Pharmacokinetic Plasma Concentrations by Cohort - Phase I
NCT02732119 (12) [back to overview]Everolimus Pharmacokinetic Plasma Concentrations - Phase II
NCT02732119 (12) [back to overview]Clinical Benefit Rate as Per Central Review by Group- Phase II
NCT02732119 (12) [back to overview]Clinical Benefit Rate as Per Central Review by Dose Cohort - Phase I
NCT02732119 (12) [back to overview]Clinical Benefit Rate as Per Central Review by Dose Cohort - Phase I
NCT02732119 (12) [back to overview]Best Overall Responses (BOR) Summary Table as Per Central Review by Group - Phase II
NCT02732119 (12) [back to overview]Time to Definitive Deterioration of ECOG Performance Status in One Category of the Score by Group - Phase II
NCT02732119 (12) [back to overview]Summary of Progression-free Survival (PFS) (Months), as Per Central Review by Group - Phase II
NCT02732119 (12) [back to overview]Overall Survival (OS) by Group - Phase II
NCT02732119 (12) [back to overview]Best Overall Responses (BOR) Summary Table as Per Central Review by Cohort - Phase I
NCT02736227 (1) [back to overview]Amount of Treg Cells Observed in Peripheral Blood in Patients With and Without Rejection
NCT02811861 (1) [back to overview]Progression-free Survival (PFS) by Independent Imaging Review (IIR)
NCT02864706 (8) [back to overview]Measured Glomerular Filtration Rate (mGFR)
NCT02864706 (8) [back to overview]Progression of Cardiac Allograft Vasculopathy (CAV) Recorded by Intravascular Ultrasound (IVUS)
NCT02864706 (8) [back to overview]Change From Baseline in Visual Analog Scale (VAS)
NCT02864706 (8) [back to overview]Change From Baseline in the Euro Quality of Life 5D
NCT02864706 (8) [back to overview]Number of Participants With Beck Depression Inventory (BDI)
NCT02864706 (8) [back to overview]Quality of Life by SF-36 Change From Pre-transplantation to 5-7 Year Follow-up
NCT02864706 (8) [back to overview]Percent of Participants With Incidence of Coronary Allograft Vasculopathy (CAV)
NCT02864706 (8) [back to overview]Myocardial Structure and Function
NCT02871791 (5) [back to overview]Duration of Response (DOR) [Phase 2a]
NCT02871791 (5) [back to overview]Median Progression Free Survival (PFS) [Phase 2a]
NCT02871791 (5) [back to overview]Overall Response Rate (ORR) [Phase 2a]
NCT02871791 (5) [back to overview]Clinical Benefit Rate (CBR) [Phase 2a]
NCT02871791 (5) [back to overview]Disease Control Rate (DCR) [Phase 2a]
NCT02915783 (3) [back to overview]Overall Survival (OS)
NCT02915783 (3) [back to overview]Objective Response Rate (ORR)
NCT02915783 (3) [back to overview]Progression-free Survival (PFS)
NCT02954198 (4) [back to overview]Percent of Participants Experiencing Acute Allograft Rejection
NCT02954198 (4) [back to overview]Subject Specific Change on Medication Side Effect Scale
NCT02954198 (4) [back to overview]Self-reported Medication Adherence From Baseline to 6 Months.
NCT02954198 (4) [back to overview]Percent of Participants Who Experienced Kidney Transplant Graft Loss
NCT03112603 (24) [back to overview]Percentage of Participants With a ≥ 50% Reduction in Daily Corticosteroid Dose
NCT03112603 (24) [back to overview]t1/2 of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Tmax of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Vz/F of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Rate of Failure-free Survival (FFS)
NCT03112603 (24) [back to overview]CL/F of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Cmax of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Change From Baseline in Functional Assessment of Cancer Therapy - Bone Marrow Transplantation (FACT-BMT)
NCT03112603 (24) [back to overview]Change From Baseline in EQ-5D-5L
NCT03112603 (24) [back to overview]AUClast of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Best Overall Response (BOR) at Cycle 7 Day 1
NCT03112603 (24) [back to overview]BOR During Cross-over Treatment With Ruxolitinib
NCT03112603 (24) [back to overview]Duration of Response Through Study Completion
NCT03112603 (24) [back to overview]Efficacy of Ruxolitinib Versus Investigator's Choice Best Available Therapy (BAT) in Participants With Moderate or Severe Steroid Refractory Chronic Graft Versus Host Disease (SR-cGvHD) Assessed by Overall Response Rate (ORR) at the Cycle 7 Day 1 Visit
NCT03112603 (24) [back to overview]Number of Participants With Any Treatment-emergent Adverse Event (TEAE)
NCT03112603 (24) [back to overview]AUCinf of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses
NCT03112603 (24) [back to overview]Utilization of Medical Resources
NCT03112603 (24) [back to overview]Rate of Participants With Clinically Relevant Improvement of the Modified Lee cGvHD Symptom Scale Score
NCT03112603 (24) [back to overview]ORR at the End of Cycle 3
NCT03112603 (24) [back to overview]Rate of FFS at Study Completion
NCT03112603 (24) [back to overview]Overall Survival (OS)
NCT03112603 (24) [back to overview]Cumulative Incidence of Malignancy Relapse/Recurrence (MR)
NCT03112603 (24) [back to overview]Cumulative Incidence of Non-relapse Mortality (NRM)
NCT03112603 (24) [back to overview]Percentage of Participants Successfully Tapered Off of All Corticosteroids
NCT03163667 (2) [back to overview]Overall Survival (OS)
NCT03163667 (2) [back to overview]Progression Free Survival (PFS)
NCT03173560 (15) [back to overview]Health-Related Quality of Life (HRQoL) Assessed by Functional Assessment of Cancer Therapy Kidney Syndrome Index-Disease-Related Symptoms (FKSI-DRS) Scores
NCT03173560 (15) [back to overview]Health-Related Quality of Life (HRQoL) Assessed by Functional Assessment of Cancer Therapy Kidney Syndrome Index-Disease-Related Symptoms (FKSI-DRS) Scores
NCT03173560 (15) [back to overview]HRQoL Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30 Scores
NCT03173560 (15) [back to overview]HRQoL Assessed by European Quality of Life (EuroQol) Five-Dimensional, 3-Level (EQ-5D-3L) Index Score and Visual Analogue Scale (VAS)
NCT03173560 (15) [back to overview]Overall Survival (OS)
NCT03173560 (15) [back to overview]Number of Participants With TEAEs and Serious TEAEs
NCT03173560 (15) [back to overview]HRQoL Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30 Scores
NCT03173560 (15) [back to overview]Objective Response Rate (ORR)
NCT03173560 (15) [back to overview]HRQoL Assessed by European Quality of Life (EuroQol) Five-Dimensional, 3-Level (EQ-5D-3L) Index Score and Visual Analogue Scale (VAS)
NCT03173560 (15) [back to overview]Objective Response Rate at Week 24 (ORR24W)
NCT03173560 (15) [back to overview]Percentage of Participants With Intolerable Grade 2 or Any Grade >=Grade 3 TEAEs Within 24 Weeks
NCT03173560 (15) [back to overview]Percentage of Participants Who Discontinued Treatment Due to Toxicity
NCT03173560 (15) [back to overview]Progression-free Survival (PFS)
NCT03173560 (15) [back to overview]Progression-free Survival After Next Line of Therapy (PFS2)
NCT03173560 (15) [back to overview]Time to Treatment Failure Due to Toxicity
NCT03176238 (5) [back to overview]Percentage of Participants Response Rates (Best Overall and Overall)
NCT03176238 (5) [back to overview]Summary of Number of Participants With Treatment Emergent Adverse Events (TEAE) - All Grades
NCT03176238 (5) [back to overview]Percent of Participants Event-free Probability Estimates of Deterioration of Eastern Cooperative Oncology Group (ECOG) Performance Status
NCT03176238 (5) [back to overview]Percentage of Participants Clinical Benefit Rate
NCT03176238 (5) [back to overview]Progression Free Survival (PFS)
NCT03245151 (18) [back to overview]Phase 2: Objective Response Rate (ORR)
NCT03245151 (18) [back to overview]Phase 2: Number of Participants With Any Treatment-emergent Serious Adverse Event (TESAE)
NCT03245151 (18) [back to overview]Phase 2: Number of Participants With Any Treatment-emergent Adverse Event (TEAE)
NCT03245151 (18) [back to overview]Phase 1: Disease Control Rate (DCR)
NCT03245151 (18) [back to overview]Phase 2: Duration of Response (DOR)
NCT03245151 (18) [back to overview]Phase 2: Disease Control Rate (DCR)
NCT03245151 (18) [back to overview]Phase 2: Clinical Benefit Rate (CBR)
NCT03245151 (18) [back to overview]Phase 1: Recommended Phase 2 Dose (RP2D) of Lenvatinib in Combination With Everolimus
NCT03245151 (18) [back to overview]Phase 1: Objective Response Rate (ORR)
NCT03245151 (18) [back to overview]Phase 1: Number of Participants With Any Treatment-emergent Serious Adverse Event (TESAE)
NCT03245151 (18) [back to overview]Phase 1: Number of Participants With Any Treatment-emergent Adverse Event (TEAE)
NCT03245151 (18) [back to overview]Phase 1: Maximum Tolerated Dose (MTD) of Lenvatinib in Combination With Everolimus
NCT03245151 (18) [back to overview]Phase 1: Clinical Benefit Rate (CBR)
NCT03245151 (18) [back to overview]Phase 1: Trough Concentrations (Ctrough) of Everolimus When Administered in Combination With Lenvatinib
NCT03245151 (18) [back to overview]Phase 1: Time to Reach Maximum Plasma Concentration (Cmax) of Lenvatinib (Tmax)
NCT03245151 (18) [back to overview]Phase 1: Maximum Plasma Concentration of Lenvatinib (Cmax)
NCT03245151 (18) [back to overview]Phase 1: Area Under the Plasma Concentration-time Curve From Time Zero to Time of Last Quantifiable Concentration of Lenvatinib (AUC[0-t Hours])
NCT03245151 (18) [back to overview]Phase 2: Objective Response Rate (ORR) at Week 16
NCT03352427 (5) [back to overview]Progression-free Survival in Participants With Newly Diagnosed High-grade Glioma (HGG)
NCT03352427 (5) [back to overview]Progression-free Survival in Participants With Newly Diagnosed Diffuse Intrinsic Pontine Glioma (DIPG)
NCT03352427 (5) [back to overview]Overall Survival
NCT03352427 (5) [back to overview]Overall Response Rate (OR) (Partial Response or Better) in Participants With Refractory or Recurrent Glioma
NCT03352427 (5) [back to overview]Overall Survival
NCT03387020 (8) [back to overview]Ribociclib Half-Life as Obtained From the Phase 1 Study
NCT03387020 (8) [back to overview]Maximum Tolerated Dose of Ribociclib and Everolimus
NCT03387020 (8) [back to overview]Average Tumor and Plasma Concentrations of Ribociclib for the Surgical Study
NCT03387020 (8) [back to overview]Everolimus Half Life as Obtained From the Phase 1 Study
NCT03387020 (8) [back to overview]Objective Responses (Complete Response + Partial Response)
NCT03387020 (8) [back to overview]Percent Change in ki67 Between Archival and Post-treatment Tissue
NCT03387020 (8) [back to overview]Everolimus AUC (Area Under the Plasma Concentration Versus Time Curve) as Obtained From the Phase I Study
NCT03387020 (8) [back to overview]Ribociclib AUC (Area Under the Plasma Concentration Versus Time Curve) as Obtained From the Patients Who Received Phase I Defined Treatment
NCT03468478 (1) [back to overview]Incidence of Cytomegalovirus Infection or Disease
NCT03525834 (5) [back to overview]Percentage of Participants With Best Overall Response (BOR) Status of Angiomyolipoma (AML) Response During Maximum Treatment Duration of 48 Weeks
NCT03525834 (5) [back to overview]Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT03525834 (5) [back to overview]Percentage of Participants With Best Overall Response Status of Angiomyolipoma (AML) Progression During Maximum Treatment Duration of 48 Weeks
NCT03525834 (5) [back to overview]Percentage of Participants With Severe Renal Impairment
NCT03525834 (5) [back to overview]Percentage of Participants With NCI CTCA Grade 3/4 Serum Creatinine
NCT03659136 (6) [back to overview]Duration of Disease Control (DC)
NCT03659136 (6) [back to overview]Overall Survival (OS)
NCT03659136 (6) [back to overview]Progression Free Survival (PFS)
NCT03659136 (6) [back to overview]Time to Pain Progression or Intensification of Pain Palliation
NCT03659136 (6) [back to overview]Number of Participants With Objective Response (OR)
NCT03659136 (6) [back to overview]Number of Patients With Disease Control (DC)
NCT03839940 (7) [back to overview]Severity of Mouth Pain Scores by Ethnicity
NCT03839940 (7) [back to overview]Severity of Mouth Pain Scores by Sex
NCT03839940 (7) [back to overview]Incidence Rate of the mTOR Inhibitor-associated Stomatitis (mIAS)-Related Pain Based on Daily Self-reports Via the Numerical Analogue Scale by Ethnicity.
NCT03839940 (7) [back to overview]Incidence Rate of the mTOR Inhibitor-associated Stomatitis (mIAS)-Related Pain Based on Daily Self-reports Via the Numerical Analogue Scale by Race
NCT03839940 (7) [back to overview]Incidence Rate of the mTOR Inhibitor-associated Stomatitis (mIAS)-Related Pain Based on Daily Self-reports Via the Numerical Analogue Scale by Sex
NCT03839940 (7) [back to overview]Severity of Mouth Pain Scores
NCT03839940 (7) [back to overview]Severity of Mouth Pain Scores by Race
NCT04177095 (6) [back to overview]Incidence of Acute Rejection
NCT04177095 (6) [back to overview]Incidence of de Novo Donor Specific Antibodies
NCT04177095 (6) [back to overview]Rate of New-onset Proteinuria
NCT04177095 (6) [back to overview]Survival
NCT04177095 (6) [back to overview]Incidence of Acute Rejection
NCT04177095 (6) [back to overview]Increase in eGFR

Overall Objective Response

Response will be evaluated in this study using the new international criteria Response Evaluation Criteria in Solid Tumors (RECIST) (NCT00085566)
Timeframe: 2 years

,,
Interventionparticipants (Number)
Partial Response (PR)Stable Disease (SD)Progression of Disease (POD)
RAD 001 30 mg003
RAD 001 50 mg012
RAD 001 70 mg21526

[back to top]

Clinical Benefit Rate

Clinical benefit rate (CBR) is defined as the objective response rate plus the proportion of participants with prolonged stable disease (SD), e.g. nonprogression at 20 weeks. Objective response rate (ORR), determined by tumor assessments from radiological tests or physical examination using Response Evaluation Criteria In Solid Tumors (RECIST). (NCT00087685)
Timeframe: 20 weeks

Interventionpercentage of participants (Number)
RAD00121

[back to top]

Number of Participants With Objective Response Plus Stable Disease Rate (CR + PR + SD)

Response determined by tumor assessments from radiological tests or physical examination using Response Evaluation Criteria In Solid Tumors (RECIST). Complete Response (CR): Disappearance of all target and non-target lesions and no evidence of new lesions documented by two disease assessments at least 4 weeks apart. Partial Response (PR): At least 30% decrease in sum of the longest dimensions (LD) of all target measurable lesions taking as reference the baseline sum of LD. Stable Disease (SD): Any condition not meeting the above criteria. The minimum duration for the SD will be 8 weeks. If the participant has stable disease at the time of the first radiographic evaluation, he/she will be considered to have stable disease. Progressive Disease (PD): At least a 20% increase in the sum of LD of target lesions taking as reference the smallest sum LD or the appearance of new lesions within 8 weeks of study entry. (NCT00087685)
Timeframe: 8 weeks

Interventionparticipants (Number)
Complete Response (CR)Partial Response (PD)Stable Disease (SD)Progressive Disease (PD)
RAD001001417

[back to top]

Overall Objective Response

Determine efficacy of the combination oral daily gefitinib and oral daily RAD001 in patients with advanced NSCLC. Response and progression will be evaluated in this study using the international criteria proposed by the Response Evaluation Criteria in Solid Tumors (RECIST). (NCT00096486)
Timeframe: 2 years

,,,
Interventionparticipants (Number)
Partial Response (PR)Stable Disease (SD)Progression of Disease (POD)Not Evaluable/ Evaluable for Toxicity Only
Phase I: RAD001 10 mg, Gefitinib 250 mg1111
Phase I: RAD001 5 mg, Gefitinib 250 mg1140
Phase II: Cohort I no Prior Conventional Chemotherapy39111
Phase II: Cohort II One or More Prior Chemotherapy215181

[back to top]

Progression Free Survival (PFS)

PFS is measured from date of trial entry until documented progression of disease or death from any cause. PFS is measured by computed tomography (CT) scans or magnetic resonance imaging (MRI) performed at baseline and after every three cycles. (NCT00113360)
Timeframe: PFS assessed every 12 weeks (at the end of every 3 cycles) or more frequently if clinically indicated

InterventionWeeks (Median)
RAD001 Plus Octreotide Depot60

[back to top]

Calculated Creatinine Clearance at 6 Month and 12 Month

"Creatinine clearance calculated by Cockcroft-Gault formula and summarized by mean, and standard deviation. Cockcroft-Gault formula to calculate Creatinine Clearance (CrCl[mL/min]) is shown below:~CrCl[mL/min] = (140 - A) * W / (72 * C) * R. Where A is age at sample date [years], W is body weight at specific visit [kg], C is the serum concentration of creatinine [mg/dL], R = 1 if the patient is male and = 0.85 if female." (NCT00154284)
Timeframe: 6 month and 12 months

,
InterventionmL/min (Mean)
6 Month12 Month
Everolimus (Certican) With Cyclosporine (Neoral) Continuation63.665.6
Everolimus (Certican) With Cyclosporine (Neoral) Withdrawal72.972.3

[back to top]

Number of Participants With Biopsy-proven Acute Rejection (BPAR) Episodes, Graft Loss, Death or Loss to Follow-up

Renal biopsies were collected for all cases of suspected acute rejection. For these cases, regardless of initiation of anti-rejection treatment, a graft core biopsy had been performed within 48 hours. These biopsies were listed on the Kidney Allograft Biopsy eCRF and the results used for patient management for BPAR. Graft loss was defined as the allograft was presumed to be lost on the day the patient started dialysis and was not able to subsequently be removed from dialysis as well as re-transplant. BPAR, graft loss, death, or loss to follow-up was analyzed by means of frequency tables. (NCT00154284)
Timeframe: Month 12

,
InterventionParticipants (Number)
Biopsy-proven Acute Rejection (BPAR)Graft LossDeathLoss to Follow-up
Everolimus (Certican) With Cyclosporine (Neoral) Continuation10000
Everolimus (Certican) With Cyclosporine (Neoral) Withdrawal10000

[back to top]

Renal Function Measured by Calculated Glomerular Filtration Rate (GFR Calculated According to the Nankivell Formula)

"Nankivell's formula for calculated GFR is shown below:~GFR [mL/min] = 6.7/C + W/4 - UREA/2 - 100/H2+ 35 (25 for females). Where W is body weight at specific visit [kg], H is height at specific visit [m], C is the serum concentration of creatinine [mmol/L], and UREA is the serum concentration of urea [mmol/L]. UREA was calculated from blood urea nitrogen (BUN) lab data by: UREA = 2.1441*BUN. If a GFR value from Nankivell formula was less than 10 [mL/min], then the value was assigned as 10 [mL/min]." (NCT00154284)
Timeframe: At Month 3 and Month 12

,
InterventionmL/min per 1.73 m^2 (Mean)
GFR at 3 months (Pre-randomization)GFR at 12 months
Everolimus (Certican) With Cyclosporine (Neoral) Continuation68.563.6
Everolimus (Certican) With Cyclosporine (Neoral) Withdrawal69.268.3

[back to top]

Serum Creatinine at Month 6 and 12

serum creatinine summarized by mean and standard deviation (NCT00154284)
Timeframe: 6 month and 12 months

,
Interventionµmol/L (Mean)
6 Month12 Month
Everolimus (Certican) With Cyclosporine (Neoral) Continuation139.1135.6
Everolimus (Certican) With Cyclosporine (Neoral) Withdrawal120.1123.0

[back to top]

Number of Participants With Any Wound Healing Disorder During the 12-month Treatment Period

A wound was considered healed if all the suture material and staples were removed and the wound was intact by 3 weeks. Any wound opened beyond this point, infected, drained fluid or herniated was considered not healed. (NCT00154297)
Timeframe: Month 12

,
InterventionParticipants (Number)
Any wound healing disorder -TotalWound healing disorder related to transplantWound healing disorder unrelated to transplant
Delayed Everolimus32286
Immediate Everolimus28262

[back to top]

Number of Participants Considered in Failure for the Primary Failure Endpoint at 6 Months Post-transplantation.

"The primary efficacy variable was the primary failure endpoint at 6 months defined as the occurrence of one or more of the following events within the first 6 months:~delayed graft function (DGF), defined as the need for dialysis within the first 7 days post-transplantation excluding the first day post-transplantation~efficacy failure (biopsy proven acute rejection (BPAR), graft loss, death or loss to follow-up)~wound healing disorder related to initial transplant surgery" (NCT00154297)
Timeframe: at 6 Month post-transplantation

,
InterventionParticipants (Number)
Failure at month 6 - TotalDialysis within first 7 daysEfficacy failure (Total)--BPAR--Graft Loss--Death--Lost to follow-up (composite efficacy endpoint)Wound healing disorderLoss to follow-up for the primary failure endpoint
Delayed Everolimus48182213424283
Immediate Everolimus39161710540250

[back to top]

Number of Participants Considered in Failure for the Primary Failure Endpoint at 3 Months

"In Failure, is at least one of these events occurred within the first 3 months: delayed graft function(DGF), (need for dialysis within the first 7 days,minus day one,post-transplantation); Biopsy proven acute rejection (BPAR), Graft loss, (allograft was presumed lost on the day the patient started and not removable from dialysis). Death; Loss to follow-up; Wound healing disorder(Any wound related to the kidney transplantation being opened beyond 3 weeks, or infected, or drained fluid or herniated was considered not healed)." (NCT00154297)
Timeframe: Month 3

,
InterventionParticipants (Number)
Failure at month 3 - TotalDialysis within first 7 daysEfficacy failure (Total)--BPAR--Graft Loss--Death--Lost to follow-up (composite efficacy endpoint)Wound healing disorderLoss to follow-up for the primary failure endpoint
Delayed Everolimus4718157324283
Immediate Everolimus3616147540240

[back to top]

Number of Participants Considered in Failure for the Primary Failure Endpoint at 12 Months Post-transplantation.

"The primary efficacy variable was the primary failure endpoint at 12 months defined as the occurrence of one or more of the following events within the first 12 months:~delayed graft function (DGF), defined as the need for dialysis within the first 7 days post-transplantation excluding the first day post-transplantation~efficacy failure (biopsy proven acute rejection (BPAR), graft loss, death or loss to follow-up)~wound healing disorder related to initial transplant surgery" (NCT00154297)
Timeframe: at 12 Month post-transplantation

,
InterventionParticipants (Number)
Failure at month 12 - TotalDialysis within first 7 daysEfficacy failure (Total)--BPAR--Graft Loss--Death--Lost to follow-up (composite efficacy endpoint)Wound healing disorderLoss to follow-up for the primary failure endpoint
Delayed Everolimus49182515525283
Immediate Everolimus42162213650260

[back to top]

Number of Participants Who Underwent Any Dialysis Within the 12-month Treatment Period

The number of patients who underwent any dialysis within the 12-month treatment period. (NCT00154297)
Timeframe: Month 12

InterventionParticipants (Number)
Immediate Everolimus16
Delayed Everolimus24

[back to top]

Duration of Dialysis

The mean duration in days of any dialysis session that occurred within the 12 month treatment period. (NCT00154297)
Timeframe: 12 months

InterventionDays (Mean)
Immediate Everolimus11.9
Delayed Everolimus7.8

[back to top]

Number of Participants With Occurrence of Treatment Failures

Treatment failures defined as a composite endpoint of biopsy proven acute rejection, graft loss, death, loss to follow up and discontinuations due to lack of efficacy or toxicity, or conversion to another regimen (at least one condition must be present). (NCT00154310)
Timeframe: up to or at Month 12

,
InterventionParticipants (Number)
Treatment failure: YesTreatment failure: No
Cyclosporine + Mycophenolate Sodium23123
Everolimus + Mycophenolate Sodium29125

[back to top]

Number of Participants With Occurrence of Biopsy Proven Acute Rejection (BPAR), Graft Loss or Death

The number of participants with occurrence of biopsy proven acute rejection (BPAR), graft loss, or death up to Month 12 during the randomized treatment period. BPAR was defined as a biopsy graded IA, IB, IIA, IIB or III according to Banff 97 classification. A graft core biopsy was performed prior to 24 hours following initiation of graft rejection therapy. The allograft is presumed to be lost on the day the patient starts dialysis and was not able to subsequently be removed from dialysis. If the patient underwent a graft nephrectomy, then the day of nephrectomy was the day of graft loss. (NCT00154310)
Timeframe: Up to Month 12

,
InterventionParticipants (Number)
BPAR: YesBPAR: NoGraft Loss: YesGraft Loss: NoDeath: YesDeath: No
Cyclosporine + Mycophenolate Sodium514101461145
Everolimus + Mycophenolate Sodium1513901540154

[back to top]

Number of Participants Who Experienced an Adverse Event or Serious Adverse Event

Additional information about the number of participants who experienced Adverse Events (greater than 5%) or Serious Adverse Events can be found in the Adverse Event section. (NCT00154310)
Timeframe: Aes from end of core study period (month 12) to end of follow-up period (month 60)

,
InterventionParticipants (Number)
Adverse EventsSerious Adverse Events
Cyclosporine + Mycophenolate Sodium14586
Everolimus + Mycophenolate Sodium15595

[back to top]

Changes in Cardiovascular Risk From Month 4.5 to Final Assessment at Month 12

An updated 1991 Framingham coronary prediction algorithm was used to estimate the total risk of developing coronary heart diseases (CHD) over the course of 10 years. Risk was calculated separately for male and females. To calculate risk, points were assigned for each of the following risk factors: age, levels of LDL cholesterol, HDL cholesterol, blood pressure, cigarette smoking, and diabetes mellitus. The sum of the individual risk factor points gives a total point score, which ranges from -5 to 18 for men and -16 to 24 for women. Higher points indicate a higher risk for CHD. (NCT00154310)
Timeframe: Month 4.5 and Month 12

,
InterventionPoints (Mean)
Male (n= 55, 37)Female (n= 22, 35)Total Population (n= 77, 72)
Cyclosporine + Mycophenolate Sodium0.10.80.4
Everolimus + Mycophenolate Sodium0.50.00.4

[back to top]

Renal Function (Nankivell Formula) at Month 12 Post Transplantation.

Renal function at the end of the trial assessed as mean absolute values of the glomerular filtration rate (GFR) calculated by Nankivell formula 12 months after renal transplantation. The Nankivell formula: GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)^2 + C ; where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kg, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. Estimated GFR is expressed in mL/min per 1.73m^2. (NCT00154310)
Timeframe: at Month 12 post transplantation

InterventionmL/min /1.73m^2 (Mean)
Everolimus + Mycophenolate Sodium71.84
Cyclosporine + Mycophenolate Sodium61.24

[back to top]

Number of Participants With Safety Parameters

The selected safety parameters (such as hypertension, hyperlipidemia, diabetes mellitus, anemia, malignancies ) were derived based on adverse events preferred terms defined in the analysis plan. (NCT00170846)
Timeframe: 24 months

,,
InterventionParticipants (Number)
Hypertension, YesHypertension, NoHyperlipidemia, YesHyperlipidemia, NoDiabetes mellitus, YesDiabetes mellitus, NoAnemia, YesAnemia, NoMalignancies, YesMalignancies, No
Group A: No RAD61176117411925987116
Group B : CNI Withdrawal1311418109612145829118
Group C: CNI Reduction9135111337137469811133

[back to top]

Change in mGFR by Baseline Calculated Creatinine Clearance (Cockcroft-Gault Formula)

"Cockcroft-Gault formula (CrCl):~Creatinine Clearance [mL/min] = CrCl (males) = (140 - A) * W / (72 * C) (males), CrCl (females) = CrCl (males) * 0.85,~Where:~A is age [years]~W is body weight [kg]~C is the serum concentration of creatinine [mg/dL]" (NCT00170846)
Timeframe: Baseline and 24 months

,,
InterventionmL/min (Mean)
Baseline CrCl(CG) ≤ 50 :(n= 32, 28, 32):- ChangeBaseline CrCl(CG) > 50 : (n=31, 29, 39):-Change
Group A: No RAD1.55-2.55
Group B : CNI Withdrawal-5.757.32
Group C: CNI Reduction0.82-0.24

[back to top]

Renal Function Assessed by Measured GFR (mGFR)

The acceptable methods for GFR measurement were Chromium 51-Ethylenediaminetetra acetic acid (Cr-EDTA), Technetium 99-Diethylenetriaminepentacetic acid (Tc-DTPA), Iohexol clearance Inuline clearance and Iothalamate clearance. The method should have been consistent for a given patient at every time point. (NCT00170846)
Timeframe: 24 months

InterventionmL/min/1.73m^2 (Mean)
Group A: No RAD46.02
Group B : CNI Withdrawal48.00
Group C: CNI Reduction46.60

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 5 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS20.3
TAXUS® EXPRESS2™ ECSS26.6

[back to top]

% Volume Obstruction (% VO)

Defined as stent intimal hyperplasia and calculated as 100*(Stent Volume - Lumen Volume)/Stent Volume by IVUS. (NCT00180479)
Timeframe: at 240 days

Interventionpercent of volume obstruction (Mean)
XIENCE V® EECSS6.91
TAXUS® EXPRESS2™ ECSS11.21

[back to top]

Acute Success: Clinical Device

Successful delivery and deployment of 1st implanted study stent/s @ the intended target lesion and successful withdrawal of the stent delivery system with final residual stenosis < 50%. (NCT00180479)
Timeframe: In-hospital

Interventionpercentage of participants (Number)
XIENCE V® EECSS98.3
TAXUS® EXPRESS2™ ECSS98.7

[back to top]

Acute Success: Clinical Procedure

Successful delivery and deployment of study stent/s @ the intended target lesion and successful withdrawal of the stent delivery system with final residual stenosis < 50%. (NCT00180479)
Timeframe: In-hospital

Interventionpercentage of participants (Number)
XIENCE V® EECSS98.5
TAXUS® EXPRESS2™ ECSS97.3

[back to top]

Distal Late Loss

Distal MLD post-procedure minus distal MLD at follow-up (distal defined as within 5 mm of healthy tissue distal to stent placement) (NCT00180479)
Timeframe: 240 days

Interventionmillimeters (Mean)
XIENCE V® EECSS0.09
TAXUS® EXPRESS2™ ECSS0.10

[back to top]

In-segment % Angiographic Binary Restenosis (% ABR) Rate

Percent of subjects with a follow-up in-segment percent diameter stenosis of ≥ 50% per QCA (NCT00180479)
Timeframe: 240 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS4.7
TAXUS® EXPRESS2™ ECSS8.9

[back to top]

In-segment % Diameter Stenosis (% DS)

Within the margins of the stent, 5 mm proximal and 5 mm distal to the stent, the value calculated as 100 * (1 - in-segment MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00180479)
Timeframe: 240 days

Interventionpercent of in-segment diameter stenosis (Mean)
XIENCE V® EECSS18.77
TAXUS® EXPRESS2™ ECSS22.82

[back to top]

In-stent % Angiographic Binary Restenosis (% ABR) Rate

Percent of subjects with a follow-up in-stent percent diameter stenosis of ≥ 50% per quantitative coronary angiography (QCA) (NCT00180479)
Timeframe: at 240 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS2.3
TAXUS® EXPRESS2™ ECSS5.7

[back to top]

In-stent % Diameter Stenosis (% DS)

In-stent: Within the margins of the stent, the value calculated as 100 * (1 - in-stent MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00180479)
Timeframe: at 240 days

Interventionpercent diameter stenosis (Mean)
XIENCE V® EECSS5.92
TAXUS® EXPRESS2™ ECSS10.30

[back to top]

In-stent Late Loss

In-stent MLD post-procedure minus in-stent MLD at follow-up (in-stent defined as within the margins of the stent) (NCT00180479)
Timeframe: at 240 days

Interventionmillimeters (Mean)
XIENCE V® EECSS0.16
TAXUS® EXPRESS2™ ECSS0.30

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 1 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS6.0
TAXUS® EXPRESS2™ ECSS10.3

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS5.0
TAXUS® EXPRESS2™ ECSS8.8

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 3 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS9.7
TAXUS® EXPRESS2™ ECSS16.4

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS1.3
TAXUS® EXPRESS2™ ECSS3.0

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 4 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS12.8
TAXUS® EXPRESS2™ ECSS18.5

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 5 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS14.4
TAXUS® EXPRESS2™ ECSS22.0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 1 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS3.4
TAXUS® EXPRESS2™ ECSS5.6

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS1.5
TAXUS® EXPRESS2™ ECSS2.1

[back to top]

Ischemia Driven Major Adverse Cardiac Event(MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 2 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS7.7
TAXUS® EXPRESS2™ ECSS13.8

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 2 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS5.7
TAXUS® EXPRESS2™ ECSS9.2

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS2.7
TAXUS® EXPRESS2™ ECSS5.0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 3 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS5.7
TAXUS® EXPRESS2™ ECSS9.2

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS0.4
TAXUS® EXPRESS2™ ECSS0.3

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 4 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS8.0
TAXUS® EXPRESS2™ ECSS10.6

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"Revascularization @ target lesion associated w/ any of following:~(+) functional ischemia study Ischemic symptoms & angiographic diameter stenosis ≥50% by core lab quantitative coronary angiography (QCA) Revascularization of a target lesion w/ angiographic diameter stenosis ≥70% by core laboratory QCA without angina or (+) functional study" (NCT00180479)
Timeframe: 5 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS8.9
TAXUS® EXPRESS2™ ECSS12.9

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 1 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS3.1
TAXUS® EXPRESS2™ ECSS4.7

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS1.2
TAXUS® EXPRESS2™ ECSS1.8

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 2 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS4.9
TAXUS® EXPRESS2™ ECSS6.6

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS2.9
TAXUS® EXPRESS2™ ECSS4.1

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 3 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS6.7
TAXUS® EXPRESS2™ ECSS8.9

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS0.3
TAXUS® EXPRESS2™ ECSS0.9

[back to top]

Ischemia Driven Major Adverse Cardiac Event (MACE)

"The composite endpoint comprised of:~Cardiac death~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI" (NCT00180479)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS2.9
TAXUS® EXPRESS2™ ECSS5.2

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 4 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS7.8
TAXUS® EXPRESS2™ ECSS9.6

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"Revascularization at the target vessel associated with any of the following~Positive functional ischemia study~Ischemic symptoms and angiographic diameter stenosis ≥ 50% by core laboratory QCA~Revascularization of a target vessel with angiographic diameter stenosis ≥ 70% by core laboratory QCA without angina or positive functional study~Derived from Non-Hierarchical Subject Counts of Adverse Events" (NCT00180479)
Timeframe: 5 years

Interventionpercentage of participants (Number)
XIENCE V® EECSS8.8
TAXUS® EXPRESS2™ ECSS11.9

[back to top]

Major Secondary Endpoint: Ischemia Driven Target Vessel Failure (ID-TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS7.2
TAXUS® EXPRESS2™ ECSS9.0

[back to top]

Persisting Incomplete Stent Apposition, Late-acquired Incomplete Stent Apposition, Aneurysm, Thrombosis, and Persisting Dissection

"Incomplete Apposition (Persisting & Late acquired): Failure to completely appose vessel wall w/ ≥1 strut separated from vessel wall w/ blood behind strut per ultrasound. Aneurysm: Abnormal vessel expansion ≥ 1.5 of reference vessel diameter. Thrombus: Protocol & ARC definition.~Persisting dissection @ follow-up, present post-procedure." (NCT00180479)
Timeframe: at 240 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS24.4
TAXUS® EXPRESS2™ ECSS14.0

[back to top]

Primary Endpoint: In-segment Late Loss (LL)

In-segment minimal lumen diameter (MLD) post-procedure minus (-) in segment MLD at 240 day follow-up and 5 mm proximal and 5mm distal to the stent equals Late Loss. MLD defined: The average of two orthogonal views (when possible) of the narrowest point within the area of assessment. (NCT00180479)
Timeframe: 240 days

Interventionmillimeters (Mean)
XIENCE V® EECSS0.14
TAXUS® EXPRESS2™ ECSS0.28

[back to top]

Proximal Late Loss

Proximal Minimum Lumen Diameter (MLD) post-procedure minus proximal MLD at follow-up (proximal defined as within 5 mm of healthy tissue proximal to stent placement) (NCT00180479)
Timeframe: at 240 days

Interventionmillimeters (Mean)
XIENCE V® EECSS0.12
TAXUS® EXPRESS2™ ECSS0.20

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 1 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS8.6
TAXUS® EXPRESS2™ ECSS11.6

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS4.1
TAXUS® EXPRESS2™ ECSS5.5

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 2 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS11.3
TAXUS® EXPRESS2™ ECSS16.4

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 3 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS14.3
TAXUS® EXPRESS2™ ECSS20.0

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V® EECSS1.6
TAXUS® EXPRESS2™ ECSS3.3

[back to top]

Target Vessel Failure (TVF)

"The composite endpoint comprised of:~Cardiac death (death in which a cardiac cause cannot be excluded)~Myocardial infarction (MI, classified as Q-wave and non-Q wave)~Ischemia-driven target lesion revascularization (TLR) by CABG or PCI~Ischemia-driven target vessel revascularization (TVR) by CABG or PCI" (NCT00180479)
Timeframe: 4 year

Interventionpercentage of participants (Number)
XIENCE V® EECSS18.5
TAXUS® EXPRESS2™ ECSS22.5

[back to top]

Number of Participants With Composite Efficacy Endpoints - 12 Month Analysis

The primary efficacy endpoint was the 12 month analysis of primary efficacy failure defined as a composite endpoint including treated biopsy proven acute rejection (BPAR), graft loss, death or loss to follow-up. A treated BPAR episode was defined as a biopsy graded IA, IB, IIA, IIB, or III that was treated with anti-rejection therapy. Graft loss was defined as the allograft was presumed to be lost on the day the patient started dialysis and was not able to subsequently be removed from dialysis as well as re-transplant. (NCT00251004)
Timeframe: 12 months

,,
InterventionParticipants (Number)
Composite Endpoint (n)--Death--Graft Loss--Treated BPAR--Lost to follow up
Control Group7069509
High-dose Everolimus Group60913386
Low-dose Everolimus Group758134812

[back to top]

Percentage of Participants With the Composite Incidence of Graft Loss, Death or Loss to Follow up at 12 Months Post-transplantation

"Graft loss was defined as graft loss (the allograft was presumed to be lost on the day the patient started dialysis and was not able to subsequently be removed from dialysis) and re-transplant.~A loss to follow-up patient in the composite endpoint of graft loss, death or loss to follow-up (the main secondary efficacy endpoint) was a patient who did not experience graft loss or death from day 1 and whose last day of contact was prior to study day 316." (NCT00251004)
Timeframe: 12 months

InterventionPercentage of participants (Number)
Low-dose Everolimus Group11.6
High-dose Everolimus Group9.7
Control Group9.4

[back to top]

Non-inferiority Analysis of Renal Function, Calculated by Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula

"Modification of Diet in Renal Disease (MDRD) formula is:~GFR [mL/min/1.73m^2] = 186.3*(C^-1.154)*(A^-0.203)*G*R where~C is the serum concentration of creatinine [mg/dL],~A is patient age at sample collection date [years],~G=0.742 when gender is female, otherwise G=1,~R=1.21 when race is black, otherwise R=1" (NCT00251004)
Timeframe: at 12 months

InterventionmL/min/1.73m^2 (Number)
Low-dose Everolimus Group54.66
High-dose Everolimus Group51.41
Control Group52.24

[back to top]

Non-inferiority Analysis on Percentage of Participants With Composite Efficacy Endpoints

The primary efficacy endpoint was the 12 month analysis of primary efficacy failure defined as a composite endpoint including treated biopsy proven acute rejection (BPAR), graft loss, death or loss to follow-up. In the definition of composite efficacy failure, loss to follow-up includes patients who did not experience treated BPAR, graft loss or death on or after day 1 and whose last day of contact was prior to day 316, the start day of the 12 month visit window. (NCT00251004)
Timeframe: 12 months

InterventionPercentage of Participants (Number)
Low-dose Everolimus Group27.1
High-dose Everolimus Group21.5
Control Group25.3

[back to top]

Percentage of Patients With Efficacy Failure (Biopsy Proven Acute Rejection [BPAR], Graft Loss or Death)

The composite efficacy failure endpoint encompasses at least one of: biopsy proven acute rejection, graft loss, or death for the patient. BPAR was defined as a clinically suspected acute rejection confirmed by biopsy. Acute rejection episodes were recorded as Liver Allograft Rejection. The allograft was presumed to be lost if a patient had a liver retransplant or died. (NCT00267189)
Timeframe: 6 months

,
InterventionPercentage of patients (Number)
Composite efficacy failure (total)Biopsy proven acute rejectionGraft LossDeath
Group 1 (Everolimus)2.81.401.4
Group 2 (Control)1.41.400

[back to top]

Mean Change From Baseline in Cockcroft-Gault Calculated Creatinine Clearance (CrCl)

"The primary variable was renal function assessed by calculated creatinine clearance using the Cockcroft-Gault formula, and was assessed at all visits.~CrCl[mL/min] = (140 - A) * W / (72 * C) * R. Where A is age at sample date [years], W is body weight at specific visit [kg], C is the serum concentration of creatinine [mg/dL], R = 1 if the patient is male and = 0.85 if female." (NCT00267189)
Timeframe: From baseline to 6 months

InterventionmL/min (Mean)
Group 1 (Everolimus)0.99
Group 2 (Control)2.26

[back to top]

Number of Patients With Discontinuation of Study Medication

(NCT00267189)
Timeframe: 6 months

,
InterventionPatients (Number)
Total # of discontinuation of study medicationAdverse EventPatient withdrew consentAbnormal laboratory value(s)Administrative problems
Group 1 (Everolimus)1814211
Group 2 (Control)10100

[back to top]

Percentage of Participants With Biopsy-proven Acute Rejection (BPAR of ISHLT Grade ≥ 3A), Acute Rejection (AR) Associated With Hemodynamic Compromise (HDC), Graft Loss/Re-transplant and Death at Month 24

"Identification of acute rejections was based on the local pathologist's evaluation of endomyocardial biopsy slides.~Hemodynamic compromise was present if 1 or more of the following were met: Ejection fraction ≤ 30% or 25% lower than Baseline or Fractional shortening ≤ 20% or 25% lower than Baseline, and/ or use of inotropic treatment." (NCT00300274)
Timeframe: 24 Months

,,
InterventionPercentage of participants (Number)
AR associated with HDCBPAR of ISHLT grade ≥ 3ADeathGraft loss/re-transplant
Everolimus 1.5 mg4.324.110.62.5
Everolimus 3.0 mg3.628.611.93.0
Mycophenolate Mofetil5.227.39.23.7

[back to top]

Renal Function Measured by Glomerular Filtration Rate (GFR) at 12 Months

"GFR was calculated using the Modification of Diet and Renal Disease (MDRD) formula:~GFR [mL/min/1.73m^2] = 186.3*(C^-1.154)*(A^-0.203)*G*R where C is the serum concentration of creatinine [mg/dL] A is age [years] G=0.742 when gender is female, otherwise G=1 R=1.21 when race is black, otherwise R=1" (NCT00300274)
Timeframe: 12 Months

InterventionmL/min/1.73^2 (Mean)
Everolimus 1.5 mg59.21
Everolimus 3.0 mg59.78
Mycophenolate Mofetil64.37

[back to top]

Renal Function Calculated by Glomerular Filtration Rate (GFR) at 24 Months

"GFR was calculated using the Modification of Diet and Renal Disease (MDRD) formula:~GFR [mL/min/1.73m^2] = 186.3*(C^-1.154)*(A^-0.203)*G*R~C is the serum concentration of creatinine [mg/dL] A is age [years] G=0.742 when gender is female, otherwise G=1 R=1.21 when race is black, otherwise R=1" (NCT00300274)
Timeframe: 24 Months

InterventionmL/min/1.73^2 (Mean)
Everolimus 1.5 mg59.50
Everolimus 3.0 mg61.84
Mycophenolate Mofetil64.52

[back to top]

Percentage of Participants With Graft Loss/Re-transplant, Death or Loss to Follow-up at 24 Months

Loss to follow-up for this composite endpoint included participants who did not experience graft loss/re-transplant or death and whose last day of contact was prior to Day 631 (start day of 24 Month visit window). (NCT00300274)
Timeframe: 24 Months

InterventionPercentage of participants (Number)
Everolimus 1.5 mg15.2
Everolimus 3.0 mg16.1
Mycophenolate Mofetil15.1

[back to top]

Percentage of Participants With Graft Loss/Re-transplant, Death or Loss to Follow-up at 12 Months

Loss to follow-up for this composite endpoint included participants who did not experience graft loss/re-transplant or death and whose last day of contact was prior to Day 316 (start day of the Month 12 visit window). (NCT00300274)
Timeframe: 12 Months

InterventionPercentage of participants (Number)
Everolimus 1.5 mg11.7
Everolimus 3.0 mg11.9
Mycophenolate Mofetil8.9

[back to top]

Percentage of Participants With Composite Efficacy Failure at 24 Months

"Composite efficacy failure was defined as Biopsy Proven Acute Rejection (BPAR) of International Society for Heart and Lung Transplantation grade ≥ 3A, Acute Rejection associated with Hemodynamic Compromise, Graft loss/Retransplant, Death or Loss to follow-up.~Identification of acute rejections was based on the local pathologist's evaluation of endomyocardial biopsy slides.~Hemodynamic compromise was present if 1 or more of the following were met: Ejection fraction ≤ 30% or 25% lower than Baseline or Fractional shortening ≤ 20% or 25% lower than Baseline and/or use of inotropic treatment." (NCT00300274)
Timeframe: 24 Months

InterventionPercentage of participants (Number)
Everolimus 1.5 mg39.4
Everolimus 3.0 mg41.1
Mycophenolate Mofetil41.3

[back to top]

Percentage of Participants With Composite Efficacy Failure at 12 Months

"Composite efficacy failure was defined as Biopsy Proven Acute Rejection(BPAR) of International Society for Heart and Lung Transplantation(ISHLT) grade ≥3A, Acute Rejection associated with Hemodynamic Compromise, Graft loss/Retransplant, Death or Loss to follow-up.~Identification of acute rejection was based on the local pathologist's evaluation of endomyocardial biopsy slides.~Hemodynamic compromise was present if 1 or more of the following were met: Ejection fraction ≤30% or 25% lower than Baseline or Fractional shortening ≤20% or 25% lower than Baseline and/or use of inotropic treatment." (NCT00300274)
Timeframe: 12 Months

InterventionPercentage of participants (Number)
Everolimus 1.5 mg35.1
Everolimus 3.0 mg35.1
Mycophenolate Mofetil33.6

[back to top]

Percentage of Participants With Cardiac Allograft Vasculopathy (CAV) at Month 12

Cardiac allograft vasculopathy is defined as a 0.5 mm increase in maximum intimal thickness as measured by Intravascular Ultrasound (IVUS) in at least one matched slice between baseline and Month 12. (NCT00300274)
Timeframe: 12 Months

InterventionPercentage of participants (Number)
Everolimus 1.5 mg12.5
Everolimus 3.0 mg21.6
Mycophenolate Mofetil26.7

[back to top]

Percentage of Participants With Biopsy-proven Acute Rejection (BPAR of ISHLT Grade ≥ 3A), Acute Rejection Associated With Hemodynamic Compromise (HDC), Graft Loss/Re-transplant and Death at Month 12

"Identification of acute rejections was based on the local pathologist's evaluation of endomyocardial biopsy slides.~Hemodynamic compromise was present if 1 or more of the following were met: Ejection fraction ≤ 30% or 25% lower than Baseline or Fractional shortening ≤ 20% or 25% lower than Baseline, and/or use of inotropic treatment." (NCT00300274)
Timeframe: 12 Months

,,
InterventionPercentage of participants (Number)
AR associated with HDCBPAR of ISHLT ≥ 3ADeathGraft loss/re-transplant
Everolimus 1.5 mg3.922.37.81.4
Everolimus 3.0 mg3.025.610.13.0
Mycophenolate Mofetil2.624.74.81.8

[back to top]

Change From Baseline in the Average Maximum Intimal Thickness at Month 12

Maximum intimal thickness was assessed using Intravascular Ultrasound (IVUS). IVUS is a technique for taking ultrasound pictures of the wall of an artery from inside the artery itself. It shows the thickness of the artery wall and any narrowing of the artery. (NCT00300274)
Timeframe: Baseline, Month 12

Interventionmm (Mean)
Everolimus 1.5 mg0.03
Everolimus 3.0 mg0.04
Mycophenolate Mofetil0.07

[back to top]

Ischemia Driven Major Adverse Cardiac Events (MACE)

Patients determined to have had a MACE event, defined as one of the following events: Cardiac death, myocardial infarction, and TLR (NCT00307047)
Timeframe: 30 days

InterventionPercentage of participants (Number)
XIENCE V®1.6
TAXUS™ EXPRESS 2™2.7

[back to top]

Ischemia Driven Target Lesion Failure (TLF)

Percentage of participants with the determination of TLF. TLF is the composite of cardiac death, target vessel myocardial infarction, and ischemic driven target lesion revascularization (TLR). (NCT00307047)
Timeframe: 1 year

InterventionPercentage of participants (Number)
XIENCE V®4.2
TAXUS™ EXPRESS 2™6.8

[back to top]

Ischemia Driven Target Lesion Failure (TLF)

Percentage of participants with the determination of TLF. TLF is the composite of cardiac death, target vessel myocardial infarction, and ischemic driven target lesion revascularization (TLR). (NCT00307047)
Timeframe: 180 days

InterventionPercentage of participants (Number)
XIENCE V®2.5
TAXUS™ EXPRESS 2™5.1

[back to top]

Ischemia Driven Target Lesion Failure (TLF)

Percentage of participants with the determination of TLF. TLF is the composite of cardiac death, target vessel myocardial infarction, and ischemic driven target lesion revascularization (TLR). (NCT00307047)
Timeframe: 2 years

InterventionPercentage of participants (Number)
XIENCE V®7.0
TAXUS™ EXPRESS 2™10.0

[back to top]

Ischemia Driven Target Lesion Failure (TLF)

Percentage of participants with the determination of TLF. TLF is the composite of cardiac death, target vessel myocardial infarction, and ischemic driven target lesion revascularization (TLR). (NCT00307047)
Timeframe: 270 days

InterventionPercentage of participants (Number)
XIENCE V®3.4
TAXUS™ EXPRESS 2™6.1

[back to top]

Ischemia Driven Target Lesion Failure (TLF)

Percentage of participants with the determination of TLF. TLF is the composite of cardiac death, target vessel myocardial infarction, and ischemic driven target lesion revascularization (TLR). (NCT00307047)
Timeframe: 30 days

InterventionPercentage of participants (Number)
XIENCE V®1.6
TAXUS™ EXPRESS 2™2.7

[back to top]

Ischemia Driven Target Lesion Revascularization (TLR)

"Revascularization of a target lesion associated with any of the following:~positive functional ischemia study~ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 1 year

Interventionpercentage of participants (Number)
XIENCE V®2.5
TAXUS™ EXPRESS 2™4.6

[back to top]

Ischemia Driven Target Lesion Revascularization (TLR)

"Revascularization of a target lesion associated with any of the following:~positive functional ischemia study~ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V®1.1
TAXUS™ EXPRESS 2™3.2

[back to top]

Ischemia Driven Target Lesion Revascularization (TLR)

"Revascularization of a target lesion associated with any of the following:~positive functional ischemia study~ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 2 years

Interventionpercentage of participants (Number)
XIENCE V®4.4
TAXUS™ EXPRESS 2™6.9

[back to top]

Ischemia Driven Target Lesion Revascularization (TLR)

"Revascularization of a target lesion associated with any of the following:~positive functional ischemia study~ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V®1.9
TAXUS™ EXPRESS 2™4.1

[back to top]

Ischemia Driven Target Lesion Revascularization (TLR)

"Revascularization of a target lesion associated with any of the following:~positive functional ischemia study~ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 3 years

Interventionpercentage of participants (Number)
XIENCE V®6.3
TAXUS™ EXPRESS 2™7.9

[back to top]

Ischemia Driven Target Lesion Revascularization (TLR)

"Revascularization of a target lesion associated with any of the following:~positive functional ischemia study~ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V®0.4
TAXUS™ EXPRESS 2™1.1

[back to top]

Ischemia Driven Target Vessel Failure (TVF)

Defined as the composite endpoint comprised of cardiac death (CD), myocardial infarction (MI), TLR, and TVR (NCT00307047)
Timeframe: 1 year

Interventionpercentage of participants (Number)
XIENCE V®5.6
TAXUS™ EXPRESS 2™7.9

[back to top]

Ischemia Driven Target Vessel Failure (TVF)

Defined as the composite endpoint comprised of cardiac death (CD), myocardial infarction (MI), TLR, and TVR (NCT00307047)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V®3.4
TAXUS™ EXPRESS 2™6.2

[back to top]

Ischemia Driven Target Vessel Failure (TVF)

Defined as the composite endpoint comprised of cardiac death (CD), myocardial infarction (MI), TLR, and TVR (NCT00307047)
Timeframe: 2 years

Interventionpercentage of participants (Number)
XIENCE V®9.6
TAXUS™ EXPRESS 2™11.8

[back to top]

Ischemia Driven Target Vessel Failure (TVF)

Defined as the composite endpoint comprised of cardiac death (CD), myocardial infarction (MI), TLR, and TVR (NCT00307047)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V®4.6
TAXUS™ EXPRESS 2™7.2

[back to top]

Ischemia Driven Target Vessel Failure (TVF)

Defined as the composite endpoint comprised of cardiac death (CD), myocardial infarction (MI), TLR, and TVR (NCT00307047)
Timeframe: 3 years

Interventionpercentage of participants (Number)
XIENCE V®13.3
TAXUS™ EXPRESS 2™14.5

[back to top]

Ischemia Driven Target Vessel Failure (TVF)

Defined as the composite endpoint comprised of cardiac death (CD), myocardial infarction (MI), TLR, and TVR (NCT00307047)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V®1.9
TAXUS™ EXPRESS 2™3.1

[back to top]

Ischemia Driven Target Vessel Revascularization (TVR)

"Revascularization of a lesion within the target vessel associated with any of the following:~positive functional ischemia study~ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 1 year

Interventionpercentage of participants (Number)
XIENCE V®3.9
TAXUS™ EXPRESS 2™5.9

[back to top]

Ischemia Driven Target Vessel Revascularization (TVR)

"Revascularization of a lesion within the target vessel associated with any of the following:~positive functional ischemia study~ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 180 days

Interventionpercentage of participants (Number)
XIENCE V®1.9
TAXUS™ EXPRESS 2™4.3

[back to top]

All Cause Mortality

(NCT00307047)
Timeframe: 3 years

InterventionPercentage of participants (Number)
XIENCE V®3.4
TAXUS™ EXPRESS 2™5.2

[back to top]

All Cause Mortality

(NCT00307047)
Timeframe: 270 days

InterventionPercentage of participants (Number)
XIENCE V®0.7
TAXUS™ EXPRESS 2™0.9

[back to top]

All Cause Mortality

(NCT00307047)
Timeframe: 2 years

InterventionPercentage of participants (Number)
XIENCE V®2.1
TAXUS™ EXPRESS 2™2.7

[back to top]

All Cause Mortality

(NCT00307047)
Timeframe: 180 days

InterventionPercentage of participants (Number)
XIENCE V®0.5
TAXUS™ EXPRESS 2™0.6

[back to top]

All Cause Mortality

(NCT00307047)
Timeframe: 1 year

InterventionPercentage of participants (Number)
XIENCE V®1.0
TAXUS™ EXPRESS 2™1.3

[back to top]

Acute Success (Clinical Procedure)

Successful delivery and deployment of the study stent or stents at the intended target lesion and successful withdrawal of the stent delivery system with attainment of final residual stenosis of less than 50% of the target lesion by QCA (by visual estimation if QCA unavailable) and/or using any adjunctive device without the occurrence of major adverse cardiac event (MACE) during the hospital stay with a maximum of first seven days following the index procedure. In multiple lesion setting all lesions must meet clinical procedure success. (NCT00307047)
Timeframe: Acute: At time of index procedure

InterventionPercentage of success (Number)
XIENCE V®98.6
TAXUS™ EXPRESS 2™98.1

[back to top]

Acute Success (Clinical Device)

Successful delivery and deployment of the first implanted study stent (in overlapping stent setting a successful delivery and deployment of the first and second study stents) at the intended target lesion and successful withdrawal of the stent delivery system with attainment of final residual stenosis of less than 50% of the target lesion by QCA (by visual estimation if QCA unavailable). Bailout subjects will be included as device success only if the above criteria for clinical device are met. (NCT00307047)
Timeframe: Acute: At time of index procedure

InterventionPercent of success (Number)
XIENCE V®92.0
TAXUS™ EXPRESS 2™90.7

[back to top]

Ischemia Driven Target Vessel Revascularization (TVR)

"Revascularization of a lesion within the target vessel associated with any of the following:~positive functional ischemia study~ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 2 years

Interventionpercentage of participants (Number)
XIENCE V®7.0
TAXUS™ EXPRESS 2™8.9

[back to top]

Ischemia Driven Target Vessel Revascularization (TVR)

"Revascularization of a lesion within the target vessel associated with any of the following:~positive functional ischemia study~ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 270 days

Interventionpercentage of participants (Number)
XIENCE V®3.0
TAXUS™ EXPRESS 2™5.3

[back to top]

Ischemia Driven Target Vessel Revascularization (TVR)

"Revascularization of a lesion within the target vessel associated with any of the following:~positive functional ischemia study~ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 3 years

Interventionpercentage of participants (Number)
XIENCE V®10.1
TAXUS™ EXPRESS 2™10.6

[back to top]

Ischemia Driven Target Vessel Revascularization (TVR)

"Revascularization of a lesion within the target vessel associated with any of the following:~positive functional ischemia study~ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~angiographic diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study" (NCT00307047)
Timeframe: 30 days

Interventionpercentage of participants (Number)
XIENCE V®0.7
TAXUS™ EXPRESS 2™1.6

[back to top]

Ischemia Driven Major Adverse Cardiac Events (MACE)

Patients determined to have had a MACE event, defined as one of the following events: Cardiac death, myocardial infarction, and TLR (NCT00307047)
Timeframe: 3 years

InterventionPercentage of participants (Number)
XIENCE V®9.8
TAXUS™ EXPRESS 2™12.3

[back to top]

Protocol Defined Stent Thrombosis Rate

"ST will be categorized as acute (≤ 1day), subacute (>1 day to ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of ST~In the absence of angiography, any unexplained death, or acute MI (S-T segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific S-T/T changes, and cardiac enzyme elevations do not suffice) Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00307047)
Timeframe: 0-393 days

InterventionPercentage of participants (Number)
XIENCE V®0.17
TAXUS™ EXPRESS 2™0.85

[back to top]

Protocol Defined Stent Thrombosis Rate

"ST will be categorized as acute (≤ 1day), subacute (>1 day to ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of ST~In the absence of angiography, any unexplained death, or acute MI (S-T segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific S-T/T changes, and cardiac enzyme elevations do not suffice) Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00307047)
Timeframe: 0-30 days

InterventionPercentage of participants (Number)
XIENCE V®0.12
TAXUS™ EXPRESS 2™0.57

[back to top]

Protocol Defined Stent Thrombosis Rate

"ST will be categorized as acute (≤ 1day), subacute (>1 day to ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of ST~In the absence of angiography, any unexplained death, or acute MI (S-T segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific S-T/T changes, and cardiac enzyme elevations do not suffice) Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00307047)
Timeframe: 0-758 days

InterventionPercentage of participants (Number)
XIENCE V®0.52
TAXUS™ EXPRESS 2™1.23

[back to top]

Protocol Defined Stent Thrombosis Rate

"ST will be categorized as acute (≤ 1day), subacute (>1 day to ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of ST~In the absence of angiography, any unexplained death, or acute MI (S-T segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific S-T/T changes, and cardiac enzyme elevations do not suffice) Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00307047)
Timeframe: 31-393 days

InterventionPercentage of participants (Number)
XIENCE V®0.04
TAXUS™ EXPRESS 2™0.34

[back to top]

All Myocardial Infarction (MI)

(NCT00307047)
Timeframe: 30 days

InterventionPercentage of participants (Number)
XIENCE V®1.5
TAXUS™ EXPRESS 2™2.1

[back to top]

Ischemia Driven Target Lesion Failure (TLF)

Percentage of participants with the determination of TLF. TLF is the composite of cardiac death, target vessel myocardial infarction, and ischemic driven target lesion revascularization (TLR). (NCT00307047)
Timeframe: 3 years

InterventionPercentage of participants (Number)
XIENCE V®9.5
TAXUS™ EXPRESS 2™11.9

[back to top]

Protocol Defined Stent Thrombosis Rate

"ST will be categorized as acute (≤ 1day), subacute (>1 day to ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of ST~In the absence of angiography, any unexplained death, or acute MI (S-T segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific S-T/T changes, and cardiac enzyme elevations do not suffice) Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00307047)
Timeframe: 0-1123 days

InterventionPercentage of participants (Number)
XIENCE V®0.79
TAXUS™ EXPRESS 2™1.99

[back to top]

Ischemia Driven Major Adverse Cardiac Events (MACE)

Patients determined to have had a MACE event, defined as one of the following events: Cardiac death, myocardial infarction, and TLR (NCT00307047)
Timeframe: 270 days

InterventionPercentage of participants (Number)
XIENCE V®3.5
TAXUS™ EXPRESS 2™6.2

[back to top]

Ischemia Driven Major Adverse Cardiac Events (MACE)

Patients determined to have had a MACE event, defined as one of the following events: Cardiac death, myocardial infarction, and TLR (NCT00307047)
Timeframe: 2 years

InterventionPercentage of participants (Number)
XIENCE V®7.2
TAXUS™ EXPRESS 2™10.2

[back to top]

Ischemia Driven Major Adverse Cardiac Events (MACE)

Patients determined to have had a MACE event, defined as one of the following events: Cardiac death, myocardial infarction, and TLR (NCT00307047)
Timeframe: 180 days

InterventionPercentage of participants (Number)
XIENCE V®2.6
TAXUS™ EXPRESS 2™5.3

[back to top]

Ischemia Driven Major Adverse Cardiac Events (MACE)

Patients determined to have had a MACE event, defined as one of the following events: Cardiac death, myocardial infarction, and TLR (NCT00307047)
Timeframe: 1 years

InterventionPercentage of participants (Number)
XIENCE V®4.2
TAXUS™ EXPRESS 2™6.9

[back to top]

Definite + Probable Stent Thrombosis Rate Based on ARC Definition

"ARC: Academic Research Consortium-defines ST as a cumulative value at the different time points and with the different seperate time points. Time 0 is defined as the time point after the guiding catheter has been removed. Acute*: 0-24 hours post implantation Subacute*: >24 hours-30 days post Late†: 30 days-1 year post Very late stent thrombosis†: >1 year post~* Acute/subacute can also be replaced by early ST. Early ST (0-30 days) is currently used in the community.~† Including primary as well as secondary late ST; secondary late ST is after a target segment revascularization." (NCT00307047)
Timeframe: 31-393 days

InterventionPercentage of participants (Number)
XIENCE V®0.13
TAXUS™ EXPRESS 2™0.42

[back to top]

Definite + Probable Stent Thrombosis Rate Based on ARC Definition

"ARC: Academic Research Consortium-defines ST as a cumulative value at the different time points and with the different seperate time points. Time 0 is defined as the time point after the guiding catheter has been removed. Acute*: 0-24 hours post implantation Subacute*: >24 hours-30 days post Late†: 30 days-1 year post Very late stent thrombosis†: >1 year post~* Acute/subacute can also be replaced by early ST. Early ST (0-30 days) is currently used in the community.~† Including primary as well as secondary late ST; secondary late ST is after a target segment revascularization." (NCT00307047)
Timeframe: 0-758 days

InterventionPercentage of participants (Number)
XIENCE V®0.48
TAXUS™ EXPRESS 2™1.32

[back to top]

Definite + Probable Stent Thrombosis Rate Based on ARC Definition

"ARC: Academic Research Consortium-defines ST as a cumulative value at the different time points and with the different seperate time points. Time 0 is defined as the time point after the guiding catheter has been removed. Acute*: 0-24 hours post implantation Subacute*: >24 hours-30 days post Late†: 30 days-1 year post Very late stent thrombosis†: >1 year post~* Acute/subacute can also be replaced by early ST. Early ST (0-30 days) is currently used in the community.~† Including primary as well as secondary late ST; secondary late ST is after a target segment revascularization." (NCT00307047)
Timeframe: 0-1123 days

InterventionPercentage of participants (Number)
XIENCE V®0.62
TAXUS™ EXPRESS 2™1.73

[back to top]

Definite + Probable Stent Thrombosis Rate Based on ARC Definition

"ARC: Academic Research Consortium-defines ST as a cumulative value at the different time points and with the different seperate time points. Time 0 is defined as the time point after the guiding catheter has been removed. Acute*: 0-24 hours post implantation Subacute*: >24 hours-30 days post Late†: 30 days-1 year post Very late stent thrombosis†: >1 year post~* Acute/subacute can also be replaced by early ST. Early ST (0-30 days) is currently used in the community.~† Including primary as well as secondary late ST; secondary late ST is after a target segment revascularization." (NCT00307047)
Timeframe: 0 -393 days

InterventionPercentage of participants (Number)
XIENCE V®0.29
TAXUS™ EXPRESS 2™1.10

[back to top]

Definite + Probable Stent Thrombosis Rate Based on Academic Research Consortium (ARC) Definition

"ARC: Academic Research Consortium-defines ST as a cumulative value at the different time points and with the different seperate time points. Time 0 is defined as the time point after the guiding catheter has been removed. Acute*: 0-24 hours post implantation Subacute*: >24 hours-30 days post Late†: 30 days-1 year post Very late stent thrombosis†: >1 year post~* Acute/subacute can also be replaced by early ST. Early ST (0-30 days) is currently used in the community.~† Including primary as well as secondary late ST; secondary late ST is after a target segment revascularization." (NCT00307047)
Timeframe: 0-30 days

InterventionPercentage of participants (Number)
XIENCE V®0.16
TAXUS™ EXPRESS 2™0.74

[back to top]

Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)

(NCT00307047)
Timeframe: 30 days

InterventionPercentage of participants (Number)
XIENCE V®2.4
TAXUS™ EXPRESS 2™3.6

[back to top]

Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)

(NCT00307047)
Timeframe: 3 years

InterventionPercentage of participants (Number)
XIENCE V®21.4
TAXUS™ EXPRESS 2™22.5

[back to top]

Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)

(NCT00307047)
Timeframe: 270 days

InterventionPercentage of participants (Number)
XIENCE V®7.2
TAXUS™ EXPRESS 2™9.3

[back to top]

Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)

(NCT00307047)
Timeframe: 2 years

InterventionPercentage of participants (Number)
XIENCE V®15.5
TAXUS™ EXPRESS 2™16.2

[back to top]

Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)

(NCT00307047)
Timeframe: 180 days

InterventionPercentage of participants (Number)
XIENCE V®5.5
TAXUS™ EXPRESS 2™7.5

[back to top]

Composite Endpoint of All Deaths, All MI, All Revascularizations (DMR)

(NCT00307047)
Timeframe: 1 year

InterventionPercentage of participants (Number)
XIENCE V®9.0
TAXUS™ EXPRESS 2™10.5

[back to top]

Cardiac Death or Target Vessel MI Rate

(NCT00307047)
Timeframe: 30 days

InterventionPercentage of participants (Number)
XIENCE V®1.5
TAXUS™ EXPRESS 2™2.1

[back to top]

Cardiac Death or Target Vessel MI Rate

(NCT00307047)
Timeframe: 3 years

InterventionPercentage of participants (Number)
XIENCE V®4.1
TAXUS™ EXPRESS 2™5.5

[back to top]

Cardiac Death or Target Vessel MI Rate

(NCT00307047)
Timeframe: 270 days

InterventionPercentage of participants (Number)
XIENCE V®2.1
TAXUS™ EXPRESS 2™3.0

[back to top]

Cardiac Death or Target Vessel MI Rate

(NCT00307047)
Timeframe: 2 years

InterventionPercentage of participants (Number)
XIENCE V®3.2
TAXUS™ EXPRESS 2™4.3

[back to top]

Cardiac Death or Target Vessel MI Rate

(NCT00307047)
Timeframe: 180 days

InterventionPercentage of participants (Number)
XIENCE V®1.8
TAXUS™ EXPRESS 2™2.8

[back to top]

Cardiac Death or Target Vessel MI Rate

(NCT00307047)
Timeframe: 1 year

InterventionPercentage of participants (Number)
XIENCE V®2.2
TAXUS™ EXPRESS 2™3.2

[back to top]

All MI

(NCT00307047)
Timeframe: 3 years

InterventionPercentage of participants (Number)
XIENCE V®3.1
TAXUS™ EXPRESS 2™4.7

[back to top]

All MI

(NCT00307047)
Timeframe: 270 days

InterventionPercentage of participants (Number)
XIENCE V®1.8
TAXUS™ EXPRESS 2™3.0

[back to top]

All MI

(NCT00307047)
Timeframe: 2 years

InterventionPercentage of participants (Number)
XIENCE V®2.6
TAXUS™ EXPRESS 2™3.9

[back to top]

All MI

(NCT00307047)
Timeframe: 180 days

InterventionPercentage of participants (Number)
XIENCE V®1.6
TAXUS™ EXPRESS 2™2.9

[back to top]

All MI

(NCT00307047)
Timeframe: 1 year

InterventionPercentage of participants (Number)
XIENCE V®1.9
TAXUS™ EXPRESS 2™3.1

[back to top]

All Cause Mortality

(NCT00307047)
Timeframe: 30 days

InterventionPercentage of participants (Number)
XIENCE V®0.0
TAXUS™ EXPRESS 2™0.2

[back to top]

Clinical Benefit Response Rate (CBR)

Efficacy measured by the clinical benefit response rate (CBR), defined as confirmed Complete Response (CR) plus Partial Response (PR) at any time plus Persistent Stable Disease (pSD). Confirmed CR is defined as disappearance of all target lesions at the time of radiographic evaluation; pSD was defined as SD lasting 24 weeks. Complete Response (CR): disappearance of all target lesions, and Partial Response (PR): at least a 30% decrease in the sum of longest diameter (LD) of target lesions taking as a reference the baseline sum LD. Stable Disease (SD): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease taking as references the smallest sum LD since the treatment started. (NCT00317720)
Timeframe: 6 weeks

Interventionpercentage of partcipants (Number)
CBRCRPRpSD
Trastuzumab + RAD0013401519

[back to top]

Optimal Dose of RAD001 in Combination With Trastuzumab (Phase I)

"In Phase I, two dose levels of RAD001 were studied: 10 mg (dose level 1) and 5 mg (dose level -1) where each dose was evaluated after cycle 1. At MDACC, the Continual Reassessment Method (CRM) for determining Maximum Tolerated Dose (MTD) was applied to the two predefined RAD001 dose levels; and at DFCI/BIDMC, a 3 x 3 study design was utilized.~Optimal dose defined as the dose most closely associated with a toxicity rate of 0.20, and toxicity defined as any grade 3 or 4 toxicity (based on Common Terminology Criteria (CTC) version 3.0 except fatigue. Participants underwent clinical evaluation every 3 weeks (one cycle) and radiologic evaluations every 6 weeks. After the second cycle, participants underwent a radiologic evaluation using the same imaging technique used at initial evaluation (ie, computed tomography or magnetic resonance imaging)." (NCT00317720)
Timeframe: Following two 3 week cycles of therapy

Interventionmg (Number)
Trastuzumab + RAD00110

[back to top]

Progression Free Survival (PFS), the Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Worsening of Their Disease

(NCT00323739)
Timeframe: 18 months

InterventionMonths (Median)
Bevacizumab and Everolimus8.1

[back to top]

Overall Survival (OS), the Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Death

(NCT00323739)
Timeframe: 18 months

InterventionMonths (Median)
Bevacizumab and Everolimus18.5

[back to top]

Median Time to Progression

(NCT00331409)
Timeframe: Time to progression

Interventionmonths (Median)
Everolimus and Imatinib Mesylate2.9

[back to top]

Number of Participants With Adverse Events

"Toxicity assessments will be obtained as follows:~Cycle 1: Weeks 1,2,3 Cycle 2: Weeks 6,9 Cycle 3: Weeks 12, 15 Cycle 4: Weeks 18, 21 Cycle 5: Weeks 24, 27 Cycle 6+: Every visit during these cycles~Safety assessments will consist of evaluating adverse events and serious adverse events." (NCT00331409)
Timeframe: Duration of study, Up to 4 years

Interventionparticipants (Number)
Everolimus and Imatinib Mesylate19

[back to top]

Number of Subjects That Demonstrated a Reduction in Tumor Measurements.

Number of subjects that received at least one post-baseline scan that demonstrated a reduction in sum target lesions per Response Evaluation Criteria in Solid Tumors (RECIST) criteria. (NCT00331409)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Everolimus and Imatinib Mesylate5

[back to top]

Overall Number of Participants Who Achieve a Response Rate (Complete Response, Partial Response, and Stable Disease) at 3 Months

(NCT00331409)
Timeframe: Up to 4 years

Interventionparticipants (Number)
Everolimus and Imatinib Mesylate18

[back to top]

Progression-free Survival at 3 Months

(NCT00331409)
Timeframe: 3 months post 1st dose

Interventionmonths (Median)
Everolimus and Imatinib Mesylate2.9

[back to top]

Number of Participants Who Experienced Adverse Events and Death

Participants were monitored for adverse events, serious adverse events and deaths thorughout the prospective and follow-up phases of the study. (NCT00332839)
Timeframe: 12 months

,
InterventionParticipants (Number)
Adverse events (serious and non-serious)Serious adverse eventsDeaths
Calcineurin Inhibitor (CNI) Group44111
Certican Group44121

[back to top]

Everolimus Trough Level Determination by Pharmacokinetics Parameter in Both Strata (Stratum 1 and 2)

For all patients in both strata, a blood sample for everolimus trough level determination will be collected immediately prior to the everolimus administration on Cycle 1 Day 15, Cycle 2 Day 1, and every month thereafter. A treatment cycle was defined as 28 days of consecutive daily treatment with everolimus and treatment continued until tumor progression. It is critical that patients not take their daily everolimus dose before the sample is drawn. (NCT00363051)
Timeframe: Cycle 1 Day 15

Interventionng/ml (Mean)
Stratum 1: Everolimus 10 mg15.7
Stratum 2: Everolimus 10 mg + Octreotide Depot17.3

[back to top]

Objective Response Rate: Percentage of Participants With Best Over All Response of Complete Response or Partial Response by Central Radiology Review (Stratum 1) Based on Response Evaluation Criteria in Solid Tumors (RECIST)

Objective response rate was defined by RECIST criteria: Partial response (PR) must have ≥ 30% decrease in the sum of longest diameter of all target lesions, from the baseline sum. Complete response (CR) must have disappearance of all target and non-target lesions. For CR or PR, tumor measurements must be confirmed by 2nd assessments within 4 weeks. Progression = 20% increase in the sum of longest diameter of all target lesions, from smallest sum of longest diameter of all target lesions recorded at or after baseline; or a new lesion; or progression of non-target lesions. (NCT00363051)
Timeframe: from date of randomization/start of treatment until first documented response confirmed 4 weeks later( at least 3 months)

Interventionpercentage of participants (Number)
Stratum 1: Everolimus 10 mg9.6

[back to top]

Objective Response Rate: Percentage of Participants With Best Over All Response of Complete Response or Partial Response by Central Radiology Review (Stratum 2) Based on Response Evaluation Criteria in Solid Tumors (RECIST)

Objective response rate was defined by RECIST criteria: Partial response (PR) must have ≥ 30% decrease in the sum of longest diameter of all target lesions, from the baseline sum. Complete response (CR) must have disappearance of all target and non-target lesions. For CR or PR, tumor measurements must be confirmed by 2nd assessments within 4 weeks. Progression = 20% increase in the sum of longest diameter of all target lesions, from smallest sum of longest diameter of all target lesions recorded at or after baseline; or a new lesion; or progression of non-target lesions. (NCT00363051)
Timeframe: from date of randomization/start of treatment until first documented response confirmed 4 weeks later (at least 3 months)

Interventionpercentage of participants (Number)
Stratum 2: Everolimus 10 mg + Octreotide Depot4.4

[back to top]

Time to Overall Survival (OS) (Stratum 2)

"Overall survival measures the time of survival , with any response or disease progression, until death. The OS is defined as the time from date of start of treatment to date of death due to any cause.~If a patient is not known to have died, survival was censored at the date of last contact. In each treatment stratum, the Kaplan-Meier estimate of the overall survival function was constructed." (NCT00363051)
Timeframe: from randomisation to dates of disease progression, death from any cause, reported between day of first patient randomised, 26 June 2006, until cut-off date 13 April 2012

Interventionmonths (Median)
Stratum 2: Everolimus 10 mg + Octreotide Depot38.77

[back to top]

Time to Progression Free Survival (PFS) Per Central Radiology Review (Stratum 2)

"Progression free survival (PFS) is defined as the time from randomization to the date of first documented disease progression or death from any cause. The Kaplan-Meier estimate of the PFS survival function was constructed.~Median PFS was obtained and displayed along with 95% confidence intervals." (NCT00363051)
Timeframe: from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 26 June 2006, until cut-off date 13 September 2010

InterventionMonths (Median)
Stratum 2: Everolimus 10 mg + Octreotide Depot16.69

[back to top]

Time to Overall Survival (OS)(Stratum 1)

"Overall survival measures the time of survival , with any response or disease progression, until death. The OS is defined as the time from date of start of treatment to date of death due to any cause.~If a patient is not known to have died, survival was censored at the date of last contact. In each treatment stratum, the Kaplan-Meier estimate of the overall survival function was constructed." (NCT00363051)
Timeframe: from randomisation to dates of disease progression, death from any cause, reported between day of first patient randomised, 26 June 2006, until cut-off date 13 April 2012

Interventionmonths (Median)
Stratum 1: Everolimus 10 mg28.78

[back to top]

Time to Progression Free Survival (PFS) Per Central Radiology Review (Stratum 1)

"Progression free survival (PFS) is defined as the time from randomization to the date of first documented disease progression or death from any cause. The Kaplan-Meier estimate of the PFS survival function was constructed.~Median PFS was obtained and displayed along with 95% confidence intervals." (NCT00363051)
Timeframe: from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 26 June 2006, until cut-off date 13 September 2010

InterventionMonths (Median)
Stratum 1: Everolimus 10 mg9.69

[back to top]

Effect of Octreotide Depot on the Trough Concentrations of Everolimus

The effect of Octreotide Depot on the trough concentrations of everolimus was assessed at Cycle 1 Day 15. (NCT00363051)
Timeframe: Cycle 1 Day 1, Cycle 2 Day 1

Interventionng/ml (Mean)
Cycle 1 Day 1 (pre-treatment baseline) (n=37)Cycle 2 Day 1 (n= 38)
Stratum 2: Everolimus 10 mg + Octreotide Depot3.23.7

[back to top]

Number of Participants With Adverse Events (AEs), Death, Serious Adverse Events (SAEs) [Stratum 2]

Adverse events are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00363051)
Timeframe: on or after the day of the first intake of study treatment to starting no later than 28 days after study treatment discontinuation, at least every month

InterventionParticipants (Number)
Adverse EventsDeathSerious Adverse Events
Stratum 2: Everolimus 10 mg + Octreotide Depot45227

[back to top]

Number of Participants With Adverse Events (AEs), Death, Serious Adverse Events (SAEs)[Stratum 1]

Adverse events are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00363051)
Timeframe: on or after the day of the first intake of study treatment to starting no later than 28 days after study treatment discontinuation, at least every month

InterventionParticipants (Number)
Adverse EventsDeathSerious Adverse Events
Stratum 1: Everolimus 10 mg1151063

[back to top]

Duration of Overall Response (Stratum 1) Based on Response Evaluation Criteria in Solid Tumors (RECIST)- Central Radiology Review

"Duration of overall response applies only to patients whose best overall response was complete response (CR) or partial response (PR):~Complete Response (CR) = at least two determinations of CR at least 4 weeks apart before progression.~Partial response (PR) = at least two determinations of PR or better at least 4 weeks apart before progression.~Progression = 20% increase in the sum of the longest diameter of all target lesions, from the smallest sum of longest diameter of all target lesions recorded at or after baseline; or a new lesion; or progression of non-target lesions" (NCT00363051)
Timeframe: from date of first documented confirmed response to time to progression, at least 3 months

InterventionMonths (Median)
Stratum 1: Everolimus 10 mg10.64

[back to top]

Number of Participants With Incidence of Biopsy-proven Acute Rejection (BPAR)

Biopsy-proven acute rejection (BPAR) was defined as a clinically suspected acute rejection confirmed by biopsy (performed by the local pathologist). For all clinically suspected rejection episodes a graft core biopsy must have been performed before or within a 24 hour period from the initiation of anti-rejection therapy. (NCT00369161)
Timeframe: from Month 4 through to Month 12

Interventionparticipants (Number)
Very Low Dose Tacrolimus2
Low Dose Tacrolimus1

[back to top]

Renal Function Assessed by Calculated Glomerular Filtration Rate (cGFR)

"Renal function was assessed by calculated glomerular filtration rate (cGFR) using Modification of Diet in Renal Disease (MDRD)formula.~GFR [mL/min/1.73m^2] = 186.3*(C-1.154)*(A-0.203)*G*R, where:~C is the serum concentration of creatinine [mg/dL],~A is patient age at sample collection date [years],~G=0.742 when gender is female, otherwise G=1,~R=1.21 when race is black, otherwise R=1" (NCT00369161)
Timeframe: 12 months post -transplant

InterventionmL/min/1.73m^2 (Mean)
Very Low Dose Tacrolimus57.07
Low Dose Tacrolimus51.73

[back to top]

Percentage of Participants With Efficacy Failure

Efficacy failure was a composite of BPAR, graft loss, death or lost to follow-up. BPAR was defined as a clinically suspected acute rejection confirmed by biopsy (performed by the local pathologist). For all clinically suspected rejection episodes a graft core biopsy must have been performed before or within a 24 hour period from the initiation of anti-rejection therapy. An allograft was presumed to be lost on the day a patient started dialysis and was unable to subsequently be removed from dialysis. If the patient underwent a graft nephrectomy, the day of nephrectomy was the day of graft loss. (NCT00369161)
Timeframe: Month 12

,
Interventionpercentage of participants (Number)
Efficacy failure (Composite)BPARGraft loss or deathGraft LossDeathLost To Follow-up
Low Dose Tacrolimus4.31.12.21.11.11.1
Very Low Dose Tacrolimus6.72.74.01.32.70.0

[back to top]

Number of Participants With Wound Problems(12 Months Analysis)

Patients with any wound healing problem such as infection related to kidney surgery, dehiscence, lymphocele, hernia, seroma, hematoma, ureteral anastomotic complication and other were reported in this analysis. (NCT00371826)
Timeframe: At Month 12

,,
InterventionParticipants (Number)
Any wound healing problemInfection related to kidney surgeryDehiscenceLymphoceleHerniaSeromaHematomaUreteral anastomotic complicationOther
Calcineurin Inhibitor (CNI) Withdrawal1623334423
CNI+MPA+ Steroid1542429011
Steroid Withdrawal932313210

[back to top]

Number of Participants With Sub Clinical Acute Rejection (36 Months Analysis)

"Based on Banff 97 criteria, sub clinical acute rejection can be:~GRADE IA - Cases with significant interstitial infiltration (>25% of parenchyma affected) and foci of moderate tubulitis (>4 mononuclear cells/tubular cross section or group of 10 tubular cells).~GRADE IB - Cases with significant interstitial infiltration (>25% of parenchyma affected) and foci of moderate tubulitis (>10 mononuclear cells/tubular cross section or group of 10 tubular cells).~GRADE IIA - Cases with significant interstitial infiltration and mild to moderate intimal arteritis (v1).~GRADE IIB - Cases with moderate to severe intimal arteritis comprising >25% of the luminal area (v2).~GRADE III - Cases with transmural arteritis or fibrinoid change and necrosis of medial smooth muscle cells (v3).~Borderline' category is used when no intimal arteritis is present, but there are foci of mild tubulitis (1 to 4 mononuclear cells/tubular cross section)." (NCT00371826)
Timeframe: At Month 36

,,
InterventionParticipants (Number)
Month 36: NOMonth 36: BorderlineMonth 36: Grade IAMonth 36: Grade IBMonth 36: Grade IIAMonth 36: Grade IIBMonth 36: Grade IIIMonth 36: Not Done
Calcineurin Inhibitor (CNI) Withdrawal91000008
CNI+MPA+ Steroid1511000014
Steroid Withdrawal20000001

[back to top]

Number of Participants With Sub Clinical Acute Rejection (12 Months Analysis)

"Based on Banff 97 criteria, sub clinical acute rejection can be:~GRADE IA - Cases with significant interstitial infiltration (>25% of parenchyma affected) and foci of moderate tubulitis (>4 mononuclear cells/tubular cross section or group of 10 tubular cells).~GRADE IB - Cases with significant interstitial infiltration (>25% of parenchyma affected) and foci of moderate tubulitis (>10 mononuclear cells/tubular cross section or group of 10 tubular cells).~GRADE IIA - Cases with significant interstitial infiltration and mild to moderate intimal arteritis (v1).~GRADE IIB - Cases with moderate to severe intimal arteritis comprising >25% of the luminal area (v2).~GRADE III - Cases with transmural arteritis or fibrinoid change and necrosis of medial smooth muscle cells (v3).~Borderline category is used when no intimal arteritis is present, but there are foci of mild tubulitis (1 to 4 mononuclear cells/tubular cross section)." (NCT00371826)
Timeframe: At Month 12

,,
InterventionParticipants (Number)
NOBorderlineGrade IAGrade IBGrade IIAGrade IIBGrade IIINot Done
Calcineurin Inhibitor (CNI) Withdrawal202100007
CNI+MPA+ Steroid292010008
Steroid Withdrawal52000003

[back to top]

Number of Participants With Post Transplant Diabetes Mellitus (PTDM) and Impaired Fasting Glucose (12 Months Analysis)

"The symptoms of post transplant diabetes mellitus (PTDM) and impaired fasting glucose are defined as any of the following conditions:~Patients receiving glucose lowering treatment~Fasting plasma glucose (FPG) >= 126 mg/dL on 2 separate occasions~Hemoglobin subtype A1c (HbA1c) > 6.5%~Diabetes reported as treatment emergent AE with end date > Day 15" (NCT00371826)
Timeframe: At Month 12

,,
InterventionParticipants (Number)
Glucose lowering treatmentFPG >= 126 mg/dL on 2 separate occasionsHbA1c > 6.5%Diabetes as treatment emergent AEAny of the above symptoms
Calcineurin Inhibitor (CNI) Withdrawal10612819
CNI+MPA+ Steroid903210
Steroid Withdrawal55538

[back to top]

Number of Participants With Notable Abnormalities in Total Cholesterol and Triglycerides as Measurement of Effect of Treatment on Cardiovascular Health (36 Months Analysis)

"Notable abnormal total cholesterol is defined as : High: >= 9.1 mmol/L , normal range is 0.00 - 5.17 mmol/L~Notable abnormal triglycerides is defined as : High: >= 8.5 mmol/L, normal range is 0.30 - 2.00 mmol/L" (NCT00371826)
Timeframe: Overall Post Baseline up to month 36

,,
InterventionParticipants (Number)
Total Cholesterol: HighTriglycerides : High
Calcineurin Inhibitor (CNI) Withdrawal60
CNI+MPA+ Steroid01
Steroid Withdrawal10

[back to top]

Number of Participants With Notable Abnormalities in Total Cholesterol and Triglycerides as Measurement of Effect of Treatment on Cardiovascular Health (12 Months Analysis)

"Notable abnormal total cholesterol is defined as : High: >= 9.1 mmol/L , normal range is 0.00 - 5.17 mmol/L~Notable abnormal triglycerides is defined as : High: >= 8.5 mmol/L, normal range is 0.30 - 2.00 mmol/L" (NCT00371826)
Timeframe: Overall post baseline up to month 12

,,
InterventionParticipants (Number)
Total Cholesterol: High (n = 48, 47,30)Triglycerides : High (n= 48, 46, 30)
Calcineurin Inhibitor (CNI) Withdrawal61
CNI+MPA+ Steroid00
Steroid Withdrawal30

[back to top]

Number of Participants With Notable Abnormal Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP) as Measurement of Effect of Treatment on Cardiovascular Health (36 Months Analysis)

"Notable abnormal systolic blood pressure is defined as :~Either an increase of >=30 that results in >=180 or >200 (mm/Hg)~OR a decrease of >=30 that results in <=90 or <75 (mm/Hg) from baseline~Notable abnormal diastolic blood pressure is defined as :~Either an increase of >=20 that results in >=105 or >115 (mm/Hg)~OR a decrease of >=20 that results in <=50 or <40 (mm/Hg) from baseline" (NCT00371826)
Timeframe: Baseline, Overall post baseline up to Month 36

,,
InterventionParticipants (Number)
SBP: >=180 mm/Hg or 200 mm/HgSBP: < = 90 mm/Hg or < 200 mm/HgDBP : >=105 mm/Hg or >115 mm/HgDBP: <=50 mm/Hg or <40 mm/Hg
Calcineurin Inhibitor (CNI) Withdrawal3121
CNI+MPA+ Steroid4054
Steroid Withdrawal1010

[back to top]

Number of Participants With Notable Abnormal Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP) as Measurement of Effect of Treatment on Cardiovascular Health (12 Months Analysis)

"Notable abnormal systolic blood pressure is defined as :~Either an increase of >=30 that results in >=180 or >200 (mm/Hg)~OR a decrease of >=30 that results in <=90 or <75 (mm/Hg)from baseline~Notable abnormal diastolic blood pressure is defined as :~Either an increase of >=20 that results in >=105 or >115 (mm/Hg)~OR a decrease of >=20 that results in <=50 or <40 (mm/Hg) from baseline" (NCT00371826)
Timeframe: Baseline, Overall post-baseline up to 12 month

,,
InterventionParticipants (Number)
SBP: >=180 mm/Hg or 200 mm/HgSBP: < = 90 mm/Hg or < 200 mm/HgDBP : >=105 mm/Hg or >115 mm/HgDBP: <=50 mm/Hg or <40 mm/Hg
Calcineurin Inhibitor (CNI) Withdrawal5131
CNI+MPA+ Steroid5064
Steroid Withdrawal7050

[back to top]

Number of Participants With New Onset Diabetes Mellitus After Transplantation (NODAT) and Impaired Fasting Glucose (36 Months Analysis)

"The symptoms of new onset diabetes mellitus after transplantation (NODAT) and impaired fasting glucose are defined as any of the following conditions:~Patients receiving glucose lowering treatment~2 fasting plasma glucose (FPG) values >= 126 mg/dL or 2 random plasma glucose (RPG) values >= 200 mg/dL or FPG value >= 126 mg/dL and 1 RPG value >= 200 mg/dL~Diabetes reported as treatment emergent AE with end date > Day 15" (NCT00371826)
Timeframe: At Month 36

,,
InterventionParticipants (Number)
Glucose lowering treatmentFPG or RPGDiabetes as treatment emergent AEAny of the above symptoms
Calcineurin Inhibitor (CNI) Withdrawal5356
CNI+MPA+ Steroid102411
Steroid Withdrawal2122

[back to top]

Number of Participants With Histological Evidence Chronic Allograft Nephropathy (CAN) (36 Months Analysis)

"Chronic rejection is characterized by a slow progressive decline in renal function and is typically preceded by the histological picture of chronic allograft nephropathy. The presence of biopsy confirmed Grade I, II or III chronic allograft nephropathy by Banff 97 criteria was assessed on all optional biopsies obtained for clinical suspicion of chronic rejection.~Data summarized by 3 categories. Yes - Patients with histological evidence of CAN ; No - Patients with histological evidence of CAN and Not Done - Central protocol defined kidney allograft biopsies were not done." (NCT00371826)
Timeframe: At Month 36

,,
InterventionParticipants (Number)
YESNONot Done
Calcineurin Inhibitor (CNI) Withdrawal288
CNI+MPA+ Steroid11614
Steroid Withdrawal111

[back to top]

Number of Participants With Histological Evidence Chronic Allograft Nephropathy (CAN) (12 Months Analysis)

"A per-protocol biopsy was performed at Baseline and Month 12 and read by an independent blinded pathologist in order to assess chronic allograft nephropathy. Chronic rejection is characterized by a slow progressive decline in renal function and is typically preceded by the histological picture of chronic allograft nephropathy. The presence of biopsy confirmed Grade I, II or III chronic allograft nephropathy by Banff 97 criteria was assessed on all optional biopsies obtained for clinical suspicion of chronic rejection.~Data summarized by 3 categories. Yes - Patients with histological evidence of CAN ; No - Patients with histological evidence of CAN and Not Done - Central protocol defined kidney allograft biopsies were not done." (NCT00371826)
Timeframe: At Month 12

,,
InterventionParticipants (Number)
YESNONot Done
Calcineurin Inhibitor (CNI) Withdrawal8157
CNI+MPA+ Steroid6268
Steroid Withdrawal253

[back to top]

Number of Participants With Employment Status (36 Months Analysis)

"The various employment status reported are:~Employed/self employed full time~Employed part time~Unemployed~Homemaker~Volunteer~Permanently disabled~Non-permanently disable~Retired~Other" (NCT00371826)
Timeframe: At screening (at day 0 +/- 7 days ), At Month 36

,,
InterventionParticipants (Number)
Screening visit: Employed/self employed full timeMonth 36: Employed/self employed full timeScreening visit: Employed part timeMonth 36: Employed part timeScreening visit: UnemployedMonth 36: UnemployedScreening visit: HomemakerMonth 36 : HomemakerScreening visit: Permanently disabledMonth 36: Permanently disabledScreening visit: Non-permanently disabledMonth 36: Non-permanently disabledScreening visit: RetiredMonth 36: RetiredScreening visit: OtherMonth 36: Other
Calcineurin Inhibitor (CNI) Withdrawal5443533300000011
CNI+MPA+ Steroid101074763311112200
Steroid Withdrawal1100101100000000

[back to top]

Number of Participants With Employment Status (12 Months Analysis)

"The various employment status reported are:~Employed/self employed full time~Employed part time~Unemployed~Homemaker~Volunteer~Permanently disabled~Non-permanently disable~Retired~Other" (NCT00371826)
Timeframe: At screening (at day 0 +/- 7 days ), At Month 12

,,
InterventionParticipants (Number)
Screening visit: Employed/self employed full timeMonth 12: Employed/self employed full timeScreening visit: Employed part timeMonth 12: Employed part timeScreening visit: UnemployedMonth 12: UnemployedScreening visit: HomemakerMonth 12 : HomemakerScreening visit: VolunteerMonth 12: VolunteerScreening visit: Permanently disabledMonth 12: Permanently disabledScreening visit: Non-permanently disabledMonth 12: Non-permanently disabledScreening visit: RetiredMonth 12: RetiredScreening visit: OtherMonth 12: Other
Calcineurin Inhibitor (CNI) Withdrawal18693106321121114111
CNI+MPA+ Steroid21147581550022202200
Steroid Withdrawal9240106520000001010

[back to top]

Number of Participants With Composite Endpoint of Treatment Failure (36 Months Analysis)

"Composite endpoint of treatment failure includes biopsy-proven acute rejection (BPAR), graft loss, death and loss-to-follow-up. A BPAR is defined as a biopsy graded IA, IB, IIA, IIB, or III as per Banff 97 classification.~The allograft was presumed to be lost on the day the patient started dialysis and was not able to subsequently be removed from dialysis. If the patient underwent a graft nephrectomy, then the day of nephrectomy was the day of graft loss." (NCT00371826)
Timeframe: At Month 12, 24 and 36

,,
InterventionParticipants (Number)
At Month 12At Month 24At Month 36
Calcineurin Inhibitor (CNI) Withdrawal467
CNI+MPA+ Steroid4610
Steroid Withdrawal000

[back to top]

Number of Participants With Biopsy Proven Acute Rejection (BPAR) Per Treatment Group (36 Months Analysis)

A biopsy-proven acute rejection is defined as a biopsy graded IA, IB, IIA, IIB, or III as per Banff 97 classification. (NCT00371826)
Timeframe: At Month 12, 24 and 36

,,
InterventionParticipants (Number)
At Month 12At Month 24At Month 36
Calcineurin Inhibitor (CNI) Withdrawal456
CNI+MPA+ Steroid458
Steroid Withdrawal000

[back to top]

Number of Participants With Biopsy Proven Acute Rejection (BPAR) Influenced by Demographic Characteristics and Morbidities (36 Months Analysis)

The influence of demographic characteristics and comorbidities on incidence of BPAR were analyzed in the following way: Demographic characteristics were age (<55 years, ≥55 years), Expanded Criteria Donor [ECD] organ (donor age >60 years or donor non heart-beating and donor age >50), gender, living vs. deceased donor, Body Mass Index (BMI) classes (underweight <18.5, normal 18.5 - <25.0, overweight 25.0 - <30.0, obesity 30.0 and above), years on dialysis before transplantation (<1, 1-5, >5 years). Comorbidities were diabetes, hypertension, cardiovascular diseases/events, nephrosclerosis, glomerulonephritis/glomerular disease, polycystic disease, and Cytomegalovirus status. (NCT00371826)
Timeframe: At Month 36

,,
InterventionParticipants (Number)
Age < 55 yearsAge > = 55 yearsECD OrganNO ECD OrganMaleFemaleLiving donorDeceased donorBMI < 18.5BMI 18.5 - <25BMI 25 - <30BMI >= 30Years on dialysis before transplantation: None< 1 Year on dialysis before transplantation1 - 5 Years on dialysis before transplantation> 5 Years on dialysis before transplantationDiabetes mellitus: NoDiabetes mellitus: YesHypertension: NoHypertension: YesCardiovascular disease : NoCardiovascular disease: YesNephrosclerosis: NoNephrosclerosis: YesGlomerulonephritis/glomerular disease : NoGlomerulonephritis/glomerular disease: YesCytomegalovirus : NegativeCytomegalovirus : Positive
Calcineurin Inhibitor (CNI) Withdrawal6006425111400321331533604215
CNI+MPA+ Steroid7108716203411421621753717144
Steroid Withdrawal0000000000000000000000000000

[back to top]

Number of Participants With Biopsy Proven Acute Rejection (BPAR) Influenced by Demographic Characteristics and Morbidities (12 Months Analysis)

The influence of demographic characteristics and comorbidities on incidence of BPAR were analyzed in the following way: Demographic characteristics were age (<55 years, ≥55 years), Expanded criteria Donor (ECD) organ (donor age >60 years or donor non heart-beating and donor age >50), gender, living vs. deceased donor, Body Mass Index (BMI) classes (underweight <18.5, normal 18.5 - <25.0, overweight 25.0 - <30.0, obesity 30.0 and above), years on dialysis before transplantation (<1, 1-5, >5 years). Comorbidities were diabetes, hypertension, cardiovascular diseases/events, nephrosclerosis, glomerulonephritis/glomerular disease, polycystic disease, and Cytomegalovirus status. (NCT00371826)
Timeframe: At Month 12

,,
InterventionParticipants (Number)
Age < 55 yearsAge > = 55 yearsECD OrganNO ECD OrganMaleFemaleLiving donorDeceased donorBMI < 18.5BMI 18.5 - <25BMI 25 - <30BMI >= 30Years on dialysis before transplantation: None< 1 Year on dialysis before transplantation1 - 5 Years on dialysis before transplantation> 5 Years on dialysis before transplantationDiabetes mellitus: NoDiabetes mellitus: YesHypertension: NoHypertension: YesCardiovascular disease : NoCardiovascular disease: YesNephrosclerosis: NoNephrosclerosis: YesGlomerulonephritis/glomerular disease : NoGlomerulonephritis/glomerular disease: YesCytomegalovirus : NegativeCytomegalovirus : Positive
Calcineurin Inhibitor (CNI) Withdrawal114312114960582138310521341115087213
CNI+MPA+ Steroid5106606012300312421533605124
Steroid Withdrawal4123412303110122410523413205

[back to top]

Number of Participants With Any Wound Problems (36 Months Analysis)

Patients with any wound healing problem such as infection related to kidney surgery, dehiscence, lymphocele, hernia, seroma, hematoma, ureteral anastomotic complication and other were reported in this analysis. (NCT00371826)
Timeframe: At Month 12, 24 and 36

,,
InterventionParticipants (Number)
At Month 12At Month 24At Month 36
Calcineurin Inhibitor (CNI) Withdrawal6910
CNI+MPA+ Steroid111313
Steroid Withdrawal222

[back to top]

Number of Participants With Antibody-mediated Rejection Per Treatment Group (36 Months Analysis)

(NCT00371826)
Timeframe: At Month 12, 24 and 36

,,
InterventionParticipants (Number)
At Month 12At Month 24At Month 36
Calcineurin Inhibitor (CNI) Withdrawal111
CNI+MPA+ Steroid002
Steroid Withdrawal000

[back to top]

Mean Urine Albumin/Creatinine Ratio [ACR] as Measurement of Proteinuria (36 Months Analysis)

Proteinuria is measured by spot morning urine Albumin/Creatinine Ratio [ACR]. When the ACR is more than or equal to 30 mg/mmol then it is known as proteinuria. (NCT00371826)
Timeframe: At Month 12, 18, 24 and 36

,,
Interventionmg/mmol (Mean)
At 12 Month (n = 18, 32, 2)At 18 Month (n= 17, 31, 2)At 24 Month ( n= 16, 29, 1)At 36 Month ( n= 15, 25, 0)
Calcineurin Inhibitor (CNI) Withdrawal23.528.230.030.6
CNI+MPA+ Steroid8.18.16.623.4
Steroid Withdrawal4.85.37.0NA

[back to top]

Mean Short-form 36 Health Survey (SF-36) Score as a Measure of Quality of Life Assessment (36 Months Analysis)

"SF-36 measures impact of disease on overall quality of life (QoL). 36-item survey has 8 subscales. The 8 subscales are : Physical functioning (PF), Role-physical (RP), Bodily pain (BP), General health (GH), Vitality (VT), Social functioning (SF), Role-emotional (RE) and Mental health (MH).~Score for each sub-scale has been standardized with the use of norm-based methods based on assessment of the general U.S. population free of chronic conditions. Scores range from 1-100 with a mean=50 and a standard deviation=10. A higher score indicates less impact on QoL." (NCT00371826)
Timeframe: At Month 24

,,
Interventionunits on a scale (Mean)
Physical functioning (PF)Role-physical (RP)Bodily pain (BP)General health (GH)Vitality (VT)Social Functioning (SF)Role-emotional (RE)Mental health (MH)
Calcineurin Inhibitor (CNI) Withdrawal84.375.081.671.670.085.972.977.5
CNI+MPA+ Steroid87.677.485.069.872.780.683.977.9
Steroid Withdrawal90.0100.084.067.060.068.833.362.0

[back to top]

Mean Short-form 36 Health Survey (SF-36) Score as a Measure of Quality of Life Assessment (12 Months Analysis)

"SF-36 measures impact of disease on overall quality of life (QoL). 36-item survey has 8 subscales. The 8 subscales are: Physical functioning (PF), Role-physical (RP), Bodily pain (BP), General health (GH), Vitality (VT), Social functioning (SF), Role-emotional (RE) and Mental health (MH).~Score for eash sub-scale has been standardized with the use of norm-based methods based on assessment of the general U.S. population free of chronic conditions. Scores range from 1-100 with a mean=50 and a standard deviation=10. A higher score indicates less impact on QoL." (NCT00371826)
Timeframe: At Month 12

,,
Interventionunits on a scale (Mean)
Physical functioning (PF)Role-physical (RP)Bodily pain (BP)General health (GH)Vitality (VT)Social Functioning (SF)Role-emotional (RE)Mental health (MH)
Calcineurin Inhibitor (CNI) Withdrawal78.772.973.667.162.774.572.273.3
CNI+MPA+ Steroid76.978.281.571.272.481.181.179.1
Steroid Withdrawal82.0100.096.886.885.097.5100.092.8

[back to top]

Mean Serum Creatinine (36 Months Analysis)

(NCT00371826)
Timeframe: At Month 12, 18, 24 and 36

,,
Interventionumol/L (Mean)
At Month 12 (n= 23, 39, 4)At Month 18 (n= 22, 36, 4)At Month 24 (n = 23, 36, 4)At Month 36 (n= 19, 35, 3)
Calcineurin Inhibitor (CNI) Withdrawal112.7113.6119.9119.4
CNI+MPA+ Steroid123.0121.4123.7131.8
Steroid Withdrawal146.8146.3146.5176.0

[back to top]

Creatinine Clearance Calculated by the Cockcroft-Gault Formula (36 Months Analysis)

"Creatinine clearance were calculated according to the Cockcroft-Gault formula:~CrCl (males) = (140-A) × BW/(72 × Cr) CrCl (females) = CrCl (males) × 0.85 where A is age [years], BW is body weight [kg], and Cr is the serum concentration of creatinine [mg/dL].~The Cockcroft-Gault formula estimates creatinine clearance based on serum creatinine level, body weight, and age." (NCT00371826)
Timeframe: At Month 12, 24 and 36

,,
InterventionmL/min (Mean)
At Month 12 (n= 23, 39, 4)At Month 18 (n=22, 36, 4)At Month 24 (n= 23, 36, 4)At Month 36 (n= 23, 39, 4)
Calcineurin Inhibitor (CNI) Withdrawal69.869.965.666.3
CNI+MPA+ Steroid71.773.772.867.5
Steroid Withdrawal63.766.263.762.7

[back to top]

Change in Bone Mineral Density Between Week 2 and Month 24 (36 Months Analysis)

Measurements of bone mineral density (BMD) by Dual Energy X-ray Absorptiometry (DEXA) were done at Week 2 and Month 24. Change in BMD between week 2 and Month 24 were done for neck of femur and lumbar spine. (NCT00371826)
Timeframe: Week 2, Month 24

,,
Interventiong/cm^2 (Mean)
Neck of Femur (Month 24- Week 2)Lumbar Spine (Month 24 - Week 2)
Calcineurin Inhibitor (CNI) Withdrawal0.2-0.0
CNI+MPA+ Steroid-0.1-0.1
Steroid Withdrawal-0.1-0.0

[back to top]

Calculated Glomerular Filtration Rate (cGFR) After Kidney Transplant to Evaluate Kidney Function (36 Months Analysis)

The glomerular filtration rate (GFR) was calculated by the Nankivell formula: GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)^ 2 + C where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kg, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. (NCT00371826)
Timeframe: At Month 24 and 36

,,
InterventionmL/min per 1.73 m^2 (Mean)
Month 24 (n= 23, 36, 4)Month 36 (n= 19, 35, 3)
Calcineurin Inhibitor (CNI) Withdrawal69.571.6
CNI+MPA+ Steroid71.869.1
Steroid Withdrawal67.061.0

[back to top]

Number of Participants With Erythropoietin Usage (36 Months Analysis)

(NCT00371826)
Timeframe: Month 36

Interventionparticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal5
CNI+MPA+ Steroid7
Steroid Withdrawal2

[back to top]

Number of Participants With Erythropoietin Usage (12 Months Analysis)

(NCT00371826)
Timeframe: Month 12

Interventionparticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal20
CNI+MPA+ Steroid10
Steroid Withdrawal8

[back to top]

Number of Participants With Composite Endpoint of Treatment Failure (12 Months Analysis)

"Composite endpoint of treatment failure includes biopsy-proven acute rejection (BPAR), graft loss, death and loss-to-follow-up. A BPAR is defined as a biopsy graded IA, IB, IIA, IIB, or III as per Banff 97 classification.~The allograft was presumed to be lost on the day the patient started dialysis and was not able to subsequently be removed from dialysis. If the patient underwent a graft nephrectomy, then the day of nephrectomy was the day of graft loss." (NCT00371826)
Timeframe: Month 12

InterventionParticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal16
CNI+MPA+ Steroid8
Steroid Withdrawal11

[back to top]

Number of Participants With Biopsy Proven Acute Rejection (BPAR) Per Treatment Group (12 Months Analysis)

A biopsy-proven acute rejection is defined as a biopsy graded IA, IB, IIA, IIB, or III as per Banff 97 classification. (NCT00371826)
Timeframe: At Month 12

InterventionParticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal15
CNI+MPA+ Steroid6
Steroid Withdrawal5

[back to top]

Number of Participants With Antibody-mediated Rejection Per Treatment Group (12 Months Analysis)

(NCT00371826)
Timeframe: At Month 12

InterventionParticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal5
CNI+MPA+ Steroid2
Steroid Withdrawal2

[back to top]

Number of Participants Hospitalized for Reasons Other Than Primary Transplantation (36 Months Analysis)

This analysis is reporting number of participants hospitalized for reasons (such as acute rejection, infection, gastrointestinal (GI) events, cardiovascular event, metabolic disorder and Other) other than primary transplantation. (NCT00371826)
Timeframe: Month 36

InterventionParticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal16
CNI+MPA+ Steroid30
Steroid Withdrawal1

[back to top]

Number of Participants Hospitalized for Reasons Other Than Primary Transplantation (12 Months Analysis)

This analysis is reporting number of participants hospitalized for reasons (such as acute rejection, infection, gastrointestinal (GI) events, cardiovascular event, metabolic disorder and Other) other than primary transplantation. (NCT00371826)
Timeframe: Month 12

InterventionParticipants (Number)
Calcineurin Inhibitor (CNI) Withdrawal34
CNI+MPA+ Steroid31
Steroid Withdrawal13

[back to top]

Mean Urine Albumin/Creatinine Ratio (ACR) as Measurement of Proteinuria (12 Months Analysis)

Proteinuria is measured by spot morning urine Albumin/Creatinine Ratio [ACR]. When the ACR is more than or equal to 30 mg/mmol then it is known as proteinuria. (NCT00371826)
Timeframe: At Month 12

Interventionmg/mg (Mean)
Calcineurin Inhibitor (CNI) Withdrawal0.2
CNI+MPA+ Steroid0.1
Steroid Withdrawal0.1

[back to top]

Mean Serum Creatinine (12 Months Analysis)

(NCT00371826)
Timeframe: At Month 12

Interventionumol/L (Mean)
Calcineurin Inhibitor (CNI) Withdrawal118.9
CNI+MPA+ Steroid161.0
Steroid Withdrawal148.6

[back to top]

Creatinine Clearance (CrCl) Calculated by the Cockcroft-Gault Formula (12 Months Analysis)

"Creatinine clearance were calculated according to the Cockcroft-Gault formula:~CrCl (males) = (140-A) × BW/(72 × Cr) CrCl (females) = CrCl (males) × 0.85 where A is age [years], BW is body weight [kg], and Cr is the serum concentration of creatinine [mg/dL].~The Cockcroft-Gault formula estimates creatinine clearance based on serum creatinine level, body weight, and age." (NCT00371826)
Timeframe: At Month 12

InterventionmL/min (Mean)
Calcineurin Inhibitor (CNI) Withdrawal66.1
CNI+MPA+ Steroid67.8
Steroid Withdrawal63.2

[back to top]

Calculated Glomerular Filtration Rate (cGFR) After Kidney Transplant to Evaluate Kidney Function (12 Months Analysis)

The glomerular filtration rate (GFR) was calculated by the Nankivell formula: GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)^ 2 + C where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kg, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. (NCT00371826)
Timeframe: At Month 12

InterventionmL/min per 1.73 m^2 (Mean)
Calcineurin Inhibitor (CNI) Withdrawal65.2
CNI+MPA+ Steroid69.3
Steroid Withdrawal66.9

[back to top]

Number of Patient Survival and Graft Survival (36 Months Analysis)

(NCT00371826)
Timeframe: At Month 12, 24 and 36

,,
InterventionParticipants (Number)
Month 12: Patient SurvivalMonth 12: Graft SurvivalMonth 24: Patient SurvivalMonth 24: Graft SurvivalMonth 36: Patient SurvivalMonth 36: Graft Survival
Calcineurin Inhibitor (CNI) Withdrawal232323232323
CNI+MPA+ Steroid393939393939
Steroid Withdrawal444444

[back to top]

Number of Patient Survival and Graft Survival (12 Months Analysis)

(NCT00371826)
Timeframe: At Month 12

,,
InterventionParticipants (Number)
Patient SurvivalGraft Survival
Calcineurin Inhibitor (CNI) Withdrawal4949
CNI+MPA+ Steroid4645
Steroid Withdrawal3030

[back to top]

Progression-free Survival

(NCT00374140)
Timeframe: From entry into trial to up to 60 months

Interventionmonths (Median)
RAD001 (Everolimus)1.3

[back to top]

Overall Survival

(NCT00374140)
Timeframe: From entry in trial to up to 60 months

Interventionmonths (Median)
RAD001 (Everolimus)6.7

[back to top]

Objective Response Rate

Number of patients for which response to treatment was observed / total number of patients. (NCT00374140)
Timeframe: From beginning of treatment up to 60 months

Interventionpercentage of participants (Number)
RAD001 (Everolimus)3

[back to top]

Determine the Proportion of Previously Treated Small Cell Lung Cancer (SCLC) Patients Whose Disease Has Not Progressed Following 6-weeks (2 Cycles) of Treatment With RAD001.

(NCT00374140)
Timeframe: Two cycles of treatment with RAD001 (~6 weeks)

Interventionpercentage of participants (Number)
RAD001 (Everolimus)26

[back to top]

Number of Patients With Biopsy-proven Acute Rejection From Month 12 to End of Study (Month 24)

Biopsy-proved acute rejection was defined as a treated acute rejection confirmed by biopsy, graded locally according to the International Society for Heart & Lung Transplantation (ISHLT) criteria. A treated acute rejection was defined as an acute rejection clinically suspected, whether biopsy-proven or not, which had been treated and confirmed by the investigator according to the response to therapy. (NCT00377962)
Timeframe: Month 12 to end of study (Month 24)

InterventionParticipants (Number)
Everolimus + CNI Reduction6
Control5

[back to top]

Number of Patients in Need of Dialysis From Month 12 to End of Study (Month 24)

(NCT00377962)
Timeframe: Month 12 to end of study (Month 24)

InterventionParticipants (Number)
Everolimus + CNI Reduction0
Control2

[back to top]

Mean Days of Hospitalization From Baseline to End of Study (Month 24)

(NCT00377962)
Timeframe: Baseline to end of study (Month 24)

InterventionDays (Mean)
Everolimus + CNI Reduction8.5
Control16.2

[back to top]

Change in Forced Vital Capacity (FVC) From Baseline to End of Study (Month 24) in the Lung Transplant Subgroup

Forced vital capacity (FVC) was measured by spirometry conducted according to internationally accepted standards. FVC is the volume delivered during an expiration made as forcefully and completely as possible starting from full inspiration. A positive change score indicates improved lung function. (NCT00377962)
Timeframe: Baseline to end of study (Month 24)

InterventionLiters (Mean)
Everolimus + CNI Reduction-0.2
Control-0.1

[back to top]

Change in Forced Expiratory Volume in 1 Second (FEV1) From Baseline to End of Study (Month 24) in the Lung Transplant Subgroup

Forced expiratory volume in 1 second (FEV1) was measured by spirometry conducted according to internationally accepted standards. FEV1 is the volume delivered in the first second of a forced vital capacity (FVC) maneuver. A positive change score indicates improved lung function. (NCT00377962)
Timeframe: Baseline to end of study (Month 24)

InterventionLiters (Mean)
Everolimus + CNI Reduction-0.2
Control-0.1

[back to top]

Number of Patients Who Died and Number of Patients With Graft Loss From Month 12 to End of Study (Month 24)

Number of patients not alive and number of patients with loss of their graft. (NCT00377962)
Timeframe: Month 12 to end of study (Month 24)

,
InterventionParticipants (Number)
DeathGraft Loss
Control00
Everolimus + CNI Reduction30

[back to top]

Change in Measured Glomerular Filtration Rate (mGFR) From Baseline to Month 12

Renal function was assessed by determining the measured glomerular filtration rate (mGFR) using creatinine ethylenediamine tetraacetic acid (Cr-EDTA) clearance or an equivalent method. A positive change score indicates improved renal function. (NCT00377962)
Timeframe: Baseline to Month 12

,
InterventionmL/min (Mean)
BaselineMonth 12Change from Baseline
Control48.047.5-0.5
Everolimus + CNI Reduction48.653.24.6

[back to top]

Change in Left Ventricular Function (Diameter and Thickness Parameters) From Baseline to End of Study (Month 24) in the Heart Transplant Subgroup

Left ventricular function was assessed by echocardiography which was performed according to local routine practice. Echocardiography parameters were left ventricular end diastolic diameter (LVEDD), left ventricular end systolic diameter (LVESD), interventricular septal wall thickness (IVSTd), and posterior wall thickness (PWTd). A positive change score indicates improved left ventricular function. (NCT00377962)
Timeframe: Baseline to end of study (Month 24)

,
Interventioncm (Mean)
LVEDDLVESDIVSTdPWTd
Control-0.00.1-0.1-0.1
Everolimus + CNI Reduction-0.10.1-0.4-0.5

[back to top]

Change in Left Ventricular Function (Filling and Ejection Fraction Parameters) From Baseline to End of Study (Month 24) in the Heart Transplant Subgroup

Left ventricular function was assessed by echocardiography which was performed according to local routine practice. Echocardiography parameters were filling fraction (FF) and ejection fraction (EF). A positive change score indicates improved left ventricular function. (NCT00377962)
Timeframe: Baseline to end of study (Month 24)

,
Interventionpercentage (Mean)
EFFF
Control0.10
Everolimus + CNI Reduction-0.60

[back to top]

Change in Measured Glomerular Filtration Rate (mGFR) From Baseline to End of Study (Month 24)

Renal function was assessed by determining the measured glomerular filtration rate (mGFR) using creatinine ethylenediamine tetraacetic acid (Cr-EDTA) clearance or an equivalent method. A positive change score indicates improved renal function. (NCT00377962)
Timeframe: Baseline to end of study (Month 24)

,
InterventionmL/min (Mean)
Month 0Month 24Change
Control49.146.8-2.4
Everolimus + CNI Reduction49.352.53.2

[back to top]

Number of Patients Discontinued From the Study Due to Adverse Events From Month 12 to End of Study (Month 24)

(NCT00377962)
Timeframe: Month 12 to end of study (Month 24)

,
InterventionParticipants (Number)
Total discontinued due to AE(s)Pulmonary embolismSkin problemsHypercholesterolemiaStrokeMuscular painDiarrheaEdema
Control00000000
Everolimus + CNI Reduction82111111

[back to top]

Change in Serum Creatinine From Baseline to End of Study (Month 24)

Renal function was assessed by determining serum creatinine using standard laboratory methods. A positive change score indicates improved renal function. (NCT00377962)
Timeframe: Baseline to end of study (Month 24)

,
Interventionμmol/L (Mean)
Month 0Month 24Change
Control1291323
Everolimus + CNI Reduction1261260

[back to top]

Number of Patients Who Experienced Adverse Events, Serious Adverse Events and Death

Patients with all (serious and non-serious) adverse events, serious adverse events and death were reported. (NCT00378014)
Timeframe: Month 12 to Month 59 post-baseline

,
InterventionNumber of participants (Number)
Adverse Events (serious and non-serious)Serious Adverse EventsDeaths
Calcineurin Inhibitor (CNI)40282
Everolimus41261

[back to top]

Incidence of Efficacy Failure

Efficacy failure was defined as the composite endpoint of biopsy-proven acute rejection (BPAR), graft loss, death, lost to follow-up (from any reason), whichever occurred first. Incidence of efficacy failure was estimated using crude rate estimation (relative frequency). (NCT00378014)
Timeframe: Month 11

InterventionPercentage of participants (Number)
Everolimus20.8
Calcineurin Inhibitor (CNI)20.4

[back to top]

Incidence of the Need for a Change in the Immunosuppressive Regimen

The incidence of any changes in the immunosuppressive regimen other than allowed in the study protocol (for example, introduction of Mycophenolic acid (MPA) or sirolimus) was estimated using crude rate estimation (relative frequency). (NCT00378014)
Timeframe: Month 11

InterventionPercentage of participants (Number)
Everolimus80.2
Calcineurin Inhibitor (CNI)73.5

[back to top]

Patient and Graft Survival

Patient survival was defined as the time from date of randomization to date of death from any cause. If a patient was not known to have died, patient survival was censored as the date of last contact. Graft survival was defined as the time from the date of randomization to the date of graft loss. If a patient was not known to suffer from a graft loss or died without graft loss, time to graft loss was censored with date of last contact or date of death, respectively. Patient and graft survival were analyzed using the Kaplan Meier method. (NCT00378014)
Timeframe: Month 11

,
InterventionPercentage of Participants (Number)
DeathGraft loss
Calcineurin Inhibitor (CNI)4.12.1
Everolimus4.32.2

[back to top]

Renal Function (cGFR)

This outcome measure evaluated renal function by assessing the calculated GFR based on the Cockcroft-Gault formula. (NCT00378014)
Timeframe: Month 5

InterventionmL/min (Mean)
Everolimus88.7
Calcineurin Inhibitor (CNI)82.9

[back to top]

Hepatitis C Virus (HCV) Replication in HCV-positive Patients

HCV ribonucleic acid (RNA) was measured by real time reverse transcriptase polymerase chain reaction (PCR; copies per mL). (NCT00378014)
Timeframe: Baseline, Month 5

,
InterventionPercentage of participants (Number)
Baseline, negativeBaseline, positiveBaseline, not doneMonth 5, negativeMonth 5, positiveMonth 5, not done
Calcineurin Inhibitor (CNI)55.30.044.738.213.248.7
Everolimus64.50.035.562.91.635.5

[back to top]

Number of Patients Who Experienced Adverse Events, Serious Adverse Events and Death

Patients with all (serious and non-serious) adverse events, serious adverse events and death were reported. (NCT00378014)
Timeframe: From randomization to Month 11

,
InterventionNumber of participants (Number)
Adverse Events (serious and non-serious)Serious Adverse EventsDeaths
Calcineurin Inhibitor (CNI)96584
Everolimus98674

[back to top]

Incidence of Treated BPAR

The incidence of treated BPAR was estimated using crude rate estimation (relative frequency). (NCT00378014)
Timeframe: Month 11

InterventionPercentage of Participants (Number)
Everolimus13.5
Calcineurin Inhibitor (CNI)10.2

[back to top]

Calculated Glomerular Filtration Rate (cGFR)

This outcome measure evaluated renal function by assessing the calculated GFR based on the Cockcroft-Gault formula. (NCT00378014)
Timeframe: Month 11

InterventionmL/min (Mean)
Everolimus87.9
Calcineurin Inhibitor (CNI)84.1

[back to top]

Response Rate: The Total Number of Participants With Progression of Disease

"To determine response rate and time to tumor progression of patients with colorectal cancer and mutations in the PI3KCA gene who are treated with RAD001. Response and progression will be evaluated in this study using the international criteria proposed by the Response Evaluation Criteria in Solid Tumors (RECIST) Committee.~Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions assessed by CT (or MRI): Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesion. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesion.~The outcome measure will be the total number of subjects who show progression of disease." (NCT00390364)
Timeframe: 1 month

Interventionparticipants with progression of disease (Number)
RAD00011

[back to top]

Progression-Free Survival for Patients Treated at MTD/Phase II Dose Level

Progression is Defined Using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% Increase in the Sum of the Longest Diameter of Target Lesions, or a Measurable Increase in a Non-target Lesion, or the Appearance of New Lesions. Progression-free survival was defined as the interval from the date of study entry until the date of tumor progression or death for the patients treated at the MTD/Phase II dose level. This outcome measure is reported for all patients treated at the same dose level and is not separated into Phase I and Phase II. The phase I and phase II results are not separated out as the timing of their enrollment (early in phase 1 or later phase II) is not relevant to the outcome measure. (NCT00392821)
Timeframe: 18 months

InterventionMonths (Median)
RAD001 and Sorafenib5.45

[back to top]

Overall Response Rate (ORR) of Patients Treated at MTD/Phase II Dose Level

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI or CT: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response Rate (ORR) = Percentage of Patients Who Experience an Objective Response (CR+ PR) to Treatment. In oncology, this outcome measure is reported for all patients treated at the same dose level and is not separated into Phase I and Phase II. The phase I and phase II results are not separated out as the timing of their enrollment (early in phase 1 or later phase II) is not relevant to the outcome measure. (NCT00392821)
Timeframe: 18 months

Interventionpercentage of evaluable patients (Number)
RAD001 and Sorafenib13

[back to top]

Clinical Response as Assessed Metabolically by Changes in Positron Emission Tomography (PET) Scan Between Baseline and Immediately Prior to Surgery.

All patients had baseline imaging in a fasted state with 18F-2-fluoro-2-deoxy-D-glucose fluorodeoxyglucose (18FDG)-PET scan and a repeat scan at 3 to 4 weeks later using routine clinical protocol for patient preparation, radiotracer administration and data acquisition. The repeat imaging occurred no longer than 24 hours before surgical resection. (NCT00401778)
Timeframe: Day 21

,,
Interventionpercentage of patients (Number)
Stable metabolic disease (SMD)Progressive metabolic disease (PM)
Control7822
Everolimus 10 mg5050
Everolimus 5 mg6436

[back to top]

Effects of RAD001 on the Regulation of Key Proteins Involved With the Mammalian Target of Rapamycin (mTOR) Axis in Tumor Specimens and Buccal Mucosa in Patients With Operable Non-small Cell Lung Cancer (NSCLC).

Changes in the expression of key signaling proteins in the mTOR/phosphatidylinositol 3-kinase (PI3K) pathway were determined by immunohistochemistry using previously published protocols and manufacturers' recommendations for antigen retrieval and antibody dilution along with positive and negative controls. Two investigators assessed protein expression jointly by light microscopy. The degree of expression was assessed by intensity (0, 1+, 2+, 3+) and percentage of cell staining in line with published algorithm. A derivative score (immunoscore) ranging between 0 and 300 was calculated as the product of intensity and percent cell staining. (NCT00401778)
Timeframe: 6 months

,,
Intervention% change in immunoscore (Mean)
S6pS6pS6/S6pMTORp4E-BP1p70s6kp70s6k Cytoplasmicp70s6k Nuclearpe1f4ePAKT NuclearPAKT CytoplasmicBim
Control-36.06-41.25-85.75-6.58-95.3725-19.17-3.33-50-33.33200-11.76
Everolimus 10 mg-77.03-47.21128.57-63.82-78.75-78.62305.37223.43-27.130-25-21.76
Everolimus 5 mg-13.69-61.57-10030150137.140.561352.78NANANA

[back to top]

Duration of Hospital Stay Following Surgery.

(NCT00401778)
Timeframe: 6 months

Interventiondays (Median)
Control5
Everolimus 5 or 10 mg5

[back to top]

Time to Progression:Time Period (in Months) From Study Entry Until Disease Progression, Death, or Last Date of Contact.

"Period from study entry until disease progression, death, or last date of contact.~Progressive Disease: Appearance of one or more new lesions and/or unequivocal progression of existing non-target lesions." (NCT00406276)
Timeframe: 6 months

InterventionMonths (Median)
Docetaxel/RAD0014.42

[back to top]

Number of Subjects Showing Partial Response and Stable Disease With the Combination of RAD001 and Docetaxel.

"Partial response (PR): At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum of LD.~Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for Progressive disease (PD), taking as reference the smallest sum Longest diameter(LD) since treatment started." (NCT00406276)
Timeframe: 6 weeks

Interventionparticipants (Number)
Partial responseStable diseaseDisease Progression
Docetaxel/RAD0012157

[back to top]

To Assess the Safety of RAD001 in Patients With Metastatic Pancreatic Cancer

Patients were followed for the duration of their time on treatment and 30 days after their last dose of RAD001. The number of patients with treatment-related adverse events are reported. (NCT00409292)
Timeframe: Patients were followed for the duration of their time on treatment and 30 days after their last dose of RAD001.

Interventionparticipants (Number)
RAD00133

[back to top]

to Assess Overall Survival Associated With RAD001 in This Patient Population.

Overall survival was defined as the time from study entry until death from any cause. (NCT00409292)
Timeframe: 2 years

Interventionmonths (Median)
RAD0014.5

[back to top]

To Assess Progression-free Survival of RAD001 at Two Months in Patients With Metastatic Pancreatic Cancer Whose Disease Has Progressed on Gemcitabine Chemotherapy.

"The Outcome Measure is reporting the number of participants experiencing Progression-free Survival at 2 months after treatment.~The study was designed with a primary end point of progression-free survival (PFS), defined as the time from study entry to documentation of progressive disease or death from any cause. On the basis of prior studies of second-line treatment in metastatic pancreatic cancer, we estimated that such treatment has been associated with a median PFS of 2 months. Our study design used a one-stage design with a target accrual of 35 eligible patients, with the assumption that an improvement in PFS at 2 months from 50% to 71% would warrant further study in this patient population." (NCT00409292)
Timeframe: two months

Interventionparticipants (Number)
RAD00123

[back to top]

to Assess Response Rate Associated With RAD001 in This Patient Population.

The secondary objectives of the study were to assess tumor response rate and overall survival. Patients were required to have measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST). Per RECIST, for target lesions assessed by CT: Complete Response (CR) is Disappearance of all target lesions; Partial Response (PR) is >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) equals CR + PR. (NCT00409292)
Timeframe: 2 years

Interventionparticipants (Number)
Stable Disease as Best ResponseProgressive Disease as Best Response
RAD001722

[back to top]

Time to Definitive Deterioration of the FKS-DRS Risk Score by at Least 2 Score Units Using Kaplan-Meier Method, by Treatment.

"The Functional Assessment of Cancer Therapy - Kidney Symptom Index, Disease Related Symptoms (FKSI-DRS) is a set of items to assess symptoms experienced by patients with advanced kidney cancer. These symptoms include fatigue, pain, weight loss, dyspnea, cough, fever and hematuria. There were 4 response categories (1=Not at all, 2= A little, 3=Quite a bit, 4=Very much), sum of item responses can range from 0 to 36. 0= severely symptomatic patient and the highest score is an asymptomatic patient. Definitive deterioration of the FKSI-DRS score was defined as a decrease by at least 2 units compared to baseline, with no later increase above this threshold observed during the study. A single measure reporting a decrease of at least 2 units was considered definitive only if it is the last one available for the patient. Time to definitive deterioration is the number of days between the date of randomization and the date of the assessment at which definitive deterioration is seen." (NCT00410124)
Timeframe: "Baseline and every 28 days under treatment and at discontinuation from RAD001 until 28Feb2008 cutoff date"

Interventionmonths (Median)
RAD001 +BSC4.76
Placebo + BSC3.84

[back to top]

Progressive Free Survival (PFS) in Patients Who Receive RAD001 Plus Best Supportive Care(BSC) Versus Patients Who Receive Matching Placebo Plus BSC

Progression Free survival is defined as the time from randomization to the date of first documented disease progression or death from any cause. The primary statistical analysis of PFS was based on central radiological assessments using a one-sided stratified log-rank test. Radiological assessments: every 8 weeks (+/-1 week) during the first year and every 12 weeks (+/- 1 week) during the second year and thereafter and at the end of the study. Kaplan-Meier methodology was used to estimate the median PFS for each treatment group. (NCT00410124)
Timeframe: Time from randomization to dates of disease progression, death from any cause or last tumor assessment reported between date of first patient randomized until 28Feb2008 cut of date.

InterventionMonths (Median)
RAD001 +BSC4.90
Placebo + BSC1.87

[back to top]

Overall Survival (OS) Assessed by the Monthly Overall Survival Assessments

Overall survival (OS) was defined as the time from date of randomization to date of death due to any cause. Kaplan-Meier methodology was used to estimate the median overall survival for each treatment group (NCT00410124)
Timeframe: Assessed every month up to 2 years after the last patient was randomized into the study from the date of randomization to the time of death. (Data cutoff was 15Nov2009)

InterventionMonths (Median)
RAD001 +BSC13.57
Placebo + BSC13.01

[back to top]

Pharmacokinetics of RAD001: Time of the Last Quantifiable Concentration in a Dosing Interval - (Tlast)

Blood samples will be collected by direct venipuncture during regularly scheduled visits according to the collection plan provided in the study protocol. (NCT00410124)
Timeframe: At pre-dose and post-dose: 1 hour, 2 hour, 5 hour, 24 hour of Cycle 1 Day 1, Cycle 1 Day 15 and at pre-dose from Cycle 2 (Day 1) and all subsequent treatment cycles until data cut-off 28Feb2008.

Interventionhour (Median)
Day 124.0
Day 1524.0

[back to top]

Pharmacokinetics of RAD001: Time at Which C-Max Occurs (t-Max)

Blood samples will be collected by direct venipuncture during regularly scheduled visits according to the collection plan provided in the study protocol. (NCT00410124)
Timeframe: At pre-dose and post-dose: 1 hour, 2 hour, 5 hour, 24 hour of Cycle 1 Day 1, Cycle 1 Day 15 and at pre-dose of From Cycle 2 (Day 1) and all subsequent treatment cycles until data cut-off 28Feb2008.

Interventionh (Median)
Day 11.0
Day 151.0

[back to top]

Pharmacokinetics of RAD001: Normalized to Body Surface Area (CL/F)

Blood samples will be collected by direct venipuncture during regularly scheduled visits according to the collection plan provided in the study protocol. (NCT00410124)
Timeframe: At pre-dose and post-dose: 1 hour, 2 hour, 5 hour, 24 hour of Cycle 1 Day 1, Cycle 1 Day 15 and at pre-dose from Cycle 2 (Day 1) and all subsequent treatment cycles until data cut-off 28Feb2008.

InterventionL/hour/m^2 (Mean)
Day 157.5

[back to top]

Pharmacokinetics of RAD001: Area Under Curve (AUC) in a Dosing Interval From Time-zero to Time of the Last Quantifiable Concentration. (AUC 0-tlast)

Blood samples will be collected by direct venipuncture during regularly scheduled visits according to the collection plan provided in the study protocol. (NCT00410124)
Timeframe: At pre-dose and post-dose: 1 hour, 2 hour, 5 hour, 24 hour of Cycle 1 Day 1, Cycle 1 Day 15 and at pre-dose from Cycle 2 (Day 1) and all subsequent treatment cycles until data cut-off 28Feb2008.

Interventionng.h/mL (Mean)
Day 1455.0
Day 15729.1

[back to top]

Duration of Response in Patients Who Receive RAD001 Plus BSC Versus Placebo Plus BSC

Duration of overall response (CR or PR) applies only to patients whose Best Overall Response (BOR) was Complete Response (CR) or Partial Response (PR). The start date is the date of first documented response (CR or PR) and the end date is the date of event defined as the first documented progression or death. Radiological assessments: every 8 weeks (+/-1 week) during the first year and every 12 weeks (+/- 1 week) during the second year and thereafter and at the end of the study. (NCT00410124)
Timeframe: Time from randomization to dates of disease progression, death from any cause or last tumor assessment reported, between date of first patient randomized until 28Feb2008 cutoff date

Interventionmonths (Median)
RAD001 +BSCNA

[back to top]

Best Overall Response Rate in Patients Who Receive RAD001 Plus BSC Versus Matching Placebo Plus BSC

The Best Overall Response rate (BOR) is defined as the percentage of patients having achieved confirmed Complete Response + Partial Response. Complete Response (CR) = at least two determinations of CR at least 4 weeks apart before progression. • Partial response (PR) = at least two determinations of PR or better at least 4 weeks apart before progression. Radiological assessments: every 8 weeks (+/-1 week) during the first year and every 12 weeks (+/- 1 week) during the second year and thereafter and at the end of the study. (NCT00410124)
Timeframe: Time from randomization to dates of disease progression, death from any cause or last tumor assessment reported, between date of first patient randomized until 28Feb2008 cutoff date

InterventionPercentage of Participants (Number)
RAD001 +BSC1.8
Placebo + BSC0.0

[back to top]

Analysis of Time to Definitive Deterioration of the Global Health Status/QoL Scale(QL) Scores of the EORTC QLQ-30 Questionnaire by at Least 10 Percent Using Kaplan Meier Method, by Treatment.

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items. Global health status / QoL scale (QL), consisting of 2 questions each scored from 1 (very poor) to 7 (excellent), and with possible scores ranging from 2 to 14. Higher score indicates better functioning. Definitive deterioration by at least 10% is defined as a decrease in score by at least 10% compared to baseline, with no increase above this threshold observed during the course of the study. A single measure reporting a decrease of at least 10% is considered definitive only if it is the last one available for the patient. Time to definitive deterioration is the number of days between the date of randomization and date of assessment at which definitive deterioration is seen. (NCT00410124)
Timeframe: "Baseline and every 28 days under treatment and at discontinuation from RAD001 until 28Feb2008 cutoff date"

Interventionmonths (Median)
RAD001 +BSC4.76
Placebo + BSC3.91

[back to top]

Pharmacokinetics of RAD001: Apparent Systemic Clearance From Blood Following Extravascular Administration (CL/F)

Blood samples will be collected by direct venipuncture during regularly scheduled visits according to the collection plan provided in the study protocol.Apparent oral clearance of RAD001 (CL/F) was calculated using AUC in a dosing interval of 24 hours (AUC0-24hours) value on Day 15 as: CL/F = dose/ AUC0-τ (NCT00410124)
Timeframe: At pre-dose and post-dose: 1 hour, 2 hour, 5 hour, 24 hour of Cycle 1 Day 1, Cycle 1 Day 15 and at pre-dose from Cycle 2 (Day 1) and all subsequent treatment cycles until data cut-off 28Feb2008.

InterventionL/hour (Mean)
Day 1515.4

[back to top]

Pharmacokinetics of RAD001:Peak Concentration in a Dosing Interval (C-max); Pre-dose Concentration at 24-h Time Point in Dosing Interval (C-min) and Average Concentration in a Dosing Interval =(C-avg)

Blood samples will be collected by direct venipuncture during regularly scheduled visits according to the collection plan provided in the study protocol. C-avg= Area under curve (AUC) in a dosing interval from time-zero to time of the last quantifiable concentration (AUC0-tlast)/ time of the last quantifiable concentration in a dosing interval (tlast) (NCT00410124)
Timeframe: At pre-dose and post-dose: 1 hour, 2 hour, 5 hour, 24 hour of Cycle 1 Day1, Cycle 1 Day 15 and at pre-dose from Cycle 2(day1) and all subsequent treatment cycles up until data cut-off 28 Feb 2008.

,
Interventionng/mL (Mean)
C-maxC-minC-avg
Day 168.17.919.0
Day 1576.719.830.4

[back to top]

Time to Definitive Deterioration of the Physical Functioning Scale (PF)Score of the EORTC QLQ-C30 Questionnaire by at Least 10 Percent Using Kaplan_Meier Method, by Treatment.

The EORTC QLQ-C30 contains 30 items. These include five functional scales (physical, role, emotional, social and cognitive functioning), three symptom scales fatigue, pain, nausea, and vomiting), a global health status/QoL scale, and six single items (dyspnea, diarrhea, constipation, anorexia, insomnia and financial impact). Physical Functioning (PF) sub-scale, consisting of 5 questions each scored from 1 (not at all) to 4 (very much), and with possible values ranging from 5 to 20. Definitive deterioration by at least 10% is defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the course of the study. A single measure reporting a decrease of at least 10% is considered definitive only if it is the last one available for the patient. Time to definitive deterioration is the number of days between the date of randomization and the date of the assessment at which definitive deterioration is seen. (NCT00410124)
Timeframe: "Baseline and every 28 days under treatment and at discontinuation from RAD001 until 28Feb2008 cutoff date"

Interventionmonths (Median)
RAD001 +BSC5.06
Placebo + BSC4.57

[back to top]

Overall Reduction in SEGA Tumor Volume.

(NCT00411619)
Timeframe: During the entire study

Interventionparticipants (Number)
# with reductions > 30% relative to baseline# with reductions > 50% relative to baseline
Everolimus1412

[back to top]

Number With Observed Adverse Side Effects

(NCT00411619)
Timeframe: During the entire study

Interventionparticipants (Number)
Upper Respiratory Tract InfectionStomatitisSinusitisMouth UlcerationCellulitisDiarrhoeaGastroenteritisOtitis MediaPyrexiaVomitingAcneConvulsionNasopharyngitisRhinitis AllergicConjunctivitisPharyngitisRashConstipationCoughDermatitis AcneiformDermatitis ContactExcoriationLacerationNasal CongestionOtitis ExternaAbnormal BehaviourBody TineaDry SkinPneumoniaSkin InfectionUrinary Tract Infection
Everolimus26251514121212111010999988877777777666666

[back to top]

Overall Survival Using Kaplan-Meier Methodology

Overall survival was defined as the time from the date of randomization to the date of death from any cause. If a patient was not known to have died, survival was censored at the date of last contact. Kaplan-Meier methodology was used to estimate the median overall survival for each treatment group. (NCT00412061)
Timeframe: Months 12, 24, 36, 48

,
InterventionPercentage of Participants (Number)
12 Months24 Months36 Months48 Months
Octreotide+ Everolimus80.557.042.938.0
Octreotide+ Placebo Followed by Open Label Arm81.863.648.541.6

[back to top]

Progression Free Survival (PFS) as Per Adjudicated Central Review by Baseline 5-hydroxyindoleacetic Acid (5-HIAA) Level

5-HIAA levels in urine are frequently elevated in patients with advanced carcinoid tumors. Baseline levels of 5-HIAA in urine were defined as 'High' if they exceeded the median value, and 'Low' if they were lower than or equal to the median. (NCT00412061)
Timeframe: If elevated at baseline, evaluated every cycle visit (28 days/cycle) reported between day of first patient randomised, 10 January 2007, until cut-off date 02 April 2010

,
InterventionMonths (Median)
5-HIAA <=median (n=93,96)5-HIAA > median (n=94,95)
Octreotide+ Everolimus21.7513.83
Octreotide+ Placebo13.908.41

[back to top]

Number of Patients With Adverse Events (AEs), Clinically Notable AE, Death, Serious Adverse Events (SAEs) (Double-Blind Phase)

AEs are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. SAEs are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00412061)
Timeframe: From first day of treatment up to 28 days after last day of treatment in double blind

,
InterventionPatients (Number)
Clinically notable AEGrade 3-4 Adverse EventsOn treatment deathSerious adverse events
Octreotide+ Everolimus20816219126
Octreotide+ Placebo1461091174

[back to top]

Progression Free Survival (PFS) as Per Adjudicated Central Radiology Review

Progression free survival (PFS) is defined as the time from randomization to the date of first documented disease progression or death from any cause. The primary analysis of PFS was based on the independent central adjudicated assessment using Kaplan-Meier method. (NCT00412061)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 10 January 2007, until cut-off date 02 April 2010

InterventionMonths (Median)
Octreotide+ Everolimus16.43
Octreotide+ Placebo11.33

[back to top]

Best Overall Response Rate as Per Adjudicated Central Radiology Review Based on Response Evaluation Criteria in Solid Tumors (RECIST)

The best overall response rate is defined as the percentage of patients having achieved confirmed Complete Response + Partial Response. Complete Response (CR) = at least two determinations of CR at least 4 weeks apart before progression. • Partial response (PR) = at least two determinations of PR or better at least 4 weeks apart before progression. (NCT00412061)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 10 January 2007, until cut-off date 02 April 2010

InterventionPercentage of patients (Number)
Octreotide+ Everolimus2.3
Octreotide+ Placebo1.9

[back to top]

Progression Free Survival (PFS) as Per Adjudicated Central Review by Baseline Chromogranin A (CgA)

"Serum CgA levels in urine are frequently elevated in patients with advanced carcinoid tumors. Baseline levels of serum CgA were characterized relative to the upper limited of normal (ULN). CgA levels exceeding 2 x ULN were considered to be 'Elevated'; otherwise considered as Non-elevated." (NCT00412061)
Timeframe: If elevated at baseline, evaluated every cycle visit (28 days/cycle) reported between day of first patient randomised, 10 January 2007, until cut-off date 02 April 2010

,
InterventionMonths (Median)
CgA<=2x ULN (n=60,78)CgA>2x ULN (n=152,130)
Octreotide+ Everolimus31.3113.93
Octreotide+ Placebo20.078.41

[back to top]

Number of Patients With Adverse Events (AEs), Clinically Notable AE, Death, Serious Adverse Events (SAEs) (Open Label Phase)

AEs are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. SAEs are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00412061)
Timeframe: From first day of treatment up to 28 days after last day of treatment in double blind

InterventionPatients (Number)
Clinically notable AEGrade 3-4 Adverse EventsOn treatment deathSerious adverse events
Everolimus Open Label Arm1541152293

[back to top]

Course of Calculated GFR (mL/Min/1.73 m^2) From Month 24 to Month 60

Course of calculated GFR (mL/min/1.73 m^2) at Months 24, 36, 48 and 60 (NCT00414440)
Timeframe: Months 24, 36, 48 and 60

,
InterventionmL/min/1.73 m^2 (Mean)
Month 24Month 36Month 48Month 60
Everolimus43.942.639.637.7
Placebo48.844.942.737.6

[back to top]

Changes in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)

Changes in systolic blood pressure (SBP) and diastolic blood pressure (DBP), at baseline and then months 12 and 24 (NCT00414440)
Timeframe: Baseline, Months 12 and 24

,
InterventionmmHG (Mean)
Baseline SBPMonth 12 SBPMonth 24 SBPBaseline DBPMonth 12 DBPMonth 24 DBP
Everolimus136134134888685
Placebo135134134888685

[back to top]

Calculated GFR, Change From Baseline at Month 60 by Baseline cGFR

Change in renal function was assessed by the estimated Glomerular Filtration Rate (eGFR) using the abbreviated (4 variables) Modification of Diet in Renal Disease (MDRD-4) formula which was developed by the MDRD Study Group and has been validated in patients with chronic kidney disease. The MDRD-4 formula used for the eGFR calculation is: eGFR (mL/min/1.73m^2) = 186.3*(C^-1.154)*(A^-0.203)*G*R, where C is the serum concentration of creatinine (mg/dL), A is age (years), G=0.742 when gender is female, otherwise G=1, R=1.21 when race is black, otherwise R=1. The changes in renal function were analyzed via analysis of covariance (ANCOVA) with treatment, pre-transplant hepatitis C virus status and randomization eGFR as covariates. Based on these ANCOVA analyses, the least-squares mean and standard errors of change were reported. (NCT00414440)
Timeframe: Months 24, 36, 48 and 60

,
InterventionmL/min/1.73 m^2 (Mean)
>70 mL/min/1.73 m^2 n=11, 18≤70 mL/min/1.73 m^2 n=53, 57>60 mL/min/1.73 m^2 n=21.26≤60 mL/min/1.73 m^2 n=43,49>50 mL/min/1.73 m^2 n=36,39≤50 mL/min/1.73 m^2 n=28, 36
Everolimus-14.8-16.1-17.0-15.3-19.0-11.8
Placebo-20.5-15.6-20.0-15.1-19.4-14.0

[back to top]

Primary Efficacy Analysis of Total Kidney Volume (mITT Set, Multiple Imputation)

Everolimus (RAD001) compared to placebo with respect to the change from baseline in total kidney volume at Month 24. (NCT00414440)
Timeframe: Baseline, Month 24

InterventionmL (Mean)
Everolimus230.1
Placebo300.8

[back to top]

Calculated GFR (mL/Min/1.73 m^2), Change From Baseline by Visit

Change in renal function was assessed by the Glomerular Filtration Rate (GFR) using the abbreviated (4 variables) Modification of Diet in Renal Disease (MDRD-4) formula which was developed by the MDRD Study Group and has been validated in patients with chronic kidney disease. The MDRD-4 formula used for the eGFR calculation is: eGFR (mL/min/1.73m^2) = 186.3*(C^-1.154)*(A^-0.203)*G*R, where C is the serum concentration of creatinine (mg/dL), A is age (years), G=0.742 when gender is female, otherwise G=1, R=1.21 when race is black, otherwise R=1. The changes in renal function were analyzed via analysis of covariance (ANCOVA) with treatment, pre-transplant hepatitis C virus status and randomization eGFR as covariates. Based on these ANCOVA analyses, the least-squares mean and standard errors of change were reported. (NCT00414440)
Timeframe: Months 3, 6, 9, 12, 18 and 24

,
InterventionmL/min/1.73m^2 (Mean)
Week 1Week 2Week 4Month 3Month 6Month 9Month 12Month 18Month 24
Everolimus2.01.70.6-0.5-2.3-4.6-5.4-7.7-8.9
Placebo-0.9-0.9-1.2-2.4-2.2-2.4-3.2-5.5-7.7

[back to top]

Biomarkers Predictive of Clinical Benefit (DCR by PTEN ) on Everolimus (RAD001) 10mg/Day

The efficacy variable that was compared in the biomarker analysis is DCR within the Everolimus (RAD001) 10mg/day arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: PTEN loss (imunohistochemistry, IHC), phosphoAKT (IHC), phosphoS6 (IHC), and p53 (IHC). From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

Interventionpercentage of participants (Number)
PTEN Expression Low/Everolimus (RAD001) 10 mg/Day26.9
PTEN Expression Normal/Everolimus (RAD001) 10 mg/Day26.1

[back to top]

Biomarker Predictive of Clinical Benefit (DCR by KRAS) on Everolimus (RAD001) 70 mg/Week

The efficacy variable compared in this biomarker analysis is disease control rate (DCR) within the 70mg/week arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: KRAS gene mutation. From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

Interventionpercentage of participants (Number)
KRAS Mutation/Everolimus (RAD001) 70 mg/Week33.3
KRAS Wildtype/Everolimus (RAD001) 70 mg/Week21.7

[back to top]

Biomarkers Predictive of Clinical Benefit (Median PFS and OS by PTEN ) on Everolimus (RAD001) 10mg/Day

The efficacy variable that was compared in the biomarker analysis are PFS & OS within the Everolimus (RAD001) 10mg/day arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: PTEN loss (imunohistochemistry, IHC), phosphoAKT (IHC), phosphoS6 (IHC), and p53 (IHC). From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

,
Interventionmonths (Median)
Median PFSMedian OS
PTEN Expression Low/Everolimus (RAD001) 10 mg/Day1.8110.38
PTEN Expression Normal/Everolimus (RAD001) 10 mg/Day1.686.34

[back to top]

Biomarker Predictive of Clinical Benefit (DCR by KRAS) on Everolimus (RAD001) 10 mg/Day

The efficacy variable compared in this biomarker analysis is disease control rate (DCR) within the 10mg/day arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: KRAS gene mutation. From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

Interventionpercentage of participants (Number)
KRAS Mutation/Everolimus (RAD001) 10 mg/Day17.1
KRAS Wildtype/Everolimus (RAD001) 10 mg/Day35.0

[back to top]

Number of Patients Who Died, Had an Serious Adverse Event (SAE), Had Grade 3 to 4 Adverse Event (AE), Discontinued Due to an AE, or Had a Clinical Notable AE by Treatment (tr).

Toxicity assessed using the NIH-NCI Common Terminology Criteria for Adverse Events, Version 3.0 (CTCAEv3.0). On treatment death defined as deaths occurring no more than 28 days after the discontinuation of study treatment. (NCT00419159)
Timeframe: From the first day of treatment until 28 days after discontinuation of study treatment

,
InterventionParticipants (Number)
DeathsOn-treatment DeathSAE regardless of relationship to study treatmentSAE with suspected relationship to study treatmentAE Grade 3-4, regardless of relationship to studyAE Grade 3-4, suspected relationship to study trAE leading to discontinuationClinically notable adverse event
Everolimus (RAD001) 10 mg/Day651422650262076
Everolimus (RAD001) 70 mg/Week86113475831985

[back to top]

Disease Control Rate (DCR) and Objective Response Rate (ORR) According to the Response Evaluation Criteria in Solid Tumors (RECIST)

"RECIST (Response Evaluation Criteria In Solid Tumors) is a set of published rules that define when cancer patients improve (respond), stay the same (stable) or worsen (progression) during treatments.~Disease Control Rate (DCR) defined as the percentage of participants with Disease Control best overall response (complete response, partial response or stable disease)and Objective Response Rate (ORR) defined as the percentage of participants with best overall Objective Response (complete response or partial response)." (NCT00419159)
Timeframe: Imaging every 8 weeks

,
InterventionPercentage of participants (Number)
Disease Control Rate (DCR)Objective Response Rate (ORR)
Everolimus (RAD001) 10 mg/Day32.40
Everolimus (RAD001) 70 mg/Week31.00

[back to top]

Biomarkers Predictive of Clinical Benefit (Median PFS and OS by PTEN ) on Everolimus (RAD001) 70mg/Week

The efficacy variable that was compared in the biomarker analysis are PFS & OS within the Everolimus (RAD001) 70mg/Week arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: PTEN loss (imunohistochemistry, IHC), phosphoAKT (IHC), phosphoS6 (IHC), and p53 (IHC). From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

,
Interventionmonths (Median)
Median PFSMedian OS
PTEN Expression Low/Everolimus (RAD001) 70 mg/Week1.715.98
PTEN Expression Normal/Everolimus (RAD001) 70 mg/Week1.774.37

[back to top]

The Number of Participants With Best Overall Response According to Response Evaluation Criteria in Solid Tumors (RECIST)

"RECIST (Response Evaluation Criteria In Solid Tumors) is a set of published rules that define when cancer patients improve (respond), stay the same (stable) or worsen (progression) during treatments.~Best Over Response (BOR): Complete Response (CR, No lesions), Partial Response (PR, 30% decrease in lesions), and Stable Disease (SD, none of the above)" (NCT00419159)
Timeframe: Imaging every 8 weeks

,
InterventionParticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Disease Control (CR or PR or SD)Objective Response (CR or PR)Unknown
Everolimus (RAD001) 10 mg/Day00265526019
Everolimus (RAD001) 70 mg/Week00255825016

[back to top]

Biomarkers Predictive of Clinical Benefit (Median PFS and OS by KRAS) on Everolimus (RAD001) 10mg/Day

The efficacy variable that was compared in the biomarker analysis are Median progression free survival (PFS) and overall survival (OS) within the Everolimus (RAD001) 10mg/day arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: KRAS gene mutation. From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

,
Interventionmonths (Median)
Median PFSMedian OS
KRAS Mutation/Everolimus (RAD001) 10 mg/Day1.715.59
KRAS Wildtype/Everolimus (RAD001) 10 mg/Day1.777.06

[back to top]

Progression-free Survival (PFS)

Duration in months from the date of first study treatment to the date of the first documented disease progression or death due to any cause. (NCT00419159)
Timeframe: Imaging every 8 weeks

InterventionMonths (Median)
Everolimus (RAD001) 70 mg/Week1.77
Everolimus (RAD001) 10 mg/Day1.77

[back to top]

Biomarkers Predictive of Clinical Benefit (Median PFS and OS by KRAS ) on Everolimus (RAD001) 70mg/Week

The efficacy variable that was compared in the biomarker analysis are Median progression free survival (PFS) and overall survival (OS) within the Everolimus (RAD001) 70mg/week arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: KRAS gene mutation. From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

,
Interventionmonths (Median)
Median PFSMedian OS
KRAS Mutation/Everolimus (RAD001) 70 mg/Week1.776.18
KRAS Wildtype/Everolimus (RAD001) 70 mg/Week1.714.90

[back to top]

Overall Survival (OS)

Overall survival defined as the time from date of first study treatment to the date of death due to any cause. (NCT00419159)
Timeframe: Every 3 months

InterventionMonths (Median)
Everolimus (RAD001) 70 mg/Week4.90
Everolimus (RAD001) 10 mg/Day5.88

[back to top]

Biomarkers Predictive of Clinical Benefit (DCR by PTEN ) on Everolimus (RAD001) 70mg/Week

The efficacy variable that was compared in the biomarker analysis is DCR within the Everolimus (RAD001) 70mg/week arm in the analysis not adjusted for prognostic factors: from archival tumor tissue (and additionally from a biopsy at a metastatic site, if available), collected during screening: PTEN loss (imunohistochemistry, IHC), phosphoAKT (IHC), phosphoS6 (IHC), and p53 (IHC). From blood plasma, collected at screening then on day 1 at cycles 2, 3 and 4, and at the end of treatment: blood lactate dehydrogenase (LDH) isoenzyme fractionation and serum levels of sVEGFR2, bFGF, PLGF and VEGF. (NCT00419159)
Timeframe: Screening and Day 1 of cycles 2, 3, 4 and end of treatment

Interventionpercentage of participants (Number)
PTEN Expression Low/Everolimus (RAD001) 70 mg/Week20.9
PTEN Expression Normal/Everolimus (RAD001) 70 mg/Week33.3

[back to top]

Number of Participants With Biopsy-proven Acute Rejection (BPAR) at Month 6 and Month 12.

(NCT00425308)
Timeframe: Month 6 and 12

,
Interventionparticipants (Number)
Yes: treatment failure Month 6No: treatment failure Month 6Yes: treatment failure Month 12No: treatment failure Month 12
Cyclosporine00112
Enteric-coated Mycophenolate Sodium (EC-MPS)00014

[back to top]

Number of Participants With Treatment Failures Assessed by Biopsy-proven Acute Rejection (BPAR), Graft Loss/Re-transplantation, Death or Lost to Follow-up at Month 12.

(NCT00425308)
Timeframe: Month 12

,
Interventionparticipants (Number)
BPAR 12 monthsGraft Loss 12 monthsDeath 12 monthsLost to Follow-up 12 months
Cyclosporine1010
Enteric-coated Mycophenolate Sodium (EC-MPS)0000

[back to top]

Change in the Glomerular Filtration Rate Estimated by Iohexol Plasma Clearance 12 Months After Randomization Between the 2 Groups of Patients Who Completed Trial

Primary efficacy endpoint: between treatment analysis of change in iohexol plasmatic clearance (mL/min) from baseline to Month 12 (M12) (NCT00425308)
Timeframe: From Baseline to Month 12

Intervention(mL/min) (Least Squares Mean)
Cyclosporine1.46
Enteric-coated Mycophenolate Sodium (EC-MPS)12.00

[back to top]

Change in Glomerular Filtration Rate Estimated by Iohexol Plasma Clearance 12 Months After Randomization Between the 2 Groups of Patients.

Primary efficacy endpoint: between treatment analysis of change in iohexol plasmatic clearance (mL/min) from baseline to Month 12 (M12) (NCT00425308)
Timeframe: From Baseline to Month 12

Intervention(mL/min) (Least Squares Mean)
Cyclosporine-4.07
Enteric-coated Mycophenolate Sodium (EC-MPS)10.30

[back to top]

Assessing Cardiovascular Risk Factors Based on Fasting C-reactive Protein (CRP).

Blood chemistry - C-reactive Protein (CRP) (mg/L) (NCT00425308)
Timeframe: From Baseline to Month 3, 6, and 12

,
Interventionmg/L (Mean)
Baseline [N = 13; N = 14]Month 3 [N = 11; N = 11]Month 6 [N = 11; N = 9]Month 12 [N = 14; N = 12]
Cyclosporine11.46.38.034.2
Enteric-coated Mycophenolate Sodium (EC-MPS)3.010.35.612.6

[back to top]

Assessing Cardiovascular Risk Factors Based on Fasting Triglycerides.

(NCT00425308)
Timeframe: From Baseline to Month 1, 3, 6, 9, and 12

,
Interventionmmol/L (Mean)
Baseline [N = 11; N = 14]Month 1 [N = 10; N = 14]Month 3 [N = 13; N = 14]Month 6 [N = 12; N = 12]Month 9 [N = 10; N = 12]Month 12 [N = 12; N = 11]
Cyclosporine2.42.12.01.91.92.3
Enteric-coated Mycophenolate Sodium (EC-MPS)1.82.02.22.12.22.3

[back to top]

Assessing Cardiovascular Risk Factors Based on Fasting Total Cholesterol.

Blood chemistry - total cholesterol (mmol/L) (NCT00425308)
Timeframe: From Baseline to Month 1, 3, 6, 9, and 12

,
Interventionmmol/L (Mean)
Baseline [N = 13; N = 14]Month 1 [N = 11; N = 14]Month 3 [N = 13; N = 14]Month 6 [N = 12; N = 12]Month 9 [N = 9; N = 12]Month 12 [N = 12; N = 11]
Cyclosporine5.75.05.35.55.45.6
Enteric-coated Mycophenolate Sodium (EC-MPS)6.05.75.65.75.45.8

[back to top]

Assessing Cardiovascular Risk Factors Based on Fasting High-density Lipoprotein (HDL) Cholesterol, Low-density Lipoprotein (LDL) Cholesterol.

(NCT00425308)
Timeframe: From Baseline to Month 3, 6, and 12

,
Interventionmmol/L (Mean)
Baseline LDL [N = 10; N = 14]Baseline HDL [N = 10; N = 14]Month 3 LDL [N = 12; N = 14]Month 3 HDL [N = 12; N = 13]Month 6 LDL [N = 12; N = 12]Month 6 HDL [N = 12; N = 12]Month 12 LDL [N = 10; N = 11]Month 12 HDL [N = 10; N = 11]
Cyclosporine3.41.53.21.73.21.63.01.7
Enteric-coated Mycophenolate Sodium (EC-MPS)3.11.53.41.53.31.43.21.5

[back to top]

Change in Renal Function Assessed by Creatinine Clearance at Month 3, Month 6 and Month 12

Change in creatinine clearance, Nankivell formula (mL/min/1.73m²) from baseline to M12 (NCT00425308)
Timeframe: From Baseline to Month 3, 6, and 12

,
InterventionmL/min/1.73m² (Mean)
Baseline [N = 12; N = 13]Month 3 [N = 10; N = 14]Month 6 [N = 10; N = 12]Month 12 [N = 7; N = 11]
Cyclosporine56.357.350.348.7
Enteric-coated Mycophenolate Sodium (EC-MPS)60.367.168.170.1

[back to top]

Change in Renal Function Assessed by Proteinuria at Month 3, Month 6 and Month 12

Change in proteinuria (g/24h) from baseline to M12 (NCT00425308)
Timeframe: From Baseline to Month 3, 6, and 12

,
Interventiong/24h (Mean)
Baseline [N = 9; N = 10]Month 3 [N = 9; N = 8]Month 6 [N = 6; N = 5]Month 12 [N = 7; N = 10]
Cyclosporine0.440.530.440.48
Enteric-coated Mycophenolate Sodium (EC-MPS)0.470.240.330.75

[back to top]

Change in Renal Function Assessed by Serum Creatinine at Month 3, Month 6 and Month 12

(NCT00425308)
Timeframe: From Baseline to Month 3, 6, and 12

,
Interventionµmol/L (Mean)
Baseline [N = 13; N = 14]Month 3 [N = 12; N = 14]Month 6 [N = 12; N = 13]Month 12 [N = 13; N = 14]
Cyclosporine161.7156.6170.8227.7
Enteric-coated Mycophenolate Sodium (EC-MPS)153.2138.9139.8130.4

[back to top]

Assessing Cardiovascular Risk Factors Based on Fasting Glucose.

Blood chemistry - fasting glycemia (mmol/L) (NCT00425308)
Timeframe: From Baseline to Month 1, 3, 6, 9, and 12

,
Interventionmmol/L (Mean)
Baseline [N = 15; N = 15]Month 1 [N = 15; N = 15]Month 3 [N = 14; N = 15]Month 6 [N = 14; N = 14]Month 9 [N = 12; N = 13]Month 12 [N = 14; N = 13]
Cyclosporine5.55.25.65.55.34.8
Enteric-coated Mycophenolate Sodium (EC-MPS)5.25.35.25.15.65.4

[back to top]

Phase I: Best Overall Response (BOR)

BOR was determined based on investigator assessment of overall lesion response using RECIST criteria guidelines. BOR = objective responses rate (ORR), disease control rate (DCR) or clinical benefit rate (CBR). ORR = (complete response (CR) or partial response(PR); DCR = (CR or PR or Stable disease (SD); CBR = (CR or PR or SD >= 24 weeks).CR = Disappearance of all target lesions; PR = At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters; SD = Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for partial disease (PD). PD = At least a 20% increase in the sum of the longest diameter of all measured target lesions, taking as reference the smallest sum of longest diameter of all target lesions recorded at or after baseline. (NCT00426556)
Timeframe: every 8 - 9 weeks until disease progression or a new lesion is identified

,,
InterventionPercentage of Participants (Number)
Objective Response Rate (ORR)Disease Control Rate (DCR)Clinical Benefit Rate (CBR)
Phase I - RAD001 10mg + PT, Daily23.582.447.1
Phase I - RAD001 30mg + PT, Weekly30.080.070.0
Phase I - RAD001 5mg + PT, Daily83.383.383.3

[back to top]

Phase II: Progression Free Survival (PFS)

PFS is defined as the time from start of treatment to the date of first documented progression or death due to any cause. If a patient has not had an event, PFS will be censored at the date of last adequate tumor assessment. (NCT00426556)
Timeframe: every 8 - 9 weeks until disease progression or a new lesion is identified

InterventionMonths (Median)
Phase II - RAD001 10mg + PT, Daily5.52

[back to top]

Phase II: Overall Survival (OS)

Overall survival (OS) is defined as the time from start of treatment to the date of death due to any cause. If a patient is not known to have died, survival was censored at the last date of contact. OS was to be reported at extension and after 3-year follow-up. The Kaplan-Meier median was used to analyze the OS. (NCT00426556)
Timeframe: every 3 months until death

InterventionMonths (Median)
Phase II - RAD001 10mg + PT, Daily18.07

[back to top]

Phase II: Overall Response Rate

The primary objective of this phase II study was to evaluate the efficacy of the dose level/regimen of everolimus recommended from the Phase I with trastuzumab and paclitaxel (PT) therapy in patients with HER2-overexpressing metastatic breast cancer whose disease progressed on/after trastuzumab mono-and/or combination therapy based on the evaluation of objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors (RECIST). Objective response rate (ORR) was defined as the proportion of patients with a best overall response (BOR) of complete response (CR) or partial response (PR). Only patients with measurable disease (the presence of at least one measurable lesion) at baseline were included in the study. CR = Disappearance of all target lesions; PR = At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters. (NCT00426556)
Timeframe: every 8 - 9 weeks until disease progression or a new lesion is identified

InterventionPercentage of Participants (Number)
Phase II - RAD001 10mg + PT, Daily21.8

[back to top]

Progression-free Survival

"Progression-free survival is defined as the time from registration to the time of progression or death due to any cause. Progression-free survival was estimated using the method of Kaplan-Meier.~Progression is defined as the following:~CLL (subset of patients in the Relapsed Indolent Non-Hodgkin Lymphoma group): >=50% increase in nodes from nadir or >=50% increase in liver/spleen size from nadir.~Waldenstrom (subset of patients in the Uncommon Lymphomas group): >50% lymph node increase in SPD of > 1 node or new nodes, or >50% liver/spleen size increase, or > 25% IgM (by SPEP) increase, or lymphocyte morphology transformation to a more aggressive histology.~All Others: New lesions or >=50% lymph nodes." (NCT00436618)
Timeframe: 5 years

Interventionyears (Median)
Relapsed Aggressive Non-Hodgkin Lymphoma0.18
Relapsed Indolent Non-Hodgkin Lymphoma0.60
Uncommon Lymphomas0.79

[back to top]

Tumor Response, Defined by Disease: Chronic Lymphocytic Leukemia(CLL): Clinical Complete or Complete or Nodular Partial or Partial Remission, Waldenstrom: Complete or Partial Response, All Others: Complete or Complete Unconfirmed or Partial Response.

"CLL (subset of patients in the Relapsed Indolent Non-Hodgkin Lymphoma group): 50% decrease in peripheral blood lymphocytes, lymphadenopathy, liver/spleen size, presence/absence of constitutional symptoms; plus ≥1 of the following: ≥1500/μL polymorphonuclear leukocytes, >100000/μL platelets, >11.0 g/dL hemoglobin or 50% improvement for these parameters without transfusions.~Waldenstrom (subset of patients in the Uncommon Lymphomas group): >50% reduction in serum immunoglobulin M(IgM) levels (by serum protein electrophoresis (SPEP)) during any point while in this study, and no appearance of new lesions.~All others: at least a 50% decrease in the sum of the products of the greatest diameters (SPD) of the six largest dominant nodes or nodal masses and no increase in the size of other nodes, liver, or spleen and splenic and hepatic nodules must regress by at least 50% in the SPD and no new sites of disease." (NCT00436618)
Timeframe: 5 years

Interventionpercentage of patients in group (Number)
Relapsed Aggressive Non-Hodgkin Lymphoma26
Relapsed Indolent Non-Hodgkin Lymphoma35
Uncommon Lymphomas49

[back to top]

Time to Progression

The time to progression is defined as the time from registration to the time of progression. The distribution of time to progression was estimated using the method of Kaplan-Meier. (NCT00436618)
Timeframe: 5 years

Interventionyears (Median)
Relapsed Aggressive Non-Hodgkin Lymphoma0.18
Relapsed Indolent Non-Hodgkin Lymphoma0.60
Uncommon Lymphomas0.90

[back to top]

Overall Survival

The overall survival or survival time is defined as the time from registration to death due to any cause. The distribution of overall survival was estimated using the method of Kaplan-Meier. (NCT00436618)
Timeframe: 5 years

Interventionyears (Median)
Relapsed Aggressive Non-Hodgkin Lymphoma0.66
Relapsed Indolent Non-Hodgkin Lymphoma2.45
Uncommon Lymphomas3.41

[back to top]

Number of Participants With Objective Response

Efficacy reported as objective response. Objective response defined as change in serum tryptase level or bone marrow mast cell percentage. (NCT00449748)
Timeframe: Monthly for first 3 months, then every 3 months

InterventionParticipants (Number)
RAD0010

[back to top]

Clinical Response Rate

Best response on treatment was based on RECIST 1.0 criteria with overall clinical response defined as achieving stable disease (SD), partial response (PR) or complete response (CR). Per RECIST 1.0 for target lesions, CR is complete disappearance of all target lesions and PR is at least a 30% decrease in the sum of longest diameter (LD) of target lesions, taking as reference baseline sum LD. To be assigned a status of CR or PR, changes in tumor measurements must be confirmed by repeat assessments performed no fewer than 4 weeks after the response. SD is neither CR/PR or progressive disease (PD). PD is at least a 20% increase in sum LD, takings as reference smallest sum LD since treatment started. (NCT00458237)
Timeframe: Disease assessments occurred every 9 weeks (3 cycles) on treatment. Treatment continued until disease progression or unacceptable toxicity. Median duration of treatment was 2.4 months.

Interventionproportion of participants (Number)
Phase I: Everolimus (Dose Level 2) and Trastuzumab.67
Phase II: Everolimus (Maximum Tolerated Dose) and Trastuzumab.80

[back to top]

Maximum Tolerated Dose (MTD)

"The MTD is determined by the number of patients who experience a dose limiting toxicity (DLT). The MTD is defined as the highest dose at which fewer than one-third of patients experience a DLT. If no DLTs are observed, the MTD is not reached. Dose Limiting Toxicities (DLTs) were defined as follows (CTCAE v4.0):~Any grade 4 hematologic toxicity, excluding anemia.~Any grade 3 or 4 nonhematologic toxicity, except for nausea, vomiting, diarrhea, or hyperlipidemia that responds promptly (within 24 hours for nausea, vomiting, and diarrhea and within 1 week for hyperlipidemia) to appropriate treatment, and except for cardiac toxicity which will be assessed after 12 weeks of treatment.~Need to hold >1 dose of trastuzumab or > 7 doses of RAD001 within the first 3 weeks because of the presence of toxicity." (NCT00458237)
Timeframe: Cycle One (first 21 days of treatment)

Interventionparticipants with DLT (Number)
Phase I: Everolimus (Dose Level 1) and Trastuzumab0
Phase I: Everolimus (Dose Level 2) and Trastuzumab0

[back to top]

Number of Patients Free of Dose Limiting Toxicity

"A dose limiting toxicity was defined as an adverse event or laboratory abnormality that occurs to patients on the Phase I portion of the trial, during the first 21 days following the first dose of RAD001/docetaxel during cycle 1, judged to be related to RAD001/docetaxel and meeting any of the following criteria:~Hematologic Toxicity:~CTCAE grade 4 neutropenia > 7 days or any Grade 3 or 4 neutropenia with fever Or CTCAE grade 3 or 4 thrombocytopenia > 7 days~Non-hematologic toxicity:~The occurrence of non-hematologic CTCAE grade 3 or 4 adverse events will be considered dose limiting, except for the following:~CTCAE grade 3 nausea or grade 3 or 4 vomiting CTCAE grade 3 or 4 vomiting will only be considered dose limiting if it occurs despite the use of standard anti-emetics.~CTCAE grade 3 or 4 fever identified with a source (i.e. infection, tumor)~CTCAE grade 3 or 4 alkaline phosphatase." (NCT00459186)
Timeframe: 21 days

Interventionparticipants (Number)
RAD001 Followed by RAD001 + Docetaxel14

[back to top]

Response Based on PET Scan

Patients were scanned using Positron Emission Tomography (PET) before and after receiving single agent RAD001. Patients were classified as having partial metabolic response, stable metabolic disease, or progressive metabolic disease based on changes in PET imaging from baseline to post-treatment. A positive FDG-PET for the purposes of this study consisted of a visualized area of abnormal increased FDG uptake that matched the anatomic location of an abnormality seen on bone scan or CT. Metabolic response was assessed for percent change in SUVmax according to the criteria of the European Organization for Research and Treatment of Cancer (EORTC) : partial metabolic response (PMR) ≤ -25%; stable metabolic disease (SMD) -25% + 25%; progressive metabolic disease (PMD) > 25%. (NCT00459186)
Timeframe: 10 to 14 days after study entry

Interventionpercentage of participants (Number)
Partial Metabolic ResponseStable Metabolic DiseaseProgressive Metabolic Disease
RAD001 Followed by RAD001 + Docetaxel226711

[back to top]

Survival Time

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT00474929)
Timeframe: Up to 3 years from registration

Interventionmonths (Median)
Sorafenib 200 mg Twice Daily, RAD001 5mg Every Day13.5

[back to top]

Progression Free Survival

Progression Free Survival is defined as the time from registration to the earliest date documentation of disease progression or death. The distribution of time to disease progression will be estimated using the method of Kaplan-Meier. (NCT00474929)
Timeframe: Up to 3 years from registration

Interventionmonths (Median)
Sorafenib 200 mg Twice Daily, RAD001 5mg Every Day4.6

[back to top]

Number of Participants Reporting a Dose Limiting Toxicity (DLT)

"The Maximum Tolerated Dose (MTD) is defined as the dose level below the lowest dose that induces dose-limiting toxicity (DLT) in at least one-third of patients (at least 2 of a maximum of 6 new patients). Dose-limiting toxicity (DLT) is defined as an adverse event attributed (definitely, probably, or possibly) in first cycle to the study treatment and meeting the following criteria:~Grade 4 infection~Grade 4 ANC or PLT~Grade 3 or higher non-hematologic adverse event.~NCI Common Terminology Criteria for Adverse Events (CTCAE) v3.0 will be used to assess adverse events. For this endpoint, the number of patients reporting a DLT event are tabulated." (NCT00474929)
Timeframe: First cycle (28 days) of study treatment

Interventionparticipants (Number)
Phase I, Dose Level 01
Phase I, Dose Level 10
Phase I, Dose Level 21
Phase I, Dose Level 32

[back to top]

Proportion of Confirmed Tumor Responses

"A confirmed response is defined to be a CR or PR noted as the objective status for eligible patients during cycles 1-12.~Complete Response (CR):~Multiple Myeloma (MM): Negative immunofixation of serum and urine, disappearance of soft tissue plasmacytomas, and normalization of free light chain (FLC) ratio.~Lymphoma: Disappearance of all clinical and radiographic evidence of disease, all lymph nodes of normal size (1.5 cm or less), no splenomegaly.~Partial Response (PR):~MM: 50% reduction of serum or urine M-protein or to less than 200mg/day, a 50% reduction in the difference between involved and uninvolved FLC, and 50% reduction in the size of soft tissue plasmacytoma.~Lymphoma: 50% or greater reduction in sum of the products of the dimension for nodal masses; no increase in liver, spleen or node size; no new sites of disease; and a 50% decrease in lymphocyte count if followed at baseline." (NCT00474929)
Timeframe: Up to 12 cycles of treatment

Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Sorafenib 200 mg Twice Daily, RAD001 5mg Every Day8.515.9

[back to top]

Participant Responses Per Treatment Arm at 12 Weeks

Radiographic criteria of response based on regional ultrasound examination (decrease in size of the primary tumor and/or fatty replacement in regional lymph nodes), and includes partial response and complete response. A decrease in size of the product of the two largest dimensions =/> 50% considered a partial response (PR), and a complete disappearance of the primary tumor by physical exam and or ultrasound and normalization of the lymph nodes by ultrasound will be considered a complete clinical response (CR). Stable Disease (SD) is carcinoma neither decreasing nor increasing in extent or severity, and Progression of disease (PD) defined as 30% increase in size primary tumor and/or lymph nodes on physical exam and/or ultrasound. (NCT00499603)
Timeframe: 12 weeks

,
Interventionparticipants (Number)
CRPRSDPD
Paclitaxel + FEC35163
Paclitaxel + RAD001 + FEC011111

[back to top]

Number Participants With Inhibition of PI3K/PTEN/AKT Pathway at 48 Hours

Number of participants with inhibition of the PI3K/PTEN/AKT pathway at 48 hours after the start of treatment, regardless of the status of the pathway at the time of randomization. Molecular changes (inhibition/activation) of the PI3K/PTEN/AKT pathway evaluated using reverse phase protein arrays (RPPA) where fine-needle aspirations (FNAs) from the primary breast cancer obtained pretreatment, and at 48 hours. Bioinformatics cluster analysis of arrays used to define molecular changes as inhibition or activation where pathways called 'active' with presence of 2 or more phosphorilated pathway proteins (pAKT, pmTOR, pGSK3, pS6K1, pS6), and 'inhibited' with one or none phosphorilated pathway proteins present. (NCT00499603)
Timeframe: 48 hours after start of treatment

Interventionparticipants (Number)
Paclitaxel + FEC27
Paclitaxel + RAD001 + FEC22

[back to top]

Participant Responses Per Treatment Arm at 24 Weeks

Radiographic criteria of response based on regional ultrasound examination (decrease in size of the primary tumor and/or fatty replacement in regional lymph nodes), and includes partial response and complete response. A decrease in size of the product of the two largest dimensions =/> 50% considered a partial response (PR), and a complete disappearance of the primary tumor by physical exam and or ultrasound and normalization of the lymph nodes by ultrasound will be considered a complete clinical response (CR). Stable Disease (SD) is carcinoma neither decreasing nor increasing in extent or severity, and Progression of disease (PD) defined as 30% increase in size primary tumor and/or lymph nodes on physical exam and/or ultrasound. (NCT00499603)
Timeframe: 24 weeks

,
Interventionparticipants (Number)
CRPRSDPD
Paclitaxel + FEC41670
Paclitaxel + RAD001 + FEC21173

[back to top]

Progression Free Survival According to Neuron Specific Enolase Tumor Marker (NSE) Baseline Level and According to NSE Early Response

"Baseline levels of serum NSE were characterized towards PFS as per local investigator assessment, relative to the upper limited of normal (ULN). NSE levels exceeding ULN were considered to be 'Elevated' otherwise considered as Non-elevated. An 'early response' (applicable to only those patients with elevated levels at baseline) was defined as a decrease of greater than or equal to 30% from baseline to Cycle 2 Day 1 or normalization by Cycle 2 Day 1. NSE is widely expressed in well-differentiated pancreatic NET. NSE is usually expressed in the cytoplasm. Pancreatic NET patients often present with elevated circulating levels of NSE in their blood. Baseline levels of these biomarkers are considered as prognostic factors." (NCT00510068)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 17 August 2007, until cut-off date 28 February 2010

,
InterventionMonths (Median)
NSE Levels at baseline: <= ULN (n: 155, 138)NSE levels at baseline: > ULN (n: 48, 56)Early NSE response: Response (n: 24, 16)Early NSE response: Non-Response (n:16, 27)
Everolimus 10 mg/Day13.868.118.113.79
Placebo5.362.833.062.58

[back to top]

Progression Free Survival According to Ki-67 Levels Categorized as: Less Than or Equal to 2%, > 2% to Less Than or Equal to 5% and > 5%

The level of Ki 67 expression for evaluable tumor samples were analyzed towards progression free survival (PFS) as per local investigator assessment. The Ki-67 protein is a cellular marker for proliferation. It is strictly associated with cell proliferation. During interphase, the Ki-67 antigen can be exclusively detected within the cell nucleus, whereas in mitosis most of the protein is relocated to the surface of the chromosomes. Baseline Ki 67 levels were categorized as: less than or equal to 2%, > 2% to less than or equal to 5% and > 5%. (NCT00510068)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 17 August 2007, until cut-off date 28 February 2010

,
InterventionMonths (Median)
Ki67 <=2% (n: 7, 17)2% Ki67 >5% (n: 20, 22)
Everolimus 10 mg/Day12.5210.947.69
Placebo3.688.483.15

[back to top]

Progression Free Survival According to Chromogramin A Tumor Marker (CgA) Baseline Level and According to CgA Early Response

"Baseline levels of serum CgA SE were characterized towards progression free survival (PFS) as per local investigator assessment, relative to the upper limited of normal (ULN). CgA levels exceeding 2 x ULN were considered to be 'Elevated' otherwise considered as Non-elevated. An 'early response' (applicable to only those patients with elevated levels at baseline) was defined as a decrease of greater than or equal to 30% from baseline to Cycle 2 Day 1 or normalization by Cycle 2 Day 1. CgA is widely expressed in well-differentiated pancreatic NET. CgA is present in the secretory granules of neuroendocrine cells. Pancreatic NET patients often present with elevated circulating levels of CgA in their blood. Baseline levels of these biomarkers are considered as prognostic factors." (NCT00510068)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 17 August 2007, until cut-off date 28 February 2010

,
InterventionMonths (Median)
CgA Levels at baseline: CgA <= 2x ULN (n:121, 97)CgA levels at baseline: CgA > 2x ULN (n:84, 103)Early CgA response: Response (n: 48, 22)Early CgA response: Non-Response (n:40, 82)
Everolimus 10 mg/Day11.178.548.5411.14
Placebo4.904.345.703.19

[back to top]

Plasma Angiogenesis Marker: Vascular Endothelial Growth Factor (VEGF)

This biomarker is related to angiogenesis pathway, was analyzed to determine the effects of everolimus on plasma antiangiogenic molecules. (NCT00510068)
Timeframe: Baseline, Cycle 2 Day 1, Cycle 3 Day 1, Cycle 4 Day 1

,
Interventionpg/mL (Mean)
Baseline (n:198, 195)Cycle 2 Day 1 (n: 185, 184)Cycle 3 Day 1 (n: 185, 174)Cycle 4 Day 1 (n: 171, 159)
Everolimus 10 mg/Day265.09243.03280.18283.51
Placebo326.16326.78292.27319.60

[back to top]

Plasma Angiogenesis Marker: Soluble Vascular Endothelial Growth Factor Receptor 1 (sVEGFR1)

This biomarker is related to angiogenesis pathway, was analyzed to determine the effects of everolimus on plasma antiangiogenic molecules. (NCT00510068)
Timeframe: Baseline, Cycle 2 Day 1, Cycle 3 Day 1, Cycle 4 Day 1

,
Interventionpg/mL (Mean)
Baseline (n:198, 195)Cycle 2 Day 1 (n: 185, 184)Cycle 3 Day 1 (n: 185, 174)Cycle 4 Day 1 (n: 171, 159)
Everolimus 10 mg/Day264.18307.46263.81258.03
Placebo256.69299.03253.37242.17

[back to top]

Plasma Angiogenesis Marker: Placental Growth Factor (PLGF)

This biomarker is related to angiogenesis pathway, was analyzed to determine the effects of everolimus on plasma antiangiogenic molecules. (NCT00510068)
Timeframe: Baseline, Cycle 2 Day 1, Cycle 3 Day 1, Cycle 4 Day 1

,
Interventionpg/mL (Mean)
Baseline (n:198, 195)Cycle 2 Day 1 (n: 185, 184)Cycle 3 Day 1 (n: 185, 174)Cycle 4 Day 1 (n: 171, 159)
Everolimus 10 mg/Day45.8225.7826.5525.69
Placebo32.9235.3833.8435.47

[back to top]

Evaluation of Pharmacokinetics (PK) Parameter: CL/F

The PK parameters for a full PK profile at steady-state were determined in blood using non compartmental methods. The PK parameter clearance of distribution expressed as a function of bioavailability (CL/F). (NCT00510068)
Timeframe: Day 1 of every cycle (28 days/cycle) throughout the study

InterventionL/h (Mean)
Everolimus 10 mg/Day20.2
Everolimus 5 mg/Day10.7

[back to top]

Plasma Angiogenesis Marker: Basic Fibroblast Growth Factor (bFGF)

This biomarker is related to angiogenesis pathway, was analyzed to determine the effects of everolimus on plasma antiangiogenic molecules. (NCT00510068)
Timeframe: Baseline, Cycle 2 Day 1, Cycle 3 Day 1, Cycle 4 Day 1

,
Interventionpg/mL (Mean)
Baseline (n:198, 195)Cycle 2 Day 1 (n: 185, 184)Cycle 3 Day 1 (n: 185, 174)Cycle 4 Day 1 (n: 171, 159)
Everolimus 10 mg/Day52.5938.4351.9751.28
Placebo51.4958.3359.0854.58

[back to top]

Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) (Open-label Period)

Adverse events are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00510068)
Timeframe: on or after the start of open-label study medication until no later than 28 days after open-label study medication discontinuation

InterventionParticipants (Number)
Adverse events (AEs)DeathSerious Adverse Events
Everolimus 10 mg/Day221122108

[back to top]

Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs)

Adverse events are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00510068)
Timeframe: on or after the start of double-blind study medication until no later than 28 days after double-blind study medication discontinuation

,
InterventionParticipants (Number)
Adverse events (AEs)DeathSerious Adverse Events
Everolimus 10 mg/Day20311184
Placebo1982352

[back to top]

Evaluation of Pharmacokinetics (PK) Parameters: Cmax, Cmin

The PK parameters for a full PK profile at steady-state were determined in blood using non compartmental methods. The PK parameter: maximum (peak) drug concentration (Cmax) and minimum (trough) drug concentration (Cmin). (NCT00510068)
Timeframe: Day 1 of every cycle (28 days/cycle) throughout the study

,
Interventionng/mL (Mean)
CmaxCmin
Everolimus 10 mg/Day62.49.80
Everolimus 5 mg/Day27.412.2

[back to top]

Evaluation of Pharmacokinetics (PK) Parameter: Tmax -Time to Maximum (Peak) Drug Concentration

The PK parameters for a full PK profile at steady-state were determined in blood using non compartmental methods. Values for tmax where summarized in median (range). (NCT00510068)
Timeframe: Day 1 of every cycle (28 days/cycle) throughout the study

Interventionh (Median)
Everolimus 10 mg/Day1.17
Everolimus 5 mg/Day3.0

[back to top]

Overall Survival

Overall survival (OS) was defined as the time from date of randomization to the date of death due to any cause. Analyses were performed using all deaths in the FAS population regardless of whether they were observed during the double-blind treatment period, the open-label treatment period, the post-treatment evaluations, or the survival follow-up period. (NCT00510068)
Timeframe: Baseline, to death- no time limit

InterventionMonths (Median)
Everolimus 10 mg/Day44.02
Placebo37.68

[back to top]

Percentage of Participants With Objective Response Rate ( CR {Complete Response} OR PR {Partial Response})

Objective Response defined by RECIST criteria: Partial response (PR) must have ≥ 30% decrease in the sum of the longest diameter of all target lesions, from the baseline sum. Complete response (CR) must have disappearance of all target and non-target lesions. For CR or PR, tumor measurements must be confirmed by 2nd assessments within 4 weeks . Progression = 20% increase in the sum of the longest diameter of all target lesions, from the smallest sum of longest diameter of all target lesions recorded at or after baseline; or a new lesion; or progression of non-target lesions (NCT00510068)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 17 August 2007, until cut-off date 28 February 2010

InterventionPercentage of participants (Number)
Everolimus 10 mg/Day4.8
Placebo2.0

[back to top]

Time to Progression Free Survival (PFS) Based as Per Investigator Using Kaplan-Meier Methodology

Progression of disease is defined as the time from study start to the date of first documented progression of disease or death due to any cause. Progression of disease is defined by RECIST criteria: Progression = 20% increase in the sum of the longest diameter of all target lesions, from the smallest sum of longest diameter of all target lesions recorded at or after baseline; or a new lesion; or progression of non-target lesions. (NCT00510068)
Timeframe: Time from randomisation to dates of disease progression, death from any cause or last tumor assessment, reported between day of first patient randomised, 17 August 2007, until cut-off date 28 February 2010

InterventionMonths (Median)
Everolimus 10 mg/Day11.04
Placebo4.60

[back to top]

Analysis of Time to Definitive Deterioration of WHO Performance Status Using Kaplan-Meier

Time to definitive worsening is defined as a definitive increase in performance status from a baseline of 0 or 1 to WHO >= 2, or from a baseline value of 2 to WHO >= 3. If no earlier deterioration, patients were censored at the end of follow-up or at the start of further antineoplastic therapy. Rates of patients with no deterioration at 3 and 6 months were computed using Kaplan-meier method. Grade 0: Able to carry out all activity without restriction; Grade 1: Restricted in physically strenuous activity but ambulatory & able to do light work; Grade 2: Ambulatory & capable of all self-care but unable to carry out any work. Up & about more than 50% of waking hours; Grade 3: Capable of only limited self-care, confined to bed or chair more than 50% of waking hours; Grade 4: Completely disabled & cannot carry on any self-care; totally confined to bed or chair. (NCT00510068)
Timeframe: 3 months, 6 months

,
Intervention% of participants with no deterioration (Number)
Month 3Month 6
Everolimus 10 mg/Day94.490.6
Placebo91.886.3

[back to top]

Evaluation of Pharmacokinetics (PK) Parameter: AUC0-t Last

The PK parameters for a full PK profile at steady-state were determined in blood using non compartmental methods. This PK parameter is area under the concentration-time curve from time zero to the time of the last quantifiable concentration (AUC0-t last). (NCT00510068)
Timeframe: Day 1 of every cycle (28 days/cycle) throughout the study

Interventionng.h/mL (Mean)
Everolimus 10 mg/Day594
Everolimus 5 mg/Day481

[back to top]

Plasma Angiogenesis Marker: Soluble Vascular Endothelial Growth Factor Receptor 2 (sVEGFR2)

This biomarker is related to angiogenesis pathway, was analyzed to determine the effects of everolimus on plasma antiangiogenic molecules. (NCT00510068)
Timeframe: Baseline, Cycle 2 Day 1, Cycle 3 Day 1, Cycle 4 Day 1

,
Interventionpg/mL (Mean)
Baseline (n:197, 193)Cycle 2 Day 1 (n: 185, 183)Cycle 3 Day 1 (n: 185, 173)Cycle 4 Day 1 (n: 172, 158)
Everolimus 10 mg/Day30061.3022691.1822021.2321218.17
Placebo31299.6130223.2129264.6728308.58

[back to top]

Efficacy Event Data After Month 12 to Month 60

Efficacy events were: Biopsy-proven acute rejection (BPAR), graft loss, death, and treatment failure (defined as composite endpoint of BPAR, graft loss, death, loss to follow-up, discontinuation due to lack of efficacy or due to toxicity). (NCT00514514)
Timeframe: Events starting after Month 12

,,
InterventionParticipants (Number)
BPARGraft lossDeathLost to follow-upDiscontinuation due to adverse eventTherapy failure (composite endpoint)
CNI Free Regimen137415835
CNI Low Regimen123913436
Standard Regimen1377171038

[back to top]

Efficacy Event Data Baseline 2 (Month 3) to Month 12

Efficacy events were: Biopsy-proven acute rejection (BPAR), graft loss, death, and treatment failure (defined as composite endpoint of BPAR, graft loss, death, loss to follow-up, discontinuation due to lack of efficacy or due to toxicity). (NCT00514514)
Timeframe: From Baseline 2 (Month 3) to Month 12

,,
InterventionParticipants (Number)
BPARGraft lossDeathLost to follow-upDiscontinuation due to lack of efficacyDiscontinuation due to adverse eventTherapy failure composite
CNI Free Regimen2022034458
CNI Low Regimen1312012735
Standard Regimen1313022534

[back to top]

Efficacy Event Data From Baseline 2 (Month 3) to Month 6

Efficacy events were: Biopsy-proven acute rejection (BPAR), graft loss, death, and treatment failure (defined as composite endpoint of BPAR, graft loss, death, loss to follow-up, discontinuation due to lack of efficacy or due to toxicity). (NCT00514514)
Timeframe: From Baseline 2 (Month 3) to Month 6

,,
InterventionParticipants (Number)
BPARGraft lossDeathLost to follow-upDiscontinuation due to lack of efficacyDiscontinuation due to adverse eventTherapy failure composite
CNI Free Regimen1511022637
CNI Low Regimen1010011319
Standard Regimen60101814

[back to top]

Change From BL2 (Month 3) to Month 12 in Cardiovascular Risk (Framingham Score; 10-year Cardiovascular Risk)

The Framingham Score (based on LDL cholesterol level) estimates the coronary heart disease risk (%) of developing one of the following coronary heart diseases: angina pectoris, myocardial infarction, or coronary disease death, over the course of 10 years. (NCT00514514)
Timeframe: From Baseline 2 (Month 3) to Month 12

,,
InterventionPercent risk (Mean)
Baseline 1/Visit 1 (n=165,171,161)Baseline 2/Month 3 (n=165,171,161)Month 12 (n=158,166,156)Change from Baseline 2 to Month 12 (n=158,166,156)
CNI Free Regimen10.28.89.10.4
CNI Low Regimen9.59.38.7-0.7
Standard Regimen10.910.39.4-0.7

[back to top]

GFR at Month 60 Utilizing Cockcroft-Gault Formula

Cockcroft-Gault formula: For men: GFR= ((140-age) × body weight in kg)∕(72 x serum creatinine in mg∕dl) For women: GFR= (0.85×(140-age) × body weight in kg)∕(72 x serum creatinine in mg/dl), ), last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 60 post-transplant

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen55.92
CNI Free Regimen61.6
CNI Low Regimen52.91

[back to top]

GFR at Month 12 Utilizing Cockcroft-Gault Formula

Cockcroft-Gault formula: For men: GFR= ((140-age) × body weight in kg)∕(72 x serum creatinine in mg∕dl)For women: GFR= (0.85×(140-age) × body weight in kg)∕(72 x serum creatinine in mg/dl), ), last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 12 post-transplant

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen60.18
CNI Free Regimen64.87
CNI Low Regimen61.16

[back to top]

GFR at Month 12 Utilizing Modification of Diet in Renal Disease (MDRD) Method

"Change in GFR (Modification of Diet in Renal Disease calculated using the -MDRD formulat:~For men: GFR = 170 × (serum creatinine -0,999)×(age-0,176) x (urea nitrogen -0,17) × (albumin0,318) For women: GFR = 170 × (serum creatinine -0,999) × (age-0,176) × (urea nitrogen -0,17) x (albumin0,318) × 0.762 with urea nitrogen = urea / 2.144. ), last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model, with treatment, center, donor type (deceased vs. living) as factors and BL2-value at V4/M3/BL2 as covariate." (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 12 post-transplant

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen50.23
CNI Free Regimen56.36
CNI Low Regimen50.24

[back to top]

Mean Change in Serum Creatinine From Month 3 to Month 60

Change in venous blood serum creatinine. Last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 60 post-transplant

Interventionmg/dl (Least Squares Mean)
Standard Regimen1.94
CNI Free Regimen1.69
CNI Low Regimen2.01

[back to top]

GFR at Month 60 Utilizing Modification of Diet in Renal Disease (MDRD) Method

"Change in GFR (Modification of Diet in Renal Disease calculated using the -MDRD formulat:~For men: GFR = 170 × (serum creatinine -0,999)×(age-0,176) x (urea nitrogen -0,17) × (albumin0,318) For women: GFR = 170 × (serum creatinine -0,999) × (age-0,176) × (urea nitrogen -0,17) x (albumin0,318) × 0.762 with urea nitrogen = urea / 2.144. ), last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model, with treatment, center, donor type (deceased vs. living) as factors and BL2-value at V4/M3/BL2 as covariate." (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 60 post-transplant

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen47.56
CNI Free Regimen53.41
CNI Low Regimen44.79

[back to top]

GFR Calculated Via Nankivell Formula at Month 60

Change in GFR using the Nankivell formula (GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)² + C where where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kilograms, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. The calculated GFR is expressed in mL/min per 1.73m², last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model, with treatment, center, donor type (deceased vs. living) as factors and BL2-value at V4/M3/BL2 as covariate. (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 60

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen60.24
CNI Free Regimen66.98
CNI Low Regimen58.74

[back to top]

GFR Via Nankivell Formula at Month 12 - All Regimens

Change in GFR using the Nankivell formula (GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)² + C where where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kilograms, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. The calculated GFR is expressed in mL/min per 1.73m², last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model, with treatment, center, donor type (deceased vs. living) as factors and BL2-value at V4/M3/BL2 as covariate. (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 12 post-transplant

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen63.03
CNI Free Regimen68.59
CNI Low Regimen63.08

[back to top]

GFR Via Nankivell Method at Month 12 - CNI-Free vs Standard Regimen

Demonstrate superiority of CNI-Free vs Standard Regimen in GFR using the Nankivell formula (GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)² + C where where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kilograms, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. The calculated GFR is expressed in mL/min per 1.73m², last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model, with treatment, center, donor type (deceased vs. living) as factors and BL2-value at V4/M3/BL2 as covariate. P-values are not adjusted (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 12 post-transplant

Interventionml/min/1.73m² (Least Squares Mean)
Standard Regimen63.03
CNI Free Regimen68.59
CNI Low Regimen63.08

[back to top]

Mean Change in Serum Creatinine From Month 3 to Month 12

Change in venous blood serum creatinine. Last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT00514514)
Timeframe: From randomization at BL2 (Month 3) to Month 12 post-transplant

Interventionmg/dl (Least Squares Mean)
Standard Regimen1.66
CNI Free Regimen1.58
CNI Low Regimen1.76

[back to top]

Surgery Group: Progression-free Survival

Progression-free survival (PFS) was assessed using Gadolinium chelate-enhanced Magnetic Resonance Imaging (MRI) and based on the Neuro-Oncology Criteria for Tumor Response for CNS tumors. PFS was measured from the first day of treatment after surgery to disease progression or death and is derived using the Kaplan-Meier method. (NCT00515086)
Timeframe: After surgery (within 96 hours), Weeks 4 and 8 and then every 8 weeks after restarting treatment until study discontinuation (Up to 28 weeks)

InterventionWeeks (Median)
Everolimus 10 mg + Surgery25.9
Everolimus 5 mg + Surgery9.1
Everolimus 0 mg + SurgeryNA
Total14.9

[back to top]

No Surgery Group: Progression Free Survival

Progression-free survival (PFS) was assessed using Gadolinium chelate-enhanced Magnetic Resonance Imaging (MRI) and based on the Neuro-Oncology Criteria for Tumor Response for CNS tumors. PFS is reported for participants with 1 previous relapse and participants with ≥2 previous relapses. PFS was measured from the first day of treatment to disease progression or death and is derived using the Kaplan-Meier method. (NCT00515086)
Timeframe: First day of treatment to study discontinuation (up to 60 weeks)

InterventionWeeks (Median)
No Surgery (1 Previous Relapse)4.14
No Surgery ( ≥ 2 Previous Relapses)4.14
Total : No Surgery4.14

[back to top]

Surgery Group: Number of Participants With Adverse Events

The number of participants with any adverse event by System Organ Class. Additional information about Adverse Events can be found in the Adverse Event Section. (NCT00515086)
Timeframe: First day of treatment to study discontinuation (Up to 28 weeks)

,,
InterventionParticipants (Number)
Gastrointestinal disordersNervous system disordersGeneral disorders & administration site conditionsMetabolism and nutrition disordersBlood and lymphatic system disordersInfections and infestationsPsychiatric disordersSkin and subcutaneous tissue disordersInvestigationsInjury, poisoning, and procedural complicationsMusculoskeletal and connective tissue disordersRespiratory, thoracic and mediastinal disordersRenal and urinary disordersVascular disordersImmune system disordersEye disordersEar and labyrinth disordersReproductive system and breast disordersCardiac disordersEndocrine disordersSurgical and medical proceduresNeoplasms benign, malignant and unspecified
Everolimus 0 mg + Surgery3553532210222210001000
Everolimus 10 mg + Surgery5446445435100211200110
Everolimus 5 mg + Surgery5544223213121111010000

[back to top]

No Surgery Group: Best Overall Tumor Response

The best overall tumor response is reported for participants with 1 previous relapse and participants with ≥2 previous relapses according to the following categories: Complete Response, Partial Response, Stable Disease and Progressive Disease. Gadolinium chelate-enhanced Magnetic Resonance Imaging (MRI) was used for tumor analysis. The objective assessment of tumor response was evaluated at each site by the designated pathologist based on the Neuro-Oncology criteria for Tumor Response for Central Nervous System (CNS) tumors. (NCT00515086)
Timeframe: First day of treatment to study discontinuation (up to 60 weeks)

,
InterventionParticipants (Number)
Complete response + Partial responseComplete responsePartial responseStable diseaseProgressive disease
No Surgery ( ≥ 2 Previous Relapses)00066
No Surgery (1 Previous Relapse)00037

[back to top]

Time to Progression

3.9 months with a CI of 21- (NCT00516165)
Timeframe: 2 years

Interventionmonth (Median)
RAD0013.9

[back to top]

Progression-free Survival Rate at 24 Weeks

"Progression was defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions~This information will be collected during two years of patient participation." (NCT00516165)
Timeframe: 2 years

Interventionmonths (Median)
RAD0013.8

[back to top]

Overall Survival

The median overall survival was 8.4 months (95% CI, 3.9-21.1 months). Only 2 ((8 %) patients were progression-free at 24 weeks. The study did not proceed to the second stage of the phase 2 portion of the study. (NCT00516165)
Timeframe: 2 years

Interventionmonths (Median)
RAD0018.4

[back to top]

Number of Patients With Adverse Events Who Were Treated With RAD001 for Advanced HCC

Everolimus given at 10 mg/day as a single agent was well tolerated in patients with advanced HCC. (NCT00516165)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
RAD00128

[back to top]

Maximum Tolerated Dose of RAD001 in Patients With Advanced Hepatocellular Carcinoma (HCC).

(NCT00516165)
Timeframe: 2 years

Interventionmg (Number)
RAD00110

[back to top]

Overall Response Rate

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT00516165)
Timeframe: 2 years

Interventionpercentage of patient response (Number)
RAD0014

[back to top]

Time to Disease Progression

Time to disease progression is defined as the time from registration to the earliest date documentation of disease progression. If a patient dies without a documentation of disease progression the patient will be considered to have had tumor progression at the time of their death unless there is sufficient documented evidence to conclude no progression occurred prior to death. The distribution of time to progression will be estimated using the method of Kaplan-Meier. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00521001)
Timeframe: Time from registration to the earliest date documentation of disease progression; Up to 5 years

Interventionmonths (Median)
Everolimus + Temozolomide2.4

[back to top]

9-week Progression-free Survival Rate

The primary endpoint of this trial is the 9 week PFS rate. A patient is a success if they are progression free at their cycle 2 evaluation (approximately 9 weeks post registration). All patients, who meet the eligibility criteria, sign a consent form, and start treatment will be included in the evaluation of the 9-week PFS rate (evaluable patients). The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated using the properties of the binomial distribution. If some patients are lost to follow up prior to their cycle 2 evaluation, the Kaplan-Meier method will be used to estimate the 9 week PFS rate. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00521001)
Timeframe: at 9 weeks

Interventionproportion of patients (Number)
Everolimus + Temozolomide0.44

[back to top]

Confirmed Response Rate (Complete Response and Partial Response)

Confirmed response rates will be evaluated by dividing the number of confirmed responders (i.e. patients that achieve a CR or PR on consecutive evaluations) by the total number of evaluable patients. Confidence intervals for the true response rate will be calculated using the properties of the binomial distribution. (NCT00521001)
Timeframe: Up to 5 years

Interventionpercentage of confirmed responses (Number)
Everolimus + Temozolomide8.3

[back to top]

Survival Time

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT00521001)
Timeframe: Time from registration to death due to any cause; Up to 5 years

Interventionmonths (Median)
Everolimus + Temozolomide8.6

[back to top]

Change in PSA

Time-to-event data, such as change in PSA will be summarized using the method of Kaplan and Meier. (NCT00526591)
Timeframe: Up to 16 weeks after start of study

Interventionng/mL (Median)
High-dose Cohort3.5
Low-dose Cohort3.04

[back to top]

Proportion of Patients Who Are P0 (i.e., no Clinically Detectable Tumor in the Pathologic Specimen) at Surgery

Specimens are fixed in formalin for 24 hours.Specimens are the cut at 3 mm intervals perpendicular to the rectal surface and the sections are examined grossly and microscopically on routine Hematoxylin and Eosin stain (H&E) (pathologic complete response or P0) will be defined as responders. (NCT00526591)
Timeframe: After 8 weeks of therapy at the time of prostatectomy

Interventionparticipants (Number)
Low-dose Cohort0
High-dose Cohort0

[back to top]

Toxicity Profile of Each Dose (Number of Patients With Worst Grade Toxicity)

Toxicity will be assessed using the NIH-NCI Common Terminology Criteria for Adverse Events, version 3.0 (CTCAEv3.0) (NCT00526591)
Timeframe: at daily dose for 8 weeks

,
Interventionparticipants (Number)
Patients with Grade 3 toxicityPatients with Grade 4 toxicity
High-dose Cohort10
Low-dose Cohort20

[back to top]

Percent Change in FDG-PETUptake Following 2 Weeks of Therapy

The secondary objective was to explore whether an early change in FDG-PET uptake is associated with tumor shrinkage. Change in FDG-PET uptake was calculated using the baseline and 2-week FDG-PET scans. (NCT00529802)
Timeframe: 2 weeks

Intervention% change in avgSUVmax at 2 weeks (Mean)
Patients Evaluable for Secondary Outcome-29.7

[back to top]

Relative Tumor Size Change Following 8 Weeks of Therapy.

The primary objective is to determine whether high SUV uptake on FDG-PET is associated with greater tumor shrinkage. Tumor size is defined as the sum of unidimensional tumor measurements from standard CT imaging calculated according to RECIST criteria. Tumor size is measured at baseline and after eight weeks of therapy. Tumor shrinkage is the relative change (%) in tumor size from baseline. (NCT00529802)
Timeframe: 8 weeks

InterventionPercent change from baseline (Mean)
Low Uptake1.4
High Uptake-0.57

[back to top]

Revascularizations

(TLR/TVR/any revascularization)both ischemia-driven and not ischemia-driven. (NCT00531011)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
XIENCE V0
TAXUS® Liberté™0

[back to top]

Distal Minimum Lumen Diameter (MLD).

Distal refers to the immediate 5 mm outside of the distal end of the stent. (NCT00531011)
Timeframe: at 9 months.

Interventionmillimeters (Mean)
XIENCE V2.28
TAXUS® Liberté™2.25

[back to top]

In-stent Late Loss (LL)

Full Analysis Set (FAS). LL is defined as the difference between the post-procedure (immediately post placement of the stent) minimal lumen diameter (MLD) and the follow-up MLD (at 270 days). In stent is measured within the confines of the stent edges. (NCT00531011)
Timeframe: at 270 days

Interventionmillimeters (Mean)
XIENCE V0.052
TAXUS® Liberté™0.238

[back to top]

In-stent Minimum Lumen Diameter (MLD).

(NCT00531011)
Timeframe: at 9 months.

Interventionmillimeters (Mean)
XIENCE V2.29
TAXUS® Liberté™2.17

[back to top]

Composite Rate of Cardiac Death, Myocardial Infarction (MI, Both Q-wave and Non Q-wave), and Ischemia-driven Target Lesion Revascularization (TLR) .

This measure is a calculation of the percentage of participants who experience any of the components of this composite measure. (NCT00531011)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
XIENCE V1
TAXUS® Liberté™2.1

[back to top]

Composite Rate of All Death, MI (Q-wave and Non Q-wave), and Target Vessel Revascularization (TVR).

(NCT00531011)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
XIENCE V1
TAXUS® Liberté™2.1

[back to top]

Device Success

defined as achievement of a final residual in-stent diameter stenosis of < 30% (visual assessment) using the assigned device only. (NCT00531011)
Timeframe: at the time of PCI

Interventionpercentage of participants (Number)
XIENCE V97.7
TAXUS® Liberté™98.2

[back to top]

Composite Endpoint of All Death, MI (Q-wave and Non Q-wave), and TVR.

(NCT00531011)
Timeframe: 9 months

Interventionpercentage of participants (Number)
XIENCE V6.8
TAXUS® Liberté™12.4

[back to top]

Procedural Success

defined as: residual in-stent %DS of < 30% using a percutaneous method, without cardiac death, Q-wave MI, non Q-wave MI, or repeat revasc of the target during hospitalization. (NCT00531011)
Timeframe: at the time of PCI

Interventionpercentage of participants (Number)
XIENCE V98.1
TAXUS® Liberté™97

[back to top]

Adjudicated Stent Thrombosis.

(NCT00531011)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
XIENCE V0
TAXUS® Liberté™0

[back to top]

Adjudicated Stent Thrombosis.

(NCT00531011)
Timeframe: 9 months

Interventionpercentage of participants (Number)
XIENCE V0
TAXUS® Liberté™0

[back to top]

Composite Endpoint of Cardiac Death, MI (Q-wave and Non Q-wave), and Ischemia-driven TLR .

ITT (NCT00531011)
Timeframe: 9 months

Interventionpercentage of participants (Number)
XIENCE V4.9
TAXUS® Liberté™7.2

[back to top]

Lesion Success

defined as attainment of < 30% residual in-stent stenosis (by visual assessment) using any percutaneous method. (NCT00531011)
Timeframe: at the time of PCI

InterventionPercentage of participants (Number)
XIENCE V99.2
TAXUS® Liberté™99.6

[back to top]

In-segment Binary Restenosis Rate

"This measures the percentage of patients who have > 50% diameter stenosis of the assessed vessel, within the stent edges~In-segment is measured within the confines of the stent edges plus within 5 mm on either side of the stent." (NCT00531011)
Timeframe: at 9 months

Interventionpercentage of participants (Number)
XIENCE V6.2
TAXUS® Liberté™4.6

[back to top]

In-segment Late Loss (LL)

LL is defined as the difference between the post-procedure (immediately post placement of the stent) minimal lumen diameter (MLD) and the follow-up MLD (at 270 days). In segment LL is measured within the confines of the stent edges and within 5 mm of those edges. (NCT00531011)
Timeframe: at 9 months

Interventionmillimeters (Mean)
XIENCE V-0.100
TAXUS® Liberté™0.037

[back to top]

In-segment Minimum Lumen Diameter (MLD).

(NCT00531011)
Timeframe: at 9 months.

Interventionmillimeters (Mean)
XIENCE V2.10
TAXUS® Liberté™2.04

[back to top]

In-stent Binary Restenosis Rate

This measures the percentage of patients who have > 50% diameter stenosis of the assessed vessel, within the stent edges. (NCT00531011)
Timeframe: at 9 months

Interventionpercentage of participants (Number)
XIENCE V5.8
TAXUS® Liberté™3.6

[back to top]

Proximal Minimum Lumen Diameter (MLD).

Proximal refers to the immediate 5 mm outside of the proximal end of the stent. (NCT00531011)
Timeframe: at 9 months.

Interventionmillimeters (Mean)
XIENCE V2.67
TAXUS® Liberté™2.61

[back to top]

Revascularizations

(TLR/TVR/any revascularization)both ischemia-driven and not ischemia-driven. (NCT00531011)
Timeframe: 9 months

Interventionpercentage of participants (Number)
XIENCE V5.8
TAXUS® Liberté™11.3

[back to top]

Overall Survival Time

Overall survival: The overall survival or survival time is defined as the time from registration to death due to any cause. The distribution of overall survival will be estimated using the method of Kaplan-Meier method. (NCT00553150)
Timeframe: Up to 15 years

Interventionmonths (Median)
Phase II15.8

[back to top]

Maximum Tolerated Dose (MTD) of Everolimus (RAD001) in Combination With Temozolomide (TMZ) and 3D-conformal Radiotherapy (RT) or Intensity-modulated Radiotherapy (IMRT) Followed by Adjuvant TMZ With or Without RAD001 (Phase I)

Patients were assessed during RT for dose-limiting toxicities (DLT), which were defined as failure to deliver greater than 75% of the planned doses of TMZ or RAD001 during RT, interruption of RT for more than 5 days because of toxicity, or the following: >= Grade 3 diarrhea or skin rash; >= Grade 4 neutropenia, leukopenia, or thrombocytopenia; >= Grade 4 hypertriglyceridemia, hypercholesterolemia, or hyperglycemia despite optimal medial management, other >= 3 non-hematologic events; or >= Grade 4 radiation dermatitis. Maximum tolerated dose (MTD) was defined a priori as the highest dose level at which 0 or 1 of 6 patients developed DLTs. The number of patients who developed DLTs are reported here by dose level, with the MTD reported in the statistical analysis section. (NCT00553150)
Timeframe: Up to 49 days

Interventionparticipants who developed DLTs (Number)
Phase I: Dose Level 01
Phase I: Dose Level 11
Phase I: Dose Level 21

[back to top]

Progression-free-survival at 6 Months (Phase II)

Progression-free-survival at 6 months: is the proportion of patients alive and progression-free at 6 months after start of regimen. This proportion will be estimated using the binomial point estimator and the binomial 95% confidence interval estimated. Progression is defined as at least a 25% increase in product of perpendicular diameters of contrast enhancement or mass or unequivocal increase in size of contrast enhancement or increase in mass effect as agreed upon independently by primary physician and quality control physicians or appearance of new lesions. (NCT00553150)
Timeframe: at 6 months

Interventionproportion of participants (Number)
Phase II0.52

[back to top]

Response Rate, as Measured in Patients Receiving FLT-PET Imaging (Phase II)

The response rate is defined as the percentage of patients receiving F-fluorothymidine positron emission tomography (FLT-PET) imaging whose cancer shrinks or disappears after treatment. A reduction in standardized uptake value (SUV) of 30% or greater in the T1-post-gadolinium scan volume of interest (T1-gad VOI) or the total tumor VOI will be considered a responsive tumor. (NCT00553150)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Phase II44.4

[back to top]

Overall Survival at 12 Months (Phase II)

"The primary endpoint is overall survival at 12 months (OS12) after entry into this study. The proportion of successes will be estimated using the binomial point estimator (number of successes divided by the total number of evaluable patients) and the binomial 95% confidence interval estimated. A patient who is evaluable and survive more than 12 months (i.e. 365 days or more) after start of therapy will be classified as a success. Patients who die within 12 months after start of therapy will be considered to have failed." (NCT00553150)
Timeframe: at 12 months

Interventionproportion of participants (Number)
Phase II0.64

[back to top]

Time to Progression (Phase II)

Time-to-disease progression is defined as the time from start of study therapy to documentation of disease progression. Patients who die without documentation of progression will be considered to have had tumor progression at the time of death unless there is documented evidence that no progression occurred before death. Patients who fail to return for evaluation after beginning therapy will be censored for progression on the last day of therapy. Patients who experience major treatment violations will be censored for progression on the date of treatment violation occurred. The time-to-progression distribution will be estimated using the Kaplan-Meier method. Progression is defined as at least a 25% increase in product of perpendicular diameters of contrast enhancement or mass or unequivocal increase in size of contrast enhancement or increase in mass effect as agreed upon independently by primary physician and quality control physicians or appearance of new lesions. (NCT00553150)
Timeframe: Up to 5 years

Interventionmonths (Median)
Phase II6.4

[back to top]

Time to Progression

(NCT00570921)
Timeframe: Duration of time start of treatment to time of documented progression or death

Interventionmonths (Median)
Fulvestrant + Everolimus7.4

[back to top]

Objective Response Rates

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT00570921)
Timeframe: Evaluated 60 days after therapy start

Interventionparticipants (Number)
Fulvestrant + Everolimus4

[back to top]

Clinical Benefit Rate

Clinical benefit rate is defined as a complete response, partial response, or stable disease (CR, PR, SD) by Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for a minimum of at least 24 weeks or more. (NCT00570921)
Timeframe: Duration of response or stable disease for 24 weeks or more

Interventionparticipants (Number)
Fulvestrant + Everolimus15

[back to top]

Response Rate

To determine the objective response rate by RECIST criteria of RAD001 in combination with temozolomide in patients with advanced pancreatic neuroendocrine tumors. Partial response (PR) by these criteria is defined as at least a 30% decrease in the sum of the longest diameter of target lesions, taking as reference the baseline sum longest diameter. Progressive disease (PD) is defined as at least a 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since the treatment started or the appearance of one or more new lesions. Stable Disease (SD) is defined as neither sufficient decrease to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum longest diameter since the treatment started. (NCT00576680)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Temozolomide and Everolimus16

[back to top]

Progression-free Survival

To determine progression-free survival when RAD001 is given in combination with temozolomide in patients with advanced pancreatic neuroendocrine tumors. Progression-free survival is defined as time from start of therapy until disease progression, as defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST), as 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since the treatment started or the appearance of one or more new lesions, or death. (NCT00576680)
Timeframe: 2 years

Interventionmonths (Median)
Temozolomide and Everolimus15.4

[back to top]

To Determine the Safety and Tolerability of This Drug Combination.

To determine the safety and tolerability of RAD001 when given in combination with temozolomide in patients with advanced pancreatic neuroendocrine tumors. (NCT00576680)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Grade 3 or 4 LymphopeniaGrade 3 or 4 thrombocytopeniaGrade 3 or 4 mucositisGrade 3 or 4 hyperglycemiaGrade 3 or 4 AST IncreaseGrade 3 or 4 Leukocyte decrease
Temozolomide and Everolimus1971847

[back to top]

Percentage of Patients in Each Study Arm With Increase of ≥1 Point in the Ishak-Knodell Staging Score in Fibrosis

Ishak-Knodell Score: 0=No fibrosis; 01=Fibrous expansion of some portal areas, with or without short fibrous septa; 02=Fibrous expansion of most portal areas, with or without short fibrous septa; 03=Fibrous expansion of most portal areas, with occasional portal to portal (P-P) bridging; 04=Fibrous expansion of portal areas, with marked bridging (portal to portal (P-P) as well as portal to central (P-C)); 05=Marked bridging (P-P and/or P-C) with occasional nodules (incomplete cirrhosis); 06=Cirrhosis, probable or definite. (NCT00582738)
Timeframe: baseline to month 24

Interventionpercentage of participants (Number)
Standard Treatment38.9
Everolimus7.1

[back to top]

Change From Baseline in Fibrosis Staging Score (Measured by the Ishak-Knodell Staging Score) Between Baseline and 24 Months Post-transplant.

"Ishak-Knodell Score: 0=No fibrosis; 01=Fibrous expansion of some portal areas, with or without short fibrous septa; 02=Fibrous expansion of most portal areas, with or without short fibrous septa; 03=Fibrous expansion of most portal areas, with occasional portal to portal (P-P) bridging; 04=Fibrous expansion of portal areas, with marked bridging (portal to portal (P-P) as well as portal to central (P-C)); 05=Marked bridging (P-P and/or P-C) with occasional nodules (incomplete cirrhosis); 06=Cirrhosis, probable or definite~Decrease in score from baseline indicates improvement" (NCT00582738)
Timeframe: baseline, 24 Months

InterventionScore on Scale (Median)
CsA-TAC-0.5
Everolimus0.0

[back to top]

Comparison of Renal Function (Glomerular Filtration Rate [GFR] Calculated Using the Modification of Diet in Renal Disease Study Group [MDRD] Formula) Between Study Groups

"GFR Month 9 value if available, otherwise minimal first year post-randomization available value. Imputation rule of missing Month 24 GFR values: GFR Month 18 value if available, otherwise Month 12 GFR is used.~Least square means are from an ANCOVA model containing treatment as factor and baseline eGFR as a covariate." (NCT00582738)
Timeframe: 12 months, 24 months/EOS

,
InterventionmL/min/1.73^2 (Least Squares Mean)
12 Months (n=21, 22)24 months (n=20, 18)
CsA-TAC62.265.5
Everolimus65.671.6

[back to top]

Percentage of Patients With Death, Graft Loss and Biopsy Proven Acute Rejection (BPAR) Between Study Groups

(NCT00582738)
Timeframe: 24 Months

,
InterventionPercentage of Participants (Number)
DeathGraft LossAcute Rejection (BPAR)
CsA/TAC000
Everolimus4.54.50.0

[back to top] [back to top]

Comparison of the Effect of Both Regimens on the Inflammatory (Acti-test) and Fibrosis (Fibro-test) Components of Fibrosure, and on Fibrosis Area Assessed by Histomorphometry

"The Fibrosure test is the combination of Fibro-test + Acti-test.~FibroTest (FT) was for the assessment of fibrosis. Fibro test was calculated using an original combination of five highly concentrated serum biochemical markers; alpha2macroglobulin, haptoglobin, apolipoprotein A1, total bilirubin and gammaglutamyltransferase (GGT). FibroTest scores range from 0.00 to 1.00 where 0.0-0.21 is no fibrosis and >= 0.59 is cirrhosis.~Acti-test was calculated using 6 serum biochemical markers; alpha2macroglobulin, haptoglobin, apolipoprotein A1, total bilirubin, GGT and alanine aminotransferase (ALT). ActiTest (AT) was used for the assessment of necroinflammatory activity. Test score ranges from 0.00 to 1.00, where 0.00-0.17 indicates no necrosis and >= 0.61 indicates severe necrosis~If 12-month Actitest value was the last available assessment, the value is used to impute the final staging score(End of Study)" (NCT00582738)
Timeframe: baseline, 12 and 24 months

,,,
Interventionunits on a scale (Median)
Baseline (n=21,21,21,21)month 12 (n=20,17, 20, 17)month 24 (n=1,2,1,2)end of study [month 24] (n=20,21,20,21)
Everolimus - Summary of Fibrotest by Treatment0.800.801.00.70
Everolimus -Summary of Actitest by Treatment0.500.500.700.50
Standard Treatment - Summary of Actitest by Treatment0.400.600.600.60
Standard Treatment -Summary of Fibrotest by Treatment0.800.800.900.80

[back to top]

Comparison of the Effect of Both Regimens in the Necroinflammatory Grading Score (Ishak-Knodell) (Portal Inflammation)

Ishak-Knodell Score: 0=No fibrosis; 01=Fibrous expansion of some portal areas, with or without short fibrous septa; 02=Fibrous expansion of most portal areas, with or without short fibrous septa; 03=Fibrous expansion of most portal areas, with occasional portal to portal (P-P) bridging; 04=Fibrous expansion of portal areas, with marked bridging (portal to portal (P-P) as well as portal to central (P-C)); 05=Marked bridging (P-P and/or P-C) with occasional nodules (incomplete cirrhosis); 06=Cirrhosis, probable or definite (NCT00582738)
Timeframe: baseline, 12 months, 24 months

,
InterventionScore on a scale (Mean)
Baseline (n= 18, 14)Month 12 (n = 18, 14)Month 24 (n = 8, 5)
CsA-TAC1.81.92.1
Everolimus2.41.92.0

[back to top]

Change From Baseline in Hepatitis C Virus (HCV) Ribonucleic Acid (RNA) Viral Load at 12 and 24 Months Post Randomization

End of Study (EOS) endpoint is the last available assessment on or after Month 12. A reduction of at least two logs in HCV RNA viral load was considered as success (NCT00582738)
Timeframe: baseline, 12 months, 24 months/EOS

,
Interventionlog10 copies/ml (Mean)
baseline12 months (n=20, 20)24 months/EoS (n=20, 20)Month 12 - baseline (n=20,20)Monh 24 - baseline (n=20,20)
CsA-TAC6.66.56.9-0.10.3
Everolimus6.46.66.70.20.3

[back to top]

Change From Baseline in Fibrosis Metavir Scoring at 12 and 24 Months Post Randomization

Metavir Score: F0=No fibrosis; F1=Portal fibrosis without septa; F2=Portal fibrosis with rare septa; F3=Numerous septa without cirrhosis Decrease in score from baseline indicates improvement (NCT00582738)
Timeframe: Baseline, 12 months, 24 months

,
InterventionScores on a Scale (Median)
Baseline (n=18, 14)Month 12 (n=18, 14)Month 24 (n=8, 5)Month 12-Baseline (n=18, 14)Month 24-Baseline (n=8, 5)
CsA-TAC1.01.01.00.00.0
Everolimus1.51.01.00.00.0

[back to top]

Objective Response Rate (ORR)

The percentage of patients who experience an objective benefit from treatment (CR+PR). The response categories were assigned using RECIST criteria. Complete Response (CR) = Disappearance of all target lesions ; Partial Response (PR) = >=30% decrease in the sum of the longest diameter of target lesions. (NCT00591734)
Timeframe: 13 months

InterventionParticipants (Count of Participants)
Intervention7

[back to top]

Overall Survival Rate

Overall Survival is defined as the length of time, in months, that patients were alive from their first date of protocol treatment until death. For patients who were alive at the time of calculation, follow-up time was censored at date of last contact. The percentage of patients who were alive at 1 year is reported here. This was estimated using the Kaplan Meier method. (NCT00591734)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Intervention43

[back to top]

Progression-free Survival

Length of time, in months, that patients were alive from their first date of protocol treatment until worsening of their disease (NCT00591734)
Timeframe: 13 months

Interventionmonths (Median)
Intervention4

[back to top]

Progression Free Survival (PFS)

8 week PFS (NCT00597506)
Timeframe: interval between start of treatment and 8-week

Interventionproportion of participants (Number)
Bevacizumab and Everolimus0.58

[back to top]

Overall Response

Overall response is composed of complete responses and partial responses. Complete response (CR): disappearance of all target lesions; Partial response: at least a 30 percent decrease in the sum of the longest diameter of the target lesions taking as reference the baseline sum longest diameter. Response is assessed at each subject's restaging, approximately ever 2 months. (NCT00597506)
Timeframe: Measured 1 month after the last treated subject came off treatment

Interventionpercentage of participants (Number)
Bevacizumab and Everolimus0

[back to top]

Net Change Relative to Baseline in Tumor Blood Flow

Tumor blood flow (ml/min/100gm) determined by functional computed tomography (CT). Functional computed tomography (CT) at baseline, after first and third cycles (21 day cycles). Change (percentage) calculated as tumor blood flow measured at baseline compared to tumor blood flow measurement taken at end of Cycle 1, week 3 (21 days), and again at end of Cycle 3, Week 9 (63 days). (NCT00607113)
Timeframe: Baseline to end of Cycle 3 (63 days)

,
Interventionml/min/100gm (Mean)
Baseline to Week 3Baseline to Week 9
Avastin-0.44-0.60
RAD001-0.12-0.40

[back to top]

In-Stent Percent Diameter Stenosis

In Stent Percent Diameter Stenosis at thirteen months. In Stent Percent Diameter Stenosis: measured percent of diameter stenosis at the region of the stent (calculated as 100x(RVD-MLD)/RVD using the mean values from 2 orthogonal views by QCA. RVD (Reference Vessel Diameter): average of normal segments within 10mm proximal and distal to target lesion from 2 orthogonal views using QCA. MLD (Minimal Lumen Diameter): average of 2 orthogonal views of the narrowest point wihtin the area of assessment. MLD measured during QCA by the angiographic core laboratory. (NCT00617084)
Timeframe: 13 Months

InterventionPercentage diameter stenosis (Mean)
1. Resolute - 12-13 Months21.65
2. XIENCE V - 12-13 Months19.76

[back to top]

Target Lesion Failure

Percentage of participants that had either Cardiac Death, Myocardial Infarction (not clearly attributable to a non-target vessel)or Target Lesion Revascularization (TLR, clinically indicated) after one year. MI: Q MI if new pathological Q waves and chest pain, non Q MI if CK elevated more than two times normal, troponin elevated more than normal, according to ARC definitions. TLR, clinically indicated if associated with ischemic symptoms and angiographic min lumen diameter bigger than fifty percent by QCA or without symptoms and min lumen diameter bigger than seventy percent. Measure average. (NCT00617084)
Timeframe: 12 months

Interventionpercentage of participants (Number)
1. Resolute - 12-13 Months8.2
2. XIENCE V - 12-13 Months8.3

[back to top]

Incidence Rate of Composite Efficacy Failure From Randomization to Month 24

"Composite efficacy failure was defined as treated biopsy proven acute rejection (tBPAR), graft loss, or death.~The incidence rates of composite efficacy failure were estimated with a Kaplan-Meier product-limit formula." (NCT00622869)
Timeframe: Randomization to Month 24

InterventionPercentage (Number)
Everolimus + Reduced Tacrolimus10.3
Tacrolimus Elimination26.0
Tacrolimus Control Arm12.5

[back to top]

Incidence Rate of Composite Efficacy Failure From Randomization to Month 12

"Composite efficacy failure was defined as treated biopsy proven acute rejection (tBPAR), graft loss, or death. A BPAR was defined as an acute rejection confirmed by biopsy with a Rejection Activity Index (RAI) score ≥ 3. tBPAR was defined as a BPAR which was treated with anti-rejection therapy. The RAI is used to score liver biopsies with acute rejection and is composed of 3 categories (portal inflammation, bile duct inflammation damage, venous endothelial inflammation) each scored on a scale of 0 (absent) to 3 (severe) by a trained pathologist. The total RAI score = the sum of the scores of the 3 categories and ranges from 0 to 9, with a higher score indicating greater rejection. The graft was presumed to be lost on the day the patient was newly listed for a liver graft, they received a graft re-transplant, or they died.~The incidence rates of composite efficacy failure were estimated with a Kaplan-Meier product-limit formula." (NCT00622869)
Timeframe: Randomization to Month 12

InterventionPercentage of participants (Number)
Everolimus + Reduced Tacrolimus6.7
Tacrolimus Elimination24.2
Tacrolimus Control Arm9.7

[back to top]

Incidence Rate of Treated Biopsy Proven Acute Rejection (tBPAR) at Months 12 and 24

"tBPAR was defined as an acute rejection confirmed by biopsy with a Rejection Activity Index (RAI) score ≥ 3, which was treated with anti-rejection therapy. Liver biopsies were collected for all cases of suspected acute rejection preferably within 24 hours, at the latest within 48 hours, whenever clinically possible. The RAI is used to score liver biopsies with acute rejection and is composed of 3 categories (portal inflammation, bile duct inflammation damage, venous endothelial inflammation) each scored on a scale of 0 (absent) to 3 (severe) by a trained pathologist. The total RAI score = the sum of the scores of the 3 categories and ranges from 0 to 9, with a higher score indicating greater rejection. The graft was presumed to be lost on the day the patient was newly listed for a liver graft, they received a graft re-transplant, or they died.~The incidence rates of tBPAR were estimated with a Kaplan-Meier product-limit formula." (NCT00622869)
Timeframe: Randomization to Month 24

,,
InterventionPercentage (Number)
Month 12Month 24
Everolimus + Reduced Tacrolimus3.04.8
Tacrolimus Control Arm7.27.7
Tacrolimus Elimination18.819.9

[back to top]

Change in Renal Function From Randomization to Months 12 and 24

"Change in renal function was assessed by the estimated Glomerular Filtration Rate (eGFR) using the abbreviated (4 variables) Modification of Diet in Renal Disease (MDRD-4) formula which was developed by the MDRD Study Group and has been validated in patients with chronic kidney disease. The MDRD-4 formula used for the eGFR calculation is: eGFR (mL/min/1.73m^2) = 186.3*(C^-1.154)*(A^-0.203)*G*R, where C is the serum concentration of creatinine (mg/dL), A is age (years), G=0.742 when gender is female, otherwise G=1, R=1.21 when race is black, otherwise R=1.~The changes in renal function were analyzed via analysis of covariance (ANCOVA) with treatment, pre-transplant hepatitis C virus status and randomization eGFR as covariates. Based on these ANCOVA analyses, the least-squares mean and standard errors of change were reported." (NCT00622869)
Timeframe: Randomization to Month 24

,,
InterventionmL/min/1.73m^2 (Least Squares Mean)
Month 12 (N=244, 231, 243)Month 24 (N=245, 231, 243)
Everolimus + Reduced Tacrolimus-2.23-7.94
Tacrolimus Control Arm-10.73-14.60
Tacrolimus Elimination-1.51-4.19

[back to top]

Biochemical Response Rate

Number of participants with 50% decline in serum PSA from baseline was pre-set as the primary measure of disease response. (NCT00629525)
Timeframe: Patients were followed for a median of 315 days

Interventionparticipants (Number)
RAD0010

[back to top]

Clinical Response

"The percentage of participants with a complete or partial response as defined by RECIST 1.0. Response Criteria are defined below:~Complete Response: Disappearance of all target lesions Partial Response: At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD Progressive Disease: At least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions Stable Disease: Neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum LD" (NCT00629525)
Timeframe: Patients were followed for a median of 315 days

Interventionparticipants (Number)
RAD0010

[back to top]

Molecular Response

Functional extent of mTOR inhibition by changes in the phosphorylation status of pS6 in prostate tumors. (NCT00629525)
Timeframe: Patients were followed for a median of 315 days

Interventionpercentage of decrease (Mean)
RAD00160.11

[back to top]

Progression Free Survival

Time in months from the start of study treatment to the date of first progression according to RECIST 1.0, or to death due to any cause. Patients alive who had not progressed as of the last follow-up had PFS censored at the last follow-up date. Median PFS was estimated using a Kaplan-Meier curve. (NCT00629525)
Timeframe: Patients were followed for a median of 315 days, with the last patient censored at 1309 days.

Interventionmonths (Median)
RAD0013.58

[back to top]

Pathologic Response

Number of participants with either a 50% or greater decrease in proliferation index or a 50% or greater increase in apoptotic index (NCT00629525)
Timeframe: Patients were followed for a median of 315 days

Interventionparticipants (Number)
RAD0010

[back to top] [back to top] [back to top]

Time to Progression (TTP)

TTP estimated with Kaplan-Meier methods is defined as the time from treatment start to when PSA progression criteria is first met, or the date of measurable or non-measurable disease progression (PD). Absent progression, patients are censored at the date of the last PSA measurement. PSA progression is a ≥25% increase over baseline or nadir PSA, whichever is lowest with a minimum increase of 5 ng/mL. If PSA declines ≥50%, PSA progression is a ≥50% PSA increase above nadir with a minimum increase of 5 ng/mL or back to pretreatment baseline, whichever is lowest. PSA progression requires 2 week confirmation. Per RECIST, PD is at least a 20% increase in sum LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or appearance of new lesions. Non-measurable PD is defined as a worsening bone scan, as indicated by the appearance of two or more new lesions, the appearance of new non-bony metastases or a requirement for radiation therapy. (NCT00630344)
Timeframe: PSA was measured monthly and measurable disease on imaging assessed every 2 cycles in first 8 weeks and every 3 cycles thereafter. In this study cohort, patients were followed on treatment up to approximately 1 year.

Interventionweeks (Median)
RAD001 + Bicalutamide8.7

[back to top] [back to top] [back to top]

Overall Response Rate

"Overall response rate is the percentage of patients achieving response taking into consideration measurable disease, bone metastases, and PSA. PSA declines in the absence of both measurable disease and the appearance of new bone lesions or a response in measurable disease without an increase in PSA or the appearance of new bone lesions. Patients with stable disease (SD) lasting at least 6 months will also be considered responders.~Per RECIST guidelines, for target lesions, complete response (CR) is complete disappearance of all target lesions and partial response (PR) is at least a 30% decrease in the sum of longest diameter (LD) of target lesions, taking as reference baseline sum LD. CR or PR confirmation is required within 4 weeks. Per modified PSAWG2 criteria (Scher H, Halabi S, Tannock I et al. JCO 2008) PSA response is defined as PSA decline ≥ 50% from baseline confirmed by a second measurement at least 4 weeks later." (NCT00630344)
Timeframe: PSA was measured monthly and measurable disease on imaging assessed every 2 cycles in first 8 weeks and every 3 cycles thereafter. In this study cohort, patients were followed on treatment up to approximately 1 year.

Interventionpercentage of patients (Number)
RAD-001 + Bicalutamide6

[back to top]

Progression of Measured Glomerular Filtration Rate

Change in renal progression measured by mean mGFR from week 7 to Month 36 (NCT00634920)
Timeframe: Week 7, Week 52, Month 36

,
InterventionmL/min/1.73m^2 (Mean)
Week 7Week 52Change from week 7 to Week 52Month 36Change from week 7 to Month 36
Control (CsA)47.847.80.046.1-1.7
Everolimus (CNI-free)46.351.55.648.21.3

[back to top]

Percentage of Participants With Treatment Failures

Treatment failure was defined as graft loss or death. (NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionPercentage of participants (Number)
Month 12: No FailureMonth 12: FailureMonth 24: No FailureMonth 24: FailureMonth 36: No FailureMonth 36: Failure
Control (CsA)100.00.098.81.296.73.3
Everolimus (CNI-free)100.00.098.81.298.81.2

[back to top]

Percentage of Participants With Graft Loss or Death

The allograft was presumed to be lost on the day the patient started dialysis and was not able to subsequently be removed from dialysis. If the patient underwent a graft nephrectomy, the day of nephrectomy was the day of graft loss. Graft loss was considered an SAE (serious adverse event). (NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionPercentage of participants (Number)
Month 12: Event First YearMonth 12: No Event First YearMonth 24: Event Second YearMonth 24: No Event Second YearMonth 36: Event Third YearMonth 36: No Event Third Year
Control (CsA)0.0100.01.198.92.297.8
Everolimus (CNI-free)0.0100.01.198.90.0100.0

[back to top]

Percentage of Participants With Biopsy Proven Acute Rejection (BPAR)

A BPAR was defined as a biopsy graded IA, IB, IIA, IIB, or III (Banff 97 classification). Biopsy graded IA: Significant interstitial infiltration (> 25% of parenchyma) and foci of moderate tubulitis (> 4 mononuclear cells/tubular cross section or group of 10 tubular cells). Biopsy grade IB: Significant interstitial infiltration (> 25% of parenchyma) and foci of severe tubulitis (> 10 mononuclear cells/tubular cross section or group of 10 tubular cells). Biopsy grade IIA: Mild to moderate intimal arteritis. Biopsy graded IIB: Severe intimal arteritis comprising > 25% of the lumenal area. (NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionPercentage of participants (Number)
Month 12: lAMonth 12: lBMonth 12: llAMonth 12: llBMonth 24: lAMonth 24: lBMonth 36: lAMonth 36: lB
Control (CsA)4.40.02.21.14.43.31.10.0
Everolimus (CNI-free)19.610.92.22.25.41.12.21.1

[back to top]

Percentage of Participants on Lipid-lowering Drugs

(NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionPercentage of participants (Number)
Month 12Month 24Month 36
Control (CsA)60.065.063.0
Everolimus (CNI-free)75.078.073.0

[back to top]

Percentage of Participants Who Had Donor Specific Antibodies (DSA)

Venous blood was drawn for donor specific (DSA) measurements prior to transplantation and at the final visit (36 months). The blood sample was first screened for the presence of PRA i.e. donor specific Immunoglobulin-G antibodies against specific HLA antigens. If PRA antibodies were detected, the blood sample was tested for specific DSAs on single antigen Luminex beads (coated with single HLA class I or II molecules). In this way, the specificity of these antibodies could be determined. (NCT00634920)
Timeframe: Month 36

,
InterventionPercentage of participants (Number)
ND (not done)NegativePositive
Control (CsA)9.070.021.0
Everolimus (CNI-free)7.078.015.0

[back to top]

Percentage of Participants Who Developed CAN (Chronic Allograft Nephropathy)

Assessed by protocol biopsies findings (Banff 1997 lesion scores and morphometry of the interstitial space) (NCT00634920)
Timeframe: Month 12, Month 36

,
InterventionPercentage of participants (Number)
Month 12Month 36
Control (CsA)1.064.0
Everolimus (CNI-free)1.059.0

[back to top]

Time to First Malignancy

This is the time to first diagnosed malignancy. Malignancies (skin- or solid cancer) were listed whether they reoccurred in situ, were metastatic or de novo. This is shown as mean time. (NCT00634920)
Timeframe: Months 12, 24, 36

InterventionMonths (Mean)
Everolimus (CNI-free)35.5
Control (CsA)35.1

[back to top]

Measured Glomerular Filtration Rate

Progression of renal function measured by mean mGFR at 36 months after renal TX. The mGFR was measured using Iohexol or Cr-EDTA clearance according to local practice. (NCT00634920)
Timeframe: Month 36

InterventionmL/min/1.73m^2 (Mean)
Everolimus (CNI-free)48.2
Control (CsA)46.1

[back to top]

Calculated Glomerular Filtration Rate

The GFR was calculated according to the Modification of Diet in Renal Disease Study Group (MDRD) method, the Cockcroft-Gault method, and the Nankivell formula. cGFR was calculated from blood samples collected at predefined time points. (NCT00634920)
Timeframe: Months 12, 36

,
InterventionmL/min/1.73m^2 (Mean)
MDRD M12MDRD M36Cockcroft-Gault M12Cockcroft-Gault M36Nankivel M12Nankivel M36
Control (CsA)60.157.445.642.161.858.9
Everolimus (CNI-free)65.059.445.443.166.361.8

[back to top] [back to top]

Lipid Profile for Apolipoprotein

Blood lipid levels of patients in both groups for Apolipoprotein (Apo) A1 and B. (NCT00634920)
Timeframe: Months 12, 24, 36

,
Interventiong/L (Mean)
Month 12: Apolipoprotein A1Month 24: Apolipoprotein A1Month 36: Apolipoprotein A1Month 12: Apolipoprotein BMonth 24: Apolipoprotein B (Month 36: Apolipoprotein B
Control (CsA)1.461.361.560.9231.0580.934
Everolimus (CNI-free)1.591.551.700.9351.1780.984

[back to top]

Lipid Profile for HDL-C, LDL-C,Total Cholesterol, and Triglycerides

Blood lipid levels of patients in both groups: HDL-C, LDL-C,Total cholesterol, and triglycerides. (NCT00634920)
Timeframe: Months 12, 24, 36

,
Interventionmmol/L (Mean)
Month 12: HDL CholesterolMonth 24: HDL CholesterolMonth 36: HDL CholesterolMonth 12: LDL CholesterolMonth 24: LDL CholesterolMonth 36: LDL CholesterolMonth 12: Total CholesterolMonth 24: Total CholesterolMonth 36: Total CholesterolMonth 12: TriglyceridesMonth 24: TriglyceridesMonth 36: Triglycerides
Control (CsA)1.4191.4091.5293.1302.9252.8225.3185.1124.8301.8681.7571.580
Everolimus (CNI-free)1.4861.4771.4953.5693.3813.2066.0915.8235.5952.4612.2882.164

[back to top]

Number of Lipid-lowering Drugs Taken

(NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionNumber of lipid-lowering drugs (Mean)
Month 12Month 24Month 36
Control (CsA)0.80.90.8
Everolimus (CNI-free)0.91.00.9

[back to top]

Percentage of Participants on Antihypertensive Drugs

(NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionPercentage of participants (Number)
Month 12: No antihypertensive drugsMonth 12: Has antihypertensive drugsMonth 24: No antihypertensive drugsMonth 24: Has antihypertensive drugsMonth 36: No antihypertensive drugsMonth 36: Has antihypertensive drugs
Control (CsA)3.396.75.394.712.887.2
Everolimus (CNI-free)9.290.84.295.815.684.4

[back to top]

Time to Treatment Failure

Treatment failure was defined as graft loss or death.Time to treatment failure is shown as mean time to treatment failure. (NCT00634920)
Timeframe: Months 12, 24, 36

InterventionDays (Mean)
Everolimus (CNI-free)972.7
Control (CsA)959.5

[back to top]

Measured Glomerular Filtration Rate

To compare the efficacy between treatment regimens by assessing the difference in renal function evaluated by mean measured glomerular filtration rate (mGFR) 12 months after renal transplantation (TX). The mGFR was measured using Iohexol or Cr-EDTA clearance according to local practice. (NCT00634920)
Timeframe: Month 12

InterventionmL/min/1.73m^2 (Mean)
Everolimus (CNI-free)51.5
Control (CsA)47.8

[back to top]

Proteinuria (Measured as Urine Albumin/Creatinine Ratio (mg/mmol))

"Proteinuria is when a large amount of protein, that should remain circulating in a person's blood, is spilled into their urine and eliminated from the body." (NCT00634920)
Timeframe: Months 12, 24, 36

,
Interventionmg/mmol (Mean)
Month 12Month 24Month 36
Control (CsA)11.2724.5580.73
Everolimus (CNI-free)17.3162.8378.78

[back to top]

Number of Antihypertensive Drugs Taken

(NCT00634920)
Timeframe: Months 12, 24, 36

,
InterventionNumber of antihypertensive dugs (Mean)
Month 12Month 24Month 36
Control (CsA)2.52.42.2
Everolimus (CNI-free)2.52.52.0

[back to top]

Number of Participants Surviving at 6 Months

Overall survival (OS) at 6 months in participants receiving a combination of Erlotinib and RAD001 who have received previous treatment for advanced pancreatic cancer. OS at 6 months is number of participants alive at 6 months. (NCT00640978)
Timeframe: 6 months

InterventionParticipants (Number)
Erlotinib + RAD00115

[back to top]

Progression-free Survival (PFS)

Progression-free survival (PFS) per RECIST criteria (NCT00651482)
Timeframe: 24 months

Interventionmonths (Median)
Bevacizumab + RAD001 (Everolimus)5.1

[back to top]

Overall Survival (OS)

(NCT00651482)
Timeframe: 44 months

Interventionmonths (Median)
Bevacizumab + RAD001 (Everolimus)21.0

[back to top] [back to top]

Objective Response (OR)

Number of subjects with objective response (OR) (NCT00651482)
Timeframe: 24 months

Interventionparticipants (Number)
Bevacizumab + RAD001 (Everolimus)1

[back to top]

Objective Response (OR) Duration

(NCT00651482)
Timeframe: 24 months

Interventionweeks (Median)
Bevacizumab + RAD001 (Everolimus)21.7

[back to top]

Time-to-Treatment Failure (TTF)

(NCT00651482)
Timeframe: 24 months

Interventionmonths (Median)
Bevacizumab + RAD001 (Everolimus)5.1

[back to top] [back to top]

Treatment Discontinuation Due to Disease Progression

Number of subjects whose treatment was discontinued due to disease progression (NCT00651482)
Timeframe: 24 months

Interventionparticipants (Number)
Bevacizumab + RAD001 (Everolimus)6

[back to top]

Treatment Discontinuation Due to Toxicity

Number of subjects whose treatment was discontinued due to toxicity (NCT00651482)
Timeframe: 24 months

Interventionparticipants (Number)
Bevacizumab + RAD001 (Everolimus)4

[back to top]

Core Study: Number of Patients With Composite Efficacy Endpoint

"The composite efficacy endpoint consisted of treated biopsy proven acute rejection (BPAR) episodes, graft loss, death or loss to follow-up. A treated BPAR was defined as a biopsy graded IA, IB, IIA, IIB, or III and which was treated with anti-rejection therapy. The allograft was presumed to be lost on the day the patient starts dialysis and was not able to stop dialysis. If the patient underwent a graft nephrectomy, then the day of nephrectomy was considered as the day of graft loss.~For the individual components (including loss of follow-up)of the composite endpoint, patients are counted for the first event to occur." (NCT00658320)
Timeframe: 12 months

,
InterventionParticipants (Number)
Composite Efficacy EndpointTreated BPARGraft LossDeathLoss to follow up (see caveats)
Everolimus + Reduced Dose of Cyclosporine73004
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine75002

[back to top]

Extension Study: Renal Function Measured by Calculated Glomerular Filtration Rate (cGFR) Using the Modification of Diet in Renal Disease (MDRD) Formula

"Renal function was assessed by glomerular filtration rate (GFR) using the MDRD formula:~GFR [mL/min/1.73m2] = 186.3*(C-1.154)*(A-0.203)*G*R C is the serum concentration of creatinine [mg/dL] A is age [years] G = 0.742 when gender is female, otherwise G=1 R = 1.21 when race is black, otherwise R=1~Loss to follow up (in In Primary Core Outcome Measure) is composite efficacy failure and contains incidence of treated BPAR, graft loss, death or loss to follow-up" (NCT00658320)
Timeframe: Month 24

InterventionmL/min/1.73m^2 (Median)
Everolimus + Reduced Dose of Cyclosporine58.90
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine54.95

[back to top]

Core Study: Renal Function Measured by Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula

"Modification of Diet in Renal Disease (MDRD) formula is:~Calculated GFR [mL/min/1.73m^2] = 186.3*(C^-1.154)*(A^-0.203)*G*R where~C is the serum concentration of creatinine [mg/dL],~A is patient age at sample collection date [years],~G=0.742 when gender is female, otherwise G=1,~R=1.21 when race is black, otherwise R=1" (NCT00658320)
Timeframe: Month 12

InterventionmL/min/1.73m^2 (Median)
Everolimus + Reduced Dose of Cyclosporine58.00
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine55.25

[back to top]

Core Study: Number Participants With Combined Graft Loss, Death or Loss to Follow-up

"The allograft was presumed to be lost on the day the patient starts dialysis and was not able to stop dialysis. If the patient underwent a graft nephrectomy, then the day of nephrectomy was considered as the day of graft loss.~A loss to follow-up in graft loss, death or loss to follow-up is a patient who did not experience graft loss or death and whose last day of contact was prior to Day 316, i.e. prior to the Month 12 visit window." (NCT00658320)
Timeframe: 12 months

InterventionParticipants (Number)
Everolimus + Reduced Dose of Cyclosporine5
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine3

[back to top]

Extension Study: Renal Function Measured by Calculated Glomerular Filtration Rate (cGFR) Using the Modification of Diet in Renal Disease (MDRD) Formula

"Renal function was assessed by glomerular filtration rate (GFR) using the MDRD formula:~GFR [mL/min/1.73m2] = 186.3*(C-1.154)*(A-0.203)*G*R C is the serum concentration of creatinine [mg/dL] A is age [years] G = 0.742 when gender is female, otherwise G=1 R = 1.21 when race is black, otherwise R=1" (NCT00658320)
Timeframe: Month 48

InterventionmL/min/1.73m^2 (Median)
Everolimus + Reduced Dose of Cyclosporine59.80

[back to top]

Extension Study: Renal Function Measured by Calculated Glomerular Filtration Rate (GFR) Using the Nankivell Formula

"The Nankivell formula was used to calculate GFR at Month 24:~GFR[mL/min]=6.7/C + W/4 - UREA/2 - 100/H^2 + 35 (25 for females) W= body weight [kg] H= height [m] C= serum creatinine [mmol/L] UREA= serum urea [mmolL]" (NCT00658320)
Timeframe: Month 24, Month 48

,
InterventionmL/min (Mean)
Month 24Month 48 (9,0)
Everolimus + Reduced Dose of Cyclosporine63.4960.16
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine59.24NA

[back to top]

Extension Study: Number of Participants With Combined Efficacy Endpoint: Graft Loss, Death, Loss to Follow-up and/or Treated Biopsy Proven Acute Rejection (BPAR)

"Graft loss was defined as the day the patient started dialysis and was not able to subsequently be removed from dialysis or re-transplant.~Loss-to-follow was a patient who did not experience a treated BPAR, graft loss or death and whose last day of contact was prior to Month 24.~A Graft Biopsy was done within 48 hours of suspect rejection. Biopsies were read by the local pathologist according to the updated Banff '97 criteria.~Treated BPAR was based on local laboratory biopsy results and was defined as a biopsy Banff criteria graded IA to III that was treated with anti-rejection therapy." (NCT00658320)
Timeframe: 24 Months

,
InterventionParticipants (Number)
Combined Efficacy EndpointTreated BPARGraft LossDeathLoss to follow-up
Everolimus + Reduced Dose of Cyclosporine43001
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine55000

[back to top]

Extension Study: Number of Participants With Adverse Events and Serious Adverse Events

Additional information about Adverse Events can be found in the Adverse Event Section. (NCT00658320)
Timeframe: 24 Months

,
InterventionParticipants (Number)
Adverse EventsSerious Adverse Events
Everolimus + Reduced Dose of Cyclosporine5030
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine5030

[back to top]

Extension Study: Everolimus Trough Levels

Blood was collected at all visits after Day 3 for trough (collected 5 minutes before study drug dose) everolimus levels and was analyzed at a central laboratory using liquid chromatography mass spectrometry. (NCT00658320)
Timeframe: Month 24, Month 48

Interventionng/mL (Mean)
Month 24Month 48 (n=8)
Everolimus + Reduced Dose of Cyclosporine5.2584.408

[back to top]

Extension Study: Cyclosporine Trough Levels

Blood was collected at all visits after Day 3 for trough (collected 5 minutes before study drug dose) cyclosporine levels and was analyzed at a central laboratory using immunoassay. (NCT00658320)
Timeframe: Month 24, Month 48

,
Interventionng/mL (Mean)
Month 24Month 48 (n=7,0)
Everolimus + Reduced Dose of Cyclosporine54.134.2
Mycophenolate Mofetil (MMF) + Standard Dose of Cyclosporine105.5NA

[back to top]

Death (Cardiac Death, Vascular Death, and Non-cardiovascular Death)

(NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS0.4

[back to top]

Death (Cardiac Death, Vascular Death, and Non-cardiovascular Death)

(NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS1.4

[back to top]

Death (Cardiac Death, Vascular Death, and Non-cardiovascular Death)

(NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS2.6

[back to top]

Dual Antiplatelet Medication Usage

"Patient is included if medications (both aspirin and thienopyridine) were taken for at least 1 day during the visit window. The visit window for 14-day visit is 7-21 days, 30-day visit is 23-37 days, 180-day visit is 166-194 days, 1-year visit is 323-407 days, and 2-year visit is 688-772 days.~Adjunctive antiplatelet therapy includes: Aspirin & Thienopyridines (Clopidogrel/Ticlopidine/Prasugrel).~Compliance refers to subjects following prescribed instructions for taking these medications. Therapy interruptions refer to any intervals during which the subject stops taking one or all of the prescribed medications." (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS86.4

[back to top]

Composite Rate of Cardiac Death, Any MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Target Lesion Revascularization (TLR) (PCI and CABG)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS3.5

[back to top]

Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS6.3

[back to top]

Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS4.7

[back to top]

Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS3.3

[back to top]

Dual Antiplatelet Medication Usage

"Patient is included if medications (both aspirin and thienopyridine) were taken for at least 1 day during the visit window. The visit window for 14-day visit is 7-21 days, 30-day visit is 23-37 days, 180-day visit is 166-194 days, 1-year visit is 323-407 days, and 2-year visit is 688-772 days.~Adjunctive antiplatelet therapy includes: Aspirin & Thienopyridines (Clopidogrel/Ticlopidine/Prasugrel).~Compliance refers to subjects following prescribed instructions for taking these medications. Therapy interruptions refer to any intervals during which the subject stops taking one or all of the prescribed medications." (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS89.2

[back to top]

Dual Antiplatelet Therapy Non-compliance Through 1 Year

Defined as patients who had at least 1 day without using either aspirin or thienopyridine from 1 to 407 days post index procedure. (NCT00676520)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS18.0

[back to top]

Major Bleeding Complications

by TIMI flow (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS2.8

[back to top]

Major Bleeding Complications

by TIMI flow (NCT00676520)
Timeframe: at 14 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS0.6

[back to top]

Major Bleeding Complications

by TIMI flow (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS1.8

[back to top]

Major Bleeding Complications

by TIMI flow (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS0.7

[back to top]

Composite Rate of Cardiac Death, Any MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Target Lesion Revascularization (TLR) (PCI and CABG)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS9.6

[back to top]

Composite Rate of Cardiac Death and MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Clinically-indicated Target Lesion Revascularization (CI-TLR) (PCI and CABG) (This Composite Endpoint is Also Denoted as TLF)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS3.5

[back to top]

Patient Health Status, Physical Limitations Assessed Using the SAQ (Seattle Angina Questionaire)

"SAQ: 19-item, 5-6-point Likert, questionnaire measuring 5 dimensions of coronary artery disease:~Anginal Stability: whether a patient's symptoms are changing over time. Anginal Frequency: how often a patient is having symptoms now Physical Limitation: how much a patient's condition is hampering his ability to do what he wants to do.~Treatment Satisfaction: how well a patient understands her care and what she thinks of it.~Disease Perception: the overall impact of a patient's condition on a patient's interpersonal relationships and state of mind.~Each dimension is assigns each response an ordinal value, beginning with 1 for the response at the lowest level of functioning, and summing across items within each of the 5 scales. Scale scores then transformed to 0-100 range by subtracting the lowest possible scale score, dividing by the range of the scale and multiplying by 100." (NCT00676520)
Timeframe: at 1 year

Interventionunits on the SAQ scale (Mean)
Subjects Receiving the XIENCE V EECSS76.0

[back to top]

Patient Health Status, Physical Limitations Assessed Using the SAQ (Seattle Angina Questionaire)

"SAQ: 19-item, 5-6-point Likert, questionnaire measuring 5 dimensions of coronary artery disease:~Anginal Stability: whether a patient's symptoms are changing over time. Anginal Frequency: how often a patient is having symptoms now Physical Limitation: how much a patient's condition is hampering his ability to do what he wants to do.~Treatment Satisfaction: how well a patient understands her care and what she thinks of it.~Disease Perception: the overall impact of a patient's condition on a patient's interpersonal relationships and state of mind.~Each dimension is assigns each response an ordinal value, beginning with 1 for the response at the lowest level of functioning, and summing across items within each of the 5 scales. Scale scores then transformed to 0-100 range by subtracting the lowest possible scale score, dividing by the range of the scale and multiplying by 100." (NCT00676520)
Timeframe: at 180 days

Interventionunits on the SAQ scale (Mean)
Subjects Receiving the XIENCE V EECSS75.8

[back to top]

SAQ (Seattle Angina Questionaire)

"SAQ: 19-item, 5-6-point Likert, questionnaire measuring 5 dimensions of coronary artery disease:~Anginal Stability: whether a patient's symptoms are changing over time. Anginal Frequency: how often a patient is having symptoms now Physical Limitation: how much a patient's condition is hampering his ability to do what he wants to do.~Treatment Satisfaction: how well a patient understands her care and what she thinks of it.~Disease Perception: the overall impact of a patient's condition on a patient's interpersonal relationships and state of mind.~Each dimension is assigns each response an ordinal value, beginning with 1 for the response at the lowest level of functioning, and summing across items within each of the 5 scales. Scale scores then transformed to 0-100 range by subtracting the lowest possible scale score, dividing by the range of the scale and multiplying by 100." (NCT00676520)
Timeframe: 1 year

Interventionunits on the SAQ scale (Mean)
Physical Limitations76.0
Angina Stability54.3
Angina Frequency90.7
Treatment Satisfaction92.2
Perception of Disease/Quality of Life78.0

[back to top]

SAQ (Seattle Angina Questionaire)

"SAQ: 19-item, 5-6-point Likert, questionnaire measuring 5 dimensions of coronary artery disease:~Anginal Stability: whether a patient's symptoms are changing over time. Anginal Frequency: how often a patient is having symptoms now Physical Limitation: how much a patient's condition is hampering his ability to do what he wants to do.~Treatment Satisfaction: how well a patient understands her care and what she thinks of it.~Disease Perception: the overall impact of a patient's condition on a patient's interpersonal relationships and state of mind.~Each dimension is assigns each response an ordinal value, beginning with 1 for the response at the lowest level of functioning, and summing across items within each of the 5 scales. Scale scores then transformed to 0-100 range by subtracting the lowest possible scale score, dividing by the range of the scale and multiplying by 100." (NCT00676520)
Timeframe: 180 days

Interventionunits on the SAQ scale (Mean)
Physical Limitations75.8
Angina Stability57.3
Angina Frequency90.1
Treatment Satisfaction91.4
Perception of Disease/Quality of Life76.8

[back to top]

Composite Rate of Cardiac Death and MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Clinically-indicated Target Lesion Revascularization (CI-TLR) (PCI and CABG) (This Composite Endpoint is Also Denoted as TLF)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS6.2

[back to top]

Composite Rate of Cardiac Death and MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Clinically-indicated Target Lesion Revascularization (CI-TLR) (PCI and CABG) (This Composite Endpoint is Also Denoted as TLF)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS9.4

[back to top]

Composite Rate of Cardiac Death and Any Myocardial Infarction (MI)

MI= ARC (Academic Research Constortium) defined (NCT00676520)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS7.2

[back to top]

Composite Rate of Cardiac Death and Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS3.6

[back to top]

Composite Rate of All Death, Any MI (Q-wave and Non Q-wave) and Any Repeat Revascularization (Percutaneous Coronary Intervention [PCI] and Coronary Artery Bypass Graft [CABG])

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS4.3

[back to top]

SAQ (Seattle Angina Questionaire)

"SAQ: 19-item, 5-6-point Likert, questionnaire measuring 5 dimensions of coronary artery disease:~Anginal Stability: whether a patient's symptoms are changing over time. Anginal Frequency: how often a patient is having symptoms now Physical Limitation: how much a patient's condition is hampering his ability to do what he wants to do.~Treatment Satisfaction: how well a patient understands her care and what she thinks of it.~Disease Perception: the overall impact of a patient's condition on a patient's interpersonal relationships and state of mind.~Each dimension is assigns each response an ordinal value, beginning with 1 for the response at the lowest level of functioning, and summing across items within each of the 5 scales. Scale scores then transformed to 0-100 range by subtracting the lowest possible scale score, dividing by the range of the scale and multiplying by 100." (NCT00676520)
Timeframe: at baseline

Interventionunits on the SAQ scale (Mean)
Physical Limitations70.0
Angina Stability42.0
Angina Frequency73.1
Treatment Satisfaction98.1
Perception of Disease/Quality of Life55.3

[back to top]

Composite Rate of All Death, Any MI (Q-wave and Non Q-wave) and Any Repeat Revascularization (Percutaneous Coronary Intervention [PCI] and Coronary Artery Bypass Graft [CABG])

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS9.4

[back to top]

Composite Rate of All Death, Any MI (Q-wave and Non Q-wave) and Any Repeat Revascularization (Percutaneous Coronary Intervention [PCI] and Coronary Artery Bypass Graft [CABG])

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS14.9

[back to top]

Patient Health Status, Physical Limitations Assessed Using the SAQ (Seattle Angina Questionaire)

"SAQ: 19-item, 5-6-point Likert, questionnaire measuring 5 dimensions of coronary artery disease:~Anginal Stability: whether a patient's symptoms are changing over time. Anginal Frequency: how often a patient is having symptoms now Physical Limitation: how much a patient's condition is hampering his ability to do what he wants to do.~Treatment Satisfaction: how well a patient understands her care and what she thinks of it.~Disease Perception: the overall impact of a patient's condition on a patient's interpersonal relationships and state of mind.~Each dimension is assigns each response an ordinal value, beginning with 1 for the response at the lowest level of functioning, and summing across items within each of the 5 scales. Scale scores then transformed to 0-100 range by subtracting the lowest possible scale score, dividing by the range of the scale and multiplying by 100." (NCT00676520)
Timeframe: at baseline

Interventionunits on the SAQ scale (Mean)
Subjects Receiving the XIENCE V EECSS70.0

[back to top]

Composite Rate of All Death and Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS3.6

[back to top]

Composite Rate of All Death and Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS5.8

[back to top]

Composite Rate of Cardiac Death, Any MI (Q-wave and Non Q-wave) Attributed to the Target Vessel, and Target Lesion Revascularization (TLR) (PCI and CABG)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS6.3

[back to top]

Procedural Success

(NCT00676520)
Timeframe: acute: post index procedure until hospital discharge

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS97.3

[back to top]

Revascularization (Target Lesion, Target Vessel [TVR], and Non-target Vessel) (PCI and CABG)

(NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS9.2

[back to top]

Revascularization (Target Lesion, Target Vessel [TVR], and Non-target Vessel) (PCI and CABG)

(NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS5.0

[back to top]

Composite Rate of All Death and Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS8.3

[back to top]

Composite Rate of Cardiac Death and Any MI (Q-wave and Non Q-wave)

MI= Academic Research Consortium (ARC) defined (NCT00676520)
Timeframe: at 180 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS5.4

[back to top]

Clinical Device Success

(NCT00676520)
Timeframe: acute: post index procedure until hospital discharge

Interventionpercentage of lesions (Number)
Subjects Receiving the XIENCE V EECSS99.8

[back to top]

Dual Antiplatelet Medication Usage

"Patient is included if medications (both aspirin and thienopyridine) were taken for at least 1 day during the visit window. The visit window for 14-day visit is 7-21 days, 30-day visit is 23-37 days, 180-day visit is 166-194 days, 1-year visit is 323-407 days, and 2-year visit is 688-772 days.~Adjunctive antiplatelet therapy includes: Aspirin & Thienopyridines (Clopidogrel/Ticlopidine/Prasugrel).~Compliance refers to subjects following prescribed instructions for taking these medications. Therapy interruptions refer to any intervals during which the subject stops taking one or all of the prescribed medications." (NCT00676520)
Timeframe: at 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS79.9

[back to top]

Stent Thrombosis (Definite and Probable) Rate as Defined by ARC (Academic Research Constortium).

"ARC Defines Stent Thrombosis in the following way:~Definite Stent Thrombosis: Angiographic or pathologic confirmation of partial or total thrombotic occlusion within the peri-stent region AND at least ONE of the following, additional criteria:~Acute ischemic symptoms Ischemic ECG changes Elevated cardiac biomarkers~Probable Stent Thrombosis: Any unexplained death within 30 days of stent implantation or any myocardial infarction, which is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis and in the absence of any other obvious cause~Possible Stent Thrombosis Any unexplained death beyond 30 days~For further information on ARC definitions, please refer to the following website: http://circ.ahajournals.org/content/115/17/2344.full#sec-1" (NCT00676520)
Timeframe: up to 1 year

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS0.81

[back to top]

Dual Antiplatelet Medication Usage

"Patient is included if medications (both aspirin and thienopyridine) were taken for at least 1 day during the visit window. The visit window for 14-day visit is 7-21 days, 30-day visit is 23-37 days, 180-day visit is 166-194 days, 1-year visit is 323-407 days, and 2-year visit is 688-772 days.~Adjunctive antiplatelet therapy includes: Aspirin & Thienopyridines (Clopidogrel/Ticlopidine/Prasugrel).~Compliance refers to subjects following prescribed instructions for taking these medications. Therapy interruptions refer to any intervals during which the subject stops taking one or all of the prescribed medications." (NCT00676520)
Timeframe: at 14 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS89.4

[back to top]

Revascularization (Target Lesion, Target Vessel [TVR], and Non-target Vessel) (PCI and CABG)

(NCT00676520)
Timeframe: at 30 days

Interventionpercentage of participants (Number)
Subjects Receiving the XIENCE V EECSS1.1

[back to top]

Overall Survival (OS) for Per Protocol (PP) and ITT Sets

OS was defined as the time from first study drug administration to death from any cause. If a patient was not known to have died at date of database closure, overall survival was censored at the date of last contact. (NCT00688623)
Timeframe: baseline up to approximately 15 months

Interventiondays (Mean)
OS for Per protocol setOS for Intent to treat set
Everolimus451.8437.1

[back to top]

Percentage of Participants With a Overall Response Rate With a Complete Response (CR) or Partial Response (PR) at 12 Months ITT Set

"The best overall response (BOR) was calculated on basis of the tumor of overall lesion response evaluated at each visit. To be assigned a status of PR or CR, changes in tumor measurements had to be confirmed by repeat assessments that should have been performed not less than 4 weeks after the criteria for response were first met. Assessments was based on RECIST criteria 1.0. Measurable disease lesions had to be accurately measured in at least one dimension with longest diameter ≥ 20 mm using conventional techniques or ≥ 10 mm with spiral CT scan (with minimum lesion size no less than double the slice thickness). PR required at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters.~CR required disappearance of all target and non-target lesions." (NCT00688623)
Timeframe: baseline up to approximately 12 months

Interventionpercentage of participants (Number)
Complete response (CR)Partial responseStable disease (SD)Progressive disease (PD)Unknown
Everolimus0.00.074.016.49.6

[back to top]

Percentage of Participants With Disease Control Rate (DCR) at 12 Months for Per Protocol (PP) and ITT Sets

DCR was based on central radiologic review and is defined as the percentage of patients with a best overall response of 'Complete response' (CR), 'Partial response' (PR) or 'Stable disease' (SD). Relative frequencies together with their exact 2-sided 80% confidence intervals were presented (NCT00688623)
Timeframe: baseline up to approximately 12 months

Interventionpercentage of participants (Number)
DCR for Per protocol setDCR for Intent to treat set
Everolimus56.750.7

[back to top]

Percentage of Participants' Best Overall Response Rate at 12 Months - Per Protocol Set (PP)

Overall response rate (ORR) was based on RECIST central assessment and defined as the percentage of patients with best overall response (BOR) of a confirmed complete response (CR) or partial response (PR). The BOR was calculated on basis of the tumor of overall lesion response evaluated at each visit. To be assigned a status of PR or CR, changes in tumor measurements had to be confirmed by repeat assessments obtained within 4 weeks after the criteria for response were first met. Assessments was based on RECIST criteria 1.0. Measurable disease lesions had to be accurately measured in at least one dimension with longest diameter ≥ 20 mm using conventional techniques or ≥ 10 mm with spiral CT scan (with minimum lesion size no less than double the slice thickness). PR required at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters. CR required disappearance of all target and non-target lesions. (NCT00688623)
Timeframe: baseline up to approximately 12 months

Interventionpercentage of participants (Number)
Complete response (CR)Partial responseStable disease (SD)Progressive disease (PD)Unknown
Everolimus0.00.056.743.30.0

[back to top]

Duration of Progression Free Survival (PFS) for Per Protocol (PP) and ITT Sets

Duration of PFS was defined as the time from first study drug administration to objective tumor progression or death from any cause. Observations from patients not experiencing tumor progression or death at date of database closure were censored with the date of their last adequate tumor assessment. Progression was either 1) a 20% increase in the sum of the longest diameter of all target lesions, taking as reference the smallest sum of longest diameter of all target lesions recorded at or after baseline or 2) the appearance of a new lesion or 3) the unequivocal progression of non-target lesions. (NCT00688623)
Timeframe: baseline up to approximately 12 months

Interventiondays (Median)
PFS days for Per protocol setPFS days for Intent to treat set
Everolimus185190

[back to top]

Percentage of Participants With Objective Response Rate at 12 Months - Per Protocol Set (PP)

Overall Response Rate (ORR) was calculated for total PP population based on central review as confirmatory, primary analysis as well as for ITT population as sensitivity analysis. It was presented with relative frequencies and the exact 2-sided 80% confidence limit (CI) computed using the Clopper-Pearson method). If the lower limit of the CI did not include p0=5%, the hypothesis that p ≤ 5% was rejected. The primary analysis was based on the PP Set (NCT00688623)
Timeframe: baseline up to approximately 12 months

Interventionpercentage of participants (Number)
Everolimus0.0

[back to top]

Percentage of Participants With Objective Response Rate at 12 Months ITT Set

Overall Response Rate (ORR) was presented for ITT population as sensitivity analysis. It was presented with relative frequencies and the exact 2-sided 80% confidence limit (CL; computed using the Clopper-Pearson method). If the lower limit of the CI did not include p0=5%, the hypothesis that p ≤ 5% was rejected. (NCT00688623)
Timeframe: baseline up to approximately 12 months

Interventionpercentage of participants (Number)
Everolimus0.0

[back to top]

Objective Response Rate

ORR is defined as the proportion of patients with tumor size reduction of a predefined amount and for a minimum time period. Response duration usually is measured from the time of initial response until documented tumor progression (NCT00688753)
Timeframe: End of trial

Intervention% participants (Number)
PP SetITT Set
RAD0011.51.2

[back to top]

To Evaluate Efficacy of RAD001 as Monotherapy for the Treatment of Papillary Renal Cancer. Efficacy is Defined as the Percentage of Patients Progression-free at 6 Months.

PFSR at 6 months based on central review (NCT00688753)
Timeframe: 6 mos

Intervention% participants (Number)
(PPFF Set, N=44)(PPSet, N=66)(ITT Set, N=86)
RAD00134.133.332.6

[back to top]

Duration of Response

The DOR analysis applied only to patients whose overall response was CR or PR and was defined as the time from onset of response (CR/PR) to progression or death from any cause. (NCT00688753)
Timeframe: End of trial

Interventiondays (Median)
local review PP setlocal review ITT set
RAD001169226

[back to top]

Median Progression Free Survival

PFS was defined as the time from first study drug administration to objective tumor progression or death from any cause. (NCT00688753)
Timeframe: End of trial

Interventiondays (Median)
PP setITT set
RAD001118113

[back to top]

Disease Control Rate (SD + PR + CR)

DCR was defined as the proportion of patients with a best overall response of CR, PR or SD and ORR as the percentage of patients with CR or PR (NCT00688753)
Timeframe: 6 mos

Intervention% Participants (Number)
PP setITT set
RAD00165.265.1

[back to top]

Incidence of Adverse Events, Serious Adverse Events, and Death.

(NCT00688753)
Timeframe: End of trial

Intervention% participants (Number)
Patients with any AEAE with suspected relation to study drugAE leading to dose adjustment or interruptionAE leading to permanent discontinuationAE requiring concomitant medicationPatients with serious adverse event (SAE)SAE suspected relation to study drugSAE leading to permanent discontinuationPatients died
RAD00110097.8353.2627.1790.2246.7423.9110.8710.87

[back to top]

Disease Control Rate (DCR)

Disease Control Rate was defined as the percentage of participants with best overall disease response of CR or PR or stable disease (SD). CR was defined as complete disappearance of all extranodal lesions. PR was defined as at least a 50% decrease in SPD of all index nodal and extranodal lesions. SD was defined failure to attain the criteria needed for CR or PR and failure to fulfill the criteria for at least a 50% increase in the SPD of all index nodal and extranodal (including splenic and/or hepatic nodules) lesions, taking as reference the smallest sum of the product of the diameters of all index lesions recorded at or after baseline. (NCT00702052)
Timeframe: From date of start of treatment up to disease progression or death (approximately up to 3.8 years)

Interventionpercentage of participants (Number)
Everolimus69.0

[back to top]

Progression Free Survival (PFS)

PFS was defined as the time from the date of start of treatment to the date of event defined as the first documented progression or death due to any cause. (NCT00702052)
Timeframe: From date of start of treatment up to disease progression or death (approximately up to 3.8 years)

Interventionmonths (Median)
Everolimus4.40

[back to top]

Overall Survival

Overall survival was defined as the time from the date of start of study treatment to the date of death due to any cause. (NCT00702052)
Timeframe: From date of start of treatment up to disease progression or death (approximately up to 3.8 years)

Interventionmonths (Median)
Everolimus16.85

[back to top]

Overall Response Rate (ORR)

Overall response rate was defined as the percentage of participants with a best overall disease response of complete response (CR) or partial response (PR). CR was defined as complete disappearance of all extranodal lesions. PR was defined as at least a 50% decrease in the sum of the product of diameter (SPD) of all index nodal and extranodal lesions. (NCT00702052)
Timeframe: From date of enrollment up to disease progression or death (approximately 3.8 years)

Interventionpercentage of participants (Number)
Everolimus8.6

[back to top]

Number of Participants With At Least One Adverse Event (AE) and Serious Adverse Event (SAE)

An AE was defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease associated with the use of study drug, whether or not considered related to the study drug or worsening of pre-existing medical condition, whether or not related to study drug. A SAE was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. (NCT00702052)
Timeframe: From the start of the study and 28 days after study drug discontinuation (approximately up to 18 months)

InterventionParticipants (Count of Participants)
AESAE
Everolimus5829

[back to top]

Best Overall Response in Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab

Overall response is defined as the number of participants having achieved confirmed Complete Response (CR) + Partial Response (PR). Confirmed CR = at least two determinations of CR at least 4 weeks apart before progression. Confirmed PR = at least two determinations of PR or better at least 4 weeks apart before progression. CR required a disappearance of all target and non-target lesions. PR required at least a 30% decrease in the sum of the longest diameters of all target lesions, taking as a reference the baseline sum of the longest diameters. Disease progression was defined as: 1) a 20% increase in the sum of the longest diameter of all target lesions, taking as reference the smallest sum of the longest diameters of all target lesions recorded at or after baseline or 2) the appearance of a new lesions or 3) the unequivocal progression of non-target lesions overall. (NCT00719264)
Timeframe: Time from first participant randomized until 31Dec2011, cutoff date.

,
InterventionNumber of participants (Number)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Unknown responseOverall objective response (CR+PR)
Bevacizumab, Interferon Alfa-2a (IFN)15084262251
Bevacizumab, RAD001 (Everolimus)04990251849

[back to top] [back to top]

Response Duration Differences in Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab

The duration of response, applied only to participants with best overall response at CR or PR, is defined as the number of days between the date of first documented response (CR or PR) and the date of the event: radiological progression as per central review or death due to underlying cancer, whichever occurs first. If no event, participant is censored at the last adequate assessment. (NCT00719264)
Timeframe: Time from first documented response date of radiological progressive disease as per independent central review, death due to underlying cancer, or last tumor assessment, reported between date of first participant randomized until 31Dec2011, cut-off date.

InterventionMonths (Median)
Bevacizumab, RAD001 (Everolimus)13.3
Bevacizumab, Interferon Alfa-2a (IFN)11.3

[back to top]

Overall Survival (OS) Treatment Effect in Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab

Overall survival (OS) was defined as the time of randomization to the date of death due to any cause. (NCT00719264)
Timeframe: Time from randomization to the date of death from any cause, reported between date of first participant randomized and up to 2 years after the last participant randomized (data cutoff: 30Aug2012)

InterventionMonths (Median)
Bevacizumab, RAD001 (Everolimus)27.1
Bevacizumab, Interferon Alfa-2a (IFN)27.1

[back to top]

Duration of Exposure of RAD001 in Participants Randomized to the Treatment Combination of RAD001 and Bevacizumab

This outcome measure was assessed continuously. (NCT00719264)
Timeframe: From the date of the first participant treated until the last patient discontinued the study treatment + 28 days

Interventionweeks (Median)
Bevacizumab, RAD001 (Everolimus)37.0

[back to top]

Progression-free Survival (PFS) of Participants Who Received RAD001 Plus Bevacizumab Versus Participants Who Received IFN Plus Bevacizumab

Tumor response and disease progression were assessed using response evaluation criteria in solid tumors (RECIST), version 1.0. All target and non-target lesions identified at baseline were assessed using the same method, CT scan with contrast or MRI with contrast, throughout the trial. All scans were reviewed by independent, central radiology. Disease progression was defined as: 1) a 20% increase in the sum of the longest diameter of all target lesions, taking as reference the smallest sum of the longest diameters of all target lesions recorded at or after baseline or 2) the appearance of a new lesions or 3) the unequivocal progression of non-target lesions overall. (NCT00719264)
Timeframe: Time from randomization to the date of radiological progressive disease as per independent central review, death from any cause, or last tumor assessment, reported between date of first participant randomized until 31Dec2011, cutoff date.

InterventionMonths (Median)
Bevacizumab, RAD001 (Everolimus)9.3
Bevacizumab, Interferon Alfa-2a (IFN)10.0

[back to top]

Time to Definitive Deterioration of the Global Health Status and the Physical Functioning (PF) Subscale Scores of the European Organization for the Research and Treatment of Cancer (EORTC)-Core Quality of Life Questionnaire (QLQ-C30) by at Least 10%

The EORTC QLQ-C30 contains 30 items. These include five functional scales (physical, role, emotional, social and cognitive functioning), three symptom scales (fatigue, pain, nausea, and vomiting), a global health status/QoL scale, and six single items (dyspnea, diarrhea, constipation, anorexia, insomnia and financial impact). The PF subscale consists of 5 questions each scored from 1 (not at all) to 4 (very much). The score for the PF subscale and global health status range from 0 to 100, with a higher score representing a high level of functioning/high quality of life. Definitive deterioration by at least 10% is defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the course of the study. A single measure reporting a decrease of at least 10% is considered definitive only if it is the last one available for the participant. (NCT00719264)
Timeframe: Time from randomization to the date of definitive deterioration (defined as no later increase above the threshold observed during the study), or date of last assessment, reported between date of first participant randomized until 31Dec2011

,
InterventionMonths (Median)
Global health status/QoLPhysical functioning
Bevacizumab, Interferon Alfa-2a (IFN)7.89.0
Bevacizumab, RAD001 (Everolimus)7.48.5

[back to top]

Number of Participants Who Experienced Adverse Events (AEs), Serious Adverse Events and Deaths

Participants were monitored for adverse events, serious adverse events and deaths throughout the study. Participants were assessed continuously at each 28-day cycle. (NCT00719264)
Timeframe: From the first participant randomized until the last patient discontinued the study treatment + 28 days

,
InterventionParticipants (Number)
Adverse events (serious and non-serious)Serious adverse eventsDeaths
Bevacizumab, Interferon Alfa-2a (IFN)1807695
Bevacizumab, RAD001 (Everolimus)1797993

[back to top]

Number of Participants With Adverse Events

(according to National Cancer Institute Common Terminology Criteria for Adverse Events version 3.0) (NCT00729482)
Timeframe: up to 24 weeks

Interventionparticipants (Number)
RAD00154

[back to top]

Overall Survival

(NCT00729482)
Timeframe: 1 year

InterventionMonths (Median)
RAD0018.3

[back to top]

Progression-free Survival Rate at 4-month (16 Weeks)

progression is definced as a more than 20% increase in one or more lesions or the appearance of any new lesion (NCT00729482)
Timeframe: 4 months (16 weeks)

Interventionpercentage of participants (Number)
RAD00118.4

[back to top]

Response Rate

Tumor response is evaluated according to the new guidelines by RECIST criteria (See Appendix D). A complete response (CR) is defined as the disappearance of all evidence of cancer for 4 weeks or longer. A partial response (PR) is defined as a 30% or more reduction in the sum of the longest diameters of target lesions for 4 weeks or longer without any evidence of new lesions or progression of any lesions. Stable disease is defined as less than a 30% reduction or less than a 20% increase in the longest diameters of target lesions without any evidence of new lesions. Progressive disease is defined as a more than 20% increase in one or more lesions or the appearance of any new lesion. (NCT00729482)
Timeframe: 2years

Interventionpercentage of participants (Number)
RAD0013.7

[back to top]

Objective Tumor Response Rates (Complete Response and Partial Response) at Week 16 (ITT)

The best overall response is the best response recorded from the start of the treatment until disease progression/recurrence (taking as reference for PD the smallest measurements recorded since the treatment started). In general, the patient's best response assignment will depend on the achievement of both measurement and confirmation criteria. Best lesion response was defined by (Resist Criteria V1 for target and non-target lesions). (NCT00767819)
Timeframe: Baseline up to approximately 16 weeks

,,
Interventionpercentage of participants (Number)
Complete responsePartial response
Arm 100
Arm 200
Arm 300

[back to top]

Best Overall Response Rates by Week 16 (ITT)

"The best overall response is the best response recorded from treatment start until disease progression/recurrence (both measurement and confirmation criteria). Best lesion response was defined by (Resist Criteria V1 for target and non-target lesions): Complete Response (CR)=at least two determinations of CR 4 weeks apart before progression, Partial Response (PR)=at least two determinations of CR 4 weeks apart before progression (and not qualifying for a CR), Stable Disease (SD)=at least one SD assessment >6 weeks after start of treatment and Progressive Disease (PD)=Progression or death due to underlying cancer ≤16 weeks after start of treatment. PD without radiologic evidence were classified as progression only, when clear evidence of clinical deterioration was available and patient discontinued due to disease progression. Unknown (UNK) = all other cases." (NCT00767819)
Timeframe: Baseline up to 16 weeks

,,
Interventionpercentage of participants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Arm 10040.551.48.1
Arm 20033.362.54.2
Arm 300100.000

[back to top]

Time to Progression (TTP) (ITT)

Time to progression (TTP) was defined as the time from the date of start of treatment to the date of event defined as the first documented progression or death from underlying disease. If a patient had not had an event, TTP was censored at the date of the last adequate tumor assessment, which was the date of Visit 6 (Week 16) for the core phase and the last available tumor assessment for the follow-up phase. TTP was explored by using the Kaplan-Meier method. (NCT00767819)
Timeframe: Baseline up to 16 weeks

Interventiondays (Median)
Arm 157
Arm 257
Arm 3499

[back to top]

Percentage of Participants With Progression-free Survival (PFS) at 16 Weeks

Progression-free Survival (PFS) was defined as the time from the date of start of treatment to the date of event defined as the first documented progression or death from any cause. If a patient had not had an event, PFS was censored at the date of the last adequate tumor assessment at week 16 (NCT00767819)
Timeframe: 16 weeks

Interventionpercentage of participants (Number)
Arm 135.1
Arm 230.5
Arm 3100.0

[back to top]

Percentage of Participants With Overall Survival (OS) at Week 16 (ITT)

Overall survival (OS) was defined as the time from the date of start of treatment to death from any cause. If a patient was not known to have died (was alive), OS was censored at the date of the last contact, which was the date of Visit 6 (Week 16) for the core phase and the last available visit for the follow-up phase. OS was explored by using the Kaplan-Meier method. One death occurred in Arm 3 after Week 16. (NCT00767819)
Timeframe: Baseline up to 16 weeks

Interventionpercentage of participants (Number)
Arm 168.8
Arm 256.9
Arm 3100.0

[back to top]

Percentage of Participants With Duration of Response (CR, PR, SD) at 16 Weeks.

Duration of response (CR, PR or SD) applied only to those patients whose best overall response was CR, PR or SD based on local radiologic assessments and was defined as the time from start of treatment to progression or death from underlying disease. Patients not experiencing progression or death at 16 weeks were censored with the date of their last tumor assessment. Duration of response was explored using the Kaplan-Meier method. (NCT00767819)
Timeframe: Baseline up to 16 weeks

Interventionpercentage of participants (Number)
Arm 160
Arm 262.5
Arm 3100

[back to top]

Frequency and Severity of Toxicities

(NCT00770120)
Timeframe: Weekly during the first 8 weeks of treatment, then every 4 weeks while on treatment, then every 8 weeks until disease progression, then every 6 months thereafter.

Interventionparticipants (Number)
AnorexiaConfusionDehydrationDiarrheaDyspnea (shortness of breath)Fatigue (asthenia, lethargy, malaise)Glucose, serum-high (hyperglycemia)HemoglobinINR (of prothrombin time)Inf (clin/microbio) w/Gr 3-4 neuts - LungInfection with unknown ANC - Lung (pneumonia)Left ventricular systolic dysfunctionLymphopeniaMucositis/stomatitis (clinical exam) - Oral cavityMucositis/stomatitis (functional/symp) - Oral cavMuscle weakness, not d/t neuropathy - body/generalPneumonitis/pulmonary infiltratesRash/desquamationTriglyceride, serum-high (hypertriglyceridemia)
Everolimus1121363411312112212

[back to top]

Response

A response was defined as either a confirmed or unconfirmed complete or partial responses as defined by RECIST. A complete response (CR) was defined as the disappearance of all disease. A partial response (PR) was defined as a >= 30% decrease in the sum of longest diameters of target lesions. A CR or PR was considered confirmed if two consecutive determinations were made at least 4 weeks apart. (NCT00770120)
Timeframe: Every 8 weeks until disease progression, up to 3 years.

Interventionpercentage of overall response rate (Number)
Everolimus2

[back to top]

Progression-Free Survival

Progression-Free Survival was defined as the duration from the date of registration until the date of disease progression per RECIST or death due to any cause. Patients known to be alive without evidence of disease progression were censored at the date of last contact. Disease progression was defined as a >= 20% increase over nadir in the sum of longest diameters of target lesions, unequivocal progression of non-target lesions in the opinion of the treating investigator, appearance of new lesions, symptomatic deterioration, or death due to disease (NCT00770120)
Timeframe: Every 8 weeks until disease progression, up to 3 years.

Interventionmonths (Median)
Everolimus3.0

[back to top]

Overall Survival

Overall survival was defined as the duration between the date of enrollment and the date of death due to any cause. Patients last known to be alive were censored at the date of last contact. (NCT00770120)
Timeframe: Every 8 weeks until disease progression, up to 3 years.

Interventionmonths (Median)
Everolimus6.3

[back to top]

Overall Response

Overall response is classified as complete response (CR), partial response (PR), stable disease (SD) or Progressive Disease (PD) on therapy.Description: Overall response is classified as complete response (CR), partial response (PR), stable disease (SD) or Progressive Disease (PD) on therapy. Response for target lesions (up to5) is based on 3 dimensions with an elliptical model volume used: 0.5L*W*T; (L) tumor extent in plane perpendicular to the selected plane; (W) longest measurement of the tumor width; (T) transverse measurement perpendicular to the width. CR is disappearance all target and non-target lesions and no new lesions. PR is >/= 65% decrease in sum of the products (referent baseline). PD 40% or more increase in any target lesion (referent smallest product observed on therapy). SD is none of the above. PR and SD classification as long as absent new lesions and unequivocal progression for non-target lesions else PD. (NCT00782626)
Timeframe: Disease evaluations (MRI brain, including volumetric analysis) occurred at baseline, at the end of course 1, every 3 courses during treatment up to 12 courses and at early treatment discontinuation.

Interventionparticipants (Number)
Partial ResponseStable Disease
Everolimus221

[back to top]

Cardiac Death/MI

This endpoint is a composite of cardiac death and all myocardial infarction per protocol definition. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.9

[back to top]

Cardiac Death/ All MI /CI-TLR

This endpoint is a composite of cardiac death, all myocardial infarction (MI)per protocol definition, and clinically-indicated target lesion revascularization (CI-TLR). (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.1

[back to top]

Cardiac Death/ All MI /CI-TLR

This endpoint is a composite of cardiac death, all myocardial infarction (MI)per protocol definition, and clinically-indicated target lesion revascularization (CI-TLR). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®12.1

[back to top]

Cardiac Death/ All MI /CI-TLR

This endpoint is a composite of cardiac death, all myocardial infarction (MI)per protocol definition, and clinically-indicated target lesion revascularization (CI-TLR). (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®7.2

[back to top]

Cardiac Death/ All MI /CI-TLR

This endpoint is a composite of cardiac death, all myocardial infarction (MI)per protocol definition, and clinically-indicated target lesion revascularization (CI-TLR). (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®8.3

[back to top]

Cardiac Death/ All MI /CI-TLR

This endpoint is a composite of cardiac death, all myocardial infarction (MI)per protocol definition, and clinically-indicated target lesion revascularization (CI-TLR). (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®8.1

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)

Target vessel myocardial infarction (MI) (MI not clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®1.5

[back to top]

All TVR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) in the target vessel from the index procedure. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.4

[back to top]

All TVR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) in the target vessel from the index procedure. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®8.6

[back to top]

Clinically Indicated Target Lesion Revascularization (CI-TLR)

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery) of the target lesion. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Clinically Indicated Target Vessel Revascularization

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery), of the target vessel. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms (per protocol). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®12.1

[back to top]

Clinically Indicated Target Vessel Revascularization

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery), of the target vessel. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®8.8

[back to top]

Clinically Indicated Target Vessel Revascularization

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery), of the target vessel. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®9.8

[back to top]

Clinically Indicated Target Vessel Revascularization

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery), of the target vessel. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®7.2

[back to top]

All TVR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) in the target vessel from the index procedure. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®11.3

[back to top]

Clinically Indicated Target Vessel Revascularization (TVR)

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery), of the target vessel. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.4

[back to top]

Composite Rate of Cardiac Death, Target Vessel Myocardial Infarction (MI) (Per Protocol Definition) & Clinically Indicated Target Lesion Revascularization (CI-TLR).

This endpoint is a composite of cardiac death, target vessel myocardial infarction per protocol definition, and clinically-indicated target lesion revascularization. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®8.1

[back to top]

All TVR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) in the target vessel from the index procedure. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®10.3

[back to top]

All TVR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) in the target vessel from the index procedure (per protocol). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®13.6

[back to top]

All TLR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) of the target lesion from the index procedure. This includes interventions classified as clinically indicated, and also includes interventions classified as not clinically indicated. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.7

[back to top]

All TLR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) of the target lesion from the index procedure. This includes interventions classified as clinically indicated, and also includes interventions classified as not clinically indicated. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®5.8

[back to top]

All TLR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) of the target lesion from the index procedure. This includes interventions classified as clinically indicated, and also includes interventions classified as not clinically indicated. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®6.8

[back to top]

All TLR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) of the target lesion from the index procedure. This includes interventions classified as clinically indicated, and also includes interventions classified as not clinically indicated. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®6.6

[back to top]

All TLR (CI and Non-CI)

Includes any repeat revascularization intervention after the index procedure by any means (percutaneous or bypass surgery) of the target lesion from the index procedure. This includes interventions classified as clinically indicated, and also includes interventions classified as not clinically indicated (per protocol). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®8.3

[back to top]

All Death/ All MI/All Coronary Revascularization

This endpoint is a composite of all death, all myocardial infarction per protocol definition, and all revascularization. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®3.5

[back to top]

All Death/ All MI/All Coronary Revascularization

This endpoint is a composite of all death, all myocardial infarction per protocol definition, and all revascularization. (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®26.5

[back to top]

All Death/ All MI/All Coronary Revascularization

This endpoint is a composite of all death, all myocardial infarction per protocol definition, and all revascularization. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®15.1

[back to top]

All Death/ All MI/All Coronary Revascularization

This endpoint is a composite of all death, all myocardial infarction per protocol definition, and all revascularization. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®22.6

[back to top]

All Death/ All MI/All Coronary Revascularization

This endpoint is a composite of all death, all myocardial infarction per protocol definition, and all revascularization. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®16.9

[back to top]

All Death (Cardiac, Vascular, Non-cardiovascular)

All death, including death from cardiac, vascular, and non-cardiovascular causes. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.7

[back to top]

All Death (Cardiac, Vascular, Non-cardiovascular)

All death, including death from cardiac, vascular, and non-cardiovascular causes. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.4

[back to top]

All Death (Cardiac, Vascular, Non-cardiovascular)

All death, including death from cardiac, vascular, and non-cardiovascular causes. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®1.5

[back to top]

All Death (Cardiac, Vascular, Non-cardiovascular)

All death, including death from cardiac, vascular, and non-cardiovascular causes. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.5

[back to top]

All Death (Cardiac, Vascular, Non-cardiovascular)

All death, including death from cardiac, vascular, and non-cardiovascular causes (per protocol). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®3.8

[back to top]

All Coronary Revascularization (TVR and Non-TVR)

Includes any revascularization intervention after the index procedure by any means (percutaneous or bypass surgery), including intervention to the target vessel, and intervention to a vessel other than the target vessel. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.1

[back to top]

All Coronary Revascularization (TVR and Non-TVR)

Includes any revascularization intervention after the index procedure by any means (percutaneous or bypass surgery), including intervention to the target vessel, and intervention to a vessel other than the target vessel. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®12.9

[back to top]

All Coronary Revascularization (TVR and Non-TVR)

Includes any revascularization intervention after the index procedure by any means (percutaneous or bypass surgery), including intervention to the target vessel, and intervention to a vessel other than the target vessel. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®20.3

[back to top]

All Coronary Revascularization (TVR and Non-TVR)

Includes any revascularization intervention after the index procedure by any means (percutaneous or bypass surgery), including intervention to the target vessel, and intervention to a vessel other than the target vessel. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®14.7

[back to top]

All Coronary Revascularization (TVR and Non-TVR)

Includes any revascularization intervention after the index procedure by any means (percutaneous or bypass surgery), including intervention to the target vessel, and intervention to a vessel other than the target vessel (per protocol). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®23.5

[back to top]

Clinically Indicated Target Lesion Revascularization (CI-TLR)

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery) of the target lesion. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®5.3

[back to top]

Composite Rate of Cardiac Death, Target Vessel Myocardial Infarction (MI) (Per Protocol Definition) & Clinically Indicated Target Lesion Revascularization (CI-TLR).

This endpoint is a composite of cardiac death, target vessel myocardial infarction per protocol definition, and clinically-indicated target lesion revascularization. (NCT00783796)
Timeframe: 3 years

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®12.1

[back to top]

Device Success (Per Lesion Basis, for Target Lesions Treated by 2.25 mm XIENCE V EECS With or Without Planned Overlap)

Successful delivery and deployment of the first study stent intended to be implanted at the intended target lesion (or intended first and second investigational stents for overlapping stents), successful withdrawal of the stent delivery system, and attainment of final residual stenosis of <50%. (NCT00783796)
Timeframe: From start of index procedure to end of index procedure

InterventionPercentage of Lesions (Number)
2.25mm XIENCE V®95.21

[back to top]

Distal % Diameter Stenosis

Value calculated as 100*(1-MLD/RVD) where MLD is minimum lumen diameter and RVD is reference vessel diameter in 5 mm of healthy tissue distal to stent placement. (NCT00783796)
Timeframe: 240 days

InterventionPercentage (Mean)
2.25mm XIENCE V®10.4

[back to top]

Distal Angiographic Binary Restenosis (ABR) Rate

Percentage of patients with target lesions with ≥ 50% diameter stenosis in 5 mm of healthy tissue distal to stent placement at angiographic follow-up. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Distal Late Loss

Distal minimum lumen diameter (MLD) post-procedure minus distal MLD at angiographic follow-up (distal defined as 5 mm of healthy tissue distal to stent placement). (NCT00783796)
Timeframe: 240 days

InterventionMillimeter (Mean)
2.25mm XIENCE V®0.00

[back to top]

In-segment % Diameter Stenosis

Value calculated as 100*(1-MLD/RVD) where MLD is in-segment minimum lumen diameter and RVD is in-segment reference vessel diameter. (NCT00783796)
Timeframe: 240 days

InterventionPercentage (Mean)
2.25mm XIENCE V®20.85

[back to top]

In-segment Angiographic Binary Restenosis (ABR) Rate

Percentage of patients with target lesions with ≥ 50% in-segment % diameter stenosis at angiographic follow-up. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®9.6

[back to top]

In-segment Late Loss (LL)

In-segment minimum lumen diameter (MLD) post-procedure minus in-segment MLD at angiographic follow-up. (NCT00783796)
Timeframe: 240 Days

InterventionMillimeters (Mean)
2.25mm XIENCE V®0.16

[back to top]

In-stent % Diameter Stenosis

Value calculated as 100*(1-MLD/RVD) where MLD is in-stent minimum lumen diameter and RVD is in-stent reference vessel diameter. (NCT00783796)
Timeframe: 240 days

InterventionPercentage (Mean)
2.25mm XIENCE V®12.86

[back to top]

In-stent Angiographic Binary Restenosis (ABR) Rate

Percentage of patients with target lesions with ≥ 50% in-stent % diameter stenosis at angiographic follow-up. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®3.8

[back to top]

In-Stent Late Loss

In-stent minimum lumen diameter (MLD) post-procedure minus in-stent MLD at angiographic follow-up. (NCT00783796)
Timeframe: 240 days

InterventionMillimeters (Mean)
2.25mm XIENCE V®0.20

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)

Target vessel myocardial infarction (MI) (MI not clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.4

[back to top]

Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)

Non target vessel myocardial infarction (MI) (MI clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)

Non target vessel myocardial infarction (MI) (MI clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)

Non target vessel myocardial infarction (MI) (MI clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Clinically Indicated Target Lesion Revascularization (CI-TLR)

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery) of the target lesion. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®4.3

[back to top]

Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)

Non target vessel myocardial infarction (MI) (MI clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Non Target Vessel MI (Q-wave, Non Q-wave)(Per Protocol)

Non target vessel myocardial infarction (MI) (MI clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)

ARC defined non-target vessel MI (MI clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 1 year

InterventionPercentage of participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)

ARC defined non-target vessel MI (MI clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Clinically Indicated Target Lesion Revascularization (CI-TLR)

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery) of the target lesion. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms. (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®5.1

[back to top]

Clinically Indicated Target Lesion Revascularization (CI-TLR)

Includes clinically indicated repeat revascularization after the index procedure by any means (percutaneous or bypass surgery) of the target lesion. Classification as clinically indicated is done prospectively and verified by angiographic core lab measurement, and requires ≥50% diameter stenosis with ischemic signs or symptoms (positive history of angina pectoris or objective signs of ischemia at rest (ECG changes) or during exercise test or abnormal invasive cardiac functional diagnostic test), or ≥70% diameter stenosis in the absence of the above-mentioned ischemic signs or symptoms (per protocol). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®6.8

[back to top]

Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)

ARC defined non-target vessel MI (MI clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 240 days

InterventionPercentage of participants (Number)
2.25mm XIENCE V®1.4

[back to top]

Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)

ARC defined non-target vessel MI (MI clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®4.5

[back to top]

Non Target Vessel MI- Q-wave, Non Q-wave (Per ARC)

ARC defined non-target vessel MI (MI clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 30 days

InterventionPercentage of participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Procedural Success (Per Subject Basis, for ALL Target and Non-target Lesions)

Achievement of a final in-stent diameter stenosis of <50% using the study device, without the occurence of cardiac death, target vessel myocardial infarction per protocol definition, or repeat revascularization of the target lesion during the hospital stay up to 7 days. (NCT00783796)
Timeframe: From the start of index procedure to end of index procedure

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®97.93

[back to top]

Proximal % Diameter Stenosis

Value calculated as 100*(1-MLD/RVD) where MLD is minimum lumen diameter and RVD is reference vessel diameter in 5 mm of healthy tissue proximal to stent placement. (NCT00783796)
Timeframe: 240 days

InterventionPercentage (Mean)
2.25mm XIENCE V®14.31

[back to top]

Proximal Angiographic Binary Restenosis (ABR) Rate

Percentage of patients with target lesions with ≥ 50% diameter stenosis in 5 mm of healthy tissue proximal to stent placement at angiographic follow-up. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.7

[back to top]

Proximal Late Loss

Proximal minimum lumen diameter (MLD) post-procedure minus proximal MLD at angiographic follow-up (proximal defined as 5 mm of healthy tissue proximal to stent placement). (NCT00783796)
Timeframe: 240 days

InterventionMillimeter (Mean)
2.25mm XIENCE V®0.21

[back to top]

Stent Thrombosis (ARC Defined)

ARC defined: Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 0 to 1 day (Acute)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Stent Thrombosis (ARC Defined)

ARC defined: Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: greater than 1 day to 30 days (Subacute)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.7

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: >1 year (Very late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 31 days - 393 days (Late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 394 - 1123 days (Very Late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.78

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 394 - 758 days (Very Late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: Overall (0 - 1123 days)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®3.08

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: Overall (0 - 393 days)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.17

[back to top]

Stent Thrombosis (ARC Defined)

Stent Thrombosis as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344-2351). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: Overall (0 - 758 days)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.29

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: > 1 day to 30 days (Subacute)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.7

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 0 to 1 day (Acute)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®0.0

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 31 - 1123 days (Late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.3

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 31 - 758 days (Late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: 31 days to 393 days (Late)

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: Overall (0 - 1123 days)

Interventionpercentage of participants (Number)
2.25mm XIENCE V®3.1

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: Overall (0 - 393 days)

Interventionpercentage of participants (Number)
2.25mm XIENCE V®2.2

[back to top]

Stent Thrombosis (Protocol Defined)

Stent Thrombosis per protocol categorized as acute (≤1 day), subacute (>1 day and ≤30 days), and late (>30 days), and defined as clinical presentation of acute coronary syndrome with angiographic evidence of stent thrombosis, or in absence of angiography, any unexplained death at any time or acute myocardial infarction* (ST segment elevation or new Q-wave) in the distribution of the target lesion within 30 days of the index procedure. (*Non-specific ST/T changes and cardiac enzymes do not suffice.). Result includes Definite/Probable/Possible. (NCT00783796)
Timeframe: Overall (0 - 758 days)

Interventionpercentage of participants (Number)
2.25mm XIENCE V®2.27

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per ARC)

ARC defined target vessel MI (MI not clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®6.6

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per ARC)

ARC defined target vessel MI (MI not clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®6.7

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per ARC)

ARC defined target vessel MI (MI not clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®6.5

[back to top]

Cardiac Death/MI

This endpoint is a composite of cardiac death and all myocardial infarction per protocol definition. (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.1

[back to top]

Cardiac Death/MI

This endpoint is a composite of cardiac death and all myocardial infarction per protocol definition. (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®5.3

[back to top]

Cardiac Death/MI

This endpoint is a composite of cardiac death and all myocardial infarction per protocol definition. (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®2.9

[back to top]

Cardiac Death/MI

This endpoint is a composite of cardiac death and all myocardial infarction per protocol definition. (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®3.0

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per ARC)

ARC defined target vessel MI (MI not clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 3 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®7.5

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per ARC)

ARC defined target vessel MI (MI not clearly attributable to a non-target vessel): Myocardial Infarction as per Academic Research Consortium standardized definitions (Circulation 2007;115:2344- 2351). (NCT00783796)
Timeframe: 30 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®6.3

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)

Target vessel myocardial infarction (MI) (MI not clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 1 year

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)

Target vessel myocardial infarction (MI) (MI not clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 2 years

Interventionpercentage of participants (Number)
2.25mm XIENCE V®1.5

[back to top]

Target Vessel MI - Q-wave and Non Q-wave (Per Protocol)

Target vessel myocardial infarction (MI) (MI not clearly attributable to a non-target vessel), including Q-wave MI (new pathologic Q waves) and Non Q-wave MI (elevation of CK to ≥ two times the upper limit normal with elevated CK-MB in the absence of new pathological Q waves). (NCT00783796)
Timeframe: 240 days

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®1.4

[back to top]

Composite Rate of Cardiac Death, Target Vessel Myocardial Infarction (MI) (Per Protocol Definition) & Clinically Indicated Target Lesion Revascularization (CI-TLR).

This endpoint is a composite of cardiac death, target vessel myocardial infarction per protocol definition, and clinically-indicated target lesion revascularization. (NCT00783796)
Timeframe: 2 years

InterventionPercentage of Participants (Number)
2.25mm XIENCE V®8.3

[back to top]

Everolimus Blood Concentration (C2h) at 2 Hours Post Dose

The participants were assessed for everolimus blood concentration at 2 hours time point after dose administration on the same day, if the participant did not vomit between previous dose and blood sample collection. Tandem liquid chromatography-mass spectrometry method was used for evaluation. C2h values were categorized as < 20 ng/mL, 20-50 ng/mL, and > 50 ng/mL, concentrations below the lower limit of quantification were entered as 0 ng/mL. (NCT00789828)
Timeframe: 2 hours post dose on Week 6, Week 24, Week 48, Week 96, Week 144, and Week 240

,
Interventionng/mL (Mean)
Week 6 (n= 37, 47)Week 24 (n= 11, 13)Week 48 (n= 1, 3)Week 96 (n= 0, 6)Week 144 (n= 0, 6)Week 240 (n= 0, 0)
Everolimus (Core Period)27.5238.723.2NANANA
Everolimus (Extension Period)27.7439.2549.7331.6326.33NA

[back to top]

Everolimus Trough Concentrations (Cmin) at 24 Hours After Last Dose

The participants were assessed for everolimus trough concentration (Cmin) at 24 hours time point after previous dose administration, at a steady state following 5 days of consistent dosing, if the participant did not vomit within 4 hours of previous dose. Tandem liquid chromatography-mass spectrometry method was used for evaluation. Cmin values were categorized as <5 ng/mL, 5-10 ng/mL, and >10 ng/mL, concentrations below the lower limit of quantification were entered as 0 ng/mL. (NCT00789828)
Timeframe: 24 hours post dose on Week 6, Week 24, Week 48, Week 72, Week 96, Week 144, and Week 240

,
Interventionng/mL (Mean)
Week 6 (n= 64, 94)Week 24 (n= 64, 89)Week 48 (n= 23, 86)Week 72 (n= 4, 92)Week 96 (n= 0, 83)Week 144 (n= 0, 69)Week 240 (n= 0, 13)
Everolimus (Core Period)5.86.597.286.08NANANA
Everolimus (Extension Period)6.096.867.077.257.097.285.85

[back to top]

Percentage of Participants With Renal Impairment During Core Period

Renal function was assessed using glomerular filtration rate (GFR) based on age measure; Modification of Diet in Renal Disease (MDRD) formula for participants aged 18 years or older, defined as GFR equal to 32788*(serum creatinine (micromol/L)^-1.154)*(age^-0.203 )*(0.742, if female)*(1.210, if black), and Schwartz formula for participants less than 18 years defined as GFR equal to 0.41*height (cm)/ Serum creatinine (mg/dL). Participants with severe renal impairment defined as GFR < 30 mL/min/1.73 m^2 and participants with National Cancer Institute's Common Terminology Criteria for Adverse Events (NCI-CTCAE) grade 3/4 serum creatinine were reported. (NCT00789828)
Timeframe: Day 1 up to 28 days after end of treatment (Core period)

,
InterventionPercentage of participants (Number)
Grade 3 or 4Grade 1 or 2Grade 0
Everolimus (Core Period)03.896.2
Placebo (Core Period)00100

[back to top]

Change From Baseline in Frequency of Total Seizure Events Per 24 Hours at Week 24 in Both Core and Extension Period

Seizure frequency per 24 hours was defined as the number of seizures in the electroencephalography (EEG) divided by the number of hours in the EEG, multiplied by 24. Seizure frequency was evaluated using a 24-hour video-EEG. Seizure frequency was listed as missing if the actual EEG recording duration was < 18 hours. (NCT00789828)
Timeframe: Baseline (Core period) to Week 24 (Core period), Baseline (Extension period, Week 24 post-core baseline) to Week 24 (Extension period, Week 48 post-core baseline)

InterventionSeizure frequency (Mean)
Everolimus (Core Period)-1.24
Placebo (Core Period)-0.24
Everolimus (Extension Period)-6.07

[back to top]

Duration of SEGA Response

Duration of SEGA response was defined as time from the date of the first documented SEGA response until the date of the first documented SEGA progression. Duration of SEGA response was evaluated only for participants who achieved a SEGA response. The time to SEGA progression was censored if SEGA progression was not observed before the first to occur out of (i) analysis cut-off date (ii) the date when systemic anti-SEGA medication is started, (iii) the date of a SEGA-related surgery or (iv) the date of death. Since, no case of SEGA progression was observed in core study which resulted in censored duration of SEGA response. Only 5 SEGA responders experienced a SEGA progression in extension period. (NCT00789828)
Timeframe: Baseline up to week 48 (end of core period), and end of extension period (up to 4 years)

Interventionmonths (Median)
Everolimus (Core Period)NA
Everolimus (Extension Period)NA

[back to top]

Duration of Skin Lesion Response in Everolimus Treated Participants

Duration of skin lesion response was defined as the time from the first skin lesion response until the first skin lesion progression, defined as worsening of lesion by > 25% or more from baseline. (NCT00789828)
Timeframe: Baseline up to week 48 (end of core period), and end of extension period (up to 4 years)

Interventionmonths (Median)
Everolimus (Core Period)NA
Everolimus (Extension Period)NA

[back to top]

Percentage of Participants With Best Overall Subependymal Giant Cell Astrocytomas (SEGA) Response

Participants were assessed for SEGA response, defined as 50% reduction from baseline in SEGA volume (where SEGA volume was the sum of the volumes of all target SEGA lesions identified at baseline, and confirmed with a second scan performed approximately 12 weeks later), no unequivocal worsening of non-target SEGA lesions, no new SEGA lesions (≥ 1 cm in longest diameter), and no new or worsening hydrocephalus. Multi-phase brain MRI was utilized to identify SEGA lesions. SEGA response rate was defined as the percentage of participants whose best overall status was SEGA response as determined by Independent Central Radiology Review. The Kaplan-Meier estimate was used for determining time to SEGA response. (NCT00789828)
Timeframe: End of core period (Week 48), and end of extension period (up to 4 years)

InterventionPercentage of participants (Number)
Everolimus (Core Period)34.6
Placebo (Core Period)0.0
Everolimus (Extension Period)57.7

[back to top]

Percentage of Participants With Skin Lesions Assessed Using Physician's Global Assessement Overall Score

Skin lesions included hypomelanotic macules, the shagreen patch, periungual or subungual fibromas, facial angiofibromas and/or forehead plaques. Response was evaluated using the Physician's Global Assessment of Clinical Condition (PGA) on a 7-point scale: Grade 0 = complete clinical response, indicated absence of disease, Grade 1, 2, and 3 = partial response, indicated improvements of ≥ 50% but < 100%, Grade 4, 5 = stable disease, indicated some or no improvements of 25% - < 50% and 6 = progressive disease, indicated worse than at baseline evaluation by > 25%. Response rate was determined for participants with ≥ 1 skin lesion at baseline, defined as the percentage of participants with overall status as complete clinical response or partial response. (NCT00789828)
Timeframe: End of core period (Week 48), and end of extension period (up to 4 years)

InterventionPercentage of participants (Number)
Everolimus (Core Period)41.7
Placebo (Core Period)10.5
Everolimus (Extension Period)58.1

[back to top]

Time to SEGA Progression

Time to SEGA progression was defined as time between randomisation to time to first SEGA progression. SEGA progression was defined as either one or more of the following criteria: 1. increase from nadir of ≥ 25% in SEGA volume to a value greater than baseline SEGA volume (where SEGA volume is the sum of the volumes of all target SEGA lesions identified at baseline, and nadir is the lowest SEGA volume obtained for the participant previously in the trial), 2. unequivocal worsening of non-target SEGA lesions, 3. appearance of new SEGA lesion ≥ 1.0 cm in longest diameter, 4. new or worsening hydrocephalus. The median TTSP based on central radiology review was not reached in any treatment arms; Only 6 events of SEGA progressions were observed in the placebo group of core period. (NCT00789828)
Timeframe: Baseline up to week 48 (end of core period), and end of extension period (up to 4 years)

Interventionmonths (Median)
Everolimus (Core Period)NA
Placebo (Core Period)NA
Everolimus (Extension Period)NA

[back to top]

Time to SEGA Response

Participants were assessed for time to SEGA response, defined as 50% reduction from baseline in SEGA volume (where SEGA volume was the sum of the volumes of all target SEGA lesions identified at baseline, and confirmed with a second scan performed approximately 12 weeks later), no unequivocal worsening of non-target SEGA lesions, no new SEGA lesions (≥ 1 cm in longest diameter), and no new or worsening hydrocephalus. Multi-phase brain MRI was utilised to identify SEGA lesions. SEGA response rate was defined as the percentage of participants whose best overall status was SEGA response as determined by Independent Central Radiology Review. The Kaplan-Meier estimate was used for determining time to SEGA response. (NCT00789828)
Timeframe: Baseline up to week 48 (end of core period), and end of extension period (up to 4 years)

Interventionmonths (Median)
Everolimus (Core Period)2.99
Everolimus (Extension Period)5.32

[back to top]

Time to SEGA Worsening

Time to SEGA worsening was defined as the time from the start of everolimus to date of the first SEGA worsening. SEGA worsening was defined as either; increase from nadir of ≥ 25% in SEGA volume or unequivocal worsening of non-target SEGA lesions, or appearance of new SEGA lesion ≥ 1.0 cm in longest diameter, or new or worsening hydrocephalus. The median value was not reached in either treatment arm of core period as SEGA worsening was observed in less participants (everolimus - 7 and placebo - 8). (NCT00789828)
Timeframe: Baseline up to week 48 (end of core period), and end of extension period (up to 4 years)

Interventionmonths (Median)
Everolimus (Core Period)NA
Placebo (Core Period)NA
Everolimus (Extension Period)55.72

[back to top]

Disease-free Survival (DFS)

DFS was defined as the time from date of randomization to the date of event defined as the first documented relapse of the disease or death due to any cause. Relapse was based on investigator assessment and was assigned only if: It was documented according to Cheson guidelines by an objective radiological assessment method; It was documented by a biopsy proven lymphoma including new or recurrent bone marrow involvement; A new anticancer therapy for lymphoma started with subsequent confirmation of the relapse within 4 weeks of the start of this anticancer therapy (NCT00790036)
Timeframe: From date of randomization to the date of event defined as the first documented recurrence of the disease, or death due to any cause and up to 6 years

InterventionPercentage of participants (Number)
RAD001 (Everolimus)177.8
Placebo77.0

[back to top]

Lymphoma-specific Survival (LSS)

LSS was defined as time from randomization to death as a result of lymphoma. (NCT00790036)
Timeframe: From randomization to death documented as a result of lymphoma up to 7 years

,
InterventionPercentage of participants (Number)
2 years3 years4 years5 years6 years
Placebo90.588.886.985.485.4
RAD001 (Everolimus)194.993.191.689.489.4

[back to top]

Overall Survival (OS)

OS was defined as the time from date of randomization to date of death due to any cause. If the patient was not known to have died, survival was censored at the date of the last contact. (NCT00790036)
Timeframe: From date of randomization to date of death due to any cause up to around 7 years

,
InterventionPercentage of participants (Number)
2 years3 years4 years5 years6 years
Placebo88.383.780.777.477.4
RAD001 (Everolimus)190.788.085.483.480.3

[back to top]

Angiomyolipoma Response Rate as Per Central Radiology Review

"Angiomyolipoma response defined as the combination of the following criteria: reduction in angiomyolipoma volume of ≥ 50% relative to baseline, where angiomyolipoma volume was sum of volumes of all target lesions identified at baseline, and with a confirmatory scan performed approximately 12 weeks later (no sooner than 8 weeks later); no new angiomyolipoma lesions ≥ 1.0 cm in longest diameter were identified; there were no kidney increases in volume > 20% from nadir. The patient did not have any angiomyolipoma-related bleeding of ≥ grade 2.~For the everolimus (core/extension periods) treatment group, the baseline means the latest value on or before starting everolimus." (NCT00790400)
Timeframe: From date of randomization until the earliest date of first documented AML progression, date of further anti-AML medication (including open-label Everolimus)/surgery or up to 5.7 years

InterventionPercentage of Participants (Number)
Everolimus Randomized (Core Period)41.8
Placebo Randomized (Core Period)0
Everolimus (Core and/or Extension Period)58.0

[back to top]

Duration of Angiomyolipoma Response - Only Everolimus Patients With Angiomyolipoma Response

Duration of angiomyolipoma response was defined as the time from the date of the first documented angiomyolipoma response until the date of the first documented angiomyolipoma progression . Angiomyolipoma response was defined as the combination of the following criteria: reduction in angiomyolipoma volume of ≥ 50% relative to baseline, where angiomyolipoma volume was sum of volumes of all target lesions identified at baseline, and with a confirmatory scan performed approximately 12 weeks later (no sooner than 8 weeks later); no new angiomyolipoma lesions ≥ 1.0 cm in longest diameter were identified; there were no kidney increases in volume > 20% from nadir. The patient did not have any angiomyolipoma-related bleeding of ≥ grade 2. (NCT00790400)
Timeframe: From date of randomization until the earliest date of first documented AML progression, date of further anti-AML medication (including open-label Everolimus)/surgery or up to about 5.7 years

Interventionmonths (Median)
Everolimus Randomized (Core Period)NA
Everolimus (Core and/or Extension Period)NA

[back to top]

Duration of Skin Lesion Response - Only Everolimus Patients With Best Overall Skin Lesion Response of Complete Clinical Response (CCR) or Partial Response (PR)

Duration of skin lesion response is defined as the time from the date of the first skin lesion response until the date of the first skin lesion progression, according to the PGA (physician's global assessment of clinical condition). A progression is when the disease is worse than at baseline evaluation by >=25% or more. (NCT00790400)
Timeframe: From date of randomization until the earliest date of first documented AML progression, date of further anti-AML medication (including open-label Everolimus)/surgery or up to about 5.7 years

Interventionmonths (Median)
Everolimus Randomized (Core Period)NA
Everolimus (Core and/or Extension Period)NA

[back to top]

Skin Lesion Response Rate as Per Investigator (Only Patients With at Least One Skin Lesion at Baseline)

Skin lesion response rate in the double-blind period was determined only among patients with at least one skin lesion at baseline, and is the percentage of this group of patients with a best overall skin lesion response on the Physician's Global Assessment of Clinical Condition (PGA) of either complete clinical response (CCR) or partial response (PR). A complete clinical response (CCR) requires a grading of 0 indicating the absence of disease (histological confirmation is not required). Grades 1, 2, and 3 constitute partial response, indicating improvement of at least 50 percent, but less than 100 percent improvement. For the everolimus (core/extension periods) treatment group, the baseline means the latest value on or before starting everolimus. (NCT00790400)
Timeframe: From date of randomization until the earliest date of first documented AML progression, date of further anti-AML medication (including open-label Everolimus)/surgery or up to 5.7 years

InterventionPercentage of participants (Number)
Everolimus Randomized (Core Period)26
Placebo Randomized (Core Period)0
Everolimus (Core and/or Extension Period)68.2

[back to top]

Time to Angiomyolipoma Progression as Per Central Radiology Review

"Time to angiomyolipoma progression (TTAP) is defined as time from date of randomization to date of first documented angiomyolipoma progression. Angiomyolipoma progression was defined as one or more of the following: Increase from nadir of ≥ 25% in angiomyolipoma volume to value greater than baseline; the appearance of a new angiomyolipoma ≥ 1.0 cm in longest diameter; an increase from nadir of 20% or more in the volume of either kidney to a value greater than baseline; angiomyolipoma-related bleeding grade ≥ 2.~For the everolimus (core/extension periods) treatment group, the time to angiomyolipoma progression is defined starting from the start of everolimus. The baseline means the latest value on or before starting everolimus." (NCT00790400)
Timeframe: From date of randomization until the earliest date of first documented AML progression, date of further anti-AML medication (including open-label Everolimus)/surgery or up to about 5.7 years

Interventionmonths (Median)
Everolimus Randomized (Core Period)NA
Placebo Randomized (Core Period)11.37
Everolimus (Core and/or Extension Period)NA

[back to top]

Time to Angiomyolipoma Response - Only Everolimus Patients With Angiomyolipoma Response

"Time to angiomyolipoma response was defined as the time from the date of randomization until the date of the first documented angiomyolipoma response. Angiomyolipoma response defined as the combination of the following criteria: reduction in angiomyolipoma volume of ≥ 50% relative to baseline, where angiomyolipoma volume was sum of volumes of all target lesions identified at baseline, and with a confirmatory scan performed approximately 12 weeks later (no sooner than 8 weeks later); no new angiomyolipoma lesions ≥ 1.0 cm in longest diameter were identified; no kidney increases in volume > 20% from nadir; no angiomyolipoma-related bleeding of ≥ grade 2.~For the everolimus (core/extension periods) treatment group, the time to angiomyolipoma response is from the start of everolimus. The baseline in the response definition means the latest value on or before starting everolimus." (NCT00790400)
Timeframe: From date of randomization until the earliest date of first documented AML progression, date of further anti-AML medication (including open-label Everolimus)/surgery or up to 5.7 years

Interventionmonths (Median)
Everolimus Randomized (Core Period)2.86
Everolimus (Core and/or Extension Period)2.89

[back to top]

Change From Baseline in Plasma Angiogenic Molecules - Vascular Endothelial Growth Factor (VEGF) Marker

Blood samples for biomarker assessment were collected immediately prior to study administration. On-treatment samples was compared to baseline samples with the change from baseline. (NCT00790400)
Timeframe: 4 weeks, 12 weeks, 24 weeks, 36 weeks 48 weeks, 60 weeks, 72 weeks

,
Interventionpg/mL (Mean)
Week 4 (n:56, 28)Week 12 (n:56, 29)Week 24 (n:53, 29)Week 36 (n:26, 18)Week 48 (n:16, 8)Week 60 (n:0, 1)Week 72 (n:0, 1)
Everolimus Randomized (Core Period)38.743.431.118.055.3NANA
Placebo Randomized (Core Period)17.6-6.1-4.35.43.1-4.12-6.1

[back to top]

Everolimus Blood Concentrations (C2h) at 2 Hours Post-dose

C2h values collected 1-3 hours after dose administration on the same study day, at steady state, and patient did not vomit between taking previous dose and blood collection. Samples collected during the first 4 days of dosing will be excluded from all analyses. (NCT00790400)
Timeframe: 2 hours post-dose administration at Weeks 2, 4, 12, 24, 48

Interventionng/mL (Mean)
2 hours post dose administration at Week 2 (n:55)2 hours post dose administration at Week 4 (n:49)2 hours post dose administration at Week 12 (n:56)2 hours post dose administration at Week 24 (n:50)2 hours post dose administration at Week 48 (n:14)
Everolimus Randomized (Core Period)33.3830.8934.4839.2733.20

[back to top]

Everolimus Trough Concentrations (Cmin)

Cmin values collected prior to dose administration on the same study day and at 20-28 hours after previous dose, at steady state, and patient did not vomit within 4 hours of previous dose. Samples collected during the first 4 days of dosing were excluded from all analyses. (NCT00790400)
Timeframe: Prior to dosing at weeks 2, 4, 12, 24, 48

Interventionng/mL (Mean)
Prior to dosing at Week 2 (n:43)Prior to dosing Week 4 (n:44)Prior to dosing Week 12 (n:49)Prior to dosing Week 24 (n:46)Prior to dosing Week 48 (n:15)
Everolimus Randomized (Core Period)7.637.728.799.3711.49

[back to top]

Percentage of Participants With Renal Impairment

Renal Impairment was measured by glomerular filtration rate which was calculated using the Modification of Diet in Renal Disease formula. Percentage of participants with renal impairment was reported. Severe renal impairment was defined as a GFR of <30ml/min/1.73m2. (NCT00790400)
Timeframe: Day 1 up to 28 days after end of treatment

,,
InterventionPercentage of Participants (Number)
Glomerular filtration rate <30 ml/min/1.73m^2Glomerular filtration rate≥ 30 ml/min/1.73m^2
Everolimus (Core and/or Extension Period)7.192.9
Everolimus Randomized (Core Period)2.597.5
Placebo Randomized (Core Period)7.792.3

[back to top]

Proportion of Pretreatment Primary Tumor Samples With mTOR Pathway Markers

To assess markers for activated mTOR pathway (including phospho-S6 and phospho-4E BP1) in all pre-treatment tissue specimens and correlate with response to treatment and PFS. (NCT00805129)
Timeframe: 25 months

InterventionProportion of pretreatment primary tumor (Number)
Everolimus0.62

[back to top]

Number of Participants Evaluated for Toxicity

To determine the safety and toxicity of Everolimus (RAD001) in this patient population with toxicities being evaluated by CTCAE v3.0 (NCT00805129)
Timeframe: through study completion, up to 25 months

InterventionParticipants (Count of Participants)
Everolimus46

[back to top]

Overall Survival of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen

Overall survival was defined as the interval from the first day of study treatment until the date of death. (NCT00805961)
Timeframe: 18 months

InterventionMonths (Number)
Overall Study13.9

[back to top]

Progression-free Survival (PFS)

Progression-free survival is defined as the duration of time from start of treatment to time of progression or death, whichever comes first. (NCT00805961)
Timeframe: 18 months

InterventionMonths (Median)
Overall Study11.3

[back to top]

Number of Participants Experiencing Toxicity After This Novel Multimodality Regimen

The toxicity assessments were made according to the common terminology criteria for adverse events (CTCAE version 3.0) of the National Cancer Institute. Number of participants with Grade 1 to 5 adverse events are reported here. (NCT00805961)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Overall Study53

[back to top]

Objective Response Rate of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen

Response to treatment was assessed by MRI using the MacDonald criteria based on the assessment of the MRI scan for measurable, evaluable, and new lesions. The objective response rate is defined as the proportion of patients with improvement and or decreased extent of lesions compared to baseline. (NCT00805961)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Overall Study31

[back to top]

Number of Participants With Bone Marrow Morphology and Cytogenetics Pre- and Post-therapy

Number of Participants with change in bone marrow morphology and cytogenetics (NCT00809185)
Timeframe: at 2 years of treatment

InterventionParticipants (Count of Participants)
RAD001(Everolimus)0

[back to top]

Number of Patients With Either a Major or Minor Erythroid Response (Hemoglobin Change From Baseline Measure)

"Major erythroid response: (1) For patients with a baseline hemoglobin less than 11 g/dL, a major erythroid response is defined as a > 2 g/dL increase in hemoglobin from baseline; or (2) 100% decrease in red blood cell transfusion requirements.~Minor erythroid response: (1) For patients with baseline hemoglobin less than 11 g/dL, a minor erythroid response is defined as an increase in hemoglobin greater than 1 g/dL but less than 2 g/dL from baseline; or (2) > 50% decrease in red blood cell transfusion requirements." (NCT00809185)
Timeframe: 2 years of treatment

Interventionparticipants (Number)
Number of Patients with a Minor ResponseNumber of Patients with a Major Response
RAD001(Everolimus)00

[back to top]

Number of Dose- and Non-dose-limiting Toxicities

Number of Dose- and Non-dose-limiting Toxicities at the end of cycle 1 associated with RAD001 (see AE/SAE section for details). (NCT00809185)
Timeframe: at end of one cycle (28 days)

Interventionevents (Number)
RAD001 (Everolimus)26

[back to top]

Progression-free Survival

Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT00814788)
Timeframe: Up to 2 years

Interventionmonths (Median)
Bicalutamide + Everolimus9.4

[back to top]

Overall Survival

Overall survival was estimated using the Kaplan-Meier method. (NCT00814788)
Timeframe: Up to 3 years

Interventionmonths (Median)
Bicalutamide + Everolimus28

[back to top]

PSA Response Rate

The PSA response rate was defined as a 30% reduction in the PSA level from baseline. PSA Working Group consensus criteria combined with radiographic studies were used to determine the proportion of patients with PSA decline. (NCT00814788)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Bicalutamide + Everolimus18

[back to top]

Progression-free Survival at 6 Months.

Number of patients alive without progressive disease at 6 months. Assessment of progression was defined by RANO as a 25% increase in the sum of all the products of measurable lesions, clear worsening of any evaluable disease or any new lesion (NCT00823459)
Timeframe: At 6 months after treatment start

InterventionParticipants (Count of Participants)
Grade IIGrade III/IV
Daily Intervention With RAD001396

[back to top]

To Assess the Correlation of Activation of the PI3K/mTOR Pathway With Survival

Survival of patients with p-S6 staining of >19% (activated pathway) (NCT00823459)
Timeframe: 5 years

Interventionyears (Median)
Daily Intervention With RAD0013.9

[back to top]

Objective Response Rate (ORR) in Patients Treated With RAD001.

Objective response is defined as complete or partial response as defined by RANO criteria as determined by MRI and steroid requirement. Complete response was defined by disappearance of all measurable disease with minimal or no steroids; a partial response was defined as 50% in sum of all products in perpendicular diameters of all measurable lesions with no new lesions on stable or decreasing steroids. (NCT00823459)
Timeframe: 12 months

Interventionparticipants (Number)
Daily Intervention With RAD0010

[back to top]

Overall Survival (OS) in Patients Treated With RAD001.

Number of years from the day the patient started treatment until the date of death, an average of 5 years. (NCT00823459)
Timeframe: Time from registration till death, an average of 5 years

Interventionyears (Median)
Daily Intervention With RAD0015.2

[back to top]

RAD001 Safety Profile in Patients With Recurrent LLG

Grade 4-5 treatment related adverse events as defined by CTCAE 3.0 (NCT00823459)
Timeframe: 13 months

InterventionParticipants (Count of Participants)
Daily Intervention With RAD0010

[back to top]

Best Overall Response by PI3K-AKT-MTOR Mutation

Next generation sequencing was used to identify participants with PI3K-AKT-MTOR mutations. Best overall response rate was analyzed with mutant versus wild-type pathways. Please see Best Overall Response Rate for response definition. (NCT00827359)
Timeframe: Evaluated while on treatment. Up to 36 months

Interventionpercentage of participants (Number)
MutationNo Mutations
Treatment5.60

[back to top]

Median Progression Free Survival

"Progression free survival (PFS) is defined as the time from start of treatment to disease progression (PD) or death from any cause as estimated by Kaplan Meier methods. Patients who have not progressed and are alive are censored at the date the patient is known to be progression-free. Progression is defined by Response Evaluation Criteria In Solid Tumors Criteria 1.1 (RECIST) as follows:~- >20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (including baseline if it's the smallest). The sum must also demonstrate an increase of >5 mm.~OR~-Appearance of new lesions and/or unequivocal progression of non-target lesions. It must be representative of overall disease status change, not a single lesion increase. For patients with PD at the first on-treatment imaging assessment, patients will be allowed to remain on study until confirmation at the next assessment at investigator discretion if patient is benefiting from treatment." (NCT00827359)
Timeframe: Follow-up time was up to 39 months from treatment start date.

Interventionmonths (Median)
Treatment3.8

[back to top]

Best Overall Response Rate

"The best overall response rate is the percentage of participants achieving complete response (CR) or partial response (PR) as the best response recorded on treatment based on Response Evaluation Criteria In Solid Tumors Criteria (RECIST 1.1) criteria.~CR and PR must meet the following lesion criteria without having any new lesions as well:~Target Lesion:~(CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.~(PR): At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters.~Non-Target Lesion:~(CR): Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis).~Non-CR/Non-Progressive Disease: Persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits. Must have PR in target lesion." (NCT00827359)
Timeframe: Evaluated while on treatment. Up to 36 months.

Interventionpercentage of participants (Number)
Treatment4.2

[back to top]

Disease Progression Diagnosed by Biopsy

Clinical progression validated by biopsy of metastatic site. Progression-free survival (PFS) will be measured from the post-op treatment start date to either the date the patient is first recorded as having disease progression, or the date of death if the patient dies due to any causes before progression. If a patient is lost to follow-up or removed for toxicities, the patient will be censored as of the last date of contact. Patients who start a new treatment before they progress will be censored as of the date of start of the new treatment. If a patient has not progressed or died, PFS is censored at the date of last follow-up. Patients removed from therapy with everolimus due to toxicities will not be included in the PFS estimation. (NCT00831480)
Timeframe: up to one year

Interventionparticipants (Number)
"Neoadjuvant Everolimus RAD001 for Advanced RCC Before Cytore"9

[back to top]

Objective Response Rate (ORR)

Objective response is defined as complete response (CR) or partial response (PR) assessed using Response Evaluation Criteria in Solid Tumors (RECIST) criteria. The primary analysis population is based on efficacy-evaluable patients, defined as those patients who receive at least one cycle of RAD001 plus erlotinib and undergo at least one post-baseline response assessment or die while on therapy. (NCT00843531)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
RAD001 Plus Erlotinib0

[back to top]

Number of Patients With Dose-limiting Toxicity (DLT)

Primary safety analysis will be conducted after the first 16 patients have had potential for completion of two cycles of protocol therapy. All patients receiving at least one dose of protocol therapy will be included in the analyses. All patients with treatment-related, Grade 3 or higher adverse events according to the NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3 resulting in a DLT will be reported. (NCT00843531)
Timeframe: Up to 9 months

InterventionParticipants (Count of Participants)
RAD001 and Erlotinib8

[back to top]

Duration of Objective Response

Duration of objective response will be defined as the time from the initial documentation of response to documented disease progression or death from any cause on study (NCT00843531)
Timeframe: Up to 2 years

Interventionmonths (Number)
RAD001 and ErlotinibNA

[back to top]

In-scaffold Percent Diameter Stenosis (%DS)

Percent Diameter Stenosis is defined as the value calculated as 100 * (1 - MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00856856)
Timeframe: 2 years

Interventionpercentage of diameter stenosis (Mean)
Absorb BVS20.94

[back to top]

In-scaffold Percent Diameter Stenosis (%DS)

Percent Diameter Stenosis is defined as the value calculated as 100 * (1 - MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00856856)
Timeframe: 3 years

Interventionpercentage of diameter stenosis (Mean)
Absorb BVS23.16

[back to top]

In-scaffold Percent Diameter Stenosis (%DS)

Percent Diameter Stenosis is defined as the value calculated as 100 * (1 - MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00856856)
Timeframe: 5 years

Interventionpercentage of diameter stenosis (Mean)
Absorb BVS22.74

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Absorb BVS4.0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS2.0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 270 days

InterventionPercentage of participants (Number)
Absorb BVS2.0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Absorb BVS7.0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 30 days

Interventionpercentage of participants (Number)
Absorb BVS0

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Absorb BVS7.1

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Absorb BVS8.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS4.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS2.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 2 years

InterventionPercentage of participants (Number)
Absorb BVS8.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 270 days

InterventionPercentage of participants (Number)
Absorb BVS2.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 3 years

InterventionPercentage of participants (Number)
Absorb BVS10.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 30 days

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 4 years

InterventionPercentage of participants (Number)
Absorb BVS10.0

[back to top]

Ischemia Driven Target Vessel Revascularization (ID-TVR)

"ID-TVR is the revascularization in the target vessel associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and an angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target vessel with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 5 years

InterventionPercentage of participants (Number)
Absorb BVS11.0

[back to top]

Late Incomplete Apposition

"Late-Acquired Incomplete Apposition is defined as incomplete apposition of the scaffold at follow-up, which was not present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS3.6

[back to top]

Late Incomplete Apposition

"Late-Acquired Incomplete Apposition is defined as incomplete apposition of the scaffold at follow-up, which was not present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS7.5

[back to top]

Late Incomplete Apposition

"Late-Acquired Incomplete Apposition is defined as incomplete apposition of the scaffold at follow-up, which was not present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 2 year

InterventionPercentage of participants (Number)
Absorb BVS2.6

[back to top]

Late Incomplete Apposition

"Late-Acquired Incomplete Apposition is defined as incomplete apposition of the scaffold at follow-up, which was not present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 3 year

InterventionPercentage of participants (Number)
Absorb BVS7.0

[back to top]

Luminal Volume

(NCT00856856)
Timeframe: 1 year

Interventionmm^3 (Mean)
Absorb BVS106.33

[back to top]

Luminal Volume

(NCT00856856)
Timeframe: 2 years

Interventionmm^3 (Mean)
Absorb BVS107.66

[back to top]

Luminal Volume

(NCT00856856)
Timeframe: 3 years

Interventionmm^3 (Mean)
Absorb BVS103.31

[back to top]

Luminal Volume

(NCT00856856)
Timeframe: 5 years

Interventionmm^3 (Mean)
Absorb BVS106.36

[back to top]

Mean Flow Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS5.90

[back to top]

Mean Flow Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS6.00

[back to top]

Mean Flow Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS6.06

[back to top]

Mean Flow Area

(NCT00856856)
Timeframe: 5 years

Interventionmm^2 (Mean)
Absorb BVS6.21

[back to top]

Mean Luminal Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS5.92

[back to top]

Mean Luminal Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS6.00

[back to top]

Mean Luminal Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS6.06

[back to top]

Mean Luminal Area

(NCT00856856)
Timeframe: 5 years

Interventionmm^2 (Mean)
Absorb BVS6.22

[back to top]

Mean Luminal Diameter

(NCT00856856)
Timeframe: 1 year

Interventionmm (Mean)
Absorb BVS2.71

[back to top]

Mean Luminal Diameter

(NCT00856856)
Timeframe: 2 years

Interventionmm (Mean)
Absorb BVS2.72

[back to top]

Mean Luminal Diameter

(NCT00856856)
Timeframe: 3 years

Interventionmm (Mean)
Absorb BVS2.74

[back to top]

Mean Luminal Diameter

It is measured during QCA by the Angiographic Core Lab. (NCT00856856)
Timeframe: 5 years

Interventionmm (Mean)
Absorb BVS2.77

[back to top]

Mean Reference Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS6.34

[back to top]

Mean Reference Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS6.29

[back to top]

Mean Reference Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS6.24

[back to top]

Mean Reference Area

(NCT00856856)
Timeframe: 5 years

Interventionmm^2 (Mean)
Absorb BVS6.13

[back to top]

Mean Scaffold Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS8.14

[back to top]

Mean Scaffold Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS8.50

[back to top]

Mean Scaffold Diameter

(NCT00856856)
Timeframe: 2 years

Interventionmm (Mean)
Absorb BVS3.19

[back to top]

Mean Scaffold Diameter

(NCT00856856)
Timeframe: 3 years

Interventionmm (Mean)
Absorb BVS3.26

[back to top]

Mean Stent Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS7.42

[back to top]

Mean Stent Diameter

(NCT00856856)
Timeframe: 1 year

Interventionmm (Mean)
Absorb BVS3.06

[back to top]

Mean Strut Core Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS0.17

[back to top]

Mean Strut Core Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS0.15

[back to top]

Mean Strut Core Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS0.19

[back to top]

Minimum Flow Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS4.28

[back to top]

Minimum Flow Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS4.29

[back to top]

Minimum Flow Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS4.45

[back to top]

Minimum Flow Area

(NCT00856856)
Timeframe: 5 years

Interventionmm^2 (Mean)
Absorb BVS4.15

[back to top]

Minimum Luminal Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS4.29

[back to top]

Minimum Luminal Area

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS4.29

[back to top]

Minimum Luminal Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS4.45

[back to top]

Minimum Luminal Area

(NCT00856856)
Timeframe: 5 years

Interventionmm^2 (Mean)
Absorb BVS4.15

[back to top]

Minimum Luminal Diameter

The average of two orthogonal views (when possible) of the narrowest point within the area of assessment - in lesion, in scaffold or in segment. MLD is visually estimated during angiography by the Investigator; it is measured during QCA by the Angiographic Core Lab. (NCT00856856)
Timeframe: 2 years

Interventionmm (Mean)
Absorb BVS2.30

[back to top]

Minimum Luminal Diameter

The average of two orthogonal views (when possible) of the narrowest point within the area of assessment - in lesion, in scaffold or in segment. MLD is visually estimated during angiography by the Investigator; it is measured during QCA by the Angiographic Core Lab. (NCT00856856)
Timeframe: 3 years

Interventionmm (Mean)
Absorb BVS2.34

[back to top]

Minimum Luminal Diameter

The average of two orthogonal views (when possible) of the narrowest point within the area of assessment - in lesion, in scaffold or in segment. MLD is visually estimated during angiography by the Investigator; it is measured during QCA by the Angiographic Core Lab. (NCT00856856)
Timeframe: 5 years

Interventionmm (Mean)
Absorb BVS2.25

[back to top]

Minimum Luminal Diameter (MLD)

The average of two orthogonal views (when possible) of the narrowest point within the area of assessment - in lesion, in scaffold or in segment. MLD is visually estimated during angiography by the Investigator; it is measured during QCA by the Angiographic Core Lab. (NCT00856856)
Timeframe: 1 year

Interventionmm (Mean)
Absorb BVS2.30

[back to top]

Minimum Scaffold Area

(NCT00856856)
Timeframe: 2 year

Interventionmm^2 (Mean)
Absorb BVS6.33

[back to top]

Minimum Scaffold Area

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS6.66

[back to top]

Minimum Scaffold Diameter

(NCT00856856)
Timeframe: 2 years

Interventionmm (Mean)
Absorb BVS2.82

[back to top]

Minimum Scaffold Diameter

(NCT00856856)
Timeframe: 3 years

Interventionmm (Mean)
Absorb BVS2.89

[back to top]

Minimum Stent Area

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS5.96

[back to top]

Minimum Stent Diameter

(NCT00856856)
Timeframe: 1 year

Interventionmm (Mean)
Absorb BVS2.74

[back to top]

Myocardial Infarction

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT00856856)
Timeframe: 1 year

Interventionpercentage of participants (Number)
ABSORB Stent3.0

[back to top]

Myocardial Infarction

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT00856856)
Timeframe: 2 years

Interventionpercentage of participants (Number)
ABSORB Stent3.0

[back to top]

Myocardial Infarction

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT00856856)
Timeframe: 3 years

Interventionpercentage of participants (Number)
ABSORB Stent3.0

[back to top]

Myocardial Infarction

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT00856856)
Timeframe: 4 years

Interventionpercentage of participants (Number)
ABSORB Stent3.0

[back to top]

Myocardial Infarction

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT00856856)
Timeframe: 5 years

Interventionpercentage of participants (Number)
ABSORB Stent3.0

[back to top]

Myocardial Infarction

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT00856856)
Timeframe: 30 days

Interventionpercentage of participants (Number)
ABSORB Stent2.0

[back to top]

Number of Struts in Side Branch

(NCT00856856)
Timeframe: 1 year

InterventionNumber of struts (Mean)
Absorb BVS0.1

[back to top]

Number of Struts in Side Branch

(NCT00856856)
Timeframe: 2 years

InterventionNumber of struts (Mean)
Absorb BVS0.2

[back to top]

Number of Struts in Side Branch

(NCT00856856)
Timeframe: 3 years

InterventionNumber of struts (Mean)
Absorb BVS0.2

[back to top]

Number of Struts in Side Branch

(NCT00856856)
Timeframe: 5 years

InterventionNumber of struts (Mean)
Absorb BVS0.0

[back to top]

Number of Struts Per BVS

(NCT00856856)
Timeframe: 1 year

InterventionNumber of Struts (Mean)
Absorb BVS162.1

[back to top]

Number of Struts Per BVS

(NCT00856856)
Timeframe: 2 years

InterventionNumber of Struts (Mean)
Absorb BVS160.9

[back to top]

Number of Struts Per BVS

(NCT00856856)
Timeframe: 3 years

InterventionNumber of Struts (Mean)
Absorb BVS145.2

[back to top]

Number of Struts Per BVS

(NCT00856856)
Timeframe: 5 years

InterventionNumber of Struts (Mean)
Absorb BVS7.1

[back to top]

Percent (%) Lumen Area Stenosis

(NCT00856856)
Timeframe: 1 year

InterventionPercentage of Lumen Area Stenosis (Mean)
Absorb BVS28.70

[back to top]

Percent (%) Lumen Area Stenosis

(NCT00856856)
Timeframe: 2 years

InterventionPercentage of Lumen Area Stenosis (Mean)
Absorb BVS28.75

[back to top]

Percent (%) Lumen Area Stenosis

(NCT00856856)
Timeframe: 3 years

InterventionPercentage of Lumen Area Stenosis (Mean)
Absorb BVS28.01

[back to top]

Percent (%) Lumen Area Stenosis

(NCT00856856)
Timeframe: 5 years

InterventionPercentage of Lumen Area Stenosis (Mean)
Absorb BVS34.32

[back to top]

Persisting Dissection

Dissection at follow-up that was present post-procedure. (NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Persisting Dissection

Dissection at follow-up that was present post-procedure. (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Persisting Dissection

Dissection at follow-up that was present post-procedure. (NCT00856856)
Timeframe: 2 years

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Persisting Dissection

Dissection at follow-up that was present post-procedure. (NCT00856856)
Timeframe: 3 years

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Persisting Dissection

Dissection at follow-up that was present post-procedure. (NCT00856856)
Timeframe: 5 years

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Persisting Incomplete Apposition

"Persisting incomplete apposition is defined as incomplete apposition at follow-up that was present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS3.5

[back to top]

Persisting Incomplete Apposition

"Persisting incomplete apposition is defined as incomplete apposition at follow-up that was present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS2.3

[back to top]

Persisting Incomplete Apposition

"Persisting incomplete apposition is defined as incomplete apposition at follow-up that was present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 2 year

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Persisting Incomplete Apposition

"Persisting incomplete apposition is defined as incomplete apposition at follow-up that was present post-procedure.~Incomplete Apposition: Failure of the scaffold to completely appose to the vessel wall after placement is defined as one or more scaffold strut separated from the vessel wall with evidence of blood speckles behind the strut in the ultrasound image." (NCT00856856)
Timeframe: 3 year

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 1 Year

Proximal Late Loss: proximal MLD post-procedure - proximal MLD at follow-up (proximal defined as within 5 mm of healthy tissue proximal to scaffold placement). (NCT00856856)
Timeframe: 1 year

InterventionMillimeter (Mean)
Absorb BVS0.12

[back to top]

Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 180 Days

Proximal Late Loss: proximal MLD post-procedure - proximal MLD at follow-up (proximal defined as within 5 mm of healthy tissue proximal to scaffold placement). (NCT00856856)
Timeframe: 180 days

InterventionMillimeter (Mean)
Absorb BVS0.07

[back to top]

Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 2 Years

Proximal Late Loss: proximal MLD post-procedure - proximal MLD at follow-up (proximal defined as within 5 mm of healthy tissue proximal to scaffold placement). (NCT00856856)
Timeframe: 2 years

InterventionMillimeter (Mean)
Absorb BVS0.12

[back to top]

Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 3 Years

Proximal Late Loss: proximal MLD post-procedure - proximal MLD at follow-up (proximal defined as within 5 mm of healthy tissue proximal to scaffold placement). (NCT00856856)
Timeframe: 3 years

InterventionMillimeter (Mean)
Absorb BVS0.14

[back to top]

Proximal Late Loss: Proximal MLD Post-procedure - Proximal MLD at 5 Years

Proximal Late Loss: proximal MLD post-procedure - proximal MLD at follow-up (proximal defined as within 5 mm of healthy tissue proximal to scaffold placement). (NCT00856856)
Timeframe: 5 years

InterventionMillimeter (Mean)
Absorb BVS0.14

[back to top]

Scaffold Thrombosis

"Scaffold thrombosis will be categorized as acute (≤ 1day), subacute (>1day ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of scaffold thrombosis (angiographic appearance of thrombus within or adjacent to a previously treated target lesion)~In the absence of angiography, any unexplained death, or acute MI (ST segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific ST /T changes and cardiac enzyme elevations do not suffice)~Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00856856)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Absorb BVS0

[back to top]

Scaffold Thrombosis

"Scaffold thrombosis will be categorized as acute (≤ 1day), subacute (>1day ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of scaffold thrombosis (angiographic appearance of thrombus within or adjacent to a previously treated target lesion)~In the absence of angiography, any unexplained death, or acute MI (ST segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific ST /T changes and cardiac enzyme elevations do not suffice)~Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00856856)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Absorb BVS0

[back to top]

Scaffold Thrombosis

"Scaffold thrombosis will be categorized as acute (≤ 1day), subacute (>1day ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of scaffold thrombosis (angiographic appearance of thrombus within or adjacent to a previously treated target lesion)~In the absence of angiography, any unexplained death, or acute MI (ST segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific ST /T changes and cardiac enzyme elevations do not suffice)~Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00856856)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Absorb BVS0

[back to top]

Scaffold Thrombosis

"Scaffold thrombosis will be categorized as acute (≤ 1day), subacute (>1day ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of scaffold thrombosis (angiographic appearance of thrombus within or adjacent to a previously treated target lesion)~In the absence of angiography, any unexplained death, or acute MI (ST segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific ST /T changes and cardiac enzyme elevations do not suffice)~Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00856856)
Timeframe: 30 days

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

Scaffold Thrombosis

"Scaffold thrombosis will be categorized as acute (≤ 1day), subacute (>1day ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of scaffold thrombosis (angiographic appearance of thrombus within or adjacent to a previously treated target lesion)~In the absence of angiography, any unexplained death, or acute MI (ST segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific ST /T changes and cardiac enzyme elevations do not suffice)~Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00856856)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Absorb BVS0

[back to top]

Scaffold Thrombosis

"Scaffold thrombosis will be categorized as acute (≤ 1day), subacute (>1day ≤ 30 days) and late (>30 days) and will be defined as any of the following:~Clinical presentation of acute coronary syndrome with angiographic evidence of scaffold thrombosis (angiographic appearance of thrombus within or adjacent to a previously treated target lesion)~In the absence of angiography, any unexplained death, or acute MI (ST segment elevation or new Q-wave)* in the distribution of the target lesion within 30 days *(Non-specific ST /T changes and cardiac enzyme elevations do not suffice)~Any thromboses that occur less than 30 days after the index procedure will not be counted as restenosis." (NCT00856856)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Absorb BVS0

[back to top]

Scaffold Volume

(NCT00856856)
Timeframe: 2 years

Interventionmm^3 (Mean)
Absorb BVS145.78

[back to top]

Scaffold Volume

(NCT00856856)
Timeframe: 3 years

Interventionmm^3 (Mean)
Absorb BVS145.07

[back to top]

Stent Volume

(NCT00856856)
Timeframe: 1 year

Interventionmm^3 (Mean)
Absorb BVS132.90

[back to top]

Strut Volume

(NCT00856856)
Timeframe: 1 year

Interventionmm^3 (Mean)
Absorb BVS3.02

[back to top]

Strut Volume

(NCT00856856)
Timeframe: 2 years

Interventionmm^3 (Mean)
Absorb BVS2.61

[back to top]

Strut Volume

(NCT00856856)
Timeframe: 3 years

Interventionmm^3 (Mean)
Absorb BVS3.28

[back to top]

Thrombus

(NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Thrombus

(NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Thrombus

(NCT00856856)
Timeframe: 2 years

InterventionPercentage of participants (Number)
Absorb BVS2.6

[back to top]

Thrombus

(NCT00856856)
Timeframe: 3 years

InterventionPercentage of participants (Number)
Absorb BVS2.08

[back to top]

Thrombus

(NCT00856856)
Timeframe: 5 years

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Tissue Coverage Area BVS (Neointimal Area)

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS1.38

[back to top]

Tissue Coverage Area BVS (Neointimal Area)

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS1.99

[back to top]

Tissue Coverage Area BVS (Neointimal Area)

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS2.25

[back to top]

Tissue Coverage Area Classical

(NCT00856856)
Timeframe: 1 year

Interventionmm^2 (Mean)
Absorb BVS1.54

[back to top]

Tissue Coverage Area Classical

(NCT00856856)
Timeframe: 2 years

Interventionmm^2 (Mean)
Absorb BVS2.13

[back to top]

Tissue Coverage Area Classical

(NCT00856856)
Timeframe: 3 years

Interventionmm^2 (Mean)
Absorb BVS2.44

[back to top]

Tissue Coverage Obstruction Volume BVS

(NCT00856856)
Timeframe: 1 year

InterventionPercent of Tissue Coverage Obstruction (Mean)
Absorb BVS19.40

[back to top]

Tissue Coverage Obstruction Volume BVS

(NCT00856856)
Timeframe: 2 years

InterventionPercent of Tissue Coverage Obstruction (Mean)
Absorb BVS25.22

[back to top]

Tissue Coverage Obstruction Volume BVS

(NCT00856856)
Timeframe: 3 years

InterventionPercent of Tissue Coverage Obstruction (Mean)
Absorb BVS27.01

[back to top]

Ischemia Driven Target Lesion Revascularization (ID-TLR)

"ID-TLR is defined as the revascularization at the target lesion associated with any of the following:~Positive functional ischemia study~Ischemic symptoms and angiographic minimal lumen diameter stenosis ≥ 50% by core laboratory quantitative coronary angiography (QCA)~Revascularization of a target lesion with diameter stenosis ≥ 70% by core laboratory QCA without either ischemic symptoms or a positive functional study." (NCT00856856)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Absorb BVS6.0

[back to top]

Tissue Coverage Obstruction Volume Classical

(NCT00856856)
Timeframe: 2 years

InterventionPercent of Tissue Coverage Obstruction (Mean)
Absorb BVS27.10

[back to top]

Tissue Coverage Obstruction Volume Classical

(NCT00856856)
Timeframe: 3 years

InterventionPercent of Tissue Coverage Obstruction (Mean)
Absorb BVS29.36

[back to top]

Tissue Coverage Volume BVS

(NCT00856856)
Timeframe: 1 year

Interventionmm^3 (Mean)
Absorb BVS24.40

[back to top]

Tissue Coverage Volume BVS

(NCT00856856)
Timeframe: 2 years

Interventionmm^3 (Mean)
Absorb BVS35.51

[back to top]

Tissue Coverage Volume BVS

(NCT00856856)
Timeframe: 3 years

Interventionmm^3 (Mean)
Absorb BVS38.47

[back to top]

Tissue Coverage Volume Classical

(NCT00856856)
Timeframe: 1 year

Interventionmm^3 (Mean)
Absorb BVS27.27

[back to top]

Tissue Coverage Volume Classical

(NCT00856856)
Timeframe: 2 years

Interventionmm^3 (Mean)
Absorb BVS38.12

[back to top]

Tissue Coverage Volume Classical

(NCT00856856)
Timeframe: 3 years

Interventionmm^3 (Mean)
Absorb BVS41.76

[back to top]

Volume Obstruction (VO)

Defined as scaffold intimal hyperplasia and calculated as 100*(Scaffold Volume - Lumen Volume)/Scaffold Volume by IVUS. (NCT00856856)
Timeframe: 1 year

Interventionpercent of scaffold volume (Mean)
Absorb BVS1.47

[back to top]

Volume Obstruction (VO)

Defined as scaffold intimal hyperplasia and calculated as 100*(Scaffold Volume - Lumen Volume)/Scaffold Volume by IVUS. (NCT00856856)
Timeframe: 180 days

Interventionpercent of scaffold volume (Mean)
Absorb BVS1.22

[back to top]

Volume Obstruction (VO)

Defined as scaffold intimal hyperplasia and calculated as 100*(Scaffold Volume - Lumen Volume)/Scaffold Volume by IVUS. (NCT00856856)
Timeframe: 2 year

Interventionpercent of scaffold volume (Mean)
Absorb BVS3.33

[back to top]

Volume Obstruction (VO)

Defined as scaffold intimal hyperplasia and calculated as 100*(Scaffold Volume - Lumen Volume)/Scaffold Volume by IVUS. (NCT00856856)
Timeframe: 3 year

Interventionpercent of scaffold volume (Mean)
Absorb BVS4.19

[back to top]

Vasomotion Analysis: In-scaffold Mean Luminal Diameter

Vasomotion function was assessed in reaction to nitrate administration. (NCT00856856)
Timeframe: 5 years

InterventionMillimeter (Mean)
Pre-NitroPost-Nitro
Absorb BVS2.492.56

[back to top]

Cardiac Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)" (NCT00856856)
Timeframe: 5 years

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

Clinical Device Success (Per Lesion)

Successful delivery and deployment of the Clinical Investigation scaffold at the intended target lesion and successful withdrawal of the scaffold delivery system with attainment of final residual stenosis of less than 50% of the target lesion by QCA (by visual estimation if QCA unavailable). Standard pre-dilation catheters and post-dilatation catheters (if applicable) may be used. Bailout patients will be included as device success only if the above criteria for clinical device are met. (NCT00856856)
Timeframe: On day 0 (the day of procedure)

Interventionpercentage of lesions (Number)
Absorb BVS100.0

[back to top]

Clinical Procedure Success (Per Patient)

Successful delivery and deployment of the Clinical Investigation scaffold at the intended target lesion and successful withdrawal of the scaffold delivery system with attainment of final residual stenosis of less than 50% of the target lesion by QCA (by visual estimation if QCA unavailable) and/or using any adjunctive device without the occurrence of ischemia-driven major adverse cardiac event (MACE) during the hospital stay with a maximum of first seven days post index procedure. (NCT00856856)
Timeframe: On day 0 (the day of procedure)

Interventionpercentage of participants (Number)
ABSORB Stent98.0

[back to top]

Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 1 Year

Distal Late Loss: distal MLD post-procedure - distal MLD at follow-up (distal defined as within 5 mm of healthy tissue distal to scaffold placement). (NCT00856856)
Timeframe: 1 year

InterventionMillimeter (Mean)
Absorb BVS0.07

[back to top]

Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 180 Days

Distal Late Loss: distal MLD post-procedure - distal MLD at follow-up (distal defined as within 5 mm of healthy tissue distal to scaffold placement). (NCT00856856)
Timeframe: 180 days

InterventionMillimeter (Mean)
Absorb BVS0.07

[back to top]

Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 2 Years

Distal Late Loss: distal MLD post-procedure - distal MLD at follow-up (distal defined as within 5 mm of healthy tissue distal to scaffold placement). (NCT00856856)
Timeframe: 2 years

InterventionMillimeter (Mean)
Absorb BVS0.04

[back to top]

Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 3 Years

Distal Late Loss: distal MLD post-procedure - distal MLD at follow-up (distal defined as within 5 mm of healthy tissue distal to scaffold placement). (NCT00856856)
Timeframe: 3 years

InterventionMillimeter (Mean)
Absorb BVS0.08

[back to top]

Distal Late Loss: Distal MLD Post-procedure - Distal MLD at 5 Years

Distal Late Loss: distal MLD post-procedure - distal MLD at follow-up (distal defined as within 5 mm of healthy tissue distal to scaffold placement). (NCT00856856)
Timeframe: 5 years

InterventionMillimeter (Mean)
Absorb BVS0.11

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Absorb BVS6.9

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 180 days

Interventionpercentage of participants (Number)
Absorb BVS5.0

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Absorb BVS9.0

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 270 days

Interventionpercentage of participants (Number)
Absorb BVS5.0

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Absorb BVS10.0

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 30 days

Interventionpercentage of participants (Number)
Absorb BVS2.0

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Absorb BVS10.1

[back to top]

Hierarchical Major Adverse Cardiac Event (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction,and clinically indicated target lesion revascularization (CI-TLR). (NCT00856856)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Absorb BVS11.0

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Absorb BVS6.9

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
ABSORB Stent5.0

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Absorb BVS11.0

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 270 days

InterventionPercentage of participants (Number)
Absorb Stent5.0

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Absorb BVS13.0

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 30 days

Interventionpercentage of participants (Number)
Absorb BVS2.0

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Absorb BVS13.1

[back to top]

Hierarchical Target Vessel Failure (TVF)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT00856856)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Absorb BVS14.0

[back to top]

In-scaffold Angiographic Binary Restenosis (ABR)

Percent of patients with a followup percent diameter stenosis of >=50% per QCA. (NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS3.5

[back to top]

In-scaffold Angiographic Binary Restenosis (ABR)

Percent of patients with a followup percent diameter stenosis of >=50% per QCA. (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

In-scaffold Angiographic Binary Restenosis (ABR)

Percent of patients with a followup percent diameter stenosis of >=50% per QCA. (NCT00856856)
Timeframe: 2 years

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

In-scaffold Angiographic Binary Restenosis (ABR)

Percent of patients with a followup percent diameter stenosis of >=50% per QCA. (NCT00856856)
Timeframe: 3 years

InterventionPercentage of participants (Number)
Absorb BVS7.8

[back to top]

In-scaffold Angiographic Binary Restenosis (ABR)

Percent of patients with a followup percent diameter stenosis of >=50% per QCA. (NCT00856856)
Timeframe: 5 years

InterventionPercentage of participants (Number)
Absorb BVS7.8

[back to top]

In-scaffold Late Loss (LL): In-scaffold MLD Post-procedure - In-scaffold MLD at 2 Years

In-scaffold Late Loss: in-scaffold MLD post-procedure - in-scaffold MLD at follow-up. (NCT00856856)
Timeframe: 2 years

InterventionMillimeter (Mean)
Absorb BVS0.27

[back to top]

In-scaffold Late Loss (LL): In-scaffold MLD Post-procedure - In-scaffold MLD at 3 Years

In-scaffold Late Loss: in-scaffold MLD post-procedure - in-scaffold MLD at follow-up. (NCT00856856)
Timeframe: 3 years

InterventionMillimeter (Mean)
Absorb BVS0.29

[back to top]

In-scaffold Late Loss (LL): In-scaffold MLD Post-procedure - In-scaffold MLD at 5 Years

In-scaffold Late Loss: in-scaffold MLD post-procedure - in-scaffold MLD at follow-up. (NCT00856856)
Timeframe: 5 years

InterventionMillimeter (Mean)
Absorb BVS0.26

[back to top]

In-scaffold Late Loss: In-scaffold MLD Post-procedure - In-scaffold MLD at 1 Year

In-scaffold Late Loss: in-scaffold MLD post-procedure - in-scaffold MLD at follow-up (NCT00856856)
Timeframe: 1 year

InterventionMillimeter (Mean)
Absorb BVS0.27

[back to top]

In-scaffold Late Loss: In-scaffold MLD Post-procedure - In-scaffold MLD at 180 Days

In-scaffold Late Loss: in-scaffold MLD post-procedure - in-scaffold MLD at follow-up. (NCT00856856)
Timeframe: 180 days

InterventionMillimeter (Mean)
Absorb BVS0.19

[back to top]

Tissue Coverage Obstruction Volume Classical

(NCT00856856)
Timeframe: 1 year

InterventionPercent of Tissue Coverage Obstruction (Mean)
Absorb BVS21.61

[back to top]

% of Acutely Covered Struts

(NCT00856856)
Timeframe: 2 years

InterventionPercent of Covered Struts (Mean)
Absorb BVS98.07

[back to top]

% of Acutely Covered Struts

(NCT00856856)
Timeframe: 3 years

InterventionPercent of Covered Struts (Mean)
Absorb BVS97.90

[back to top]

% of Covered Struts (150 µm)

(NCT00856856)
Timeframe: 1 year

InterventionPercent of Covered Struts (Mean)
Absorb BVS96.86

[back to top]

% of Uncovered Struts (150 µm)

(NCT00856856)
Timeframe: 1 year

InterventionPercent of Uncovered Struts (Mean)
Absorb BVS3.14

[back to top]

% of Uncovered Struts (150 µm)

(NCT00856856)
Timeframe: 2 years

InterventionPercent of Uncovered Struts (Mean)
Absorb BVS1.93

[back to top]

% of Uncovered Struts (150 µm)

(NCT00856856)
Timeframe: 3 years

InterventionPercent of Uncovered Struts (Mean)
Absorb BVS2.10

[back to top]

Aneurysm

An abnormal expansion or protrusion of a coronary blood vessel resulting from a disease or weakening of the vessel's wall (all three layers) that exceeds the RVD of the vessel by 1.5 times. (NCT00856856)
Timeframe: 1 year

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Aneurysm

An abnormal expansion or protrusion of a coronary blood vessel resulting from a disease or weakening of the vessel's wall (all three layers) that exceeds the RVD of the vessel by 1.5 times. (NCT00856856)
Timeframe: 180 days

InterventionPercentage of participants (Number)
Absorb BVS2.4

[back to top]

Aneurysm

An abnormal expansion or protrusion of a coronary blood vessel resulting from a disease or weakening of the vessel's wall (all three layers) that exceeds the RVD of the vessel by 1.5 times. (NCT00856856)
Timeframe: 2 years

InterventionPercentage of participants (Number)
Absorb BVS2.6

[back to top]

Aneurysm

An abnormal expansion or protrusion of a coronary blood vessel resulting from a disease or weakening of the vessel's wall (all three layers) that exceeds the RVD of the vessel by 1.5 times. (NCT00856856)
Timeframe: 3 years

InterventionPercentage of participants (Number)
Absorb BVS2.08

[back to top]

Aneurysm

An abnormal expansion or protrusion of a coronary blood vessel resulting from a disease or weakening of the vessel's wall (all three layers) that exceeds the RVD of the vessel by 1.5 times. (NCT00856856)
Timeframe: 5 years

InterventionPercentage of participants (Number)
Absorb BVS0.0

[back to top]

Cardiac Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)" (NCT00856856)
Timeframe: 1 year

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

Cardiac Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)" (NCT00856856)
Timeframe: 2 years

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

Cardiac Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)" (NCT00856856)
Timeframe: 3 years

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

Cardiac Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)" (NCT00856856)
Timeframe: 30 days

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

Cardiac Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)" (NCT00856856)
Timeframe: 4 years

Interventionpercentage of participants (Number)
ABSORB Stent0

[back to top]

In-scaffold Percent Diameter Stenosis (%DS)

Percent Diameter Stenosis is defined as the value calculated as 100 * (1 - MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00856856)
Timeframe: 1 year

Interventionpercentage of diameter stenosis (Mean)
Absorb BVS21.35

[back to top]

In-scaffold Percent Diameter Stenosis (%DS)

Percent Diameter Stenosis is defined as the value calculated as 100 * (1 - MLD/RVD) using the mean values from two orthogonal views (when possible) by QCA. (NCT00856856)
Timeframe: 180 days

Interventionpercentage of diameter stenosis (Mean)
Absorb BVS19.21

[back to top]

Change in Visual Acuity From Baseline to Week 4 in Patients Treated With Everolimus

Best corrected visual acuity (BCVA) was assessed on both eyes. BCVA measurements were taken in sitting position using Early Treatment Diabetic Retinopathy Study (ETDRs)-like visual acuity testing charts at an initial testing distance specific to test charts. BCVA is measured from the number of letters the patient can read on the eye chart. (NCT00857259)
Timeframe: Baseline and week 4

,,
InterventionLetters (Mean)
Baseline4 WeeksChange in baseline
Everolimus 5 mg4643-3
Everolimus 5 mg and Ranibizumab 0.5 mg55.363.24.4
Ranibizumab 0.5 mg67.069.02.0

[back to top]

Change in Central Retinal Thickness From Baseline to Week 4, as Measured by Optical Coherence Tomography (OCT)

Central retinal thickness was assessed by Optical coherence tomography (OCT). The primary thickness endpoint was the mean thickness of the foveal field of the macula map produced by the analysis of the sequence of six radial scans. Foveal field thickness was the average thickness of a circular field with a diameter of 1 mm. OCT images were analyzed by a central reading center. (NCT00857259)
Timeframe: Baseline and 4 weeks

,,
Interventionµm (Mean)
BaselineWeek 4Change from Baseline
Everolimus 5 mg and Ranibizumab 0.5 mg244.3217.6-41.0
Oral Everolimus 5 mg454.8492.237.3
Ranibizumab 0.5 mg306.0308.02.0

[back to top]

Occurrence of Treatment Failures From Month 6 to 9 and Month 9 to 18

Treatment failure was defined as composite endpoint of biopsy proven acute rejection of ISHLT 1990 grade ≥ 3A resp. ISHLT 2004 grade ≥ 2R, acute rejection episodes associated with hemodynamic compromise, graft loss / re-transplant, death, loss to follow up (at least one condition must be present). If participant had an occurrence in each period it was counted for each period. (NCT00862979)
Timeframe: Month 6 to Month 9; Month 9 to Month 18

,
InterventionOccurences (Number)
Month 6 to Month 9 Treatment failure - all reasonsMonth 9 to Month 18 Treatment failure-all reasons
CNI-free-regimen415
CNI-regimen13

[back to top]

Calculated Glomerular Filtration Rate (cGFR) According to Cockcroft-Gault at Month 12 and 18

Calculated Glomerular Filtration Rate (cGFR) according to Cockcroft-Gault at Month 12 and 18. For men: GFR=(140-Age) x Body weight (kg) / 72 x Serum Creatinine (mg/dl) For women: GFR=0.85 (140 -Age) x Body weight(kg)/ 72 x Serum Creatinine (mg/dl) (NCT00862979)
Timeframe: Month 12 and 18

,
InterventionmL/min (Mean)
Month 12Month 18
CNI-free-regimen69.866.9
CNI-regimen54.254.2

[back to top]

Serum Creatinine at Month 6, 8, 9, 10 12 and 18

Serum Creatinine is an indicator of renal function measured in the blood (NCT00862979)
Timeframe: Month 6, 8, 9, 10 12 and 18

,
Interventionμmol/L (Mean)
Month 6Month 8Month 9Month 10Month 12Month 18
CNI-free-regimen1.491.271.241.201.221.27
CNI-regimen1.531.441.581.531.531.50

[back to top]

Reciprocal Creatinine Slope Between Month 6 and Month 18

Reciprocal Creatinine Slope is an indication of renal function over time with a higher slope value indicating an improvement in renal function. (NCT00862979)
Timeframe: Between Month 6 and Month 18

Intervention1/(μmol/L)/(hour) (Mean)
CNI-regimen0.045
CNI-free-regimen0.403

[back to top]

Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula at Month 18

Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula at Month 18 cGFR (in mL/min/1.73 m2) = 186.3*(C-1.154)*(A-0.203)*G*R where C = the serum concentration of creatinine (mg/dL), A = age (years), G = 0.742 when gender is female, otherwise G = 1, R = 1.21 when race is black, otherwise R = 1. (NCT00862979)
Timeframe: Month 18

InterventionmL/min (Mean)
CNI-regimen54.2
CNI-free-regimen66.9

[back to top]

Occurrence of Major Cardiac Events (MACE) From Month 6 to 18

Major cardiac events (MACE) was defined as one of the following: any death, myocardial infarction, coronary artery bypass grafting (NCT00862979)
Timeframe: Month 6 to Month 18

,
InterventionOccurences (Number)
Myocardial infarctionDeath
CNI-free-regimen01
CNI-regimen10

[back to top]

Overall Response Rate (ORR)

Overall response rate (ORR) is the percentage of patients with a best overall response of complete response (CR) or partial response (PR) according to RECIST 1.0. Per RECIST criteria 1.0, CR: Disappearance of all target lesions; PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters. (NCT00863655)
Timeframe: up to 21 months

InterventionPercentage of participants (Number)
Everolimus + Exemestane9.5
Placebo + Exemestane0.4

[back to top]

Estradiol Plasma Concentrations

Compare estradiol concentrations from baseline to week 4 in both treatment arms. (NCT00863655)
Timeframe: Baseline, Week 4

,
Interventionpg/mL (Mean)
Baseline (n: = 41, 14)Week 4 (n: 38, 15)
Everolimus + Exemestane5.623.50
Placebo + Exemestane4.095.17

[back to top]

Clinical Benefit Rate (CBR)

CBR is defined as the percentage of patients with best overall response of either complete response (CR), a partial response (PR) or stable disease (SD) >= 24 weeks, according to RECIST 1.0. Per RECIST criteria 1.0, CR: Disappearance of all target lesions; PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters; SD = Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for PD; PD = At least a 20% increase in the sum of the longest diameter of all measured target lesions, taking as reference the smallest sum of longest diameter of all target lesions recorded at or after baseline. (NCT00863655)
Timeframe: up to 21 months

InterventionPercentage of participants (Number)
Everolimus + Exemestane33.4
Placebo + Exemestane18.0

[back to top]

Duration of Response (Among Participants With Best Overall Response of CR or PR) Estimated Per Kaplan-Meier

Duration of response of CR or PR based on investigator applies only to patients whose best overall response was CR or PR (RECIST 1.0). The start date was the date of first documented response (CR or PR) and the end date and censoring is defined the same as that for time to progression. Per RECIST criteria 1.0, CR: Disappearance of all target lesions; PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters. (NCT00863655)
Timeframe: 21 months

InterventionMonths (Median)
Everolimus + Exemestane8.21
Placebo + ExemestaneNA

[back to top]

Overall Survival (OS) by Number of Deaths

Overall survival, the key secondary endpoint in this study, is defined as the time from date of randomization to the date of death due to any cause. If a patient is not known to have died, survival was censored at the date of last contact. (NCT00863655)
Timeframe: up to 53 months

InterventionParticipants (Number)
Everolimus + Exemestane267
Placebo + Exemestane143

[back to top]

Progression-free Survival (PFS) Based on Local Radiology Review of Tumor Assessments.

Progression-free survival, the primary endpoint in this study, is defined as the time from the date of randomization to the date of first documented radiological progression or death due to any cause. Disease progression was based on the tumor assessment by the local radiologist or investigator using RECIST 1.0 criteria. If a patient did not progress or known to have died at the date of the analysis cut-off or start of another antineoplastic therapy, the PFS date was censored to the date of last adequate tumor assessment prior to cut-off date or start of antineoplastic therapy. For patients with lytic or mixed (lytic+sclerotic) bone lesions, the following is considered progression: appearance of ≥1 new lytic lesions in bone; the appearance of ≥ new lesions outside of bone and unequivocal progression of existing bone lesions. (NCT00863655)
Timeframe: date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first, reported between day of first patient randomized up to about 19 months

Interventionmonths (Median)
Everolimus + Exemestane6.93
Placebo + Exemestane2.83

[back to top]

Overall Survival (OS) by Median

Overall survival, the key secondary endpoint in this study, is defined as the time from date of randomization to the date of death due to any cause. (NCT00863655)
Timeframe: up to 53 months

InterventionMonths (Median)
Everolimus + Exemestane30.98
Placebo + Exemestane26.55

[back to top]

Proportion of Patients With no Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG PS) Using Kaplan-Meier

The ECOG PS (Eastern Cooperative Oncology Group Performance Scale) is a standard criteria for measuring how treatment of cancer impacts level of functioning in terms of the ability to care for oneself, daily activity, & physical ability (walking, working, etc.). Scale score ranges:0 to 5, 5 being the worst. Scale index: 0: Fully active, able to carry on all pre-disease performance without restriction. 1: Restricted in physically strenuous activity but ambulatory & able to carry out work of a light or sedentary nature. 2: Ambulatory & capable of all self-care but unable to carry out any work activities. Up & about more than 50% of waking hours. 3: Capable of only limited self-care, confined to bed or chair more than 50% of waking hours. 4 - Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair. 5 - Dead. A deterioration of ECOG is an increase of 1 of the ECOG PS without improvement back to initial level at a subsequent time of measurement. (NCT00863655)
Timeframe: 2, 4, 6, 9 months

,
InterventionProportion of patients (Number)
2 Months4 Months6 Months9 Months
Everolimus + Exemestane0.840.740.640.57
Placebo + Exemestane0.870.800.670.47

[back to top]

Proportion of Patients With Having no Overall Response Based on Investigator Assessment

overall response = complete response (CR) + partial response (PR) per RECIST 1.0 Time to overall response (CR or PR) based on investigator is the time between date of randomization/start of treatment until first documented response (CR or PR). This analysis included all patients/responders. Patients who did not achieve a confirmed PR or CR were censored at last adequate tumor assessment date when they did not progress. Per RECIST criteria 1.0, CR: Disappearance of all target lesions; PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters. (NCT00863655)
Timeframe: 2, 4, 6, 9 months

,
InterventionProportion of patients (Number)
2 months4 months6 months9 months
Everolimus + Exemestane0.960.930.920.90
Placebo + Exemestane1.001.001.001.00

[back to top]

Patient-reported Outcomes (PROs): Time to Deterioration of PRO Scores Using Kaplan Meier - EORTC QLQ-C30

"The QLQ-C30 is composed of both multi-item scales and single-item measures. These include 5 functional scales, 3 symptom scales, a global health status - QoL scale, and 6 single items. Each of the multi-item scales includes a different set of items - no item occurs in more than 1 scale. All of the scales measures range in score from 0 to 100. A high scale score = higher response level. Thus a high score for a functional scale represents a healthy level of function, a high score for the global health status / QoL represents a high quality of life but a high score for a symptom scale / item represents a high level of symptomatology / problems. The principle for scoring these scales: 1.) Estimate the average of the items that contribute to the scale = raw score. 2.) Linear transformation to standardize the raw score, so that scores range from 0 to 100; a higher score represents a higher (better) level of functioning, or a higher (worse) level of symptoms." (NCT00863655)
Timeframe: Up to 21 months

,
InterventionMonths (Median)
Deterioration global health status score ≥ 5%Deterioration in PF domain score of ≥ 5%Deterioration in EF domain score of ≥ 5%Deterioration in SF domain score of ≥ 5%
Everolimus + Exemestane4.534.836.938.34
Placebo + Exemestane4.404.376.937.03

[back to top]

Exemestane Concentrations at Week 4

Characterize the PK of exemestane in combination with or without everolimus using Cmin and C2h at week 4 in a small group of patients. (NCT00863655)
Timeframe: predose, 2 hours post-dose

,
Interventionng/mL (Mean)
Pre-dose (Cmin) (n: 34, n: 22)2 hours post-dose (C2h) (n: 39, n: 22)
Everolimus + Exemestane0.6323.16
Placebo + Exemestane0.4313.30

[back to top]

Everolimus Concentrations at Week 4

Characterize the pharmacokinetics (PK) of everolimus in combination with exemestane using Cmin (pre-dose) and C2h (post-dose) at week 4 in a small group of patients. (NCT00863655)
Timeframe: pre-dose, 2 hours post-dose

Interventionng/mL (Mean)
Pre-dose (Cmin) (n:22)2 hours post-dose (C2h) (n:24)
Everolimus + Exemestane16.0446.50

[back to top]

Progression-free Survival

Progression-free survival is defined as the duration of time from start of treatment to time of documentation of progression or death (NCT00869999)
Timeframe: 2 years

Interventionmonths (Median)
Everolimus/Rituximab2.9

[back to top]

Duration of Overall Response

Duration of overall response is measured from the time measurement criteria are met for complete response or partial response until the first date that recurrent or progressive disease is objectively documented. (NCT00869999)
Timeframe: 2 years

Interventionmonths (Median)
Everolimus/Rituximab8.1

[back to top]

Overall Response Rate

Complete response plus partial response after 6 cycles. Response rate will be evaluated by using the modified Cheson criteria for lymphoma response. Complete response requires all of the following: 1) PET positive prior to therapy: mass of any size permitted if PET negative. Variable FDG-avid or PET negative prior to therapy: regression to normal size on CT (/= 1.5 cm before therapy) 2) Spleen (if enlarged before therapy) must have regressed in size and must not be palpable, 3) If bone marrow is known to be involved, repeat biopsy documents clearance. Partial response requires 1) >/= 50% decrease in SPD, 2) No new sites of disease or increase in the size of other nodes, liver or spleen, 3) Splenic and hepatic nodules must regress by at least 50% in SPD (NCT00869999)
Timeframe: Assessed at the conclusion of cycle 2, cycle 4 and cycle 6

Interventionpercentage of participants (Number)
Everolimus/Rituximab38

[back to top]

Clinical Benefit Rate (CBR) Equal to or Greater Than 24 Weeks - Full Population

CBR is defined as the percentage of participants whose best overall response is either complete response (CR), a partial response (PR) or stable disease (SD) lasting for at least 24 weeks, according to RECIST. This was assessed in the full patient population. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

InterventionPercentage of participants (Number)
Everolimus + Paclitaxel + Trastuzumab75.8
Placebo + Paclitaxel + Trastuzumab81.2

[back to top]

Clinical Benefit Rate (CBR) Equal to or Greater Than 24 Weeks - HR-negative Population

CBR is defined as the percentage of participants whose best overall response is either complete response (CR), a partial response (PR) or stable disease (SD) lasting for at least 24 weeks, according to RECIST. This was assessed in a subset of patients with Hormone Receptor Negative disease. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

InterventionPercentage of participants (Number)
Everolimus + Paclitaxel + Trastuzumab78.8
Placebo + Paclitaxel + Trastuzumab79.6

[back to top]

Overall Response Rate (ORR) - HR-negative Population

ORR is defined as the percentage of participants whose best overall response is either complete response (CR) or partial response (PR) according to RECIST. This was assessed in a subset of patients with Hormone Receptor Negative disease. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

InterventionPercentage of participants (Number)
Everolimus + Paclitaxel + Trastuzumab73.1
Placebo + Paclitaxel + Trastuzumab70.9

[back to top]

Overall Survival (OS) - Full Population

OS is defined as the time from date of randomization to the date of death due to any cause. For patients with documented progression, survival follow up was performed either by telephone or clinic visit at least every 3 months. Additional survival updates were requested prior to interim or final analysis or prior to providing data to the health authorities. This was assessed in the full patient population. (NCT00876395)
Timeframe: up to about 76 months

InterventionMonths (Median)
Everolimus + Paclitaxel + Trastuzumab48.56
Placebo + Paclitaxel + Trastuzumab49.97

[back to top]

Time to Overall Response Based on Investigator - HR-negative Population

Time to overall response defined as the time between date of randomization until first documented response Complete reseponse (CR) or partial response (PR) ), according to RECIST. This was assessed in the full patient population and in a subset of patients with Hormone Receptor Negative disease. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

Interventionmonths (Median)
Everolimus + Paclitaxel + Trastuzumab1.94
Placebo + Paclitaxel + Trastuzumab1.97

[back to top]

Time to Overall Response Based on Investigator - Full Population

Time to overall response defined as the time between date of randomization until first documented response Complete reseponse (CR) or partial response (PR) ), according to RECIST. This was assessed in the full patient population and in a subset of patients with Hormone Receptor Negative disease. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

Interventionmonths (Median)
Everolimus + Paclitaxel + Trastuzumab2.10
Placebo + Paclitaxel + Trastuzumab2.00

[back to top]

Everolimus Blood Level Concentrations at Steady States for Everolimus

Blood levels at steady states for everolimus 10 mg/day and 5 mg/day. Only valid samples are included. Some patients had dose reduction to 5 mg daily dose therefore the everolimus blood concentration for them have been summarized separately. Cycle = 28 days (NCT00876395)
Timeframe: predose, 2 hours post-dose at Cycle 2/Day 1, Cycle 2/Day 15, Cycle 2/ Day 22

,
Interventionng/mL (Mean)
Pre-dose (Cmin) @ C2D12 hrs post administration (C2h) @ C2D1Pre-dose (Cmin) @ C2D152 hrs post administration (C2h) @ C2D15Pre-dose (Cmin) @ C2D222 hrs post administration (C2h) @ C2D22
Everolimus 10 mg/Day14.38044.48513.20643.49413.43243.947
Everolimus 5 mg/Day7.95923.4495.47320.3297.49422.192

[back to top]

Overall Response (OR) - Full Population

OR applies only to patients whose best OR was CR or PR. Start date = date of first documented response (CR or PR) and end date = date of documented response (CR or PR) and end date = date of event defined as the first documented progression or death due to underlying cause. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

,
InterventionPercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Everolimus + Paclitaxel + Trastuzumab5.661.5
Placebo + Paclitaxel + Trastuzumab5.963.2

[back to top]

Overall Response (OR) - HR-negative Population

OR applies only to patients whose best OR was CR or PR. Start date = date of first documented response (CR or PR) and end date = date of documented response (CR or PR) and end date = date of event defined as the first documented progression or death due to underlying cause. This was assessed in the HR-negative patient population. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

,
InterventionPercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Everolimus + Paclitaxel + Trastuzumab7.765.4
Placebo + Paclitaxel + Trastuzumab2.968.0

[back to top]

Paclitaxel Plasma Concentrations

Blood levels at steady states for everolimus/placebo (NCT00876395)
Timeframe: Cycle 2/Day 15 (Pre-infusion and end of infusion)

,
Interventionng/mL (Mean)
Pre-infusion (Cmin) @ C2D15End of infusion (Cmax) @ C2D15
Everolimus1.4245159.338
Everolimus Placebo04296.697

[back to top]

Time to Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG-PS) Score - HR-negative Population

Time to definitive deterioration of the ECOG PS by one category of the score from baseline will be performed. Baseline is the last available assessment on or before randomization date. A deterioration is considered definitive if no improvements in the ECOG PS status is observed at a subsequent time of measurement during the treatment period following the time point where the deterioration is observed. (NCT00876395)
Timeframe: up to about 56 months

Interventionmonths (Median)
Everolimus + Paclitaxel + TrastuzumabNA
Placebo + Paclitaxel + TrastuzumabNA

[back to top]

Time to Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG-PS) Score - Full Population

Time to definitive deterioration of the ECOG PS by one category of the score from baseline will be performed. Baseline is the last available assessment on or before randomization date. A deterioration is considered definitive if no improvements in the ECOG PS status is observed at a subsequent time of measurement during the treatment period following the time point where the deterioration is observed. (NCT00876395)
Timeframe: up to about 56 months

Interventionmonths (Median)
Everolimus + Paclitaxel + Trastuzumab39.20
Placebo + Paclitaxel + TrastuzumabNA

[back to top]

Progression-free Survival (PFS) Per Investigators' Assessment Based on Local Radiology Review - Full Population

PFS is defined as the time from the date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first. This was assessed in the full patient population. (NCT00876395)
Timeframe: date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first, reported between day of first patient randomized up to about 56 months

Interventionmonths (Median)
Everolimus + Paclitaxel + Trastuzumab14.95
Placebo + Paclitaxel + Trastuzumab14.49

[back to top]

Trastuzumab Serum Concentrations

Blood levels at steady states for everolimus/placebo (NCT00876395)
Timeframe: Cycle 4/Day 1 (Pre-infusion and end of infusion)

,
Interventionmicrogram/ml (Mean)
Pre-infusion (Cmin) @ C4D1End of infusion (Cmax) @ C4D1
Everolimus + Trastuzumab26.60664.296
Everolimus Placebo29.18067.643

[back to top]

Overall Response Rate (ORR) - Full Population

ORR is defined as the percentage of participants whose best overall response is either complete response (CR) or partial response (PR) according to RECIST. This was assessed in the full patient population. Complete response is achieved when all lesions evaluated at Baseline are absent at subsequent visit. (NCT00876395)
Timeframe: up to about 23 months

InterventionPercentage of participants (Number)
Everolimus + Paclitaxel + Trastuzumab67.1
Placebo + Paclitaxel + Trastuzumab69.0

[back to top]

Progression-free Survival (PFS) Per Investigators' Assessment Based on Local Radiology Review - (Hormone Receptor (HR)-Negative Population

PFS is defined as the time from the date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first. This was assessed in the HR-negative patient population. (NCT00876395)
Timeframe: date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first, reported between day of first patient randomized up to about 56 months

InterventionMonths (Median)
Everolimus + Paclitaxel + Trastuzumab20.27
Placebo + Paclitaxel + Trastuzumab13.08

[back to top]

Overall Survival (OS) - HR-negative Population

OS is defined as the time from date of randomization to the date of death due to any cause. For patients with documented progression, survival follow up was performed either by telephone or clinic visit at least every 3 months. Additional survival updates were requested prior to interim or final analysis or prior to providing data to the health authorities. This was assessed in the HR-negative patient population. (NCT00876395)
Timeframe: up to about 76 months

InterventionMonths (Median)
Everolimus + Paclitaxel + Trastuzumab56.97
Placebo + Paclitaxel + Trastuzumab41.63

[back to top]

Patient Reported Outcome (PRO): Time to Definitive Deterioration of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire 30 (EORTC QLQ-C30) Scores

The EORTC QLQ-C30 global health status/quality of life sub-scale (QL) was pre-specified as the primary domain of interest, followed by physical functioning (PF), social functioning (SF) and emotional functioning (EF).The EORTC QLQ-C30 questionnaire, along with a module specific for gastric cancer patients (EORTC QLQ-STO22), was used to evaluate PRO. The QLQ-C30 has five function scales (physical, role, cognitive, emotional and social), three symptom scales (fatigue, pain and nausea/vomiting) and a global health status/quality of life scale. In addition, there are questions that assess specific symptoms. The QLQ-STO22 consists of 22 questions that make up five multi-item scales (dysphagia, pain, reflux, eating and anxiety) and four single-item scales (dry mouth, tasting, body image and hair loss). (NCT00879333)
Timeframe: 2.5 years

,
InterventionMonths (Median)
In QL score by at least 5 % compared to baselineIn PF score by at least 5 % compared to baselineIn SF score by at least 5 % compared to baselineIn EF score by at least 5 % compared to baseline
Everolimus 10mg/Daily1.511.351.871.84
Placebo1.451.151.871.71

[back to top]

Overall Response Rate (ORR)

ORR was defined as the proportion of patients with measurable disease in whom best overall response (OR) was either complete response (CR) or partial response (PR) according to RECIST criteria. (NCT00879333)
Timeframe: 2.5 years

,
InterventionParticipants (Number)
Measurable DiseaseComplete Response (CR)Partial Response (PR)Overall Response Rate (ORR)
Everolimus 10mg/Daily37911617
Placebo191044

[back to top]

Progression Free Survival (PFS)

Progression free survival was defined as the time from the date of randomization to the date of the first documented disease progression or death due to any cause, where progression was based on Investigator assessment of baseline and post-baseline scans according to RECIST. Progression free survival was censored if no PFS event was observed before the first to occur out of (i) the cut-off date, or (ii) the date when a further anticancer therapy was started. The censoring date was the date of the last adequate tumor assessment before either of these two events occurred. If a PFS event was observed after two or more missing or non-evaluable tumor assessments, then the date of progression was censored at the date of the last adequate tumor assessment; for a PFS event observed after a single missing or non-evaluable tumor assessment, the actual date of disease progression was used. Anslsis was done using Kaplan-Meier estimates method. (NCT00879333)
Timeframe: 2.5 years

InterventionMonths (Median)
Everolimus 10mg/Daily1.68
Placebo1.41

[back to top]

Overall Survival (OS)

The primary objective of this study was to compare OS between everolimus + best supportive care (BSC) and placebo + BSC. OS, was defined as the time from date of randomization to the date of death due to any cause. If at the analysis cut-off date a patient was not known to have died, survival was censored at the date of the last contact. OS was analyzed using the Kaplan Meier estimates method. (NCT00879333)
Timeframe: 2.5 years

InterventionMonths (Median)
Everolimus 10mg/Daily5.39
Placebo4.34

[back to top]

Time to Definitive Deterioration of Eastern Cooperative Oncology Group Performance Status (ECOG PS) Score

The ECOG PS scale was used to classify patients according to their functional impairment, with scores ranging from 0 (fully active) to 5 (dead). An analysis of the time to definitive deterioration of the ECOG PS by one category of the score from baseline was performed. Definitive deterioration was defined as a definitive increase by one category from baseline in ECOG PS, with no later improvements observed during the course of the study. A single measure reporting an increase in ECOG PS is sufficient to consider it as a definitive worsening only if it was the last one available for the patient. Kaplan-Meier method was used to estimate the distribution function of time to definitive worsening. (NCT00879333)
Timeframe: 2.5 years

InterventionMonths (Median)
Everolimus 10mg/Daily2.30
Placebo2.23

[back to top]

Everolimus Steady State Concentraions at Predose (Cmin) and Cmax at Week 5

Cmin is the minimum (trough) steady-state drug concentration in the blood during multiple dosing and Cmax is the maximum (peak) blood drug concentration after dose administration. Cmax is estimated as the maximum of C1h and C2H. C1h is 1 hour post-dose blood concentration and C2h is 2 hour post-dose blood concentration. Only valid pre-dose (Cmin), C1h, and C2h everolimus samples were included in the analysis. Valid pre-dose samples were confirmed blood samples collected at steady-state, collected immediately prior to dosing on the same study day, and collected at approximately 24 ± 4 hours after the previous dose and with no vomiting within the first 4 hours following the last dose. Valid C1h and C2h samples were confirmed blood samples collected at steady-state and within ± 1 hour window and with no vomiting within the first 4 hours following the current and previous dose. (NCT00879333)
Timeframe: Week 5

,
Interventionng/mL (Mean)
Pre-dose (Cmin) (n: 201,18)Cmax (n: 218,16)
Everolimus 10mg/Daily16.14372.775
Everolimus 5 mg/Day10.49837.269

[back to top]

Everolimus Steady State Concentraions at Predose (Cmin) and Cmax by Region Asia vs. Rest of the World (ROW) at Week 5

Cmin is the minimum (trough) steady-state drug concentration in the blood during multiple dosing and Cmax is the maximum (peak) blood drug concentration after dose administration. Cmax is estimated as the maximum of C1h and C2H. C1h is 1 hour post-dose blood concentration and C2h is 2 hour post-dose blood concentration. Only valid pre-dose (Cmin), C1h, and C2h everolimus samples were included in the analysis. Valid pre-dose samples were confirmed blood samples collected at steady-state, collected immediately prior to dosing on the same study day, and collected at approximately 24 ± 4 hours after the previous dose and with no vomiting within the first 4 hours following the last dose. Valid C1h and C2h samples were confirmed blood samples collected at steady-state and within ± 1 hour window and with no vomiting within the first 4 hours following the current and previous dose. (NCT00879333)
Timeframe: Week 5

,
Interventionng/mL (Mean)
Asia: Pre-dose (n:127, 11)Asia: Cmax (n:132, 10)Rest of the World: Pre-dose (n:74, 7)Rest of the World: Cmax (n:86, 6)
Everolimus 10mg/Daily16.80473.56815.00971.558
Everolimus 5mg/Day9.92134.58011.40641.750

[back to top]

Progression-free Survival

The time from randomization until disease progression, death, or date of last contact. Endpoints are progression or death. Patients who are not observed with an endpoint are censored. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) as an 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00886691)
Timeframe: Duration of time from start of treatment to time of progression, approximately 4 years and 6 months.

Interventionmonths (Median)
Everolimus (RAD001) Plus Bevacizumab5.9
Placebo Plus Bevacizumab4.5

[back to top]

The Proportion of Patients With Measurable Disease Who Have Objective Tumor Responses by Treatment.

Complete and Partial Tumor Response by RECIST 1.0 (NCT00886691)
Timeframe: Up to approximately 4 years and 6 months

Interventionpercentage of participants responding (Number)
Everolimus (RAD001) Plus Bevacizumab22.2
Placebo Plus Bevacizumab12.1

[back to top]

Percentage of Participants With at Least One Cancer Antigen 125 (CA-125) Response

Response as evaluated by CA-125 levels.A CA 125 test measures the amount of the protein CA 125 (cancer antigen 125) in blood.CA 125 is a tumor marker recommended for clinical use in the diagnosis and management of ovarian cancer. CA-125 responses were assessed with Rustin criteria. Initial values had to be 2x ULN (upper limit of normal) within 2 weeks of starting therapy to be considered evaluable.Patient were evaluated by using best overall response while receiving study therapy. (NCT00886691)
Timeframe: Prior to each cycle of treatment. Then follow-up every three months for 2 years , then every 6 months for 3 years. Duration was approximately 4 years and 6 months.

Interventionpercentage of participants (Number)
Everolimus (RAD001) Plus Bevacizumab27.3
Placebo Plus Bevacizumab15.4

[back to top]

Characterize and Compare Progression-free Survival and Overall Survival in Patients With Measurable Disease (RECIST Criteria) and Patients With Detectable (Non-measurable) Disease

Progression-free survival and overall survival broken down by measurable disease status (NCT00886691)
Timeframe: Continued until disease progression, assessed up to approximately 4 years and 6 months

InterventionMonths (Median)
PFS (Progression Free Survival) in Non-measurable Patients3.94
PFS (Progression Free Survival) in Measurable Patients5.91
OS (Overall Survival) in Non-measurable Patients11.10
OS (Overall Survival) in Measurable Patients16.92

[back to top]

Incidence of Adverse Events as Assessed by Common Terminology Criteria for Adverse Events Version 4.0 (CTCAE v4.0)

Number of participants with a grade of 3 or higher during the treatment period. (NCT00886691)
Timeframe: All Adverse Events (AEs) and Serious Adverse Events (SAEs) occurring during treatment and up to 30 days after stopping the study treatment, approximately 4 years and 6 months.

InterventionParticipants (Count of Participants)
Everolimus (RAD001) Plus Bevacizumab43
Placebo Plus Bevacizumab20

[back to top]

Time to Definitive Deterioration of the Global Health Status/QoL Scores of the EORTC QLQ-C30 by First-Line Drug

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items.The standardized score for the PF, fatigue subscales and global health status ranges from 0 to 100, with a higher score representing a high level of functioning/high level of symptom/high quality of life. Definitive deterioration by at least 10% was defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the first line of treatment. A single measure reporting a decrease of at least 10% was considered definitive only if it was the last one available for the participant. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L7.92
Sunitinib 1L/Everolimus 2L12.25

[back to top]

Overall Response Rate (ORR) - First -Line (1-L)

ORR was defined as the number of participants with best overall response (BOR) of complete response (CR) or partial response (PR) and was based on investigator assessment of radiology data per RECIST. Participants with best overall response of 'Unknown' were treated as non-responders in the calculation of the ORR. Confirmed CR = at least two determinations of CR at least 4 weeks apart before progression. Confirmed PR = at least two determinations of PR or better at least 4 weeks apart before progression. CR required a disappearance of all target and non-target lesions. PR required at least a 30% decrease in the sum of the longest diameters of all target lesions, taking as a reference the baseline sum of the longest diameters. Radiological assessments : every 12 weeks until disease progression, the start of another antineoplastic therapy or for any other reason. (NCT00903175)
Timeframe: based on radiological assessments every 3 months until disease progression, start of another antineoplastic therapy or for any other reason up to 35 months

,
Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)Overall Response Rate (CR + PR)
Everolimus 1L/Sunitinib 2L11819
Sunitinib 1L/Everolimus 2L35962

[back to top]

Time to Definitive Deterioration of the Physical Functioning Scale of the EORTC QLQ-C30 - by First and Second-Line Drugs Combined

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items.The standardized score for the PF, fatigue subscales and global health status ranges from 0 to 100, with a higher score representing a high level of functioning/high level of symptom/high quality of life. Definitive deterioration by at least 10% was defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the first line or second line treatment. A single measure reporting a decrease of at least 10% was considered definitive only if it was the last one available for the participant. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L12.25
Sunitinib 1L/Everolimus 2L14.03

[back to top]

Time to Definitive Deterioration of the Global Health Status/QoL Scores of the EORTC QLQ-C30 by First and Second-Line Drugs Combined

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items. The standardized score for the PF, fatigue subscales and global health status ranges from 0 to 100, with a higher score representing a high level of functioning/high level of symptom/high quality of life. Definitive deterioration by at least 10% was defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the first line or second line treatment. A single measure reporting a decrease of at least 10% was considered definitive only if it was the last one available for the participant. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L10.84
Sunitinib 1L/Everolimus 2L12.71

[back to top]

Time to Definitive Deterioration of the FKSI-DRS Risk Score by at Least 3 Score Units by First-line Drug

The Functional Assessment of Cancer Therapy - Kidney Symptom Index, Disease Related Symptoms (FKSI-DRS) is a set of items to assess symptoms experienced by patients with advanced kidney cancer. These symptoms include fatigue, pain, weight loss, dyspnea, cough, fever and hematuria. Each item is scored on a 5-point scale (0 = not at all; 4 = very much). The FKSI-DRS total score ranges from 0 (most severe symptoms) to 36 (no symptoms). Definitive deterioration was defined as a decrease by at least 3 units compared to baseline, with no later increase above this threshold observed during the 1-L of treatment. A single measure reporting a decrease of at least 3 units was considered definitive only if it was the last one available for the patient. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L12.65
Sunitinib 1L/Everolimus 2L16.66

[back to top]

Time to Definitive Deterioration of the FKSI-DRS Risk Score by at Least 3 Score Units by First and Second-line Drugs Combined

The Functional Assessment of Cancer Therapy - Kidney Symptom Index, Disease Related Symptoms (FKSI-DRS) is a set of items to assess symptoms experienced by patients with advanced kidney cancer. These symptoms include fatigue, pain, weight loss, dyspnea, cough, fever and hematuria. Each item is scored on a 5-point scale (0 = not at all; 4 = very much). The FKSI-DRS total score ranges from 0 (most severe symptoms) to 36 (no symptoms). Definitive deterioration was defined as a decrease by at least 3 units compared to baseline, with no later increase above this threshold observed during the 1-L or 2-L treatment. A single measure reporting a decrease of at least 3 units was considered definitive only if it was the last one available for the patient. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L14.23
Sunitinib 1L/Everolimus 2L15.97

[back to top]

Time to Definitive Deterioration of the Fatigue Scale of the EORTC QLQ-C30 by First-Line Drug

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items.The standardized score for the PF, fatigue subscales and global health status ranges from 0 to 100, with a higher score representing a high level of functioning/high level of symptom/high quality of life. Definitive deterioration by at least 10% was defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the first line of treatment. A single measure reporting a decrease of at least 10% was considered definitive only if it was the last one available for the participant. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L9.20
Sunitinib 1L/Everolimus 2L11.37

[back to top]

Time to Definitive Deterioration of the Fatigue Scale of the EORTC QLQ-C30 by First and Second-Line Drugs Combined

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items. The standardized score for the PF, fatigue subscales and global health status ranges from 0 to 100, with a higher score representing a high level of functioning/high level of symptom/high quality of life. Definitive deterioration by at least 10% was defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the first line or second line treatment. A single measure reporting a decrease of at least 10% was considered definitive only if it was the last one available for the participant. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Mean)
Everolimus 1L/Sunitinib 2L11.56
Sunitinib 1L/Everolimus 2L13.34

[back to top]

Progression-free Survival Combined (PFS-C)

PFS-C (1L and 2L study drugs combined) was a composite endpoint which combined both lines of study treatment. It was defined as the time from the date of randomization to the first of the following: date of death due to any cause, or date of the first radiologically documented progression disease during or after the second-line treatment period for patients with a radiologically documented progression disease in the first-line treatment period and who had crossed-over to second-line treatment no more than 6 weeks after progression. (NCT00903175)
Timeframe: based on radiological assessments every 3 months until disease progression, start of another antineoplastic therapy or for any other reason up to about 56 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L21.68
Sunitinib 1L/Everolimus 2L22.18

[back to top]

Progression Free Survival First-Line (PFS 1-L)

PFS_1L based on investigator assessment of radiology data by RECIST 1.0, was defined as the time from the date of randomization to the date of the first documented disease progression or death due to any cause during or after first-line treatment with everolimus or sunitinib. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00903175)
Timeframe: based on radiological assessments every 3 months until disease progression, start of another antineoplastic therapy or for any other reason up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L7.85
Sunitinib 1L/Everolimus 2L10.71

[back to top]

Overall Survival (OS)

Overall survival was defined as the time from date of randomization to date of death due to any cause. The analysis of OS included all deaths in the FAS regardless of when they were observed. (NCT00903175)
Timeframe: Every 2 months from randomization up to 3 years after last patient randomized

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L22.41
Sunitinib 1L/Everolimus 2L29.47

[back to top]

Duration of Response (DoR) - First-Line (1-L)

Duration of overall response (CR or PR) applies only to patients whose Best Overall Response (BOR) was Complete Response (CR) or Partial Response (PR) during the first-line treatment period. The start date was the date of first documented response (CR or PR) during the first-line treatment and the end date was the date of the event defined as the first documented progression or death due to underlying cancer during or after the same treatment line. (NCT00903175)
Timeframe: based on radiological assessments every 3 months until disease progression, start of another antineoplastic therapy or for any other reason up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L13.37
Sunitinib 1L/Everolimus 2L17.25

[back to top]

Time to Definitive Deterioration of the Physical Functioning (PF) Scale of the EORTC QLQ-C30 - by First-Line (1L) Drug

The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) contains 30 items. These include a global health status/QoL scale, five functional scales, three symptom scales, and six single items. The standardized score for the PF, fatigue subscales and global health status ranges from 0 to 100, with a higher score representing a high level of functioning/high level of symptom/high quality of life. Definitive deterioration by at least 10% was defined as a decrease in score by at least 10% compared to baseline, with no later increase above this threshold observed during the first line of treatment. A single measure reporting a decrease of at least 10% was considered definitive only if it was the last one available for the participant. (NCT00903175)
Timeframe: <=14 days prior to the first dose of study medication, on day 1, day 28 of every cycle, at the end of treatment visit, at the 28 day FUP visit and monthly thereafter for up to 3 months or until initiation of another anticancer therapy up to 35 months

InterventionMonths (Median)
Everolimus 1L/Sunitinib 2L13.47
Sunitinib 1L/Everolimus 2L14.03

[back to top]

Progression-free Survival (PFS) Until First Progression

Median PFS will be calculated based on time to first progression or death. (NCT00912340)
Timeframe: Every 3 to 4 weeks after study start, until progression or death, assessed up to 5 years

Interventionmonths (Median)
Trastuzumab2.0
Everolimus5.7

[back to top]

Progression-free Survival (PFS) in Patients Who Crossed Over

Median PFS will be calculated based on time to first progression or death. (NCT00912340)
Timeframe: Every 3 to 4 weeks after study start, until progression or death, assessed up to 5 years

Interventionmonths (Median)
Trastuzumab6.3
Everolimus3.1

[back to top] [back to top]

Duration of Response (DOR)

Defined as time between date of objective response and date of response to disease progression or death, as defined by RECIST v1.1 criteria. Objective response is defined as either complete response [CR] or partial response [PR]. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT00915603)
Timeframe: every 8 weeks until treatment discontinuation, expected average 6 months

Interventionmonths (Median)
Paclitaxel/Bevacizumab/Everolimus7.8
Paclitaxel/Bevacizumab/Placebo6.0

[back to top]

Overall Response Rate (ORR)

The number of patients with observed complete response [CR] or partial response [PR]. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR (NCT00915603)
Timeframe: every 8 weeks until treatment discontinuation, expected average of 18 months

Interventionparticipants (Number)
Paclitaxel/Bevacizumab/Everolimus35
Paclitaxel/Bevacizumab/Placebo32

[back to top]

Overall Survival (OS)

Assessed from Day 1 of study drug administration to date of death due to any cause. (NCT00915603)
Timeframe: every 8 weeks until treatment discontinuation, expected average 6 months

Interventionmonths (Median)
Paclitaxel/Bevacizumab/Everolimus17.5
Paclitaxel/Bevacizumab/Placebo19.6

[back to top]

Progression-Free Survival (PFS)

Progression-free survival will be measured from Day 1 of study drug administration to disease progression defined by Response Evaluation Criteria in Solid Tumors Criteria (RECIST v1.1) as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT00915603)
Timeframe: every 8 weeks until progressive disease, expected average of 18 months

Interventionmonths (Median)
Paclitaxel/Bevacizumab/Everolimus9.1
Paclitaxel/Bevacizumab/Placebo7.1

[back to top]

Number of Phase I Participants With Dose-Limiting Toxicity Events (Phase I)

The Maximum Tolerated Dose (MTD) is defined as the dose level below the lowest dose that induces dose-limiting toxicity (DLT) in at least one-third of patients graded according to NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Dose-limiting toxicities include non-hematologic events graded 3 or higher and deemed at least possibly related to treatment. A total of 6 patients treated at the MTD will be sufficient to identify common toxicities at the MTD. The number of patients reporting a dose-limiting event are reported. (NCT00918333)
Timeframe: 4 weeks

InterventionParticipants (Count of Participants)
Phase I (Panobinostat + Everolimus)0

[back to top]

Overall Response Rate (Phase II)

For myeloma, a complete response(CR, defined as Negative immunofixation(IFE) of the serum and urine, < 5% plasma cells in bone marrow(BM), Disappearance of plasmacytomas), stringent CR(sCR, defined as CR plus Normal serum FLC ratio, Absence of clonal cells in BM), very good partial response(VGPR, defined as PR plus Serum and urine M-component detectable by IFE but not on electrophoresis), partial response(PR, defined as a ≥ 50% reduction of serum M-protein and/or reduction in 24-h urinary M-protein by ≥ 90% or to <200 mg per 24 h), or minor response(MR, defined as ≥25% but < 49% reduction of serum M-protein and reduction in 24h urine M-protein by 50-89%) noted as the objective status. For lymphoma, a CR(defined as no evidence of measurable disease), or PR(defined as regression of measurable disease and no new sites of disease) noted as the objective status. Percentage of successes will be estimated by 100 times the number of successes divided by the total number of evaluable patients. (NCT00918333)
Timeframe: Up to 12 courses

Interventionpercentage of patients (Number)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)39
Phase II (Myeloma Patients Receiving 20 mg LBH589)7
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)20
Phase II (Myeloma Patients Receiving 30/40 mg LBH589)0

[back to top]

Duration of Response (Phase II)

Duration of response is defined as the time from the date at which the objective status is first noted to be (for myeloma) a complete response (CR, defined as Negative immunofixation(IFE) of the serum and urine, < 5% plasma cells in bone marrow(BM), Disappearance of plasmacytomas), stringent CR(sCR, defined as CR plus Normal serum FLC ratio, Absence of clonal cells in BM), very good partial response(VGPR, defined as PR plus Serum and urine M-component detectable by IFE but not on electrophoresis), partial response(PR, defined as a ≥ 50% reduction of serum M-protein and/or reduction in 24-h urinary M-protein by ≥ 90% or to <200 mg per 24 h) or minor response(MR, defined as ≥25% but < 49% reduction of serum M-protein and reduction in 24h urine M-protein by 50-89%); (for lymphoma) a CR(defined as no evidence of measurable disease), or PR(defined as regression of measurable disease and no new sites of disease) noted as the objective status. Estimated using the method of Kaplan-Meier. (NCT00918333)
Timeframe: The time from the date at which the patient's objective status is first noted to be a CR, sCR, VGPR, PR, or minor response to the earliest date progression is documented, assessed up to 2 years post-treatment

Interventionmonths (Median)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)9.1
Phase II (Myeloma Patients Receiving 20 mg LBH589)NA
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)12.9

[back to top]

Progression-free Survival (Phase II)

Progression-free survival time is defined as the time from registration to progression or death due to any cause. Progression is defined for myeloma as Any one or more of the following: Increase of 25% from lowest value in, Serum M-component (absolute increase must be ≥ 0.5 g/dl), Serum M-component increase ≥ 1 g/dl, if lowest M component was ≥ 5 g/dl,Urine M-component (absolute increase must be ≥ 200 mg/24 h), Bone marrow plasma cell percentage (absolute % must be ≥10%) Or any one or more of the following felt related to the underlying clonal plasma cell proliferative disorder: Development of new soft tissue plasmacytomas or bone lesions, Hypercalcemia (≥11.5 mg/dl) Decrease in hemoglobin of ≥2 g/dl, Serum creatinine level ≥2 mg/dl. Progression is defined for lymphoma as any new lesion or increase by ≥50% of previously involved sites from nadir. The median and 95% confidence intervals are estimated using the method of Kaplan-Meier. (NCT00918333)
Timeframe: Time from registration to progression or death due to any cause, assessed up to 2 years post-treatment

Interventionmonths (Median)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)3.7
Phase II (Myeloma Patients Receiving 20 mg LBH589)2.3
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)4.2
Phase II (Myeloma Patients Receiving 30/40 mg LBH589)4.3

[back to top]

Overall Survival Time (Phase II)

Overall survival time is defined as the time from registration to death due to any cause. The median and 95% confidence intervals will be estimated using the method of Kaplan-Meier. (NCT00918333)
Timeframe: Time from registration to death due to any cause, assessed up to 2 years post-treatment

Interventionmonths (Median)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)17.1
Phase II (Myeloma Patients Receiving 20 mg LBH589)16.6
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)35.4
Phase II (Myeloma Patients Receiving 30/40 mg LBH589)21.7

[back to top]

Number of Patients With Pathological Complete Response

Pathological complete response is defined as no residual tumor on histopathological analysis of both breast and axillary contents. (NCT00930930)
Timeframe: at time of surgery, week 15-18

Interventionparticipants (Number)
Cisplatin and Paclitaxel + RAD00134
Cisplatin and Paclitaxel + Placebo17

[back to top]

Clinical Tumor Response to Neoadjuvant Therapy as Measured by Ultrasound Immediately Before Surgery

Per RECIST criteria v. 1.0: measurable lesions: complete response (CR) disappearance of target lesions, partial response (PR) > 30% decrease in the sum of the longest diameter (LD) of target lesions, progressive disease (PD) > 20% increase in the sum of the LD of target lesions or appearance of new lesions, stable disease (SD) neither sufficient decrease nor increase of the sum of smallest sum of the LD of target lesions (NCT00930930)
Timeframe: After treatment, week 12-15

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot Evaluable
Cisplatin and Paclitaxel + Placebo2319501
Cisplatin and Paclitaxel + RAD00148271821

[back to top]

Number of Patients With Each Worst-grade Toxicity Response

Tables represent the number of patients with their worst-grade toxicity at each of five grades (grade 1, least severe to grade 5, most severe) following NCI Common Toxicity Criteria. Not all participants necessarily have an adverse event, thus not everyone will be accounted for in worst-grade toxicities. Likewise, one participant can potentially have more than one event in various grades 1-5 which accounts for the difference in number of patients analyzed and total number in the worst-grade toxicity tables. (NCT00930930)
Timeframe: week 12

,
Interventionparticipants (Number)
Number of patients with worst-grade toxicity 1Number of patients with worst-grade toxicity 2Number of patients with worst-grade toxicity 3Number of patients with worst-grade toxicity 4Number of patients with worst-grade toxicity 5
Cisplatin and Paclitaxel + Placebo528910
Cisplatin and Paclitaxel + RAD0019403930

[back to top]

Number of Patients That Underwent Breast Conservation Surgery

Defined as patients that did not undergo complete removal of a cancerous breast (mastectomy). (NCT00930930)
Timeframe: at the time of surgery, week 15-18

Interventionparticipants (Number)
Cisplatin and Paclitaxel + RAD00138
Cisplatin and Paclitaxel + Placebo19

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of RAD001 in Combination of Weekly Abraxane and Determine the Phase II Dose of RAD001.

(NCT00934895)
Timeframe: 5 yrs

Interventionmg (Number)
All Participants5

[back to top]

Progression-free Survival

Progression-free survival time is defined as the time from registration to progression or death due to any cause. The distribution of progression-free survival will be estimated using the method of Kaplan-Meier (NCT00935792)
Timeframe: up to 5 years

InterventionMonths (Median)
All Evaluable Patients4.9

[back to top]

Clinical Response (Complete or Partial Remission)

CR requires all of the following for a period of at least 2months:Absence of lymphadenopathy.No hepatomegaly or splenomegaly.Absence of constitutional symptoms.• Neutrophils>1500/ul•Platelets>100,000/ul • Hemoglobin >11.0gm/dl• Peripheral blood lymphocytes <4000/uLBonemarrow. normocellular with<30%of nucleated cells being lymphocytes.PR requires two for 2+months.≥50%decrease in peripheral blood lymphocyte count from the pretreatment baseline value.≥ 50%reduction in the sum of the products of the maximal perpendicular diameters of the largest measured node or nodal masses in the right and left cervical, axillary, and inguinal lymph node regions.≥ 50%reduction in size of liver and/or spleen noting the maximal distance below the respective costal margins of palpable hepatosplenomegaly during rest.Neutrophils>1500/ul or50%improvement over baseline. Platelets>100,000/ul or50%increase over baseline. Hemoglobin>11.0 gm/dl or50%increase over baseline without transfusions (NCT00935792)
Timeframe: After 2 courses of treatment

,,
Interventionparticipants (Number)
Complete ResponsePartial Response
Phase 1, Dose Level 103
Phase 1, Dose Level 211
Phase 2, Dose Level 103

[back to top]

Survival Time

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier (NCT00935792)
Timeframe: up to 5 years

InterventionMonths (Median)
All Evaluable Patients29.5

[back to top]

Time to Subsequent Therapy

(NCT00935792)
Timeframe: up to 5 years

InterventionMonths (Median)
All Evaluable Patients13.9

[back to top]

Duration of Response

Duration of response is defined for all evaluable patients who have achieved a clinical response as the date at which the patient's objective status is first noted to be a Complete Response or Partial Response to the earliest date progression is documented. The distribution of duration of response will be estimated using the method of Kaplan-Meier (NCT00935792)
Timeframe: up to 5 years

InterventionMonths (Median)
All Evaluable Patients4.6

[back to top]

Test the Safety and Tolerability of the Combination of Everolimus and Alemtuzumab.

The number and severity of all adverse events will be tabulated and summarized in this patient population. The grade 3+ adverse events will also be described and summarized in a similar fashion. This will provide an indication of the level of tolerance for this treatment combination in this patient group. Below is the number of patients that experienced a grade 3+ Adverse event that was at least possibly related to Treatment. (NCT00935792)
Timeframe: Up to 12 months past final treatment

Interventionparticipants (Number)
All Evaluable Patients16

[back to top]

Number of Participants With Dose-Limiting Toxicities

"The maximum tolerated dose is the dose level below the lowest dose that induces dose-limiting toxicity in at least one-third of patients. A total of 6 patients treated at the MTD will be sufficient to identify common toxicities at the MTD. Dose-limiting toxicity will be defined as an adverse event attributed (definitely, probably, or possibly) to the study treatment and meeting the following criteria.~Hematologic: ANC ≤ 0.3 x 109/L or platelet count < 10 x 109/L Other nonhematologic: ≥grade 3 as per NCI Common Terminology Criteria for Adverse Events v3.0 except for fatigue, hyperlipidemia, and hyperglycemia." (NCT00935792)
Timeframe: 1 Month

Interventionparticipants with DLTs (Number)
Phase 1, Dose Level 11
Phase 1, Dose Level 22

[back to top]

Duration of Response

Duration of response was defined for all evaluable participants who have achieved an objective response as the date at which the participant's objective status is first noted to be either CR or PR to the date progression is documented. (NCT00936702)
Timeframe: Up to 3 years

Interventionmonths (Median)
Treatment (Carboplatin, Paclitaxel, and Everolimus)5.8

[back to top]

Overall Survival

Overall survival was defined as the time from study enrollment to the time of death from any cause or last follow-up. (NCT00936702)
Timeframe: Time from registration to death or last follow-up (up to 3 years)

Interventionmonths (Median)
Treatment (Carboplatin, Paclitaxel, and Everolimus)10.1

[back to top]

Percentage of Participants With Confirmed Tumor Responses

"Confirmed tumor response was defined to be either a complete response (CR) or partial response (PR) noted as the objective status on 2 consecutive evaluations at least 4 weeks apart.~Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria:~Complete Response (CR): disappearance of all target lesions;~Partial Response (PR) 30% decrease in sum of longest diameter of target lesions;" (NCT00936702)
Timeframe: First 6 Cycles of treatment (an average of 6 months)

Interventionpercentage of participants (Number)
Treatment (Carboplatin, Paclitaxel, and Everolimus)36

[back to top]

Progression-free Survival

The progression-free survival (PFS) was defined as the time from date of registration to the documentation of disease progression or death as a result of any cause, whichever comes first. (NCT00936702)
Timeframe: Time from registration to the disease progression or death (up to 3 years)

Interventionmonths (Median)
Treatment (Carboplatin, Paclitaxel, and Everolimus)4.1

[back to top]

Time to Treatment Failure

Time to treatment failure was defined to be the time from the date of registration to the date at which the participant is removed from treatment due to progression, adverse events, or refusal. (NCT00936702)
Timeframe: Up to 3 years

Interventionmonths (Median)
Treatment (Carboplatin, Paclitaxel, and Everolimus)3.1

[back to top]

Median Overall Survival

Overall Survival (OS) is defined as the time from randomization (or registration) to death due to any cause, or censored at date last known alive. (NCT00936858)
Timeframe: Every 2 months for first 24 months, then every 3 months from >24 to 60 months; up to 5 years post study registration.

Interventionmonths (Median)
Arm A32.7

[back to top]

Median Progression Free Survival

Progression free survival (PFS) is defined as the time from start of treatment to disease progression or death from any cause as estimated by Kaplan Meier methods. Progression is measured using RECIST 1.1 criteria, defined as at least a 20% increase in size in target lesion and/or unequivocal progression of non-target lesions and/or appearance of new lesions. Patients who have not progressed and are alive are censored at the date the patient is known to be progression-free. (NCT00936858)
Timeframe: Every 2 months for first 24 months, then every 3 months from >24 to 60 months; up to 48 months.

Interventionmonths (Median)
Arm A12.5

[back to top]

Objective Response Rate

"The objective response rate is the proportion of participants achieving complete response (CR) or partial response (PR) on treatment based on Response Evaluation Criteria In Solid Tumors Criteria (RECIST 1.1) criteria.~PR or better is achieved if the following are true:~Target Lesions:~-At least a 30% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm.~Non-target Lesions:~No progression. No appearance new lesions or unequivocal progression of existing non-target lesions. Unequivocal progression should not normally trump target lesion status. It must be representative of overall disease status change, not a single lesion increase.~Bone Lesions:~->50% increase in lesions.~-No new lesions." (NCT00936858)
Timeframe: Every 2 months for first 24 months, then every 3 months from >24 to 60 months; up to 48 months.

Interventionpercentage of participants (Number)
Arm A6

[back to top]

Mean Change in Quality of Life [Medullary Thyroid Cancer Population Only]

"The M. D. Anderson Symptom Inventory (MDASI) questionnaire was used to assess quality of life. Questions 1 to 19 were scored. Each of the 19 questions have a response range of 0 - 10, where 0 represents not present and 10 represents as bad as you can imagine. The 19 responses are averaged together to create a mean score, with a lower score indicating a better quality of life. All questionnaire mean scores at each timepoint are averaged together to give a mean score at that timepoint (baseline and week 8) The mean change in quality of life is calculated by subtracting the baseline mean score from the week 8 mean score." (NCT00936858)
Timeframe: Measured at baseline and then again at cycle 8 (8 months).

Interventionmean change in score on the MDASI scale (Mean)
Arm A-0.0621

[back to top]

12-Week Progression-Free Survival (PFS)

Tumor assessments performed by Computed Tomography (CT) scan or Magnetic Resonance Imaging (MRI) after 4 weeks, 12 weeks, then every 8 weeks thereafter. Participants who received at least one dose of RAD001+erlotinib and who die before 12 weeks, counted as having progressive disease. Response Evaluation Criteria in Solid Tumors (RECIST) defined as Complete Response (CR): Disappearance of all target lesions; Partial Response (PR): >30% decrease in sum of longest diameter (LD) of target lesions, taking as reference baseline sum LD. Progression-free survival defined as stable disease or better. Progressive Disease (PD): >20% increase in sum of LD of target lesions, taking as reference smallest sum LD recorded since treatment started or appearance of one or more new lesions; Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference smallest sum LD since treatment started. (NCT00942734)
Timeframe: 12 weeks

InterventionPercentage of Participants (Number)
RAD001 + Erlotinib48.57

[back to top]

Part 1: Number of Participants Who Experienced One or More Adverse Events (AEs)

An AE is any unfavorable and unintended sign, symptom, or disease temporally associated with the use of study drug, whether or not considered related to this study drug. AEs may include the onset of new illness and the exacerbation of pre-existing conditions. (NCT00954512)
Timeframe: Up to ~30 days after the final dose of robatumumab (Up to ~14 months)

InterventionParticipants (Number)
Regimen A: FOLFIRI (± Cetuximab) + Robatumumab2
Regimen B: Carboplatin + Paclitaxel + Robatumumab3
Regimen D: Trastuzumab + Robatumumab2
Regimen E: mTor Inhibitor (Everolimus) + Robatumumab4
Regimen F: Gemcitabine (± Erlotinib) + Robatumumab4

[back to top]

Part 2: Number of Participants With Each Type of Response Evaluation Criteria in Solid Tumors (RECIST)-Determined Overall Best Response

Overall best response was determined by RECIST criteria. Types of overall response could be: Complete Response (CR), Partial Response (PR), Stable Disease (SD), Progressive Disease (PD), Not Assessable (NA) or Incomplete Response/Stable Disease (IR/SD). (NCT00954512)
Timeframe: Up to ~30 days after the final dose of robatumumab (Up to ~14 months)

,,,,
InterventionParticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Not Assessable (NA)Incomplete Response/Stable Disease (IR/SD)
Regimen A: FOLFIRI (± Cetuximab) + Robatumumab000200
Regimen B: Carboplatin + Paclitaxel + Robatumumab002100
Regimen D: Trastuzumab + Robatumumab001100
Regimen E: mTor Inhibitor (Everolimus) + Robatumumab002100
Regimen F: Gemcitabine (± Erlotinib) + Robatumumab001100

[back to top]

Number of Participants Who Experienced Adverse Events, Serious Adverse Events and Death

Participants with adverse events (serious plus non-serious), serious adverse events and death were reported. (NCT00956293)
Timeframe: Months 6, 12, 24, 36, 48 and 60

,
InterventionParticipants (Number)
Adverse events (serious and non-serious)Serious adverse eventsDeaths
Control Group21110
Everolimus Group50280

[back to top]

Change in Renal Function (Creatinine Slope)

X(slope)=(1/value of creatinine). (NCT00965094)
Timeframe: 3 months, 5 months, 7 months, 9 months

,
Interventionmg/dl per month (Mean)
(ITT) Baseline 2: Month 3 N=15, 15(ITT) Month 3 (1st week) N=15, 4(ITT) Month 3 (2nd week) N=15, 4(ITT) Month 3 (3rd week) N=13, 2(ITT) Month 3 (4th week) N=12, 0*(ITT) Month 5 N=14, 15(ITT) Month 7 N=14, 14(ITT) Month 9 N=11, 15(PP) Baseline 2: Month 3 N=11, 15(PP) Month 3 (first week) N=11, 4(PP) Month 3 (second week) N=11, 4(PP) Month 3 (third week) N=10, 2(PP) Month 3 (fourth week) N=9, 0*(PP) Month 5 N=11, 15(PP) Month 7 N=11, 14(PP) Month 9 N=11, 15
Everolimus0.70.70.80.70.80.80.70.80.70.70.80.70.80.80.80.8
Reference Therapy0.70.80.90.9NA0.70.70.80.70.80.90.9NA0.70.70.8

[back to top]

Participants Who Had Occurrence of Biopsy Proven Acute Rejection, Graft Loss or Death.

Biopsy-proven acute rejection was defined as a biopsy gradeed IA, IB, IIA, IIB, or III. The allograft was presumed to be lost if the patient started dialysis and was not able to subsequently be removed from dialysis. If the patient underwent a graft nephrectomy, then the day of nephrectomy was the day of graft loss. (NCT00965094)
Timeframe: 9 months

,
InterventionParticipants (Number)
NoYes
Everolimus141
Reference Therapy141

[back to top]

Participants Who Had Occurrence of Treatment Failure.

Treatment failure was defined as a composite endpoint of biopsy-proven acute rejection, graft loss, death, loss to follow up, discontinuation due to lack of efficacy or toxicity or conversion to another regimen. (NCT00965094)
Timeframe: 9 months

,
InterventionParticipants (Number)
NoYes
Everolimus105
Reference Therapy141

[back to top]

Assessment of GFR by the Cockcroft-Gault Method (LOCF)

the GFR was also calculated using the Cockcroft-Gault method (Cockcroft and Gault 1976) and the Modification of Diet in Renal Disease (MDRD) method (Levey et al., 1999, Rodrigo et al., 2003; Pierrat et al., 2003).Cockcroft-Gault formula For men: GFR= (140-Age)X Body Weight[kg]/72X Serum Creatinine[mg/dl] For women: GFR= 0,85x(140-Age) x Body Weight[kg]/72x Serum Creatinine [mg/dl] The Last Observation Carried Forward (LOCF) imputation technique was used for this analysis. (NCT00965094)
Timeframe: 9 months

InterventionmL/min (Mean)
Everolimus72.6
Reference Therapy72.7

[back to top]

Renal Function Assessed as Glomerula Filtration Rate (GFR) - Nankivell Method - 9 Months After Renal Transplantation (LOCF)

The glomerular filtration rate (GFR) is the best clinical estimate of renal function in health and disease, and correlates well with the clinical severity of renal function disturbances. The GFR, calculated according to the Nankivell formula, was used as the primary outcome measure in this study. This equation has been validated in renal transplant patients against the true GFR measured by a radionuclide method and has been confirmed as a very accurate method to calculate the GFR in this specific population (Gaspari et al., 2004)GFR = 6.7 / Scr + BW / 4 - Surea / 2-100 / (height)² + C Scr = serum creatinine concentration expressed in mmol/L. BW = body weight in kg, Surea = serum urea in mmol/L and Height is expressed in meters.The Last Observation Carried Forward (LOCF) imputation technique was used for this analysis. (NCT00965094)
Timeframe: 9 months

InterventionmL/min/1.73m^2 (Mean)
Everolimus15.0
Reference Therapy16.6

[back to top]

Maximum Tolerated Dose (MTD) of Everolimus With Panobinostat

MTD of the novel combination of Everolimus + Panobinostat (LBH589) in a phase-I study in participants with relapsed lymphoma (Hodgkin and non-Hodgkin) where MTD is defined as the highest dose at which no more than 1 in 6 of the participants in the cohort experiences one or more dose limiting toxicities (DLTs) in the first 28 day treatment cycle. Thirty patients were enrolled onto four dose levels: Everolimus (mg, orally) 5, 5, 10, 10 daily or Panobinostat (mg, orally) 10, 20, 20, 30 three times per week. The MTD was established without the use of colony stimulating factor in cycle 1. (NCT00967044)
Timeframe: 28 day treatment cycle

Interventionmg, orally (Number)
Everolimus (daily)Panobinostat (three times weekly)
Panobinostat + Everolimus1020

[back to top]

Overall Response Rate (OR) Where OR = CR + CRp + CRi

Number of participants out of total treated who experienced a complete response response according to RECIST criteria either (CR + CRp) CR Without Platelet Recovery. Response (CR + CRp) defined as Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x10^9/L, platelet count > 100 x10^9/L, and blasts < 5% in a normocellular or hypercellular marrow. Complete remission without platelet recovery (CRp): Peripheral blood and marrow parameters as for CR, but with platelet count > 20 x 10^9/L and < 100 x 10^9/L in the absence of platelet transfusions. CR with incomplete blood count recovery (CRi): Same as CR but platelets ≤ 100,000/mcl and/or neutrophils ≤ 1,000/mcl. (NCT00968253)
Timeframe: 8 courses of treatment, up to 24 weeks

Interventionpercentage of participants (Number)
Phase I: RAD001 5 mg + Combination Chemo33
Phase I: RAD001 10 mg + Combination Chemo33
Phase II: MTD RAD001 + Combination Chemo33

[back to top]

Participant Responses by Daily Dose Level Assignment (RAD001 5 mg, 10 mg and MTD 5 mg)

Response defined as Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x10^9/L, platelet count > 100 x10^9/L, and blasts < 5% in a normocellular or hypercellular marrow. Complete remission without platelet recovery (CRp): Peripheral blood and marrow parameters as for CR, but with platelet count > 20 x 10^9/L and < 100 x 10^9/L in the absence of platelet transfusions. CR with incomplete blood count recovery (CRi): Same as CR but platelets ≤ 100,000/mcl and/or neutrophils ≤ 1,000/mcl. Partial remission (PR): Peripheral blood count recovery as for CR, with decrease in marrow blasts by > 50% from pretreatment values with no more than 25% leukemia/lymphoma cells in the marrow. Nonresponder, Other: All other responses will be considered failures. (NCT00968253)
Timeframe: Up to 20 cycles of study drugs (21 day cycles) or till disease progression

,,
Interventionparticipants (Number)
Complete RemissionComplete Remission without platelet recoveryCR with incomplete blood count recoveryPartial RemissionNonresponder
Phase I: RAD001 10 mg + Combination Chemo20115
Phase I: RAD001 5 mg + Combination Chemo10011
Phase II: MTD RAD001 + Combination Chemo31008

[back to top]

Maximum Tolerated Dose [MTD] Determination by Number of Participants With Dose Limiting Toxicity (DLT)

"The Maximum tolerated dose (MTD) was the highest dose level at which fewer than 2 of 6 patients developed a dose limiting toxicity (DLT) in the first two cycles of therapy. A 3 by 3 design was used for dose escalation in the phase I portion of the study.~A dose-limiting toxic effect (DLT) was defined as a clinically significant adverse event or abnormal laboratory value directly attributable to everolimus and assessed as unrelated to disease progression, intercurrent illness, or concomitant medications, occurring during the first or second cycle of therapy, that met any of the following criteria: CTCAE version 3.0 grade 3 increased AST or ALT for 7 days, CTCAE grade 4 increased AST or ALT of any duration, or any other clinically significant CTCAE grade 3 or 4 toxic effect. Electrolyte abnormalities (changes in glucose, chemistries, liver enzymes, pancreatic enzymes) correctable by optimal therapy and without clinical impact were not considered DLTs." (NCT00968253)
Timeframe: Following first two dose cycles (21 days/each), up to 42 days

InterventionParticipants (Count of Participants)
Phase I: RAD001 5 mg + Combination Chemo0
Phase I: RAD001 10 mg + Combination Chemo1

[back to top]

Progression-free Survival (PFS), in the Treatment of Patients With Refractory Meningioma.

Progression-free survival (PFS) is defined as the time from randomization until objective tumor progression (PD) or death. Progression is defined per MacDonald criteria for response as ≥25% increase in size of enhancing tumor or any new tumor on MRI scan, neurologically worse, and steroids stable or increased. (NCT00972335)
Timeframe: 18 months

Interventionmonths (Median)
Combination Therapy22

[back to top]

To Evaluate the Toxicity of Bevacizumab/Everolimus in Patients With Recurrent Meningioma.

(NCT00972335)
Timeframe: 18 months

Interventionparticipants (Number)
ThrombocytopeniaAnemiaLeukopeniaNeutropeniaHypercholesterolemiaMucositisFatigueProteinuriaHypertriglyceridemiaRash/desquamationDiarrheaHypertensionEdema-limbVomitingAnorexiaNauseaHyperglycemiaPain - oral cavityEpistaxisHeadacheArthralgia
Combination Therapy962299866665544433333

[back to top] [back to top]

Overall Response Rate of RAD001 in Patients With Previously Untreated WM

"Overall Response = Complete Response + Near Complete Response + Very Good Partial Response + Partial Response + Minor Response Complete Response: resolution of all symptoms, normalization of serum IgM levels with complete disappearance of IgM paraprotein by immunofixation, and resolution of any adenopathy or splenomegaly. A near CR (nCR) is defined as fulfilling all CR criteria in the presence of positive immunofixation test for an IgM paraprotein.~Very Good Partial Response: > 90% reduction in serum IgM levels. Partial Response: > 50% reduction in serum IgM levels. Minor Response: 25-49% reduction in serum IgM levels Progressive Disease: greater than 25% increase in serum IgM level occurs from the lowest attained response value or progression of clinically significant disease related symptom(s).~Stable Disease: < 25% change in serum IgM levels, in the absence of new or increasing adenopathy or splenomegaly and/or other progressive signs or symptoms of WM" (NCT00976248)
Timeframe: End of Treatment, an average of 16 months

Interventionparticipants (Number)
RAD00122

[back to top]

Time to Progression With Single Agent RAD001 Therapy in Previously Untreated WM.

Progression is defined as a 25% increase in serum IgM from the lowest attained response value or progression of clinically significant disease related symptoms. (NCT00976248)
Timeframe: End of Treatment, an average of 16 months

InterventionMonths (Median)
RAD00121

[back to top]

Confirmed Tumor Response Rate (Complete Response [CR] or Partial Response [PR]) According to Response Evaluation Criteria in Solid Tumors (RECIST) Criteria

Confirmed Tumor Response: A confirmed tumor response is defined to be a CR or PR (by the RECIST criteria) noted as the objective status on 2 consecutive evaluations at least 8 weeks apart. The proportion of tumor responses will be estimated by the number of confirmed tumor responses divided by the total number of evaluable patients. A ninety percent confidence interval for the true proportion of confirmed tumor responses will be calculated assuming that the number of confirmed tumor responses follows a binomial distribution. (NCT00976573)
Timeframe: Up to 5 years

Interventionpercentage of patients (Number)
Arm A (Bevacizumab, Paclitaxel, and Carboplatin)13
Arm B (Bevacizumab, Paclitaxel, Carboplatin, and Everolimus)23

[back to top]

Progression-free Survival

The primary endpoint is progression-free survival (PFS) defined as the time from randomization to documentation of disease progression or death without documentation of progression. The distribution of PFS times will be estimated using the Kaplan-Meier method. Progression is defined using the RECIST Criteria as at least a 20% increase in the sum of diameters of target lesions taking as reference the smallest sum of diameters recorded on study (this includes the baseline sum if that is the smallest on study) or the appearance of one or more new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm of target lesions, or the appearance of one or more new lesions, unequivocal progression of existing non-target lesions, although unequivocal progression should not normally trump target lesion status. It must be representative of overall disease status change, not a single lesion increase. (NCT00976573)
Timeframe: Time from randomization to documentation of disease progression or death without documentation of progression;Up to 5 years

Interventionmonths (Median)
Arm A (Bevacizumab, Paclitaxel, and Carboplatin)5.6
Arm B (Bevacizumab, Paclitaxel, Carboplatin, and Everolimus)5.1

[back to top]

Overall Survival Time

Overall survival time is defined as the time from registration to death due to any cause. The distribution of survival times will be estimated using the method of Kaplan-Meier. (NCT00976573)
Timeframe: up to 5 years

Interventionmonths (Median)
Arm A (Bevacizumab, Paclitaxel, and Carboplatin)14.5
Arm B (Bevacizumab, Paclitaxel, Carboplatin, and Everolimus)10.8

[back to top]

Toxicity

For this secondary endpoint, toxicity is defined as a grade 3 or higher adverse events that is classified as either possibly, probably, or definitely related to study treatment. The assignment of attribution to study treatment and grade (or degree of severity) of the adverse event are classified using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. The percentage of participants reporting a grade 3 or higher toxicity is reported. For a list of all reported adverse events, please refer to the Adverse Events Section below. (NCT00976573)
Timeframe: Up to 5 years

,
Interventionpercentage of participants (Number)
NeutropeniaLeukopeniaFatigue
Arm A (Bevacizumab, Paclitaxel, and Carboplatin)351711
Arm B (Bevacizumab, Paclitaxel, Carboplatin, and Everolimus)582417

[back to top]

Overall Response Rate

Overall Response Rate is the number of participants with a partial and complete response assessed by the Updated Cheson criteria for lymphoma. A complete response is complete disappearance of all detectable clinical and radiographic evidence of disease and disappearance of all disease-related symptoms if present before therapy and normalization of those biochemical abnormalities. If a subject has residual lesions on CT scan and the disease was fluorodeoxyglucose (FDG) avid pre-treatment, then that subject will be considered to be in a complete response. Partial response is a greater than or equal to 50% decrease in the sum of the products of the greatest diameters of 6 largest dominant nodes or nodal masses. No increase in size of nodes, liver or spleen and no new sites of disease (NCT00978432)
Timeframe: after 2 cycles of each study drug or after 2 cycles of doublet, up to 24 weeks

Interventionparticipants (Number)
Arm 1a (RAD001 Followed by LBH589)1
Arm 1b (LBH589 Followed by RAD001)0
Doublet (Combination RAD001 and LBH589)4

[back to top]

Summary of Adverse Events (AEs)

Counts of adverse events or treatment-emergent adverse events (TEAE, defined as newly occurring or worsening after first dose) experienced by patients on study drug(s). National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE version 3.0) is used to assess severity. Relatedness to study drug is assessed by the investigator. (NCT00978432)
Timeframe: From the time of first dose of study drug until 4 weeks after participant has stopped study drug; up to 1 year

,,
Interventionevents (Number)
Blood and lymphatic AEsGastrointestinal AEsGeneral Disorder AEsInvestigation (Lab) AEsMetabolism and Nutrition AEsMusculoskeletal and Connective Tissue AEsNervous System AEsRenal and Urinary AEsReproductive System AEsRespiratory, thoracic and mediastinal AEsSkin and Subcutaneous Tissue AEsVascular AEs
Arm 1a (RAD001 Followed by LBH589)1830665413001082
Arm 1b (LBH589 Followed by RAD001)6105321311421
Doublet (Combination RAD001 and LBH589)423919234525411531

[back to top]

Biomarker Correlations: Time to Progression

Potential correlations between time to progression and S6 protein and mTOR-related proteins in tumor tissue samples from these patients. (NCT00985192)
Timeframe: 30 months

Interventionmonths (Median)
p-S6 level 0-2p-S6 level >2p-mTOR level 0-2p-mTOR level >2
Everolimus1.753.41.82.6

[back to top]

Efficacy in Terms of Progression Free Response

Progression-free survival (PFS), was defined as the time from the date of initial treatment to first objective documentation of disease progression, or death. Estimated using the Kaplan-Meier method. Complete response (CR) + partial response (PR) + stable disease (SD) were determined according to Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Radiologic disease assessments were utilized. (NCT00985192)
Timeframe: evry 3 months in year 1, every 6 months after that

Interventionmonths (Median)
Everolimus1.8

[back to top]

Overall Disease-control Rate in Patients With Previously Treated Unresectable or Metastatic Adenocarcinoma of the Upper Gastrointestinal Tract Treated With Everolimus.

Disease control rate (DCR), defined as complete response (CR) + partial response (PR) + stable disease (SD) according to Response Evaluation Criteria In Solid Tumors (RECIST) criteria. (NCT00985192)
Timeframe: Radiologic disease assessment was performed every 8 weeks (14 days = 1 cycle) treatment discontinuation.

Interventionpercent subjects with disease control (Number)
Everolimus40

[back to top]

Overall Survival

Overall Survival (OS), defined as the time from date of initial treatment to date of death. Survival function was estimated using the Kaplan-Meier method. (NCT00985192)
Timeframe: 2.5 year

Interventionmonths (Median)
Everolimus3.4

[back to top]

Biomarker Correlations: Progression Free Survival

Potential correlations between progression free survival and S6 protein and mTOR-related proteins in tumor tissue samples from these patients. (NCT00985192)
Timeframe: 30 months

Interventionmonths (Median)
p-S6 level 0-2p-S6 level >2p-mTOR level 0-2p-mTOR level >2
Everolimus1.83.51.82.6

[back to top]

Observed Biomarkers

Potential correlations between clinical outcome and biomarkers of interest, including S6 protein overexpression and/or other mTOR-related proteins in tumor tissue samples from these patients. (NCT00985192)
Timeframe: 30 months

Interventionnumber of tissue blocks (Number)
Tumor Grade 0, p-S6Tumor Grade 1+, p-S6Tumor Grade 2+, p-S6Tumor Grade 3+, p-S6Tumor Grade 4+, p-S6Tumor Grade 0, p-mTORTumor Grade 1+, p-mTORTumor Grade 2+, p-mTORTumor Grade 3+, p-mTORTumor Grade 4+, p-mTOR
Everolimus415893082290

[back to top]

Vinorelbine Blood Concentrations by Leading Dose and Time Point

Pre-infusion (Cmin) and end of infusion (C2h) vinorelbine PK blood samples were collected at Cycle 2 Day 1. Only valid vinorelbine PK blood samples collected at steady state were used in the analyses. (NCT01007942)
Timeframe: Cycle 2, Day 1

,
Interventionng/ml (Mean)
Pre-infusion - dose (Cmin) (n: 76, 64)End of infusion (Cmax) (n: 58, 49)
Everolimus11.085867.147
Everolimus Placebo0.0611068.51

[back to top]

Trastuzumab Blood Concentrations by Leading Dose and Time Point

Pre-infusion (Cmin) and end of infusion (C2h) trastuzumab PK blood samples were collected at Cycle 3 Day 1. Only valid trastuzumab PK blood samples collected at steady state were used in the analyses. (NCT01007942)
Timeframe: Cycle 3, Day 1

,
Interventionng/ml (Mean)
Pre-infusion - dose (Cmin) (n: 73, 57)End of infusion (Cmax) (n: 75, 59)
Everolimus23.35164.279
Everolimus Placebo24.52660.576

[back to top]

Progressive-free Survival (PFS) Per Investigator Assessment

PFS was defined as the time from the date of randomization to the date of first radiologically documented tumor progression or death from any cause, whichever occurs first. PFS primary analysis performed when 415 events were reached. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01007942)
Timeframe: Every 6 weeks until disease progression or death which ever occurred first up to about 41 months

Interventionmonths (Median)
Everolimus + Vinorelbine + Trastuzumab7.00
Placebo + Vinorelbine + Trastuzumab5.78

[back to top]

Overall Survival (OS)

OS was defined as the time from date of randomization to the date of death from any cause. Final OS was conducted when 388 deaths occurred. (NCT01007942)
Timeframe: Every 3 months until death up to 41 months

Interventionmonths (Median)
Everolimus + Vinorelbine + Trastuzumab23.46
Placebo + Vinorelbine + Trastuzumab24.08

[back to top]

Overall Response Rate (ORR)

ORR was defined as the percentage of participants whose best overall response was either complete response (CR) or partial response (PR) according to RECIST version 1.0. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT01007942)
Timeframe: Every 6 weeks until disease progression or death which ever occurred first up to about 41 months

InterventionPercentage of participants (Number)
Everolimus + Vinorelbine + Trastuzumab40.8
Placebo + Vinorelbine + Trastuzumab37.2

[back to top]

Median Time to Deterioration of the ECOG Performance Status Score

Time to deterioration of ECOG performance status score was summarized at time of assessment. ECOG (Eastern Cooperative Oncology Group)performance scale is a standard criteria for measuring how treatment of cancer impacts their level of functioning in terms of their ability to care for themselves, daily activity, & physical ability (walking, working, etc.). Scale score ranges from 0 to 5, 5 being the worst. ECOG scale index: 0 - Fully active, able to carry on all pre-disease performance without restriction. 1 - Restricted in physically strenuous activity but ambulatory & able to carry out work of a light or sedentary nature, e.g., light housework, office work. 2 - Ambulatory & capable of all self-care but unable to carry out any work activities. Up & about more than 50% of waking hours. 3 - Capable of only limited self-care, confined to bed or chair more than 50% of waking hours. 4 - Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair. 5 - Dead (NCT01007942)
Timeframe: baseline, until disease progression or death up to about 41 months

Interventionmonths (Median)
Everolimus + Vinorelbine + Trastuzumab32.66
Placebo + Vinorelbine + Trastuzumab21.55

[back to top]

Clinical Benefit Rate (CBR)

CBR was defined as the percentage of participants whose best overall response, according to RECIST, was either complete response (CR), a partial response (PR) or stable disease (SD) lasting for at least 24 weeks. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; SD = Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for PD; PD = At least a 20% increase in the sum of the longest diameter of all measured target lesions, taking as reference the smallest sum of longest diameter of all target lesions recorded at or after baseline. (NCT01007942)
Timeframe: Every 6 weeks until disease progression or death which ever occurred first up to about 41 months

InterventionPercentage of participants (Number)
Everolimus + Vinorelbine + Trastuzumab59.2
Placebo + Vinorelbine + Trastuzumab53.3

[back to top]

Everolimus Blood Concentrations by Leading Dose and Time Point

Pre-dose (Cmin) and 2 hours post-dose (C2h) everolimus PK blood samples were collected at Cycle 2 Day 1. Only valid everolimus PK blood samples collected at steady state were used in the analyses. (NCT01007942)
Timeframe: Cycle 2, Day 1

,
Interventionng/ml (Mean)
Pre-dose (Cmin) (n: 7, 32)2 hours post administration (C2h) (n:10, 43)
Everolimus5.65222.005
Everolimus 2.5 mg2.92813.035

[back to top]

PRO: Time to Deterioration in Global Health Status/QoL Domain Score of the European Organization for the Research and Treatment of Cancer (EORTC)-Core Quality of Life Questionnaire (QLQ-C30) (by at Least 10%)

PRO = patient reported outcomes; Time to deterioration (≥ 10% worsening from baseline), in the global health status of EORTC QLQ-C30 scale was done in the 3 functional scales (emotional, physical, & social functioning [EF, PF, & SF]). It contains 30 items & is composed of multi-item scales & single-item measures. These include 5 functional scales (physical, role, emotional, social & cognitive functioning), 3 symptom scales (fatigue, pain, nausea, & vomiting), a global health status/QoL scale, and 6 single items (dyspnea, diarrhea, constipation, anorexia, insomnia & financial impact). Each of the multi-item scale includes a different set of items - no item occurs in more than 1 scale. Each item in the EORTC QLQ-C30 has 4 response categories (1=Not at all, 2= A little, 3= Quite a bit, 4= Very much) with the higher number representing a worse outcome. The global health domain score of the QLQ-C30 questionnaire was pre-specified as the primary QoL domain of interest & disclosed here. (NCT01007942)
Timeframe: Baseline, until disease progression or death up to about 41 months

,
Interventionmonths (Median)
Deterioration - global QoL domain by at least 10%Deterioration in the PF domain by at least 10%Deterioration in the EF domain by at least 10%Deterioration in the SF domain by at least 10%
Everolimus + Vinorelbine + Trastuzumab8.3111.9615.1811.33
Placebo + Vinorelbine + Trastuzumab7.2912.4812.4513.11

[back to top]

Progression Free Survival (PFS) of RAD001 in Combination With Weekly Cetuximab and Cisplatin.

Median number of months for which participants are free of progression after initiating treatment with RAD001 in combination with weekly cetuximab and cisplatin. (NCT01009346)
Timeframe: 2 years

Interventionmonths (Median)
Study Arm (RAD001)2.8

[back to top]

Maximum Tolerated Dose (MTD) of RAD001 in Combination With Cetuximab and Cisplatin.

MTD will be defined as a) the dose of RAD001 producing DLT in 0-1 out of 6 patients, or b) the dose level below the dose which produced DLT in <2 out of 6 patients, or c) the dose of 10mg po qd with less than 33% rate of DLT (NCT01009346)
Timeframe: 6 months

Interventionmg (Number)
Study Arm (RAD001, Cetuximab, Cisplatin or Carboplatin)10

[back to top]

Overall Survival (OS)

Overall survival (OS) is defined as the time from randomization until death from any cause. (NCT01014351)
Timeframe: 18 months

InterventionMonths (Median)
Paclitaxel/Carboplatin/Everolimus10.12

[back to top]

Objective Response Rate (ORR)

Objective Response Rate (ORR) is defined as the Percentage of Patients Who Experience an Objective Benefit From Treatment. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI or CT: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT01014351)
Timeframe: 18 months

Interventionpercentage of patients (Number)
Paclitaxel/Carboplatin/Everolimus17

[back to top]

Progression-free Survival (PFS)

Progression-free survival (PFS) is defined as the time from randomization until objective tumor progression (PD) or death. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01014351)
Timeframe: 18 months

InterventionMonths (Median)
Paclitaxel/Carboplatin/Everolimus4.04

[back to top]

Absolute and Percent Frequencies of Patients With LDL ≥ 100 mg/mL at 1, 3 and 6 Months, by Treatment Group

LDL = low density lipoprotein (NCT01017029)
Timeframe: 6 months

,
Interventionparticipants (Number)
Month 1Month 3Month 6
Delayed Introduction of Everolimus383736
Immediate Introduction of Everolimus413734

[back to top]

Participants With at Least One Occurrence of Safety Composite Endpoint After 6 Months by Treatment Group

Comparison of 6-month cumulative incidence of safety composite endpoint (wound healing delay) related to initial transplant surgery, pleural/pericardial effusions and occurrence of acute renal insufficiency, defined as estimated glomerular filtration rate (eGFR) ≤ 30 mL/min/1.73 m2, between delayed everolimus arm and immediate everolimus arm (NCT01017029)
Timeframe: 6 months

Interventionparticipants (Number)
Immediate Introduction of Everolimus40
Delayed Introduction of Everolimus30

[back to top]

Participants With at Least One Occurrence of Composite Treatment Failure Events

Comparison of 6-months cumulative incidence of composite treatment failure events (BPAR ≥ 2R, rejection with hemodynamic compromise, graft loss, or death) between delayed everolimus arm and immediate everolimus arm (NCT01017029)
Timeframe: 6 months

Interventionparticipants (Number)
Immediate Introduction of Everolimus33
Delayed Introduction of Everolimus26

[back to top]

Hazard Cox's Model Analysis of Pericardial/Pleural Effusions

Pericardial effusions: any pericardial effusion defined as at least moderate (i.e. measuring at least 2.0 cm in diastole, in the point of largest distance between the pericardial leaflets), with or without signs of hemodynamic compromise, or leading to drainage or to prolonged hospitalization. Pleural effusions: need for surgical drainage tubes for longer than 7 days after surgery and subsequent pleural effusions leading to drainage. CI = confidence interval, HR = hazard ratio, MDRD = Modification of Diet in Renal Disease (NCT01017029)
Timeframe: 6 months

Interventionparticipants (Number)
Immediate Introduction of Everolimus30
Delayed Introduction of Everolimus18

[back to top]

Partcipants With at Least One Occurrence of Each Safety Composite Endpoint Event After 6 Months by Treatment Group

(NCT01017029)
Timeframe: 6 months

,
Interventionparticipants (Number)
Wound healing complicationPleural effusionPericardial effusioneGFR ≤ 30 mL/min/1.73 m2
Delayed Introduction of Everolimus81188
Immediate Introduction of Everolimus101307

[back to top]

Participants With CMV Infection and CMV Syndrome/Disease After 6 Months by Treatment Group

CMV infection is defined as pp65 antigenemia or DNAemia (NCT01017029)
Timeframe: 6 months

,
Interventionparticipants (Number)
CMV infectionsCMV syndrome/disease
Delayed Introduction of Everolimus636
Immediate Introduction of Everolimus463

[back to top]

Overall Response Rate (ORR) Based on the Assessments by Investigator

ORR: % of patients whose overall disease response was a complete response (CR) or a partial response (PR) in 8 cycles CR: Complete normalization of all index nodal & extranodal lesions: Radiological regression to normal size of all lymph nodes & nodal masses & complete disappearance of all lesions PR: At least a 50% decrease in the SPD of all index nodal & extranodal lesions FDG-avid or PET positive prior to therapy: one or more PET positive at previously involved site.At least a 50% increase in the SPD of all index nodal & extranodal lesions, taking as reference the smallest sum of the product of the diameters of all index lesions recorded at or after baseline . Lesions PET positive if FDG-avid lymphoma or PET positive prior to therapy. Unknown (UNK): Progression not documented & one or more of the index lesions not assessed or assessed using a different method than baseline at the time of radiologic evaluation. Each cycle was 28 days. (NCT01022996)
Timeframe: at screening and every threee months beginning at cycle 3 until end of treatment due to progression of disease, unacceptable toxicity, death or discontinuation from the study for any other reason

Interventionpercentage of participants (Number)
Complete response (CR)Partial response (PR)Response of CR or PR
RAD0018.836.845.6

[back to top]

Disease Control Rate (DCR)

The disease control rate was defined as the percentage of patients with a best overall response of CR, PR or stable disease (SD). (NCT01022996)
Timeframe: Every three months beginning at Cycle 3 until end of treatment due to progression of disease, unacceptable toxicity, death or discontinuation from the study for any other reason

InterventionPercentage of participants (Number)
RAD00180.7

[back to top]

Duration of Disease Control

The duration of overall response (CR/PR) was applied only to patients whose best overall response was CR or PR. Duration of overall response was calculated from the date of the first documented response of CR or PR to the date of first documented disease progression or death due to any cause or start of a new antineoplastic therapy. (NCT01022996)
Timeframe: Every three months beginning at Cycle 3 until end of treatment due to progression of disease, unacceptable toxicity, death or discontinuation from the study for any other reason

InterventionDays (Mean)
RAD001321.9

[back to top]

Progression Free Survival (PFS) by Kaplan-Meier Estimate

Progression-free survival (PFS) was defined as the time from the first date of treatment to the date of first documented disease progression or death due to any cause or start of a new antineoplastic therapy. An event for PFS was defined as a documented disease progression or death due to any cause or start of a new antineoplastic therapy, whichever occurred first. Cycle = 28 days. (NCT01022996)
Timeframe: Every three months beginning at Cycle 3 until end of treatment due to progression of disease, unacceptable toxicity, death or discontinuation from the study for any other reason

InterventionDays (Median)
RAD0018.0

[back to top]

Time to Overall Response (TTR) Per Kaplan-Meier Estimate

Time to overall response was defined as the time from the first date of treatment to the date of first documented response of CR or PR. Time to overall response is applied to patients whose best overall response is CR or PR. Patients who drop-out or did not have a response (CR or PR) will be treated as censored at the date of last adequate tumor assessment. (NCT01022996)
Timeframe: Every three months beginning at Cycle 3 until end of treatment due to progression of disease, unacceptable toxicity, death or discontinuation from the study for any other reason

InterventionDays (Median)
RAD001NA

[back to top]

Duration of Overall Response (DoR)

The duration of overall response was calculated from the date of first documented response (CR or PR) to the date of first documented disease progression or death due to any cause or start of a new antineoplastic therapy. This only applies to patients whose best overall response is CR or PR. (NCT01022996)
Timeframe: Every three months beginning at Cycle 3 until end of treatment due to progression of disease, unacceptable toxicity, death or discontinuation from the study for any other reason

InterventionDays (Mean)
RAD001350.8

[back to top]

Treatment Failure Rate

Occurrence or not of treatment failure in each patient. Treatment failure was defined as a composite endpoint of biopsy-proven acute rejection (a biopsy graded IA, IB, IIA, IIB or III according to Banff '97 grading with 2007 update), graft loss, death or lost to follow-up occurring after randomization (V5) and within M12 (V9). (NCT01023815)
Timeframe: Between randomization (Month 3) and Month 12

InterventionParticipants (Number)
Group A - Once-a-day Regimen3
Group B - Steroid Withdrawal Group10
Group C - Standard Twice-a-day Group2

[back to top]

Number of Participants With Graft and Patient Survival After Randomization

"Graft Survival, calculated from the date of transplantation to the date of irreversible graft failure signified by return to long-term retransplantation or the date of the last follow-up during the period when the transplant was still functioning or to the date of death.~Patient survival, calculated from the date of transplantation to the date of death or the date of the last follow-up." (NCT01023815)
Timeframe: Month 3 to Month 12

InterventionParticipants (Number)
Group B - Steroid Withdrawal Group68
Group C - Standard Twice-a-day Group71

[back to top]

Changes in the Estimated Glomerular Filtration Rate (eGFR) Between Randomization (Month 3) and Month 12

eGFR by Nankivell, in terms of descriptive statistics and change vs randomization visit - to compare the changes in the estimated GFR (Nankivell) between randomization and Month 12 in the steroid withdrawal group (Group B) to the change observed in the standard twice-a-day group (Group C), for non-inferiority (NCT01023815)
Timeframe: Month 3 to Month 12

InterventionmL/min (Mean)
Group B - Steroid Withdrawal Group-1.7
Group C - Standard Twice-a-day Group2.5

[back to top]

Biopsy Proven Acute Rejection (BPAR) Rate Between Randomization and Month 12

"Occurrence of BPAR (after randomization) between arm B (steroid withdrawal group) and arm c (standard twice-a-day group).~BPAR was defined as a biopsy graded IA, IB, IIA, IIB, or III according to Banff 1997 grading with 2007 update." (NCT01023815)
Timeframe: Month 3 to Month 12

InterventionParticipants (Number)
Group B - Steroid Withdrawal Group9
Group C - Standard Twice-a-day Group2

[back to top]

Change in Serum Creatinine

Serum creatinine (a blood measurement) is an important indicator of renal health because it is an easily-measured by-product of muscle metabolism. Measuring serum creatinine is a simple test and it is the most commonly used indicator of renal function. (NCT01023815)
Timeframe: M3, M12

,
Interventionmg/dL (Mean)
Serum Creatinine @ month 3Serum Creatinine @ month 12
Group B - Steroid Withdrawal Group1.41.5
Group C - Standard Twice-a-day Group1.41.4

[back to top]

Change in Estimated Creatine Clearance

At each visit, estimated creatinine clearance was measured in the local laboratory to analyze the evolution of the renal function. The following indirect measures of renal function were computed: estimated creatinine clearance according to Cockcroft and Gault formula and MDRD formula. (NCT01023815)
Timeframe: M3, M12

,
InterventionmL/min (Mean)
Using Cockcroft and Gault model @ month 3Using Cockcroft and Gault model @ month 12Using MDRD-4 formular @ month 3Using MDRD-4 formular @ month 12
Group B - Steroid Withdrawal Group64.862.357.953.6
Group C - Standard Twice-a-day Group63.066.958.861.8

[back to top]

To Determine the Rate of Clinical Benefit (i.e. Rate of Complete or Partial Response Plus Stable Disease) at 16 Weeks for Patients With Malignant Mesothelioma Treated With Everolimus as Second or Third Line Therapy.

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR (NCT01024946)
Timeframe: 16 weeks

Interventionparticipants (Number)
Stable DiseaseProgression of Disease
Patients Getting Everolimus42

[back to top]

Number of Participants With Incidence of Proteinuria Events

Number of participants with Incidence of proteinuria events indicating chronic kidney disease (NCT01025817)
Timeframe: Baseline and 12 Months

,
InterventionParticipants (Number)
Baseline: Proteinuria (>=300 mg/g)Month 12, Day 316-450: Proteinuria (>=300 mg/g)
Everolimus and Low Dose Tacrolimus24336
Mycophenolate Mofetil and Standard Dose Tacrolimus25035

[back to top]

Number of Participants With Incidence Rates of BKV Viremia, BKV Viruria, or BKV Nephropathy

Participants with Incidence of BKV (viremia, viruria, or nephropathy). BKV is Polyomavirus type BK. (NCT01025817)
Timeframe: 12 Months

,
InterventionParticipants (Number)
Lab evidence of BKV ViremiaLab evidence of BKV ViruriaBKV Disease (Nephropathy)
Everolimus and Low Dose Tacrolimus19195
Mycophenolate Mofetil and Standard Dose Tacrolimus27155

[back to top]

Number of Participants With Incidence of Adverse Events, Serious Adverse Events, and Tacrolimus-associated Adverse Events

Incidence of adverse events, serious adverse events, and tacrolimus-associated adverse events by System Organ Class (NCT01025817)
Timeframe: 12 Months

,
InterventionParticipants (Number)
Any system organ classMetabolism and nutrition disordersGastrointestinal disordersInjury, poisoning and procedural complicationsGeneral disorders &administration site conditionsInfections and infestationsInvestigationsRenal and urinary disordersVascular disordersBlood and lymphatic system disordersNervous system disordersRespiratory, thoracic and mediastinal disordersMusculoskeletal and connective tissue disordersSkin and subcutaneous tissue disordersPsychiatric disordersReproductive system and breast disordersCardiac disordersEye disordersImmune system disordersEndocrine disordersNeoplasms benign,malignant,other incl cysts/polypsEar and labyrinth disordersHepatobiliary disordersSurgical and medical proceduresCongenital, familial and genetic disordersSocial circumstances
Everolimus and Low Dose Tacrolimus3032662332231991841501411311301251221101099656512613121076200
Mycophenolate Mofetil and Standard Dose Tacrolimus30226324720217719614316012116315013411410810640473611815113061

[back to top]

Number of Participants With Incidence of CMV (Viremia, Syndrome and Disease)

Participants with incidence of CMV (viremia, syndrome and disease). CMV is cytomegalovirus. (NCT01025817)
Timeframe: 12 Months

,
InterventionParticipants (Number)
CMV syndrome eventLab evidence of CMV ViremiaCMV Disease
Everolimus and Low Dose Tacrolimus972
Mycophenolate Mofetil and Standard Dose Tacrolimus13108

[back to top]

Estimated Glomerular Filtration Rate (eGFR)

Renal function was assessed by estimated Glomerular Filtration Rate (eGFR) using the Modification of Diet in Renal Disease (MDRD) formula. MDRD formula: GFR [mL/min/1.73m˄2] = 186.3*(C˄-1.154)*(A˄-0.203)*G*R. DEFINITIONS: C = serum concentration of creatinine [mg/dL]; A = age [years]; G = 0.742 when gender is female, otherwise G = 1; R = 1.21 when race is black, otherwise R = 1 (NCT01025817)
Timeframe: 12 Months

InterventionmL/min/1.73m˄2 (Mean)
Everolimus and Low Dose Tacrolimus63.14
Mycophenolate Mofetil and Standard Dose Tacrolimus63.06

[back to top]

Number of Participants With Incidence of Composite Efficacy Failure

Efficacy failure rate used the composite endpoint of: (1) treated biopsy-proven acute rejection (BPAR)*, (2) graft loss**, (3) participant death or(4) loss to follow-up. *A treated BPAR was defined as a biopsy graded IA, IB, IIA, IIB, or III and which was treated with anti-rejection therapy. **Graft loss is defined as when the allograft was presumed lost on the day the participant started dialysis and was not able to subsequently be removed from dialysis. (NCT01025817)
Timeframe: 12 Months

,
InterventionParticipants (Number)
Composite EndpointTreated Biopsy-proven Acute rejection (BPAR)Graft LossDeathLoss to follow up
Everolimus and Low Dose Tacrolimus7659469
Mycophenolate Mofetil and Standard Dose Tacrolimus623412517

[back to top]

Number of Participants With Incidence of New Onset of Diabetes Mellitus

Incidence of new onset diabetes mellitus defined as non-diabetic patients before transplantation, who are receiving glucose lowering treatment for more than 30 days post-transplant, or with a random plasma glucose ≥200 mg dL (11.1 mmol/L) with 2 fasting plasma glucose values ≥126 mg/dL (7 mmol/L) (NCT01025817)
Timeframe: 12 Months

,
InterventionParticipants (Number)
Any New Onset DiabetesrandomGlucose≥200mg/dL w/2 fastingGlucose≥126mg/dLConcomitant Diabetes medicine for 30 days or more
Everolimus and Low Dose Tacrolimus251513
Mycophenolate Mofetil and Standard Dose Tacrolimus221214

[back to top]

Total Number of Participants Experienced a Response (Complete Response+Partial Response+Stable Disease)

The number participants who experienced Complete Response+Partial Response+Stable Disease per Response Evaluation Criteria in Solid Tumors (RECIST v1.1). (NCT01031381)
Timeframe: Within 4 weeks (28 days) of study treatment initiation (baseline)

Interventionparticipants (Number)
RAD001 + Bevacizumab4

[back to top]

Progression-free Survival (PFS) at 6-months

The percentage of participants who were alive with the disease (cancer) at 6 months after treatment, but whose disease had not worsened/progressed per Response Evaluation Criteria in Solid Tumors (RECIST v1.1). (NCT01031381)
Timeframe: Up to 36 months (data collection period for the cohort); Up to 6 months for participant

Interventionpercentage of participants (Number)
RAD001 + Bevacizumab28

[back to top]

Patients With Overall Response

Per Response Evaluation Criteria in Solid Tumor (RECIST) criteria v. 1.0: measurable lesions: complete response (CR) disappearance of target lesions, partial response (PR) > 30% decrease in the sum of the longest diameter (LD) of target lesions, progressive disease (PD) > 20% increase in the sum of the LD of target lesions or appearance of new lesions, stable disease (SD) neither sufficient decrease nor increase of the sum of smallest sum of the LD of target lesions (NCT01031446)
Timeframe: every 12 weeks

Interventionparticipants (Number)
Complete ResponsePartial ResponseProgressive DiseaseStable Disease
RAD001 and Cisplatin and Paclitaxel1121127

[back to top]

Maximum Feasible Dose in Milligrams Per Meter Squared of Body Surface Area (mg/m2) of Cisplatin and Paclitaxel for Women With Metastatic Breast Cancer

The recommended dose for the Phase II trial will be the most prevalent dose delivered per week in Phase I that allows for safe and feasible administration of the medications.The maximum tolerated dose (MTD) is defined as the dose preceding that at which 2 or more of 3 patients experience dose-limiting toxicity (DLT) during the initial cycle of therapy. DLTs include Common Toxicity Criteria (CTC) Grade 4 neutropenia (absolute neutrophil count [ANC] < 0.5 x 10 9/L for > 5 days), febrile neutropenia (ANC < 1.0 x 10 0/L with fever > 38.5 degrees Centigrade) or documented infection associated with Grade 3-4 neutropenia, CTC Grade 4 thrombocytopenia < 25 x 10 9/L or CTC Grade 3 < 50-25 x 10 9/L thrombocytopenia with bleeding, and Grade 3-4 non-hematologic toxicity despite symptomatic therapy. (NCT01031446)
Timeframe: at 8 weeks

Interventionmg/m2 (Number)
CisplatinPaclitaxel
RAD001 and Cisplatin and Paclitazel2580

[back to top]

Time to Progression in Patients With Metastatic Basal-like Breast Cancer.

Median duration in months from on-study to disease progression in patients with metastatic basal-like breast cancer. All patients with basal-like breast cancer are negative for estrogen, progesterone, and human epidermal growth factor (HER2) receptors. (NCT01031446)
Timeframe: Up to 64 weeks

Interventionmonths (Median)
RAD001 and Cisplatin and Paclitaxel4.0

[back to top]

Time to Progression

Duration in months from date on-study to date patient exhibited progressive disease (NCT01031446)
Timeframe: Up to 64 weeks

Interventionmonths (Median)
RAD001 and Cisplatin and Paclitaxel4.0

[back to top]

Patients With Progression-free Survival

Patients who had not experienced disease progression and who were alive at 6 months after study entry (NCT01031446)
Timeframe: at 6 months

Interventionparticipants (Number)
RAD001 and Cisplatin and Paclitaxel21

[back to top]

Maximum Feasible Dose in mg of RAD001 (Everolimus)for Women With Metastatic Breast Cancer

The recommended dose for the Phase II trial will be the most prevalent dose delivered per day in Phase I that allows for safe and feasible administration the medication. The MTD is defined as the dose preceding that at which 2 or more of 3 patients experience dose-limiting toxicity (DLT) during the initial cycle of therapy. DLTs include Common Toxicity Criteria (CTC) Grade 4 neutropenia (absolute neutrophil count [ANC] < 0.5 x 10 9/L for > 5 days), febrile neutropenia (ANC < 1.0 x 10 0/L with fever > 38.5 degrees Centigrade) or documented infection associated with Grade 3-4 neutropenia, CTC Grade 4 thrombocytopenia < 25 x 10 9/L or CTC Grade 3 < 50-25 x 10 9/L thrombocytopenia with bleeding, and Grade 3-4 non-hematologic toxicity despite symptomatic therapy (NCT01031446)
Timeframe: at 8 weeks

Interventionmg (Number)
RAD001 and Cisplatin and Paclitaxel5

[back to top]

Exploratory Objective: Correlation of PFS With Biomarkers

Exploratory analysis of serum biomarkers were undertaken to generate a potential signature for response. The correlation with 6 month progression free survival P value for four plasma biomarkers is reported. (NCT01034631)
Timeframe: 6 months

InterventionCorrelation with PFS P Value (Number)
Serum Amyloid P-Component (SAP)Sex Hormone-Binding Globulin (SHBG)Matrix Metalloproteinase-9 (MMP-9)Stem Cell Factor (SCF)
Phase II Participants With Sufficient Correlative Samples0.01840.00630.04210.0291

[back to top]

Phase II: Response Rate With Combination Therapy Compared to Everolimus Alone

Objective response is defined as a confirmed CR or PR per RECIST criteria. Complete Response (CR): Disappearance of all target lesions. Partial Response (PR): At least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LD. Progressive Disease (PD): At least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum LD since the treatment started. (NCT01034631)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Combination Arm A: Everolimus + BNC105P1
Sequential Arm B:Everolimus Followed by BNC105P Monotherapy1

[back to top]

Phase I: Toxicities of BNC105P in Combination With Everolimus.

Determine the toxicities of BNC105P in combination with everolimus. Drug-related treatment emergent adverse events by CTCAE grade 2 or greater are reported (NCT01034631)
Timeframe: Until disease progression or unacceptable toxicity, up to 24 cycles or 24 months

Interventionparticipants (Number)
AST-SGOT Grade 2Cough Grade 2Dyspepsia Grade 2Hemoglobin Grade 2Hemoglobin Grade 3Hypomagnesia Grade 2Low Platelets Grade 2Weight Loss Grade 2Fatigue Grade 2Left ventricular systolic dysfunction Grade 2Mucositis (oral) Grade 2Nail Infection Grade 2Diaphoresis Grade 2Pericardial effusion Grade 3Pleural effusion Grade 2
Phase I Participants111411112121111

[back to top]

Phase II: Adverse Events of Everolimus and BNC105P When Administered as a Combination or Sequential Regimen.

Determine adverse events of everolimus and BNC105P when administered as a combination or sequential regimen. Total number of serious and non-serious adverse events for Arm A and Arm B are summarized. Complete adverse event information is supplied in the Adverse Events reporting section. (NCT01034631)
Timeframe: 12 months

,
Interventionnumber of adverse events (Number)
Non-Serious Adverse EventsSerious Adverse Events
Combination Arm A: Everolimus + BNC105P141939
Sequential Arm B:Everolimus Followed by BNC105P Monotherapy165450

[back to top]

Phase I: Response Rate of BNC105P in Combination With Everolimus.

Number of objective responses per RECIST criteria. Complete Response (CR): Disappearance of all target lesions. Partial Response (PR): At least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LD. Progressive Disease (PD): At least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum LD since the treatment started. (NCT01034631)
Timeframe: Until disease progression or unacceptable toxicity, up to 24 cycles or 24 months

InterventionParticipants (Count of Participants)
CRPRSDPDUnknown
Phase I Participants00843

[back to top]

Geometric Mean Half-life of BNC105 and BNC105P in Combination With Everolimus.

Determine the PK Profile for BN105P in combination with everolimus by calculating the geometric mean half-life of BNC105P (NCT01034631)
Timeframe: 12 months

Interventionhours (Geometric Mean)
Half-life of BNC105Half-life of BNC105P
Phase I Participants.32.08

[back to top]

Phase I: Maximum Tolerated Dose of BNC105P in Combination With Everolimus.

Phase I (NCT01034631)
Timeframe: Until disease progression or unacceptable toxicity, up to 24 cycles or 24 months

Interventionmg/m^2 (Number)
Phase I Participants16

[back to top]

Phase II: 6-month Progression Free Survival (PFS) With the Addition of BNC105P to Everolimus.

Improvement in 6-month PFS with the addition of BNC105P to everolimus. Progression is defined using RECIST criteria as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions (NCT01034631)
Timeframe: 6 months

Interventionprobability of 6MPFS (Number)
Combination Arm A: Everolimus + BNC105P.3382
Sequential Arm B:Everolimus Followed by BNC105P Monotherapy.3030

[back to top]

Phase II: Overall Survival

Determine overall survival probability, up to a maximum of 5 years from registration for protocol therapy. (NCT01034631)
Timeframe: 60 months

Interventionprobability of OS at 60 months. (Number)
Combination Arm A: Everolimus + BNC105P.15
Sequential Arm B:Everolimus Followed by BNC105P Monotherapy.21

[back to top]

Phase II: Progression Free Survival (PFS) With BNC105P Alone in Patients After Progressing on Everolimus.

Median time to progression for arm P participants who crossed over to BNC105P monotherapy after progression. Progression is defined per RECIST criteria as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions (NCT01034631)
Timeframe: 12 months

Interventionmonths (Median)
Arm B Participants Who Crossed Over to BNC105P Monotherapy1.8

[back to top]

Time to Tumor Progression (TTP)

TTP was defined as the time from the date of randomization to the date of the first documented radiologic confirmation of disease progression. Since the study did not meet the primary objective, TTP was not formally tested. (NCT01035229)
Timeframe: Until all patients have disease progression or leave study due to intolerable adverse events- Estimate of 1 year for each patient

InterventionMonths (Median)
Everolimus + Best Supportive Care (BSC)2.96
Placebo + Best Supportive Care2.60

[back to top]

Time to Definitive Deterioration of EORTC QLQ-C30 Scores

The primary quality of life endpoint was the time to definitive 5% deterioration from baseline in the global health status/quality of life scale of the EORTC QLQ-C30 questionnaire. Definitive deterioration by at least 5% is defined as a decrease in score by at least 5% compared to baseline, with no later observed increase above this threshold. The EORTC quality of life questionnaire (QLQ) is an integrated system for assessing the healthrelated quality of life (QoL) of cancer patients participating in international clinical trials. All of the scales and single-item measures range in score from 0 to 100. A high scale score represents a higher response level. Thus a high score for a functional scale represents a high / healthy level of functioning, a high score for the global health status / QoL represents a high QoL, but a high score for a symptom scale / item represents a high level of symptomatology / problems. (NCT01035229)
Timeframe: Until all patients have disease progression or leave study due to intolerable adverse events - Estimate of 1 year for each patient.

InterventionMonths (Median)
Everolimus + Best Supportive Care (BSC)2.86
Placebo + Best Supportive Care3.45

[back to top]

Time to Definitive Deterioration of ECOG Performance Score (PS) Score

Change in Eastern Cooperative Oncology Group (ECOG) were assessed by time to definitive performance status deterioration by at least one category on the ECOG scale. Deterioration was considered definitive if no improvement in the ECOG PS was observed at a subsequent measurement. ECOG PS: 0=Fully active, able to carry on all pre-disease performance without restriction, 1=Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature, e.g., light house work, office work; 2=Ambulatory and capable of all selfcare but unable to carry out any work activities. Up and about more than 50% of waking hours; 3=Capable of only limited selfcare, confined to bed or chair more than 50% of waking hours; 4=Completely disabled. Cannot carry on any selfcare. Totally confined to bed or chair; 5=Dead (NCT01035229)
Timeframe: Until all patients have disease progression or leave study due to intolerable adverse events- Estimate of 1 year for each patient.

InterventionMonths (Median)
Everolimus + Best Supportive Care (BSC)4.27
Placebo + Best Supportive Care4.47

[back to top]

Pharmacokinetics Assessments - Cmin

Cmin is the pre-dose blood concentration at steady-state (ng/mL). Pre-dose (Cmin) blood samples were collected from all patients in both arms at Visit 3. Steady-state for the Cmin sample was defined as continuous administration of the same dose in the last 4 days prior to the collection of the Cmin sample. Steady-state for the 5 mg every other day regimen was defined as the state when the 5 mg dose was taken 2 days and 4 days before sampling. PK samples were only drawn at visit 3, and only analyzed for patients receiving everolimus at steady state (if patients had received the dose the previous 4 days). In addition summary statistics were only done for each everolimus dose when 3 samples were available. Only valid pre-dose (Cmin) everolimus samples were included in the analysis. (NCT01035229)
Timeframe: Until all patients have disease progression or leave study due to intolerable adverse events - Estimate of 1 year for each patient.

Interventionng/mL (Mean)
Everolimus 7.5mg + Best Supportive Care (BSC)16.141
Everolimus 5mg + Best Supportive Care (BSC)9.318

[back to top]

Pharmacokinetics Assessments - Cmax

Cmax is the maximum (peak) blood drug concentration after dose administration (ng/mL) calculated as the maximum of C1h and C2h. C1h was 1 hour post-dose blood concentration (ng/mL) and C2h was 2 hour post-dose blood concentration (ng/mL). C1h and C2h post-dose samples were collected from all patients in both arms at Visit 3. Steady-state for the C1h and C2h samples was defined as continuous administration of the same dose in the previous 4 days and the day on which the C1h and C2h samples were collected. Steady-state for the 5 mg every other day regimen was defined as the state when the 5 mg dose was taken 2 days and 4 days before sampling. PK samples were only drawn at visit 3, and only analyzed for patients receiving everolimus at steady state (if patients had received the dose the previous 4 days). In addition summary statistics were only done for each everolimus dose when 3 samples were available. Only valid C1h and C2h everolimus samples were included in the analysis. (NCT01035229)
Timeframe: Until all patients have disease progression or leave study due to intolerable adverse events- Estimate of 1 year for each patient.

Interventionng/mL (Mean)
Everolimus 7.5mg + Best Supportive Care (BSC)47.881
Everolimus 5mg + Best Supportive Care (BSC)31.592

[back to top]

Percentage of Participants With Disease Control Rate (DCR)

DCR is defined as the proportion of participants with a best objective response (BOR) of complete response (CR) or partial response (PR) or stable disease (SD) according to RECIST. The BOR was the best response recorded from the start of the treatment until disease progression. CR is disappearance of all target lesions; PR is at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters; SD is neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for PD. PD is at least a 20% increase in the sum of the longest diameter of all measured target lesions, taking as reference the smallest sum of longest diameter of all target lesions recorded at or after baseline. (NCT01035229)
Timeframe: Until all patients have disease progression or leave study due to intolerable adverse events- Estimate of 1 year for each patient

InterventionPercentage of Participants (Number)
Everolimus + Best Supportive Care (BSC)56.1
Placebo + Best Supportive Care45.1

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of randomization to the date of death from any cause. The comparison of OS between the 2 arms was done using a stratified log-rank test at one-sided 2.5% level of significance. (NCT01035229)
Timeframe: When 454 OS events were observed

InterventionMonths (Median)
Everolimus + Best Supportive Care (BSC)7.56
Placebo + Best Supportive Care7.33

[back to top]

Evaluate Safety of the Combination at a Daily Dosing of 2.5mg RAD001, 5 mg RAD001 or 10 mg RAD001 (Phase 1 Part)

Number of patients who experienced a Dose Limiting Toxicity (DLT). DLT will be assessed in the first 28 days of dosing. Patients need to get dosed with 2 rounds/sessions of all chemotherapy agents in the first 28 days in order to be evaluable for DLT assessment. The primary endpoint is safety as summarized by dose limiting toxicity (DLT). (NCT01047293)
Timeframe: December 2011

Interventionparticipants (Number)
ARM 1 RAD001 5 mg QOD0
ARM 2 5mg RAD001 QD1
ARM 3 10mg RAD001 QD1

[back to top]

Progression Free Survival at Six Months

(NCT01047293)
Timeframe: 6 months

Interventionpercentage of participants (Number)
All Patients87

[back to top]

PSA Response Rate

PSA response rate with response defined as => a 30% reduction in PSA (NCT01051570)
Timeframe: Day 1 of each cycle (every 21 days), through study completion, an average of 6 months

Interventionpercentage of participants (Number)
Carboplatin, RAD 001 & Prednisone15

[back to top]

Association of PSA Response Rate With Correlative Markers (Phospho mTOR, pAKT, and p70S6)

PSA response defined as a decrease of 30% or more will be tabled against mTOR, pAKT, and p70S6 (1+, 2+, 3+ vs ND) (NCT01051570)
Timeframe: Archival tissue will be collected if available. Optional biopsies pre-treatment and 24 hours after first everolimus and carboplatin dose

Interventionparticipants (Number)
pAKT(ND) vs RespondermTOR(ND) vs Responderp70S6(ND) vs Responder
Carboplatin, RAD 001 & Prednisone101

[back to top]

Overall Survival

Overall Survival as measured by the Kaplan-Meier method (NCT01051570)
Timeframe: After treatment, participants will be contacted every 3 months up to 4 years

Interventionmonths (Median)
Carboplatin, RAD 001 & Prednisone12.5

[back to top]

Pharmacokinetics: Observed Carboplatin AUC Was Estimated Based on the Concentration in the 2.75-h Sample.

Using a limited sampling model (i.e., AUC = 0.52 × C2.75h + 0.92) (Sorensen et al., 1993), observed carboplatin AUC was estimated based on the concentration in the 2.75-h sample. (NCT01051570)
Timeframe: Samples were collected Cycle 2, Day 1

Interventionmg/ml*min (Mean)
Carboplatin, RAD 001 & Prednisone5.8

[back to top]

Time to Progression (TTP)

Progression defined as at least a 20% increase in the sum of the longest diameter (LD) of target lesions taking as references the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. (NCT01051570)
Timeframe: Up to 63 days while on treatment, then up 90 days thereafter. From date of registration to date of progressive disease.

Interventionmonths (Median)
Carboplatin, RAD 001 & Prednisone2.5

[back to top]

Number of Participants With Toxicity as Measured by NCI CTCAE v3.0 Criteria

Number of Participants with Grade 3/4 Toxicity as measured by NCI CTCAE v3.0 criteria (NCT01051570)
Timeframe: Day 1 of each cycle (every 21 days), through study completion, an average of 6 months

InterventionParticipants (Count of Participants)
AnemiaThrombocytopeniaLymphopeniaLeukopeniaInfection without neutropeniaHypophosphatemiaNeutropeniaDehydrationHyperglycemiaHyponatremiaPulmonary embolismFatigueHypercholesterolemiaRashASTHypomagnesemiaHypokalemia
Carboplatin, RAD 001 & Prednisone109644433332211111

[back to top]

Overall Survival (OS)

(NCT01051791)
Timeframe: Up to 60 months

Interventionmonths (Median)
Everolimus 10 mg Daily4.5

[back to top]

Progression Free Survival (PFS)

(NCT01051791)
Timeframe: Up to 60 months

Interventionmonths (Median)
Everolimus 10 mg Daily1.5

[back to top]

Objective Response Rate (ORR)

The number of patients with recurrent/refractory SCCHN (head and neck squamous cell carcinoma) treated with single-agent everolimus that, experience a Complete Response (CR) + number of patients that experience a Partial Response (PR ) / total evaluable patients. (NCT01051791)
Timeframe: Up to 60 months

Interventionpercentage of participants (Number)
Everolimus 10 mg Daily0

[back to top]

Clinical Benefit Rate (CBR)

The number of patients with recurrent/refractory SCCHN (head and neck squamous cell carcinoma) treated with single-agent everolimus that, experience a Complete Response (CR) + number of patients that experience a Partial Response (PR ) + number of patients that experience Stable Disease (SD) / total evaluable patients. (NCT01051791)
Timeframe: Up to 60 months

Interventionpercentage of participants (Number)
Everolimus 10 mg Daily28

[back to top]

Response

(NCT01057277)
Timeframe: 1 year

Interventionparticipants (Number)
RAD001(Afinitor)NA

[back to top]

Overall Survival (OS) [Phase II]

OS based on the Kaplan-Meier method is defined as the time from study entry to death or date last known alive. (NCT01058655)
Timeframe: Long-term follow-up for survival was not specified per protocol. Participants were followed for up to 20 months on this study.

Interventionmonths (Median)
Phase II: Everolimus 10 mg + Tivozanib 1 mg [Evaluable]5.6

[back to top]

Everolimus Maximum Tolerated Dose (MTD) [Phase I]

The everolimus MTD in combination with tivozanib is determined by the number of patients who experience a dose limiting toxicity (DLT). See subsequent primary outcome measure for the DLT definition. The MTD is defined as the highest dose at which fewer than one-third of patients experience a DLT. If no DLTs are observed, the MTD is not reached. (NCT01058655)
Timeframe: Patients were assessed continuously for toxicity while on study. The observation period for MTD evaluation was the first 28 days (cycle 1) of treatment.

Interventionmg daily for 4 weeks of a 4 week cycle (Number)
Phase I: Evaluable10

[back to top]

Dose Limiting Toxicity (DLT) [Phase I]

A DLT was defined as a treatment-related (attribution possible, probable, definite) adverse event that meets any of the following criteria: Grade 3 (G3) or higher non-hematologic toxicity (excluding, nausea, vomiting, diarrhea, alopecia, hypertension, hypercholesterolemia, or hypertriglyceridemia); G3 diarrhea, nausea or vomiting lasting > 48 hours or leading to hospitalization, despite aggressive anti-diarrheal or anti-emetic medications; G4 diarrhea, despite aggressive anti-diarrheal medications; G4 vomiting, despite aggressive anti-emetic medications; G3 hypertension, for which blood pressure cannot be reduced to <150/100 with anti-hypertensive therapies; G4 hypertension or severe hypertension, as defined by systolic blood pressure >180 mmHg or diastolic blood pressure > 110 mmHg; G4 hypercholesterolemia or hypertriglyceridemia lasting > 7 days, despite appropriate use of anti-hyperlipidemic medications; G4 hematologic toxicity lasting for >5 days, including leukopenia, neutropen (NCT01058655)
Timeframe: Patients were assessed continuously for toxicity while on study. The observation period for DLT evaluation was the first 28 days (cycle 1) of treatment.

InterventionParticipants with DLT (Number)
Phase I Cohort 1: Everolimus 5 mg + Tivozanib 1 mg0
Phase I Cohort 2: Everolimus 10 mg + Tivozanib 1 mg0
Phase I Cohort 3: Everolimus 10 mg + Tivozanib 1.5 mg2

[back to top]

Disease Control Rate (DCR) [Phase II]

Disease Control Rate is defined as the percentage of patients who achieve confirmed stable disease (SD) or better on treatment based on RECIST 1.0 criteria. Per RECIST 1.0 for target lesions, complete response (CR) is disappearance of all target lesions and partial response (PR) is at least a 30% decrease in the sum of longest diameter (LD) of target lesions, taking as reference baseline sum LD. Progressive disease (PD) is at least a 20% increase in sum LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started. SD is neither PR nor PD. For non-target lesions, PD is the appearance of one or more new lesions and/or unequivocal progression of existing non-target lesions. (NCT01058655)
Timeframe: Disease was assessed every 2 cycles on treatment. Median treatment duration on this study cohort was 2 months (range 1-16).

Interventionpercentage of participants (Number)
Phase II: Everolimus 10 mg + Tivozanib 1 mg [Evaluable]50

[back to top]

Tivozanib Maximum Tolerated Dose (MTD) [Phase I]

The tivozanib MTD in combination with everolimus is determined by the number of patients who experience a dose limiting toxicity (DLT). See subsequent primary outcome measure for the DLT definition. The MTD is defined as the highest dose at which fewer than one-third of patients experience a DLT. If no DLTs are observed, the MTD is not reached. (NCT01058655)
Timeframe: Patients were assessed continuously for toxicity while on study. The observation period for MTD evaluation was the first 28 days (cycle 1) of treatment.

Interventionmg daily for 3 weeks of a 4 week cycle (Number)
Phase I: Evaluable1.0

[back to top]

Progression-Free Survival (PFS) [Phase II]

PFS based on the Kaplan-Meier method is defined as the time from study entry to the earliest documentation of disease progression (PD) or death. Participants alive without evidence of PD were censored at the earliest date of last disease assessment. Per RECIST 1.0 criteria: progressive disease (PD) is at least a 20% increase in the sum of longest diameter (LD) of target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. PD for the evaluation of non-target lesions is the appearance of one or more new lesions and/or unequivocal progression of non-target lesions. (NCT01058655)
Timeframe: Disease was assessed radiographically to document clinical progression every 2 cycles on treatment. Participants were followed for up to 16 months since study entry.

Interventionmonths (Median)
Phase II: Everolimus 10 mg + Tivozanib 1 mg [Evaluable]3.0

[back to top]

Change From Baseline in Forced Vital Capacity (FVC)

All spirometry evaluation followed recommendations of the Standardization of Lung Function Testing. All spirometry maneuvers were performed in sitting position whilst wearing nose clips. At least three acceptable maneuvers were performed for each time point, and results met within-test and between-test criteria for acceptability. The highest value was obtained of FVC from any of the three maneuvers that met acceptability criteria. Change from baseline in FVC was compared between everolimus and historical placebo data from multicenter trial of sirolimus in LAM. (MILES NCT00414648) due to absence of placebo group in this current study on a rare lung disease. These two studies had different study designs, so the change from baseline to 6 months (26 weeks) in MILES study was estimated from the publicly reported rate of change per month to provide a meaningful comparison. This historical control be can be viewed on the Novartis Clinical Trial Results website. (NCT01059318)
Timeframe: Baseline, 26 weeks

InterventionmL (Mean)
Everolimus10

[back to top]

Change From Baseline in Forced Expiratory Volume in 1 Second (FEV1)

"All spirometry evaluation followed the recommendations of the Standardization of Lung Function Testing. All spirometry maneuvers were performed in the sitting position whilst wearing nose clips. At least three acceptable maneuvers were performed for each time point, and the results met within-test and between-test criteria for acceptability. The highest value was obtained of FEV1 from any of the three maneuvers that met acceptability criteria.~Change from baseline in FEV1 was compared between everolimus and historical placebo data from the MILES study (NCT00414648) due to the absence of a placebo group in this current study on a rare lung disease. These two studies had different study designs, so the change from baseline to 6 months (26 weeks) in MILES study was estimated from the publicly reported rate of change per month in order to provide a meaningful comparison. This historical control can be viewed on the Novartis Clinical Trial Results website." (NCT01059318)
Timeframe: Baseline, 26 weeks

InterventionmL (Mean)
Everolimus114

[back to top]

Change From Baseline in Extended Pulmonary Function Testing

Extended pulmonary function testing such as Total Lung Capacity (TLC), Thoracic Gas Volume (TGV), Residual Volume (RV) were measured using a body plethysmograph (NCT01059318)
Timeframe: Baseline, 26 weeks

InterventionLiters (L) (Mean)
TLCTGVRV
Everolimus0.1990.2670.169

[back to top]

Change From Baseline in 6-minute Walk Test Score to Measure Exercise Capacity

A standardized 6-minute walk test (6MWT) was performed in accordance with the guidelines of the American Thoracic Society 2002. The test was done about the same time of day to avoid diurnal variation in an environment that had an adequate temperature to avoid additional burden to the patient due to heat or cold air. The distance walked in six minutes (6MWD) was recorded. (NCT01059318)
Timeframe: Baseline, 26 weeks

Interventionmeters (m) (Mean)
Everolimus46.9

[back to top]

Change From Baseline in Carbon Monoxide Diffusing Capacity (DLCO)

Carbon Monoxide Diffusing Capacity (DLCO) one of the extended pulmonary function testing which was also measured using a body plethysmograph. (NCT01059318)
Timeframe: Baseline, 26 weeks

Interventionml/min/mmHg (Mean)
Everolimus-0.782

[back to top]

Mean Trough (C0,ss) and Peak (C2,ss) Drug Concentration at Steady State

Venous blood samples (2 mL) for pharmacokinetic evaluation were collected pre dose and 2 hours post dose at preselected visits. (NCT01059318)
Timeframe: pre-dose and at 2 hour post dose at week 26

,,
Interventionng/mL (Mean)
Trough (C0, ss)Peak (C2, ss)
Everolimus 10 mg/Day11.045.7
Everolimus 2.5 mg/Day3.112.1
Everolimus 5 mg/Day5.822.6

[back to top]

Change From Baseline in Vascular Endothelial Growth Factor-D (VEGF-D) Concentrations

Blood samples (1 mL) for determination of VEGF-D were collected from a forearm vein (direct venipuncture or from an indwelling cannula) and 2 aliquots of serum were collected. VEGF-D levels were determined from only 1 of the 2 serum aliquots, with the second acting as a back-up. A serum VEGF-D >800 pg/mL level supports a diagnosis of Lymphangioleiomyomatosis (LAM) (NCT01059318)
Timeframe: Baseline, 26 weeks

Interventionpg/mL (Mean)
Everolimus 2.5 mg/Day-464.3
Everolimus 5 mg/Day-1113.2
Everolimus 10 mg/Day-1771.7

[back to top]

Change From Baseline in Oxygen Saturation

Oxygen saturation means the amount of oxygen in blood stream. Oxygen saturation was measured by using a Pulse Oximeter. (NCT01059318)
Timeframe: Baseline, 26 weeks

Interventionpercentage of oxygen (Mean)
Everolimus0.8

[back to top]

Phase II: Distribution of Worst Adverse Event Grade

The worst/highest grade of any adverse event reported was determined for each patient. The percentage of patients in each grade level is reported. Adverse events are graded using CTCAE v4.0. Grade refers to the severity of the AE. The CTCAE v4.0 assigns Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grade 1 Mild AE, Grade 2 Moderate AE, Grade 3 Severe AE, Grade 4 Life-threatening or disabling AE, Grade 5 Death related to AE. (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

,
Interventionpercentage of patients (Number)
Grade 1Grade 2Grade 3Grade 4Grade 5
Ph II: RT + TMZ00332
Ph II: RT + TMZ + RAD00103501

[back to top]

Phase I: Number of Patients With Dose-limiting Toxicity (DLT)

DLT is defined as any of the following events occurring during the first 8 weeks of treatment with RAD001 and temozolomide and attributable to the study drugs: any grade 3 or 4 thrombocytopenia, grade 4 anemia, or grade 4 neutropenia lasting more than 7 days; any non-hematologic grade 3 or greater adverse event (AE), excluding alopecia, despite maximal medical therapy; any grade 4 radiation-induced skin changes; failure to recover from adverse events to be eligible for re-treatment with RAD001 and temozolomide within 14 days of the last dose of either drug; or any episode of non-infectious pneumonitis grade 2, 3, or 4 of any duration. Adverse events are graded using CTCAE v4.0. Grade refers to the severity of the AE. The CTCAE v4.0 assigns Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grade 1 Mild AE, Grade 2 Moderate AE, Grade 3 Severe AE, Grade 4 Life-threatening or disabling AE, Grade 5 Death related to AE (NCT01062399)
Timeframe: From start of treatment to eight weeks.

InterventionParticipants (Count of Participants)
Ph I: RT + TMZ + RAD001 2.5 mg/Day2
Ph I: RT + TMZ + RAD001 5 mg/Day2
Ph I: RT + TMZ + RAD001 10 mg/Day2

[back to top]

Phase II: Overall Survival (OS)

Overall survival time is defined as time from/randomization to the date of death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

Interventionmonths (Median)
Ph II: RT + TMZ21.2
Ph II: RT + TMZ + RAD00116.5

[back to top]

Phase II: Progression-free Survival (PFS)

Using the Response Assessment in Neuro- Oncology (RANO) criteria, the progression is defined by any of the following: > 25% increase in sum of the products of perpendicular diameters of enhancing lesions compared to the smallest tumor measurement obtained either at baseline (if no decrease) or best response, on stable or increasing doses of corticosteroids; Significant increase in T2/FLAIR non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation of therapy, not due to co-morbid events; Any new lesion; Clear clinical deterioration not attributable to other causes apart from the tumor or changes in corticosteroid dose; Failure to return for evaluation due to death or deteriorating condition; Clear progression of non-measurable disease. PFS time is defined as time from registration to date of progression, death, or last known follow-up (censored). PFS rates are estimated using the Kaplan-Meier method. (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

Interventionmonths (Median)
Ph II: RT + TMZ10.2
Ph II: RT + TMZ + RAD0018.2

[back to top]

Phase I: Distribution of Worst Adverse Event Grade

"AE reporting in Phase I was split up by treatment timing: concurrent treatment (RT, TMZ, RAD001); post-RT treatment (TMZ, RAD001) along with all AE's reported in follow-up.~The worst/highest grade of any adverse event reported in each time period was determined for each patient. The percentage of patients in each grade level is reported. Adverse events are graded using CTCAE v4.0. Grade refers to the severity of the AE. The CTCAE v4.0 assigns Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grade 1 Mild AE, Grade 2 Moderate AE, Grade 3 Severe AE, Grade 4 Life-threatening or disabling AE, Grade 5 Death related to AE." (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

,,
Interventionpercentage of participants (Number)
Concurrent treatment: Grade 1Concurrent treatment: Grade 2Concurrent treatment: Grade 3Concurrent treatment: Grade 4Concurrent treatment: Grade 5Post-RT treatment: Grade 1Post-RT treatment: Grade 2Post-RT treatment: Grade 3Post-RT treatment: Grade 4Post-RT treatment: Grade 5
Ph I: RT + TMZ + RAD001 10 mg/Day12.50.087.50.00.00.0100.00.00.00.0
Ph I: RT + TMZ + RAD001 2.5 mg/Day0.00.050.050.00.00.00.071.414.314.3
Ph I: RT + TMZ + RAD001 5 mg/Day11.133.344.411.10.00.042.928.60.00.0

[back to top]

Number of Participants With Disease Progression

(NCT01068249)
Timeframe: from study entry (1st treatment) to date of tumor progression, date of death, or, for patients alive without tumor progression, date of last follow-up, up to 2 years

InterventionParticipants (Count of Participants)
RAD001 + Letrozole31

[back to top]

Number of Participants With Objective Response Rate

Objective response or stable disease rate monitored as patients accrue and are evaluated following the example of Thall and Simon. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR (NCT01068249)
Timeframe: at 8 weeks of treatment, then every 12 weeks, up to 2 years

InterventionParticipants (Count of Participants)
Complete Response (CR)Partial Response (PR)
RAD001 + Letrozole92

[back to top]

Number of Participants With Adverse Events (All Grades)

Toxicity was assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events (version 3.0). (NCT01068249)
Timeframe: Adverse events were reported from first dose of study treatment until end of study treatment plus 30 days post treatment

InterventionParticipants (Count of Participants)
RAD001 + Letrozole28

[back to top]

Median Overall Survival (OS)

OS is defined as the interval from first dose of study treatment to the date of death, irrespective of the cause of death; subjects still alive will be censored at the date of the last contact (NCT01068249)
Timeframe: from (1st treatment) to death or, for living patients, date of last contact, up to 24.4 months

Interventionmonths (Median)
RAD001 + Letrozole14

[back to top]

Median Progression Free Survival (PFS)

PFS is the time from first dose of study treatment until the first date of either objective disease progression or death due to any cause. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 30% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. Median PFS was estimated using the Kaplan-Meier method. (NCT01068249)
Timeframe: from study entry (1st treatment) to date of tumor progression, date of death, or, for patients alive without tumor progression, date of last follow-up, assessed up to 2 years

Interventionmonths (Median)
RAD001 + Letrozole3

[back to top]

Reduction in Seizure Frequency

The primary efficacy endpoint was the percentage of participants demonstrating a 50% or greater reduction in seizure frequency at the end of the maintenance phase (weeks 13-16) compared to baseline (weeks 1-4) (NCT01070316)
Timeframe: Baseline (Weeks 1-4), Week 16

Interventionpercentage (Number)
Everolimus60

[back to top]

Number of Participants Continuing Study Medication Over Time

(NCT01070316)
Timeframe: Individual subjects will be assessed every 6 months for up to 48 months; aggregate analysis will take place at end of study

Interventionparticipants (Number)
16 weeksEntered Extension Phase (4 months)6 months12 months18 months24 months30 months36 months42 months48 months
Everolimus20181817161616151414

[back to top]

Number of Patients Reporting Dose-Limiting Toxicity (DLT) (Phase I)

The number of dose-limiting toxic events (DLT) for this combination of drug treatment will determine the Maximum Tolerated Dose (MTD) in subsequent phases of this study. The following events were defined as a DLT: a grade 4+ Neutropenia or platelet count decrease, a grade 4 infection, or any grade 3+ non-hematologic event as assessed using Common Terminology Criteria for Adverse Events (CTCAE) CTEP Version 4.0. Here, the number of patients reporting a DLT are reported (NCT01075321)
Timeframe: After one 28 day cycle

InterventionParticipants (Count of Participants)
Phase I: Dose Level -11
Phase I: Dose Level 02
Phase I: Dose Level 13
Phase II: Dose Level 00

[back to top]

Duration of Response for All Eligible Patients

Duration of response is defined for all evaluable patients who have achieved an objective response as the date at which the patient's earliest objective status is first noted to be either a CR or PR to the earliest date progression is documented. (NCT01075321)
Timeframe: Up to 5 years

Interventionmonths (Median)
All Patients (Everolimus and Lenalidomide)14.7

[back to top]

Overall Survival for All Eligible Patients

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT01075321)
Timeframe: Up to 5 years

Interventionmonths (Median)
All Patients (Everolimus and Lenalidomide)20.3

[back to top]

Time to Treatment Failure for All Eligible Patients

Time to treatment failure is defined to be the time from registration to the date at which the patient is removed from treatment due to progression, adverse events, or refusal. The distribution of time to treatment failure will be estimated using the method of Kaplan-Meier. (NCT01075321)
Timeframe: Up to 5 years

Interventionmonths (Median)
All Patients (Everolimus and Lenalidomide)4.7

[back to top]

Progression-Free Survival For All Eligible Patients

Progression-free survival time is defined as the time from registration to the earliest date of documentation of disease progression. The distribution of progression-free survival time will be estimated using the method of Kaplan-Meier. (NCT01075321)
Timeframe: Up to 5 years

Interventionmonths (Median)
All Patients (Everolimus and Lenalidomide)5.3

[back to top]

Change in Interstitial Fibrosis/Tabular Atrophy (IF/TA) Grade

Difference (M12 - M3) in IF/TA grade. IF/TA grade was assessed during centralized reading according to the Banff 2005/2007 classification comprising grade I (<25%), grade II (25-50%) and grade III (>50% of lesions). (NCT01079143)
Timeframe: M3 and M12 post transplantation

,,,
InterventionNumber of Participants (Number)
Difference in IF/TA grade -1Difference in IF/TA grade 0Difference in IF/TA grade 1Difference in IF/TA grade 2Difference in IF/TA grade 3
Certican EMT-121183NA
Certican EMT+59732
Neoral EMT-233135NA
Neoral EMT+791150

[back to top]

Number of Participants With Progression of Renal Graft Fibrosis (Primary Comparison - ITT Population

"Progression of Interstitial Fibrosis/Tabular Atrophy (IF/TA) is the percentage (%) of participants with an increase >= 1 in IF/TA grade according to Banff (2005 - 2007)according to Epithelial-mesenchymal transition (EMT) profile and by treatment groups.~Grade I (the better): mild interstitial fibrosis and tubular atrophy (<25% of cortical area) Grade II : moderate interstitial fibrosis and tubular atrophy (26-50% of cortical area) Grade III (the worse) : severe interstitial fibrosis and tubular atrophy (>50% of cortical area)" (NCT01079143)
Timeframe: Month 3 (M3) and Month 12 (M12) post transplantation

,
InterventionParticipants (Number)
Participants with an IF/TA grade <= II at M3Participants with Fibrosis progression, M3 to M12
Certican EMT+2612
Neoral EMT+3116

[back to top]

Number of Participants With Progression of Renal Fibrosis Using Numerical Quantification

Comparisons according to epithelial-mesenchymal transition (EMT) profile and immunosuppressive treatment (NCT01079143)
Timeframe: M3 to M12 post transplantation

,,,
InterventionParticipants (Number)
Fibrosis Progression - No (n=24, 42, 31, 53)Fibrosis progression - Yes (n=24, 42,31,43)Fibrosis progression - Missing (n=24, 42, 31, 53)
Certican EMT-30121
Certican EMT+1862
Neoral EMT-42110
Neoral EMT+19120

[back to top]

Number of Participants With Epithelial-mesenchymal Transition (EMT) Status

Incidence and severity of EMT status. EMT status was determined centrally using the graft biopsy taken at 3 months. The result was quickly obtained (within 7 to 15 days) and sent to the company in charge of randomization in order to allocate each patient to a treatment group and to provide the investigator with this information without confirming EMT status. (NCT01079143)
Timeframe: M3 and M12 post transplantation

,,,
InterventionParticipants (Number)
EMT Status at M3-Negative (n=26,43,32,53)EMT Status at M3- Positive (n=26,43,32,53)EMT Status at M3 - Not done (n=26,43,32,53)EMT Status at M12- Negative (n=25,41,32,53)EMT Status at M12- Positive (n=25,41,32,53)EMT Status at M12- Not done (n=25,41,32,53)
Certican EMT-430024170
Certican EMT+02608170
Neoral EMT-530036170
Neoral EMT+03209230

[back to top]

Number of Participants With Epithelial-mesenchymal Transition (EMT) Score

"Incidence and severity of EMT score. The intensity of EMT markers expression present and detectable in the renal graft at an early stage following transplantation is known as EMT score.~EMT score 0 (the best): <1 EMT score 1 (the better): 1-10% of tubular atrophy EMT score 2: 10-25% of tubular atrophy EMT score 3: 25-50% of tubular atrophy EMT score 4 (the worst): >50% of tubular atrophy" (NCT01079143)
Timeframe: M3 and M12 post transplantation

,,,
Interventionparticipants (Number)
EMT Score 0 at M3 (n= 26,43,32, 53)EMT Score 1 at M3 (n= 26,43,32, 53)EMT Score 2 at M3 (n= 26,43,32, 53)EMT Score 3 at M3 (n= 26,43,32, 53)EMT Score 4 at M3 (n= 26,43,32, 53)EMT Score 0 at M12 (n= 25,41,32, 53)EMT Score 1 at M12 (n= 25,41,32, 53)EMT Score 2 at M12 (n= 25,41,32, 53)EMT Score 3 at M12 (n= 25,41,32, 53)EMT Score 4 at M12 (n= 25,41,32, 53)EMT Missing Score at M12 (n= 25,41,32, 53)
Certican EMT-2617000131111422
Certican EMT+0017901710431
Neoral EMT-2825000191712410
Neoral EMT+0020840910940

[back to top]

Interstitial Fibrosis/Tabular Atrophy (IF/TA)

Incidence and severity of IF/TA according to 2005/2007 Banff classification system grades (grades l to lll). IF/TA grade was assessed during centralized reading according to the Banff 2005/2007 classification comprising grade I (<25%), grade II (25-50%) and grade III (>50% of lesions). (NCT01079143)
Timeframe: M3 and M12 post transplantation

,,,
InterventionNumber of participants (Number)
Interstitial Fibrosis/Tubular Atrophy (IF/TA)IF/TA grade at M3 at grade 1IFTA grade at M3 at grade IIIF/TA grade at M3 at grade IIIIF/TA grade at M12 at grade 0IT/TA grade at M12 at grade IIF/TA grade at M12 at grade IIIF/TA grade at M12 at grade III
Certican EMT-38500201850
Certican EMT+1311209863
Neoral EMT-44900311561
Neoral EMT+13171171591

[back to top]

Incidence (Number) of BPAR

A biopsy proven acute rejection (BPAR) is defined as a biopsy graded IA, IB, IIA, IIB or III as per 2005/2007 Banff histological classification system. (NCT01079143)
Timeframe: M6 and M12 post transplantation

,,,
InterventionNumber of participants (Number)
M6 - NoM6 - YesM12 - NoM12 - Yes
Certican EMT-5284317
Certican EMT+333297
Neoral EMT-590563
Neoral EMT+390372

[back to top]

Change in EMT Score

"Change (M12 - M3) in EMT Score. Progression in EMT score is defined as an increase by >=1 of EMT score from M3 to M12.~EMT score 0 (the best): <1 EMT score 1 (the better) : 1-10% of tubular atrophy EMT score 2 : 10-25% of tubular atrophy EMT score 3 : 25-50% of tubular atrophy EMT score 4 (the worst): >50% of tubular atrophy" (NCT01079143)
Timeframe: M3 and M12 post transplantation

Interventionscores on a scale (Mean)
Certican EMT+-0.3
Certican EMT-0.9
Neoral EMT+-0.3
Neoral EMT-0.6

[back to top]

Incidence (Number) of Subclinical Rejections and Borderline Lesions

"Incidence of subclinical rejections and borderline lesions with regards to histological evidence of rejection with clinical findings.~Subclinical rejections: rejection without clinical symptoms which is diagnosed by chance when a graft biopsy is performed.~Clinically suspected BPAR: rejection suspected because of the presence of clinical symptoms (fever, pain, increase of creatinine) and then confirmed by the graft biopsy.~Borderline lesions: suspicious for acute T-cell mediated rejection. This category is used when no intimal arteritis is present, but there are foci of tubulitis with minor interstitial infiltration or interstitial infiltration with mild tubulitis." (NCT01079143)
Timeframe: M3

,
InterventionParticipants (Number)
Subclinical rejections-No (n=68, 84)Subclinical rejections- Yes (n=68, 84)Subclinical rejections- missing (n=68, 84)Clinically suspected BPAR - No (N=68, 84)Clinically suspected BPAR - Yes (N=68, 84)Clinically suspected BPAR - Missing (N=68, 84)Borderline lesions - No (n=68,84)Borderline lesions - Yes (n=68,84)Borderline lesions - Missing (n=68,84)
Certican671168016261
Neoral8401840171131

[back to top]

Change in Urine Protein/Creatinine Ratio (Without Imputation)

One of the factors associated with EMT progression between M3 and M12 according to univariate analysis (ITT biopsies M3 & M12). (NCT01079143)
Timeframe: Month 3 (baseline), Month 12

Interventionmg/mmol (Mean)
Certican EMT+44.4
Certican EMT-3.5
Neoral EMT+16.0
Neoral EMT-29.8

[back to top]

Change From Baseline (M3) in Estimated Glomerular Filtration Rate (eGFR)

"eGFR was calculated according to the abbreviated Modification of Diet in Renal Disease (MDRD) Formula and creatinine clearance according to the Cockcroft-Gault formula (mL/min/1.73m²).~LOCF = Last observation carried forward" (NCT01079143)
Timeframe: M3 (baseline) to M12 post transplantation

,
InterventionmL/min/1.73m^2 (Least Squares Mean)
without imputationimputation by LOCF(96, 97)
Certican6.995.96
Neoral2.542.15

[back to top]

Change in Estimated Glomerular Filtration Rate (eGFR) at M12 From Baseline (M3) - ANCOVA Model

The average patient renal function was evaluated on the basis of eGFR was calculated according to the abbreviated MDRD formula and creatinine clearance according to the Cockcroft-Gault formula (mL/min/1.73m²). (NCT01079143)
Timeframe: Baseline (M3), M12

,,,
InterventionmL/min/1.73m² (Mean)
without imputationimputation by LOCF (36, 60, 39, 58)
Certican EMT-5.627.3
Certican EMT+8.6-11.9
Neoral EMT-3.8-4.9
Neoral EMT+1.55.1

[back to top]

Incidence (Number) of Participants With Graft Losses

If a participant underwent a graft nephrectomy, then the day of nephrectomy was considered as the day of graft loss. (NCT01079143)
Timeframe: M6 and M12 post transplantation

,,,
InterventionParticipants (Number)
M6 - NoM6 - YesM12 - NoM12 - Yes
Certican EMT-600564
Certican EMT+360351
Neoral EMT-590590
Neoral EMT+390381

[back to top]

Type of Biopsy Proven Acute Rejection (BPAR)

"A biopsy proven acute rejection (BPAR) is defined as a biopsy graded IA, IB, IIA, IIB or III as per 2005/2007 Banff histological classification system.~Biopsy graded IA: Significant interstitial infiltration (> 25% of parenchyma) and foci of moderate tubulitis (> 4 mononuclear cells/tubular cross section or group of 10 tubular cells).~Biopsy grade IB: Significant interstitial infiltration (> 25% of parenchyma) and foci of severe tubulitis (> 10 mononuclear cells/tubular cross section or group of 10 tubular cells).~Biopsy grade IIA: Mild to moderate intimal arteritis. Biopsy graded IIB: Severe intimal arteritis comprising > 25% of the lumenal area.~Biopsy graded III: Transmural (full vessel wall thickness) arteritis and/or arterial fibrinoid change and necrosis of medial smooth muscle cells (with accompanying lymphocytic inflammation)." (NCT01079143)
Timeframe: M6 and M12 post transplantation

,,,
InterventionParticipants (Number)
Cellular AR - NoCellular AR - Yes
Certican EMT-4614
Certican EMT+306
Neoral EMT-581
Neoral EMT+381

[back to top]

Treatment Failures

A treatment failure is defined as biopsy proven acute rejection (BPAR), a graft loss, a death or a loss to follow up. It was assessed between randomization and 6 and 12 months post-transplantation. (NCT01079143)
Timeframe: M6 and M12 post transplantation

,,,
InterventionNumber of Participants (Number)
M6: Treatment Failure- NoM6: Treatment Failure- YesM12: Treatment Failure- NoM12: Treatment Failure- YesM6: BPAR - NoM6: BPAR -YesM12: BPAR - NoM12: BPAR - YesM6: Graft Loss - NoM6: Graft Loss - YesM12: Graft Loss - NoM12: Graft Loss - YesM6: Death - NoM6: Death - YesM12: Death - NoM12: Death - YesM6: Loss to follow-up - NoM6: Loss to follow-up - YesM12: Loss to follow-up - NoM12: Loss to follow-up - Yes
Certican EMT-52842185284317600564600600600591
Certican EMT+333297333297360351360360360360
Neoral EMT-590563590563590590590590590590
Neoral EMT+390363390372390381390390390390

[back to top]

Change in Percentage of Interstitial Fibrosis (IF) by Numerical Quantification

Percentage of IF measured by numerical quantification. The IF percentage was transposed in grade according to the following rule: grade 0 for an IF % ≤5%, grade I for an IF % ranging from >5% to < 25%, grade II for an IF % ranging from 25 to 50%, grade III for an IF % >50%. Interstitial graft fibrosis has been identified as the primary cause of graft loss following death with a functional graft [3-5]. (NCT01079143)
Timeframe: M3 and M12 post transplantation

,,,
InterventionPercentage of IF (Mean)
Percentage of IF at M3 (n=26,43,32,53)Percentage of IF at M12 (n=24,42,32,53)Change in Percentage of IF (n=24,42,32,53)
Certican EMT-15.920.94.9
Certican EMT+22.827.65.1
Neoral EMT-17.820.42.7
Neoral EMT+23.427.43.9

[back to top]

Severity of BPAR

"A biopsy proven acute rejection (BPAR) is defined as a biopsy graded IA, IB, IIA, IIB or III as per 2005/2007 Banff histological classification system.~Biopsy graded IA: Significant interstitial infiltration (> 25% of parenchyma) and foci of moderate tubulitis (> 4 mononuclear cells/tubular cross section or group of 10 tubular cells).~Biopsy grade IB: Significant interstitial infiltration (> 25% of parenchyma) and foci of severe tubulitis (> 10 mononuclear cells/tubular cross section or group of 10 tubular cells).~Biopsy grade IIA: Mild to moderate intimal arteritis. Biopsy graded IIB: Severe intimal arteritis comprising > 25% of the lumenal area.~Biopsy graded III: Transmural (full vessel wall thickness) arteritis and/or arterial fibrinoid change and necrosis of medial smooth muscle cells (with accompanying lymphocytic inflammation)." (NCT01079143)
Timeframe: M6 and M12 post transplantation

,,,
InterventionParticipants (Number)
M6: Banff type Grade IAM6: Banff type Grade IBM6: Banff type Grade IIAM6: Banff type Grade IIBM6: Banff type Grade IIIM12: Banff type Grade IAM12: Banff type Grade IBM12: Banff type Grade IIAM12: Banff type Grade IIBM12: Banff type Grade III
Certican EMT-2401075110
Certican EMT+0010022100
Neoral EMT-0000000100
Neoral EMT+0000010000

[back to top]

Risk Factors of IF/TA Progression

"Composite factors regarding fibrosis progression at 12 months using a logistic regression model; the parameters initially considered concerned the demographic characteristics of both recipient and donor, transplantation characteristics, hypertension, diabetes, study treatments, immunosuppression, EMT, IF/TA, function at M3, the onset of acute rejection, the presence of anti-donor antibodies at M12, infections and BK virus viremia.~Arteriolar hyaline thickening is thickening of the walls of arterioles by the deposition of homogeneous pink hyaline material.~BPAR is biopsy proven acute rejection. TEM progression is the increase ≥ 1 of TEM score between Month 3 and Month 12" (NCT01079143)
Timeframe: M12 post transplantation

,
InterventionParticipants (Number)
Donor Sex - MaleDonor Sex - FemaleDonor Age - <=50 yearsDonor Age - >50 yearsExpanded Criteria donor: NOExpanded Criteria Donor: YesDelayed graft function: NoDelayed graft function: YesCC at M3 <50 mL/min/1.73m^2CC at M3 >=50 mL/min/1.73m^2 (n=67, 85)Mesangial matrix increase at M3 - 0 (n=64, 85)Mesangial matrix (mm) increase at M3: 1 (n=64, 85)Mesangial matrix increase at M3: 2 (n=64, 85)Interstitial fibrosis (ci) at M3: 0 (n=66, 85)Interstitial fibrosis (ci) at M3: 1 (n=66, 85)Interstitial fibrosis (ci) at M3: 2 (n=66, 85)Arteriolar hyaline thickening (ah) at M3: 0Arteriolar hyaline thickening (ah) at M3: 1Arteriolar hyaline thickening (ah) at M3: 2Arteriolar hyaline thickening (ah) at M3: 3BPAR - NoBPAR - YesTEM Progression fron M3-M12: No (n=67, 83)TEM Progression fron M3-M12: Yes (n=67, 83)TEM Progression fron M3-M12: Missing (n=67, 83)
No- No Fibrosis Progression4838483863237610355084105527356218179757263
Yes- Fibrosis Progression452228394027521540275923521403615106551226410

[back to top]

Plasma Concentration of Lenalidomide

(NCT01088048)
Timeframe: Predose, 1.5 hours postdose at Week 1 and predose at Week 5

Interventionng/mL (Mean)
Pre-dose (Week 1)1.5 hr post-dose (Week 1)Pre-dose (Week 5)
Lenalidomide (Cohort 7a, 7b, 7c)NA51.6NA

[back to top]

Plasma Concentration of Everolimus

(NCT01088048)
Timeframe: Predose, 1.5 hours postdose at Weeks 0 and 4

Interventionng/mL (Mean)
Pre-dose (Week 0)1.5 hr post-dose (Week 0)Pre-dose (Week 4)1.5 hr post-dose (Week 4)
Everolimus (Cohort 5a)NA93.03.056.3

[back to top]

Plasma Concentration of Bendamustine

(NCT01088048)
Timeframe: Predose, 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 2.0, 3.0, 4.0, 5.0, 6.0 hours postdose at Week 0

Interventionng/mL (Mean)
Pre-dose (Week 0)0.25 hr post-dose (Week 0)0.5 hr post-dose (Week 0)0.75 hr post-dose (Week 0)1.0 hr post-dose (Week 0)1.25 hr post-dose (Week 0)1.5 hr post-dose (Week 0)2.0 hr post-dose (Week 0)3.0 hr post-dose (Week 0)4.0 hr post-dose (Week 0)5.0 hr post-dose (Week 0)6.0 hr post-dose (Week 0)
Bendamustine (Cohorts 1b, 2b, 3a, 3b, 3f, 3g, 4b, 5c)NA3484.14694.73433.22847.21916.41211.2514.0463.478.515.34.4

[back to top]

Toxicity of Administration of IDELA

Percentage of participants experiencing toxicities of administration of IDELA were measured according to the Common Terminology Criteria for Adverse Events v4.02 (NCT01088048)
Timeframe: First dose date up to 5 years

Interventionpercentage of participants (Number)
Idelalisib + Rituximab100.0
Idelalisib + Bendamustine100.0
Idelalisib + Everolimus100.0
Idelalisib + Bortezomib83.33
Idelalisib + Rituximab + Bendamustine100.0
Idelalisib + Ofatumumab100.0
Idelalisib + Fludarabine100.0
Idelalisib + Chlorambucil100.0
Idelalisib + Rituximab + Chlorambucil100.0
Idelalisib + Rituximab + Lenalidomide100.0

[back to top]

Time to Response

Time to response (TTR) was defined as the interval from the start of study drug to the first documentation of CR or PR. (NCT01088048)
Timeframe: Up to 5 years

Interventionmonths (Median)
Idelalisib + Rituximab1.9
Idelalisib + Bendamustine1.9
Idelalisib + Everolimus1.9
Idelalisib + Bortezomib1.9
Idelalisib + Rituximab + Bendamustine1.9
Idelalisib + Ofatumumab1.9
Idelalisib + Fludarabine1.9
Idelalisib + Chlorambucil1.9
Idelalisib + Rituximab + Chlorambucil1.9
Idelalisib + Rituximab + Lenalidomide3.0

[back to top]

Progression-free Survival

"Progression free survival (PFS) was defined as the interval from the start of study drug to the earlier of the first documentation of disease progression or death from any cause.~The response definitions were based on the following standard criteria established for each indication:~CLL: International Workshop on chronic lymphocytic leukemia (IWCLL), 2008~iNHL & MCL: Cheson, 2007" (NCT01088048)
Timeframe: Up to 5 years

Interventionmonths (Median)
Idelalisib + RituximabNA
Idelalisib + BendamustineNA
Idelalisib + Everolimus4.3
Idelalisib + Bortezomib8.1
Idelalisib + Rituximab + BendamustineNA
Idelalisib + OfatumumabNA
Idelalisib + FludarabineNA
Idelalisib + ChlorambucilNA
Idelalisib + Rituximab + ChlorambucilNA
Idelalisib + Rituximab + LenalidomideNA

[back to top]

Overall Survival

Overall Survival (OS) was defined as the interval from the start of study drug to death from any cause. (NCT01088048)
Timeframe: Up to 5 years

Interventionmonths (Median)
Idelalisib + RituximabNA
Idelalisib + BendamustineNA
Idelalisib + EverolimusNA
Idelalisib + BortezomibNA
Idelalisib + Rituximab + BendamustineNA
Idelalisib + OfatumumabNA
Idelalisib + FludarabineNA
Idelalisib + ChlorambucilNA
Idelalisib + Rituximab + ChlorambucilNA
Idelalisib + Rituximab + LenalidomideNA

[back to top]

Overall Response Rate

"Overall Response Rate (ORR) was defined as the percentage of participants achieving a complete response (CR) or partial response (PR).~The response definitions were based on the following standard criteria established for each indication:~CLL: International Workshop on chronic lymphocytic leukemia (IWCLL),2008~iNHL & MCL: Cheson, 2007" (NCT01088048)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Idelalisib + Rituximab78.4
Idelalisib + Bendamustine84.3
Idelalisib + Everolimus44.4
Idelalisib + Bortezomib61.1
Idelalisib + Rituximab + Bendamustine81.8
Idelalisib + Ofatumumab71.4
Idelalisib + Fludarabine91.7
Idelalisib + Chlorambucil66.7
Idelalisib + Rituximab + Chlorambucil93.3
Idelalisib + Rituximab + Lenalidomide71.4

[back to top]

Duration of Response

Duration of response (DOR) was defined as the interval from the first documentation of CR or PR to the earlier of the first documentation of disease progression or death from any cause. (NCT01088048)
Timeframe: Up to 5 years

Interventionmonths (Median)
Idelalisib + RituximabNA
Idelalisib + BendamustineNA
Idelalisib + Everolimus5.6
Idelalisib + Bortezomib9.3
Idelalisib + Rituximab + BendamustineNA
Idelalisib + OfatumumabNA
Idelalisib + FludarabineNA
Idelalisib + ChlorambucilNA
Idelalisib + Rituximab + ChlorambucilNA
Idelalisib + Rituximab + LenalidomideNA

[back to top]

Duration of Exposure to IDELA

Duration of exposure to IDELA was summarized using descriptive statistics. (NCT01088048)
Timeframe: First dose date up to 12 months

Interventionmonths (Mean)
Idelalisib + Rituximab8.1
Idelalisib + Bendamustine7.6
Idelalisib + Everolimus4.2
Idelalisib + Bortezomib5.1
Idelalisib + Rituximab + Bendamustine8.0
Idelalisib + Ofatumumab8.3
Idelalisib + Fludarabine8.9
Idelalisib + Chlorambucil8.8
Idelalisib + Rituximab + Chlorambucil8.7
Idelalisib + Rituximab + Lenalidomide7.7

[back to top]

Plasma Concentration of IDELA (Cohort 6)

(NCT01088048)
Timeframe: Predose, 1.5 hours postdose at Weeks 0, 4, 12 and 24

Interventionng/mL (Mean)
Pre-dose (Week 0)1.5 hr post-dose (Week 0)Pre-dose (Week 4)1.5 hr post-dose (Week 4)Pre-dose (Week 12)1.5 hr post-dose (Week 12)Pre-dose (Week 24)1.5 hr post-dose (Week 24)
Idelalisib 150 mg (Cohort 6)0.01930.7530.81869.8677.91733.0346.81710.7

[back to top]

Plasma Concentration of IDELA (Cohort 7)

(NCT01088048)
Timeframe: Predose, 1.5 hours postdose at Weeks 0, 5 and 13

Interventionng/mL (Mean)
Pre-dose (Week 0)1.5 hr post-dose (Week 0)Pre-dose (Week 5)1.5 hr post-dose (Week 5)Pre-dose (Week 13)1.5 hr post-dose (Week 13)
Idelalisib 150 mg (Cohort 7)NA1603.120.71621.3354.8592.7

[back to top]

Sub-study: Plasma Concentration of IDELA (Cohorts 1-4)

(NCT01088048)
Timeframe: pre dose and 0.5, 1, 1.5, 2.0, 3.0, 4.0, and 6.0 hours post dose

,
Interventionng/mL (Mean)
Pre-dose0.5 hr post-dose1.0 hr post-dose1.5 hr post-dose2.0 hr post-dose3.0 hr post-dose4.0 hr post-dose6.0 hr post-dose
Idelalisib 100 mg (Cohort 1a, 1b)1.5437.61022.41264.71282.71001.0788.4523.7
Idelalisib 150 mg (Cohorts 2a, 2b, 3a, 3c, 3d, 3e, 3f, 3g, 4a, 4b)0.01222.11723.11599.21646.21238.5879.8489.3

[back to top]

Plasma Concentration of IDELA (Cohort 4)

(NCT01088048)
Timeframe: Predose at Week 0; predose, 0.5, 1.0, 1.5, 2.0, 3.0, 4.0, 6.0 hours postdose at Week 4; predose, 1.5 hours postdose at Week 12; and predose, 1.5 hours postdose at Week 24

Interventionng/mL (Mean)
Pre-dose (Week 0)Pre-dose (Week 4)0.5 hr post-dose (Week 4)1.0 hr post-dose (Week 4)1.5 hr post-dose (Week 4)2.0 hr post-dose (Week 4)3.0 hr post-dose (Week 4)4.0 hr post-dose (Week 4)6.0 hr post-dose (Week 4)Pre-dose (Week 12)1.5 hr post-dose (Week 12)Pre-dose (Week 24)1.5 hr post-dose (Week 24)
Idelalisib 150 mg (Cohort 4)0.00.01119.5994.51758.2737.0605.0547.0524.0401.92018.2752.52251.1

[back to top]

Plasma Concentration of IDELA (Cohort 1, Cohorts 2 and 3, Cohort 5)

(NCT01088048)
Timeframe: Predose, 0.5, 1.0, 1.5, 2.0, 3.0, 4.0, 6.0 hours postdose at Week 0; predose, 1.5 hours postdose at Weeks 4, 12, and 24

,,
Interventionng/mL (Mean)
Pre-dose (Week 0)0.5 hr post-dose (Week 0)1.0 hr post-dose (Week 0)1.5 hr post-dose (Week 0)2.0 hr post-dose (Week 0)3.0 hr post-dose (Week 0)4.0 hr post-dose (Week 0)6.0 hr post-dose (Week 0)Pre-dose (Week 4)1.5 hr post-dose (Week 4)Pre-dose (Week 12)1.5 hr post-dose (Week 12)Pre-dose (Week 24)1.5 hr post-dose (Week 24)
Idelalisib 100 mg (Cohort 1)0.4437.61022.41434.01282.71001.0788.4523.7416.51297.2361.91309.5369.91061.6
Idelalisib 150 mg (Cohort 5)0.01380.01600.01564.71400.01170.0795.0517.0408.01877.9433.91426.8549.1885.6
Idelalisib 150 mg (Cohorts 2 and 3)68.81231.41789.32017.31732.81296.1910.0486.0364.11808.1351.41883.0419.71840.1

[back to top]

Percentage of Participants With Stable Disease (SD)

Percentage of participants with stable disease during treatment is defined as stable disease [SD] by RECIST 1.1 criteria as calculated in each treatment arm. Stable Disease (SD) is defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. (NCT01108445)
Timeframe: Baseline to 36 months

Interventionpercentage of participants (Number)
RAD00159.6
Sunitinib64.7

[back to top]

Percentage of Participants With Adverse Events

To assess toxicities associated with everolimus or sunitinib using NCI CTC version 4.0 criteria (NCT01108445)
Timeframe: 24 months

Interventionpercentage of participants (Number)
RAD00163.2
Sunitinib80.4

[back to top]

Overall Response Rate

Defined as complete response [CR] and partial response [PR] by RECIST 1.1 criteria in each treatment arm.Overall Response Rate (ORR) = CR + PR. Complete Response (CR) is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial Response (PR) is at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. (NCT01108445)
Timeframe: 24 months

Interventionpercentage of participants (Number)
RAD0018.8
Sunitinib17.6

[back to top]

Median OS

To compare the median OS in each treatment arm. (NCT01108445)
Timeframe: Up to 40 months

Interventionmonths (Median)
RAD00113.2
Sunitinib31.5

[back to top]

12 Week Clinical Benefit Rate as Percentage

Rate of complete or partial response or stable disease by the RECIST 1.1 criteria lasting ≥ 12 weeks prior to progression. Benefit rate is defined as complete response [CR] and partial response [PR] and stable disease [SD] by RECIST 1.1 criteria in each treatment arm. Benefit rate = CR + PR + SD. Complete Response (CR) is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial Response (PR) is at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. (NCT01108445)
Timeframe: Baseline to 36 months

Interventionpercentage of particpants (Number)
RAD00124.6
Sunitinib41.2

[back to top]

Median Duration of Response (CR, PR, and SD)

To compare the median duration of response (CR, PR, and SD) in each treatment arm. According to RECIST 1.1, Complete Response (CR) is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial Response (PR) is at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. (NCT01108445)
Timeframe: 24 months

Interventionmonths (Median)
RAD0013.9
Sunitinib8.3

[back to top]

Change in Quality-of-life

To compare change in quality-of-life, as measured by the FACT-KSI scale at baseline and end of treatment per subject in each treatment arm. Functional Assessment of Cancer Therapy Kidney Symptom Index. FKSI is a questionnaire for FACT-Kidney Symptom Index used to assess QoL/participant-reported outcomes for participants diagnosed with renal cell cancer. The FKSI contained 15 questions each ranging from 0 (not at all) to 4 (very much) so that FKSI ranged between 0-60 where higher scores reflects better functioning and fewer symptoms. (NCT01108445)
Timeframe: baseline, up to 40 months

Interventionunits on a scale (Mean)
RAD001-6.6
Sunitinib-6.4

[back to top]

Change in Quality-of-life

To compare change in quality-of-life, as measured by the FACT-KSI scale at baseline and cycle 6 day1 per subject in each treatment arm. Functional Assessment of Cancer Therapy Kidney Symptom Index. FKSI is a questionnaire for FACT-Kidney Symptom Index used to assess QoL/participant-reported outcomes for participants diagnosed with renal cell cancer. The FKSI contained 15 questions each ranging from 0 (not at all) to 4 (very much) so that FKSI ranged between 0-60 where higher scores reflects better functioning and fewer symptoms. (NCT01108445)
Timeframe: baseline, cycle 6 day 1

Interventionunits on a scale (Mean)
RAD001-4.4
Sunitinib-2.1

[back to top]

Progression Free Survival Rates

6-, 12-, and 24-month rates of PFS in each arm will be compared for each treatment arm. Progressive disease is defined as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progression). (NCT01108445)
Timeframe: 6, 12 and 24 months

,
Interventionpercentage of participants (Number)
6 Months12 Months24 Months
RAD00140.317.09.3
Sunitinib55.037.722.8

[back to top]

PFS Expressed in Months

Progression-free survival (PFS) expressed in months as compared to an historic control (interferon-treated clear cell RCC control arm from the sunitinib phase III study). Progressive disease is defined as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progression). (NCT01108445)
Timeframe: 24 months

InterventionMonths (Median)
RAD0015.6
Sunitinib8.3

[back to top]

Best Tumor Shrinkage as a Percentile in Each Arm

To compare the best tumor shrinkage as a percentile in each treatment arm. The percentile change at each follow up visit is calculated by measuring the percentage change in the Sum of lesion measurement from baseline. The best tumor shrinkage is lowest percentile change. A decrease is indicated by a negative percentage. (NCT01108445)
Timeframe: 24 months

Interventionpercentile decrease (Median)
RAD001-2.1
Sunitinib-10.7

[back to top]

Time-to-new Metastatic Disease in Each Treatment Arm

To compare the time-to-new metastatic disease in each treatment arm, defined from the date of first study agent administration to the onset of a new evaluable site of disease, excluding the primary site and all sites documented at baseline (NCT01108445)
Timeframe: 36 months

Interventionmonths (Median)
RAD00119.4
Sunitinib36

[back to top]

Anti-tumor Activity as Measured by Median Progression Free Survival Time

The primary objective will be to compare the anti-tumor activity of everolimus and sunitinib in subjects with mRCC with non-clear cell pathology, as measured by progression-free survival (PFS) following treatment initiation according to RECIST 1.1 criteria. Progressive disease is defined as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progression). (NCT01108445)
Timeframe: 24 Months

InterventionMonths (Median)
RAD0015.6
Sunitinib8.3

[back to top]

Overall Survival Rates

To compare overall survival (OS) rates at 6, 12, 24, and 36 months and over time in each treatment arm. (NCT01108445)
Timeframe: 6, 12, 24, 36 months

,
Interventionpercentage probability (Number)
6 months12 months24 months36 months
RAD00183.557.740.835.7
Sunitinib85.474.751.346.2

[back to top]

Change in Quality-of-life

To compare change in quality-of-life, as measured by the FACT-KSI scale at baseline and cycle 3 day 1 per subject in each treatment arm. Functional Assessment of Cancer Therapy Kidney Symptom Index. FKSI is a questionnaire for FACT-Kidney Symptom Index used to assess QoL/participant-reported outcomes for participants diagnosed with renal cell cancer. The FKSI contained 15 questions each ranging from 0 (not at all) to 4 (very much) so that FKSI ranged between 0-60 where higher scores reflects better functioning and fewer symptoms. (NCT01108445)
Timeframe: baseline, cycle 3 day 1

Interventionunits on a scale (Mean)
RAD001-3.5
Sunitinib-0.9

[back to top]

Site of Progression: Distant

Number of patients with distant progression (NCT01111058)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Everolimus (RAD001)2
Placebo2

[back to top]

Site of Progression: Local-regional

Number of patients with local-regional progression (NCT01111058)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Everolimus (RAD001)2
Placebo5

[back to top]

Site of Progression: Unknown

Number of patients with unknown site of progression (NCT01111058)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Everolimus (RAD001)1
Placebo1

[back to top]

2 Year Progression Free Survival Rate

Time to disease progression or death from any cause--2 year rate (NCT01111058)
Timeframe: 2 years

Interventionpercentage of patients (Number)
Everolimus (RAD001)56.1
Placebo55.9

[back to top]

Number of Participants With Toxicity

Adverse event rate, any type, any grade regardless of attribution (NCT01111058)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Everolimus (RAD001)25
Placebo16

[back to top]

Estimated Glomerular Filtration Rate (eGFR)

Assessment of renal function by comparing change from randomization to Month 12 in eGFR (MDRD4) between treatment arms (Full analysis set). Renal function was assessed by estimated Glomerular Filtration Rate (eGFR) using the Modification of Diet in Renal Disease (MDRD) formula. MDRD formula: GFR [mL/min/1.73m˄2] = 186.3*(C˄-1.154)*(A˄-0.203)*G*R. DEFINITIONS: C = serum concentration of creatinine [mg/dL]; A = age [years]; G = 0.742 when gender is female, otherwise G = 1; R = 1.21 when race is black, otherwise R = 1 (NCT01114529)
Timeframe: Month 12

InterventionmL/min/1.73m^2 (Mean)
Everolimus64.1
Standard CNI (Tac)61.5
Standard CNI (CsA)58.4

[back to top]

Incidence of Composite Efficacy Endpoint for Each Arm at Month 12 and Month 24

Efficacy failure rate used the composite endpoint of: (1) treated biopsy-proven acute rejection (BPAR)*, (2) graft loss**, or (3) death . *A treated BPAR was defined as a biopsy graded IA, IB, IIA, IIB, or III and which was treated with anti-rejection therapy. **Graft loss is defined as when the allograft was presumed lost on the day the participant started dialysis and was not able to subsequently be removed from dialysis or re-transplanted. (NCT01114529)
Timeframe: at 12 months and month 24 post-transplantation

,,
InterventionNumber of incidence (Number)
Month 12Month 24
Everolimus2127
Standard CNI (CsA)88
Standard CNI (Tac)48

[back to top]

Comparison of Incidence Rates of Efficacy Endpoints Between Treatment Arms (Full Analysis Set - 24 Month Analysis)

(treated BPAR ≥ IB, graft loss or death)A comparison of the incidence rates for the individual components of the composite efficacy endpoint between treatment arms (NCT01114529)
Timeframe: at 24 months post-transplantation

,,
InterventionNumber of incidence (Number)
Composite failure: tBPAR>=IB, graft loss, deathComposite tBPAR>=IB, graft loss, death, loss f/uComposite of graft loss or DeathtBPAR>=IBGraft lossDeathSuspected Acute rejectionSubclinical Acute rejectionAcute rejection (AR)Treated Acute rejection (tAR)biopsy proven acute rejection (BPAR)treated biopsy proven acute rejection (tBPAR)tBPAR=IAtBPAR=IBtBPAR=IIAtBPAR=IIBtBPAR=IIIAntibody tBPARAntibody mediated rejection (AMR)
Everolimus2735101848610524037321614320216
Standard CNI (CsA)8154524190151413138510013
Standard CNI (Tac)8146325270149873300014

[back to top]

Change in Left Ventricular Mass Index (LVMi) From Randomization to Month 12 and Month 24

Evolution of left ventricular mass and hypertrophy were evaluated by left ventricular mass index (LVMi) assessed by echocardiography. LVMi is derived using a standard formula from dimensional measurements on the echocardiogram. Analysis of covariance was applied with treatment, center (as a random effect), and donor type as factors and LVMi at Randomization as covariate. (NCT01114529)
Timeframe: Randomization, Month 12 and Month 24

,,
Interventiong/m^2.7 (Mean)
RandomizationMonth 12Month 24
Everolimus50.3049.9546.66
Standard CNI (CsA)51.1350.9647.91
Standard CNI (Tac)51.0848.9845.63

[back to top]

Percentage of Participants Achieving Complete Response (CR) or Partial Response (PR) [Response Rate (RR)]

Response was determined using Response Evaluation Criteria In Solid Tumors (RECIST v1.1) criteria. CR was defined as the disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeters (mm) and normalization of tumor marker level of non-target lesions; PR was defined as at least a 30% decrease in sum of longest diameter of target lesions. (NCT01115803)
Timeframe: Baseline to disease progression or death or up to 6 cycles of 28 days

Interventionpercentage of participants (Number)
Arm A - 50 mg LY2584702 QD + 150 mg Erlotinib QD0
Arm A - 50 mg LY2584702 (BID) + 150 mg Erlotinib QD0
Arm A - 100 mg LY2584702 BID + 150 mg Erlotinib QD0
Arm A - 75 mg LY2584702 BID + 150 mg Erlotinib QD0
Arm B - 50 mg LY2584702 QD + 10 mg Everolimus QD0
Arm B - 100 mg LY2584702 QD + 10 mg Everolimus QD0
Arm B - 50 mg LY2584702 BID + 10 mg Everolimus QD0

[back to top]

Number of Participants With Complete Response (CR), Partial Response (PR) or Stable Disease (SD) [Best Overall Response (BOR) (CR+PR+SD)]

BOR was determined using Response Evaluation Criteria In Solid Tumors (RECIST v1.1) criteria. CR was defined as the disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeters (mm) and normalization of tumor marker level of non-target lesions; PR was defined as at least a 30% decrease in sum of longest diameter (LD) of target lesions; Progressive Disease (PD) was defined as at least 20% increase in sum of LD of target lesions and minimum 5 mm increase over nadir. SD was defined as small changes that did not meet above criteria. (NCT01115803)
Timeframe: Baseline up to 112 Days

InterventionParticipants (Count of Participants)
Arm A - 50 mg LY2584702 QD + 150 mg Erlotinib QD1
Arm A - 50 mg LY2584702 BID + 150 mg Erlotinib QD1
Arm A - 100 mg LY2584702 BID + 150 mg Erlotinib QD1
Arm A - 75 mg LY2584702 BID + 150 mg Erlotinib QD1
Arm B - 50 mg LY2584702 QD + 10 mg Everolimus QD0
Arm B - 100 mg LY2584702 QD + 10 mg Everolimus QD2
Arm B - 50 mg LY2584702 BID + 10 mg Everolimus QD2

[back to top]

Number of Participants Who Died Due to Progressive Disease Within 30 Days of Study Drug Discontinuation

(NCT01115803)
Timeframe: Within 30 days of study drug discontinuation

InterventionParticipants (Count of Participants)
Arm A - 50 mg LY2584702 QD + 150 mg Erlotinib QD0
Arm A - 50 mg LY2584702 BID + 150 mg Erlotinib QD0
Arm A - 100 mg LY2584702 BID + 150 mg Erlotinib QD0
Arm A - 75 mg LY2584702 BID + 150 mg Erlotinib QD0
Arm B - 50 mg LY2584702 QD + 10 mg Everolimus QD1
Arm B - 100 mg LY2584702 QD + 10 mg Everolimus QD0
Arm B - 50 mg LY2584702 BID + 10 mg Everolimus QD0

[back to top]

Pharmacokinetics, Maximum Observed Plasma Concentration (Cmax) of LY2584702

(NCT01115803)
Timeframe: Cycle 1 Day 1 (C1 D1): predose, 0.5, 1, 2, 3, 5, 8 hours postdose and Cycle 1 Day 8 (C1 D8): predose, 0.5, 1, 2, 3, 5, and 8 hours postdose of 28-day cycle

,,,,,,
Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
C1 D1, single doseC1 D8, steady state
Arm A - 100 mg LY2584702 BID + 150 mg Erlotinib QD2421.642709.61
Arm A - 50 mg LY2584702 BID + 150 mg Erlotinib QD999.541636.71
Arm A - 50 mg LY2584702 QD + 150 mg Erlotinib QD918.241125.46
Arm A - 75 mg LY2584702 BID + 150 mg Erlotinib QD1768.591967.04
Arm B - 100 mg LY2584702 QD + 10 mg Everolimus QD2286.222260.71
Arm B - 50 mg LY2584702 BID + 10 mg Everolimus QD1569.242109.62
Arm B - 50 mg LY2584702 QD + 10 mg Everolimus QD1192.461169.95

[back to top]

Pharmacokinetics, Area Under the Concentration Time Curve (AUC)

AUC from time 0 to 8 hours (AUC0-8) and AUC from time 0 to infinity (AUC0-∞). (NCT01115803)
Timeframe: Cycle 1 Days 1 (C1 D1) and Cycle 1 Day 8 (C1 D8) of 28-day cycle

,,,,,,
Interventionnanograms*hours per milliliter (ng*h/mL) (Geometric Mean)
AUC0-8, D1, single doseAUC0-8, D8, steady stateAUC0-∞, D1, single dose
Arm A - 100 mg LY2584702 BID + 150 mg Erlotinib QD8610.5415225.0820813.83
Arm A - 50 mg LY2584702 BID + 150 mg Erlotinib QD4308.987948.437627.50
Arm A - 50 mg LY2584702 QD + 150 mg Erlotinib QD4436.605395.968699.54
Arm A - 75 mg LY2584702 BID + 150 mg Erlotinib QD8091.9313959.7416254.07
Arm B - 100 mg LY2584702 QD + 10 mg Everolimus QD11410.1811327.3322343.07
Arm B - 50 mg LY2584702 BID + 10 mg Everolimus QD6097.4410527.688764.13
Arm B - 50 mg LY2584702 QD + 10 mg Everolimus QD5699.8565460.8011386.03

[back to top]

Clinically Significant Effects (Number of Participants With Adverse Events)

Clinically significant events were defined as serious adverse events (SAEs) and other non-SAEs regardless of causality. A summary of serious and other non SAEs regardless of causality is located in the Reported Adverse Event module. (NCT01115803)
Timeframe: Baseline up to 7 months

,,,,,,
InterventionParticipants (Count of Participants)
SAEsNon-SAEs
Arm A - 100 mg LY2584702 BID + 150 mg Erlotinib QD23
Arm A - 50 mg LY2584702 BID + 150 mg Erlotinib QD25
Arm A - 50 mg LY2584702 QD + 150 mg Erlotinib QD14
Arm A - 75 mg LY2584702 BID + 150 mg Erlotinib QD35
Arm B - 100 mg LY2584702 QD + 10 mg Everolimus QD23
Arm B - 50 mg LY2584702 BID + 10 mg Everolimus QD26
Arm B - 50 mg LY2584702 QD + 10 mg Everolimus QD23

[back to top] [back to top]

PTEN Mutation Rate [Phase II]

The PTEN mutation rate is the percentage of patients with PTEN mutation identified in pre-therapy and post-therapy bone marrow samples per established methods. (NCT01125293)
Timeframe: Samples were collected pre-therapy before the beginning therapy (baseline) and post-therapy after finishing the 6th treatment cycle (168 days).

Interventionpercentage of participants (Number)
MYD88/L265P mutationCXCR4/C1013G mutation
Phase II805

[back to top]

Very-good-partial-response-or-better Rate [Phase II]

"Very-good-partial-response-or-better rate is the percentage of participants with complete response (CR) or very good partial response (VGPR) on to the combination of everolimus/bortezomib/rituximab. The combination regimen was received for up to 6 cycles.~CR:~Absence of serum monoclonal IgM protein by immunofixation~Normal serum IgM level~Complete resolution of extramedullary disease, i.e., lymphadenopathy and splenomegaly if present at baseline~Morphologically normal bone marrow aspirate and trephine biopsy~VGPR:~Monoclonal IgM protein is detectable ≥90% reduction in serum IgM level from baseline*~Complete resolution of extramedullary disease, i.e." (NCT01125293)
Timeframe: Up to 6 cycles (Day 168)

Interventionpercentage of participants (Number)
Phase II4.3

[back to top]

Treatment-Emergent Sensory Neuropathy Rate [Phase I]

Percentage of participants experiencing Grade 1 - Grade 3 treatment-emergent peripheral (sensory) neuropathy events based on CTCAEv3 as reported on case report forms for phase I participants. (NCT01125293)
Timeframe: Adverse events were assessed each cycle (ever 28 days) for 6 cycles then every 3 months on maintenance. Duration of therapy for the Phase I study up to 41 months.

Interventionpercentage of participants (Number)
Phase I Stage A Level 133
Phase I Stage A Level 233
Phase I Stage B Level 1100
Phase I Stage B Level 267
Phase I Dose Expansion50
Phase II48

[back to top]

Phase II Duration of Response (DoR)

"The DoR is defined as the elapsed time from date when the measurement criteria are first met for a complete or partial response (whichever status is recorded first) until the date of first observation of objective disease progression. Analysis of DoR will be as follows:~For responding patients who die without objective PD (including death from study disease), DoR will be censored at the date of the last objective progression-free disease assessment.~For responding patients not known to have died as of the data cut-off date and who do not have objective PD, DoR will be censored at the date of the last objective progression-free disease assessment.~For responding patients who receive subsequent systemic anticancer therapy (after discontinuation from the study chemotherapy) prior to objectively determined disease progression, DoR will be censored at the date of the last objective progression-free disease assessment prior to post discontinuation therapy." (NCT01125293)
Timeframe: Patients were assessed for disease every cycle while on treatment (168 days) and every three months while on maintenance therapy. In long-term follow-up, disease was monitored every 3 months. Study cohort median (range) follow-up was 15 (1 - 23) months.

Interventionmonths (Median)
Phase II14

[back to top]

Phase II Overall Response Rate

"Overall response rate is percentage of participants with complete (CR), very good partial (VGPR), partial (PR), or minimal response (MR) as best response during treatment.~CR:~No serum monoclonal IgM protein by immunofixation~Normal IgM level~Complete resolution of extramedullary disease, i.e., lymphadenopathy and splenomegaly if present at baseline~Morphologically normal bone marrow aspirate and trephine biopsy~VGPR:~Monoclonal IgM protein is detectable ≥90% reduction in IgM level from baseline~Complete resolution of extramedullary disease, i.e., lymphadenopathy/splenomegaly if present at baseline~No new signs/symptoms of active disease~PR:~Monoclonal IgM protein is detectable ≥50% but <90% reduction in IgM level from baseline~Reduction in extramedullary disease~No new signs/symptoms of active disease~MR:~Monoclonal IgM protein is detectable ≥25% but <50% reduction in IgM level from baseline~No new signs/symptoms of active disease" (NCT01125293)
Timeframe: Up to 6 cycles (Day 168)

Interventionpercentage of participants (Number)
Phase II87

[back to top]

2 Year Progression-free-survival [Phase II]

2-year progression free survival (PFS) is the probability of participants alive and progression free at 2 years from study entry estimated using Kaplan-Meier methods. PFS is defined as the time from enrollment to progressive disease (PD) or death. PD is defined as ≥25% increase in serum IgM level from lowest nadir and/or progression in clinical features attributable the disease. Patients alive without PD and not reporting use of non-protocol therapy prior to PD are censored at date of last objective progression-free disease assessment. Patients reporting use of non-protocol therapy prior to PD are censored at date of last objective progression-free disease assessment date prior to post-discontinuation therapy. (NCT01125293)
Timeframe: Patients were assessed for disease every cycle while on treatment (168 days) and every three months while on maintenance therapy. In long-term follow-up, disease was monitored every 3 months. Study cohort median (range) follow-up was 15 (1 - 23) months.

Interventionpercent probability of PFS (Number)
Phase II28

[back to top]

2-year Time-to-progression Probability (TTP) [Phase II]

2-year TTP probability is based on Kaplan-Meier methods. TTP is defined as time from enrollment to the date of progressive disease (PD). PD is defined as ≥25% increase in serum IgM level from lowest nadir and/or progression in clinical features attributable the disease. Patients without PD and not reporting use of non-protocol therapy prior to PD are censored at date of last objective progression-free disease assessment. Patients reporting use of non-protocol therapy prior to PD are censored at date of last objective progression-free disease assessment date prior to post-discontinuation therapy. (NCT01125293)
Timeframe: Patients were assessed for disease every cycle while on treatment (168 days) and every three months while on maintenance therapy. In long-term follow-up, disease was monitored every 3 months. Study cohort median (range) follow-up was 15 (1 - 23) months.

Interventionpercent probability of progression (Number)
Phase II28

[back to top]

Everolimus Dose Limiting Toxicity (DLT) [Phase I]

"The following qualify as dose limiting toxicities:~Grade 3 or greater non-hematologic toxicity, considered by the investigator to be related to study drugs.~Grade 4 hematologic toxicity defined as: thrombocytopenia with platelets <10,000 µ/L on more than one occasion despite transfusion support; grade 4 neutropenia occurring for more than 7 days and/or resulting in neutropenic fever with elevated temperature (defined as > 101 degrees F). Lymphopenia, a recognized side effect of bortezomib, is not considered a DLT.~Inability to receive Day 1 dose for Cycle 2 due to toxicity" (NCT01125293)
Timeframe: Assessed within the first cycle (28 days) of the study.

Interventionparticipants (Number)
Phase I Stage A Level 10
Phase I Stage A Level 20
Phase I Stage B Level 10
Phase I Stage B Level 20

[back to top]

Everolimus Maximum Tolerated Dose (MTD) Stage A [Phase I]

The MTD of Everolimus/Rituximab combination is determined by the number of patients who have dose limiting toxicity (DLT). See DLT primary outcome measure for definition. MTD is defined as the highest dose where <1/3 participants experience a DLT. If no DLT's are observed on Level 1, 3 subjects will be enrolled in the Level 2. If >1/3 subjects in a cohort have DLT, that dose will not be considered safe, with no escalation (MTD exceeded). If 1/3 subjects experience DLT, the cohort will be expanded to 6 subjects. If <2 subjects with a DLT among the expanded cohort of 6 evaluable subjects a cohort of 3 subjects will be enrolled in the next higher dose level. If 2 or more subjects with a DLT among the expanded cohort of 6 subjects, that dose level will not be considered safe, no escalation (MTD exceeded). If no DLT's are observed, then the MTD is not reached. The MTD was not reached with 0/3 participants experiencing a DLT in the highest dose level. Higher doses were not planned/tested. (NCT01125293)
Timeframe: Assessed within the first cycle (28 days) of the study.

Interventionmg (Number)
Phase I Stage A Level 1 and 210

[back to top]

Everolimus Maximum Tolerated Dose (MTD) Stage B [Phase I]

The MTD of Everolimus/Bortezomib/Rituximab combination is determined by the number of patients who have dose limiting toxicity (DLT). See DLT primary outcome measure for definition. MTD is defined as the highest dose where <1/3 participants experience a DLT. If no DLT's are observed on Level 1, 3 subjects will be enrolled in the Level 2. If >1/3 subjects in a cohort have DLT, that dose will not be considered safe, with no escalation (MTD exceeded). If 1/3 subjects experience DLT, the cohort will be expanded to 6 subjects. If <2 subjects with a DLT among the expanded cohort of 6 evaluable subjects a cohort of 3 subjects will be enrolled in the next higher dose level. If 2 or more subjects with a DLT among the expanded cohort of 6 subjects, that dose level will not be considered safe, no escalation (MTD exceeded).If no DLT's observed, then the MTD is not reached. The MTD was not reached with 0/3 participants experiencing a DLT in the highest level. Higher doses were not planned/tested. (NCT01125293)
Timeframe: Assessed within the first cycle (28 days) of the study.

Interventionmg (Number)
Phase I Stage B Level 1 & 2 and Dose Expansion10

[back to top]

Median Progression-free Survival Time

Progression-free survival time is defined as the time from first day of treatment to the first date of disease progression or death as a result of any cause. Progression was assessed every 2 cycles of treatment (6 weeks) by CT, CT/PET, or MRI. Progression is defined using RECIST 1.0, as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01127763)
Timeframe: up to 1 year

Interventionmonths (Median)
RAD001+Carboplatin3

[back to top]

Toxicity Profile-Non Hematological

Reported as percentage of patients who experienced grade 3 and higher non-hematological AEs related to the study drugs. (NCT01127763)
Timeframe: treatment period (up to 1 year) plus 30 days off treatment

Interventionpercentage of patients (Number)
NauseaVomitingDehydrationMucositisHypersensitivity
RAD001+Carboplatin (All Patients)44444

[back to top]

Toxicity Profile-Hematological

Reported as percentage of patients who experienced grade 3 and higher hematological adverse events (AEs) related to the study drugs. (NCT01127763)
Timeframe: treatment period (up to 1 year) plus 30 days off treatment

,,
Interventionpercentage of patients (Number)
AnemiaThrombocytopeniaLeukopeniaNeutropenia
RAD001+Carboplatin (All Patients)428412
RAD001+Carboplatin (AUC 4)51105
RAD001+Carboplatin (AUC 6 and 5)0711429

[back to top]

Clinical Benefit Rate (Complete Response, Partial Response, and Stable Disease That Lasts More Than 6 Months)

Clinical benefit rate is defined as the number of patients with complete response (CR), partial response (PR), or stable disease (SD) that lasts at least 6 months. Response was assessed every 2 cycles of treatment (6 weeks) by computed tomography (CT), CT/positron emission tomography (PET), or magnetic resonance imaging (MRI). Overall response evaluation is based on Response Evaluation Criteria In Solid Tumors 1.0 (RECIST 1.0). Per RECIST 1.0 for target lesions: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT01127763)
Timeframe: up to 1 year

Interventionpercentage of patients (Number)
RAD001+Carboplatin36

[back to top]

Tumor Responses

Change in tumor size (sum of longest diameters of target lesions) after two cycles of induction therapy, expressed as log of ratio of post-treatment to baseline measure. (NCT01133678)
Timeframe: Baseline and 2 months

Interventiondimensionless (log ratio) (Mean)
Everolimus-0.609
Placebo-0.796

[back to top]

Summary of Plasma Concentrations of Lenvatinib for Sparse Pharmacokinetic (PK) Sampling for Phase 1b and Phase 2

Blood samples were collected during the Randomization Phase. Most participants had 6 samples taken over 3 cycles of treatment (sparse sampling - 2 samples taken per cycle, one at predose and one at 2 to 8 hours postdose). Plasma concentrations of lenvatinib were measured and concentration data were summarized. The summary statistics at time points with one or more below the limit of quantitation (BLQ) values were calculated by assigning zero for each BLQ value. (NCT01136733)
Timeframe: Cycle 1 (Day 1), Cycle 2 (Day 1), Cycle 3 (Day 1)

Interventionng/mL (Geometric Mean)
Cycle 1, Day 1 (0 Hours)5.6
Cycle 1, Day 1 (2-8 Hours)197
Cycle 2, Day 1 (0 Hours)66.9
Cycle 2, Day 1 (2-8 Hours)237
Cycle 3, Day 1 (0 Hours)37.0
Cycle 3, Day 1 (2-8 Hours)180

[back to top]

Maximum Concentration of Everolimus (Cmax) in Blood When Administered Alone or in Combination With Lenvatinib

Cmax for everolimus was defined as the maximum observed concentration of everolimus in blood following administration of study treatment on Cycle 1 Day 15 and was obtained directly from the measured blood concentration-time curves. (NCT01136733)
Timeframe: Phase 2: Cycle 1 Day 15

Interventionng/mL (Mean)
Phase 2: 18 mg Lenvatinib + 5 mg Everolimus38
Phase 2: 10 mg Everolimus54

[back to top]

Phase 2: Objective Response Rate (ORR)

The ORR was defined as the percentage of participants who had the best overall response (BOR) of complete response (CR) or partial response (PR) as determined by the investigator, using RECIST 1.1 in the evaluation of MRI or CT scans of targeted lesions. Tumor assessments were performed every 8 weeks (or sooner if there was evidence of progressive disease). The BOR was defined as the best response recorded from the start of the study treatment until discontinuation from the study. CR was defined as disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) had to have reduction in short axis to less than 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. ORR = CR + PR was calculated with exact 95% CIs using the method of Clopper and Pearson. (NCT01136733)
Timeframe: Randomization (Cycle 1 Day 1) until first evidence of disease progression, assessed up to the data cutoff date (13 Jun 2014), or up to approximately 2 years and 3 months

InterventionPercentage of participants (Number)
Phase 2 (Arm A): 18 mg Lenvatinib Plus 5 mg Everolimus43.1
Phase 2 (Arm B): 24 mg Lenvatinib26.9
Phase 2 (Arm C): 10 mg Everolimus6.0

[back to top]

Phase 2: Overall Survival (OS)

OS was defined as the time (in months) from the date of randomization until date of death from any cause. Median survival time was calculated using K-M estimate for each treatment arm and presented with 2-sided 95% CIs. Participants who were lost to follow-up or alive at the data cutoff date (10 Dec 2014) were censored at the date the participants were last known to be alive. (NCT01136733)
Timeframe: Randomization (Cycle 1 Day 1) until date of death from any cause, assessed up to the data cutoff date (10 Dec 2014), up to approximately 2 years and 9 months

InterventionMonths (Median)
Phase 2 (Arm A): 18 mg Lenvatinib Plus 5 mg Everolimus25.5
Phase 2 (Arm B): 24 mg Lenvatinib19.1
Phase 2 (Arm C): 10 mg Everolimus15.4

[back to top]

Phase 1b: Number of Participants With Dose-limiting Toxicity (DLT)

A DLT was defined as either a treatment-related failure to administer greater than or equal to (>=) 75% of the planned dosage of lenvatinib/everolimus or a specific National Cancer Institute Common Toxicity Criteria (NCI CTC) >= Grade 3 (severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self-care daily living activities) hematologic or nonhematologic toxicities considered to be possibly related to lenvatinib and/or everolimus therapy assessed during the first treatment cycle of each dose level. Higher grade indicates more severe toxicity. (NCT01136733)
Timeframe: First dose of study drug (Cycle 1 Day 1) to end of first 4 weeks of therapy (Cycle 1)

,,
InterventionParticipants (Number)
Grade 3 abdominal painGrade 2 fatigue with Grade 1 GI reflux & anorexiaGrade 3 nauseaGrade 2 stomatitis
Phase 1b (Cohort 1): 12 mg Lenvatinib Plus 5 mg Everolimus1000
Phase 1b (Cohort 2): 18 mg Lenvatinib Plus 5 mg Everolimus0100
Phase 1b (Cohort 3): 24 mg Lenvatinib Plus 5 mg Everolimus0011

[back to top]

Area Under the Blood Concentration-Time Curve From 0 to 24 Hours for Everolimus When Administered Alone or in Combination With Lenvatinib

Between 9 and 12 participants in each of the 3 treatment arms participated in an optional substudy where instead of the sparse sampling, 9 samples were to be taken over 1 single 24-hour period (i.e., intensive sampling) for full PK profiling. Blood samples were analyzed for study drug using standardized methods. PK parameters for everolimus were derived from everolimus concentration data using non-compartmental methods. Data were compared via descriptive statistics between single agent and combination therapy. (NCT01136733)
Timeframe: Phase 2: Cycle 1 Day 15 immediately predose, and 30 minutes, 1, 2, 3, 4, 8, 12 (optional), and 24 hours postdose (predose on Day 16)

Interventionng·hr/mL (Mean)
Phase 2: 18 mg Lenvatinib + 5 mg Everolimus378
Phase 2: 10 mg Everolimus463

[back to top] [back to top]

Area Under the Plasma Concentration-Time Curve From 0 to 24 Hours (AUC(0-24)) for Lenvatinib When Administered Alone or in Combination With Everolimus

Between 9 and 12 participants in each of the 3 treatment arms participated in an optional substudy where instead of the sparse sampling, 9 samples were to be taken over 1 single 24-hour period (i.e., intensive sampling) for full PK profiling. Blood samples were analyzed for study drug using standardized methods. PK parameters for lenvatinib were derived from lenvatinib concentration data using non-compartmental methods. Data were compared via descriptive statistics between single agent and combination therapy. (NCT01136733)
Timeframe: Phase 2: Cycle 1 Day 15 immediately predose, and 30 minutes, 1, 2, 3, 4, 8, 12 (optional), and 24 hours postdose (predose on Day 16)

Interventionng·hr/mL (Mean)
Phase 2: 18 mg Lenvatinib + 5 mg Everolimus3185
Phase 2: 24 mg Lenvatinib5252

[back to top]

Phase 2: Progression-Free Survival (PFS)

PFS was defined as the time (in months) from the date of first dose of study drug to the first documentation of disease progression or death, whichever occurred first. Kaplan-Meier (K-M) estimates were used to estimate median PFS, presented with 2-sided 95% confidence intervals (CIs). Tumor assessments were performed every 8 weeks (or sooner if there was evidence of progressive disease using computed tomography (CT) or magnetic resonance imaging (MRI) and scan acquisition techniques (including use or nonuse of intravenous (IV) contrast). Tumor response was determined at the site by the investigator and radiologist using Response Evaluation Criteria In Solid Tumors (RECIST) 1.1 in the evaluation of the tumor assessment scans. The date of objective disease progression was defined as the earliest date of radiological disease progression. Participants removed from therapy due to clinical progression with no radiologic confirmation were censored at their last radiologic assessment date. (NCT01136733)
Timeframe: Date of randomization into Phase 2 (Cycle 1 Day 1) to the date of first documentation of disease progression or death (whichever occurred first), assessed up to data cutoff date (13 Jun 2014), up to approximately 2 years and 3 months

InterventionMonths (Median)
Phase 2 (Arm A): 18 mg Lenvatinib Plus 5 mg Everolimus14.6
Phase 2 (Arm B): 24 mg Lenvatinib7.4
Phase 2 (Arm C): 10 mg Everolimus5.5

[back to top]

Summary of Blood Concentrations of Everolimus for Sparse PK Sampling for Phase 1b and Phase 2

Blood samples were collected during the Randomization Phase. Most participants had 6 samples taken over 3 cycles of treatment (sparse sampling - 2 samples taken per cycle, one at predose and one at 2 to 8 hours postdose). Whole blood concentrations of everolimus were measured and concentration data were summarized. The summary statistics at time points with one or more BLQ values were calculated by assigning zero for each BLQ value. (NCT01136733)
Timeframe: Cycle 1 (Day 1), Cycle 2 (Day 1), Cycle 3 (Day 1)

Interventionng/mL (Geometric Mean)
Cycle 1, Day 1 (0 Hours)0.0
Cycle 1, Day 1 (2-8 Hours)19.4
Cycle 2, Day 1 (0 Hours)10.0
Cycle 2, Day 1 (2-8 Hours)24.3
Cycle 3, Day 1 (0 Hours)6.8
Cycle 3, Day 1 (2-8 Hours)26.4

[back to top]

Clinical Benefit Rate (CBR)

The CBR was defined as the percentage of participants who had BOR of CR, PR, or durable SD (duration of SD was greater than or equal to 23 weeks) and was based on investigator review data using RECIST 1.1. The BOR was defined as the best response recorded from the start of study treatment until discontinuation from the study. There was no requirement for confirmatory measurement of PR or CR to deem either one the BOR. The 95% CI was constructed using the method of Clopper and Pearson. CBR = CR + PR + SD greater than or equal to 23 weeks. (NCT01136733)
Timeframe: Baseline (Randomization) to first evidence of disease progression, assessed up to the data cutoff date (13 Jun 2014), or up to approximately 2 years and 3 months

InterventionPercentage of participants (Number)
Phase 2 (Arm A): 18 mg Lenvatinib Plus 5 mg Everolimus68.6
Phase 2 (Arm B): 24 mg Lenvatinib65.4
Phase 2 (Arm C): 10 mg Everolimus42.0

[back to top]

Disease Control Rate (DCR)

The DCR was defined as the percentage of participants who had a BOR of CR or PR or SD (minimum duration from randomization to SD greater than or equal to 7 weeks). Assessments were performed every 8 weeks and were based on investigator review data using RECIST 1.1. The 95% CI was constructed using the method of Clopper and Pearson. DCR = CR + PR + SD greater than or equal to 7 weeks. (NCT01136733)
Timeframe: Baseline (Randomization) to first evidence of disease progression, assessed up to the data cutoff date (13 Jun 2014), or up to approximately 2 years and 3 months

InterventionPercentage of participants (Number)
Phase 2 (Arm A): 18 mg Lenvatinib Plus 5 mg Everolimus84.3
Phase 2 (Arm B): 24 mg Lenvatinib78.8
Phase 2 (Arm C): 10 mg Everolimus68.0

[back to top]

Time to Cmax (Tmax) for Everolimus When Administered Alone or in Combination With Lenvatinib

Tmax for everolimus was the amount of time taken after administration of study treatment on Cycle 1 Day 15 to reach the maximum concentration (Cmax) of everolimus in blood. (NCT01136733)
Timeframe: Phase 2: Cycle 1 Day 15

InterventionHours (Median)
Phase 2: 18 mg Lenvatinib + 5 mg Everolimus1.0
Phase 2: 10 mg Everolimus1.0

[back to top]

Durable Stable Disease (SD) Rate

The durable SD rate was defined as the percentage of participants whose BOR was SD and the duration of SD was greater than or equal to 23 weeks. The durable SD was based on investigator review data using RECIST 1.1. The 95% CI was constructed using the method of Clopper and Pearson. (NCT01136733)
Timeframe: Baseline (Randomization) to first evidence of disease progression, assessed up to the data cutoff date (13 Jun 2014), or up to approximately 2 years and 3 months

InterventionPercentage of participants (Number)
Phase 2 (Arm A): 18 mg Lenvatinib Plus 5 mg Everolimus25.5
Phase 2 (Arm B): 24 mg Lenvatinib38.5
Phase 2 (Arm C): 10 mg Everolimus36.0

[back to top]

Maximum Concentration (Cmax) of Lenvatinib in Plasma When Administered Alone or in Combination With Everolimus

Cmax for lenvatinib was defined as the maximum observed concentration of lenvatinib in plasma following administration of study treatment on Cycle 1 Day 15 and was obtained directly from the measured plasma concentration-time curves. (NCT01136733)
Timeframe: Phase 2: Cycle 1 Day 15

Interventionng/mL (Mean)
Phase 2: 18 mg Lenvatinib + 5 mg Everolimus327
Phase 2: 24 mg Lenvatinib403

[back to top]

Time to Cmax (Tmax) for Lenvatinib When Administered Alone or in Combination With Everolimus

Tmax for lenvatinib was the amount of time taken after administration of study treatment on Cycle 1 Day 15 to reach maximum concentration (Cmax) of lenvatinib in plasma. (NCT01136733)
Timeframe: Phase 2: Cycle 1 Day 15

InterventionHours (Median)
Phase 2: 18 mg Lenvatinib + 5 mg Everolimus2.0
Phase 2: 24 mg Lenvatinib4.0

[back to top]

Effect of RAD001 on Pharmacokinetics AUC(0-inf) of JI-101

Determine the mean percent change that RAD001 has on the peak concentration as determined by calculating the AUC (0-inf) of JI101 in the presence and absence of RAD001 (NCT01149434)
Timeframe: pre-dose and at 0.5, 1, 2, 4, 6, 8, 10, and 24 hours after dosing (Cycle 1 Day 8 for RAD001 + JI101 and Cycle 1 Day 15 for JI-101 alone

Interventionpercentage change (Mean)
Pharmacokinetic Arm Phase 195.3

[back to top]

Progression Free-Survival in the Ovarian Cancer Cohort

progression-free survival at 2 months. We define progression as using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), to detect a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01149434)
Timeframe: 2 months

Interventionpercentage of participants (Number)
Pharmacodynamic Arm Phase 271.4

[back to top]

Safety and Tolerability of JI-101

Number of patients experiencing a grade 2 or higher Adverse Event related to JI101 (NCT01149434)
Timeframe: 2 years

InterventionParticipants (Number)
Pharmacokinetic Arm Phase 14
Pharmacodynamic Arm Phase 212

[back to top]

Effect of JI 101 on Pharmacokinetics Area Under Curve (AUC) (0-inf) of RAD001

Determine the mean percent change that JI-101 has on the peak concentration as determined by calculating the AUC (0-inf) of RAD001 in the presence and absence of JI-101 (NCT01149434)
Timeframe: pre-dose and at 0.5, 1, 2, 4, 6, 8, 10, and 24 hours after dosing (Cycle 1 Day 1 for RAD001 alone and Cycle 1 Day 8 for RAD001 + JI-101

Interventionpercentage change (Mean)
Pharmacokinetic Arm165.6

[back to top]

Tumor Response in the Ovarian Cancer Cohort

Response Rate determined by the sum of patients achieving complete or partial response to JI-101 as defined by Response Evaluation Criteria in Solid Tumors (NCT01149434)
Timeframe: 2 years

InterventionParticipants (Number)
Pharmacodynamic Arm Phase 20

[back to top]

Tumor Response

Response Rate determined by the sum of patients achieving complete or partial response to JI-101 as defined by Response Evaluation Criteria in Solid Tumors. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR (NCT01149434)
Timeframe: 2 years

Interventionparticipants (Number)
Pharmacokinetic Arm Phase 10

[back to top]

Incidence Rate of Composite Efficacy Failure Defined as Treated Biopsy Proven Acute Rejection (tBPAR ), Graft Loss or Death

The number of participants who experienced composite efficacy failure was analyzed. Composite efficacy failure was defined as treated biopsy proven acute rejection (tBPAR), graft loss, or death. A BPAR was defined as an acute rejection confirmed by biopsy with a Rejection Activity Index (RAI) score ≥ 3. tBPAR was defined as a BPAR which was treated with anti-rejection therapy. The RAI is used to score liver biopsies with acute rejection and is composed of 3 categories (portal inflammation, bile duct inflammation damage, and venous endothelial inflammation) each scored on a scale of 0 (absent) to 3 (severe) by a trained pathologist. The total RAI score = the sum of the scores of the 3 categories and ranges from 0 to 9, with a higher score indicating greater rejection. The graft was presumed to be lost on the day the patient was newly listed for a liver graft, received a graft re-transplant, or died. (NCT01150097)
Timeframe: from months 24 to 36

InterventionParticipants (Number)
Everolimus + Reduced Tacrolimus2
Tacrolimus Elimination1
Tacrolimus Control3

[back to top]

Incidence Rate of Composite Efficacy Failure Defined as Graft Loss or Death

The number of participants who experienced graft loss or death was analyzed. The graft was presumed to be lost on the day the patient was newly listed for a liver graft, received a graft re-transplant, or died. (NCT01150097)
Timeframe: from months 36 - 48

InterventionParticipants (Number)
Everolimus + Reduced Tacrolimus0
Tacrolimus Elimination0

[back to top]

Change in Renal Function

Change in renal function was assessed by the estimated Glomerular Filtration Rate (eGFR) using the abbreviated (4 variables) Modification of Diet in Renal Disease (MDRD-4) formula which was developed by the MDRD Study Group and has been validated in patients with chronic kidney disease. The MDRD-4 formula used for the eGFR calculation is: eGFR (mL/min/1.73m^2) = 186.3*(C^-1.154)*(A^-0.203)*G*R, where C is the serum concentration of creatinine (mg/dL), A is age (years), G=0.742 when gender is female, otherwise G=1, R=1.21 when race is black, otherwise R=1. (NCT01150097)
Timeframe: from months 24 to 36

InterventionmL/min/1.73m^2 (Mean)
Everolimus + Reduced Tacrolimus-0.9
Tacrolimus Elimination2.5
Tacrolimus Control-3.3

[back to top]

Incidence Rate of Composite Efficacy Failure Defined as Graft Loss or Death

The number of participants who experienced graft loss or death was analyzed. The graft was presumed to be lost on the day the patient was newly listed for a liver graft, received a graft re-transplant, or died. (NCT01150097)
Timeframe: from months 24 to 36

InterventionParticipants (Number)
Everolimus + Reduced Tacrolimus2
Tacrolimus Elimination0
Tacrolimus Control1

[back to top]

Incidence Rate of tBPAR

The number of participants who had a tBPAR was analyzed. tBPAR was defined as a BPAR which was treated with anti-rejection therapy. The RAI is used to score liver biopsies with acute rejection and is composed of 3 categories (portal inflammation, bile duct inflammation damage, and venous endothelial inflammation) each scored on a scale of 0 (absent) to 3 (severe) by a trained pathologist. The total RAI score = the sum of the scores of the 3 categories and ranges from 0 to 9, with a higher score indicating greater rejection. (NCT01150097)
Timeframe: from months 24 - 36

InterventionParticipants (Number)
Everolimus + Reduced Tacrolimus0
Tacrolimus Elimination1
Tacrolimus Control2

[back to top]

Incidence Rate of Composite Efficacy Failure Defined as Treated Biopsy Proven Acute Rejection (tBPAR ), Graft Loss or Death

The number of participants who experienced composite efficacy failure was analyzed. Composite efficacy failure was defined as treated biopsy proven acute rejection (tBPAR), graft loss, or death. A BPAR was defined as an acute rejection confirmed by biopsy with a Rejection Activity Index (RAI) score ≥ 3. tBPAR was defined as a BPAR which was treated with anti-rejection therapy. The RAI is used to score liver biopsies with acute rejection and is composed of 3 categories (portal inflammation, bile duct inflammation damage, and venous endothelial inflammation) each scored on a scale of 0 (absent) to 3 (severe) by a trained pathologist. The total RAI score = the sum of the scores of the 3 categories and ranges from 0 to 9, with a higher score indicating greater rejection. The graft was presumed to be lost on the day the patient was newly listed for a liver graft, received a graft re-transplant, or died. (NCT01150097)
Timeframe: from months 36 to 48

InterventionParticipants (Number)
Everolimus + Reduced Tacrolimus1
Tacrolimus Elimination0

[back to top]

Progression-Free Survival (PFS)

Progression-free survival (PFS) is defined as the time between Day 1 Cycle 1 and date of first documented recurrence or death. Patients who do not exhibit progression while on trial will be censored at their last known assessment. Progression is defined per RECIST criteria as either 1) at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest (nadir) sum since the treatment started, or the appearance of one or more new lesions. Requires not only 20% increase, but absolute increase of a minimum of 5 mm over sum. OR 2) Appearance of one or more new lesions and/or unequivocal progression of existing non-target lesions. (NCT01154335)
Timeframe: 18 Months

Interventionmonths (Mean)
Dose Level 12.8
Dose Level 23.6
Dose Level 2a1.6

[back to top]

Response Rate

Response rate (RR) will be estimated as the proportion of patients exhibiting complete response or partial response out of all evaluable cases. Complete Response is defined per RECIST as disappearance of all target/non-target lesions and normalization of tumor markers. Partial Response is defined per RECIST as At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD. (NCT01154335)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Dose Level 10
Dose Level 20
Dose Level 2a0

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of the Combination of OSI-906 and Everolimus for the Treatment of Patients With Refractory Metastatic Colorectal Cancer.

The MTD of the drug combination will be determined as the highest dose at which ≤1 of 6 subjects experiences a Grade 3 or Grade 4 DLT according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) v4.0 (NCT01154335)
Timeframe: 18 Months

Interventionmilligrams (Number)
OSI-906 BID DoseEverolimus QD Dose
All Patients505

[back to top]

Overall Survival (OS)

Overall survival (OS) is defined as the time between Day 1 Cycle 1 to the date of death from any cause. Those remaining alive will be censored at their last known assessment or follow-up. (NCT01154335)
Timeframe: 18 months

Interventionweeks (Mean)
Dose Level 15.1
Dose Level 25.1
Dose Level 2a3.3

[back to top]

Common Terminology Criteria for Adverse Events (CTCAE) Events

Number of participants experiencing serious CTCAE events in the study. (NCT01158651)
Timeframe: approximately 48 weeks

Interventionparticipants (Number)
RAD001 (Everolimus) Active Therapy23

[back to top]

RAD001 Response Rate Based on 2D MRI Change From Baseline

Outcome is 2D MRI target tumor volume compared to baseline volume. Success criteria is volume less than 80% of baseline. (NCT01158651)
Timeframe: Approximately 48 weeks

Interventionparticipants (Number)
RAD001 (Everolimus) Active Therapy15

[back to top]

Change From Baseline in Cardiovascular Biomarkers, C-reactive Protein (CRP)

Blood samples were collected to analyze CRP. A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, month 6, month 24

,
Interventionmg/dl (Mean)
Month 6 (n=27,30)Month 24 (n=24,24)
Everolimus0.326-0.040
Tacrolimus0.5120.100

[back to top]

Percentage of Participants With Biopsy-proven Acute Rejection (BPAR), Graft Loss, Death and Lost to Follow up

The incidence of BPAR, graft loss, death and lost to follow-up events was calculated using relative frequency. (NCT01169701)
Timeframe: Month 24

,
InterventionPercentage of participants (Number)
BPARGraft lossDeathsLost to follow-up
Everolimus0.000.000.000.00
Tacrolimus0.000.000.003.13

[back to top]

Change From Baseline in the Cardiovascular Biomarker, Type 1 Procollagen Amino-terminal-propeptide (PINP)

Blood samples were collected to analyze PCR. A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, month 6, month 24

,
Interventionug/l (Mean)
Month 6 (n=27,30)Month 24 (n=24,24)
Everolimus-33.36-28.17
Tacrolimus-13.82-13.65

[back to top]

Renal Function as Measured by Estimated Glomerular Filtration Rate (eGFR)

Estimated GFR was caluclated using the modification of diet in renal disease (MDRD) formula. (NCT01169701)
Timeframe: Month 6, month 12, month 24

,
InterventionmL/min/1.73m^2 (Mean)
Month 6 (n=33,29)Month 12 (n=32,28)Month 24 (n=31,26)
Everolimus63.78161.22560.779
Tacrolimus55.64857.75757.727

[back to top]

Renal Function Measured by Serum Creatinine

Serum samples were collected to analyze serum creatinine. (NCT01169701)
Timeframe: Month 6, month 12, month 24

,
Interventionmg/dl (Mean)
Month 6 (n=33,29)Month 12 (n=32,28)Month 24 (n= 31,26)
Everolimus1.2341.2561.260
Tacrolimus1.2321.2311.217

[back to top]

Change From Baseline in the Cardiovascular Biomarker, N-terminal Pro-brain Natriuretic Peptide Fraction (NT-proBNP)

Blood samples were collected to analyze NT-proBNP. A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, month 6, month 24

,
Interventionpg/mL (Mean)
Month 6 (n=27,30)Month 24 (n=24,24)
Everolimus-79.10-193.3
Tacrolimus21.604-80.20

[back to top]

Change From Baseline in the Cardiovascular Biomarker, Glycosylated Hemoglobin (HbA1c)

Blood samples were collected to analyze HbA1c. A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, month 6, month 24

,
InterventionPercentage of HbA1c (Mean)
Month 6 (n=20,22)Month12 (n=22,20)
Everolimus0.1590.185
Tacrolimus0.0150.045

[back to top]

Change From Baseline in Cardiovascular Biomarkers: Troponin I and Collagen Type 1 C-telopeptide (ICTP)

Blood samples were collected to analyze Troponin I and collagen type 1 C-telopeptide (ICTP). A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, month 6, month 24

,
Interventionng/ml (Mean)
Troponin 1, Month 6 (n=27,30)Troponin 1, Month 24 (n=24,24)ICTP, Month 6 (n=27,30)ICTP, Month 24 (n=24,24)
Everolimus-0.006-0.007-0.195-0.125
Tacrolimus0.0000.0030.049-0.035

[back to top]

Change From Baseline in the Cardiovascular Biomarker, Myeloperoxidase (MPO)

Blood samples were collected to analyze MPO. A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, month 6, month 24

,
InterventionU/mL (Mean)
Month 6 (n=27,30)Month 24 (n=24,24)
Everolimus-0.093-0.642
Tacrolimus0.433-0.329

[back to top]

Renal Function as Measured by Creatinine Clearance

Creatinine clearance was calculated using the Cockroft-Gault formula. (NCT01169701)
Timeframe: Month 6, month 12, month 24

,
Interventionmg/min (Mean)
Month 6 (n=29,25)Month 12 (n=28,25)Month 24 (n=28,24)
Everolimus76.61873.36372.910
Tacrolimus64.84165.03766.933

[back to top]

Pulse Wave Velocity (PWV)

Utilizing the SphygmoCor Device, ECG leads placed at the carotid and femoral arteries provided the measure of the pulse wave at that particular arterial location. The distance between the two vascular beds divided by the pulse wave time shift provided a measure of the pulse wave velocity. (NCT01169701)
Timeframe: Month 6, month 24

,
Interventionm/sec (Mean)
Month 6 (n=31,30)Month 24 (n=28,25)
Everolimus7.407.06
Tacrolimus7.017.58

[back to top]

Change From Baseline in Left Ventricular Mass Index (LVMI)

Left ventricular hypertrophy grade was assessed by echocardiogram where the left ventricular mass index was calculated. The presence of LVM was defined as > 49.2 g/m^2.7 in men and >46.7 g/m^2.7 in women. A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, Month 24

Interventiong/m^2.7 (Mean)
Tacrolimus-6.071
Everolimus-4.008

[back to top]

Percentage of Participants With Major Cardiovascular Events (MACE)

The percentage of participants who experienced MACE were reported. MACE included acute myocardial infarction, insertion or replacement of implantable defibrillator, peripheral vascular disorders, congestive heart failure, coronary artery bypass, other events, percutaneous coronary intervention and stroke. (NCT01169701)
Timeframe: Month 24

InterventionPercentage of participants (Number)
Tacrolimus0.00
Everolimus0.00

[back to top]

Change From Baseline in Mean 24 Hour Systolic and Diastolic Blood Pressure

Blood pressure was measured using ambulatory blood pressure monitoring (ABPM). A negative change from baseline indicates improvement. (NCT01169701)
Timeframe: Baseline, Month 6, month 12, month 24

,
InterventionmmHg (Mean)
Month 6 (n=31,29)Month 12 (n=28,24)Month 24 (n=29,24)
Everolimus3.22.72.0
Tacrolimus-0.62.12.2

[back to top] [back to top]

Dose Limiting Toxicity (DLT) [Phase I Dose Finding]

A DLT was defined as an adverse event (a) with treatment attribution of possible, probable, or definite, and (b) occurs during cycle 1, and (c) meets any of the following criteria: Grade 3 or 4 non-hematologic toxicity excluding: nausea/vomiting controlled with antiemetics, Grade 3 hypertension that resolves to ≤150/90 within 7 days with supportive care, and Grade 3 asymptomatic, clinically insignificant laboratory abnormalities (LDH, alkaline phosphatase due to bone metastases, and asymptomatic hypophosphatemia). Hematologic toxicity: Grade 4 thrombocytopenia, Grade 3 thrombocytopenia with bleeding, Grade 4 neutropenia which persists for >7 days, Grade 4 neutropenia associated with fever >38.5C; Hematologic toxicity excluded lymphopenia or anemia of any grade. Missing more than 5 days of planned doses for drug-related intolerable grade 2 toxicity;Toxicity related to therapy severe enough to require a dose-reduction. (NCT01184326)
Timeframe: Participants were assessed for adverse events on days 1 and 15 for cycles 1-2 and every cycle thereafter; The observation period for DLT evaluation was the first 28 days of treatment.

InterventionParticipants (Count of Participants)
Dose Level 0: Everolimus 5mg + Pazopanib 600 mg2
Dose Level -1: Everolimus 5mg + Pazopanib 400 mg0

[back to top]

Objective Response Rate [Expansion]

The objective response rate (ORR) was defined as the percentage of participants achieving complete response (CR) or partial response (PR) based on RECIST 1.1 criteria on treatment. Per RECIST 1.1 for target lesions: CR is complete disappearance of all target lesions and PR is at least a 30% decrease in the sum of longest diameter (LD) of target lesions, taking as reference baseline sum LD. PR or better overall response assumes at a minimum incomplete response/stable disease (SD) for the evaluation of non-target lesions and absence of new lesions. (NCT01184326)
Timeframe: TDisease was evaluated radiologically at baseline and every 8 weeks on treatment; Treatment duration was up to 6 cycles in the Dose Level 0 cohort, 14 cycles in the Dose Level -1 cohort and 10 cycles in the expansion cohort (1 cycle=28 days).

Interventionpercentage of participants (Number)
All Phase I Dose Expansion Participants21.4

[back to top]

Median Progression-Free Survival [Expansion]

Progression-free survival based on the Kaplan-Meier method is defined as the duration of time from study entry to documented disease progression (PD) or death. Participants alive without PD are censored at date of last disease assessment. Per RECIST 1.1 for target lesions: PD is at least a 20% increase in sum longest diameter (LD), taking as reference the smallest sum on study with at least 5 mm absolute increase or the appearance of one or more new lesions. For non-target lesions, PD is appearance of one or more new lesions or unequivocal progression of existing non-target lesions. (NCT01184326)
Timeframe: Disease was evaluated radiologically at baseline and every 8 weeks on treatment; PFS follow-up was up to 9.2 months in the expansion cohort.

Interventionmonths (Median)
All Phase I Dose Expansion Participants2.9

[back to top]

Mean Duration of Response [Expansion]

Duration of response was calculated as the time from achieving complete response (CR) or partial response (PR) based on RECIST 1.1 criteria on treatment until disease progression. Per RECIST 1.1 for target lesions: CR is complete disappearance of all target lesions and PR is at least a 30% decrease in the sum of longest diameter (LD) of target lesions, taking as reference baseline sum LD. PR or better overall response assumes at a minimum incomplete response/stable disease (SD) for the evaluation of non-target lesions and absence of new lesions. For target lesions: PD is at least a 20% increase in sum longest diameter (LD), taking as reference the smallest sum on study with at least 5 mm absolute increase or the appearance of one or more new lesions. For non-target lesions, PD is appearance of one or more new lesions or unequivocal progression of existing non-target lesions. (NCT01184326)
Timeframe: Disease was evaluated radiologically at baseline and every 8 weeks on treatment; PFS follow-up was up to 9.2 months in the expansion cohort.

Interventionmonths (Mean)
All Phase I Dose Expansion Participants4.9

[back to top] [back to top]

Maximum Tolerated Dose (MTD) [Phase I Dose Finding]

The MTD of Pazopanib in combination with Everolimus 5 mg PO QD was determined by the number of patients who experience a dose limiting toxicity (DLT). See subsequent primary outcome measure for the DLT definition. The MTD is defined as the highest dose at which fewer than one-third of patients experience a DLT. If no DLTs are observed, the MTD is not reached. (NCT01184326)
Timeframe: Participants were assessed for adverse events on days 1 and 15 for cycles 1-2 and every cycle thereafter; The observation period for MTD evaluation was the first 28 days of treatment.

Interventionmg (Number)
All Phase I Dose Finding Participants400

[back to top]

Progression Free Survival (PFS) for First Line Medication

The amount of time after the first line medication begins until the cancer gets worse progresses. Progression is measured by an increase in measures tumors of at least 20 %, or overall increase in all the tumors, or the presence of new tumors. (NCT01185366)
Timeframe: 7 months

Interventionmonths (Median)
Everolimus4.1
Sunitinib6.1

[back to top]

Number of Participants Who Experienced Either a Grade 3 or 4 Adverse Event

Side effects, also called adverse events, that were related to either drug were documented and graded for severity according to the Common Terminology Criteria for Adverse Events (CTCAE) version 3. Grades increases in severity from 1 - 5. Grade one is a mild change which only requires monitoring. Grade 2 is a moderate change and may require some medication. Grade 3 is severe and requires hospitalization. Grade 4 is life threatening. Grade 5 is death. (NCT01185366)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Everolimus18
Sunitinib27

[back to top]

Number of Participants With Best Overall Response for First Line Medication

The best overall response for each participant was determined by using the Response Evaluation Criteria for Solid Tumors (RECIST). The responses are Complete Response (CR), Partial Response (PR), Stable Disease (SD), and Progression Disease (PD). CR is the disappearance of the cancer everywhere in the participant. PR is at least a 30 % reduction in the measured tumors from baseline. SD is no discernible change in the presence of cancer. Progressive disease is an increase of at least 20% of the measured cancer from the cancer's smallest measure. (NCT01185366)
Timeframe: 7 months

,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Everolimus01268
Sunitinib03219

[back to top]

Overall Survival of the First Line Therapy

The amount of time each participant is alive from the start of the first line therapy. (NCT01185366)
Timeframe: 17 months

Interventionmonths (Median)
Everolimus14.9
Sunitinib16.2

[back to top]

Progression Free Survival (PFS) for Crossover Medication

The amount of time after the crossover medication begins until the cancer gets worse progresses. Progression is measured by an increase in measures tumors of at least 20 %, or overall increase in all the tumors, or the presence of new tumors. (NCT01185366)
Timeframe: 4 months

Interventionmonths (Median)
Everolimus1.8
Sunitinib1.8

[back to top]

Toxicities, Graded According to the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0

The number of patients reporting grade 3 or higher adverse events as graded by the NCI's Common Toxicity Criteria (CTCAE) Version 4 are reported here. A complete list of all reported adverse events is reported in the Adverse Events section of this report. (NCT01198158)
Timeframe: Up to 30 days after completion of study treatment

,
InterventionParticipants (Count of Participants)
Grade 3 Adverse eventGrade 4 Adverse event
Arm I (Everolimus)211
Arm II (Everolimus With Bevacizumab)203

[back to top]

Progression-free Survival (PFS)

Progression free survival (PFS) is defined as the time from the date of randomization to the date of disease progression or death resulting from any cause, whichever comes first. Progression is defined according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01198158)
Timeframe: The time from randomization to disease progression or death from any cause, assessed up to 5.5 years

Interventionmonths (Median)
Arm I (Everolimus)7.2
Arm II (Everolimus With Bevacizumab)6.9

[back to top]

Objective Response Rate (CR + PR)

The response rate (percentage) is the percent of patients whose best response was Complete Response (CR) or Partial Response (PR) as defined by RECIST 1.1 criteria. Percentage of successes will be estimated by 100 times the number of successes divided by the total number of evaluable patients. (NCT01198158)
Timeframe: Up to 5.5 years

Interventionpercentage of participants (Number)
Arm I (Everolimus)17.1
Arm II (Everolimus With Bevacizumab)16.7

[back to top]

Overall Survival (OS)

Overall survival time is defined as the time from randomization to death due to any cause. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01198158)
Timeframe: The time from date of randomization to date of death due to any cause, assessed up to 5.5 years

Interventionmonths (Median)
Arm I (Everolimus)27.5
Arm II (Everolimus With Bevacizumab)21.8

[back to top]

Disease Control Rate (Stable Disease [SD] + Partial Response [PR] + Complete Response [CR]);

The disease control rate was based on the data as per local radiological review following the RECIST criteria. The disease control rate is defined as the proportion of patients with CR, PR, or SD and was summarized in terms of percentage with 95% exact Clopper-Pearson CIs (NCT01206764)
Timeframe: Approximately 4 years

InterventionPercentage of Participants (Number)
RAD00174.6

[back to top]

PFS (Progression-Free Survival)

the time from the date of the start of RAD001 treatment to the date of the first documented disease progression or death due to any cause (NCT01206764)
Timeframe: Approximately 4 years

InterventionWeeks (Median)
RAD00127.71

[back to top]

Overall Survival

Overall survival is defined as the time from date of start of treatment to date of death due to any cause. If a patient is not known to have died, survival will be censored at the date of last contact. (NCT01206764)
Timeframe: Approximately 4 years

InterventionWeeks (Median)
RAD001NA

[back to top]

Objective Response Rate (ORR; Where ORR = CR + PR)

The overall tumor response was based on the data as per local radiological review, following RECIST criteria. The ORR is defined as the proportion of patients with CR or PR and was summarized in terms of percentage with 95% exact Clopper-Pearson CIs (NCT01206764)
Timeframe: Approximately 4 years

InterventionPercentage of participants (Number)
RAD00117

[back to top]

Duration of Response (DOR)

The DOR analysis applied only to patients whose overall response was CR or PR and was defined as the time from onset of response (CR/PR) to progression or death from any cause (NCT01206764)
Timeframe: Approximately 4 years

InterventionWeeks (Median)
RAD001169

[back to top]

Number of Adverse Events as a Measure of Safety and Tolerability

To determine the safety of everolimus and everolimus plus paclitaxel in this patient population. A summary with the count of events per grade is provided. (NCT01215136)
Timeframe: 5 months

,
InterventionNumber of Events (Number)
Grade 1 Adverse EventsGrade 2 Adverse EventsGrade 3 Adverse EventsGrade 4 Adverse EventsGrade 5 Adverse Events
Cohort 1109400
Cohort 2116753913

[back to top]

Response Rate of Single-Agent Everolimus and Everolimus + Paclitaxel

To evaluate clinical benefit rate (complete response, partial response, and stable disease) at 4 months from initiation of treatment. (NCT01215136)
Timeframe: 4 months

Interventionpercentage of participants (Number)
Cohort 114.3
Cohort 237.9

[back to top]

Progression Free Survival

To determine median progression free survival per RECIST 1.1, per cohort. (NCT01215136)
Timeframe: 4 months

Interventionmonths (Median)
Cohort 12.3327
Cohort 25.8480

[back to top]

Overall Survival

To determine median overall survival at 1-year from the initiation of treatment. (NCT01215136)
Timeframe: 12 months

Interventionmonths (Median)
Cohort 14.5010
Cohort 210.8747

[back to top]

Progression Free Survival

Progression Free Survival (PFS) was defined as the time from study entry until disease progression or death, whichever occurs first. The median PFS was estimated using the Kaplan-Meier method. Progression was assessed per RECIST criteria, and defined as at least a 20% increase in the sum of the longest diameters of target lesions (and an absolute increase of at least 0.5 cm) or the appearance of new lesions. (NCT01229943)
Timeframe: From study entry to the date of documented progression or death from any cause, up to 3 years

Interventionmonths (Median)
Arm I (Octreotide Acetate and Everolimus)14.0
Arm II (Octreotide Acetate, Everolimus, and Bevacizumab)16.7

[back to top]

Overall Survival (OS)

Overall survival (OS) is defined as the time from study entry to death from any cause. The median OS was estimated using the Kaplan-Meier method. (NCT01229943)
Timeframe: From registration to time of death, assessed up to 3 years

Interventionmonths (Median)
Arm I (Octreotide Acetate and Everolimus)35.0
Arm II (Octreotide Acetate, Everolimus, and Bevacizumab)36.7

[back to top]

Overall Response Rate

The proportion of patients who respond (completely or partially) to each combination regimen will be estimated. Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria: Complete Response (CR): disappearance of all target lesions; Partial Response (PR) 30% decrease in sum of longest diameter of target lesions. (NCT01229943)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Arm I (Octreotide Acetate and Everolimus)12
Arm II (Octreotide Acetate, Everolimus, and Bevacizumab)31

[back to top]

Progression-free Survival

Estimated using the product-limit method of Kaplan and Meier. From the date treatment started until the date of first documented progression or date of death from any cause, whichever came first. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01231399)
Timeframe: up to 5 years

InterventionMonths (Median)
Dose Level 1 - 2.5 mg Everolimus Daily14.5

[back to top]

Overall Survival

Estimated using the product-limit method of Kaplan and Meier. From the date treatment started until the date of death from any cause. (NCT01231399)
Timeframe: Up to 5 years.

InterventionMonths (Median)
Dose Level 1 - 2.5 mg Everolimus Daily20.3

[back to top]

Number of Subject With Overall Response

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR (NCT01231399)
Timeframe: Up to 5 years

Interventionparticipants (Number)
Dose Level 1 - 2.5 mg Everolimus Daily5

[back to top]

Maximum Tolerated Dose (MTD) of Everolimus

The highest dose tested in which fewer than 33% of patients experience an attributable DLT to the study drug, when at least 6 patients are treated at that dose and are evaluable for toxicity. Toxicities will be assessed using Common Terminology Criteria for Adverse Events (CTCAE) v4.0. (NCT01231399)
Timeframe: Course 1 (first 28 days)

Interventionmg (Number)
Dose Level 1 - 2.5 mg Everolimus2.5

[back to top]

Disease Control Rate (DCR)

Disease Control Rate (DCR) was defined as the proportion of patients whose best overall response was either: Complete Response (CR), Partial Response (PR) or Stable Disease (SD). Disease Control Rate was calculated only for patients with measurable disease at baseline and was summarized using descriptive statistics. (NCT01231659)
Timeframe: From the date of randomization until the date of the first documented disease progression or date of death from any cause whichever came first, assessed for approximately 66 months

InterventionPercentage of Participants (Number)
Everolimus + Letrozole85.7

[back to top]

Median Time to Overall Survival (OS)

Overall Survival (OS) was defined as the time from the date of study entry to date of death due to any cause. If a death had not observed by the date of analysis, then OS was censored at the date of last contact. Distribution of OS was estimated using the Kaplan Meier method. The median OS along with 95% CI was presented. (NCT01231659)
Timeframe: From Date of randomization up to approximately 66 months

InterventionMonths (Median)
Everolimus + Letrozole22.3

[back to top]

Median Time to Progression-Free Survival (PFS)

Progression Free Survival (PFS) was defined as the time from the date of study entry to the date of first documented tumor progression or death from any cause, whichever occurred first. If a patient did not have an event, PFS was censored at the last date of tumor assessment. For patients with measurable disease at baseline, progression was determined according to the RECIST 1.0 criteria. Only descriptive analysis done. (NCT01231659)
Timeframe: Date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first, reported between day of first patient randomized up to 66 months

InterventionMonths (Median)
Everolimus + Letrozole9.0

[back to top]

Percentage of Participants With Overall Response Rate (ORR)

Overall Response Rate (ORR) was defined as the proportion of patients whose best overall response was either complete response (CR) or partial response (PR) according to RECIST 1.0 for target lesions and assessed by CT: Complete Response (CR), disappearance of all target lesions for a period of at least one month; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (ORR) = CR + PR. Only descriptive statistics. (NCT01231659)
Timeframe: From the date of randomization until the date of the first documented disease progression or date of death from any cause whichever came first, assessed for approximately 15 months

InterventionPercentage of Participants (Number)
Everolimus + Letrozole37.2

[back to top]

All Collected Deaths

"On treatment deaths were collected from FPFT up to 28 days after study drug discontinuation, for a maximum duration of 2002 days (treatment duration ranged from 1 to 1974 days).~Deaths post treatment survival follow up were collected after the on- treatment period, up to approximately 2092 days. Patients who didn't die during the on-treatment period and had not stopped study participation at the time of data cut-off (end of study) were censored." (NCT01231659)
Timeframe: up to 2002 days (on-treatment), up to approximately 2092 days (study duration)

InterventionParticipants (Count of Participants)
On-treatment deathsPost-treatment deathsAll deaths
Everolimus + Letrozole25052

[back to top]

Long-term Safety and Tolerability

The assessment of safety was based mainly on the frequency of AEs and on the number of laboratory values that fell outside of pre-determined ranges. Other safety data (e.g. ECG, vital signs) were considered as appropriate. Only descriptive analysis done. (NCT01231659)
Timeframe: From Date of first dose up to approximately 66 months

InterventionParticipants (Number)
AEs - AllAEs - Grade 3AEs - Grade 4
Everolimus + Letrozole72397

[back to top]

Median Overall Survival (OS) in Months

Overall survival reported in months as time interval between the date of treatment and the date of death or last follow-up. (NCT01239342)
Timeframe: Time interval between the date of treatment and the date of death or last follow-up, assessed up to 5 years

InterventionMonths (Median)
MK220623.5
Everolimus15.7

[back to top]

Summary of Selected Toxicities Grade 3 or Greater Toxicity Based on the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0

Adverse Events (AEs) list of reported events with associated intervention agent in a uniform presentation of events. The method of Thall, Simon and Estey (1995, 1996) was used to collect study participants' safety data summarized by treatment arm, category, severity and relevance. Comprehensive listing of AEs collected on study can be found in Adverse Event section separated by severity, Serious and Other AEs and represented by treatment arm, organ system-category within defined severity. (NCT01239342)
Timeframe: Up to 5 years

,
InterventionToxicities (Number)
HyperglycemiaHypertriglyceridemiaHyperuricemiaLymphocyte count decreasedMucositis oralRash maculo-papularPruritusProteinuriaPancreatitisLipase increasedGeneralized muscle weaknessDyspneaDehydration
Everolimus1101000000000
MK22067122181111112

[back to top]

Median Progression Free Survival (PFS) in Months

PFS defined as Time interval between date of treatment and date of disease progression, date of death or last follow-up date, whichever occurs first. Progression defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a nontarget lesion, or the appearance of new lesions. (NCT01239342)
Timeframe: Time interval between date of treatment and date of disease progression, date of death or last follow-up date, whichever occurs first, assessed up to 5 years

InterventionMonths (Median)
MK22063.68
Everolimus5.98

[back to top]

Overall Response Rate (ORR) Defined as Complete Response (CR) + Partial Response (PR)

Response for CR + PR defined by RECIST version 1.1. Repeat radiologic studies to evaluate disease progression or response (in accordance with restaging of disease) every 8 weeks. Complete Response (CR): Disappearance all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial Response (PR): >30% decrease in sum diameters of target lesions, reference baseline sum diameters. Progressive Disease (PD): >20% increase in sum diameters of target lesions, reference smallest sum on study (includes baseline sum if is smallest on study). In addition to relative increase of 20%, sum must demonstrate absolute increase >5 mm. (Note: appearance 1/+ new lesions considered progressions). Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, reference smallest sum diameters. (NCT01239342)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
MK220614.3
Everolimus0

[back to top]

Evaluation of Renal Function

Evaluation of renal function (serum creatinine) 90 days after transplant and 365 days after transplant. (NCT01239472)
Timeframe: 90 days after transplant and 365 days after transplant

,
Interventionmg/dL (Median)
Cr 90daysCr 365days
Everolimus1.31.4
Tacrolimus1.61.4

[back to top]

Cytokines Evaluation

Cd106(VCAM-1) , IP-10/CXCL10, MIG/CXCL9, MCP-1/CCL2, IL-1, RANTES, IL-8, IL12p70, TNF, IL-10, IL-6, IL-1, VEGF, FGF, CD54(ICAM-1) were analysed in urine 90 days after transplant (before randomization), and 365 days after transplant. Material were conserved at -80 Celsius, and analysed by ELISA at same time. Data was shown in MFI units (NCT01239472)
Timeframe: Urine and biopsy data are collected 90 days and 365 days after transplant.

,
InterventionMFI (Median)
IP10 90daysIP10 365daysMCP1 90daysMCP1 365daysMIG 90daysMIG 365daysRANTES 90daysRANTES 365daysIL8 90daysIL8 365daysIL12 90daysIL12 365daysTNF 90daysTNF 365daysIL10 90daysIL10 365 daysIL6 90daysIL6 365daysIL1 90daysIL1 365daysVEGF 90daysVEGF 365daysFGF 90daysFGF 365daysCD106 90daysCD106 365daysCD54 90daysCD54 365days
Everolimus8931474119137543358014515562443512412414615716418234926221522870454329285641961412931664
Tacrolimus380763234913063962871481444956411161241361531551792822812122288284393043222183241512696

[back to top]

Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)

Dual Anti-platelet Therapy (DAPT) is Aspirin & Clopidogrel/Ticlopidine/Other. (NCT01249027)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Observational934

[back to top]

Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)

Dual Anti-platelet Therapy (DAPT) is Aspirin & Clopidogrel/Ticlopidine/Other. (NCT01249027)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Observational879

[back to top]

Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)

Dual Anti-platelet Therapy (DAPT) is Aspirin & Clopidogrel/Ticlopidine/Other. (NCT01249027)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Observational800

[back to top]

Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)

Dual Anti-platelet Therapy (DAPT) is Aspirin and Clopidogrel/Ticlopidine/Other. (NCT01249027)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Observational2231

[back to top]

Number of Participants Compliance With Dual Anti-platelet Therapy (DAPT)

Dual Anti-platelet Therapy (DAPT) is Aspirin and Clopidogrel/Ticlopidine/Other. (NCT01249027)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Observational1085

[back to top]

Number of Participants Experiencing Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above)." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational75

[back to top]

Number of Participants Experiencing Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above)." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational98

[back to top]

Number of Participants Experiencing Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above)." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational34

[back to top]

Number of Participants Experiencing Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above)." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational64

[back to top]

Number of Participants With All Target Lesion Revascularization

Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold. (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational30

[back to top]

Number of Participants With All Target Lesion Revascularization

Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold. (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational33

[back to top]

Number of Participants With All Target Lesion Revascularization

Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold. (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational21

[back to top]

Number of Participants With All Target Lesions Revascularization

Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold. (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational25

[back to top]

Number of Participants With All Target Lesions Revascularization

Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold. (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational12

[back to top]

Number of Participants With All Target Vessel Revascularization

Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion. (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational42

[back to top]

Number of Participants With All Target Vessel Revascularization

Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion. (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational53

[back to top]

Number of Participants With All Target Vessel Revascularization

Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion. (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational60

[back to top]

Number of Participants With All Target Vessel Revascularization

Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion. (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational20

[back to top]

Number of Participants With All Target Vessel Revascularization

Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion. (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational36

[back to top]

Number of Participants With Any MI

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational23

[back to top]

Number of Participants With Any MI

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational28

[back to top]

Number of Participants With Any MI

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational31

[back to top]

Number of Participants With Any MI

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational20

[back to top]

Number of Participants With Any MI

"Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational13

[back to top]

Number of Participants With Composite Rate of All Death and Any MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational92

[back to top]

Number of Participants With Composite Rate of All Death and Any MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational120

[back to top]

Number of Participants With Composite Rate of All Death and Any MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational137

[back to top]

Number of Participants With Composite Rate of All Death and Any MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational46

[back to top]

Number of Participants With Composite Rate of All Death and Any MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational80

[back to top]

Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational191

[back to top]

Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational235

[back to top]

Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational271

[back to top]

Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational90

[back to top]

Number of Participants With Composite Rate of All Death, Any MI, and Any Repeat Revascularization

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational154

[back to top]

Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)

"Cardiac Death:Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational71

[back to top]

Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)

"Cardiac Death:Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational87

[back to top]

Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)

"Cardiac Death:Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational95

[back to top]

Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)

"Cardiac Death:Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational35

[back to top]

Number of Participants With Composite Rate of Cardiac Death, MI Attributed to the Target Vessel (TV-MI), and All Target Lesion Revascularization (TLR)

"Cardiac Death:Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational62

[back to top]

Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational63

[back to top]

Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational76

[back to top]

Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational84

[back to top]

Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational33

[back to top]

Number of Participants With Incidence of the Composite Rate of Cardiac Death and Any Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational55

[back to top]

Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))

"Ischemia-Driven Target Vessel Failure (TVF) is the composite endpoint comprised of~Cardiac death,~Myocardial infarction (Q wave and Non-Q wave),~Ischemia-driven target lesion revascularization by Coronary artery bypass grafting (CABG) or Percutaneous coronary intervention (PCI),~Ischemia-driven target vessel revascularization by CABG or PCI." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational100

[back to top]

Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))

"Ischemia-Driven Target Vessel Failure (TVF) is the composite endpoint comprised of~Cardiac death,~Myocardial infarction (Q wave and Non-Q wave),~Ischemia-driven target lesion revascularization by Coronary artery bypass grafting (CABG) or Percutaneous coronary intervention (PCI),~Ischemia-driven target vessel revascularization by CABG or PCI." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational123

[back to top]

Number of Participants Experiencing Death

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery).~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above)." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational113

[back to top]

Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))

"Ischemia-Driven Target Vessel Failure (TVF) is the composite endpoint comprised of~Cardiac death,~Myocardial infarction (Q wave and Non-Q wave),~Ischemia-driven target lesion revascularization by Coronary artery bypass grafting (CABG) or Percutaneous coronary intervention (PCI),~Ischemia-driven target vessel revascularization by CABG or PCI." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational50

[back to top]

Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))

"Ischemia-Driven Target Vessel Failure (TVF) is the composite endpoint comprised of~Cardiac death,~Myocardial infarction (Q wave and Non-Q wave),~Ischemia-driven target lesion revascularization by Coronary artery bypass grafting (CABG) or Percutaneous coronary intervention (PCI),~Ischemia-driven target vessel revascularization by CABG or PCI." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational86

[back to top]

Number of Participants With Major Bleeding Complications (According to GUSTO Classification)

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events:~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise Mild: Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational26

[back to top]

Number of Participants With Major Bleeding Complications (According to GUSTO Classification)

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events:~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise Mild: Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational31

[back to top]

Number of Participants With Major Bleeding Complications (According to GUSTO Classification)

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events:~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise Mild: Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational18

[back to top]

Number of Participants With Major Bleeding Complications (According to GUSTO Classification)

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events:~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise Mild: Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational18

[back to top]

Number of Participants With Major Bleeding Complications (According to GUSTO Classification)

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events:~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention.~Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise.~Mild: Bleeding that does not meet criteria for either moderate or severe bleeding." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational14

[back to top]

Number of Participants With Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational49

[back to top]

Number of Participants With Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational111

[back to top]

Number of Participants With Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational132

[back to top]

Number of Participants With Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational154

[back to top]

Number of Participants With Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational83

[back to top]

Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01249027)
Timeframe: 0 to 1137 days

InterventionParticipants (Count of Participants)
Observational69

[back to top]

Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01249027)
Timeframe: 0 to 1502 days

InterventionParticipants (Count of Participants)
Observational85

[back to top]

Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational93

[back to top]

Number of Participants With Target Lesion Failure (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01249027)
Timeframe: 0 to 772 days

InterventionParticipants (Count of Participants)
Observational60

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Composite Rate of Cardiac Death, TV-MI and Ischemia-driven TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01249027)
Timeframe: 0 to 407 days

InterventionParticipants (Count of Participants)
Observational35

[back to top]

Number of Participants With Stent Thrombosis

"Definite Stent Thrombosis (ST) occurred by either angiographic/pathologic confirmation of ST.~Angiographic confirmation:The presence of a thrombus that originates in the stent/in the segment 5mm proximal/distal to the stent&presence of at least 1 of the following criteria within 48hours:~Acute onset of ischemic symptoms at rest~New ischemic ECG changes~Typical rise&fall in cardiac biomarkers~Nonocclusive &occlusive thrombus~Pathological confirmation:Evidence of recent thrombus within the stent determined at autopsy/via examination of tissue retrieved following thrombectomy.~Probable ST may occur due to:~Unexplained death within first 30 days~Irrespective of the time after the index procedure,any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of ST&in the absence of any other obvious cause." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
DefiniteDefinite & Probable
Observational510

[back to top]

Number of Participants With Ischemia-driven Target Vessel Failure (ID-TVF) (Composite Rate of Cardiac Death, All MI and Target Vessel Revascularization (TVR))

"Ischemia-Driven Target Vessel Failure (TVF) is the composite endpoint comprised of~Cardiac death,~Myocardial infarction (Q wave and Non-Q wave),~Ischemia-driven target lesion revascularization by Coronary artery bypass grafting (CABG) or Percutaneous coronary intervention (PCI),~Ischemia-driven target vessel revascularization by CABG or PCI." (NCT01249027)
Timeframe: 0 to 1867 days

InterventionParticipants (Count of Participants)
Observational136

[back to top]

Number of Participants With Stable Disease (SD)

For patients with metastatic uveal melanoma treated with RAD001 and pasireotide LAR. (NCT01252251)
Timeframe: at 16 weeks

InterventionParticipants (Count of Participants)
RAD001 and Pasireotide LAR7

[back to top]

Median Progression Free Survival(PFS)

(NCT01252251)
Timeframe: Up to 3 years

Interventionweeks (Median)
RAD001 and Pasireotide LAR16

[back to top]

Median Overall Survival (OS)

(NCT01252251)
Timeframe: Up to 3 years

Interventionmonths (Median)
RAD001 and Pasireotide LAR11

[back to top]

Survival of Patients Treated

With Everolimus in combination with intravesical gemcitabine. Overall survival following start of therapy will be estimated using Kaplan-Meier methods. (NCT01259063)
Timeframe: 1 year

Intervention% of pts with CR who relapsed in a year (Number)
Pts Who Failed or Relapsed After Intravesical BCG21

[back to top]

Complete Response (CR) Rate

Complete Response (CR) is defined as no evidence of disease (by cytology and cystoscopy). Partial Response is defined as negative cystoscopy and positive cytology. Progression is defined as the development of invasion or metastasis. If the participant develops a recurrence, they will be considered having failed treatment. Participants will also be considered having failed treatment if there is no response to treatment after two cycles. (NCT01259063)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseDid not achieve CR or PR
Pts Who Failed or Relapsed After Intravesical BCG1788

[back to top]

Phase I - Dose-limiting Toxicity (DLT)

Everolimus (mg) given in conjunction with intravesical gemcitabine (NCT01259063)
Timeframe: 8 weeks

InterventionDLT (Number)
Pts Who Failed or Relapsed After Intravesical BCG0

[back to top]

Phase II - Patients Who Are Free of Disease at 1 Year

following start of therapy. (NCT01259063)
Timeframe: 1 year

Interventionpercentage of pts disease free at 1 year (Number)
Pts Who Failed or Relapsed After Intravesical BCG21

[back to top]

Phase I - Maximum Tolerated Dose (MTD)

of Everolimus given in conjunction with intravesical gemcitabine (NCT01259063)
Timeframe: 8 weeks

Interventionmg daily of everolimus (Number)
Pts Who Failed or Relapsed After Intravesical BCG10

[back to top]

Number of Rejections Leading to Hemodynamic Compromise

Number of all rejections were recorded through the duration of the study with the intent to identify rejections leading to hemodynamic compromise. (NCT01266148)
Timeframe: 52 weeks

,
Interventionrejections (Number)
Total rejectionTreated rejectionRejections with hemodynamic compromise
Control128170
Everolimus185430

[back to top]

Measured Glomerular Filtration Rate (mGFR), 12 Months After Heart Transplantation

Measured Glomerular Filtration Rate (mGFR) describes the flow rate of filtered fluid through the kidney. GFR is equal to the clearance rate when any solute is freely filtered and is neither reabsorbed nor secreted by the kidneys. The rate therefore measured is the quantity of the substance in the urine that originated from a calculable volume of blood. Participants' urine was used for this assessment at week 52 after heart transplant. (NCT01266148)
Timeframe: Week 52

InterventionmGFR ml/min (Mean)
Everolimus79.8
Control61.5

[back to top]

Average Level of Protenuria at Week 52

Proteinuria is measured as the ratio of albumin/creatinine mg/mmol. Measurements were taken from participants urine samples. (NCT01266148)
Timeframe: 52 weeks

Interventionmg/mmol (Mean)
Everolimus7.2
Control1.2

[back to top]

Change in Quality of Life - Euro Quality of Life 5D (EQ-5D)

Change in Quality of Life was assessed via the EQ-5D questionnaire which consists of: EQ-5D-5L descriptive system and EQ Visual Analogue scale (EQ VAS). The EQ-5D-5L comprises 5 dimensions (mobility, self-care, usual activities, pain/discomfort, anxiety/depression). Each dimension has 5 levels (no problems, slight problems, moderate problems, severe problems, and extreme problems). The patient indicates his/her health state by checking the most appropriate statement. This decision results in a 1-digit number expressing the level selected for that dimension. The digits for 5 dimensions are combined in a 5-digit number describing the respondent's health state. The possible score is 1 to 5 where a lower number indicates improvement. The EQ VAS records the patient's self-rated health on a 20 cm vertical, visual analogue scale with endpoints labelled 'the best health you can imagine' and 'the worst health you can imagine'. The score is 0 to 100 where a higher score represents improvement. (NCT01266148)
Timeframe: Pre transplant and 52 weeks

,
InterventionUnits on a scale (Mean)
EQ-5D-5L pre- transplant (n=45,49)EQ-5D-5L at week 52 (n=41,44)EQ VAS pre- transplant (n=41,48)EQ VAS at week 52 (n=41,42)
Control0.50690.836738.979.4
Everolimus0.57500.832946.080.0

[back to top]

Change in Calculated Glomerular Filtration Rate From Pre-transplantation to Week 52

Change in calculated glomerular filtration rate from pre-transplantation to week 52 was calculated according to the Modification of Diet in Renal Disease (MDRD) method. Measurements were taken prior to transplant (day 1), between weeks 7 to 11 and end week 52. (NCT01266148)
Timeframe: Day 1, weeks 7 to 11(baseline) and of week 52

,
InterventionmGFR mL/min (Mean)
Day 1 (n= 43, 47)Weeks 7 to 11 (n= 46, 49)Week 52 (n= 42, 53)
Control-12.6-6.8-8.0
Everolimus-13.07.427.8

[back to top]

Progression of Chronic Allograft Vasculopathy (CAV) Based on Maximal Intimal Thickness (MIT) From Baseline to Week 52

The progression of chronic allograft vasculopathy (CAV) was assessed by intravascular ultrasound (IVUS) examinations and measured Maximal Intimal Thickness (MIT)(in mm). A major coronary epicardial artery (preferentially the left-anterior descending coronary artery) was imaged, and the MIT parameters were recorded at baseline and at week 52. (NCT01266148)
Timeframe: Baseline and week 52

,
Interventionmm (Mean)
Baseline (Week 7)Week 52
Control0.560.65
Everolimus0.520.55

[back to top]

Progression of Chronic Allograft Vasculopathy (CAV) Based on Incidence of Chronic Allograft Vasculopathy (CAV) From Baseline to Week 52

the progression of chronic allograft vasculopathy (CAV) assessed by intravascular ultrasound (IVUS) examinations, measured the incidence of CAV (in percent of patients) at baseline and at week 52. Incidence of CAV represents percent of patients having a MIT (maximal intima thickness) > 0.5 mm. (NCT01266148)
Timeframe: Baseline and week 52

,
InterventionPercentage of patients (Number)
No CAV at baseline (week 7)CAV at baseline (week 7)No CAV at week 52CAV at week 52
Control47.952.135.464.6
Everolimus43.556.55050

[back to top]

Occurrence of Treatment Failures up to 12 Months After Transplant

Treatment failure was defined as the number of participants who died or lost their graft at any timepoint througout the duration of the study. (NCT01266148)
Timeframe: 52 weeks

,
Interventionparticipants (Number)
Treatment failure (death)Graff lossTotal treatment failures (graft loss + death
Control303
Everolimus202

[back to top]

Calculated Glomerular Filtration Rate From Pre-Transplantation to Week 52

Calculated Glomerular Filtration Rate from pre-transplantation to week 52 was calculated according to the Modification of Diet in Renal Disease (MDRD) method. Measurements were taken prior to transplant (day 1), between weeks 7 to 11 and end of week 52. (NCT01266148)
Timeframe: Day 1, weeks 7 to 11 and of week 52

,
InterventionmGFR mL/min (Mean)
Day 1 (n= 46, 48)Weeks 7 to 11 (n=50, 51)Week 52 (n= 45, 55)
Control66.169.669.3
Everolimus65.484.9104.5

[back to top]

Lipid Profile at 12 Months

Total Cholesterol, LDL-Chol, HDL-Chol and TG at week 52. Measurements were taken via participants blood samples. (NCT01266148)
Timeframe: 52 weeks

,
Interventionmmol/L (Mean)
Total cholesterolLDL-CHDL-C
Control5.12.81.6
Everolimus5.32.91.6

[back to top]

Change in Quality of Life Assessed by SF-36 (Minnesota Living With Heart Failure Questionnaire ([MLHF)]) From Pre-transplant to Week 52 of Treatment

Change in Quality of Life was assessed via the SF-36 (Minnesota Living with Heart Failure questionnaire ([MLHF)]) before transplant surgery and at week 52 of treatment. The SF-36 is a validated, self-administered questionnaire. The questionnaire, which includes 36 questions measures 8 dimensions of health: physical function, role-physical, bodily pain, general health, vitality, social function, role-emotional, and mental health. Scores can be summarized in 2 summary components assessing physical and mental health. Items in each dimension are coded, aggregated, summed, and transformed into a scale ranging from 0 (worse health) to 100 (best health). (NCT01266148)
Timeframe: Pre transplant and 52 weeks

,
InterventionUnits on a scale (Mean)
Physical Health pre- transplantPhysical Health at week 52Mental Health pre- transplantMental Health at week 52
Control32.948.838.753.9
Everolimus30.848.846.251.5

[back to top]

Number of Participants With Progression-free Survival

"Time from starting treatment until disease progression as defined using Response Evaluation CriteriaIn Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions, or death." (NCT01270321)
Timeframe: Through study completion, an average of 1 year

InterventionParticipants (Count of Participants)
Arm A (Everolimus Alone)17
Arm B (Pasireotide Alone)9
Arm C (Everolimus + Pasireotide)11

[back to top]

Number of Participants With Response Per Responsive Evaluation Criteria in Solid Tumors Criteria (RECIST v1.0)

"Number of participants with response per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR." (NCT01270321)
Timeframe: Through study completion, an average of 1 year

InterventionParticipants (Count of Participants)
Arm A (Everolimus Alone)17
Arm B (Pasireotide Alone)9
Arm C (Everolimus + Pasireotide)11

[back to top]

All Collected Deaths

On treatment deaths were collected from FPFT up to 28 days after study drug discontinuation, for a maximum duration of 35.52 months (treatment duration ranged from 0.36 to to 35.52 months for the phase I part) and a maximum duration of 21.88 months (treatment duration of 0.16 to 21.88 months for the phase II part). Deaths post treatment survival follow up were collected after the on treatment period, up to 70 months. (NCT01275222)
Timeframe: approx. 35.52 months, approx. 70 months

,,,,,
InterventionParticipants (Number)
Total DeathsDeaths on-treatment
Phase l: RAD001 2.5mg/Day + Glivec 600mg/Day83
Phase l: RAD001 2.5mg/Day + Glivec 800mg/Day32
Phase l: RAD001 20mg/Week122
Phase l: RAD001 5mg/Day + Glivec 600mg/Day30
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day93
Phase ll: Stratum ll (Post Second-line Therapy)226

[back to top]

4-Month Progression-free Survival (PFS) Rate - Phase II

Assessed clinical efficacy of the combination regimen in this patient population per Progression-free survival (PFS). Progression-free survival (PFS) was the time from date of start of treatment to the date of first documented progression or death due to any cause. Per protocol (PP) population includes patients with no known overall lesion response during 4 months +/- 2 weeks, or who later had response of CR/PR/stable disease (SD). PP population excludes patients with no known response during 4 months + / -2 weeks and no subsequent CR/PR/SD. (NCT01275222)
Timeframe: about 4 months

InterventionPercentage of participants (Number)
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day17.4
Phase ll: Stratum ll (Post Second-line Therapy)37.1

[back to top]

Best Overall Response (BOR) as Assessed by Overall Response (Complete Response (CR) & Partial Response (PR)) - Phase I & II

Assessed clinical efficacy of the combination regimen in this patient population per BOR by Overall response (CR + PR). Complete response: Disappearance of all target lesions; Partial response: ≥ 30% decrease in the sum of the longest diameter of target lesions compared to baseline (and not ≥20% increase compared to smallest sum). (NCT01275222)
Timeframe: 6 - 8 weeks

InterventionPercentage of Participants (Number)
Phase l: RAD001 20mg/Week0
Phase l: RAD001 2.5mg/Day + Glivec 600mg/Day1
Phase l: RAD001 5mg/Day + Glivec 600mg/Day0
Phase l: RAD001 2.5mg/Day + Glivec 800mg/Day0.0
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day0
Phase ll: Stratum ll (Post Second-line Therapy)1

[back to top]

Number of Participants With Adverse Events (AEs): Phase I & II

Assessed safety and tolerability of the combination administration of RAD001 and imatinib when given to patients with imatinib-refractory gastro-intestinal stromal tumors (GIST) by number of participants with adverse events. (NCT01275222)
Timeframe: 4 - 8 weeks

InterventionParticipants (Number)
Phase l: RAD001 20mg/Week13
Phase l: RAD001 2.5mg/Day + Glivec 600mg/Day13
Phase l: RAD001 5mg/Day + Glivec 600mg/Day5
Phase l: RAD001 2.5mg/Day + Glivec 800mg/Day11
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day27
Phase ll: Stratum ll (Post Second-line Therapy)47

[back to top]

Overall Survival (OS) - Phase I & II

Assessed clinical efficacy of the combination regimen in this patient population per Overall Survival (OS). Overall survival was defined as the time from date of start of treatment to date of death due to any cause. For patients still receiving study medication at the time of the analysis, survival was censored at the date of last examination. If a patient was not known to have died but was no longer continuing with study medication, survival was censored at the date of last contact. (NCT01275222)
Timeframe: about 60 months

Interventionmonths (Median)
Phase l: RAD001 20mg/Week9.4
Phase l: RAD001 5mg/Day + Glivec 600mg/Day10.9
Phase l: RAD001 2.5mg/Day + Glivec 600mg/Day18.7
Phase l: RAD001 2.5mg/Day + Glivec 800mg/DayNA
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day14.9
Phase ll: Stratum ll (Post Second-line Therapy)10.7

[back to top]

Progression-free Survival (PFS) - Phase II

"Assessed clinical efficacy of the combination regimen in this patient population per Progression-free survival (PFS). Progression-free survival (PFS) was the time from date of start of treatment to the date of first documented progression or death due to any cause. If a patient had not progressed or died, progression-free survival was censored at the time of last adequate tumor assessment.~According to the study protocol no tumor assessments were performed after discontinuation of treatment with study drug. Therefore, for all patients who discontinued treatment without a documented progression but died thereafter, death was considered as PFS event only if it occurred within the regular safety follow-up of 28 days after discontinuation. Otherwise, the PFS time was censored at the last adequate tumor assessment." (NCT01275222)
Timeframe: about 60 months

Interventionmonths (Median)
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day1.9
Phase ll: Stratum ll (Post Second-line Therapy)3.5

[back to top]

Trough Concentrations for RAD001 and for Imatinib - Phase II

Assessed the pharmacokinetics (PK) of the combination administration of RAD001 and imatinib in this patient population. (NCT01275222)
Timeframe: Part II Daily regimen: Baseline, Days 8, 15 and thereafter approximately every two months starting at month 3 whenever possible

Interventionng/mL (Median)
Week 2: RAD001Week 3: RAD001Week 9: RAD001Month 4: RAD001Baseline: ImatinibWeek 2: ImatinibWeek 3: ImatinibWeek 9: ImatinibMonth 4: ImatinibMonth 6: ImatinibMonth 8: ImatinibMonth 12: Imatinib
Phase ll: Stratum ll (Post Second-line Therapy)12.10012.75010.9507.840501.50540.00718.00899.0446.002040.00575.00324.00

[back to top]

Trough Concentrations for RAD001 and for Imatinib - Phase II

Assessed the pharmacokinetics (PK) of the combination administration of RAD001 and imatinib in this patient population. (NCT01275222)
Timeframe: Part II Daily regimen: Baseline, Days 8, 15 and thereafter approximately every two months starting at month 3 whenever possible

Interventionng/mL (Median)
Week 2: RAD001Week 3: RAD001Baseline: ImatinibWeek 2: ImatinibWeek 3: ImatinibMonth 4: ImatinibMonth 6: Imatinib
Phase ll - Stratum l (First-line Resistant/Refractory): RAD001 2.5mg/Day + Glivec 600mg/Day24.30019.900472.00626.50572.00333.00181.00

[back to top]

Renal Function Assessed by Creatinine Clearance

Renal function was assessed by measuring serum creatinine and by computing creatinine clearance using the formula of Cockcroft-Gault. (NCT01276457)
Timeframe: Month 12, Month 18, and Month 24

,
InterventionmL/min (Mean)
Month 12 (n=111, 111)Month 18 (n=108, 108)Month 24 (n=106, 110)
Standard Everolimus Blood Target + Low Dose Cyclosporine62.5062.8063.76
Upper Everolimus Blood Target + Very Low Dose Cyclosporine61.2660.9061.92

[back to top]

Number of Participants Who Died, Number of Participants Who Lost Their Graft, and Number of Participants Who Died or Lost Their Graft

A participant lost his graft if he/she started dialysis and was not able to subsequently be removed from dialysis or underwent graft nephrectomy. (NCT01276457)
Timeframe: Baseline to end of study (Month 24)

,
InterventionParticipants (Number)
DiedLost graftDied or lost graft
Standard Everolimus Blood Target + Low Dose Cyclosporine31518
Upper Everolimus Blood Target + Very Low Dose Cyclosporine268

[back to top]

Number of Participants With Adverse Events (AE), Serious Adverse Events (SAE) or Deaths

Safety was assessed using reports of adverse events of all participants in this study. Serious adverse events are those events that resulted in death, were life threatening, required inpatient hospitalization or prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity, or was a congenital anomaly/birth defect. (NCT01276457)
Timeframe: Baseline to end of study (Month 24)

,
InterventionPartcipants (Number)
AEsInfectionClinically significant AEsSAEsDied
Standard Everolimus Blood Target + Low Dose Cyclosporine14310153903
Upper Everolimus Blood Target + Very Low Dose Cyclosporine1429558852

[back to top]

Number of Participants With Biopsy-proven Acute Rejection

A graft core biopsy was performed on all suspected acute rejection episodes within 48 hours. Biopsies were read by the local pathologist according to the 1997 Banff criteria. A biopsy-proven acute rejection was be defined as a biopsy graded IA, IB, IIA, IIB, or III. (NCT01276457)
Timeframe: Baseline to end of study (Month 24)

InterventionParticipants (Number)
Upper Everolimus Blood Target + Very Low Dose Cyclosporine21
Standard Everolimus Blood Target + Low Dose Cyclosporine21

[back to top]

Response Rate (CR + PR) Assessed by RECIST 1.1 (Phase II)

(NCT01281865)
Timeframe: At 8 weeks

,
Interventionparticipants (Number)
Progression of DiseaseStable Disease
Arm 1-Treatment (Everolimus and Imatinib Mesylate)45
Arm 2-Treatment (Everolimus and Imatinib Mesylate)10

[back to top]

Progression-free Survival (PFS)

Objective tumor responses were assessed every 2 cycles of chemotherapy with computed tomography or positron emission tomography/ computed tomography scans in accordance with Response Evaluation Criteria In Solid Tumors (RECIST) criteria. (NCT01283334)
Timeframe: The time of PFS was calculated as the time from study enrollment to the disease progression date, death date, or last contact, whichever came first, up to 25 months

Interventionmonths (Median)
Carboplatin, Cetuximab and Everolimus8.15

[back to top]

To Measure the Safety and Clinical Effectiveness of the Combination of Carboplatin, Cetuximab and RAD001 in Patients With Advanced (Recurrent or Metastatic) Head and Neck Cancer

This phase 1 clinical trial used a standard 3 + 3 design. Four dose levels of everolimus were planned to be evaluated, and the standard 3 + 3 design with dose de-escalation was used in the trial. Namely, 3 patients were assigned to starting dose level 1. If no dose-limiting toxicity (DLT) was observed, the trial proceeded to the next dose level, and another cohort of 3 patients was enrolled. If at least 2 of the 3 patients experienced at least 1 DLT, then the dose level decreased; otherwise, if only 1 patient experienced DLT, then 3 more patients were enrolled at the same dose level. If none of the 3 additional patients experienced DLT, the dose was escalated; otherwise, the dose level decreased. Dose reduction continued until a dose level was reached at which 6 patients had been treated and at most 1 DLT was observed. (NCT01283334)
Timeframe: Within the first 21 days of therapy

Interventionparticipants (Number)
Everolimus Dose Level 1 DLT2
Everolimus Dose Level -1 DLT1

[back to top]

Evaluation of the Efficacy of RAD001 on Neurocognition in Patients With TSC Compared With Placebo.

"Baseline and 6 month Timepoint scores are reported for the following primary outcome measures:~Peabody Picture Vocabulary Test 4 (PPVT-4; Receptive Language Measure). Scores reported as (mean, SD). Range=40-160, higher scores are better.~Expressive Vocabulary Test 2 (EVT-2; Expressive Language Measure). Scores reported as (mean, SD). Range=40-160, higher scores are better.~Wide Range Assessment of Memory and Learning 2 (WRAML2; Measure of Verbal Memory and Attention ). Scores reported as (mean, SD). Range=1-19, higher scores are better.~Vineland Adaptive Behavior Scales-II (VABS-II; Measure of Adaptive Behavior). Scores reported as (mean, SD). Range = 40-160, higher scores are better.~Purdue Pegboard Test (Measure of Fine Motor Speed and Coordination). Scores reported as (mean, SD). Range = 40-160, higher scores are better." (NCT01289912)
Timeframe: 6 months

,,,
Interventionunits on a scale (Mean)
PPVT4 (Receptive Language)EVT2 (Expressive Language)WRAML2 Verbal LearningWRAML2 Verbal RecallVABS2 Adaptive Behavior CompositeVABS2 CommunicationVABS2 SocializationVABS2 Daily Living SkillsVABS2 Motor SkillsPegboard, Dominant HandPegboard, Non-Dominant Hand
Placebo - 6 Months88.5489.158.468.2379.0081.7780.3881.6992.6989.8392.15
Placebo - Baseline86.0085.478.007.0778.7981.2779.0778.8089.87101.33105.27
RAD001 - 6 Months82.582.557.858.0874.7276.7976.8676.4187.7997.25112.78
RAD001 - Baseline81.0690.007.147.5272.0673.6874.1074.6878.83103.06119.30

[back to top]

Evaluation of the Safety of RAD001 on Neurocognition in Patients With TSC Compared With Placebo in Patients With TSC.

Evaluation of the safety of RAD001 compared with placebo in patients with TSC focusing on NCI CTCAE Grade 3 and 4 adverse events, serious adverse events, and Grade 3 and 4 laboratory toxicities. (NCT01289912)
Timeframe: 6 months

InterventionAdverse Events (Number)
RAD001325
Placebo147

[back to top]

Comparison of Autism Spectrum Disorders Features Between Patients Taking RAD001 vs. Placebo

Scores for the Baseline and 6 Month Timepoints are reported. The secondary outcome measure was the Social Responsiveness Scale (SRS). Standard scores are reported with a mean of 100 and standard deviation of 15. The range is 40-160 with higher scores indicating a better outcome. (NCT01289912)
Timeframe: 6 months

,,,
Interventionunits on a scale (SRS) (Mean)
SRS Social Responsiveness (range 40(worse) -160)SRS Social AwarenessSRS Social CognitionSRS Social CommunicationSRS Social MotivationSRS Autism Mannerisms
Placebo - 6 Months80.6267.3881.1576.8572.7779.77
Placebo - Baseline77.1467.4076.4374.3669.8076.71
RAD001 - 6 Months75.6964.8676.1771.6967.2478.66
RAD001 - Baseline76.4362.9075.7072.2369.6080.03

[back to top]

Comparison of Behavioral Problems Between Patients Taking RAD001 vs Placebo

"Scores for Baseline and 6 Month Timepoints are reported for the following secondary outcome measures:~Behavior Rating Inventory of Executive Function (BRIEF) (Measure of executive functions) T-scores are reported, with a mean of 50 and a standard deviation of 10. The range is 30-100 with higher scores indicating a worse outcome.~Behavioral Assessment System for Children (BASC) (Measure of emotional and behavioral problems) T-scores are reported (mean of 50, SD of 10). The range is 30-100 with higher scores indicating a worse outcome. Conversely, on the Adaptive Skills subscale of the BASC, lower scores indicate a poorer outcome.~Strengths and Difficulties Questionnaire (SDQ). Includes questions related to emotional symptoms, conduct problems, inattention/hyperactivity, peer relationship problems and prosocial behavior. Responses to these items are summed to comprise a Total Difficulties Score, which ranges from 0-40, with lower scores indicating a better outcome" (NCT01289912)
Timeframe: 6 months

,,,
Interventionunits on a scale (Mean)
BRIEF Global Executive CompositeBRIEF Behavioral Regulation IndexBRIEF Metacognition IndexBASC2 Behavioral Symptoms IndexBASC2 Externalizing ProblemsBASC2 Internalizing ProblemsBASC2 Adaptive SkillsSDQ Total Difficulties Scale
Placebo - 6 Months62.2560.8361.8357.5452.6256.0042.0814.42
Placebo - Baseline70.5070.9368.0762.2957.3658.4338.7916.00
RAD001 - 6 Months64.6862.5764.5755.4152.3451.4540.0013.20
RAD001 - Baseline69.4866.5368.9358.9753.2451.5536.7215.46

[back to top]

Evaluation of the Efficacy of RAD001 on Neurocognition (Cambridge Neuropsychological Test Automated Battery) in Patients With TSC Compared With Placebo.

"Scores are reported for baseline and 6 month timepoints on the Cambridge Neuropsychological Test Automated Battery (CANTAB) subscales below. For all subscales, scores are reported as the mean difference between the study subjects and a normative population matched for age, gender and IQ (e.g., subject subscale score - mean of matched normative group = reported score). Higher scores represent a better outcome.~Spatial Span (SSP) (spatial memory span) Range: -3 to 3~Spatial Working Memory (working memory) Range: -3 to 3~Pattern Recognition Memory (PRM) (visual pattern recognition memory) Range: -3 to 3~Spatial Recognition Memory (SRM) (spatial recognition memory) Range: -4 to 4~Rapid Visual Information Processing (RVIP) (sustained attention) Range: -4 to 4~Stockings of Cambridge (SOC) (spatial planning) Range: -4 to 4~Intra-Extra Dimensional Set Shift (IDED) (cognitive flexibility) Range: -5 to 5~Reaction Time (processing speed) Range: -5 to 5" (NCT01289912)
Timeframe: 6 months

,,,
Interventionunits on a scale (Mean)
CANTAB Pattern Recognition MemoryCANTAB Spatial Recognition MemoryCANTAB Spatial SpanCANTAB Spatial Working MemoryCANTAB Reaction TimeCANTAB Rapid Visual ProcessingCANTAB Stockings of CambridgeCANTAB IDED
Placebo - 6 Months-.91-2.62-1.29-.73-.88-.97-.58-1.05
Placebo - Baseline-1.48-2.33-1.32-.66-2.24-.96-1.38-1.02
RAD001 - 6 Months-1.09-2.19-1.32-.27-2.91-1.2-1.84-2.24
RAD001 - Baseline-1.23-2.24-1.02-.67-1.15-1.41-1.89-1.48

[back to top]

Comparison of Academic Skills Between Patients Taking RAD001 vs. Placebo

Scores are reported for Baseline and 6 Month Timepoints. The secondary outcome measure was the Wide Range Achievement Test 4 (WRAT4), which was used to assess academic skills. The Reading and Math subtests were used. Standard scores are reported which have a mean of 100 and a standard deviation of 15 (range=40-160 where higher is better). (NCT01289912)
Timeframe: 6 months

,,,
Interventionunits on a scale (Mean)
Word Reading (WRAT 4)Math Computation (WRAT 4)
Placebo - 6 Months91.1577.31
Placebo - Baseline86.8076.00
RAD001 - 6 Months84.4573.81
RAD001 - Baseline84.2874.60

[back to top]

Extracranial Response

"Extracranial response was measured using RECIST 1.1 criteria and defined as the number of subjects achieving CR or PR.~Complete Response (CR) - Disappearance of all target and nontarget lesions Partial Response (PR) - at least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum longest diameter AND an absolute decrease of at least 5mm in at least one target lesion.~Stable Disease (SD) - neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum of the longest diameter since the treatment started.~Progressive Disease (PD) - at least a 20% increase in the sum LD of target lesions, taking as reference the smallest sum of the longest diameter recorded since the treatment started AND an absolute increase in size of at least 5 mm in at least one target lesion OR the appearance of one or more new lesions of at least 6 mm in size." (NCT01305941)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Everolimus +Vinorelbine + Trastuzumab5

[back to top]

Time to Intracranial Progression.

"Time to intracranial progression after administration of everolimus in combination with trastuzumab and vinorelbine as defined via modified RECIST criteria.~Progressive Disease (PD) - at least a 20% increase in the sum LD of target lesions, taking as reference the smallest sum of the longest diameter recorded since the treatment started AND an absolute increase in size of at least 5 mm in at least one target lesion OR the appearance of one or more new lesions of at least 6 mm in size." (NCT01305941)
Timeframe: 3 years

Interventionmonths (Median)
Everolimus +Vinorelbine + Trastuzumab3.93

[back to top]

Toxicity

"Grade 3 or higher toxicities of interest are reported. Toxicity was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) v4.~The NCI CTCAE is a descriptive terminology which can be utilized for Adverse Event (AE) reporting. A grading (severity) scale is provided for each AE term. Grade 1 Mild; asymptomatic or mild symptoms; clinical or diagnostic observations only; intervention not indicated. Grade 2 Moderate; minimal, local or noninvasive intervention indicated; limiting age-appropriate instrumental Activities of Daily Living (ADL). Grade 3 Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self care ADL. Grade 4 Life-threatening consequences; urgent intervention indicated. Grade 5 Death related to AE." (NCT01305941)
Timeframe: 24 weeks

InterventionParticipants (Count of Participants)
Alkaline phosphatase increasedAnemiaAnorexiaAsparate Aminotransferase IncreasedDiarrheaFebrile NeutropeniaHyperkalemiaLymphocyte Count DecreasedMucositis OralNeutrophil Count DecreasedPneumonitisSepsisWhite Blood Cell Decreased
Everolimus +Vinorelbine + Trastuzumab151212135131311

[back to top]

Overall Survival

Overall survival (OS) after administration of everolimus in combination with trastuzumab and vinorelbine (NCT01305941)
Timeframe: 3 years

Interventionyears (Median)
Everolimus +Vinorelbine + Trastuzumab1.01

[back to top]

Intracranial Response Rate- MacDonald Criteria

"Intracranial tumor lesions were evaluated via gadolinium-enhanced brain MRI using the MacDonald criteria. Measurable disease is defined as at least 1 measurable brain lesion accurately measured in at least 2 dimensions (longest diameter) as ≥5.0 mm. Tumor size is the product of the 2 longest bi-dimensional lines.~Complete Response (CR)- Disappearance of all tumor on consecutive CT or MRI scans at least 1 month apart, off steroids for treatment of neurological symptoms, and neurologically stable or improved.~Partial Response (PR)- ≥50% reduction in size of tumor on consecutive CT or MRI scans at least 1 month part, steroids stable or reduced, and neurologically stable or improved.~Progressive Disease (PD)- ≥25% increase in size of tumor or any new tumor on CT or MRI scans, or neurologically worse, and steroids stable or increased due to neurologic symptoms.~Stable Disease (SD)- all other situations Overall Response Rate (ORR) is the sum of partial responses (PRs) and CRs." (NCT01305941)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Everolimus +Vinorelbine + Trastuzumab1

[back to top]

Functional Assessment of Cancer Therapy- Brain (FACT-Br) Change From Baseline to Assess Impact of Everolimus in Combination With Trastuzumab and Vinorelbine on Quality of Life

The FACT-Br is a 23-question self-report questionnaire subscale administered with the Functional Assessment of Cancer Therapy- General (FACT-G) which contains concerns relevant to patients with brain tumors . Each question has a value 0-4. For some questions a higher indicates better outcome and others are the opposite. The former are summed as is, the latter are reversed in value before adding, such that each domain ranges from 0 to 4 times the number of questions in the domain, with 0 indicating worst and the highest possible value indicating best outcome. Total scores on the FACT-Br subscale range from 0 to 92 with lower scores indicating declining quality of life. The change from baseline is the difference in scores between the baseline and 9 week assessments. (NCT01305941)
Timeframe: 9 weeks

Interventionunits on a scale (Median)
Everolimus +Vinorelbine + Trastuzumab-1.0

[back to top]

Intracranial Objective Response Rate- Modified RECIST Criteria

"response will be evaluated via gadolinium-enhanced brain MRI using modified RECIST criteria.~Complete Response (CR) - Disappearance of all target and nontarget lesions~Partial Response (PR) - at least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum longest diameter AND an absolute decrease of at least 5mm in at least one target lesion.~Stable Disease (SD) - neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum of the longest diameter since the treatment started.~Progressive Disease (PD) - at least a 20% increase in the sum LD of target lesions, taking as reference the smallest sum of the longest diameter recorded since the treatment started AND an absolute increase in size of at least 5 mm in at least one target lesion OR the appearance of one or more new lesions of at least 6 mm in size." (NCT01305941)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Everolimus +Vinorelbine + Trastuzumab01178

[back to top]

Functional Assessment Cancer Therapy- Breast (FACT-B) From Baseline to 9 Weeks of Treatment to Assess Impact of Everolimus in Combination With Trastuzumab and Vinorelbine on Quality of Life

The FACT-B is a 10-question self-report questionnaire subscale administered with the Functional Assessment of Cancer Therapy- General (FACT-G) which contains concerns relevant to patients with breast cancer. Each question has a value 0-4. For some questions a higher indicates better outcome and others are the opposite. The former are summed as is, the latter are reversed in value before adding, such that each domain ranges from 0 to 4 times the number of questions in the domain, with 0 indicating worst and the highest possible value indicating best outcome. Total scores on the FACT-B subscale range from 0 to 40 with lower scores indicating declining quality of life. The change from baseline is the difference in scores between the baseline and 9 week assessments. (NCT01305941)
Timeframe: 9 weeks

Interventionunits on a scale (Median)
Everolimus +Vinorelbine + Trastuzumab1.0

[back to top]

Extracranial Time to Progression

"To evaluate the extracranial time to progression as determined by RECIST 1.1 criteria after administration of everolimus in combination with trastuzumab and vinorelbine.~Progressive Disease (PD) - at least a 20% increase in the sum LD of target lesions, taking as reference the smallest sum of the longest diameter recorded since the treatment started AND an absolute increase in size of at least 5 mm in at least one target lesion OR the appearance of one or more new lesions of at least 6 mm in size." (NCT01305941)
Timeframe: 3 years

Interventionmonths (Median)
Everolimus +Vinorelbine + Trastuzumab4.01

[back to top]

Number of Participants Without New Bone Lesions After 12 Weeks of Treatment

(NCT01313559)
Timeframe: After 12 weeks of treatment

Interventionparticipants (Number)
Cohort A (Pasireotide)0
Cohort B (Pasireotide and Everolimus)0

[back to top]

Number of Participants With > 50% Decline From Baseline PSA Level

(NCT01313559)
Timeframe: After 12 weeks of treatment

Interventionparticipants (Number)
Cohort A (Pasireotide)2
Cohort B (Pasireotide and Everolimus)0

[back to top]

Number of Participants With Progression Free Survival (PFS) Based on RECIST 1.1 Criteria

Progression free survival (PFS) based on primary outcome criteria for disease progression. Patients without radiographic disease progression who permanently discontinue the study drugs will be censored (NCT01313559)
Timeframe: Assessed up to 30 days after completion of study treatment

Interventionparticipants (Number)
Cohort A (Pasireotide)0
Cohort B (Pasireotide and Everolimus)0

[back to top]

Number of Participants Alive and Progression Free After 12 Weeks of Treatment

Progression of disease is defined as disease progression by RECIST 1.1 criteria on CT scan (X-ray computed tomography), or appearance of > 2 new bone lesions on bone scan, or prostate-specific antigen (PSA) progression by Prostate Cancer Clinical Trials Working Group (PCWG2) criteria or death from any cause. (NCT01313559)
Timeframe: 12 weeks after treatment

Interventionparticipants (Number)
Cohort A (Pasireotide)0
Cohort B (Pasireotide and Everolimus)0

[back to top]

Percentage of Participants Progression-free

Tumors were assessed according to Response Evaluation Criteria in Solid tumors (RECIST) to determine progression-free status. Complete response (CR) is disappearance of all lesions (i.e. all evidence of disease, not just the target lesions) determined by 2 observations not less than 4 weeks apart; Partial response (PR) is > 30% decrease in the sum of longest diameters of target lesions compared to baseline, with response or stable disease observed in non-target lesions, and no new lesions; Stable disease (SD) is neither sufficient shrinkage to qualify for response or sufficient increase to qualify for progressive disease in target lesions, with response or stable disease observed in non-target lesions, and no new lesions; and Progressive disease (PD is: > 20% increase in the sum of longest diameters of target lesions compared to smallest sum longest diameter recorded. In addition, the sum must also demonstrate an absolute increase of at least 5mm. (NCT01317615)
Timeframe: 6 months

InterventionPercentage of participants (Number)
RAD001 Plus Paclitaxel/Carboplatin8.2

[back to top]

Overall Survival (OS)

OS was defined as the time from date of start of treatment to date of death due to any cause. (NCT01317615)
Timeframe: 12 months

InterventionDays (Median)
RAD001 Plus Paclitaxel/Carboplatin298

[back to top]

Percentage of Participants With Disease Control Rate (DCR)

DCR was defined as is the percentage of participants with a best overall response of CR or PR or SD. Complete response (CR) is disappearance of all lesions (i.e. all evidence of disease, not just the target lesions) determined by 2 observations not less than 4 weeks apart; Partial response (PR) is > 30% decrease in the sum of longest diameters of target lesions compared to baseline, with response or stable disease observed in non-target lesions, and no new lesions; Stable disease (SD) is neither sufficient shrinkage to qualify for response or sufficient increase to qualify for progressive disease in target lesions, with response or stable disease observed in non-target lesions, and no new lesions; and Progressive disease (PD is: > 20% increase in the sum of longest diameters of target lesions compared to smallest sum longest diameter recorded. In addition, the sum must also demonstrate an absolute increase of at least 5mm. (NCT01317615)
Timeframe: 3 months

InterventionPercentage of participants (Number)
RAD001 Plus Paclitaxel/Carboplatin73.5

[back to top]

Progression Free Survival (PFS)

PFS was defined as the time from the date of start of treatment to date of event defined as the first documented progression or death due to any cause. (NCT01317615)
Timeframe: 6 months

InterventionDays (Median)
RAD001 Plus Paclitaxel/Carboplatin132

[back to top]

Percentage of Participants With Overall Response Rate (ORR)

ORR was defined as is the proportion of participants with a best overall response of CR or PR. CR is disappearance of all lesions (i.e. all evidence of disease, not just the target lesions) determined by 2 observations not less than 4 weeks apart; Partial response. PR is > 30% decrease in the sum of longest diameters of target lesions compared to baseline, with response or stable disease observed in non-target lesions, and no new lesions. (NCT01317615)
Timeframe: 3 months

InterventionPercentage of participants (Number)
RAD001 Plus Paclitaxel/Carboplatin44.9

[back to top]

Percentage of Participants Progression-free

Tumors were assessed according to Response Evaluation Criteria in Solid tumors (RECIST) to determine progression-free status. Complete response (CR): disappearance of all lesions (i.e. all evidence of disease, not just the target lesions) determined by 2 observations not less than 4 weeks apart; Partial response (PR): > 30% decrease in the sum of longest diameters of target lesions compared to baseline, with response or stable disease observed in non-target lesions, and no new lesions; Stable disease (SD): neither sufficient shrinkage to qualify for response or sufficient increase to qualify for progressive disease in target lesions, with response or stable disease observed in non-target lesions, and no new lesions; Progressive disease (PD): > 20% increase in the sum of longest diameters of target lesions compared to smallest sum longest diameter recorded. In addition, the sum must also demonstrate an absolute increase of at least 5mm. (NCT01317615)
Timeframe: 3 months

InterventionPercentage of participants (Number)
RAD001 Plus Paclitaxel/Carboplatin49.0

[back to top]

Any Bleeding

(NCT01347554)
Timeframe: Two year

Interventionparticipants (Number)
Xience V Stent Group7
Endeavor Resolute Group11

[back to top]

Device-oriented Composite Outcome

defined as a composite of all-cause mortality, any MI (includes non-target vessel territory) and repeat revascularization (includes all target and non-target vessel) (NCT01347554)
Timeframe: Two years

Interventionparticipants (Number)
Xience V Stent Group20
Endeavor Resolute Group24

[back to top]

Device-oriented Composite Outcome

defined as a composite of cardiac death, Myocardial infarction not clearly attributable to a nontarget vessel and target lesion revascularization (NCT01347554)
Timeframe: Two year

Interventionparticipants (Number)
Xience V Stent Group12
Endeavor Resolute Group13

[back to top]

Stent Thrombosis

Definite and probable stent thrombosis (NCT01347554)
Timeframe: Two years

Interventionparticipants (Number)
Xience V Stent Group0
Endeavor Resolute Group7

[back to top]

Number of Patients With Objective Radiographic Responses Based on Volumetric MRI Measurements (In Stratum 2 Only)

"Response was assessed at the time that a follow up volumetric MRI scan is performed (after course 6 and then every 6 months and at the end of treatment).~Complete response (CR): complete resolution of all measurable or palpable PN for ≥ 28days and no appearance of new lesions.~Partial response (PR): A ≥ 20% reduction in the sum of the volume of all index PN lesions for ≥ 28days.~Stable disease (SD): A < 20% increase and < 20% decrease in the sum of the volume of all index PN lesions for ≥ 28days." (NCT01365468)
Timeframe: Screening, after course #6, then every 6 months and end of treatment(1 course=28days)

InterventionPatients (Number)
Complete ResponsePartial ResponseStable Disease
Stratum 2005

[back to top]

Number of Patients With Adverse Events Assessed by Common Toxicity Criteria for Adverse Events (CTCAE) V.04

Adverse events were assessed according to the NCI Common Toxicity Criteria for Adverse Events (CTCAE) version 4.0. If CTCAE grading does not exist for an adverse event, the severity of mild, moderate, severe, and life-threatening, corresponding to grades 1 - 4 respectively, were used. CTCAE grade 5 (death) was not used in this study. (NCT01365468)
Timeframe: From the time ICF was signed until 28 days after End of Treatment (up to a maximum of 25 months)

,
InterventionPatients (Number)
At least one Grade 1 AEAt least one Grade 2 AEAt least one Grade 3 AEAt least one Grade 4 AE
Stratum 14411
Stratum 25500

[back to top]

Time to Disease Progression (TTP) Based on Change in Volumetric MRI Measurements in Children and Adults (In Stratum I Only)

This endpoint was planned to be analyzed for only Stratum 1 patients. Progression of disease defined as a ≥ 20% increase in the volume (by volumetric MRI) of at least one of the index plexiform neurofibromas (PN) compared to the pretreatment volume measured prior to the start of the current treatment phase. (NCT01365468)
Timeframe: Screening, after course #6, #12, #18, #24, End of Treatment(1 course=28days)

InterventionDays (Median)
Stratum 1NA

[back to top]

Overall Survival (OS) Using Kaplan Meier Method

Overall survival was defined as the time from date of randomization/start of treatment to date of death due to any cause. If a patient is not known to have died, survival was to be censored at the date of last contact. (NCT01374451)
Timeframe: Once 80 PFS events had occurred

,
InterventionPercentage of participants (Number)
6 months12 months18 months24 months
Everolimus93.786.175.571.0
Paseriotide LAR + Everolimus93.585.581.477.0

[back to top]

Progression-free Survival (PFS) Per Local Radiological Review

PFS per RECIST 1.0. (Response Evaluation Criteria in Solid Tumors). PFS was defined as the time from the date of randomization to the date of the first radiologically documented disease progression or death due to any cause. (NCT01374451)
Timeframe: Once 80 PFS events had occurred aproximately after 24 months

Interventionmonths (Median)
Paseriotide LAR + Everolimus16.82
Everolimus16.59

[back to top]

Summary of Pharmacokinetics (PK) for Everolimus for AUClast

(NCT01374451)
Timeframe: Cycle 2 Day 1

Interventionng*hr/mL (Mean)
Paseriotide LAR + Everolimus511
Everolimus378

[back to top]

Summary of Pharmacokinetics (PK) for Everolimus for CL/F

(NCT01374451)
Timeframe: Cycle 2 Day 1

InterventionL/hr (Mean)
Paseriotide LAR + Everolimus20.0
Everolimus29.0

[back to top]

Summary of Pharmacokinetics (PK) for Everolimus for Cmax and Cmin

(NCT01374451)
Timeframe: Cycle 2 Day 1

,
Interventionng/mL (Mean)
CmaxCmin
Everolimus60.27.67
Paseriotide LAR + Everolimus58.714.7

[back to top]

Summary of Pharmacokinetics (PK) for Everolimus for Tmax

(NCT01374451)
Timeframe: Cycle 2 Day 1

Interventionhr (Median)
Paseriotide LAR + Everolimus1.00
Everolimus0.500

[back to top]

Safety and Tolerability Profile of Everolimus Alone or in Combination With Pasireotide LAR

Consisted of monitoring and recording the rate, type, severity, and causal relationship of adverse events (AEs) and serious AEs (SAEs) to treatment. The safety analysis was based mainly on the frequency of AEs or SAEs and on the number of laboratory values that fell outside of pre-determined range. (NCT01374451)
Timeframe: Once 80 PFS events had occurred

,
InterventionParticipants (Number)
DeathsSerious Adverse EventsAdverse Events
Everolimus104981
Paseriotide LAR + Everolimus74178

[back to top]

Summary of Pasireotide Concentrations Following Intramuscular Injection of Pasireotide LAR 60mg

(NCT01374451)
Timeframe: Cycle 1 Day 21, Cycle 2 Day 29

Interventionng/mL (Mean)
Cycle 1 Day 21 (n:69)Cycle 2 Day 29 (n: 64)
Paseriotide LAR + Everolimus21.619.9

[back to top]

Objective Response Rate (ORR) as Per Radiology Review

"Objective response was determined by the local radiologist according to the RECIST Version 1.0. ORR is the percentage of patients with a best overall response of complete response (CR) or partial response (PR). This is also referred to as Overall response rate.~CR: Disappearance of all nontarget lesions. PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the smallest sum of the longest diameter of all target lesions recorded at or after baseline." (NCT01374451)
Timeframe: Once 80 PFS events had occurred

InterventionPercentage of participants (Number)
Paseriotide LAR + Everolimus20.3
Everolimus6.2

[back to top]

Disease Control Rate (DCR) as Per Radiology Review

Disease control rate is the percentage of patients with a best overall response of CR or PR or stable disease (SD) determined by the local radiologist according to the Response Evaluation Criteria In Solid Tumors Criteria (RECIST) Version 1.0. CR: Disappearance of all nontarget lesions. PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the smallest sum of the longest diameter of all target lesions recorded at or after baseline. SD: Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for progressive disease (PD). PD: Any progression ≤ 18 weeks after randomization (and not qualifying for CR, PR or stable disease SD. (NCT01374451)
Timeframe: Once 80 PFS events had occurred

InterventionPercentage of participants (Number)
Paseriotide LAR + Everolimus77.2
Everolimus82.7

[back to top]

Number of Participants With Adverse Events

Number of participants with adverse events as a measure of safety and tolerability (NCT01376310)
Timeframe: Until 30 days after the last dose of study treatment. Subjects may have continued to receive study treatment until disease progression, death, unacceptable toxicity or until locally commercially available. The maximum duration of exposure was 76 months.

,
InterventionParticipants (Count of Participants)
Adverse EventsTreatment-Related Adverse EventsSerious Adverse EventsTreatment-Related Serious Adverse Events
Cohort A (GSK1120212 < 24 Weeks)119101268
Cohort B (GSK1120212 >= 24 Weeks)3026134

[back to top]

Time to Progression (TTP) Based on the Modified RECIST Criteria

"Time to Progression (TTP) defined as the time from the date of randomization to the date of first documented radiological confirmation of disease progression based on modified RECIST criteria. Progressive Disease: >20% increase in sum of the longest diameters (SLD) of viable target lesion (arterial phase enhancement)" (NCT01379521)
Timeframe: 3, 6, 12, 18 and 24 months

Interventionmonths (Median)
Everolimus + TACE6.3
Placebo + TACE6.4

[back to top]

Percentage of Participants With a Decrease in the Sum of the of Longest Diameters (SLD) of Target Lesions From Baseline to 30 Months

Percentage of participants with a decrease in the sum of the of longest diameters (SLD) of target lesions from Baseline to 30 months (NCT01379521)
Timeframe: baseline, 30 months

InterventionPercentage of participants (Number)
Everolimus + TACE93.8
Placebo + TACE88.0

[back to top]

Overall Survival (OS)

Overall survival was defined as the time from date of randomization to date of death due to any cause. If death had not occurred at the date of the analysis cut-off then OS was censored at the date of the last contact. (NCT01379521)
Timeframe: 6, 12, 18, 24, 30 months

Interventionmonths (Median)
Everolimus + TACE29.9
Placebo + TACE21.7

[back to top]

Participants Evaluated for Toxicity

Toxicity will be assessed using Common Terminology Criteria for Adverse Events (CTCAE) v4.0 (Gompertz 1825),. Toxicities will be summarized by type and grade using frequencies and rates. (NCT01399918)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Everolimus and Bevacizumab55

[back to top]

Secondary Endpoint Will be the Overall Response Rate (ORR)

per the international criteria defined by the Response Evaluation Criteria in Solid Tumors Committee (NCT01399918)
Timeframe: 1 year

Interventionpercent of participants with ORR (Number)
Everolimus and Bevacizumab35

[back to top]

To Evaluate the Efficacy of Combining Everolimus and Bevacizumab in Patients With Advanced RCC of Non-clear Cell Histology

the percent of patients alive and progression-free after 6 months of therapy. (NCT01399918)
Timeframe: 6 months

Interventionpercent of participants (Number)
Everolimus and Bevacizumab78

[back to top]

Incidence of Bacterial, Viral, and Fungal Infections at Month 12

Incidence of bacterial, viral, and fungal infections at Month 12 (NCT01404325)
Timeframe: Month 12

,
Interventionincidences (Number)
Bacterial infectionsViral infectionsFungal infections
Centre Specific Triple IS Regimen4200
Quadruple Low Level IS Regimen3143

[back to top]

Incidence of Acute Rejection Episodes at Month 6 and Month 12

"Incidence of acute rejection episodes at Month 6 and Month 12. This table counts multiple occurrences of rejection in the same patient as different incidents. Every incident is associated with both an A and a B classification.~Classification A: Acute Rejection Grade 0 - none, Grade 1-minimal, Grade 2- mild, Grade 3-moderate, Grade 4-Severe; Classification B: Airway Inflammation-Grade 0-none, Grade 1R-low grade, Grade 2R-high grade, Grade X-ungradeable" (NCT01404325)
Timeframe: Month 6, Month 12

,
Interventionincidences (Number)
Up to 6 months: Classification AUp to 6 months: Classification B6 to 12 months: Classification A6 to 12 months: Classification B
Centre Specific Triple IS Regimen131399
Quadruple Low Level IS Regimen11111010

[back to top]

High-Density Lipoprotein (HDL)Cholesterol Levels at Month 1, 3, 6, 9, 12

HDL Cholesterol levels at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionmmol/L (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen-0.1-0.0-0.0-0.0-0.0
Quadruple Low Level IS Regimen0.10.10.10.00.0

[back to top]

Exercise Capacity 6-Minute Walk Test(6MWT) at Month 6 and Month 12

Exercise capacity (6MWT) at Month 6 and Month 12 Measured by the Borg Scale. THE BORG SCALE 0 is Nothing at all, 0.5 is Very, very slight (just noticeable); 1 is Very slight, 2 is Slight (light); 3 is Moderate; 4 is Somewhat severe; 5 is Severe (heavy), 7 is Very severe, 10 is Very, very severe (maximal)The higher the Borg score implies increased shortness of breath and/or increased fatigue. (NCT01404325)
Timeframe: Month 6, Month 12

,
InterventionScores on a scale (Mean)
Month 6-Borg score - Before start of testMonth 6-Borg score - At end of testMonth 12-Borg score - Before start of testMonth 12-Borg score - At end of test
Centre Specific Triple IS Regimen0.391.640.422.06
Quadruple Low Level IS Regimen0.471.750.382.08

[back to top]

Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula at Month 1, 3, 6, 9, 12

Calculated Glomerular Filtration Rate (cGFR) Using Modification of Diet in Renal Disease (MDRD) Formula at Month 1, 3, 6, 9, 12 cGFR (in mL/min/1.73 m2) = 186.3*(C-1.154)*(A-0.203)*G*R where C = the serum concentration of creatinine (mg/dL), A = age (years), G = 0.742 when gender is female, otherwise G = 1, R = 1.21 when race is black, otherwise R = 1. (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
InterventionmL/min (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen65.863.362.361.760.5
Quadruple Low Level IS Regimen72.068.767.667.467.0

[back to top]

Calculated Glomerular Filtration Rate (cGFR) According to Cystatin C-based Hoek's Formula at Month 1, 3, 6, 9, 12

Calculated Glomerular Filtration Rate (cGFR) according to Cystatin C-based Hoek's formula at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
InterventionmL/min (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen61.860.358.757.757.5
Quadruple Low Level IS Regimen68.565.763.061.860.8

[back to top]

Calculated Glomerular Filtration Rate (cGFR) According to Chronic Kidney Disease Epidemiology Collaboration (CKD EPI) at Month 1, 3, 6, 9, 12

Calculated Glomerular Filtration Rate (cGFR) according to Chronic Kidney Disease Epidemiology Collaboration (CKD EPI) at Month 1, 3, 6, 9 and 12. The CKD-EPI equation, expressed as a single equation, is GFR = 141 × min(Scr/κ, 1)α × max(Scr/κ, 1)^-1.209 × 0.993Age × 1.018 (if female) × 1.159 (if black), where Scr is serum creatinine, κ is 0.7 for females and 0.9 for males, α is -0.329 for females and -0.411 for males, min indicates the minimum of Scr/ĸ or 1, and max indicates the maximum of Scr/κ or 1 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
InterventionmL/min (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen67.164.363.162.361.2
Quadruple Low Level IS Regimen73.570.469.269.068.3

[back to top]

Adherence to Target Ranges of Tacrolimus at Month 1, 3, 6, 9, 12

Adherence to target ranges of Tacrolimus at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionparticipant adherence (Number)
Month 1 Below Target RangeMonth 1 Within Target RangeMonth 1 Above Target RangeMonth 3 Below Target RangeMonth 3 Within Target RangeMonth 3 Above Target RangeMonth 6 Below Target RangeMonth 6 Within Target RangeMonth 6 Above Target RangeMonth 9 Below Target RangeMonth 9 Within Target RangeMonth 9 Above Target RangeMonth 12 Below Target RangeMonth 12 Within Target RangeMonth 12 Above Target Range
Centre Specific Triple IS Regimen04500470145004600470
Quadruple Low Level IS Regimen3241732318523122181832315

[back to top]

Adherence to Target Ranges of Everolimus at Month 1, 3, 6, 9, 12

Adherence to target ranges of everolimus at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

Interventionparticipant adherence (Number)
Month 1 Below Target RangeMonth 1 Within Target RangeMonth 1 Above Target RangeMonth 3 Below Target RangeMonth 3 Within Target RangeMonth 3 Above Target RangeMonth 6 Below Target RangeMonth 6 Within Target RangeMonth 6 Above Target RangeMonth 9 Below Target RangeMonth 9 Within Target RangeMonth 9 Above Target RangeMonth 12 Below Target RangeMonth 12 Within Target RangeMonth 12 Above Target Range
Quadruple Low Level IS Regimen1042145411573912534133389

[back to top]

Adherence to Target Ranges of Cyclosporine A (CsA) at Month 1, 3, 6, 9, 12

Adherence to target ranges of Cyclosporine A (CsA) at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionparticipant adherence (Number)
Month 1 Below Target RangeMonth 1 Within Target RangeMonth 1 Above Target RangeMonth 3 Below Target RangeMonth 3 Within Target RangeMonth 3 Above Target RangeMonth 6 Below Target RangeMonth 6 Within Target RangeMonth 6 Above Target RangeMonth 9 Below Target RangeMonth 9 Within Target RangeMonth 9 Above Target RangeMonth 12 Below Target RangeMonth 12 Within Target RangeMonth 12 Above Target Range
Centre Specific Triple IS Regimen88041006804903100
Quadruple Low Level IS Regimen01830182018201610165

[back to top]

Calculated Glomerular Filtration Rate (cGFR) According to Chronic Kidney Disease Epidemiology Collaboration (CKD EPI) at 12 Months

Calculated Glomerular Filtration Rate (cGFR) according to Chronic Kidney Disease Epidemiology Collaboration (CKD EPI) at 12 months The CKD-EPI equation, expressed as a single equation, is GFR = 141 × min(Scr/κ, 1)α × max(Scr/κ, 1)^-1.209 × 0.993Age × 1.018 (if female) × 1.159 (if black), where Scr is serum creatinine, κ is 0.7 for females and 0.9 for males, α is -0.329 for females and -0.411 for males, min indicates the minimum of Scr/ĸ or 1, and max indicates the maximum of Scr/κ or 1 (NCT01404325)
Timeframe: Month 12

InterventionmL/min (Least Squares Mean)
Quadruple Low Level IS Regimen64.5
Centre Specific Triple IS Regimen54.6

[back to top]

Calculated Glomerular Filtration Rate (cGFR) According to Cockcroft-Gault at Month 1, 3, 6, 9, 12

Calculated Glomerular Filtration Rate (cGFR) according to Cockcroft-Gault at Month 1, 3, 6, 9, 12 For men: GFR=(140-Age) x Body weight (kg) / 72 x Serum Creatinine (mg/dl) For women: GFR=0.85 (140 -Age) x Body weight(kg)/ 72 x Serum Creatinine (mg/dl) (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
InterventionmL/min (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen72.170.169.169.068.4
Quadruple Low Level IS Regimen78.676.176.276.276.2

[back to top]

Trough Levels of Everolimus at Month 1, 3, 6, 9, 12

Trough levels of everolimus at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

Interventionng/mL (Mean)
Month 1Month 3Month 6Month 9Month 12
Quadruple Low Level IS Regimen4.24.44.14.54.3

[back to top]

Trough Levels of Tacrolimus at Month 1, 3, 6, 9, 12

Trough levels of Tacrolimus at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionng/mL (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen10.4410.5310.0910.679.66
Quadruple Low Level IS Regimen5.075.184.705.785.19

[back to top]

Incidence of Patients Experiencing a Decline in GFR of < 10, 10-15, 15-20, 20-25 and > 25 mL/Min From Baseline to Month 6 and 12.

Incidence of patients experiencing a decline in GFR of < 10, 10-15, 15-20, 20-25 and > 25 mL/min from Baseline to Month 6 and 12calculated by the CKD-EPI method. Participants are counted in each decline level observed for that participant; this means participants may be counted in more than 1 decline level. (NCT01404325)
Timeframe: Baseline, Month 6, Month 12

,
Interventionincidences (Number)
Up to Month 6: < 10 mL/minUp to Month 6: 10 - < 15 mL/minUp to Month 6: 15 - < 20 mL/minUp to Month 6: 20 - < 25 mL/minUp to Month 6: > 25 mL/minUp to Month 12: < 10 mL/minUp to Month 12: 10 - < 15 mL/minUp to Month 12: 15 - < 20 mL/minUp to Month 12: 20 - < 25 mL/minUp to Month 12: > 25 mL/min
Centre Specific Triple IS Regimen4418117753271899
Quadruple Low Level IS Regimen2896233610887

[back to top]

Trough Levels of Cyclosporine A (CsA) at Month 1, 3, 6, 9, 12

Trough levels of Cyclosporine A (CsA) at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionng/mL (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen106109103107108
Quadruple Low Level IS Regimen6159.6558.5057.5969.33

[back to top]

Triglyceride Levels at Month 1, 3, 6, 9, 12

Triglyceride levels at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionmmol/L (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen2.02.12.02.02.1
Quadruple Low Level IS Regimen2.62.72.62.72.7

[back to top]

Total Cholesterol Levels at Month 1, 3, 6, 9, 12

Total Cholesterol levels at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionmmol/L (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen-0.2-0.1-0.2-0.1-0.1
Quadruple Low Level IS Regimen0.81.01.11.10.8

[back to top]

Quality of Life (QoL, SF36) at Month 6 and Month 12

Quality of Life (QoL, SF36) at Month 6 and Month 12 Scores can range from a minimum of 0 (maximum disability) to a maximum of 100 (no disability). (NCT01404325)
Timeframe: Month 6, Month 12

,
Interventionscores on a scale (Mean)
Month 6: Physical component summary scoreMonth 6: Mental component summary scoreMonth 6: Physical functioningMonth 6: Role-PhysicalMonth 6: Bodily painMonth 6: General health perceptionMonth 6: VitalityMonth 6:Social functioningMonth 6: Role-EmotionalMonth 6: Mental healthMonth 12: Physical component summary scoreMonth 12: Mental component summary scoreMonth 12: Physical functioningMonth 12: Role-PhysicalMonth 12: Bodily PainMonth 12: General health perceptionMonth 12: VitalityMonth 12: Social functioningMonth 12: Role-EmotionalMonth 12: Mental health
Centre Specific Triple IS Regimen48.952.684.177.181.370.670.088.984.881.047.753.882.578.080.165.869.990.586.281.8
Quadruple Low Level IS Regimen46.951.578.871.176.465.467.484.184.778.547.249.777.468.376.866.765.782.379.475.4

[back to top]

Incidence of Treated Arterial Hypertension up to Month 12

Incidence of treated of arterial hypertension up to Month 12 (NCT01404325)
Timeframe: up to Month 12

,
Interventionincidences (Number)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen11111
Quadruple Low Level IS Regimen22234

[back to top]

Incidence of Renal Replacement Therapy at Month 6 and Month 12

Incidence of renal replacement therapy at Month 6 and Month 12: There were no incidences in either group where renal replacement therapy was required (NCT01404325)
Timeframe: Month 6, Month 12

,
Interventionincidences (Number)
up to Month 6up to Month 12
Centre Specific Triple IS Regimen00
Quadruple Low Level IS Regimen00

[back to top]

Incidence of Graft Loss/Re-transplantation at Month 6 and Month 12

Incidence of graft loss/re-transplantation at Month 6 and Month 12 (NCT01404325)
Timeframe: Month 6, Month 12

,
Interventionincidences (Number)
Month 6Month 12
Centre Specific Triple IS Regimen01
Quadruple Low Level IS Regimen01

[back to top]

Low-Density Lipoprotein (LDL)Cholesterol Levels at Month 1, 3, 6, 9, 12

LDL Cholesterol levels at Month 1, 3, 6, 9, 12 (NCT01404325)
Timeframe: Month 1, 3, 6, 9, 12

,
Interventionmmol/L (Mean)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen0.0-0.0-0.00.10.1
Quadruple Low Level IS Regimen0.40.60.70.80.5

[back to top]

Incidence of Diabetes Mellitus up to Month 12

Incidence of Diabetes Mellitus up to Month 12 (NCT01404325)
Timeframe: up to Month 12

,
Interventionincidences (Number)
Month 1Month 3Month 6Month 9Month 12
Centre Specific Triple IS Regimen00111
Quadruple Low Level IS Regimen00011

[back to top]

Incidence of Death at Month 6 and Month 12

Incidence of death at Month 6 and Month 12 (NCT01404325)
Timeframe: Month 6, Month 12

,
Interventionincidences (Number)
Month 6Month 12
Centre Specific Triple IS Regimen01
Quadruple Low Level IS Regimen03

[back to top]

Incidence of Bronchiolitis Obliterans Syndrome (BOS) at Month 6 and Month 12

Incidence of Bronchiolitis obliterans syndrome (BOS) at Month 6 and Month 12 BOS 0 is FEV1 > 90% of baseline and FEF25-75% > 75% of baseline; BOS 0-p is FEV1 81-90% of baseline and/or FEF25-75% ≤ 75% of baseline; BOS 1 is FEV1 66-80% of baseline; BOS 2 is FEV1 51-65% of baseline; BOS 3 is FEV1 ≤ 50% of baseline (NCT01404325)
Timeframe: Month 6, Month 12

,
Interventionincidences (Number)
Month 6 - Total (all grades)Month 6 - BOS 0Month 6 - BOS 0pMonth 6 - BOS 1Month 6 - BOS 2Month 6 - BOS 3Month 6 - BOS (>/=1)Month 12 - Total (all grades)Month 12 - BOS 0Month 12 - BOS 0pMonth 12 - BOS 1Month 12 - BOS 2Month 12 - BOS 3Month 12 - BOS (>/=1)
Centre Specific Triple IS Regimen61401450276138145139
Quadruple Low Level IS Regimen66362631046635263115

[back to top]

Change From Baseline in Serum Creatinine - ITT

Blood samples were collected to assess serum creatinine measurements. A negative change from baseline indicates improvement. (NCT01410448)
Timeframe: baseline, 3 months

Interventionmg/dL (Mean)
Immediate Everolimus (IE)-4.79
Delayed Everolimus (DE)-5.13

[back to top]

Change From Baseline in Serum Creatinine - Modified ITT

Blood samples were collected to assess serum creatinine measurements. A negative change from baseline indicates improvement. (NCT01410448)
Timeframe: baseline, 12 months

Interventionmg/dL (Mean)
Immediate Everolimus (IE)-4.96
Delayed Everolimus (DE)-5.22

[back to top]

Percentage of Participants With Proteinuria

Incidence of proteinuria (>1,000 mg/day in urine collected in 24 hours or > 1.0 if measured on the urine protein/creatinine concentration ratio in a spot urine sample) was assessed. (NCT01410448)
Timeframe: 3 months

,
InterventionPercentage of participants (Number)
YesNoMissing
Delayed Everolimus (DE)4.2168.4227.37
Immediate Everolimus (IE)4.1568.9126.94

[back to top]

Percentage of Participants Who Experienced Treatment Failure - Worst-case Scenario

The percentage of participants who experienced treatment failure was assessed. Treatment failure was defined as the occurrence of at least one failure event among death, graft loss or biopsy-proven acute rejection (BPAR). In the worst-case scenario, treatment failure was identified in one of the following cases: occurrence of at least one treatment failure event or study discontinuation due to any reason. (NCT01410448)
Timeframe: 3 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)11.40
Delayed Everolimus (DE)21.05

[back to top]

Percentage of Participants With a New Onset of Diabetes

The percentage of participants with a new onset of diabetes was assessed. (NCT01410448)
Timeframe: 12 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)3.14
Delayed Everolimus (DE)4.05

[back to top]

Percentage of Participants With a New Onset of Malignancy

The percentage of participants with a new onset of malignancy was assessed. (NCT01410448)
Timeframe: 12 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)0
Delayed Everolimus (DE)0.68

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate (eGFR) (Calculated With Modified Diet in Renal Disease (MDRD)-4 Formula - ITT

Renal function was assessed by measuring serum creatinine and serum urea and by calculating creatinine clearance using the MDRD-4 formula. eGFR = 186.3*(serum creatinine [mg/dL])^-1.154 * (age at screening) -0.203 * (0.742 if female) * (1.21 if African American). A positive change from baseline indicates improvement. (NCT01410448)
Timeframe: baseline, 3 Months

InterventionmL/min (Mean)
Immediate Everolimus (IE)38.64
Delayed Everolimus (DE)39.13

[back to top]

Percentage of Participants With Acute Rejection (AR)

AR was defined as an episode of increased serum creatinine >30% that was clinically diagnosed as an acute rejection but was not biopsy proven. (NCT01410448)
Timeframe: 12 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)12.44
Delayed Everolimus (DE)10.53

[back to top]

Participant/Graft Survival Rate: Percentage of Participants With Failure Events of Death or Graft Loss - Worst-case Scenario

The percentage of participants who experienced death or graft loss was assessed. In the worst-case scenario, failure, i.e. participants death or graft loss, was identified in one of the following cases: occurrence of at least one failure event or study discontinuation due to any reason. (NCT01410448)
Timeframe: 3 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)6.74
Delayed Everolimus (DE)18.42

[back to top]

Percentage of Participants Without Wound Healing Complications - Worst-case Scenario

The percentage of participants without wound healing complications was assessed. Wound healing complications consisted of lymphorrhea, fluid collections, wound dehiscence, wound infections and incisional hernia. In the worst-case scenario, failure, i.e. at least one healing complication occurrence, was identified in one of the following cases: wound complication occurrence, missing information about wound complication occurrence or study discontinuation due to any reason for participants who did not complete the 12 month follow-up visit. (NCT01410448)
Timeframe: 12 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)65.80
Delayed Everolimus (DE)59.47

[back to top]

Duration of DGF

The duration of DGF was defined as the elapsed time from first to last day of post-transplant dialysis. (NCT01410448)
Timeframe: 3 months

InterventionDays (Median)
Immediate Everolimus (IE)8.50
Delayed Everolimus (DE)5.50

[back to top]

Patient Survival Rate: Percentage of Deaths - Worst-case Scenario

The percentage of deaths was assessed. In the worst-case scenario, failure, i.e. death, was identified in one of the following cases: participant's death or study discontinuation due to any reason. (NCT01410448)
Timeframe: 3 Months, 12 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)6.22
Delayed Everolimus (DE)18.42

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate (eGFR) (Calculated With Modified Diet in Renal Disease (MDRD)-4 Formula - Modified ITT

Renal function was assessed by measuring serum creatinine and serum urea and by calculating creatinine clearance using the MDRD-4 formula. eGFR = 186.3*(serum creatinine [mg/dL])^-1.154 * (age at screening) -0.203 * (0.742 if female) * (1.21 if African American). A positive change from baseline indicates improvement. (NCT01410448)
Timeframe: baseline, 12 months

InterventionmL/min (Mean)
Immediate Everolimus (IE)41.26
Delayed Everolimus (DE)41.56

[back to top]

Percentage of Participants Without Wound Healing Complications - Worst-case Scenario

The percentage of participants without wound healing complications was assessed. Wound healing complications consisted of lymphorrhea, fluid collections, wound dehiscence, wound infections and incisional hernia. In the worst-case scenario, failure, i.e. at least one healing complication occurrence, was identified in one of the following cases: wound complication occurrence, missing information about wound complication occurrence, or study discontinuation due to any reason. (NCT01410448)
Timeframe: 3 months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)68.39
Delayed Everolimus (DE)61.58

[back to top]

Graft Survival Rate: Percentage of Participants With Graft Loss - Worst-case Scenario

The percentage of participants who experienced graft loss was assessed. In the worst-case scenario, failure, i.e. graft loss, was identified in one of the following cases: occurrence of graft loss or discontinuation due to any reason. (NCT01410448)
Timeframe: 3 months, 12 months

,
InterventionPercentage of participants (Number)
3 months12 months
Delayed Everolimus (DE)18.4219.47
Immediate Everolimus (IE)6.747.25

[back to top]

Percentage of Participants With BPAR - Worst-case Scenario

A biopsy-proven acute rejection was defined as a biopsy graded IA, IB, IIA, IIB or III. In the worst-case scenario, failure, i.e. BPAR, was identified in one of the following cases: occurrence of BPAR or study discontinuation due to any reason. (NCT01410448)
Timeframe: 3 Months, 12 months

,
InterventionPercentage of participants (Number)
3 monts12 months
Delayed Everolimus (DE)21.0524.74
Immediate Everolimus (IE)11.4015.54

[back to top]

Percentage of Participants With Delayed Graft Function (DGF) -

DGF was defined as the need for dialysis in the first week after transplant, excluding Renal Replacement Therapy within the first 24 hours after transplantation. (NCT01410448)
Timeframe: 3 Months

InterventionPercentage of participants (Number)
Immediate Everolimus (IE)23.83
Delayed Everolimus (DE)31.58

[back to top]

Audiologic Response

"Defined as improvement in speech discrimination score (SDS), defined as an improvement in the score above the 95% critical difference threshold, compared to baseline audiogram at initiation of treatment. Audiologic worsening: decrease in SDS score below the 95% critical difference threshold, compared to baseline audiogram at initiation of treatment.~Patients with vestibular schwannomas will receive baseline audiograms within 28 days before enrollments and subsequent audiograms at the time of each MRI." (NCT01419639)
Timeframe: 1 Year

Interventionparticipants (Number)
RAD0010

[back to top]

Change in Tumor Size From Baseline

(NCT01419639)
Timeframe: 1 Year

Intervention% of change in tumor size from baseline (Median)
RAD0015.72

[back to top]

Radiographic Response

To estimate the objective response rates to RAD001 in patients with NF2-related tumors including cranial nerve schwannomas, meningiomas and ependymomas. Radiographic response for study purposes = greater than or equal to 15% reduction in tumor volume in any of the target tumors (partial response). Complete disappearance of any of the target tumors = complete response. MRI of the brain and spine will be performed every 3 months. If an objective response (15% reduction in tumor volume compared to baseline) is observed in any target tumor or stable disease, drug will be continued. (NCT01419639)
Timeframe: 1 Year

Interventionparticipants (Number)
RAD0010

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™57
XIENCE™34

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™1
XIENCE™0

[back to top]

Number of Participants With All Revascularization

Revascularization: Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold. Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion. Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion. Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™59
XIENCE™34

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™76
XIENCE™40

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™80
XIENCE™41

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization. (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™24
XIENCE™15

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization. (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™19
XIENCE™9

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization. (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™38
XIENCE™21

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization. (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™68
XIENCE™39

[back to top]

Number of Participants With DMR (All Death, All MI, All Revascularization)

DMR is the composite of All Death, All MI, All Revascularization. (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™4

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™17
XIENCE™5

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™3

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™25
XIENCE™7

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™38
XIENCE™11

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™2

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™40
XIENCE™12

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™41
XIENCE™13

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE) (Cardiac Death, All MI, ID-TLR)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and ischemic driven target lesion revascularization (ID-TLR). (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™6
XIENCE™6

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™4
XIENCE™3

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™22
XIENCE™19

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™1
XIENCE™0

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™29
XIENCE™20

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™0
XIENCE™0

[back to top]

Number of Participants With Non-Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™30
XIENCE™20

[back to top]

Number of Participants With Procedural Success

Achievement of final in-scaffold/stent residual stenosis of less than 50% by QCA with successful delivery and deployment of at least one study scaffold/stent at the intended target lesion and successful withdrawal of the delivery system for all target lesions without the occurrence of cardiac death, target vessel MI or repeat TLR during the hospital stay. (NCT01425281)
Timeframe: From the start of index procedure to end of index procedure

InterventionParticipants (Count of Participants)
Absorb BVS™322
XIENCE™164

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, Target Vessel Myocardial Infarction (TV-MI), ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™16
XIENCE™5

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™3

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™23
XIENCE™5

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™34
XIENCE™8

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™37
XIENCE™9

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death, TV-MI, ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™38
XIENCE™9

[back to top]

Number of Participants With Target Lesion Failure (TLF) (Cardiac Death,(Target Vessel Myocardial Infarction(TV-MI), ID-TLR)

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™12
XIENCE™2

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated (CI) or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™2
XIENCE™0

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated (CI) or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™1
XIENCE™0

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated [CI] or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™4
XIENCE™3

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated [CI] or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™2
XIENCE™1

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated [CI] or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™9
XIENCE™3

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated [CI] or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™24
XIENCE™8

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated [CI] or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™27
XIENCE™8

[back to top]

Number of Participants With Target Lesion Revascularization (TLR)

"Target Lesion Revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. All TLR should be classified prospectively as clinically indicated [CI] or not clinically indicated by the investigator prior to repeat angiography. An independent angiographic core laboratory should verify that the severity of percent diameter stenosis meets requirements for clinical indication and will overrule in cases where investigator reports are not in agreement.~The target lesion is defined as the treated segment from 5 mm proximal to the stent and to 5 mm distal to the scaffold/stent." (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™28
XIENCE™8

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR) (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™45
XIENCE™21

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR) (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™47
XIENCE™22

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™18
XIENCE™8

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™15
XIENCE™5

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™28
XIENCE™11

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™41
XIENCE™20

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™3

[back to top]

Number of Participants With Target Vessel Failure (TVF) (Cardiac Death, All MI, ID-TVR)

Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or Ischemic-Driven Target Vessel Revascularization (ID-TVR). (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™8
XIENCE™8

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™4
XIENCE™4

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™15
XIENCE™9

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™33
XIENCE™19

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™2
XIENCE™2

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™38
XIENCE™19

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™41
XIENCE™19

[back to top]

Number of Participants With Target Vessel Revascularization (TVR)

Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself. (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™1
XIENCE™0

[back to top]

Number of Participants With Acute Stent/Scaffold Thrombosis

"Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points.~Timings:~Acute:0-24 hours;Subacute:>24 hours-30 days;Late:30 days-1 year;Very late:>1 year.~Definite stent thrombosis occurred by either angiographic/pathologic confirmation.~Angiographic confirmation:The presence of a thrombus that originates in the stent or in the segment 5 mm proximal or distal to the stent&presence of at least 1 of the following criteria within a 48-hour time window~-Acute onset of ischemic symptoms at rest;New ischemic ECG changes;Typical rise&fall in cardiac biomarkers;Nonocclusive/Occlusive thrombus~Pathological confirmation:Evidence of recent thrombus.~Probable stent thrombosis may occur after intracoronary stenting due to:~Unexplained death within first 30 days~Any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis&in the absence of any other obvious cause." (NCT01425281)
Timeframe: <=1 day

,
InterventionParticipants (Count of Participants)
DefiniteProbable
Absorb BVS™10
XIENCE™00

[back to top]

Number of Participants With Acute/Subacute Stent/Scaffold Thrombosis

"Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points.~Timings:~Acute:0-24 hours;Subacute:>24 hours-30 days;Late:30 days-1 year;Very late:>1 year.~Definite stent thrombosis occurred by either angiographic/pathologic confirmation.~Angiographic confirmation:The presence of a thrombus that originates in the stent or in the segment 5 mm proximal or distal to the stent&presence of at least 1 of the following criteria within a 48-hour time window~-Acute onset of ischemic symptoms at rest;New ischemic ECG changes;Typical rise&fall in cardiac biomarkers;Nonocclusive/Occlusive thrombus~Pathological confirmation:Evidence of recent thrombus.~Probable stent thrombosis may occur after intracoronary stenting due to:~Unexplained death within first 30 days~Any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis&in the absence of any other obvious cause." (NCT01425281)
Timeframe: 0-30 days

,
InterventionParticipants (Count of Participants)
DefiniteProbable
Absorb BVS™20
XIENCE™00

[back to top]

Number of Participants With Cumulative Stent/Scaffold Thrombosis

"Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points.~Timings:~Acute:0-24 hours;Subacute:>24 hours-30 days;Late:30 days-1 year;Very late:>1 year.~Definite stent thrombosis occurred by either angiographic/pathologic confirmation.~Angiographic confirmation:The presence of a thrombus that originates in the stent or in the segment 5 mm proximal or distal to the stent&presence of at least 1 of the following criteria within a 48-hour time window~-Acute onset of ischemic symptoms at rest;New ischemic ECG changes;Typical rise&fall in cardiac biomarkers;Nonocclusive/Occlusive thrombus~Pathological confirmation:Evidence of recent thrombus.~Probable stent thrombosis may occur after intracoronary stenting due to:~Unexplained death within first 30 days~Any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis&in the absence of any other obvious cause." (NCT01425281)
Timeframe: 0-1853 days

,
InterventionParticipants (Count of Participants)
DefiniteProbable
Absorb BVS™81
XIENCE™00

[back to top]

Number of Participants With Late Stent/Scaffold Thrombosis

"Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points.~Timings:~Acute:0-24 hours;Subacute:>24 hours-30 days;Late:30 days-1 year;Very late:>1 year.~Definite stent thrombosis occurred by either angiographic/pathologic confirmation.~Angiographic confirmation:The presence of a thrombus that originates in the stent or in the segment 5 mm proximal or distal to the stent&presence of at least 1 of the following criteria within a 48-hour time window~-Acute onset of ischemic symptoms at rest;New ischemic ECG changes;Typical rise&fall in cardiac biomarkers;Nonocclusive/Occlusive thrombus~Pathological confirmation:Evidence of recent thrombus.~Probable stent thrombosis may occur after intracoronary stenting due to:~Unexplained death within first 30 days~Any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis&in the absence of any other obvious cause." (NCT01425281)
Timeframe: 31-365 days

,
InterventionParticipants (Count of Participants)
DefiniteProbable
Absorb BVS™01
XIENCE™00

[back to top]

Number of Participants With Subacute Stent/Scaffold Thrombosis

"Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points.~Timings:~Acute:0-24 hours;Subacute:>24 hours-30 days;Late:30 days-1 year;Very late:>1 year.~Definite stent thrombosis occurred by either angiographic/pathologic confirmation.~Angiographic confirmation:The presence of a thrombus that originates in the stent or in the segment 5 mm proximal or distal to the stent&presence of at least 1 of the following criteria within a 48-hour time window~-Acute onset of ischemic symptoms at rest;New ischemic ECG changes;Typical rise&fall in cardiac biomarkers;Nonocclusive/Occlusive thrombus~Pathological confirmation:Evidence of recent thrombus.~Probable stent thrombosis may occur after intracoronary stenting due to:~Unexplained death within first 30 days~Any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis&in the absence of any other obvious cause." (NCT01425281)
Timeframe: > 1-30 days

,
InterventionParticipants (Count of Participants)
DefiniteProbable
Absorb BVS™10
XIENCE™00

[back to top]

Number of Participants With Very Late Stent/Scaffold Thrombosis

"Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points.~Timings:~Acute:0-24 hours;Subacute:>24 hours-30 days;Late:30 days-1 year;Very late:>1 year.~Definite stent thrombosis occurred by either angiographic/pathologic confirmation.~Angiographic confirmation:The presence of a thrombus that originates in the stent or in the segment 5 mm proximal or distal to the stent&presence of at least 1 of the following criteria within a 48-hour time window~-Acute onset of ischemic symptoms at rest;New ischemic ECG changes;Typical rise&fall in cardiac biomarkers;Nonocclusive/Occlusive thrombus~Pathological confirmation:Evidence of recent thrombus.~Probable stent thrombosis may occur after intracoronary stenting due to:~Unexplained death within first 30 days~Any MI that is related to documented acute ischemia in the territory of the implanted stent without angiographic confirmation of stent thrombosis&in the absence of any other obvious cause." (NCT01425281)
Timeframe: > 365 days

,
InterventionParticipants (Count of Participants)
DefiniteProbable
Absorb BVS™60
XIENCE™00

[back to top]

Number of Participants With Non Target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel. (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™10
XIENCE™11

[back to top]

Absolute Difference (3 Years Post Nitrate- 3 Years Pre Nitrate) In-Scaffold Mean Lumen Diameter (MLD)

In-scaffold:Within the margins of the scaffold. (NCT01425281)
Timeframe: 3 years

Interventionmm (Mean)
Absorb BVS™0.05
XIENCE™0.06

[back to top]

Absolute Difference (3 Years Post-nitrate - Post Procedure Post-nitrate) In-Scaffold Minimum Lumen Diameter

In-scaffold:Within the margins of the scaffold. (NCT01425281)
Timeframe: 3 years

Interventionmm (Mean)
Absorb BVS™-0.37
XIENCE™-0.25

[back to top]

Device Success

Successful delivery and deployment of the first study scaffold/stent the intended target lesion and successful withdrawal of the delivery system with attainment of final in-scaffold/stent residual stenosis of less than 50% by quantitative coronary angiography (QCA). (NCT01425281)
Timeframe: From the start of index procedure to end of index procedure

InterventionPercentage of lesions (Number)
Absorb BVS™99.5
XIENCE™100

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™0
XIENCE™1

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 180 Days

InterventionParticipants (Count of Participants)
Absorb BVS™0
XIENCE™1

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™4
XIENCE™1

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™8
XIENCE™6

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™0
XIENCE™0

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™11
XIENCE™7

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™7

[back to top]

Number of Participants Experiencing All Death (Cardiac, Vascular, Non-Cardiovascular)

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™0
XIENCE™0

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™3

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™2

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma. Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™35
XIENCE™12

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™22
XIENCE™5

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™33
XIENCE™11

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma. Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™37
XIENCE™13

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI) Q wave MI Development of new, pathological Q wave on the ECG. Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants Experiencing All Death/All MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™15
XIENCE™3

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI) Q wave MI: Development of new, pathological Q wave on the ECG. Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™29
XIENCE™9

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI) Q wave MI: Development of new, pathological Q wave on the ECG. Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™29
XIENCE™10

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI) Q wave MI: Development of new, pathological Q wave on the ECG. Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves" (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™15
XIENCE™2

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™2

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™20
XIENCE™4

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™28
XIENCE™8

[back to top]

Number of Participants Experiencing Cardiac Death/All MI

"Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), un witnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™2

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™15
XIENCE™2

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™2

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™19
XIENCE™4

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™27
XIENCE™5

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated Creatine kinase-MB (CK-MB) in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™14
XIENCE™2

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated Creatine kinase-MB (CK-MB) in the absence of new pathological Q waves." (NCT01425281)
Timeframe: In-hospital (≤ 7 days of post index procedure)

InterventionParticipants (Count of Participants)
Absorb BVS™13
XIENCE™2

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Absorb BVS™27
XIENCE™5

[back to top]

Number of Participants With All Myocardial Infarction (Per Protocol Definition)

"Myocardial Infarction (MI)~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01425281)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Absorb BVS™27
XIENCE™6

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Absorb BVS™12
XIENCE™12

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: 180 days

InterventionParticipants (Count of Participants)
Absorb BVS™7
XIENCE™6

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Absorb BVS™22
XIENCE™18

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Absorb BVS™49
XIENCE™33

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT01425281)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Absorb BVS™2
XIENCE™2

[back to top]

Maximum Tolerated Dose of Sorafenib for Participants With Recurrent Malignant Gliomas

Maximum tolerated dose of sorafenib for participants with recurrent malignant gliomas. MTD is defined as the dose at which fewer than one-third of participants experience a dose limiting toxicity (DLT) to one of the study drugs. A DLT is any grade 3 thrombocytopenia, grade 4 anemia, grade 4 neutropenia, or febrile neutropenia of any grade. Any non-hematologic grade 3 or grade 4 toxicity, excluding alopecia and/or hand/foot reaction. Failure to recover from toxicities (to grade 1 or less) to be eligible for re-treatment with sorafenib within 14 days of the last dose of sorafenib. (NCT01434602)
Timeframe: First 28 days of treatment only (cycle 1)

Interventionmg (Number)
All Participants400

[back to top]

Median Amount of Time a Participant Survives After Therapy for Participants With Recurrent Malignant Glioma (Group A, Group B, and Group C) Treated With Everolimus and Sorafenib Measured From the Time of Study Enrollment

Median amount of time a participant survives after therapy for participants with recurrent malignant glioma (Group A, Group B, and Group C) treated with everolimus and sorafenib measured from the time of study enrollment. (NCT01434602)
Timeframe: From the time of study enrollment up to 1.5 years

InterventionMonths (Median)
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily7.75
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily4.77
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily11.97

[back to top]

Median Amount of Time Participants Survives Without Disease Progression for Groups A, B, and C Treated With Everolimus and Sorafenib

Median amount of time participants survives without disease progression for Groups A, B, and C treated with everolimus and sorafenib. Progression was measured by brain magnetic resonance imaging (MRI) tumor measurements. Progression is defined as >25% increase in the sum of products of perpendicular diameters of enhancing lesions (over baseline if no decrease) on stable or increasing dose of corticosteroids; and/or significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation or therapy, not due to co-morbid events (radiation therapy, demyelination, ischemic injury, infection, seizures, postoperative changes, or other treatment effects). Any new lesion. (NCT01434602)
Timeframe: Up to 6 months

InterventionMonths (Median)
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily1.92
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily1.17
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily1.87

[back to top]

Number of Participants With a Dose-limiting Toxicity (DLT)

A DLT is any grade 3 thrombocytopenia, grade 4 anemia, grade 4 neutropenia, or febrile neutropenia of any grade. Any non-hematologic grade 3 or grade 4 toxicity, excluding alopecia and/or hand/foot reaction. Failure to recover from toxicities (to grade 1 or less) to be eligible for re-treatment with sorafenib and everolimus within 14 days of the last dose of sorafenib and everolimus. (NCT01434602)
Timeframe: First 28 days of treatment only (cycle 1)

InterventionParticipants (Count of Participants)
Phase I Dose Level I Sorafenib 400mg Twice Daily + Everolimus 5mg Daily3
Phase I Dose Level -1 Sorafenib 400mg Twice Daily, 7 Days on, 7 Off + Everolimus 5mg Daily1

[back to top]

Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0)

Here is the number of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0). A non-serious adverse event is any untoward medical occurrence. A serious adverse event is an adverse event or suspected adverse reaction that results in death, a life-threatening adverse drug experience, hospitalization, disruption of the ability to conduct normal life functions, congenital anomaly/birth defect or important medical events that jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the previous outcomes mentioned. (NCT01434602)
Timeframe: Date treatment consent signed to date off study, approximately 21 months and 18 days, 30 months and twelve days, 83 months and 8 days, and 84 months and 26 days for each group respectively.

InterventionParticipants (Count of Participants)
Phase I Dose Level I Sorafenib 400mg Twice Daily + Everolimus 5mg Daily6
Phase I Dose Level -1 Sorafenib 400mg Twice Daily, 7 Days on, 7 Off + Everolimus 5mg Daily7
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily12
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily38
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily20

[back to top]

Percentage of Participants Who Survived Without Disease Progression at 3 Months After Treatment for Glioblastoma Participants With Prior Bevacizumab Exposure

Percentage of participants who survived without disease progression at 3 months after treatment for glioblastoma participants with prior bevacizumab exposure. Progression was measured by brain magnetic resonance imaging (MRI) tumor measurements. Progression is defined as >25% increase in the sum of products of perpendicular diameters of enhancing lesions (over baseline if no decrease) on stable or increasing dose of corticosteroids; and/or significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation or therapy, not due to co-morbid events (radiation therapy, demyelination, ischemic injury, infection, seizures, postoperative changes, or other treatment effects). Any new lesion. (NCT01434602)
Timeframe: 3 months

InterventionPercentage of participants (Number)
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily12.9

[back to top]

Rate of Participants Symptom Interference With Function

To evaluate the occurrence of symptoms and correlate to disease progression and tolerance to treatment using the MD Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) self-reporting tool. The MDASI-BT consists of 23 symptoms rated on an 11-point scale (0 - did not interfere, to 10 - interfered completely) to indicate the symptoms that interfere with a participant's life in the last 24 hours. We calculated the mean symptom interference at the time of clinical evaluation. (NCT01434602)
Timeframe: Completed at baseline, before a brain or spine magnetic resonance imaging (MRI)/clinical evaluation, and at cycle 1-2, cycle 4, cycle 6, cycle 8, cycle 10 and cycle 12 (each cycle is 28 days)

Interventionscore on a scale (Mean)
BaselineCycle 1Cycle 2
Phase I Dose Level I Sorafenib 400mg Twice Daily + Everolimus 5mg Daily5.04.25.0

[back to top]

Percentage of Participants Who Survived Without Disease Progression at 6 Months After Treatment for Anaplastic Glioma (AG) Participants With no Prior Bevacizumab Exposure

Percentage of participants who survived without disease progression at 6 months after treatment for anaplastic glioma (AG) participants with no prior bevacizumab exposure. Progression was measured by brain magnetic resonance imaging (MRI) tumor measurements. Progression is defined as >25% increase in the sum of products of perpendicular diameters of enhancing lesions (over baseline if no decrease) on stable or increasing dose of corticosteroids; and/or significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation or therapy, not due to co-morbid events (radiation therapy, demyelination, ischemic injury, infection, seizures, postoperative changes, or other treatment effects). Any new lesion. (NCT01434602)
Timeframe: 6 months

InterventionPercentage of participants (Number)
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily26.7

[back to top]

Percentage of Participants Who Survived Without Disease Progression at 6 Months After Treatment for Glioblastoma Participants With no Prior Bevacizumab Exposure

Percentage of participants who survived without disease progression at 6 months after treatment for glioblastoma participants with no prior bevacizumab exposure. Progression was measured by brain magnetic resonance imaging (MRI) tumor measurements. Progression is defined as >25% increase in the sum of products of perpendicular diameters of enhancing lesions (over baseline if no decrease) on stable or increasing dose of corticosteroids; and/or significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation or therapy, not due to co-morbid events (radiation therapy, demyelination, ischemic injury, infection, seizures, postoperative changes, or other treatment effects). Any new lesion. (NCT01434602)
Timeframe: 6 months

InterventionPercentage of participants (Number)
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily8.33

[back to top]

Objective Response for Participants With Recurrent Malignant Gliomas (Group A, Group B, and Group C) Treated With Everolimus and Sorafenib

Objective response for participants with recurrent malignant gliomas treated with everolimus and sorafenib was assessed by brain magnetic resonance imaging (MRI) tumor measurements. Complete Response is complete disappearance of all CE measurable and evaluable disease. Partial Response is greater than or equal to 50% decrease under baseline in the sum of products of perpendicular diameters of the two largest CE measurable lesions, sustained for at least 4 weeks. Progressive Disease >25% increase in the sum of products of perpendicular diameters of enhancing lesions (over baseline if no decrease) on stable or increasing dose of corticosteroids, significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation or therapy, not due to co-morbid events, or any new lesion. Stable Disease does not qualify for CR, PR, or progression. (NCT01434602)
Timeframe: After 1 cycle of treatment, or those who exhibit objective progression prior the end of cycle 1 (one cycle = 28 days).

,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseProgressive DiseaseStable DiseaseNot Evaluable
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on 7 Off + Everolimus 5mg Daily00480
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily0017167
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily027102

[back to top]

Rate of Participants Symptom Severity

To evaluate the occurrence of symptoms and correlate to disease progression and tolerance to treatment using the MD Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) self-reporting tool. The MDASI-BT consists of 23 symptoms rated on an 11-point scale (0 - not present, to 10 - as bad as you can imagine) to indicate the presence and severity of the symptom. We calculated the mean core symptom severity at the time of clinical evaluation. (NCT01434602)
Timeframe: Completed at baseline, before a brain or spine magnetic resonance imaging (MRI)/clinical evaluation, and at cycle 1-2, cycle 4, cycle 6, cycle 8, cycle 10 and cycle 12 (each cycle is 28 days)

Interventionscore on a scale (Mean)
BaselineCycle 1Cycle 2Cycle 4Cycle 6Cycle 8Cycle 10Cycle 12
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily1.31.81.51.81.41.81.51.4

[back to top]

Rate of Participants Symptom Severity

To evaluate the occurrence of symptoms and correlate to disease progression and tolerance to treatment using the MD Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) self-reporting tool. The MDASI-BT consists of 23 symptoms rated on an 11-point scale (0 - not present, to 10 - as bad as you can imagine) to indicate the presence and severity of the symptom. We calculated the mean core symptom severity at the time of clinical evaluation. (NCT01434602)
Timeframe: Completed at baseline, before a brain or spine magnetic resonance imaging (MRI)/clinical evaluation, and at cycle 1-2, cycle 4, cycle 6, cycle 8, cycle 10 and cycle 12 (each cycle is 28 days)

,,
Interventionscore on a scale (Mean)
BaselineCycle 1Cycle 2Cycle 4
Phase 1 Dose Level -1 Sorafenib 400mg Twice Daily 7 Days On, & Days Off + Everolimus Daily1.31.61.90.4
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on 7 Off + Everolimus 5mg Daily1.52.22.31.3
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily2.02.53.43.5

[back to top]

Rate of Participants Symptom Severity

To evaluate the occurrence of symptoms and correlate to disease progression and tolerance to treatment using the MD Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) self-reporting tool. The MDASI-BT consists of 23 symptoms rated on an 11-point scale (0 - not present, to 10 - as bad as you can imagine) to indicate the presence and severity of the symptom. We calculated the mean core symptom severity at the time of clinical evaluation. (NCT01434602)
Timeframe: Completed at baseline, before a brain or spine magnetic resonance imaging (MRI)/clinical evaluation, and at cycle 1-2, cycle 4, cycle 6, cycle 8, cycle 10 and cycle 12 (each cycle is 28 days)

Interventionscore on a scale (Mean)
BaselineCycle 1Cycle 2
Phase 1 Dose LEvel 1 Sorafenib 400mg Twice Daily + Everolimus 5mg Daily3.93.83.5

[back to top]

Rate of Participants Symptom Interference With Function

To evaluate the occurrence of symptoms and correlate to disease progression and tolerance to treatment using the MD Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) self-reporting tool. The MDASI-BT consists of 23 symptoms rated on an 11-point scale (0 - did not interfere, to 10 - interfered completely) to indicate the symptoms that interfere with a participant's life in the last 24 hours. We calculated the mean symptom interference at the time of clinical evaluation. (NCT01434602)
Timeframe: Completed at baseline, before a brain or spine magnetic resonance imaging (MRI)/clinical evaluation, and at cycle 1-2, cycle 4, cycle 6, cycle 8, cycle 10 and cycle 12 (each cycle is 28 days)

Interventionscore on a scale (Mean)
BaselineCycle 1Cycle 2Cycle 4Cycle 6Cycle 8Cycle 10Cycle 12
Phase 2 Group C Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily2.12.51.83.32.02.41.61.2

[back to top]

Rate of Participants Symptom Interference With Function

To evaluate the occurrence of symptoms and correlate to disease progression and tolerance to treatment using the MD Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) self-reporting tool. The MDASI-BT consists of 23 symptoms rated on an 11-point scale (0 - did not interfere, to 10 - interfered completely) to indicate the symptoms that interfere with a participant's life in the last 24 hours. We calculated the mean symptom interference at the time of clinical evaluation. (NCT01434602)
Timeframe: Completed at baseline, before a brain or spine magnetic resonance imaging (MRI)/clinical evaluation, and at cycle 1-2, cycle 4, cycle 6, cycle 8, cycle 10 and cycle 12 (each cycle is 28 days)

,,
Interventionscore on a scale (Mean)
BaselineCycle 1Cycle 2Cycle 4
Phase 2 Group A Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on 7 Off + Everolimus 5mg Daily2.33.34.32.8
Phase 2 Group B Dose Level -1 Sorafenib 400mg Twice Daily 7 Days on, 7 Off + Everolimus 5mg Daily3.44.16.44.2
Phase I Dose Level -1 Sorafenib 400mg Twice Daily, 7 Days on, 7 Off + Everolimus 5mg Daily1.52.32.80.3

[back to top]

Maximum Tolerated Dose of Everolimus for Participants With Recurrent Malignant Gliomas

Maximum tolerated dose of everolimus for participants with recurrent malignant gliomas. MTD is defined as the dose at which fewer than one-third of participants experience a dose limiting toxicity (DLT) to one of the study drugs. A DLT is any grade 3 thrombocytopenia, grade 4 anemia, grade 4 neutropenia, or febrile neutropenia of any grade. Any non-hematologic grade 3 or grade 4 toxicity, excluding alopecia and/or hand/foot reaction. Failure to recover from toxicities (to grade 1 or less) to be eligible for re-treatment with everolimus within 14 days of the last dose of everolimus. (NCT01434602)
Timeframe: First 28 days of treatment only (cycle 1)

Interventionmg (Number)
All Participants5

[back to top]

Number of Individuals Experiencing Toxicity

Safety determinations are based on the rate of drug-related adverse events (AEs) reported based upon the toxicity as measured by the NCI Common Terminology Criteria for Adverse Events version 4.0 (CTCAE v4.0). (NCT01488487)
Timeframe: 3.5 years

Interventionparticipants (Number)
Dose Limiting Toxicities (DLT)Any adverse event
Everolimus + Pasireotide124

[back to top]

Objective Response Rate (ORR)

"ORR is the rate of complete responses (CRs) + partial responses (PRs) as determined by RECIST (v1.1) and modified HCC RECIST criteria. Responses defined as follows:~CR: disappearance of all clinical/radiological evidence of tumor including any intratumoral arterial enhancement in all target lesions.~Partial Response (PR): at least a 30% decrease in the sum of diameters of viable (contrast enhancement in the arterial phase) target lesions, referencing the baseline sum.~Stable Disease (SD): any cases that do not qualify for either partial response or progressive disease.~Progressive Disease (PD): an increase of at least 20% in the sum of the diameters of viable target lesions, referencing the nadir sum, and/or the appearance of one or more new lesions. A new hepatic nodule signals PD when the longest diameter is at least 10 mm and the nodule shows the typical vascular pattern of HCC on dynamic imaging or if at least 1-cm interval growth is seen in subsequent scans." (NCT01488487)
Timeframe: 3.5 years

Interventionpercentage of participants (Number)
Radiographic response (CR or PR)Stable disease
Everolimus + Pasireotide045.5

[back to top]

Time to Progression (TTP)

Time to progression is defined as the time from study enrollment until radiological progression in a previously embolized lobe, development of new lesions in an untreated lobe, or evidence of extrahepatic progression (based on modified Hepatocellular Carcinoma (HCC) Response Evaluation Criteria In Solid Tumors (RECIST) criteria). Patients will be followed until death. Patients that die of causes unrelated to the study drug without evidence of progression will be censored. (NCT01488487)
Timeframe: 3.5 years

Interventionmonths (Median)
Everolimus + Pasireotide3.5

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from study enrollment until death. (NCT01488487)
Timeframe: 3.5 years

Interventionmonths (Median)
Everolimus + Pasireotide6.7

[back to top]

Duration of Response (DoR)

DoR was defined as the time from the first occurrence of PR or CR (as per local radiological review) until the date of the first documented disease progression or death due to underlying cancer. (NCT01491672)
Timeframe: 20 months

Interventionmonths (Median)
Prior Sunitinib10.8
Other Prior Vascular Endothelial Growth Factor (VEGF)7.4
Prior Cytokines9.2
All Participants9.2

[back to top]

Clinical Benefit Rate (CBR)

CBR was defined as the proportion of patients with best overall response of complete response (CR) or partial response (PR) or stable disease based on the local radiological data according to the RECIST 1.0 criteria. (NCT01491672)
Timeframe: 20 months

InterventionParticipants (Number)
Prior Sunitinib41
Other Prior Vascular Endothelial Growth Factor (VEGF)48
Prior Cytokines11
All Participants100

[back to top]

Duration of PFS for Each First-line Treatment Cohort

Duration of PFS during second-line treatment was defined as the time from the date of enrollment to the date of the first documented disease progression or death due to any cause. Participants' assessment was based on the local radiological data according to the RECIST 1.0 Criteria. (NCT01491672)
Timeframe: 20 months

Interventionmonths (Median)
Prior Sunitinib5.6
Other Prior Vascular Endothelial Growth Factor (VEGF)7.8
Prior Cytokines12.9

[back to top]

Progression-free Survival (PFS) - All Participants

PFS during second-line treatment was defined as the time from the date of enrollment to the date of the first documented disease progression or death due to any cause. The primary analysis of PFS was based on a local radiology review of CT scans and MRI collected until the participant experienced disease progression according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.0. (NCT01491672)
Timeframe: 20 months

Interventionmonths (Median)
All Participants7.4

[back to top]

Overall Survival (OS)

OS was defined as the time from date of enrollment to date of death due to any cause. (NCT01491672)
Timeframe: 28 months

Interventionmonths (Median)
Prior Sunitinib23.8
Other Prior Vascular Endothelial Growth Factor (VEGF)17.2
Prior CytokinesNA
All Participants23.8

[back to top]

Objective Response Rate (ORR)

ORR was defined as the proportion of participants with best overall response of CR or PR based on the local radiological data according to the RECIST 1.0 Criteria (NCT01491672)
Timeframe: 20 months

InterventionParticipants (Number)
Prior Sunitinib4
Other Prior Vascular Endothelial Growth Factor (VEGF)3
Prior Cytokines3
All Participants10

[back to top]

Clinical Benefit Rate of Patients Treated With the Combination of Letrozole and Lapatinib and Then After Progression, Treated With Everolimus, Letrozole and Lapatinib.

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Progression, as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions; Stable Disease (SD), neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for disease progression.~Clinical benefit rate is defined as complete response+partial response+ stable disease. All participants will be treated with the combination of letrozole and lapatinib. Once the participant progresses on this regimen, the participant will be treated with everolimus, letrozole and lapatinib until they progress." (NCT01499160)
Timeframe: From date of study entry until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 24 months

InterventionParticipants (Count of Participants)
Letrozole in Combination With Lapatinib Followed by Everolimus6

[back to top]

PROGRESSION FREE SURVIVAL TUMOR ASSESSMENT

Patients treated with the combination of Letrozole and Lapatinib will provide tumor biopsy sample (NCT01499160)
Timeframe: From date of study entry until 4 weeks after removal from study or until death (whichever occurs first) up to 24 months.

InterventionParticipants (Count of Participants)
Letrozole in Combination With Lapatinib Followed by Everolimus6

[back to top] [back to top]

Maximum Observed Everolimus Blood Concentration (Cmax)

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point) and at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after completion of implantation of the last study stent (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

Interventionng/mL (Mean)
Everolimus Dose of 95.4 µg0.71
Everolimus Dose of 102.4 µg0.67
Everolimus Dose of 138.6 µg0.91

[back to top]

Definite + Probable Stent Thrombosis Rate Based on Academic Research Consortium (ARC) Definition

DEFINITE ST: acute coronary syndrome and angiographic or pathologic evidence of stent thrombosis; PROBABLE ST: unexplained death within 30 days or target-vessel infarction without angiographic information ARC ST is reported as a cumulative value at different time points and within the different separate time points. Time 0 is the time point after the guide catheter has been removed. Acute ST: 0-24 hours after stent implantation; Subacute ST: >24 hours to 30 days post; late ST: >30 days to 1 year post; Very late ST: >1 year post; NOTE: Acute/subacute can be replaced by early ST (0-30 days). (NCT01510327)
Timeframe: >30 days-1 year

Interventionpercentage of participants (Number)
PROMUS Element0.0

[back to top]

Definite + Probable Stent Thrombosis Rate Based on Academic Research Consortium (ARC) Definition

DEFINITE ST: acute coronary syndrome and angiographic or pathologic evidence of stent thrombosis; PROBABLE ST: unexplained death within 30 days or target-vessel infarction without angiographic information ARC ST is reported as a cumulative value at different time points and within the different separate time points. Time 0 is the time point after the guide catheter has been removed. Acute ST: 0-24 hours after stent implantation; Subacute ST: >24 hours to 30 days post; late ST: >30 days to 1 year post; Very late ST: >1 year post; NOTE: Acute/subacute can be replaced by early ST (0-30 days). (NCT01510327)
Timeframe: >24 hours-30 days

Interventionpercentage of participants (Number)
PROMUS Element0.0

[back to top]

Definite + Probable Stent Thrombosis (ST) Rate Based on Academic Research Consortium (ARC) Definition

DEFINITE ST: acute coronary syndrome and angiographic or pathologic evidence of stent thrombosis; PROBABLE ST: unexplained death within 30 days or target-vessel infarction without angiographic information ARC ST is reported as a cumulative value at different time points and within the different separate time points. Time 0 is the time point after the guide catheter has been removed. Acute ST: 0-24 hours after stent implantation; Subacute ST: >24 hours to 30 days post; late ST: >30 days to 1 year post; Very late ST: >1 year post; NOTE: Acute/subacute can be replaced by early ST (0-30 days). (NCT01510327)
Timeframe: 24 hours

Interventionpercentage of participants (Number)
PROMUS Element0.0

[back to top]

Area Under the Concentration Versus Time Curve (AUC 0-t) Everolimus

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point) and at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after completion of implantation of the last study stent; t is the last time at which concentration can be quantified (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

Interventionng*hr/mL (Mean)
Everolimus Dose of 95.4 µg7.27
Everolimus Dose of 102.4 µg6.45
Everolimus Dose of 138.6 µg10.87

[back to top]

Area Under the Concentration Versus Time Curve (AUC 0-infinity) Everolimus

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point), 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after implantation of the last study stent. (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

Interventionng*hr/mL (Mean)
Everolimus Dose of 95.4 µg19.26
Everolimus Dose of 102.4 µg12.95
Everolimus Dose of 138.6 µg60.74

[back to top]

Area Under the Concentration Versus Time Curve (AUC 0-24), Everolimus

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point) and at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after completion of implantation of the last study stent (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

Interventionng*hr/mL (Mean)
Everolimus Dose of 95.4 µg6.83
Everolimus Dose of 102.4 µg6.14
Everolimus Dose of 138.6 µg9.51

[back to top]

All Death

Number of participants no longer alive (NCT01510327)
Timeframe: 6 months

Interventionpercentage of participants who died (Number)
PROMUS Element0.0

[back to top]

Time of Occurrence of Maximum Everolimus Concentration (Tmax)

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point) and at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after completion of implantation of the last study stent (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

Interventionhours (Mean)
Everolimus Dose of 95.4 µg0.47
Everolimus Dose of 102.4 µg0.62
Everolimus Dose of 138.6 µg0.52

[back to top]

Total Blood Clearance - Everolimus (CL)

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point), 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after completion of implantation of the last study stent. (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

InterventionL/h (Mean)
Everolimus Dose of 95.4 µg6445
Everolimus Dose of 102.4 µg8044
Everolimus Dose of 138.6 µg2511

[back to top]

Terminal Phase Half-life (t1/2) Everolimus

Venous blood draw up to 24 hours prior to implantation of the first study stent (predose time point), at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours after completion of implantation of the last study stent. (NCT01510327)
Timeframe: Predose <24 hours; post dose at 30 minutes, 1, 2, 4, 6, 12, 24, 48, and 72 hours

InterventionHours (Mean)
Everolimus Dose of 95.4 µg34.19
Everolimus Dose of 102.4 µg22.83
Everolimus Dose of 138.6 µg136.06

[back to top]

Target Vessel Revascularization (TVR)

TVR is any ischemia-driven repeat percutaneous intervention to improve blood flow, or bypass surgery of not previously existing lesions with diameter stenosis ≥50% by quantitative coronary angiography in the target vessel, including the target lesion. (NCT01510327)
Timeframe: 6 months

Interventionpercentage of participants (Number)
PROMUS Element0.0

[back to top]

Target Lesion Revascularization (TLR)

TLR is any ischemia-driven repeat percutaneous intervention to improve blood flow of the successfully treated target lesion or bypass surgery of the target vessel with a graft distally to the successfully treated target lesion. (NCT01510327)
Timeframe: 6 months

Interventionpercentage of participants (Number)
PROMUS Element0.0

[back to top]

Overall Survival (OS) of Patients Treated With Everolimus After Failure of First-line Sunitinib or Pazopanib Therapy up to 48 Months

Overall survival (OS) was defined as the time from date of start of treatment to date of death due to any cause. If a patient was not known to have died, survival will be censored at the date of last contact. (NCT01514448)
Timeframe: 48 months

Interventionmonths (Median)
1st Line SUN14.8
1st Line PAZ20.4

[back to top]

Percentage of Patients With Overall Response Rate (ORR) Treated With Everolimus After Failure of First-line Sunitinib or Pazopanib Therapy at Month 6

Overall response rate (ORR) is the Percentage of patients with a best overall response of complete response (CR) or partial response (PR) by month 6. ORR was assessed according to RECIST 1.1 criteria. Partial response (PR) required at least a 30% decrease in the sum of the longest diameters of all target lesions, taking as reference the baseline sum of the longest diameters. Complete response (CR) required a disappearance of all target and non-target lesions. (NCT01514448)
Timeframe: Month 6

Interventionpercentage of participants (Number)
1st Line SUN0.0
1st Line PAZ0.0

[back to top]

Percentage of Progression-free Patients by Month 6

Percentage of progression-free patients by month 6 after starting everolimus treatment. For the purpose of the binomial design of the study, a patient being 'progression-free' will be defined as a patient without disease progression by month 6 whereas a subject with progressive disease by month 6 will not be counted as 'progression-free'. The primary variable was derived from radiologic tumor assessments according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1.) Disease progression was either 1) a 20% increase in the sum of the longest diameter of all target lesions, taking as reference the smallest sum of the longest diameters of all target lesions recorded at or after baseline (minimum absolute increase 5 mm in sum) or 2) the appearance of a new lesion or 3) the unequivocal progression of non-target lesions overall. (NCT01514448)
Timeframe: Month 6

InterventionPercentage of participants (Number)
1st Line SUN8.3
1st Line PAZ46.2

[back to top]

Progression-Free Survival (PFS) as the Time Interval Between First Intake of Everolimus and First Documented Disease Progression or Death Due to Any Cause at 24 Months

Progression-free survival (PFS) is the time from date of start of treatment to the date of event defined as the first documented progression or death due to any cause. If a patient did not have an event, progression-free survival was censored at the date of last adequate tumor assessment (NCT01514448)
Timeframe: 24 months

Interventionmonths (Median)
1st Line SUN2.8
1st Line PAZ8.0

[back to top]

Time to Definitive Deterioration in Functional Assessment of Cancer Therapy - General (FACT-G) Questionnaire Total Score

"FACT-G is a self-assessed health-related quality of life questionnaire. The questionnaire is comprised of 27 questions examining physical, social/family, emotional, and functional well-being. Participants responded to the items on a five-point scale, ranging from 0: Not at all to 4: Very much. The total score ranges from 0 to 108, with higher scores indicating a better patient-reported outcome/quality of life.~Definitive deterioration is defined as a decrease in the total score by at least 7 points compared to baseline with no further improvement.~Death was considered as worsening of the FACT-G total score if it occurred close to the last available assessment, where close was defined as twice the planned period between two assessments. Patients without definitive worsening prior to analysis cut-off or prior to start of another anticancer therapy were censored at the date of their last assessment." (NCT01524783)
Timeframe: From randomization to definitive deterioration of FACT-G total score, assessed up to approximately 3 years

InterventionMonths (Median)
Everolimus + BSC13.01
Placebo + BSC9.23

[back to top]

Overall Survival (OS)

OS is defined as the time from the date of randomization to date of death due to any cause. If a death had not been observed by the date of analysis cut-off, then OS was censored at the date of last contact. All participants randomized to placebo arm who crossed over to everolimus were censored. (NCT01524783)
Timeframe: From date of randomization to date of death, assessed up to approximately 8 years

InterventionMonths (Median)
Everolimus + BSC43.1
Placebo + BSC41.76

[back to top]

Disease Control Rate (DCR) Based on Modified RECIST 1.0 and as Per Central Radiology Assessment

"DCR is defined as the proportion of subjects with best overall response of CR or PR or stable disease based on modified RECIST 1.0 and as per central radiology assessment.~CR: disappearance of all target lesions. PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters.~Stable disease: Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for progression." (NCT01524783)
Timeframe: From randomization until end of treatment, assessed up to approximately 2.5 years

InterventionPercentage of participants (Number)
Everolimus + BSC82.4
Placebo + BSC64.9

[back to top]

Pharmacokinetics (PK): Predose Concentration (Cmin) of Everolimus at Day 29

A pre-dose blood sample at day 29 was collected to determine the exposure of everolimus at the steady-state pre-dose concentration (Cmin). Cmin is provided for participants randomized to everolimus+BSC who received 10mg of everolimus daily and also for participants randomized to everolimus+BSC who received 5mg of everolimus daily which was required for a number of participants in the study experiencing adverse events requiring dose modifications (NCT01524783)
Timeframe: Pre-dose at Day 29.

Interventionnanogram/milliliter (ng/mL) (Mean)
10mg daily dose5mg daily dose
Everolimus + BSC16.3824.700

[back to top]

Overall Response Rate (ORR) as Per Modified RECIST 1.0 According to Central Evaluation

"ORR is defined as the proportion of patients with best overall response (BOR) of complete response (CR) or partial response (PR), according to central evaluation and as per modified RECIST 1.0.~CR: disappearance of all target lesions. PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters." (NCT01524783)
Timeframe: From randomization until end of treatment, assessed up to approximately 2.5 years

InterventionPercentage of participants (Number)
Everolimus + BSC2.0
Placebo + BSC1.0

[back to top]

Time to Definitive Deterioration in World Health Organization (WHO) Performance Status (PS) Change

"WHO PS is a scale rated from 0 (fully active) to 5 (death) by a healthcare professional to assess the overall status of a patient: a lower score represents a higher ability to perform daily tasks. Deterioration is defined as an increase of at least one point compared to baseline. Deterioration is considered definitive if no improvements in the WHO PS status is observed at a subsequent time of measurement during the treatment period following the time point where the deterioration is observed. Death was considered as worsening of the WHO PS if it occurred close to the last available assessment, where close was defined as twice the planned period between two assessments. Patients without definitive worsening prior to analysis cut-off or prior to start of another anticancer therapy were censored at the date of their last assessment." (NCT01524783)
Timeframe: From randomization to definitive deterioration of WHO performance status, assessed up to approximately 3 years

InterventionMonths (Median)
Everolimus + BSC24.08
Placebo + BSC24.15

[back to top]

Probability of Participants Remaining Event-Free in Progression-Free Survival (PFS) Based on Central Radiology Assessment

"PFS is defined as the time from randomization to the date of the first documented tumor progression or death from any cause, whichever comes first.~Progression was defined using modified RECIST 1.0 and as per central radiology assessment as at least a 20% increase in the sum of diameter of all measured target lesions, taking as reference the smallest sum of diameter of all target lesions recorded at or after baseline. Progression was assessed by cat scan (CT) and/or magnetic resonance imaging (MRI).~For participants who had not progressed or died at the analysis cut-off date, PFS was censored at the date of the last adequate tumor evaluation date. An adequate tumour assessment is a tumour assessment with an overall response other than unknown.~The percentage event-free probability estimate is the estimated probability that a patient will remain event-free in PFS up to the specified time point." (NCT01524783)
Timeframe: From date of randomization to progression or death, whichever comes first, assessed up to 27 months

,
InterventionPercent event-free probability in PFS (Number)
2 months4 months6 months8 months10 months12 months15 months18 months21 months24 months27 months
Everolimus + BSC90.181.272.162.451.744.440.131.827.622.0NA
Placebo + BSC74.649.140.135.831.328.126.424.417.417.417.4

[back to top]

Change From Baseline in Neuron Specific Enolase (NSE) Levels

NSE is a potential biomarker for tumor response. Blood samples were collected for assessment of NSE levels. Change from Baseline at a particular visit was calculated as the NSE level at that visit minus Baseline. (NCT01524783)
Timeframe: From baseline (every 4 weeks) up to Week 116

,
Interventionmicrogram/liter (ug/L) (Mean)
Week 4Week 8Week 12Week 16Week 20Week 24Week 28Week 32Week 36Week 40Week 44Week 48Week 52Week 56Week 60Week 64Week 68Week 72Week 76Week 80Week 84Week 88Week 92Week 96Week 100Week 104Week 108Week 112Week 116
Everolimus + BSC0.10.3-0.32.00.81.71.11.52.01.20.82.01.53.62.44.41.11.61.72.98.02.24.04.31.82.95.52.810.6
Placebo + BSC-2.2-4.215.54.15.36.70.43.12.01.21.20.60.70.90.10.91.90.70.60.70.10.60.10.0-0.2-1.7-0.5-3.20.2

[back to top]

Change From Baseline in Chromogranin A (CgA) Levels

CgA is a potential biomarker for tumor response. Blood samples were collected for assessment of CgA levels. Change from Baseline at a particular visit was calculated as the CgA level at that visit minus Baseline. (NCT01524783)
Timeframe: From baseline (every 4 weeks) up to 116 weeks

,
Interventionmicrogram/liter (ug/L) (Mean)
Week 4Week 8Week 12Week 16Week 20Week 24Week 28Week 32Week 36Week 40Week 44Week 48Week 52Week 56Week 60Week 64Week 68Week 72Week 76Week 80Week 84Week 88Week 92Week 96Week 100Week 104Week 108Week 112Week 116
Everolimus + BSC434.8162.3396.7263.5162.1253.2587.2508.594.7511.4552.81326.21196.21920.61722.03811.01413.3239.53256.55421.04379.9571.2765.5984.6935.31466.42245.82817.02951.8
Placebo + BSC1154.32697.61176.01450.43640.11783.91350.32000.7180.9207.6140.6129.0124.2134.9120.9141.6222.6233.7210.9286.3175.4-9.463.7146.5-12.7-21.7215.9514.599.6

[back to top]

All Collected Deaths

"Deaths on-treatment were collected from first dose of study treatment until end of study treatment plus 30 days post treatment, up to maximum duration of approximately 8 years.~Total Deaths were collected from first dose of study treatment until end of post-treatment survival follow, up to maximum duration of approximately 8 years." (NCT01524783)
Timeframe: On-treatment deaths: up to approximately 8 years. All deaths: up to approximately 8 years

,,,
InterventionParticipants (Number)
On-treatment deathsTotal deaths
Everolimus + BSC (Throughout the Study)10126
Everolimus +BSC (Crossover)01
Everolimus+BSC (All)10127
Placebo + BSC (Blinded Period)557

[back to top]

Incidence of Biopsy Proven Antibody Mediated Rejection.

To evaluate the proportion of patients with the following efficacy events: biopsy proven antibody mediated rejection/Steroid resistant BPAR and BPAR treated with T cell depleting therapy. (NCT01544491)
Timeframe: at 12 and 36 months post-transplantation

,
InterventionParticipants (Count of Participants)
patients with BPAR at 12 monthsBPAR Steroid resistant,12 monthsBPAR, T Cell depleting therapy 12 monthspatients with BPAR at 36 monthsBPAR Steroid resistant,36 monthsBPAR, T Cell depleting therapy 36 months
EVR+rTAC711612
MMF+sTAC8011221

[back to top]

Number of Participants Having Reached the Composite Efficacy Endpoint of Biopsy-proven Acute Rejection

To estimate the rate of the composite efficacy endpoint of biopsy-proven acute rejection (BPAR), graft loss or death at 12 months post transplantation in primary paediatric kidney transplant recipients converted at 4-6 weeks post-transplantation from MMF + standard TAC regimen and steroids, to everolimus + reduced dose TAC regimen and steroid withdrawal at 6 months, versus continuation of MMF + standard TAC regimen and steroids. (NCT01544491)
Timeframe: 12 months, 36 months

,
InterventionParticipants (Count of Participants)
12 months36 months
EVR+rTAC55
MMF+sTAC35

[back to top]

To Evaluate Renal Function, Assessed by Glomerular Filtration Rate (eGFR) and Estimated by the Schwartz Formula (Extended), at Month 12

To evaluate renal function assessed by Glomerular Filtration Rate (eGFR) estimated by the Schwartz Formula (extended) (Schwartz, 2009). Results given as change from randomization (NCT01544491)
Timeframe: 12 months post-transplantation

InterventionmL/min/1.73m2 (Mean)
EVR+rTAC4.6
MMF+sTAC-0.0

[back to top]

Chronic Allograft Nephropathy / Interstitial Fibrosis and Tubular Atrophy

To evaluate the proportion of patients with chronic allograft nephropathy (interstitial fibrosis and tubular atrophy, IF/TA) by histopathology and its progression.The term chronic allograft nephropathy was used inappropriately in the protocol and therefore, replaced by interstitial fibrosis and tubular atrophy (NCT01544491)
Timeframe: at 12 and 36 months post-transplantation.

,
InterventionParticipants (Count of Participants)
12 months36 months
EVR+rTAC311
MMF+sTAC37

[back to top]

Composite Efficacy Endpoint

To evaluate the proportion of patients with the following efficacy events: Biopsy Proven Acute Rejection (BPAR), graft loss or death. The efficacy events will be descriptively summarized by treatment group. (NCT01544491)
Timeframe: at 12 and 36 months post-transplantation

,
InterventionParticipants (Count of Participants)
month 12 composite efficacy endpointgraft loss 12 monthsdeath 12 monthsacute rejection 12 monthstreated acute rejection 12 monthsBiopsy proven acute rejection 12 monthstreated Biopsy proven acute rejection 12 monthsmonth 36 composite efficacy endpointgraft loss 36 monthsdeath 36 monthsacute rejection 36 monthsBiopsy proven acute rejection 36 monthstreated Biopsy proven acute rejection 36 months
EVR+rTAC5005555510555
MMF+sTAC3004433520755

[back to top]

Evaluation of Evolution of Renal Allograft Function Over Time

results given as eGFR values by time interval (NCT01544491)
Timeframe: baseline, 6 months, 12 months , 24 months, 36 months

,
Interventionml/min/1.73m2 (Mean)
baselinemonth 6month 12month 24month 36
EVR+rTAC14.276.676.972.968.2
MMF+sTAC13.168.367.868.669.6

[back to top]

Growth/Development : Weight, Height, BMI : Change From Baseline

Evaluation of the potential effects upon the bone growth. The Z-score is a statistical tool which helps to assess data (here child growth parameters) relative to a reference or standard population. The Z-score describes the distance and direction of an observation away from the population median (or mean, however, here the median was used). A negative Z-score shows that data are lower than the median of the standard population, a positive Z-score shows that data are higher than the median of the standard population, and a Z-score of zero shows that the data are equal to the median of the standard population. The more the Z-score is distant from 0, the more expressed is for example underweight or overweight. (NCT01544491)
Timeframe: month 12 , month 36 post transplantation.

,
Interventionz score (Mean)
Height month 12Height month 36Weight month 12Weight month 36BMI month 12BMI month 36
EVR+rTAC0.370.720.300.610.000.02
MMF+sTAC0.200.390.420.820.240.47

[back to top]

To Evaluate Renal Function, Assessed by Glomerular Filtration Rate (eGFR) and Estimated by the Schwartz Formula (Abbreviated), at Month 12 and 36

To evaluate renal function assessed by Glomerular Filtration Rate (eGFR) estimated by the Schwartz Formula (abbreviated) (Schwartz, 2009). (NCT01544491)
Timeframe: 12 months and 36 months post-transplantation

,
InterventionmL/min/1.73m2 (Mean)
12 months36 months
EVR+rTAC76.768.1
MMF+sTAC71.767.3

[back to top]

To Evaluate the Severity of BPAR (Acute T-cell Mediated Rejection Only) (Banff 2009)

"T-cell mediated rejection severity :~Type IA - Significant interstitial infiltration (> 25% of parenchyma) and foci of moderate tubulitis (> 4 mononuclear cells/tubular cross section or group of 10 tubular cells).~Type IB - Significant interstitial infiltration (> 25% of parenchyma) and foci of severe tubulitis (> 10 mononuclear cells/tubular cross section or group of 10 tubular cells).~Type IIA - Mild to moderate intimal arteritis Type IIB - Severe intimal arteritis comprising > 25% of the lumenal area Type III - Transmural (full vessel wall thickness) arteritis and/or arterial fibrinoid change and necrosis of medial smooth muscle cells (with accompanying lymphocytic inflammation)" (NCT01544491)
Timeframe: month 12, month 36

,
InterventionParticipants (Count of Participants)
month 12 grade IAmonth 12 grade IBmonth 12 grade IIAmonth 12 grade IIBmonth 12 grade IIImonth 36 grade IAmonth 36 grade IBmonth 36 grade IIAmonth 36 grade IIBmonth 36 grade III
EVR+rTAC3100032000
MMF+sTAC1020010111

[back to top]

To Evaluate the Time to Event of BPAR

Time to incidence of Event, given in terms of number of participants with an Event according to time interval up to 36 months (NCT01544491)
Timeframe: 36 months

,
InterventionParticipants (Count of Participants)
day 1-7day 8-14day 15-28day 29-56day 57-84day 85-150day 151- 240day 241-330day 331- 510day 511-690day 691-870day 871-1050after day 1050
EVR+rTAC0010020101000
MMF+sTAC0000020210000

[back to top]

Proteinuria (Urinary Protein/Creatinine Ratio)

The urinary protein/creatinine ratio will be descriptively summarized by treatment group at each visit. The incidence rate of patients with proteinuria will be categorized in <0.2 g/mg/mg, 0.2<2.0 mg/mg and ≥ 2.0 mg/mg and summarized by treatment groups at each visit. (NCT01544491)
Timeframe: at 12 and 36 months post-transplantation

,
InterventionParticipants (Count of Participants)
Baseline < 200mg/gBaseline 200 - < 2000 mg/gBaseline >= 2000 mg/gMonth 12 < 200mg/gMonth 12 200 - < 2000 mg/gMonth 12 >= 2000 mg/gMonth 36 < 200mg/gMonth 36 200 - < 2000 mg/gMonth 36 >= 2000 mg/g
EVR+rTAC112141913023101
MMF+sTAC212152811023110

[back to top]

Overall Survival From the Start (OSS) of Study Treatment

Time from first pazopanib dose until death due to any cause in patients who received at least one dose of pazopanib followed by everolimus (NCT01545817)
Timeframe: Throughout the study, up to 4 years

InterventionMonths (Median)
Pazopanib35.7

[back to top]

Overall Survival of Everolimus (OSE)

Time from first everolimus dose until death due to any cause (NCT01545817)
Timeframe: Throughout the study period, up to 4 years

Interventionmonths (Median)
Everolimus15.6

[back to top]

Progression Free Survival (PFS) Rates

PFS rates 3 and 6 months after date of first dose of second-line everolimus treatment. (NCT01545817)
Timeframe: 3 months, 6 months

InterventionSurvival probability (Number)
3 months6 months
All Patients0.7370.355

[back to top]

Objective Response Rate (ORR) for the Pazopanib Treatment Period Using RECIST

"Percentage of patients with Complete or Partial Response at any time following the start of first-line pazopanib treatment.~RECIST Complete Response is defined to be a disappearance of all target lesion(s). RECIST Partial Response is defined to be at least a 30% decrease in the sum of the target lesion LDs." (NCT01545817)
Timeframe: Throughout the study period, up to 4 years

InterventionPercentages of participants (Number)
Complete responsePartial response
All Patients6.839.2

[back to top]

Objective Response Rate (ORR) for the Everolimus Treatment Period Using RECIST

"Percentage of patients with Complete or Partial Response at any time following the start of second-line everolimus treatment as per RECIST.~RECIST Complete Response is defined to be a disappearance of all target lesion(s). RECIST Partial Response is defined to be at least a 30% decrease in the sum of the target lesion LDs." (NCT01545817)
Timeframe: Throughout the study, up to 4 years

InterventionPercentages of Participants (Number)
Complete responsePartial response
All Patients0.07.9

[back to top]

Progression Free Survival (PFS) for the Everolimus Treatment Period Using RECIST

"Time between the date of first everolimus dose and date of disease progression or death (whichever comes first) in patients treated initially with pazopanib.~Disease progression is measured by RECIST (Response Evaluation Criteria in Solid Tumors), which is at least a 20% increase in the sum of the target lesion longest diameters (LDs)." (NCT01545817)
Timeframe: Throughout the study period, up to 4 years

Interventionmonths (Median)
Everolimus5.1

[back to top]

PFS for the Pazopanib Treatment Period Using RECIST

Time from first pazopanib dose until disease progression or death from any cause (whichever occurred earlier), provided this occurred prior to the start of everolimus and within 6 months of last dose of pazopanib (NCT01545817)
Timeframe: Throughout the study period, up to 4 years

InterventionMonths (Number)
Pazopanib11.0

[back to top] [back to top]

Estimated GFR - PP Set

This was a sensitivity analysis for the primary outcome measure based on the per-protocol set of patients. (NCT01551212)
Timeframe: month 12

InterventionmL/min (Mean)
Everolimus/Tacrolimus74.83
Tacrolimus70.65

[back to top]

Estimated Glomerular Filtration Rate (GFR)

The estmated GFR was calculated using MDRD-4 formula (Modification of Diet in Renal Disease Study Group). (NCT01551212)
Timeframe: month 12

InterventionmL/min (Mean)
Everolimus/Tacrolimus73.46
Tacrolimus71.95

[back to top]

Incidence of de Novo HCC Malignancies

Incidence of de novo Hepatocellular Carcinoma (HCC) malignancies assessed as treatment emergent adverse events of special interest (NCT01551212)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Everolimus/Tacrolimus0
Tacrolimus2

[back to top]

Number of Participants With HCV

Number of Participants with HCV (hepatitis C virus) assessed as treatment emergent adverse events of special interest (NCT01551212)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Everolimus/Tacrolimus6
Tacrolimus12

[back to top]

Percentage of Participants With Treated Biopsy Proven Acute Rejection (BPAR), Graft Loss or Death

Percentage of Participants with Treated Biopsy Proven Acute Rejection (BPAR), Graft Loss or Death at Month 12 (NCT01551212)
Timeframe: 12 months

InterventionPercent of Patients (Number)
Everolimus/Tacrolimus9.5
Tacrolimus7.9

[back to top]

Biochemical Response Rate (BRR) for 5HIAA Levels

The percentages are the biochemical response rates i.e. percentage of patients showing normalization i.e. return to within normal ranges, or a decrease of >= 50% from baseline of 5HIAA concentrations. (NCT01563354)
Timeframe: Baseline up Week 52

,,
Interventionpercentage of participants (Number)
Week 12Week 24Week 36Week 48Week 52
Everolimus11.111.111.100
Pasireotide LAR20.05.05.05.05.0
Pasireotide LAR and Everolimus Combination10.020.05.05.010.0

[back to top]

Duration of Biochemical Response (DBR), by Treatment (Full Analysis Set)

Time from the first documentation of biochemical response to the first documentation of biochemical progression or to death due to any cause, whichever occurred first. Biochemical progression is defined as an increase of serum CgA levels ≥ 25% compared to baseline. (NCT01563354)
Timeframe: Baseline up to Month 18

Interventionmonths (Median)
Pasireotide LAR14.75
Everolimus2.00
Pasireotide LAR and Everolimus Combination8.38

[back to top]

Summary of Duration of Response (Months)

Date of first objective tumor response to date of tumor progression or death due to any cause. (NCT01563354)
Timeframe: Every 3 months up to Year 1

,,
Interventionmonths (Number)
25th percentileMedian75th percentile
EverolimusNANANA
Pasireotide LARNANANA
Pasireotide LAR and Everolimus CombinationNANANA

[back to top]

Percentage of Participants Progression-free at 9 Months Based on Response Evaluation Criteria In Solid Tumors v1.1 (RECIST v1.1)

"Patients with Complete Response (CR), Partial Response (PR), or Stable Disease (SD) at Month 9 were to be considered as progression-free based on RECIST v1.1. Patients with missing tumor assessment, or with overall lesion response unknown at Month 9 were considered as non progression-free, unless any of the following assessments at Week 48 or Week 52 indicate CR, PR, or SD, in which case the patient was to be considered as progression-free at Month 9. Patients discontinuing the study for any reason prior to the 9 month assessment were to be considered as non progression-free." (NCT01563354)
Timeframe: Baseline up to 9 months

,,
Interventionpercentage of participants (Number)
Complete responsePartial responseStable diseaseProgression-free (PF) at Month 9
Everolimus02.431.033.3
Pasireotide LAR02.434.139.0
Pasireotide LAR and Everolimus Combination02.448.858.5

[back to top]

12-month Disease Control Rate (DCR) and Objective Response Rate (ORR)

Objective response rate (ORR) was defined as the percentage of patients showing a best overall response (BOR) of CR or PR during the core study according to RECIST v1.1 criteria. The best overall response is interpreted as the best response recorded from the start of the treatment until disease progression/recurrence, death from any cause or until the patient withdraws consent, whichever is earliest. DCR was was defined as the percentage of participants with a best overall response of complete response, partial response or stable disease during 12 months of treatment according to RECIST v1.1. (NCT01563354)
Timeframe: Baseline up to Month 12

,,
Interventionpercentage of participants (Number)
Objective response (CR+PR)Disease control rate (CR+PR+SD)Complete response (CR)Partial response (PR)Stable diseaseProgressive diseaseUnknownNot assessedDiscontinued before month 12
Everolimus2.473.802.471.44.84.816.764.3
Pasireotide LAR2.480.502.478.014.62.42.468.3
Pasireotide LAR and Everolimus Combination4.978.004.973.27.3014.663.4

[back to top]

Summary of Progression-free Survival (PFS) Based on RECIST v1.1

Time from first study drug administration to objective tumor progression or death from any cause according to RECIST v1.1 (NCT01563354)
Timeframe: Baseline, every 3 months up to 69 months

Interventionmonths (Median)
Pasireotide LAR8.51
Everolimus12.48
Pasireotide LAR and Everolimus Combination16.53

[back to top]

Biochemical Response Rate (BRR) for Chromogranin A (CgA) Levels

Percentage of patients showing normalization or a decrease of ≥ 30% of serum CgA concentrations compared to baseline. (NCT01563354)
Timeframe: Baseline up to Week 52

,,
Interventionpercentage of participants (Number)
Week 12Week 24Week 36Week 48Week 52
Everolimus7.47.43.700
Pasireotide LAR20.68.88.88.85.9
Pasireotide LAR and Everolimus Combination17.120.011.411.45.7

[back to top]

Kaplan-Meier Estimates of Progression-free Survival (PFS)

Percent (%) event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. Percent event-free probability estimates are obtained from the Kaplan-Meier survival estimates. Events are time from first study drug administration to objective tumor progression or death from any cause according to RECIST v1.1. (NCT01563354)
Timeframe: Baseline, every 3 months up to 69 months

,,
Interventionevent free probability estimates (Number)
3 months6 months9 months12 months15 months18 months21 months24 months27 months30 months33 months36 months39 months42 months45 months48 months51 months54 months57 months60 months63 months66 months69 months
Everolimus91.263.556.950.246.838.629.419.619.69.89.89.89.89.89.89.8NANANANANANANA
Pasireotide LAR83.668.249.639.932.621.814.514.514.510.910.910.910.910.910.910.910.910.910.910.910.9NANA
Pasireotide LAR and Everolimus Combination88.685.579.255.551.242.738.028.528.519.019.014.214.214.214.214.214.214.27.17.17.17.17.1

[back to top]

Summary of Time to Response (Months)

Time from start of treatment to the first observed objective tumor response (partial response or complete response) observed according to RECIST v1.1. (NCT01563354)
Timeframe: Every 3 months up to Year 1

,,
Interventionmonths (Number)
25th percentileMedian75th percentile
EverolimusNANANA
Pasireotide LARNANANA
Pasireotide LAR and Everolimus CombinationNANANA

[back to top]

Kaplan-Meier Event-free Probability Estimate for Biochemical Progression-free Survival Based on CgA Levels

Percent (%) Event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. Percent event-free probability estimates are obtained from the Kaplan-Meier survival estimates. Events are biochemical progressions, i.e., an increase of CgA levels >= 25% compared to baseline or deaths due to any cause. (NCT01563354)
Timeframe: Baseline, every 3 months up to Month 24

,,
Interventionevent free probability estimates (Number)
3 months6 months9 months12 months15 months18 months21 months24 months
Everolimus35.417.711.07.4NANANANA
Pasireotide LAR43.129.518.518.518.513.813.8NA
Pasireotide LAR and Everolimus Combination77.144.529.726.418.118.118.118.1

[back to top]

Kaplan-Meier Event-free Probability Estimate Based on CgA Levels

Kaplan Meier estimates are for Duration of biochemical response (DBR) outcome measure. Events are biochemical progressions i.e. an increase of CgA levels >= 25% compared to baseline or deaths due to any cause. Percent (%) Event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. (NCT01563354)
Timeframe: Baseline, every 3 months up to Month 18

,,
Interventionevent free probability estimates (Number)
3 months6 months9 months12 months15 months18 months
Everolimus37.5NANANANANA
Pasireotide LAR75.056.356.356.337.537.5
Pasireotide LAR and Everolimus Combination77.877.844.444.444.444.4

[back to top]

Summary of Biochemical Progression-free Survival Based on CgA Levels by Treatment

Time from the first documentation of biochemical response to the first documentation of biochemical progression or to death due to any cause, whichever occurred first. Biochemical progression is defined as an increase of serum CgA levels ≥ 25% compared to baseline. (NCT01563354)
Timeframe: Baseline up Month 24

Interventionmonths (Median)
Pasireotide LAR2.89
Everolimus2.86
Pasireotide LAR and Everolimus Combination5.62

[back to top]

Number of Participants With an Adverse Event.

Number of participants with an adverse event. Please refer to the adverse event reporting for more detail. (NCT01582009)
Timeframe: The time from registration up to 3 years

InterventionParticipants (Count of Participants)
Arm I: Oral Panobinostat and Oral Everolimus26

[back to top]

Number of Participants With Clinical Response

Number of participants with clinical response. Response will be evaluated in this study using the new international criteria proposed by the Response Evaluation Criteria in Solid Tumors ver 1.0 Committee [JNCI 92(3):205-216, 2000]. Changes in only the largest diameter (unidimensional measurement) of the tumor lesions are used in the RECIST ver. 1.0 criteria. (NCT01582009)
Timeframe: The time from registration up to 3 years

InterventionParticipants (Count of Participants)
Arm I: Oral Panobinostat and Oral Everolimus0

[back to top]

Progression-free Survival (PFS)

6 month PFS survival rate. Calculated as the total number of failures (deaths or progression) divided by the total follow-up or exposure time of patients on study. Assessed using Kaplan Meier and Proportional Hazards. (NCT01582009)
Timeframe: The time from registration to documentation of disease progression up to 3 years

Interventionpercentage of participants (Number)
Arm I: Oral Panobinostat and Oral Everolimus28

[back to top]

Median Progression Free Survival

Median progression free survival. Assessed using Kaplan Meier and Proportional Hazards. (NCT01582009)
Timeframe: The time from registration up to 3 years

Interventionmonths (Median)
Arm I: Oral Panobinostat and Oral Everolimus5.3

[back to top]

6-month Overall Survival Rate

6-month overall survival rate (NCT01582009)
Timeframe: The time from registration up to 3 years

Interventionpercentage of participants (Number)
Arm I: Oral Panobinostat and Oral Everolimus96

[back to top]

Mean EORTC QLQ-G.I. NET21 Score (Core)

The EORTC QLQ-G.I. NET21 contains 21 questions and has three defined multi-item symptom scales (endocrine - 3 questions, gastrointestinal - 5 questions, and treatment related side effects - 3 questions), two single item symptoms (bone/muscle pain and concern about weight loss), two psychosocial scales (social function - 3 questions and disease-related worries - 3 questions) and two other single items (sexuality and communication). For each of the 9 domains, final scores are transformed such that they range from 0-100, where higher scores indicate worsening. (NCT01595009)
Timeframe: Baseline, weeks 4, 8, 20, 32, 44, and end of treatment (EOT) up to week 82

Interventionunits on a scale (Mean)
Endocrine, BaselineEndocrine, week 4Endocrine, week 8Endocrine, week 20Endocrine, week 32Endocrine, week 44Endocrine, EOT up to week 82G.I., BaselineG.I., week 4G.I., week 8G.I., week 20G.I., week 32G.I., week 44G.I., EOT up to week 82Treatment, baselineTreatment, week 4Treatment, week 8Treatment, week 20Treatment, week 32Treatment, week 44Treatment, EOT up to week 82Social function, BaselineSocial function, week 4Social function, week 8Social function, week 20Social function, week 32Social function, week 44Social function, EOT up to week 82Disease-related worries, BaselineDisease-related worries, week 4Disease-related worries, week 8Disease-related worries, week 20Disease-related worries, week 32Disease-related worries, week 44Disease-related worries, EOT up to week 82Muscle/bone pain, BaselineMuscle/bone pain, week 4Muscle/bone pain, week 8Muscle/bone pain, week 20Muscle/bone pain, week 32Muscle/bone pain, week 44Muscle/bone pain, EOT up to week 82Sexual function, BaselineSexual function, week 4Sexual function, week 8Sexual function, week 20Sexual function, week 32Sexual function, EOT up to week 82Communication function, BaselineCommunication function, week 4Communication function, week 8Communication function, week 20Communication function, week 32Communication function, week 44Communication function, EOT up to week 82Body image, BaselineBody image, week 4Body image, week 8Body image, week 20Body image, week 32Body image, week 44Body image, EOT up to week 82
pNET (Core)12.9913.2113.4712.5016.670.009.4923.6923.0222.3425.6325.000.0025.859.2617.7216.1828.7027.7816.6722.0639.5636.5238.3540.2838.8933.3338.8943.1039.2040.5637.5038.8933.3342.8926.6724.0731.1841.6750.0033.3334.1335.4621.0528.7933.3311.1125.817.588.1812.2214.298.330.0012.7015.9015.0921.5116.670.000.0023.02

[back to top]

Change in EORTC QLQ-G.I. NET21 Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)

The EORTC QLQ-G.I. NET21 contains 21 questions and has three defined multi-item symptom scales (endocrine - 3 questions, gastrointestinal - 5 questions, and treatment related side effects - 3 questions), two single item symptoms (bone/muscle pain and concern about weight loss), two psychosocial scales (social function - 3 questions, disease-related worries - 3 questions) and two other single items (sexuality and communication). For each of the 9 domains, final scores are transformed such that they range from 0-100, where higher scores indicate worsening outcomes. A positive change from baseline indicates worsening. (NCT01595009)
Timeframe: baseline, every 12 weeks and up to 4 years

Interventionunits on a scale (Mean)
Social functionDisease-related worriesSexual functionCommunicative functioningEndocrine scaleG.I. scaleTreatment scaleMuscle/bone pain symptomBody image
GI NET7.41-0.9350.0011.115.5614.445.5633.330.00

[back to top]

Number and Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) and Deaths During the Extension Phase (E1)

The number of participants with AEs, SAEs and deaths were assessed. (NCT01595009)
Timeframe: from the first day of treatment in the extension up to 4 years

,
InterventionParticipants (Count of Participants)
AEsSAEsDeaths
GI NET1141
Lung NET (E1)410

[back to top]

Investigator-assessed Best Overall Response During the Extension Phase (E1)

Best overall response was determined from the sequence of investigator overall lesions responses according to Response Evaluation Criteria in Solid Tumors (RECIST). Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR; Progression, a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01595009)
Timeframe: from the start of treatment, every 12 weeks for the first year and then every 6 months up to 4 years

,
InterventionParticipants (Count of Participants)
Complete responsePartial responseStable diseaseProgressive diseaseUnknown
GI NET01730
Lung NET (E1)00220

[back to top]

Number and Percentage of Participants With Ratings of 'no Problem, 'Some Problem' and 'Extreme Problem' in the EuroQol Five Dimensions Questionnaire (EQ-5D) (Core)

"The EQ-5D is divided into two distinct sections. The first section includes one item addressing each of five dimensions (mobility, self-care, usual activity, pain/discomfort, and anxiety/depression). Patients rate each of these items from no problem, some problem, or extreme problem. A composite health index is then defined by combining the levels for each dimension. The second section of the questionnaire measures self-rated (global) health status utilizing a vertically oriented visual analogue scale where 100 represents the best possible health state and 0 represents the worst possible health state. Respondents are asked to rate their current health by placing a mark along this continuum. The scores from each section are then transformed into a single health utility score. Overall scores range from 0 to 1 with lower scores representing a higher level of dysfunction." (NCT01595009)
Timeframe: Baseline, weeks 4, 8, 20, 32, 44, and end of treatment (EOT) up to week 82

InterventionParticipants (Count of Participants)
Baseline, Mobility, No problemBaseline, Mobility, Some problemBaseline, Mobility, Extreme problemBaseline, Self-care, No problemBaseline, Self-care, Some problemBaseline, Usual activities, No problemBaseline, Usual activities, Some problemBaseline, Usual actvities, Extreme problemBaseline, Pain/Discomfort, No problemBaseline, Pain/discomfort, Some problemBaseline, Pain/discomfort, Extreme problemBaseline, Anxiety/depression, No problemBaseline, Anxiety/depression, Some problemBaseline, Anxiety/depression, Extreme problemWeek 4, Mobility, No problemWeek 4, Mobility, Some problemWeek 4, Mobility, Extreme problemWeek 4, Self-care, No problemWeek 4, Self-care, Some problemWeek 4, Self-care, Extreme problemWeek 4, Usual activities, No problemWeek 4, Usual activities, Some problemWeek 4, Usual activities, Extreme problemWeek 4, Pain/discomfort, No problemWeek 4, Pain/discomfort, Some problemWeek 4, Pain/discomfort, Extreme problemWeek 4, Anxiety/depression, No problemWeek 4, Anxiety depression, Some problemWeek 4, Anxiety/depression, Extreme problemWeek 8, Mobility, No problemWeek 8, Mobility, Some problemWeek 8, Mobility, Extreme problemWeek 8, Self-care, No problemWeek 8, Self-care, Some problemWeek 8, Self-care, Extreme problemWeek 8, Usual activities, No problemWeek 8, Usual activities, Some problemWeek 8, Usual activties, Extreme problemWeek 8, Pain/discomfort, No problemWeek 8, Pain/discomfort, Some problemWeek 8, Pain/discomfort, Extreme problemWeek 8, Anxiety/depression, No problemWeek 8, Anxiety/depression, Some problemWeek 8, Anxiety/depression, Extreme problemWeek 20, Mobility, No problemWeek 20, Mobility, Some problemWeek 20, Mobility, Extreme problemWeek 20, Self-care, No problemWeek 20, Self-care, Some problemWeek 20, Usual activities, No problemWeek 20 Usual activities, Some problemWeek 20, Usual activities, Extreme problemWeek 20, Pain/discomfort, No problemWeek 20, Pain/discomfort, Some problemWeek 20, Pain/discomfort, Extreme problemWeek 20, Anxiety/depression, No problemWeek 20, Anxiety/depression, Some problemWeek 20, Anxiety/depression, Extreme problemWeek 32, Mobility, No problemWeek 32, Mobility, Some problemWeek 32, Mobility, Extreme problemWeek 32, Self-care, No problemWeek 32, Self-care, Some problemWeek 32, Self-care, Extreme problemWeek 32, Usual activities, No problemWeek 32, Usual activities, Some problemWeek 32, Usual activities, Extreme problemWeek 32, Pain/discomfort, No problemWeek 32, Pain/discomfort, Some problemWeek 32, Pain/discomfort, Extreme problemWeek 32, Anxiety/depression, No problemWeek 32, Anxiety/depression, Some problemWeek 32, Anxiety/depression, Extreme problemWeek 44, Mobility, No problemWeek 44, Mobility, Some problemWeek 44, Mobility, Extreme problemWeek 44, Self-care, No problemWeek 44, Self-care, Some problemWeek 44, Usual activities, No problemWeek 44, usual activities, Some problemWeek 44, Pain/discomfort, No problemWeek 44, Pain/discomfort, Some problemWeek 44, Anxiry/depression, No problemWeek 44, Anxiety/depression, Some problemWeek 44, Anxiety/depression, Extreme problemEOT up to Week 82, Mobility, No problemEOT up to Week 82, Mobility, Some problemEOT up to Week 82, Mobility, Extreme problemEOT up to Week 82, Self-care, No problemEOT up to Week 82, Self-care, Some problemEOT up to Week 82, Self-care, Extreme problemEOT up toWeek 82, Usual activities, No problemEOT up to Week 82, Usual activities, Some problemEOT up to Week 82, Usual activities, Extreme prob.EOT up to Week 82, Pain/discomfort, No problemEOT up to Week 82, Pain/discomfort, Some problemEOT up to Week 82, Pain/discomfort, Extreme prob.EOT up to Week 82, Anxiety/depression, No problemEOT up to Week 82, Anxiety/depression, Some prob.EOT up toWeek 82, Anxiety/depression, Extreme prob
Non-pNET (Core)7635110210604664066753546663718616255427306485743345373701514041525521038418371714963022219342272352512232322214310272201459411226768761423145619129361118481131406

[back to top]

Number and Percentage of Participants With Ratings of 'no Problem, 'Some Problem' and 'Extreme Problem' in the EuroQol Five Dimensions Questionnaire (EQ-5D) (Core)

"The EQ-5D is divided into two distinct sections. The first section includes one item addressing each of five dimensions (mobility, self-care, usual activity, pain/discomfort, and anxiety/depression). Patients rate each of these items from no problem, some problem, or extreme problem. A composite health index is then defined by combining the levels for each dimension. The second section of the questionnaire measures self-rated (global) health status utilizing a vertically oriented visual analogue scale where 100 represents the best possible health state and 0 represents the worst possible health state. Respondents are asked to rate their current health by placing a mark along this continuum. The scores from each section are then transformed into a single health utility score. Overall scores range from 0 to 1 with lower scores representing a higher level of dysfunction." (NCT01595009)
Timeframe: Baseline, weeks 4, 8, 20, 32, 44, and end of treatment (EOT) up to week 82

InterventionParticipants (Count of Participants)
Baseline, Mobility, No problemBaseline, Mobility, Some problemBaseline, Mobility, Extreme problemBaseline, Self-care, No problemBaseline, Self-care, Some problemBaseline, Self-care, Extreme problemBaseline, Usual activities, No problemBaseline, Usual activities, Some problemBaseline, Usual actvities, Extreme problemBaseline, Pain/Discomfort, No problemBaseline, Pain/discomfort, Some problemBaseline, Pain/discomfort, Extreme problemBaseline, Anxiety/depression, No problemBaseline, Anxiety/depression, Some problemBaseline, Anxiety/depression, Extreme problemWeek 4, Mobility, No problemWeek 4, Mobility, Some problemWeek 4, Mobility, Extreme problemWeek 4, Self-care, No problemWeek 4, Self-care, Some problemWeek 4, Self-care, Extreme problemWeek 4, Usual activities, No problemWeek 4, Usual activities, Some problemWeek 4, Usual activities, Extreme problemWeek 4, Pain/discomfort, No problemWeek 4, Pain/discomfort, Some problemWeek 4, Pain/discomfort, Extreme problemWeek 4, Anxiety/depression, No problemWeek 4, Anxiety depression, Some problemWeek 4, Anxiety/depression, Extreme problemWeek 8, Mobility, No problemWeek 8, Mobility, Some problemWeek 8, Mobility, Extreme problemWeek 8, Self-care, No problemWeek 8, Self-care, Some problemWeek 8, Self-care, Extreme problemWeek 8, Usual activities, No problemWeek 8, Usual activities, Some problemWeek 8, Usual activties, Extreme problemWeek 8, Pain/discomfort, No problemWeek 8, Pain/discomfort, Some problemWeek 8, Pain/discomfort, Extreme problemWeek 8, Anxiety/depression, No problemWeek 8, Anxiety/depression, Some problemWeek 8, Anxiety/depression, Extreme problemWeek 20, Mobility, No problemWeek 20, Mobility, Some problemWeek 20, Self-care, No problemWeek 20, Self-care, Some problemWeek 20, Usual activities, No problemWeek 20 Usual activities, Some problemWeek 20, Usual activities, Extreme problemWeek 20, Pain/discomfort, No problemWeek 20, Pain/discomfort, Some problemWeek 20, Pain/discomfort, Extreme problemWeek 20, Anxiety/depression, No problemWeek 20, Anxiety/depression, Some problemWeek 20, Anxiety/depression, Extreme problemWeek 32, Mobility, No problemWeek 32, Mobility, Some problemWeek 32, Self-care, No problemWeek 32, Usual activities, No problemWeek 32, Usual activities, Some problemWeek 32, Pain/discomfort, No problemWeek 32, Pain/discomfort, Some problemWeek 32, Anxiety/depression, No problemWeek 32, Anxiety/depression, Some problemWeek 44, Mobility, No problemWeek 44, Mobility, Some problemWeek 44, Self-care, No problemWeek 44, usual activities, Some problemWeek 44, Pain/discomfort, Some problemWeek 44, Anxiry/depression, No problemWeek 44, Anxiety/depression, Some problemEOT up to Week 82, Mobility, No problemEOT up to Week 82, Mobility, Some problemEOT up to Week 82, Mobility, Extreme problemEOT up to Week 82, Self-care, No problemEOT up to Week 82, Self-care, Some problemEOT up to Week 82, Self-care, Extreme problemEOT up toWeek 82, Usual activities, No problemEOT up to Week 82, Usual activities, Some problemEOT up to Week 82, Usual activities, Extreme prob.EOT up to Week 82, Pain/discomfort, No problemEOT up to Week 82, Pain/discomfort, Some problemEOT up to Week 82, Pain/discomfort, Extreme prob.EOT up to Week 82, Anxiety/depression, No problemEOT up to Week 82, Anxiety/depression, Some prob.EOT up toWeek 82, Anxiety/depression, Extreme prob
pNET (Core)84312100152714345952665475722728910366314395835446148242617646234343274330126325418101121611612132541144121333125923469122542454336548333

[back to top]

Change in EORTC QLQ-G.I. NET21 Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)

The EORTC QLQ-G.I. NET21 contains 21 questions and has three defined multi-item symptom scales (endocrine - 3 questions, gastrointestinal - 5 questions, and treatment related side effects - 3 questions), two single item symptoms (bone/muscle pain and concern about weight loss), two psychosocial scales (social function - 3 questions, disease-related worries - 3 questions) and two other single items (sexuality and communication). For each of the 9 domains, final scores are transformed such that they range from 0-100, where higher scores indicate worsening outcomes. A positive change from baseline indicates worsening. (NCT01595009)
Timeframe: baseline, every 12 weeks and up to 4 years

Interventionunits on a scale (Mean)
Social functionDisease-related worriesCommunicative functioningEndocrine scaleG.I. scaleTreatment scaleBody image
Lung NET (E1)11.1144.440.0011.110.00-16.670.00

[back to top]

Investigator-assessed Progression Free Survival (PFS) (E1)

PFS was defined as the time from the date of the start of therapy in the extension study to the date of the first radiologically documented disease progression or death due to any cause. If a participant had not progressed or died at the analysis cut-off date or when he/she received any further anti-neoplastic therapy, PFS was censored at the time of the last adequate tumor evaluation before the cut-off date or the anti-neoplastic therapy start date. (NCT01595009)
Timeframe: from first date of treatment in the extension up to 4 years

InterventionDays (Median)
Lung NET (E1)159
GI NET (E1)655

[back to top]

Investigator-assessed Progression Free Survival (PFS) (Core)

PFS was defined as the time from the date of the first dose to the date of the first radiologically documented disease progression or death due to any cause. If a participant had not progressed or died at the study end date or when he/she received any further anti-neoplastic therapy, PFS was censored at the time of the last tumor assessment before the end of study date. Progression was defined,using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01595009)
Timeframe: from the day of first treatment up to 19 months

InterventionMonths (Median)
pNET (Core)7.62
Non-pNET (Core)10.78

[back to top]

Change in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-C30 (EORTC QLQ-C30) Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)

"The EORTC QLQ-C30 contains 30 questions assessed by the participant. There are 9 multiple-item scales: 5 scales that assess aspects of functioning (physical, role, cognitive, emotional, and social); 3 symptom scales (Fatigue, Pain, and Nausea and Vomiting); and a global health status and QOL scale. There are 5 single-item measures assessing additional symptoms (i.e., dyspnea, loss of appetite, insomnia, constipation, and diarrhea) and a single item concerning perceived financial impact of the disease. All but two questions have 4-point scales ranging from Not at all to Very much. The two questions concerning global health status and QOL have 7 point scales with ratings ranging from Very poor to Excellent. For each of the 14 domains, changes are calculated as value at later time point minus value at baseline, and final scores are transformed such that they range from 0-100, where higher scores indicate better outcomes. A positive change from baseline indicates improvement." (NCT01595009)
Timeframe: baseline, every 12 weeks and up to 4 years

,
Interventionscore on a scale (Mean)
Global health status/QoLFunctional scale: Physical functioningFunctional scale: Role functioningFunctional scale: Emotional functioningFunctional scale: Cognitive functioningFunctional scale: Social functioningSymptom scale: FatigueSymptom scale: Nausea and vomitingSymptom scale: PainSymptom scale: DyspneaSymptom scale: InsomniaSymptom scale: Appetite lossSymptom scale: ConstipationSymptom scale: DiarrheaSymptom scale: Financial difficulties
GI NET-0.000.005.56-1.85-16.67-8.33-3.700.0016.67-5.5611.110.005.56-16.6716.67
Lung NET (E1)0.006.67-16.67-25.00-66.670.000.000.000.000.0033.330.000.000.00-33.33

[back to top]

Mean European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-C30 (EORTC QLQ-C30) Score (Core)

"The EORTC QLQ-C30 contains 30 questions assessed by the participant. There are 9 multiple-item scales: 5 scales that assess aspects of functioning (physical, role functioning, cognitive, emotional, and social); 3 symptom scales (Fatigue, Pain, and Nausea and Vomiting); and a global health status/Quality of Life (QOL) scale. There are 5 single-item measures assessing additional symptoms (i.e., dyspnea, loss of appetite, insomnia, constipation, and diarrhea) and a single item concerning perceived financial impact of the disease. All but two questions have 4-point scales ranging from Not at all to Very much. The two questions concerning global health status/ QOL have 7 point scales with ratings ranging from Very poor to Excellent. For each of the 14 domains, final scores are transformed such that they range from 0-100, where higher scores indicate improvement." (NCT01595009)
Timeframe: Baseline, weeks 4, 8, 20, 32, 44, and end of treatment (EOT) up to week 82

,
Interventionunits on a scale (Mean)
Global health status/QOL, BaselineGlobal health status/QOL, Week 4Global health status/QOL, Week 8Global health status/QOL,Week 20Global health status/QOL, Week 32Global health status/QOL, Week 44Global health status/QOL, EOT up to Week 82Physical functioning, BaselinePhysical functioning, week 4Physical functioning, week 8Physical functioning, week 20Physical functioning, week 32Physical functioning, week 44Physical functioning, EOT up to week 82Role functioning, BaselineRole functioning, week 4Role functioning, week 8Role functioning, week 20Role functioning, week 32Role functioning, week 44Role functioning, EOT up to week 82Emotional functioning, BaselineEmotional functioning, week 4Emotional functioning, week 8Emotional functioning, week 20Emotional functioning, week 32Emotional functioning, week 44Emotional functioning, EOT up to week 82Cognitive functioning, BaselineCognitive functioning, week 4Cognitive functioning, week 8Cognitive functioning, week 20Cognitive functioning, week 32Cognitive functioning, week 44Cognitive functioning, EOT up to week 82Social functioning, BaselineSocial functioning, week 4Social functioning, week 8Social functioning, week 20Social functioning, week 32Social functioning, week 44Social functioning, EOT up to week 82Fatigue, baselineFatigue, week 4Fatigue, week 8Fatigue, week 20Fatigue, week 32Fatigue, week 44Fatigue, EOT up to week 82Nausea and vomiting, BaselineNausea and vomiting, week 4Nausea and vomiting, week 8Nausea and vomiting, week 20Nausea and vomiting, week 32Nausea and vomiting, week 44Nausea and vomiting, EOT up to week 82Pain, BaselinePain, week 4Pain, week 8Pain, week 20Pain, week 32Pain, week 44Pain, EOT up to week 82Dyspnea, BaselineDyspnea, week 4Dyspnea, week 8Dyspnea, week 20Dyspnea, week 32Dyspnea, week 44Dyspnea, EOT up to week 82Insomnia, BaselineInsomnia, week 4Insomnia, week 8Insomnia, week 24Insomnia, week 32Insomnia, week 48Insomnia, EOT up o week 82Appetite loss, BaselineAppetitie loss, week 4Appetitie loss, week 8Appetitie loss, week 24Appetitie loss, week 36Appetitie loss, week 44Appetitie loss, EOT up to week 82Constipation, BaselineConstipation, week 4Constipation, week 8Constipation, week 20Constipation, week 32Constipation, week 44Constipation, EOT up to week 82Diarrhea, BaselineDiarrhea, week 4Diarrhea, week 8Diarrhea, week 20Diarrhea, week 32Diarrhea, week 44Diarrhea, EOT up to week 82Financial difficulties, BaselineFinancial difficulties, week 4Financial difficulties, week 8Financial difficulties, week 20Financial difficulties, week 32Financial difficulties, week 44Financial difficulties, EOT up to week 82
Non-pNET (Core)60.5358.9156.5959.6556.2057.7448.8672.9768.6267.4873.7171.6276.1960.9467.2762.2558.1458.3355.2666.6749.1367.2066.6964.0263.1661.1165.6757.6380.6178.8076.3378.6575.2184.5273.4667.4267.8065.3166.0766.2472.6252.8942.3943.5848.8644.4448.7238.8956.499.467.4310.346.145.984.7615.8031.2130.8837.5533.3334.6226.1943.9024.3226.1429.1228.0735.9021.4333.7728.1832.3629.0235.0933.3328.5734.6318.6221.9025.6720.4723.9326.1933.7713.2111.337.588.774.272.3810.3936.6738.1942.4231.5840.1740.4839.0416.5216.6716.8619.8821.3719.0527.63
pNET (Core)64.9363.1559.7263.1050.061.1161.4079.8977.0276.2279.7768.0068.8975.7075.2872.4971.8379.7666.6761.1167.8474.7976.3173.3876.1981.6775.0074.6184.1786.1181.9483.9386.6794.4481.3575.5678.7175.6973.2160.0066.6775.1031.3035.2236.9134.2942.2237.0438.8311.2510.529.598.056.6711.119.6921.5725.9125.2327.9843.3338.8927.3316.1117.8021.0011.4913.3322.2225.1924.7227.7832.8824.1426.6722.2232.1720.5625.1624.6618.3920.0011.1124.1413.0611.0010.506.900.000.0012.7919.8920.9227.4016.6713.3322.2225.9718.7716.6719.4421.4340.0022.2221.71

[back to top]

Mean EQ-5D Visual Analogue Scale (VAS) Score (Core)

"The EQ-5D is divided into two distinct sections. The first section includes one item addressing each of five dimensions (mobility, self-care, usual activity, pain/discomfort, and anxiety/depression). Patients rate each of these items from no problem, some problem, or extreme problem. A composite health index is then defined by combining the levels for each dimension. The second section of the questionnaire measures self-rated (global) health status utilizing a vertically oriented visual analogue scale where 100 represents the best possible health state and 0 represents the worst possible health state. Respondents are asked to rate their current health by placing a mark along this continuum. The scores from each section are then transformed into a single health utility score. Overall scores range from 0 to 1 with lower scores representing a higher level of dysfunction." (NCT01595009)
Timeframe: Baseline, weeks 4, 8, 20, 32, 44, and end of treatment (EOT) up to week 82

,
Interventionunits on a scale (Mean)
BaselineWeek 4Week 8Week 20Week 32Week 44EOT up to Week 82
Non-pNET (Core)63.963.861.264.662.068.655.3
pNET (Core)68.869.364.667.442.666.766.5

[back to top]

Mean EORTC QLQ-G.I. NET21 Score (Core)

The EORTC QLQ-G.I. NET21 contains 21 questions and has three defined multi-item symptom scales (endocrine - 3 questions, gastrointestinal - 5 questions, and treatment related side effects - 3 questions), two single item symptoms (bone/muscle pain and concern about weight loss), two psychosocial scales (social function - 3 questions and disease-related worries - 3 questions) and two other single items (sexuality and communication). For each of the 9 domains, final scores are transformed such that they range from 0-100, where higher scores indicate worsening. (NCT01595009)
Timeframe: Baseline, weeks 4, 8, 20, 32, 44, and end of treatment (EOT) up to week 82

Interventionunits on a scale (Mean)
Endocrine, BaselineEndocrine, week 4Endocrine, week 8Endocrine, week 20Endocrine, week 32Endocrine, week 44Endocrine, EOT up to week 82G.I., BaselineG.I., week 4G.I., week 8G.I., week 20G.I., week 32G.I., week 44G.I., EOT up to week 82Treatment, baselineTreatment, week 4Treatment, week 8Treatment, week 20Treatment, week 32Treatment, week 44Treatment, EOT up to week 82Social function, BaselineSocial function, week 4Social function, week 8Social function, week 20Social function, week 32Social function, week 44Social function, EOT up to week 82Disease-related worries, BaselineDisease-related worries, week 4Disease-related worries, week 8Disease-related worries, week 20Disease-related worries, week 32Disease-related worries, week 44Disease-related worries, EOT up to week 82Muscle/bone pain, BaselineMuscle/bone pain, week 4Muscle/bone pain, week 8Muscle/bone pain, week 20Muscle/bone pain, week 32Muscle/bone pain, week 44Muscle/bone pain, EOT up to week 82Sexual function, BaselineSexual function, week 4Sexual function, week 8Sexual function, week 20Sexual function, week 32Sexual function, week 44Sexual function, EOT up to week 82Communication function, BaselineCommunication function, week 4Communication function, week 8Communication function, week 20Communication function, week 32Communication function, week 44Communication function, EOT up to week 82Body image, BaselineBody image, week 4Body image, week 8Body image, week 20Body image, week 32Body image, week 44Body image, EOT up to week 82
Non-pNET (Core)23.5217.8516.7915.8713.747.1415.2026.1923.3927.5322.3126.8420.9530.4818.4422.2220.2719.7424.4918.1824.8147.4245.2846.4942.6648.5440.0854.2253.9446.3347.3246.8353.5144.0555.0429.6633.0033.3334.5240.3526.1940.8939.3240.8943.9238.1029.1747.6248.949.886.469.6910.307.894.7612.6122.4322.7926.1030.3028.8330.9534.70

[back to top]

Number and Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) and Deaths (Core)

The number of participants with AEs, SAEs and deaths were assessed. (NCT01595009)
Timeframe: from the day of first treatment up to 19 months

,
InterventionParticipants (Count of Participants)
Adverse eventsSerious adverse eventsDeaths
Non-pNET (Core)1095910
pNET (Core)90255

[back to top]

Change in EuroQol Five Dimensions Questionnaire (EQ-5D) Score at the End of Treatment From Baseline (Baseline = First Day of Treatment in the Extension) (E1)

The EQ-5D contains 2 sections:1st section has 1 item addressing each of 5 dimensions (mobility, self-care, usual activity, pain/discomfort, anxiety/depression). Each dimension has 3 levels: no problems, some problems, extreme problems, 1-3, respectively. A health state is defined by combining 1 level from each dimension. 243 health states are possible. Each state is referred to in a 5 digit code. E.g., state 11111 indicates no problems on any dimension, while state 11223 indicates no problems with mobility and self-care, some problems with performing usual activities, moderate pain or discomfort and extreme anxiety or depression. The 2nd section measures self-rated health status using a visual analogue scale (VAS) where 100 represents best possible health and 0 represents worst possible health. Patients are asked to rate their current health by placing a mark on the VAS. Scores from each section are then transformed into an overall score of 0 or 1, with 0= higher level of dysfunction. (NCT01595009)
Timeframe: baseline, every 12 weeks and up to 4 years

,
Interventionunits on a scale (Mean)
Composite health indexSelf-rated health status
GI NET-8.58-4.17
Lung NET (E1)7.26-10.00

[back to top]

Investigator-assessed Best Overall Response (Core)

Best overall response was determined from the sequence of investigator overall lesions responses according to Response Evaluation Criteria in Solid Tumors (RECIST). Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR; Progression, a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01595009)
Timeframe: from the start of treatment, every 12 weeks for the first year and then every 6 months up to 19 months

,
InterventionParticipants (Count of Participants)
Partial response (PR)Stable disease (SD)Progressive disease (PD)UnknownObjective response rate (complete response or PR)
Non-pNET (Core)1848271
pNET (Core)2749412

[back to top]

Estimated Glomerular Filtration Rate (eGFR) at Day 8 and Week 24

(MDRDa formula) with imputation by last observation carried forward (LOCF) (NCT01596062)
Timeframe: Day 8, Week 24

,,
InterventionmL/min/1.73m^2 (Mean)
Day 8Week 24 (n= 3, 6, 6)
Simulect 40mg + Neoral + Myfortic + Steroids35.031.9
Simulect 80mg + Certican + Myfortic + Steroids49.954.4
Simulect 80mg + Neoral + Myfortic + Steroids53.855.7

[back to top]

AUC of Basiliximab Binding to CD25 Receptors From Day 0 to Day 84

Mean AUC was calculated only for patients who received two Simulect injections. (NCT01596062)
Timeframe: Day 84 (Week 12) post-transplantation

InterventionWeeks * Percentage of T cells (Mean)
Simulect 40mg + Neoral + Myfortic + Steroids7.0
Simulect 80mg + Neoral + Myfortic + Steroids9.9
Simulect 80mg + Certican + Myfortic + Steroids8.4

[back to top]

Area Under the Curve (AUC) of CD25 Saturation by Basiliximab From Day 0 to Day 84

CD25 saturation is the percentage of T cells expressing CD25. Mean AUC of CD25 was calculated only for patients who received two Simulect® injections. (NCT01596062)
Timeframe: Day 84 (Week 12) after transplantation

InterventionWeeks * Percentage of saturated CD25 (Mean)
Simulect 40mg + Neoral + Myfortic + Steroids8.4
Simulect 80mg + Neoral + Myfortic + Steroids11.1
Simulect 80mg + Certican + Myfortic + Steroids9.7

[back to top]

Saturation Rate of CD25 Antigen Saturation by Basiliximab

CD25 saturation is the percentage of T cells expressing CD25 (NCT01596062)
Timeframe: Day 0, Day 1, Day 4, Day 6, Day 14, Day 21, Day 28, Day 42, Day 56 and Day 84 (Week 12) post-transplantation

,,
Interventionpercentage of T cells (Mean)
Day 0 (before injection)Day 0 (2 hours after injection)Day 1Day 4 (before injection); (n= 3, 6, 6)Day 4 (2 hours after injection): (n= 3, 6, 5)Day 6: (n= 3, 6, 6)Day 14: (n= 3, 6, 6)Day 21: (n= 3, 6, 6)Day 28: (n= 3, 6, 6)Day 42: (n= 3, 6, 5)Day 56: (n= 3, 6, 5)Day 84 (Week 12): (n= 3, 6, 5)
Simulect 40mg + Neoral + Myfortic + Steroids0.093.795.7100.096.798.798.3100.095.378.365.00.0
Simulect 80mg + Certican + Myfortic + Steroids0.094.799.093.784.894.097.392.592.799.294.214.2
Simulect 80mg + Neoral + Myfortic + Steroids0.096.797.796.296.3100.096.794.5100.0100.093.367.5

[back to top]

Percentage of Participants With Biopsy Proven Acute Rejection (BPAR) According to Type and Severity

Antibody mediated acute rejection: C4d deposition, presence of circulating antidonor antibody, morphologic evidence of acute tissue injury such as acute tubular necrosis-like minimal inflammation or capillary and/or glomerular inflammation and/or thromboses or arterial inflammation. Cellular acute rejection: acute T-cell mediated rejection Type IA: Significant interstitial infiltration (> 25% of parenchyma) and foci of moderate tubulitis (> 4 mononuclear cells/tubular cross section or group of 10 tubular cells) Type IB: Significant interstitial infiltration (> 25% of parenchyma) and foci of severe tubulitis (> 10 mononuclear cells/tubular cross section or group of 10 tubular cells) Type IIA: Mild to moderate intimal arteritis. Type IIB: Severe intimal arteritis comprising > 25% of the lumenal area. Type III: Transmural (full vessel wall thickness) arteritis and/or arterial fibrinoid change and necrosis of medial smooth muscle cells (with accompanying lymphocytic inflammation). (NCT01596062)
Timeframe: Day 84 (Week 12), Week 24 post-transplantation

,,
InterventionPercentage of participants (Number)
Day 84 (Week 12): Antibody mediated AR - NoDay 84 (Week 12): Antibody mediated AR - YesDay 84 (Week 12): Cellular AR - NoDay 84 (Week 12): Cellular AR - YesDay 84 (Week 12): Banff type lADay 84 (Week 12): Banff type lBDay 84 (Week 12): Banff type llADay 84 (Week 12): Banff type llBDay 84 (Week 12): Banff type lll (C AR)Week 24: Antibody mediated AR - NoWeek 24: Antibody mediated AR - YesWeek 24: Cellular AR - NoWeek 24: Cellular AR - YesWeek 24: Banff type lAWeek 24: Banff type lBWeek 24: Banff type llAWeek 24: Banff type llBWeek 24: Banff type lll
Simulect 40mg + Neoral + Myfortic + Steroids66.733.3100.00.00.00.00.00.00.066.733.3100.00.00.00.00.00.00.0
Simulect 80mg + Certican + Myfortic + Steroids85.714.357.142.914.314.30.014.30.085.714.357.142.90.028.60.014.30.0
Simulect 80mg + Neoral + Myfortic + Steroids100.00.083.316.70.016.70.00.00.0100.00.083.316.70.016.70.00.00.0

[back to top]

Percentage of Participants With of Biopsy Proven Acute Rejection (BPAR)

BPAR is one of the components of treatment failure. One assessment of efficacy was BPAR. Renal graft biopsies were performed and the renal tissue was examined to determine if there was acute rejection of the renal transplant. (NCT01596062)
Timeframe: Day 84 (Week 12), Week 24 post-transplantation

,,
InterventionPercentage of participants (Number)
Day 84 (Week 12): NoDay 84 (Week 12):YesWeek 24: NoWeek 24:Yes
Simulect 40mg + Neoral + Myfortic + Steroids66.733.366.733.3
Simulect 80mg + Certican + Myfortic + Steroids42.957.142.957.1
Simulect 80mg + Neoral + Myfortic + Steroids83.316.783.316.7

[back to top]

Percentage of T-cells That Bind Basiliximab to CD25 Receptors

This is the percentage of T cells binding basiliximab at all timepoints. (NCT01596062)
Timeframe: Day 0, Day 1, Day 4, Day 6, Day 14, Day 21, Day 28, Day 42, Day 56 and Day 84 (Week 12) post-transplantation

,,
InterventionPercentage of T cells (Mean)
Day 0 (before injection)Day 0 (2 hours after injection)Day 1 (n= 3, 5, 7)Day 4 (before injection): (n= 3, 6, 6)Day 4 (2 hours after injection): (n= 3, 5, 5)Day 6: (n= 3, 6, 6)Day 14: (n= 3, 6, 6)Day 21: (n= 3, 6, 6)Day 28: (n= 3, 6, 6)Day 42: (n= 3, 6, 5)Day 56: (n= 3, 6, 5)Day 84 (Week 12): (n= 3, 6, 5)
Simulect 40mg + Neoral + Myfortic + Steroids0.095.370.0100.0100.091.7100.089.076.347.353.06.7
Simulect 80mg + Certican + Myfortic + Steroids4.781.691.453.081.298.393.885.882.778.875.215.8
Simulect 80mg + Neoral + Myfortic + Steroids0.096.264.859.083.499.089.087.791.084.088.857.7

[back to top]

Proportion of CD3+, CD4+, CD8+, CD19+ and CD56+ T Cells

Cell counts of various subpopulations of T, B and NK lymphocytes (CD3, CD4, CD8, CD19 and CD56) (flow cytometry). (NCT01596062)
Timeframe: Day 0, Day 6, Day 42, Day 84 (Week 12)

,,
Intervention10^9 cells/L (Mean)
CD3 cells count at Day 0 (before injection)CD3 cells count at Day 6: (n= 3, 6, 6)CD3 cells count at Day 42: (n= 3, 6, 5)CD3 cells count at Day 84 (Week 12): (n= 3, 6, 6)CD4 cells count at Day 0 (before injection)CD4 cells count at Day 6: (n= 3, 6, 6)CD4 cells count at Day 42: (n= 3, 6, 5)CD4 cells count at Day 84 (Week 12): (n= 3, 6, 6)CD8 cells count at Day 0 (before injection)CD8 cells count at Day 6: (n= 3, 6, 6)CD8 cells count at Day 42: (n= 3, 6, 5)CD8 cells count at Day 84 (Week 12): (n= 3, 6, 6)CD19 cells count at Day 0 (before injection)CD19 cells count at Day 6: (n=3, 6, 6)CD19 cells count at Day 42: (n= 3, 6, 5)CD19 cells count at Day 84 (Week 12): (n= 3, 6, 6)CD56 cells count at Day 0 (before injection)CD56 cells count at Day 6: (n= 3, 6, 6)CD56 cells count at Day 42: (n= 3, 6, 5)CD56 cells count at Day 84 (Week 12): (n= 3, 6, 6)
Simulect 40mg + Neoral + Myfortic + Steroids0.50.60.50.80.40.50.30.60.10.20.10.20.10.10.10.20.10.10.10.1
Simulect 80mg + Certican + Myfortic + Steroids0.70.91.11.10.50.60.80.70.20.20.40.30.10.20.20.10.20.20.20.1
Simulect 80mg + Neoral + Myfortic + Steroids0.91.11.21.20.60.70.80.80.30.30.30.30.10.20.20.10.20.10.20.1

[back to top]

Percentage of Participants With of Treatment Failures

Treatment failure was defined either as a BPAR, a graft loss, a death or a loss to follow-up. An extended treatment failure was also defined including treated borderline lesions, BPAR, graft loss, death or loss to follow-up. Treated borderline lesions were considered as acute rejection by investigators and DMC experts. (NCT01596062)
Timeframe: Day 84 (Week 12), Week 24

,,
InterventionPercentage of participants (Number)
Day84 (Week 12): BPAR and/or borderline lesions-NoDay84 (Week 12):BPAR and/or borderline lesions-YesDay 84 (Week 12): Graft loss - NoDay 84 (Week 12): Graft loss - YesDay 84 (Week 12): Death - NoDay 84 (Week 12): Death - YesDay 84 (Week 12): Loss to follow-up - NoDay 84 (Week 12): Loss to follow-up - YesWeek 24: BPAR or borderline lesions - NoWeek 24: Graft loss - NoWeek 24: Death - NoWeek 24: Loss to follow-up - NoWeek 24: Loss to follow-up - YesWeek 24: BPAR or borderline lesions - YesWeek 24: Graft loss - YesWeek 24: Death - Yes
Simulect 40mg + Neoral + Myfortic + Steroids66.733.366.733.3100.00.0100.00.066.766.7100.0100.00.033.333.30.0
Simulect 80mg + Certican + Myfortic + Steroids14.385.7100.00.0100.00.0100.00.014.3100.0100.0100.00.085.70.00.0
Simulect 80mg + Neoral + Myfortic + Steroids66.733.3100.00.0100.00.0100.00.066.7100.0100.0100.00.033.30.00.0

[back to top]

Growth Development - Height at Baseline and Month 24

"Individual growth measurements were compared with the gender and age-specific growth percentiles in the CDC growth charts for the US population. Each value observed is thus represented by the (approximated) percentage of subjects with a lower value in the reference population. Changes were calculated on this scale and thus express the change in growth measurements relative to the percentiles in the CDC growth charts.~Patients were classified into growth percentile categories (<=5, >5-25, >25-50, >50-75, >75-95 and >95% percentile)." (NCT01598987)
Timeframe: Baseline, Month 24

,,,,,,
InterventionPercentages (Number)
DecreaseNo ChangeIncrease >3 to 5%Increase >5 to 10%Increase >10%
<=5% Percentile0.050.033.30.016.7
>25% - 50% Percentile33.30.00.033.333.3
>5% - 25% Percentile12.512.512.50.062.5
>50% - 75% Percentile0.00.00.00.0100.0
>75% - 95% Percentile0.050.050.00.00.0
>95% Percentile0.0100.00.00.00.0
Total9.0027.318.24.540.9

[back to top]

Growth Development - Height at Baseline and Month 12

"Individual growth measurements were compared with the gender and age-specific growth percentiles in the CDC growth charts for the US population. Each value observed is thus represented by the (approximated) percentage of subjects with a lower value in the reference population. Changes were calculated on this scale and thus express the change in growth measurements relative to the percentiles in the CDC growth charts.~Patients were classified into growth percentile categories (<=5, >5-25, >25-50, >50-75, >75-95 and >95% percentile)." (NCT01598987)
Timeframe: Baseline, Month 12

,,,,,,
InterventionPercentages (Number)
DecreaseNo ChangeIncrease >3 to 5%Increase >5 to 10%Increase >10%
<=5% Percentile0.068.812.50.018.8
>25% - 50% Percentile16.733.30.016.733.3
>5% - 25% Percentile26.720.06.720.026.7
>50% - 75% Percentile14.328.60.014.342.9
>75% - 95% Percentile25.050.025.00.00.0
>95% Percentile0.0100.00.00.00.0
Total14.044.08.010.024.0

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate - Month 12

Evolution of renal function assessed by estimated Glomerular Filtration Rate (eGFR) calculated by the Chronic Kidney Disease in Children (CKiD) Schwartz formula (Schwartz 2009), expressed in mean change in eGFR of CKiD between start of study (baseline assessment) and Month 12. (NCT01598987)
Timeframe: Baseline, Month 12

InterventionmL/min/1.73m^2 (Mean)
Everolimus Based Regimen6.2

[back to top]

Change From Baseline in Estimated Glomerular Filtration Rate - Month 24

Evolution of renal function assessed by estimated Glomerular Filtration Rate (eGFR) calculated by the Chronic Kidney Disease in Children (CKiD) Schwartz formula (Schwartz 2009), expressed in mean change in eGFR of CKiD between start of study (baseline assessment) and Month 24. (NCT01598987)
Timeframe: Baseline, Month 24

InterventionmL/min/1.73m2 (Mean)
Everolimus Based Regimen4.5

[back to top]

Kaplan-Meier Estimates for Failure Rates of Efficacy Endpoints

"The proportion of patients with composite efficacy failure (treated biopsy proven acute rejection[tBPAR], graft loss [GL] , death [D]) before/at Month 12 and Month 24, estimated with Kaplan-Meier (KM) methods and the proportion of patients who experienced any of the components of composite efficacy failure (tBPAR, GL, D) before/at Month 12 and Month 24, separately for each component.~AR: acute rejection; BPAR: biopsy proven acute rejection. Rate = Kaplan-Meier estimate for failure in %; CI = confidence interval for failure rate." (NCT01598987)
Timeframe: At 12-month and 24-month after start of study drug

InterventionPercentages (Number)
Month 12: tBPAR,GL,or DMonth 12: tBPAR,GL,D,or loss to follow-upMonth 12: Treated BPARMonth 12: Graft lossMonth 12: DeathMonth 12: Graft loss or deathMonth 12: BPARMonth 12: Treated ARMonth 24: tBPAR,GL,or DMonth 24: tBPAR,GL,D,or loss to follow-upMonth 24: Treated BPARMonth 24: Graft LossMonth 24: DeathMonth 24: Graft loss or deathMonth 24: BPARMonth 24: Treated AR
Everolimus Based Regimen1.91.91.90.00.00.03.73.65.99.75.90.00.00.011.97.7

[back to top]

Growth Development - Weight at Baseline and Month 24

"Individual growth measurements were compared with the gender and age-specific growth percentiles in the CDC growth charts for the US population. Each value observed is thus represented by the (approximated) percentage of subjects with a lower value in the reference population. Changes were calculated on this scale and thus express the change in growth measurements relative to the percentiles in the CDC growth charts.~Patients were classified into growth percentile categories (<=5, >5-25, >25-50, >50-75, >75-95 and >95% percentile)." (NCT01598987)
Timeframe: Baseline, Month 24

,,,,,
InterventionPercentages (Number)
DecreaseNo ChangeIncrease >3 to 5%Increase >5 to 10%Increase >10%
<=5% Percentile0.050.016.716.716.7
>25% - 50% Percentile100.00.00.00.00.0
>5% - 25% Percentile40.00020.040.0
>50% - 75% Percentile12.525.012.512.537.5
>95% Percentile0.0100.00.00.00.0
Total22.727.39.113.627.3

[back to top]

Growth Development - Weight at Baseline and Month 12

"Individual growth measurements were compared with the gender and age-specific growth percentiles in the CDC growth charts for the US population. Each value observed is thus represented by the (approximated) percentage of subjects with a lower value in the reference population. Changes were calculated on this scale and thus express the change in growth measurements relative to the percentiles in the CDC growth charts.~Patients were classified into growth percentile categories (<=5, >5-25, >25-50, >50-75, >75-95 and >95% percentile)." (NCT01598987)
Timeframe: Baseline, Month 12

,,,,,,
InterventionPercentages (Number)
DecreaseNo ChangeIncrease >3 to 5%Increase >5 to 10%Increase >10%
<=5% Percentile0.033.30.026.740.0
>25% - 50% Percentile42.90.00.00.057.1
>5% - 25% Percentile20.020.00.00.060.0
>50% - 75% Percentile72.718.20.09.10.0
>75% - 95% Percentile16.733.316.70.033.3
>95% Percentile0.0100.00.00.00.0
Total28.024.02.010.036.0

[back to top]

Evidence of Reduction of BK Viruria and/or Clearance of BK Viremia

composite outcome of a 50% or greater reduction in BKV urine levels and/or complete clearance of BKV viremia by 3 months after randomization (NCT01624948)
Timeframe: 3 months post-randomization

Interventionparticipants (Number)
Everolimus+Tacrolimus/Prednisone11
Standard of Care: 50% Reduction of MPA8

[back to top]

Proteinuria

(NCT01624948)
Timeframe: 3 months post-randomization

Interventiong/g creatinine (Mean)
Everolimus+Tacrolimus/Prednisone0.16
Standard of Care: 50% Reduction of MPA0.23

[back to top]

p70S6 Kinase Phosphorylation

(NCT01624948)
Timeframe: 3 months post-randomization

InterventionMean Fluorescence Intensity (Median)
Reached Primary Endpoint5002
Failed to Reach Primary Endpoint4353

[back to top]

Cholesterol

(NCT01624948)
Timeframe: 3 months post-randomization

Interventionmg/dL (Mean)
Everolimus+Tacrolimus/Prednisone212
Standard of Care: 50% Reduction of MPA170

[back to top]

Evaluation for the Development of BK Virus Nephropathy or Doubling of BK Viremia Levels

A doubling of BK viremia levels or the development of BKV nephropathy in subjects enrolled in the experimental study arm will prompt a conversion to standard care therapy. We will closely monitor BKV levels in the urine and blood and assess renal function monthly, as per our usual standard of care. Based on BKV results as well as renal function, as assessed by serum Cr, biopsies may be done for cause. Patients will have a final visit at month 4 to monitor for adverse events. (NCT01624948)
Timeframe: 3 months post-randomization

Interventionparticipants (Number)
Everolimus+Tacrolimus/Prednisone2
Standard of Care: 50% Reduction of MPA2

[back to top]

Median Between the Calculated Mean Residual Expression of NFAT-regulated Genes

"Measurement of the expression of three NFAT-regulated genes IL-2, interferon gamma, and GM-CSF, to predict a rejection episode.~The residual gene expression after Tacrolimus intake was calculated as T1.5/T0*100, where T0 is the adjusted number of transcripts at Tacrolimus pre-dose level and T1.5 is the number of transcripts 1.5 hours after drug intake. For all three genes the residual expression was averaged and presented as MRE of NFAT-regulated genes." (NCT01624948)
Timeframe: 3 months post-randomization

Interventionpercent residual expression (Median)
Rejection46.35
No Rejection29.12

[back to top]

Change From Baseline (Randomization) in Serum Creatinine

"Change in serum creatinine concentrations from baseline (randomization) to week 24 post-randomization was one of the efficacy assessments of renal function.~Baseline was Day 28 visit." (NCT01625377)
Timeframe: Baseline, Week 24

Interventionµmol/L (Mean)
Tacrolimus7.2
Everolimus (RAD001)-1.3

[back to top]

Number of Patients With Treated or Not Treated Biopsy Proven Acute Rejection (BPAR)

Biopsy proven acute rejection was defined as a clinically suspected acute rejection confirmed by biopsy. (NCT01625377)
Timeframe: at 12 week and 24 week

,
InterventionPatients (Number)
Week 12, Treated BPARWeek 12, Not treated BPARWeek 24, Treated BPARWeek 24, Not Treated BPAR
Everolimus (RAD001)2081
Tacrolimus2020

[back to top]

Change From Baseline (Randomization) in Creatinine Clearance Estimated Using the Adjusted Cockcroft-Gault Formula

Creatinine clearance by the Cockcroft-Gault formula is computed in mL/min/1.73m^2 from the creatinine clearance in mL/min by multiplying it by 1.73 and dividing it by the body surface area Baseline was Day 28 visit. (NCT01625377)
Timeframe: Baseline, Week 24

InterventionmL/min/1.73m^2 (Mean)
Tacrolimus-9.0
Everolimus (RAD001)0.7

[back to top]

Change From Baseline (Randomization) in Glomerular Filtration Rate Estimated by CKD-EPI Formula

"GFR estimated by using the Chronic kidney disease- epidemiology (CKD-EPI) formula:~eGFR (mL/min/1.73m^2) = 141 * min(C/K,1)^ α * max(C/K,1)^-1.209 * 0.993^A * 1.1018 (if male) * 1.159 (if black) where C = serum creatinine (in mg/dL) ; A = Age (in years); K = 0.7 for women and 0.9 for men; α = -0.329 for women and -0.411 for men. Baseline was Day 28 visit." (NCT01625377)
Timeframe: Baseline, Week 24

InterventionmL/min/1.73m^2 (Mean)
Tacrolimus-6.9
Everolimus (RAD001)2.4

[back to top]

Change From Baseline (Randomization) in Renal Function

"Change in renal function was measured by change in glomerular filtration rate (GFR). GFR calculated using the abbreviated modification of diet in renal disease (aMDRD) formula.~GFR in mL/min/1.73m^2 for men of non-black ethnicity: 186 * [C/88]^-1.154 * [A]^-0.023*G*R ; C = serum creatinine (in μmol/L); A = Age (in years). G = 0.742 when the patient is a women; Otherwise G=1 R= 1.21 when the patient was of black ethnicity; Otherwise R = 1 Baseline was Day 28 visit." (NCT01625377)
Timeframe: Baseline, Week 24

InterventionmL/min/1.73m^2 (Least Squares Mean)
Tacrolimus-13.29
Everolimus (RAD001)1.05

[back to top]

Change From Baseline (Randomization) in Urine Protein/Creatinine Ratio

Change in urine protein/creatinine ratio from baseline (randomization) to week 24 post-randomization was one of the efficacy assessments of renal function. Baseline was Day 28 visit. (NCT01625377)
Timeframe: Baseline, week 24

Interventionmg/mmol (Mean)
Tacrolimus-2.3
Everolimus (RAD001)21.9

[back to top]

Change From Baseline (Randomization) in Glomerular Filtration Rate Estimated by Abbreviated Modification of Diet in Renal Disease (MDRD) Formula

"Change in glomerular filtration rate was calculated using the MDRD abbreviated formula.~GFR in mL/min/1.73m^2 for men of non-black ethnicity: 186 * [C/88]^-1.154 * [A]^-0.023*G*R ; C = serum creatinine (in μmol/L); A = Age (in years). G = 0.742 when the patient is a women; Otherwise G=1 R= 1.21 when the patient was of black ethnicity; Otherwise R = 1 Baseline was Day 28 visit." (NCT01625377)
Timeframe: Baseline, Week 24

InterventionmL/min/1.73m^2 (Mean)
Tacrolimus-11.8
Everolimus (RAD001)0.1

[back to top]

Number of Patients in Different Stages of Chronic Kidney Diseases According to the K/DOQI Classification System

"Kidney disease outcomes quality initiative (K/DOQI) classification is based on glomerular filtration rate (GFR), abbreviated MDRD formula (mL/min/1.73m^2) :~Stage 1 : GFR >= 90; Stage 2 = GFR was between 60-89; Stage 3 = GFR was between 30-59 ; Stage 4 = GFR was between 15-29; Stage 5 = GFR was < 15 (or dialysis)" (NCT01625377)
Timeframe: At Week 24

,
InterventionPatients (Number)
Stage 1Stage 2Stage 3Stage 4Stage 5
Everolimus (RAD001)4129400
Tacrolimus24342800

[back to top]

Number of Patients Reported With Different Categories of Severity of BPAR According to Banff Classification

"Biopsy proven acute rejection was defined as a clinically suspected acute rejection confirmed by biopsy.~The severity of BPAR was categorized as :~Mild (Banff grade I, RAI = 4 and 5) Moderate (Banff grade II, RAI = 6 and 7) Severe (Banff grade III, RAI = 8 and 9) Banff Rejection Activity Index (RAI) comprises 3 components scored from 0 to 3: venous endothelial inflammation; bile duct inflammation damage; and portal inflammation; the scores are combined to an overall score (the RAI) ranging from 0 to 9. An overall score of 0-3 is considered indeterminate, score of 4-5 is mild acute, score of 6-7 is moderate acute , and score of 8-9 is severe acute. Only the episode with the highest total RAI score for each participant was counted." (NCT01625377)
Timeframe: at 12 week and 24 week

,
InterventionPatients (Number)
Week 12: MildWeek 12: ModerateWeek 12: SevereWeek 24: MildWeek 24: ModerateWeek 24: Sever
Everolimus (RAD001)110530
Tacrolimus110110

[back to top]

Number of Patients With Any Adverse Events, Serious Adverse Events, Death and Premature Discontinuation

Baseline was Day 28 visit. This endpoint reports patients with total adverse events (any), serious adverse events, death and premature discontinuation. (NCT01625377)
Timeframe: Baseline to 24 weeks

,
InterventionPatients (Number)
Any Adverse eventsSerious Adverse eventsDeathAt least one AE led to premature discontinuation
Everolimus (RAD001)8142218
Tacrolimus852814

[back to top]

Number of Patients With Death or Graft Loss

The graft was presumed to be lost on the day the patient was registered again on the waiting list, or the day he/she received a new graft. (NCT01625377)
Timeframe: at week 24

,
InterventionPatients (Number)
Graft LossDeath
Everolimus (RAD001)01
Tacrolimus11

[back to top]

Number of Patients With Treated or Untreated BPAR With RAI Score Greater Than 3

"Biopsy proven acute rejection (BPAR) was defined as a clinically suspected acute rejection confirmed by biopsy. The Banff Rejection Activity Index (RAI) comprises 3 components scored from 0 to 3: venous endothelial inflammation; bile duct inflammation damage; and portal inflammation; the scores are combined to an overall score (the RAI) ranging from 0 to 9. An overall score of 0-3 is considered indeterminate, score of 4-5 is mild acute, score of 6-7 is moderate acute , and score of 8-9 is severe acute. Only the episode with the highest total RAI score for each participant was counted.~The patients with treated or untreated BPAR having RAI score > 3 were reported in this end point." (NCT01625377)
Timeframe: At 24 weeks

,
InterventionPatients (Number)
Not Treated BPAR: RAI score >3Not Treated BPAR: Missing RAI scoreTreated BPAR: RAI score >3
Everolimus (RAD001)018
Tacrolimus002

[back to top]

Number of Patients With Treatment Failures

"Incidence of treatment failures, assessed with composite criterion including treated biopsy proven acute rejection (tBPAR) with a rejection activity index (RAI) according to Banff classification >3, graft loss or death at 6 months.~Biopsy proven acute rejection (BPAR) was defined as a clinically suspected acute rejection confirmed by biopsy. The Banff Rejection Activity Index (RAI) comprises 3 components scored from 0 to 3: venous endothelial inflammation; bile duct inflammation damage; and portal inflammation; the scores are combined to an overall score (the RAI) ranging from 0 to 9. An overall score of 0-3 is considered indeterminate, score of 4-5 is mild acute, score of 6-7 is moderate acute , and score of 8-9 is severe acute. Only the episode with the highest total RAI score for each participant was counted.~The graft was presumed to be lost on the day the patient was registered again on the waiting list, or the day he/she received a new graft." (NCT01625377)
Timeframe: At week 12 and week 24

,
InterventionPatients (Number)
week 12, Treatment failures - NOweek 12, Treatment failures - YESweek 24, Treatment failures - NOweek 24, Treatment failures - YES
Everolimus (RAD001)882819
Tacrolimus912894

[back to top]

Overall Survival (OS)

Overall survival (OS) was defined as the time from study enrollment to death from any cause. Data on vital status were collected after study completion through May 1, 2018 and were used in the determination of OS. (NCT01627067)
Timeframe: From the registration until disease progression, death, unacceptable toxicity, or withdraw of study consent, whichever occurred first assessed up to 67 months

Interventionmonths (Median)
Everolimus + Exemestane + Metformin28.8

[back to top]

Progression-Free Survival (PFS)

The primary end point of this study was progression-free survival (PFS), defined as the time from study enrollment to disease progression or death from any cause, whichever occurred first. Data for PFS were censored at the time of a patient's removal from study. All other analyses were post-hoc and should be regarded as such. (NCT01627067)
Timeframe: From the registration until disease progression, death, unacceptable toxicity, or withdraw of study consent, whichever occurred first assessed up to 67 months

Interventionmonths (Median)
Everolimus + Exemestane + Metformin6.3

[back to top]

Compare Overall Survival (OS) Between the Number of Obese and Overweight Participants

Overall survival (OS) was defined as the time from study enrollment to death from any cause. Data on vital status were collected after study completion through May 1, 2018 and were used in the determination of OS. Compare OS between obese participants ( n=11; BMI >/=25 kg/m2) and overweight participants (n=11; BMI NCT01627067)
Timeframe: From the registration until disease progression, death, unacceptable toxicity, or withdraw of study consent, whichever occurred first assessed up to 67 months

InterventionParticipants (Count of Participants)
Obese participantsOverweight participants
Compare OS Between Obese Patient and Overweight Participants1111

[back to top]

Compare Progression Free Survival (PFS) Between the Number of Obese and Overweight Participants

The primary end point of this study was progression-free survival (PFS), defined as the time from study enrollment to disease progression or death from any cause, whichever occurred first. Data for PFS were censored at the time of a participants removal from study. Compare PFS between overweight patients (n=11; BMI /=25 kg/m2) on univariable cox regression analysis. (NCT01627067)
Timeframe: From the registration until disease progression, death, unacceptable toxicity, or withdraw of study consent, whichever occurred first assessed up to 67 months

InterventionParticipants (Count of Participants)
Obese participantsOverweight participants
Compare PFS Between Obese and Overweight Participants1111

[back to top]

Number of Participants With Response

Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. The clinical benefit rate (CBR) was defined as the proportion of patients with a best overall response of CR, PR or SD (non-CR/non-PD for patients without measurable disease) lasting at least 24 weeks. For patients with a best overall response of CR or PR, the duration of response (DOR) was defined as the time from first documentation of CR or PR to the time of disease progression. Data on the DOR and PFS were censored at the time of a patient's removal from the study. For patients with a best overall response of SD (non-CR/non-PD for patients without measurable disease),the duration of SD (non-CR/non-PD for patients without measurable disease) was defined as the time from study enrollment to the time of disease progression. (NCT01627067)
Timeframe: From the registration until disease progression, death, unacceptable toxicity, or withdraw of study consent, whichever occurred first assessed up to 67 months

InterventionParticipants (Count of Participants)
Complete ResponsePartial responseStable Disease lasting >=24 wksStable Disease lasting <24 wksProgressive diseaseOverall response (CR+PR)Clinical benefit (CR+PR+SD >/= 24 wks
Everolimus + Exemestane + Metformin05755512

[back to top]

Progression Free Survival (PFS)

PFS is defined as the time from the date of randomization until the date of the first radiologically documented disease progression or death due to any cause. PFS is based on local investigator assessment. Patients will be followed up for the duration of the study and for an expected average of every 12 weeks after randomization. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of all target lesions, or unequivocal progression of non-target lesions, or the appearance of new lesions. (NCT01628913)
Timeframe: up to approx. 18 months

InterventionMonths (Median)
BEZ2358.2
Everolimus10.8

[back to top]

Efficacy of Treatment

Determine the efficacy of everolimus in the treatment of CTCL as overall response rate (ORR) (NCT01637090)
Timeframe: 12 months after beginning treatment

Intervention ()
All Patients on Study0

[back to top]

Progression-free Survival

Determine progression-free survival of CTCL patients treated with everolimus (NCT01637090)
Timeframe: two years after discontinuing study treatment

Intervention ()
All Patients on Study0

[back to top]

Effect of mTOR on Tumors

Determine mTOR (mammilian target of rapamycin) pathway activation and number of regulatory T cells (Tregs) in pre-treated tumor tissue and evaluate changes following treatment (NCT01637090)
Timeframe: one year

Intervention ()
All Patients on Study0

[back to top]

Time to Response

Determine time to response (TTR)/duration of objective response (DOR) (NCT01637090)
Timeframe: three months

Intervention ()
All Patients on Study0

[back to top]

Number of Participants With Adverse Events as a Measure of Safety and Tolerability

Determine the adverse event profile and tolerability of everolimus in patients with CTCL (NCT01637090)
Timeframe: Up to one year

Interventionparticipants (Number)
All Patients on Study3

[back to top]

Median PFS

Kaplan-Meier PFS will be measured from the date that study therapy is initiated until the date of first evidence of radiographic disease progression, global clinical deterioration as determined by study investigators, or death. (NCT01642186)
Timeframe: 2 years

Interventionmonths (Median)
Arm A Everolimus2.6
Arm B Letrozole Plus Leuprolide2.7
Arm C Combination Everolimus, Letrozole and Leuprolide2.4

[back to top]

Number of Participants With One or More Adverse Events/Toxicity

Adverse events/toxicity will be monitored and recorded using the CTCAE version 4.0 and summarized descriptively. (NCT01642186)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm A Everolimus9
Arm B Letrozole Plus Leuprolide9
Arm C Combination Everolimus, Letrozole and Leuprolide10

[back to top]

Number of Participants With Tissue Biomarkers Collected

Associations between baseline tissue biomarkers and PFS6 will be assessed using Fisher's exact test for categorical biomarkers, trend tests for ordinal biomarkers and Wilcoxon rank-sum test for continuous biomarkers. For patients undergoing surgery, changes in tissue biomarkers from baseline to surgery will be summarized descriptively in an exploratory fashion. (NCT01642186)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm A Everolimus9
Arm B Letrozole Plus Leuprolide9
Arm C Combination Everolimus, Letrozole and Leuprolide10

[back to top]

Percentage of Participants With Stable Disease

Objective responses will be reported using RECIST guidelines (version 1.1). Objective response will be estimated using binomial proportions and exact 95% CIs will be provided. Stable Disease is defined as neither sufficient shrinkage (compared to baseline) to qualify for Partial Reponse nor sufficient increase (taking as reference the smallest sum diameters while on study) to qualify for Progressive Disease. (NCT01642186)
Timeframe: 2 years

InterventionPercentage of pts with stable disease (Number)
Arm A Everolimus55
Arm B Letrozole Plus Leuprolide37
Arm C Combination Everolimus, Letrozole and Leuprolide11

[back to top]

Median Overall Survival (OS)

Kaplan-Meier OS will be measured from the date that study therapy was commenced until the date of death. (NCT01642186)
Timeframe: From date of randomization until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 100 months

Interventionmonths (Median)
Arm A Everolimus12.5
Arm B Letrozole Plus Leuprolide14.0
Arm C Combination Everolimus, Letrozole and Leuprolide10.6

[back to top]

Change in T Cell & B Cell Generation

Evaluate the change in regulatory T cell generation and review the relationship of the newly generated T cells with their function in the two maintenance immunosuppressive regimens at baseline, 3 and 12 months post-transplant. (NCT01653847)
Timeframe: Baseline, 3 months, and 12 months post-transplant

,
InterventionMean % of Treg cells in peripheral blood (Mean)
Baseline3 Months12 Months
Group 1: Tacrolimus With MMF.1.050.80.81
Group 2: Tacrolimus With Everolimus0.931.121.18

[back to top]

Renal Allograft Survival

The number of subjects with renal allograft survival. (NCT01653847)
Timeframe: 12 months post-transplant

InterventionParticipants (Count of Participants)
Group 1: Tacrolimus With MMF.20
Group 2: Tacrolimus With Everolimus20

[back to top]

Patient Survival

The number of patients who were alive at 2 years post transplant (NCT01653847)
Timeframe: baseline - 24 months post transplant

InterventionParticipants (Count of Participants)
Group 1: Tacrolimus With MMF.20
Group 2: Tacrolimus With Everolimus20

[back to top]

Acute Rejection

Number of subjects who experience acute rejection of the renal allograft. (NCT01653847)
Timeframe: 12 months post transplant

InterventionParticipants (Count of Participants)
Group 1: Tacrolimus With MMF.4
Group 2: Tacrolimus With Everolimus0

[back to top]

Change in Glomerular Filtration Rate (GFR)

Evaluate the change in graft function (as measured by GFR) at 12 months post-transplant from baseline. (NCT01653847)
Timeframe: 3 months, 6 months, and 12 months post-transplant

,
Interventionml/minutes per 1.73 meters^2 (Mean)
3 months6 months12 months
Group 1: Tacrolimus With MMF.636465
Group 2: Tacrolimus With Everolimus666472

[back to top]

Vascular Endothelial Growth Factor (VEGF) and Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) Levels at Baseline and Pre-Cycles 3 and 5

To assess changes in Vascular Endothelial Growth Factor (VEGF) and Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) Levels in peripheral blood specimens during treatment. (NCT01661283)
Timeframe: Baseline, Pre-Cycle 3, Pre-Cycle 5

Interventionpg/mL (Median)
VEGF- BaselineVEGF- Pre Cycle 3VEGF- Pre Cycle 5VEGFR2- BaselineVEGFR2 Pre Cycle 3VEGFR2 Pre Cycle 5
Treatment103.58184.24192.491363.76911.89863.85

[back to top]

Clinical Benefit Rate (Complete Response, Partial Response, and Stable Disease at ≥ 4 Months Using World Health Organization (WHO) Criteria) of Everolimus in Combination With Bevacizumab

"Evaluate if the combination of the mTOR inhibitor everolimus combined with the angiogenesis inhibitor bevacizumab would result in a modest clinical benefit rate, which included confirmed partial and complete responses and disease stability for four or more treatment cycles based on WHO Response Criteria. Per WHO for target lesions: Complete Response (CR): Disappearance of all known disease, determined by two consecutive observations not less than 4 weeks apart.~Partial Response (PR): A > 50% decrease in the total tumor load of the lesions that have been measured to determine the effect of therapy not less than four weeks apart. The observations must be consecutive. Stable Disease (SD): A 50% decrease in total tumor area cannot be established nor has a 25% increase in the size of one or more measurable lesions been demonstrated." (NCT01661283)
Timeframe: Assessed at Baseline and prior to Cycle 3, 5, 7, 9, etc., for up to 2 years. 1 cycle =28 days

InterventionParticipants (Count of Participants)
Treatment3

[back to top]

Number of Participants With Response Stratified by Individuals With Sporadic or NF1 Associated MPNST

To explore differences in the response rate to everolimus in combination with bevacizumab in individuals with sporadic and NF1 associated MPNST. Responses include confirmed partial and complete responses and disease stability for four or more treatment cycles based on WHO Response Criteria. Per WHO for target lesions: Complete Response (CR): Disappearance of all known disease, determined by two consecutive observations not less than 4 weeks apart. Partial Response (PR): A > 50% decrease in the total tumor load of the lesions that have been measured to determine the effect of therapy not less than four weeks apart. The observations must be consecutive. Stable Disease (SD): A 50% decrease in total tumor area cannot be established nor has a 25% increase in the size of one or more measurable lesions been demonstrated. (NCT01661283)
Timeframe: Assessed at Baseline and prior to Cycle 3, 5, 7, 9, etc., for up to 2 years. 1 cycle =28 days

InterventionParticipants (Count of Participants)
NF1 associated MPNSTSporadic MPNST
Treatment21

[back to top]

Safety as Assessed by Avoidance of Grade 3-4 Adverse Events

Number of participants who did not experience at least one grade 3-4 adverse event by CTCAE 4.0. (NCT01665768)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Everolimus and Rituximab1

[back to top]

Event Free Survival (EFS)

Percentage of participants alive without disease progression. As defined by Cheson criteria, disease progression is a new lesion or >= 50% increase in the size of previously identified sites of disease. EFS was estimated using Kaplan-Meier survival analysis. (NCT01665768)
Timeframe: 2.5 years

Interventionpercentage of participants (Number)
Everolimus and Rituximab58

[back to top]

Number of Participants With Abnormal Hematology and Serum Chemistry Laboratory Parameters by Worse CTC Grade - SI Units

Common Terminology Criteria (CTC) version 4.0 in International System of Units (SI); Gr 3=Severe, Gr 4= Potentially Life-threatening or disabling. Hematology parameters=Hemoglobin (Gr 3: < 8.0 g/dL), Platelet Count (Gr 3: 25.0 -< 50.0*10^9 c/L; Gr 4: < 25.0*10^9 c/L), Leukocyte Count (Gr 3: 1.0 -< 2.0*10^3 c/µL; Gr4: < 1.0*10^3 c/µL), Absolute Lymphocyte Count (Gr 3: 0.2 -< 0.5*10^3 c/µL; Gr 4: < 0.2*10^3 c/µL), Absolute Neutrophil Count (Gr 3: 0.5 - < 1.0*10^3 c/µL; Gr 4: < 0.5*10^3 c/µL). Liver Function parameters=Alkaline Phosphatase (Gr 3: > 5.0 - 20.0 U/L * ULN; Gr 4: > 20.0 U/L * ULN), AST (Gr 3: > 5.0 - 20.0 U/L * ULN; Gr 4: > 20.0 U/L * ULN), ALT (Gr 3: > 5.0 - 20.0 U/L * ULN; Gr 4: > 20.0 U/L * ULN), tBIL (Gr 3: > 3.0 - 10.0 mg/dL * ULN; Gr 4: > 10.0 mg/dL * ULN). Renal parameter=Creatinine (Grade: Gr3: > 3.0 - 6.0 mg/dL *ULN; Gr4: > 6.0 mg/dL *ULN). Cells per microliter (c/µL). Cells per Liter (c/L). Grams per deciliter (g/dL). Milligrams per deciliter (mg/dL). (NCT01668784)
Timeframe: Day 1 to 30 days post study completion (approximately 106 months)

,
InterventionParticipants (Count of Participants)
Hemoglobin, Grade 3Platelet Count, Grade 3Platelet Count, Grade 4Leukocytes, Grade 3Leukocytes, Grade 4Lymphocytes (absolute), Grade 3Lymphocytes (absolute), Grade 4Absolute Neutrophil Count, Grade 3Absolute Neutrophil Count, Grade 4Alkaline Phosphatase, Grade 3Aspartate Aminotransferase, Grade 3Aspartate Aminotransferase, Grade 4Alanine Aminotransferase, Grade 3Alanine Aminotransferase, Grade 4Bilirubin Total, Grade 3Creatinine, Grade 3Creatinine, Grade 4Hypercalcemia, Grade 3Hypercalcemia, Grade 4Hypocalcemia, Grade 3Hypocalcemia, Grade 4Hyperkalemia, Grade 3Hyperkalemia, Grade 4Hypokalemia, Grade 3Hypermagnesmia, Grade 3Hypomagnesmia, Grade 3Hyponatremia, Grade 3Hyponatremia, Grade 4
Everolimus6161104362136030251114070300221
Nivolumab3310012830011921213537421113531261

[back to top]

Number of Participants Meeting Marked Laboratory Abnormality Criteria in Specific Liver and Thyroid Tests

Aspartate aminotransferase, AST. Alanine aminotransaminase, ALT. Total bilirubin, tBIL. Thyroid stimulating hormone, TSH. Upper limit of normal (ULN). Units per Liter (U/L). Results reported in International System of Units (SI). (NCT01668784)
Timeframe: Day 1 to 30 days post study completion (approximately 106 months)

,
InterventionParticipants (Count of Participants)
ALT or AST > 3.0*ULNALT or AST > 5.0*ULNALT or AST > 10.0*ULNALT or AST > 20.0*ULNTotal Bilirubin > 2.0*ULNALT or AST>3.0*ULN, tBIL>2.0 ULN, 1dayALT or AST>3.0*ULN, tBIL>2.0 ULN,30dayTSH > ULNTSH < LLN
Everolimus157102017860
Nivolumab32209263415960

[back to top]

Overall Survival (OS) at Primary Endpoint

Overall Survival (OS) was defined as the time from randomization to the date of death. Participants that had not died were censored at last known date alive. Median OS time was calculated using Kaplan-Meier Estimates. Interim analysis for the Primary Endpoint occurred after 398 deaths (70% of the total OS events needed for final analysis). At that time the data monitoring committee noted that the pre-specified boundary for OS (nominal significance level p < 0.0148) was crossed while no new safety signals that would affect continuation of the study were found. The study was stopped early by the Sponsor, Bristol-Myers Squibb (BMS) and the interim analysis became the final analysis. As a result, participants in the everolimus groups could be assessed for a crossover to nivolumab treatment if they met all inclusion criteria. (NCT01668784)
Timeframe: Randomization until 398 deaths, up to May 2015 (approximately 30 months)

Interventionmonths (Median)
Nivolumab25.00
Everolimus19.55

[back to top]

Investigator-assessed Objective Response Rate (ORR)

ORR is defined as Percentage of participants with a best response of complete response (CR) or partial response (PR) divided by number of randomized participants. CR=Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.; PR=At least a 30% decrease in the sum of diameters of target lesions, the baseline sum diameters used as reference. Tumor assessments began at 8 weeks following randomization and continued every 8 weeks for the first year, then every 12 weeks thereafter until disease progression or death. CIs used Clopper and Pearson. (NCT01668784)
Timeframe: from randomization up to disease progression or death (approximately up to 105 Months)

Interventionpercentage of participants (Number)
Nivolumab25.9
Everolimus6.1

[back to top]

Extended Collection to Post Hoc Overall Survival (OS)

"Overall Survival (OS) was defined as the time from randomization to the date of death. Participants that had not died were censored at last known date alive. Median OS time was calculated using Kaplan-Meier Estimates. Interim analysis for the Primary Endpoint occurred after 398 deaths (70% of the total OS events needed for final analysis). At that time the data monitoring committee noted that the pre-specified boundary for OS (nominal significance level p < 0.0148) was crossed while no new safety signals that would affect continuation of the study were found. The study was stopped early by the Sponsor, Bristol-Myers Squibb (BMS) and the interim analysis became the final analysis. As a result, participants in the everolimus groups could be assessed for a crossover to nivolumab treatment if they met all inclusion criteria.~This outcome measure represents an updated version of the primary endpoint to include additional data collection that has occurred after the primary completion date. (NCT01668784)
Timeframe: from randomization up to disease progression or death (approximately up to 105 months)

InterventionMonths (Median)
Nivolumab25.82
Everolimus19.55

[back to top]

Investigator-assessed Duration of Objective Response

Duration of objective response is defined as the time from study start date to response, CR or partial response, PR) to the date of the first documented tumor progression as determined by the investigator (per RECIST 1.1 criteria or clinical) or death due to any cause, whichever occurred first. For participants who neither progress nor die, the duration of objective response were censored at the same time they were censored for the primary definition. CR=Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.; PR=At least a 30% decrease in the sum of diameters of target lesions, the baseline sum diameters used as reference. Based on Kaplan-Meier Estimates. (NCT01668784)
Timeframe: From randomization to date of disease progression or death or censoring if no progression or death occurred (approximately 105 months)

Interventionmonths (Median)
Nivolumab13.11
Everolimus10.18

[back to top]

Number of Participants With Serious Adverse Events, Death, Discontinuation Due to Adverse Events

Adverse event (AE) defined: any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. Serious adverse event (SAE) defined: a medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency/abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. (NCT01668784)
Timeframe: Day of first dose to 30 days post study completion (approximately 106 months)

,
InterventionParticipants (Count of Participants)
DeathsSAEsDrug related SAEsDrug related AEsDiscontinued due to Drug-related AEsDiscontinued due to AEsAdverse Events
Everolimus342177553535182387
Nivolumab324211513274085398

[back to top]

Investigator-assessed Time of Progression-free Survival (PFS)

PFS=time from randomization to date of first documented tumor progression as determined by investigator (per RECIST 1.1 criteria or clinical) or death due to any cause, whichever occurred first. Participants who die without a reported prior progression and without subsequent anti-cancer therapy were considered to have progressed on the date of their death. Participants who did not progress or die were censored on the date of their last evaluable tumor assessment. Participants who did not have any on-study tumor assessments and did not die were censored on the date they were randomized. Participants who received any subsequent anti-cancer therapy without a prior reported progression were censored at the last evaluable tumor assessment prior to or on initiation date of the subsequent anti-cancer therapy. Progressive disease: >=20% increase in sum of target lesion diameters and sum must show absolute increase of >=5mm; smallest sum on study as reference. Based on Kaplan-Meier Estimates. (NCT01668784)
Timeframe: from randomization up to disease progression or death (approximately up to 105 Months)

Interventionmonths (Median)
Nivolumab4.21
Everolimus4.50

[back to top] [back to top]

Overall Survival (OS) by Programmed Death-Ligand 1 (PD-L1) Expression Level

Quantifiable PD-L1 expression=percent of tumor cell membrane staining in a minimum of 100 evaluable tumor cells per Dako PD-L1 IHC assay. If the PD-L1 staining could not be quantified it was classified as: indeterminate=tumor cell membrane staining hampered for reasons attributed to biology of tumor biopsy specimen and not due to improper sample preparation or handling; not evaluable=tumor biopsy specimen was not optimally collected or prepared. Not evaluable determined from H&E process before the tumor biopsy specimen was sent for evaluation or from H&E process during PD-L1 evaluation; baseline PD-L1 expression=if more than one tumor biopsy specimen was available, the most recently collected specimen with a quantifiable result. If all specimens for a given participant are either indeterminate or not evaluable, then the PD-L1 expression was considered indeterminate as long as at least one specimen is indeterminate. Otherwise, PD-L1 expression was considered not evaluable. (NCT01668784)
Timeframe: Randomization to date of death or date of last contact for patients without documentation of death, up to May 2015 (approximately 30 months)

,
Interventionmonths (Median)
Participant PD-L1 expression >=1%Participant PD-L1 expression <1%
Everolimus4.174.67
Nivolumab5.363.94

[back to top]

Investigator-assessed Time to Objective Response

Time to objective response is defined as the time from randomization to first response (complete response, CR or partial response, PR). CR=Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.; PR=At least a 30% decrease in the sum of diameters of target lesions, the baseline sum diameters used as reference. (NCT01668784)
Timeframe: Randomization to date of first response (approximately 105 months)

Interventionmonths (Median)
Nivolumab3.55
Everolimus3.71

[back to top]

Distant Recurrence-Free Survival (DRFS)

Time from date of registration to date of invasive distant disease recurrence, second invasive primary cancer (breast or not) or death due to any cause. Patients last known to be alive who have not experienced distant recurrence, or second primary cancer are censored at their last contact date. The results were presented as 5-year DRFS estimate. (NCT01674140)
Timeframe: 5 years after last accrual

Interventionpercentage of participants (Number)
Placebo75.7
Everolimus76.9

[back to top]

Invasive Disease-Free Survival (IDFS)

Time from date of registration to date of first invasive recurrence (local, regional or distant), second invasive primary cancer (breast or not), or death due to any cause. Patients last known to be alive who have not experienced recurrence or second primary cancer are censored at their last contact date. The results were presented as 5-year IDFS estimate. (NCT01674140)
Timeframe: Up to 5 years post registration

Interventionpercentage of participants (Number)
Placebo74.4
Everolimus74.9

[back to top]

Overall Survival (OS)

Time from date of registration to date of death due to any cause. Patients last known to be alive are censored at their last contact date. The results were presented as 5-year OS estimate. (NCT01674140)
Timeframe: 5 years after last accrual

Interventionpercentage of participants (Number)
Placebo85.8
Everolimus88.1

[back to top]

Toxicity Based on Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0, Assessed up to 10 Years.

Toxicity based on Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Only adverse events that are possibly, probably, or definitely related to study drug are reported. (NCT01674140)
Timeframe: Every 6 weeks while on protocol therapy. Adverse events (AEs) that occurred during follow up after protocol treatment were reported as late AEs, for every 6 months for the first 2 years and then yearly until 10 years after registration.

,
InterventionParticipants (Number)
Abdominal painAlanine aminotransferase increasedAllergic reactionAnemiaAnxietyAppendicitisAppendicitis perforatedArthralgiaAscitesAspartate aminotransferase increasedBack painBone painBreast infectionCardiac arrestCholecystitisCholesterol highColitisCoughDehydrationDepressionDiarrheaDizzinessDyspneaEdema faceEdema limbsEye disorders - Other, specifyEye infectionFatigueFeverGallbladder infectionGallbladder perforationGastrointestinal disorders - Other, specifyGeneral disorders and admin site conditions - OtherGeneralized muscle weaknessHeadacheHeart failureHip fractureHot flashesHyperglycemiaHyperhidrosisHyperkalemiaHypertensionHypertriglyceridemiaHypokalemiaHyponatremiaHypophosphatemiaHypoxiaImmune system disorders - Other, specifyInfections and infestations - Other, specifyInfusion related reactionInsomniaInvestigations - Other, specifyIrregular menstruationJoint infectionKidney infectionLeft ventricular systolic dysfunctionLipase increasedLung infectionLymphedemaLymphocyte count decreasedMenorrhagiaMucositis oralMuscle weakness lower limbMuscle weakness upper limbMusculoskeletal and connective tiss disorder - OtherMyalgiaNasal congestionNauseaNeck painNervous system disorders - Other, specifyNeuralgiaNeutrophil count decreasedObesityOtitis mediaPainPain in extremityPapulopustular rashParoxysmal atrial tachycardiaPeriorbital edemaPeripheral sensory neuropathyPlatelet count decreasedPleural effusionPneumonitisPortal vein thrombosisPostoperative hemorrhageProductive coughPruritusPsychiatric disorders - Other, specifyPulmonary edemaRadiation recall reaction (dermatologic)Rash acneiformRash maculo-papularRash pustularRespiratory failureSepsisSeromaSkin and subcutaneous tissue disorders - OtherSkin infectionSkin ulcerationSoft tissue infectionSore throatSuicidal ideationSuicide attemptThromboembolic eventTooth infectionUpper respiratory infectionUrinary tract infectionVascular access complicationVomitingWeight gainWeight lossWhite blood cell decreasedWound complicationWound dehiscenceWound infection
Everolimus74210012516016019123213271111231112114410332115355212151311122061360601012121012201001022427111201214207138221115112110220546
Placebo1100120101101100100030200006000000100210064000004000100012051201000011031011010110100011000001000300000300101202010

[back to top]

Incidence of Chronic Allograft Nephropathy (CAI) at 12 Months Post-transplant

Incidence of (biopsy-proven) chronic allograft nephropathy (CAI) [interstitial fibrosis and tubular atrophy, using standard Banff criteria] at 12 months post-transplant. (NCT01680861)
Timeframe: 1 year

Interventionparticipants (Number)
Tacrolimus/Everolimus3
Tacrolimus/EC-MPS3

[back to top]

BPAR (Biopsy-proven Acute Rejection) Incidence During the First 12 Months Post-transplant

BPAR (biopsy-proven acute rejection) incidence during the first 12 months post-transplant. Grading is determined using standard Banff criteria. (NCT01680861)
Timeframe: 1 year

Interventionparticipants (Number)
Tacrolimus/Everolimus1
Tacrolimus/EC-MPS3

[back to top]

Discontinuance of Any Study Medication (Tacrolimus, Everolimus, or EC-MPS)

(NCT01680861)
Timeframe: during the first 12 months post-transplant

Interventionparticipants (Number)
Tacrolimus/Everolimus0
Tacrolimus/EC-MPS1

[back to top]

eGFR (Calculated Glomerular Filtration Rate), i.e., Renal Function, at 1 Month Post-transplant.

using the abbreviated MDRD formula. (NCT01680861)
Timeframe: at 1 month post-transplant

Interventionml/min per 1.73 m2 (Mean)
Tacrolimus/Everolimus82.1
Tacrolimus/EC-MPS62.1

[back to top]

eGFR (Renal Function) at 6 Months Post-transplant

using the abbreviated MDRD formula. (NCT01680861)
Timeframe: at 6 months post-transplant

Interventionml/min per 1.73 m2 (Mean)
Tacrolimus/Everolimus66.7
Tacrolimus/EC-MPS63.7

[back to top]

eGFR (Renal Function) at Month 3 Post-transplant

Renal function as determined by the estimated glomerular filtration rate (eGFR) at 3 months post-transplant, using the abbreviated MDRD formula. (NCT01680861)
Timeframe: at 3 months post-transplant

Interventionml/min per 1.73 m^2 (Mean)
Tacrolimus/Everolimus75.7
Tacrolimus/EC-MPS65.6

[back to top]

Graft Loss (Return to Permanent Dialysis or Death)

(NCT01680861)
Timeframe: during the first 12 months post-transplant

Interventionparticipants (Number)
Tacrolimus/Everolimus0
Tacrolimus/EC-MPS0

[back to top]

First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Overall Health at the End of the First Stomatitis Episode

"The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The second item asked the participant to rate their overall health from 0 (worst possible) to 10 (perfect health). The overall health OSDQ score are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment.~Only participants who were randomized in the stomatitis sub-study were included in this analysis." (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
Dexamethasone: From 2 (Day 1) to 5 (end of first stomatitis)Dexamethasone: From 4 (Day 1) to 3 (end of first stomatitis)Dexamethasone: From 5 (Day 1) to 5 (end of first stomatitis)Dexamethasone: From 5 (Day 1) to 6 (end of first stomatitis)Dexamethasone: From 5 (Day 1) to 7 (end of first stomatitis)Dexamethasone: From 6 (Day 1) to 5 (end of first stomatitis)Dexamethasone: From 6 (Day 1) to 7 (end of first stomatitis)Dexamethasone: From 7 (Day 1) to 3 (end of first stomatitis)Dexamethasone: From 8 (Day 1) to 8 (end of first stomatitis)Dexamethasone: From 8 (Day 1) to 9 (end of first stomatitis)Dexamethasone: From 9 (Day 1) to 9 (end of first stomatitis)SoC: From 0 (Day 1) to 5 (end of first stomatitis)SoC: From 4 (Day 1) to 3 (end of first stomatitis)SoC: From 4 (Day 1) to 8 (end of first stomatitis)SoC: From 5 (Day 1) to 5 (end of first stomatitis)SoC: From 6 (Day 1) to 7 (end of first stomatitis)SoC: From 7 (Day 1) to 3 (end of first stomatitis)SoC: From 8 (Day 1) to 9 (end of first stomatitis)SoC: From 8 (Day 1) to 10 (end of first stomatitis)SoC: From 9 (Day 1) to 4 (end of first stomatitis)SoC: From 9 (Day 1) to 9 (end of first stomatitis)SoC: From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)1111111111111111111122

[back to top]

First-line Treatment: Progression-free Survival (PFS)

PFS in the first line setting is defined as the time from the date of enrollment to the date of first documented progression based on local radiology review or death due to any cause. If a participant did not progress or was not known to have died at the date of the analysis cut-off or start of another antineoplastic therapy, the PFS date was censored to the date of last adequate tumor assessment prior to cut-off date or start of antineoplastic therapy. The median PFS was estimated and presented along with 95% confidence intervals. The primary analysis of PFS for first line was performed 12 months after the last patient's recruitment. (NCT01698918)
Timeframe: From the date of enrollment to the date of first documented progression or deaths, assessed up to approximately 2.8 years

InterventionMonths (Median)
Everolimus+Letrozole (First-line Treatment)NA

[back to top]

First-line Treatment: Overall Response Rate (ORR)

"ORR in first line setting is defined as the percentage of participants while on first-line treatment with best overall response of complete response (CR) or partial response (PR) according to RECIST version 1.0 based on local review. Confidence intervals were calculated based on the Exact Clopper-Pearson method.~ORR while on first-line treatment was assessed up to 24 months after the last patient's recruitment.~CR: disappearance of all target lesions PR: at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters." (NCT01698918)
Timeframe: From the date of enrollment until discontinuation of first-line treatment, assessed up to approximately 3.8 years

InterventionPercentage of participants (Number)
Everolimus+Letrozole (First Line Treatment)45.0

[back to top]

First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness Limiting Swallowing, Drinking, Eating, Talking and Sleeping at the End of the First Stomatitis Episode

"The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The fourth item asked the participant to rate how much their mouth and throat soreness limited them in 1) swallowing, 2) drinking, 3) eating, 4) talking and 5) sleeping. For each activity, mouth and throat soreness scores ranged from 0 (not limited) to 4 (unable to do). Scores are presented as the shift from Day 1 of first stomatitis stomatitis value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first episode value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment.~Only participants who were randomized in the stomatitis sub-study were included in this analysis." (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
Swallowing (dexamethasone): From 0 (Day 1) to 0 (end of first stomatitis)Swallowing (dexamethasone): From 1 (Day 1) to 0 (end of first stomatitis)Swallowing (dexamethasone): From 1 (Day 1) to 1 (end of first stomatitis)Swallowing (dexamethasone): From 2 (Day 1) to 0 (end of first stomatitis)Swallowing (dexamethasone): From 2 (Day 1) to 2 (end of first stomatitis)Swallowing (SoC): From 0 (Day 1) to 0 (end of first stomatitis)Swallowing (SoC): From 1 (Day 1) to 2 (end of first stomatitis)Swallowing (SoC): From 2 (Day 1) to 0 (end of first stomatitis)Swallowing (SoC): From 2 (Day 1) to 1 (end of first stomatitis)Swallowing (SoC): From 3 (Day 1) to 0 (end of first stomatitis)Swallowing (SoC): From 3 (Day 1) to 1 (end of first stomatitis)Swallowing (SoC): From missing value (Day 1) to missing value (end of first stomatitis)Drinking (dexamethasone): From 0 (Day 1) to 0 (end of first stomatitis)Drinking (dexamethasone): From 1 (Day 1) to 0 (end of first stomatitis)Drinking (dexamethasone): From 1 (Day 1) to 1 (end of first stomatitis)Drinking (dexamethasone): From 2 (Day 1) to 0 (end of first stomatitis)Drinking (dexamethasone): From 2 (Day 1) to 2 (end of first stomatitis)Drinking (SoC): From 0 (Day 1) to 0 (end of first stomatitis)Drinking (SoC): From 0 (Day 1) to 1 (end of first stomatitis)Drinking (SoC): From 2 (Day 1) to 0 (end of first stomatitis)Drinking (SoC): From 2 (Day 1) to 1 (end of first stomatitis)Drinking (SoC): From 3 (Day 1) to 0 (end of first stomatitis)Drinking (SoC): From 3 (Day 1) to 1 (end of first stomatitis)Drinking (SoC): From missing value (Day 1) to missing value (end of first stomatitis)Eating (dexamethasone): From 0 (Day 1) to 0 (end of first stomatitis)Eating (dexamethasone): From 1 (Day 1) to 0 (end of first stomatitis)Eating (dexamethasone): From 1 (Day 1) to 1 (end of first stomatitis)Eating (dexamethasone): From 2 (Day 1) to 0 (end of first stomatitis)Eating (dexamethasone): From 2 (Day 1) to 1 (end of first stomatitis)Eating (dexamethasone): From 2 (Day 1) to 2 (end of first stomatitis)Eating (dexamethasone): From 3 (Day 1) to 1 (end of first stomatitis)Eating (SoC): From 1 (Day 1) to 0 (end of first stomatitis)Eating (SoC): From 1 (Day 1) to 1 (end of first stomatitis)Eating (SoC): From 2 (Day 1) to 0 (end of first stomatitis)Eating (SoC): From 3 (Day 1) to 0 (end of first stomatitis)Eating (SoC): From 3 (Day 1) to 1 (end of first stomatitis)Eating (SoC): From 3 (Day 1) to 2 (end of first stomatitis)Eating (SoC): From 4 (Day 1) to 1 (end of first stomatitis)Eating (SoC): From missing value (Day 1) to missing value (end of first stomatitis)Talking (dexamethasone): From 0 (Day 1) to 0 (end of first stomatitis)Talking (dexamethasone): From 1 (Day 1) to 0 (end of first stomatitis)Talking (dexamethasone): From 2 (Day 1) to 0 (end of first stomatitis)Talking (dexamethasone): From 2 (Day 1) to 1 (end of first stomatitis)Talking (dexamethasone): From 2 (Day 1) to 2 (end of first stomatitis)Talking (SoC): From 0 (Day 1) to 0 (end of first stomatitis)Talking (SoC): From 0 (Day 1) to 1 (end of first stomatitis)Talking (SoC): From 1 (Day 1) to 0 (end of first stomatitis)Talking (SoC): From 2 (Day 1) to 0 (end of first stomatitis)Talking (SoC): From 2 (Day 1) to 1 (end of first stomatitis)Talking (SoC): From 3 (Day 1) to 0 (end of first stomatitis)Talking (SoC): From 3 (Day 1) to 2 (end of first stomatitis)Talking (SoC): From missing value (Day 1) to missing value (end of first stomatitis)Sleeping (dexamethasone): From 0 (Day 1) to 0 (end of first stomatitis)Sleeping (dexamethasone): From 1 (Day 1) to 0 (end of first stomatitis)Sleeping (dexamethasone): From 2 (Day 1) to 0 (end of stomatitis)Sleeping (dexamethasone): From 2 (Day 1) to 1 (end of first stomatitis)Sleeping (dexamethasone): From 2 (Day 1) to 2 (end of first stomatitis)Sleeping (SoC): From 0 (Day 1) to 0 (end of first stomatitis)Sleeping (SoC): From 0 (Day 1) to 2 (end of first stomatitis)Sleeping (SoC): From 1 (Day 1) to 1 (end of first stomatitis)Sleeping (SoC): From 2 (Day 1) to 0 (end of first stomatitis)Sleeping (SoC): From 3 (Day 1) to 0 (end of first stomatitis)Sleeping (SoC): From 3 (Day 1) to 1 (end of first stomatitis)Sleeping (SoC): From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)6211151121126211132311123121211211123126121151111112711115112112

[back to top]

First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity Affecting Daily Activities at the End of the First Stomatitis Episode

"The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The sixth item asked the participant to rate their mouth pain severity affecting daily activities score from 0 (no effect on daily activities) to 10 (completely prevented from doing daily activities). The mouth pain severity affecting daily activities OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment.~Only participants who were randomized in the stomatitis sub-study were included in this analysis." (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
Dexamethasone: From 0 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 1 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 2 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 3 (Day 1) to 2 (end of first stomatitis)Dexamethasone: From 5 (Day 1) to 3 (end of first stomatitis)Dexamethasone: From 6 (Day 1) to 1 (end of first stomatitis)SoC: From 0 (Day 1) to 0 (end of first stomatitis)SoC: From 1 (Day 1) to 4 (end of first stomatitis)SoC: From 2 (Day 1) to 2 (end of first stomatitis)SoC: From 3 (Day 1) to 0 (end of first stomatitis)SoC: From 3 (Day 1) to 1 (end of first stomatitis)SoC: From 5 (Day 1) to 2 (end of first stomatitis)SoC: From 5 (Day 1) to 5 (end of first stomatitis)SoC: From 9 (Day 1) to 0 (end of first stomatitis)SoC: From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)512111411111112

[back to top]

First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity at the End of the First Stomatitis Episode

"The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The fifth item asked the participant to rate their mouth pain severity from 0 (no pain) to 10 (unbearable pain). The mouth pain severity OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment.~Only participants who were randomized in the stomatitis sub-study were included in this analysis." (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
Dexamethasone: From 0 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 1 (Day 1) to 2 (end of first stomatitis)Dexamethasone: From 2 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 3 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 3 (Day 1) to 1 (end of first stomatitis)Dexamethasone: From 3 (Day 1) to 2 (end of first stomatitis)Dexamethasone: From 5 (Day 1) to 2 (end of first stomatitis)Dexamethasone: From 5 (Day 1) to 3 (end of first stomatitis)Dexamethasone: From 7 (Day 1) to 1 (end of first stomatitis)SoC: From 1 (Day 1) to 0 (end of first stomatitis)SoC: From 1 (Day 1) to 1 (end of first stomatitis)SoC: From 4 (Day 1) to 1 (end of first stomatitis)SoC: From 5 (Day 1) to 5 (end of first stomatitis)SoC: From 7 (Day 1) to 0 (end of first stomatitis)SoC: From 8 (Day 1) to 2 (end of first stomatitis)SoC: From 8 (Day 1) to 4 (end of first stomatitis)SoC: From 8 (Day 1) to 6 (end of first stomatitis)SoC: From 9 (Day 1) to 0 (end of first stomatitis)SoC: From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)1122111111121112112

[back to top]

First-line Treatment: Duration of First Stomatitis Based on OSDQ

The duration of the first stomatitis episode was calculated using the start and end date recorded in the OSDQ. The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis. The first item of the questionnaire asked the participants the date when they experienced the first symptoms of stomatitis. Start date of the first occurrence of stomatitis is defined as the first date ever recorded for this item in the questionnaire. Stop date of the first stomatitis episode is defined as the last date the OSDQ was completed for this episode. Participants were censored if they died before resolution of stomatitis, received a new anticancer therapy, discontinued the study treatment with no resolution of the stomatitis or the stomatitis event was still on-going at the cut-off. PROs were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From start date of first stomatitis episode in first line until its resolution, assessed up to 3.8 years

InterventionWeeks (Median)
Everolimus+Letrozole (First-line Treatment)12.3

[back to top]

First-line Treatment (Stomatitis Sub-study): Duration of First Stomatitis Based on OSDQ

The duration of the first stomatitis was calculated using the start and end date reported in the OSDQ. The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis. The first item asked the participants the date when they experienced the first symptoms of stomatitis. Start date of the first stomatitis is defined as the first date ever recorded for this item in the questionnaire. Stop date of the first stomatitis is defined as the last date the OSDQ was completed for this episode. Participants were censored if they died before resolution of stomatitis, received a new anticancer therapy, discontinued the study treatment with no resolution of the stomatitis or the stomatitis was still on-going at the cut-off. PROs were assessed up to 24 months after last patient's recruitment. Only participants who were randomized in the stomatitis sub-study were included in this analysis. (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionWeeks (Median)
DexamethasoneStandard of Care
Everolimus+Letrozole (First-line Treatment)NA13.7

[back to top]

First-line Treatment (Stomatitis Sub-study): Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness at the End of the First Stomatitis Episode

"The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The third item asked the participant to rate their mouth and throat soreness from 0 (no soreness) to 4 (extreme soreness). The mouth and throat soreness OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment.~Only participants who were randomized in the stomatitis sub-study were included in this analysis." (NCT01698918)
Timeframe: From start date of stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
Dexamethasone: From 0 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 1 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 1 (Day 1) to 1 (end of first stomatitis)Dexamethasone: From 2 (Day 1) to 0 (end of first stomatitis)Dexamethasone: From 2 (Day 1) to 1 (end of first stomatitis)Dexamethasone: From 3 (Day 1) to 1 (end of first stomatitis)SoC: From 1 (Day 1) to 0 (end of first stomatitis)SoC: From 1 (Day 1) to 1 (end of first stomatitis)SoC: From 2 (Day 1) to 1 (end of first stomatitis)SoC: From 3 (Day 1) to 0 (end of first stomatitis)SoC: From 3 (Day 1) to 1 (end of first stomatitisSoC: From 3 (Day 1) to 2 (end of first stomatitisSoC: From 4 (Day 1) to 1 (end of first stomatitisSoC: From 4 (Day 1) to 2 (end of first stomatitisSoC: From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)132131111213112

[back to top]

First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness Limiting Swallowing, Drinking, Eating, Talking and Sleeping at the End of the First Stomatitis Episode

The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The fourth item asked the participant to rate how much their mouth and throat soreness limited them in 1) swallowing, 2) drinking, 3) eating, 4) talking and 5) sleeping. For each activity, mouth and throat soreness scores ranged from 0 (not limited) to 4 (unable to do). Scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
Swallowing: From 0 (Day 1) to 0 (end of first stomatitis)Swallowing: From 0 (Day 1) to 1 (end of first stomatitis)Swallowing: From 0 (Day 1) to 2 (end of first stomatitis)Swallowing: From 1 (Day 1) to 0 (end of first stomatitis)Swallowing: From 1 (Day 1) to 1 (end of first stomatitis)Swallowing: From 1 (Day 1) to 2 (end of first stomatitis)Swallowing: From 1 (Day 1) to 3 (end of first stomatitis)Swallowing: From 2 (Day 1) to 0 (end of first stomatitis)Swallowing: From 2 (Day 1) to 1 (end of first stomatitis)Swallowing: From 2 (Day 1) to 2 (end of first stomatitis)Swallowing: From 3 (Day 1) to 0 (end of first stomatitis)Swallowing: From 3 (Day 1) to 1 (end of first stomatitis)Swallowing: From 3 (Day 1) to 3 (end of first stomatitis)Swallowing: From missing value (Day 1) to missing value (end of first stomatitis)Drinking: From 0 (Day 1) to 0 (end of first stomatitis)Drinking: From 0 (Day 1) to 1 (end of first stomatitis)Drinking: From 0 (Day 1) to 2 (end of first stomatitis)Drinking: From 1 (Day 1) to 0 (end of first stomatitis)Drinking: From 1 (Day 1) to 1 (end of first stomatitis)Drinking: From 1 (Day 1) to 2 (end of first stomatitis)Drinking: From 2 (Day 1) to 0 (end of first stomatitis)Drinking: From 2 (Day 1) to 1 (end of first stomatitis)Drinking: From 2 (Day 1) to 2 (end of first stomatitis)Drinking: From 2 (Day 1) to 3 (end of first stomatitis)Drinking: From 3 (Day 1) to 0 (end of first stomatitis)Drinking: From 3 (Day 1) to 1 (end of first stomatitis)Drinking: From 3 (Day 1) to 3 (end of first stomatitis)Drinking: From missing value (Day 1) to missing value (end of first stomatitis)Eating: From 0 (Day 1) to 0 (end of first stomatitis)Eating: From 0 (Day 1) to 1 (end of first stomatitis)Eating: From 0 (Day 1) to 2 (end of first stomatitis)Eating: From 1 (Day 1) to 0 (end of first stomatitis)Eating: From 1 (Day 1) to 1 (end of first stomatitis)Eating: From 1 (Day 1) to 2 (end of first stomatitis)Eating: From 2 (Day 1) to 0 (end of first stomatitis)Eating: From 2 (Day 1) to 1 (end of first stomatitis)Eating: From 2 (Day 1) to 2 (end of first stomatitis)Eating: From 2 (Day 1) to 3 (end of first stomatitis)Eating: From 3 (Day 1) to 0 (end of first stomatitis)Eating: From 3 (Day 1) to 1 (end of first stomatitis)Eating: From 3 (Day 1) to 2 (end of first stomatitis)Eating: From 3 (Day 1) to 3 (end of first stomatitis)Eating: From 4 (Day 1) to 1 (end of first stomatitis)Eating: From missing value (Day 1) to missing value (end of first stomatitis)Talking: From 0 (Day 1) to 0 (end of first stomatitis)Talking: From 0 (Day 1) to 1 (end of first stomatitis)Talking: From 1 (Day 1) to 0 (end of first stomatitis)Talking: From 1 (Day 1) to 1 (end of first stomatitis)Talking: From 2 (Day 1) to 0 (end of first stomatitis)Talking: From 2 (Day 1) to 1 (end of first stomatitis)Talking: From 2 (Day 1) to 2 (end of first stomatitis)Talking: From 2 (Day 1) to 3 (end of first stomatitis)Talking: From 3 (Day 1) to 0 (end of first stomatitis)Talking: From 3 (Day 1) to 2 (end of first stomatitis)Talking: From 3 (Day 1) to 3 (end of first stomatitis)Talking: From missing value (Day 1) to missing value (end of first stomatitis)Sleeping: From 0 (Day 1) to 0 (end of first stomatitis)Sleeping: From 0 (Day 1) to 1 (end of first stomatitis)Sleeping: From 0 (Day 1) to 2 (end of first stomatitis)Sleeping: From 1 (Day 1) to 0 (end of first stomatitis)Sleeping: From 1 (Day 1) to 1 (end of first stomatitis)Sleeping: From 1 (Day 1) to 2 (end of first stomatitis)Sleeping: From 2 (Day 1) to 0 (end of first stomatitis)Sleeping: From 2 (Day 1) to 1 (end of first stomatitis)Sleeping: From 2 (Day 1) to 2 (end of first stomatitis)Sleeping: From 3 (Day 1) to 0 (end of first stomatitis)Sleeping: From 3 (Day 1) to 1 (end of first stomatitis)Sleeping: From 3 (Day 1) to 2 (end of first stomatitis)Sleeping: From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First Line Treatment)4031763164282274841105143326119172114112834276341745512252314139484182243611111

[back to top]

First-line Treatment: Clinical Benefit Rate (CBR)

"CBR in first line is defined as the percentage of participants while on first-line treatment with best overall response of CR, PR or stable disease (SD) with a duration of 24 weeks or longer, according to RECIST version 1.0 based on local review. Confidence intervals (CI) were calculated based on the Exact Clopper-Pearson method.~CBR while on first-line treatment was assessed up to 24 months after the last patient's recruitment.~CR: disappearance of all target lesions PR: at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters.~SD: Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for progressive disease" (NCT01698918)
Timeframe: From the date of enrollment until first-line treatment discontinuation, assessed up to approximately 3.8 years

InterventionPercentage of participants (Number)
Everolimus+Letrozole (First-line Treatment)74.3

[back to top]

All Collected Deaths

"On-treatment deaths in first line were collected from first dose of first-line treatment until the date of last administration of the first-line study treatment plus 28 days for participants not entering second-line or the minimum between the date of last administration of the first-line study treatment plus 28 days and the date of first administration of second-line study treatment minus one day for participants entering second line, up to 7.3 years. On-treatment deaths in second line were collected from first dose of second-line treatment to 28 days after the last administration of second-line treatment, up to 5.4 years.~Post-treatment survival follow-up deaths were collected from day 29 after last dose of study treatment to end of study, up to 7.3 years.~All deaths refer to the sum of on-treatment deaths and post-treatment deaths." (NCT01698918)
Timeframe: On-treatment deaths in first line: Up to 7.3 years. On-treatment deaths in second line: Up to 5.4 years. Post-treatment and all deaths: Up to 7.3 years

InterventionParticipants (Number)
On -treatment deaths in first lineOn-treatment deaths in second linePost-treatment survival follow-up deathsAll deaths
Everolimus + Letrozole/Exemestane (First-line and Second-line Treatment)1124962

[back to top]

Second-line Treatment: Progression-free Survival (PFS)

"PFS in the second line setting is defined as the time interval between the start of the second-line treatment and documented disease progression based on local radiology review or death due to any cause reported during or after second-line treatment period. If a participant did not progress or was not known to have died at the date of the analysis cut-off or start of another antineoplastic therapy, the PFS date was censored to the date of last adequate tumor assessment prior to cut-off date or start of antineoplastic therapy. The median PFS was estimated and presented along with 95% confidence intervals.~PFS while on second-line treatment was assessed up to 24 months after the last patient's recruitment." (NCT01698918)
Timeframe: From the start of the second-line treatment until the date of first documented progression or death, assessed up to approximately 2.4 years

InterventionMonths (Median)
Everolimus+Exemestane (Second Line Treatment)3.7

[back to top]

First-line Treatment (Stomatitis Sub-study): Time to First Stomatitis Episode as Assessed by the OSDQ

"The time to first occurrence of stomatitis based on OSDQ is defined as time from first study treatment administration in the first line to start date of the first stomatitis episode recorded in the OSDQ. The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The first item asked the participants when they experienced the first symptoms of stomatitis. Start date of the first occurrence of stomatitis is defined as the first date ever recorded for this item in the questionnaire. PROs were assessed up to 24 months after last patient's recruitment.~Only participants who were randomized in the stomatitis sub-study were included in this analysis." (NCT01698918)
Timeframe: From first study treatment administration in the first line until first stomatitis episode, assessed up to approximately 3.8 years

InterventionWeeks (Median)
DexamethasoneStandard of care
Everolimus+ Letrozole (First-line Treatment)1.42.3

[back to top]

First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth and Throat Soreness at the End of the First Stomatitis Episode

The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The third item asked the participant to rate their mouth and throat soreness from 0 (no soreness) to 4 (extreme soreness). The mouth and throat soreness OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
From 0 (Day 1) to 0 (end of first stomatitis)From 1 (Day 1) to 0 (end of first stomatitis)From 1 (Day 1) to 1 (end of first stomatitis)From 1 (Day 1) to 2 (end of first stomatitis)From 2 (Day 1) to 0 (end of first stomatitis)From 2 (Day 1) to 1 (end of first stomatitis)From 2 (Day 1) to 2 (end of first stomatitis)From 2 (Day 1) to 3 (end of first stomatitis)From 3 (Day 1) to 0 (end of first stomatitis)From 3 (Day 1) to 1 (end of first stomatitis)From 3 (Day 1) to 2 (end of first stomatitis)From 3 (Day 1) to 3 (end of first stomatitis)From 3 (Day 1) to 4 (end of first stomatitis)From 4 (Day 1) to 1 (end of first stomatitis)From 4 (Day 1) to 2 (end of first stomatitis)From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)3221231374194511214

[back to top]

First-line Treatment: Time to First Stomatitis Episode as Assessed by the Oral Stomatitis Daily Questionnaire (OSDQ)

The time to first occurrence of stomatitis based on OSDQ is defined as time from first-line treatment administration to start date of the stomatitis episode recorded in the OSDQ in the first line. The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The first item asked the participants when they experienced the first symptoms of stomatitis. Start date of the first occurrence of stomatitis is defined as the first date ever recorded for this item in the questionnaire. Patient reported outcomes (PROs) were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From first-line treatment administration until first stomatitis episode in the first line, assessed up to approximately 3.8 years

InterventionWeeks (Median)
Everolimus+Letrozole (First-line Treatment)1.7

[back to top]

First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity Affecting Daily Activities at the End of the First Stomatitis Episode

The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The sixth item asked the participant to rate their mouth pain severity affecting daily activities score from 0 (no effect on daily activities) to 10 (completely prevented from doing daily activities). The mouth pain severity affecting daily activities OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
From 0 (Day 1) to 0 (end of first stomatitis)From 0 (Day 1) to 1 (end of first stomatitis)From 0 (Day 1) to 5 (end of first stomatitis)From 1 (Day 1) to 0 (end of first stomatitis)From 1 (Day 1) to 1 (end of first stomatitis)From 1 (Day 1) to 4 (end of first stomatitis)From 2 (Day 1) to 0 (end of first stomatitis)From 2 (Day 1) to 1 (end of first stomatitis)From 2 (Day 1) to 2 (end of first stomatitis)From 2 (Day 1) to 3 (end of first stomatitis)From 2 (Day 1) to 4 (end of first stomatitis)From 3 (Day 1) to 0 (end of first stomatitis)From 3 (Day 1) to 1 (end of first stomatitis)From 3 (Day 1) to 2 (end of first stomatitis)From 3 (Day 1) to 3 (end of first stomatitis)From 3 (Day 1) to 4 (end of first stomatitis)From 4 (Day 1) to 0 (end of first stomatitis)From 5 (Day 1) to 0 (end of first stomatitis)From 5 (Day 1) to 1 (end of first stomatitis)From 5 (Day 1) to 2 (end of first stomatitis)From 5 (Day 1) to 3 (end of first stomatitis)From 5 (Day 1) to 5 (end of first stomatitis)From 5 (Day 1) to 6 (end of first stomatitis)From 6 (Day 1) to 1 (end of first stomatitis)From 6 (Day 1) to 5 (end of first stomatitis)From 6 (Day 1) to 8 (end of first stomatitis)From 7 (Day 1) to 0 (end of first stomatitis)From 8 (Day 1) to 0 (end of first stomatitis)From 8 (Day 1) to 8 (end of first stomatitis)From 8 (Day 1) to 10 (end of first stomatitis)From 9 (Day 1) to 0 (end of first stomatitis)From 9 (Day 1) to 8 (end of first stomatitis)From 10 (Day 1) to 10 (end of first stomatitis)From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)33117426131133111221111111111111114

[back to top]

First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Mouth Pain Severity at the End of the First Stomatitis Episode

The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily until the resolution of the stomatitis episode. The fifth item asked the participant to rate their mouth pain severity from 0 (no pain) to 10 (unbearable pain). The mouth pain severity OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
From 0 (Day 1) to 0 (end of first stomatitis)From 1 (Day 1) to 0 (end of first stomatitis)From 1 (Day 1) to 1 (end of first stomatitis)From 1 (Day 1) to 2 (end of first stomatitis)From 1 (Day 1) to 3 (end of first stomatitis)From 1 (Day 1) to 5 (end of first stomatitis)From 2 (Day 1) to 0 (end of first stomatitis)From 2 (Day 1) to 3 (end of first stomatitis)From 3 (Day 1) to 0 (end of first stomatitis)From 3 (Day 1) to 1 (end of first stomatitis)From 3 (Day 1) to 2 (end of first stomatitis)From 3 (Day 1) to 3 (end of first stomatitis)From 3 (Day 1) to 4 (end of first stomatitis)From 4 (Day 1) to 0 (end of first stomatitis)From 4 (Day 1) to 1 (end of first stomatitis)From 4 (Day 1) to 4 (end of first stomatitis)From 4 (Day 1) to 5 (end of first stomatitis)From 5 (Day 1) to 0 (end of first stomatitis)From 5 (Day 1) to 1 (end of first stomatitis)From 5 (Day 1) to 2 (end of first stomatitis)From 5 (Day 1) to 3 (end of first stomatitis)From 5 (Day 1) to 4 (end of first stomatitis)From 5 (Day 1) to 5 (end of first stomatitis)From 5 (Day 1) to 6 (end of first stomatitis)From 5 (Day 1) to 7 (end of first stomatitis)From 5 (Day 1) to 10 (end of first stomatitis)From 6 (Day 1) to 0 (end of first stomatitis)From 6 (Day 1) to 1 (end of first stomatitis)From 6 (Day 1) to 2 (end of first stomatitis)From 6 (Day 1) to 5 (end of first stomatitis)From 6 (Day 1) to 6 (end of first stomatitis)From 6 (Day 1) to 8 (end of first stomatitis)From 7 (Day 1) to 0 (end of first stomatitis)From 7 (Day 1) to 1 (end of first stomatitis)From 7 (Day 1) to 2 (end of first stomatitis)From 7 (Day 1) to 7 (end of first stomatitis)From 8 (Day 1) to 0 (end of first stomatitis)From 8 (Day 1) to 2 (end of first stomatitis)From 8 (Day 1) to 4 (end of first stomatitis)From 8 (Day 1) to 5 (end of first stomatitis)From 8 (Day 1) to 6 (end of first stomatitis)From 8 (Day 1) to 9 (end of first stomatitis)From 9 (Day 1) to 0 (end of first stomatitis)From 9 (Day 1) to 9 (end of first stomatitis)From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)5106111617121242113111111112111112211312111214

[back to top]

Second-line Treatment: Overall Response Rate (ORR)

"ORR in second line is defined as the percentage of participants receiving second-line study treatment with best overall response of complete response (CR) or partial response (PR) according to RECIST version 1.0 based on local review. Confidence intervals (CI) were calculated based on the Exact Clopper-Pearson method.~ORR while on second-line treatment was assessed up to 24 months after the last patient's recruitment.~CR: disappearance of all target lesions PR: at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters" (NCT01698918)
Timeframe: From the start of the second-line treatment up to approximately 2.4 years

InterventionPercentage of participants (Number)
Everolimus+Exemestane (Second-line Treatment)6.0

[back to top]

First-line Treatment: Number of Participants With Shift of Response in OSDQ Score on Overall Health at the End of the First Stomatitis Episode

The OSDQ is a patient self-administered 6-item questionnaire with a 24-hour recall period. Participants completed the questionnaire daily up to the resolution of the stomatitis episode. The second item asked the participant to rate their overall health from 0 (worst possible) to 10 (perfect health). The overall health OSDQ scores are presented as the shift from Day 1 of first stomatitis episode value to the value at the end of the first episode of stomatitis. Day 1 is defined as the first OSDQ questionnaire recorded. End of first stomatitis value is defined as the last OSDQ questionnaire of the first episode of stomatitis. PROs were assessed up to 24 months after last patient's recruitment. (NCT01698918)
Timeframe: From start date of first stomatitis episode until its resolution, assessed up to 3.8 years

InterventionParticipants (Count of Participants)
From 0 (Day 1) to 0 (end of first stomatitis)From 0 (Day 1) to 1 (end of first stomatitis)From 0 (Day 1) to 5 (end of first stomatitis)From 1 (Day 1) to 1 (end of first stomatitis)From 1 (Day 1) to 2 (end of first stomatitis)From 2 (Day 1) to 5 (end of first stomatitis)From 2 (Day 1) to 8 (end of first stomatitis)From 3 (Day 1) to 0 (end of first stomatitis)From 3 (Day 1) to 3 (end of first stomatitis)From 3 (Day 1) to 5 (end of first stomatitis)From 3 (Day 1) to 10 (end of first stomatitis)From 4 (Day 1) to 3 (end of first stomatitis)From 4 (Day 1) to 5 (end of first stomatitis)From 4 (Day 1) to 7 (end of first stomatitis)From 4 (Day 1) to 8 (end of first stomatitis)From 5 (Day 1) to 0 (end of first stomatitis)From 5 (Day 1) to 3 (end of first stomatitis)From 5 (Day 1) to 4 (end of first stomatitis)From 5 (Day 1) to 5 (end of first stomatitis)From 5 (Day 1) to 6 (end of first stomatitis)From 5 (Day 1) to 7 (end of first stomatitis)From 5 (Day 1) to 9 (end of first stomatitis)From 5 (Day 1) to 10 (end of first stomatitis)From 6 (Day 1) to 5 (end of first stomatitis)From 6 (Day 1) to 6 (end of first stomatitis)From 6 (Day 1) to 7 (end of first stomatitis)From 6 (Day 1) to 9 (end of first stomatitis)From 7 (Day 1) to 3 (end of first stomatitis)From 7 (Day 1) to 4 (end of first stomatitis)From 7 (Day 1) to 5 (end of first stomatitis)From 7 (Day 1) to 7 (end of first stomatitis)From 7 (Day 1) to 9 (end of first stomatitis)From 8 (Day 1) to 1 (end of first stomatitis)From 8 (Day 1) to 7 (end of first stomatitis)From 8 (Day 1) to 8 (end of first stomatitis)From 8 (Day 1) to 9 (end of first stomatitis)From 8 (Day 1) to 10 (end of first stomatitis)From 9 (Day 1) to 0 (end of first stomatitis)From 9 (Day 1) to 4 (end of first stomatitis)From 9 (Day 1) to 8 (end of first stomatitis)From 9 (Day 1) to 9 (end of first stomatitis)From 10 (Day 1) to 3 (end of first stomatitis)From 10 (Day 1) to 9 (end of first stomatitis)From missing value (Day 1) to missing value (end of first stomatitis)
Everolimus+Letrozole (First-line Treatment)21111121322211213174312232221131114521218113

[back to top]

Second-line Treatment: Clinical Benefit Rate (CBR)

"CBR in second line is defined as the percentage of participants receiving second-line study treatment with best overall response of CR, PR or stable disease (SD) with a duration of 24 weeks or longer, according to RECIST version 1.0 based on local review. Confidence intervals (CI) were calculated based on the Exact Clopper-Pearson method.~CBR while on second-line treatment was assessed up to 24 months after the last patient's recruitment.~CR: disappearance of all target lesions PR: at least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters.~SD: Neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for progressive disease" (NCT01698918)
Timeframe: From the start of the second-line treatment up to approximately 2.4 years

InterventionPercentage of participants (Number)
Everolimus+Exemestane (Second-line Treatment)28.0

[back to top]

Number of Participants With Clinical Benfit During Extension Phase

Number of participants with clinical benefit as judged by the investigator during the extension phase. The extension phase for up to 3 years for participants who were continuing to benefit from study treatment following the end of the core study phase (24 months after last patient's recruitment) was added in the amendment 5 (dated 14-Feb-2017). Results are presented by line of treatment (NCT01698918)
Timeframe: From the end of core phase (upon approval of amendment 5) up to approximately 3 years

,
InterventionParticipants (Count of Participants)
Extension Week 1Extension Week 12Extension Week 24Extension Week 36Extension Week 48Extension Week 60Extension Week 72Extension Week 84Extension Week 96Extension Week 108Extension Week 120Extension Week 132Extension Week 144Extension End of treatment
Everolimus+Exemestane (Second-line Treatment)65433333332111
Everolimus+Letrozole (First-line Treatment)343028232018161312121110811

[back to top]

Overall Survival (OS)

"OS following first-line treatment with everolimus + letrozole is defined as the time from the date of receiving first line study treatment to date of death due to any cause (including first-line and second-line treatment periods). If a participant was not known to have died, survival was censored at the date of last contact.~OS following first-line treatment was assessed up to 24 months after last patient's recruitment." (NCT01698918)
Timeframe: From the date of enrollment to date of death, assessed up to approximately 3.8 years

InterventionMonths (Median)
Everolimus+Letrozole/Exemestane (First-line Treatment and Second-line Treatment)NA

[back to top]

Seizure Free Rates by Time Window

Percentage of seizure-free participants for each 12-week time window. (NCT01713946)
Timeframe: Weeks 18, 30, 42, 54, 66, 78, 90 & 102

InterventionPercentage of seizure-free participants (Number)
Week 18Week 30Week 42Week 54Week 66Week 78Week 90Week 102
Everolimus Long Term Evaluation (LTE)3.986.878.448.7010.9913.4914.8615.18

[back to top]

Long Term Evaluation: Relationship Between Seizure Frequency and Time-normalized Everolimus Concentration at Trough (Cmin,TN) - Repeated Measures Analysis

A repeated measures analysis considering fixed 2-week intervals and including the level of exposure (time-normalized Cmin values), the time on-treatment and the seizure frequency at baseline quantified the estimated percentage change over 2 weeks in seizure frequency associated with a double exposure to everolimus, 15 days more on treatment and half the seizure frequency at baseline. A positive percentage change means a reduction in seizure frequency whereas a negative percentage change means an increase in seizure frequency. (NCT01713946)
Timeframe: During everolimus treatment from start of everolimus up to the end of the extension phase, an average of 1.7 year

Intervention% change over 2-wk in seizures freq. (Mean)
By doubling the time-normalized Cmin (log scale)By reporting half the seizure freq. at baselineBy adding 12 weeks on treatmen
Everolimus Long Term Evaluation (LTE)8.9348.826.42

[back to top]

Core Phase: Median Percentage Change From Baseline in Seizure Frequency by Time Normalized Minimum Concentration

Percentage change from baseline in average weekly seizure frequency during the maintenance period of the Core phase is calculated as follow: (SFcfb) = 100 × (SFB - SFM) ÷ SFB where SFB is the average weekly seizure frequency in the Baseline phase and SFM is the average weekly seizure frequency in the maintenance period of the Core phase. A positive percentage change from baseline (SFcfb) means a reduction in seizure frequency whereas a negative percentage change from baseline (SFcfb) means an increase in seizure frequency. (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionPercentage change (Median)
<3 ng/mL20.55
3-7 ng/mL35.56
>7-<9 ng/mL39.72
9-15 ng/mL47.69
>15 ng/mL61.56

[back to top]

Core Phase: Food & Drug Administration (FDA): Percentage Change From Baseline in Partial Onset-seizure Frequency

"Comparison of median percent change from baseline in weekly seizure frequency in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm during maintenance period of the core phase. Percentage change from baseline in average weekly seizure frequency during the maintenance period of the Core phase (SFcfb) = 100 × (SFB - SFM) ÷ SFB where:~SFB is the average weekly seizure frequency in the Baseline phase SFM is the average weekly seizure frequency in the maintenance period of the Core phase A positive percentage change from baseline (SFcfb) means a reduction in seizure frequency whereas a negative percentage change from baseline (SFcfb) means an increase in seizure frequency." (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionPercentage change from baseline (Median)
Everolimus LT Target of 3 to 7 ng/mL29.29
Everolimus HT Target of 9 to 15 ng/mL39.55
Placebo14.86

[back to top]

Core Phase: Percentage of Patients With at Least a 25% Reduction in Seizure Frequency

Comparison of percentage of patients with at least ≥ 25% reduction in seizure frequency in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm during maintenance period of the core phase. At least 25% reduction from baseline in partial-onset seizure frequency during maintenance period of the core phase. (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionPercentage of participants (Number)
Everolimus LT Target of 3 to 7 ng/mL52.1
Everolimus HT Target of 9 to 15 ng/mL70.0
Placebo37.8

[back to top]

Core Phase: Impact of Everolimus on Anti-epileptic Drugs (AEDs) Concentrations

Impact of everolimus on AED concentrations at trough. Pre-dose plasma samples to measure AED concentrations were measured at at Visits 1 (Screening), 2 (Baseline), 3, and 5. Effects of everolimus on the exposure of antiepileptic drugs was assessed by comparing the anti-epileptic drug concentrations at Visits 1 and 2 (AEDs alone) and at Visits 3 and 5 (AEDs plus everolimus). (NCT01713946)
Timeframe: Baseline, Weeks 1 & 3

Interventionng/mL (Geometric Mean)
Valporic Acid0.962
Carbamazepine1.108
Clobazam1.093
N-desmethylclobazam1.071
Topiramate0.983
TRI4771.086
TRI4761.194
Clonazepam1.065
Zonisamide1.028
Phenobarbital0.957
Phenytoin1.020

[back to top]

Percentage of Seizure-free Patients During the Maintenance Period of the Core Phase

Comparison of seizure freedom (100% reduction in seizure frequency) in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm during maintenance period of the core phase. Seizure free means a 100% reduction from baseline in partial-onset seizure frequency during maintenance period of the core phase. (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionPercentage of seizure-free participants (Number)
Everolimus LT Target of 3 to 7 ng/mL5.1
Everolimus HT Target of 9 to 15 ng/mL3.8
Placebo0.8

[back to top]

Core Phase: Change From Baseline in the Overall Vineland-II Adaptive Behavior Composite (ABC) Score

Comparison of adaptive functioning using the VABS-II composite score in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm. The Vineland II assesses an individual's development of personal independence & social responsibility. The questionnaire contains 433 items which assess 15 subdomains organized into the five domains of Communication, Daily Living Skills, Socialization, Motor Skills and Maladaptive Behavior. The overall Adaptive Behavior Composite (ABC) score is obtained by summing the standard scores of the first four domain scores for patients aged less than 7 years, or the first 3 domain scores for patients aged 7 or older (the Maladaptive Behavior domain is optional). The ABC standard score ranges from 20 to 160 with a mean of 100 and a standard deviation of 15. Higher scores correspond to improved adaptive level. Note that 2 questionnaires with ABC scores<20 (data issues) were included in this analysis. (NCT01713946)
Timeframe: Baseline, 18 weeks

,,
Interventionscores on a scale (Median)
BaselineEnd of Core PhaseChange from Baseline
Everolimus HT Target of 9 to 15 ng/mL56.5055.000.00
Everolimus LT Target of 3 to 7 ng/mL58.0054.00-1.00
Placebo55.0055.000.00

[back to top]

Long Term Evaluation: Incidence of Suicide Attempt, Suicidal Ideation or Behavior During Core Phase Per Columbia Suicide Severity Rating Scale (C-SSRS) Outcomes

The C-SSRS was completed at each visit. The table below presents the number of patients who reported at least one completed suicide, one suicide attempt, one preparatory action toward imminent suicidal behavior, one suicidal ideation and one self-injurious behavior without suicidal intent at any time point after starting everolimus. (NCT01713946)
Timeframe: During everolimus treatment from start of everolimus up to permanent discontinuation of everolimus, an average of 2.3 years

InterventionParticipants (Number)
Completed suicideSuicidal attemptPrep. actions toward imminent suicidal behaviorSuicidal ideationSelf-injurious behavior without suicide intent
Everolimus Long Term Evaluation (LTE)01271

[back to top]

Core Phase: Response Rate in Seizure Frequency by Time Normalized Minimum Concentration

Comparison of response rate in seizure frequency for 5 categories of time-normalized minimum concentration (Cmin, TN) (< 3 ng/mL; 3-7 ng/mL; >7-<9 ng/mL; 9-15 ng/mL; >15 ng/mL). Response rate is the percentage of patients with ≥ 50% reduction from baseline in average weekly partial-onset seizure frequency during the maintenance period of the Core phase. (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionPercentage of responders (Median)
<3 ng/mL14.3
3-7 ng/mL29.9
>7-<9 ng/mL44.2
9-15 ng/mL50.0
>15 ng/mL50.0

[back to top]

Core Phase: European Medicine Agency (EMA): Seizure Frequency Response Rate

Comparison of response rates in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm. Response means at least a 50% reduction from baseline in partial-onset seizure frequency during the maintenance period of the core phase. (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionPercentage of responders (Number)
Everolimus LT Target of 3 to 7 ng/mL28.2
Everolimus HT Target of 9 to 15 ng/mL40.0
Placebo15.1

[back to top]

Core Phase: Changes From Baseline in Number of Seizure-free Days

Comparison of seizure-free days relative to baseline in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm during maintenance period of the core phase (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

InterventionNumber of seizure-free days -per 28 days (Median)
Everolimus LT Target of 3 to 7 ng/mL2.00
Everolimus HT Target of 9 to 15 ng/mL4.01
Placebo0.47

[back to top]

Core Phase: Change From Baseline in the QOLIE-31-P Overall Quality-of-life Score for Patients Aged >=18 Years

Comparison of quality of life (from 3 age specific questionnaires) in the everolimus low-trough treatment arm (3-7 ng/mL), hightrough treatment arm (9-15 ng/mL) and placebo arm at the end of the core phase. The Quality of Life in Epilepsy Inventory-31-Problems (QOLIE-31-P) is a survey of health-related quality of life for adults with epilepsy. The QOLIE-31-P is completed by the patient. It contains 39 items, of which a total of 30 are used to make up 7 different subscales. Scores range from 0-100, with higher scores indicating a greater level of functioning and QoL. The overall quality of life score is obtained by summing a linear combination of the 7 subscale scores, where each subscale is multiplied by a relative weight that is obtained from the patient's answer to 7 items of this questionnaire. (NCT01713946)
Timeframe: Baseline, Week 18

,,
Interventionscores on a scale (Median)
BaselineEnd of Core PhaseChange from Baseline
Everolimus HT Target of 9 to 15 ng/mL43.237.10.4
Everolimus LT Target of 3 to 7 ng/mL39.548.6-0.5
Placebo43.654.85.3

[back to top]

Long Term Evaluation: Effect of Everolimus Over Time in the Overall Wechsler Nonverbal Composite Score

The brief version of the WNV consists of a 2-subtest battery: only Matrices and Recognition subtests for patients under 8, and Matrices and Spatial Span subtests for patients aged 8 to 21. Based on the raw scores obtained from the subtests, standardized z-scores were calculated for each subtest using the following formula: Zscore = (X - b)/Sb where X is the raw score of the subtest, b and Sb represent the mean and standard deviation respectively of the subtest score recorded at baseline for the study population. The composite WNV score was computed by summing up the Z-scores of the 3 subtests of the WNV (i.e. matrices, recognition, and coding for patients aged <8 years and matrices, spatial span, and coding for patients aged 8 to 21 years). The composite WNV score has no range (NCT01713946)
Timeframe: Baseline, Weeks 18, 42, 66 and 90

Interventionscores on a scale (Median)
BaselineWeek 18Week 42Week 66Week 90
Everolimus Long Term Evaluation (LTE)-0.53-0.44-0.360.13-0.06

[back to top]

Long Term Evaluation: Effect of Everolimus Over Time in the Overall Vineland-II Adaptive Behavior Composite (ABC) Score

Comparison of adaptive functioning using the VABS-II composite score in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm. The Vineland II assesses an individual's development of personal independence & social responsibility. The questionnaire contains 433 items which assess 15 subdomains organized into the five domains of Communication, Daily Living Skills, Socialization, Motor Skills and Maladaptive Behavior. The overall Adaptive Behavior Composite (ABC) score is obtained by summing the standard scores of the first four domain scores for patients aged less than 7 years, or the first 3 domain scores for patients aged 7 or older (the Maladaptive Behavior domain is optional). The ABC standard score ranges from 20 to 160 with a mean of 100 and a standard deviation of 15. Higher scores correspond to improved adaptive level. Note that 2 questionnaires with ABC scores<20 (data issues) were included in this analysis. (NCT01713946)
Timeframe: Baseline, Weeks 18, 42, 66 and 90

Interventionscores on a scale (Median)
BaselineWeek 18Week 42Week 66Week 90
Everolimus Long Term Evaluation (LTE)56.0053.5055.5057.0049.50

[back to top]

Core Phase: Probability That a Patient Remains On-treatment up to a Specified Time Point

"Comparison of time to treatment discontinuation in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm during the core phase. Treatment duration is defined as the time from randomization until the date of permanent study treatment discontinuation (for any reason) at any time during the Core phase.~The percentage event-free probability estimate is the estimated probability that a patient will remain on-treatment up to a specified time point (Week 6, 12, 18)" (NCT01713946)
Timeframe: Week 6, Week 12, Week 18

,,
InterventionPercentage event-free prob. estimates (Number)
Week 6Week 12Week 18
Everolimus HT Target of 9 to 15 ng/mL96.295.471.5
Everolimus LT Target of 3 to 7 ng/mL97.495.770.1
Placebo99.297.575.6

[back to top]

Core Phase: Incidence of Suicide Attempt, Suicidal Ideation or Behavior During Core Phase Per Columbia Suicide Severity Rating Scale (C-SSRS) Outcomes

"Comparison of suicidality using the C-SSRS in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm. The Columbia-Suicide Severity Rating Scale (C-SSRS) is a questionnaire used for suicide assessment developed by multiple institutions, including Columbia University, with NIMH support. The scale is evidence-supported and is part of a national and international public health initiative involving the assessment of suicidality. There are different scoring systems depending on the population. The important elements to note are that the higher the scores on the individual items and the more yes items, the higher the suicide risk." (NCT01713946)
Timeframe: Baseline, Week 18

,,
InterventionParticipants (Number)
Completed suicideSuicide attemptPrep actions toward imminent suicidal behaviorSuicidal ideationSelf-injurious behavior without suicide intent
Everolimus HT Target of 9 to 15 ng/mL00010
Everolimus LT Target of 3 to 7 ng/mL01230
Placebo00000

[back to top]

Core Phase: Distribution of Reduction From Baseline in Seizure Frequency

Comparison of percentage of patients in six categories of seizure reduction from baseline (≤ -25% (exacerbation); > -25% to < 25% (no change); ≥ 25% to < 50%; ≥ 50% to < 75%; ≥ 75% to < 100%; 100% (seizure-freedom)) in the everolimus low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm during maintenance period of the core phase (NCT01713946)
Timeframe: Baseline (8-week period before randomization), Week 7 to 18 (12-week maintenance period of the core phase)

,,
InterventionPercentage of participants (Number)
100% (seizure free)≥ 75 to <100 (75% responder)≥ 50 to <75 (50% responder)≥ 25 to <50 (25% responder)>-25 to <25 (No change)≤ -25 (Exacerbation)Missing (missing)
Everolimus HT Target of 9 to 15 ng/mL3.815.420.830.018.511.50.0
Everolimus LT Target of 3 to 7 ng/mL5.16.017.123.935.012.80.0
Placebo0.85.09.222.741.220.20.8

[back to top]

Core Phase: Change From Baseline in the QOLIE-AD-48 Overall Quality-of-life Score for Patients >=11 to 18 Years

Comparison of quality of life (from 3 age specific questionnaires) in the everolimus low-trough treatment arm (3-7 ng/mL), hightrough treatment arm (9-15 ng/mL) and placebo arm at the end of the core phase. The Quality of Life in Epilepsy Inventory for Adolescents-48 (QOLIE-AD-48) is a survey of health-related quality of life for adolescents 11 to 18 years of age with epilepsy. The QOLIE-AD-48 is completed by the patient. It contains 48 items which assess 8 subscales. Scores range from 0-100, with higher scores corresponding to improved QoL. The overall quality of life score is obtained by summing a linear combination of the 8 subscale scores, where each subscale is multiplied by a relative weight that is provided in the original publication. (NCT01713946)
Timeframe: Baseline, Week 18

,,
Interventionscores on a scale (Median)
BaselineEnd of Core PhaseChange from Baseline
Everolimus HT Target of 9 to 15 ng/mL58.860.75.8
Everolimus LT Target of 3 to 7 ng/mL56.158.54.7
Placebo59.665.47.2

[back to top]

Core Phase: Change From Baseline in Wechsler Nonverbal Composite Score

The brief version of the WNV consists of a 2-subtest battery: only Matrices and Recognition subtests for patients under 8, and Matrices and Spatial Span subtests for patients aged 8 to 21. Based on the raw scores obtained from the subtests, standardized z-scores were calculated for each subtest using the following formula: Zscore = (X - b)/Sb where X is the raw score of the subtest, b and Sb represent the mean and standard deviation respectively of the subtest score recorded at baseline for the study population. The composite WNV score was computed by summing up the Z-scores of the 3 subtests of the WNV (i.e. matrices, recognition, and coding for patients aged <8 years and matrices, spatial span, and coding for patients aged 8 to 21 years). The composite WNV score has no range. (NCT01713946)
Timeframe: Baseline, Week 18

,,
Interventionscores on a scale (Median)
BaselineEnd of Core PhaseChange from Baseline
Everolimus HT Target of 9 to 15 ng/mL-0.69-0.890.00
Everolimus LT Target of 3 to 7 ng/mL-0.48-0.040.00
Placebo-1.11-0.510.00

[back to top]

Long Term Evaluation: Percentage Change From Start of Everolimus in Seizure Frequency by Time Window

"Percentage change from start of everolimus in average weekly seizure frequency (SFcfe) = 100 × (SFe - SFtw) ÷ SFe where:~SFe is the average weekly seizure frequency in the 8-week period before start of everolimus SFtw is the average weekly seizure frequency in a 12-week time window A positive percentage change from start of everolimus (SFcfe) means a reduction in seizure frequency whereas a negative percentage change from start of everolimus (SFcfe) means an increase in seizure frequency." (NCT01713946)
Timeframe: Baseline (8-week period before start of everolimus), Week 7 to 18, Week 19 to 30, and 12 weeks thereafter up to Week 102

InterventionPercent change (Median)
Week 18Week 30Week 42Week 54Week 66Week 78Week 90Week 102
Everolimus Long Term Evaluation (LTE)31.6535.7442.8646.0549.0751.6957.3359.69

[back to top]

Core Phase: Change From Baseline in the QOLCE Overall Quality-of-life Score for Patients <11 Years

Comparison of quality of life in the everolimus (from 3 age specific questionnaires) low-trough treatment arm (3-7 ng/mL), high-trough treatment arm (9-15 ng/mL) and placebo arm at the end of the core phase. The Quality of Life Childhood Epilepsy (QOLCE) questionnaire, used for patients < 11 years at baseline, was completed by the patient's parent or caregiver. It consists of 16 subscales (13 multi-item scales and 3 single item scales) and one overall quality-of-life score. Scores range from 0-100, with higher scores corresponding to improved QoL. The Overall Quality of Life Score is computed by adding each subscale score for each individual and then dividing by 16. (NCT01713946)
Timeframe: Baseline, Week 18

,,
Interventionscores on a scale (Median)
BaselineEnd of Core PhaseChange from Baseline
Everolimus HT Target of 9 to 15 ng/mL56.559.50.0
Everolimus LT Target of 3 to 7 ng/mL52.353.60.2
Placebo55.357.21.0

[back to top]

Percentage of Participants With Progression Free Survival at 6 Months

Response was be determined by bi-dimensional diameters. RECIST criteria will be collected and used for secondary evaluation. Patients will have brain MRI scans with and without gadolinium performed prior to therapy, after every second course in the first year, after every third course in the second year, and at the End of Study visit (if not done within prior 3 months). Spine MRIs should be performed prior to therapy and at the same time points as standard brain MRIs if clinically indicated. (NCT01734512)
Timeframe: Up to 6 months

Interventionpercentage (Number)
Everolimus67

[back to top]

Overall Survival (OS) Events (Number of Deaths) - FAS

Overall survival (OS) is defined as the time from date of start of treatment to date of death due to any cause. If a patient is not known to have died, survival will be censored at the date of last contact. Time to median OS was not estimable. (NCT01743560)
Timeframe: Start of treatment to the date of death up to approximately 48 weeks

InterventionNumber of events (Number)
DeathsNumber of censored observations
Everolimus and Exemestane841

[back to top]

Overall Survival (OS) - % Event-free Probability Estimate - FAS

Overall survival (OS) is defined as the time from date of start of treatment to date of death due to any cause. If a patient is not known to have died, survival will be censored at the date of last contact. (NCT01743560)
Timeframe: Start of treatment to the date of death up to approximately 48 weeks

InterventionPercentage of participants (Number)
Event free at 12 weeksEvent free at 24 weeksEvent free at 36 weeksEvent free at 48 weeks
Everolimus and Exemestane93.383.974.274.2

[back to top]

Change From Baseline EORTC Quality of Life Questionnaire of Cancer Patients QLQ-C30 at Each Time Point

The QLQ-C30 is composed of multi-item scales and single-item measures including 5 functional scales, 3 symptom scales, a global health status-QoL scale, and 6 single items. Each of the multi-item scales includes a different set of items - no item occurs in more than 1 scale. High scale score=higher response level; a high score for a functional scale=a healthy level of function, high score for the global health status/QoL=high quality of life but a high score for a symptom scale / item=high level of symptomatology/problems. The principle for scoring these scales: 1.) Estimate the average of the items that contribute to the scale = raw score. 2.) Linear transformation to standardize the raw score, so that scores range from 0 to 100. Results should be interpreted with caution as the numbers of patients with available data over time were limited, and because of high variances as evidenced by large standard deviations (NCT01743560)
Timeframe: Baseline 12,24,36,48 weeks

,,,
Interventionscores (Mean)
Global health status/QoLPhysical functioningRole functioningEmotional functioningCognitive functioningSocial functioningFatigueNausea/ vomitingPainDyspneaInsomniaAppetite lossConstipationDiarrheaFinancial problems
Week 12-9.0-5.1-4.52.6-8.6-9.98.6-1.21.218.53.730.94.94.9-1.2
Week 24-3.6-1.03.1-3.1-5.2-9.49.52.93.98.32.023.511.88.3-10.4
Week 361.94.98.37.51.33.80.9-3.8-1.32.6012.810.3-5.1-10.3
Week 48-8.3-10.8-12.30.6-3.7-14.77.3-2.40.610.7-3.616.78.37.4-2.6

[back to top]

Progression-free Survival (PFS) - % Event-free Probability Estimate - FAS

Progression-free survival (PFS) is the time from date of start of treatment to the date of event defined as the first documented progression or death due to any cause. If a patient has not had an event, progression-free survival is censored at the date of last adequate tumor assessment. Tumor assessment and response was evaluated according to RECIST v1.1Response was assessed by local radiology review. The PFS was analyzed using the Kaplan Meier method. (NCT01743560)
Timeframe: Start of treatment to the date of event defined as first documented progression due to any cause up to approximately 48 weeks

InterventionPercentage of participants (Number)
Event free at 12 weeksEvent free at 24 weeksEvent free at 36 weeksEvent free at 48 weeks
Everolimus and Exemestane67.949.128.918.4

[back to top]

Progression-free Survival (PFS) Events as Per Investigators - FAS

Progression-free survival (PFS) is the time from date of start of treatment to the date of event defined as the first documented progression or death due to any cause. If a patient has not had an event, progression-free survival is censored at the date of last adequate tumor assessment. Tumor assessment and response was evaluated according to RECIST v1.1Response was assessed by local radiology review. (NCT01743560)
Timeframe: Start of treatment to the date of event defined as first documented progression due to any cause up to approximately 48 weeks

InterventionNumber of events (Number)
DeathsProgression of diseaseNumber of censored observations
Everolimus and Exemestane82516

[back to top]

Change From Baseline in EuroQoL 5-dimension Visual Analogue Scores - FAS

EuroQoL Quality of Life Scale (EQ-5D) is a standardized instrument to assess health state values. The EQ-5D essentially consists of 2 pages: the EQ-5D descriptive system and the EQ visual analogue scale (VAS). For the visual analogue scale, participants draw a line from a box to the point on the thermometer-like scale corresponding to their health state, 0-100 (100 = Best health state). Weights are used to score the responses to the 5 domains, with scores ranging from 0 to 1 (where a score of 1 represents a perfect state). Scores for the visual analogue scale reflect the position where participant's line crosses the thermometer-like scale. Results should be interpreted with caution as the numbers of patients with available data over time were limited, and because of high variances as evidenced by large standard deviations (NCT01743560)
Timeframe: Baseline 12,24,36,48 weeks

Interventionunits on a scale (Mean)
Week 12-7.9
Week 24-6.1
Week 36-6.3
Week 48-11.6

[back to top]

Overall Response Rate of Everolimus and Exemestane Treatment in Postmenopausal Women With Hormone Receptor Positive Locally Advanced or Metastatic Breast Cancer

The Overall Response Rate (ORR) was defined as the proportion of patients with a best OR of confirmed CR or PR by week 48. Treatment success is defined as: The best Overall Response (OR) for each patient is determined from the sequence of investigator overall lesion responses according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1.). To be assigned a best OR of Complete Responese (CR) at least two determinations of CR at least 4 weeks apart before progression are required. To be assigned a best OR of Partial Response (PR) at least two determinations of PR or better at least 4 weeks apart before progression (and not qualifying for a CR) are required. (NCT01743560)
Timeframe: At 48 weeks

InterventionPercentage of participants (Number)
Everolimus and Exemestane14.3

[back to top]

Progression-free Survival (PFS) by Median Time in Weeks as Per Investigators - FAS

Progression-free survival (PFS) is the time from date of start of treatment to the date of event defined as the first documented progression or death due to any cause. If a patient has not had an event, progression-free survival is censored at the date of last adequate tumor assessment. Tumor assessment and response was evaluated according to RECIST v1.1Response was assessed by local radiology review. (NCT01743560)
Timeframe: Start of treatment to the date of event defined as first documented progression due to any cause up to approximately 48 weeks

Interventionweeks (Median)
Everolimus and Exemestane23.6

[back to top]

Percentage of Patient Responses in EuroQoL 5-dimension Questionnaire - FAS

EuroQoL Quality of Life Scale (EQ-5D) is a standardized instrument to assess health state values is a standardized instrument to assess health state values. The EQ-5D essentially consists of 2 pages: the EQ-5D descriptive system and the EQ visual analogue scale (VAS). The EQ-5D descriptive system is comprised of the following 5 dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension has 5 levels: no problems, slight problems, moderate problems, severe problems and extreme problems. Percentage of participants' responses were presented by visits. Results should be interpreted with caution as the numbers of patients with available data over time were limited. (NCT01743560)
Timeframe: Baseline 12,24,36,48 weeks

,,,,
InterventionPercentage of participants (Number)
Mobility-no problemMobility-slight problemMobility-moderate problemMobility-severe problemMobility-unable to walkSelf-care - no problemSelf-care - slight problemSelf-care - moderate problemSelf-care -severe problemSelf-care - unableUsual activities - no problemsUsual activities - slight problemsUsual activities - moderate problemsUsual activities - severe problemsUsual activities -unable to doPain/discomfort - nonePain/discomfort - slightPain/discomfort - moderatePain/discomfort - severePain/discomfort - extremeAnxiety/depression - noneAnxiety/depression - slightAnxiety/depression - moderateAnxiety/depression - severeAnxiety/depression - extreme
Day 138.818.432.76.1069.418.46.12.0030.622.428.610.24.120.430.640.84.1044.934.716.300
Week 1220.414.320.42.0044.94.18.20014.316.322.44.1014.316.322.44.1022.422.412.200
Week 2418.410.28.20034.702.00022.42.012.20010.218.48.20018.412.26.100
Week 3614.38.22.02.0024.502.00014.38.22.02.0012.28.26.10016.310.2000
Week 4816.316.314.36.14.140.810.24.12.0018.414.312.26.16.118.420.414.34.1022.420.48.26.10

[back to top]

Best Overall Response of Everolimus and Exemestane Treatment in Postmenopausal Women With Hormone Receptor Positive Locally Advanced or Metastatic Breast Cancer

The best Overall Response (OR) for each patient is determined from the sequence of investigator overall lesion responses according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1.). To be assigned a best OR of Complete Responese (CR) at least two determinations of CR at least 4 weeks apart before progression are required. To be assigned a best OR of Partial Response (PR) at least two determinations of PR or better at least 4 weeks apart before progression (and not qualifying for a CR) are required.The Overall Response Rate (ORR) was defined as the proportion of patients with a best OR of confirmed CR or PR by week 48. (NCT01743560)
Timeframe: At 48 weeks

Interventionparticipants (Number)
Patients with measurable disease at baselinePatients with non-measurable disease at baselineBest at WK 48 - Complete Response (CR)Best at WK 48 - Partial Response (PR)Best at WK 48 - Stable Disease (SD)Best at WK 48 - Progressive Disease (PD)UnknownMissing
Everolimus and Exemestane391007181518

[back to top]

Mean Change in Treatment Satisfaction Questionnaire for Medication (TSQM) Between Week 3 and 12

TSQM was used to measure the Patients' self-reported satisfaction or dissatisfaction with the study treatment. The differences in mean scale scores between weeks 3 and 12 comparing treatment satisfaction in the different treatment arms: everolimus + exemestane combination therapy versus everolimus monotherapy, and everolimus + exemestane combination therapy versus capecitabine monotherapy. The TSQM version 1.4 domain scores range from 0 to 100 with higher scores representing a higher satisfaction on that domain. (NCT01783444)
Timeframe: Week 3, Week 12

,,
InterventionUnit on a scale (Mean)
Side-effectsEffectivenessConvenienceGlobal satisfaction
Capecitabine 1250 mg/m2-2.61.20.52.3
Everolimus 10 mg-9.11.21.01.8
Everolimus 10 mg + Exemestane 25 mg-4.8-2.2-0.6-1.0

[back to top]

Time to Eastern Cooperative Oncology Group (ECOG) Performance Deterioration

The Eastern Cooperative Oncology Group (ECOG) Performance Status is a scale used to assess physical health of subjects,ranging from 0 (most active) to 5 (least active). Definitive deterioration is defined as no improvement in the ECOG status following observation of the deterioration. (NCT01783444)
Timeframe: Baseline, every 6 weeks up to about 43 months

InterventionWeeks (Median)
Everolimus 10 mg + Exemestane 25 mg72.57
Everolimus 10 mg126.57
Capecitabine 1250 mg/m2120.00

[back to top]

Time to 10% Definitive Deterioration in the Global Health Status / Quality of Life

The global health status/QoL scale score of the QLQ-C30 is identified as the primary PRO variable of interest. Physical Functioning (PF), Emotional Functioning (EF) and Social Functioning (SF) scale scores of the QLQ-C30. The time to definitive 10% deterioration is the number of days between the date of randomization and the date of the assessment at which definitive deterioration is seen. Definitive 10% (5-point) deterioration is defined as a decrease in score by at least 10% (5-points) compared to baseline, with no later increase above this threshold observed during the course of the study. (NCT01783444)
Timeframe: Baseline, every 6 weeks up to about 43 months

InterventionWeeks (Median)
Everolimus 10 mg + Exemestane 25 mg30.86
Everolimus 10 mg23.86
Capecitabine 1250 mg/m261.29

[back to top]

Progression Free Survival (PFS) - Everolimus Plus Exemestane Versus Capecitabine Alone

Progression Free Survival (PFS) is defined as the time from date of randomization to the date of first radiologically documented progression or death due to any cause. If a patient did not have an event, PFS was censored at the date of the last adequate tumor assessment. PFS was compared between the everolimus + exemestane combination therapy with the everolimus monotherapy. (NCT01783444)
Timeframe: Date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first, reported between day of first patient randomized up to 39 months

InterventionMonths (Median)
Everolimus 10 mg + Exemestane 25 mg8.41
Capecitabine 1250 mg/m29.59

[back to top]

Progression Free Survival (PFS) - Everolimus Plus Exemestane Versus Everolimus Alone

Progression Free Survival (PFS) is defined as the time from date of randomization to the date of first radiologically documented progression or death due to any cause. If a patient did not have an event, PFS was censored at the date of the last adequate tumor assessment. PFS was compared between the everolimus + exemestane combination therapy with the everolimus monotherapy. (NCT01783444)
Timeframe: Date of randomization to the date of first documented tumor progression or death from any cause, whichever occurs first, reported between day of first patient randomized up to 39 months

InterventionMonths (Median)
Everolimus 10 mg + Exemestane 25 mg8.41
Everolimus 10 mg6.77

[back to top]

Clinical Benefit Rate (CBR)

Clinical Benefit Rate (CBR) is defined as the proportion of participants with a best overall response of complete response (CR) or partial response (PR) or stable disease (SD) or Non-CR/non-PD lasting more than 24 weeks based on local investigator's assessment according to RECIST 1.1. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR, Stable disease (SD), neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for PD; CBR = CR+PR+SD. Only descriptive statistics. (NCT01783444)
Timeframe: From the date of randomization until the date of the first documented disease progression or date of death from any cause whichever came first, assessed for approximately 43 months

InterventionPercentage of Participants (Number)
Everolimus 10 mg + Exemestane 25 mg59
Everolimus 10 mg43
Capecitabine 1250 mg/m253

[back to top]

Overall Response Rate (ORR)

Overall Response Rate (ORR) as the proportion of patients whose best overall response is either complete response (CR) or partial response (PR) according to RECIST 1.1 This was assessed in the full patient population. Complete response is achieved when all lesions evaluated at baseline are absent at subsequent visit. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. Only descriptive statistics. (NCT01783444)
Timeframe: From the date of randomization until the date of the first documented disease progression or date of death from any cause whichever came first, assessed for approximately 43 months

InterventionPercentage of Participants (Number)
Everolimus 10 mg + Exemestane 25 mg21
Everolimus 10 mg12
Capecitabine 1250 mg/m223

[back to top]

All Collected Deaths

"On treatment deaths were collected from FPFT up to 30 days after study drug discontinuation, for a maximum duration of 224 weeks (treatment duration ranged from 1.3 to 220.0 weeks).~Deaths post treatment survival follow up were collected after the on- treatment period, up to approximately 5 years. Patients who didn't die during the on-treatment period and had not stopped study participation at the time of data cut-off (end of study) were censored." (NCT01783444)
Timeframe: up to 224 weeks (on-treatment), up to approximately 5 years (study duration)

,,
InterventionParticipants (Count of Participants)
On-treatment deathsPost-treatment deathsAll deaths
Capecitabine 1250 mg/m225759
Everolimus 10 mg55560
Everolimus 10 mg + Exemestane 25 mg96271

[back to top]

Overall Survival (OS)

Overall Survival (OS) is defined as the time from date of randomization to date of death due to any cause. If a patient was not known to have died by the date of analysis cut-off, OS was censored at the date of last known date patient alive. (NCT01783444)
Timeframe: Every 3 months following end of treatment visit, assessed for approximately 54 months

InterventionMonths (Median)
Everolimus 10 mg + Exemestane 25 mg23.06
Everolimus 10 mg29.27
Capecitabine 1250 mg/m225.56

[back to top]

Disease Stabilization Rate - Phase II

"Disease stabilization rate is defined as the proportion of patients achieving a best overall response of complete response, partial response, or stable disease.~Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.~Partial Response (PR): At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters.~Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. Persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits." (NCT01784861)
Timeframe: Through completion of treatment (estimated to be 12 months)

InterventionParticipants (Count of Participants)
Phase II: X-82 + Everolimus1

[back to top]

Progression Free Survival (PFS) - Phase II

"PFS is defined as the duration of time from start of treatment to time of progression or death, whichever occurs first.~Progressive Disease (PD): At least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. Appearance of one or more new lesions and/or unequivocal progression of existing non-target lesions. Unequivocal progression should not normally trump target lesion status. It must be representative of overall disease status change, not a single lesion increase." (NCT01784861)
Timeframe: Up to 3 years

Interventiondays (Median)
Phase II: X-82 + Everolimus183

[back to top] [back to top]

Number of Participants With Toxicity - Phase II

Toxicity will be graded by NCI Common Terminology Criteria for Adverse Events (CTCAE version 4.0) (NCT01784861)
Timeframe: Through 30 days after completion of treatment (estimated to be 13 months)

InterventionParticipants (Count of Participants)
DiarrheaMucositis oralNauseaVomitingChest painEdema limbsHematomaMouth soresCreatinine increasedPlatelet count decreasedHip painLeg crampsDizzinessHeadacheAllergic rhinitisRash acneiformRash maculo-papularHypertension
Phase II: X-82 + Everolimus211111111111111111

[back to top]

Overall Toxicities - Phase I

-Toxicities will be graded by the NCI Common Terminology Criteria for Adverse Events (CTCAE version 4.0) (NCT01784861)
Timeframe: 30 days after completion of treatment (estimated to be 13 months)

,,,,
InterventionParticipants (Count of Participants)
AnemiaHearing impairedPeripheral vision/depth perception impairmentAbdominal distensionAbdominal painColonic fistulaConstipationDiarrheaDry mouthDyspepsiaFlatulenceHemorrhoidsIntra-abdominal hemorrhageLoss of tasteMucositis oralNauseaRectal fissureStomach painTaste changesVomitingChillsEdema faceEdema limbsFatigueFeverPainCold soresElevated lactate dehydrogenase totalOral thrushPeritoneal infectionSinusitisUpper respiratory infectionUrinary tract infectionBruisingSlow wound healingAlkaline phosphatase increasedBlood bilirubin increasedCreatinine increasedNeutrophil count decreasedPlatelet count decreasedWeight lossAnorexiaDehydrationHypernatremiaHypertriglyceridemiaHypophosphatemiaArthralgiaBack painFlank painMyalgiaPain to extremityAphasiaDizzinessDysgeusiaHeadacheTremorTrigeminal nerve disorderAnxietyConfusionDepressionInsomniaLibido decreasedMood swingsHematuriaUrinary frequencyTesticular painCoughDyspneaEpistaxisHiccupsHypoxiaNasal congestionSore throatAlopeciaHair color changesPruritisRash acneiformRash maculo-papularSkin hypopigmentationSkin thinningHot flashesHypertensionDysphagiaHypovolemiaIntraperitoneal bleedingNight sweatsThromboembolic eventLung infectionPancreas infectionSepsisDiabetic ketoacidosisStrokePleural effusionMesenteric ischemia
Phase I Dose Level 0: X-82 + Everolimus0100100200000021000100010000000110000000111000000001101000001000000101000000000000000100000100
Phase I Dose Level 1: X-82 + Everolimus1011001201001021000110121000211000000000010000011000100000100000000100100000011000011010011001
Phase I Dose Level 2: X-82 + Everolimus0000101200100021011110230210000000000020130000111000000111010001002110001110021001000000000000
Phase I Dose Level 3: X-82 + Everolimus1000511320110143100234352000002002111134130021021120223000113110212130010002120101000010000010
Phase I Dose Level 4: X-82 + Everolimus0000200200001021000100120101000000000100032110100000111000001000001100000000110010100001100000

[back to top]

Number of Participants With Dose Limiting Toxicities - Phase I

Tolerability of X-82 in combination with everolimus will be determined by NCI Common Terminology Criteria for Adverse Events (CTCAE version 4.0) (NCT01784861)
Timeframe: Completion of 1st cycle for all patients in Phase I portion of study (completed in approximately 20 months)

InterventionParticipants (Count of Participants)
Phase I Dose Level 0: X-82 + Everolimus0
Phase I Dose Level 1: X-82 + Everolimus0
Phase I Dose Level 2: X-82 + Everolimus0
Phase I Dose Level 3: X-82 + Everolimus1
Phase I Dose Level 4: X-82 + Everolimus0

[back to top]

Objective Response Rate (Complete Response + Partial Response) - Phase II

"Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.~Partial Response (PR): At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters." (NCT01784861)
Timeframe: Through completion of treatment (estimated to be 12 months)

InterventionParticipants (Count of Participants)
Phase II: X-82 + Everolimus0

[back to top]

Overall Survival - Phase II

Start of the treatment until death. (NCT01784861)
Timeframe: Up to 3 years

Interventiondays (Median)
Phase II: X-82 + Everolimus115

[back to top]

Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

Any sign or symptom that occurs during the study treatment plus the 30 days post treatment. All SAEs were captured in safety database from enrollment. Safety data collection was changed in the protocol amendment released in March 2016: AEs and SAEs were captured in the clinical database from protocol amendment release (18 March 2016). Hence, SAEs from both safety database and clinical database are summarized separately. (NCT01789281)
Timeframe: SAEs collected in safety database from enrollment to end of treatment (EOT) plus 30 days, up to approximately 7.2 years. AEs/SAEs collected in clinical database from protocol amendment date 18 March 2016 to EOT plus 30 days, up to approximately 4.5 years

,
InterventionParticipants (Count of Participants)
SAEs (Safety database)Treatment-related SAEs (Safety database)AEs (Clinical Database)Tretament-related AEs (Clinical Database)SAEs (Clinical Database)Treatment-related SAEs (Clinical Database)
Everolimus617420
Everolimus+Sandostatin LAR947442

[back to top]

Percentage of Patients With Clinical Benefit

Percentage of patients with clinical benefit as judged by the investigator. Investigator attestation of continued clinical benefit was collected in clinical database after protocol amendment (release date 18 March 2016). Clinical benefit assessment before protocol amendment was done retrospectively. (NCT01789281)
Timeframe: After 3 months from enrollment, every 3 months, until end of treatment, assessed up to 7.2 years

,
InterventionParticipants (Count of Participants)
At 3 monthsAt 6 monthsAt 9 monthsAt 12 monthsAt 15 monthsAt 18 monthsAt 21 monthsAt 24 monthsAt 27 monthsAt 30 monthsAt 33 monthsAt 36 monthsAt 39 monthsAt 42 monthsAt 45 monthsAt 48 monthsAt 51 monthsAt 54 monthsAt 57 monthsAt 60 monthsAt 63 monthsAt 66 monthsAt 69 monthsAt 72 monthsAt 75 monthsAt 78 monthsAt 81 monthsAt 84 months
Everolimus9888888653211111100000000000
Everolimus+Sandostatin LAR7777777777777666665544333210

[back to top]

Clinical Benefit Rate

Clinical benefit rate is defined as number of patients with objective response (complete response or partial response) or stable disease for at least 24 weeks divided by number of patients randomized in each arm. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI and/or CT: Partial Response (PR) is defined as >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD) is defined as neither sufficient shrinkage to qualify for a Partial Response nor sufficient increase to qualify for Progression of Disease; Complete Response (CR) is defined as disappearance of all target lesions. (NCT01797120)
Timeframe: Every 3 months until progression or up to 3 years

Interventionproportion of patients (Number)
Fulvestrant & Everolimus0.636
Fulvestrant & Placebo0.415

[back to top]

Objective Response Rate

Objective response rate is defined as number of patients with complete or partial response (by Physical Exam, CT or MRI) divided by number of patients randomized in each arm (NCT01797120)
Timeframe: Every 3 months until progression or up to 3 years

Interventionproportion of patients (Number)
Fulvestrant & Everolimus0.182
Fulvestrant & Placebo0.123

[back to top]

Overall Survival

Overall survival will be characterized using Kaplan-Meier plots and other descriptive metrics. (NCT01797120)
Timeframe: Every 3 months until progression or up to 3 years

Interventionmonths (Median)
Fulvestrant & Everolimus28.3
Fulvestrant & Placebo31.4

[back to top]

Progression-free Survival

Progression-free survival documented by Physical Exam, CT Scan or MRI in post-menopausal patients with hormone-receptor positive metastatic breast cancer that is resistant to aromatase inhibitor therapy treated with fulvestrant and everolimus compared to fulvestrant alone from randomization to documented disease progression or death. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01797120)
Timeframe: Every 3 months until progression or up to 3 years

Interventionmonths (Median)
Fulvestrant & Everolimus10.3
Fulvestrant & Placebo5.1

[back to top]

Number of Participants With an Objective Response

ORR is the proportion of participants with a complete response (CR) or partial response (PR) per the Response Evaluation Criteria in Solid Tumors (RECIST). Complete Response is disappearance of all tumors. Partial Response is at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. (NCT01827384)
Timeframe: Up to 30 days after completion of study treatment, up to 75 months

,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial Response
TAC1 -> TAC400
TAC200
TAC2 -> TAC100
TAC2 -> TAC300
TAC301
TAC3 -> TAC100
TAC3 -> TAC1 -> TAC400
TAC3 -> TAC400
TAC400
TAC4 -> TAC100
TAC4 -> TAC200
TAC4 -> TAC300
Treatment Assignment Code 1 (TAC1)00

[back to top]

Proportion of Participants With 4 Month Progression-free Survival (PFS)

Time from random assignment to progression or death from any cause (whichever comes first). Progression was measured by the Response Evaluation Criteria in Solid Tumors (RECIST). Progression is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study. And the appearance of one or more new lesions is also considered progressions. (NCT01827384)
Timeframe: 4 months

Interventionproportion of participants (Number)
Treatment Assignment Code 1 (TAC1)0.23
TAC1 -> TAC40.00
TAC20.22
TAC2 -> TAC10.00
TAC2 -> TAC31.00
TAC30.41
TAC3 -> TAC10.50
TAC3 -> TAC1 -> TAC40.00
TAC3 -> TAC40.00
TAC40.13
TAC4 -> TAC10.29
TAC4 -> TAC20.00
TAC4 -> TAC30.00

[back to top]

Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v5.0)

Here is the number of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria for Adverse Events (CTCAE v5.0). A non-serious adverse event is any untoward medical occurrence. A serious adverse event is an adverse event or suspected adverse reaction that results in death, a life-threatening adverse drug experience, hospitalization, disruption of the ability to conduct normal life functions, congenital anomaly/birth defect or important medical events that jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the previous outcomes mentioned. (NCT01827384)
Timeframe: Date treatment consent signed to date off study, approx. 73months (m) & 21day (d); 4m & 11d; 61m & 8d; 4m & 11d; 10m & 11d; 72m & 29d; 13m & 11d; 6m & 13d; 29m & 18d; 48m & 25d; 49m & 5d; 4m &7d; 15m & 5d; and 75m &13d, for each group respectively.

InterventionParticipants (Count of Participants)
Treatment Assignment Code 1 (TAC1)13
TAC1 -> TAC41
TAC29
TAC2 -> TAC11
TAC2 -> TAC31
TAC322
TAC3 -> TAC12
TAC3 -> TAC1 -> TAC41
TAC3 -> TAC42
TAC415
TAC4 -> TAC17
TAC4 -> TAC21
TAC4 -> TAC32
Participants Enrolled But Not Treated2

[back to top]

Percent of Participants With Delayed Graft Function by Day

Delayed graft function (DGF) was defined as the need for dialysis within the first 7 days post-transplantation, excluding the first post-transplantation day. (NCT01843348)
Timeframe: Post transplant up to day 7

,,
InterventionPercent of participants (Number)
day 1 (8,7,4)day 2 (2,2,2)day 3 (5,1,2)day 4 (4,2,4)day 5 (4,5,3)day 6 (1,1,2)day 7 (2,4,0)>7 days (9,16,21)
CycA+Certican10.55.35.310.57.95.30.055.3
TAC+Certican18.45.32.65.313.22.610.542.1
TAC+MPA22.95.714.311.411.42.95.725.7

[back to top]

Glomular Filtration Rate (GFR) mL/Min Via Cockcroft- Gault Method at Month 12 Post Transplant

Cockcroft-Gault formula: For men: GFR= ((140-age) × body weight in kg)∕(72 x serum creatinine in mg∕dl)For women: GFR= (0.85×(140-age) × body weight in kg)∕(72 x serum creatinine in mg/dl), ), last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT01843348)
Timeframe: Month 12 post transplant

InterventionmL/min per 1.73m² (Least Squares Mean)
TAC+MPA60.26
TAC+Certican52.25
CycA+Certican51.30

[back to top]

Glomular Filtration Rate (GFR) Via Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) Method at Month 12 Post Transplant

Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) method = GFR=141 x min(Scr/κ, 1)α x max(Scr/κ, 1)1.209 x 0.993Age x 1.018 [if female] x 1.159 [if black] where Scr is serum creatinine, κ is 0.7 for females and 0.9 for males, α is 0.329 for females and 0.411 for males, min indicates the minimum of Scr/κ or 1, and max indicates the maximum of Scr/κ or 1. last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT01843348)
Timeframe: Month 12 post transplant

InterventionmL/min per 1.73m² (Least Squares Mean)
TAC+MPA51.62
TAC+Certican44.42
CycA+Certican42.44

[back to top]

Glomular Filtration Rate (GFR) Via Modification of Diet in Renal Disease (MDRD) Method at Month 12 Post Transplant

Modification of Diet in Renal Disease (MDRD) = For men: GFR = 170 x (serum creatinine -0,999) x (age-0,176) x (urea nitrogen -0,17) x (albumin0,318) For women: GFR = 170 x (serum creatinine -0,999) x (age-0,176) x (urea nitrogen -0,17) x albumin0,318) x 0.762 with urea nitrogen = urea / 2.144. last observation carried forward (LOCF) was used for imputation of missing values, ANCOVA model (NCT01843348)
Timeframe: Month 12 post transplant

InterventionmL/min per 1.73m² (Least Squares Mean)
TAC+MPA53.24
TAC+Certican45.72
CycA+Certican43.47

[back to top]

Duration of Wound Healing

A wound will be considered healed if all the suture material and staples are removed and the wound is intact. Number of participants is based on all patients of the respective treatment group in the safety set, excluding patients with no answer (unknown). (NCT01843348)
Timeframe: Post transplant until individual reporting

Interventiondays (Mean)
TAC+MPA42.4
TAC+Certican54.1
CycA+Certican85.3

[back to top]

Glomular Filtration Rate (GFR) mL/Min Via Nankivell Method at Month 12 - Standard Regimen vs Certican Regimens

"To demonstrate non-inferiority in renal function assessed by glomerular filtration rate (Nankivell formula) in at least one of the Certican® treatment regimens compared to the standard regimen group at month 12 post-transplantation in renal transplant patients. Nankivell formula:~GFR = 6.7/Scr + BW/4 - Surea/2 - 100/(height)² + C where Scr is the serum creatinine concentration expressed in mmol/L, BW the body weight in kg, Surea the serum urea in mmol/L, height in m, and the constant C is 35 for male and 25 for female patients. The eGFR is expressed in mL/min per 1.73m². If a patient was on dialysis at the time of urea or creatinine assessment, the eGFR was set to 0. Analysis set = per protocol set" (NCT01843348)
Timeframe: One year post transplant

,,
InterventionmL/min per 1.73m² (Mean)
Month 1 - Day 1 to 60 (146,111,78)Month 3 - Day 61 to 136 (143,108,79)Month 6 - Day 137 to 228 (142,108,76)Month 9 - Day 229 to 319 (140,106,77)Month12 - Day 320 to 450 (147,111, 80)
CycA+Certican59.4762.2263.1762.8961.51
TAC+Certican60.5461.2162.7664.6863.34
TAC+MPA62.6266.3668.0569.4770.41

[back to top]

Percent of Participants With Delayed Graft Function and Slow Graft Function

Delayed graft function (DGF) was defined as the need for dialysis within the first 7 days post-transplantation, excluding the first post-transplantation day. Slow graft function (SGF) was defined as a serum creatinine >3.0 mg/dL at Day 5 post-transplantation. Full analysis set (NCT01843348)
Timeframe: Post transplant to month 12

,,
InterventionPercent of participants (Number)
Delayed graft function (197,187,172)Slow graft function (195,187,171)
CycA+Certican22.149.7
TAC+Certican20.348.7
TAC+MPA17.846.2

[back to top]

Percent of Participants With Viral Infections

Viral infections for BKV Virus Humane Polyomavirus 1 and Cytomegalovirus (NCT01843348)
Timeframe: Post transplant to month 12

,,
InterventionPercent of participants (Number)
Viral infections - CMVMissingViral infections - CMVAsymptomaticViral infections - CMVMildViral infections - CMVModerateViral infections - CMVSevereViral infections - BKVAsymptomaticViral infections - BKVMildViral infections - BKVModerateViral infections - BKVSevere
CycA+Certican1.01.01.01.00.05.03.01.00
TAC+Certican0.01.02.01.00.08.07.02.00
TAC+MPA1.07.05.06.01.010.05.07.00

[back to top]

Percent of Participants With Wound Healing Complications During Study

Information collected to report wound healing process which included percentage of participants with complications, fluid collections detected and occurrence of lymphoceles (NCT01843348)
Timeframe: Post transplant until individual reporting

,,
InterventionPercent of participants (Number)
Wound healing complicationFluids detectedOccurrence of lymphoceles
CycA+Certican22.227.821.8
TAC+Certican19.126.818.2
TAC+MPA14.318.711.8

[back to top]

Percentage of Participants With Composite Treatment Failure Endpoints - Difference Between Groups at Month 12

Combined endpoint included: biopsy proven acute rejection (BPAR) defined as a rejection which was acute and proven by biopsy, graft loss (GL) defined as: allograft was presumed to be lost on the day the patient starts dialysis and not able to be removed from dialysis or death. Patients who prematurely discontinued the study: if the patient did not suffer from an event before discontinuation and reason was not related to efficacy, the patient was assessed as having had no event, otherwise the patient was assessed as having had an event. Full analysis set (FAS) (NCT01843348)
Timeframe: Month 12 post transplant

,,,,
InterventionPercentage of participants (Number)
BPAR or graft loss or deathBPAR, graft loss, death, or loss of follow-up
CycA+Certican24.632.7
CycA+Certican -Tac+MPA - Difference Between Groups14.917.1
TAC+Certican13.022.6
Tac+Certican - Tac+MPA - Difference Between Groups3.27.0
TAC+MPA9.815.6

[back to top]

Percentage of Participants With Treatment Failure Endpoints at Month 12

Treatment failure endpoints: biopsy proven acute rejection (BPAR) defined as a rejection which was acute and proven by biopsy, graft loss (GL) defined as: allograft was presumed to be lost on the day the patient starts dialysis and not able to be removed from dialysis or death. Patients who prematurely discontinued the study: if the patient did not suffer from an event before discontinuation and reason was not related to efficacy, the patient was assessed as having had no event, otherwise the patient was assessed as having had an event. Full analysis set (FAS) (NCT01843348)
Timeframe: Month 12 post transplant

,,
InterventionPercentage of participants (Number)
Biopsy proven acute rejection (BPAR)Treated BPAR (tBPAR)Graft lossDeath
CycA+Certican24.623.69.06.5
TAC+Certican12.011.56.36.3
TAC+MPA9.38.85.44.9

[back to top]

Objective Response Rate (ORR)

Objective Response Rate (ORR) is the number of participants with a best response of complete response (CR) or partial response (PR) divided by number of randomized participants. ORR was assessed by the Independent Radiology Committee (IRC) per RECIST 1.1 which was confirmed by a subsequent visit >= 28 days later, and was analyzed in the Intent to Treat (ITT) population at the time of the primary analysis of Progression Free Survival (PFS). The data cutoff date was 22 May 2015. (NCT01865747)
Timeframe: ORR was assessed at 8 weeks post-randomization, every 8 weeks for 12 months, and every 12 weeks until date of disease progression or death, up to May 2015 (approximately 21 months)

Interventionpercentage of participants (Number)
Cabozantinib (XL184)17
Everolimus (Afinitor)3

[back to top]

Overall Survival (OS)

Overall Survival (OS) is defined as the time from randomization to the date of death. Participants that had not died were censored at last known date alive. Median OS was calculated using Kaplan-Meier estimates. Interim analyses for OS occurred after 320 deaths (78% of the total OS events needed for final analysis). (NCT01865747)
Timeframe: OS was measured from the time of randomization until 320 deaths, approximately 28 months

Interventionmonths (Number)
Cabozantinib (XL184)21.4
Everolimus (Afinitor)16.5

[back to top]

Progression-free Survival (PFS)

The primary analysis of PFS is the time from randomization to date of first documented tumor progression as determined by investigator (per RECIST 1.1 criteria) or death due to any cause, whichever occurred first. A Kaplan-Meier analysis was performed to estimate the median duration. (NCT01865747)
Timeframe: PFS is measured from the date of randomization until the date of first documented disease progression or date of death from any cause as determined by the Independent Radiology Committee (IRC) per RECIST 1.1, assessed for up to 17 months.

Interventionmonths (Number)
Cabozantinib (XL184)7.4
Everolimus (Afinitor)3.8

[back to top]

Composite Efficacy Failure of Treated Biopsy in Everolimus With Reduced Tacrolimus Group Compared to Standard Tacrolimus in Patients From Japan Only

"Rate of composite efficacy failure of treated biopsy in everolimus with reduced tacrolimus group compared to standard tacrolimus from randomization in core study up to 36 months in the extension study.~Composite endpoint = treated BPAR, graft loss or death. AR = Acute rejection; tAR = treated AR; BPR = biopsy proven rejection; BPAR = biopsy proven acute rejection; tBPAR = treated BPAR" (NCT01888432)
Timeframe: randomization, 36 months post transplantion

,
InterventionParticipants (Number)
Composite endpointOn-treatment composite endpointGraft loss/deathtBPARGraft lossDeathARtARBPRBPAR
EVR+Reduced TAC2112013263
TAC Control1010102032

[back to top]

Number of Participants With Composite of tBPAR, Graft Loss, and Death

Compare between the treatment group EVR with rTAC vs standard TAC: incidence of a composite of tBPAR, graft loss, death (NCT01888432)
Timeframe: Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
tBPAR/graft loss/deathOn-treatment tBPAR/graft loss/death
EVR+Reduced TAC127
TAC Control119

[back to top]

Number of Participants With Time to Recurrence of HCC in Subjects With a Diagnosis of HCC at the Time of Liver Transplantation

Patients transplanted for HCC or with HCC diagnosed at time of transplantation were monitored for HCC recurrence according to local practice. For example routine laboratory monitoring/tests, tumor markers, hepatic ultrasound, computed tomography scans (CAT, CT) or MRI (especially Fe-MRI) on a regular basis per local practice. (NCT01888432)
Timeframe: Month 12 and Month 24

,
InterventionParticipants (Count of Participants)
HCC recurrence (n/M) - month 12HCC recurrence (n/M) - month 24
EVR+Reduced TAC01
TAC Control56

[back to top]

Compare Renal Function Over Time Assessed by the Change by eGFR, Post-randomization

Change in renal function from randomization to month 24 assessed by the change in estimated GFR (MDRD-4). Rate of change of renal function. (NCT01888432)
Timeframe: From randomziation to month 24

InterventionmL/min/1.73 m2 (Least Squares Mean)
EVR+Reduced TAC-11.01
TAC Control-14.26

[back to top]

Renal Function by Estimated Glomerular Filtration Rate (eGFR) From Randomization

Renal function (change in estimated glomerular filtration rate (eGFR)) from randomization to Month 12 post transplantation with everolimus (EVR) in combination with reduced tacrolimus (rTAC) compared to standard exposure tacrolimus (TAC) in living donor liver transplant recipients. (NCT01888432)
Timeframe: From randomization to month 12

InterventionmL/min/1.73 m^2 (Least Squares Mean)
EVR+Reduced TAC-7.94
TAC Control-12.09

[back to top]

Number of Subjects Experiencing Adverse Events/Infections by SOC

(NCT01888432)
Timeframe: Month 24

,
InterventionParticipants (Count of Participants)
Any AE/infectionBlood and lymphatic system disordersCardiac disordersCongenital, familial and genetic disordersEar and labyrinth disordersEndocrine disordersEye disordersGastrointestinal disordersGeneral disorders&admin site conditionsHepatobiliary disordersImmune system disordersInfections and infestationsInjury, poisoning&proced. complicationsInvestigationsMetabolism and nutrition disordersMusculoskeletal and connective tissue disordersNeoplasms benign, malig&unspecified (cysts&polyps)Nervous system disordersProduct issues#Psychiatric disordersRenal and urinary disordersReproductive system&breast dis.Respiratory, thoracic&mediastinal dis.Skin&subcutaneous tissue disordersVascular disorders
EVR+Reduced TAC140441512117984844884366187301038133469343938
TAC Control13632122521574424011702868604317441263612394430

[back to top]

Number of Participants With Composite Efficacy Failure of Treated Biopsy Proven Acute Rejection, Graft Loss or Death in Everolimus With Reduced Tacrolimus Group Compared to Standard Tacrolimus

Rate of composite efficacy failure of treated biopsy proven acute rejection (tBPAR ≥ RAI score 3), graft loss (GL) or death (D) in everolimus with reduced tacrolimus group compared to standard tacrolimus at 12 months (NCT01888432)
Timeframe: 12 months post transplantation

InterventionParticipants (Count of Participants)
EVR+Reduced TAC7
TAC Control8

[back to top]

Compare Incidence of tAR

Compare between the treatment group EVR with rTAC vs standard TAC: incidence of treated acute rejection (tAR). (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
Month 12Month 24
EVR+Reduced TAC57
TAC Control67

[back to top]

Renal Function by Estimated Glomerular Filtration Rate (All Extension Patients)

Renal function (change in estimated glomerular filtration rate (eGFR)) from randomization to Month 36 post transplantation with everolimus (EVR) in combination with reduced tacrolimus (rTAC) compared to standard exposure tacrolimus (TAC) in living donor liver transplant recipients in Japan (NCT01888432)
Timeframe: randomization, at 36 months post transplantation

InterventionmL/min/1.73m2 (Least Squares Mean)
EVR+Reduced TAC-26.88
TAC Control-16.87

[back to top]

Compare Incidence of a Composite of Death or Graft Loss

Compare between the treatment group EVR with rTAC vs standard TAC: Incidence of a composite of death or graft loss (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
Month 12Month 24
EVR+Reduced TAC48
TAC Control35

[back to top]

Compare Incidence of AR

Compare between the treatment group EVR with rTAC vs standard TAC: incidence of acute rejection (AR) (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
Month 12Month 24
EVR+Reduced TAC912
TAC Control89

[back to top]

Compare Incidence of BPAR

Compare between the treatment group EVR with rTAC vs standard TAC: incidence of a composite of biopsy proven acute rejection (BPAR) (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
Month 12Month 24
EVR+Reduced TAC78
TAC Control67

[back to top]

Compare Incidence of Death

Compare between the treatment group EVR with rTAC vs standard TAC: incidence of death (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
Month 12Month 24Month 24 (on-treatment death)
EVR+Reduced TAC484
TAC Control343

[back to top]

Compare Incidence of Graft Loss

Compare between the treatment group EVR with rTAC vs standard TAC: incidence of graft loss (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
Month 12month 24month 24 (on-treatment graft loss)
EVR+Reduced TAC000
TAC Control010

[back to top]

Compare Incidence of Notable Safety Events (SAEs, Infections and Serious Infections Leading to Premature Discontinuation)

Notable events include death, Serious AE/infection,, and AE/infection leading to discontinuation of study medication. (NCT01888432)
Timeframe: Month 24

,
InterventionParticipants (Count of Participants)
Any notable eventsDeathSerious AE/InfectionAE/Infection lead. to premature disc of study med
EVR+Reduced TAC8688321
TAC Control8247818

[back to top]

Compare Incidence of tBPAR

Compare between the treatment group EVR with rTAC vs standard TAC: Incidence of tBPAR (NCT01888432)
Timeframe: Month 12 and Month 24 post transplantation

,
InterventionParticipants (Count of Participants)
tBPAR - month 12tBPAR - month 24On-treatment tBPAR - month 24
EVR+Reduced TAC343
TAC Control566

[back to top]

Number of All Death (Cardiac, Vascular, and Non-cardiovascular)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.) This study as no primary or secondary endpoints, all endpoints are of equal weight.~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above).~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)91

[back to top]

Number of All Deaths, Myocardial Infarction, Any Repetitive Revascularization Composite Endpoints

"All deaths include Cardiac death, Cardiovascular death and Non-cardiovascular death.~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)330

[back to top]

Number of Participants With All Target Vessel Revascularization (TVR)

"Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)26

[back to top]

Number of Participants With All TLR

This study has no primary or secondary endpoints, all endpoints are of equal weight. (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)1

[back to top]

Number of Participants With All TLR

This study has no primary or secondary endpoints, all endpoints are of equal weight. (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)58

[back to top]

Number of Participants With ID-TVR (TLR and TVR, Non-target Lesion)

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Revascularization includes TLR, TVR, non-target lesion, and non TVR." (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)59

[back to top]

Number of Participants With Cardiac Death and All Myocardial Infarction (MI) (Q-wave and Non-Q Wave) Composite Endpoint

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)7

[back to top]

Number of Participants With All TVR (TLR and TVR, Non-target Lesion)

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~All TVR (TLR and TVR, non-target lesion)" (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)1

[back to top]

Number of Participants With ID-TVR (TLR and TVR, Non-target Lesion)

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Revascularization includes TLR, TVR, non-target lesion, and non TVR." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)1

[back to top]

Number of Participants With Target Lesion Failure (TLF)

"Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR).~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)5

[back to top]

Number of Participants With Sub-acute Stent Thrombosis

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points and with the different separate time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Acute scaffold/stent thrombosis : 0 - 24 hours post stent implantation Subacute scaffold/stent thrombosis: >24 hours - 30 days post stent implantation Late scaffold/stent thrombosis: 30 days - 1 year post stent implantation Extremely late scaffold/stent thrombosis: >1 year post stent implantation" (NCT01894152)
Timeframe: > 1 day to 30 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)2

[back to top]

Number of Participants With ID-TVR, Non-target Lesion

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Revascularization includes TLR, TVR, non-target lesion, and non TVR." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)0

[back to top]

Number of Participants With ID-TVR, Non-target Lesion

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Revascularization includes TLR, TVR, non-target lesion, and non TVR." (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)22

[back to top]

Number of Participants With Late Stent Thrombosis

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points and with the different separate time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Acute scaffold/stent thrombosis : 0 - 24 hours post stent implantation Subacute scaffold/stent thrombosis: >24 hours - 30 days post stent implantation Late scaffold/stent thrombosis: 30 days - 1 year post stent implantation Extremely late scaffold/stent thrombosis: >1 year post stent implantation" (NCT01894152)
Timeframe: 31 to 365 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)1

[back to top]

Number of Participants With Overall Stent Thrombosis

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points and with the different separate time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Acute scaffold/stent thrombosis : 0 - 24 hours post stent implantation Subacute scaffold/stent thrombosis: >24 hours - 30 days post stent implantation Late scaffold/stent thrombosis: 30 days - 1 year post stent implantation Extremely late scaffold/stent thrombosis: >1 year post stent implantation" (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)3

[back to top]

Number of All Death (Cardiac, Vascular, and Non-cardiovascular)

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.) This study as no primary or secondary endpoints, all endpoints are of equal weight.~- Non-cardiac death is defined as a death not due to cardiac causes (as defined above).~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)3

[back to top]

Number of Participants With Early Stent Thrombosis

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points and with the different separate time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Acute scaffold/stent thrombosis : 0 - 24 hours post stent implantation Subacute scaffold/stent thrombosis: >24 hours - 30 days post stent implantation Late scaffold/stent thrombosis: 30 days - 1 year post stent implantation Extremely late scaffold/stent thrombosis: >1 year post stent implantation" (NCT01894152)
Timeframe: 0 - 30 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)2

[back to top]

Number of All Deaths, Myocardial Infarction, Any Repetitive Revascularization Composite Endpoints

"All deaths include Cardiac death, Cardiovascular death and Non-cardiovascular death.~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)8

[back to top]

Number of Participants With All TVR (TLR and TVR, Non-target Lesion)

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~All TVR (TLR and TVR, non-target lesion)" (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)84

[back to top]

Number of Participants With Target Lesion Failure (TLF)

"Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR).~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)100

[back to top]

Number of Participants With Very Late Stent Thrombosis

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points and with the different separate time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Acute scaffold/stent thrombosis : 0 - 24 hours post stent implantation Subacute scaffold/stent thrombosis: >24 hours - 30 days post stent implantation Late scaffold/stent thrombosis: 30 days - 1 year post stent implantation Extremely late scaffold/stent thrombosis: >1 year post stent implantation" (NCT01894152)
Timeframe: > 365 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)0

[back to top]

Number of Participants With Acute Stent Thrombosis

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Scaffold/Stent thrombosis should be reported as a cumulative value at the different time points and with the different separate time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Acute scaffold/stent thrombosis : 0 - 24 hours post stent implantation Subacute scaffold/stent thrombosis: >24 hours - 30 days post stent implantation Late scaffold/stent thrombosis: 30 days - 1 year post stent implantation Extremely late scaffold/stent thrombosis: >1 year post stent implantation" (NCT01894152)
Timeframe: 0 to 1 day

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)0

[back to top]

Number of Participants With Target Vessel Failure (ID-TVF) (Cardiac Death, All Myocardial Infarctions and Ischemia-driven Target Vessel Revascularization)

"Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or ischemia-driven Target Vessel Revascularization (ID-TVR).~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)7

[back to top]

Number of Participants With Target Vessel ARC MI

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG. Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)23

[back to top]

Number of Participants With Target Vessel ARC MI

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG. Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)2

[back to top]

Number of Participants With Composite Rate of All Deaths and Myocardial Infarctions (MI)

"All deaths include Cardiac death, Cardiovascular death and Non-cardiovascular death.~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)8

[back to top]

Number of Participants With ID-TLR

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Revascularization includes TLR, TVR, non-target lesion, and non TVR." (NCT01894152)
Timeframe: 0 to 1885 days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)37

[back to top]

Number of Participants With ID-TLR

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~Revascularization includes TLR, TVR, non-target lesion, and non TVR." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)1

[back to top]

Number of Participants With Target Vessel Failure (ID-TVF) (Cardiac Death, All Myocardial Infarctions and Ischemia-driven Target Vessel Revascularization)

"Target Vessel Failure (TVF) is the composite of Cardiac Death, Myocardial infarction (MI) or ischemia-driven Target Vessel Revascularization (ID-TVR).~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)180

[back to top]

Number of Participants With Cardiogenic Death, Target Vessel Blood Flow Myocardial Infarction, Target Lesion Revascularization Composite Endpoint

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI): Patient diagnosed with myocardial infarction, but its relation with target vessel not clear, therefore considered target vessel myocardial infarction.~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)5

[back to top]

Number of Participants With Cardiogenic Death, Target Vessel Blood Flow Myocardial Infarction, Target Lesion Revascularization Composite Endpoint

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.) Myocardial Infarction (MI): Patient diagnosed with myocardial infarction, but its relation with target vessel not clear, therefore considered target vessel myocardial infarction.~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)120

[back to top]

Number of Participants With Composite Rate of All Deaths and Myocardial Infarctions (MI)

"All deaths include Cardiac death, Cardiovascular death and Non-cardiovascular death.~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)156

[back to top]

Number of Participants With All Target Vessel Revascularization (TVR)

"Target Vessel Revascularization is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion which includes upstream and downstream branches and the target lesion itself.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)0

[back to top]

Number of Participants With All Revascularization (Target Lesion, Target Vessel, and Non-target Vessel) (PCI and Coronary Artery Bypass Graft [CABG])

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~All Revascularization includes Coronary artery bypass grafting and Percutaneous coronary intervention" (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)223

[back to top]

Number of Participants With All Revascularization (Target Lesion, Target Vessel, and Non-target Vessel) (PCI and Coronary Artery Bypass Graft [CABG])

"This study has no primary or secondary endpoints, all endpoints are of equal weight.~All Revascularization includes Coronary artery bypass grafting and Percutaneous coronary intervention" (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)1

[back to top]

Number of Participants With All Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)75

[back to top]

Number of Participants With All Myocardial Infarction (MI) (Including Q-wave and Non-Q-wave)

"Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: ≤ 7 days after index procedure (Hospitalization)

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)6

[back to top]

Number of Participants With Cardiac Death and All Myocardial Infarction (MI) (Q-wave and Non-Q Wave) Composite Endpoint

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded.~(This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality, cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI):~Q wave MI: Development of new, pathological Q wave on the ECG.~Non-Q wave MI: Elevation of Creatine kinase (CK) levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves.~This study has no primary or secondary endpoints, all endpoints are of equal weight." (NCT01894152)
Timeframe: 0 through 1885 Days

InterventionParticipants (Count of Participants)
XIENCE PRIME Everolimus Eluting Coronary Stent System (EECSS)114

[back to top]

The Number of Polyoma BK Viremia Patients

Patients will be monitored at regular interval for the development of Polyomavirus Viremia. (NCT01911546)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Everolimus + Low-dose Tacrolimus5

[back to top]

The Number of CMV Viremia

The number of patients with CMV viremia (NCT01911546)
Timeframe: 12 Months

InterventionParticipants (Count of Participants)
Everolimus + Low-dose Tacrolimus0

[back to top]

Incidence of Cell Mediated Rejection (CMR)

Patients will be monitored for any episodes of CMR. (NCT01911546)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Everolimus + Low-dose Tacrolimus2

[back to top]

Incidence of Antibody Mediated Rejection (ABMR)

Protocol biopsies were obtained at T0 and 6 months post transplant. (NCT01911546)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Everolimus + Low-dose Tacrolimus2

[back to top]

Frequency of Seizures Assessed by Total Number of Seizures

Parents will be asked to document the frequency of their child's seizures using a manual or electronic (seizuretracker.com) seizure diary. The total number of seizures at baseline for all participants. (NCT01929642)
Timeframe: at baseline

Interventionseizures (Number)
Everolimus or Sirolimus0

[back to top]

Feasibility Measurements of Parental Stress

"Measurements of stress will be administered. Specifically, we will use the Parental Stress Index. Quantifying stress, as well as compliance with the study protocol, will allow investigators to objectively assess the feasibility of a larger clinical trial of sirolimus in patients with TSC.~Parental stress index maximum score: 180 Parental stress index minimum score: 36 higher raw scores indicate higher levels of stress." (NCT01929642)
Timeframe: Change from baseline to EOT visit 12 week 53

Interventionunits on a scale (Mean)
Everolimus or Sirolimus111

[back to top]

Cognitive Function as Assessed by the Capute Scale

"Score range maximum: 100 Score range minimum: 0~High values represent a high cognitive function Below 70 is abnormal. 70-100 is the normal range." (NCT01929642)
Timeframe: 1 year

Interventionscore on a scale (Mean)
Everolimus or Sirolimus15

[back to top]

Caregiver Burden

"The Caregiver Burden Scale is a standard set of questions which will be used to measure the non-medical impact of TSC on caregivers and how it affects the feasibility of study completion.~The Caregiver's Burden Scale (CBS) is a 22-item scale that assess subjectively experienced burden by caregiver's to chronically disabled persons. maximum scores: 88 & Minumum scores: 22~High values represent a worse outcome" (NCT01929642)
Timeframe: Change from baseline to EOT visit 12 week 53

Interventionunits on a scale (Mean)
Everolimus or Sirolimus61

[back to top]

Number of Participants With Compliance to the Treatment Protocol.

One outcome measurement of feasibility will include family/patient compliance with the treatment protocol, which will be assessed and documented at every study visit and telephone follow-up call, by the physician and/or study team member. This was calculated by calculating dividing the total number of study visits and study assessments completed by the total number of study visits and study assessments indicated by the treatment protocol. (NCT01929642)
Timeframe: Change from baseline to EOT visit 12 week 53

Interventionpercentage of completed visits/measures (Number)
Participant 002Participant 003Participant 001
Everolimus or Sirolimus1005858

[back to top]

Total Number of Aggressions or Self-injuries

This is the total number of aggressions or self-injuries for all participants. (NCT01929642)
Timeframe: 1 year

InterventionNumber of aggressions or self-injuries (Number)
Everolimus or Sirolimus82

[back to top]

Repetitive Behavior

"Repetitive behavior will be assessed using the Repetitive Behavior Scale - revised, a questionnaire to characterize several domains of repetitive behavior including ritualistic behavior, stereotypic behavior, self-injurious behavior, compulsive behavior, and restricted interests.~There are 36 items on the scale. Behaviors are rated on a 4-point scale: 0-Behavior does not occur, 1-Behavior occurs and is a mild problem, 2-Behavior occurs and is a moderate problem, 3-Behavior occurs and is a severe problem.~Maximum score: 108 & minimum score: 0 A high score represents the worse outcome" (NCT01929642)
Timeframe: 1 year

Interventionscore on a scale (Mean)
Everolimus or Sirolimus69

[back to top]

Tumor Response

"Evaluate tumor response using RECIST criteria after 12 weeks of treatment at definitive surgery.~Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR., or similar definition that is accurate and appropriate." (NCT01931163)
Timeframe: tumor response at 12 weeks after treatment

InterventionParticipants (Count of Participants)
Everolimus Plus Cisplatin22

[back to top]

Leukopenia

The number of patients with leukopenia as indicated by white blood cell count less than 1.0, absolute neutrophil count less than 500 or the need for exogenous granulocyte stimulating factor administration (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus1

[back to top]

Infection Requiring Hospitalization

The number of patients with serious infections as defined by need for hospitalization (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus11

[back to top]

Impaired Glucose Tolerance

The number of patients with impaired glucose tolerance as indicated by fasting blood glucose levels, Hemoglobin A1C (HgbA1C) levels and the need for hypoglycemic medications (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus14

[back to top]

Graft Survival

Graft survival is defined as the percentage of kidney transplants still functioning at 2 years post baseline visit . One patient died of natural causes at 12 months with a functioning graft. (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus49

[back to top]

Gastrointestinal Complaints

The number of patients with gastrointestinal complaints as indicated by abdominal pain, nausea, vomiting or diarrhea not accounted for by a specific episode of illness such as gastroenteritis (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus22

[back to top]

Development of Donor Specific Antibody

The number of patients with incidence of development of donor specific antibody (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus2

[back to top]

Cytomegalovirus

The number of patients with Incidence of cytomegalovirus infection as defined by need for hospitalization (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus0

[back to top]

BK Nephropathy

The number of patients with BK nephropathy as defined by biopsy. Note that biopsies were not required as part of the study but were only done as part of the patient's standard of care if rejection was suspected (i.e. if the serum creatinine increased by 25% and was not associated with elevated tacrolimus levels or clinical signs of dehydration/illness to account for elevated creatinine) (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus0

[back to top]

BK Infection

The number of patients with BK infection as defined by blood titers requiring reduction in immunosuppressive dose (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus7

[back to top]

Biopsy Proven Acute Rejection

The percentage of patients with a treated biopsy-proven acute rejection (a co-primary endpoint) within the 2 year study time period (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus1

[back to top]

Cardiovascular Complications

The number of patients with cardiovascular complications as indicated by conditions such as dysrhythmias, coronary artery disease requiring intervention or myocardial infarction (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus1

[back to top]

Lipid Levels

The number of patients with hyperlipidemia as defined by the development of new onset hyperlipidemia in the baseline negative patients and the number of baseline positive patients who required starting a new lipid-lowering medication or an increase in dose of their lipid-lowering medication over the course of the study (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Baseline negative patients with new onset hyperlipidemiaBaseline positive patients started on new lipid-lowering medication or needing increase in dose
Arm 1 Everolimus/Reduced Dose Tacrolimus719

[back to top]

Thrombocytopenia

The number of patients with thrombocytopenia as defined by platelet count less than 50 (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus0

[back to top]

Renal Function

"Renal function in patients will be assessed using glomerular filtration rate (GFR) as measured by the Modified Diet Renal Disease (MDRD) estimation.~Glomerular filtration is the process by which the kidneys filter the blood, removing excess wastes and fluids. Glomerular filtration rate (GFR) is a calculation that determines how well the blood is filtered by the kidneys, which is one way to measure remaining kidney function. GFR is also used to find the stage of chronic kidney disease. Glomerular filtration rate is usually calculated using a mathematical formula that compares a person's size, age, sex, and race to serum creatinine levels. The higher the GFR number, the better the kidney function; the lower the GFR number, the worse the kidney function. A GFR of 60 or higher is in the normal range. A GFR below 60 may mean kidney disease. A GFR of 15 or lower may mean kidney failure." (NCT01935128)
Timeframe: 2 years

InterventionmL/min/1.73 m2 (Mean)
Arm 1 Everolimus/Reduced Dose Tacrolimus65.70

[back to top]

Proteinuria

The number of patients with proteinuria as defined by spot urine protein to creatinine ratio greater than 1.0 (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus7

[back to top]

Pneumonitis

The number of patients with pneumonitis as demonstrated by lung inflammation symptoms such as shortness of breath and/or cough requiring clinical intervention and management (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus3

[back to top]

Patient Survival

Patient survival is defined as the percentage of patients still surviving at 2 years post baseline visit (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus49

[back to top]

Neurotoxicity

The number of patients with neurotoxicity as evidenced by incidence of new onset seizure activity or tremors (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus1

[back to top]

Mouth Ulcers

The number of patients with stomatitis/aphthous ulcer (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus14

[back to top]

Malignancies

The number of patients developing malignancies including post-transplant lymphoproliferative disorders (NCT01935128)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm 1 Everolimus/Reduced Dose Tacrolimus2

[back to top]

Recipient and Donor Genotyping for Selected Variants of CYP3A5, ABCB1 (MDR1), and CYP4A Genes

A blood sample was obtained from recipients and donors to measure gene polymorphism effects on metabolism of calcineurin inhibitor and everolimus. The polymorphisms are represented as the number of SNP occurrences for the CYP3A5, ABCB1 (MDR1) gene, and CYP4A4*22 genes. (NCT01936519)
Timeframe: 2 years

,
InterventionParticipants (Count of Participants)
Recipient Genotypes : rs776746 (CYP3A5 gene)Recipient Genotypes : rs1045642 (ABCB1 gene)Recipient Genotypes : rs1128503 (ABCB1 gene)Recipient Genotypes : rs2032582 (ABCB1 gene)Recipient Genotypes : rs35599367 (CYP4A4*22 gene)Donor Genotypes : rs776746 (CYP3A5 gene)Donor Genotypes : rs1045642 (ABCB1 gene)Donor Genotypes : rs1128503 (ABCB1 gene)Donor Genotypes : rs2032582 (ABCB1 gene)Donor Genotypes : rs35599367 (Cyp4A4*22)
Calcineurin Inhibitor and Mycophenolic Acid11671131000
Everolimus and Mycophenolic Acid11680105001

[back to top]

Renal Function as Measured by 24 Hour Urine Creatinine Clearance

Renal Function was assessed by 24 hr urine collection creatinine clearance measured (mL/min). 24 Hr urine collection was assessed at baseline, 6 months, 1 year, and 2 years post transplant. (NCT01936519)
Timeframe: 6 months, 1 year, and 2 years

,
InterventionmL/min (Mean)
24hr Urine Creatinine Clearance at 6 months24hr Urine Creatinine Clearance at 1 year24hr Urine Creatinine Clearance at 2 years
Calcineurin Inhibitor With Mycophenolic Acid68.0968.0961.54
Everolimus With Mycophenolic Acid70.7586.890.63

[back to top]

Renal Function as Measured by Cockcroft Gault Creatinine Clearance

The Cockcroft-Gault formula for estimating creatinine clearance was determined at 6 months, 1 year, and 2 years post transplant (NCT01936519)
Timeframe: 6 months, 1 year, and 2 years

,
InterventionmL/min/1.73m2 (Mean)
Cockcroft Gault Creatinine Clearance at 6 monthsCockcroft Gault Creatinine Clearance at 1 yearCockcroft Gault Creatinine Clearance at 2 years
Calcineurin Inhibitor and Mycophenolic Acid79.8486.8480.85
Everolimus and Mycophenolic Acid100.17113.47108.16

[back to top]

Renal Function as Measured by Iothalamate Clearance

Iothalamate Clearance was assessed at 6 months, 1 year, and 2 years post transplant. (NCT01936519)
Timeframe: 6 months, 1 year, and 2 years

,
InterventionmL/min/1.73m2 (Mean)
Iothalamate Clearance at 6 monthsIothalamate Clearance at 1 yearIothalamate Clearance at 2 years
Calcineurin Inhibitor and Mycophenolic Acid67.9966.6557.19
Everolimus and Mycophenolic Acid74.23104.0179.41

[back to top]

Renal Function as Measured by Modification of Diet in Renal Disease (MDRD) Estimated Glomerular Filtration Rate (eGFR)

Modification of Diet in Renal Disease (MDRD) estimated Glomerular Filtration Rate (eGFR) was assessed at 6 months, 1 year, and 2 years post transplant. (NCT01936519)
Timeframe: 6 months, 1 year, and 2 years

,
InterventionmL/min/1.73 m2 (Mean)
MDRD eGFR at 6 monthsMDRD eGFR at 1 yearMDRD eGFR at 2 years
Calcineurin Inhibitor and Mycophenolic Acid62.1860.6353.29
Everolimus and Mycophenolic Acid81.2788.0187.37

[back to top]

Renal Function as Measured by Serum Creatinine Level

Serum creatinine levels were assessed at 6 months, 1 year, and 2 years post transplant (NCT01936519)
Timeframe: 6 months, 1 year, and 2 years

,
Interventionmg/dL (Mean)
Serum Creatinine at 6 monthsSerum Creatinine at 1 yearSerum Creatinine at 2 years
Calcineurin Inhibitor and Mycophenolic Acid1.291.291.51
Everolimus and Mycophenolic Acid1.020.950.95

[back to top]

Number of Participants With Composite Endpoint of Graft Survival (Non-death Censored) and Biopsy Proven Acute Rejection at 1 Year

"The primary objective of this pilot study will be to determine equivalency of Zortress® as compared to Rapamune® when used in our de novo immunosuppression regimen following renal transplantation. The primary endpoint will be a composite endpoint of graft survival (non-death censored) and biopsy proven acute rejection at 1 year. The primary outcome of immunosuppressive protection would be studied in our Thymoglobulin and rapid steroid discontinuation protocol, with half-dose Neoral as described above." (NCT01976390)
Timeframe: 1 Year

,
InterventionParticipants (Count of Participants)
graft survival (non-death censored) at 1 yearbiopsy proven acute rejection at 1 year
Rapamune (Sirolimus)1919
Zortress (Everolimus)4038

[back to top]

Number of Pariticipants With Adverse Events as a Measure of Safety and Tolerability

Any sign or symptom that occurs during the study treatment plus the 30 days post treatment. (NCT02017860)
Timeframe: Adverse events were collected from first dose of study treatment until end of study treatment plus 30 days post treatment, up to maximum duration of approximately 5 years.

InterventionCount of Participants (Number)
Participants who had at least one Adverse EventParticipants with Grade 3/4 Adverse Event
Everolimus42

[back to top]

Rate of Reduction in Seizures

To assess the reduction in seizure rate the investigator will compare the seizure rate on study to that experienced one month prior to enrolling in the study. The RANO low grade gliomas (LGG) guideline will be used to assess reduction of seizures, which calls a 50% or more reduction number of monthly seizures an 'improvement'; a 50% or more increase a worsening'; and anything less than 50% in either direction a 'stable seizure rate'. (NCT02023905)
Timeframe: Up to 36 Months

Interventionpercentage of participants (Number)
Arm 1: Everolimus25
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)50

[back to top]

Progression-Free Survival Rate (PFS) (Arms 1 and 2)

Participants will be analyzed based on intention to treat using the Response Assessment in Neuro-Oncology (RANO) criteria for progression. Based on RANO criteria, a responder is defined using both radiographic and clinical criteria. Complete response (CR) or Partial Response (PR) will be first assessed by radiographic changes as determined by an improvement of the bi-dimensional evaluation of the tumor size. In addition, changes in neurologic function and steroid use will be considered. Analyses will be performed after all enrolled participants in arm 1 or arm 2 have completed 33 months on study, or whenever the progression status of all participants has been established, whichever comes first. Kaplan-Meier estimates and the associated 95% confidence intervals (CI) will be calculated for the 33-month PFS separately for the two arms. (NCT02023905)
Timeframe: Up to 33 Months

Interventionpercentage of participants (Number)
Arm 1: Everolimus31
Arm 2: Everolimus and TemozolomideNA

[back to top]

Progression-Free Survival Rate (PFS) (Arm 3)

Participants will be analyzed based on intention to treat using the Response Assessment in Neuro-Oncology (RANO) criteria for progression. Based on RANO criteria, a responder is defined using both radiographic and clinical criteria. Complete response (CR) or Partial Response (PR) will be first assessed by radiographic changes as determined by an improvement of the bi-dimensional evaluation of the tumor size. In addition, changes in neurologic function and steroid use will be considered. Analyses will be performed after all participants enrolled in arm 3 have completed 38 months on study, or whenever the progression status of all participants has been established, whichever comes first. Kaplan-Meier estimates and the associated 95% CIs will be calculated for the 38-month PFS. (NCT02023905)
Timeframe: Up to 38 Months

Interventionpercentage of participants (Number)
Arm 3: Everolimus (1p/19q Co-deletion Present)55.5

[back to top]

Overall Survival Rate (OS)

Participants will be analyzed based on an intention to treat model. Overall survival is defined as the first day of treatment until death or study completion, whichever comes first. Kaplan-Meier estimates and the associated 95% CIs will be calculated by treatment arm. (NCT02023905)
Timeframe: Up to 84 Months

Interventionpercentage of participants (Number)
Arm 1: EverolimusNA
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)NA

[back to top]

Median Progression Free Survival (PFS)

Participants will be analyzed based on an intention to treat model. PFS is defined as the time from first objective response to the time of disease progression or death using the Response Assessment in Neuro-Oncology (RANO) criteria to determine progression. If the participant does not have an event of disease progression or recurrence nor has the patient died, the participant's data will be censored at the date of last contact with the patient. Kaplan-Meier estimates and the associated 95% CIs will be calculated. (NCT02023905)
Timeframe: Up to 84 Months

Interventionmonths (Median)
Arm 1: Everolimus25.8
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)43.6

[back to top]

Objective Response Rate (ORR)

Overall response rate (ORR) is the percentage of patients who achieved a best response of complete response (CR) or partial response (PR) out of all assigned patients. Based on the best objective status as assessed by the Response Assessment in Neuro-Oncology (RANO) criteria. Using RANO criteria, a responder is defined by radiographic and clinical criteria. Complete response or PR will be first assessed by radiographic changes as determined by an improvement of the bi-dimensional evaluation of the tumor size. In addition, changes in neurologic function and steroid use will be considered. The point estimate and the associated 2-sided 95% CI for the response rate separately for the three arms. (NCT02023905)
Timeframe: Up to 36 Months

Interventionpercentage of participants (Number)
Arm 1: Everolimus0
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)11

[back to top]

Overall Survival (OS)

OS is defined as time from randomization to death from any cause. Patients who are still alive are censored at the last date of known alive. (NCT02031536)
Timeframe: assessed every 3 months if < 2 years from study entry, and every 6 months up to 5 years from study entry or death, whichever occurred first

Interventionyears (Median)
Arm A (Everolimus)2.05

[back to top]

Disease Free Survival (DFS)

DFS is defined as time from randomization to disease recurrence or death, whichever occurred first. Patients who are still alive are censored at the last date of known free of recurrence. (NCT02031536)
Timeframe: assessed every 12 weeks while on treatment and then every 3 months if < 2 years from study entry; every 6 months up to 5 years from study entry or recurrence, whichever occurred first

Interventionyears (Median)
Arm A (Everolimus)1.3

[back to top]

Safety of Simvastatin in the Treatment of LAM-S and LAM-TS Patients

Safety is a primary outcome measure which will be assessed by any major changes or deterioration in patient health. (NCT02061397)
Timeframe: 5 months

Interventionparticipants (Number)
Single Simvastatin Treatment Arm0

[back to top]

Percent Predicted FEV1

Lung function will be measured by FEV1: forced expiratory volume in 1s mean and calculated as % predicted +_ SD (standard deviation). (NCT02061397)
Timeframe: 5 months

Interventionpercent predicted FEV1 (Mean)
Single Simvastatin Treatment Arm-2.9

[back to top]

Time to Resolution of Stomatitis From Grade 2 or Greater to Grade 1 or Less

The number of days to achieve resolution of stomatitis from grade 2 or greater to grade 1 or less was assessed. Grade 1 = minimal symptoms, normal diet; grade 2 = symptomatic, but able to swallow a modified diet; grade 3 = symptomatic and unable to aliment or hydrate orally; and grade 4 = symptoms associated with life-threatening consequences. (NCT02069093)
Timeframe: 56 days

InterventionDays (Median)
Dexamethasone Based MouthwashNA

[back to top]

Blood Concentration of Everolimus and Exemestane

Blood samples were collected and analyzed. (NCT02069093)
Timeframe: 28 days (pre-dose)

Interventionng/mL (Geometric Mean)
Everolimus (n=42)Exemestane (n=48)
Dexamethasone Based Mouthwash12.0470.657

[back to top]

Dose Intensity of Everolimus and Exemestane

The dose intensity was calculated as the cumulative dose of everolimus or exemestane divided by the length of time on treatment during the first 56 days of treatment. (NCT02069093)
Timeframe: 56 days

Interventionmg/day (Median)
EverolimusExemestane
Dexamethasone Based Mouthwash10.025.0

[back to top]

Number of Participants With All Grades of Stomatitis

The number of participants with all grades of stomatitis was defined as the number of participants who had stomatitis grade 1 or higher. Grade 1 = minimal symptoms, normal diet; grade 2 = symptomatic, but able to swallow a modified diet; grade 3 = symptomatic and unable to aliment or hydrate orally; and grade 4 = symptoms associated with life-threatening consequences. (NCT02069093)
Timeframe: 56 days

InterventionParticipants (Number)
Stomatitis all gradeNot evaluable
Dexamethasone Based Mouthwash181

[back to top]

Number of Participants With Stomatitis Grade ≥ 2

The incidence of grade ≥ 2 stomatitis was reported. Grade 1 = minimal symptoms, normal diet; grade 2 = symptomatic, but able to swallow a modified diet; grade 3 = symptomatic and unable to aliment or hydrate orally; and grade 4 = symptoms associated with life-threatening consequences. (NCT02069093)
Timeframe: 56 days

InterventionParticipants (Number)
Stomatitis grade >=2: YesStomatitis grade >=2: NoStomatitis grade >=2: Not evaluable
Dexamethasone Based Mouthwash2831

[back to top]

Median Number of Mouthwashes Per Day

The median number of mouthwashes per day was assessed. (NCT02069093)
Timeframe: 56 days

InterventionNumber of mouthwashes (Median)
Dexamethasone Based Mouthwash3.95

[back to top]

Percentage of Healthy Tissue Coverage That Was Greater Than 40 Micrometers

The secondary efficacy endpoint is mechanistic Optical coherence tomography (OCT) healthy level of intimal tissue coverage, determined by the OCT core laboratory at 1 year for subjects in Cohorts A and B. This reports the percentage of healthy tissue coverage that was great than 40 micrometers. (NCT02073565)
Timeframe: 1 year

InterventionHealthy Tissue Strut Coverage (>40 µm) % (Mean)
Combo91.27
Everolimus Eluting Stent (EES)74.82

[back to top]

Number of Patients Exhibiting Human Antimurine Antibody (HAMA) Reaction

Serum will be assessed for HAMA development at index, 30 days, and 12 months in Cohort B subjects. Human antimurine antibody plasma assessment will be with blood draws performed during index procedure, 30 day follow-up visit, and 1 year catheterizations. (NCT02073565)
Timeframe: Day of device implantation, 30 days, 12 months

,
InterventionParticipants (Count of Participants)
Baseline HAMA Responders30 day HAMA Responders1 Year HAMA Responders
Combo000
Everolimus Eluting Stent (EES)000

[back to top]

Number of Participants With Target Vessel Failure (TVF)

The primary clinical endpoint of Target Vessel Failure (TVF), defined as cardiac death, target-vessel myocardial infarction (MI), or ischemia-driven Target Vessel Revascularization(TVR) by percutaneous or surgical methods, at 1 year. (NCT02073565)
Timeframe: 1 year follow-up

InterventionParticipants (Count of Participants)
Combo20
Everolimus Eluting Stent (EES)12

[back to top]

Number of Patients With Clinically and Functionally Ischemia-Driven Target Lesion Revascularization (TLR)

Clinically and functionally ischemia-driven target lesion revascularization (TLR), including use of target-vessel Fractional Flow Reserve (FFR), analyzed dichotomously using the Fractional Flow Reserve (FFR) vs. Angiography in Multivessel Evaluation (FAME) study criteria of 0.8 during a 2 minute infusion of adenosine or adenosine triphosphate.34 Abnormal FFR-driven interventions at 1 year will be included in the evaluation of ischemia-driven TLR. (NCT02073565)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Combo12
Everolimus Eluting Stent (EES)8

[back to top]

Adverse Drug Reactions

Measure and compare the incidence of significant immunosuppressant-related adverse drug reactions for each group. (NCT02096107)
Timeframe: 2 year

InterventionParticipants (Count of Participants)
Low Intensity Tacrolimus11
Standard of Care6

[back to top]

Percentage of Participants Discontinuing or Modifying Immunosuppressant Use

Measure and compare the rates of immunosuppressant discontinuation and modification for each group. (NCT02096107)
Timeframe: 2 year

Interventionpercentage of participants (Number)
Low Intensity Tacrolimus37
Standard of Care20

[back to top]

Kidney Allograft Survival

Compare the patient and graft survival rates at one-year post-transplant in each group. (NCT02096107)
Timeframe: 1 year

Interventionpercentage of patients graft survival (Number)
Low Intensity Tacrolimus100
Standard of Care100

[back to top]

Infection

Compare the rates of post-transplant infections, including CMV, BK, and admissions to the hospital for infectious causes in each group. (NCT02096107)
Timeframe: 2 year

Interventionpercentage of subjects with infection (Number)
Low Intensity Tacrolimus17
Standard of Care30

[back to top]

Renal Function Measured by Estimated Glomerular Filtration Rate (eGFR)

Measure and compare the estimated GFR (using the 4-variable modified MDRD equation) in patients converted to everolimus, low intensity tacrolimus and prednisone versus the standard of care tacrolimus, mycophenolate and prednisone regimen, both at one year and two years post-transplant. (NCT02096107)
Timeframe: 1 year

InterventionmL/min/1.73 m^2 (Mean)
Low Intensity Tacrolimus59
Standard of Care56

[back to top]

Kidney Allograft Fibrosis Assessment

"Measure and compare the change in interstitial fibrosis (morphometric analysis of trichrome stained slides) at one-year post-transplant in patients converted to everolimus, low intensity tacrolimus and prednisone versus the standard of care tacrolimus, mycophenolate and prednisone regimen.~The scale is a percentage of the tissue that demonstrates fibrosis. The scale range is 0 to 100%. The higher the percentage, the worse the fibrosis and the poorer the prognosis. A lower score (percentage) is therefore better." (NCT02096107)
Timeframe: 1 year

,
Intervention% of fibrosis (Mean)
BaselineEnd of Follow-up
Low Intensity Tacrolimus22.922.9
Standard of Care20.827.8

[back to top]

In Stent: Mean Thickness of Strut Coverage at Follow up

Mean thickness of strut coverage at follow up (In-Stent safety endpoint). Struts have been considered as covered when tissue overlying the struts is >0 μm by optical coherence tomography (OCT) (NCT02098876)
Timeframe: 1 year

Interventionmm^2 (Mean)
Resolute Integrity DES0.118
Xience Xpedition DES0.096

[back to top]

In Stent Mean Cross-sectional Area of Neo-intimal Tissue Coverage

Cross-sectional area in neointimal hyperplasia by Optical Coherence Tomography (OCT) at 1 year following stent placement (NCT02098876)
Timeframe: 1 year

Interventionmm^2 (Mean)
Resolute Integrity DES1.381
Xience Xpedition DES0.949

[back to top]

Stent Edge -Change in Plaque Area (Efficacy Endpoint) at 5 mm Proximal and Distal to Stent.

Change in plaque area at the stent edges will be calculated from the change in plaque area in the 5 mm proximal and distal segments by intravascular ultrasound (IVUS); calculated as follow-up values minus baseline values (NCT02098876)
Timeframe: 1 year

Interventionmm^2 (Mean)
Resolute Integrity DES-0.8
Xience Xpedition DES-0.5

[back to top]

In Stent: Plaque Prolapse Post-PCI (In-Stent Mechanistic Endpoint)

Plaque will be identified by Optical Coherence Tomography (OCT)- In-Stent: Plaque prolapse post-PCI (In-Stent mechanistic endpoint) (NCT02098876)
Timeframe: Immediately after stent implantation

Interventionmm^2 (Mean)
Resolute Integrity DES0.12
Xience Xpedition DES0.13

[back to top]

Stent Edge: Percent Area With Low Wall Shear Stress (WSS) at Stent Edges Post-PCI (Mechanistic Endpoint)

The % area of low wall shear stress in the 5 mm proximal and distal segments immediately after stent implantation will be measured by intravascular ultrasound (IVUS). (NCT02098876)
Timeframe: Immediately after stent implantation

Interventionpercentage of area (Mean)
Resolute Integrity DES0.6
Xience Xpedition DES0.64

[back to top]

In-Stent: Degree of Vascular Straightening Post-percutaneous Coronary Intervention (PCI) (In-Stent Mechanistic Endpoint)

post PCI angulation by Angio: In-Stent: Degree of vascular straightening post-percutaneous coronary intervention (PCI) (In-Stent mechanistic endpoint) (NCT02098876)
Timeframe: Immediately after stent implantation

InterventionDegree (Mean)
Resolute Integrity DES40
Xience Xpedition DES36

[back to top]

In-Stent: Percent Area of Low Wall Shear Stress (WSS)-(In-Stent Mechanistic Endpoint)

The % area of low wall shear stress immediately after stent implantation will be measured by Optical Coherence Tomography (OCT) (NCT02098876)
Timeframe: Immediately after stent implantation

Interventionpercentage of area (Mean)
Resolute Integrity DES0.9
Xience Xpedition DES0.9

[back to top]

Stent Edge: Degree of Vascular Straightening Post-percutaneous Coronary Intervention (PCI) at the Stent Edges (Stent Edge Mechanistic Endpoint)

Degree of vascular straightening post-percutaneous coronary intervention (PCI) at the stent edges (Stent Edge mechanistic endpoint) will be measured post PCI angulation by Angio (NCT02098876)
Timeframe: Immediately after stent implantation

InterventionDegrees (Mean)
Resolute Integrity DES22
Xience Xpedition DES21

[back to top]

Renal Function Assessed by Estimated Glomerular Filtartion Rate (eGFR)

"Renal function was assessed by eGFR using the MDRD-4 formula at 12 months after transplant:~eGFR = 186.3 * (serum creatinine)-1.154 * age-0.203 * (0.742 for women) * (1.21 if African American) where serum creatinine was in mg/dL and age in years." (NCT02115113)
Timeframe: At 12 months post-transplant

InterventionmL/min/1.73m^2 (Least Squares Mean)
Group A (Tacrolimus Elimination Arm)85.50
Group B (Tacrolimus Minimization Arm)80.26

[back to top]

Percentage of Participants With Treated Biopsy Proven Acute Rejection (tBPAR) Acute Rejection (AR), Graft Loss (GL) or Death (D)

Participants were assessed for tBPAR, AR, GL or death. For all suspected rejection episodes, regardless of initiation of anti-rejection treatment, a liver biopsy was to be performed preferably within 24 hours, latest within 48 hours whenever clinically possible. A treated biopsy proven acute rejection was considered an episode of acute rejection when demonstrated by local pathology reading with a rejection activity index of at least 3 or greater of acute rejection index and when treated with anti-rejection therapy. The allograft was considered lost on the day the subject was re-transplanted or died due to liver failure. (NCT02115113)
Timeframe: At 12 and 18 months post-transplant

,
InterventionParticipants (Count of Participants)
tBPAR, 12 monthsAR, 12 monthsGL, 12 monthsDeath, 12 monthstBPAR, 18 monthsAR, 18 monthsGL, 18 monthsDeath, 18 months
Group A (Tacrolimus Elimination Arm)22012201
Group B (Tacrolimus Minimization Arm)11001101

[back to top]

Change From Baseline (Randomization) in Serum Creatinine at 12 Months Post-transplant

Blood samples were collected to assess serum creatinine. (NCT02115113)
Timeframe: baseline, 12 months post-transplant

Interventionmg/dL (Least Squares Mean)
Group A (Tacrolimus Elimination Arm)-0.11
Group B (Tacrolimus Minimization Arm)-0.05

[back to top]

Number of Subjects With Disease Progression Using the Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 (Phase IB)

Progression determined using response evaluation criteria in solid tumors (RECIST) version 1.1 or showed clinical progression. RECIST criteria for progression includes: At least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started (including the baseline scan if that is the smallest), and at least a 5mm increase or the appearance of new lesions. Rates and associated 95% confidence limits will be estimated. (NCT02120469)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Dose Level B110
Dose Level A18
Dose Level A23

[back to top]

Number of Participants With Grade 3 or Higher Toxicities (Phase IB)

Will be graded according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Rates and associated 95% exact Clopper and Pearson binomial confidence limits will be estimated for grade 3 or higher toxicities attributed to eribulin mesylate or everolimus. (NCT02120469)
Timeframe: On treatment, 21 days per cycle up to 2 years

InterventionParticipants (Count of Participants)
Dose Level B18
Dose Level A15
Dose Level A23

[back to top] [back to top]

Median Progression Free Survival (Phase IB)

Kaplan Meier methods will be used to estimate the median and 95% confidence limits over all dose levels per protocol plan. (NCT02120469)
Timeframe: Form the date treatment begins until the first date on which recurrence, progression, or death due to any cause, assessed up to 2 years

Interventionmonths (Median)
Treatment (Everolimus, Eribulin Mesylate)2.6

[back to top]

Median Overall Survival (Phase IB)

Kaplan Meier methods will be used to estimate the median and 95% confidence limits across all dose levels per protocol plan. (NCT02120469)
Timeframe: From start of treatment to death due to any cause, assessed up to 2 years

Interventionmonths (Median)
Treatment (Everolimus, Eribulin Mesylate)8.3

[back to top]

Absolute Values of Blood Pressure: Mean

Absolute (mean and median) values for SBP and DBP at 3, 6, 12 and 24 months posttransplant; (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionmmHg (Mean)
Diastolic Month 3Systolic Month 3Diastolic Month 6Systolic Month 6Diastolic Month 12Systolic Month 12Diastolic Month 24Systolic Month 24
Treatment A78.7134.277.4128.178.7131.078.1130.9
Treatment B77.7131.079.4133.080.1131.078.5131.7

[back to top]

Absolute Calculated Glomerular Filtration Rate (cGFR): Mean

Absolute (mean and median) cGFR values at 3, 6, 12 and 24 months post-transplant, as determined from the 4-variable Modification of Diet in Renal Disease (MDRD) formula (NCT02137239)
Timeframe: Up 24 Months post-transplant

,
InterventionmL/min/1.73 m^2 (Mean)
At 3 MonthsAt 6 MonthsAt 12 MonthsAt 24 Months
Treatment A69.266.066.271.8
Treatment B62.263.962.068.7

[back to top]

Time to Clinically-suspected Biopsy-proven Acute Rejection (CSBPAR).

Time to Clinically suspected biopsy proven acute rejection (NCT02137239)
Timeframe: Up to 24 Months

InterventionMonths (Mean)
Treatment ANA
Treatment BNA

[back to top]

Percentage of Participants With Serious Adverse Events (SAEs)

Percentage of participants with SAEs up to 24 months post-transplant (NCT02137239)
Timeframe: Up to 24 months Post-Transplant

InterventionPercentage of participants with SAEs (Number)
Treatment A52.0
Treatment B60.6

[back to top]

Percentage of Participants With Adverse Events (AEs)

Percentage of participants with AEs up to 24 months post-transplant (NCT02137239)
Timeframe: Up to 24 months Post-Transplant

InterventionPercentage of participants with AEs (Number)
Treatment A100.0
Treatment B97.0

[back to top]

Percentage of Particpants With Laboratory Test Abnormalities (LTAs)

Percentage of participants with laboratory tests with marked laboratory abnormalities (NCT02137239)
Timeframe: At 24 Months

,
InterventionPercentage of participants (Number)
Hemoglobin (Low)Leukocytes (low)Lymphocyte (Absolute) (low)Neutrophils (Absolute) (low)Aspartate Aminotransferase (High)Creatinine (High)Inorganic Phosphorus (low)Potassium (high)Sodium (low)Albumin (low)Glucose (high)Triglycerides (high)Uric Acid (high)
Treatment A12.0084.004.016.024.04.04.008.012.08.0
Treatment B6.13.069.73.003.012.1003.012.100

[back to top]

Urine Protein Creatinine Ratio (UPr/Cr)

Urine protein to creatinine ratio (UPr/Cr) at 3, 6, 12 and 24 months post-transplant. (NCT02137239)
Timeframe: Up 24 Months post-transplant

,
Interventionmg Protein/mg Creatinine (Mean)
At 3 MonthsAt 6 MonthsAt 12 MonthsAt 24 Months
Treatment A0.31460.38960.28350.3940
Treatment B0.14120.14610.18490.1685

[back to top]

Treatment Differences in Therapeutic Modalities

Treatment Received for Biopsy Proven Acute Rejection (Banff Grade IA or Higher), or Humoral (Antibody Mediated) Rejection Treatment regimen: Categorical analysis of CSBPAR episodes by treatment received. (NCT02137239)
Timeframe: at 6, 12 and 24 Months

,
InterventionPercentage of participants with CSBPARs (Number)
Corticosteroids (6 months)Lymphocyte depleting agent (6 months)Plasmapheresis (6 months)IVIG (6 months)Rituximab (6 months)Corticosteroids (12 months)Lymphocyte depleting agent (12 months)Plasmapheresis (12 months)IVIG (12 months)Rituximab (12 months)Corticosteroids (24 months)Lymphocyte depleting agent (24 months)Plasmapheresis (24 months)IVIG (24 months)Rituximab (24 months)
Treatment A7.7000015.43.800019.23.8000
Treatment B9.46.303.1012.56.303.1012.56.303.10

[back to top]

Time to Event: Graft Loss and Death

The Number of days to participant Graft Loss and death for any reason (NCT02137239)
Timeframe: Up to 728 Days

,
InterventionDays (Number)
Graft Loss
Treatment A107
Treatment B2

[back to top]

Percentage of Participants With New Onset Diabetes After Transplant

Percentage of participants with New Onset Diabetes After Transplantation (NODAT) at 6, 12, and 24 months post-transplant. (NCT02137239)
Timeframe: up to 24 months

,
InterventionPercentage of participants (Number)
Up to 6 MonthsUp to 12 MonthsUp to 24 Months
Treatment A11.511.515.4
Treatment B6.36.312.5

[back to top]

Percentage of Participants With Events of Special Interest (ESIs)

"Percentage of participants which have one of the following events of special interest:~Serious Infections Post-Transplant Lymphoproliferative Disorder (PTLD) Progressive multifocal leukoencephalopathy (PML) Malignancies (Other than PTLD) including non-melanoma skin carcinomas (Malignancies) Tuberculosis Infections Central Nervous System (CNS) Infections Viral Infections Infusion Related reactions within 24 hours since belatacept infusion" (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionPercentage of participants with ESIs (Number)
Serious InfectionsPTLDPMLMalignanciesTBCNS InfectionsViral InfectionsInfusion Related Reactions
Treatment A164.004.00001
Treatment B15.23.003.00000

[back to top]

Percentage of Participants With Donor Specific Anti-HLA Antibodies (DSA)

Percentage of participants with, and titers of pre-existing (pre-transplant) DSA on Day 1 (pre-transplant, pre-dose), and at Months 12 and 24 posttransplant (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionPercentage of Participants (Number)
Baseline Class 1 DSABaseline Class 2 DSABaseline Both Class 1 and 2 DSA12 Month Class 1 DSA12 Month Class 2 DSA12 Month Both Class 1 and 2 DSA24 Month Class 1 DSA24 Month Class 2 DSA24 Month Both Class 1 and 2 DSA
Treatment A10008.00008.0000
Treatment B00003.0303.033.030

[back to top]

Percentage of Participants With De Novo Donor Specific Anti-HLA Antibodies (DSA)

Characterization of any de novo DSA detected by IgM and IgG subclasses, and by the presence or absence of complement fixing properties. (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionPercentage (Number)
Baseline Class 1 DSABaseline Class 2 DSABaseline Both Class 1 and 2 DSADe Novo 12 Month Class 1 DSADe Novo 12 Month Class 2 DSADe Novo 12 Month Both Class 1 and 2 DSADe Novo 24 Month Class 1 DSADe Novo 24 Month Class 2 DSADe Novo 24 Month Both Class 1 and 2 DSA
Treatment A1000000000
Treatment B0000003.4500

[back to top]

Percentage of Participants With BANFF Grade by Severity Grades. BANFF Type (Grade) for Acute/Active Rejection

"Treatment differences in the severity grades to treat all episodes of CSBPAR at 6, 12, and 24 months post-transplant.~Type 1A - Cases with significant interstitial infiltration (>25% of parenchyma affected) and foci of moderate tubulitis (>4 mononuclear cells/Tubular cross section or group of 10 Tubular cell). Type 1B - Cases with significant interstitial infiltration (>25% of parenchyma affected) and foci of moderate tubulitis (>10 mononuclear cells/Tubular cross section or group of 10 Tubular cell).Type 2A - Cases with mild to moderate intimal arteritis.Type 2B - Cases with severe intimal arteritis comprising >25% of the luminal area. Type 3 - Cases with transmural arteritis and/or arterial fibrinoid change and necrosis of medial smooth muscle cells (v3 with accompanying lymphocytic inflammation)" (NCT02137239)
Timeframe: At 6, 12 and 24 Months

,
InterventionPercentage of Participants (Number)
6 Months: Mild Acute (1A)6 Months: Mild Acute (1B)6 Months: Moderate Acute (2A)6 Months: Moderate Acute (2B)6 Months: Severe Acute12 Months: Mild Acute (1A)12 Months: Mild Acute (1B)12 Months: Moderate Acute (2A)12 Months: Moderate Acute (2B)12 Months: Severe Acute24 Months: Mild Acute (1A)24 Months: Mild Acute (1B)24 Months: Moderate Acute (2A)24 Months: Moderate Acute (2B)24 Months: Severe Acute
Treatment A3.807.7007.707.70011.507.700
Treatment B03.16.3003.13.16.3003.13.16.300

[back to top]

Percentage of Clinically-suspected Biopsy-proven Acute Rejection (CSBPAR) at 6 Months

Number of Participants with Clinically-suspected biopsy-proven acute rejection (CSBPAR) at 6 Months (NCT02137239)
Timeframe: 6 Months

InterventionPercentage of participants (Number)
Treatment A7.7
Treatment B9.4

[back to top]

Number of Participants Who Survive With a Functioning Graft

Number of all participants who survive with a functioning graft at 6, 12 and 24 months post transplant (NCT02137239)
Timeframe: At 6, 12 and 24 months

,
InterventionParticipants (Count of Participants)
At 6 MonthsAt 12 MonthsAt 24 Months
Treatment A252525
Treatment B313131

[back to top]

Number of Participants Who Experience Graft Loss Post Transplant

Number of all participants who experience graft loss at 6, 12 and 24 months post transplant (NCT02137239)
Timeframe: At 6, 12 and 24 months

,
InterventionParticipants (Count of Participants)
At 6 MonthsAt 12 MonthsAt 24 Months
Treatment A111
Treatment B111

[back to top]

Number of Participants Deaths Post Transplant

Number of participant deaths at 6, 12 and 24 months post transplant (NCT02137239)
Timeframe: up to 24 months

,
InterventionParticipants (Count of Participants)
At 6 MonthsAt 12 MonthsAt 24 Months
Treatment A000
Treatment B000

[back to top]

Median Calculated Glomerular Filtration Rate (cGFR)

Median cGFR values at 3, 6, 12 and 24 months post-transplant, as determined from the 4-variable Modification of Diet in Renal Disease (MDRD) formula (NCT02137239)
Timeframe: Up 24 Months post-transplant

,
InterventionmL/min/1.73 m^2 (Median)
At 3 MonthsAt 6 MonthsAt 12 MonthsAt 24 Months
Treatment A64.064.066.073.5
Treatment B62.067.062.568.0

[back to top]

Mean Changes From Baseline Values of Lipid Values

"Mean changes from baseline values in the following:~Serum total cholesterol (TC) Serum high density lipoprotein (HDL) cholesterol Serum low density lipoprotein (LDL) cholesterol Serum triglycerides (TG)" (NCT02137239)
Timeframe: at months 12 and 24

,
Interventionmg/dL (Mean)
TC Month 12TC Month 24HDL Month 12HDL Month 24LDL Month 12LDL Month 24TG Month 12TG Month 24
Treatment A25.726.65.46.225.717.43.3106.8
Treatment B-2.810.01.94.810.815.7-86.1-13.6

[back to top]

Mean Changes From Baseline Values for Blood Pressure

Mean changes from baseline values for SBP and DBP at 6, 12 and 24 months post-transplant (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionmmHg (Mean)
Diastolic Month 6Systolic Month 6Diastolic Month 12Systolic Month 12Diastolic Month 24Systolic Month 24
Treatment A1.0-4.02.3-1.10.9-2.3
Treatment B4.8-0.75.4-3.22.1-4.2

[back to top]

Mean Change From Month 3 in cGFR

The mean change from Month 3 cGFR at 3, 6, 12 and 24 months post-transplant (NCT02137239)
Timeframe: Up 24 Months post-transplant

,
InterventionmL/min/1.73 m^2 (Mean)
At 3 MonthsAt 6 MonthsAt 12 MonthsAt 24 Months
Treatment A0-3.2-3.13.1
Treatment B02.81.46.3

[back to top]

Mean and Mean Change From Baseline in Whole Blood HbA1c

Mean whole blood HbA1C concentrations, and mean changes from baseline values at Months 6, 12 and 24 months post-transplant. (NCT02137239)
Timeframe: Up to 24 months

,
Interventionmg/dL (Mean)
Mean Value at 6 monthsChange from baseline at 6 monthsMean Value at 12 monthsChange from baseline at 12 monthsMean Value at 24 monthsChange from baseline at 24 months
Treatment A6.110.346.180.476.240.66
Treatment B6.130.486.210.326.290.41

[back to top]

Mean and Mean Change From Baseline in Blood Glucose

Mean fasting blood glucose levels, and mean changes from baseline values at Months 6, 12 and 24 months post- transplant (NCT02137239)
Timeframe: Up to 24 months

,
Interventionmg/dL (Mean)
Mean Value at 6 monthsChange from baseline at 6 monthsMean Value at 12 monthsChange from baseline at 12 monthsMean Value at 24 monthsChange from baseline at 24 months
Treatment A107.24.9101.1-1.3127.522.3
Treatment B107.24.8127.520.6111.815.0

[back to top]

Clinically-suspected Biopsy-proven Acute Rejection (CSBPAR) at 6, 12 and 24 Months

"Clinically-suspected biopsy-proven acute rejection (CSBPAR) at 6, 12 and 24 Months~Change in the incidence of CSBPAR at 6, 12 and 24 months post transplant, in the belatacept + EVL(Treatment A) as compared to TAC + MMF (Treatment B)." (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionPercentage of CSBPAR (Number)
CSBPAR at 6 MonthsCSBPAR at 12 monthsCSBPAR at 24 Months
Treatment A7.711.515.4
Treatment B9.412.512.5

[back to top]

Absolute Values of Fasting Lipid Values: Median

"Absolute (mean and median) values at 3, 6, 12 and 24 months post-transplant for the following:~Serum total cholesterol (TC) Serum high density lipoprotein (HDL) cholesterol Serum low density lipoprotein (LDL) cholesterol Serum triglycerides (TG)" (NCT02137239)
Timeframe: Up to 24 Months

,
Interventionmg/dL (Median)
TC Month 3TC Month 6TC Month 12TC Month 24HDL Month 3HDL Month 6HDL Month 12HDL Month 24LDL Month 3LDL Month 6LDL Month 12LDL Month 24TG Month 3TG Month 6TG Month 12TG Month 24
Treatment A167.0187.0184.0193.045.045.550.049.089.099.5104.0103.5147.0157.5154.0159.0
Treatment B173.0178.0171.5166.049.049.047.049.095.0100.091.586.0128.0126.0130.0114.0

[back to top]

Absolute Values of Fasting Lipid Values: Mean

"Absolute (mean and median) values at 3, 6, 12 and 24 months post-transplant for the following:~Serum total cholesterol (TC) Serum high density lipoprotein (HDL) cholesterol Serum low density lipoprotein (LDL) cholesterol Serum triglycerides (TG)" (NCT02137239)
Timeframe: Up to 24 Months

,
Interventionmg/dL (Mean)
TC Month 3TC Month 6TC Month 12TC Month 24HDL Month 3HDL Month 6HDL Month 12HDL Month 24LDL Month 3LDL Month 6LDL Month 12LDL Month 24TG Month 3TG Month 6TG Month 12TG Month 24
Treatment A181.2197.7189.0193.250.646.449.450.196.9115.2107.597.9171.6180.0162.4263.4
Treatment B174.4175169.9168.250.453.949.651.396.593.788.091.5137.8138.3161.3145.0

[back to top]

Absolute Values of Blood Pressure: Median

Absolute (mean and median) values for SBP and DBP at 3, 6, 12 and 24 months posttransplant; (NCT02137239)
Timeframe: Up to 24 Months

,
InterventionmmHg (Median)
Diastolic Month 3Systolic Month 3Diastolic Month 6Systolic Month 6Diastolic Month 12Systolic Month 12Diastolic Month 24Systolic Month 24
Treatment A78.5135.575.5127.077.0130.078.0130.0
Treatment B80.0131.080.0131.081.0126.079.0130.0

[back to top]

Progression-free Survival (Fulvestrant Versus Fulvestrant + Everolimus + Anastrozole)

From date of registration to date of first documentation of progression or death due to any cause. Participants last known to be alive without report of progression are censored at date of last contact. (NCT02137837)
Timeframe: up to 5 years

Interventionmonths (Median)
Arm 1: Fulvestrant + Everolimus Placebo + Anastrozole Placebo14.0
Arm 3: Fulvestrant + Everolimus + Anastrozole9.9

[back to top]

Progression Free Survival (Fulvestrant + Everolimus vs Fulvestrant + Everolimus + Anastrozole)

From date of registration to date of first documentation of progression or death due to any cause. Participants last known to be alive without report of progression are censored at date of last contact. (NCT02137837)
Timeframe: up to 5 years

Interventionmonths (Median)
Arm 2: Fulvestrant + Everolimus + Anastrozole Placebo11.3
Arm 3: Fulvestrant + Everolimus + Anastrozole9.90

[back to top]

Overall Survival

From date of registration to date of death due to any cause. Participants last known to be alive without report of progression are censored at date of last contact. (NCT02137837)
Timeframe: up to 5 years

Interventionmonths (Median)
Arm 1: Fulvestrant + Everolimus Placebo + Anastrozole Placebo46.8
Arm 2: Fulvestrant + Everolimus + Anastrozole Placebo56.6
Arm 3: Fulvestrant + Everolimus + Anastrozole33.6

[back to top]

Molecular Determinants of Response in Circulating Tumor Cells: CTC-ETI

CTC-Endocrine Therapy Index (CTC-ETI) on the CellSearch® platform. Based on enumeration of CTC/7.5 mL of whole blood, with >= 5 being elevated. (Due to limited samples collected, full analysis was not able to be performed as planned, so outcome measure reported here is number with elevated Day 1 CTC.) (NCT02137837)
Timeframe: Day 1, Day 29, time of progression (Day 29 to be collected only if Day 1 CTC was elevated.)

InterventionParticipants (Count of Participants)
Arm 1: Fulvestrant + Everolimus Placebo + Anastrozole Placebo1
Arm 2: Fulvestrant + Everolimus + Anastrozole Placebo1
Arm 3: Fulvestrant + Everolimus + Anastrozole0

[back to top]

Clinical Benefit Rate

Proportion of participants who have confirmed and unconfirmed partial response, complete response or stable disease. (NCT02137837)
Timeframe: assessed every 12 weeks, up to 5 years

Interventionpercentage of participants (Number)
Arm 1: Fulvestrant + Everolimus Placebo + Anastrozole Placebo69
Arm 2: Fulvestrant + Everolimus + Anastrozole Placebo83
Arm 3: Fulvestrant + Everolimus + Anastrozole67

[back to top] [back to top]

Response Rate

Proportion of participants who have confirmed or unconfirmed partial or complete response to therapy (NCT02137837)
Timeframe: assessed every 12 weeks, up to 5 years

Interventionpercentage of participants (Number)
Arm 1: Fulvestrant + Everolimus Placebo + Anastrozole Placebo22
Arm 2: Fulvestrant + Everolimus + Anastrozole Placebo60
Arm 3: Fulvestrant + Everolimus + Anastrozole44

[back to top]

Progression-Free Survival (Fulvestrant vs Fulvestrant + Everolimus )

From date of registration to date of first documentation of progression or death due to any cause. Participants last known to be alive are censored at date of last contact. (NCT02137837)
Timeframe: up to 5 years

Interventionmonths (Median)
Arm 1: Fulvestrant + Everolimus Placebo + Anastrozole Placebo14.0
Arm 2: Fulvestrant + Everolimus + Anastrozole Placebo11.3

[back to top]

Progression Free Survival

Progression Free Survival (PFS) was defined as the time from randomization to the first of either disease progression or death from any cause. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT02143726)
Timeframe: 4 years and 4 months

InterventionMonths (Median)
Arm 1 (Sorafenib)9.4
Arm 2 (Sorafenib and Everolimus)24.7

[back to top]

Overall Survival

(NCT02143726)
Timeframe: 5 years

InterventionMonths (Median)
Arm 1 (Sorafenib)NA
Arm 2 (Sorafenib and Everolimus)40.1

[back to top]

Confirmed Response Rate

A patient will be classified as a confirmed response per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT02143726)
Timeframe: 4 years 4 months

InterventionProportion of participants (Number)
Arm 1 (Sorafenib)0.188
Arm 2 (Sorafenib and Everolimus)0.235

[back to top]

Number of Participants With Grade 3 or Higher Adverse Events

The maximum grade for each type of adverse event will be summarized using CTCAE version 4.0. The frequency and percentage of grade 3+ adverse events will be compared between the 2 treatment arms. (NCT02143726)
Timeframe: 4 years 3 months

InterventionParticipants (Count of Participants)
Arm 1 (Sorafenib)10
Arm 2 (Sorafenib and Everolimus)12

[back to top]

Percent of Participants With Grade 3 or Higher Infectious Adverse Event(s)

"The severity of infectious adverse events (AEs) was classified into grades as follows:~Grade 1 = asymptomatic; clinical or diagnostic observation only; intervention with oral antibiotic, antifungal, or antiviral agent only; no invasive intervention required~Grade 2 = symptomatic; intervention with intravenous antibiotic, antifungal, or antiviral agent; invasive intervention may be required~Grade 3 = any infection associated with hemodynamic compromise requiring pressors; any infection necessitating intensive care unit level of care; any infection necessitating operative intervention; any infection involving the central nervous system; any infection with a positive fungal blood culture; any proven or probable aspergillus infection; any tissue invasive fungal infection; any pneumocystis jiroveci infection~Grade 4 = life-threatening infection~Grade 5 = death resulting from infection" (NCT02188719)
Timeframe: Transplantation to 40 Weeks Post Transplantation

InterventionPercent of Participants (Number)
Cohort 10
Cohort 211.1

[back to top]

Percent of Participants With Adverse Events (AEs) Attributable to the Donor Alloantigen Reactive Tregs (darTregs) Infusion

"AEs classified by the site investigator/clinician as possibly or definitely related to the study treatment, the Donor Alloantigen Reactive Tregs (darTregs) infusion. These AEs include:~infusion reaction~Grade 3 or higher cytokine release syndrome (Reference: National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.0 (4/28/2009) grading criteria~malignant cellular transformation." (NCT02188719)
Timeframe: Transplantation to 40 Weeks Post Transplantation

InterventionPercent of Participants (Number)
Cohort 20

[back to top]

Percent of Participants With Grade 2 or Higher Hematologic Adverse Events (AEs) of Anemia, Neutropenia, and/or Thrombocytopenia

"The severity of adverse events (AEs) was classified into grades using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.0 (4/28/2009):~Grade 1 = mild AE~Grade 2 = moderate AE~Grade 3 = severe and undesirable AE~Grade 4 = life-threatening or disabling AE~Grade 5 = death" (NCT02188719)
Timeframe: Transplantation to 40 Weeks Post Transplantation

,
InterventionPercent of Participants (Number)
AnemiaNeutropeniaThrombocytopenia
Cohort 166.716.716.7
Cohort 222.200

[back to top]

Percent of Participants With Biopsy-Proven Acute and/or Chronic Rejection

"Biopsy-proven acute rejection graded as Mild, Moderate or Severe, per 1997 Banff classification. Chronic Rejection graded using Banff 2000 classification.~References: 1.) Banff Schema for Grading Liver Allograft Rejection: An International Consensus Document developed by an international panel of experts in liver transplantation pathology, hepatology, and surgery (Hepatology 1997; 25(3): 658-663). 2.) Update of the International Banff Schema for Liver Allograft Rejection: Working Recommendations for the Histopathologic Staging and Reporting of Chronic Rejection (Hepatology 2000; 31(3): 792-799)." (NCT02188719)
Timeframe: Transplantation to 40 Weeks Post Transplantation

,
InterventionPercent of Participants (Number)
Mild Acute RejectionModerate Acute RejectionSevere Acute RejectionChronic Rejection
Cohort 10000
Cohort 211.1000

[back to top]

Percent of Participants With Grade 3 or Higher Wound Complication(s) Adverse Event(s)

"The severity of adverse events (AEs) was classified into grades using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.0 (4/28/2009):~Grade 3 wound complications are defined as Hernia without evidence of strangulation; fascial disruption/dehiscence; primary wound closure or revision by operative intervention indicated~Grade 4 complications are defined as Hernia with evidence of strangulation; major reconstruction flap, grafting, resection, or amputation indicated" (NCT02188719)
Timeframe: Transplantation to 40 Weeks Post Transplantation

InterventionPercent of Participants (Number)
Cohort 10
Cohort 20

[back to top]

Progression-free Survival

Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT02201212)
Timeframe: Baseline, Up to 2 Years

Interventionmonths (Median)
Everolimus2.0

[back to top]

Duration of Response

Duration of Response Rate (NCT02201212)
Timeframe: Baseline, Every 8 weeks, 2 Years

Interventionmonths (Median)
Everolimus12.7

[back to top]

Toxicity Rate

CTCAE v4.0 Toxicity Rate Grade 3 or higher (NCT02201212)
Timeframe: 2 Years

InterventionParticipants (Count of Participants)
Everolimus3

[back to top]

Objective Response Rate

"RECIST 1.1 criteria for Objective Response:~Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by scan: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR" (NCT02201212)
Timeframe: Baseline, Every 8 weeks, 2 Years

InterventionParticipants (Count of Participants)
Everolimus2

[back to top]

Overall Survival

Overall Survival Rate (NCT02201212)
Timeframe: 4 Years

Interventionmonths (Median)
Everolimus7.27

[back to top]

Median Survival

Survival is defined as the duration alive from study entry until death. (NCT02228681)
Timeframe: Following treatment discontinuation, patients are followed quarterly for 2 years, semi-annually for 3 more years, annually thereafter.

InterventionMonths (Median)
Everolimus and LetrozoleNA
Tamoxifen and Medroxyprogesterone Acetate16.6

[back to top]

Frequency of Response

"A confirmed complete or partial response as defined by RECIST 1.1 was considered a response. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR" (NCT02228681)
Timeframe: From date of randomization until the date of first documented progression or date of death , up to 3 years.

InterventionNumber of participants (Number)
Everolimus and Letrozole9
Tamoxifen and Medroxyprogesterone Acetate8

[back to top]

Frequency and Severity of CTCAE (Common Toxicity Criteria for Adverse Events) Version 4

Maximum grade of physician assessed adverse events graded and categorized using Common Toxicity Criteria for Adverse Events (CTCAE) version 4 (NCT02228681)
Timeframe: Assessed throughout the treatment period and for 30 days after discontinuation of treatment. Treatment continues until progression of disease.

,
InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4Grade 5
Everolimus and Letrozole262720
Tamoxifen and Medroxyprogesterone Acetate2141721

[back to top]

Median Progression-free Survival

Progression-free Survival is defined as the duration alive from study entry until progression is documented, or death; whichever comes sooner. Progressive disease is defined as at least a 5 mm absolute increase and a 20% relative increase in the sum of measurable target lesions' longest dimensions relative to the smallest sum at baseline or on study or the appearance of new lesions or unequivocal progression of existing non-target lesions. (NCT02228681)
Timeframe: Tumor measurements were done at 8 and 16 weeks after initiating treatment and then every 12 weeks and compared with baseline measurements prior to treatment. Measurements are continued until disease progression is documented or death.

Interventionmonths (Median)
Everolimus and Letrozole6.4
Tamoxifen and Medroxyprogesterone Acetate3.7

[back to top]

PEPI Score

To obtain the PEPI score, risk points for relapse-free survival (RFS) are assigned depending on the hazard ratio (HR) from the multivariable analysis. The total PEPI score assigned to each patient is the sum of the risk points derived from the pT stage, pN stage, Ki67 level, and estrogen receptor status of the surgical specimen. A HR in the range of 1 to 2 receives one risk point; a HR in the 2 to 2.5 range, two risk points; a HR greater than 2.5, three risk points. The total risk point score for each patient is the sum of all the risk points accumulated from the four factors in the model, ranges from 0 (best possible outcome) to 12 (worst possible outcome). (NCT02236572)
Timeframe: up to 26 weeks

Interventionunits on a scale (Median)
Aromatase Inhibitor Plus Everolimus3

[back to top]

Participant Ability to Tolerate Study Treatment With Minimal Side Effects

Adverse events (using CTCAE (4.0)) that begin or worsen after informed consent were recorded. Adverse events are itemized in the adverse events module (in terms of total events and total participants experiencing events). Presented are the total number of participants that experience adverse events. For clarification, counts of participants that experienced serious adverse events and counts of participants that experienced 'other' (not serious) adverse events are presented separately. (NCT02236572)
Timeframe: Up to 26 weeks

InterventionParticipants (Count of Participants)
Participants with Serious Adverse EventsParticipants with Other (Not Including Serious) Adverse Events
Aromatase Inhibitor Plus Everolimus517

[back to top]

Symptomatic Skeletal Event-free Survival (SSE-FS)

Time from date of randomization to occurrence of one of the following, whichever happened earlier: 1) an on study SSE, which was defined as the use of external beam radiotherapy (EBRT) to relieve skeletal symptoms, the occurrence of new symptomatic pathological bone fractures (vertebral or nonvertebral), the occurrence of spinal cord compression, a tumor related orthopedic surgical intervention; or 2) death from any cause. Per Protocol Amendment 10, following primary analysis completion, further assessments were focused on safety, and only limited efficacy data including SSE and survival were collected and not designed to support reconsideration of the primary analysis efficacy conclusions. Accordingly, no formal statistical analyses were performed for primary and secondary efficacy outcomes in the final analysis. All primary and secondary efficacy outcome measures presented in this document came from the primary completion analysis. (NCT02258451)
Timeframe: Up to 55 months

InterventionMonths (Median)
Radium-223 + EXE/EVE21.1
Placebo + EXE/EVE19.9

[back to top]

Number of Participants With Hematological Toxicities: Worst Grade Under Treatment (From First Dosing Till Primary Analysis)

(NCT02258451)
Timeframe: From first dosing till primary analysis cutoff date, up to 55 months

InterventionParticipants (Count of Participants)
Anemia72094292Anemia72094293Leukocytosis72094292Leukocytosis72094293Hemoglobin increased72094292Hemoglobin increased72094293Lymphocyte count decreased72094292Lymphocyte count decreased72094293Lymphocyte count increased72094293Lymphocyte count increased72094292Neutrophil count decreased72094293Neutrophil count decreased72094292Platelet count decreased72094293Platelet count decreased72094292White blood cell decreased72094293White blood cell decreased72094292
NormalGrade 1Grade 2Grade 3Grade 4
Radium-223 + EXE/EVE61
Placebo + EXE/EVE57
Radium-223 + EXE/EVE43
Placebo + EXE/EVE55
Radium-223 + EXE/EVE21
Placebo + EXE/EVE11
Radium-223 + EXE/EVE14
Placebo + EXE/EVE14
Radium-223 + EXE/EVE139
Placebo + EXE/EVE137
Radium-223 + EXE/EVE0
Radium-223 + EXE/EVE1
Radium-223 + EXE/EVE138
Radium-223 + EXE/EVE20
Placebo + EXE/EVE33
Radium-223 + EXE/EVE63
Placebo + EXE/EVE45
Placebo + EXE/EVE24
Radium-223 + EXE/EVE11
Placebo + EXE/EVE35
Placebo + EXE/EVE0
Radium-223 + EXE/EVE2
Placebo + EXE/EVE2
Radium-223 + EXE/EVE137
Placebo + EXE/EVE135
Radium-223 + EXE/EVE24
Placebo + EXE/EVE29
Radium-223 + EXE/EVE47
Placebo + EXE/EVE28
Radium-223 + EXE/EVE19
Radium-223 + EXE/EVE49
Placebo + EXE/EVE78
Radium-223 + EXE/EVE60
Placebo + EXE/EVE61
Radium-223 + EXE/EVE7
Radium-223 + EXE/EVE64
Placebo + EXE/EVE74
Radium-223 + EXE/EVE37
Placebo + EXE/EVE47
Radium-223 + EXE/EVE59
Placebo + EXE/EVE36
Radium-223 + EXE/EVE17
Placebo + EXE/EVE3
Placebo + EXE/EVE1
Radium-223 + EXE/EVE25
Placebo + EXE/EVE50

[back to top]

Number of Participants With Hematological Toxicities: Worst Grade Under Treatment

(NCT02258451)
Timeframe: From first dosing up to 30 days after the last administration of study treatments, up to 72.6 months

InterventionParticipants (Count of Participants)
Anemia72094292Anemia72094293Leukocytosis72094293Leukocytosis72094292Hemoglobin increased72094292Hemoglobin increased72094293Lymphocyte count decreased72094293Lymphocyte count decreased72094292Lymphocyte count increased72094293Lymphocyte count increased72094292Neutrophil count decreased72094293Neutrophil count decreased72094292Platelet count decreased72094293Platelet count decreased72094292White blood cell decreased72094293White blood cell decreased72094292
Grade 3NormalGrade 1Grade 2Grade 4
Radium-223 + EXE/EVE61
Placebo + EXE/EVE57
Radium-223 + EXE/EVE43
Placebo + EXE/EVE55
Radium-223 + EXE/EVE21
Placebo + EXE/EVE11
Radium-223 + EXE/EVE14
Placebo + EXE/EVE14
Radium-223 + EXE/EVE139
Placebo + EXE/EVE0
Radium-223 + EXE/EVE138
Placebo + EXE/EVE137
Radium-223 + EXE/EVE20
Placebo + EXE/EVE33
Radium-223 + EXE/EVE63
Placebo + EXE/EVE45
Placebo + EXE/EVE24
Radium-223 + EXE/EVE2
Radium-223 + EXE/EVE11
Placebo + EXE/EVE35
Placebo + EXE/EVE2
Radium-223 + EXE/EVE137
Placebo + EXE/EVE135
Radium-223 + EXE/EVE23
Placebo + EXE/EVE30
Radium-223 + EXE/EVE48
Placebo + EXE/EVE28
Radium-223 + EXE/EVE19
Radium-223 + EXE/EVE0
Radium-223 + EXE/EVE49
Placebo + EXE/EVE77
Radium-223 + EXE/EVE60
Placebo + EXE/EVE60
Radium-223 + EXE/EVE7
Radium-223 + EXE/EVE64
Placebo + EXE/EVE74
Radium-223 + EXE/EVE37
Placebo + EXE/EVE48
Radium-223 + EXE/EVE59
Placebo + EXE/EVE37
Radium-223 + EXE/EVE17
Placebo + EXE/EVE3
Radium-223 + EXE/EVE1
Placebo + EXE/EVE1
Radium-223 + EXE/EVE25

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs) (From First Dosing Till Primary Analysis)

An AE was any untoward medical occurrence (i.e. any unfavorable and unintended sign [including abnormal laboratory findings], symptom or disease) in a patient or clinical investigation participant after providing written informed consent for participation in the study. AEs were considered to be treatment-emergent if they started or worsened after first application of study intervention up to 30 days after end of treatment with study intervention. A serious adverse event (SAE) was an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening; persistent or significant disability/incapacity; congenital anomaly; another medical important serious event as judged by the investigator and an occurrence of any additional malignancies, including acute myelocytic leukemia or hematological conditions. (NCT02258451)
Timeframe: From first dosing till primary analysis cutoff date, up to 55 months

,
InterventionParticipants (Count of Participants)
Any TEAESerious TEAERadium-223/Placebo-related TEAEsExemestane-related TEAEsEverolimus-related TEAEs
Placebo + EXE/EVE136535064125
Radium-223 + EXE/EVE139577375130

[back to top]

Radiological Progression-free Survival (rPFS)

Time from the date of randomization to the date of confirmed radiological progression in either soft tissue, viscera or bone, or death (if death occurs before progression). Progression is defined using the modified RECIST 1.1 criteria (the modification refers to bone lesions assessment). Progression is defined as a 20% increase in the sum of the longest diameter of target lesions, or an unequivocal increase in non-target lesions, or the appearance of new lesions. All bone lesions are considered non-measurable and new bone lesions identified by bone scan should be confirmed by further imaging (CT/MRI). If a new bone lesion or unequivocal increase in size of bone lesions is only visible on a CT/MRI and not visible on a technetium-99m bone scan, progression should be declared without further confirmation. (NCT02258451)
Timeframe: Up to 55 months

InterventionMonths (Median)
Radium-223 + EXE/EVE7.9
Placebo + EXE/EVE6.7

[back to top]

Number of Participants With Post-treatment Chemotherapy Related Adverse Events (From First Dosing Till Primary Analysis)

(NCT02258451)
Timeframe: From post-treatment till primary analysis cutoff date, up to 55 months

,
InterventionParticipants (Count of Participants)
All system organ classes_Any_Grade 3All system organ classes_Any_Grade 4Blood and lymphatic system disorders_Any_Grade 3Blood and lymphatic system disorders_Febrile neutropenia_Grade 3Investigations_Any_Grade 4Investigations_Neutrophil count decreased_Grade 4
Placebo + EXE/EVE000000
Radium-223 + EXE/EVE111111

[back to top]

Number of Participants With Post-treatment Chemotherapy Related Adverse Events

According to protocol amendment 10, all participants who completed the EOT visit will be transferred to a separate extended safety follow-up study for their remaining follow-up. Thus, no further post-treatment data were collected after protocol amendment 10. (NCT02258451)
Timeframe: From post-treatment till end of study, up to 45.8 months

,
InterventionParticipants (Count of Participants)
All system organ classes_Any_Grade 3All system organ classes_Any_Grade 4Blood and lymphatic system disorders_Any_Grade 3Blood and lymphatic system disorders_Febrile neutropenia_Grade 3Investigations_Any_Grade 4Investigations_Neutrophil count decreased_Grade 4
Placebo + EXE/EVE000000
Radium-223 + EXE/EVE111111

[back to top]

Number of Participants With New Primary Malignancies

(NCT02258451)
Timeframe: From first dosing till end of study, up to 72.6 months

InterventionParticipants (Count of Participants)
Radium-223 + EXE/EVE1
Placebo + EXE/EVE0

[back to top]

Number of Participants With New Primary Malignancies During Study Treatment Till Primary Analysis

(NCT02258451)
Timeframe: From first dosing till primary analysis cutoff date, up to 55 months

InterventionParticipants (Count of Participants)
Radium-223 + EXE/EVE1
Placebo + EXE/EVE0

[back to top]

Time to Cytotoxic Chemotherapy

Time from the date of randomization to the date of the first cytotoxic chemotherapy (NCT02258451)
Timeframe: Up to 55 months

InterventionMonths (Median)
Radium-223 + EXE/EVE13.7
Placebo + EXE/EVE11.6

[back to top]

Time to Opiate Use for Cancer Pain

Interval from the date of randomization to the date of opiate use (NCT02258451)
Timeframe: Up to 55 months

InterventionMonths (Median)
Radium-223 + EXE/EVE21.4
Placebo + EXE/EVE18.4

[back to top]

Overall Survival

The time from the date of randomization to the date of death due to any cause. Per Protocol Amendment 10, following primary analysis completion, further assessments were focused on safety, and only limited efficacy data including SSE and survival were collected and not designed to support reconsideration of the primary analysis efficacy conclusions. Accordingly, no formal statistical analyses were performed for primary and secondary efficacy outcomes in the final analysis. All primary and secondary efficacy outcome measures presented in this document came from the primary completion analysis. (NCT02258451)
Timeframe: Up to 55 months

InterventionMonths (Median)
Radium-223 + EXE/EVE25.0
Placebo + EXE/EVE26.4

[back to top]

Percentage of Participants With Pain Improvement

In the percentage of participants with confirmed pain improvement. Confirmed pain improvement is defined a 2-point decrease or more in BPI-SF WPS from baseline over 2 consecutive measurements conducted at least 4 weeks apart, without an increase in pain management (IPM). An IPM is defined as the initiation of any opioid in participants not taking opioids at baseline, the initiation of a strong opioid in participants taking a weak opioid at baseline, or the initiation of an additional strong opioid in participants taking a strong opioid at baseline. (NCT02258451)
Timeframe: Up to 55 months

Interventionpercentage of participants (Number)
Radium-223 + EXE/EVE38.5
Placebo + EXE/EVE34.4

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs)

An AE was any untoward medical occurrence (i.e. any unfavorable and unintended sign [including abnormal laboratory findings], symptom or disease) in a patient or clinical investigation participant after providing written informed consent for participation in the study. AEs were considered to be treatment-emergent if they started or worsened after first application of study intervention up to 30 days after end of treatment with study intervention. A serious adverse event (SAE) was an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening; persistent or significant disability/incapacity; congenital anomaly; another medical important serious event as judged by the investigator and an occurrence of any additional malignancies, including acute myelocytic leukemia or hematological conditions. (NCT02258451)
Timeframe: From first dosing up to 30 days after the last administration of study treatments, up to 72.6 months

,
InterventionParticipants (Count of Participants)
Any TEAESerious TEAERadium-223/Placebo-related TEAEsExemestane-related TEAEsEverolimus-related TEAEs
Placebo + EXE/EVE136555064125
Radium-223 + EXE/EVE139577375130

[back to top]

Time to Pain Progression

"Time from randomization to the first date a participant experienced pain progression based on WPS. Pain progression was defined as an increase of 2 or more points in the BPI-SF Worst pain in 24 hours score from baseline observed at 2 consecutive evaluations ≥4 weeks apart or an increase in pain management (IPM) with respect to baseline, whichever occurred first. An IPM is defined as the initiation of any opioid in participants not taking opioids at baseline, the initiation of a strong opioid in participants taking a weak opioid at baseline, or the initiation of an additional strong opioid in participants taking a strong opioid at baseline." (NCT02258451)
Timeframe: Up to 55 months

InterventionMonths (Median)
Radium-223 + EXE/EVE7.6
Placebo + EXE/EVE5.7

[back to top]

Mean of Number of Tumor Assessment Visits - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

(NCT02264665)
Timeframe: During 2 years of prospective follow-up

InterventionVisits (Mean)
Targeted Therapy at Inclusion5.8
Other Treatments at Inclusion5.5

[back to top]

Number of Participants With Adverse Events Leading to Death - Based on Treatment Received At-least Once During the Study

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. Adverse events leading to death are reported in this outcome measure. Participants in this outcome measure were grouped according to type of treatment received at least once during study irrespective of treatment they initiated or were receiving at time of inclusion. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

InterventionParticipants (Count of Participants)
Everolimus During Study3
Sunitinib During Study2
Chemotherapy During Study11
Somatostatin Analogues During Study at Inclusion12
Metabolic Radiotherapy During Study2
Other Than Study Treatments During Study1

[back to top]

OS Rate at 2 Years- Based on Targeted Therapy Group and Other Treatments Group at Inclusion

OS was the percentage of participants who were alive at 2 years of prospective follow-up. OS was defined as the time (in months) from the date of start of treatment (of the treatment line ongoing at the time of inclusion in the study) to the date of death due to any cause. Participants with no event or lost to follow-up or alive at the end of the study were censored at their last follow-up date (last follow-up date or last news). Analysis was performed using Kaplan-Meier method. (NCT02264665)
Timeframe: At 2 years of prospective follow-up

InterventionPercentage of participants (Number)
Targeted Therapy at Inclusion63.9
Other Treatments at Inclusion71.7

[back to top]

Number of Participants Who Had a Change in Their Treatment - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

In this outcome measure, number of participants with a change in treatment compared to treatment at inclusion is reported. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

InterventionParticipants (Count of Participants)
Targeted Therapy at Inclusion44
Other Treatments at Inclusion68

[back to top]

Overall Survival (OS) Rate at 2 Years - Based on Type of Treatment at Inclusion

OS was the percentage of participants who were alive at 2 years of prospective follow-up. OS was defined as the time (in months) from the date of start of treatment (of the treatment line ongoing at the time of inclusion in the study) to the date of death due to any cause. Participants with no event or lost to follow-up or alive at the end of the study were censored at their last follow-up date (last follow-up date or last news). Analysis was performed using Kaplan-Meier method. (NCT02264665)
Timeframe: At 2 years of prospective follow-up

InterventionPercentage of participants (Number)
Everolimus (Targeted Therapy) at Inclusion60.2
Sunitinib (Targeted Therapy) at Inclusion68.4
Chemotherapy (Other Treatment) at Inclusion69.6
Somatostatin Analogues (Other Treatment) at Inclusion79.7
Metabolic Radiotherapy (Other Treatment) at InclusionNA

[back to top]

PFS at 2 Years Assessed by Investigator Per RECIST v1.1 - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

PFS was defined as time (in months) from date of start of treatment (the treatment line ongoing at the time of inclusion in the study) to first documentation of PD or date of death due to any cause, when receiving the main treatment at the time of inclusion. RECIST v1.1, PD: at least a 20% increase in sum of diameters of target lesions, taking as reference smallest sum on study treatment (this included baseline sum if that is smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. Unequivocal progression of existing non-target lesions. Appearance of 1 or more new target or non-target lesions. If a participant did not have an event, data censoring was done at the last recorded time to PD or the last tumor assessment, last disease assessment. Analysis was performed using Kaplan-Meier method. (NCT02264665)
Timeframe: At 2 years of prospective follow-up

InterventionPercentage of participants (Number)
Targeted Therapy at Inclusion19.9
Other Treatments at Inclusion29.0

[back to top] [back to top]

Number of Participants With Adverse Events Leading to Discontinuation of Treatment - Based on Treatment Received At-least Once During the Study

An AE was any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. Participants in this outcome measure were grouped according to type of treatment received at least once during study irrespective of treatment they initiated or were receiving at time of inclusion. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

,,,,,
InterventionParticipants (Count of Participants)
Temporary discontinuation of treatmentPermanent discontinuation of treatment
Chemotherapy During Study1112
Everolimus During Study1514
Metabolic Radiotherapy During Study00
Other Than Study Treatment During Study11
Somatostatin Analogues During Study at Inclusion812
Sunitinib During Study910

[back to top]

Number of Participants With Different Types of Investigation Used for Tumor Assessment - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

The different types of investigations were performed for assessment of tumor regardless of treatment received. Investigations performed were Computed Tomography (CT) scan, Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) scan, Octreoscan, Ultrasound. All the investigations were used at least once during the study for all treatments. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

,
InterventionParticipants (Count of Participants)
CT ScanMRIPET ScanOctreoscanUltrasoundImaging (CT scan and/or MRI)
Other Treatments at Inclusion65451515474
Targeted Therapy at Inclusion48281113652

[back to top]

Number of Participants With Types of Main Lines of Treatment Received During the Follow-up - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

The types of main lines of treatment included 1st line, 2nd line, 3rd line, 4th line, 5th line, 6th line, 7th line and 8th line. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

,
InterventionParticipants (Count of Participants)
1st line2nd line3rd line4th line5th line6th line7th line8th line
Other Treatments at Inclusion3843302615210
Targeted Therapy at Inclusion827252815742

[back to top]

Number of Main Lines of Treatment Received During the Study - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

The number of main lines of treatment received during the study treatment are reported in this outcome measure. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

InterventionTreatment lines (Mean)
Targeted Therapy at Inclusion3.7
Other Treatments at Inclusion2.8

[back to top]

Number of Combined Main Lines of Treatments Received - Based on Targeted Therapy Group and Other Treatments Group at Inclusion

The number of lines of combined main treatment administered during the study were assessed. (NCT02264665)
Timeframe: During 2 years of prospective follow-up

InterventionTreatment lines (Mean)
Targeted Therapy at Inclusion3.9
Other Treatments at Inclusion2.9

[back to top]

Progression-Free Survival (PFS) at 2 Years Assessed by Investigator Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1 - Based on Type of Treatment at Inclusion

PFS was defined as time (in months) from date of start of treatment (the treatment line ongoing at the time of inclusion in the study) to first documentation of disease progression (PD) or date of death due to any cause, when receiving the main treatment at the time of inclusion. RECIST v1.1, PD: at least a 20% increase in sum of diameters of target lesions, taking as reference smallest sum on study treatment (this included baseline sum if that is smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 millimeters (mm). Unequivocal progression of existing non-target lesions. Appearance of 1 or more new target or non-target lesions. If a participant did not have an event, data censoring was done at the last recorded time to PD or the last tumor assessment, last disease assessment. Analysis was performed using Kaplan-Meier method. (NCT02264665)
Timeframe: At 2 years of prospective follow-up

InterventionPercentage of participants (Number)
Everolimus (Targeted Therapy) at Inclusion26.5
Sunitinib (Targeted Therapy) at Inclusion19.3
Chemotherapy (Other Treatment) at Inclusion36.5
Somatostatin Analogues (Other Treatment) at Inclusion38.6
Metabolic Radiotherapy (Other Treatment) at InclusionNA

[back to top]

Progression Free Survival (PFS)

"PFS will be estimated using the method of Kaplan-Meier. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), Disease progression in evaluable patients will be defined as one or more of the following:~Any new disease and/or clear progression of evaluable disease; OR 2 fold elevation in CA-125 from its lowest level (either initial level or nadir, whichever is lowest, since study enrollment) combined with CA-125 elevation confirmed by re-assay at any time." (NCT02283658)
Timeframe: Time from registration to the first of either disease progression or death from any cause, assessed up to 2 years

InterventionMonths (Median)
Treatment (Everolimus and Letrozole)3.9

[back to top]

Percentage of Participants With CA-125 Response

CA-125 response: The key secondary endpoint of the study will be a CA-125 response, defined as a 50% or greater reduction in baseline CA-125. The null hypothesis will be set at CA-125 response rate of 8.3%, based on the response of single agent letrozole, as reported by Bowman et al (7). The treatment of letrozole and everolimus will be considered promising, based on CA-125, if the observed CA-125 response rate is 30% or more. (NCT02283658)
Timeframe: Up to 2 years

Interventionpercentage of patients (Number)
Treatment (Everolimus and Letrozole)21

[back to top]

Percentage of Patients Alive and Progression Free Survival at 12 Weeks

"The percentage of PFS12 successes will be estimated by the number of successes divided by the total number of evaluable patients. Ninety-five percent confidence intervals for the true success proportion will be calculated according to the exact binomial method. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), Disease progression in evaluable patients will be defined as one or more of the following:~Any new disease and/or clear progression of evaluable disease; OR~2 fold elevation in CA-125 from its lowest level (either initial level or nadir, whichever is lowest, since study enrollment) combined with CA-125 elevation confirmed by re-assay at any time." (NCT02283658)
Timeframe: 12 weeks

Interventionpercentage of patients (Number)
Treatment (Everolimus and Letrozole)47

[back to top]

Confirmed Response Rate, Estimated Using RECIST 1.1 Criteria

"A confirmed tumor response is defined to be either a complete response or partial response noted as the objective status on 2 consecutive evaluations at least 4 weeks apart. A complete response (CR) in evaluable patients will be defined as:~Disappearance of any sign of (evaluable) disease, AND~Normalization of CA-125; if CA-125 normalizes, it should be confirmed at any time.~A partial response (PR) in evaluable patients will be defined as:~Decrement in CA-125 by >50%, and~Improvement in any additional evaluable disease (if present) as assessed by the enrolling physician." (NCT02283658)
Timeframe: Up to 24 weeks

Interventionpercentage of patients (Number)
Treatment (Everolimus and Letrozole)16

[back to top]

Overall Suravival(OS)

OS will be estimated using the method of Kaplan-Meier. (NCT02283658)
Timeframe: Time from registration to death from any cause, assessed up to 2 years

InterventionMonths (Median)
Treatment (Everolimus and Letrozole)13

[back to top]

Number of Participants Experiencing Adverse Events

The number of patients with adverse events.The maximum grade for each type of adverse event (AE) will be recorded for each patient, and frequency tables will be reviewed to determine AE patterns. These tables are reported in the adverse events section of this report. (NCT02283658)
Timeframe: Up to 30 days post-treatment

InterventionParticipants (Count of Participants)
Treatment (Everolimus and Letrozole)19

[back to top]

Median Time From First Occurrence of CR or PR to Disease Progression or Death Also Called Duration of Response (DOR)

Only those patients who achieved Complete Response or Partial Response will be included in the summaries of DOR. DOR is defined as time from first date of response of CR or PR to disease progression or death as defined by RECIST v1.1 criteria. Participants who are alive and free from disease progression will be censored at the date of last tumor assessment. Patients who receive non-protocol therapy (subsequent therapy) prior to incurring an event will be censored at the date of last tumor assessment prior to the start of subsequent therapy. A CR is the complete disappearance of all target lesions. A PR is a decrease of 30% or more of the diameter(s) of all target lesions from the baseline sum of diameters. (NCT02291913)
Timeframe: every 8 weeks until discontinuation, up to 20 months

Interventionmonths (Median)
Everolimus8.8

[back to top]

Median Progression Free Survival (PFS)

PFS is defined as the time from Day 1 of study drug administration to disease progression as defined by RECIST (Response Evaluation Criteria in Solid Tumors) version 1.1 criteria, or death on study. Participants who are alive and free from disease progression will be censored at the date of last radiologic tumor assessment. Participants who receive non-protocol therapy (subsequent therapy) prior to incurring an event will be censored at the date of last tumor assessment prior to the start of subsequent therapy. Participants who do not have a post-baseline tumor assessment will be censored at the date of first treatment (Day 1). (NCT02291913)
Timeframe: up to 3 years

Interventionmonths (Median)
Everolimus7.2

[back to top]

Number of Patients With Adverse Events (AEs) as a Measure of Safety and Tolerability

Assessments were made through analysis of the reported incidence of treatment-emergent AEs. All participants who received at least one dose of protocol treatment were followed for safety. Adverse events were collected from day of first dose to 30 days after last protocol treatment and graded according to National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.03. (NCT02291913)
Timeframe: Up to 20 months

InterventionParticipants (Count of Participants)
Everolimus48

[back to top]

Number of Patients With an Objective Response (CR or PR) Also Called the Overall Response Rate (ORR).

Defined as the number of patients with objective evidence of complete or partial response (CR or PR) using RECIST version 1.1. A CR is the complete disappearance of all target lesions. A PR is a decrease of 30% or more of the diameter(s) of all target lesions from the baseline sum of diameters. (NCT02291913)
Timeframe: every 8 weeks until discontinuation, up to 20 months

InterventionParticipants (Count of Participants)
Everolimus2

[back to top]

Number of Participants With CR, PR, or 6 Months of SD Also Called Clinical Benefit Rate (CBR)

The proportion of patients with Complete Response (CR) or Partial Response (PR) or 6 months or more of Stable Disease (SD). A CR is the complete disappearance of all target lesions. A PR is a decrease of 30% or more of the diameter(s) of all target lesions from the baseline sum of diameters. SD is not meeting the criteria for PR or a 20% increase in target lesions called Progressive Disease (PD). (NCT02291913)
Timeframe: Up to 20 months

InterventionParticipants (Count of Participants)
Everolimus12

[back to top]

Median Overall Survival (OS)

Defined as the time from date of first study treatment to date of death due to any cause. Patients who are alive will be censored at the date of last known date alive. (NCT02291913)
Timeframe: up to 3 years from first treatment

Interventionmonths (Median)
Everolimus26.7

[back to top]

Overall Survival

Overall survival is defined as the time from treatment start date until date of death or date last known alive following therapy. (NCT02315625)
Timeframe: Up to approximately 4 years

InterventionMonths (Median)
Sunitinib First, Everolimus Second25.5
Everolimus First, Sunitinib Second10.7

[back to top]

Number of Participants With Serious and Non-Serious Adverse Events

Here is the count of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0). A non-serious adverse event is any untoward medical occurrence. A serious adverse event is an adverse event or suspected adverse reaction that results in death, a life threatening adverse drug experience, hospitalization, disruption of the ability to conduct normal life functions, congenital anomaly/birth defect or important medical events that jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the previous outcomes mentioned. (NCT02315625)
Timeframe: Date treatment consent signed to date off study approximately 49 months and 9 days.

InterventionParticipants (Count of Participants)
Sunitinib15
Everolimus11

[back to top]

Number of Participants With an Overall Response

Overall response was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST) is defined as Complete Response (CR), Partial Response (PR), and Stable Disease (SD). Complete Response is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial Response is at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. Stable Disease is neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for Progressive Disease (PD) (e.g. appearance of one or more new lesions), taking as reference the smallest sum of diameters while on study. (NCT02315625)
Timeframe: Every 3 months until disease progression, up to 12 months

InterventionParticipants (Count of Participants)
Sunitinib First, Everolimus Second1
Everolimus First, Sunitinib Second0

[back to top]

Median Survival Time (MST)

MST is the amount of time a subject survives after therapy. (NCT02315625)
Timeframe: Death, an average of 12 months follow up

InterventionMonths (Median)
Sunitinib First, Everolimus Second9.5
Everolimus First, Sunitinib Second5.5

[back to top]

Median Amount of Time Subject Survives Without Disease Progression After Treatment

Median amount of time subject survives without disease progression after treatment. Progression was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST) and is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered progressions. (NCT02315625)
Timeframe: Up to approximately 2 years

Interventionmonths (Median)
Sunitinib First, Everolimus Second7
Everolimus First, Sunitinib Second18

[back to top]

3D Photographic Measurement of Surface Volume of Cutaneous Neurofibroma Lesion

Photographs of selected lesions to measure surface volume. (NCT02332902)
Timeframe: 6 months

Interventionmm^3 (Mean)
Baseline6 months
Intervention32.5433.93

[back to top]

Number of Participants With Grade 3-4 Adverse Events

Determination if orally administered Afinitor is safe in patients a indicated by lack of Grade 3-4 adverse events during the trial period. (NCT02332902)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Intervention0

[back to top]

Mutations Associated With Therapeutic Response

-To correlate mutations in the mTOR pathway with therapeutic response with everolimus (NCT02352844)
Timeframe: Completion of treatment (estimated average of 6 months)

InterventionParticipants (Count of Participants)
STK11 c.375-2delSTK11 c.375-8C>G
Arm 1 (Everolimus)11

[back to top]

Response Rate (RR)

The primary endpoint will be to describe the response rate using RECIST 1.1. Response rate will be defined as complete response (disappearance of all target lesion) plus partial response (a least a 30% decrease in the sum of diameters of target lesions). (NCT02352844)
Timeframe: Completion of treatment (estimated average of 6 months)

InterventionParticipants (Count of Participants)
Arm 1 (Everolimus)1

[back to top]

Genetic Changes Associated With Disease Progression

-To investigate the genetic changes associated with disease progression following treatment with everolimus. (NCT02352844)
Timeframe: Completion of treatment (estimated average of 6 months)

InterventionParticipants (Count of Participants)
NF1 p.E8*STK11 p.E199*NF1 p.Y489CSTK11 exon 1 E33XSTK11 L282fs*5NF1 c.7190C>TNF1 c.7253C>T
Arm 1 (Everolimus)1111111

[back to top]

Number of Lesions With Lesion Success

Defined as attainment of < 30% residual stenosis of the target lesion using any percutaneous method. (NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure)

InterventionLesions (Count of Units)
Orsiro Sirolimus Coronary Stent System1102
Xience Everolimus Coronary Stent System579

[back to top]

Number of Participants With Procedure Success

Defined as attainment of < 30% residual stenosis of the target lesion using the assigned study stent only without occurrence of in-hospital major adverse cardiac events (MACE; composite of all-cause death, Q-wave or non-Q-wave MI, and any clinically-driven TLR). (NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure)

InterventionParticipants (Count of Participants)
Orsiro Sirolimus Coronary Stent System827
Xience Everolimus Coronary Stent System401

[back to top]

Number of Participants With TLF and Individual TLF Components

TLF events are defined as cardiac death, target vessel Q-wave or non-Q-wave MI, or any clinically-driven TLR. The number of participants with any TLF event is provided, as well as number of participants with each of the individual components. (NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure), 1, 6, 12 months, 2, 3, 4 and 5 years

,
InterventionParticipants (Count of Participants)
6-24 hours post-index TLF Total6-24 hours post-index TLF Component: Cardiac Death6-24 hours post-index TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI6-24 hours post-index TLF Component: Clinically-driven TLR1-month TLF Total1-month TLF Component: Cardiac Death1-month TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI1-month TLF Component: Clinically-driven TLR6-month TLF Total6-month TLF Component: Cardiac Death6-month TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI6-month TLF Component: Clinically-driven TLR12-month TLF Total12-month TLF Component: Cardiac Death12-month TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI12-month TLF Component: Clinically-driven TLR2-year TLF Total2-year TLF Component: Cardiac Death2-year TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI2-year TLF Component: Clinically-driven TLR3-year TLF Total3-year TLF Component: Cardiac Death3-year TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI3-year TLF Component: Clinically-driven TLR4-year TLF Total4-year TLF Component: Cardiac Death4-year TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI4-year TLF Component: Clinically-driven TLR5-year TLF Total5-year TLF Component: Cardiac Death5-year TLF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI5-year TLF Component: Clinically-driven TLR
Orsiro Sirolimus Coronary Stent System351342371364471381352139176454521729462791174938104215648
Xience Everolimus Coronary Stent System31129232130339234841335104823919565412560544286684532

[back to top]

Percentage of Participants With Target Lesion Failure (TLF) at 12 Months Post-Index Procedure by Bayesian Estimation

TLF is defined as all cardiac death, target vessel Q-wave or non-Q-wave myocardial infarction (MI), or clinically driven target lesion revascularization (TLR). (NCT02389946)
Timeframe: 12-Months

InterventionPercentage of participants (Mean)
Orsiro Sirolimus Coronary Stent System6.32
Xience Everolimus Coronary Stent System8.90

[back to top]

Number of Participants With MACE and Individual MACE Components

MACE events are defined as all-cause death, Q-wave or non-Q-wave MI, or any clinically-driven TLR. The number of participants with any MACE event is provided, as well as number of participants with each of the individual components. (NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure), 1, 6, 12 months, 2, 3, 4 and 5 years

,
InterventionParticipants (Count of Participants)
6-24 hours post-index MACE Total6-24 hours post-index MACE Component: Death6-24 hours post-index MACE Component: Protocol defined Q-Wave or non-Q-wave MI6-24 hours post-index MACE Component: Clinically-driven TLR1-month MACE Total1-month MACE Component: Death1-month MACE Component: Protocol defined Q-Wave or non-Q-wave MI1-month MACE Component: Clinically-driven TLR6-month MACE Total6-month MACE Component: Death6-month MACE Component: Protocol-defined Q-Wave or non-Q-wave MI6-month MACE Component: Clinically-driven TLR12-month MACE Total12-month MACE Component: Death12-month MACE Component: Protocol-defined Q-Wave or non-Q-wave MI12-month MACE Component: Clinically-driven TLR2-year MACE Total2-year MACE Component: Death2-year MACE Component: Protocol-defined Q-Wave or non-Q-wave MI2-year MACE Component: Clinically-driven TLR3-year MACE Total3-year MACE Component: Death3-year MACE Component: Protocol-defined Q-Wave or non-Q-wave MI3-year MACE Component: Clinically-driven TLR4-year MACE Total4-year MACE Component: Death4-year MACE Component: Protocol-defined Q-Wave or non-Q-wave MI4-year MACE Component: Clinically-driven TLR5-year MACE Total5-year MACE Component: Death5-year MACE Component: Protocol-defined Q-Wave or non-Q-wave MI5-year MACE Clinically-driven TLR
Orsiro Sirolimus Coronary Stent System3513423913845364013597411785165621102276027133456638155567648
Xience Everolimus Coronary Stent System31130232131342436844637105694219731749258122522892275832

[back to top]

Number of Participants With Myocardial Infarction

(NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure), 1, 6, 12 months, 2, 3, 4 and 5 years

,
InterventionParticipants (Count of Participants)
6-24 hours post-index procedure1-month post-index procedure6-month12-month2-year3-year4-year5-year
Orsiro Sirolimus Coronary Stent System3438404156606676
Xience Everolimus Coronary Stent System3031363742495258

[back to top]

Number of Participants With Myocardial Infarction or Cardiac Death

(NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure), 1, 6, 12 months, 2, 3, 4 and 5 years

,
InterventionParticipants (Count of Participants)
Hospital Discharge (6-24 hours post-index procedure)1-month6-month12-month2-year3-year4-year5-year
Orsiro Sirolimus Coronary Stent System3539414261688294
Xience Everolimus Coronary Stent System3031373943515461

[back to top]

Number of Participants With Stent Thrombosis

Stent thrombosis according to the Academic Research Consortium criteria. (NCT02389946)
Timeframe: 24 hours, 30 days, 1 year, and 5 years post-index procedure

,
InterventionParticipants (Count of Participants)
Possible ARC Stent Thrombosis TotalPossible ARC Stent Thrombosis Acute (<=24 hours)Possible ARC Stent Thrombosis Sub-acute (>24 hours and <=30 days)Possible ARC Stent Thrombosis Late (>30 days and <=1 year)Possible ARC Stent Thrombosis Very Late (>1 year)Definite/Probable ARC Stent Thrombosis TotalDefinite/Probable ARC Stent Thrombosis Acute (<=24 hours)Definite/Probable ARC Stent Thrombosis Sub-acute (>24 hours and <=30 days)Definite/Probable ARC Stent Thrombosis Late (>30 days and <=1 year)Definite/Probable ARC Stent Thrombosis Very Late (>1 year)
Orsiro Sirolimus Coronary Stent System110001151211
Xience Everolimus Coronary Stent System6001570124

[back to top]

Number of Participants With Target Vessel Failure (TVF) and Individual TVF Components

TVF events are defined as cardiac death, target vessel Q-wave or non-Q-wave MI, or any clinically-driven TVR. The number of participants with any TVF event is provided, as well as number of participants with each of the individual components. (NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure), 1, 6, 12 months, 2, 3, 4 and 5 years

,
InterventionParticipants (Count of Participants)
6-24 hours post-index TVF Total6-24 hours post-index TVF Component: Cardiac Death6-24 hours post-index TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI6-24 hours post-index TVF Component: Clinically-driven TVR1-month TVF Total1-month TVF Component: Cardiac Death1-month TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI1-month TVF Component: Clinically-driven TVR6-month TVF Total6-month TVF Component: Cardiac Death6-month TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI6-month TVF Component: Clinically-driven TVR12-month TVF Total12-month TVF Component: Cardiac Death12-month TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI12-month TVF Component: Clinically-driven TVR2-year TVF Total2-year TVF Component: Cardiac Death2-year TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI2-year TVF Component: Clinically-driven TVR3-year TVF Total3-year TVF Component: Cardiac Death3-year TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI3-year TVF Component: Clinically-driven TVR4-year TVF Total4-year TVF Component: Cardiac Death4-year TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI4-year TVF Component: Clinically-driven TVR5-year TVF Total5-year TVF Component: Cardiac Death5-year TVF Component: Protocol-defined Target Vessel Q-wave or non-Q-Wave MI5-year TVF Component: Clinically-driven TVR
Orsiro Sirolimus Coronary Stent System3513423813655113818601392774545378494645110174963127215678
Xience Everolimus Coronary Stent System311292321304402341045335155623930685414074544468184551

[back to top]

Number of Lesions With Device Success

Defined as attainment of < 30% residual stenosis of the target lesion using the assigned study stent only. (NCT02389946)
Timeframe: Hospital Discharge (6-24 hours post-index procedure)

InterventionLesions (Number)
Orsiro Sirolimus Coronary Stent System1082
Xience Everolimus Coronary Stent System566

[back to top]

Plasma HIV RNA

Plasma HIV RNA was quantified in a highly sensitive single-copy assay (SCA) using repetitive sampling in the Panther system (Hologic) at the Blood Systems Research Institute. Up to 18 replicates were tested for each sample in order to determine plasma RNA levels as low as 0.18 copies/mL. (NCT02429869)
Timeframe: Baseline, Month 2, Month 6, Month 12 (6 months post discontinuation of everolimus)

Interventionnucleic acid copies per million cells (Median)
BaselineMonth 2Month 6Month 12
Everolimus0.2910.5841.0420.941

[back to top]

Cell-associated Total HIV RNA

Peripheral blood mononuclear cells were isolated from whole blood using the Ficoll density gradient technique. Peripheral blood CD4 T cells were enriched by negative selection using antibody-coupled magnetic beads (Stem Cell Technologies) prior to simultaneous RNA and DNA isolation using cell-sparing protocols (AllPrep, Qiagen). Bulk CD4+ T cell or PBMC-associated HIV DNA and unspliced RNA were quantified using real-time PCR methods.The primer and probe sequences targeted conserved regions to enable quantification of a broad range of HIV subtypes. Values were normalized to DNA quantification of a human housekeeping gene (CCR5) in order to determine nucleic acid copies per million CD4+ T cell or PBMC as described. In addition to traditional quantitative PCR, a novel single-cell-in-droplet (scd)PCR method was used to quantify the absolute number or frequency of individual purified CD4+ T cells that express unspliced HIV RNA. (NCT02429869)
Timeframe: Baseline, Month 2, Month 6, Month 12 (6 months post discontinuation of everolimus)

Interventionnucleic acid copies per million cells (Median)
Baseline HIV RNA CD4+Month 2 HIV RNA CD4+Month 6 HIV RNA CD4+Month 12 HIV RNA CD4+
Everolimus423310702354

[back to top]

Cell-associated HIV DNA

Peripheral blood mononuclear cells were isolated from whole blood using the Ficoll density gradient technique. Peripheral blood CD4 T cells were enriched by negative selection using antibody-coupled magnetic beads (Stem Cell Technologies) prior to simultaneous RNA and DNA isolation using cell-sparing protocols (AllPrep, Qiagen). Bulk CD4+ T cell or PBMC-associated HIV DNA and unspliced RNA were quantified using real-time PCR methods.The primer and probe sequences targeted conserved regions to enable quantification of a broad range of HIV subtypes. Values were normalized to DNA quantification of a human housekeeping gene (CCR5) in order to determine nucleic acid copies per million CD4+ T cell or PBMC as described. In addition to traditional quantitative PCR, a novel single-cell-in-droplet (scd)PCR method was used to quantify the absolute number or frequency of individual purified CD4+ T cells that express unspliced HIV RNA. (NCT02429869)
Timeframe: Baseline, Month 2, Month 6, Month 12 (6 months post discontinuation of everolimus)

Interventionnucleic acid copies per million cells (Median)
BaselineMonth 2Month 6Month 12
Everolimus35438116

[back to top]

Blood Total VEGF Levels (Not Only VEGF-D)

(NCT02451696)
Timeframe: 28 days

Interventionpg/ml (Mean)
Treated Subjects56.5
Reference Subjects50.85

[back to top]

mTOR Brain Tissue-S6 Phosphate by Western Blot

(NCT02451696)
Timeframe: 28 days

Interventionnormalized val-protein relative to actin (Mean)
Treated Subjects0.49
Reference Subjects0.81

[back to top]

Number of Patients With Adverse Events

.Adverse event monitoring should be continued for at least 30 days (or 5 half-lives, whichever is longer) following the last dose of study treatment (NCT02451696)
Timeframe: 6 weeks

InterventionParticipants (Count of Participants)
Treated Subjects0
Reference Subjects0

[back to top]

HMGB1 Expression in Brain Tissue

HMGB1 expression is measured through label-free quantification (LFQ). LFQ is a method in mass spectroscopy that determines the relative amount of proteins in biological samples. The unit of measure is LFQ intensity; a higher LFQ intensity indicates greater HMGB1 expression. (NCT02451696)
Timeframe: 28 days

InterventionLFQ intensity (Mean)
Treated Subjects14.85
Reference Subjects15.06

[back to top]

Blood Everolimus Levels

mTOR signaling in blood (NCT02451696)
Timeframe: 28 days

Interventionng/ml (Mean)
Treated Subjects12.35
Reference Subjects2

[back to top]

Objective Response Rate (ORR)

ORR was defined as the percentage of participants who achieved BOR of CR or PR. ORR was assessed using RECIST 1.1. (NCT02454478)
Timeframe: From first dose of study drug until PD, development of unacceptable toxicity, participant requests to discontinue, withdrawal of consent or study termination (up to approximately 23 months)

Interventionpercentage of participants (Number)
Lenvatinib 18 mg + Everolimus 5 mg71.4

[back to top]

Tss,Max: Time to Reach the Maximum Plasma Concentration (Cmax) at Steady State for Levatinib and Everolimus

(NCT02454478)
Timeframe: Cycle 1 Day 15 predose and at 1, 2, 4 ,8, and 24 hours postdose (Cycle length=28 days)

Interventionhour (Median)
Lenvatinib: Cycle 1 Day 15Everolimus: Cycle 1 Day 15
Lenvatinib 18 mg + Everolimus 5 mg3.770.97

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

(NCT02454478)
Timeframe: Baseline up to 30 days after the last dose of study drug (up to approximately 21.5 months)

Interventionparticipants (Number)
TEAESAE
Lenvatinib 18 mg + Everolimus 5 mg73

[back to top]

Number of Participants With the Minimum Percent Change From Baseline in the Sum of Diameters of Target Lesions

(NCT02454478)
Timeframe: Baseline up to first tumor assessment at which diameter of target lesions were available (up to approximately 23 months)

Interventionparticipants (Number)
Less than or equal to (<=) -30%Greater than (>) -30% to less than (<) 20%Greater than or equal to (>=) 20%
Lenvatinib 18 mg + Everolimus 5 mg511

[back to top]

Disease Control Rate (DCR)

DCR was defined as the percentage of participants who achieved BOR of CR, PR, or SD. DCR was assessed based on RECIST 1.1. (NCT02454478)
Timeframe: From first dose of study drug until PD, development of unacceptable toxicity, participant requests to discontinue, withdrawal of consent or study termination (up to approximately 23 months)

Interventionpercentage of participants (Number)
Lenvatinib 18 mg + Everolimus 5 mg85.7

[back to top]

Number of Participants With Best Overall Response (BOR)

BOR included complete response (CR), partial response (PR), stable disease (SD), and PD (progressive disease). BOR was assessed using Response Evaluation Criteria in Solid Tumor (RECIST) 1.1 (NCT02454478)
Timeframe: From first dose of study drug until PD, development of unacceptable toxicity, participant requests to discontinue, withdrawal of consent or study termination (up to approximately 23 months)

Interventionparticipants (Number)
CRPRSDPD
Lenvatinib 18 mg + Everolimus 5 mg0511

[back to top]

Css,Max: Maximum Observed Plasma Concentration at Steady State for Levatinib and Everolimus

(NCT02454478)
Timeframe: Cycle 1 Day 15 predose and at 1, 2, 4 ,8, and 24 hours postdose (Cycle length=28 days)

Interventionng/mL (Mean)
Lenvatinib: Cycle 1 Day 15Everolimus: Cycle 1 Day 15
Lenvatinib 18 mg + Everolimus 5 mg25741.5

[back to top]

Cmax: Maximum Observed Plasma Concentration for Levatinib and Everolimus

(NCT02454478)
Timeframe: Cycle 1 Day 1 predose and at 1, 2, 4 ,8, and 24 hours postdose (Cycle length=28 days)

Interventionnanogram per milliliter (ng/mL) (Mean)
Lenvatinib: Cycle 1 Day 1Everolimus: Cycle 1 Day 1
Lenvatinib 18 mg + Everolimus 5 mg28939.1

[back to top]

Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Levatinib and Everolimus

(NCT02454478)
Timeframe: Cycle 1 Day 1 predose and at 1, 2, 4 ,8, and 24 hours postdose (Cycle length=28 days)

Interventionhour (Median)
Lenvatinib: Cycle 1 Day 1Everolimus: Cycle 1 Day 1
Lenvatinib 18 mg + Everolimus 5 mg3.870.92

[back to top]

Number of Participants Who Experienced Any Dose Limiting Toxicity (DLT)

DLT was defined as toxicity related to the combination therapy and was graded according to Common Terminology Criteria for Adverse Events version 4.03 (CTCAE v4.03). (NCT02454478)
Timeframe: From first dose of study drug up to Cycle 1 Day 28 (Cycle length=28 days)

Interventionparticipants (Number)
Lenvatinib 18 mg + Everolimus 5 mg0

[back to top]

AUC 0-t: Area Under the Concentration-time Curve From Zero (Pre-dose) to Time of Last Quantifiable Concentration for Levatinib and Everolimus

(NCT02454478)
Timeframe: Cycle 1 Day 1 and Cycle 1 Day 15 predose and at 1, 2, 4 ,8, and 24 hours postdose (Cycle length=28 days)

Interventionhour * nanogram per milliliter (h*ng/mL) (Mean)
Lenvatinib: Cycle 1 Day 1Everolimus: Cycle 1 Day 1Lenvatinib: Cycle 1 Day 15Everolimus: Cycle 1 Day 15
Lenvatinib 18 mg + Everolimus 5 mg27702113220401

[back to top]

Count of Participants With Non-serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0)

Here is the count of participants with non-serious adverse events assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0). A non-serious adverse event is any untoward medical occurrence. (NCT02504892)
Timeframe: Date treatment consent signed to date off study, approximately 8 months and 28 days.

InterventionParticipants (Count of Participants)
Birt-Hogg-Dube Syndrome3

[back to top]

Progression-free Survival (PFS)

Progression-free survival is defined as the time interval from start of treatment to documented evidence of disease progression. Progression is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes baseline sum if that is the smallest on study). The sum must also demonstrate an absolute increase of at least 5mm. Note: the appearance of one or more new lesions is also progression. Progressive disease was assessed by the Response Evaluation in Solid Tumors (RECIST) criteria v1.1. (NCT02504892)
Timeframe: median follow-up time: 9 months

InterventionMonths (Median)
Birt-Hogg-Dube SyndromeNA

[back to top]

Overall Response Rate With Everolimus Treatment.

Overall best response is defined as the best response recorded from the start of the treatment until disease progression/recurrence (taking as reference for progressive disease the smallest measurements recorded since the treatment started). Response was assessed by the Response Evaluation in Solid Tumors (RECIST) criteria v1.1. Progressive disease is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes baseline sum if that is the smallest on study). The sum must also demonstrate an absolute increase of at least 5mm. Note: the appearance of one or more new lesions is also progression. Complete response is disappearance of all target lesions. Partial response is at least a 30% decrease in the sum of the diameters of target lesions. Stable disease is neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum of diameters while on study. (NCT02504892)
Timeframe: End of treatment: every 12 weeks up to 1 year

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Birt-Hogg-Dube Syndrome0030

[back to top]

Overall Survival (OS)

Overall Survival is defined as the time between the first day of treatment to the day of death. (NCT02504892)
Timeframe: From the first day of treatment to the day of death, up to 1 year

InterventionMonths (Median)
Birt-Hogg-Dube SyndromeNA

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent59

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent73

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent78

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent85

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent88

[back to top]

Number of Participants With All Revascularization

"Revascularization:~Target Lesion Revascularization (TLR) is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. The target lesion is defined as the treated segment from 5 mm proximal to the scaffold and to 5 mm distal to the test scaffold.~Target Vessel Revascularization (TVR) is defined as any repeat percutaneous intervention or surgical bypass of any segment of the target vessel. The target vessel is defined as the entire major coronary vessel proximal and distal to the target lesion.~Non Target Lesion Revascularization (Non-TLR) is any revascularization in the target vessel for a lesion other than the target lesion.~Non Target Vessel Revascularization (Non-TVR)is any revascularization in a vessel other than the target vessel." (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent34

[back to top]

Number of Participants With Myocardial Infarction

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent2

[back to top]

Number of Participants With Myocardial Infarction

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent3

[back to top]

Number of Participants With Cardiac Death or MI

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.) Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent5

[back to top]

Number of Participants With Cardiac Death or MI

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent6

[back to top]

Number of Participants With Cardiac Death or MI

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent8

[back to top]

Number of Participants With Cardiac Death or MI

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent8

[back to top]

Number of Participants With Cardiac Death or MI

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent12

[back to top]

Number of Participants With Cardiac Death or MI

"Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery.)~Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent5

[back to top]

Number of Participants With Cardiac Death or Target Vessel MI

Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery). (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With Cardiac Death or Target Vessel MI

Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery). (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent6

[back to top]

Number of Participants With Cardiac Death or Target Vessel MI

Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery). (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent6

[back to top]

Number of Participants With Cardiac Death or Target Vessel MI

Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery). (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent9

[back to top]

Number of Participants With Target Vessel Failure

Target vessel failure includes cardiac death, MI and ischemia driven TLR; ischemia driven TVR, non TLR; ischemia driven TVR (TLR or TVR, nonTLR). (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent22

[back to top]

Number of Participants With Myocardial Infarction

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent2

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and Clinically indicated target lesion revascularization (CI-TLR). (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent9

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and clinically indicated target lesion revascularization (CI-TLR). (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent28

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and clinically indicated target lesion revascularization (CI-TLR). (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent23

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and clinically indicated target lesion revascularization (CI-TLR). (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent23

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and clinically indicated target lesion revascularization (CI-TLR). (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent18

[back to top]

Number of Participants With Major Adverse Cardiac Events (MACE)

Major adverse cardiac events (MACE) is defined as the composite of cardiac death, all myocardial infarction, and clinically indicated target lesion revascularization (CI-TLR). (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent12

[back to top]

Number of Participants With Hemorrhage

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events.~Severe or life-threatening:~Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention~Moderate:~Bleeding that requires blood transfusion but does not result in hemodynamic compromise~Mild:~Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent0

[back to top]

Number of Participants With Hemorrhage

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events.~Severe or life-threatening:~Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention~Moderate:~Bleeding that requires blood transfusion but does not result in hemodynamic compromise~Mild:~Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent2

[back to top]

Number of Participants With Hemorrhage

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events.~Severe or life-threatening:~Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention~Moderate:~Bleeding that requires blood transfusion but does not result in hemodynamic compromise~Mild:~Bleeding that does not meet criteria for either moderate or severe bleeding" (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent1

[back to top]

Number of Participants With Hemorrhage

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events.~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention; Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise; Mild: Bleeding that does not meet criteria for either moderate or severe bleeding." (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With Hemorrhage

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events.~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention; Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise; Mild: Bleeding that does not meet criteria for either moderate or severe bleeding." (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With Hemorrhage

"Bleeding complications will be defined according to the GUSTO (Global Use of Strategies to Open Occluded Coronary Arteries) classification of severe, moderate, and mild bleeding events.~Severe or life-threatening: Either intracranial hemorrhage or bleeding that causes hemodynamic compromise and requires intervention; Moderate: Bleeding that requires blood transfusion but does not result in hemodynamic compromise; Mild: Bleeding that does not meet criteria for either moderate or severe bleeding." (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With Death or MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent6

[back to top]

Number of Participants With Death or MI

"All deaths includes~Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI)~Q wave MI Development of new, pathological Q wave on the ECG.~Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent9

[back to top]

Number of Participants With Death or MI

"All deaths includes Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI) Q wave MI Development of new, pathological Q wave on the ECG. Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves" (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent33

[back to top]

Number of Participants With Death or MI

"All deaths includes Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI) Q wave MI Development of new, pathological Q wave on the ECG. Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves" (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent27

[back to top]

Number of Participants With Death or MI

"All deaths includes Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI) Q wave MI Development of new, pathological Q wave on the ECG. Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves" (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent22

[back to top]

Number of Participants With Death or MI

"All deaths includes Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma.~Myocardial Infarction (MI) Q wave MI Development of new, pathological Q wave on the ECG. Non-Q wave MI Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves" (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent14

[back to top]

Number of Participants With Cardiac Death or Target-Vessel MI

Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery). (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Net Gain: In-stent, In-segment

Difference between acute gain and late loss. (NCT02513719)
Timeframe: Post-Procedure (on day 0)

InterventionMillimeter (Mean)
StentSegment
XIENCE PRIME SV Everolimus Eluting Coronary Stent1.311.08

[back to top]

Late Loss(LL): In-stent,In-segment,Proximal, and Distal

Proximal and distal late loss was calculated by [post-procedure minimum lumen diameter (MLD)] - [MLD at 8 months]. (NCT02513719)
Timeframe: 8 months

InterventionMillimeter (Mean)
StentProximalDistalSegment
XIENCE PRIME SV Everolimus Eluting Coronary Stent0.230.13-0.030.09

[back to top]

Acute Gain: In-stent,In-segment

The acute gain was defined as the difference between post- and preprocedural minimal lumen diameter (MLD). (NCT02513719)
Timeframe: Pre procedure to post procedure (on day 0)

InterventionMillimeter (Mean)
StentSegment
XIENCE PRIME SV Everolimus Eluting Coronary Stent1.561.16

[back to top]

Success Rate: XIENCE PRIME Implant Success by Patient

"The stent lengths used were 8 mm, 12 mm, and 15 mm,18 mm, 23 mm, and 28 mm and the stent diameter was 2.25 mm.~Implant success is assessed as per physicians decision, but means that the stent could be implanted at the intended location." (NCT02513719)
Timeframe: < or = 1 day

Interventionparticipants (Number)
XIENCE PRIME SV Everolimus Eluting Coronary Stent100

[back to top]

Success Rate: Percentage of Lesions With Procedural Success

Achievement of final in-scaffold/stent residual stenosis of less than 50% by QCA with successful delivery and deployment of at least one study scaffold/stent at the intended target lesion and successful withdrawal of the delivery system for all target lesions without the occurrence of cardiac death, target vessel MI or repeat TLR during the hospital stay (less than or equal to 7 days). (NCT02513719)
Timeframe: < or = 1 day

InterventionPercentage of lesions (Number)
XIENCE PRIME SV Everolimus Eluting Coronary Stent100

[back to top]

Success Rate: Percentage of Devices With Implant Success

"The stent lengths used were 8 mm, 12 mm, and 15 mm,18 mm, 23 mm, and 28 mm and the stent diameter was 2.25mm.~Successful delivery and deployment of the first study scaffold/stent the intended target lesion and successful withdrawal of the delivery system with attainment of final in-scaffold/stent residual stenosis of less than 50% by quantitative coronary angiography (QCA)." (NCT02513719)
Timeframe: < or = 1 day

Interventionpercentage of devices (Number)
XIENCE PRIME SV Everolimus Eluting Coronary Stent100

[back to top]

Percent Diameter Stenosis (%DS)

The value calculated as 100 * (1- minimum lumen diameter/reference vessel diameter) (MLD/RVD) using the mean values from two orthogonal views (when possible) by quantitative coronary angiography (QCA). (NCT02513719)
Timeframe: Pre-procedure

InterventionPercent Diameter stenosis (Mean)
XIENCE PRIME SV Everolimus Eluting Coronary Stent73.62

[back to top]

Percent Diameter Stenosis (%DS)

The value calculated as 100 * (1- minimum lumen diameter/reference vessel diameter) (MLD/RVD) using the mean values from two orthogonal views (when possible) by quantitative coronary angiography (QCA). (NCT02513719)
Timeframe: post procedure (on day 0)

InterventionPercent Diameter stenosis (Mean)
XIENCE PRIME SV Everolimus Eluting Coronary Stent26.03

[back to top]

Percent Diameter Stenosis (%DS)

The value calculated as 100 * (1- minimum lumen diameter/reference vessel diameter) (MLD/RVD) using the mean values from two orthogonal views (when possible) by quantitative coronary angiography (QCA). (NCT02513719)
Timeframe: 8 months

InterventionPercent Diameter stenosis (Mean)
XIENCE PRIME SV Everolimus Eluting Coronary Stent28.31

[back to top]

Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)

The number of patient with Ischemia-driven Target vessel revascularization (TLR or TVR, non-TLR) (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent42

[back to top]

Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)

Target vessel revascularization (TLR or TVR, non-TLR) includes ischemia driven TVR (TLR or TVR, non-TLR) or non-ischemia driven TVR (TLR or TVR, non-TLR) (NCT02513719)
Timeframe: 0-5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent51

[back to top]

Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)

Target vessel revascularization (TLR or TVR, non-TLR) includes ischemia driven TVR (TLR or TVR, non-TLR) or non-ischemia driven TVR (TLR or TVR, non-TLR) (NCT02513719)
Timeframe: 0-4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent47

[back to top]

Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)

Target vessel revascularization (TLR or TVR, non-TLR) includes ischemia driven TVR (TLR or TVR, non-TLR) or non-ischemia driven TVR (TLR or TVR, non-TLR) (NCT02513719)
Timeframe: 0-3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent45

[back to top]

Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)

Target vessel revascularization (TLR or TVR, non-TLR) includes ischemia driven TVR (TLR or TVR, non-TLR) or non-ischemia driven TVR (TLR or TVR, non-TLR) (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent21

[back to top]

Number of Participants With Target Vessel Revascularization (TLR or TVR, Non-TLR)

Target vessel revascularization (TLR or TVR, non-TLR) includes ischemia driven TVR (TLR or TVR, non-TLR) or non-ischemia driven TVR (TLR or TVR, non-TLR) (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent34

[back to top]

Number of Participants With Target Vessel Failure

Target vessel failure includes cardiac death, MI, ischemia driven TLR, ischemia driven TVR, non TLR and ischemia driven TVR (TLR or TVR, nonTLR). (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent16

[back to top]

Number of Participants With Target Vessel Failure

Target vessel failure includes cardiac death, MI and ischemia driven TLR; ischemia driven TVR, non TLR; ischemia driven TVR (TLR or TVR, nonTLR). (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent45

[back to top]

Number of Participants With Target Vessel Failure

Target vessel failure includes cardiac death, MI and ischemia driven TLR; ischemia driven TVR, non TLR; ischemia driven TVR (TLR or TVR, nonTLR). (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent39

[back to top]

Number of Participants With Target Vessel Failure

Target vessel failure includes cardiac death, MI and ischemia driven TLR; ischemia driven TVR, non TLR; ischemia driven TVR (TLR or TVR, nonTLR). (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent35

[back to top]

Number of Participants With Target Vessel Failure

Target vessel failure includes cardiac death, MI and ischemia driven TLR; ischemia driven TVR, non TLR; ischemia driven TVR (TLR or TVR, nonTLR). (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent30

[back to top]

Number of Participants With Target Lesion Revascularization

Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. Target lesion revascularization (TLR) includes ischemia driven TLR and non-ischemia driven TLR (NCT02513719)
Timeframe: 0-5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent27

[back to top]

Number of Participants With Target Lesion Revascularization

Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. Target lesion revascularization (TLR) includes ischemia driven TLR and non-ischemia driven TLR (NCT02513719)
Timeframe: 0-4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent26

[back to top]

Number of Participants With Target Lesion Revascularization

Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. Target lesion revascularization (TLR) includes ischemia driven TLR and non-ischemia driven TLR (NCT02513719)
Timeframe: 0-3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent25

[back to top]

Number of Participants With Target Lesion Revascularization

Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. Target lesion revascularization (TLR) includes ischemia driven TLR and non-ischemia driven TLR (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent9

[back to top]

Number of Participants With Target Lesion Revascularization

Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. Target lesion revascularization (TLR) includes ischemia driven TLR and non-ischemia driven TLR (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent22

[back to top]

Number of Participants With Target Lesion Revascularization

Target lesion revascularization is defined as any repeat percutaneous intervention of the target lesion or bypass surgery of the target vessel performed for restenosis or other complication of the target lesion. Target lesion revascularization (TLR) includes ischemia driven TLR and non-ischemia driven TLR (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent16

[back to top]

Number of Participants With Target Lesion Failure

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent8

[back to top]

Number of Participants With Cardiac Death or Target Vessel-MI

Cardiac death is defined as any death in which a cardiac cause cannot be excluded. (This includes but is not limited to acute myocardial infarction, cardiac perforation/pericardial tamponade, arrhythmia or conduction abnormality,cerebrovascular accident within 30 days of the procedure or cerebrovascular accident suspected of being related to the procedure, death due to complication of the procedure, including bleeding, vascular repair, transfusion reaction, or bypass surgery). (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent5

[back to top]

Number of Participants With Target Lesion Failure

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent25

[back to top]

Number of Participants With Target Lesion Failure

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent21

[back to top]

Number of Participants With Target Lesion Failure

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent21

[back to top]

Number of Participants With Target Lesion Failure

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent16

[back to top]

Number of Participants With Target Lesion Failure

Target Lesion Failure is composite of Cardiac death/ Target Vessel Myocardial Infarction (TV-MI)/ Ischemic-Driven Target Lesion Revascularization (ID-TLR). (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent11

[back to top]

Number of Participants With Stent Thrombosis: Very Late

"Stent/Scaffold Thrombosis (per ARC): Stent/Scaffold Thrombosis should be reported as a cumulative value over time and at various individual time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Very late stent thrombosis : >1 year after stent implantation." (NCT02513719)
Timeframe: >1 year post stent implantation

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent0

[back to top]

Number of Death

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent7

[back to top]

Number of Death

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent11

[back to top]

Number of Death

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT02513719)
Timeframe: 0-3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent20

[back to top]

Number of Death

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT02513719)
Timeframe: 0-4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent25

[back to top]

Number of Death

"All deaths are considered cardiac unless an unequivocal non-cardiac cause can be established. Specifically, any unexpected death even in subjects with coexisting potentially fatal non-cardiac disease (e.g. cancer, infection) should be classified as cardiac.~• Cardiac death: Any death due to proximate cardiac cause (e.g. MI, low-output failure, fatal arrhythmia), unwitnessed death and death of unknown cause, all procedure related deaths including those related to concomitant treatment.~• Vascular death: Death due to non-coronary vascular causes such as cerebrovascular disease, pulmonary embolism, ruptured aortic aneurysm, dissecting aneurysm, or other vascular cause.~• Non-cardiovascular death: Any death not covered by the above definitions such as death caused by infection, malignancy, sepsis, pulmonary causes, accident, suicide or trauma." (NCT02513719)
Timeframe: 0-5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent30

[back to top]

Number of Death

Death includes cardiac death, non-cardiac death and non-coronary death. (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With All Death/All MI/All Revascularization

(NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent66

[back to top]

Number of Participants With All Death/All MI/All Revascularization

(NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent84

[back to top]

Number of Participants With All Death/All MI/All Revascularization

(NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent96

[back to top]

Number of Participants With All Death/All MI/All Revascularization

(NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent106

[back to top]

Number of Participants With All Death/All MI/All Revascularization

(NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent111

[back to top]

Number of Participants With Stent Thrombosis: Subacute

"Stent/Scaffold Thrombosis (per ARC): Stent/Scaffold Thrombosis should be reported as a cumulative value over time and at various individual time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Subacute stent thrombosis : >24 hours to 30 days after stent implantation" (NCT02513719)
Timeframe: >24 hours to 30 days post stent implantation

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent0

[back to top]

Number of Participants With Stent Thrombosis: Late

"Stent/Scaffold Thrombosis (per ARC): Stent/Scaffold Thrombosis should be reported as a cumulative value over time and at various individual time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timings:~Late stent thrombosis : >30 days to 1 year after stent implantation" (NCT02513719)
Timeframe: 30 days to 1 year post stent implantation

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent1

[back to top]

Number of Participants With Stent Thrombosis: Acute

"Stent/Scaffold Thrombosis (per ARC): Stent/Scaffold Thrombosis should be reported as a cumulative value over time and at various individual time points. Time 0 is defined as the time point after the guiding catheter has been removed and the subject left the catheterization lab.~Timing:~Acute stent thrombosis: 0 to 24 hours after stent implantation" (NCT02513719)
Timeframe: 0-24 hours post stent implantation

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent1

[back to top]

Number of Participants With Non-target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent16

[back to top]

Number of Participants With Non-target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT02513719)
Timeframe: 0 to 5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent58

[back to top]

Number of Participants With Non-target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT02513719)
Timeframe: 0 to 4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent55

[back to top]

Number of Participants With Non-target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT02513719)
Timeframe: 0 to 3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent45

[back to top]

Number of Participants With Non-target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT02513719)
Timeframe: 0 to 2 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent42

[back to top]

Number of Participants With Non-target Vessel Revascularization (Non-TVR)

Non Target Vessel Revascularization (Non-TVR) is any revascularization in a vessel other than the target vessel. (NCT02513719)
Timeframe: 0 to 1 year

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent32

[back to top]

Number of Participants With Myocardial Infarction

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0-5 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent5

[back to top]

Number of Participants With Myocardial Infarction

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0-4 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With Myocardial Infarction

"Myocardial Infarction (MI) - Q wave MI: Development of new, pathological Q wave on the ECG.~-Non-Q wave MI: Elevation of CK levels to ≥ two times the upper limit of normal (ULN) with elevated CK-MB in the absence of new pathological Q waves." (NCT02513719)
Timeframe: 0-3 years

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent4

[back to top]

Number of Participants With All Death/All MI/All Revascularization

(NCT02513719)
Timeframe: 0 to 8 months

InterventionParticipants (Count of Participants)
XIENCE PRIME SV Everolimus Eluting Coronary Stent38

[back to top]

Number of Participants Who Experienced Dose-limiting Toxicities

Evaluate the number of participants who had a dose limiting toxicity with the combination of letrozole with everolimus and TRC105 per treatment arm (NCT02520063)
Timeframe: 4 weeks

Interventionparticipants (Number)
Phase I Cohort 11
Phase I Cohort 20

[back to top]

Number of Patients With Tumor Volume Reduction Greater Than 25%

Measuring the efficacy and tolerability of everolimus by measuring the number of patients with tumor volume reduction greater than 25% in sporadic AMLs as measured by DCE MRI (NCT02539459)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Treatment (Everolimus)5

[back to top]

Safety and Tolerability of Everolimus in Patients With Sporadic AML

Number of participants with treatment-related adverse events requiring dose reduction or study withdrawal as assessed by Common Toxicity Criteria for Adverse Effects (CTCAE v4.0) (NCT02539459)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Treatment (Everolimus)10

[back to top]

Number of Participants Who Progressed

"The PFS is defined as the time elapsed between treatment initiation and tumor progression or death from any cause, with censoring of patients who are lost to follow-up. Progression is defined using the Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1): At least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progression)." (NCT02560012)
Timeframe: From date of enrollment until the date of first documented progression, date of death from any cause, or date that the study was stopped, whichever came first, an average of 16 months

InterventionParticipants (Count of Participants)
Personalized Therapy0

[back to top]

Phase 1b: Terminal Elimination Rate Constant (λz) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The λz is the apparent elimination rate constant obtained by linear regression of three or more log-transformed data points in the terminal phase (not including Cmax). (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,
Intervention1/h (Mean)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus0.1250.0870
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel0.1010.137
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel0.1950.149
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel0.1350.118
Cohort 4 (CRC): Ibrutinib 560 mg + CetuximabNA0.0897
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab0.1320.0942

[back to top]

Phase 1b: Area Under the Concentration-Time Curve to Last Observed Time Point (AUClast) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The AUClast was calculated by the linear trapezoidal method. (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,,
Interventionng∙h/mL (Mean)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus1316548
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus25681810
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel1180859
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel32272380
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel11071165
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab8471527
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab26472178

[back to top]

Phase 1b: Apparent Total Clearance at Steady-State (CLss/F) for Ibrutinib in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. Apparent total CLss/F (Cycle 2 Day 1) was calculated as dose/AUC0-24h. (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

InterventionL/h (Mean)
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus1103
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus622
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel994
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel1067
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel1102
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab645
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab595

[back to top]

Phase 1b: Terminal Elimination Rate Constant (λz) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The λz is the apparent elimination rate constant obtained by linear regression of three or more log-transformed data points in the terminal phase (not including Cmax). (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

Intervention1/h (Mean)
Ibrutininb
Cohort 1 (RCC): Ibrutinib 560 mg + EverolimusNA

[back to top]

Phase 1b: Terminal Elimination Half-Life (t1/2term) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The apparent t1/2term was calculated by ln(2)/λz, where λz is the apparent elimination rate constant obtained by linear regression of three or more log-transformed data points in the terminal phase (not including Cmax). (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,,
Interventionhours (Mean)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 560 mg + EverolimusNANA
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus6.288.00
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel7.245.08
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel4.316.79
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel5.546.48
Cohort 4 (CRC): Ibrutinib 560 mg + CetuximabNA8.10
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab5.528.02

[back to top]

Phase 1b: Observed Maximum Concentration (Cmax) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The Cmax was noted as observed. (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,,
Interventionng/mL (Mean)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus17251.2
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus351147
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel260127
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel424198
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel164106
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab172154
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab371177

[back to top]

Phase 1b: Disease Control Rate (DCR) in Cohorts 1 to 6

DCR is is defined as the percentage of participants who have a best response of PR, CR, or stable disease (SD) to therapy in accordance with RECIST 1.1 criteria. CR: The disappearance of all target and non-target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Normalization of tumor marker level, if relevant. All lymph nodes must be non-pathological in size (< 10 mm short axis). PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD since the treatment started (baseline or after). For SD, tthere was no need for confirmation by a subsequent imaging assessment. (NCT02599324)
Timeframe: Maximum time on study (Phase 1b only) for Cohort 1 was 37.4 months; for Cohort 2 was 44.7 months; for Cohort 3 was 41.9 months; for Cohort 4 was 34.2 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months.

Interventionpercentage of participants (Number)
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus66.7
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus71.4
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel75.0
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel50.0
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel77.8
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab75.0
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab80.0
Cohort 5 (UC): Ibrutinib 840 mg37.5
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab76.9

[back to top]

Phase 1b: Number of Participants With Dose-Limiting Toxicities (DLTs) in Cohorts 1 to 6

A DLT was defined as any Grade 3 (severe) or higher non-hematologic or Grade 4 (life-threatening) hematologic adverse event (AE) occurring during the DLT observation period that was considered to be at least possibly related to the study treatment (ibrutinib or drug combination). (NCT02599324)
Timeframe: 21 days after the initiation of therapy at the start of Cycle 1

InterventionParticipants (Count of Participants)
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus0
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus1
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel0
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel0
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel2
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab0
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab0
Cohort 5 (UC): Ibrutinib 840 mg0
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab0

[back to top]

Phase 1b: ORR in Cohorts 1 to 6

ORR is defined as the percentage of participants who have a best response of partial response (PR) or complete response (CR) to therapy in accordance with RECIST 1.1 criteria. CR: The disappearance of all target and non-target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Normalization of tumor marker level, if relevant. All lymph nodes must be non-pathological in size (< 10 mm short axis). PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. (NCT02599324)
Timeframe: Maximum time on study (Phase 1b only) for Cohort 1 was 37.4 months; for Cohort 2 was 44.7 months; for Cohort 3 was 41.9 months; for Cohort 4 was 34.2 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months.

Interventionpercentage of participants (Number)
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus0
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus0
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel50.0
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel20.0
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel11.1
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab12.5
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab0
Cohort 5 (UC): Ibrutinib 840 mg12.5
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab38.5

[back to top]

Phase 1b/2 RP2D: DCR in Cohorts 1 to 6

DCR is is defined as the percentage of participants who have a best response of PR, CR, or SD to therapy in accordance with RECIST 1.1 criteria. CR: The disappearance of all target and non-target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Normalization of tumor marker level, if relevant. All lymph nodes must be non-pathological in size (< 10 mm short axis). PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD since the treatment started (baseline or after). For SD, tthere was no need for confirmation by a subsequent imaging assessment. (NCT02599324)
Timeframe: Maximum time on study (Phase 1b/2 RP2D) for Cohort 1 was 37.4 months; for Cohort 2 was 44.7 months; for Cohort 3 was 41.9 months; for Cohort 4 was 23.5 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months. (Reverse Kaplan-Meier estimates)

Interventionpercentage of participants (Number)
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus80.6
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel66.7
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel74.4
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab83.0
Cohort 5 (UC): Ibrutinib 840 mg48.3
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab71.4

[back to top]

Phase 1b/2 RP2D: Duration of Response (DOR) in Cohorts 1 to 6

"DOR is defined for confirmed responders (PR or better) as time from the date of initial response (PR or better) to the date of first documentation of PD (according to RECIST 1.1) or death, whichever occurs first, regardless of use of subsequent anti-cancer treatment. Confirmed responders without documentation of PD or death or with unknown status at the data extraction were censored at the last adequate post-baseline disease assessment showing no evidence of PD. PD was defined as at least a 20% increase in the size of target lesions with an absolute increase of at least 5 mm, unequivocal progression of existing non-target lesions, or the appearance of one or more new lesions.~Per protocol, participants in Phase 1b receiving the Phase 2 RP2D and participants in Phase 2 RP2D were analyzed together." (NCT02599324)
Timeframe: Maximum time on study (Phase 1b/2 RP2D) for Cohort 1 was 37.4 months; for Cohort 2 was 44.7 months; for Cohort 3 was 41.9 months; for Cohort 4 was 23.5 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months. (Reverse Kaplan-Meier estimates)

Interventionmonths (Median)
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus3.1
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel4.4
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel5.5
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab11.1
Cohort 5 (UC): Ibrutinib 840 mg3.5
Cohort 6 (UC): Ibrutinib 560 mg + PembrolizumabNA

[back to top]

Phase 1b/2 RP2D: ORR in Cohorts 1 and 2

ORR is defined as the percentage of participants who have a best response to therapy of PR or CR in accordance with RECIST 1.1 criteria. CR: The disappearance of all target and non-target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Normalization of tumor marker level, if relevant. All lymph nodes must be non-pathological in size (< 10 mm short axis). PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. (NCT02599324)
Timeframe: Maximum time on study for Cohort 1 (Phase 1b/2 RP2D) was 37.4 months; for Cohort 2 (Phase 1b/2 RP2D) was 44.7 months. (Reverse Kaplan-Meier estimates)

Interventionpercentage of participants (Number)
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus2.8
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel26.3

[back to top]

Phase 1b/2 RP2D: Overall Response Rate (ORR) as Assessed by Investigator in Cohorts 3 to 6

ORR is defined as the percentage of participants who have a best response of partial response (PR) or complete response (CR) to therapy in accordance with RECIST 1.1 criteria. CR: The disappearance of all target and non-target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Normalization of tumor marker level, if relevant. All lymph nodes must be non-pathological in size (< 10 mm short axis). PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. (NCT02599324)
Timeframe: Maximum time on study (Phase 1b/2 RP2D) for Cohort 3 was 41.9 months; for Cohort 4 was 23.5 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months.

Interventionpercentage of participants (Number)
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel17.9
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab14.9
Cohort 5 (UC): Ibrutinib 840 mg6.9
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab35.7

[back to top]

Phase 1b/2 RP2D: Overall Survival (OS) in Cohorts 1 to 6

OS is defined as the time from the date of first dose of study treatment to the date of death from any cause. Subjects who were not known to have died at the data extraction will be censored at date last known alive. (NCT02599324)
Timeframe: Maximum time on study (Phase 1b/2 RP2D) for Cohort 1 was 37.4 months; for Cohort 2 was 44.7 months; for Cohort 3 was 41.9 months; for Cohort 4 was 23.5 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months. (Reverse Kaplan-Meier estimates)

Interventionmonths (Median)
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus21.0
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel8.2
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel7.3
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab15.0
Cohort 5 (UC): Ibrutinib 840 mg8.2
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab15.7

[back to top]

Phase 1b/2 RP2D: PFS in Cohorts 3 to 6

PFS is defined as the time from the date of first dose of study treatment to the date of first documentation of PD or date of death from any cause, whichever occurs first, regardless of the use of subsequent anti-cancer treatment. PD was defined in accordance with RECIST 1.1 criteria. (NCT02599324)
Timeframe: Maximum time on study (Phase 1b/2 RP2D) for Cohort 3 was 41.9 months; for Cohort 4 was 23.5 months; for Cohort 5 was 17.3 months; for Cohort 6 was 20.1 months. (Reverse Kaplan-Meier estimates)

Interventionmonths (Median)
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel4.0
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab5.4
Cohort 5 (UC): Ibrutinib 840 mg1.6
Cohort 6 (UC): Ibrutinib 560 mg + Pembrolizumab2.9

[back to top]

Phase 1b/2 RP2D: Progression-Free Survival (PFS) as Assessed by Investigator in Cohorts 1 and 2

PFS is defined as the time from the date of first dose of study treatment to the date of first documentation of progressive disease (PD) or date of death from any cause, whichever occurs first, regardless of the use of subsequent anti-cancer treatment. PD was defined in accordance with Response Evaluation Criteria In Solid Tumors (RECIST) criteria version 1.1. PD: At least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study), and an absolute increase of ≥ 5 mm, or unequivocal progression of existing non-target lesions or the appearance of new lesions. (NCT02599324)
Timeframe: Maximum time on study for Cohort 1 (Phase 1b/2 RP2D) was 37.4 months; for Cohort 2 (Phase 1b/2 RP2D) was 44.7 months.

Interventionmonths (Median)
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus5.6
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel4.1

[back to top]

Phase 1b: Area Under the Concentration-Time Curve From Time 0 to Hour 24 (AUC0-24h) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The AUC0-24h was calculated by the linear trapezoidal method. (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,,
Interventionng∙h/mL (Mean)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus1467646
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus25681810
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel21941146
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel35032604
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel12531254
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab9801527
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab28072178

[back to top]

Phase 1b: Time of Last Observed Concentration (Tlast) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The tlast was noted as observed. (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,,
Interventionhours (Median)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus15.015.0
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus24.024.0
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel24.024.0
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel24.024.0
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel24.024.0
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab24.024.0
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab24.024.0

[back to top]

Phase 1b: Time to Cmax (Tmax) for Ibrutinib and Its Metabolite PCI-45227 in Cohorts 1 to 4

Actual collection times relative to ibrutinib administration were used for the calculation of pharmacokinetic parameters of ibrutinib and PCI-45227. For actual predose collection times that were < 0, these values were set equal to 0. Predose concentrations were applied as 24 hour concentrations in order to calculate steady-state (Cycle 2 Day 1) pharmacokinetic parameters for ibrutinib and PCI-45227. The tmax was noted as observed. (NCT02599324)
Timeframe: Cycle 2 Day 1: predose, 1 h ± 15 min, 2 h ± 15 min, 4 h ± 15 min, 6 h ± 15 min postdose

,,,,,,
Interventionhours (Median)
IbrutininbPCI-45227
Cohort 1 (RCC): Ibrutinib 560 mg + Everolimus3.013.03
Cohort 1 (RCC): Ibrutinib 840 mg + Everolimus2.003.93
Cohort 2 (UC): Ibrutinib 560 mg + Paclitaxel3.714.00
Cohort 2 (UC): Ibrutinib 840 mg + Paclitaxel4.004.00
Cohort 3 (GA): Ibrutinib 560 mg + Docetaxel3.894.00
Cohort 4 (CRC): Ibrutinib 560 mg + Cetuximab2.892.99
Cohort 4 (CRC): Ibrutinib 840 mg + Cetuximab2.034.00

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs)

An AE was defined as any untoward medical occurrence in participants administered a pharmaceutical product; the untoward medical occurrence does not necessarily have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal (investigational) product whether or not it is related to the medicinal product. TEAE was defined as the event that occur after administration of the first dose of study drug and through 30 days after the last dose of study drug. (NCT02724020)
Timeframe: From first dose of study drug through 30 days after the last dose of study drug (approximately up to 31 months)

InterventionParticipants (Count of Participants)
Arm A: Single-agent Everolimus 10 mg QD32
Arm B: Single-agent MLN0128 30 mg QW30
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD31

[back to top]

Objective Response Rate (ORR)

ORR was defined as the percentage of participants among response evaluable analysis set who achieve a best overall response of complete response (CR) or partial response (PR) based on investigators assessment of response following RECIST 1.1. CR was defined as disappearance of all target lesions, non-target lesions, no new lesions, and normalization of tumor marker level. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, no progression in non-target lesion, and no new lesions. (NCT02724020)
Timeframe: From first dose of study drug to disease progression or death (up to 51 months)

Interventionpercentage of participants (Number)
Arm A: Single-agent Everolimus 10 mg QD15.6
Arm B: Single-agent MLN0128 30 mg QW0
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD6.5

[back to top]

Overall Survival (OS)

Overall survival in months was defined as the time from the date of randomization to the date of death. (NCT02724020)
Timeframe: From first dose of study drug through 30 days after the last dose of study drug (up to 51 months)

Interventionmonths (Median)
Arm A: Single-agent Everolimus 10 mg QD22.4
Arm B: Single-agent MLN0128 30 mg QW16.2
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD18.1

[back to top]

Progression-Free Survival (PFS)

PFS was defined as the time from the date of randomization to the date of first documentation of progressive disease (PD) or death due to any cause, whichever occurs first. Per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 criteria. PD was defined as at least a 20% increase in the sum of the longest diameter (LD) of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. (NCT02724020)
Timeframe: From first dose of study drug up to disease progression or death, assessed up to 43 months

Interventionmonths (Median)
Arm A: Single-agent Everolimus 10 mg QD3.8
Arm B: Single-agent MLN0128 30 mg QW3.6
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD3.1

[back to top]

Time-to-progression (TTP)

TTP in months is defined as the time from the date of randomization to the date of first documentation of progression. Per RECIST v1.1, PD was defined as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. (NCT02724020)
Timeframe: From first dose of study drug up to disease progression or death (up to 51 months)

Interventionmonths (Median)
Arm A: Single-agent Everolimus 10 mg QD3.8
Arm B: Single-agent MLN0128 30 mg QW3.5
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD3.7

[back to top]

Clinical Benefit Rate (CBR)

CBR is defined as the percentage of participants who achieve a best response of CR, PR or stable disease (SD) of any duration. CR was defined as disappearance of all target lesions, non-target lesions, no new lesions, and normalization of tumor marker level. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, no progression in non-target lesion, and no new lesions. SD was defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT02724020)
Timeframe: From first dose of study drug up to disease progression or death (up to 51 months)

Interventionpercentage of participants (Number)
Arm A: Single-agent Everolimus 10 mg QD62.5
Arm B: Single-agent MLN0128 30 mg QW50.0
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD54.8

[back to top]

CBR With SD Duration of at Least 16 Weeks

CBR with SD duration of at least 4 months (CBR-16) was defined as the percentage of participants who achieve CR or PR of any duration or have SD with duration of at least 16 weeks. CR was defined as disappearance of all target lesions, non-target lesions, no new lesions, and normalization of tumor marker level. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, no progression in non-target lesion, and no new lesions. SD was defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT02724020)
Timeframe: Up to Week 16

Interventionpercentage of participants (Number)
Arm A: Single-agent Everolimus 10 mg QD40.6
Arm B: Single-agent MLN0128 30 mg QW25.0
Arm C: Combination of MLN0128 4 mg QD + MLN1117 200 mg QD29.0

[back to top]

Duration of Overall Response (DOR) by Group - Phase II

Patients whose best response is complete response (CR) or partial response (PR). The start date is the date of first documented response (CR or PR) and the end date is the date of first documented progression or death due to underlying cancer. (NCT02732119)
Timeframe: Baseline up to approximately 16 months

Interventionmonths (Median)
Group 114.6
Group 26.4

[back to top]

Participants With Dose Limiting Toxicities by Preferred Term in Cycle 1 (28 Days) - in Phase I

A dose-limiting toxicity (DLT) is defined as an adverse event or abnormal laboratory value assessed as having a reasonably possible relationship to the study medication(s) and is unrelated to disease, disease progression, inter-current illness, or concomitant medications that occurs within the first 28 days of treatment (cycle 1) and meets any of the criteria defined in the protocol. National Cancer Institute Common Terminology Criteria for Adverse events (NCI CTCAE) version 4.03 will be used for all grading. (NCT02732119)
Timeframe: Baseline up to 28 days

,,
InterventionParticipants (Count of Participants)
Febrile neutropenia Grade 3Neutropenia Grade 4Thrombocytopenia Grade 4Cardiac tamponade Grade 4Diarrhea Grade 3Hyperglycemia Grade 4
Cohort A100110
Cohort B111000
Cohort C000001

[back to top]

Everolimus Pharmacokinetic Plasma Concentrations by Cohort - Phase I

Plasma concentrations; below limit of quantitation values set to zero (NCT02732119)
Timeframe: Cycle 1,2: Day 1 and 15 - pre-dose, 2 and 4 hour post dose, Cycle 3 , Day 1 - pre-dose

,,
Interventionng/mL (Median)
Cycle 1, Day 15 Pre-doseCycle 1, Day 15 2 H, post doseCycle 1, Day 15 4 H, post doseCycle 2, Day 1 Pre-doseCycle 2, Day 15 Pre-doseCycle 2, Day 15 2 H post-doseCycle 2, Day 15 4 H post-doseCycle 3, Day 1 4 H pre-dose
Cohort A7.8215.217.76.475.8921.813.74.7
Cohort B8.311716.26.224.2614.59.136.97
Cohort C8.2836.221.57.767.1722.126.96.33

[back to top]

Everolimus Pharmacokinetic Plasma Concentrations - Phase II

Plasma concentrations; below limit of quantitation values set to zero (NCT02732119)
Timeframe: Cycle 1,2: Day 1 and 15 - pre-dose, 2 and 4 hour post dose, Cycle 3 , Day 1 - pre-dose

,
Interventionng/mL (Median)
Cycle 1, Day 15 pre doseCycle 1, Day 15 2 H post-doseCycle 1, Day 15 4 H post-doseCycle 2 Day 1 pre-doseCycle 2, Day 15 pre-doseCycle 2, Day 15 2 H post-doseCycle 2, Day 15 4 H post-doseCycle 3, Day 1 pre-dose
Group 1214644583199245652642193
Group 2132372323125170442406198

[back to top]

Clinical Benefit Rate as Per Central Review by Group- Phase II

"Clinical Benefit Rate (CBR) is defined as the percentage of participants with a complete response (CR) or partial response (PR) or with stable disease (SD) as per Response Evaluation Criteria in Solid Tumors (RECIST) V1.1 or Non-Complete Response or Non-Progressive Disease (NCRNPD) during first 24 weeks of first dose. CR=disappearance of all non-nodal target lesions, PR=at least a 30% decrease in the sum of diameter of all target lesions, SD=neither sufficient shrinkage to qualify for PR or CR nor an increase in lesions which would qualify for progressive disease (PD), PD=at least a 20% increase in the sum of diameter of all target lesions.~The hypothesis was to be rejected if the lower limit of the 95% Confidence Interval for Clinical Benefit Rate (CBR) was greater than 10%." (NCT02732119)
Timeframe: From baseline up to 24 weeks

,
Interventionpercentage of participants (Number)
Overall response rate (CR+PR):CBR - CR+PR+SD (NCRNPD) by 24 weeksDisease control rate (CR+PR+SD(NCRNPD)
Group 16.565.265.2
Group 29.459.459.4

[back to top]

Clinical Benefit Rate as Per Central Review by Dose Cohort - Phase I

Clinical Benefit Rate (CBR) is defined as the percentage of patients with a complete response (CR) or partial response (PR) or with stable disease (SD) at 24 weeks as per Response Evaluation Criteria in Solid Tumors (RECIST) V1.1 or Non-Complete Response Non-Progressive Disease (NCRNPD) up to Week 24 and at Week 24. (NCT02732119)
Timeframe: Baseline up to 24 weeks and at week 24

,
Interventionpercentage of participants (Number)
Overall response rate (CR+PR):CBR - CR+PR+SD (NCRNPD) by 24 weeksCBR - CR+PR+SD (NCRNPD) at 24 weeksDisease control rate (CR+PR+SD(NCRNPD)
Cohort B10.080.00.080.0
Cohort C28.6100.028.6100

[back to top]

Clinical Benefit Rate as Per Central Review by Dose Cohort - Phase I

Clinical Benefit Rate (CBR) is defined as the percentage of patients with a complete response (CR) or partial response (PR) or with stable disease (SD) at 24 weeks as per Response Evaluation Criteria in Solid Tumors (RECIST) V1.1 or Non-Complete Response Non-Progressive Disease (NCRNPD) up to Week 24 and at Week 24. (NCT02732119)
Timeframe: Baseline up to 24 weeks and at week 24

Interventionpercentage of participants (Number)
CBR - CR+PR+SD (NCRNPD) by 24 weeksCBR - CR+PR+SD (NCRNPD) at 24 weeksDisease control rate (CR+PR+SD(NCRNPD)
Cohort A62.5062.5

[back to top]

Best Overall Responses (BOR) Summary Table as Per Central Review by Group - Phase II

Complete response (CR) or partial response (PR), or stable disease (SD) at 24 weeks as per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 or Non-CR Non-PD (NCRNPD) at Week 24. (NCT02732119)
Timeframe: Baseline up to 24 weeks and at 24 weeks

,
InterventionParticipants (Count of Participants)
Participants with measurable disease at baselineParticipants with non-measurable disease at baselineComplete response (CR)Partial Response (PR)Stable disease (SD)Progressive disease (PD)NCRNPD - Non-complete response non-progressive diseaseUnknown
Group 13511031714102
Group 2211103612101

[back to top]

Time to Definitive Deterioration of ECOG Performance Status in One Category of the Score by Group - Phase II

Time to definitive deterioration of ECOG performance status in one category of score is defined as the time from the date of randomization to the date of event, which is defined as at least one score lower than the baseline. (NCT02732119)
Timeframe: Baseline up to approximately 8 months

Interventionmedian (Number)
Group 1NA
Group 212.0

[back to top]

Summary of Progression-free Survival (PFS) (Months), as Per Central Review by Group - Phase II

Progression is defined as <= 12 weeks after randomization/ start of treatment (and not qualifying for CR, PR or SD). (NCT02732119)
Timeframe: Baseline up to approximately 32 months

Interventionmonths (Median)
Group 18.0
Group 24.7

[back to top]

Overall Survival (OS) by Group - Phase II

Overall survival is the time from date of first treatment to the date of death due to any cause. If a patient is not known to have died, survival will be censored at the last date of contact. (NCT02732119)
Timeframe: Baseline up to approximately 32 months

Interventionmonths (Median)
Group 127.4
Group 2NA

[back to top]

Best Overall Responses (BOR) Summary Table as Per Central Review by Cohort - Phase I

Complete response (CR) or partial response (PR), or stable disease (SD)as per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 or Non-CR Non-PD (NCRNPD) (NCT02732119)
Timeframe: From baseline up to 24 weeks

,,
InterventionParticipants (Count of Participants)
Participants with measurable disease at baselineParticipants with non-measurable disease at baselineComplete response (CR)Partial Response (PR)Stable disease (SD)Progressive disease (PD)NCRNPD - Non-complete response non-progressive diseaseUnknown
Cohort A53003320
Cohort B73016012
Cohort C43022030

[back to top]

Amount of Treg Cells Observed in Peripheral Blood in Patients With and Without Rejection

flow cytometry immunophenotyping and gene expression microassays to assess amount of Treg cells and mRNA in peripheral blood in patients with and without rejection (after Tacc withdrwal) (NCT02736227)
Timeframe: Six months post transplantation

Interventionpercentage of CD4 positive cell (Mean)
Graft Rejection After Tacrolimus Was Discontinued (REJ)10
No Graft Rejection After Tacrolimus Weaning (Non REJ)5

[back to top]

Progression-free Survival (PFS) by Independent Imaging Review (IIR)

PFS assessed by IIR was defined as the time from the date of randomization to the date of the first documentation of progressive disease (PD) or death (whichever occurred first) using Response Evaluation Criteria in Solid Tumors (RECIST 1.1). PD was defined as at least a 20 percent (%) increase or 5 millimeter (mm) increase in the sum of diameters of target lesions (taking as reference the smallest sum on study) recorded since the treatment started or the appearance of 1 or more new lesions. PFS was analyzed using Kaplan-Meier method. (NCT02811861)
Timeframe: From the date of randomization to the date of the first documentation of PD or date of death, whichever occurred first or up to data cutoff date 28 Aug 2020 (up to approximately 46 months)

Interventionmonths (Median)
Lenvatinib 18 mg Plus Everolimus 5 mg14.7
Lenvatinib 20 mg Plus Pembrolizumab 200 mg23.9
Sunitinib 50 mg9.2

[back to top]

Measured Glomerular Filtration Rate (mGFR)

Renal function as assessed by measured Glomerular Filtration Rate (mGFR) (Cr-EDTA or iohexol clearance). Baseline Visit 1 and Patient 4252 excluded from the intent treat analysis set. (NCT02864706)
Timeframe: at the 5-7 year follow-up visit

InterventionmL/min/1.73m2 (Least Squares Mean)
Everolimus74.7
Control62.4

[back to top]

Progression of Cardiac Allograft Vasculopathy (CAV) Recorded by Intravascular Ultrasound (IVUS)

"Cardiac Allograft Vasculopathy (CAV) was defined as mean maximal intimal thickness (MIT)~≥0.5 mm, measured for the entire matched pullback recording by intravascular ultrasound (IVUS). The incidence of CAV at 5-7 years was compared between groups using the Cochran-Mantel-Haenszel test with stratification according to baseline distribution of CAV incidence." (NCT02864706)
Timeframe: within 5-7 years

Interventionmm (Mean)
Everolimus0.13
Control0.23

[back to top]

Change From Baseline in Visual Analog Scale (VAS)

"Change in visual analog scale (VAS) from baseline to the 5 to 7 Year follow up visit.~0 is no pain; and 10 is the worst possible pain" (NCT02864706)
Timeframe: baseline, at the 5-7 year visit

Interventionmm (Mean)
Everolimus35.6
Control34.0

[back to top]

Change From Baseline in the Euro Quality of Life 5D

"Change from baseline in Euro Quality of Life-5D from 3 Year Follow-Up to 5 to 7 Year Follow-Up Baseline Visit 1 (ITT Set)~Euro Quality of Life 5D (EQ-5D): is a descriptive system of healthrelated quality of life states consisting of five dimensions (mobility, self-care, usual activities, pain/discomfort and anxiety/depression) each of which can be assessed as one of three levels of severity (no problems/some or moderate problems/extreme problems). A Visual Analogue Scale (VAS)-scale is also included in the EQ-5D questionnaire.~The EQ-5D index is calculated based on the United Kingdom Time Trade-Off (TTO) N3 value set which converts the five dimensions scores into a single measure with a possible range from -0.163 (worst possible health state) to +1 (perfect health). A positive change from baseline indicates an improvement in Quality of Life." (NCT02864706)
Timeframe: Baseline, 5-7 year visit

Interventionscores on the scale (Mean)
Everolimus0.2323
Control0.2982

[back to top]

Number of Participants With Beck Depression Inventory (BDI)

Beck Depression Inventory (BDI) Score has the following categories of depression. Normal, Mild, Moderate Severe and Missing. (NCT02864706)
Timeframe: at the 5-7 year visit

,
InterventionParticipants (Count of Participants)
NormalMildModerateSevereMissing
Control134505
Everolimus156213

[back to top]

Quality of Life by SF-36 Change From Pre-transplantation to 5-7 Year Follow-up

This Quality of life Short Form Survey with 36 items (Minnesota Living with Heart Failure Questionnaire)was administered to patients pre-transplantation and after transplantation at the 5-7 year visit. This data represents the change. The survey consist of scores on a scale. Each form is scaled from 0 t 100. 0 = maximum disability and 100 equals no disability. (NCT02864706)
Timeframe: at the 5-7 year visit

,
Interventionscores on a scale (Mean)
Physical Health SummaryMental Health SummaryPhysical FunctioningRole PhysicalBodily PainGeneral HealthVitalitySocial FunctioningRole EmotionalMental Health
Control13.215.340.855.17.223.428.943.942.814.4
Everolimus16.810.436.750.210.325.730.039.923.88.6

[back to top]

Percent of Participants With Incidence of Coronary Allograft Vasculopathy (CAV)

"Cardiac Allograft Vasculopathy (CAV) was defined as mean maximal intimal thickness (MIT)~≥0.5 mm, measured for the entire matched pullback recording by intravascular ultrasound (IVUS). The incidence of CAV at 5-7 years was compared between groups using the Cochran-Mantel-Haenszel test with stratification according to baseline distribution of CAV incidence." (NCT02864706)
Timeframe: at the 5-7 year follow-up

Interventionpercent of participants (Number)
Everolimus53
Control74

[back to top]

Myocardial Structure and Function

Myocardial structure and function by echocardiography assessment measured by ventricular end systolic diameter. (NCT02864706)
Timeframe: within 5-7 years

,
Interventioncm (Mean)
LVESD (left ventricular end systolic diameter)LVEDD (left ventricular end diastolic diameter)
Control3.14.9
Everolimus3.14.7

[back to top]

Duration of Response (DOR) [Phase 2a]

Response will be assessed among participants eligible for the phase IIa part of the study who received at least one dose of the study drugs at the MTD/RP2D and have measurable disease at screening. DOR defined based on the duration of stable disease. (NCT02871791)
Timeframe: Disease evaluations were performed every 8 weeks (within 24 weeks of initiation of study treatment) or every 12 weeks (greater than 24 weeks of initiation of study treatment). Treatment duration has a median of 111 days and maximum of 681 days.

Interventionmonths (Median)
Palbociclib, Everolimus, Exemestane4.24

[back to top]

Median Progression Free Survival (PFS) [Phase 2a]

Progression-free survival based on the Kaplan-Meier method is defined as the duration of time from study entry to documented disease progression (PD) or death. Per RECIST 1.1 criteria: progressive disease (PD) is at least a 20% increase in the sum of longest diameter (LD) of target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. PD for the evaluation of non-target lesions is the appearance of one or more new lesions and/or unequivocal progression of non-target lesions. (NCT02871791)
Timeframe: Disease is evaluated and followed-up every 8 weeks, with median of 20.04 months and maximum of 35.64 months.

Interventionmonths (Median)
Palbociclib, Everolimus, Exemestane3.8

[back to top]

Overall Response Rate (ORR) [Phase 2a]

The objective response rate (ORR) was defined as the proportion of participants achieving complete response (CR) or partial response (PR) based on RECIST 1.1 criteria on treatment. (NCT02871791)
Timeframe: Disease evaluations were performed every 8 weeks (within 24 weeks of initiation of study treatment) or every 12 weeks (greater than 24 weeks of initiation of study treatment). Treatment duration has a median of 111 days and maximum of 681 days.

InterventionParticipants (Count of Participants)
Palbociclib, Everolimus, Exemestane0

[back to top]

Clinical Benefit Rate (CBR) [Phase 2a]

CBR is defined as the proportion of participants achieving complete response, partial response or stable disease for more than 6 months (CR+PR+SD ≥ 24 weeks) taking as reference the smallest measurements recorded since the treatment started, including the baseline measurements. (NCT02871791)
Timeframe: Disease evaluations were performed every 8 weeks (within 24 weeks of initiation of study treatment) or every 12 weeks (greater than 24 weeks of initiation of study treatment). Treatment duration has a median of 111 days and maximum of 681 days.

InterventionParticipants (Count of Participants)
Palbociclib, Everolimus, Exemestane6

[back to top]

Disease Control Rate (DCR) [Phase 2a]

The DCR defined as the proportion of patient that has CR+PR+SD>=12 weeks. Response will be assessed among participants eligible for the phase IIa part of the study who received at least one dose of the study drugs at the MTD/RP2D and have measurable disease at screening. (NCT02871791)
Timeframe: Disease evaluations were performed every 8 weeks (within 24 weeks of initiation of study treatment) or every 12 weeks (greater than 24 weeks of initiation of study treatment). Treatment duration has a median of 111 days and maximum of 681 days.

Interventionproportion of patients (Number)
Palbociclib, Everolimus, Exemestane0.563

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of the first dose of study drug until the date of death from any cause. OS was analyzed using Kaplan Meier method. In the absence of death before data cutoff, participants were censored either at the date last known to be alive or the date of data cutoff, whichever came earlier. Participants were followed for survival every 12 weeks after the end of treatment visit. If a clinic visit was not feasible, follow-up information was obtained via telephone or email. (NCT02915783)
Timeframe: From the date of the first dose of study drug until the date of death from any cause (up to approximately 4 years 8 months)

Interventionmonths (Median)
Lenvatinib 18 mg/Day + Everolimus 5 mg/Day16.33

[back to top]

Objective Response Rate (ORR)

ORR was assessed by the investigator based on Response Evaluation Criteria in Solid Tumors (RECIST) version (v) 1.1. ORR was defined as the percentage of participants with confirmed best overall response (BOR) of complete response (CR) or partial response (PR). CR was defined as the disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis to less than (<) 10 millimeters (mm). PR was defined as at least a 30 percent (%) decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. (NCT02915783)
Timeframe: From the date of the first dose of study drug to the date of the first documentation of disease progression or death, whichever occurred first (up to approximately 3 years 9 months)

Interventionpercentage of participants (Number)
Lenvatinib 18 mg/Day + Everolimus 5 mg/Day25.8

[back to top]

Progression-free Survival (PFS)

PFS was assessed by investigator per RECIST v1.1, defined as time from date of first dose of study drug to date of first documentation of progressive disease (PD) or death whichever occurred first. PD: greater than or equal to (>=) 20% increase in sum of diameters of target lesions, reference-smallest sum recorded in study (sum at baseline if that was smallest). Sum of diameters must have absolute increase of >=5 mm. Appearance of >=1 new lesions also considered PD. PFS was analyzed using Kaplan Meier method. PFS was censored on date of last adequate radiologic assessment prior to new anticancer therapy, more than one missed visits, treatment discontinuation, and cutoff date when no PD or death occurred before any of these (on first dose of study treatment if no adequate post baseline tumor assessment was available), per publication by Food and Drug Administration (FDA) 'Guidance for Industry Clinical Trial Endpoints for the Approval of Cancer Drugs and Biologics (2007). (NCT02915783)
Timeframe: From the date of the first dose of study drug to the date of the first documentation of disease progression or death, whichever occurred first (up to approximately 3 years 9 months)

Interventionmonths (Median)
Lenvatinib 18 mg/Day + Everolimus 5 mg/Day13.11

[back to top]

Percent of Participants Experiencing Acute Allograft Rejection

Estimate the composite of treatment failure rate, defined as acute allograft rejection with a Banff grade 1A or higher, graft loss, or death at six months post-conversion, in patients converted to a once-daily immunosuppressant regimen of Envarsus®, everolimus, and prednisone versus patients converted to a twice-daily regimen of Envarsus®, MMF, and prednisone. (NCT02954198)
Timeframe: Baseline to 6 months post conversion

InterventionParticipants (Count of Participants)
Control: Envarsus + MMF0
Intervention: Envarsus + Everoliumus0

[back to top]

Subject Specific Change on Medication Side Effect Scale

Examine subject specific change on a validated Medication Side Effect Scale at the time of the conversion versus six months post-conversion, compared between the two arms. Side effect burden scale is from 0 to 180. A lower score is less side effect burden, a higher score is more side effect burden. (NCT02954198)
Timeframe: Baseline to 6 months post conversion

,
Interventionunits on a scale (Mean)
Baseline6 month post-conversion
Control: Envarsus + MMF7193
Intervention: Envarsus + Everoliumus3738

[back to top]

Self-reported Medication Adherence From Baseline to 6 Months.

Percent of subjects reporting high medication adherence at baseline compared to 6 months post-conversion, using the Morisky Medication Adherence scale (MMAS). The MMAS rates medication adherence on a scale of 0 to 8. 0 is high adherence, 1-2 is medium adherence, and greater than or equal to 3 is low adherence. (NCT02954198)
Timeframe: 6 months post conversion

,
Intervention% of participants (Number)
Baseline6 months post-conversion
Control: Envarsus + MMF8059
Intervention: Envarsus + Everoliumus4547

[back to top]

Percent of Participants Who Experienced Kidney Transplant Graft Loss

Measure and compare time-to-event analysis between the two arms graft loss (time to event analysis) (NCT02954198)
Timeframe: Baseline to 6 months post conversion

InterventionParticipants (Count of Participants)
Control: Envarsus + MMF0
Intervention: Envarsus + Everoliumus0

[back to top]

Percentage of Participants With a ≥ 50% Reduction in Daily Corticosteroid Dose

All corticosteroid dosages prescribed to the participant and all dose changes during the study were to be recorded for assessment of participants with a ≥ 50% reduction in daily corticosteroid dose. (NCT03112603)
Timeframe: from Day 15 up to Day 182

,
Interventionpercentage of participants (Number)
Day 15 to ≤ Day 28Day 29 to ≤ Day 42Day 43 to ≤ Day 56Day 57 to ≤ Day 70Day 71 to ≤ Day 84Day 85 to ≤ Day 98Day 99 to ≤ Day 112Day 113 to ≤ Day 126Day 127 to ≤ Day 140Day 141 to ≤ Day 154Day 155 to ≤ Day 168Day 169 to ≤ Day 182
Best Available Therapy13.233.141.147.951.454.060.466.268.368.371.688.8
Ruxolitinib12.735.048.458.762.369.571.273.272.870.074.681.9

[back to top]

t1/2 of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"t1/2 was defined as the apparent terminal phase disposition half-life of ruxolitinib. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

Interventionhours (Geometric Mean)
Day 1Day 15
Ruxolitinib2.402.32

[back to top]

Tmax of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"tmax was defined as the time to reach the maximum plasma concentration of ruxolitinib. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

Interventionhours (Median)
Day 1Day 15
Ruxolitinib0.8331.00

[back to top]

Vz/F of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"Vz/F was defined as the apparent oral dose volume of distribution of ruxolitinib. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

InterventionLiters (Geometric Mean)
Day 1Day 15
Ruxolitinib54.050.9

[back to top]

Rate of Failure-free Survival (FFS)

Composite time to event endpoint incorporating the following FFS events: (i) relapse or recurrence of underlying disease or death due to underlying disease, (ii) nonrelapse mortality, or (iii) addition or initiation of another systemic therapy for cGvHD. (NCT03112603)
Timeframe: Baseline to when the last participant reached Cycle 7 Day 1 (each cycle was comprised of 4 weeks)

Interventionmonths (Median)
RuxolitinibNA
Best Available Therapy5.7

[back to top]

CL/F of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"CL/F was defined as the oral dose clearance of ruxolitinib. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

InterventionLiters/hour (Geometric Mean)
Day 1Day 15
Ruxolitinib15.615.2

[back to top]

Cmax of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"Cmax was defined as the maximum observed plasma concentration of ruxolitinib. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Day 1Day 15
Ruxolitinib167215

[back to top]

Change From Baseline in Functional Assessment of Cancer Therapy - Bone Marrow Transplantation (FACT-BMT)

"Change from Baseline was calculated as the post-Baseline value minus the Baseline value. The FACT-BMT is a 50-item self-report questionnaire that measures the effect of a therapy on domains including physical, functional, social/family, and emotional well-being, together with additional concerns relevant for bone marrow transplantation participants. The questions were based on a 5-point Likert scale, where 0 corresponds to not at all and 4 corresponds to very much. The higher the final score, the better the quality of life. The FACT-BMT total score ranges from 0 to 148." (NCT03112603)
Timeframe: Baseline; up to Cycle 39 Day 1 (each cycle was comprised of 4 weeks)

,
Interventionscores on a scale (Mean)
Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 5 Day 1Cycle 6 Day 1Cycle 7 Day 1Cycle 9 Day 1Cycle 12 Day 1Cycle 15 Day 1Cycle 18 Day 1Cycle 21 Day 1Cycle 24 Day 1Cycle 27 Day 1Cycle 30 Day 1Cycle 33 Day 1Cycle 36 Day 1Cycle 39 Day 1
Best Available Therapy-0.22-1.82-1.05-0.232.410.851.203.747.588.193.687.845.105.755.674.776.64
Ruxolitinib2.320.621.981.252.914.145.327.265.074.105.245.906.237.535.178.728.65

[back to top]

Change From Baseline in EQ-5D-5L

The EQ-5D-5L is a descriptive classification consisting of five dimensions of health: mobility, self-care, usual activities, anxiety/depression, and pain/discomfort. The five-level version (no problems, slight problems, moderate problems, severe problems, and extreme problems) uses a 5-point Likert scale, with 1 being no problems and 5 being extreme problems. (NCT03112603)
Timeframe: Baseline; up to Cycle 39 Day 1 (each cycle was comprised of 4 weeks)

,
Interventionscores on a scale (Mean)
Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 5 Day 1Cycle 6 Day 1Cycle 7 Day 1Cycle 9 Day 1Cycle 12 Day 1Cycle 15 Day 1Cycle 18 Day 1Cycle 21 Day 1Cycle 24 Day 1Cycle 27 Day 1Cycle 30 Day 1Cycle 33 Day 1Cycle 36 Day 1Cycle 39 Day 1
Best Available Therapy-0.01-0.04-0.01-0.030.01-0.00-0.020.020.030.02-0.000.010.030.010.050.040.01
Ruxolitinib0.030.030.040.020.050.070.070.070.070.070.080.070.060.050.000.060.06

[back to top]

AUClast of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"AUClast was defined as the area under the concentration-time curve up to the last measurable concentration of ruxolitinib. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

Interventionng*hour/mL (Geometric Mean)
Day 1Day 15
Ruxolitinib636945

[back to top]

Best Overall Response (BOR) at Cycle 7 Day 1

BOR was defined as the percentage of participants who achieved an overall response (CR+PR) based on cGvHD disease assessments (National Institutes of Health Consensus Criteria) without the requirement of additional systemic therapies for an earlier progression, mixed response, or non-response at any time point (up to Cycle 7 Day 1 or the start of additional systemic therapy for cGvHD). Scoring of response was relative to the organ score at the time of randomization. CR: complete resolution of all signs and symptoms of cGVHD in all evaluable organs without the initiation or addition of new systemic therapy. PR: improvement in at least one organ (e.g., improvement of 1 or more points on a 4- to 7-point scale, or an improvement of 2 or more points on a 10- to 12-point scale) without progression in other organs or sites, initiation, or addition of new systemic therapies. This analysis was based on the primary cutoff date (May 2020). (NCT03112603)
Timeframe: up to Cycle 7 Day 1 (each cycle was comprised of 4 weeks)

Interventionpercentage of participants (Number)
Ruxolitinib76.4
Best Available Therapy60.4

[back to top]

BOR During Cross-over Treatment With Ruxolitinib

BOR was defined as the percentage of participants who achieved an overall response (CR+PR) based on cGvHD disease assessments (National Institutes of Health Consensus Criteria) without the requirement of additional systemic therapies for an earlier progression, mixed response, or non-response at any time point (up to Cycle 7 Day 1 or the start of additional systemic therapy for cGvHD). Scoring of response was relative to the organ score at the time of randomization. CR: complete resolution of all signs and symptoms of cGVHD in all evaluable organs without the initiation or addition of new systemic therapy. PR: improvement in at least one organ (e.g., improvement of 1 or more points on a 4- to 7-point scale, or an improvement of 2 or more points on a 10- to 12-point scale) without progression in other organs or sites, initiation, or addition of new systemic therapies. (NCT03112603)
Timeframe: from Crossover Cycle 1 Day 1 to any time point up to and including Crossover Cycle 7 Day 1 (each cycle was comprised of 4 weeks)

Interventionpercentage of participants (Number)
Ruxolitinib Cross-Over Period81.4

[back to top]

Duration of Response Through Study Completion

DOR was defined as the time from first response until cGvHD progression, death, or the date of change/addition of systemic therapies for cGvHD and as assessed for responders only. Response was based on cGvHD disease assessments (National Institutes of Health consensus criteria). Duration of response was evaluated in participants who achieved a CR or PR at or before Cycle 7 Day 1. The analysis included a larger number of participants than the number of participants who achieved CR or PR at Cycle 7 Day 1 (82 ruxolitinib and 42 BAT) because the analysis took into account not only those participants who achieved CR or PR at Cycle 7 Day 1, but also participants who achieved CR or PR before Cycle 7 Day 1 but who may have lost their response at Cycle 7 Day 1. For this reason, the number of participants in this analysis does not align with the best overall response (BOR) at Cycle 7 Day 1. This analysis was based on the cutoff date upon study completion (December 2022). (NCT03112603)
Timeframe: from first response to LPLV (approximately 5 years)

InterventionMonths (Median)
RuxolitinibNA
Best Available Therapy6.4

[back to top]

Efficacy of Ruxolitinib Versus Investigator's Choice Best Available Therapy (BAT) in Participants With Moderate or Severe Steroid Refractory Chronic Graft Versus Host Disease (SR-cGvHD) Assessed by Overall Response Rate (ORR) at the Cycle 7 Day 1 Visit

ORR was defined as the percentage of participants in each arm demonstrating a complete response (CR) or partial response (PR) based on chronic GvHD (cGvHD) disease assessments (National Institutes of Health Consensus Criteria) without the requirement of additional systemic therapies for an earlier progression, mixed response, or non-response. Scoring of response was relative to the organ score at the time of randomization. CR: complete resolution of all signs and symptoms of cGVHD in all evaluable organs without the initiation or addition of new systemic therapy. PR: improvement in at least one organ (e.g., improvement of 1 or more points on a 4- to 7-point scale, or an improvement of 2 or more points on a 10- to 12-point scale) without progression in other organs or sites, initiation, or addition of new systemic therapies. (NCT03112603)
Timeframe: Cycle 7 Day 1 (each cycle was comprised of 4 weeks)

Interventionpercentage of participants (Number)
Ruxolitinib49.7
Best Available Therapy25.6

[back to top]

Number of Participants With Any Treatment-emergent Adverse Event (TEAE)

Adverse events were defined as the appearance of (or worsening of any pre-existing) undesirable signs, symptoms, or medical conditions that occurred after the participant's signed informed consent was obtained. Abnormal laboratory values or test results occurring after informed consent constituted adverse events only if they induced clinical signs or symptoms, were considered clinically significant, required therapy (e.g., hematologic abnormality that required transfusion or hematological stem cell support), or required changes in study medication(s). TEAEs were defined as those AEs that started or worsened during the on-treatment period (either randomized or cross-over period). (NCT03112603)
Timeframe: from Baseline to LPLV (approximately 5 years)

InterventionParticipants (Count of Participants)
Ruxolitinib165
Best Available Therapy148
Ruxolitinib Cross-Over Period70

[back to top]

AUCinf of Ruxolitinib After Single (Cycle 1 Day 1) and Multiple (Cycle 1 Day 15) Doses

"AUCinf was defined as the area under the concentration-time curve from time 0 to infinity. Early enrolling participants (approximately the first 8 adult and first 4 adolescent participants) randomized to ruxolitinib arm followed an extensive PK sampling schedule. Subsequent participants randomized to ruxolitinib, any randomized participants receiving ruxolitinib after Cycle 6, and any randomized participants receiving BAT that cross over to ruxolitinib followed the sparse PK sampling schedule." (NCT03112603)
Timeframe: Extensive Sampling Schedule: Cycle 1 Days 1 and 15: predose; 0.5, 1, 1.5, 4, 6, and 9 hours post-dose. Sparse Sampling Schedule: Cycle 1 Days 1 and 15: predose; 1.5 hours post-dose

Interventionng*hour/mL (Geometric Mean)
Day 1
Ruxolitinib642

[back to top]

Utilization of Medical Resources

The percentage of participants with at least one submission to healthcare encounter was assessed. (NCT03112603)
Timeframe: from Baseline to LPLV (approximately 5 years)

Interventionpercentage of participants (Number)
Ruxolitinib57.0
Best Available Therapy65.8

[back to top]

Rate of Participants With Clinically Relevant Improvement of the Modified Lee cGvHD Symptom Scale Score

"To assess improvement of symptoms based on the total symptom score (TSS); a responder was defined as having achieved a clinically relevant reduction from Baseline of the TSS. The scale consists of 30 items in 7 subscales (skin, eye, mouth, lung, nutrition, energy, and psychological). Participants reported their level of symptom bother over the previous month on a 5-point likert scale: not at all, slightly, moderately, quite a bit, or extremely. Subscale scores and the summary score range from 0 to 100, with a higher score indicating worse symptoms." (NCT03112603)
Timeframe: Baseline; Cycle 7 Day 1 (each cycle was comprised of 4 weeks)

Interventionpercentage of participants (Number)
Ruxolitinib24.2
Best Available Therapy11.0

[back to top]

ORR at the End of Cycle 3

ORR was defined as the percentage of participants in each arm demonstrating a CR or PR based on cGvHD disease assessments (National Institutes of Health Consensus Criteria) without the requirement of additional systemic therapies for an earlier progression, mixed response, or non-response. Scoring of response was relative to the organ score at the time of randomization. CR: complete resolution of all signs and symptoms of cGVHD in all evaluable organs without the initiation or addition of new systemic therapy. PR: improvement in at least one organ (e.g., improvement of 1 or more points on a 4- to 7-point scale, or an improvement of 2 or more points on a 10- to 12-point scale) without progression in other organs or sites, initiation, or addition of new systemic therapies. (NCT03112603)
Timeframe: Cycle 4 Day 1 (each cycle was comprised of 4 weeks)

Interventionpercentage of participants (Number)
Ruxolitinib54.5
Best Available Therapy31.1

[back to top]

Rate of FFS at Study Completion

Composite time to event endpoint incorporating the following FFS events: (i) relapse or recurrence of underlying disease or death due to underlying disease, (ii) nonrelapse mortality, or (iii) addition or initiation of another systemic therapy for cGvHD. (NCT03112603)
Timeframe: From Baseline to Last Participant Last Visit (LPLV) (approximately 5 years)

Interventionmonths (Median)
Ruxolitinib38.4
Best Available Therapy5.7

[back to top]

Overall Survival (OS)

Overall survival was defined as the time from the date of randomization to the date of death due to any cause. (NCT03112603)
Timeframe: from the date of randomization to the date of death due to any cause up to LPLV (approximately 5 years)

Interventionmonths (Median)
RuxolitinibNA
Best Available TherapyNA

[back to top]

Cumulative Incidence of Malignancy Relapse/Recurrence (MR)

Defined as the cumulative incidence rate from competing risk analysis of MR from the date of randomization to hematologic malignancy relapse/recurrence. (NCT03112603)
Timeframe: Months 3, 6, 12, 18, 24, 30, and 36

,
Interventionpercentage of participants (Number)
0 to < 3 months3 to < 6 months6 to < 12 months12 to < 18 months18 to < 24 months24 to <30 months30 to <36 months
Best Available Therapy1.312.656.086.086.086.787.50
Ruxolitinib1.923.225.187.828.488.488.48

[back to top]

Cumulative Incidence of Non-relapse Mortality (NRM)

Defined as the cumulative incidence rate from competing risk analysis for NRM from the date of randomization to the date of death not preceded by underlying disease relapse/recurrence. (NCT03112603)
Timeframe: Months 3, 6, 12, 18, 24, 30, and 36

,
Interventionpercentage of participants (Number)
Month 3Month 6Month 12Month 18Month 24Month 30Month 36
Best Available Therapy4.446.4315.1216.4819.2219.2222.0
Ruxolitinib5.459.1315.3015.9317.8317.8317.83

[back to top]

Percentage of Participants Successfully Tapered Off of All Corticosteroids

All corticosteroid dosages prescribed to the participant and all dose changes during the study were to be recorded for assessment of participants who successfully tapered off of all corticosteroids. Participants who completely tapered off corticosteroids refer to those who permanently discontinued steroids as per the dose administration panel and who did not restart steroids in the same interval. Participants who were tapered off and continued follow-up were also counted as being tapered off with 0 dose in subsequent intervals until they discontinued from the main treatment period or restarted steroid treatment. (NCT03112603)
Timeframe: up to Day 179

,
Interventionpercentage of participants (Number)
Day 1 to ≤ Day 28Day 29 to ≤ Day 56Day 57 to ≤ Day 84Day 85 to ≤ Day 112Day 113 to ≤ Day 140Day 141 to ≤ Day 168Day 169 to ≤ Day 179
Best Available Therapy2.65.48.510.312.416.815.9
Ruxolitinib2.59.614.016.319.724.224.1

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from randomization to the date of death from any cause. Participants with no documentation of death on-study were censored at the date at which they were last known to be alive. (NCT03163667)
Timeframe: As of the data cutoff date of 30 Sep 2020; maximum duration of follow-up for OS was 30.4 months.

Interventionmonths (Median)
Placebo + Everolimus9.72
CB-839 + Everolimus14.37

[back to top]

Progression Free Survival (PFS)

"PFS was defined as the time from randomization to the date of documented disease progression (assessed by Investigator per Response Evaluation Criteria in Solid Tumors [RECIST] v1.1) within 2 scheduled scan intervals following previous evaluable radiologic tumor assessment or death for any cause, whichever occurred first. Participants with no documentation of disease progression or death on-study were censored at the date of last available tumor assessment.~Progressive Disease (PD) per RECIST 1.1: At least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study. In addition, the sum must also demonstrate an absolute increase of at least 5 mm." (NCT03163667)
Timeframe: As of the primary data cutoff date of 26 Apr 2019; maximum duration of follow-up for PFS was 11.2 months.

Interventionmonths (Median)
Placebo + Everolimus1.91
CB-839 + Everolimus3.81

[back to top] [back to top] [back to top]

HRQoL Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30 Scores

The EORT QLQ-C30 consisted of 30 questions comprising 9 multiple-item scales and 6 single items. Multiple-item scales of QLQ-C30 consisted of 5 functional scales (physical, role, emotional, cognitive, and social) and 3 symptom scales (fatigue, nausea and vomiting, pain) and a global health status/QOL score. Six single-item scales of QLQ-C30 involved dyspnea, insomnia, appetite loss, constipation, diarrhea and financial difficulties. First 28 questions used a 4-point scale (1 = Not at all to 4 = Very much); and last 2 questions used a 7-point scale (1 = Very poor to 7 = Excellent). Scores for all scales range from 0 to 100. For the overall HRQoL and functioning scales, a higher score was correlated with better HRQoL, whereas a higher score for symptom scales represented worse HRQoL. (NCT03173560)
Timeframe: At baseline (prior to first dose of study drug), on Day 1 of each subsequent cycle (cycle length =28 days), and at the Off-treatment visit (up to 29 months)

Interventionscore on a scale (Mean)
Global Health Status/QoL Score; BaselineGlobal Health Status/QoL Score; Cycle 2 Day 1Global Health Status/QoL Score; Cycle 3 Day 1Global Health Status/QoL Score; Cycle 4 Day 1Global Health Status/QoL Score; Cycle 5 Day 1Global Health Status/QoL Score; Cycle 6 Day 1Global Health Status/QoL Score; Cycle 7 Day 1Global Health Status/QoL Score; Cycle 8 Day 1Global Health Status/QoL Score; Cycle 9 Day 1Global Health Status/QoL Score; Cycle 10 Day 1Global Health Status/QoL Score; Cycle 11 Day 1Global Health Status/QoL Score; Cycle 12 Day 1Global Health Status/QoL Score; Cycle 13 Day 1Global Health Status/QoL Score; Cycle 14 Day 1Global Health Status/QoL Score; Cycle 15 Day 1Global Health Status/QoL Score; Cycle 16 Day 1Global Health Status/QoL Score; Cycle 17 Day 1Global Health Status/QoL Score; Cycle 18 Day 1Global Health Status/QoL Score; Cycle 19 Day 1Global Health Status/QoL Score; Cycle 20 Day 1Global Health Status/QoL Score; Cycle 21 Day 1Global Health Status/QoL Score; Cycle 22 Day 1Global Health Status/QoL Score; Cycle 23 Day 1Global Health Status/QoL Score; Cycle 24 Day 1Global Health Status/QoL Score; Cycle 25 Day 1Global Health Status/QoL Score; Cycle 26 Day 1Global Health Status/QoL Score; Cycle 27 Day 1Global Health Status/QoL Score; Cycle 28 Day 1Global Health Status/QoL Score; Cycle 29 Day 1Global Health Status/QoL Score; Cycle 30 Day 1Global Health Status/QoL Score; Off-treatment VisitPhysical Functioning Score; BaselinePhysical Functioning Score; Cycle 2 Day 1Physical Functioning Score; Cycle 3 Day 1Physical Functioning Score; Cycle 4 Day 1Physical Functioning Score; Cycle 5 Day 1Physical Functioning Score; Cycle 6 Day 1Physical Functioning Score; Cycle 7 Day 1Physical Functioning Score; Cycle 8 Day 1Physical Functioning Score; Cycle 9 Day 1Physical Functioning Score; Cycle 10 Day 1Physical Functioning Score; Cycle 11 Day 1Physical Functioning Score; Cycle 12 Day 1Physical Functioning Score; Cycle 13 Day 1Physical Functioning Score; Cycle 14 Day 1Physical Functioning Score; Cycle 15 Day 1Physical Functioning Score; Cycle 16 Day 1Physical Functioning Score; Cycle 17 Day 1Physical Functioning Score; Cycle 18 Day 1Physical Functioning Score; Cycle 19 Day 1Physical Functioning Score; Cycle 20 Day 1Physical Functioning Score; Cycle 21 Day 1Physical Functioning Score; Cycle 22 Day 1Physical Functioning Score; Cycle 23 Day 1Physical Functioning Score; Cycle 24 Day 1Physical Functioning Score; Cycle 25 Day 1Physical Functioning Score; Cycle 26 Day 1Physical Functioning Score; Cycle 27 Day 1Physical Functioning Score; Cycle 28 Day 1Physical Functioning Score; Cycle 29 Day 1Physical Functioning Score; Cycle 30 Day 1Physical Functioning Score; Off-treatment VisitRole Functioning Score; BaselineRole Functioning Score; Cycle 2 Day 1Role Functioning Score; Cycle 3 Day 1Role Functioning Score; Cycle 4 Day 1Role Functioning Score; Cycle 5 Day 1Role Functioning Score; Cycle 6 Day 1Role Functioning Score; Cycle 7 Day 1Role Functioning Score; Cycle 8 Day 1Role Functioning Score; Cycle 9 Day 1Role Functioning Score; Cycle 10 Day 1Role Functioning Score; Cycle 11 Day 1Role Functioning Score; Cycle 12 Day 1Role Functioning Score; Cycle 13 Day 1Role Functioning Score; Cycle 14 Day 1Role Functioning Score; Cycle 15 Day 1Role Functioning Score; Cycle 16 Day 1Role Functioning Score; Cycle 17 Day 1Role Functioning Score; Cycle 18 Day 1Role Functioning Score; Cycle 19 Day 1Role Functioning Score; Cycle 20 Day 1Role Functioning Score; Cycle 21 Day 1Role Functioning Score; Cycle 22 Day 1Role Functioning Score; Cycle 23 Day 1Role Functioning Score; Cycle 24 Day 1Role Functioning Score; Cycle 25 Day 1Role Functioning Score; Cycle 26 Day 1Role Functioning Score; Cycle 27 Day 1Role Functioning Score; Cycle 28 Day 1Role Functioning Score; Cycle 29 Day 1Role Functioning Score; Cycle 30 Day 1Role Functioning Score; Off-treatment VisitEmotional Functioning Score; BaselineEmotional Functioning Score; Cycle 2 Day 1Emotional Functioning Score; Cycle 3 Day 1Emotional Functioning Score; Cycle 4 Day 1Emotional Functioning Score; Cycle 5 Day 1Emotional Functioning Score; Cycle 6 Day 1Emotional Functioning Score; Cycle 7 Day 1Emotional Functioning Score; Cycle 8 Day 1Emotional Functioning Score; Cycle 9 Day 1Emotional Functioning Score; Cycle 10 Day 1Emotional Functioning Score; Cycle 11 Day 1Emotional Functioning Score; Cycle 12 Day 1Emotional Functioning Score; Cycle 13 Day 1Emotional Functioning Score; Cycle 14 Day 1Emotional Functioning Score; Cycle 15 Day 1Emotional Functioning Score; Cycle 16 Day 1Emotional Functioning Score; Cycle 17 Day 1Emotional Functioning Score; Cycle 18 Day 1Emotional Functioning Score; Cycle 19 Day 1Emotional Functioning Score; Cycle 20 Day 1Emotional Functioning Score; Cycle 21 Day 1Emotional Functioning Score; Cycle 22 Day 1Emotional Functioning Score; Cycle 23 Day 1Emotional Functioning Score; Cycle 24 Day 1Emotional Functioning Score; Cycle 25 Day 1Emotional Functioning Score; Cycle 26 Day 1Emotional Functioning Score; Cycle 27 Day 1Emotional Functioning Score; Cycle 28 Day 1Emotional Functioning Score; Cycle 29 Day 1Emotional Functioning Score; Cycle 30 Day 1Emotional Functioning Score; Off-treatment VisitCognitive Functioning Score; BaselineCognitive Functioning Score; Cycle 2 Day 1Cognitive Functioning Score; Cycle 3 Day 1Cognitive Functioning Score; Cycle 4 Day 1Cognitive Functioning Score; Cycle 5 Day 1Cognitive Functioning Score; Cycle 6 Day 1Cognitive Functioning Score; Cycle 7 Day 1Cognitive Functioning Score; Cycle 8 Day 1Cognitive Functioning Score; Cycle 9 Day 1Cognitive Functioning Score; Cycle 10 Day 1Cognitive Functioning Score; Cycle 11 Day 1Cognitive Functioning Score; Cycle 12 Day 1Cognitive Functioning Score; Cycle 13 Day 1Cognitive Functioning Score; Cycle 14 Day 1Cognitive Functioning Score; Cycle 15 Day 1Cognitive Functioning Score; Cycle 16 Day 1Cognitive Functioning Score; Cycle 17 Day 1Cognitive Functioning Score; Cycle 18 Day 1Cognitive Functioning Score; Cycle 19 Day 1Cognitive Functioning Score; Cycle 20 Day 1Cognitive Functioning Score; Cycle 21 Day 1Cognitive Functioning Score; Cycle 22 Day 1Cognitive Functioning Score; Cycle 23 Day 1Cognitive Functioning Score; Cycle 24 Day 1Cognitive Functioning Score; Cycle 25 Day 1Cognitive Functioning Score; Cycle 26 Day 1Cognitive Functioning Score; Cycle 27 Day 1Cognitive Functioning Score; Cycle 28 Day 1Cognitive Functioning Score; Cycle 29 Day 1Cognitive Functioning Score; Cycle 30 Day 1Cognitive Functioning Score; Off-treatment VisitSocial Functioning Score; BaselineSocial Functioning Score; Cycle 2 Day 1Social Functioning Score; Cycle 3 Day 1Social Functioning Score; Cycle 4 Day 1Social Functioning Score; Cycle 5 Day 1Social Functioning Score; Cycle 6 Day 1Social Functioning Score; Cycle 7 Day 1Social Functioning Score; Cycle 8 Day 1Social Functioning Score; Cycle 9 Day 1Social Functioning Score; Cycle 10 Day 1Social Functioning Score; Cycle 11 Day 1Social Functioning Score; Cycle 12 Day 1Social Functioning Score; Cycle 13 Day 1Social Functioning Score; Cycle 14 Day 1Social Functioning Score; Cycle 15 Day 1Social Functioning Score; Cycle 16 Day 1Social Functioning Score; Cycle 17 Day 1Social Functioning Score; Cycle 18 Day 1Social Functioning Score; Cycle 19 Day 1Social Functioning Score; Cycle 20 Day 1Social Functioning Score; Cycle 21 Day 1Social Functioning Score; Cycle 22 Day 1Social Functioning Score; Cycle 23 Day 1Social Functioning Score; Cycle 24 Day 1Social Functioning Score; Cycle 25 Day 1Social Functioning Score; Cycle 26 Day 1Social Functioning Score; Cycle 27 Day 1Social Functioning Score; Cycle 28 Day 1Social Functioning Score; Cycle 29 Day 1Social Functioning Score; Cycle 30 Day 1Social Functioning Score; Off-treatment VisitFatigue Score; BaselineFatigue Score; Cycle 2 Day 1Fatigue Score; Cycle 3 Day 1Fatigue Score; Cycle 4 Day 1Fatigue Score; Cycle 5 Day 1Fatigue Score; Cycle 6 Day 1Fatigue Score; Cycle 7 Day 1Fatigue Score; Cycle 8 Day 1Fatigue Score; Cycle 9 Day 1Fatigue Score; Cycle 10 Day 1Fatigue Score; Cycle 11 Day 1Fatigue Score; Cycle 12 Day 1Fatigue Score; Cycle 13 Day 1Fatigue Score; Cycle 14 Day 1Fatigue Score; Cycle 15 Day 1Fatigue Score; Cycle 16 Day 1Fatigue Score; Cycle 17 Day 1Fatigue Score; Cycle 18 Day 1Fatigue Score; Cycle 19 Day 1Fatigue Score; Cycle 20 Day 1Fatigue Score; Cycle 21 Day 1Fatigue Score; Cycle 22 Day 1Fatigue Score; Cycle 23 Day 1Fatigue Score; Cycle 24 Day 1Fatigue Score; Cycle 25 Day 1Fatigue Score; Cycle 26 Day 1Fatigue Score; Cycle 27 Day 1Fatigue Score; Cycle 28 Day 1Fatigue Score; Cycle 29 Day 1Fatigue Score; Cycle 30 Day 1Fatigue Score; Off-treatment VisitNausea and Vomiting Score; BaselineNausea and Vomiting Score; Cycle 2 Day 1Nausea and Vomiting Score; Cycle 3 Day 1Nausea and Vomiting Score; Cycle 4 Day 1Nausea and Vomiting Score; Cycle 5 Day 1Nausea and Vomiting Score; Cycle 6 Day 1Nausea and Vomiting Score; Cycle 7 Day 1Nausea and Vomiting Score; Cycle 8 Day 1Nausea and Vomiting Score; Cycle 9 Day 1Nausea and Vomiting Score; Cycle 10 Day 1Nausea and Vomiting Score; Cycle 11 Day 1Nausea and Vomiting Score; Cycle 12 Day 1Nausea and Vomiting Score; Cycle 13 Day 1Nausea and Vomiting Score; Cycle 14 Day 1Nausea and Vomiting Score; Cycle 15 Day 1Nausea and Vomiting Score; Cycle 16 Day 1Nausea and Vomiting Score; Cycle 17 Day 1Nausea and Vomiting Score; Cycle 18 Day 1Nausea and Vomiting Score; Cycle 19 Day 1Nausea and Vomiting Score; Cycle 20 Day 1Nausea and Vomiting Score; Cycle 21 Day 1Nausea and Vomiting Score; Cycle 22 Day 1Nausea and Vomiting Score; Cycle 23 Day 1Nausea and Vomiting Score; Cycle 24 Day 1Nausea and Vomiting Score; Cycle 25 Day 1Nausea and Vomiting Score; Cycle 26 Day 1Nausea and Vomiting Score; Cycle 27 Day 1Nausea and Vomiting Score; Cycle 28 Day 1Nausea and Vomiting Score; Cycle 29 Day 1Nausea and Vomiting Score; Cycle 30 Day 1Nausea and Vomiting Score; Off-treatment VisitPain Score; BaselinePain Score; Cycle 2 Day 1Pain Score; Cycle 3 Day 1Pain Score; Cycle 4 Day 1Pain Score; Cycle 5 Day 1Pain Score; Cycle 6 Day 1Pain Score; Cycle 7 Day 1Pain Score; Cycle 8 Day 1Pain Score; Cycle 9 Day 1Pain Score; Cycle 10 Day 1Pain Score; Cycle 11 Day 1Pain Score; Cycle 12 Day 1Pain Score; Cycle 13 Day 1Pain Score; Cycle 14 Day 1Pain Score; Cycle 15 Day 1Pain Score; Cycle 16 Day 1Pain Score; Cycle 17 Day 1Pain Score; Cycle 18 Day 1Pain Score; Cycle 19 Day 1Pain Score; Cycle 20 Day 1Pain Score; Cycle 21 Day 1Pain Score; Cycle 22 Day 1Pain Score; Cycle 23 Day 1Pain Score; Cycle 24 Day 1Pain Score; Cycle 25 Day 1Pain Score; Cycle 26 Day 1Pain Score; Cycle 27 Day 1Pain Score; Cycle 28 Day 1Pain Score; Cycle 29 Day 1Pain Score; Cycle 30 Day 1Pain Score; Off-treatment VisitDyspnea Score; BaselineDyspnea Score; Cycle 2 Day 1Dyspnea Score; Cycle 3 Day 1Dyspnea Score; Cycle 4 Day 1Dyspnea Score; Cycle 5 Day 1Dyspnea Score; Cycle 6 Day 1Dyspnea Score; Cycle 7 Day 1Dyspnea Score; Cycle 8 Day 1Dyspnea Score; Cycle 9 Day 1Dyspnea Score; Cycle 10 Day 1Dyspnea Score; Cycle 11 Day 1Dyspnea Score; Cycle 12 Day 1Dyspnea Score; Cycle 13 Day 1Dyspnea Score; Cycle 14 Day 1Dyspnea Score; Cycle 15 Day 1Dyspnea Score; Cycle 16 Day 1Dyspnea Score; Cycle 17 Day 1Dyspnea Score; Cycle 18 Day 1Dyspnea Score; Cycle 19 Day 1Dyspnea Score; Cycle 20 Day 1Dyspnea Score; Cycle 21 Day 1Dyspnea Score; Cycle 22 Day 1Dyspnea Score; Cycle 23 Day 1Dyspnea Score; Cycle 24 Day 1Dyspnea Score; Cycle 25 Day 1Dyspnea Score; Cycle 26 Day 1Dyspnea Score; Cycle 27 Day 1Dyspnea Score; Cycle 28 Day 1Dyspnea Score; Cycle 29 Day 1Dyspnea Score; Cycle 30 Day 1Dyspnea Score; Off-treatment VisitInsomnia Score; BaselineInsomnia Score; Cycle 2 Day 1Insomnia Score; Cycle 3 Day 1Insomnia Score; Cycle 4 Day 1Insomnia Score; Cycle 5 Day 1Insomnia Score; Cycle 6 Day 1Insomnia Score; Cycle 7 Day 1Insomnia Score; Cycle 8 Day 1Insomnia Score; Cycle 9 Day 1Insomnia Score; Cycle 10 Day 1Insomnia Score; Cycle 11 Day 1Insomnia Score; Cycle 12 Day 1Insomnia Score; Cycle 13 Day 1Insomnia Score; Cycle 14 Day 1Insomnia Score; Cycle 15 Day 1Insomnia Score; Cycle 16 Day 1Insomnia Score; Cycle 17 Day 1Insomnia Score; Cycle 18 Day 1Insomnia Score; Cycle 19 Day 1Insomnia Score; Cycle 20 Day 1Insomnia Score; Cycle 21 Day 1Insomnia Score; Cycle 22 Day 1Insomnia Score; Cycle 23 Day 1Insomnia Score; Cycle 24 Day 1Insomnia Score; Cycle 25 Day 1Insomnia Score; Cycle 26 Day 1Insomnia Score; Cycle 27 Day 1Insomnia Score; Cycle 28 Day 1Insomnia Score; Cycle 29 Day 1Insomnia Score; Cycle 30 Day 1Insomnia Score; Off-treatment VisitAppetite Loss Score; BaselineAppetite Loss Score; Cycle 2 Day 1Appetite Loss Score; Cycle 3 Day 1Appetite Loss Score; Cycle 4 Day 1Appetite Loss Score; Cycle 5 Day 1Appetite Loss Score; Cycle 6 Day 1Appetite Loss Score; Cycle 7 Day 1Appetite Loss Score; Cycle 8 Day 1Appetite Loss Score; Cycle 9 Day 1Appetite Loss Score; Cycle 10 Day 1Appetite Loss Score; Cycle 11 Day 1Appetite Loss Score; Cycle 12 Day 1Appetite Loss Score; Cycle 13 Day 1Appetite Loss Score; Cycle 14 Day 1Appetite Loss Score; Cycle 15 Day 1Appetite Loss Score; Cycle 16 Day 1Appetite Loss Score; Cycle 17 Day 1Appetite Loss Score; Cycle 18 Day 1Appetite Loss Score; Cycle 19 Day 1Appetite Loss Score; Cycle 20 Day 1Appetite Loss Score; Cycle 21 Day 1Appetite Loss Score; Cycle 22 Day 1Appetite Loss Score; Cycle 23 Day 1Appetite Loss Score; Cycle 24 Day 1Appetite Loss Score; Cycle 25 Day 1Appetite Loss Score; Cycle 26 Day 1Appetite Loss Score; Cycle 27 Day 1Appetite Loss Score; Cycle 28 Day 1Appetite Loss Score; Cycle 29 Day 1Appetite Loss Score; Cycle 30 Day 1Appetite Loss Score; Off-treatment VisitConstipation Score; BaselineConstipation Score; Cycle 2 Day 1Constipation Score; Cycle 3 Day 1Constipation Score; Cycle 4 Day 1Constipation Score; Cycle 5 Day 1Constipation Score; Cycle 6 Day 1Constipation Score; Cycle 7 Day 1Constipation Score; Cycle 8 Day 1Constipation Score; Cycle 9 Day 1Constipation Score; Cycle 10 Day 1Constipation Score; Cycle 11 Day 1Constipation Score; Cycle 12 Day 1Constipation Score; Cycle 13 Day 1Constipation Score; Cycle 14 Day 1Constipation Score; Cycle 15 Day 1Constipation Score; Cycle 16 Day 1Constipation Score; Cycle 17 Day 1Constipation Score; Cycle 18 Day 1Constipation Score; Cycle 19 Day 1Constipation Score; Cycle 20 Day 1Constipation Score; Cycle 21 Day 1Constipation Score; Cycle 22 Day 1Constipation Score; Cycle 23 Day 1Constipation Score; Cycle 24 Day 1Constipation Score; Cycle 25 Day 1Constipation Score; Cycle 26 Day 1Constipation Score; Cycle 27 Day 1Constipation Score; Cycle 28 Day 1Constipation Score; Cycle 29 Day 1Constipation Score; Cycle 30 Day 1Constipation Score; Off-treatment VisitDiarrhea Score; BaselineDiarrhea Score; Cycle 2 Day 1Diarrhea Score; Cycle 3 Day 1Diarrhea Score; Cycle 4 Day 1Diarrhea Score; Cycle 5 Day 1Diarrhea Score; Cycle 6 Day 1Diarrhea Score; Cycle 7 Day 1Diarrhea Score; Cycle 8 Day 1Diarrhea Score; Cycle 9 Day 1Diarrhea Score; Cycle 10 Day 1Diarrhea Score; Cycle 11 Day 1Diarrhea Score; Cycle 12 Day 1Diarrhea Score; Cycle 13 Day 1Diarrhea Score; Cycle 14 Day 1Diarrhea Score; Cycle 15 Day 1Diarrhea Score; Cycle 16 Day 1Diarrhea Score; Cycle 17 Day 1Diarrhea Score; Cycle 18 Day 1Diarrhea Score; Cycle 19 Day 1Diarrhea Score; Cycle 20 Day 1Diarrhea Score; Cycle 21 Day 1Diarrhea Score; Cycle 22 Day 1Diarrhea Score; Cycle 23 Day 1Diarrhea Score; Cycle 24 Day 1Diarrhea Score; Cycle 25 Day 1Diarrhea Score; Cycle 26 Day 1Diarrhea Score; Cycle 27 Day 1Diarrhea Score; Cycle 28 Day 1Diarrhea Score; Cycle 29 Day 1Diarrhea Score; Cycle 30 Day 1Diarrhea Score; Off-treatment VisitFinancial Difficulties Score; BaselineFinancial Difficulties Score; Cycle 2 Day 1Financial Difficulties Score; Cycle 3 Day 1Financial Difficulties Score; Cycle 4 Day 1Financial Difficulties Score; Cycle 5 Day 1Financial Difficulties Score; Cycle 6 Day 1Financial Difficulties Score; Cycle 7 Day 1Financial Difficulties Score; Cycle 8 Day 1Financial Difficulties Score; Cycle 9 Day 1Financial Difficulties Score; Cycle 10 Day 1Financial Difficulties Score; Cycle 11 Day 1Financial Difficulties Score; Cycle 12 Day 1Financial Difficulties Score; Cycle 13 Day 1Financial Difficulties Score; Cycle 14 Day 1Financial Difficulties Score; Cycle 15 Day 1Financial Difficulties Score; Cycle 16 Day 1Financial Difficulties Score; Cycle 17 Day 1Financial Difficulties Score; Cycle 18 Day 1Financial Difficulties Score; Cycle 19 Day 1Financial Difficulties Score; Cycle 20 Day 1Financial Difficulties Score; Cycle 21 Day 1Financial Difficulties Score; Cycle 22 Day 1Financial Difficulties Score; Cycle 23 Day 1Financial Difficulties Score; Cycle 24 Day 1Financial Difficulties Score; Cycle 25 Day 1Financial Difficulties Score; Cycle 26 Day 1Financial Difficulties Score; Cycle 27 Day 1Financial Difficulties Score; Cycle 28 Day 1Financial Difficulties Score; Cycle 29 Day 1Financial Difficulties Score; Cycle 30 Day 1Financial Difficulties Score; Off-treatment Visit
Lenvatinib 18 mg + Everolimus 5 mg63.5860.9361.8562.6263.7663.7362.5462.1163.7766.4465.8666.9468.9470.3566.8364.5467.7168.3866.6766.4071.1366.0566.6765.7969.7960.9071.4377.787566.6756.0576.6075.3876.1575.4077.8577.7678.4179.1180.6081.2081.1383.0180.2482.3980.9281.7482.3381.5281.5782.8084.2982.7281.3983.8684.1771.7985.719086.6710070.4175.7269.3372.1873.6072.4473.7573.9775.6075.9176.8979.1180.918081.1374.8479.0883.7582.3577.4579.5781.5577.7879.8683.3382.2969.2390.4897.2296.6710063.5180.0682.0682.9084.1984.5284.3884.2984.8885.9687.8487.1586.6787.2787.5087.2587.4187.0886.7684.8086.5688.9989.2087.1587.2890.6385.2695.2410010010073.8789.5188.8587.4790.3287.4387.6887.9488.4987.6890.4488.8988.6189.7087.1883.9989.3689.5890.2089.2291.9488.1090.1288.1992.1198.9692.3195.2410010010080.4178.2977.1577.1977.3379.6378.7177.3079.0478.6285.1485.1983.8983.9483.9781.3780.8080.4282.8480.8881.7282.1482.1083.3384.2184.3876.9295.2410010010066.8931.7636.7436.6232.6835.1733.1930.7930.8130.8027.1127.4623.4825.0525.5828.9825.532524.8427.4527.2423.0226.7529.1726.9025.6937.6120.6316.6715.5611.1138.896.289.569.7412.1711.4210.148.5710.149.967.788.696.996.977.558.829.2259.317.3510.227.748.646.257.892.0814.1016.678.33006.7623.7727.042522.8722.9722.5023.6523.2021.0119.3318.0618.5518.4816.0417.6518.4417.5016.6719.6119.3517.8622.8418.0614.9115.6321.7911.902.786.67027.7022.2219.5619.7217.5215.7518.8915.8714.0914.4911.5615.0212.9010.9110.6912.429.931010.789.8011.837.1412.3511.1112.288.3315.3814.2916.6720021.6222.8419.912320.1922.0517.502019.5916.6718.2217.3713.9813.3311.951614.8913.3311.7613.7312.9010.716.1711.1112.2812.5020.519.5211.1113.33028.8318.3127.5629.8130.9030.6726.1126.9825.0025.3622.2222.0718.8217.2817.6124.1822.7022.5022.2220.5921.5115.4817.2820.8315.7918.7523.089.5211.110025.2312.5511.1110.809.809.878.477.447.229.428.007.989.689.097.699.339.935.007.849.808.607.147.4111.118.7718.7515.38000012.616.7917.6627.1929.4129.3326.8931.7531.2728.6229.3326.8522.7823.0324.3630.0723.4029.1725.4921.5722.5821.4324.6922.2219.3018.7517.9533.3322.2220.00012.1620.3715.0118.2016.4218.7817.8015.8717.5316.6717.3516.2016.1119.1417.3119.6117.0219.1719.6120.5920.4320.2420.9923.6119.3016.6720.51000025.11

[back to top]

HRQoL Assessed by European Quality of Life (EuroQol) Five-Dimensional, 3-Level (EQ-5D-3L) Index Score and Visual Analogue Scale (VAS)

"The EQ-5D-3L is a health profile questionnaire assessing quality of life along 5 dimensions. Participants rate 5 dimensions of health (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) by choosing from 3 answering options (1=no problems; 2=some problems; 3=extreme problems). The summed score ranges from 5-15 with 5 corresponding to no problems and 15 corresponding to severe problems in the 5 dimensions. The EQ-5D index was calculated by applying preference-based weights (tariffs) to the scores of the five health state dimensions. Index values can range from -1 to 1, with 0 representing a health state equivalent to death and 1 representing perfect health. EQ-5D-3L also included an EQ visual analogue scale (VAS) that ranges between 100 (best imaginable health) and 0 (worst imaginable health). Decrease from baseline in EQ-5D-3L signifies improvement. Total index EQ-5D-3L summary score was weighted with a range of -0.594 (worst) to 1.0 (best)." (NCT03173560)
Timeframe: At baseline (prior to first dose of study drug), on Day 1 of each subsequent cycle (cycle length =28 days), and at the Off-treatment visit (up to 29 months)

Interventionscore on a scale (Mean)
EQ-5D Index Score; BaselineEQ-5D Index Score; Cycle 2 Day 1EQ-5D Index Score; Cycle 3 Day 1EQ-5D Index Score; Cycle 4 Day 1EQ-5D Index Score; Cycle 5 Day 1EQ-5D Index Score; Cycle 6 Day 1EQ-5D Index Score; Cycle 7 Day 1EQ-5D Index Score; Cycle 8 Day 1EQ-5D Index Score; Cycle 9 Day 1EQ-5D Index Score; Cycle 10 Day 1EQ-5D Index Score; Cycle 11 Day 1EQ-5D Index Score; Cycle 12 Day 1EQ-5D Index Score; Cycle 13 Day 1EQ-5D Index Score; Cycle 14 Day 1EQ-5D Index Score; Cycle 15 Day 1EQ-5D Index Score; Cycle 16 Day 1EQ-5D Index Score; Cycle 17 Day 1EQ-5D Index Score; Cycle 18 Day 1EQ-5D Index Score; Cycle 19 Day 1EQ-5D Index Score; Cycle 20 Day 1EQ-5D Index Score; Cycle 21 Day 1EQ-5D Index Score; Cycle 22 Day 1EQ-5D Index Score; Cycle 23 Day 1EQ-5D Index Score; Cycle 24 Day 1EQ-5D Index Score; Cycle 25 Day 1EQ-5D Index Score; Cycle 26 Day 1EQ-5D Index Score; Cycle 27 Day 1EQ-5D Index Score; Cycle 28 Day 1EQ-5D Index Score; Cycle 29 Day 1EQ-5D Index Score; Cycle 30 Day 1EQ-5D Index Score; Off-treatment VisitEQ-VAS Score; BaselineEQ-VAS Score; Cycle 2 Day 1EQ-VAS Score; Cycle 3 Day 1EQ-VAS Score; Cycle 4 Day 1EQ-VAS Score; Cycle 5 Day 1EQ-VAS Score; Cycle 6 Day 1EQ-VAS Score; Cycle 7 Day 1EQ-VAS Score; Cycle 8 Day 1EQ-VAS Score; Cycle 9 Day 1EQ-VAS Score; Cycle 10 Day 1EQ-VAS Score; Cycle 11 Day 1EQ-VAS Score; Cycle 12 Day 1EQ-VAS Score; Cycle 13 Day 1EQ-VAS Score; Cycle 14 Day 1EQ-VAS Score; Cycle 15 Day 1EQ-VAS Score; Cycle 16 Day 1EQ-VAS Score; Cycle 17 Day 1EQ-VAS Score; Cycle 18 Day 1EQ-VAS Score; Cycle 19 Day 1EQ-VAS Score; Cycle 20 Day 1EQ-VAS Score; Cycle 21 Day 1EQ-VAS Score; Cycle 22 Day 1EQ-VAS Score; Cycle 23 Day 1EQ-VAS Score; Cycle 24 Day 1EQ-VAS Score; Cycle 25 Day 1EQ-VAS Score; Cycle 26 Day 1EQ-VAS Score; Cycle 27 Day 1EQ-VAS Score; Cycle 28 Day 1EQ-VAS Score; Cycle 29 Day 1EQ-VAS Score; Cycle 30 Day 1EQ-VAS Score; Off-treatment Visit
Lenvatinib 18 mg + Everolimus 5 mg0.780.760.780.780.780.770.780.800.790.810.820.830.820.830.810.810.830.840.790.820.830.850.820.840.830.730.930.970.9610.6870.0169.5270.7169.6670.3071.7370.3970.4069.9273.1873.2174.4674.3574.4072.5872.8073.8073.7171.7672.0776.4674.4473.6771.2172.8165.2377.4377.50745062.66

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of randomization until the date of death from any cause. In the absence of confirmation of death, participants will be censored either at the date that the participant was last known to be alive or the date of data cutoff for the primary analysis, whichever comes earlier. Median OS was to be calculated using Kaplan-Meier estimate and presented with 2-sided 95% confidence interval. (NCT03173560)
Timeframe: From the date of randomization until the date of death from any cause, or up to date of data cut off for the primary analysis (up to 29 months)

Interventionmonths (Median)
Lenvatinib 14 mg + Everolimus 5 mg27.0
Lenvatinib 18 mg + Everolimus 5 mgNA

[back to top]

Number of Participants With TEAEs and Serious TEAEs

TEAEs were defined as those adverse events (AEs) that occurred (or worsened, if present at Baseline) after the first dose of study drug through 28 days after the last dose of study drug. An AE was defined as any untoward medical occurrence in a participants or clinical investigation participant administered an investigational product. An AE does not necessarily have a causal relationship with medicinal product. A serious adverse event (SAE) was defined as any AE if it resulted in death or life-threatening AE or required inpatient hospitalization or prolongation of existing hospitalization or resulted in persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions or was a congenital anomaly/birth defect. (NCT03173560)
Timeframe: From date of first dose of study drug up to 28 days after last dose of study drug, or date of data cut off for the primary analysis (up to 29 months)

,
InterventionParticipants (Count of Participants)
Participants With TEAEsParticipants With Serious TEAEs
Lenvatinib 14 mg + Everolimus 5 mg17385
Lenvatinib 18 mg + Everolimus 5 mg16782

[back to top]

HRQoL Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-C30 Scores

The EORT QLQ-C30 consisted of 30 questions comprising 9 multiple-item scales and 6 single items. Multiple-item scales of QLQ-C30 consisted of 5 functional scales (physical, role, emotional, cognitive, and social) and 3 symptom scales (fatigue, nausea and vomiting, pain) and a global health status/QOL score. Six single-item scales of QLQ-C30 involved dyspnea, insomnia, appetite loss, constipation, diarrhea and financial difficulties. First 28 questions used a 4-point scale (1 = Not at all to 4 = Very much); and last 2 questions used a 7-point scale (1 = Very poor to 7 = Excellent). Scores for all scales range from 0 to 100. For the overall HRQoL and functioning scales, a higher score was correlated with better HRQoL, whereas a higher score for symptom scales represented worse HRQoL. (NCT03173560)
Timeframe: At baseline (prior to first dose of study drug), on Day 1 of each subsequent cycle (cycle length =28 days), and at the Off-treatment visit (up to 29 months)

Interventionscore on a scale (Mean)
Global Health Status/QoL Score; BaselineGlobal Health Status/QoL Score; Cycle 2 Day 1Global Health Status/QoL Score; Cycle 3 Day 1Global Health Status/QoL Score; Cycle 4 Day 1Global Health Status/QoL Score; Cycle 5 Day 1Global Health Status/QoL Score; Cycle 6 Day 1Global Health Status/QoL Score; Cycle 7 Day 1Global Health Status/QoL Score; Cycle 8 Day 1Global Health Status/QoL Score; Cycle 9 Day 1Global Health Status/QoL Score; Cycle 10 Day 1Global Health Status/QoL Score; Cycle 11 Day 1Global Health Status/QoL Score; Cycle 12 Day 1Global Health Status/QoL Score; Cycle 13 Day 1Global Health Status/QoL Score; Cycle 14 Day 1Global Health Status/QoL Score; Cycle 15 Day 1Global Health Status/QoL Score; Cycle 16 Day 1Global Health Status/QoL Score; Cycle 17 Day 1Global Health Status/QoL Score; Cycle 18 Day 1Global Health Status/QoL Score; Cycle 19 Day 1Global Health Status/QoL Score; Cycle 20 Day 1Global Health Status/QoL Score; Cycle 21 Day 1Global Health Status/QoL Score; Cycle 22 Day 1Global Health Status/QoL Score; Cycle 23 Day 1Global Health Status/QoL Score; Cycle 24 Day 1Global Health Status/QoL Score; Cycle 25 Day 1Global Health Status/QoL Score; Cycle 26 Day 1Global Health Status/QoL Score; Cycle 27 Day 1Global Health Status/QoL Score; Cycle 28 Day 1Global Health Status/QoL Score; Cycle 29 Day 1Global Health Status/QoL Score; Cycle 30 Day 1Global Health Status/QoL Score; Cycle 31 Day 1Global Health Status/QoL Score; Off-treatment VisitPhysical Functioning Score; BaselinePhysical Functioning Score; Cycle 2 Day 1Physical Functioning Score; Cycle 3 Day 1Physical Functioning Score; Cycle 4 Day 1Physical Functioning Score; Cycle 5 Day 1Physical Functioning Score; Cycle 6 Day 1Physical Functioning Score; Cycle 7 Day 1Physical Functioning Score; Cycle 8 Day 1Physical Functioning Score; Cycle 9 Day 1Physical Functioning Score; Cycle 10 Day 1Physical Functioning Score; Cycle 11 Day 1Physical Functioning Score; Cycle 12 Day 1Physical Functioning Score; Cycle 13 Day 1Physical Functioning Score; Cycle 14 Day 1Physical Functioning Score; Cycle 15 Day 1Physical Functioning Score; Cycle 16 Day 1Physical Functioning Score; Cycle 17 Day 1Physical Functioning Score; Cycle 18 Day 1Physical Functioning Score; Cycle 19 Day 1Physical Functioning Score; Cycle 20 Day 1Physical Functioning Score; Cycle 21 Day 1Physical Functioning Score; Cycle 22 Day 1Physical Functioning Score; Cycle 23 Day 1Physical Functioning Score; Cycle 24 Day 1Physical Functioning Score; Cycle 25 Day 1Physical Functioning Score; Cycle 26 Day 1Physical Functioning Score; Cycle 27 Day 1Physical Functioning Score; Cycle 28 Day 1Physical Functioning Score; Cycle 29 Day 1Physical Functioning Score; Cycle 30 Day 1Physical Functioning Score; Cycle 31 Day 1Physical Functioning Score; Off-treatment VisitRole Functioning Score; BaselineRole Functioning Score; Cycle 2 Day 1Role Functioning Score; Cycle 3 Day 1Role Functioning Score; Cycle 4 Day 1Role Functioning Score; Cycle 5 Day 1Role Functioning Score; Cycle 6 Day 1Role Functioning Score; Cycle 7 Day 1Role Functioning Score; Cycle 8 Day 1Role Functioning Score; Cycle 9 Day 1Role Functioning Score; Cycle 10 Day 1Role Functioning Score; Cycle 11 Day 1Role Functioning Score; Cycle 12 Day 1Role Functioning Score; Cycle 13 Day 1Role Functioning Score; Cycle 14 Day 1Role Functioning Score; Cycle 15 Day 1Role Functioning Score; Cycle 16 Day 1Role Functioning Score; Cycle 17 Day 1Role Functioning Score; Cycle 18 Day 1Role Functioning Score; Cycle 19 Day 1Role Functioning Score; Cycle 20 Day 1Role Functioning Score; Cycle 21 Day 1Role Functioning Score; Cycle 22 Day 1Role Functioning Score; Cycle 23 Day 1Role Functioning Score; Cycle 24 Day 1Role Functioning Score; Cycle 25 Day 1Role Functioning Score; Cycle 26 Day 1Role Functioning Score; Cycle 27 Day 1Role Functioning Score; Cycle 28 Day 1Role Functioning Score; Cycle 29 Day 1Role Functioning Score; Cycle 30 Day 1Role Functioning Score; Cycle 31 Day 1Role Functioning Score; Off-treatment VisitEmotional Functioning Score; BaselineEmotional Functioning Score; Cycle 2 Day 1Emotional Functioning Score; Cycle 3 Day 1Emotional Functioning Score; Cycle 4 Day 1Emotional Functioning Score; Cycle 5 Day 1Emotional Functioning Score; Cycle 6 Day 1Emotional Functioning Score; Cycle 7 Day 1Emotional Functioning Score; Cycle 8 Day 1Emotional Functioning Score; Cycle 9 Day 1Emotional Functioning Score; Cycle 10 Day 1Emotional Functioning Score; Cycle 11 Day 1Emotional Functioning Score; Cycle 12 Day 1Emotional Functioning Score; Cycle 13 Day 1Emotional Functioning Score; Cycle 14 Day 1Emotional Functioning Score; Cycle 15 Day 1Emotional Functioning Score; Cycle 16 Day 1Emotional Functioning Score; Cycle 17 Day 1Emotional Functioning Score; Cycle 18 Day 1Emotional Functioning Score; Cycle 19 Day 1Emotional Functioning Score; Cycle 20 Day 1Emotional Functioning Score; Cycle 21 Day 1Emotional Functioning Score; Cycle 22 Day 1Emotional Functioning Score; Cycle 23 Day 1Emotional Functioning Score; Cycle 24 Day 1Emotional Functioning Score; Cycle 25 Day 1Emotional Functioning Score; Cycle 26 Day 1Emotional Functioning Score; Cycle 27 Day 1Emotional Functioning Score; Cycle 28 Day 1Emotional Functioning Score; Cycle 29 Day 1Emotional Functioning Score; Cycle 30 Day 1Emotional Functioning Score; Cycle 31 Day 1Emotional Functioning Score; Off-treatment VisitCognitive Functioning Score; BaselineCognitive Functioning Score; Cycle 2 Day 1Cognitive Functioning Score; Cycle 3 Day 1Cognitive Functioning Score; Cycle 4 Day 1Cognitive Functioning Score; Cycle 5 Day 1Cognitive Functioning Score; Cycle 6 Day 1Cognitive Functioning Score; Cycle 7 Day 1Cognitive Functioning Score; Cycle 8 Day 1Cognitive Functioning Score; Cycle 9 Day 1Cognitive Functioning Score; Cycle 10 Day 1Cognitive Functioning Score; Cycle 11 Day 1Cognitive Functioning Score; Cycle 12 Day 1Cognitive Functioning Score; Cycle 13 Day 1Cognitive Functioning Score; Cycle 14 Day 1Cognitive Functioning Score; Cycle 15 Day 1Cognitive Functioning Score; Cycle 16 Day 1Cognitive Functioning Score; Cycle 17 Day 1Cognitive Functioning Score; Cycle 18 Day 1Cognitive Functioning Score; Cycle 19 Day 1Cognitive Functioning Score; Cycle 20 Day 1Cognitive Functioning Score; Cycle 21 Day 1Cognitive Functioning Score; Cycle 22 Day 1Cognitive Functioning Score; Cycle 23 Day 1Cognitive Functioning Score; Cycle 24 Day 1Cognitive Functioning Score; Cycle 25 Day 1Cognitive Functioning Score; Cycle 26 Day 1Cognitive Functioning Score; Cycle 27 Day 1Cognitive Functioning Score; Cycle 28 Day 1Cognitive Functioning Score; Cycle 29 Day 1Cognitive Functioning Score; Cycle 30 Day 1Cognitive Functioning Score; Cycle 31 Day 1Cognitive Functioning Score; Off-treatment VisitSocial Functioning Score; BaselineSocial Functioning Score; Cycle 2 Day 1Social Functioning Score; Cycle 3 Day 1Social Functioning Score; Cycle 4 Day 1Social Functioning Score; Cycle 5 Day 1Social Functioning Score; Cycle 6 Day 1Social Functioning Score; Cycle 7 Day 1Social Functioning Score; Cycle 8 Day 1Social Functioning Score; Cycle 9 Day 1Social Functioning Score; Cycle 10 Day 1Social Functioning Score; Cycle 11 Day 1Social Functioning Score; Cycle 12 Day 1Social Functioning Score; Cycle 13 Day 1Social Functioning Score; Cycle 14 Day 1Social Functioning Score; Cycle 15 Day 1Social Functioning Score; Cycle 16 Day 1Social Functioning Score; Cycle 17 Day 1Social Functioning Score; Cycle 18 Day 1Social Functioning Score; Cycle 19 Day 1Social Functioning Score; Cycle 20 Day 1Social Functioning Score; Cycle 21 Day 1Social Functioning Score; Cycle 22 Day 1Social Functioning Score; Cycle 23 Day 1Social Functioning Score; Cycle 24 Day 1Social Functioning Score; Cycle 25 Day 1Social Functioning Score; Cycle 26 Day 1Social Functioning Score; Cycle 27 Day 1Social Functioning Score; Cycle 28 Day 1Social Functioning Score; Cycle 29 Day 1Social Functioning Score; Cycle 30 Day 1Social Functioning Score; Cycle 31 Day 1Social Functioning Score; Off-treatment VisitFatigue Score; BaselineFatigue Score; Cycle 2 Day 1Fatigue Score; Cycle 3 Day 1Fatigue Score; Cycle 4 Day 1Fatigue Score; Cycle 5 Day 1Fatigue Score; Cycle 6 Day 1Fatigue Score; Cycle 7 Day 1Fatigue Score; Cycle 8 Day 1Fatigue Score; Cycle 9 Day 1Fatigue Score; Cycle 10 Day 1Fatigue Score; Cycle 11 Day 1Fatigue Score; Cycle 12 Day 1Fatigue Score; Cycle 13 Day 1Fatigue Score; Cycle 14 Day 1Fatigue Score; Cycle 15 Day 1Fatigue Score; Cycle 16 Day 1Fatigue Score; Cycle 17 Day 1Fatigue Score; Cycle 18 Day 1Fatigue Score; Cycle 19 Day 1Fatigue Score; Cycle 20 Day 1Fatigue Score; Cycle 21 Day 1Fatigue Score; Cycle 22 Day 1Fatigue Score; Cycle 23 Day 1Fatigue Score; Cycle 24 Day 1Fatigue Score; Cycle 25 Day 1Fatigue Score; Cycle 26 Day 1Fatigue Score; Cycle 27 Day 1Fatigue Score; Cycle 28 Day 1Fatigue Score; Cycle 29 Day 1Fatigue Score; Cycle 30 Day 1Fatigue Score; Cycle 31 Day 1Fatigue Score; Off-treatment VisitNausea and Vomiting Score; BaselineNausea and Vomiting Score; Cycle 2 Day 1Nausea and Vomiting Score; Cycle 3 Day 1Nausea and Vomiting Score; Cycle 4 Day 1Nausea and Vomiting Score; Cycle 5 Day 1Nausea and Vomiting Score; Cycle 6 Day 1Nausea and Vomiting Score; Cycle 7 Day 1Nausea and Vomiting Score; Cycle 8 Day 1Nausea and Vomiting Score; Cycle 9 Day 1Nausea and Vomiting Score; Cycle 10 Day 1Nausea and Vomiting Score; Cycle 11 Day 1Nausea and Vomiting Score; Cycle 12 Day 1Nausea and Vomiting Score; Cycle 13 Day 1Nausea and Vomiting Score; Cycle 14 Day 1Nausea and Vomiting Score; Cycle 15 Day 1Nausea and Vomiting Score; Cycle 16 Day 1Nausea and Vomiting Score; Cycle 17 Day 1Nausea and Vomiting Score; Cycle 18 Day 1Nausea and Vomiting Score; Cycle 19 Day 1Nausea and Vomiting Score; Cycle 20 Day 1Nausea and Vomiting Score; Cycle 21 Day 1Nausea and Vomiting Score; Cycle 22 Day 1Nausea and Vomiting Score; Cycle 23 Day 1Nausea and Vomiting Score; Cycle 24 Day 1Nausea and Vomiting Score; Cycle 25 Day 1Nausea and Vomiting Score; Cycle 26 Day 1Nausea and Vomiting Score; Cycle 27 Day 1Nausea and Vomiting Score; Cycle 28 Day 1Nausea and Vomiting Score; Cycle 29 Day 1Nausea and Vomiting Score; Cycle 30 Day 1Nausea and Vomiting Score; Cycle 31 Day 1Nausea and Vomiting Score; Off-treatment VisitPain Score; BaselinePain Score; Cycle 2 Day 1Pain Score; Cycle 3 Day 1Pain Score; Cycle 4 Day 1Pain Score; Cycle 5 Day 1Pain Score; Cycle 6 Day 1Pain Score; Cycle 7 Day 1Pain Score; Cycle 8 Day 1Pain Score; Cycle 9 Day 1Pain Score; Cycle 10 Day 1Pain Score; Cycle 11 Day 1Pain Score; Cycle 12 Day 1Pain Score; Cycle 13 Day 1Pain Score; Cycle 14 Day 1Pain Score; Cycle 15 Day 1Pain Score; Cycle 16 Day 1Pain Score; Cycle 17 Day 1Pain Score; Cycle 18 Day 1Pain Score; Cycle 19 Day 1Pain Score; Cycle 20 Day 1Pain Score; Cycle 21 Day 1Pain Score; Cycle 22 Day 1Pain Score; Cycle 23 Day 1Pain Score; Cycle 24 Day 1Pain Score; Cycle 25 Day 1Pain Score; Cycle 26 Day 1Pain Score; Cycle 27 Day 1Pain Score; Cycle 28 Day 1Pain Score; Cycle 29 Day 1Pain Score; Cycle 30 Day 1Pain Score; Cycle 31 Day 1Pain Score; Off-treatment VisitDyspnea Score; BaselineDyspnea Score; Cycle 2 Day 1Dyspnea Score; Cycle 3 Day 1Dyspnea Score; Cycle 4 Day 1Dyspnea Score; Cycle 5 Day 1Dyspnea Score; Cycle 6 Day 1Dyspnea Score; Cycle 7 Day 1Dyspnea Score; Cycle 8 Day 1Dyspnea Score; Cycle 9 Day 1Dyspnea Score; Cycle 10 Day 1Dyspnea Score; Cycle 11 Day 1Dyspnea Score; Cycle 12 Day 1Dyspnea Score; Cycle 13 Day 1Dyspnea Score; Cycle 14 Day 1Dyspnea Score; Cycle 15 Day 1Dyspnea Score; Cycle 16 Day 1Dyspnea Score; Cycle 17 Day 1Dyspnea Score; Cycle 18 Day 1Dyspnea Score; Cycle 19 Day 1Dyspnea Score; Cycle 20 Day 1Dyspnea Score; Cycle 21 Day 1Dyspnea Score; Cycle 22 Day 1Dyspnea Score; Cycle 23 Day 1Dyspnea Score; Cycle 24 Day 1Dyspnea Score; Cycle 25 Day 1Dyspnea Score; Cycle 26 Day 1Dyspnea Score; Cycle 27 Day 1Dyspnea Score; Cycle 28 Day 1Dyspnea Score; Cycle 29 Day 1Dyspnea Score; Cycle 30 Day 1Dyspnea Score; Cycle 31 Day 1Dyspnea Score; Off-treatment VisitInsomnia Score; BaselineInsomnia Score; Cycle 2 Day 1Insomnia Score; Cycle 3 Day 1Insomnia Score; Cycle 4 Day 1Insomnia Score; Cycle 5 Day 1Insomnia Score; Cycle 6 Day 1Insomnia Score; Cycle 7 Day 1Insomnia Score; Cycle 8 Day 1Insomnia Score; Cycle 9 Day 1Insomnia Score; Cycle 10 Day 1Insomnia Score; Cycle 11 Day 1Insomnia Score; Cycle 12 Day 1Insomnia Score; Cycle 13 Day 1Insomnia Score; Cycle 14 Day 1Insomnia Score; Cycle 15 Day 1Insomnia Score; Cycle 16 Day 1Insomnia Score; Cycle 17 Day 1Insomnia Score; Cycle 18 Day 1Insomnia Score; Cycle 19 Day 1Insomnia Score; Cycle 20 Day 1Insomnia Score; Cycle 21 Day 1Insomnia Score; Cycle 22 Day 1Insomnia Score; Cycle 23 Day 1Insomnia Score; Cycle 24 Day 1Insomnia Score; Cycle 25 Day 1Insomnia Score; Cycle 26 Day 1Insomnia Score; Cycle 27 Day 1Insomnia Score; Cycle 28 Day 1Insomnia Score; Cycle 29 Day 1Insomnia Score; Cycle 30 Day 1Insomnia Score; Cycle 31 Day 1Insomnia Score; Off-treatment VisitAppetite Loss Score; BaselineAppetite Loss Score; Cycle 2 Day 1Appetite Loss Score; Cycle 3 Day 1Appetite Loss Score; Cycle 4 Day 1Appetite Loss Score; Cycle 5 Day 1Appetite Loss Score; Cycle 6 Day 1Appetite Loss Score; Cycle 7 Day 1Appetite Loss Score; Cycle 8 Day 1Appetite Loss Score; Cycle 9 Day 1Appetite Loss Score; Cycle 10 Day 1Appetite Loss Score; Cycle 11 Day 1Appetite Loss Score; Cycle 12 Day 1Appetite Loss Score; Cycle 13 Day 1Appetite Loss Score; Cycle 14 Day 1Appetite Loss Score; Cycle 15 Day 1Appetite Loss Score; Cycle 16 Day 1Appetite Loss Score; Cycle 17 Day 1Appetite Loss Score; Cycle 18 Day 1Appetite Loss Score; Cycle 19 Day 1Appetite Loss Score; Cycle 20 Day 1Appetite Loss Score; Cycle 21 Day 1Appetite Loss Score; Cycle 22 Day 1Appetite Loss Score; Cycle 23 Day 1Appetite Loss Score; Cycle 24 Day 1Appetite Loss Score; Cycle 25 Day 1Appetite Loss Score; Cycle 26 Day 1Appetite Loss Score; Cycle 27 Day 1Appetite Loss Score; Cycle 28 Day 1Appetite Loss Score; Cycle 29 Day 1Appetite Loss Score; Cycle 30 Day 1Appetite Loss Score; Cycle 31 Day 1Appetite Loss Score; Off-treatment VisitConstipation Score; BaselineConstipation Score; Cycle 2 Day 1Constipation Score; Cycle 3 Day 1Constipation Score; Cycle 4 Day 1Constipation Score; Cycle 5 Day 1Constipation Score; Cycle 6 Day 1Constipation Score; Cycle 7 Day 1Constipation Score; Cycle 8 Day 1Constipation Score; Cycle 9 Day 1Constipation Score; Cycle 10 Day 1Constipation Score; Cycle 11 Day 1Constipation Score; Cycle 12 Day 1Constipation Score; Cycle 13 Day 1Constipation Score; Cycle 14 Day 1Constipation Score; Cycle 15 Day 1Constipation Score; Cycle 16 Day 1Constipation Score; Cycle 17 Day 1Constipation Score; Cycle 18 Day 1Constipation Score; Cycle 19 Day 1Constipation Score; Cycle 20 Day 1Constipation Score; Cycle 21 Day 1Constipation Score; Cycle 22 Day 1Constipation Score; Cycle 23 Day 1Constipation Score; Cycle 24 Day 1Constipation Score; Cycle 25 Day 1Constipation Score; Cycle 26 Day 1Constipation Score; Cycle 27 Day 1Constipation Score; Cycle 28 Day 1Constipation Score; Cycle 29 Day 1Constipation Score; Cycle 30 Day 1Constipation Score; Cycle 31 Day 1Constipation Score; Off-treatment VisitDiarrhea Score; BaselineDiarrhea Score; Cycle 2 Day 1Diarrhea Score; Cycle 3 Day 1Diarrhea Score; Cycle 4 Day 1Diarrhea Score; Cycle 5 Day 1Diarrhea Score; Cycle 6 Day 1Diarrhea Score; Cycle 7 Day 1Diarrhea Score; Cycle 8 Day 1Diarrhea Score; Cycle 9 Day 1Diarrhea Score; Cycle 10 Day 1Diarrhea Score; Cycle 11 Day 1Diarrhea Score; Cycle 12 Day 1Diarrhea Score; Cycle 13 Day 1Diarrhea Score; Cycle 14 Day 1Diarrhea Score; Cycle 15 Day 1Diarrhea Score; Cycle 16 Day 1Diarrhea Score; Cycle 17 Day 1Diarrhea Score; Cycle 18 Day 1Diarrhea Score; Cycle 19 Day 1Diarrhea Score; Cycle 20 Day 1Diarrhea Score; Cycle 21 Day 1Diarrhea Score; Cycle 22 Day 1Diarrhea Score; Cycle 23 Day 1Diarrhea Score; Cycle 24 Day 1Diarrhea Score; Cycle 25 Day 1Diarrhea Score; Cycle 26 Day 1Diarrhea Score; Cycle 27 Day 1Diarrhea Score; Cycle 28 Day 1Diarrhea Score; Cycle 29 Day 1Diarrhea Score; Cycle 30 Day 1Diarrhea Score; Cycle 31 Day 1Diarrhea Score; Off-treatment VisitFinancial Difficulties Score; BaselineFinancial Difficulties Score; Cycle 2 Day 1Financial Difficulties Score; Cycle 3 Day 1Financial Difficulties Score; Cycle 4 Day 1Financial Difficulties Score; Cycle 5 Day 1Financial Difficulties Score; Cycle 6 Day 1Financial Difficulties Score; Cycle 7 Day 1Financial Difficulties Score; Cycle 8 Day 1Financial Difficulties Score; Cycle 9 Day 1Financial Difficulties Score; Cycle 10 Day 1Financial Difficulties Score; Cycle 11 Day 1Financial Difficulties Score; Cycle 12 Day 1Financial Difficulties Score; Cycle 13 Day 1Financial Difficulties Score; Cycle 14 Day 1Financial Difficulties Score; Cycle 15 Day 1Financial Difficulties Score; Cycle 16 Day 1Financial Difficulties Score; Cycle 17 Day 1Financial Difficulties Score; Cycle 18 Day 1Financial Difficulties Score; Cycle 19 Day 1Financial Difficulties Score; Cycle 20 Day 1Financial Difficulties Score; Cycle 21 Day 1Financial Difficulties Score; Cycle 22 Day 1Financial Difficulties Score; Cycle 23 Day 1Financial Difficulties Score; Cycle 24 Day 1Financial Difficulties Score; Cycle 25 Day 1Financial Difficulties Score; Cycle 26 Day 1Financial Difficulties Score; Cycle 27 Day 1Financial Difficulties Score; Cycle 28 Day 1Financial Difficulties Score; Cycle 29 Day 1Financial Difficulties Score; Cycle 30 Day 1Financial Difficulties Score; Cycle 31 Day 1Financial Difficulties Score; Off-treatment Visit
Lenvatinib 14 mg + Everolimus 5 mg63.3558.6056.4358.7159.3858.7558.7258.6858.9763.1360.8757.8159.3961.5662.9861.5960.8859.6858.3358.9557.0562.726064.4458.3359.3859.7258.3356.2554.1733.3351.0476.3673.5472.7470.9072.8072.1773.0272.7774.5475.0673.7275.5274.5575.7875.4573.4671.8575.4876.6773.0974.6280.3579.5677.3372.7876.6776.6778.3378.3376.678064.5073.7868.256766.6767.3268.1666.5168.2067.5868.5269.5770.5169.6471.0972.7370.3365.7467.7467.2870.3769.2375.4473.3378.8977.787577.7879.177587.5066.6760.9779.3579.4178.4778.2675.8580.6077.1879.0878.7580.0482.2581.5179.3281.3580.3081.5079.6381.9081.1780.2581.7385.0986.6788.8983.3377.0880.5675757566.6769.6287.9187.1584.4583.5883.9984.7182.4081.9783.7085.3984.0685.4283.9385.7184.8583.7482.8783.8780.2582.1081.4188.6083.3384.4484.7289.5883.3391.6791.6787.5083.3375.2177.2973.4672.0471.4371.7870.8071.7070.2472.1672.4373.9176.5672.9272.7972.7373.1770.3776.8874.0775.3173.7279.8282.2283.3387.5083.3380.5683.3379.1770.8310064.5831.3239.2140.0940.2137.5438.8038.0636.1435.9034.5732.3732.8235.5230.8428.0333.3333.3334.0537.0434.5734.1926.3226.6728.1533.3326.3927.7822.2222.222533.3343.477.6011.5014.8615.8012.0712.7412.4613.1012.2715.0211.1112.8212.2014.9714.0211.3814.3510.7516.6716.6712.827.025.566.6713.894.175.5612.5016.678.3316.6713.3323.3926.7927.6326.6729.0426.2928.0427.3827.1126.9527.2926.4124.4022.1124.2429.6727.7825.2731.4829.6326.9223.6821.1122.2227.7827.082525252533.3328.3321.1818.9020.6319.9019.1616.6719.3719.2415.9317.9218.6313.5413.6915.6515.1516.2615.7416.1315.3823.4615.3817.5413.332022.2216.6711.1116.672516.6733.3324.6823.0826.1130.3926.5723.0224.8625.0826.4625.5624.5824.0223.4421.4318.0618.9420.3317.5927.9624.6925.9323.0821.0526.6724.4416.6716.6722.2225252533.3332.0515.7926.7931.9732.0930.4730.3529.2829.2529.3027.5724.1522.0523.2119.7317.4223.5820.3719.3519.7523.4623.0817.5417.7813.3319.4429.1716.678.3316.678.33029.5815.9810.409.5211.1112.248.4711.019.626.236.586.765.643.645.445.435.9811.117.539.888.648.977.026.674.445.568.3316.6725.0025.0016.6733.3313.336.4322.2932.2135.5928.6131.9633.6429.5534.4329.2228.0228.4926.1926.5329.5520.3321.3021.5123.4619.7520.5117.5415.5611.1119.4425.0033.3316.678.3316.6733.3318.3321.7621.0823.1322.9022.9325.0030.1625.4325.5622.5026.2624.8723.0320.8321.7121.6723.8117.2018.5216.0519.2324.5617.7817.7816.6720.8316.6725.0025.0025.00031.65

[back to top]

Objective Response Rate (ORR)

ORR was defined as the percentage of participants with a BOR of CR or PR at the at the end of treatment based on investigator assessment according to RECIST v1.1. CR: defined as the disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis to <10 mm. PR: defined as at least a 30 % decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. To be considered a BOR, all responses had to be confirmed no less than 4 weeks after the initial assessment of response. (NCT03173560)
Timeframe: From date of randomization up to first documentation of PD or date of death, whichever occurred first or up to the date of data cut off for the primary analysis (up to 29 months)

Interventionpercentage of participants (Number)
Lenvatinib 14 mg + Everolimus 5 mg34.6
Lenvatinib 18 mg + Everolimus 5 mg40.6

[back to top]

HRQoL Assessed by European Quality of Life (EuroQol) Five-Dimensional, 3-Level (EQ-5D-3L) Index Score and Visual Analogue Scale (VAS)

"The EQ-5D-3L is a health profile questionnaire assessing quality of life along 5 dimensions. Participants rate 5 dimensions of health (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) by choosing from 3 answering options (1=no problems; 2=some problems; 3=extreme problems). The summed score ranges from 5-15 with 5 corresponding to no problems and 15 corresponding to severe problems in the 5 dimensions. The EQ-5D index was calculated by applying preference-based weights (tariffs) to the scores of the five health state dimensions. Index values can range from -1 to 1, with 0 representing a health state equivalent to death and 1 representing perfect health. EQ-5D-3L also included an EQ visual analogue scale (VAS) that ranges between 100 (best imaginable health) and 0 (worst imaginable health). Decrease from baseline in EQ-5D-3L signifies improvement. Total index EQ-5D-3L summary score was weighted with a range of -0.594 (worst) to 1.0 (best)." (NCT03173560)
Timeframe: At baseline (prior to first dose of study drug), on Day 1 of each subsequent cycle (cycle length =28 days), and at the Off-treatment visit (up to 29 months)

Interventionscore on a scale (Mean)
EQ-5D Index Score; BaselineEQ-5D Index Score; Cycle 2 Day 1EQ-5D Index Score; Cycle 3 Day 1EQ-5D Index Score; Cycle 4 Day 1EQ-5D Index Score; Cycle 5 Day 1EQ-5D Index Score; Cycle 6 Day 1EQ-5D Index Score; Cycle 7 Day 1EQ-5D Index Score; Cycle 8 Day 1EQ-5D Index Score; Cycle 9 Day 1EQ-5D Index Score; Cycle 10 Day 1EQ-5D Index Score; Cycle 11 Day 1EQ-5D Index Score; Cycle 12 Day 1EQ-5D Index Score; Cycle 13 Day 1EQ-5D Index Score; Cycle 14 Day 1EQ-5D Index Score; Cycle 15 Day 1EQ-5D Index Score; Cycle 16 Day 1EQ-5D Index Score; Cycle 17 Day 1EQ-5D Index Score; Cycle 18 Day 1EQ-5D Index Score; Cycle 19 Day 1EQ-5D Index Score; Cycle 20 Day 1EQ-5D Index Score; Cycle 21 Day 1EQ-5D Index Score; Cycle 22 Day 1EQ-5D Index Score; Cycle 23 Day 1EQ-5D Index Score; Cycle 24 Day 1EQ-5D Index Score; Cycle 25 Day 1EQ-5D Index Score; Cycle 26 Day 1EQ-5D Index Score; Cycle 27 Day 1EQ-5D Index Score; Cycle 28 Day 1EQ-5D Index Score; Cycle 29 Day 1EQ-5D Index Score; Cycle 30 Day 1EQ-5D Index Score; Cycle 31 Day 1EQ-5D Index Score; Off-treatment VisitEQ-VAS Score; BaselineEQ-VAS Score; Cycle 2 Day 1EQ-VAS Score; Cycle 3 Day 1EQ-VAS Score; Cycle 4 Day 1EQ-VAS Score; Cycle 5 Day 1EQ-VAS Score; Cycle 6 Day 1EQ-VAS Score; Cycle 7 Day 1EQ-VAS Score; Cycle 8 Day 1EQ-VAS Score; Cycle 9 Day 1EQ-VAS Score; Cycle 10 Day 1EQ-VAS Score; Cycle 11 Day 1EQ-VAS Score; Cycle 12 Day 1EQ-VAS Score; Cycle 13 Day 1EQ-VAS Score; Cycle 14 Day 1EQ-VAS Score; Cycle 15 Day 1EQ-VAS Score; Cycle 16 Day 1EQ-VAS Score; Cycle 17 Day 1EQ-VAS Score; Cycle 18 Day 1EQ-VAS Score; Cycle 19 Day 1EQ-VAS Score; Cycle 20 Day 1EQ-VAS Score; Cycle 21 Day 1EQ-VAS Score; Cycle 22 Day 1EQ-VAS Score; Cycle 23 Day 1EQ-VAS Score; Cycle 24 Day 1EQ-VAS Score; Cycle 25 Day 1EQ-VAS Score; Cycle 26 Day 1EQ-VAS Score; Cycle 27 Day 1EQ-VAS Score; Cycle 28 Day 1EQ-VAS Score; Cycle 29 Day 1EQ-VAS Score; Cycle 30 Day 1EQ-VAS Score; Cycle 31 Day 1EQ-VAS Score; Off-treatment Visit
Lenvatinib 14 mg + Everolimus 5 mg0.760.760.730.730.730.730.710.750.730.760.770.770.760.770.760.730.730.760.690.700.730.740.740.750.700.720.770.730.740.610.870.6168.5766.0566.2265.6965.8665.5365.7464.1665.5166.9267.7466.4665.6466.4167.0566.5465.7864.7062.1562.7062.1265.2167.7365.2760.5864.1361.8354.2552.7551.753558.48

[back to top]

Objective Response Rate at Week 24 (ORR24W)

ORR24W was defined as the percentage of participants with a best overall response (BOR) of complete response (CR) or partial response (PR) at the Week 24 (after randomization) time point, during treatment or within 28 days after the last dose date but on or prior to the start of new anticancer therapy based on investigator assessment according to Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1. CR: defined as the disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis to less than (<) 10 millimeters (mm). PR: defined as at least a 30 percent (%) decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. To be considered a BOR, all responses had to be confirmed no less than 4 weeks after the initial assessment of response. (NCT03173560)
Timeframe: At Week 24

Interventionpercentage of participants (Number)
Lenvatinib 14 mg + Everolimus 5 mg32.1
Lenvatinib 18 mg + Everolimus 5 mg34.8

[back to top]

Percentage of Participants With Intolerable Grade 2 or Any Grade >=Grade 3 TEAEs Within 24 Weeks

TEAE was defined as an adverse event (AE) with an onset that had occurred after receiving study drug. A severity grade was defined by the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.03. As per NCI-CTCAE, Grade 1 scales as Mild; Grade 2 scales as Moderate; Grade 3 scales as severe or medically significant but not immediately life threatening; Grade 4 scales as life-threatening consequences; and Grade 5 scales as death related to AE. (NCT03173560)
Timeframe: Up to Week 24

Interventionpercentage of participants (Number)
Lenvatinib 14 mg + Everolimus 5 mg82.8
Lenvatinib 18 mg + Everolimus 5 mg79.6

[back to top]

Percentage of Participants Who Discontinued Treatment Due to Toxicity

Percentage of participants who discontinued treatment due to toxicity, defined as the percentage of participants who discontinued study treatment due to TEAEs. Toxicity (except hypertension and non-infectious pneumonitis) was assessed according to NCI-CTCAE v4.03. (NCT03173560)
Timeframe: From date of first dose of study drug up to 28 days after last dose of study drug, or date of data cut off for the primary analysis (up to 29 months)

Interventionpercentage of participants (Number)
Lenvatinib 14 mg + Everolimus 5 mg15.7
Lenvatinib 18 mg + Everolimus 5 mg17.5

[back to top]

Progression-free Survival (PFS)

PFS was defined as the time from the date of randomization to the date of the first documentation of PD by investigator assessment or date of death, whichever occurred first according to RECIST v1.1. PD: at least 20% increase (including an absolute increase of at least 5 millimeter [mm]) in the sum of diameter (SOD) of target lesions, taking as reference the smallest sum and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. Median PFS was analyzed using the Kaplan-Meier product-limit estimates for each treatment group and presented with 2-sided 95% confidence interval (CI). (NCT03173560)
Timeframe: From the date of randomization to the date of the first documentation of PD or date of death, whichever occurred first or up to date of data cutoff for the primary analysis (up to 29 months)

Interventionmonths (Median)
Lenvatinib 14 mg + Everolimus 5 mg11.1
Lenvatinib 18 mg + Everolimus 5 mg14.7

[back to top] [back to top]

Time to Treatment Failure Due to Toxicity

Time to treatment failure due to toxicity was defined as the time from the date of randomization to the date that a participant discontinued study treatment due to TEAEs. Toxicity (except hypertension and non-infectious pneumonitis) was assessed according to CTCAE v4.03. (NCT03173560)
Timeframe: From the date of randomization to the date of discontinuation of study treatment due to TEAEs, or date of data cut off for the primary analysis (up to 29 months)

Interventionmonths (Median)
Lenvatinib 14 mg + Everolimus 5 mg3.15
Lenvatinib 18 mg + Everolimus 5 mg5.70

[back to top]

Percentage of Participants Response Rates (Best Overall and Overall)

The best overall response for each patient is determined from the sequence of investigator overall lesion responses according to RECIST 1.1: Complete Response (CR)=disappearance of target lesions, Partial Response (PR) was >=30% decrease in sum of diameter of lesions, Progressive Disease (PD) was >=20% decrease in sum of diameter of lesions, Stable Disease (SD) does not qualify for PR, CR or PD, Unknown=not documented or assessed. To be assigned a best overall response of CR at least two determinations of CR at least 4 weeks apart before progression are required. To be assigned a best overall response of PR at least two determinations of PR or better at least 4 weeks apart before progression (and not qualifying for a CR) are required. The Overall response rate (ORR) is the percentage of patients with a best overall response of confirmed complete (CR) or partial (PR) response. The 95% confidence intervals (CI) were computed using the Clopper-Pearson method (NCT03176238)
Timeframe: Baseline up to approximately 43 weeks for Asian countires and 32 weeks for Non-Asian countries

,,
InterventionPercentage of participants (Number)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Unknown (UNK)Overall response rate (ORR: CR+PR)
Asian Everolimus + Exemestane1.520.151.318.19.021.6
Non-Asian Everolimus + Exemestane0.008.363.919.48.38.3
Total1.318.353.218.38.919.6

[back to top]

Summary of Number of Participants With Treatment Emergent Adverse Events (TEAE) - All Grades

Adverse events (AEs), serious adverse events (SAEs), changes from baseline in vital signs and laboratory results (hematology, blood chemistry, lipid profile) qualifying and reported as AEs. Although a patient might had two or more adverse events the patient is only counted once in a category. The same patient might appear in different categories. AESI: Adverse events of special interest. (NCT03176238)
Timeframe: Baseline up to approximately 43 weeks for Asian countires and 32 weeks for Non-Asian countries including a 30 day post treatment follow up period

,,
InterventionParticipants (Count of Participants)
no TEAEat least 1 TEAEat least 1 drug-related TEAEat least 1 serious TEAE (STEAE)STEAE leading to deathNon-fatal STEAEat least 1 drug-related STEAEat least 1 drug-related STEAE - deathat least 1 drug-related non-fatal STEAETEAE leading to permanent tx discontinuation1 TE AESI
Asian Everolimus + Exemestane4195185596532822625172
Non-Asian Everolimus + Exemestane03633164128171134
Total42312187510653633336206

[back to top]

Percent of Participants Event-free Probability Estimates of Deterioration of Eastern Cooperative Oncology Group (ECOG) Performance Status

Time to deterioration of ECOG performance status, from baseline will be assessed using the ECOG Performance Status Scale (Oken, 1982). Time to deterioration is the time from date of start of treatment to the date of the event defined as deterioration. Deterioration is defined as an increase in performance status from 0 to 2 or greater, an increase in performance status from 1-2 to 3 or greater, or death due to any cause. Event-free probability estimate is the estimated probability that a patient will remain event-free up to the specified time point. Event-free probability estimates were are obtained from the Kaplan-Meier survival estimates. (NCT03176238)
Timeframe: Baseline up to approximately 50 weeks

,,
Interventionpercent of participants (Number)
Week 0 n=199,36,235Week 20 n=134,18,152Week 50 n=58,7,65
Asian Everolimus + Exemestane100.095.090.6
Non-Asian Everolimus + Exemestane100.079.870.4
Total100.0092.787.6

[back to top]

Percentage of Participants Clinical Benefit Rate

Clinical benefit rate: Patients with best overall response rate of CR (any duration), PR (any duration) and SD with duration of 24 weeks or longer according to RECIST 1.1 criteria: Complete Response (CR)=disappearance of target lesions, Partial Response (PR) was >=30% decrease in sum of diameter of lesions, Progressive Disease (PD) was >=20% decrease in sum of diameter of lesions, Stable Disease (SD) does not qualify for PR, CR or PD, Unknown=not documented or assessed. Best overall response of CR = at least two determinations of CR at least 4 weeks apart before progression are required. Best overall response of PR = at least two determinations of PR or better at least 4 weeks apart before progression (and not qualifying for a CR) are required. The Overall response rate (ORR) is the percentage of patients with a best overall response of confirmed complete (CR) or partial (PR) response. The 95% confidence intervals (CI) were computed using the Clopper-Pearson method. (NCT03176238)
Timeframe: Baseline up to approximately 43 weeks for Asian countires and 32 weeks for Non-Asian countries

InterventionPercentage of participants (Number)
Asian Everolimus + Exemestane48.2
Non-Asian Everolimus + Exemestane30.6
Total45.5

[back to top]

Progression Free Survival (PFS)

"PFS is time from date of start of treatment to date of disease progression or death due to any cause, whichever occurs first.~b Percentiles with 95% CIs are calculated from PROC LIFETEST output using method of Brookmeyer and Crowley (1982)" (NCT03176238)
Timeframe: Baseline up to approximately 43 weeks for Asian countires and 40 weeks for Non-Asian countries

Interventionweeks (Median)
Asian Everolimus and Exemestane40.6
Non-Asian Everolimus + Exemestane37.0
Total40.6

[back to top]

Phase 2: Objective Response Rate (ORR)

ORR was defined as the percentage of participants with BOR of CR or PR based on investigator assessment according to RECIST version 1.1 for non-HGG cohorts and RANO for HGG cohort. CR was defined as disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR was defined as at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. (NCT03245151)
Timeframe: From the date of the first dose of study drug to the date of first documentation of disease progression or death, which ever occurred first (up to 6.5 months)

Interventionpercentage of participants (Number)
Phase 2: Cohort 1, Ewing Sarcoma0.0
Phase 2: Cohort 2, Rhabdomyosarcoma10.0
Phase 2: Cohort 3, HGG0.0

[back to top]

Phase 2: Number of Participants With Any Treatment-emergent Serious Adverse Event (TESAE)

A TESAE was any untoward medical occurrence that at any dose: resulted in death; life threatening condition; required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity; was a congenital anomaly/birth defect or was medically important due to other reasons than the above mentioned criteria. An adverse event was defined as any untoward medical occurrence in a participant administered an investigational product. (NCT03245151)
Timeframe: From date of first dose up to 28 days after the last dose of study treatment (up to 7.5 months)

InterventionParticipants (Count of Participants)
Phase 2: Cohort 1, Ewing Sarcoma6
Phase 2: Cohort 2, Rhabdomyosarcoma8
Phase 2: Cohort 3, HGG8

[back to top]

Phase 2: Number of Participants With Any Treatment-emergent Adverse Event (TEAE)

A TEAE was defined as an adverse event that emerged during treatment, having been absent at pretreatment or reemerged during treatment, having been present at pretreatment but stopped before treatment, or worsened in severity during treatment relative to the pretreatment state, when the adverse event is continuous. An adverse event was defined as any untoward medical occurrence in a participant administered an investigational product. (NCT03245151)
Timeframe: From date of first dose up to 28 days after the last dose of study treatment (up to 7.5 months)

InterventionParticipants (Count of Participants)
Phase 2: Cohort 1, Ewing Sarcoma10
Phase 2: Cohort 2, Rhabdomyosarcoma19
Phase 2: Cohort 3, HGG11

[back to top]

Phase 1: Disease Control Rate (DCR)

DCR was defined as percentage of participants with a confirmed CR, PR, or stable disease (SD) (SD duration >=7 weeks since the first dose of study treatment) divided by number of participants in analysis set. DCR was assessed by an investigator based on RECIST v1.1 for non-HGG participants or RANO for HGG participants. CR: disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR: at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD. PD: at least 20% increase (including an absolute increase of at least 5 mm) in the SOD of target lesions, taking as reference the smallest sum and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT03245151)
Timeframe: From first dose of study drug until PD or death, whichever occurred first (up to 16.5 months)

Interventionpercentage of participants (Number)
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^220.0
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^250.0

[back to top]

Phase 2: Duration of Response (DOR)

DOR was defined as the time (in months) from the date of first observation of confirmed response (PR or CR) to the date of the first observation of progression based on the investigator's assessment utilizing RECIST 1.1 for non-HGG cohorts and RANO for HGG cohort, or date of death, whatever the cause. CR: disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR: at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. PD was defined as at least 20% increase (including an absolute increase of at least 5 mm) in the SOD of target lesions, taking as reference the smallest sum and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT03245151)
Timeframe: From date of the first observation of CR or PR until the date of first observation of progression or date of death (up to 6.5 months)

Interventionmonths (Median)
Phase 2: Cohort 2, Rhabdomyosarcoma2.4

[back to top]

Phase 2: Disease Control Rate (DCR)

DCR was defined as percentage of participants with confirmed CR, PR, or SD (SD duration greater than or equal to [>=] 7 weeks since first dose of study treatment) divided by number of participants in analysis set. DCR was assessed by investigator based on RECIST v1.1 for non-HGG cohorts or RANO for HGG cohort. CR: disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR: at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD. PD: at least 20% increase (including an absolute increase of at least 5 mm) in the SOD of target lesions, taking as reference the smallest sum and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT03245151)
Timeframe: From first dose of study drug until PD or death, whichever occurred first (up to 6.5 months)

Interventionpercentage of participants (Number)
Phase 2: Cohort 1, Ewing Sarcoma40.0
Phase 2: Cohort 2, Rhabdomyosarcoma40.0
Phase 2: Cohort 3, HGG30.0

[back to top]

Phase 2: Clinical Benefit Rate (CBR)

CBR was defined as percentage of participants who had BOR of CR, PR, or durable SD (SD duration >=23 weeks since the first dose of study treatment) divided by number of participants in analysis set. CBR was assessed by an investigator based on RECIST v1.1 for non-HGG cohorts or RANO for HGG cohort. CR: disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR: at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD. PD: at least 20% increase (including an absolute increase of at least 5 mm) in the SOD of target lesions, taking as reference the smallest sum and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT03245151)
Timeframe: From first dose of study drug until PD or death, whichever occurred first (up to 6.5 months)

Interventionpercentage of participants (Number)
Phase 2: Cohort 1, Ewing Sarcoma20.0
Phase 2: Cohort 2, Rhabdomyosarcoma10.0
Phase 2: Cohort 3, HGG0.0

[back to top] [back to top]

Phase 1: Objective Response Rate (ORR)

ORR was defined as the percentage of participants with BOR of CR or PR based on investigator assessment according to RECIST version 1.1 for non-HGG cohorts and RANO for HGG cohort. CR was defined as disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR was defined as at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. (NCT03245151)
Timeframe: From the date of the first dose of study drug to the date of first documentation of disease progression or death, whichever occurred first (up to 16.5 months)

Interventionpercentage of participants (Number)
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^20.0
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^20.0

[back to top]

Phase 1: Number of Participants With Any Treatment-emergent Serious Adverse Event (TESAE)

A TESAE was any untoward medical occurrence that at any dose: resulted in death; life threatening condition; required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity; was a congenital anomaly/birth defect or was medically important due to other reasons than the above mentioned criteria. An adverse event was defined as any untoward medical occurrence in a participant administered an investigational product. (NCT03245151)
Timeframe: From date of first dose up to 28 days after the last dose of study treatment (Up to 17.5 months)

InterventionParticipants (Count of Participants)
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^22
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^212

[back to top]

Phase 1: Number of Participants With Any Treatment-emergent Adverse Event (TEAE)

A TEAE was defined as an adverse event that emerged during treatment, having been absent at pretreatment or reemerged during treatment, having been present at pretreatment but stopped before treatment, or worsened in severity during treatment relative to the pretreatment state, when the adverse event is continuous. An adverse event was defined as any untoward medical occurrence in a participant administered an investigational product. (NCT03245151)
Timeframe: From date of first dose up to 28 days after the last dose of study treatment (Up to 17.5 months)

InterventionParticipants (Count of Participants)
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^25
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^218

[back to top]

Phase 1: Maximum Tolerated Dose (MTD) of Lenvatinib in Combination With Everolimus

MTD was defined as the highest dose level at which no more than 1/6 participants experienced a dose limiting toxicity (DLTs), with the next higher dose having at least 0 of 3 or 1 of 6 participants experiencing DLTs. DLT was graded according to common terminology criteria for adverse events (CTCAE) version 4.03. (NCT03245151)
Timeframe: Cycle 1 (Each cycle was of 28 days)

Interventionmilligram per square meter (mg/m^2) (Number)
Phase 1: Lenvatinib + Everolimus (All Participants)11

[back to top]

Phase 1: Clinical Benefit Rate (CBR)

CBR was defined as percentage of participants who had BOR of CR, PR, or durable SD (SD duration >=23 weeks since the first dose of study treatment) divided by number of participants in analysis set. CBR was assessed by an investigator based on RECIST v1.1 for non-HGG participants or RANO for HGG participants. CR: disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis <10 mm. PR: at least a 30% decrease in the SOD of target lesions, taking as reference the baseline sum diameters. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD. PD: at least 20% increase (including an absolute increase of at least 5 mm) in the SOD of target lesions, taking as reference the smallest sum and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT03245151)
Timeframe: From first dose of study drug until PD or death, whichever occurred first (up to 16.5 months)

Interventionpercentage of participants (Number)
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^220.0
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^222.2

[back to top]

Phase 1: Trough Concentrations (Ctrough) of Everolimus When Administered in Combination With Lenvatinib

Trough concentrations of everolimus was quantified using validated liquid LC-MS/MS methods. (NCT03245151)
Timeframe: Cycle 1 Days 1, 2, 15 and 22: Pre-dose (Cycle length=28 days)

,
Interventionng/mL (Mean)
Cycle 1 Day 1: Pre-doseCycle 1 Day 2: Pre-doseCycle 1 Day 15: Pre-doseCycle 1 Day 22: Pre-dose
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^20.02.25.14.2
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^20.02.13.22.8

[back to top]

Phase 1: Time to Reach Maximum Plasma Concentration (Cmax) of Lenvatinib (Tmax)

Tmax of lenvatinib was quantified using validated liquid LC-MS/MS methods. (NCT03245151)
Timeframe: Cycle 1 Days 1 and 15: 0-8 hours post-dose (Cycle length=28 days)

,
Interventionhours (Median)
Cycle 1 Day 1Cycle 1 Day 15
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^22.8902.950
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^23.0003.950

[back to top]

Phase 1: Maximum Plasma Concentration of Lenvatinib (Cmax)

Cmax of lenvatinib was quantified using validated liquid LC-MS/MS methods. (NCT03245151)
Timeframe: Cycle 1 Days 1 and 15: 0-8 hours post-dose (Cycle length=28 days)

,
Interventionnanogram per milliliter (ng/mL) (Mean)
Cycle 1 Day 1Cycle 1 Day 15
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^2404.13447.62
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^2240.20314.20

[back to top]

Phase 1: Area Under the Plasma Concentration-time Curve From Time Zero to Time of Last Quantifiable Concentration of Lenvatinib (AUC[0-t Hours])

AUC0-t of lenvatinib was quantified using validated liquid chromatography tandem mass spectrometry (LC-MS/MS) methods. (NCT03245151)
Timeframe: Cycle 1 Days 1 and 15: 0-8 hours post-dose (Cycle length=28 days)

,
Interventionnanogram*hour per milliliter (ng*hr/mL) (Mean)
Cycle 1 Day 1Cycle 1 Day 15
Phase 1: Lenvatinib 11 mg/m^2 + Everolimus 3 mg/m^23281.12139.8
Phase 1: Lenvatinib 8 mg/m^2 + Everolimus 3 mg/m^22338.01328.0

[back to top]

Phase 2: Objective Response Rate (ORR) at Week 16

ORR at Week 16 was defined as the percentage of participants with a best overall response (BOR) of complete response (CR) or partial response (PR) at Week 16 based on investigator assessment according to response evaluation criteria in solid tumors (RECIST) version 1.1 for non-HGG cohorts and response assessment in neuro-oncology (RANO) for HGG cohort. CR was defined as disappearance of all target and non-target lesions (non-lymph nodes). All pathological lymph nodes (whether target or non-target) must have a reduction in their short axis less than (<) 10 millimeter (mm). PR was defined as at least a 30 percent (%) decrease in the sum of diameter (SOD) of target lesions, taking as reference the baseline sum diameters. (NCT03245151)
Timeframe: Week 16

Interventionpercentage of participants (Number)
Phase 2: Cohort 1, Ewing Sarcoma0.0
Phase 2: Cohort 2, Rhabdomyosarcoma10.0
Phase 2: Cohort 3, HGG0

[back to top]

Progression-free Survival in Participants With Newly Diagnosed High-grade Glioma (HGG)

Percentage of participants without progression, defined as 25% increase in the size of the tumor or appearance of new lesions. (NCT03352427)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Dasatinib+Everolimus0

[back to top]

Progression-free Survival in Participants With Newly Diagnosed Diffuse Intrinsic Pontine Glioma (DIPG)

Percentage of participants without progression, defined as 25% increase in the size of the tumor or appearance of new lesions. (NCT03352427)
Timeframe: 8 months

Interventionpercentage of participants (Number)
Dasatinib+Everolimus0

[back to top]

Overall Survival

Percentage of patients alive at one year. (NCT03352427)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Dasatinib+Everolimus0

[back to top]

Overall Response Rate (OR) (Partial Response or Better) in Participants With Refractory or Recurrent Glioma

The overall response assessment will take into account response in both target and non-target lesions, as well as the appearance of new lesions. Partial Response (PR) will be defined as ≥50% decrease in size of tumor in comparison to baseline measurements. Complete Response (CR) will be defined as the disappearance of all abnormal signal. This includes return to normal size of the brain stem for brain stem lesions. Reported as percentage of participants with partial or better response at 56 days. (NCT03352427)
Timeframe: 56 Days

Interventionpercentage of participants (Number)
Dasatinib+Everolimus0

[back to top]

Overall Survival

Percentage of participants alive at 2 years. (NCT03352427)
Timeframe: up to 17 months

Interventionpercentage of participants (Number)
Dasatinib+Everolimus0

[back to top]

Ribociclib Half-Life as Obtained From the Phase 1 Study

Ribociclib plasma concentration-time data obtained from patients received Phase I defined treatment will be modeled using compartmental approaches to estimate the half-life (hrs) for each dose level on days 1 and 17 of course 1. Patients who were enrolled in the Surgical stratum but subsequently received Phase I defined treatment with PK data available will also be included in the analysis population. 5 patients from surgical stratum received dose level 1 treatment. (NCT03387020)
Timeframe: Pre-dose, 1, 2, 4, 8, 24, 32 (day 1 course 1 only), & 48 (day 1 course 1 only) hours after dose on days 1 and 17 of course 1.

Interventionhours (Median)
Dose Level 1, Day 1 of Course 111.7
Dose Level 1, Day 17 of Course 18.9
Dose Level 2, Day 1 of Course 18.5
Dose Level 2, Day 17 of Course 17.5

[back to top]

Maximum Tolerated Dose of Ribociclib and Everolimus

Defined as the highest dose level at which six patients have been treated with at most one patient experiencing a dose limiting toxicity (DLT) and the next higher dose level has been determined to be too toxic. Will be estimated using a Rolling-6 phase I design. Only patients who are evaluable for DLT will be consider for estimating of Maximum Tolerated Dose. Patient who were enrolled under Surgical cohort but received sufficient study defined treatment would also be evaluable for DLT. (NCT03387020)
Timeframe: 4 weeks

Interventionmg/m^2/day (Number)
RibociclibEverolumus
DLT Evaluable Set1201.2

[back to top]

Average Tumor and Plasma Concentrations of Ribociclib for the Surgical Study

Average tumor Ribociclib concentrations will be compared to plasma concentrations. The mean tumor-to-plasma ratio of Ribociclib at the time of surgery will be described. (NCT03387020)
Timeframe: Surgical study: Prior to starting ribociclib (day -7 or -10), prior to ribociclib dose on days -5 & -2, day 0, and at the time of surgery

Interventionratio (Median)
Surgical14.6

[back to top]

Everolimus Half Life as Obtained From the Phase 1 Study

Everolimus plasma concentration-time data obtained from patients received Phase I defined treatment will be modeled using compartmental approaches to estimate the half-life (hrs) for each dose level on day 17 of course 1 and day 1 of course 2. Patients who were enrolled in the Surgical stratum but subsequently received Phase I defined treatment with PK data available will also be included in the analysis population. 5 patients from surgical stratum received dose level 1 treatment. (NCT03387020)
Timeframe: Pre-dose, 0.5 (course 2 only), 1, 1.5 (course 2 only), 2, 4, 6 (course 2 only), 8, & 24 hours after dose on day 17 of course 1, and day 1 of course 2

Interventionhours (Median)
Dose Level 1, Day 17 of Course 113.5
Dose Level 1, Day 1 of Course 214.5
Dose Level 2, Day 17 of Course 111.2
Dose Level 2, Day 1 of Course 211.5

[back to top]

Objective Responses (Complete Response + Partial Response)

Number of patients with sustained objective responses will be reported by stratum and dose. Objective response refers to at least 50% reduction in 2 dimensional measurements of the tumor. In order to count towards this objective, any response needs to be sustained for at least 8 weeks. (NCT03387020)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Phase I, Dose Level 10
Phase I, Dose Level 20
Surgical0

[back to top]

Percent Change in ki67 Between Archival and Post-treatment Tissue

For the surgical study, Ki67 will be evaluated in paired pre-treatment tumor (diagnostic) and post-treatment tumor (recurrent) tissue in consenting patients to assess percent change. The percent of Ki67 in tumor tissues will be measured and the result is presented as the percent of Ki67 in post-treatment tumor minus the percent of Ki67 measured in pre-treatment tumor. (NCT03387020)
Timeframe: Up to 2 years

InterventionChange of Percentage of Ki67 in Tumor (Median)
Surgical-20

[back to top]

Everolimus AUC (Area Under the Plasma Concentration Versus Time Curve) as Obtained From the Phase I Study

Everolimus plasma concentration-time data obtained from patients who received Phase I defined treatment will be modeled using compartmental approaches to estimate the AUC (hr*nM) for each dose level on day 17 of course 1 and day 1 of course 2. Patients who were enrolled in the Surgical stratum but subsequently received Phase I defined treatment with PK data available will also be included in the analysis population. 5 patients from surgical stratum received dose level 1 treatment. (NCT03387020)
Timeframe: Pre-dose, 0.5 (course 2 only), 1, 1.5 (course 2 only), 2, 4, 6 (course 2 only), 8, 24 hours after dose on day 17 of course 1, and day 1 of course 2.

Interventionhr*nM (Median)
Dose Level 1, Day 17 of Course 1201
Dose Level 1, Day 1 of Course 280.1
Dose Level 2, Day 17 of Course 1147.5
Dose Level 2, Day 1 of Course 259

[back to top]

Ribociclib AUC (Area Under the Plasma Concentration Versus Time Curve) as Obtained From the Patients Who Received Phase I Defined Treatment

Ribociclib plasma concentration-time data obtained from patients who received Phase I defined treatment will be modeled using compartmental approaches to estimate the AUC (hr*μM) for each dose level on days 1 and 17 of course 1. Patients who were enrolled in the Surgical stratum but subsequently received Phase I defined treatment with PK data available will also be included in the analysis population. 5 patients from surgical stratum received dose level 1 treatment. (NCT03387020)
Timeframe: Pre-dose, 1, 2, 4, 8, 24, 32 (day 1 course 1 only), & 48 (day 1 course 1 only) hours after dose on days 1 and 17 of course 1.

Interventionhr*uM (Median)
Dose Level 1, Day 1 of Course 13.96
Dose Level 1, Day 17 of Course 110.3
Dose Level 2, Day 1 of Course 112.0
Dose Level 2, Day 17 of Course 110.15

[back to top]

Incidence of Cytomegalovirus Infection or Disease

Incidence of CMV infection/disease in three study groups (SRL, EVR anda MPS). (NCT03468478)
Timeframe: 12 months follow up

InterventionParticipants (Count of Participants)
Sirolimus +Tacrolimus9
Everolimus +Tacrolimus7
Mycophenolate +Tacrolimus39

[back to top]

Percentage of Participants With Best Overall Response (BOR) Status of Angiomyolipoma (AML) Response During Maximum Treatment Duration of 48 Weeks

"BOR status is the best status recorded from start of treatment until disease progression. AML response status: reduction in AML volume of at least 50% relative to screening AML. In addition, AML response have to satisfy: no new AML ≥ 1 cm in longest diameter are identified, neither kidney increases in volume by more than 20% from nadir (where nadir is the lowest kidney volume at the screening), the participant does not have any angiomyolipoma-related bleeding of grade equal or over 2 (as defined by NCI CTCAE, version 4.03).~Up to five of the largest measurable lesions (≥ 1 cm in longest diameter) in each kidney were measured at screening via Magnetic Resonance Imaging/Computed tomography (MRI/CT) and were defined as screening AML. The sum of the volumes of these individual lesions was defined as AML volume and it was measured at each assessment during the trial. Increases in the AML volume were evidence of worsening AML, decreases were evidence of clinical response." (NCT03525834)
Timeframe: Baseline, 48 weeks

InterventionParticipants (Count of Participants)
Everolimus28

[back to top]

Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

Percentage of participants with AEs and SAEs including significant changes from baseline in vital signs, electrocardiograms and laboratory parameters (laboratory assessments included hematology, biochemistry, coagulation and urinalysis) qualifying and reported as AEs. The percentage of participants in each category is reported in the table. (NCT03525834)
Timeframe: From the start of treatment to 30 days after end of treatment, assessed up to maximum duration of 52 weeks

InterventionParticipants (Count of Participants)
AEsTreatment-related AEsSAEsTreatment-related SAEs
Everolimus403962

[back to top]

Percentage of Participants With Best Overall Response Status of Angiomyolipoma (AML) Progression During Maximum Treatment Duration of 48 Weeks

"AML progression status is defined as one or more of the following: an increase from nadir of 25% or more in AML volume to a value greater than screening AML (where nadir is the lowest AML volume obtained for the participant previously in the trial), the appearance of a new AML ≥ 1.0 cm in longest diameter, an increase from nadir of 20% or more in the volume of either kidney to a value greater than screening (where nadir is the lowest kidney volume obtained for the participant previously in the trial), angiomyolipoma-related bleeding grade ≥2 (as defined by NCI CTCAE, version 4.03).~Up to five of the largest measurable lesions (≥ 1 cm in longest diameter) in each kidney were measured at screening via Magnetic Resonance Imaging/Computed tomography (MRI/CT) and were defined as screening AML. The sum of the volumes of these individual lesions was defined as AML volume and it was measured at each assessment during the trial. Increases in the AML volume were evidence of worsening AML." (NCT03525834)
Timeframe: Baseline, 48 weeks

InterventionParticipants (Count of Participants)
Everolimus1

[back to top]

Percentage of Participants With Severe Renal Impairment

"Renal impairment is defined as calculated Creatinine Clearance (CrCl) < 30 mL/min. CrCl was measured at baseline and at four other time points; only the baseline and the worst post-baseline value for every participant were counted (lowest value for CrCl).~Creatinine clearance was estimated using the Cockcroft-Gault formula:~creatinine clearance (mL/minute) = urine creatinine concentration (mL/dL) x volume of urine (mL/24 hour) / plasma creatinine concentration (mg/dL) x 1440 minute/24 hour" (NCT03525834)
Timeframe: Baseline, 48 weeks

InterventionParticipants (Count of Participants)
BaselinePost-baseline, 48 weeks
Everolimus01

[back to top]

Percentage of Participants With NCI CTCA Grade 3/4 Serum Creatinine

NCI CTCAE grade 3/4 serum creatinine is defined as > 3.0 x upper limits of normal (ULN). Serum creatinine was measured at baseline and at four other time points; only the worst post-baseline value for every participant was counted (highest value for serum creatinine). (NCT03525834)
Timeframe: Baseline, 48 weeks

InterventionParticipants (Count of Participants)
BaselinePost-baseline, 48 weeks
Everolimus00

[back to top]

Duration of Disease Control (DC)

"Duration of disease control (DC), defined as the time from randomisation until the earliest of disease progression (according to Response Evaluation Criteria In Solid Tumors (RECIST, version 1.1) in combination with modified MD Anderson Criteria (for bone lesion assessment) or death from any cause, among patients with DC. Duration of DC was assessed by independent reviewers.~The duration of DC was calculated as followed:~For patients with disease progression or death:~Duration of DC [days] = date of outcome - date of randomisation + 1~For patients without disease progression or death:~Duration of DC (censored) [days] = date of outcome - date of randomisation + 1" (NCT03659136)
Timeframe: From randomisation until the earliest of progressive disease or death from any cause, up to 892 days.

InterventionMonths (Median)
1000 mg Xentuzumab + 10 mg Everolimus + 25 mg Exemestane14.6
Placebo + 10 mg Everolimus + 25 mg Exemestane18.4

[back to top]

Overall Survival (OS)

"Overall survival (OS) defined as the time from randomisation until death from any cause. For patients with 'event' as an outcome for OS:~OS[days] = date of outcome - date of randomisation + 1.~For patients with 'censored' as an outcome for OS:~OS (censored)[days] = date of outcome - date of randomisation + 1." (NCT03659136)
Timeframe: From randomisation until death from any cause, up to 995 days.

InterventionMonths (Median)
1000 mg Xentuzumab + 10 mg Everolimus + 25 mg ExemestaneNA
Placebo + 10 mg Everolimus + 25 mg ExemestaneNA

[back to top]

Progression Free Survival (PFS)

Progression-free survival (PFS) defined as the time from randomisation until progressive disease (PD) according to Response Evaluation Criteria In Solid Tumors (RECIST, version 1.1) in combination with modified MD Anderson Criteria (for bone lesion assessment), based on blinded independent assessment or death from any cause, whichever occurred earlier. As per RECIST, PD is defined as at least a 20% increase in the sum of diameters of target lesions, unequivocal progression of non-target lesions or the appearance of new lesions. (NCT03659136)
Timeframe: From randomisation until the earliest of disease progression, death or the time point of primary PFS analysis, up to 892 days.

InterventionMonths (Median)
1000 mg Xentuzumab + 10 mg Everolimus + 25 mg Exemestane12.7
Placebo + 10 mg Everolimus + 25 mg Exemestane11.0

[back to top]

Time to Pain Progression or Intensification of Pain Palliation

"Time to pain progression or intensification of pain palliation was defined as the time from randomisation until the earliest of:~2 point increase from baseline in the Brief Pain Inventory - Short Form (BPI-SF), Item 3 (worst pain), without a decrease (of ≥1 point) from baseline analgesics use (via the 8-point Analgesic Quantification Algorithm [AQA]), or~2 point increase from baseline in the AQA, or Death." (NCT03659136)
Timeframe: From randomisation until the earliest of pain progression, intensification of pain palliation, death or the time point of progression free survival analysis, up to 843 days.

InterventionMonths (Median)
1000 mg Xentuzumab + 10 mg Everolimus + 25 mg Exemestane5.6
Placebo + 10 mg Everolimus + 25 mg Exemestane3.0

[back to top]

Number of Participants With Objective Response (OR)

Number of participants with objective response (OR) by independent assessment. OR is defined as a best overall response of complete response (CR) or partial response (PR). Best overall response is defined according to RECIST v1.1 in combination with modified MD Anderson Criteria (for bone lesion assessment) and will consider all tumor assessments from randomisation until the earliest of PD, start of subsequent anti-cancer therapy, loss to follow-up, withdrawal of consent or death. To be aligned with the primary endpoint derivation, tumor assessments after two or more consecutively misses assessments were not considered. (NCT03659136)
Timeframe: From randomisation until end of treatment, up to 892 days.

InterventionParticipants (Count of Participants)
1000 mg Xentuzumab + 10 mg Everolimus + 25 mg Exemestane6
Placebo + 10 mg Everolimus + 25 mg Exemestane5

[back to top]

Number of Patients With Disease Control (DC)

Disease control (DC) was defined as a best overall response (BOR) of either complete response (CR), partial response (PR), stable disease (SD) or Non-CR/No-PD. SD and Non-CR/Non-PR must have been observed up until at least week 24 tumor assessment. BOR was defined according to RECIST v1.1 in combination with modified MD Anderson Criteria (for bone lesion assessment) based on all evaluable tumor assessments from randomisation until the earliest of PD, start of subsequent anti-cancer therapy, loss to follow-up, withdrawal of consent or death. To be aligned with the primary endpoint derivation, tumor assessments after two or more consecutively misses assessments were not considered. DC was assessed by independent reviewers. (NCT03659136)
Timeframe: From randomisation until the earliest of progressive disease or death from any cause, up to 892 days.

InterventionParticipants (Count of Participants)
1000 mg Xentuzumab + 10 mg Everolimus + 25 mg Exemestane29
Placebo + 10 mg Everolimus + 25 mg Exemestane25

[back to top]

Severity of Mouth Pain Scores by Ethnicity

Average Area Under the Curve per assessment (aAUCpa) of mouth pain scores as measured by NAMPS will be computed for each patient to summarize the sequence of numerical analogue mouth pain scores over the eight week study period. Scores are reported on a 0-100 scale, where 100 = better outcome QOL (i.e. less pain). The aAUCpa is the average of each AUC between each sequential assessment from treatment-initiation to the end of the treatment at 8 weeks post-treatment-initiation and will be compared between the two treatment arms. (NCT03839940)
Timeframe: Up to 8 weeks

,
Interventionscores on scale * days (Mean)
Not Hispanic or LatinoOther
Group I (Dexamethasone)12.10.0
Group II (Placebo)4.90.0

[back to top]

Severity of Mouth Pain Scores by Sex

Average Area Under the Curve per assessment (aAUCpa) of mouth pain scores as measured by NAMPS will be computed for each patient to summarize the sequence of numerical analogue mouth pain scores over the eight week study period. Scores are reported on a 0-100 scale, where 100 = better outcome QOL (i.e. less pain). The aAUCpa is the average of each AUC between each sequential assessment from treatment-initiation to the end of the treatment at 8 weeks post-treatment-initiation and will be compared between the two treatment arms. (NCT03839940)
Timeframe: Up to 8 weeks

,
Interventionscores on scale * days (Mean)
FemaleMale
Group I (Dexamethasone)11.411.0
Group II (Placebo)2.718.8

[back to top] [back to top] [back to top] [back to top]

Severity of Mouth Pain Scores

Average Area Under the Curve per assessment (aAUCpa) of mouth pain scores as measured by NAMPS will be computed for each patient to summarize the sequence of numerical analogue mouth pain scores over the eight week study period. Scores are reported on a 0-100 scale, where 100 = better outcome QOL (i.e. less pain). The aAUCpa is the average of each AUC between each sequential assessment from treatment-initiation to the end of the treatment at 8 weeks post-treatment-initiation and will be compared between the two treatment arms. (NCT03839940)
Timeframe: Up to 8 weeks

Interventionscores on scale * days (Mean)
Group I (Dexamethasone)11.3
Group II (Placebo)4.6

[back to top]

Severity of Mouth Pain Scores by Race

Average Area Under the Curve per assessment (aAUCpa) of mouth pain scores as measured by NAMPS will be computed for each patient to summarize the sequence of numerical analogue mouth pain scores over the eight week study period. Scores are reported on a 0-100 scale, where 100 = better outcome QOL (i.e. less pain). The aAUCpa is the average of each AUC between each sequential assessment from treatment-initiation to the end of the treatment at 8 weeks post-treatment-initiation and will be compared between the two treatment arms. (NCT03839940)
Timeframe: Up to 8 weeks

,
Interventionscores on scale * days (Mean)
Black or African AmericanWhite
Group I (Dexamethasone)9.312.0
Group II (Placebo)7.94.4

[back to top]

Incidence of Acute Rejection

Biopsy-proven according to Banff 2017 criteria (NCT04177095)
Timeframe: Through study completion, 1 year

InterventionParticipants (Count of Participants)
Belatacept Treated Patients0

[back to top]

Incidence of de Novo Donor Specific Antibodies

Screened for using Luminex platform (NCT04177095)
Timeframe: At 12 months

InterventionParticipants (Count of Participants)
Belatacept Treated Patients0

[back to top]

Rate of New-onset Proteinuria

Defined as g/g creatinine, measured on random urine sample (NCT04177095)
Timeframe: At 12 months

InterventionParticipants (Count of Participants)
Belatacept Treated Patients1

[back to top]

Survival

Overall and death-censored graft survival (NCT04177095)
Timeframe: At 12 months

InterventionParticipants (Count of Participants)
Belatacept Treated Patients12

[back to top]

Incidence of Acute Rejection

Biopsy-proven according to Banff 2017 criteria (NCT04177095)
Timeframe: Enrollment through 12 months

InterventionParticipants (Count of Participants)
Belatacept Treated Patients0

[back to top]

Increase in eGFR

Calculated using CKD-EPI formula (NCT04177095)
Timeframe: From baseline to 12 months

InterventionParticipants (Count of Participants)
Belatacept Treated Patients6

[back to top]