Page last updated: 2024-12-08

lenalidomide

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 CID216326
CHEMBL ID848
CHEBI ID63791
SCHEMBL ID1980410
SCHEMBL ID32978
MeSH IDM0479831

Synonyms (116)

Synonym
AC-914
HY-A0003
AB01273975-02
3-(4-amino-1-oxo-2,3-dihydro-1h-isoindol-2-yl)piperidine-2,6-dione
revlimid
cdc-501
cc-5013 ,
revimid
cdc-5013
imid-5013
enmd-0997
imid3
lenalidomide ,
c13h13n3o3
2,6-piperidinedione, 3-(4-amino-1,3-dihydro-1-oxo-2h- isoindol-2-yl)-
imid3 cpd
lenalidomide (usan/inn)
191732-72-6
D04687
revlimid (tn)
3-(4-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione
DB00480
1-oxo-2-(2,6-dioxopiperidin-3-yl)-4-aminoisoindoline
cdc 501
2,6-piperidinedione, 3-(4-amino-1,3-dihydro-1-oxo-2h-isoindol-2-yl)-
3-(4-amino-1-oxo-1,3-dihydro-2h-isoindol-2-yl)piperidine-2,6-dione
lenalidomide [usan]
cc 5013
NCGC00167491-01
chebi:63791 ,
CHEMBL848
syp-1512
AKOS005146276
EC-000.2340
FT-0659651
nsc-747972
2, 3-(4-amino-1,3-dihydro-1-oxo-2h-isoindol-2-yl)-
nsc747972
revlimid (tn) (celgene)
3-(7-amino-3-oxo-1h-isoindol-2-yl)-piperidine-2,6-dione
STK639603
3-(7-amino-3-oxo-1h-isoindol-2-yl)piperidine-2,6-dione
NCGC00167491-03
NCGC00167491-02
tox21_112492
dtxsid8046664 ,
dtxcid6026664
cas-191732-72-6
lenalidomide (cc-5013)
revlimid (lenalidomide)
AKOS005174869
f0p408n6v4 ,
nsc 747972
unii-f0p408n6v4
lenalidomide [usan:inn:ban]
hsdb 8220
enmd 0997
BCP9000847
FT-0670758
BCPP000186
MLS003899194
smr002529986
(3s)-3-(4-amino-1-oxo-1,3-dihydro-2h-isoindol-2-yl)piperidine-2,6-dione
AM20050439
CS-0125
S1029
gtpl7331
4-amino-2-(6-hydroxy-2-oxo-2,3,4,5-tetrahydropyridin-3-yl)-2,3-dihydro-1h-isoindol-1-one
lenalidomide [jan]
lenalidomide [mi]
lenalidomide [who-dd]
lenalidomide [inn]
lenalidomide [vandf]
lenalidomide [ema epar]
lenalidomide [mart.]
lenalidomide [orange book]
SCHEMBL1980410
GOTYRUGSSMKFNF-UHFFFAOYSA-N
3-(4-amino-1-oxo-1,3-dihydro-2h-isoindol-2-yl)-2,6-dioxopiperidine
3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione
SCHEMBL32978
tox21_112492_1
NCGC00167491-04
revlimid, lenalidomide, cc-5013
KS-1207
AB01273975-01
Q-101410
3-(4-amino-1-oxo-1,3-dihydro-2h-isoindol-2-yl)-2,6-piperidinedione
AB01273975_03
mfcd07772307
SR-01000883999-1
sr-01000883999
HMS3654G07
3-(4-amino-1,3-dihydro-1-oxo-2h-isoindol-2-yl)-2,6-piperidinedione
443912-14-9
SW218084-2
Q425681
SY047646
3-(4-amino-1-oxo-2-isoindolinyl)piperidine-2,6-dione
Z1515385074
FT-0670759
bdbm65454
BCP01390
HMS3674C05
CCG-264781
lenadoamide
SB66166
BL164614
EN300-118706
BP-27972
(3rs)-3-(4-amino-1-oxo-1,3-dihydro-2h-isoindol-2-yl)piperidine-2,6-dione
lenalidomida
nsc703813
l04ax04
lenalidomide (mart.)
lenalidomidum

Research Excerpts

Overview

Lenalidomide is an immunomodulatory drug used to treat multiple myeloma that requires renal dosing adjustment based on Cockcroft-Gault (CG) It is an important medication used in induction therapy for MM and is also used for maintenance therapy for standard risk patients.

ExcerptReferenceRelevance
"Lenalidomide is a type of immunomodulatory agent with anti-tumor activity by mainly expressed in the anti-angiogenesis. "( Design, synthesis and biological evaluation of Lenalidomide derivatives as tumor angiogenesis inhibitor.
Hu, S; Li, Z; Yan, H; Yuan, L, 2017
)
2.15
"Lenalidomide is an immunomodulatory agent with multiple mechanisms of action, and treatment with lenalidomide is associated with adverse events such as thrombosis and abdominal pain; nonetheless, other rarer adverse events do exist, with few knowledge from physicians and pharmacists. "( Lenalidomide-induced arthritis: A case report and review of literature and pharmacovigilance databases.
Despas, F; Gauthier, M; Icard, C; Mocquot, P; Nogaro, JC, 2022
)
3.61
"Lenalidomide is an immunomodulatory drug."( Lenalidomide attenuates post-inflammation pulmonary fibrosis through blocking NF-κB signaling pathway.
Deng, R; Gao, S; Huang, H; Jiang, Q; Li, S; Li, X; Liang, Q; Luan, J; Ruan, H; Yang, C; Zhang, F; Zhang, R; Zhou, H, 2022
)
2.89
"Lenalidomide is an orally-administered immunomodulatory small molecule with activity in AML and a favorable safety profile in older adults with active leukemia."( Phase Ib trial of lenalidomide as post-remission therapy for older adults with acute myeloid leukemia: Safety and longitudinal assessment of geriatric functional domains.
Brenizer, T; Deal, AM; Foster, MC; Ivanova, A; Jamieson, K; Matson, M; Muss, H; Nyrop, KA; Pulley, W; Van Deventer, HW; Vohra, SN; Woods, JD; Zeidner, JF; Zhang, J, 2022
)
1.78
"Lenalidomide is an important medication used in induction therapy for MM and is also used for maintenance therapy for standard risk patients."( Three Cases of Lenalidomide Therapy for Multiple Myeloma and Subsequent Development of Secondary B-ALL.
Asawa, P; Chahine, Z; Lister, J; Sadashiv, S; Samhouri, Y; Vusqa, UT, 2022
)
1.8
"Lenalidomide is an immunomodulatory drug used to treat multiple myeloma that requires renal dosing adjustment based on Cockcroft-Gault (CG). "( Estimation of Kidney Function in Patients With Multiple Myeloma: Implications for Lenalidomide Dosing.
Gonsalves, WI; Lam, S; M Saunders, I; Momper, JD; Salama, E; Tzachanis, D, 2023
)
2.58
"Lenalidomide is a synthetic analog of thalidomide formed by the removal of one keto group (plus the addition of an amino group); it has anti-tumor activities beneficial for the treatment of hematologic malignancies. "( Low cerebrospinal fluid-to-plasma ratios of orally administered lenalidomide mediated by its low cell membrane permeability in patients with hematologic malignancies.
Ando, K; Kamiya, Y; Machida, S; Murayama, N; Ogiya, D; Saito, R; Shiraiwa, S; Suzuki, R; Tazume, K; Yamazaki, H, 2022
)
2.4
"Lenalidomide is an immunomodulatory drug widely used in the treatment of multiple myeloma. "( [A novel cause of acute kidney injury in multiple myeloma: Lenalidomide-induced acute interstitial nephritis].
Alchahin, G; Boncila, SD; Bouaka, C; Fofana, AS; Fongoro, S; Haussaire, D; Samaké, M; Sy, S; Torrents, J; Valensi, RC; Yattara, H, 2022
)
2.41
"Lenalidomide (LEN) is an immunomodulatory drug used for treating several hematological malignancies such as multiple myeloma, adult T-cell lymphoma/leukemia, and follicular lymphoma."( Early administration of lenalidomide after allogeneic hematopoietic stem cell transplantation suppresses graft-versus-host disease by inhibiting T-cell migration to the gastrointestinal tract.
Ito, T; Nasa, Y; Nomura, S; Satake, A; Tsubokura, Y; Tsuji, R; Yoshimura, H, 2022
)
1.75
"Lenalidomide (Len) is a structural analog of thalidomide, which belongs to the second generation of immunomodulators and has the functions of tumor killing, immune regulation, anti-angiogenesis and regulation of the myeloma microenvironment."( Lenalidomide attenuates IMQ-induced inflammation in a mouse model of psoriasis.
Hou, JJ; Jia, HY; Qiu, HY; Wu, Y; Zhang, MD; Zhou, ML, 2022
)
2.89
"Lenalidomide is an immunomodulatory drug and is very effective in the management of a number of malignancies, including multiple myeloma. "( Embryo-fetal exposure and developmental outcome of lenalidomide following oral administration to pregnant cynomolgus monkeys.
Fuchs, A; Hui, JY; Kumar, G, 2022
)
2.42
"Lenalidomide is a ligand of the E3 ligase substrate adapter cereblon (CRBN) that achieves its clinical effects in part by the promotion of substrate recruitment and degradation. "( Molecular and Structural Characterization of Lenalidomide-Mediated Sequestration of eIF3i.
Lin, Z; Shen, D; Woo, CM; Yang, B, 2022
)
2.42
"Lenalidomide (LND) is an oral anticancer drug used to treat various malignant hematologic diseases, including multiple myeloma. "( Evaluation of Lung Toxicity With Lenalidomide Using the Pharmacovigilance Database.
Eren, T; Murata, S; Sato, J; Shimizu, T; Uchida, M, 2022
)
2.45
"Lenalidomide (LND) is an analogue of thalidomide that is second generation immunomodulatory drugs (IMiDs). "( Development and validation of a novel chiral chromatographic method for separation of lenalidomide enantiomers in human plasma.
Dhamija, P; Handu, S; Kumar, V; Nath, UK; Prakash, A; Sahu, PL; Vardhan, G, 2023
)
2.58
"Lenalidomide is an effective maintenance agent for patients with myeloma, prolonging first remission and, in transplant eligible patients, improving overall survival (OS) compared to observation. "( The addition of vorinostat to lenalidomide maintenance for patients with newly diagnosed multiple myeloma of all ages: results from 'Myeloma XI', a multicentre, open-label, randomised, phase III trial.
Cairns, DA; Cook, G; Davies, FE; Drayson, MT; Gregory, W; Hockaday, A; Jackson, GH; Jenner, MW; Jones, JR; Kaiser, MF; Karunanithi, K; Kishore, B; Lindsay, J; Menzies, T; Morgan, GJ; Olivier, C; Owen, RG; Pawlyn, C, 2023
)
2.64
"Lenalidomide is an effective component of induction and maintenance therapy for multiple myeloma, though with a risk of secondary malignancies, including acute lymphoblastic leukemia (ALL). "( Lenalidomide-associated B-cell ALL: clinical and pathologic correlates and sensitivity to lenalidomide withdrawal.
Bhatnagar, B; Dilip, D; Geyer, MB; Glass, JL; Hassoun, H; Klein, V; Landau, H; Lozanski, G; Mims, AS; Park, JH; Roshal, M; Shaffer, BC; Xiao, W; Zhang, Y, 2023
)
3.8
"Lenalidomide (LND) is an oral antineoplastic agent used in the treatment of various malignant hematologic diseases, including multiple myeloma. "( Evaluation of the Time to Onset and Outcome of Lenalidomide-induced Thrombosis and Embolism Using Spontaneous Reporting Database.
Kawahara, Y; Sato, J; Shimizu, T; Uchida, M; Yamamoto, N,
)
1.83
"Lenalidomide is a weak substrate of P-glycoprotein (P-gp), though it is unclear whether P-gp is solely responsible for lenalidomide transport."( Development of a physiologically based pharmacokinetic model for intravenous lenalidomide in mice.
Foster, DJR; Hughes, JH; Phelps, MA; Reuter, SE; Rozewski, DM; Upton, RN, 2019
)
1.46
"Lenalidomide is a promising drug for skin lupus treatment, particularly regarding the apparent lower frequency of nerve side effects."( Thalidomide and Lenalidomide for Refractory Systemic/Cutaneous Lupus Erythematosus Treatment: A Narrative Review of Literature for Clinical Practice.
Aikawa, NE; Bonfa, E; Heise, CO; Pasoto, SG; Romiti, R; Silva, CA; Yuki, EFN, 2021
)
1.69
"Lenalidomide is an immunomodulatory drug that is administered commonly in patients with relapsed or refractory multiple myeloma (RRMM). "( Elevated eosinophil level predicted long time to next treatment in relapsed or refractory myeloma patients treated with lenalidomide.
Gunji, T; Katori, M; Masuoka, H; Nishiwaki, K; Suzuki, K; Yano, S, 2020
)
2.21
"Lenalidomide is an immunomodulatory drug approved in the United States for use with rituximab in patients with relapsed/refractory follicular lymphoma. "( Lenalidomide in follicular lymphoma.
Flowers, CR; Fowler, NH; Leonard, JP, 2020
)
3.44
"Lenalidomide is an integral, yet evolving, part of current treatment pathways for both transplant-eligible and transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM). "( The clinical management of lenalidomide-based therapy in patients with newly diagnosed multiple myeloma.
Dechow, T; Hackanson, B; Knop, S; Merz, M; Scheytt, M; Schmidt, C, 2020
)
2.3
"Lenalidomide is a cereblon modulator known for its antitumor, anti-inflammatory, and immunomodulatory properties in clinical applications. "( Design and synthesis of new lenalidomide analogs via Suzuki cross-coupling reaction.
Jin, J; Li, J; Lu, W; Wang, HL; Wang, YJ; Xiao, D; Zhou, YB, 2020
)
2.29
"Lenalidomide is an immunomodulatory agent with antitumor activity in non-Hodgkin lymphoma, with an acceptable toxicity profile and manageable side effects."( Elderly Non-GCB Diffuse Large B-Cell Lymphoma Patient Responding to Lenalidomide after Epicardial Relapse: A Case Report.
Argnani, L; Broccoli, A; Casadei, B; Cavo, M; Gentilini, M; Zinzani, PL, 2020
)
1.51
"Lenalidomide is an immunomodulatory drug approved for maintenance treatment in newly diagnosed multiple myeloma, and it has been shown to improve progression-free survival (PFS) and, in several studies, overall survival. "( Prolonged lenalidomide maintenance therapy improves the depth of response in multiple myeloma.
Alonso, R; Bahri, N; Cedena, MT; Collado-Yurrita, L; García, C; Lahuerta, JJ; López-Jiménez, J; Martin, T; Martínez-López, J; Moraleda, JM; Ríos-Tamayo, R; Sánchez, R; Shah, N; Valeri, A; Wolf, J; Wong, S, 2020
)
2.4
"Lenalidomide is a backbone agent in the treatment of multiple myeloma, but dose adjustment is required for those with renal impairment (RI). "( An open-label, pharmacokinetic study of lenalidomide and dexamethasone therapy in previously untreated multiple myeloma (MM) patients with various degrees of renal impairment - validation of official dosing guidelines.
Cao, Y; Chen, CI; Chen, E; Chen, H; Kakar, S; Kukreti, V; Lau, A; Le, LW; Levina, O; Paul, H; Prica, A; Reece, DE; Tiedemann, R; Trudel, S, 2020
)
2.27
"Lenalidomide is an immunomodulatory drug (IMiD) used to treat multiple myeloma (MM) patients. "( Development of Label-Free Impedimetric Immunosensors for IKZF1 and IKZF3 Femtomolar Detection for Monitoring Multiple Myeloma Patients Treated with Lenalidomide.
Abdulkarim, H; Siaj, M; Zourob, M, 2020
)
2.2
"Lenalidomide is an immunomodulatory agent with demonstrated activity in multiple subtypes of lymphoma including cHL, and has also been shown to improve both progression-free and overall survival as maintenance therapy after ASCT in multiple myeloma."( A Pilot Study of Lenalidomide Maintenance Therapy after Autologous Transplantation in Relapsed or Refractory Classical Hodgkin Lymphoma.
Abboud, CN; Bartlett, NL; Cashen, AF; Dipersio, JF; Fehniger, TA; Hurd, DD; Jacoby, MA; Jaglowski, SM; Shea, L; Wagner-Johnston, ND; Wan, F; Watkins, MP, 2020
)
1.62
"Lenalidomide is an immunomodulator that has shown safety and efficacy in multiple myeloma and is also approved for use in several types of lymphoma."( Lenalidomide demonstrates clinical activity in anaplastic lymphoma kinase-positive large B-cell lymphoma.
Abro, B; Bhaskar, S; Fraum, TJ; Mehta-Shah, N, 2020
)
2.72
"Lenalidomide is a centrally active thalidomide analog that has potent anti-inflammatory and antiangiogenic activities. "( The neuroprotective action of lenalidomide on rotenone model of Parkinson's Disease: Neurotrophic and supportive actions in the substantia nigra pars compacta.
Bilge, SS; Cankara, FN; Günaydın, C; Kortholt, A; Özmen, Ö, 2020
)
2.29
"Lenalidomide is an oral immunomodulatory drug which downregulates MYC and its target genes thereby providing support using lenalidomide as additional therapeutic option for MYC+ LBCL."( Treatment of patients with MYC rearrangement positive large B-cell lymphoma with R-CHOP plus lenalidomide: results of a multicenter HOVON phase II trial.
Arens, AIJ; Bakunina, K; Beeker, A; Bilgin, YM; Boersma, RS; Bohmer, LH; Burggraaff, CN; Chamuleau, MED; de Jong, D; de Jongh, E; Diepstra, A; Dierickx, D; Durian, MF; Hijmering, N; Hoekstra, O; Huijbregts, J; Kersten, MJ; Koene, HR; Lugtenburg, PJ; Mandigers, C; Marijt, EAF; Mous, R; Nijland, M; Schouten, HC; Snijders, TJF; Tick, LW; van den Berg, A; van Imhoff, GW; van Rees, B; Vermaat, JSP; Visser, O; Zijlstra, JM, 2020
)
1.5
"Lenalidomide is an effective immunomodulatory derivative drug used in the treatment of multiple myeloma (MM). "( Original Versus Generic Lenalidomide in Patients with Relapsed/Refractory Multiple Myeloma: Comparison of Efficacy and Adverse Events
Bolaman, AZ; Eroğlu Küçükerdiler, H; Ertop, Ş; Sahip, B; Sargın, G; Selim, C; Turgutkaya, A; Yavaşoğlu, İ, 2021
)
2.37
"Lenalidomide is a safe and feasible maintenance strategy in patients with high-risk AML who are not candidates for ASCT, and it has beneficial effects for patients with negative measurable residual disease."( Phase 2 study of lenalidomide maintenance for patients with high-risk acute myeloid leukemia in remission.
Abou Dalle, I; Andreeff, M; Borthakur, G; Cortes, JE; Daver, N; DiNardo, CD; Estrov, Z; Ferrajoli, A; Garcia-Manero, G; Jabbour, E; Jain, N; Jammal, N; Kadia, TM; Kantarjian, HM; Naqvi, K; Pelletier, S; Pemmaraju, N; Pierce, S; Ravandi, F; Wang, SA; Wang, X, 2021
)
2.4
"Lenalidomide is an important component of initial therapy in newly diagnosed multiple myeloma, either as maintenance therapy post-autologous stem cell transplantation (ASCT) or as first-line therapy with dexamethasone for patients' ineligible for ASCT (non-ASCT). "( Retrospective study of treatment patterns and outcomes post-lenalidomide for multiple myeloma in Canada.
Aslam, M; Atenafu, EG; Cherniawsky, H; Gul, E; Jimenez-Zepeda, VH; Kotb, R; LeBlanc, R; Louzada, ML; Masih-Khan, E; McCurdy, A; Reece, DE; Reiman, A; Sebag, M; Song, K; Stakiw, J; Venner, CP; White, D, 2021
)
2.31
"Lenalidomide is a well-tolerated, oral agent that is associated with increased overall and progression free survival when used as maintenance following ASCT. "( Safety of lenalidomide for maintenance treatment of patients with multiple myeloma following autologous stem cell transplantation.
Jackson, GH; Jones, JR; Pawlyn, C, 2021
)
2.47
"Lenalidomide-dexamethasone is a standard treatment approach for relapsed/refractory MM, and according to recent large randomized clinical trials (RCT, the standard arm of POLLUX, ASPIRE, TOURMALINE), the progression-free survival (PFS) is expected to be approximately 18 months."( Beneficial Effect of Lenalidomide-Dexamethason Treatment in Relapsed/Refractory Multiple Myeloma Patients: Results of Real-Life Data From 11 Hungarian Centers.
Alizadeh, H; Altai, E; Borbényi, Z; Csukly, Z; Dávid Tóth, A; Deák, B; Gaál-Weisinger, J; Hardi, A; Illés, Á; Kohl, Z; Kórád, K; Kosztolányi, S; Lengyel, Z; Lovas, S; Masszi, T; Mikala, G; Modok, S; Nagy, Z; Plander, M; Radványi, G; Rajnics, P; Réka Szita, V; Rencsik, A; Rottek, J; Schneider, T; Szaleczky, E; Szendrei, T; Varga, G; Váróczy, L, 2021
)
1.66
"Lenalidomide is an immunomodulatory drug (IMiD) with a unique mode of action (MOA) that may vary across disease-type. "( Lenalidomide in the treatment of chronic lymphocytic leukemia.
Brown, JR; Itchaki, G, 2017
)
3.34
"Lenalidomide is a therapeutically effective drug in chronic lymphocytic leukemia (CLL). "( Increased SHISA3 expression characterizes chronic lymphocytic leukemia patients sensitive to lenalidomide.
Benatti, S; Bulgarelli, J; Cuneo, A; Debbia, G; Fiorcari, S; Forconi, F; Gaidano, G; Laurenti, L; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Palumbo, GA; Rigolin, GM; Rizzotto, L; Rossi, D; Santachiara, R; Vallisa, D, 2018
)
2.14
"Lenalidomide is an immunomodulatory drug that is also currently used in transplant-eligible patients with multiple myeloma. "( Impact of lenalidomide-based induction therapy on the mobilization of CD34
Jantunen, E; Mäntymaa, P; Partanen, A; Pelkonen, J; Putkonen, M; Ropponen, A; Sankelo, M; Siitonen, T; Silvennoinen, R; Valtola, J; Varmavuo, V, 2017
)
2.3
"Lenalidomide is an immunomodulatory agent approved in the US for patients with relapsed/refractory MCL following bortezomib based on results from 3 multicenter phase II studies (2 including relapsed/refractory aggressive NHL and 1 focusing on MCL post-bortezomib)."( Long-term analysis of phase II studies of single-agent lenalidomide in relapsed/refractory mantle cell lymphoma.
Casadebaig Bravo, ML; Drach, J; Fu, T; Goy, A; Habermann, TM; Kalayoglu Besisik, S; Luigi Zinzani, P; Ramchandren, R; Takeshita, K; Tuscano, JM; Witzig, TE; Zhang, L, 2017
)
1.42
"Lenalidomide is an immunomodulatory drug administered orally in the treatment of multiple myeloma. "( Realistic Lenalidomide Dose Adjustment Strategy for Transplant-Ineligible Elderly Patients with Relapsed/Refractory Multiple Myeloma: Japanese Real-World Experience.
Azuma, Y; Fujita, S; Hotta, M; Ishii, K; Ito, T; Nakanishi, T; Nakaya, A; Nomura, S; Satake, A; Tsubokura, Y; Yoshimura, H, 2017
)
2.3
"Lenalidomide is an immunomodulatory agent that has demonstrated clinical benefit for patients with relapsed or refractory mantle cell lymphoma (MCL); however, despite this observed clinical activity, the mechanism of action (MOA) of lenalidomide has not been characterized in this setting. "( Activity of lenalidomide in mantle cell lymphoma can be explained by NK cell-mediated cytotoxicity.
Apollonio, B; Chiu, H; Chopra, R; Couto, S; Flynt, E; Gandhi, AK; Hagner, PR; Ortiz, M; Ramsay, AG; Thakurta, A; Trotter, M; Waldman, MF; Wang, M, 2017
)
2.28
"Lenalidomide is an active agent for the treatment of MALT lymphoma. "( Immunohistochemical expression of cereblon and MUM1 as potential predictive markers of response to lenalidomide in extranodal marginal zone B-cell lymphoma of the mucosa-associated lymphoid tissue (MALT lymphoma).
Dolak, W; Kiesewetter, B; Kornauth, C; Lukas, J; Mayerhoefer, ME; Raderer, M; Simonitsch-Klupp, I, 2018
)
2.14
"Lenalidomide is an efficacious maintenance therapy reducing the relative risk of progression in first-line patients with chronic lymphocytic leukaemia who do not achieve minimal residual disease negative disease state following chemoimmunotherapy approaches. "( Lenalidomide maintenance after first-line therapy for high-risk chronic lymphocytic leukaemia (CLLM1): final results from a randomised, double-blind, phase 3 study.
Al-Sawaf, O; Aldaoud, A; Bahlo, J; Bosch, F; Böttcher, S; Döhner, H; Dörfel, S; Eichhorst, B; Fink, AM; Fischer, K; Ghia, P; Hallek, M; Hebart, H; Jentsch-Ullrich, K; Kater, AP; Kneba, M; Kreuzer, KA; Nösslinger, T; Ritgen, M; Robrecht, S; Stilgenbauer, S; Tausch, E; Wendtner, CM, 2017
)
3.34
"Lenalidomide is an immunomodulatory drug that may modulate anti-tumor immunity."( A phase I study of lenalidomide plus chemotherapy with mitoxantrone, etoposide, and cytarabine for the reinduction of patients with acute myeloid leukemia.
Amrein, PC; Attar, EC; Avigan, D; Ballen, KK; Brunner, AM; DeAngelo, DJ; Ebert, BL; Fathi, AT; Hobbs, GS; Luptakova, K; McMasters, M; Rosenblatt, J; Sperling, AS; Steensma, DP; Stone, RM; Stroopinsky, D; Wadleigh, M; Washington, A; Werner, L, 2018
)
1.53
"Lenalidomide is a thalidomide analog with anti-angiogenic properties. "( Lenalidomide as a novel therapy for gastrointestinal angiodysplasia in von Willebrand disease.
Bull-Henry, K; Kessler, CM; Khatri, NV; Kohli, DR; Patel, B; Solomon, SS, 2018
)
3.37
"Lenalidomide (LEN) is an immunomodulatory drug with significant clinical activity against relapsed and refractory multiple myeloma (r/r MM). "( Lenalidomide plus dexamethasone for patients with relapsed or refractory multiple myeloma: Final results of a non-interventional study and comparison with the pivotal phase 3 clinical trials.
Aldaoud, A; Harde, J; Knauf, W; Losem, C; Mittermueller, J; Neise, M; Niemeier, B; Potthoff, K; Trarbach, T, 2018
)
3.37
"Lenalidomide is an immunomodulatory drug approved by the US Food and Drug Administration in 2006 for the treatment of multiple myeloma. "( Glioblastoma Multiforme in a Patient with Multiple Myeloma: A Case Report and Literature Review.
Bajaj, K; Ibrahim, M; Kapila, A; Moore, CA, 2018
)
1.92
"Lenalidomide is a second-generation oral immunomodulatory drug that has been broadly applied in the treatment of various hematological malignancies."( Durable remission in a patient of mixed phenotype acute leukemia with Philadelphia chromosome-positive treated with nilotinib and lenalidomide: A case report.
Lai, B; Mu, Q; Ouyang, G; Sheng, L; Wang, Y; Xu, K; Zhang, Y; Zhu, H, 2018
)
1.41
"Lenalidomide is an antiangiogenic and immunomodulatory agent with broad antitumor activity."( Ketoconazole plus Lenalidomide in patients with Castration-Resistant Prostate Cancer (CRPC): results of an open-label phase II study.
Barata, PC; Cooney, M; Dreicer, R; Garcia, JA; Mendiratta, P; Tyler, A, 2018
)
1.54
"Lenalidomide is an immunomodulatory imide drug used broadly in the treatment of multiple myeloma and lymphoma. "( Population pharmacokinetics of lenalidomide in patients with B-cell malignancies.
Blum, KA; Blum, W; Byrd, JC; Foster, DJR; Gao, Y; Grever, MR; Hofmeister, CC; Hughes, JH; Marcucci, G; Phelps, MA; Reuter, SE; Upton, RN, 2019
)
2.24
"Lenalidomide (LEN) is an immunomodulator with clinical activity against myeloma cells. "( Lenalidomide and dexamethasone in treatment of patients with relapsed and refractory multiple myeloma - analysis of data from the Czech Myeloma Group Registry of Monoclonal Gammopathies.
Brozova, L; Flochova, E; Gregora, E; Hajek, R; Jelinek, T; Jungova, A; Kralikova, E; Maisnar, V; Minarik, J; Pour, L; Radocha, J; Spicka, I; Stecova, N; Stefanikova, Z, 2019
)
3.4
"Lenalidomide is an effective therapeutic agent for multiple myeloma (MM). "( Impact of chromosomal abnormalities on the efficacy of lenalidomide plus dexamethasone treatment in patients with relapsed/refractory multiple myeloma.
Fujinami, H; Iida, S; Ishida, T; Ito, A; Kinoshita, S; Komatsu, H; Kusumoto, S; Marumo, Y; Masaki, A; Narita, T; Oshima, Y; Ri, M; Sasaki, H; Tachita, T; Totani, H; Yoshida, T, 2019
)
2.2
"Lenalidomide is an immunomodulatory agent that belongs to a family of IMiDs used to treat multiple myeloma. "( Slow lenalidomide desensitization protocol for patients with multiple myeloma: case series from a single center.
Bahlis, NJ; Bailey, K; Duggan, P; Jimenez-Zepeda, VH; McCulloch, S; Neri, P; Tay, J; Yau, P, 2019
)
2.47
"Lenalidomide is a known and approved treatment strategy for relapsed MM."( Lenalidomide in combination with an activin A-neutralizing antibody: preclinical rationale for a novel anti-myeloma strategy.
Cirstea, D; Eda, H; Fulciniti, M; Mahindra, A; Nemani, N; Patel, K; Raje, N; Santo, L; Scullen, T; Vallet, S; Yee, A, 2013
)
2.55
"Lenalidomide is an analog of thalidomide with similar efficacy but improved side-effect profile."( Lenalidomide and thalidomide in the treatment of chronic pain.
Asher, C; Furnish, T, 2013
)
2.55
"Lenalidomide is a thalidomide analog used for the treatment of myelodysplastic syndromes, with pleiotropic activities including induction of apoptosis, inhibition of angiogenesis and broad immunomodulatory effects."( Lenalidomide-induced regenerative macronodules infarction in a cirrhosis patient.
Arrivé, L; Carbonell, N; Cervera, P; Dangouloff-Ros, V, 2013
)
2.55
"Lenalidomide (Revlimid) is an immunomodulatory analog of thalidomide with significant T-cell stimulatory and antiangiogenic properties."( Sargramostim (GM-CSF) and lenalidomide in castration-resistant prostate cancer (CRPC): results from a phase I-II clinical trial.
Dreicer, R; Elson, P; Garcia, JA; Triozzi, P; Tyler, A, 2014
)
1.42
"Lenalidomide (LEN) is a relatively new and very effective therapy for multiple myeloma (MM). "( Evaluating the effects of lenalidomide induction therapy on peripheral stem cells collection in patients undergoing autologous stem cell transplant for multiple myeloma.
Abidi, MH; Abrams, J; Al-Kadhimi, Z; Ayash, L; Bhutani, D; Deol, A; Lum, L; Ratanatharathorn, V; Tageja, N; Uberti, J; Valent, J; Zonder, J, 2013
)
2.13
"Lenalidomide is an effective drug in low-risk myelodysplastic syndromes (MDS) with isolated del(5q), although not all patients respond. "( Response to lenalidomide in myelodysplastic syndromes with del(5q): influence of cytogenetics and mutations.
Alvarez, S; Arenillas, L; Calasanz, MJ; Cervera, J; Cigudosa, JC; Costa, D; Del Rey, M; Diez-Campelo, M; Florensa, L; González-Martínez, T; Hernández, JM; Ibáñez, M; Jerez, A; Larráyoz, MJ; Lumbreras, E; Maciejewski, J; Mallo, M; Marugán, I; Nomdedeu, M; Pedro, C; Solé, F; Such, E, 2013
)
2.21
"Lenalidomide is an active immunomodulatory and antiproliferative agent in multiple myeloma. "( Paradoxical effect of lenalidomide on cytokine/growth factor profiles in multiple myeloma.
Amiot, M; Bonnaud, S; Gomez-Bougie, P; Gratas, C; Le Gouill, S; Maïga, S; Moreau, P; Pellat-Deceunynck, C, 2013
)
2.15
"Lenalidomide is an effective treatment for MM; however, treatment-related adverse events must be considered and appropriate adjustments and/or prophylactic treatment should be initiated where possible."( Lenalidomide treatment for multiple myeloma: systematic review and meta-analysis of randomized controlled trials.
Chi, XH; Lu, XC; Yang, B; Yu, RL, 2013
)
3.28
"Lenalidomide is an immunomodulatory agent that was approved for the treatment of a monoclonal bone marrow disorders, myelodysplastic syndrome del(5q)(MDS del(5q)), in 2005; the drug was subsequently also approved for the treatment of refractory multiple myeloma, a bone marrow malignancy of the B-lymphocyte lineage. "( Can lenalidomide play a role in the management of scleritis?
Abul, N; Al-Awadhi, A; Al-Jafar, HA; Kumar, N, 2013
)
2.39
"Lenalidomide is an immunomodulatory and antiangiogenic agent that has shown clinical benefit in MF patients in several phase II clinical trials."( [Effectiveness and safety of lenalidomide in myelofibrosis patients: a case series from the Spanish compassionate use program].
Asensio, A; Blanes, M; Boqué, C; Castillo, I; Hermosilla, MM; Ojea, MA,
)
1.14
"Lenalidomide is an immunomodulatory agent which has been approved for multiple myeloma. "( Immunomodulation therapy with lenalidomide in follicular, transformed and diffuse large B cell lymphoma: current data on safety and efficacy.
Desai, M; Newberry, KJ; Ou, Z; Romaguera, J; Wang, M; Zhang, L, 2013
)
2.12
"Lenalidomide is an immunomodulatory agent with proven tumoricidal and antiproliferative activity in MCL."( Single-agent lenalidomide in patients with mantle-cell lymphoma who relapsed or progressed after or were refractory to bortezomib: phase II MCL-001 (EMERGE) study.
Cicero, S; Drach, J; Fu, T; Goy, A; Kalayoglu Besisik, S; Ramchandren, R; Sinha, R; Williams, ME; Witzig, TE; Zhang, L, 2013
)
1.48
"Lenalidomide is an immunomodulatory drug approved by the AEMPS and the EMA, in combination with dexamethasone, for the treatment of multiple myeloma in adult patients who have received at least one prior therapy. "( [Review of evidence of thalidomide and lenalidomide in different hematological diseases: chronic lymphocytic leukemia, primary amyloidosis, myelofibrosis and syndrome myelodysplastic].
Jiménez Lozano, I; Juárez Jiménez, JC,
)
1.84
"Lenalidomide is an immunomodulatory agent demonstrating antitumor and antiproliferative effects in MCL."( Long-term follow-up of lenalidomide in relapsed/refractory mantle cell lymphoma: subset analysis of the NHL-003 study.
Czuczman, MS; Haioun, C; Li, J; Polikoff, J; Prandi, K; Reeder, CB; Tilly, H; Vose, JM; Witzig, TE; Zhang, L; Zinzani, PL, 2013
)
1.42
"Lenalidomide is an immunomodulatory drug with co-stimulatory effects on NKT cells in vitro and is an approved treatment for MM, although its mode of action in that context is not well defined."( Natural killer T cell defects in multiple myeloma and the impact of lenalidomide therapy.
Berzins, SP; Chan, AC; Godfrey, DI; Harrison, SJ; Leeansyah, E; Neeson, P; Prince, HM; Quach, H; Ritchie, D; Tainton, K, 2014
)
1.36
"Lenalidomide is an immunomodulatory drug with effects on the immune system that may enhance antibody-dependent cell-mediated cytotoxicity and reverse tumor-induced immune suppression. "( Combined lenalidomide, low-dose dexamethasone, and rituximab achieves durable responses in rituximab-resistant indolent and mantle cell lymphomas.
Ahmadi, T; Aqui, NA; Chong, EA; Gordon, A; Mato, AR; Nasta, SD; Schuster, SJ; Svoboda, J, 2014
)
2.26
"Lenalidomide is an oral immunomodulatory drug used in multiple myeloma and myelodysplastic syndrome and most recently it has shown to be effective in the treatment of various lymphoproliferative disorders such as chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma. "( Lenalidomide and chronic lymphocytic leukemia.
Acebes-Huerta, A; Gonzalez, S; Gonzalez-García, E; González-Rodríguez, AP; Huergo-Zapico, L; Payer, AR; Villa-Alvarez, M, 2013
)
3.28
"Lenalidomide is an IMID immunomodulatory agent clinically active in patients with chronic lymphocytic leukemia (CLL). "( Endothelium-mediated survival of leukemic cells and angiogenesis-related factors are affected by lenalidomide treatment in chronic lymphocytic leukemia.
Bonacorsi, G; Bulgarelli, J; Castelli, I; Cuneo, A; Debbia, G; Fiorcari, S; Forconi, F; Gaidano, G; Laurenti, L; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Palumbo, GA; Rigolin, GM; Rizzotto, L; Rossi, D; Santachiara, R; Vallisa, D, 2014
)
2.06
"Lenalidomide is a drug with clinical efficacy in multiple myeloma and other B cell neoplasms, but its mechanism of action is unknown. "( Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells.
Carr, SA; Ciarlo, C; Comer, E; Ebert, BL; Grauman, P; Hartman, E; Heckl, D; Hurst, SN; Krönke, J; Li, X; McConkey, M; Munshi, N; Narla, A; Schenone, M; Schreiber, SL; Svinkina, T; Udeshi, ND, 2014
)
3.29
"Lenalidomide is an immunomodulating drug structurally similar to thalidomide. "( Pulmonary toxicity associated with the use of lenalidomide: case report of late-onset acute respiratory distress syndrome and literature review.
Beau Salinas, F; D'Halluin, P; Diot, E; Lioger, B; Lissandre, S; Mankikian, J; Marchand Adam, S; Mercier, E,
)
1.83
"Lenalidomide is an immunomodulatory agent that selectively suppresses the del(5q) clone."( Myelodysplastic syndromes with 5q deletion: pathophysiology and role of lenalidomide.
Besa, EC; Gaballa, MR, 2014
)
1.36
"Lenalidomide is an immunomodulatory agent used for the treatment of myelodysplastic syndromes and multiple myeloma. "( The interactions of lenalidomide with human uptake and efflux transporters and UDP-glucuronosyltransferase 1A1: lack of potential for drug-drug interactions.
Kumar, G; Surapaneni, S; Tong, Z; Yerramilli, U, 2014
)
2.17
"Lenalidomide is an immunomodulatory drug (IMiD), which is well established and approved in the treatment of multiple myeloma (MM) and 5q-myelodysplastic syndrome (MDS). "( Lenalidomide.
Kanz, L; Weisel, K, 2014
)
3.29
"Lenalidomide is an immunomodulatory agent with therapeutic activity in chronic lymphocytic leukemia (CLL). "( Lenalidomide and rituximab for the initial treatment of patients with chronic lymphocytic leukemia: a multicenter clinical-translational study from the chronic lymphocytic leukemia research consortium.
Barrientos, JC; Brown, JR; Castro, JE; Greaves, A; James, DF; Johnson, AJ; Kipps, TJ; Neuberg, D; Rai, KR; Rassenti, LZ; Werner, L; Wierda, WG, 2014
)
3.29
"Lenalidomide is an IMiD® immunomodulatory drug, which may warrant evaluation in urothelial carcinoma (UC)."( The preclinical activity of lenalidomide in indolent urothelial carcinoma.
Jian, W; Lerner, SP; Levitt, JM; Sonpavde, G, 2014
)
2.14
"Lenalidomide is an immunomodulatory drug (IMiD) used principally in the treatment of multiple myeloma (MM), myelodysplastic syndromes (MS) and amyloidosis. "( Cutaneous adverse reactions to lenalidomide.
Allegra, A; Calapai, G; Gangemi, S; Imbesi, S; Musolino, C,
)
1.86
"Lenalidomide is an oral immunomodulatory drug that repairs antitumor T-cell function and is showing efficacy in ongoing chronic lymphocytic leukemia (CLL) and lymphoma clinical trials."( How does lenalidomide target the chronic lymphocytic leukemia microenvironment?
Egle, A; Kater, AP; Ramsay, AG; Tonino, SH, 2014
)
1.54
"Lenalidomide is a potent drug with pleiotropic effects in patients with myelodysplastic syndrome (MDS) with deletion of the long arm of chromosome 5 [del(5q)]. "( Genome-wide miRNA profiling in myelodysplastic syndrome with del(5q) treated with lenalidomide.
Belickova, M; Cermak, J; Hrustincova, A; Jonasova, A; Klema, J; Krejcik, Z; Merkerova, MD; Michalova, K; Stara, E; Zemanova, Z, 2015
)
2.09
"Lenalidomide is an immunomodulatory drug with therapeutic activity in chronic lymphocytic leukemia (CLL). "( Lenalidomide induces immunomodulation in chronic lymphocytic leukemia and enhances antitumor immune responses mediated by NK and CD4 T cells.
Acebes-Huerta, A; Fernandez-Guizan, A; Gonzalez, S; Gonzalez-Rodriguez, AP; Huergo-Zapico, L; López-Soto, A; Payer, AR, 2014
)
3.29
"Lenalidomide is an immunomodulatory agent clinically active in chronic lymphocytic leukemia patients. "( Lenalidomide interferes with tumor-promoting properties of nurse-like cells in chronic lymphocytic leukemia.
Audrito, V; Bulgarelli, J; Colaci, E; Deaglio, S; Fiorcari, S; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Narni, F; Potenza, L; Zucchini, P, 2015
)
3.3
"Lenalidomide is an oral non-chemotherapy immunomodulator with direct and indirect effects on non-Hodgkin lymphoma (NHL) cells and with single-agent activity in relapsed/refractory aggressive and indolent B-cell NHL, including mantle cell lymphoma (MCL), diffuse large B-cell lymphoma, and follicular lymphoma. "( A comprehensive review of lenalidomide therapy for B-cell non-Hodgkin lymphoma.
Chiappella, A; Czuczman, MS; Fowler, N; Goy, A; Habermann, TM; Hernandez-Ilizaliturri, FJ; Nowakowski, GS; Vitolo, U; Witzig, TE, 2015
)
2.16
"Lenalidomide is an immunomodulatory drug (IMiD) with activity in lymphoid malignancies occurring primarily through immune modulation (eg, T-cell immune synapse enhancement and NK-cell/T-cell effector augmentation) and antiproliferative effects. "( Lenalidomide in non-Hodgkin lymphoma: biological perspectives and therapeutic opportunities.
Coyle, M; Evens, AM; Kritharis, A; Sharma, J, 2015
)
3.3
"Lenalidomide is a novel thalidomide analogue with immunomodulatory and antiangiogenic effects that has been successfully used for the treatment of low and intermediate-1 risk myelodysplastic syndromes (MDSs) with a del(5q) aberration. "( Aberrant expression of the microRNA cluster in 14q32 is associated with del(5q) myelodysplastic syndrome and lenalidomide treatment.
Beličková, M; Čermák, J; Dostálová Merkerová, M; Hruštincová, A; Jonášová, A; Kléma, J; Krejčík, Z; Michalová, K; Zemanová, Z, 2015
)
2.07
"Lenalidomide (Revlimid(®)) is a second-generation immunomodulatory drug structurally related to thalidomide, with improved efficacy and tolerability, for which the label in the EU was recently expanded to include continuous therapy in patients with previously untreated multiple myeloma not eligible for stem-cell transplantation. "( Lenalidomide: a review of its continuous use in patients with newly diagnosed multiple myeloma not eligible for stem-cell transplantation.
McCormack, PL, 2015
)
3.3
"Lenalidomide is an approved medication for relapsed mantle cell lymphoma in patients who received at least two lines of therapy, including bortezomib."( Lenalidomide for mantle cell lymphoma.
Goy, AH; Skarbnik, AP, 2015
)
2.58
"Lenalidomide is a thalidomide derivative designed for reduced toxicity and increased immunomodulatory properties."( Lenalidomide reduces microglial activation and behavioral deficits in a transgenic model of Parkinson's disease.
Anderson, S; Mante, M; Masliah, E; Rockenstein, E; Valera, E, 2015
)
2.58
"Lenalidomide is an oral immunomodulatory drug used to treat multiple myeloma and some other hematological malignancies. "( Evaluation of pharmacokinetic and pharmacodynamic interactions when lenalidomide is co-administered with warfarin in a randomized clinical trial setting.
Chen, N; Knight, R; Palmisano, M; Weiss, D; Zhou, S, 2015
)
2.1
"Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). "( Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS.
Bullinger, L; Carr, SA; Cathers, BE; Chamberlain, PP; Chopra, R; Ebert, BL; Fink, EC; Gandhi, AK; Griffiths, E; Hollenbach, PW; Hurst, SN; Järås, M; Krönke, J; MacBeth, KJ; Man, HW; Mani, DR; McConkey, M; Schneider, RK; Svinkina, T; Udeshi, ND; Wetzler, M, 2015
)
3.3
"Lenalidomide is an orally active immunomodulatory drug that has direct antineoplastic activity and indirect effects mediated through multiple types of immune cells found in the tumor microenvironment, including B, T, natural killer (NK), and dendritic cells. "( Mechanisms of Action of Lenalidomide in B-Cell Non-Hodgkin Lymphoma.
Fowler, N; Gribben, JG; Morschhauser, F, 2015
)
2.17
"Lenalidomide (LEN) is an immunomodulatory drug with US Food and Drug Administration approval for use in myelodysplastic syndromes (MDS), multiple myeloma (MM), and mantle cell lymphoma (MCL). "( Practical Management of Lenalidomide-Related Rash.
Kurtin, SE; Ridgeway, JA; Tinsley, SM, 2015
)
2.17
"Lenalidomide is an immunomodulatory agent (IMiD) clinically active in chronic lymphocytic leukemia (CLL), both in heavily pre-treated patients and upfront. "( Lenalidomide in chronic lymphocytic leukemia: the present and future in the era of tyrosine kinase inhibitors.
Colaci, E; Fiorcari, S; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Potenza, L, 2016
)
3.32
"Lenalidomide is an oral immunomodulatory drug (IMiD) approved in the United States for patients with MM."( A rare case of nasopharyngeal carcinoma in a patient with multiple myeloma after treatment by lenalidomide.
Qian, W; Wang, B; Xu, G; Yang, M, 2015
)
1.36
"Lenalidomide is an effective treatment option in relapsed refractory non hodgkin's lymphoma."( Lenalidomide in relapsed refractory non-Hodgkin's lymphoma: An Indian perspective.
Abraham, LJ; Babu, KG; Lakshmaiah, KC; Lokanatha, D; Rachan Shetty, KS; Sathyanarayanan, V,
)
3.02
"Lenalidomide is an immune-modulatory piperidine-dione that has demonstrated activity especially in the treatment of hematopoietic malignancies."( Lenalidomide in an in vitro Dendritic Cell Model for Malignant Gliomas.
Kramm, CM; Kühnöl, CD; Staege, MS, 2016
)
2.6
"Lenalidomide is a potential treatment option for refractory bleeding in AVWS secondary to MG."( Lenalidomide as a novel treatment for refractory acquired von Willebrand syndrome associated with monoclonal gammopathy.
Brophy, TM; Browne, PV; Hayden, PJ; Lavin, M; O'Connell, N; O'Donnell, JS; O'Sullivan, JM; Rawley, O; Ryan, K, 2016
)
2.6
"Lenalidomide is an oral immunomodulatory drug with significant activity in indolent B-cell and mantle cell lymphomas. "( Clinical experience with lenalidomide alone or in combination with rituximab in indolent B-cell and mantle cell lymphomas.
Martin, P; Ruan, J; Schuster, SJ; Shah, B, 2016
)
2.18
"Lenalidomide is an oral drug with immune-modulatory and anti-angiogenic activity against selected hematologic malignancies but as yet little is known regarding its effectiveness for solid tumors."( Lenalidomide normalizes tumor vessels in colorectal cancer improving chemotherapy activity.
Aglietta, M; Bertotti, A; Bussolino, F; Gammaitoni, L; Giraudo, E; Giraudo, L; Grignani, G; Leone, F; Leuci, V; Luraghi, P; Maione, F; Mesiano, G; Migliardi, G; Rotolo, R; Sangiolo, D; Sassi, F; Todorovic, M; Trusolino, L, 2016
)
2.6
"Lenalidomide is an immunomodulator and antiangiogenic agent, with limited single-agent efficacy in HCC."( Phase I Study of Lenalidomide and Sorafenib in Patients With Advanced Hepatocellular Carcinoma.
Althouse, SK; Chiorean, EG; Clark, RS; Loehrer, PJ; Shahda, S; Spittler, AJ, 2016
)
1.5
"Lenalidomide is an oral non-chemotherapy immunomodulatory agent with an acceptable toxicity profile and manageable side-effects. "( Lenalidomide for the treatment of B-cell lymphoma.
Bouabdallah, R; Coso, D; Garciaz, S; Schiano de Colella, JM, 2016
)
3.32
"Lenalidomide is an immunomodulatory agent with the capacity to stimulate immune cell cytokine production and ADCC activity."( A Phase I Trial to Evaluate Antibody-Dependent Cellular Cytotoxicity of Cetuximab and Lenalidomide in Advanced Colorectal and Head and Neck Cancer.
Bertino, EM; Carson, WE; Davis, M; Grever, M; McMichael, EL; Mo, X; Otterson, GA; Paul, B; Trikha, P, 2016
)
1.38
"Lenalidomide is a novel analogue of thalidomide and has anti‑inflammatory, immunomodulatory and anti‑angiogenic effects."( Lenalidomide induces apoptosis and inhibits angiogenesis via caspase‑3 and VEGF in hepatocellular carcinoma cells.
Ding, Y; Jiang, C; Qu, Z; Wu, J, 2016
)
2.6
"Lenalidomide is an analogue of thalidomide that is more powerful and associated with less neurotoxicity than thalidomide."( Treatment of prurigo nodularis with lenalidomide.
Arjona-Aguilera, C; Jiménez-Gallo, D; Linares-Barrios, M; Ossorio-García, L; Rodríguez-Mateos, ME, 2017
)
1.45
"Lenalidomide is an immunomodulatory compound with high clinical activity in multiple myeloma. "( IKZF1 expression is a prognostic marker in newly diagnosed standard-risk multiple myeloma treated with lenalidomide and intensive chemotherapy: a study of the German Myeloma Study Group (DSMM).
Bargou, R; Bassermann, F; Bullinger, L; Bunjes, D; Döhner, H; Einsele, H; Engelhardt, M; Greiner, A; Knop, S; Kolmus, S; Köpff, S; Krönke, J; Kuchenbauer, F; Kull, M; Langer, C; Mügge, LO; Schreder, M; Straka, C; Teleanu, V, 2017
)
2.11
"Lenalidomide is an immunomodulatory drug (IMiDs) with clinical efficacy in multiple myeloma (MM) and other late B-cell neoplasms. "( Multiple myeloma cells' capacity to decompose H
Ahmann, GJ; Bergsagel, PL; Braggio, E; Chesi, M; Fonseca, R; Panchabhai, SC; Sebastian, S; Shi, CX; Stewart, AK; Van Wier, SA; Zhu, YX, 2017
)
1.9
"Lenalidomide is a suitable candidate for long-lasting maintenance as it is an oral drug, active against DLBCL that can be taken for years with an acceptable toxicity profile."( Lenalidomide maintenance in patients with relapsed diffuse large B-cell lymphoma who are not eligible for autologous stem cell transplantation: an open label, single-arm, multicentre phase 2 trial.
Arcari, A; Bertoldero, G; Calimeri, T; Cecchetti, C; Chiozzotto, M; Couto, S; Fabbri, A; Ferreri, AJ; Frezzato, M; Govi, S; Nonis, A; Ponzoni, M; Re, A; Ren, Y; Rocco, AD; Rusconi, C; Sassone, M; Scarfò, L; Spina, M; Stelitano, C; Zaja, F; Zambello, R, 2017
)
2.62
"Lenalidomide is an immunomodulatory drug, structurally related to thalidomide, with pleiotropic activity including antiangiogenic and antineoplastic properties. "( Treatment of plasma cell dyscrasias with lenalidomide.
Dimopoulos, MA; Kastritis, E; Rajkumar, SV, 2008
)
2.05
"Lenalidomide is a safe and effective therapy for refractory metastatic RCC. "( Lenalidomide therapy for metastatic renal cell carcinoma.
Amato, RJ; Hernandez-McClain, J; Khan, M; Saxena, S, 2008
)
3.23
"Lenalidomide is an oral analogue of thalidomide that lacks the neurotoxic side effects associated with the parent drug, and has shown significant antimyeloma activity."( Lenalidomide and its role in the management of multiple myeloma.
Boccadoro, M; Cavallo, F; Falco, P; Larocca, A; Liberati, AM; Musto, P; Palumbo, A, 2008
)
2.51
"Lenalidomide (Revlimid) is an immunomodulatory drug and an analogue of thalidomide, a known teratogen. "( RevAssist: a comprehensive risk minimization programme for preventing fetal exposure to lenalidomide.
Brandenburg, NA; Bwire, R; Castaneda, CP; Freeman, J; Quigley, C; Zeldis, JB, 2008
)
2.01
"Lenalidomide is a candidate for study in AML based on its clinical activity in a related disorder, myelodysplastic syndrome (MDS), with the 5q- chromosomal abnormality."( Single-agent lenalidomide induces complete remission of acute myeloid leukemia in patients with isolated trisomy 13.
Abboud, CN; Blum, W; Byrd, JC; Fehniger, TA; Kefauver, C; Marcucci, G; Payton, JE; Vij, R, 2009
)
1.44
"Lenalidomide is an immunomodulatory drug, which has anti-myeloma activity in vitro. "( United Kingdom myeloma forum position statement on the use of lenalidomide in multiple myeloma.
Behrens, J; Bird, J; Cavenagh, J; Cook, G; Davies, F; Morgan, G; Morris, C; Schey, S; Tighe, J; Williams, C, 2009
)
2.04
"Lenalidomide is an important contemporary treatment option for patients with multiple myeloma (MM). "( A case of severe aplastic anemia secondary to treatment with lenalidomide for multiple myeloma.
Alexandrescu, DT; Dasanu, CA, 2009
)
2.04
"Lenalidomide therapy is a potential alternative or adjunctive treatment for patients with severe, chronic DLE that is refractory to standard therapies. "( Lenalidomide for the treatment of resistant discoid lupus erythematosus.
Albrecht, J; Bonilla-Martinez, Z; Okawa, J; Rose, M; Rosenbach, M; Shah, A; Werth, VP, 2009
)
3.24
"Lenalidomide is an agent that has shown great activity in patients with multiple myeloma (MM). "( Impairment of filgrastim-induced stem cell mobilization after prior lenalidomide in patients with multiple myeloma.
Alousi, A; Anderlini, P; Andersson, B; Champlin, R; de Lima, M; Giralt, S; Hosing, C; Jones, R; Kebriaei, P; Khouri, I; Körbling, M; McMannis, J; Nieto, Y; Popat, U; Qazilbash, M; Saliba, R; Shpall, E; Thandi, R; Thomas, S; Wang, M; Weber, D, 2009
)
2.03
"Lenalidomide is a 4-amino-glutamyl analogue of thalidomide that lacks the neurologic side effects of sedation and neuropathy and has emerged as a drug with activity against various hematological and solid malignancies."( Mechanism of action of lenalidomide in hematological malignancies.
Das, B; Goel, S; Heuck, C; Kotla, V; Nischal, S; Verma, A; Vivek, K, 2009
)
1.38
"Lenalidomide is an immunomodulatory agent recently reported to be effective in the treatment of transfusion-dependent anemia due to low- or intermediate-1 risk myelodysplastic syndromes (MDS) associated with a deletion 5q (del 5q) cytogenetic abnormality. "( Lenalidomide is active in Japanese patients with symptomatic anemia in low- or intermediate-1 risk myelodysplastic syndromes with a deletion 5q abnormality.
Harada, H; Kimura, A; Matsuda, A; Ozawa, K; Suzuki, K; Suzuki, T; Takatoku, M; Takeshita, K; Taniwaki, M; Tohyama, K; Tsudo, M; Watanabe, M; Yanagita, S; Yoshida, Y, 2009
)
3.24
"Lenalidomide is a potent immunomodulatory agent, with the ability to down-regulate pro-inflammatory cytokines and up-regulate anti-inflammatory cytokines."( Lenalidomide (Revlimid) administration at symptom onset is neuroprotective in a mouse model of amyotrophic lateral sclerosis.
Beal, MF; Calingasan, NY; Hensley, K; Kiaei, M; Neymotin, A; Petri, S; Schafer, P; Stewart, C; Wille, E, 2009
)
2.52
"Lenalidomide is an immunomodulatory drug (IMiD) with erythropoietic activity in myelodysplastic syndromes (MDS) that is karyotype dependent. "( Lenalidomide--a transforming therapeutic agent in myelodysplastic syndromes.
List, A, 2009
)
3.24
"Lenalidomide is a novel immunomodulatory agent with antiproliferative activities. "( Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin's Lymphoma.
Cole, C; Ervin-Haynes, A; Justice, G; Kaplan, H; Moore, T; Pietronigro, D; Reeder, C; Takeshita, K; Voralia, M; Vose, JM; Wiernik, PH; Witzig, TE; Zeldis, JB, 2009
)
3.24
"lenalidomide is a thalidomide analogue approved for treatment of myelodysplastic syndromes (MDS) associated with a cytogenetic 5q deletion. "( The clinical utility of lenalidomide in multiple myeloma and myelodysplastic syndromes.
Bonkowski, J; Kolesar, JM; Vermeulen, LC, 2010
)
2.11
"lenalidomide is an effective agent for the treatment of MDS with a 5q deletion and relapsed or refractory multiple myeloma."( The clinical utility of lenalidomide in multiple myeloma and myelodysplastic syndromes.
Bonkowski, J; Kolesar, JM; Vermeulen, LC, 2010
)
2.11
"Lenalidomide is a thalidomide analog and an immunomodulatory drug with demonstrated efficacy in various hematological malignancies. "( Distribution of lenalidomide into semen of healthy men after multiple oral doses.
Assaf, M; Chen, N; Choudhury, S; Laskin, OL; Lau, H; Wang, X, 2010
)
2.15
"Lenalidomide is a more potent and less toxic oral analog of thalidomide. "( Lenalidomide-induced interstitial lung disease.
Chen, Y; Kiatsimkul, P; Nugent, K; Raj, R, 2010
)
3.25
"Lenalidomide is an oral immunomodulatory drug that is highly effective in treating multiple myeloma, has a favorable safety profile and is now being evaluated as maintenance therapy, preventive therapy and in combination with other new agents."( Lenalidomide in multiple myeloma: current role and future directions.
Bizzari, JP; Jacques, C; Knight, RD; Tozer, A; Zeldis, JB, 2010
)
3.25
"Lenalidomide is an active treatment for multiple myeloma (MM) and is increasingly used as part of the initial treatment of this disease. "( Stem cell collection in patients with multiple myeloma: impact of induction therapy and mobilization regimen.
Cook, R; Cunningham, K; Gardler, M; Hummel, K; Luger, SM; Mangan, PA; Nazha, A; O'Doherty, U; Porter, DL; Schuster, S; Siegel, D; Stadtmauer, EA; Vogl, DT, 2011
)
1.81
"Lenalidomide is a promising new drug in the treatment of patients with multiple myeloma. "( [The use of lenalidomide in the treatment of multiple myeloma].
Hájek, R; Holánek, M, 2010
)
2.18
"Lenalidomide is a new immunomodulatory drug, FDA-approved for the treatment of the 5q-myelodysplastic syndrome and refractory or relapsed multiple myeloma (MM). "( A case of lenalidomide-induced hypersensitivity pneumonitis.
Feilchenfeldt, J; Györik, S; Lerch, E; Mazzucchelli, L; Quadri, F, 2010
)
2.21
"Lenalidomide (LDM) is an immunomodulatory and anti-angiogenic drug which has been shown to be effective in several haematological disorders (multiple myeloma [MM], myeloid metaplasia with myelofibrosis [MF] and myelodysplastic syndrome [MDS]). "( [Assessing lenalidomide for treating multiple myeloma, myelofibrosis and myelodysplastic syndrome].
Arroyo Domingo, E; Castillo Valero, I; Hernández Prats, C; Real Panisello, M; Romero Iborra, F; Sánchez Casado, MI,
)
1.96
"Lenalidomide is an immunomodulatory drug that has shown preliminary activity in the treatment of chronic lymphocytic leukemia (CLL). "( Management of patients with chronic lymphocytic leukemia treated with lenalidomide.
Chanan-Khan, A; Miller, KC; Musial, L; Whitworth, A, 2010
)
2.04
"Lenalidomide is an immunomodulatory agent used to treat plasma cell dyscrasias. "( Kidney dysfunction during lenalidomide treatment for AL amyloidosis.
Dember, LM; Fennessey, S; Finn, KT; Sanchorawala, V; Seldin, DC; Shelton, A; Specter, R; Zeldis, JB, 2011
)
2.11
"Lenalidomide is a functional and structural analogue of thalidomide with a relatively benign toxicity profile and pleiotropic anti-tumor activity, exhibiting anti-angiogenic and immunomodulatory effects. "( Lenalidomide: a synthetic compound with an evolving role in cancer management.
Grivas, PD; Saloura, V, 2010
)
3.25
"Lenalidomide is an immunomodulatory agent derived from thalidomide and is approved for the treatment of multiple myeloma and myelodysplastic syndromes. "( Myocarditis during lenalidomide therapy.
Ahmadi, T; Carver, JR; Chong, EA; Nasta, S; Schuster, SJ; Stonecypher, M; Wheeler, JE, 2010
)
2.13
"Lenalidomide is an IMiD® immunomodulatory compound, incorporating structural modification of the drug thalidomide, which is active against a wide variety of autoimmune Th-2-dependent disorders, including erythema nodosum of leprosy, leishmaniasis, as well as severe ulcerative disorders such as Behcet's syndrome."( The potential of immunomodulatory drugs in the treatment of solid tumors.
Dalgleish, A; Galustian, C, 2010
)
1.08
"Lenalidomide is a novel immunomodulatory agent with a unique dual mechanism of action: its tumoricidal effect leads to direct tumor cell death, and its immunomodulatory effect keeps the tumor in remission. "( Lenalidomide: an update on evidence from clinical trials.
Dimopoulos, MA; Terpos, E, 2010
)
3.25
"Lenalidomide is an IMiDs® immunomodulatory compound with a dual mechanism of action - tumoricidal effects rapidly reduce MM burden while long-term immunomodulatory actions maintain tumor suppression."( Future drug developments in multiple myeloma: an overview of novel lenalidomide-based combination therapies.
Morgan, G, 2010
)
1.32
"Lenalidomide is an oral immunomodulatory drug with multiple effects on the immune system and tumor cell microenvironment leading to inhibition of malignant cell growth. "( Single-agent lenalidomide in the treatment of previously untreated chronic lymphocytic leukemia.
Bergsagel, PL; Brandwein, J; Chen, CI; Dave, N; Gibson, S; Hernandez, T; Johnston, J; Kukreti, V; Lau, A; Leung-Hagesteijn, C; Li, ZH; Pantoja, M; Paul, H; Spaner, D; Trudel, S; Wei, E; Xu, W, 2011
)
2.18
"Lenalidomide is an immunomodulatory agent with antitumor activity in B-cell malignancies. "( An international phase II trial of single-agent lenalidomide for relapsed or refractory aggressive B-cell non-Hodgkin's lymphoma.
Bouabdallah, R; Buckstein, R; Czuczman, MS; Ervin-Haynes, AL; Guo, P; Haioun, C; Pietronigro, D; Polikoff, JA; Reeder, CB; Tilly, H; Vose, JM; Witzig, TE; Zinzani, PL, 2011
)
2.07
"Lenalidomide is a thalidomide analogue, designed to have improved efficacy and tolerability over the parent drug. "( Previous thalidomide therapy may not affect lenalidomide response and outcome in relapse or refractory multiple myeloma patients.
Benevolo, G; Berruti, A; Boccadoro, M; Bringhen, S; Caravita, T; Cavallo, F; Corradini, P; Gay, F; Guglielmelli, T; Montefusco, V; Offidani, M; Palumbo, A; Petrucci, MT; Piro, E; Rrodhe, S; Saglio, G, 2011
)
2.07
"Lenalidomide is an antiangiogenic drug associated with hypothyroidism. "( Thyroid abnormalities in patients treated with lenalidomide for hematological malignancies: results of a retrospective case review.
Brown, K; Clayton, W; Figaro, MK; Jagasia, S; Kassim, A; Lakhani, VT; Usoh, C, 2011
)
2.07
"Lenalidomide is an anti-angiogenic IMiD(®) immunomodulatory drug. "( A prospective clinical trial of lenalidomide with topotecan in women with advanced epithelial ovarian carcinoma.
Carter, JS; Downs, LS, 2011
)
2.1
"Lenalidomide is an immunomodulatory agent with activity in a range of haematological cancers including chronic lymphocytic leukaemia (CLL). "( Lenalidomide can be highly effective in chronic lymphocytic leukaemia despite T-cell depletion and deletion of chromosome 17p.
Arumainathan, A; Kalakonda, N; Pettitt, AR, 2011
)
3.25
"Lenalidomide is an IMiD immunomodulatory compound that has both tumouricidal and immunomodulatory activity in MM."( Lenalidomide downregulates the cell survival factor, interferon regulatory factor-4, providing a potential mechanistic link for predicting response.
Bartlett, JB; Bolomsky, A; Brady, H; Chopra, R; Daniel, T; Gaidarova, S; Gisslinger, H; Heintel, D; Hilgarth, B; Jäger, U; Lopez-Girona, A; Ludwig, H; Mendy, D; Schafer, PH; Schreder, M; Zhang, LH; Zojer, N, 2011
)
2.53
"Lenalidomide is a derivative of thalidomide and is FDA-approved for the treatment of myelodysplastic syndrome and, in combination with dexamethasone, for the treatment of relapsed multiple myeloma. "( Reversible pulmonary toxicity due to lenalidomide.
Barker, A; Coates, S; Spurgeon, S, 2012
)
2.09
"Lenalidomide is a thalidomide analogue that may serve as an adjunctive therapy for treatment-refractory cutaneous lupus erythematosus (CLE)."( Lenalidomide therapy in treatment-refractory cutaneous lupus erythematosus: histologic and circulating leukocyte profile and potential risk of a systemic lupus flare.
Braunstein, I; Goodman, NG; Kovarik, CL; Krathen, M; Luning Prak, E; Okawa, J; Rosenbach, M; Shah, A; Werth, VP, 2012
)
3.26
"Lenalidomide is a derivative of thalidomide used in the treatment of multiple myeloma."( Possible lenalidomide-induced Stevens-Johnson syndrome during treatment for multiple myeloma.
Bolesta, S; Boruah, PK; Shetty, SM, 2011
)
1.51
"Lenalidomide is an IMiDs® oral immunomodulatory compound developed for the treatment of patients with multiple myeloma (MM) and myelodysplastic syndromes (MDS). "( The clinical safety of lenalidomide in multiple myeloma and myelodysplastic syndromes.
Fenaux, P; Freeman, J; Palumbo, A; Weiss, L, 2012
)
2.13
"Lenalidomide (LEN) is a structural analogue of Thalidomide and is currently considered a promising compound among immunomodulatory drugs. "( Lenalidomide in multiple myeloma: current experimental and clinical data.
Cives, M; Dammacco, F; Milano, A; Silvestris, F, 2012
)
3.26
"Lenalidomide is an antiangiogenic and immunomodulatory agent that has been utilized in the treatment of patients with brain tumors."( Plasma and cerebrospinal fluid pharmacokinetics of thalidomide and lenalidomide in nonhuman primates.
Berg, SL; Dauser, RC; Gibson, BW; McGuffey, LH; Muscal, JA; Nuchtern, JG; Sun, Y, 2012
)
1.34
"Lenalidomide (len) is an analog of thalidomide (thal), and both are used in the treatment of a diverse group of medical conditions. "( Lenalidomide alone or lenalidomide plus dexamethasone significantly inhibit IgG and IgM in vitro... A possible explanation for their mechanism of action in treating multiple myeloma.
Greig, N; Sandoval, F; Shannon, E; Stagg, P, 2012
)
3.26
"Lenalidomide is an immunomodulatory, antiangiogenic drug that is a structural analog of thalidomide. "( [Lenalidomide in hematological malignancies---review].
Jin, X; Zhang, YZ, 2012
)
2.73
"Lenalidomide is an immunomodulator used to treat 5q-myelodysplastic syndrome, myelofibrosis, and multiple myeloma. "( Lenalidomide-induced elevated bilirubin.
Kolesar, JM; Simondsen, KA, 2012
)
3.26
"Lenalidomide is an amino-substituted analog of thalidomide with potent immunomodulatory properties. "( Plasma exchange and rituximab treatment for lenalidomide-associated cold agglutinin disease.
Akpek, G; Brauer, DL; Edelman, B; Hess, JR; Rapoport, AP, 2012
)
2.08
"Lenalidomide is a potent immunomodulatory agent capable of downregulating proinflammatory cytokines such as tumor necrosis factor-α (TNF-α) and upregulating anti-inflammatory cytokines. "( Short-term lenalidomide (Revlimid) administration ameliorates cardiomyocyte contractile dysfunction in ob/ob obese mice.
Dong, M; Hua, Y; Jones, KR; Li, L; Li, Q; Ren, J; Smith, DT; Yuan, M, 2012
)
2.21
"Lenalidomide is a second generation immunomodulatory agent (IMiD), which currently represents the standard of care for treatment of transfusion dependent lower risk myelodysplastic syndrome (MDS) patients with deletion (5q). "( Lenalidomide for treatment of myelodysplastic syndromes.
Komrokji, RS; List, AF, 2012
)
3.26
"Lenalidomide is a second-generation immunomodulatory drug that has been approved to treat relapsed or refractory multiple myeloma. "( Drug-induced interstitial pneumonitis due to low-dose lenalidomide.
Arita, M; Hotta, M; Ishida, T; Kunimasa, K; Maeda, T; Ueda, T, 2012
)
2.07
"Lenalidomide is a thalidomide analogue with immunomodulatory and anti-angiogenic properties that include altering cytokine production, activating T cells, and augmenting natural killer cell function. "( Lenalidomide in solid tumors.
Segler, A; Tsimberidou, AM, 2012
)
3.26
"Lenalidomide is an effective treatment for patients with del(5q) and myelodysplastic syndrome (MDS) The exact mechanism of lenalidomide function and its impact on the prognosis of patients is not known exactly."( Changes associated with lenalidomide treatment in the gene expression profiles of patients with del(5q).
Belickova, M; Caniga, M; Cechova, E; Cermak, J; Dostalova Merkerova, M; Jonasova, A; Krejcik, Z; Michalova, K; Neuwirtova, R; Vesela, J; Votavova, H; Zemanova, Z, 2012
)
2.13
"Lenalidomide is an active agent in the activated B cell-like (ABC) subtype of diffuse large B cell lymphoma (DLBCL), but its mechanism of action is unknown."( Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma.
Balasubramanian, S; Buggy, JJ; Ceribelli, M; Emre, NC; Ferrer, M; Guha, R; Kohlhammer, H; Mathews, LA; Platig, J; Powell, J; Shaffer, AL; Shinn, P; Staudt, LM; Thomas, C; Waldmann, TA; Wright, G; Xiao, W; Xu, W; Yang, Y; Young, RM; Zhang, M; Zhao, H, 2012
)
1.1
"Lenalidomide is a highly potent therapy in low-risk MDS with del 5q, however, the treatment carries a high risk of rapidly developing neutropenia and thrombocytopenia."( Myelodysplastic syndromes: a challenging disease for patients and physicians.
Somani, N, 2012
)
1.1
"Lenalidomide is an effective therapeutic agent for multiple myeloma that exhibits immunomodulatory properties including the activation of T and NK cells. "( Lenalidomide enhances anti-myeloma cellular immunity.
Anderson, KC; Arnason, J; Avigan, D; Glotzbecker, B; Joyce, RM; Kufe, D; Kufe, T; Levine, JD; Luptakova, K; Mills, H; Rosenblatt, J; Stroopinsky, D; Tzachanis, D; Vasir, B; Zwicker, JI, 2013
)
3.28
"Lenalidomide (LEN) is an immunomodulatory drug (IMiD) which exerts tumoricidal effects and has immunomodulatory, anti-inflammatory and anti-angiogenic properties that synergistically keep the tumor in remission. "( Novel lenalidomide-based combinations for treatment of multiple myeloma.
Brunetti, O; Cives, M; Longo, V; Silvestris, F; Simone, V, 2013
)
2.31
"Lenalidomide is an oral immunomodulatory drug, which was recently introduced for the treatment of multiple myeloma (MM). "( Evaluation of the pharmacokinetics, preclinical, and clinical efficacy of lenalidomide for the treatment of multiple myeloma.
Boccadoro, M; Bringhen, S; Cerrato, C; Gay, F; Palumbo, A; Pautasso, C, 2012
)
2.05
"Lenalidomide is a 4-amino-glutamyl analogue of thalidomide that has emerged as a drug with activity against various hematological and solid malignancies."( [Action mechanism of lenalidomide in hematological malignancies - review].
Liu, LR; Qian, SX, 2012
)
1.42
"Lenalidomide is an immunomodulatory drug with efficacy in various hematological malignancies. "( Single-dose pharmacokinetics of lenalidomide in healthy volunteers: dose proportionality, food effect, and racial sensitivity.
Chen, N; Kasserra, C; Lau, H; Liu, L; Reyes, J, 2012
)
2.11
"Lenalidomide is a synthetic derivative of thalidomide exhibiting multiple immunomodulatory activities beneficial in the treatment of several hematological malignancies. "( Pharmacokinetics and tissue disposition of lenalidomide in mice.
Byrd, JC; Gao, Y; Herman, SE; Jarjoura, D; Johnson, AJ; Li, X; Mahoney, E; Phelps, MA; Rozewski, DM; Shin, JD; Stefanovski, MR; Towns, WH; Yang, X, 2012
)
2.08
"Lenalidomide is an immunomodulatory drug frequently used for treatment of patients with multiple myeloma and myelodysplastic syndromes. "( Lenalidomide-associated hepatotoxicity--a case report and literature review.
Fontana, RJ; Konda, A; Nojkov, B; Signori, C, 2012
)
3.26
"Lenalidomide is an immunomodulatory drug derived from thalidomide, developed to maximize its anti-inflammatory and antineoplastic properties while reducing toxicity. "( Current treatment strategies with lenalidomide in multiple myeloma and future perspectives.
Boccadoro, M; Cavallo, F; Larocca, A; Mina, R; Palumbo, A, 2012
)
2.1
"Lenalidomide is a member of the immunomodulatory agents (IMiDs), and is currently approved for use in myelodysplastic syndromes and multiple myeloma. "( Lenalidomide alone and in combination for chronic lymphocytic leukemia.
Chen, CI, 2013
)
3.28
"Lenalidomide is an immunomodulatory drug active as salvage therapy for chronic lymphocytic leukemia (CLL). "( Phase II study of lenalidomide and rituximab as salvage therapy for patients with relapsed or refractory chronic lymphocytic leukemia.
Badoux, XC; Burger, JA; Faderl, S; Ferrajoli, A; Keating, MJ; O'Brien, SM; Sargent, R; Wen, S; Wierda, WG, 2013
)
2.17
"Lenalidomide is an oral immunomodulatory drug derived from thalidomide. "( Lenalidomide as a novel treatment of acute myeloid leukemia.
Borthakur, G; Chen, Y, 2013
)
3.28
"Lenalidomide is an orally bioavailable analogue of thalidomide with more potent immunomodulatory, antiangiogenic, and antitumor activities than the parent compound and with a better safety profile."( Emerging data on IMiDs in the treatment of myelodysplastic syndromes (MDS).
List, AF, 2005
)
1.05
"Lenalidomide is a potent immunomodulating drug that offers different mechanisms of action and therapeutic potential for the treatment of multiple myeloma, MDS, and other malignancies."( Role of lenalidomide in the treatment of multiple myeloma and myelodysplastic syndrome.
Hammond, JM; Maier, SK, 2006
)
2.21
"Lenalidomide (CC-5013) is an immunomodulatory thalidomide analogue licensed in the United States of America (USA) for the treatment of a subtype of myelodysplastic syndrome."( Toxicity profile of the immunomodulatory thalidomide analogue, lenalidomide: phase I clinical trial of three dosing schedules in patients with solid malignancies.
Daines, CA; Dalgleish, AG; Knight, RD; O'Byrne, KJ; Sharma, RA; Steward, WP, 2006
)
1.29
"Lenalidomide (Revlimid) is an immunomodulatory drug that has undergone rapid clinical development in multiple myeloma and was recently approved by the US FDA for use in patients with relapsed disease."( Lenalidomide in multiple myeloma.
Anderson, KC; Hideshima, T; Mitsiades, C; Richardson, PG, 2006
)
2.5
"Lenalidomide (LEN) is a structural and functional analogue of thalidomide that has demonstrated enhanced immunomodulatory properties and a more favorable toxicity profile. "( Phase II study of lenalidomide in patients with metastatic renal cell carcinoma.
Baz, RC; Bukowski, RM; Choueiri, TK; Dreicer, R; Elson, P; Garcia, JA; Jinks, HA; Mekhail, TM; Rini, BI; Thakkar, SG, 2006
)
2.11
"Lenalidomide is an immunomodulatory derivative of thalidomide with significantly greater in vitro activity and a different toxicity profile. "( Phase I trial of three-weekly docetaxel, carboplatin and oral lenalidomide (Revlimid) in patients with advanced solid tumors.
Borden, E; Bukowski, R; Cline-Burkhardt, M; Davis, M; Dowlati, A; Dreicer, R; Kalmadi, S; Mekhail, T; O'Keefe, S; Pelley, RJ, 2007
)
2.02
"Lenalidomide is an immunomodulatory agent with biological activity in several hematologic malignancies, including myelodysplastic syndrome. "( The role of lenalidomide in the treatment of patients with chromosome 5q deletion and other myelodysplastic syndromes.
Kale, V; List, A; Melchert, M, 2007
)
2.16
"Lenalidomide is an active immunomodulatory agent for the treatment of myelodysplastic syndrome with encouraging erythropoetic and cytogenetic remitting activity that is karyotype dependent."( The role of lenalidomide in the treatment of patients with chromosome 5q deletion and other myelodysplastic syndromes.
Kale, V; List, A; Melchert, M, 2007
)
2.16
"Lenalidomide is a novel analog of thalidomide with both immunomodulatory and antiangiogenic properties that are more potent than those same properties in the parent compound. "( Lenalidomide: immunomodulatory, antiangiogenic, and clinical activity in solid tumors.
Dreicer, R, 2007
)
3.23
"Lenalidomide is an immunomodulatory drug, structurally related to thalidomide, which has pleotropic activity, including antiangiogenic and antineoplastic properties. "( The evolving role of lenalidomide in the treatment of hematologic malignancies.
Dimopoulos, MA; Kastritis, E, 2007
)
2.1
"Lenalidomide is an immunomodulatory drug that was developed by modification of the first-generation immunomodulatory drug thalidomide in a drug discovery program. "( Lenalidomide: the emerging role of a novel targeted agent in malignancies.
Baz, R; Kalmadi, S; Mahindra, A, 2007
)
3.23
"Lenalidomide is an immunomodulatory agent approved for use in patients with myelodysplastic syndrome, and in combination with dexamethasone for refractory or relapsed multiple myeloma. "( Hypersensitivity pneumonitis-like syndrome associated with the use of lenalidomide.
Abonour, R; Knox, K; Smith, P; Thornburg, A; Twigg, HL, 2007
)
2.02
"Lenalidomide is a structural analogue of thalidomide."( [New treatment of multiple myeloma].
Hulin, C, 2007
)
1.06
"Lenalidomide is a potent, novel thalidomide analog that has demonstrated promising clinical activity in patients with relapsed or refractory multiple myeloma (MM). "( Lenalidomide: a new agent for patients with relapsed or refractory multiple myeloma.
Tariman, JD, 2007
)
3.23
"Lenalidomide is an analogue of thalidomide and has been shown to be more potent than thalidomide in the stimulation of T-cell, interleukin-2, and interferon-gamma production. "( Lenalidomide in the treatment of multiple myeloma.
Rao, KV, 2007
)
3.23
"Lenalidomide is a structural analogue of thalidomide with similar but more potent biologic activity. "( Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma.
Attal, M; Corso, A; Dimopoulos, M; Dmoszynska, A; Facon, T; Foà, R; Harousseau, JL; Hellmann, A; Knight, RD; Masliak, Z; Olesnyckyj, M; Patin, J; Prince, HM; San Miguel, J; Spencer, A; Yu, Z; Zeldis, JB, 2007
)
3.23
"Lenalidomide (CC-5013) is a structural derivative of thalidomide, with antiangiogenic and immunomodulatory effects. "( Phase II trial of lenalidomide in patients with metastatic renal cell carcinoma.
Bacik, J; DeLuca, J; Ishill, N; Kondagunta, GV; Motzer, RJ; Patel, PH; Russo, P; Schwartz, L, 2008
)
2.12
"Lenalidomide is a very active drug in myelodysplastic syndrome with del (5q). "( Unusual clonal evolution involving 5q in a case of myelodysplastic syndrome with deletion 5q 31 treated with lenalidomide.
Da Rocha, A; Eclache, V; Fenaux, P; Le Roux, G, 2008
)
2
"Lenalidomide is an effective new agent for the treatment of patients with myelodysplastic syndrome (MDS), an acquired hematopoietic disorder characterized by ineffective blood cell production and a predisposition to the development of leukemia. "( An erythroid differentiation signature predicts response to lenalidomide in myelodysplastic syndrome.
Bosco, J; Ebert, BL; Galili, N; Golub, TR; Ladd-Acosta, C; Mak, R; Pretz, J; Raza, A; Stone, R; Tamayo, P; Tanguturi, S, 2008
)
2.03
"Lenalidomide is a novel anticancer agent that has made a major impact in the treatment of patients with B-cell malignancies. "( Lenalidomide for the treatment of B-cell malignancies.
Chanan-Khan, AA; Cheson, BD, 2008
)
3.23
"Lenalidomide is a novel therapeutic agent with uncertain mechanism of action that is clinically active in myelodysplastic syndrome (MDS) and multiple myeloma (MM). "( Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia.
Andritsos, LA; Awan, F; Blum, W; Byrd, JC; Chen, CS; Jarjoura, D; Johnson, AJ; Kefauver, C; Knight, RD; Lapalombella, R; Lehman, A; Lozanski, G; May, SE; Raymond, CA; Ruppert, AS; Smith, LL; Wang, DS, 2008
)
2.13
"Lenalidomide is an immunomodulatory drug derived from thalidomide. "( Lenalidomide in the treatment of multiple myeloma: a review.
Armoiry, X; Aulagner, G; Facon, T, 2008
)
3.23

Effects

Lenalidomide has a central role in the treatment of multiple myeloma and results in improved survival. The drug has a manageable safety profile whether administered as a single agent or in combination with rituximab.

Lenalidomide has been used as an immunomodulatory drug with direct and indirect anti-tumor effects. It has been approved by the US FDA and the European Medicines Agency for use in combination with dexamethasone.

ExcerptReferenceRelevance
"Lenalidomide has a central role in the treatment of multiple myeloma and results in improved survival. "( Hepatitis E during lenalidomide treatment for multiple myeloma in complete remission.
Faber, LM; Kootte, RS, 2017
)
2.23
"Lenalidomide has a more favorable adverse effect profile compared to its parent compound thalidomide. "( Lenalidomide and thalidomide in the treatment of chronic pain.
Asher, C; Furnish, T, 2013
)
3.28
"Lenalidomide has a unique mechanism of action in CLL."( Lenalidomide in chronic lymphocytic leukemia: the present and future in the era of tyrosine kinase inhibitors.
Colaci, E; Fiorcari, S; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Potenza, L, 2016
)
2.6
"Lenalidomide has a manageable safety profile whether administered as a single agent or in combination with rituximab."( Clinical experience with lenalidomide alone or in combination with rituximab in indolent B-cell and mantle cell lymphomas.
Martin, P; Ruan, J; Schuster, SJ; Shah, B, 2016
)
1.46
"Lenalidomide has a predominantly renal route of excretion and in patients with RI the plasma concentration and half-life of the drug are significantly increased."( Treatment with lenalidomide and dexamethasone in patients with multiple myeloma and renal impairment.
Alegre, A; Dimopoulos, MA; Goldschmidt, H; Mark, T; Niesvizky, R; Terpos, E, 2012
)
1.45
"Lenalidomide has a better toxicity profile than thalidomide."( Novel immunomodulatory compounds in multiple myeloma.
Mahindra, A; Saini, N, 2013
)
1.11
"Lenalidomide has dramatic therapeutic effects in patients with low-risk MDS and a chromosome 5q31 deletion, resulting in complete cytogenetic remission in >60% of patients."( Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
Boultwood, J; Cattan, H; Christensson, B; Emanuelsson, EK; Forsblom, AM; Hellström-Lindberg, E; Jädersten, M; Merup, M; Nilsson, L; Pellagatti, A; Samuelsson, J; Sander, B; Wainscoat, JS, 2007
)
2.5
"Lenalidomide has response rates of 45% in relapsed transformed DLBCL."( Lenalidomide in combination with R-CHOP produces high response rates and progression-free survival in new, untreated diffuse large B-cell lymphoma transformed from follicular lymphoma: results from the Phase 2 MC078E study.
Ansell, SM; Desai, SH; Habermann, TM; Inwards, DJ; Johnston, PB; King, RL; LaPlant, B; Macon, WR; Micallef, I; Nowakowski, GS; Porrata, LF; Wang, Y; Witzig, TE, 2021
)
2.79
"Lenalidomide has been standard therapy for multiple myeloma and other haematological malignancies for more than a decade. "( Second primary malignancies in patients with haematological cancers treated with lenalidomide: a systematic review and meta-analysis.
Boyiadzis, M; Franz, J; Jones, JR; Klem, ML; Lontos, K; Saleem, K; Shaikh, N; Yabes, JG, 2022
)
2.39
"Lenalidomide has major efficacy in LR-MDS with deletion 5q."( How we manage adults with myelodysplastic syndrome.
Ades, L; Fenaux, P; Platzbecker, U, 2020
)
1.28
"Lenalidomide-rituximab has been demonstrated to be an effective chemotherapy-free therapy that improves upon single-agent rituximab and may become an alternative to chemoimmunotherapy."( Lenalidomide in follicular lymphoma.
Flowers, CR; Fowler, NH; Leonard, JP, 2020
)
2.72
"Lenalidomide, which has been safely combined with CHOP to treat B-cell lymphoma, has shown efficacy as a single agent in AITL treatment."( Integrative analysis of a phase 2 trial combining lenalidomide with CHOP in angioimmunoblastic T-cell lymphoma.
Bachy, E; Becker, S; Beldi-Ferchiou, A; Bossard, C; Bouabdallah, R; Cacheux, V; Camus, V; Cartron, G; Casasnovas, O; Cottereau, AS; de Leval, L; Delfau-Larue, MH; Delmer, A; Fataccioli, V; Fourati, S; Gaulard, P; Haioun, C; Lemonnier, F; Letourneau, A; Meignan, M; Missiaglia, E; Moles-Moreau, MP; Parrens, M; Pelletier, L; Régny, C; Robe, C; Safar, V; Voillat, L, 2021
)
1.6
"Lenalidomide has recently emerged as a therapeutic option for POEMS syndrome."( Cereblon expression is a prognostic marker in newly diagnosed POEMS syndrome treated with lenalidomide plus dexamethasone.
Cai, H; Cai, QQ; Cao, XX; Gao, XM; Le, J; Li, J; Zhao, H, 2021
)
1.56
"Lenalidomide, which has immunomodulatory effect, can enhance the activation of T-, NK-cells and endothelial cells, however there are no data available whether it can modulate bone marrow stromal cells (BMSCs)."( Lenalidomide abrogates the survival effect of bone marrow stromal cells in chronic lymphocytic leukemia.
Barna, G; Czeti, Á; Hernádfői, M; Kriston, C; Márk, Á; Matolcsy, A; Plander, M; Szabó, O; Szalóki, G; Takács, F, 2021
)
2.79
"Lenalidomide monotherapy has rarely been evaluated for newly diagnosed transplant-ineligible MM patients."( Multicenter, phase II study of response-adapted lenalidomide-based therapy for transplant-ineligible patients with newly diagnosed multiple myeloma without high-risk features.
Baz, R; Kang, HJ; Kim, JA; Kim, JS; Kim, K; Kim, SH; Kim, YS; Kwak, JY; Lee, JH; Lee, JJ; Lee, WS; Min, CK; Suh, C; Yoo, KH; Yoon, DH; Yoon, SS, 2022
)
1.7
"Lenalidomide has a central role in the treatment of multiple myeloma and results in improved survival. "( Hepatitis E during lenalidomide treatment for multiple myeloma in complete remission.
Faber, LM; Kootte, RS, 2017
)
2.23
"Lenalidomide has immunomodulatory and anti-angiogenic effects and showed moderate anti-tumour efficacy in patients with. "( Lenalidomide as second-line therapy for advanced hepatocellular carcinoma: exploration of biomarkers for treatment efficacy.
Chen, BB; Cheng, AL; Hsu, C; Hsu, CH; Lin, ZZ; Ou, DL; Shao, YY; Wang, MJ, 2017
)
3.34
"Lenalidomide has multifaceted antimyeloma properties, including direct tumoricidal and immunomodulatory effects. "( Lenalidomide in combination or alone as maintenance therapy following autologous stem cell transplant in patients with multiple myeloma: a review of options for and against.
Anderson, KC; Attal, M; Holstein, SA; McCarthy, PL; Richardson, PG; Schlossman, RL, 2017
)
3.34
"Lenalidomide has been associated with an increased risk of venous thromboembolism (VTE) in multiple myeloma. "( Venous thromboembolism in patients with non-Hodgkin lymphoma or chronic lymphocytic leukemia treated with lenalidomide: a systematic review.
Orna, E; Sancho, JM; Sorigue, M, 2018
)
2.14
"Lenalidomide has been shown to be potentially teratogenic in thalidomide-sensitive animal species. "( Association of pharmacokinetic profiles of lenalidomide in human plasma simulated using pharmacokinetic data in humanized-liver mice with liver toxicity detected by human serum albumin RNA.
Guengerich, FP; Kamiya, Y; Kusama, T; Mitsui, M; Murayama, N; Shimizu, M; Suemizu, H; Uehara, S; Yamazaki, H, 2018
)
2.19
"Lenalidomide has anti-tumor activity in CLL but can be complicated by tumor lysis syndrome (TLS) and tumor flare (TF). "( A phase 2 study of lenalidomide and dexamethasone in previously untreated patients with chronic lymphocytic leukemia (CLL).
Chen, CI; Croucher, D; Gibson, SB; Johnston, JB; Kakar, S; Lau, A; Le, LW; Paul, H; Queau, M; Sherry, B; Snitzler, S; Spaner, D; Trudel, S; Wei, EN, 2019
)
2.29
"Lenalidomide monotherapy has not been evaluated in newly diagnosed myeloma patients."( Lenalidomide-based response-adapted therapy for older adults without high risk myeloma.
Alsina, M; Baz, R; Brayer, J; Fridley, BL; Hillgruber, N; Lee, JH; Naqvi, SMH; Shain, KH; Sullivan, DM, 2019
)
2.68
"As lenalidomide has promising single agent activity in multiple relapsed HL, we replaced bleomycin in ABVD with lenalidomide in this phase-I trial."( Doxorubicin, vinblastine, dacarbazine and lenalidomide for older Hodgkin lymphoma patients: final results of a German Hodgkin Study Group (GHSG) phase-I trial.
Atta, J; Böll, B; Borchmann, P; Bürkle, C; Eichenauer, DA; Engert, A; Feuring-Buske, M; Fuchs, M; Pfreundschuh, M; Plütschow, A; Sökler, M; Thielen, I; Vogelhuber, M; von Tresckow, B, 2019
)
1.29
"Lenalidomide has modest single-agent activity comparable with other newer drugs in recurrent diffuse large B cell lymphoma with response rates between 19% and 28%. "( Checking in on Lenalidomide in Diffuse Large B Cell Lymphoma.
Bertino, JR; Cooper, DL; Goldfinger, M; Xu, M, 2019
)
2.31
"Lenalidomide has a more favorable adverse effect profile compared to its parent compound thalidomide. "( Lenalidomide and thalidomide in the treatment of chronic pain.
Asher, C; Furnish, T, 2013
)
3.28
"Lenalidomide has been linked to myelodysplastic syndrome (MDS) after autotransplants for myeloma. "( Risk factors for MDS and acute leukemia following total therapy 2 and 3 for multiple myeloma.
Abdallah, AO; Bailey, C; Barlogie, B; Chauhan, N; Cottler-Fox, M; Crowley, J; Epstein, J; Heuck, CJ; Hoering, A; Johann, D; Muzaffar, J; Petty, N; Rosenthal, A; Sawyer, J; Sexton, R; Singh, Z; Usmani, SZ; van Rhee, F; Waheed, S; Yaccoby, S, 2013
)
1.83
"Lenalidomide has significant antimyeloma activity but it is associated with a significant risk of venous thromboembolism (VTE). "( Clinical and genetic factors associated with venous thromboembolism in myeloma patients treated with lenalidomide-based regimens.
Bagratuni, T; Dimopoulos, MA; Eleutherakis-Papaiakovou, E; Gavriatopoulou, M; Kanelias, N; Kastritis, E; Kostouros, E; Politou, M; Roussou, M; Terpos, E, 2013
)
2.05
"Lenalidomide has been approved for RRMM for several years in Europe and North America, but has not been accessible to Asian patients in the past."( Lenalidomide with dexamethasone treatment for relapsed/refractory myeloma patients in Korea-experience from 110 patients.
Eom, HS; Jo, DY; Jun, HJ; Kim, JS; Kim, K; Kim, KH; Kim, SH; Kim, SJ; Kim, YS; Kwak, JY; Lee, JH; Lee, JJ; Lee, JO; Min, CK; Moon, JH; Mun, YC; Park, SK; Ryoo, HM; Suh, C; Voelter, V; Yoon, SS, 2014
)
2.57
"Lenalidomide has been linked to second primary malignancies in myeloma. "( Second primary malignancies with lenalidomide therapy for newly diagnosed myeloma: a meta-analysis of individual patient data.
Anderson, K; Barlogie, B; Boccadoro, M; Bringhen, S; Cavo, M; Ciccone, G; Dimopoulos, MA; Evangelista, A; Hajek, R; Kumar, SK; Larocca, A; Lonial, S; Lupparelli, G; McCarthy, PL; Musto, P; Nooka, AK; Offidani, M; Palumbo, A; Petrucci, MT; Richardson, P; Sonneveld, P; Spencer, A; Usmani, S; van der Holt, B; Waage, A; Zweegman, S, 2014
)
2.13
"Lenalidomide has high activity in relapsed or refractory aggressive B-cell lymphomas."( Lenalidomide plus R-CHOP21 in elderly patients with untreated diffuse large B-cell lymphoma: results of the REAL07 open-label, multicentre, phase 2 trial.
Baldi, I; Botto, B; Carella, AM; Castellino, A; Chiappella, A; Ciccone, G; Congiu, A; Dreyling, M; Fattori, PP; Franceschetti, S; Gaidano, G; Gaudiano, M; Inghirami, G; Ladetto, M; Liberati, AM; Molinari, AL; Pavone, V; Rossi, G; Salvi, F; Spina, M; Vitolo, U; Zaccaria, A; Zanni, M; Zinzani, P, 2014
)
2.57
"Lenalidomide has demonstrated clinical activity in patients with chronic lymphocytic leukemia (CLL), even though it is not cytotoxic for primary CLL cells in vitro. "( Lenalidomide inhibits the proliferation of CLL cells via a cereblon/p21(WAF1/Cip1)-dependent mechanism independent of functional p53.
Bharati, IS; Cathers, B; Corral, LG; Cui, B; Fecteau, JF; Futalan, D; Gaidarova, S; Ghia, EM; Kipps, TJ; Lopez-Girona, A; Messmer, D; Schwaederlé, M, 2014
)
3.29
"Lenalidomide has significant single-agent activity in relapsed diffuse large B-cell lymphoma (DLBCL). "( Lenalidomide combined with R-CHOP overcomes negative prognostic impact of non-germinal center B-cell phenotype in newly diagnosed diffuse large B-Cell lymphoma: a phase II study.
Ansell, SM; Foran, JM; Gascoyne, RD; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, B; Macon, WR; Micallef, IN; Nelson, GD; Nowakowski, GS; Porrata, LF; Reeder, CB; Rivera, CE; Thompson, CA; Witzig, TE, 2015
)
3.3
"Lenalidomide (Len) has been demonstrated to be one of the most efficient new treatment options."( Combination of lenalidomide with vitamin D3 induces apoptosis in mantle cell lymphoma via demethylation of BIK.
Amiot, M; Brosseau, C; Dousset, C; Le Gouill, S; Maïga, S; Moreau, P; Pellat-Deceunynck, C; Touzeau, C, 2014
)
1.48
"Lenalidomide has dual antiangiogenic and immunomodulatory properties and confirmed antitumor activity in hematologic malignancies. "( A phase II study of lenalidomide in platinum-sensitive recurrent ovarian carcinoma.
Berton-Rigaud, D; Fabbro, M; Favier, L; Lesoin, A; Lortholary, A; Mari, V; Pujade-Lauraine, E; Ray-Coquard, I; Selle, F; Sevin, E, 2014
)
2.17
"Lenalidomide has emerged as an important treatment for patients with multiple myeloma (MM). "( Efficacy and Safety of Lenalidomide in the Treatment of Multiple Myeloma: A Systematic Review and Meta-analysis of Randomized Controlled Trials.
Guo, XN; Qiao, SK; Ren, HY; Ren, JH, 2015
)
2.17
"Lenalidomide has shown protective effects in an animal model of amyotrophic lateral sclerosis, and its mechanism of action involves modulation of cytokine production and inhibition of NF-κB signaling."( Lenalidomide reduces microglial activation and behavioral deficits in a transgenic model of Parkinson's disease.
Anderson, S; Mante, M; Masliah, E; Rockenstein, E; Valera, E, 2015
)
2.58
"Lenalidomide has been approved for the treatment of lower-risk myelodysplastic syndrome (MDS) with 5q deletion (del(5q)). "( Polish experience of lenalidomide in the treatment of lower risk myelodysplastic syndrome with isolated del(5q).
Bieniaszewska, M; Butrym, A; Kumiega, B; Lech-Maranda, E; Madry, K; Mazur, G; Mital, A; Patkowska, E; Rybka, J; Torosian, T; Warzocha, K; Wichary, R, 2015
)
2.18
"Lenalidomide has been shown to produce synergistic effects in experimental models when evaluated in combination with rituximab, dexamethasone, bortezomib, and B-cell receptor signaling inhibitors, consistent with mechanisms complementary to these agents."( Mechanisms of Action of Lenalidomide in B-Cell Non-Hodgkin Lymphoma.
Fowler, N; Gribben, JG; Morschhauser, F, 2015
)
1.45
"Lenalidomide has demonstrated remarkable efficacy for therapy of lower-risk myelodysplastic syndromes (MDS) associated with 5q(-). "( Treatment of Patients With Myelodysplastic Syndrome With Lenalidomide in Clinical Routine in Austria.
Aschauer, G; Burgstaller, S; Fiegl, M; Fridrik, M; Girschikofsky, M; Greil, R; Keil, F; Linkesch, W; Nösslinger, T; Petzer, A; Stauder, R, 2015
)
2.1
"Lenalidomide has a unique mechanism of action in CLL."( Lenalidomide in chronic lymphocytic leukemia: the present and future in the era of tyrosine kinase inhibitors.
Colaci, E; Fiorcari, S; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Potenza, L, 2016
)
2.6
"Lenalidomide has synergistic anticancer effects when used with chemotherapy. "( Phase I clinical trial of lenalidomide in combination with 5-fluorouracil, leucovorin, and oxaliplatin in patients with advanced cancer.
Falchook, G; Fu, S; Hong, DS; Naing, A; Piha-Paul, S; Said, R; Tsimberidou, AM; Wheler, JJ; Ye, Y, 2016
)
2.18
"Lenalidomide has a manageable safety profile whether administered as a single agent or in combination with rituximab."( Clinical experience with lenalidomide alone or in combination with rituximab in indolent B-cell and mantle cell lymphomas.
Martin, P; Ruan, J; Schuster, SJ; Shah, B, 2016
)
1.46
"Lenalidomide has been proved to be effective for relapsed/refractory CLL as a single agent or in combination with various chemo-immunotherapeutic regimens."( Efficacy of lenalidomide in relapsed/refractory chronic lymphocytic leukemia patient: a systematic review and meta-analysis.
Hu, X; Liang, L; Liu, H; Yang, LP; Zhao, M; Zhu, YC, 2016
)
1.53
"Lenalidomide (LEN) has been used as an immunomodulatory drug with direct and indirect anti-tumor effects. "( Lenalidomide enhances the function of dendritic cells generated from patients with multiple myeloma.
Anh-NguyenThi, T; Jaya Lakshmi, T; Jung, SH; Kim, HJ; Lee, HJ; Lee, JJ; Nguyen-Pham, TN; Vo, MC, 2017
)
3.34
"Lenalidomide has been used successfully as an upfront treatment either with high or low dose dexamethasone or with melphalan and prednisone, resulting in high overall response and complete response rates and excellent 1-year survival."( Treatment of plasma cell dyscrasias with lenalidomide.
Dimopoulos, MA; Kastritis, E; Rajkumar, SV, 2008
)
1.33
"Lenalidomide has now been approved by the US FDA and the European Medicines Agency for use in combination with dexamethasone in patients with at least one prior therapy."( Lenalidomide and its role in the management of multiple myeloma.
Boccadoro, M; Cavallo, F; Falco, P; Larocca, A; Liberati, AM; Musto, P; Palumbo, A, 2008
)
2.51
"Lenalidomide has activity in a variety of hematologic malignancies, including non-Hodgkin's lymphoma (NHL)."( Lenalidomide monotherapy in relapsed or refractory aggressive non-Hodgkin's lymphoma.
Cole, CE; Ervin-Haynes, A; Habermann, TM; Justice, G; Lam, W; Lossos, IS; McBride, K; Pietronigro, D; Takeshita, K; Tuscano, JM; Vose, JM; Wiernik, PH; Wride, K; Zeldis, JB, 2008
)
2.51
"Lenalidomide has significant activity in myelodysplastic syndromes, multiple myeloma, and non-Hodgkin's lymphoma (NHL). "( lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells.
Adams, M; Bartlett, JB; Carter, T; Chen, R; Muller, G; Schafer, P; Stirling, D; Wu, L, 2008
)
3.23
"Lenalidomide has proven efficacy and safety and has been shown to reduce transfusion requirements and reverse cytogenetic abnormalities in lower-risk myelodysplastic syndromes (MDS). "( Durable long-term responses in patients with myelodysplastic syndromes treated with lenalidomide.
Kurtin, SE; List, AF, 2009
)
2.02
"Lenalidomide has been shown to be an immunomodulator, affecting both cellular and humoral limbs of the immune system."( Mechanism of action of lenalidomide in hematological malignancies.
Das, B; Goel, S; Heuck, C; Kotla, V; Nischal, S; Verma, A; Vivek, K, 2009
)
1.38
"Lenalidomide has efficacy against melanoma in animal models and safety in phase 1 trials."( Results of a multicenter, randomized, double-blind, dose-evaluating phase 2/3 study of lenalidomide in the treatment of metastatic malignant melanoma.
Agarwala, SS; Atkins, MB; Bedikian, AY; Glaspy, J; Jungnelius, JU; Knight, RD; O'Day, S; Richards, JM, 2009
)
1.3
"Lenalidomide has shown good activity in transfusion-dependent patients with the del(5q) cytogenetic abnormality and modest activity in other lower-risk patients."( Treatment of MDS: something old, something new, something borrowed...
Sekeres, MA, 2009
)
1.07
"Lenalidomide has been shown to benefit patients with multiple myeloma, myelodysplastic syndromes, and lymphoma."( Thalidomide and lenalidomide as new therapeutics for the treatment of chronic lymphocytic leukemia.
Awan, FT; Byrd, JC; Fischer, B; Hu, W; Johnson, AJ; Lapalombella, R; Lucas, M, 2010
)
1.43
"As lenalidomide has shown to be efficacious in both myelodysplastic syndromes and myeloproliferative neoplasms, we have treated 2 RARS-T patients, who were transfusion dependent, with lenalidomide."( Efficacy of single-agent lenalidomide in patients with JAK2 (V617F) mutated refractory anemia with ring sideroblasts and thrombocytosis.
de Wolf, JT; Huls, G; Mulder, AB; Rosati, S; van de Loosdrecht, AA; Vellenga, E, 2010
)
1.18
"Lenalidomide also has meaningful clinical activity in lower-risk patients without deletion 5q."( Lenalidomide in myelodysplastic syndromes: an erythropoiesis-stimulating agent or more?
Komrokji, RS; Lancet, JE; List, AF, 2010
)
2.52
"Lenalidomide has already been approved for the management of low or intermediate-1 risk myelodysplastic syndrome with chromosome 5q31 deletion and relapsed/refractory multiple myeloma in combination with dexamethasone."( Lenalidomide: a synthetic compound with an evolving role in cancer management.
Grivas, PD; Saloura, V, 2010
)
2.52
"Lenalidomide has multiple properties, including anti-inflammatory, antiangiogenic and costimulatory effects, as well as being able to inhibit T-regulatory cells, all of which are properties deemed desirable for anticancer activity."( The potential of immunomodulatory drugs in the treatment of solid tumors.
Dalgleish, A; Galustian, C, 2010
)
1.08
"Lenalidomide has demonstrated clinical activity in myelodysplastic syndromes, particularly in patients with a deletion in the long arm of chromosome 5 (del[5q] abnormality). "( Pleiotropic mechanisms of action of lenalidomide efficacy in del(5q) myelodysplastic syndromes.
Carter, T; Chopra, R; Heise, C; Schafer, P, 2010
)
2.08
"Lenalidomide has acceptable toxicity and is associated with long-term disease stabilization and PSA declines. "( Lenalidomide in nonmetastatic biochemically relapsed prostate cancer: results of a phase I/II double-blinded, randomized study.
Antonarakis, ES; Carducci, M; Denmeade, S; Drake, C; Eisenberger, M; Hudock, S; Keizman, D; Pili, R; Sinibaldi, V; Zahurak, M, 2010
)
3.25
"HD lenalidomide has evidence of clinical activity as initial therapy for older AML patients, and further study of lenalidomide in AML and MDS is warranted."( A phase 2 study of high-dose lenalidomide as initial therapy for older patients with acute myeloid leukemia.
Abboud, CN; Cashen, AF; Demland, J; DiPersio, JF; Fehniger, TA; Nelson, AD; Stockerl-Goldstein, KE; Trinkaus, K; Uy, GL; Vij, R; Westervelt, P, 2011
)
1.17
"Lenalidomide has raised concerns regarding its potential impact on the ability to collect stem cells for autologous stem cell transplantation, especially after prolonged exposure. "( Stem cell mobilization in patients with newly diagnosed multiple myeloma after lenalidomide induction therapy.
Ben-Yehuda, D; Boccadoro, M; Bringhen, S; Cafro, AM; Calabrese, E; Caravita, T; Carella, AM; Cascavilla, N; Catalano, L; Cavallo, F; Crippa, C; Liberati, AM; Lupo, B; Milone, G; Montefusco, V; Musto, P; Nagler, A; Omedè, P; Palumbo, A; Passera, R; Patriarca, F; Peccatori, J; Petrucci, MT; Rossini, F, 2011
)
2.04
"Lenalidomide has been approved for treatment of relapsed multiple myeloma, to be delivered as a continuous therapy."( Continuous treatment in multiple myeloma - ready for prime time?
Knop, S, 2011
)
1.09
"Lenalidomide has demonstrated impressive antileukaemic effects in patients with chronic lymphocytic leukaemia (CLL). "( Biological effects and clinical significance of lenalidomide-induced tumour flare reaction in patients with chronic lymphocytic leukaemia: in vivo evidence of immune activation and antitumour response.
Bangia, N; Borrello, I; Chanan-Khan, AA; Chitta, K; Czuczman, MS; Ersing, N; Lee, K; Mashtare, TL; Masood, A; Paulus, A; Sher, T; Swaika, A; Wallace, PK; Wilding, G, 2011
)
2.07
"Lenalidomide (LEN) has been shown to yield red blood cell (RBC) transfusion independence in about 25% of lower risk myelodysplastic syndromes (MDS) without del(5q), but its efficacy in patients clearly refractory to erythropoiesis-stimulating agents (ESA) is not known. "( Lenalidomide in lower-risk myelodysplastic syndromes with karyotypes other than deletion 5q and refractory to erythropoiesis-stimulating agents.
Banos, A; Baracco, F; Besson, C; Blanc, M; Cannas, G; Corm, S; Fenaux, P; Perrier, H; Prebet, T; Sibon, D; Slama, B; Vey, N; Wattel, E, 2012
)
3.26
"Lenalidomide has shown efficacy in combination with bortezomib and dexamethasone but this combination has been poorly tolerated."( A phase 2 study of pegylated liposomal doxorubicin, bortezomib, dexamethasone and lenalidomide for patients with relapsed/refractory multiple myeloma.
Berenson, JR; Bravin, E; Cartmell, A; Chen, CS; Flam, M; Hilger, JD; Kazamel, T; Nassir, Y; Swift, RA; Vescio, R; Woliver, T; Yellin, O, 2012
)
1.33
"Lenalidomide has been shown to elicit cardiovascular effects, although its impact on cardiac function remains obscure."( Short-term lenalidomide (Revlimid) administration ameliorates cardiomyocyte contractile dysfunction in ob/ob obese mice.
Dong, M; Hua, Y; Jones, KR; Li, L; Li, Q; Ren, J; Smith, DT; Yuan, M, 2012
)
1.49
"Lenalidomide has tumoricidal and immunomodulatory activity against multiple myeloma. "( Continuous lenalidomide treatment for newly diagnosed multiple myeloma.
Beksac, M; Ben Yehuda, D; Bladé, J; Cascavilla, N; Catalano, J; Cavo, M; Corso, A; Delforge, M; Dimopoulos, MA; Gisslinger, H; Hajek, R; Herbein, L; Iosava, G; Jacques, C; Kloczko, J; Kropff, M; Langer, C; Mei, J; Palumbo, A; Petrucci, MT; Plesner, T; Radke, J; Spicka, I; Weisel, K; Wiktor-Jędrzejczak, W; Yu, Z; Zodelava, M, 2012
)
2.21
"Lenalidomide has unique activity with a high transfusion independence rate observed in this subset of patients."( Lenalidomide for treatment of myelodysplastic syndromes.
Komrokji, RS; List, AF, 2012
)
2.54
"Lenalidomide has a predominantly renal route of excretion and in patients with RI the plasma concentration and half-life of the drug are significantly increased."( Treatment with lenalidomide and dexamethasone in patients with multiple myeloma and renal impairment.
Alegre, A; Dimopoulos, MA; Goldschmidt, H; Mark, T; Niesvizky, R; Terpos, E, 2012
)
1.45
"Lenalidomide (LEN) has emerged as a promising therapeutic option for the management of various hematologic malignancies. "( Impact of lenalidomide on the functional properties of human mesenchymal stromal cells.
Benath, G; Bornhäuser, M; Ehninger, G; Ferrer, RA; Hofbauer, LC; Platzbecker, U; Rauner, M; Schmitz, M; Wehner, R; Wobus, M, 2012
)
2.22
"Lenalidomide has previously been reported to have clinical efficacy in this setting; however, long-term reports are limited."( Impressive activity of lenalidomide monotherapy in refractory angioimmunoblastic T-cell lymphoma: report of a case with long-term follow-up.
Bocchia, M; Cencini, E; Defina, M; Fabbri, A; Fontanelli, G; Gozzetti, A; Mazzei, MA; Pietrini, A; Volterrani, L, 2013
)
1.42
"Lenalidomide has been licenced for the treatment of multiple myeloma and, in 2012, it is used as a standard treatment of relapses of the disease. "( [The effect of lenalidomide on rare blood disorders: Langerhans cell histiocytosis, multicentric Castleman disease, POEMS syndrome, Erdheim-Chester disease and angiomatosis].
Adam, Z; Hájek, R; Koukalová, R; Král, Z; Krejčí, M; Mayer, J; Nebeský, T; Pour, L; Rehák, Z; Szturz, P; Zahradová, L, 2012
)
2.17
"Lenalidomide has a better toxicity profile than thalidomide."( Novel immunomodulatory compounds in multiple myeloma.
Mahindra, A; Saini, N, 2013
)
1.11
"Lenalidomide has clinical activity in AML with manageable toxicity. "( Lenalidomide as a novel treatment of acute myeloid leukemia.
Borthakur, G; Chen, Y, 2013
)
3.28
"Lenalidomide has anti-inflammatory, anti-angiogenic and immunomodulatory properties, and targets tumor cells by direct cytotoxicity and, indirectly by interfering with several components of the tumor microenvironment [1]."( Lenalidomide in lymphomas and chronic lymphocytic leukemia.
Wiernik, PH, 2013
)
2.55
"Lenalidomide has hematologic activity in patients with low-risk myelodysplastic syndromes who have no response to erythropoietin or who are unlikely to benefit from conventional therapy."( Efficacy of lenalidomide in myelodysplastic syndromes.
Buresh, A; Fuchs, D; Heaton, R; Knight, R; Kurtin, S; List, A; Mahadevan, D; Rimsza, L; Roe, DJ; Zeldis, JB, 2005
)
2.15
"Lenalidomide has received fast track designation from the FDA for the treatment of MM and myelodysplastic syndromes."( Current therapeutic uses of lenalidomide in multiple myeloma.
Anderson, KC; Hideshima, T; Richardson, PG, 2006
)
1.35
"Lenalidomide efficacy has been reported in clinical trials of multiple myeloma and myelodysplastic syndromes (MDS), particularly in MDS patients with a del 5q cytogenetic abnormality, with or without other cytogenetic abnormalities."( Lenalidomide inhibits proliferation of Namalwa CSN.70 cells and interferes with Gab1 phosphorylation and adaptor protein complex assembly.
Gandhi, AK; Kang, J; Naziruddin, S; Parton, A; Schafer, PH; Stirling, DI, 2006
)
2.5
"Lenalidomide has shown impressive activity in relapsed refractory myeloma as well as newly diagnosed disease."( Thalidomide and lenalidomide in the treatment of multiple myeloma.
Kumar, S; Rajkumar, SV, 2006
)
1.4
"Lenalidomide has different toxicities than thalidomide, exhibiting greater myelosuppression but virtually no constipation, somnolence, or peripheral neuropathy."( Advances in oral therapy in the treatment of multiple myeloma.
Doss, DS, 2006
)
1.06
"Lenalidomide has particular activity in patients with transfusion-dependent del(5q) myelodysplastic syndromes (MDS), but mechanistic information is limited regarding the relationship between erythroid and cytogenetic responses. "( Lenalidomide in the context of complex karyotype or interrupted treatment: case reviews of del(5q)MDS patients with unexpected responses.
Aul, C; Giagounidis, AA; Göhring, G; Haase, S; Heinsch, M; Schlegelberger, B, 2007
)
3.23
"Lenalidomide has emerged as an effective therapeutic alternative for the management of anemia in lower-risk myelodysplastic syndromes (MDS). "( Evolving applications of lenalidomide in the management of anemia in myelodysplastic syndromes.
List, AF, 2006
)
2.08
"Lenalidomide has been approved for the treatment of transfusion-dependent low- or intermediate-1-risk myelodysplastic syndromes (MDS) associated with a chromosome 5q deletion with or without additional cytogenetic abnormalities. "( Cost effectiveness of lenalidomide in the treatment of transfusion-dependent myelodysplastic syndromes in the United States.
Goss, TF; Hellström-Lindberg, E; Jädersten, M; Knight, R; List, AF; Schaefer, C; Szende, A; Totten, PJ, 2006
)
2.09
"Lenalidomide has been approved by the US Food and Drug Administration for the treatment of patients with myelodysplastic syndromes (MDS) with an interstitial deletion of the long arm of chromosome 5 and, more recently, in combination with dexamethasone for multiple myeloma in patients who received at least one prior therapy. "( Practical considerations in the use of lenalidomide therapy for myelodysplastic syndromes.
Kurtin, S; Sokol, L, 2006
)
2.05
"Lenalidomide has shown significant activity in refractory/resistant multiple myeloma, and further studies have shown its activity in other hematologic malignancies with some very encouraging results, especially in subsets of patients with myelodysplastic syndromes."( The evolving role of lenalidomide in the treatment of hematologic malignancies.
Dimopoulos, MA; Kastritis, E, 2007
)
1.38
"Lenalidomide has also been shown to overcome thalidomide resistance in MM patients."( Lenalidomide in myelodysplastic syndrome and multiple myeloma.
Shah, SR; Tran, TM, 2007
)
2.5
"Lenalidomide has shown significant antimyeloma activity in clinical studies. "( Melphalan, prednisone, and lenalidomide treatment for newly diagnosed myeloma: a report from the GIMEMA--Italian Multiple Myeloma Network.
Ambrosini, MT; Boccadoro, M; Bringhen, S; Ciccone, G; Corradini, P; Crippa, C; Di Raimondo, F; Falco, P; Falcone, A; Foà, R; Gay, F; Giuliani, N; Knight, R; Musto, P; Omedè, P; Palumbo, A; Petrucci, MT; Zeldis, JB, 2007
)
2.08
"Lenalidomide has clinically meaningful activity in transfusion-dependent patients with low- or int-1-risk MDS who lack the deletion 5q karyotypic abnormality."( Phase 2 study of lenalidomide in transfusion-dependent, low-risk, and intermediate-1 risk myelodysplastic syndromes with karyotypes other than deletion 5q.
Bennett, JM; Curtin, PT; Deeg, HJ; Dewald, GW; Dreisbach, L; Feldman, EJ; Greenberg, PL; Klimek, VM; Knight, RD; List, AF; Raza, A; Reeves, JA; Schiffer, CA; Schmidt, M; Shammo, JM; Stone, RM; Thomas, D; Wride, K; Zeldis, JB, 2008
)
1.41

Actions

Lenalidomide can enhance the anti-tumor effects of sorafenib in HCC through its immune modulatory effects, and CD8(+) TILs play an important role. Lenalidomides also activate T cells indirectly by inhibiting regulatory T cells and myeloid derived suppressor cells (MDSC)

ExcerptReferenceRelevance
"Lenalidomide promotes an antitumoral immune microenvironment, whereas daratumumab can potentially cause NK cell fratricide."( Novel Cell and Immune Engagers in Optimizing Tumor- Specific Immunity Post-Autologous Transplantation in Multiple Myeloma.
Arora, N; Bachanova, V; Janakiram, M; Miller, JS, 2022
)
1.44
"Lenalidomide also had a lower incidence rate of infections (68.7) versus bortezomib (95.9) and thalidomide (76.0)."( A Noninterventional, Observational, European Post-Authorization Safety Study of Patients With Relapsed/Refractory Multiple Myeloma Treated With Lenalidomide.
Andreasson, B; Bacon, P; Berthou, C; Blau, IW; Caers, J; Crotty, G; Di Micco, A; Dimopoulos, M; Gamberi, B; Hájek, R; Hernandez, M; Kueenburg, E; Minnema, MC; Parreira, J; Peters, S; Remes, K; Rosettani, B; Semenzato, G; Tholouli, E, 2020
)
1.48
"Lenalidomide is known to increase the risk of venous thromboembolism in patients with hematologic malignancies. "( Frequency of venous thrombotic events in patients with myelodysplastic syndrome and 5q deletion syndrome during lenalidomide therapy.
Al-Kali, A; Alkharabsheh, OA; Alkhateeb, HB; Begna, KH; Elliott, MA; Gangat, N; Hogan, WJ; Litzow, MR; Patnaik, MS; Saadeh, SS; Zblewski, DL, 2019
)
2.17
"Lenalidomide was able to inhibit CLL survival advantage mediated by endothelial contact."( Endothelium-mediated survival of leukemic cells and angiogenesis-related factors are affected by lenalidomide treatment in chronic lymphocytic leukemia.
Bonacorsi, G; Bulgarelli, J; Castelli, I; Cuneo, A; Debbia, G; Fiorcari, S; Forconi, F; Gaidano, G; Laurenti, L; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Palumbo, GA; Rigolin, GM; Rizzotto, L; Rossi, D; Santachiara, R; Vallisa, D, 2014
)
1.34
"Lenalidomide can enhance the anti-tumor effects of sorafenib in HCC through its immune modulatory effects, and CD8(+) TILs play an important role in the anti-tumor synergism."( Potential synergistic anti-tumor activity between lenalidomide and sorafenib in hepatocellular carcinoma.
Chang, CJ; Cheng, AL; Gandhi, AK; Hsu, C; Huang, ZM; Jeng, YM; Liao, SC; Lin, YJ; Lin, ZZ; Ou, DL, 2014
)
2.1
"Lenalidomide also activate T cells indirectly by inhibiting regulatory T cells and myeloid derived suppressor cells (MDSC) which inhibit the activation of T cells, but controversy still exist about effects on regulatory T cells."( [Immunomodulatory effects of lenalidomide].
Handa, H; Murakami, H; Saitoh, T, 2015
)
1.43
"Lenalidomide does not only promotes tumor apoptosis, but also stimulates T and NK cells, thereby facilitating NK-mediated tumor recognition and killing."( Activation of NK cells and disruption of PD-L1/PD-1 axis: two different ways for lenalidomide to block myeloma progression.
Berchem, G; Giuliani, M; Janji, B, 2017
)
1.4
"Lenalidomide plays an important role in our chemotherapeutic armamentarium against multiple myeloma, in part by exerting direct anti-proliferative and pro-apoptotic effects. "( Evidence of a role for activation of Wnt/beta-catenin signaling in the resistance of plasma cells to lenalidomide.
Bjorklund, CC; Davis, RE; Kornblau, SM; Kuhn, DJ; Ma, W; Orlowski, RZ; Shah, JJ; Wang, M; Wang, ZQ, 2011
)
2.03
"Lenalidomide also acts to enhance the immune system by inducing the activation of immune effector cells, such as T cells and natural killer cells, and inducing cytokine production."( Lenalidomide: a review of its use in the treatment of relapsed or refractory multiple myeloma.
Lyseng-Williamson, KA; Scott, LJ, 2011
)
2.53
"Lenalidomide also acts to enhance the immune system by inducing the activation of immune effector cells, such as T cells and natural killer cells, and inducing cytokine production."( Spotlight on lenalidomide in relapsed or refractory multiple myeloma.
Lyseng-Williamson, KA; Scott, LJ, 2011
)
1.46
"Lenalidomide even in lower dosed combined with steroids can induce complete responses in patients with refractory AITL."( Therapy refractory angioimmunoblastic T-cell lymphoma in complete remission with lenalidomide.
Beckers, MM; Huls, G, 2013
)
1.34
"Lenalidomide and Sorafenib inhibit HUVEC ability to migrate and form tubes and when used in combination the inhibition is increased. "( Combination therapy targeting the tumor microenvironment is effective in a model of human ocular melanoma.
Blansfield, JA; Kachala, S; Libutti, SK; Lorang, D; Mangiameli, DP; Muller, GW; Schafer, PH; Stirling, DI, 2007
)
1.78
"Lenalidomide does not produce significant sedation, constipation or neuropathy, but does lead to significant myelosuppression, unlike thalidomide."( Lenalidomide in myelodysplastic syndrome and multiple myeloma.
Shah, SR; Tran, TM, 2007
)
2.5

Treatment

Lenalidomide treatment was shown to substantially inhibit tumor-induced neo-angiogenesis rather than to exert a direct cytotoxic effect on lymphoma cells. The drug can be a good treatment option for AL amyloidosis in HIV-infected patients on antiretroviral therapy.

ExcerptReferenceRelevance
"Lenalidomide treatment of MM substantially improves patient survival, although significantly increases thrombotic risk by an unknown mechanism."( Multiple myeloma and its treatment contribute to increased platelet reactivity.
Bye, AP; Gibbins, JM; Grech, H; Khan, D; Kriek, N; Laffan, M; Mitchell, JL; Ramasamy, K; Rana, RH; Sage, T; Shapiro, S; Thakurta, A; Unsworth, AJ, 2023
)
1.63
"Lenalidomide treatment was shown to substantially inhibit tumor-induced neo-angiogenesis rather than to exert a direct cytotoxic effect on lymphoma cells."( Lenalidomide improves the therapeutic effect of an interferon-α-dendritic cell-based lymphoma vaccine.
Belardelli, F; Cox, MC; Donati, S; Lapenta, C; Lattanzi, L; Macchia, I; Santini, SM; Sestili, P; Spada, M; Spadaro, F; Urbani, F, 2019
)
2.68
"Lenalidomide-dexamethasone treatment is a common treatment regimen used in refractory multiple myeloma."( A multiple myeloma patient who developed ischemic colitis during lenalidomide treatment: A rare case report.
Altuntaş, F; Batgi, H; Dal, MS; Kızıl Çakar, M; Merdin, A; Yiğenoğlu, TN, 2020
)
1.52
"Lenalidomide-treated patients with CB had longer overall survival than those who did not (P = .01)."( Retrospective Analysis of the Clinical Use and Benefit of Lenalidomide and Thalidomide in Myelofibrosis.
Al Ali, N; Castillo-Tokumori, F; Komrokji, R; Kuykendall, AT; Lancet, J; Padron, E; Sallman, D; Sweet, K; Talati, C; Yun, S, 2020
)
1.52
"Lenalidomide treatment decreased the expression of antigens on CLL cells, which mediate the interactions with the microenvironment."( Lenalidomide abrogates the survival effect of bone marrow stromal cells in chronic lymphocytic leukemia.
Barna, G; Czeti, Á; Hernádfői, M; Kriston, C; Márk, Á; Matolcsy, A; Plander, M; Szabó, O; Szalóki, G; Takács, F, 2021
)
2.79
"Lenalidomide can be a good treatment option for AL amyloidosis in HIV-infected patients on antiretroviral therapy."( Lenalidomide as a treatment for relapsed AL amyloidosis in an HIV-positive patient.
Cook, M; Denman, J; Manavi, K, 2017
)
2.62
"Lenalidomide treatment of ABC-DLBCL cells resulted in downregulation of SPIB at the level of transcription."( Lenalidomide modulates gene expression in human ABC-DLBCL cells by regulating IKAROS interaction with an intronic control region of SPIB.
Batista, CR; DeKoter, RP; Solomon, LA, 2017
)
2.62
"Lenalidomide-based treatment showed clinical activity, with no unexpected toxicities, in patients with relapsed/refractory mantle cell lymphoma who previously failed ibrutinib therapy."( Observational study of lenalidomide in patients with mantle cell lymphoma who relapsed/progressed after or were refractory/intolerant to ibrutinib (MCL-004).
Barnett, E; Bravo, MC; Ghosh, N; Goy, A; Hamadani, M; Lossos, IS; Martin, P; Phillips, T; Reeder, CB; Rule, S; Schuster, SJ; Wang, M, 2017
)
2.21
"Lenalidomide-treated patients were more likely to achieve transfusion independence (TI) ≥ 8 weeks (26.9% vs."( The Effect of Lenalidomide on Health-Related Quality of Life in Patients With Lower-Risk Non-del(5q) Myelodysplastic Syndromes: Results From the MDS-005 Study.
Almeida, A; Altincatal, A; Beach, CL; Buckstein, R; Fenaux, P; Giagounidis, A; Guo, S; Platzbecker, U; Santini, V; Wu, C, 2018
)
1.56
"Lenalidomide doses and treatment regimens differed between trials."( Lenalidomide treatment for multiple myeloma: systematic review and meta-analysis of randomized controlled trials.
Chi, XH; Lu, XC; Yang, B; Yu, RL, 2013
)
2.55
"Lenalidomide treatment for refractory or relapsed multiple myeloma in elderly patients may be feasible in an outpatient setting. "( Very low-dose lenalidomide therapy for elderly multiple myeloma patients.
Hirata, T; Inagaki, T; Iwai, M; Katayama, Y; Kawano, H; Kimura, S; Kishi, M; Koide, T; Matsui, T; Minagawa, K; Suzuki, T; Takechi, M, 2013
)
2.19
"Lenalidomide treatment resulted in a significant reduction in the number of MCL-associated macrophages."( Lenalidomide inhibits lymphangiogenesis in preclinical models of mantle cell lymphoma.
Chen, H; Fu, J; Herzog, BH; McDaniel, JM; Ruan, J; Sheng, M; Song, K; Xia, L, 2013
)
2.55
"Lenalidomide treatment does not appear to increase AML risk in this population of patients."( Multivariate time-dependent comparison of the impact of lenalidomide in lower-risk myelodysplastic syndromes with chromosome 5q deletion.
Arilla, MJ; Arrizabalaga, B; Azaceta, G; Bailén, A; Bargay, J; Brunet, S; Cerveró, C; de Paz, R; Del Cañizo, C; Diez-Campelo, M; Falantes, J; García-Pintos, M; Lorenzo, I; Luño, E; Marco-Betes, V; Nomdedeu, B; Osorio, S; Ramos, F; Sánchez-García, J; Sanz, GF; Serrano-López, J; Such, E; Tormo, M; Valcárcel, D; Xicoy, B, 2014
)
1.37
"Lenalidomide treatment did not affect glucose clearance in lean or ob/ob mice."( Tumour necrosis factor-α inhibition with lenalidomide alleviates tissue oxidative injury and apoptosis in ob/ob obese mice.
Dong, H; Hu, N; Jiang, S; Jones, KR; Kang, YM; Luo, F; Ren, J; Xiong, L; Zhu, X; Zou, Y, 2014
)
1.39
"Lenalidomide treatment resulted in significant increases in CT/FEM-derived estimates of bone strength. "( A longitudinal computed tomography study of lenalidomide and bortezomib treatment for multiple myeloma: trabecular microarchitecture and biomechanics assessed using multidetector computed tomography.
Awai, K; Date, S; Kaichi, Y; Kiguchi, M; Kuroda, Y; Sakai, A; Sakoda, Y; Takasu, M; Tani, C, 2014
)
2.11
"Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase."( Lenalidomide induces lipid raft assembly to enhance erythropoietin receptor signaling in myelodysplastic syndrome progenitors.
Basiorka, AA; Caceres, G; Clark, J; Heaton, R; Johnson, JO; List, AF; McGraw, KL; Ozawa, Y; Padron, E; Sokol, L; Wei, S, 2014
)
2.57
"Lenalidomide pre-treatment of MM cell lines reduced TReg generation and the concomitant TReg:TEff (CD4(+)CD25(+)FoxP3(-): effector T cells) ratio, as a consequence of reduced ICOSL transcription."( Downregulation of myeloma-induced ICOS-L and regulatory T cell generation by lenalidomide and dexamethasone therapy.
Carter, C; Cook, G; Parrish, C; Scott, GB; Wood, PM, 2015
)
1.37
"Lenalidomide treatment in combination with dexamethasone and/or chemotherapy is associated with a significant risk of venous thromboembolism (VTE) in patients with multiple myeloma (MM). "( Silent venous thromboembolism in multiple myeloma patients treated with lenalidomide.
Isoda, A; Koumoto, M; Matsumoto, M; Matsumoto, Y; Miyazawa, Y; Ookawa, M; Sato, N; Sawamura, M, 2015
)
2.09
"Lenalidomide treatment alone delayed symptom onset, while nanoceria treatment had no effect on symptom onset or severity, but did promote recovery; lenalidomide and nanoceria each significantly attenuated white matter pathology and associated inflammation. "( Combination therapy with lenalidomide and nanoceria ameliorates CNS autoimmunity.
Celik, H; Chigurapati, S; Chigurupati, S; Das, S; Eitan, E; Fishbein, KW; Ghosh, P; Ghosh, S; Greig, NH; Hutchison, ER; Mattson, MP; Raymick, J; Sarkar, S; Sasaki, CY; Seal, S; Spencer, RG; Tweedie, D, 2015
)
2.16
"Lenalidomide treatment was inversely associated with SPM in the nested case-control analysis (OR = 0.03, 95%CI = 0.002-0.34)."( Subsequent primary malignancies among multiple myeloma patients treated with or without lenalidomide.
Alsina, M; Baz, R; Dalton, W; Fisher, K; Fulp, W; Hampras, S; Kenvin, L; Knight, R; Komrokji, R; Lee, JH; Nishihori, T; Olesnyckyj, M; Rollison, DE; Shain, KH; Sullivan, D; Xu, Q, 2017
)
1.4
"Lenalidomide treatment also increased the proportion of activated peripheral blood T lymphocytes."( Effect of lenalidomide therapy on hematopoiesis of patients with myelodysplastic syndrome associated with chromosome 5q deletion.
Galanopoulos, A; Giannikou, K; Hatzimichael, E; Kartasis, Z; Klaus, M; Kokoris, S; Korkolopoulou, P; Liapi, D; Papadaki, HA; Pappa, V; Parcharidou, A; Pontikoglou, C; Psyllaki, M; Sambani, C; Symeonidis, A; Ximeri, M, 2010
)
1.48
"Lenalidomide is an active treatment for multiple myeloma (MM) and is increasingly used as part of the initial treatment of this disease. "( Stem cell collection in patients with multiple myeloma: impact of induction therapy and mobilization regimen.
Cook, R; Cunningham, K; Gardler, M; Hummel, K; Luger, SM; Mangan, PA; Nazha, A; O'Doherty, U; Porter, DL; Schuster, S; Siegel, D; Stadtmauer, EA; Vogl, DT, 2011
)
1.81
"Lenalidomide (25 mg daily) treatment was then initiated in a continuous dosing schedule. "( Lenalidomide induced good clinical response in a patient with multiple relapsed and refractory Hodgkin's lymphoma.
Kolonic, SO; Mandac, I, 2010
)
3.25
"Lenalidomide-based treatment is effective across the spectrum of MM disease phases, allowing for the long-term management of myeloma."( Lenalidomide: an update on evidence from clinical trials.
Dimopoulos, MA; Terpos, E, 2010
)
2.52
"Lenalidomide-treated MMECs showed changes in VEGF/VEGFR2 signaling pathway and several proteins controlling EC motility, cytoskeleton remodeling, and energy metabolism pathways."( Lenalidomide restrains motility and overangiogenic potential of bone marrow endothelial cells in patients with active multiple myeloma.
Basile, A; Berardi, S; Caivano, A; Capalbo, S; Cascavilla, N; Coluccia, AM; Dammacco, F; de Luca, E; De Luisi, A; Di Pietro, G; Ditonno, P; Ferrucci, A; Guarini, A; Maffia, M; Moschetta, M; Pieroni, L; Quarta, G; Ranieri, G; Ria, R; Ribatti, D; Urbani, A; Vacca, A, 2011
)
2.53
"Lenalidomide-based treatment is highly effective and is an attractive treatment option in patients with multiple myeloma with impaired renal function. "( Successful treatment of patients with multiple myeloma and impaired renal function with lenalidomide: results of 4 German centers.
Hahn-Ast, C; Kuhn, S; Langer, C; Oehrlein, K; Pönisch, W; Sturm, I; Weisel, KC, 2012
)
2.04
"Lenalidomide treatment demonstrated for the first time in the literature impressive and long-term clinical efficacy in a heavily pretreated chemorefractory AITL patient."( Impressive activity of lenalidomide monotherapy in refractory angioimmunoblastic T-cell lymphoma: report of a case with long-term follow-up.
Bocchia, M; Cencini, E; Defina, M; Fabbri, A; Fontanelli, G; Gozzetti, A; Mazzei, MA; Pietrini, A; Volterrani, L, 2013
)
1.42
"Lenalidomide treatment in myelodysplastic syndrome (MDS) may lead to thrombocytopenia and dose reductions/delays. "( A randomized, double-blind, placebo-controlled phase 2 study evaluating the efficacy and safety of romiplostim treatment of patients with low or intermediate-1 risk myelodysplastic syndrome receiving lenalidomide.
Gandhi, S; Hu, K; Larson, RA; Liu, D; Lyons, RM; Matei, C; Scott, B; Wang, ES; Yang, AS, 2012
)
2.01
"Lenalidomide (LEN) treatment in multiple myeloma (MM) results in a superior outcome. "( Longitudinal bone marrow evaluations for myelodysplasia in patients with myeloma before and after treatment with lenalidomide.
Dai, L; Gollin, SM; Lentzsch, S; Mapara, MY; Monaghan, SA; Normolle, DP, 2013
)
2.04
"Lenalidomide treatment may be effective in improving HRQL outcomes."( Health-related quality of life outcomes of lenalidomide in transfusion-dependent patients with Low- or Intermediate-1-risk myelodysplastic syndromes with a chromosome 5q deletion: results from a randomized clinical trial.
Brandenburg, NA; Fenaux, P; Knight, R; Muus, P; Revicki, DA; Yu, R, 2013
)
1.37
"Lenalidomide treatment resulted in both erythroid and cytogenetic responses in the majority of patients with del(5q), accompanied by reductions in inflammatory cytokine generation and marrow microvessel density and improvement in primitive hematopoietic progenitor recovery."( Lenalidomide: targeted anemia therapy for myelodysplastic syndromes.
Baker, AF; Bellamy, W; Green, S; List, AF, 2006
)
2.5
"Treatment with lenalidomide decreases the risk of progression; however, novel triplet regimens are superior, and earlier disease may be more treatment sensitive."( Carfilzomib, Lenalidomide, and Dexamethasone Followed by Lenalidomide Maintenance for Prevention of Symptomatic Multiple Myeloma in Patients With High-risk Smoldering Myeloma: A Phase 2 Nonrandomized Controlled Trial.
Bhutani, M; Braylan, R; Calvo, KR; Choyke, P; Dew, A; Dulau-Florea, A; Emanuel, M; Figg, WD; Hill, E; Kazandjian, D; Korde, N; Kwok, M; Landgren, O; Lee, MJ; Lee, S; Lindenberg, L; Mailankody, S; Manasanch, E; Maric, I; Mena, E; Morrison, C; Patel, N; Petrosyan, A; Roschewski, M; Roswarski, J; Steinberg, SM; Stetler-Stevenson, M; Tageja, N; Trepel, JB; Turkbey, B; Wang, HW; Wang, W; Yuan, C; Zhang, Y, 2021
)
1.33
"Treatment with lenalidomide and thalidomide resulted in no effects on placental weights, fetal body weights and body measurements."( Embryo-fetal exposure and developmental outcome of lenalidomide following oral administration to pregnant cynomolgus monkeys.
Fuchs, A; Hui, JY; Kumar, G, 2022
)
1.31
"Treatment by lenalidomide and pcDNA3.1(+)/NS3 improves NK cytotoxicity up to 66.80% suggesting that lenalidomide can be used in parallel with such therapeutic vaccines as cancer vaccine or virus vaccines."( Low Dose of Lenalidomide Enhances NK Cell Activity: Possible Implication as an Adjuvant.
Bamdad, T; Borhani, K; Hashempour, T, 2017
)
1.2
"We treated by lenalidomide 55 (42 female; 13 male; median age 69) chronically transfused lower risk MDS patients with del(5q) (45) and non-del(5q) (10)."( Lenalidomide treatment in lower risk myelodysplastic syndromes-The experience of a Czech hematology center. (Positive effect of erythropoietin ± prednisone addition to lenalidomide in refractory or relapsed patients).
Belickova, M; Cmunt, E; Fuchs, O; Jonasova, A; Michalova, K; Minarik, L; Moudra, A; Neuwirtova, R; Polackova, H; Siskova, M; Stopka, T; Zemanova, Z, 2018
)
2.27
"Treatment with lenalidomide, as the final therapeutic option, resolved the intractable melena and improved both the intestinal lesions and myeloma."( Therapeutic effects of lenalidomide on hemorrhagic intestinal myeloma-associated AL amyloidosis.
Aoki, K; Arima, H; Imai, H; Ishikawa, T; Kato, A; Matsushita, A; Mori, M; Nagano, S; Ono, Y; Tabata, S; Takahashi, T; Takiuchi, Y; Yanagita, S, 2013
)
1.04
"Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21)."( Lenalidomide inhibits the proliferation of CLL cells via a cereblon/p21(WAF1/Cip1)-dependent mechanism independent of functional p53.
Bharati, IS; Cathers, B; Corral, LG; Cui, B; Fecteau, JF; Futalan, D; Gaidarova, S; Ghia, EM; Kipps, TJ; Lopez-Girona, A; Messmer, D; Schwaederlé, M, 2014
)
2.18
"Treatment with lenalidomide reduced tumor vessel density (p = 0.0001) and enhanced mature pericyte coverage of residual vessels (p = 0.002). "( Lenalidomide normalizes tumor vessels in colorectal cancer improving chemotherapy activity.
Aglietta, M; Bertotti, A; Bussolino, F; Gammaitoni, L; Giraudo, E; Giraudo, L; Grignani, G; Leone, F; Leuci, V; Luraghi, P; Maione, F; Mesiano, G; Migliardi, G; Rotolo, R; Sangiolo, D; Sassi, F; Todorovic, M; Trusolino, L, 2016
)
2.23
"Treatment with lenalidomide prevented the association of Cereblon with Rabex-5."( Rabex-5 is a lenalidomide target molecule that negatively regulates TLR-induced type 1 IFN production.
Gemechu, Y; Kishimoto, T; Millrine, D; Tei, M, 2016
)
1.14
"Treatment with lenalidomide increased RhoA activity and reversed the migration and activation defects of RASGRP1-deficient lymphocytes."( RASGRP1 deficiency causes immunodeficiency with impaired cytoskeletal dynamics.
Ban, SA; Banerjee, PP; Bennett, KL; Bilic, I; Boztug, K; Cagdas, D; Dupré, L; Garncarz, W; Gribben, JG; Hons, M; Hsu, HT; Huppa, JB; Jäger, U; Mace, EM; McClanahan, F; Mukherjee, M; Orange, JS; Petronczki, ÖY; Pfajfer, L; Pickl, WF; Platzer, R; Salzer, E; Sanal, Ö; Sinha, P; Sixt, M; Stockinger, H; Supper, V; Tezcan, I; Willmann, KL; Zlabinger, GJ, 2016
)
0.77
"Treatment with lenalidomide was associated with neutropenia, thrombocytopenia, constipation, pruritus, and fatigue."( The clinical utility of lenalidomide in multiple myeloma and myelodysplastic syndromes.
Bonkowski, J; Kolesar, JM; Vermeulen, LC, 2010
)
1.01
"Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. "( Lenalidomide modulates IL-8 and anti-prostate antibody levels in men with biochemically recurrent prostate cancer.
Antonarakis, ES; Carducci, M; Drake, CG; Eisenberger, MA; Keizman, D; McNeel, DG; Smith, HA; Thoburn, CJ; Zabransky, DJ; Zahurak, M, 2012
)
2.17
"Treatment with lenalidomide resulted in the normalization of functional T-cell subsets in responders, suggesting that lenalidomide may modulate cell-mediated immunity in patients with CLL."( Treatment with lenalidomide modulates T-cell immunophenotype and cytokine production in patients with chronic lymphocytic leukemia.
Badoux, X; Cohen, EN; Estrov, Z; Faderl, SH; Ferrajoli, A; Gao, H; Keating, MJ; Lee, BN; Reuben, JM; Wierda, WG, 2011
)
1.08
"Treatment with lenalidomide plus dexamethasone did prolong overall survival by nearly half a year in this population with end-stage myeloma."( Lenalidomide in relapsed refractory myeloma patients: impact of previous response to bortezomib and thalidomide on treatment efficacy. Results of a medical need program in Belgium.
Bries, G; Delforge, M; Demuynck, H; Depryck, B; Doyen, C; Kentos, A; Meuleman, N; Michiels, A; Mineur, PO; Offner, F; Pierre, P; Ravoet, C; Schots, R; Van de Velde, A; Van Droogenbroeck, J; Vekemans, MC; Wu, KL,
)
1.91
"Treatment with lenalidomide 10 mg and thalidomide 50 mg provided median D-dimer levels of 9.32 and the disease has remained stable for 9 months."( [Chronic disseminated intravascular coagulation (DIC) markers in a patient with multiple angiomatosis during treatment with anti-angiogenics: interferon α, thalidomide and lenalidomide].
Adam, Z; Hájek, R; Koukalová, R; Král, Z; Krejčí, M; Křikavová, L; Matýšková, M; Mayer, J; Navrátil, M; Nebeský, T; Neuman, C; Pour, L; Rehák, Z; Szturz, P; Tomíška, M, 2012
)
0.91
"Treatment with lenalidomide induced complete remission on PET-CT."( [The effect of lenalidomide on rare blood disorders: Langerhans cell histiocytosis, multicentric Castleman disease, POEMS syndrome, Erdheim-Chester disease and angiomatosis].
Adam, Z; Hájek, R; Koukalová, R; Král, Z; Krejčí, M; Mayer, J; Nebeský, T; Pour, L; Rehák, Z; Szturz, P; Zahradová, L, 2012
)
1.07
"Pretreatment with lenalidomide sensitized MM cells to SGN-40-induced cell death."( Immunomodulatory drug lenalidomide (CC-5013, IMiD3) augments anti-CD40 SGN-40-induced cytotoxicity in human multiple myeloma: clinical implications.
Anderson, KC; Bae, J; Breitkreutz, I; Catley, L; Chauhan, D; Coffey, R; Grewal, IS; Hideshima, T; Li, XF; Munshi, NC; Podar, K; Richardson, P; Schlossman, R; Song, W; Tai, YT; Treon, SP, 2005
)
0.97
"Treatment with lenalidomide was recently shown to be effective in MDS, particularly in those cases with del(5q), resulting in durable cytogenetic remission and hematological responses."( Evaluation of recurring cytogenetic abnormalities in the treatment of myelodysplastic syndromes.
Le Beau, MM; Olney, HJ, 2007
)
0.68
"Treatment with lenalidomide was associated with an OR rate of 31% in patients with 11q or 17p deletion, of 24% in patients with unmutated V(H), and of 25% in patients with fludarabine-refractory disease."( Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia.
Cohen, EN; Estrov, Z; Faderl, S; Ferrajoli, A; Gao, H; Keating, MJ; Lee, BN; Li, C; O'Brien, SM; Reuben, JM; Schlette, EJ; Wen, S; Wierda, WG, 2008
)
2.13

Toxicity

Lenalidomide administered at 25 mg/d in relapsed CLL is associated with unacceptable toxicity. The rapid onset and adverse clinical effects of tumor flare represent a significant limitation of lenalidomid use in CLL at this dose.

ExcerptReferenceRelevance
" In all, 87% of adverse effects were classified as grade 1 or grade 2 according to Common Toxicity Criteria and there were no serious adverse events attributable to CC-5013 treatment."( Phase I study to determine the safety, tolerability and immunostimulatory activity of thalidomide analogue CC-5013 in patients with metastatic malignant melanoma and other advanced cancers.
Bartlett, JB; Clarke, IA; Dalgleish, AG; Dredge, K; Kristeleit, H; Michael, A; Muller, GW; Nicholson, S; Pandha, H; Polychronis, A; Stirling, DI; Zeldis, J, 2004
)
0.32
" Their toxic profile is favorable, but during the drug development process some severe (sometimes lethal) toxicities have been observed, such as interstitial lung disease in patients treated with drugs targeting the epidermal growth factor receptor."( Side effects of anti-cancer molecular-targeted therapies (not monoclonal antibodies).
Awada, A; de Castro, G, 2006
)
0.33
"To examine dermatologic adverse effects of lenalidomide in patients with amyloidosis and multiple myeloma and to determine whether the adverse effects are different when lenalidomide is used alone compared with when it is used in combination with dexamethasone."( Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma.
Davis, MD; Dispenzieri, A; Rajkumar, SV; Sviggum, HP, 2006
)
0.88
"The prevalence of dermatologic adverse effects in patients receiving lenalidomide was higher in those with amyloidosis than in those with multiple myeloma."( Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma.
Davis, MD; Dispenzieri, A; Rajkumar, SV; Sviggum, HP, 2006
)
0.85
" The majority of adverse events were grades 1-2, including fatigue (25/80 cycles), nausea/vomiting (23/80), constipation (13/80), abdominal pain (17/80), rash (12/80), neutropenia (12/80), and anemia (12/80)."( Safety and efficacy of lenalidomide (Revlimid) in recurrent ovarian and primary peritoneal carcinoma.
Chan, JK; Guo, HY; Husain, A; Teng, NN; Zhang, MM, 2007
)
0.65
"In all studies, thalidomide was selectively toxic to development."( Evaluation of the developmental toxicity of lenalidomide in rabbits.
Christian, MS; Hoberman, A; Laskin, OL; Latriano, L; Sharper, V; Stirling, DI, 2007
)
0.6
" Unlike thalidomide, lenalidomide affected embryo-fetal development only at maternally toxic dosages, confirming that structure-activity relationships may not predict maternal or developmental effects."( Evaluation of the developmental toxicity of lenalidomide in rabbits.
Christian, MS; Hoberman, A; Laskin, OL; Latriano, L; Sharper, V; Stirling, DI, 2007
)
0.92
" Serious adverse events including tumor flare and tumor lysis are summarized."( Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia.
Andritsos, LA; Awan, F; Blum, W; Byrd, JC; Chen, CS; Jarjoura, D; Johnson, AJ; Kefauver, C; Knight, RD; Lapalombella, R; Lehman, A; Lozanski, G; May, SE; Raymond, CA; Ruppert, AS; Smith, LL; Wang, DS, 2008
)
0.69
"Four consecutive patients were treated with lenalidomide; all had serious adverse events."( Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia.
Andritsos, LA; Awan, F; Blum, W; Byrd, JC; Chen, CS; Jarjoura, D; Johnson, AJ; Kefauver, C; Knight, RD; Lapalombella, R; Lehman, A; Lozanski, G; May, SE; Raymond, CA; Ruppert, AS; Smith, LL; Wang, DS, 2008
)
0.95
"Lenalidomide administered at 25 mg/d in relapsed CLL is associated with unacceptable toxicity; the rapid onset and adverse clinical effects of tumor flare represent a significant limitation of lenalidomide use in CLL at this dose."( Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia.
Andritsos, LA; Awan, F; Blum, W; Byrd, JC; Chen, CS; Jarjoura, D; Johnson, AJ; Kefauver, C; Knight, RD; Lapalombella, R; Lehman, A; Lozanski, G; May, SE; Raymond, CA; Ruppert, AS; Smith, LL; Wang, DS, 2008
)
2.13
" The most common grade 3 or 4 adverse events were neutropenia (60%), thrombocytopenia (39%), and anemia (20%), which proved manageable with dose reduction."( Safety and efficacy of single-agent lenalidomide in patients with relapsed and refractory multiple myeloma.
Anderson, KC; Badros, AZ; Bensinger, W; Berenson, J; Hussein, M; Irwin, D; Jagannath, S; Kenvin, L; Knight, R; Olesnyckyj, M; Richardson, P; Singhal, S; Vescio, R; Williams, SF; Yu, Z; Zeldis, J, 2009
)
0.63
" The most common adverse events (AEs) were haematological (49%), gastrointestinal (59%), and fatigue (55%)."( Expanded safety experience with lenalidomide plus dexamethasone in relapsed or refractory multiple myeloma.
Abonour, R; Alsina, M; Bahlis, NJ; Boccia, RV; Chen, C; Coutre, SE; Knight, RD; Kumar, S; Matous, J; Niesvizky, R; Pietronigro, D; Rajkumar, V; Reece, DE; Richardson, P; Siegel, D; Stadtmauer, EA; Vescio, R; Zeldis, JB, 2009
)
0.64
" The major adverse events associated with these treatments are somnolence (thalidomide), venous thromboembolism (thalidomide and lenalidomide), myelosuppression (lenalidomide and bortezomib), gastrointestinal disturbance, and peripheral neuropathy (thalidomide and bortezomib)."( Management of treatment-related adverse events in patients with multiple myeloma.
Mateos, MV, 2010
)
0.57
"The results from this study indicated that, with careful monitoring of the CLCr level and adverse events as well as appropriate dose adjustments, lenalidomide plus dexamethasone is an effective and well tolerated treatment option for patients with multiple myeloma who have RI."( The efficacy and safety of lenalidomide plus dexamethasone in relapsed and/or refractory multiple myeloma patients with impaired renal function.
Alegre, A; de Castro, CM; Dimopoulos, M; Goldschmidt, H; Masliak, Z; Olesnyckyj, M; Reece, D; Stadtmauer, EA; Weber, DM; Yu, Z; Zonder, JA, 2010
)
0.86
" Three patients experienced a grade 4 adverse reaction; two pulmonary emboli and one cerebral ischemia."( Phase I safety study of lenalidomide and dacarbazine in patients with metastatic melanoma previously untreated with systemic chemotherapy.
Bassett, R; Bedikian, A; Hwu, P; Hwu, WJ; Kim, KB; Knight, RD; Papadopoulos, NE; Patnana, M, 2010
)
0.67
" On the other hand, when new drugs are used it is very important to know their associated toxicity, since adequate management of the adverse effects can help to avoid unnecessary treatment interruptions - thereby undoubtedly contributing to improvement in the efficacy of therapy."( Management of the adverse effects of lenalidomide in multiple myeloma.
González Rodríguez, AP, 2011
)
0.64
" The major adverse events (AEs) associated with lenalidomide include: hematological toxicities (myelosuppression), mainly neutropenia, venous thromboembolism, gastrointestinal disturbance, skin toxicity, atrial fibrillation, asthenia, and decreased peripheral blood stem cell yield during stem cell collection when lenalidomide is used after a long period of time."( Lenalidomide treatment for patients with multiple myeloma: diagnosis and management of most frequent adverse events.
García Sánchez, PJ; González Rodríguez, AP; Mesa, MG; Pérez Persona, E, 2011
)
2.07
" However, these benefits are accompanied by increases in treatment-related adverse events (AEs), which may be particularly pronounced in older individuals."( Practical management of adverse events in multiple myeloma: can therapy be attenuated in older patients?
Bringhen, S; Mateos, MV; Palumbo, A; San Miguel, JF, 2011
)
0.37
" Febrile neutropenia was rare (4%) and there were no toxic deaths."( Lenalidomide can be safely combined with R-CHOP (R2CHOP) in the initial chemotherapy for aggressive B-cell lymphomas: phase I study.
Ansell, SM; Habermann, TM; Inwards, DJ; Johnston, PB; Klebig, RR; LaPlant, B; Macon, WR; Micallef, IN; Nowakowski, GS; Porrata, LF; Reeder, CB; Rivera, CE; Witzig, TE, 2011
)
1.81
" Common adverse events included fatigue, injection site reactions, constipation, nausea, pruritus and febrile neutropenia."( Safety, efficacy and biological predictors of response to sequential azacitidine and lenalidomide for elderly patients with acute myeloid leukemia.
Abdel-Wahab, O; Berube, C; Bhattacharya, S; Coutre, S; Figueroa, ME; Gallegos, L; Gotlib, JR; Kohrt, HE; Levine, R; Liedtke, M; Medeiros, BC; Melnick, A; Mitchell, BS; Pollyea, DA; Zehnder, J; Zhang, B, 2012
)
0.6
" Most adverse events occur early during the course of treatment and are manageable."( The clinical safety of lenalidomide in multiple myeloma and myelodysplastic syndromes.
Fenaux, P; Freeman, J; Palumbo, A; Weiss, L, 2012
)
0.69
" Serum ferritin (SF) was measured monthly, and safety assessment included monitoring of adverse events during treatment and of liver and renal parameters."( Deferasirox treatment for myelodysplastic syndromes: "real-life" efficacy and safety in a single-institution patient population.
Alimena, G; Breccia, M; Colafigli, G; Federico, V; Finsinger, P; Latagliata, R; Loglisci, G; Petrucci, L; Salaroli, A; Santopietro, M; Serrao, A, 2012
)
0.38
" These agents have specific adverse event (AE) profiles, and it is especially important to consider severe AEs that may lead to premature discontinuation, negatively affecting outcomes."( How to maintain patients on long-term therapy: understanding the profile and kinetics of adverse events.
Mateos, MV, 2012
)
0.38
" There were two serious treatment-related adverse events during the treatment period (cerebrovascular accident, placebo; worsening thrombocytopenia, romiplostim 500 μg)."( A randomized, double-blind, placebo-controlled phase 2 study evaluating the efficacy and safety of romiplostim treatment of patients with low or intermediate-1 risk myelodysplastic syndrome receiving lenalidomide.
Gandhi, S; Hu, K; Larson, RA; Liu, D; Lyons, RM; Matei, C; Scott, B; Wang, ES; Yang, AS, 2012
)
0.57
"Lenalidomide appears to be efficacious and safe in patients with refractory CLE, but clinical relapse is frequent after its withdrawal."( Efficacy and safety of lenalidomide for refractory cutaneous lupus erythematosus.
Ávila, G; Cortés-Hernández, J; Ordi-Ros, J; Vilardell-Tarrés, M, 2012
)
2.13
"Cutaneous toxicity is a frequent side effect of new anticancer targeted therapies."( Cutaneous adverse reactions linked to targeted anticancer therapies bortezomib and lenalidomide for multiple myeloma: new drugs, old side effects.
Brandi, G; Dika, E; Maibach, H; Patrizi, A; Tacchetti, P; Venturi, M, 2014
)
0.63
"This study evaluates the impact of cutaneous adverse drug reactions (cADR) of the new therapies bortezomib and lenalidomide and presents a review of their skin side effects."( Cutaneous adverse reactions linked to targeted anticancer therapies bortezomib and lenalidomide for multiple myeloma: new drugs, old side effects.
Brandi, G; Dika, E; Maibach, H; Patrizi, A; Tacchetti, P; Venturi, M, 2014
)
0.84
" Three adverse events linked to bortezomib and 4 to lenalidomide forced to a complete withdrawal of the drug, while 3 reactions due to bortezomib mandated a dose reduction."( Cutaneous adverse reactions linked to targeted anticancer therapies bortezomib and lenalidomide for multiple myeloma: new drugs, old side effects.
Brandi, G; Dika, E; Maibach, H; Patrizi, A; Tacchetti, P; Venturi, M, 2014
)
0.88
" Our study suggests that cutaneous toxicities, when researched by Dermatologists, are a side effect even more frequent than the reported data."( Cutaneous adverse reactions linked to targeted anticancer therapies bortezomib and lenalidomide for multiple myeloma: new drugs, old side effects.
Brandi, G; Dika, E; Maibach, H; Patrizi, A; Tacchetti, P; Venturi, M, 2014
)
0.63
" Low-dose lenalidomide therapy was effective and safe against MM with a bortezomib-associated lung disorder."( Safety and effectiveness of low-dose lenalidomide therapy for multiple myeloma complicated with bortezomib-associated interstitial pneumonia.
Ihara, K; Inomata, H; Kato, J; Kikuchi, S; Koyama, R; Muramatsu, H; Nagamachi, Y; Nishisato, T; Nozawa, E; Okamoto, T; Ono, K; Tanaka, S; Yamada, H; Yamauchi, N; Yano, T, 2013
)
1.07
"Patients received oral lenalidomide 25mg once daily on days 1-21 of each 28-day cycle for a maximum of 24 months, until disease progression or development of unacceptable adverse events (AEs)."( A phase 2, multicentre, single-arm, open-label study to evaluate the safety and efficacy of single-agent lenalidomide (Revlimid) in subjects with relapsed or refractory peripheral T-cell non-Hodgkin lymphoma: the EXPECT trial.
Bosly, A; Coiffier, B; Delarue, R; Fitoussi, O; Gabarre, J; Glaisner, S; Haioun, C; Li, J; Lister, J; Morschhauser, F; Quach, H; Thieblemont, C, 2013
)
0.91
" Central hypothyroidism is a well-recognized side effect of bexarotene."( Thyroid dysfunction as an unintended side effect of anticancer drugs.
Appetecchia, M; Baldelli, R; Barnabei, A; Corsello, SM; Paragliola, R; Torino, F, 2013
)
0.39
" The most common adverse events observed were neutropenia (56%), thrombocytopenia (50%), and anemia (40%)."( [Effectiveness and safety of lenalidomide in myelofibrosis patients: a case series from the Spanish compassionate use program].
Asensio, A; Blanes, M; Boqué, C; Castillo, I; Hermosilla, MM; Ojea, MA,
)
0.42
" Grade 3-4 hematologic adverse events were: neutropenia in 28% of the courses, thrombocytopenia in 9%, and anemia in 3%."( Lenalidomide plus cyclophosphamide, doxorubicin, vincristine, prednisone and rituximab is safe and effective in untreated, elderly patients with diffuse large B-cell lymphoma: a phase I study by the Fondazione Italiana Linfomi.
Baldi, I; Bottelli, C; Carella, AM; Castellino, A; Chiappella, A; Ciccone, G; De Masi, P; Gaidano, G; Ladetto, M; Liberati, AM; Orsucci, L; Palumbo, A; Pavone, V; Perticone, S; Rossi, G; Rossini, B; Salvi, F; Tucci, A; Vitolo, U; Zanni, M, 2013
)
1.83
" Adverse events were reported in 68."( "Real-world" data on the efficacy and safety of lenalidomide and dexamethasone in patients with relapsed/refractory multiple myeloma who were treated according to the standard clinical practice: a study of the Greek Myeloma Study Group.
Anagnostopoulos, N; Anargyrou, K; Briasoulis, E; Giannakoulas, N; Hatzimichael, E; Karras, G; Katodritou, E; Kotsopoulou, M; Kyriakou, D; Kyrtsonis, MC; Lalagianni, C; Maniatis, A; Matsouka, P; Michali, E; Papageorgiou, G; Spanoudakis, E; Symeonidis, A; Terpos, E; Tsakiridou, A; Tsionos, K; Vadikolia, C; Zikos, P, 2014
)
0.66
"The median age of the patients was 58 years, with 30% of the patients aged >65 years, 49% having an International Staging System stage of 2 and 3, 12% having severe renal insufficiency, and 8% demonstrating an adverse result on fluorescence in situ hybridization."( Efficacy and safety profile of long-term exposure to lenalidomide in patients with recurrent multiple myeloma.
Avet Loiseau, H; Bonnet, S; Debarri, H; Demarquette, H; Facon, T; Fouquet, G; Gay, J; Guidez, S; Herbaux, C; Hulin, C; Leleu, X; Michel, J; Miljkovic, D; Perrot, A; Serrier, C; Tardy, S, 2013
)
0.64
" Adverse events were manageable and mostly included thrombocytopenia and neutropenia."( Efficacy and safety of lenalinomide combined with rituximab in patients with relapsed/refractory diffuse large B-cell lymphoma.
Aurran-Schleinitz, T; Blaise, D; Bouabdallah, R; Broussais-Guillaumot, F; Chetaille, B; Coso, D; Esterni, B; Ivanov, V; Olive, D; Schiano, JM; Stoppa, AM, 2014
)
0.4
" In conclusion, low-dose lenalidomide plus dexamethasone therapy is an effective and safe regimen for patients with relapsed or refractory POEMS syndrome."( Efficacy and safety of low-dose lenalidomide plus dexamethasone in patients with relapsed or refractory POEMS syndrome.
Cai, H; Cai, QQ; Cao, XX; Li, J; Wang, C; Zhou, DB, 2015
)
1
" The most common grade 3 or 4 adverse events were neutropenia (38 [35%] of 110 patients), muscle pain (ten [9%]), rash (eight [7%]), cough, dyspnoea, or other pulmonary symptoms (five [5%]), fatigue (five [5%]), thrombosis (five [5%]), and thrombocytopenia (four [4%])."( Safety and activity of lenalidomide and rituximab in untreated indolent lymphoma: an open-label, phase 2 trial.
Baladandayuthapani, V; Claret, LC; Davis, RE; Fanale, MA; Fayad, LE; Feng, L; Fowler, NH; Hagemeister, FB; Kwak, LW; McLaughlin, P; Muzzafar, T; Nastoupil, L; Neelapu, SS; Oki, Y; Orlowski, RZ; Rawal, S; Romaguera, JE; Samaniego, F; Shah, J; Tsai, KY; Turturro, F; Wang, M; Westin, JR, 2014
)
0.71
" Four dose-limiting toxic events were noted in phase 1: one at a dose of ixazomib of 2·97 mg/m(2) and three at 3·95 mg/m(2)."( Safety and tolerability of ixazomib, an oral proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma: an open-label phase 1/2 study.
Berdeja, JG; Berg, D; Di Bacco, A; Estevam, J; Gupta, N; Hamadani, M; Hari, P; Hui, AM; Kaufman, JL; Kumar, SK; Laubach, JP; Liao, E; Lonial, S; Niesvizky, R; Rajkumar, V; Richardson, PG; Roy, V; Stewart, AK; Vescio, R, 2014
)
0.63
" The outcomes included overall response (OR) rate, complete response (CR) rate, 3-year progression-free survival (PFS) rate, 3-year overall survival (OS) rate, and different types of treatment-related adverse events."( Efficacy and Safety of Lenalidomide in the Treatment of Multiple Myeloma: A Systematic Review and Meta-analysis of Randomized Controlled Trials.
Guo, XN; Qiao, SK; Ren, HY; Ren, JH, 2015
)
0.73
" The most frequent adverse events ≥ grade 3 at 15 mg were 14% anemia and 43% neutropenia."( Lenalidomide is safe and active in Waldenström macroglobulinemia.
Arnulf, B; Bakala, J; Banos, A; Bories, C; Brice, P; Choquet, S; Demarquette, H; Dib, M; Fouquet, G; Guidez, S; Herbaux, C; Karlin, L; Leblond, V; LeGouill, S; Leleu, X; Louni, C; Martin, A; Morel, P; Nudel, M; Ohyba, B; Petillon, MO; Poulain, S; Salles, G; Thielemans, B; Tournilhac, O, 2015
)
1.86
" Twenty-one (49%) patients had at least one drug-related grade ≥3 adverse event (AE); the most common were neutropenia (19%), diarrhea (14%), and thrombocytopenia (12%)."( Pharmacokinetics and safety of ixazomib plus lenalidomide-dexamethasone in Asian patients with relapsed/refractory myeloma: a phase 1 study.
Chim, CS; Chng, WJ; Esseltine, DL; Goh, YT; Gupta, N; Hanley, MJ; Hui, AM; Kim, K; Lee, JH; Min, CK; Venkatakrishnan, K; Wong, RS; Yang, H, 2015
)
0.68
"0%) and lack of 3/4 grade adverse events (R(2)=1."( Efficacy and safety of lenalidomide treatment in multiple myeloma (MM) patients--Report of the Polish Myeloma Group.
Becht, R; Bołkun, Ł; Butrym, A; Błońska, D; Charliński, G; Dębski, J; Dmoszyńska, A; Druzd-Sitek, A; Dytfeld, D; Hałka, J; Hołojda, J; Hus, M; Januszczyk, J; Jurczyszyn, A; Knopińska-Posłuszny, W; Kuliczkowski, K; Kłoczko, J; Lech-Marańda, E; Legieć, W; Malenda, A; Nowicki, A; Pogrzeba, J; Rymko, M; Rzepecki, P; Stella-Hołowiecka, B; Subocz, E; Torosian, T; Urbanowicz, A; Urbańska-Ryś, H; Usnarska-Zubkiewicz, L; Zaucha, JM; Zdziarska, B; Zubkiewicz-Kucharska, A, 2016
)
0.74
"LEN is an effective and safe therapeutic option, even in intensively treated resistant and relapsed MM patients, as well as in patients with stable disease and previous treatment-induced neurological complications."( Efficacy and safety of lenalidomide treatment in multiple myeloma (MM) patients--Report of the Polish Myeloma Group.
Becht, R; Bołkun, Ł; Butrym, A; Błońska, D; Charliński, G; Dębski, J; Dmoszyńska, A; Druzd-Sitek, A; Dytfeld, D; Hałka, J; Hołojda, J; Hus, M; Januszczyk, J; Jurczyszyn, A; Knopińska-Posłuszny, W; Kuliczkowski, K; Kłoczko, J; Lech-Marańda, E; Legieć, W; Malenda, A; Nowicki, A; Pogrzeba, J; Rymko, M; Rzepecki, P; Stella-Hołowiecka, B; Subocz, E; Torosian, T; Urbanowicz, A; Urbańska-Ryś, H; Usnarska-Zubkiewicz, L; Zaucha, JM; Zdziarska, B; Zubkiewicz-Kucharska, A, 2016
)
0.74
" The most common grade ≥3 adverse events (AEs) were neutropenia and thrombocytopenia."( Safety and efficacy of different lenalidomide starting doses in patients with relapsed or refractory chronic lymphocytic leukemia: results of an international multicenter double-blinded randomized phase II trial.
Buhler, A; Chanan-Khan, A; De Bedout, S; Dürig, J; Fraser, GA; Gribben, JG; Hallek, M; Hillmen, P; Kalaycio, M; Kipps, TJ; Mei, J; Michallet, AS; Purse, B; Stilgenbauer, S; Wendtner, CM; Zhang, J, 2016
)
0.72
" All patients had ≥ 1 adverse event (AE)."( Pharmacokinetics and Safety of Elotuzumab Combined With Lenalidomide and Dexamethasone in Patients With Multiple Myeloma and Various Levels of Renal Impairment: Results of a Phase Ib Study.
Badros, A; Berdeja, J; Bleickardt, E; Gupta, M; Jagannath, S; Kaufman, JL; Lynch, M; Manges, R; Paliwal, P; Tendolkar, A; Vij, R; Zonder, J, 2016
)
0.68
" Complete response (CR), progression-free survival (PFS), overall survival (OS), and adverse events (AE) were combined."( Efficacy and Safety of Novel Agent-Based Therapies for Multiple Myeloma: A Meta-Analysis.
Li, Y; Wang, X; Yan, X, 2016
)
0.43
" LenDex was interrupted in three cases because of adverse events (infections and cutaneous events); 78 % of the patients were on thromboprophylaxis with aspirin."( Lenalidomide is effective and safe for the treatment of patients with relapsed multiple myeloma and very severe renal impairment.
Coelho, I; Esteves, GV; Esteves, S; Freitas, J; Geraldes, C; Gomes, F; João, C; Lúcio, P; Neves, M, 2016
)
1.88
" All four children completed the protocol without any significant adverse effects and remain in complete and durable remission 15-18 months posttreatment."( Excellent remission rates with limited toxicity in relapsed/refractory Langerhans cell histiocytosis with pulse dexamethasone and lenalidomide in children.
Bakane, A; Raj, R; Ramachandrakurup, S; Subburaj, D; Uppuluri, R, 2017
)
0.66
" Safety was evaluated based on the occurrence rates of grades 3-4 adverse events (AEs)."( Efficacy and Safety of Lenalidomide for Treatment of Low-/Intermediate-1-Risk Myelodysplastic Syndromes with or without 5q Deletion: A Systematic Review and Meta-Analysis.
Deng, ZQ; He, PF; Lian, XY; Lin, J; Qian, J; Wen, XM; Xu, ZJ; Yang, L; Yao, DM; Zhang, ZH, 2016
)
0.74
" We demonstrated that CK1α inactivation, while toxic for myeloma cells, is dispensable for the survival of healthy B lymphocytes and stromal cells."( Inactivation of CK1α in multiple myeloma empowers drug cytotoxicity by affecting AKT and β-catenin survival signaling pathways.
Barilà, G; Bonaldi, L; Cabrelle, A; Carrino, M; Costacurta, M; Gianesin, K; Macaccaro, P; Manni, S; Manzoni, M; Martines, A; Neri, A; Nunes, SC; Piazza, F; Semenzato, G; Taiana, E; Trentin, L; Tubi, LQ; Zambello, R, 2017
)
0.46
" The main concerns on adverse events were thrombosis/embolism events, peripheral neuropathy, and second primary malignancies."( Efficacy and safety of bortezomib, thalidomide, and lenalidomide in multiple myeloma: An overview of systematic reviews with meta-analyses.
Aguiar, PM; Colleoni, GWB; de Mendonça Lima, T; Storpirtis, S, 2017
)
0.71
" Safety data from TOURMALINE-MM1 are reviewed and guidance for managing clinically relevant adverse events associated with IRd is provided."( Management of adverse events associated with ixazomib plus lenalidomide/dexamethasone in relapsed/refractory multiple myeloma.
Avivi, I; Berg, D; Einsele, H; Esseltine, DL; Gupta, N; Hájek, R; Hari, P; Kumar, S; Liberati, AM; Lin, J; Lonial, S; Ludwig, H; Masszi, T; Mateos, MV; Minnema, MC; Moreau, P; Richardson, PG; Romeril, K; Shustik, C; Spencer, A, 2017
)
0.7
" There were no vaccine-related adverse effects."( Revaccination after Autologous Hematopoietic Stem Cell Transplantation Is Safe and Effective in Patients with Multiple Myeloma Receiving Lenalidomide Maintenance.
Chung, DJ; Copelan, O; Devlin, SM; Giralt, SA; Hassoun, H; Kenny, S; Koehne, G; Korde, NS; Landau, H; Landgren, CO; Lendvai, N; Lesokhin, AM; Mailankody, S; Maloy, M; Palazzo, M; Perales, MA; Seier, K; Shah, GL, 2018
)
0.68
" Compared with placebo, lenalidomide was associated with a higher incidence of grade 3-4 treatment-emergent adverse events (TEAEs; 86% vs."( Safety profile of lenalidomide in patients with lower-risk myelodysplastic syndromes without del(5q): results of a phase 3 trial.
Almeida, A; Beach, CL; Castaneda, C; Fenaux, P; Garcia-Manero, G; Goldberg, SL; Gröpper, S; Jonasova, A; Santini, V; Vey, N; Zhong, J, 2018
)
1.12
" One hundred thirty-seven multiple myeloma patients on maintenance lenalidomide or bortezomib post-auto-HCT who received either MMR or herpes zoster vaccine were analyzed and any adverse events documented in the medical record in the 42 days following vaccination were recorded."( Safety of live-attenuated measles-mumps-rubella and herpes zoster vaccination in multiple myeloma patients on maintenance lenalidomide or bortezomib after autologous hematopoietic cell transplantation.
Baden, LR; Hammond, SP; Issa, NC; Laubach, JP; Leblebjian, H; Marty, FM; Pandit, A; Richardson, PG, 2018
)
0.92
" Carfilzomib (CFZ) has high selectivity and minimal off-target adverse effects including lower rates of PNP."( Efficacy and toxicity profile of carfilzomib based regimens for treatment of multiple myeloma: A systematic review.
Abraham, I; Anwer, F; Iftikhar, A; Kapoor, V; Latif, A; McBride, A; Mushtaq, A; Riaz, IB; Zahid, U, 2018
)
0.48
" Adverse events (AEs) occurred in 69 (57."( Efficacy and safety of lenalidomide and dexamethasone in patients with relapsed/\ refractory multiple myeloma: a real-life experience
Duran, M; Durusoy, R; Patır, P; Şahin, F; Saydam, G; Soyer, N; Töbü, M; Tombuloğlu, M; Ünal, HD; Uysal, A; Vural, F, 2018
)
0.79
"Overall, these results suggest that the vaccine is safe and immunogenic in this patient population and support continued study of PVX-410 in SMM."( Assessment of Safety and Immunogenicity of PVX-410 Vaccine With or Without Lenalidomide in Patients With Smoldering Multiple Myeloma: A Nonrandomized Clinical Trial.
Bae, J; Kaufman, JL; Nooka, AK; Peterkin, D; Raje, NS; Richardson, PG; Wang, ML; Yee, AJ, 2018
)
0.71
" The overall incidence of adverse events (AEs) was similar in IRd and placebo-Rd groups."( [Safety and management of adverse events of ixazomib/lenalidomide/dexamethasone therapy in Japanese patients with relapsed/refractory multiple myeloma].
Aotsuka, N; Berg, D; Handa, H; Iida, S; Ishida, T; Izumi, T; Kase, Y; Komeno, T; Soeda, J; Sunami, K,
)
0.38
" The adverse events were usually mild, none leading to permanent drug interruptions."( Real World Efficacy and Safety Results of Ixazomib Lenalidomide and Dexamethasone Combination in Relapsed/Refractory Multiple Myeloma: Data Collected from the Hungarian Ixazomib Named Patient Program.
Alizadeh, H; Bátai, Á; Deák, B; Demeter, J; Illés, Á; Kosztolányi, S; Mikala, G; Nagy, Z; Pető, M; Plander, M; Schneider, T; Szendrei, T; Szomor, Á; Varga, G; Váróczy, L, 2019
)
0.77
" Common adverse events included hematological toxicities including neutropenia and lymphopenia; however, the rate of febrile neutropenia was low (3."( Evaluation of the safety and efficacy of daratumumab outside of clinical trials.
Abe, Y; Kitadate, A; Kobayashi, H; Matsue, K; Miura, D; Narita, K; Takeuchi, M; Terao, T; Tsushima, T, 2019
)
0.51
" Twenty-six (65%) patients experimented adverse events: 8 patients (20%) underwent 12 serious AE (≥grade 3)."( Efficacy and safety of autologous stem cell transplantation after induction therapy with lenalidomide, bortezomib, and dexamethasone.
Arcani, R; Azouza, W; Brunet, C; Colle, J; Costello, R; Fanciullino, R; Farnault, L; Ivanov, V; Lafage, M; Mercier, C; Pourroy, B; Roche, P; Suchon, P; Venton, G, 2019
)
0.74
" There were 10 fatal infections, and the most frequent adverse events were mild infusion-associated reactions and hematologic toxicities."( Real-world data on the efficacy and safety of daratumumab treatment in Hungarian relapsed/refractory multiple myeloma patients.
Adamkovich, N; Alizadeh, H; Bereczki, Á; Borbényi, Z; Csacsovszki, O; Csukly, Z; Egyed, M; Farkas, P; Illés, Á; Lovas, S; Masszi, T; Mikala, G; Nagy, Z; Plander, M; Rajnics, P; Rejtő, L; Schneider, T; Szaleczky, E; Szendrei, T; Szomor, Á; Varga, G; Váróczy, L; Wohner, N; Wolf, K, 2019
)
0.51
" We retrieved evidence on treatment-related grade III/IV adverse events, progression-free survival (PFS) and overall survival (OS)."( A systematic literature review and network meta-analysis of effectiveness and safety outcomes in advanced melanoma.
Franken, MG; Gheorghe, M; Haanen, JBAG; Leeneman, B; Uyl-de Groot, CA; van Baal, PHM, 2019
)
0.51
" Outcomes assessed included overall response rate (ORR), complete response (CR) rate, overall survival (OS), progression-free survival (PFS), and adverse events."( Matching-adjusted Indirect Comparisons of the Efficacy and Safety of Acalabrutinib Versus Other Targeted Therapies in Relapsed/Refractory Mantle Cell Lymphoma.
Abhyankar, S; Kabadi, SM; Signorovitch, J; Song, J; Telford, C; Yao, Z; Zhao, J, 2019
)
0.51
" This subpopulation analysis assessed the impact of lenalidomide maintenance and treatment-emergent adverse events (TEAEs) on health-related quality of life (HRQOL)."( Lenalidomide maintenance for diffuse large B-cell lymphoma patients responding to R-CHOP: quality of life, dosing, and safety results from the randomised controlled REMARC study.
Abraham, J; Bouabdallah, R; Bron, D; Caballero, D; Casadebaig, ML; Casasnovas, RO; Catalano, J; Cohen, AM; Coiffier, B; Costello, R; da Silva, MG; Daguindau, N; Deeren, D; Eisenmann, JC; Fitoussi, O; Fruchart, C; Gaulard, P; Gonzalez, H; Greil, R; Grosicki, S; Howlett, S; Le Du, K; Lionne-Huyghe, P; Morschhauser, F; Mounier, N; Nicolas-Virezelier, E; Obéric, L; Perrot, A; Pica, GM; Roulin, L; Snauwaert, S; Takeshita, K; Thieblemont, C; Tilly, H; Tricot, S; Trotman, J; van Eygen, K, 2020
)
2.25
" However, some patients discontinue VRd because of severe adverse events, despite its high efficacy."( The efficacy and safety of modified bortezomib-lenalidomide-dexamethasone in transplant-eligible patients with newly diagnosed multiple myeloma.
Abe, Y; Ishida, T; Miyazaki, K; Nashimoto, J; Ogura, M; Okazuka, K; Sato, K; Suzuki, K; Tsukada, N; Uto, Y; Yoshiki, Y, 2020
)
0.82
" Adverse events were acceptable."( The effectiveness and safety of lenalidomide and dexamethasone in patients with relapsed/refractory multiple myeloma in real-world clinical practice: a study of the Korean Multiple Myeloma Working Party (KMMWP-151 study).
Bang, SM; Cho, SH; Do, YR; Eom, HS; Jo, JC; Kim, HJ; Kim, J; Kim, JS; Kim, K; Kim, MK; Kim, SH; Lee, HS; Lee, JH; Lee, JJ; Lee, MH; Lee, WS; Lee, Y; Lim, SN; Min, CK; Mun, YC; Park, SK; Park, Y; Shin, HJ; Yi, JH; Yoon, DH; Yoon, SS, 2020
)
0.84
" In this systematic review, associations of the efficacy of each approved regimen with adverse events (AEs) and the total cost per cycle were compared with a Bayesian network meta-analysis (NMA) of phase 3 randomized controlled trials (RCTs)."( Association of adverse events and associated cost with efficacy for approved relapsed and/or refractory multiple myeloma regimens: A Bayesian network meta-analysis of phase 3 randomized controlled trials.
Aryal, M; Chhabra, S; D'Souza, A; Dhakal, B; Ghose, S; Giri, S; Hamadani, M; Hari, PN; Janz, S; Narra, RK; Pathak, LK; Smunt, TL; Szabo, A, 2020
)
0.56
" Fourteen patients (9%) discontinued IRd due to adverse events/toxicity in the absence of disease progression."( Real-world effectiveness and safety of ixazomib-lenalidomide-dexamethasone in relapsed/refractory multiple myeloma.
Aitchison, R; Dimopoulos, MA; Gavriatopoulou, M; Hajek, R; Jelinek, T; Jenner, MW; Kastritis, E; Katodritou, E; Kothari, J; Kotsopoulou, M; Maouche, N; Minarik, J; Ntanasis-Stathopoulos, I; Pika, T; Plonkova, H; Ramasamy, K; Terpos, E; Vallance, GD; Zomas, A, 2020
)
0.81
" Adverse events from these agents might affect the ability of patients to tolerate treatment over time."( Longitudinal Toxicity over Time (ToxT) analysis to evaluate tolerability: a case study of lenalidomide in the CALGB 50401 (Alliance) trial.
Atherton, PJ; Bartlett, NL; Blum, KA; Dueck, AC; Flowers, CR; Habermann, TM; Jung, SH; Leonard, JP; Novotny, PJ; Pitcher, BN; Tan, A; Thanarajasingam, G; Witzig, TE, 2020
)
0.78
" Adverse events were analyzed by incidence rates per 100 person-years to account for differences in observation length and treatment duration."( A Noninterventional, Observational, European Post-Authorization Safety Study of Patients With Relapsed/Refractory Multiple Myeloma Treated With Lenalidomide.
Andreasson, B; Bacon, P; Berthou, C; Blau, IW; Caers, J; Crotty, G; Di Micco, A; Dimopoulos, M; Gamberi, B; Hájek, R; Hernandez, M; Kueenburg, E; Minnema, MC; Parreira, J; Peters, S; Remes, K; Rosettani, B; Semenzato, G; Tholouli, E, 2020
)
0.76
" 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
)
0.82
"The incidence of grade 3 or more haematological and grade 2 or more non-haematological adverse events was lower in the dose-escalation group than in the standard-dose group, and only that of diarrhoea was significantly lower."( Safety and Effectiveness of Ixazomib Dose-escalating Strategy in Ixazomib-Lenalidomide-Dexamethasone Treatment for Relapsed/Refractory Multiple Myeloma.
Boku, S; Higai, K; Kagoo, T; Niijima, D; Ogawa, C; Ohashi, Y; Tani, K; Ueno, H; Yachi, Y; Yano, T; Yatabe, M; Yokoyama, A,
)
0.36
"A dose-escalation strategy to optimise ixazomib dosing may reduce treatment interruption due to adverse events without compromising its antitumor activity."( Safety and Effectiveness of Ixazomib Dose-escalating Strategy in Ixazomib-Lenalidomide-Dexamethasone Treatment for Relapsed/Refractory Multiple Myeloma.
Boku, S; Higai, K; Kagoo, T; Niijima, D; Ogawa, C; Ohashi, Y; Tani, K; Ueno, H; Yachi, Y; Yano, T; Yatabe, M; Yokoyama, A,
)
0.36
" In routine practice, few patients received ixazomib-based induction therapy due to reasons including (1) patients' preference on oral regimens, (2) concerns on adverse events (AEs) of other intravenous/subcutaneous regimens, (3) requirements for less center visits, and (4) fears of COVID-19 and other infectious disease exposures."( Ixazomib-based frontline therapy in patients with newly diagnosed multiple myeloma in real-life practice showed comparable efficacy and safety profile with those reported in clinical trial: a multi-center study.
Bao, L; Chen, B; Ding, K; Fu, C; Hua, L; Huang, Y; Li, B; Li, J; Liu, P; Luo, J; Wang, L; Wang, S; Wang, W; Wu, G; Xia, Z; Xu, T; Yang, W; Yang, Y; Zhang, W; Zhou, X, 2020
)
0.56
" Despite the longer treatment duration, fewer patients with ≥CR versus VGPR/PR experienced treatment-emergent adverse events that led to discontinuation of carfilzomib-based treatment in ASPIRE or ENDEAVOR."( Efficacy and safety profile of deep responders to carfilzomib-based therapy: a subgroup analysis from ASPIRE and ENDEAVOR.
Cook, G; DeCosta, L; Delforge, M; Desgraz, R; Gay, F; Mateos, MV; Moreau, P; Szabo, Z; Weisel, K, 2021
)
0.62
" Main grade 3-4 adverse events were neutropenia (21%), infections (11%), and hypertension (6%)."( A real-world efficacy and safety analysis of combined carfilzomib, lenalidomide, and dexamethasone (KRd) in relapsed/refractory multiple myeloma.
Antonioli, E; Barilà, G; Bonalumi, A; Buda, G; Cavo, M; Cea, M; di Giovanni Bezzi, C; Dozza, L; Furlan, A; Mancuso, K; Martello, M; Marzocchi, G; Pantani, L; Petrini, M; Quaresima, M; Rizzello, I; Rocchi, S; Scalese, M; Staderini, M; Tacchetti, P; Zamagni, E, 2021
)
0.86
" It is available in original and generic forms in Turkey, but there is no clinical study that has compared the effectiveness and adverse events (AEs) of the generic and original forms of lenalidomide."( Original Versus Generic Lenalidomide in Patients with Relapsed/Refractory Multiple Myeloma: Comparison of Efficacy and Adverse Events
Bolaman, AZ; Eroğlu Küçükerdiler, H; Ertop, Ş; Sahip, B; Sargın, G; Selim, C; Turgutkaya, A; Yavaşoğlu, İ, 2021
)
1.12
" Most common (≥2 patients) grade 3 or 4 adverse events were neutropenia (12 patients [27%]), rash (4 patients [9%]), lung infection (3 patients [7%]), and increased alanine aminotransferase level (2 patients [4%])."( Safety and Effectiveness of Weekly Carfilzomib, Lenalidomide, Dexamethasone, and Daratumumab Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma: The MANHATTAN Nonrandomized Clinical Trial.
Arcila, ME; Caple, J; Chansakul, A; Chung, DJ; Ciardello, A; Concepcion, I; Derkach, A; Diamond, B; Dogan, A; Giralt, SA; Harrison, A; Hassoun, H; Ho, C; Hultcrantz, M; Jones, K; Korde, N; Lahoud, OB; Landau, HJ; Landgren, O; Lesokhin, AM; Lu, SX; Maclaughlan, K; Mailankody, S; Maura, F; Murata, K; Piacentini, C; Ramanathan, L; Rispoli, J; Roshal, M; Rustad, EH; Salcedo, M; Sams, A; Schlossman, J; Scordo, M; Shah, G; Shah, UA; Shekarkhand, T; Tan, C; Tavitian, E; Thoren, K; Verducci, D; Werner, K; Yellapantula, V, 2021
)
0.88
" Through meta-analysis of adverse reactions after taking lenalidomide, 35."( The efficacy and safety of lenalidomide in the treatment of multiple myeloma patients after allo-hematopoietic stem-cell transplantation: a systematic review and meta-analysis.
Liu, M; Zhang, X; Zhong, J, 2021
)
1.16
"Lenalidomide is effective in the treatment of MM patients after allo-HSCT, and reducing the incidence of infection and peripheral neuropathy, but it is not effective in reducing GVHD and blood system adverse reactions."( The efficacy and safety of lenalidomide in the treatment of multiple myeloma patients after allo-hematopoietic stem-cell transplantation: a systematic review and meta-analysis.
Liu, M; Zhang, X; Zhong, J, 2021
)
2.36
" The most common grade 3/4 adverse events included neutropenia, leukopenia, thrombocytopenia, lung infections, diarrhea, and maculopapular rash."( Safety and Efficacy of Combination Maintenance Therapy with Ixazomib and Lenalidomide in Patients with Posttransplant Myeloma.
Bashir, Q; Feng, L; Huo, XJ; Lee, HC; Manasanch, EM; Morphey, A; Olsem, J; Orlowski, RZ; Patel, KK; Qazilbash, MH; Shah, JJ; Thomas, SK; Weber, DM, 2022
)
0.95
"The addition of ixazomib to lenalidomide maintenance demonstrated a better than expected PFS compared with historical data using lenalidomide alone and was safe and tolerable."( Safety and Efficacy of Combination Maintenance Therapy with Ixazomib and Lenalidomide in Patients with Posttransplant Myeloma.
Bashir, Q; Feng, L; Huo, XJ; Lee, HC; Manasanch, EM; Morphey, A; Olsem, J; Orlowski, RZ; Patel, KK; Qazilbash, MH; Shah, JJ; Thomas, SK; Weber, DM, 2022
)
1.25
" Among participants undergoing treatment with lenalidomide for multiple myeloma, Black patients were underrepresented and the adverse event of skin hyperpigmentation was underreported."( Underrepresentation of Black participants and adverse events in clinical trials of lenalidomide for myeloma.
Blevins, F; Hughes, D; Lerner, A; Li, KY; Mann, M; Milrod, CJ; Patel, P; Sanchorawala, V; Sloan, JM, 2022
)
1.2
" The most frequent adverse events were neutropenia, fatigue, leukopenia, and diarrhea."( AMC-070: Lenalidomide Is Safe and Effective in HIV-Associated Kaposi Sarcoma.
Aboulafia, D; Ambinder, RF; Baiocchi, R; Bui, JD; Chiao, EY; Dittmer, DP; Han, S; Martínez-Maza, O; Maurer, T; Mitsuyasu, R; Moore, P; Reid, EG; Shimabukuro, K; Wachsman, W, 2022
)
1.14
" The most common adverse events were manageable."( AMC-070: Lenalidomide Is Safe and Effective in HIV-Associated Kaposi Sarcoma.
Aboulafia, D; Ambinder, RF; Baiocchi, R; Bui, JD; Chiao, EY; Dittmer, DP; Han, S; Martínez-Maza, O; Maurer, T; Mitsuyasu, R; Moore, P; Reid, EG; Shimabukuro, K; Wachsman, W, 2022
)
1.14
" Treatment continued until disease progression or intolerable adverse events (AEs)."( Efficacy and safety of pomalidomide and low-dose dexamethasone in Chinese patients with relapsed or refractory multiple myeloma: a multicenter, prospective, single-arm, phase 2 trial.
Bai, H; Fang, BJ; Fu, WJ; Liao, AJ; Lu, J; Niu, T; Wang, YF; Zhao, HG, 2022
)
0.72
" The most common grade 3 and 4 treatment-emergent adverse events (TEAEs) were neutropenia (63."( Efficacy and safety of pomalidomide and low-dose dexamethasone in Chinese patients with relapsed or refractory multiple myeloma: a multicenter, prospective, single-arm, phase 2 trial.
Bai, H; Fang, BJ; Fu, WJ; Liao, AJ; Lu, J; Niu, T; Wang, YF; Zhao, HG, 2022
)
0.72
"Pomalidomide in combination with low-dose dexamethasone is effective and safe in Chinese RRMM patients."( Efficacy and safety of pomalidomide and low-dose dexamethasone in Chinese patients with relapsed or refractory multiple myeloma: a multicenter, prospective, single-arm, phase 2 trial.
Bai, H; Fang, BJ; Fu, WJ; Liao, AJ; Lu, J; Niu, T; Wang, YF; Zhao, HG, 2022
)
0.72
" Patients who received continuous dosing appeared to have a higher incidence of adverse events when compared to intermittent dosing with the most common adverse events being neutropenia, fatigue, and rash."( Safety and tolerability of lenalidomide maintenance dosing in patients with multiple myeloma post-autologous stem cell transplant.
Davis, JA; Gaffney, KJ; Hashmi, H; Shockley, A; Smith, D; Weeda, E, 2022
)
1.02
"1% had grade 3-4 or higher drug-related adverse reactions, mainly hematological toxicity, and no patients died of adverse reactions."( Efficacy and safety analysis of bortezomib-based triplet regimens sequential lenalidomide in newly diagnosed multiple myeloma patients.
Liu, Y; Su, J; Wen, J; Xu, F; Yue, J; Zhang, Y; Zhou, Q, 2023
)
1.14
" We aimed to comprehensively evaluate the risk of adverse events associated with carfilzomib in Korean patients with RRMM."( Carfilzomib's Real-World Safety Outcomes in Korea: Target Trial Emulation Study Using Electronic Health Records.
Jang, HY; Kim, CJ; Kim, IW; Lee, HK; Oh, JM; Yoon, SS, 2022
)
0.72
" The lenalidomide group had a significant incidence of grade ≥ 3 hematological adverse events (AEs) involving neutropenia (RR = 1."( Efficacy and safety of lenalidomide in diffuse large B-cell lymphoma: a meta-analysis of randomized controlled trials.
Chen, L; Guo, X; Liang, T; Liu, J; Mi, R; Yin, Q, 2023
)
1.73
" Non-hematologic adverse events (AEs) were more common than hematologic AEs."( Real-world toxicity and effectiveness of ixazomib, lenalidomide, and dexamethasone in Korean patients with relapsed and/or refractory multiple myeloma.
Jo, JC; Kim, HR; Kim, K; Kim, SH; Lee, JH; Lee, JJ; Lee, JY; Min, CK; Moon, JH; Shin, HJ, 2023
)
1.16
" The most common adverse events with LND are myelosuppression, thrombosis and pneumonia."( Evaluation of Lung Toxicity With Lenalidomide Using the Pharmacovigilance Database.
Eren, T; Murata, S; Sato, J; Shimizu, T; Uchida, M, 2022
)
1
" Data on lung adverse drug reactions (ADRs) were analyzed, and safety signals were estimated using reported odds ratios (RORs) and 95% confidence intervals (CIs)."( Evaluation of Lung Toxicity With Lenalidomide Using the Pharmacovigilance Database.
Eren, T; Murata, S; Sato, J; Shimizu, T; Uchida, M, 2022
)
1
" None of the patients that were treated with desensitization had severe immune-mediated or non-dermatological adverse reactions."( Desensitization protocol to lenalidomide: An effective and safe treatment modality for delayed hypersensitivity-induced rash in patients with multiple myeloma.
Aumann, S; Gatt, ME; Lebel, E; Parnasa, E; Ribak, Y; Rubin, L; Saban, R; Shamriz, O; Tal, Y; Talmon, A; Vainstein, V, 2023
)
1.2
"Desensitization to lenalidomide is safe and effective."( Desensitization protocol to lenalidomide: An effective and safe treatment modality for delayed hypersensitivity-induced rash in patients with multiple myeloma.
Aumann, S; Gatt, ME; Lebel, E; Parnasa, E; Ribak, Y; Rubin, L; Saban, R; Shamriz, O; Tal, Y; Talmon, A; Vainstein, V, 2023
)
1.53
"In conclusion, replacing Revlimid® with its generic version Rivelime® in singlet, doublet or triplet lenalidomide containing RRMM regimens seems not to compromise the efficacy of treatment, and to yield a similarly improved response rates and survival outcome and no additional toxic effects, enabling a long-term therapy."( Generic Lenalidomide Rivelime Versus Brand-name Revlimid® in the Treatment of Relapsed/Refractory Multiple Myeloma: A Retrospective Single-center Experience on Efficacy, Safety and Survival Outcome.
Beksac, M; Gurman, G; Ilhan, O; Koyun, D; Seval, GC; Topcuoglu, P; Yuksel, MK, 2023
)
1.56
" Lenalidomide-related serious adverse events were not observed."( Efficacy and safety of lenalidomide in HIV-associated cryptococcal meningitis patients with persistent intracranial inflammation: an open-label, single-arm, prospective interventional study.
Guo, Y; Huang, Y; Hui, J; Liu, X; Peng, X; Tao, R; Wan, Z; Zhu, B; Zhu, X, 2023
)
2.13
"Lenalidomide could improve persistent intracranial inflammation in HIV-CM patients significantly and was well tolerated without serious adverse events observed."( Efficacy and safety of lenalidomide in HIV-associated cryptococcal meningitis patients with persistent intracranial inflammation: an open-label, single-arm, prospective interventional study.
Guo, Y; Huang, Y; Hui, J; Liu, X; Peng, X; Tao, R; Wan, Z; Zhu, B; Zhu, X, 2023
)
2.66
" The risk of cardiac adverse events (CAEs) with PIs has been documented with bortezomib and carfilzomib; however, only a few studies have been reported on ixazomib."( Cardiac Adverse Events Associated with Multiple Myeloma Patients Treated with Proteasome Inhibitors.
Endo, M; Fujiwara, M; Goto, M; Shimizu, T; Uchida, M, 2023
)
0.91
"This study aimed to determine the safety signals of adverse events related to CAEs, the effect of concomitant medications, the time to the occurrence of CAEs, and the incidence of fatal clinical outcomes after the occurrence of CAEs for three PIs using the US Pharmacovigilance database."( Cardiac Adverse Events Associated with Multiple Myeloma Patients Treated with Proteasome Inhibitors.
Endo, M; Fujiwara, M; Goto, M; Shimizu, T; Uchida, M, 2023
)
0.91
"We examined 1,567,240 cases of 231 drugs registered as anticancer drugs in the US Food and Drug Administration Adverse Event Reporting System (FAERS) database from January 1997 to March 2021."( Cardiac Adverse Events Associated with Multiple Myeloma Patients Treated with Proteasome Inhibitors.
Endo, M; Fujiwara, M; Goto, M; Shimizu, T; Uchida, M, 2023
)
0.91
" However, no adverse event CAE signals were observed with ixazomib treatment."( Cardiac Adverse Events Associated with Multiple Myeloma Patients Treated with Proteasome Inhibitors.
Endo, M; Fujiwara, M; Goto, M; Shimizu, T; Uchida, M, 2023
)
0.91
" The most common adverse events were neutropenia and thrombocytopenia."( Adding venetoclax to lenalidomide and rituximab is safe and effective in patients with untreated mantle cell lymphoma.
Bond, D; Boonstra, P; Carty, SA; Danilov, AV; Herrera, AF; Kaminski, MS; Kandarpa, M; Karimi, YH; Kump, K; Maddocks, K; Mayer, TL; Nachar, V; Nikolovska-Coleska, Z; Phillips, TJ; Popplewell, L; Takiar, R; Wilcox, RA, 2023
)
1.23
" Patients' treatment responses, overall response rate, progression-free survival rate, and adverse events were recorded."( Real-world data on the effectiveness and safety of Ixazomib-Lenalidomide-Dexamethasone therapy in relapsed/refractory multiple myeloma patients: a multicenter experience in Turkey.
Albayrak, M; Altuntas, F; Bakırtaş, M; Başcı, S; Basturk, A; Çakar, MK; Dal, MS; Dogu, MH; Ekinci, O; Eser, B; Giden, AO; Gulturk, E; Hacıbekiroglu, T; Kalpakci, Y; Korkmaz, S; Miskioglu, M; Ozatli, D; Serin, I; Ulas, T; Yiğenoğlu, TN, 2023
)
1.15
" Frequent (≥10% incidence) grade ≥3 treatment emergent adverse events were decreased neutrophil and platelet counts (n=7 [16%] each)."( Efficacy and Safety of Ixazomib Plus Lenalidomide and Dexamethasone Following Injectable PI-Based Therapy in Relapsed/Refractory Multiple Myeloma.
Abe, Y; Chou, T; Handa, H; Ito, S; Mori, I; Sasaki, M; Shinozaki, T; Suzuki, K; Takezako, N; Yoshida, T, 2023
)
1.18

Pharmacokinetics

This study aimed to develop a population pharmacokinetic model for lenalidomide in multiple cancers, including CLL. The objective of this study was to evaluate the pharmacokinetics and pharmacodynamic drug interactions.

ExcerptReferenceRelevance
" The differences in pharmacokinetic parameters between normal renal function and mild renal impairment were minor to modest (11%-32%)."( Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis.
Chen, N; Knight, R; Kong, L; Kumar, G; Laskin, OL; Lau, H; Zeldis, JB, 2007
)
0.66
" Pharmacokinetic parameters were estimated using noncompartmental methods."( Plasma and cerebrospinal fluid pharmacokinetics of thalidomide and lenalidomide in nonhuman primates.
Berg, SL; Dauser, RC; Gibson, BW; McGuffey, LH; Muscal, JA; Nuchtern, JG; Sun, Y, 2012
)
0.62
"9 mL/min/kg, the median plasma AUC was 80 μM h, and the median terminal half-life (t(½)) was 13."( Plasma and cerebrospinal fluid pharmacokinetics of thalidomide and lenalidomide in nonhuman primates.
Berg, SL; Dauser, RC; Gibson, BW; McGuffey, LH; Muscal, JA; Nuchtern, JG; Sun, Y, 2012
)
0.62
" The terminal elimination half-life (t (½)) was 3-4 h at doses up to 50 mg and was not affected by food."( Single-dose pharmacokinetics of lenalidomide in healthy volunteers: dose proportionality, food effect, and racial sensitivity.
Chen, N; Kasserra, C; Lau, H; Liu, L; Reyes, J, 2012
)
0.66
" Murine pharmacokinetic characterization necessary for translational and further preclinical investigations has not been published."( Pharmacokinetics and tissue disposition of lenalidomide in mice.
Byrd, JC; Gao, Y; Herman, SE; Jarjoura, D; Johnson, AJ; Li, X; Mahoney, E; Phelps, MA; Rozewski, DM; Shin, JD; Stefanovski, MR; Towns, WH; Yang, X, 2012
)
0.64
"This article describes, for the first time, a highly sensitive and specific enzyme-linked immunosorbent assay (ELISA) for therapeutic monitoring and pharmacokinetic studies of lenalidomide (LND), the potent drug for treatment of multiple myeloma (MM)."( A highly sensitive polyclonal antibody-based ELISA for therapeutic monitoring and pharmacokinetic studies of lenalidomide.
Abuhejail, RM; Alzoman, NZ; Darwish, IA, 2014
)
0.81
" The analytical method was applied to support a pharmacokinetic study of simultaneous estimation of lenalidomide, ibrutinib, and its active metabolite PCI-45227 in Wistar rat."( Simultaneous quantification of lenalidomide, ibrutinib and its active metabolite PCI-45227 in rat plasma by LC-MS/MS: application to a pharmacokinetic study.
Govindarajulu, B; Rangasamy, M; Thappali, S; Vakkalanka, S; Veeraraghavan, S; Viswanadha, S, 2015
)
0.92
" The objective of this study was to evaluate the pharmacokinetic and pharmacodynamic drug interactions between lenalidomide and warfarin in healthy volunteers."( Evaluation of pharmacokinetic and pharmacodynamic interactions when lenalidomide is co-administered with warfarin in a randomized clinical trial setting.
Chen, N; Knight, R; Palmisano, M; Weiss, D; Zhou, S, 2015
)
0.86
"The 90 % confidence intervals (CI) for the ratio of AUC or Cmax geometric means between co-administration with lenalidomide and placebo were within the 80-125 % bioequivalence bounds for R-warfarin and S-warfarin."( Evaluation of pharmacokinetic and pharmacodynamic interactions when lenalidomide is co-administered with warfarin in a randomized clinical trial setting.
Chen, N; Knight, R; Palmisano, M; Weiss, D; Zhou, S, 2015
)
0.86
" This study was conducted to investigate the pharmacokinetic and safety profiles of ixazomib, administered with lenalidomide-dexamethasone, in East Asian patients with relapsed/refractory MM."( Pharmacokinetics and safety of ixazomib plus lenalidomide-dexamethasone in Asian patients with relapsed/refractory myeloma: a phase 1 study.
Chim, CS; Chng, WJ; Esseltine, DL; Goh, YT; Gupta, N; Hanley, MJ; Hui, AM; Kim, K; Lee, JH; Min, CK; Venkatakrishnan, K; Wong, RS; Yang, H, 2015
)
0.89
"A wide linearity range analytical method for the determination of lenalidomide in patients with multiple myeloma for pharmacokinetic studies is required."( A Wide Linearity Range Method for the Determination of Lenalidomide in Plasma by High-Performance Liquid Chromatography: Application to Pharmacokinetic Studies.
Climente-Martí, M; Guglieri-López, B; Martinez-Gómez, MA; Merino-Sanjuán, M; Pérez-Pitarch, A; Porta-Oltra, B, 2016
)
0.92
" A set of blood samples was taken in order to develop a pharmacokinetic model accounting for lenalidomide concentrations in this setting."( Pharmacokinetics of lenalidomide during high cut-off dialysis in a patient with multiple myeloma and renal failure.
Buclin, T; Burnier, M; Dao, K; Figg, WD; Kissling, S; Lu, Y; Peer, CJ; Stadelmann, R, 2017
)
1
" The aim of this study was to conduct a population pharmacokinetic analysis of lenalidomide in multiple myeloma patients to identify and evaluate non-studied covariates that could be used for dose individualization."( Population pharmacokinetics of lenalidomide in multiple myeloma patients.
Climente-Martí, M; Guchelaar, HJ; Guglieri-López, B; Merino-Sanjuán, M; Moes, DJ; Pérez-Pitarch, A; Porta-Oltra, B, 2017
)
0.97
" Nonlinear mixed-effects modeling was used to develop a population pharmacokinetic model and perform covariate analysis."( Population pharmacokinetics of lenalidomide in multiple myeloma patients.
Climente-Martí, M; Guchelaar, HJ; Guglieri-López, B; Merino-Sanjuán, M; Moes, DJ; Pérez-Pitarch, A; Porta-Oltra, B, 2017
)
0.74
" In addition, the developed population pharmacokinetic model can be used to individualize lenalidomide dose in multiple myeloma patients, taking into account not only creatinine clearance but also body surface area."( Population pharmacokinetics of lenalidomide in multiple myeloma patients.
Climente-Martí, M; Guchelaar, HJ; Guglieri-López, B; Merino-Sanjuán, M; Moes, DJ; Pérez-Pitarch, A; Porta-Oltra, B, 2017
)
0.96
"A population pharmacokinetic (PopPK) model of lenalidomide was developed using data pooled from 13 clinical studies (dose range, 5-400 mg) in participants who were considered to have adequate capability for renal excretion of lenalidomide (creatinine clearance [CrCl] > 50 mL/min)."( Population Pharmacokinetics of Lenalidomide in Healthy Volunteers and Patients With Hematologic Malignancies.
Chen, N; Connarn, JN; Gao, Y; Hwang, R; Palmisano, M, 2018
)
1.03
" Simulations of human plasma concentrations of lenalidomide were achieved with simplified physiologically-based pharmacokinetic models in control and humanized-liver mice or by the direct fitting analysis of reported human data, in accordance with reported lenalidomide concentrations after low dose administration in humans."( Association of pharmacokinetic profiles of lenalidomide in human plasma simulated using pharmacokinetic data in humanized-liver mice with liver toxicity detected by human serum albumin RNA.
Guengerich, FP; Kamiya, Y; Kusama, T; Mitsui, M; Murayama, N; Shimizu, M; Suemizu, H; Uehara, S; Yamazaki, H, 2018
)
1
" Population pharmacokinetic analyses were conducted to determine the pharmacokinetics of intravenous daratumumab in combination therapy versus monotherapy, evaluate the effect of patient- and disease-related covariates on drug disposition, and examine the relationships between daratumumab exposure and efficacy/safety outcomes."( Pharmacokinetics and Exposure-Response Analyses of Daratumumab in Combination Therapy Regimens for Patients with Multiple Myeloma.
Belch, A; Capra, M; Clemens, PL; Dimopoulos, MA; Gomez, D; Ho, PJ; Iida, S; Jansson, R; Leiba, M; Medvedova, E; Min, CK; Schecter, J; Sonneveld, P; Sun, YN; Xu, XS; Zhang, L, 2018
)
0.48
" This study aimed to develop a population pharmacokinetic model for lenalidomide in multiple cancers, including CLL, to identify any disease-related differences in disposition."( Population pharmacokinetics of lenalidomide in patients with B-cell malignancies.
Blum, KA; Blum, W; Byrd, JC; Foster, DJR; Gao, Y; Grever, MR; Hofmeister, CC; Hughes, JH; Marcucci, G; Phelps, MA; Reuter, SE; Upton, RN, 2019
)
1.04
" A population pharmacokinetic model was developed with NONMEM and patient demographics were tested as covariates."( Population pharmacokinetics of lenalidomide in patients with B-cell malignancies.
Blum, KA; Blum, W; Byrd, JC; Foster, DJR; Gao, Y; Grever, MR; Hofmeister, CC; Hughes, JH; Marcucci, G; Phelps, MA; Reuter, SE; Upton, RN, 2019
)
0.8
"This is the first report of a lenalidomide population pharmacokinetic model to evaluate lenalidomide pharmacokinetics in patients with CLL and compare its pharmacokinetics with other B-cell malignancies."( Population pharmacokinetics of lenalidomide in patients with B-cell malignancies.
Blum, KA; Blum, W; Byrd, JC; Foster, DJR; Gao, Y; Grever, MR; Hofmeister, CC; Hughes, JH; Marcucci, G; Phelps, MA; Reuter, SE; Upton, RN, 2019
)
1.09
"A physiologically based pharmacokinetic model was developed using the Open Systems Pharmacology Suite to explore the pharmacokinetics of lenalidomide in a variety of tissues."( Development of a physiologically based pharmacokinetic model for intravenous lenalidomide in mice.
Foster, DJR; Hughes, JH; Phelps, MA; Reuter, SE; Rozewski, DM; Upton, RN, 2019
)
0.95
" Additionally, a population pharmacokinetic (PK) analysis and simulations were used to compare the PK profiles of the split first dose regimen with the recommended single first dose regimens of daratumumab in previously approved indications."( Split First Dose Administration of Intravenous Daratumumab for the Treatment of Multiple Myeloma (MM): Clinical and Population Pharmacokinetic Analyses.
Bladé, J; Clemens, PL; Deraedt, W; Jakubowiak, A; Krishnan, A; Lonial, S; Luo, M; Moreau, P; Nnane, I; Oriol, A; Qi, M; Sun, YN; Ukropec, J; Usmani, SZ; Xu, XS; Zhou, H, 2020
)
0.56
" 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
)
0.82
" Pharmacokinetic parameters were assessed, and adverse events (AEs) were monitored throughout."( Pharmacokinetic and bioequivalence of lenalidomide in multiple myeloma patients.
Feng, P; Gao, T; Liu, X; Shen, Q; Xiang, J; Zheng, L, 2023
)
1.18
"The two formulations of lenalidomide 25 mg displayed similar pharmacokinetic profiles and were bioequivalent."( Pharmacokinetic and bioequivalence of lenalidomide in multiple myeloma patients.
Feng, P; Gao, T; Liu, X; Shen, Q; Xiang, J; Zheng, L, 2023
)
1.49

Compound-Compound Interactions

This study showed that blockade of the CXCR4-CXCL12 axis by ulocuplumab is safe with acceptable AEs and leads to a high response rate. We demonstrated that lenalidomide can be safely combined with R-CHOP.

ExcerptReferenceRelevance
" This article provides the clinical rationale for the use of PLD in combination with immunomodulatory drugs to treat patients with MM and summarizes the clinical experience with these combinations to date."( Pegylated liposomal doxorubicin and immunomodulatory drug combinations in multiple myeloma: rationale and clinical experience.
Chanan-Khan, AA; Lee, K, 2007
)
0.34
" Lenalidomide in combination with dexamethasone has been approved by the United States Food and Drug Administration and the European Medicines Agency for the treatment of patients with MM who have received at least one prior therapy."( Lenalidomide in combination with dexamethasone for the treatment of relapsed or refractory multiple myeloma.
Attal, M; Dimopoulos, M; Harousseau, JL; Hussein, M; Knop, S; Ludwig, H; Palumbo, A; San Miguel, J; Sonneveld, P; von Lilienfeld-Toal, M, 2009
)
2.71
" Food and Drug Administration (FDA) on June 29, 2006, for use in combination with dexamethasone in patients with multiple myeloma (MM) who have received at least one prior therapy."( Lenalidomide in combination with dexamethasone for the treatment of multiple myeloma after one prior therapy.
Booth, B; Dagher, R; Farrell, A; Hazarika, M; Justice, R; Pazdur, R; Rock, E; Sridhara, R; Williams, G, 2008
)
1.79
" This phase 1/2 dose-escalation study aimed to determine the maximum tolerated dose (MTD) of lenalidomide in combination with melphalan and dexamethasone (M-dex), and assess the efficacy and tolerability of this therapy for patients with de novo AL amyloidosis."( Lenalidomide in combination with melphalan and dexamethasone in patients with newly diagnosed AL amyloidosis: a multicenter phase 1/2 dose-escalation study.
Alakl, M; Benboubker, L; Bridoux, F; Fermand, JP; Harousseau, JL; Hermine, O; Jaccard, A; Leblond, V; Leleu, X; Moreau, P; Planche, L; Roussel, M; Royer, B; Salles, G, 2010
)
2.02
"We evaluated the in vitro antimyeloma activity of AT9283 alone and in combination with lenalidomide and the in vivo efficacy by using a xenograft mouse model of human MM."( Antimyeloma activity of a multitargeted kinase inhibitor, AT9283, via potent Aurora kinase and STAT3 inhibition either alone or in combination with lenalidomide.
Anderson, KC; Bandi, M; Chauhan, D; Cirstea, D; Gorgun, G; Hideshima, T; Hu, Y; Mahindra, A; Munshi, NC; Nelson, EA; Patel, K; Pozzi, S; Raje, N; Rodig, S; Santo, L; Squires, M; Unitt, C; Vallet, S, 2011
)
0.79
" We conducted a phase I trial to establish the maximum tolerated dose of lenalidomide that could be combined with R-CHOP (rituximab-cyclophosphamide, doxorubicin, vincristine, and prednisone)."( Lenalidomide can be safely combined with R-CHOP (R2CHOP) in the initial chemotherapy for aggressive B-cell lymphomas: phase I study.
Ansell, SM; Habermann, TM; Inwards, DJ; Johnston, PB; Klebig, RR; LaPlant, B; Macon, WR; Micallef, IN; Nowakowski, GS; Porrata, LF; Reeder, CB; Rivera, CE; Witzig, TE, 2011
)
2.04
"Ezatiostat was administered to 19 patients with non-deletion(5q) myelodysplastic syndrome (MDS) at one of two doses (2000 mg or 2500 mg/day) in combination with 10 mg of lenalidomide on days 1-21 of a 28-day cycle."( Phase 1 dose-ranging study of ezatiostat hydrochloride in combination with lenalidomide in patients with non-deletion (5q) low to intermediate-1 risk myelodysplastic syndrome (MDS).
Boccia, R; Brown, GL; Galili, N; Garcia-Manero, G; Lyons, RM; Mesa, RA; Mulford, D; Raza, A; Sekeres, MA; Smith, SE; Steensma, DP, 2012
)
0.8
"The tolerability and activity profile of ezatiostat co-administered with lenalidomide supports the further development of ezatiostat in combination with lenalidomide in MDS and also encourages studies of this combination in other hematologic malignancies where lenalidomide is active."( Phase 1 dose-ranging study of ezatiostat hydrochloride in combination with lenalidomide in patients with non-deletion (5q) low to intermediate-1 risk myelodysplastic syndrome (MDS).
Boccia, R; Brown, GL; Galili, N; Garcia-Manero, G; Lyons, RM; Mesa, RA; Mulford, D; Raza, A; Sekeres, MA; Smith, SE; Steensma, DP, 2012
)
0.84
" We aimed to identify the maximum tolerated dose (MTD) of lenalidomide when combined with rituximab in a phase 1 trial and to assess the efficacy and safety of this combination in a phase 2 trial in patients with relapsed or refractory MCL."( Lenalidomide in combination with rituximab for patients with relapsed or refractory mantle-cell lymphoma: a phase 1/2 clinical trial.
Badillo, M; Bejarano, M; Bell, N; Byrne, C; Cabanillas, F; Champlin, R; Chen, W; Chen, Y; Fanale, M; Fayad, L; Feng, L; Fowler, N; Hagemeister, F; Hartig, K; Kwak, L; McLaughlin, P; Neelapu, SS; Newberry, KJ; Romaguera, J; Samaniego, F; Sun, L; Wagner-Bartak, N; Wang, M; Younes, A; Young, KH; Zeldis, J; Zhang, L, 2012
)
2.07
" A total of 704 patients (390 aged <65 years, 232 aged 65-74 years, and 82 aged ≥75 years) received lenalidomide or placebo, both in combination with dexamethasone."( Lenalidomide in combination with dexamethasone improves survival and time-to-progression in patients ≥65 years old with relapsed or refractory multiple myeloma.
Borrello, I; Chanan-Khan, AA; Dimopoulos, M; Foà, R; Hellmann, A; Knight, R; Lonial, S; Swern, AS; Weber, D, 2012
)
2.04
"Preclinical and clinical studies have shown that proteasome inhibitors (PIs) have anti-MM activity in combination with dexamethasone or lenalidomide."( CEP-18770 (delanzomib) in combination with dexamethasone and lenalidomide inhibits the growth of multiple myeloma.
Berenson, JR; Chen, H; Hunter, K; Jones-Bolin, S; Li, J; Li, M; Ruggeri, B; Sanchez, E; Wang, C, 2012
)
0.82
"To evaluate the 6-mo overall survival, safety and tolerability of lenalidomide in combination with standard gemcitabine as first-line treatment for patients with metastatic pancreatic cancer."( Lenalidomide in combination with gemcitabine as first-line treatment for patients with metastatic carcinoma of the pancreas: a Sarah Cannon Research Institute phase II trial.
Arkenau, HT; Bendell, JC; Burris, HA; Hainsworth, JD; Infante, JR; Jones, GT; Lane, CM; Rubin, MS; Spigel, DR; Waterhouse, D, 2013
)
2.07
"We conducted a phase I dose escalation study to determine the maximal tolerated dose of bortezomib that could be combined with standard dose lenalidomide in patients with MDS or AML."( Phase I dose escalation study of bortezomib in combination with lenalidomide in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML).
Amrein, PC; Attar, EC; Ballen, KK; Deangelo, DJ; Fathi, AT; Foster, J; Fraser, JW; McAfee, S; Neuberg, D; Steensma, DP; Stone, RM; Wadleigh, M, 2013
)
0.83
" Furthermore, reported for the first time is the effect of lenalidomide in combination with cetuximab on T cell function, including increases in circulating naïve and central memory T cells."( Immunomodulatory effects in a phase II study of lenalidomide combined with cetuximab in refractory KRAS-mutant metastatic colorectal cancer patients.
Chopra, R; Gandhi, AK; Jungnelius, U; Li, M; Romano, A; Schafer, PH; Shi, T; Siena, S; Tabernero, J, 2013
)
0.89
" In conclusion, the poor results obtained with lenalidomide in combination with vorinostat and dexamethasone provide no arguments that could justify further investigation of this drug combination for the treatment of relapsed PTCL."( Lenalidomide in combination with vorinostat and dexamethasone for the treatment of relapsed/refractory peripheral T cell lymphoma (PTCL): report of a phase I/II trial.
Egle, A; Greil, R; Hopfinger, G; Lang, A; Linkesch, W; Melchardt, T; Nösslinger, T; Weiss, L, 2014
)
2.1
" The C max of digoxin was slightly higher (+14 %) when administered with lenalidomide versus placebo."( No clinically significant drug interactions between lenalidomide and P‑glycoprotein substrates and inhibitors: results from controlled phase I studies in healthy volunteers.
Chen, N; Kasserra, C; Kumar, G; Liu, L; Palmisano, M; Reyes, J; Wang, X; Weiss, D; Weiss, L; Zhou, S, 2014
)
0.88
" Xenografts acquired resistance to two generations of immunomodulatory drugs (IMiDs; lenalidomide and pomalidomide) in combination with dexamethasone, that was reversible after a wash-out period."( In vivo murine model of acquired resistance in myeloma reveals differential mechanisms for lenalidomide and pomalidomide in combination with dexamethasone.
Bjorklund, CC; Corchete, LA; Couto, S; Delgado, M; Díaz-Rodríguez, E; Fernández-Lázaro, D; Garayoa, M; García-Gómez, A; Gutiérrez, NC; López-Corral, L; Mateos, MV; Montero, JC; Ocio, EM; Paíno, T; Pandiella, A; San-Miguel, JF; San-Segundo, L; Wang, M, 2015
)
0.86
" We demonstrated that lenalidomide can be safely combined with R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone); this new combination is known as R2CHOP."( Lenalidomide combined with R-CHOP overcomes negative prognostic impact of non-germinal center B-cell phenotype in newly diagnosed diffuse large B-Cell lymphoma: a phase II study.
Ansell, SM; Foran, JM; Gascoyne, RD; Habermann, TM; Inwards, DJ; Johnston, PB; LaPlant, B; Macon, WR; Micallef, IN; Nelson, GD; Nowakowski, GS; Porrata, LF; Reeder, CB; Rivera, CE; Thompson, CA; Witzig, TE, 2015
)
2.17
" Bendamustine was administered with a cumulative dose of up to 200 mg/m(2)."( Bendamustine in combination with thalidomide and dexamethasone is a viable salvage option in myeloma relapsed and/or refractory to bortezomib and lenalidomide.
Aitchison, R; Blesing, N; Lau, IJ; Peniket, A; Rabin, N; Ramasamy, K; Roberts, P; Smith, D; Yong, K, 2015
)
0.62
" In a phase 1/2 trial we aimed to assess the safety, tolerability, and activity of ixazomib in combination with lenalidomide and dexamethasone in newly diagnosed multiple myeloma."( Safety and tolerability of ixazomib, an oral proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma: an open-label phase 1/2 study.
Berdeja, JG; Berg, D; Di Bacco, A; Estevam, J; Gupta, N; Hamadani, M; Hari, P; Hui, AM; Kaufman, JL; Kumar, SK; Laubach, JP; Liao, E; Lonial, S; Niesvizky, R; Rajkumar, V; Richardson, PG; Roy, V; Stewart, AK; Vescio, R, 2014
)
0.84
"In this Phase 1b study, the safety and tolerability of maintenance therapy, comprising lenalidomide (0-25 mg, days 5-25) in combination with azacitidine (50-75 mg/m(2) , days 1-5) every 28 d, was explored in 40 patients with acute myeloid leukaemia (AML) in complete remission after chemotherapy."( Maintenance lenalidomide in combination with 5-azacitidine as post-remission therapy for acute myeloid leukaemia.
Avery, S; Fleming, S; Govindaraj, C; McManus, J; Patil, S; Perruzza, S; Plebanski, M; Spencer, A; Stevenson, W; Tan, P; Wei, A, 2015
)
1.02
"On August 5, 2013, a marketing authorization valid throughout the European Union (EU) was issued for pomalidomide in combination with dexamethasone for the treatment of adult patients with relapsed and refractory multiple myeloma (MM) who have received at least two prior treatment regimens, including both lenalidomide and bortezomib, and have demonstrated disease progression on the last therapy."( The European medicines agency review of pomalidomide in combination with low-dose dexamethasone for the treatment of adult patients with multiple myeloma: summary of the scientific assessment of the committee for medicinal products for human use.
Camarero, J; Flores, B; Gisselbrecht, C; Hanaizi, Z; Hemmings, R; Laane, E; Pignatti, F; Salmonson, T; Sancho-Lopez, A, 2015
)
0.59
" The maximum tolerated dose (MTD) of lenalidomide given in combination with gemcitabine was defined as the highest dose level at which no more than one out of four (25%) subjects experiences a dose-limiting toxicity (DLT)."( A phase I dose-escalation study of lenalidomide in combination with gemcitabine in patients with advanced pancreatic cancer.
Liljefors, M; Rossmann, E; Ullenhag, GJ, 2015
)
0.97
"Treatment with dose-adjusted Len combined with low-dose Dex is an effective and safe therapy for older RRMM patients exhibiting renal impairment after receiving Bor-based therapies."( Dose-adjusted Lenalidomide Combined with Low-dose Dexamethasone Rescues Older Patients with Bortezomib-resistant Multiple Myeloma.
Kadowaki, M; Kohno, K; Okamura, S; Takase, K; Yamasaki, S, 2015
)
0.78
"Proteasome inhibitors (PIs) in combination with immunomodulatory drugs (IMiDs) have been shown to be effective against relapsed/refractory multiple myeloma (RRMM)."( Phase I study of once weekly treatment with bortezomib in combination with lenalidomide and dexamethasone for relapsed or refractory multiple myeloma.
Hagiwara, S; Iida, S; Ishida, T; Ito, A; Kanamori, T; Kato, C; Kinoshita, S; Komatsu, H; Kusumoto, S; Masuda, A; Murakami, S; Nakashima, T; Narita, T; Ri, M; Suzuki, N; Totani, H; Yoshida, T, 2016
)
0.66
"The present study evaluated the pharmacokinetics and safety of elotuzumab, a humanized IgG1 monoclonal antibody against signaling lymphocyte activation molecule-F7, combined with lenalidomide and dexamethasone, in patients with multiple myeloma (MM) and renal impairment."( Pharmacokinetics and Safety of Elotuzumab Combined With Lenalidomide and Dexamethasone in Patients With Multiple Myeloma and Various Levels of Renal Impairment: Results of a Phase Ib Study.
Badros, A; Berdeja, J; Bleickardt, E; Gupta, M; Jagannath, S; Kaufman, JL; Lynch, M; Manges, R; Paliwal, P; Tendolkar, A; Vij, R; Zonder, J, 2016
)
0.87
" We conducted a phase I study of lenalidomide in combination with FOLFOX (5-fluorouracil, leucovorin, and oxaliplatin) in patients with advanced cancer."( Phase I clinical trial of lenalidomide in combination with 5-fluorouracil, leucovorin, and oxaliplatin in patients with advanced cancer.
Falchook, G; Fu, S; Hong, DS; Naing, A; Piha-Paul, S; Said, R; Tsimberidou, AM; Wheler, JJ; Ye, Y, 2016
)
1.02
"Lenalidomide in combination with FOLFOX was well tolerated."( Phase I clinical trial of lenalidomide in combination with 5-fluorouracil, leucovorin, and oxaliplatin in patients with advanced cancer.
Falchook, G; Fu, S; Hong, DS; Naing, A; Piha-Paul, S; Said, R; Tsimberidou, AM; Wheler, JJ; Ye, Y, 2016
)
2.18
" We conducted a phase 1b trial to determine the maximum tolerated dose (MTD) of lenalidomide in combination with R-ESHAP (rituximab, etoposide, cisplatin, cytarabine, methylprednisolone) (LR-ESHAP) in patients with relapsed or refractory DLBCL."( Lenalidomide in combination with R-ESHAP in patients with relapsed or refractory diffuse large B-cell lymphoma: a phase 1b study from GELTAMO group.
Caballero, D; Canales, M; Dlouhy, I; González-Barca, E; López-Guillermo, A; Martín, A; Montes-Moreno, S; Ocio, EM; Redondo, AM; Salar, A, 2016
)
2.1
"Pomalidomide is an IMiD(®) immunomodulatory agent, which has shown clinically significant benefits in relapsed and/or refractory multiple myeloma (rrMM) patients when combined with dexamethasone, regardless of refractory status to lenalidomide or bortezomib."( Pomalidomide in combination with dexamethasone results in synergistic anti-tumour responses in pre-clinical models of lenalidomide-resistant multiple myeloma.
Bjorklund, CC; Cathers, BE; Chopra, R; Daniel, TO; Gandhi, AK; Leisten, J; Lopez-Girona, A; Lu, L; Mendy, D; Miller, K; Narla, RK; Ning, Y; Orlowski, RZ; Raymon, HK; Rychak, E; Shi, T; Thakurta, A, 2016
)
0.83
" We developed a phase I study to evaluate the safety and tolerability of Len in combination with intermediate dose AraC (1."( A phase I study of intermediate dose cytarabine in combination with lenalidomide in relapsed/refractory acute myeloid leukemia.
Brady, WE; Dickey, NM; Ford, LA; Griffiths, EA; L Bashaw, H; Sperrazza, J; Tan, W; Thompson, JE; Vigil, CE; Wang, ES; Wetzler, M, 2016
)
0.67
" Lenalidomide has a manageable safety profile whether administered as a single agent or in combination with rituximab."( Clinical experience with lenalidomide alone or in combination with rituximab in indolent B-cell and mantle cell lymphomas.
Martin, P; Ruan, J; Schuster, SJ; Shah, B, 2016
)
1.65
"To investigate the clinical efficacy of regimen consisting of lenalidomide combined with chemotherapy for acute leukemia and its impact on vascular endothilial growth factor (vEGF) and basic fibroblast growth factor (bFGF), and to analyze the relationship lenalidomide with therapeutic efficacy of leukemia."( [Clinical Curative Efficacy of Lenalidomide Combined with Chemotherapy for Acute Leukemia and Its Impact on VEGF].
Ma, LM; Ruan, LH; Yang, XW; Zhao, XQ, 2016
)
0.96
"The lenalidomide combined with chemotherapy can significantly decrease the expression level of VEGF and bFGF, and enhance the remission rate of patients with AML."( [Clinical Curative Efficacy of Lenalidomide Combined with Chemotherapy for Acute Leukemia and Its Impact on VEGF].
Ma, LM; Ruan, LH; Yang, XW; Zhao, XQ, 2016
)
1.28
" We have previously shown in a retrospective analysis that lenalidomide combined with continuous low-dose cyclophosphamide and prednisone (REP) had remarkable activity in heavily pretreated, lenalidomide-refractory MM patients."( Phase 1/2 study of lenalidomide combined with low-dose cyclophosphamide and prednisone in lenalidomide-refractory multiple myeloma.
Beeker, A; Bloem, AC; Bos, GMJ; Broijl, A; Faber, LM; Franssen, LE; Klein, SK; Koene, HR; Levin, MD; Lokhorst, HM; Mutis, T; Nijhof, IS; Oostvogels, R; Raymakers, R; Sonneveld, P; van de Donk, NWCJ; van der Spek, E; van Kessel, B; van Spronsen, DJ; van Velzen, J; Westerweel, PE; Ypma, PF; Zweegman, S, 2016
)
1.01
"Clinical trials of vorinostat, a Class I/II histone deacetylase inhibitor, in combination with proteasome inhibitors and immunomodulatory agents have shown activity in relapsed/refractory multiple myeloma."( A phase IIb trial of vorinostat in combination with lenalidomide and dexamethasone in patients with multiple myeloma refractory to previous lenalidomide-containing regimens.
Anand, P; Bilotti, E; Biran, N; Ivanovski, K; McBride, L; Richter, JR; Sanchez, L; Siegel, DS; Vesole, DH, 2017
)
0.71
"We report the first clinical investigation conducted in Japan to confirm the safety, tolerability, and pharmacokinetics of ixazomib alone and combined with lenalidomide-dexamethasone (Rd) in Japanese patients with relapsed/refractory multiple myeloma."( Phase 1 study of ixazomib alone or combined with lenalidomide-dexamethasone in Japanese patients with relapsed/refractory multiple myeloma.
Chou, T; Handa, H; Ishizawa, K; Kase, Y; Suzuki, K; Takubo, T, 2017
)
0.91
" In a previous phase 3 study in patients with relapsed/refractory multiple myeloma (RRMM), elotuzumab (10 mg/kg, ∼3-h infusion), combined with lenalidomide and dexamethasone, demonstrated durable efficacy and acceptable safety; 10% (33/321) of patients had infusion reactions (IRs; Grade 1/2: 29; Grade 3: 4)."( A phase 2 safety study of accelerated elotuzumab infusion, over less than 1 h, in combination with lenalidomide and dexamethasone, in patients with multiple myeloma.
Badarinath, S; Berenson, J; Cartmell, A; Harb, W; Lyons, R; Manges, R; McIntyre, K; Mohamed, H; Nourbakhsh, A; Rifkin, R, 2017
)
0.87
"On November 30, 2015, the FDA approved elotuzumab (Empliciti; Bristol-Myers Squibb) in combination with lenalidomide and dexamethasone for the treatment of patients with multiple myeloma who received one to three prior therapies."( FDA Drug Approval: Elotuzumab in Combination with Lenalidomide and Dexamethasone for the Treatment of Relapsed or Refractory Multiple Myeloma.
Deisseroth, A; Farrell, AT; Goldberg, KB; Gormley, NJ; Kaminskas, E; Ko, CW; Kormanik, N; Nie, L; Pazdur, R, 2017
)
0.92
" This phase I/II study was conducted to determine the maximum tolerated dose (MTD), safety, and efficacy of lenalidomide combined with panobinostat in relapsed/refractory HL."( A Phase I/II Trial of Panobinostat in Combination With Lenalidomide in Patients With Relapsed or Refractory Hodgkin Lymphoma.
Bartlett, NL; Blum, KA; Christian, BA; Devine, SM; Fehniger, TA; Jaglowski, SM; Maly, JJ; Phelps, MA; Sexton, JL; Wagner-Johnston, ND; Wei, L; Zhu, X, 2017
)
0.92
"This phase 1 study investigated the safety of the anthracycline amrubicin combined with lenalidomide and dexamethasone in adults with relapsed or refractory multiple myeloma."( A phase I, open-label, dose-escalation study of amrubicin in combination with lenalidomide and weekly dexamethasone in previously treated adults with relapsed or refractory multiple myeloma.
Berube, C; Coutré, SE; Dinner, S; Dunn, TJ; Gotlib, J; Kaufman, GP; Liedtke, M; Medeiros, BC; Price, E, 2018
)
0.93
"To evaluate the cost-effectiveness of lenalidomide in combination with dexamethasone compared to bortezomib in combination with dexamethasone in patients with multiple myeloma previously treated with bortezomib, from the perspective of the Chilean National Health Service."( Cost-effectiveness of lenalidomide in combination with dexamethasone compared to bortezomib in combination with dexamethasone for the second-line treatment of multiple myeloma in Chile.
Aceituno, S; Appierto, M; Gozalbo, I; Lizán, L, 2018
)
1.07
"Lenalidomide in combination with dexamethasone represents a potentially cost-effective alternative for the second-line treatment of patients with multiple myeloma who are not eligible for transplantation, from the perspective of the Chilean National Health Service."( Cost-effectiveness of lenalidomide in combination with dexamethasone compared to bortezomib in combination with dexamethasone for the second-line treatment of multiple myeloma in Chile.
Aceituno, S; Appierto, M; Gozalbo, I; Lizán, L, 2018
)
2.24
" The treatment outcomes of MAbs versus HDACi in combination with bortezomib or lenalidomide plus dexamethasone remain unknown."( Monoclonal Antibodies versus Histone Deacetylase Inhibitors in Combination with Bortezomib or Lenalidomide plus Dexamethasone for the Treatment of Relapsed or Refractory Multiple Myeloma: An Indirect-Comparison Meta-Analysis of Randomized Controlled Trial
Chen, X; Feng, J; Gao, G; Shen, H; Tang, H; Xu, L; Zhang, N; Zheng, Y, 2018
)
0.93
"Lenalidomide in combination with R-CHOP had an acceptable safety profile and showed anti-cancer activity in patients with previously untreated high burden follicular lymphoma."( Lenalidomide in combination with R-CHOP (R2-CHOP) as first-line treatment of patients with high tumour burden follicular lymphoma: a single-arm, open-label, phase 2 study.
Becker, S; Bouabdallah, R; Cabeçadas, J; Casasnovas, O; Feugier, P; Gabarre, J; Gouill, SL; Haioun, C; Jardin, F; Lamy, T; Molina, TJ; Morschhauser, F; Mounier, N; Salles, G; Tilly, H; Tournilhac, O, 2018
)
3.37
" These results support assessments of the efficacy of R2 combined with methotrexate-based chemotherapy as a first-line treatment of PCNSL."( Lenalidomide in combination with intravenous rituximab (REVRI) in relapsed/refractory primary CNS lymphoma or primary intraocular lymphoma: a multicenter prospective 'proof of concept' phase II study of the French Oculo-Cerebral lymphoma (LOC) Network and
Beauchesne, P; Cassoux, N; Chevrier, M; Chinot, O; Choquet, S; Ghesquieres, H; Gressin, R; Gyan, E; Hoang-Xuan, K; Houillier, C; Laadhari, M; Le Garff-Tavernier, M; Moluçon-Chabrot, C; Morschhauser, F; Nicolas-Virelizier, E; Savignoni, A; Schmitt, A; Soumelis, V; Soussain, C; Touitou, V; Turbiez, I, 2019
)
1.96
"In preclinical studies, oral azacitidine (CC-486), a hypomethylating agent, has been shown to have a direct anti-MM effect and in vitro anti-MM synergism when combined with lenalidomide (LEN)."( Oral azacitidine (CC-486) in combination with lenalidomide and dexamethasone in advanced, lenalidomide-refractory multiple myeloma (ROAR study).
Bergin, K; Bowen, KM; Couto, S; Guzman, R; Kalff, A; Khong, T; Mithraprabhu, S; Ren, Y; Reynolds, J; Spencer, A; Thakurta, A; Wang, M, 2019
)
0.97
"This phase Ib/II study aimed to determine the safety and tolerability of ulocuplumab alone and in combination with lenalidomide and dexamethasone (Arm A), or bortezomib and dexamethasone (Arm B), in patients with relapsed/refractory multiple myeloma."( A Phase Ib/II Trial of the First-in-Class Anti-CXCR4 Antibody Ulocuplumab in Combination with Lenalidomide or Bortezomib Plus Dexamethasone in Relapsed Multiple Myeloma.
Anderson, KC; Baz, R; Becker, PS; Chuma, S; Ghobrial, IM; Henrick, P; Hornburg, K; Laubach, J; Liu, CJ; Perez, RP; Redd, RA; Reyes, K; Richardson, PG; Robbins, MD; Sabbatini, P; Savell, A; Sklavenitis-Pistofidis, R; Zavidij, O, 2020
)
0.99
"This study showed that blockade of the CXCR4-CXCL12 axis by ulocuplumab is safe with acceptable AEs and leads to a high response rate in combination with lenalidomide and dexamethasone in patients with relapsed/refractory myeloma, making CXCR4 inhibitors a promising class of antimyeloma drugs that should be further explored in clinical trials."( A Phase Ib/II Trial of the First-in-Class Anti-CXCR4 Antibody Ulocuplumab in Combination with Lenalidomide or Bortezomib Plus Dexamethasone in Relapsed Multiple Myeloma.
Anderson, KC; Baz, R; Becker, PS; Chuma, S; Ghobrial, IM; Henrick, P; Hornburg, K; Laubach, J; Liu, CJ; Perez, RP; Redd, RA; Reyes, K; Richardson, PG; Robbins, MD; Sabbatini, P; Savell, A; Sklavenitis-Pistofidis, R; Zavidij, O, 2020
)
0.98
"To predict the real-world (RW) cost-effectiveness of carfilzomib in combination with lenalidomide and dexamethasone (KRd) versus lenalidomide and dexamethasone (Rd) in relapsed multiple myeloma (MM) patients after one to three prior therapies."( Methodology and results of real-world cost-effectiveness of carfilzomib in combination with lenalidomide and dexamethasone in relapsed multiple myeloma using registry data.
Agirrezabal, I; Campioni, M; Gonzalez-McQuire, S; Hajek, R; Jandova, P; Maisnar, V; Minarik, J; Pour, L; Spicka, I, 2020
)
1
" The combination of cyclophosphamide and dexamethasone (CD) is a recognised treatment option for patients with relapsed refractory multiple myeloma (RRMM) who have relapsed after treatment with bortezomib and lenalidomide, whilst also often being combined with newer proteasome inhibitors."( The MUK eight protocol: a randomised phase II trial of cyclophosphamide and dexamethasone in combination with ixazomib, in relapsed or refractory multiple myeloma (RRMM) patients who have relapsed after treatment with thalidomide, lenalidomide and a prote
Auner, HW; Bailey, L; Brown, S; Brudenell Straw, F; Cook, G; Cook, M; Dawkins, B; Flanagan, L; Hinsley, S; Kaiser, MF; McKee, S; Meads, D; Reed, S; Sherratt, D; Walker, K, 2020
)
0.93
" The primary objective of the trial is to evaluate whether ixazomib in combination with cyclophosphamide and dexamethasone (ICD) has improved clinical activity compared to CD in terms of progression-free survival (PFS)."( The MUK eight protocol: a randomised phase II trial of cyclophosphamide and dexamethasone in combination with ixazomib, in relapsed or refractory multiple myeloma (RRMM) patients who have relapsed after treatment with thalidomide, lenalidomide and a prote
Auner, HW; Bailey, L; Brown, S; Brudenell Straw, F; Cook, G; Cook, M; Dawkins, B; Flanagan, L; Hinsley, S; Kaiser, MF; McKee, S; Meads, D; Reed, S; Sherratt, D; Walker, K, 2020
)
0.74
" Previous studies investigating the safety and efficacy of ICD in patients with RRMM demonstrate a toxicity profile consistent with ixazomib in combination with lenalidomide and dexamethasone, whilst the combination showed possible activity in RRMM patients."( The MUK eight protocol: a randomised phase II trial of cyclophosphamide and dexamethasone in combination with ixazomib, in relapsed or refractory multiple myeloma (RRMM) patients who have relapsed after treatment with thalidomide, lenalidomide and a prote
Auner, HW; Bailey, L; Brown, S; Brudenell Straw, F; Cook, G; Cook, M; Dawkins, B; Flanagan, L; Hinsley, S; Kaiser, MF; McKee, S; Meads, D; Reed, S; Sherratt, D; Walker, K, 2020
)
0.94
" Recently, Phase Ib and Phase III trials evaluated the triplet drug combination daratumumab-carfilzomib-dexamethasone in the relapse setting and demonstrated strong clinical efficacy, especially in lenalidomide refractory patients."( Carfilzomib in combination with daratumumab in the management of relapsed multiple myeloma.
Antier, C; Moreau, P; Touzeau, C, 2021
)
0.81
" Lenalidomide combination with rituximab (LR) is among the most successful and extensively studied novel approaches."( An Overview of Lenalidomide in Combination with Rituximab for the Treatment of Adult Patients with Follicular Lymphoma: The Evidence to Date.
Salihoglu, A; Soysal, T; Yilmaz, U, 2021
)
1.88
"This study evaluates the cost-effectiveness of daratumumab (D) in combination with lenalidomide and dexamethasone (Rd) for treatment of relapsed and/or refractory multiple myeloma in patients who have received at least one prior therapy in Singapore."( Cost-effectiveness of daratumumab in combination with lenalidomide and dexamethasone for relapsed and/or refractory multiple myeloma.
Aziz, MIA; Chng, WJ; Ng, K; Wong, XY, 2022
)
1.19
" Despite the proven clinical activity of tafasitamab in combination with lenalidomide in the treatment of diffuse large B-cell lymphoma (DLBCL), a higher number of immune cells in cancer patients may improve the activity of tafasitamab."( Tafasitamab mediates killing of B-cell non-Hodgkin's lymphoma in combination with γδ T cell or allogeneic NK cell therapy.
Boxhammer, R; Cho, SY; Endell, J; Heitmueller, C; Her, JH; Hoeres, T; Hwang, YK; Patra-Kneuer, M; Pretscher, D; Schanzer, J; Steidl, S; Wilhelm, M, 2022
)
0.95
" An exposure-response (E-R) analysis using data from patients with relapsed/refractory multiple myeloma (RRMM) enrolled in a phase Ib clinical study who received isatuximab at doses from 5 to 20 mg/kg weekly for 1 cycle (4 weeks) followed by every 2 weeks thereafter (qw/q2w) in combination with pomalidomide/dexamethasone (n = 44) was first used to determine the optimal dose/schedule for the phase III ICARIA-MM study."( Exposure-response analyses for selection/confirmation of optimal isatuximab dosing regimen in combination with pomalidomide/dexamethasone treatment in patients with multiple myeloma.
Brillac, C; Fau, JB; Gaudel-Dedieu, N; Koiwai, K; Nguyen, L; Rachedi, F; Sebastien, B; Semiond, D; Thai, HT; van de Velde, H; Veyrat-Follet, C, 2022
)
0.72
"In combination with lenalidomide and dexamethasone (KRd), Carfilzomib has been approved for the treatment of relapsed and refractory multiple myeloma (RRMM) on ASPIRE trial."( Carfilzomib combined with lenalidomide and dexamethasone (KRd) as salvage therapy for multiple myeloma patients: italian, multicenter, retrospective clinical experience with 600 cases outside of controlled clinical trials.
Barone, M; Botta, C; Bruzzese, A; Cangialosi, C; Catalano, L; Cavo, M; Conticello, C; Di Raimondo, F; Falcone, AP; Farina, G; Gamberi, B; Garibaldi, B; Gentile, M; Iaccino, E; Mancuso, K; Martino, EA; Mele, A; Mele, G; Mendicino, F; Morabito, F; Musso, M; Musto, P; Neri, A; Palmieri, S; Pavone, V; Reddiconto, G; Rizziello, I; Rocchi, S; Tacchetti, P; Tripepi, G; Vigna, E; Vincelli, ID; Zamagni, E, 2022
)
1.34
" Therefore, the effects of a long multi-epitope peptide vaccine in combination with lenalidomide and anti-PD1 were analyzed in this study."( The long multi-epitope peptide vaccine combined with adjuvants improved the therapeutic effects in a glioblastoma mouse model.
Jung, S; Jung, TY; Kim, GE; Kim, IY; Kim, YH; Kim, YJ; Lee, CW; Lee, HJ; Lee, JJ; Lee, TK; Moon, KS; Tran, TA; Yun, H, 2022
)
0.95
" We report the first case of primary PCL successfully treated with upfront novel agents consisting of Venetoclax and daratumumab in combination with intensive chemotherapy and allogeneic transplantation."( Plasma Cell Leukemia with Successful Upfront Venetoclax in Combination with Allogeneic Transplantation.
Ahmad Asnawi, AW; Chong, SL; Koh, AZY; Lau, NS; Liew, PK; Md Fauzi, A; Selvaratnam, V; Tan, SM; Tang, ASO, 2023
)
0.91
"Ixazomib (IXA) is an oral proteasome inhibitor (PI) used in combination with lenalidomide and dexamethasone (IXA-Rd) for patients with relapsed and/or refractory multiple myeloma (RRMM)."( Real-world effectiveness of ixazomib combined with lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: the REMIX study.
Barrak, J; Benboubker, L; Calmettes, C; Charvet-Rumpler, A; Chouaid, C; Clement-Filliatre, L; Honeyman, F; Hulin, C; Karlin, L; Laribi, K; Leleu, X; Macro, M; Maloisel, F; Richez, V; Stoppa, AM; Vincent, L; Zerazhi, H, 2023
)
1.39
" Ex-vivo studies demonstrating upregulation of KMA by lenalidomide, and enhanced effector-cell cytotoxicity provided the rationale for this phase IIb study where KM or KM in combination with lenalidomide and dexamethasone (KM-Rd) was administered in relapsed, refractory κMM patients."( A sequential cohort study evaluating single-agent KappaMab and KappaMab combined with lenalidomide and low-dose dexamethasone in relapsed and/or refractory kappa light chain-restricted multiple myeloma (AMaRC 01-16).
Dunn, R; Harrison, SJ; Kalff, A; Quach, H; Reynolds, J; Shortt, J; Spencer, A; Walker, P; Wallington-Gates, C; Wellard, C, 2023
)
1.38
" Subjects received 3 cycles of lenalidomide 10 mg/day on days 1-14 of every 21-day cycle, in combination with R-ESHAP at standard doses."( Lenalidomide in combination with R-ESHAP in patients with relapsed or refractory diffuse large B-cell lymphoma: A phase 2 study from GELTAMO.
Baile, M; Bergua, J; Caballero, D; Campo, E; Dlouhy, I; Enjuanes, A; Espeso, M; González-Barca, E; Grande, C; Jarque, I; López-Guillermo, A; Martín García-Sancho, A; Montes-Moreno, S; Redondo, A; Rodríguez, G; Salar, A; Sancho, JM, 2023
)
2.64

Bioavailability

Lenalidomide (LDM) is widely used for the treatment of transfusion-dependent anaemia. It has low oral bioavailability due to its poor aqueous solubility. The drug is an orally bioavailable analogue of thalidomides.

ExcerptReferenceRelevance
" Lenalidomide is an orally bioavailable analogue of thalidomide with more potent immunomodulatory, antiangiogenic, and antitumor activities than the parent compound and with a better safety profile."( Emerging data on IMiDs in the treatment of myelodysplastic syndromes (MDS).
List, AF, 2005
)
1.24
" Studies herein define mouse plasma pharmacokinetics and tissue distribution after intravenous (IV) bolus administration and bioavailability after oral and intraperitoneal delivery."( Pharmacokinetics and tissue disposition of lenalidomide in mice.
Byrd, JC; Gao, Y; Herman, SE; Jarjoura, D; Johnson, AJ; Li, X; Mahoney, E; Phelps, MA; Rozewski, DM; Shin, JD; Stefanovski, MR; Towns, WH; Yang, X, 2012
)
0.64
" Both of these analogs displayed oral bioavailability in rat."( Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity.
Babusis, D; Capone, L; Chen, R; Corral, L; Kang, J; Man, HW; Moghaddam, MF; Muller, GW; Parton, A; Ruchelman, AL; Schafer, PH; Shirley, MA; Tang, Y; Zhang, W, 2013
)
0.71
" Neither the rate nor the capacity of lenalidomide renal excretion was affected by quinidine or temsirolimus, in addition lenalidomide absorption rate and bioavailability remained unchanged."( No clinically significant drug interactions between lenalidomide and P‑glycoprotein substrates and inhibitors: results from controlled phase I studies in healthy volunteers.
Chen, N; Kasserra, C; Kumar, G; Liu, L; Palmisano, M; Reyes, J; Wang, X; Weiss, D; Weiss, L; Zhou, S, 2014
)
0.92
"Nonobvious controlled polymeric pharmaceutical excipient, chitosan nanoparticles (CS-NPs) for lenalidomide encapsulation were geared up by the simple ionic cross linking method to get better bioavailability and to reduce under as well as overloading of hydrophobic and sparingly soluble drug lenalidomide towards cancer cells."( Studies on drug-polymer interaction, in vitro release and cytotoxicity from chitosan particles excipient.
Gomathi, T; Govindarajan, C; Imran, PK; Kim, SK; Rose H R, MH; Sudha, PN; Venkatesan, J, 2014
)
0.62
" As an orally bioavailable immunomodulator with antineoplastic, immunologic, and antiproliferative activity in B-cell lymphoma, lenalidomide has emerged as one such option."( The evolving role of lenalidomide in non-Hodgkin lymphoma.
Galanina, N; Nabhan, C; Petrich, A, 2016
)
0.96
"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
" New nanomicelles were prepared as carriers for this combination to maximize bioavailability and accumulation at the tumor site because of the enhanced permeability and retention (EPR) effect."( A Novel Nanomicellar Combination of Fenretinide and Lenalidomide Shows Marked Antitumor Activity in a Neuroblastoma Xenograft Model.
Brodeur, GM; Calonghi, N; Chorny, M; Farruggia, G; Guan, P; Kolla, V; Nguyen, F; Orienti, I, 2019
)
0.76
"Lenalidomide (LDM), widely used for the treatment of transfusion-dependent anaemia, has low oral bioavailability due to its poor aqueous solubility."( Novel pharmaceutical cocrystal of lenalidomide with nicotinamide: Structural design, evaluation, and thermal phase transition study.
Wang, L; Yan, Y; Zhang, X; Zhou, X, 2022
)
2.44

Dosage Studied

The initial daily dosing of lenalidomide is 10 mg for MDS with a 5q deletion and 25 mg for relapsed or refractory multiple myeloma. However the response rate, even using lenalidmide, was low.

ExcerptRelevanceReference
" Caution should be exercised when lenalidomide therapy is commenced and CrCl should be incorporated as a determinant of the initial dosing of lenalidomide in MM patients."( Lenalidomide-induced myelosuppression is associated with renal dysfunction: adverse events evaluation of treatment-naïve patients undergoing front-line lenalidomide and dexamethasone therapy.
Chen-Kiang, S; Christos, PJ; Coleman, M; Ely, S; Furst, JR; Jalbrzikowski, J; Jayabalan, D; Lent, R; Leonard, JP; Mark, T; Mazumdar, M; Naib, T; Niesvizky, R; Pearse, RN; Zafar, F, 2007
)
2.06
" Clinical trials of relapsed and refractory MM have shown that lenalidomide is clinically efficacious at a dosage of 25 mg/day when administered in combination with dexamethasone."( Lenalidomide in myelodysplastic syndrome and multiple myeloma.
Shah, SR; Tran, TM, 2007
)
2.02
"The pharmacology, clinical use, adverse effects, dosage and administration, and cost of lenalidomide in the treatment of multiple myeloma (MM) are reviewed."( Lenalidomide in the treatment of multiple myeloma.
Rao, KV, 2007
)
2
" Future studies with lenalidomide in CLL should focus on understanding this toxicity, investigating patients at risk, and investigating alternative safer dosing schedules."( Higher doses of lenalidomide are associated with unacceptable toxicity including life-threatening tumor flare in patients with chronic lymphocytic leukemia.
Andritsos, LA; Awan, F; Blum, W; Byrd, JC; Chen, CS; Jarjoura, D; Johnson, AJ; Kefauver, C; Knight, RD; Lapalombella, R; Lehman, A; Lozanski, G; May, SE; Raymond, CA; Ruppert, AS; Smith, LL; Wang, DS, 2008
)
1.01
" The 45 patients received a total number of 192 cycles, respectively a median of three cycles; the median dosage of dexamethasone was 240 mg per cycle."( Effective prophylaxis of thromboembolic complications with low molecular weight heparin in relapsed multiple myeloma patients treated with lenalidomide and dexamethasone.
Goldschmidt, H; Hegenbart, U; Hillengass, J; Ho, AD; Hundemer, M; Klein, U; Kosely, F; Moehler, T; Neben, K; Schmitt, S, 2009
)
0.56
" With these drug therapies has come a more targeted approach to treatment enabling not only improved antimyeloma efficacy but also the use of decreased dosing enhancing the safety and tolerability of these regimens."( New drugs in multiple myeloma.
Berenson, JR; Yellin, O, 2008
)
0.35
" The recommended phase II dosing is docetaxel 75 mg/m(2) on day 1, lenalidomide 25 mg on days 1-14, and pegfilgrastim 6 mg on day 2, given every 3 weeks."( Phase I trial of docetaxel given every 3 weeks and daily lenalidomide in patients with advanced solid tumors.
Bako, J; Bokar, JA; Brell, JM; Cooney, MM; Dowlati, A; Gibbons, J; Horvath, N; Krishnamurthi, S; Nock, C; Remick, SC; Sanborn, SL, 2009
)
0.83
"Intended therapy consisted of 48 weeks of lenalidomide (25 mg/d for 3 weeks and then 1 week off) along with rituximab (375 mg/m(2)/wk) dosed on weeks 2 to 5 and 13 to 16."( Lenalidomide and rituximab in Waldenstrom's macroglobulinemia.
Anderson, KC; Baron, AD; Branagan, AR; Chu, L; Hunter, ZR; Hyman, PM; Ioakimidis, L; Kash, JJ; Munshi, NC; Musto, P; Nawfel, EL; Nunnink, JC; Patterson, CJ; Sharon, DJ; Soumerai, JD; Terjanian, TO; Treon, SP, 2009
)
2.06
" Improvement was noted within 1 month at a dosage of 5 mg/d in one case and was maintained for 10 months before the dosage was doubled to 10 mg/d for 12 months because of a slight worsening of symptoms."( Lenalidomide for the treatment of resistant discoid lupus erythematosus.
Albrecht, J; Bonilla-Martinez, Z; Okawa, J; Rose, M; Rosenbach, M; Shah, A; Werth, VP, 2009
)
1.8
" Lenalidomide was well tolerated up to a 35-mg/d intermittent dosing schedule at doses higher than previously indicated for hematologic malignancies."( Phase I study of oral lenalidomide in patients with refractory metastatic cancer.
Aragon-Ching, JB; Arlen, PM; Dahut, WL; Figg, WD; Gulley, JL; Tohnya, TM; Ventiz, J; Woo, S; Wright, JJ, 2009
)
1.58
" The initial daily dosing of lenalidomide is 10 mg for MDS with a 5q deletion and 25 mg for relapsed or refractory multiple myeloma."( The clinical utility of lenalidomide in multiple myeloma and myelodysplastic syndromes.
Bonkowski, J; Kolesar, JM; Vermeulen, LC, 2010
)
0.96
" With dosing of lenalidomide according to renal function, LenDex can be administered to patients with RI (who may not have other treatment options) without excessive toxicity."( Lenalidomide and dexamethasone for the treatment of refractory/relapsed multiple myeloma: dosing of lenalidomide according to renal function and effect on renal impairment.
Christoulas, D; Dimopoulos, MA; Efstathiou, E; Gavriatopoulou, M; Grapsa, I; Kastritis, E; Matsouka, C; Migkou, M; Mparmparoussi, D; Psimenou, E; Roussou, M; Terpos, E, 2010
)
2.15
"Lenalidomide (25 mg daily) treatment was then initiated in a continuous dosing schedule."( Lenalidomide induced good clinical response in a patient with multiple relapsed and refractory Hodgkin's lymphoma.
Kolonic, SO; Mandac, I, 2010
)
3.25
" Pharmacokinetic analysis demonstrated that the lenalidomide area under the concentration-time curve from 0 to 24 hours and maximum plasma concentration increased with dosage over the range studied."( Phase I trial of lenalidomide in pediatric patients with recurrent, refractory, or progressive primary CNS tumors: Pediatric Brain Tumor Consortium study PBTC-018.
Blaney, SM; Boyett, JM; Fangusaro, J; Goldman, S; Jakacki, R; Kun, LE; Macdonald, T; Packer, R; Pollack, IF; Schaiquevich, P; Stewart, CF; Wallace, D; Warren, KE, 2011
)
0.96
" Of note, once-weekly bortezomib dosing (in combination with MP±T) was shown to reduce the incidence of peripheral neuropathy and gastrointestinal events compared with twice-weekly dosing, while maintaining efficacy."( Practical management of adverse events in multiple myeloma: can therapy be attenuated in older patients?
Bringhen, S; Mateos, MV; Palumbo, A; San Miguel, JF, 2011
)
0.37
" Lenalidomide was dosed 10 mg on days 1 to 21 of a 28-day schedule for a total of 24 cycles."( Lenalidomide maintenance after nonmyeloablative allogeneic stem cell transplantation in multiple myeloma is not feasible: results of the HOVON 76 Trial.
Bruijnen, CP; Cornelisse, PB; Cornelissen, JJ; Emmelot, M; Huisman, C; Huls, G; Janssen, JJ; Kersten, MJ; Kneppers, E; Lokhorst, HM; Meijer, E; Minnema, MC; Mutis, T; Sonneveld, P; van der Holt, B; Zweegman, S, 2011
)
2.72
" Although well tolerated, this phase II trial did not show superior efficacy compared with conventional dosing and scheduling of these agents."( Phase II trial of syncopated thalidomide, lenalidomide, and weekly dexamethasone in patients with newly diagnosed multiple myeloma.
Anand, P; Bello, E; Bendarz, U; Bilotti, E; McBride, L; McNeill, A; Olivo, K; Siegel, DS; Tufail, M; Vesole, DH, 2012
)
0.64
" However the response rate, even using lenalidomide at 50 mg, was not superior to standard dosing of thalidomide or lenalidomide plus dexamethasone."( Phase II trial of syncopated thalidomide, lenalidomide, and weekly dexamethasone in patients with newly diagnosed multiple myeloma.
Anand, P; Bello, E; Bendarz, U; Bilotti, E; McBride, L; McNeill, A; Olivo, K; Siegel, DS; Tufail, M; Vesole, DH, 2012
)
0.91
" A prospective cohort study in 50 patients with RRMM has reported that when Len/Dex dosing was adjusted according to renal function, response rates and survival outcomes were similar in patients with and without RI, and there was no increase in adverse events in patients with RI."( Treatment with lenalidomide and dexamethasone in patients with multiple myeloma and renal impairment.
Alegre, A; Dimopoulos, MA; Goldschmidt, H; Mark, T; Niesvizky, R; Terpos, E, 2012
)
0.73
" However, phase III trials were dosed without regard to food; therefore, clinical relevance of the food effect was minimal."( Single-dose pharmacokinetics of lenalidomide in healthy volunteers: dose proportionality, food effect, and racial sensitivity.
Chen, N; Kasserra, C; Lau, H; Liu, L; Reyes, J, 2012
)
0.66
" We observed dose-dependent kinetics over the evaluated dosing range."( Pharmacokinetics and tissue disposition of lenalidomide in mice.
Byrd, JC; Gao, Y; Herman, SE; Jarjoura, D; Johnson, AJ; Li, X; Mahoney, E; Phelps, MA; Rozewski, DM; Shin, JD; Stefanovski, MR; Towns, WH; Yang, X, 2012
)
0.64
" Lenalidomide even in lower dosed combined with steroids can induce complete responses in patients with refractory AITL."( Therapy refractory angioimmunoblastic T-cell lymphoma in complete remission with lenalidomide.
Beckers, MM; Huls, G, 2013
)
1.53
" CRd was administered on 28-day dosing cycles: carfilzomib 15 to 27 mg/m(2) on days 1, 2, 8, 9, 15, and 16; lenalidomide 10 to 25 mg on days 1 to 21; and dexamethasone 40 mg weekly."( Phase Ib dose-escalation study (PX-171-006) of carfilzomib, lenalidomide, and low-dose dexamethasone in relapsed or progressive multiple myeloma.
Alsina, M; Bensinger, WI; Kunkel, LA; Lee, S; Martin, TG; Niesvizky, R; Orlowski, RZ; Siegel, DS; Wang, M; Wong, AF, 2013
)
0.84
" The most common adverse events (incidence ≥20 %) occurring in lenalidomide recipients were thrombocytopenia and neutropenia, which were generally managed by dosage reductions and/or interruptions, and/or pharmacotherapy."( Lenalidomide: a review of its use in patients with transfusion-dependent anaemia due to low- or intermediate-1-risk myelodysplastic syndrome associated with 5q chromosome deletion.
Scott, LJ; Syed, YY, 2013
)
2.07
" The optimal dosing schedule of granulocyte colony-stimulating factor (G-CSF) is unclear."( Intermittent granulocyte colony-stimulating factor for neutropenia management in patients with relapsed or refractory multiple myeloma treated with lenalidomide plus dexamethasone.
Atenafu, EG; Chen, C; Kukreti, V; Masih-Khan, E; Reece, DE; Sun, HL; Trudel, S; Winter, A; Yeboah, E, 2015
)
0.62
" High-dose cytarabine is clearly effective and there definitely is a dose-response relationship for cytarabine and remission duration."( Optimal therapy for adult patients with acute myeloid leukemia in first complete remission.
Wiernik, PH, 2014
)
0.4
" The MTD was found to be continuous dosing of lenalidomide 10 mg/day plus sunitinib 37."( A phase I/II study of lenalidomide in combination with sunitinib in patients with advanced or metastatic renal cell carcinoma.
Beck, R; Burris, HA; Fandi, A; Garcia, JA; Infante, JR; Jungnelius, U; Li, S; Redman, B; Rini, B, 2014
)
0.98
"Adequate dosing of lenalidomide in Chronic Lymphocytic Leukemia (CLL) remains unclear."( A dose escalation feasibility study of lenalidomide for treatment of symptomatic, relapsed chronic lymphocytic leukemia.
Andritsos, LA; Browning, R; Byrd, JC; Flynn, J; Gao, Y; Harper, E; Jiang, Y; Johnson, AJ; Jones, J; Kefauver, C; Maddocks, K; Phelps, MA; Poi, M; Rozewski, DM; Ruppert, AS, 2014
)
1
" For patients with small lymphocytic lymphoma, dosing began at 10 mg/day to avoid tumour flare, with an escalation of 5 mg/month to 20 mg/day."( Safety and activity of lenalidomide and rituximab in untreated indolent lymphoma: an open-label, phase 2 trial.
Baladandayuthapani, V; Claret, LC; Davis, RE; Fanale, MA; Fayad, LE; Feng, L; Fowler, NH; Hagemeister, FB; Kwak, LW; McLaughlin, P; Muzzafar, T; Nastoupil, L; Neelapu, SS; Oki, Y; Orlowski, RZ; Rawal, S; Romaguera, JE; Samaniego, F; Shah, J; Tsai, KY; Turturro, F; Wang, M; Westin, JR, 2014
)
0.71
" Treatment-specific tools and clinical assessments can be useful for optimizing dosing and schedule adjustments to increase therapy duration, and implementing supportive care strategies (e."( Treatment-related symptom management in patients with multiple myeloma: a review.
Colson, K, 2015
)
0.42
" In addition to appropriate drug dosing and administration, effective supportive care and health maintenance are crucial for maximizing quality of life and disease control."( Treatment-related symptom management in patients with multiple myeloma: a review.
Colson, K, 2015
)
0.42
" The tolerability profile demonstrated in the dose escalation schedule of lenalidomide suggests the dosing of lenalidomide to be 25 mg daily on days 1-21 with standard dosing of gemcitabine and merits further evaluation in a phase II trial."( A phase I dose-escalation study of lenalidomide in combination with gemcitabine in patients with advanced pancreatic cancer.
Liljefors, M; Rossmann, E; Ullenhag, GJ, 2015
)
0.92
"The biologic endpoint of full KIR occupancy was achieved across the IPH2101 dosing interval."( A Phase I Trial of the Anti-KIR Antibody IPH2101 and Lenalidomide in Patients with Relapsed/Refractory Multiple Myeloma.
Andre, P; Benson, DM; Caligiuri, MA; Cohen, AD; Efebera, YA; Hofmeister, CC; Jagannath, S; Munshi, NC; Spitzer, G; Zerbib, R, 2015
)
0.67
" The 90 % CI for the ratio of area under the INR curve from time zero until 144 hours after dosing (AUCINR, 0-144) or the peak INR geometric means between co-administration with lenalidomide versus placebo was also within the 85-125 % bounds."( Evaluation of pharmacokinetic and pharmacodynamic interactions when lenalidomide is co-administered with warfarin in a randomized clinical trial setting.
Chen, N; Knight, R; Palmisano, M; Weiss, D; Zhou, S, 2015
)
0.84
" Alternate approaches such as sequential dosing need to be evaluated when considering novel combination strategies for myelofibrosis."( Ruxolitinib in combination with lenalidomide as therapy for patients with myelofibrosis.
Borthakur, G; Cortes, J; Daver, N; Jabbour, E; Kadia, T; Kantarjian, H; Newberry, K; Pemmaraju, N; Pierce, S; Ravandi, F; Sasaki, K; Verstovsek, S; Wang, X; Zhou, L, 2015
)
0.7
" We point the attention on open issues, including drug dosage and administration schedule, prediction of clinical response to lenalidomide, and combination therapeutic strategies."( Lenalidomide in chronic lymphocytic leukemia: the present and future in the era of tyrosine kinase inhibitors.
Colaci, E; Fiorcari, S; Luppi, M; Maffei, R; Marasca, R; Martinelli, S; Potenza, L, 2016
)
2.08
" This analysis of the pivotal phase 3 FIRST trial examined the impact of renally adapted dosing of lenalidomide and dexamethasone on outcomes of patients with different degrees of renal impairment."( Impact of renal impairment on outcomes with lenalidomide and dexamethasone treatment in the FIRST trial, a randomized, open-label phase 3 trial in transplant-ineligible patients with multiple myeloma.
Belhadj, K; Bensinger, W; Chen, G; Cheung, MC; Derigs, HG; Dib, M; Dimopoulos, MA; Eom, H; Ervin-Haynes, A; Facon, T; Gamberi, B; Hall, R; Jaccard, A; Jardel, H; Karlin, L; Kolb, B; Lenain, P; Leupin, N; Liu, T; Marek, J; Rigaudeau, S; Roussel, M; Schots, R; Tosikyan, A; Van der Jagt, R, 2016
)
0.91
" To determine the optimal dosing schedule of once weekly bortezomib (BTZ) combined with lenalidomide (LEN) and dexamethasone (DEX), especially in the outpatient setting, we conducted a phase I dose escalation study."( Phase I study of once weekly treatment with bortezomib in combination with lenalidomide and dexamethasone for relapsed or refractory multiple myeloma.
Hagiwara, S; Iida, S; Ishida, T; Ito, A; Kanamori, T; Kato, C; Kinoshita, S; Komatsu, H; Kusumoto, S; Masuda, A; Murakami, S; Nakashima, T; Narita, T; Ri, M; Suzuki, N; Totani, H; Yoshida, T, 2016
)
0.89
" To establish safety, lenalidomide dosing in this combination was escalated in a conventional 3 + 3 design (15, 20 and 25 mg daily for 2 weeks followed by 1 week off)."( Phase II trial of docetaxel, bevacizumab, lenalidomide and prednisone in patients with metastatic castration-resistant prostate cancer.
Adesunloye, BA; Arlen, PM; Beedie, SL; Chen, C; Chun, G; Cordes, L; Couvillon, A; Dahut, WL; Dawson, NA; Figg, WD; Gulley, JL; Harold, N; Huang, X; Karzai, FH; Lee, MJ; Lee, S; Madan, RA; McLeod, DG; Ning, YM; Rosner, I; Sissung, T; Steinberg, SM; Theoret, MR; Tomita, Y; Trepel, JB, 2016
)
1.01
" A significant correlation was observed between platelet (Plt) count prior to the start of Len therapy (pre-treatment Plt) and the difference between pre-treatment Plt and the minimum Plt up to the point in time of treatment discontinuation, prolongation, or dosage reduction (min-Plt) (r=0."( The Establishment of Indicators of Thrombocytopenia in Patients Receiving Lenalidomide Therapy.
Goto, C; Goto, H; Ichihashi, A; Kasahara, S; Nagaya, K; Osawa, T; Tachi, T; Takahashi, T; Teramachi, H; Umeda, M; Yasuda, M, 2015
)
0.65
" The present review examines the drug's pharmacokinetics, discusses the main adverse renal effects that are associated with lenalidomide treatment, and makes recommendations for dosage adjustment in patients with underlying renal impairment."( [Lenalidomide nephrotoxicity].
Izzedine, H; Kheder El-Fekih, R, 2016
)
1.55
" Conventionally dosed lenalidomide has activity in 5q-MDS."( A study of high-dose lenalidomide induction and low-dose lenalidomide maintenance therapy for patients with hypomethylating agent refractory myelodysplastic syndrome.
Cashen, AF; Cherian, MA; DiPersio, JF; Fiala, M; Fletcher, T; Gao, F; Jacoby, MA; Stockerl-Goldstein, K; Tibes, R; Uy, GL; Vij, R; Westervelt, P, 2016
)
1.07
" Dosing was based on the lenalidomide label."( Pharmacokinetics, safety, and efficacy of lenalidomide plus dexamethasone in patients with multiple myeloma and renal impairment.
Abraham, J; Arnulf, B; Bridoux, F; Chen, N; Desport, E; Fermand, JP; Jaccard, A; Moreau, S; Moumas, E, 2016
)
1
" When co-administrated with anti-CD20 mAbs, dosage of lenalidomide was not the key factor of ORR in combination therapy."( Efficacy of lenalidomide in relapsed/refractory chronic lymphocytic leukemia patient: a systematic review and meta-analysis.
Hu, X; Liang, L; Liu, H; Yang, LP; Zhao, M; Zhu, YC, 2016
)
1.06
" Overall, lenalidomide is a suitable therapeutic option for R/R DLBCL, especially in non-GCB DLBCL, and 25 mg/day dosing should be preferred."( Lenalidomide in Relapsed or Refractory Diffuse Large B-Cell Lymphoma: Is It a Valid Treatment Option?
Cuzzocrea, S; Ferrero, S; Ghione, P; Marabese, A; Mian, M; Mondello, P; Pitini, V; Steiner, N; Willenbacher, W, 2016
)
2.28
" This prompted us to explore the concept of less intense drug dosing with shorter intervals between courses with the aim of preventing inter-course relapse."( Dose-dense and less dose-intense Total Therapy 5 for gene expression profiling-defined high-risk multiple myeloma.
Alapat, D; Avery, D; Bailey, C; Barlogie, B; Crowley, J; Epstein, J; Heuck, CJ; Hoering, A; Jethava, Y; Khan, R; Mitchell, A; Morgan, G; Petty, N; Sawyer, J; Schinke, C; Smith, R; Stein, C; Steward, D; Thanendrarajan, S; Tian, E; van Rhee, F; Waheed, S; Yaccoby, S; Zangari, M, 2016
)
0.43
" Continous high dosing schedules are poorly tolerated and minimally active."( A phase 2 trial of high dose lenalidomide in patients with relapsed/refractory higher-risk myelodysplastic syndromes and acute myeloid leukaemia with trilineage dysplasia.
Carraway, HE; DeZern, A; Gojo, I; Gore, SD; Smith, BD; Zeidan, AM, 2017
)
0.75
" The developed model was used to simulate dose schedules in order to explore the need of different dosing regimens in patients with different covariate values."( Population pharmacokinetics of lenalidomide in multiple myeloma patients.
Climente-Martí, M; Guchelaar, HJ; Guglieri-López, B; Merino-Sanjuán, M; Moes, DJ; Pérez-Pitarch, A; Porta-Oltra, B, 2017
)
0.74
" Dosing comprised elotuzumab 10 mg/kg intravenously (weekly, Cycles 1-2; biweekly, Cycles 3+), lenalidomide 25 mg (daily, Days 1-21), and dexamethasone (28 mg orally and 8 mg intravenously, weekly, Cycles 1-2; 40 mg orally, weekly, Cycles 3+), in 28-day cycles."( A phase 2 safety study of accelerated elotuzumab infusion, over less than 1 h, in combination with lenalidomide and dexamethasone, in patients with multiple myeloma.
Badarinath, S; Berenson, J; Cartmell, A; Harb, W; Lyons, R; Manges, R; McIntyre, K; Mohamed, H; Nourbakhsh, A; Rifkin, R, 2017
)
0.89
" Median duration of dosing was 36."( A phase 1b study of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma.
Baz, R; Benson, DM; Campana, F; Charpentier, E; Lendvai, N; Lesokhin, AM; Martin, T; Munster, P; Vij, R; Wack, C; Wolf, J, 2017
)
0.73
" Population pharmacokinetic data from 375 patients demonstrated weight-based dosing is appropriate for elotuzumab, and that ethnicity and hepatic/renal function have minimal effects on exposure."( The Clinical Pharmacology of Elotuzumab.
Bello, A; Dodge, R; Gupta, M; Mora, J; Passey, C; Robbins, M; Roy, A; Sheng, J; Tendolkar, A, 2018
)
0.48
" The basis of the RI treatment in MM is bortizomib-based regimen, which does not require dosage adjustment in patients with dialysis or renal insufficiency."( [Expert consensus for the diagnosis and treatment of patients with renal impairment of multiple myeloma].
, 2017
)
0.46
" Although the POM dosage was reduced to 1-2 mg/day due to somnolence, which was reported as an adverse event, stringent complete response (sCR) was achieved and sustained for 10 months following 11 cycles of low-dose POM monotherapy."( [Achievement of a stringent complete response with low-dose pomalidomide monotherapy in a multiple myeloma patient].
Endo, T; Hamada, T; Hatta, Y; Iriyama, N; Koike, T; Kurihara, K; Miura, K; Nakagawa, M; Otake, S; Sato, H; Takahashi, H; Takei, M; Uchino, Y,
)
0.13
" Lenalidomide was administered at standard dosing and in combination with corticosteroids and/or chemotherapy."( Retreatment with lenalidomide is an effective option in heavily pretreated refractory multiple myeloma patients.
Adam, Z; Brozova, L; Kral, Z; Krejci, M; Pour, L; Sandecka, V; Sevcikova, S; Stork, M; Velichova, R,
)
1.38
" We compared eGFR-dependent CKD staging and lenalidomide dosage assignments."( GFR estimation in lenalidomide treatment of multiple myeloma patients: a prospective cohort study.
Engelhardt, M; Grünewald, J; Ihorst, G; Kleber, M; Reinhardt, H; Schmidts, A; Terpos, E; Walz, G; Wäsch, R; Zschiedrich, S, 2019
)
1.11
" CKD-EPI assigned 3% of patients to higher lenalidomide dosing as opposed to MDRD."( GFR estimation in lenalidomide treatment of multiple myeloma patients: a prospective cohort study.
Engelhardt, M; Grünewald, J; Ihorst, G; Kleber, M; Reinhardt, H; Schmidts, A; Terpos, E; Walz, G; Wäsch, R; Zschiedrich, S, 2019
)
1.11
"In our multiple myeloma patient cohort, CKD-EPI and MDRD led to similar CKD staging with minor differences in lenalidomide dosage assignment."( GFR estimation in lenalidomide treatment of multiple myeloma patients: a prospective cohort study.
Engelhardt, M; Grünewald, J; Ihorst, G; Kleber, M; Reinhardt, H; Schmidts, A; Terpos, E; Walz, G; Wäsch, R; Zschiedrich, S, 2019
)
1.06
" Using various dosing schedules, the majority of patients (684/694) received daratumumab at a dose of 16 mg/kg."( Pharmacokinetics and Exposure-Response Analyses of Daratumumab in Combination Therapy Regimens for Patients with Multiple Myeloma.
Belch, A; Capra, M; Clemens, PL; Dimopoulos, MA; Gomez, D; Ho, PJ; Iida, S; Jansson, R; Leiba, M; Medvedova, E; Min, CK; Schecter, J; Sonneveld, P; Sun, YN; Xu, XS; Zhang, L, 2018
)
0.48
"These data support the recommended 16 mg/kg dose of daratumumab and the respective dosing schedules in the POLLUX and CASTOR pivotal studies."( Pharmacokinetics and Exposure-Response Analyses of Daratumumab in Combination Therapy Regimens for Patients with Multiple Myeloma.
Belch, A; Capra, M; Clemens, PL; Dimopoulos, MA; Gomez, D; Ho, PJ; Iida, S; Jansson, R; Leiba, M; Medvedova, E; Min, CK; Schecter, J; Sonneveld, P; Sun, YN; Xu, XS; Zhang, L, 2018
)
0.48
" Mice dosed with INCB (30 mg/kg) showed significant reductions in tumor volume on days 28, 35, 42, 49, 56, and 63."( The anti-myeloma effects of the selective JAK1 inhibitor (INCB052793) alone and in combination in vitro and in vivo.
Berenson, JR; Chen, H; Hekmati, T; Li, M; Nosrati, JD; Patil, S; Sanchez, E; Schlossberg, RE; Soof, CM; Tanenbaum, EJ; Tang, G; Vidisheva, A; Wang, C; Zahab, B, 2019
)
0.51
" Patient adherence was calculated by dividing the number of bottle-uncapping events by the total number of doses supplied for each dosing cycle."( Effect of a Smart Pill Bottle and Pharmacist Intervention on Medication Adherence in Patients with Multiple Myeloma New to Lenalidomide Therapy.
Mathews, KB; Mauro, J; Sredzinski, ES, 2019
)
0.72
" Patients (N = 569) with ≥1 prior line received Rd (lenalidomide, 25 mg, on Days 1-21 of each 28-day cycle; dexamethasone, 40 mg, weekly) ± daratumumab at the approved dosing schedule."( Daratumumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a randomized, open-label, phase 3 study.
Bahlis, NJ; Benboubker, L; Chiu, C; Cook, G; Dimopoulos, MA; Ho, PJ; Kaufman, JL; Kim, K; Krevvata, M; Leiba, M; Moreau, P; Okonkwo, L; Qi, M; Qin, X; San-Miguel, J; Takezako, N; Trivedi, S; Ukropec, J; White, DJ, 2020
)
1.15
" Splitting the first intravenous infusion of daratumumab over 2 days is an approved alternative dosing regimen to reduce the duration of the first infusion and provide flexibility for patients and healthcare providers."( Split First Dose Administration of Intravenous Daratumumab for the Treatment of Multiple Myeloma (MM): Clinical and Population Pharmacokinetic Analyses.
Bladé, J; Clemens, PL; Deraedt, W; Jakubowiak, A; Krishnan, A; Lonial, S; Luo, M; Moreau, P; Nnane, I; Oriol, A; Qi, M; Sun, YN; Ukropec, J; Usmani, SZ; Xu, XS; Zhou, H, 2020
)
0.56
" The population PK simulations demonstrated virtually identical PK profiles after the first day of treatment for all approved indications and recommended dosing schedules of daratumumab."( Split First Dose Administration of Intravenous Daratumumab for the Treatment of Multiple Myeloma (MM): Clinical and Population Pharmacokinetic Analyses.
Bladé, J; Clemens, PL; Deraedt, W; Jakubowiak, A; Krishnan, A; Lonial, S; Luo, M; Moreau, P; Nnane, I; Oriol, A; Qi, M; Sun, YN; Ukropec, J; Usmani, SZ; Xu, XS; Zhou, H, 2020
)
0.56
" In this paper, we utilize the available clinical trial evidence to provide recommendations for patient selection and lenalidomide dosing in both the first-line setting in patients ineligible for ASCT and the maintenance setting in patients who have undergone ASCT."( The clinical management of lenalidomide-based therapy in patients with newly diagnosed multiple myeloma.
Dechow, T; Hackanson, B; Knop, S; Merz, M; Scheytt, M; Schmidt, C, 2020
)
1.06
" We evaluated the pharmacokinetics (PK) and safety of lenalidomide and dexamethasone as frontline pre-transplant induction, with doses adjusted at start of each cycle based on creatinine clearance, as per the official dosing guidelines."( An open-label, pharmacokinetic study of lenalidomide and dexamethasone therapy in previously untreated multiple myeloma (MM) patients with various degrees of renal impairment - validation of official dosing guidelines.
Cao, Y; Chen, CI; Chen, E; Chen, H; Kakar, S; Kukreti, V; Lau, A; Le, LW; Levina, O; Paul, H; Prica, A; Reece, DE; Tiedemann, R; Trudel, S, 2020
)
1.07
" The most appropriate delivery and dosing regimens of these therapies for patients at advanced age and frailty status is also unclear."( Optimising the value of immunomodulatory drugs during induction and maintenance in transplant ineligible patients with newly diagnosed multiple myeloma: results from Myeloma XI, a multicentre, open-label, randomised, Phase III trial.
Cairns, DA; Collett, C; Cook, G; Davies, FE; Drayson, MT; Garg, M; Gregory, WM; Jackson, GH; Jenner, MW; Jones, JR; Kaiser, MF; Karunanithi, K; Kishore, B; Lindsay, J; Morgan, GJ; Owen, RG; Pawlyn, C; Russell, NH; Striha, A; Taylor, C; Waterhouse, A; Williams, CD; Wilson, J, 2021
)
0.62
" Key model drivers included subsequent therapies, dosing schedule, and time horizon."( Lenalidomide as maintenance treatment for patients with multiple myeloma after autologous stem cell transplantation: A pharmaco-economic assessment.
Boersma, J; Dhanasiri, S; Lee, D; Ramsden, R; Uyl-de Groot, CA; Zweegman, S, 2020
)
2
" Here, we investigated the safety and effectiveness of ixazomib dosing schedules."( Safety and Effectiveness of Ixazomib Dose-escalating Strategy in Ixazomib-Lenalidomide-Dexamethasone Treatment for Relapsed/Refractory Multiple Myeloma.
Boku, S; Higai, K; Kagoo, T; Niijima, D; Ogawa, C; Ohashi, Y; Tani, K; Ueno, H; Yachi, Y; Yano, T; Yatabe, M; Yokoyama, A,
)
0.36
"A dose-escalation strategy to optimise ixazomib dosing may reduce treatment interruption due to adverse events without compromising its antitumor activity."( Safety and Effectiveness of Ixazomib Dose-escalating Strategy in Ixazomib-Lenalidomide-Dexamethasone Treatment for Relapsed/Refractory Multiple Myeloma.
Boku, S; Higai, K; Kagoo, T; Niijima, D; Ogawa, C; Ohashi, Y; Tani, K; Ueno, H; Yachi, Y; Yano, T; Yatabe, M; Yokoyama, A,
)
0.36
" Though peripheral sensory neuropathy was more frequent with twice weekly dosing (P = ."( Outcomes with different administration schedules of bortezomib in bortezomib, lenalidomide and dexamethasone (VRd) as first-line therapy in multiple myeloma.
Buadi, F; Cook, J; Dingli, D; Dispenzieri, A; Fonder, A; Gertz, MA; Go, R; Gonsalves, W; Hayman, S; Higgins, A; Hobbs, M; Hwa, YL; Johnson, I; Kapoor, P; Kourelis, T; Kumar, S; Kyle, R; Lacy, M; Leung, N; Rajkumar, VS; Sidana, S; Warsame, R, 2021
)
0.85
"Our simpler and slow desensitization protocol, which desensitizes the patients without reducing the effect of LEN, includes drug holidays, similar to the usual LEN dosing schedule, and moreover is recommended as a treatment option especially for elderly patients with no housemate to help with medical management."( Simple desensitization protocol for multiple myeloma patients with lenalidomide-induced skin rash: Case series.
Hagihara, M; Inoue, M; Mita, M; Ohara, S; Sugi, T; Uchida, T; Yasu, T, 2021
)
0.86
" Therefore, we investigated whether LEN-induced skin rash is affected by the duration of BOR administration and the dosing interval between BOR and LEN administration."( Preceding bortezomib administration for a certain period reduces the risk of lenalidomide-induced skin rash.
Hagihara, M; Hanai, H; Inoue, M; Kubo, K; Kushi, R; Mita, M; Ohara, S; Sugi, T; Uchida, T; Yasu, T, 2022
)
0.95
" Developed models supported the phase III isatuximab dosing regimen selection/confirmation of 10 mg/kg qw/q2w for use in combination with pomalidomide/dexamethasone in patients with RRMM."( Exposure-response analyses for selection/confirmation of optimal isatuximab dosing regimen in combination with pomalidomide/dexamethasone treatment in patients with multiple myeloma.
Brillac, C; Fau, JB; Gaudel-Dedieu, N; Koiwai, K; Nguyen, L; Rachedi, F; Sebastien, B; Semiond, D; Thai, HT; van de Velde, H; Veyrat-Follet, C, 2022
)
0.72
"Lenalidomide is an immunomodulatory drug used to treat multiple myeloma that requires renal dosing adjustment based on Cockcroft-Gault (CG)."( Estimation of Kidney Function in Patients With Multiple Myeloma: Implications for Lenalidomide Dosing.
Gonsalves, WI; Lam, S; M Saunders, I; Momper, JD; Salama, E; Tzachanis, D, 2023
)
2.58
"All participants will receive IRD induction with the dosing strategy randomised (1:1) at trial entry."(
Best, P; Bird, J; Bowcock, S; Boyd, K; Cairns, DA; Cook, G; Coulson, AB; Dawkins, B; de Tute, R; Drayson, M; Gardner, H; Henderson, R; Hockaday, A; Jackson, G; Jenner, M; Jones, J; Kaiser, M; Kishore, B; Meads, D; Olivier, C; Owen, R; Parrish, C; Pawlyn, C; Rabin, N; Royle, KL, 2022
)
0.72
" Maintenance lenalidomide dosing practices vary amongst physicians and current literature lacks comparisons on intermittent versus continuous dosing."( Safety and tolerability of lenalidomide maintenance dosing in patients with multiple myeloma post-autologous stem cell transplant.
Davis, JA; Gaffney, KJ; Hashmi, H; Shockley, A; Smith, D; Weeda, E, 2022
)
1.39
" The primary objective was to determine the incidence of dose modification, defined as any dosage reduction, delay in treatment, or discontinuation of therapy."( Safety and tolerability of lenalidomide maintenance dosing in patients with multiple myeloma post-autologous stem cell transplant.
Davis, JA; Gaffney, KJ; Hashmi, H; Shockley, A; Smith, D; Weeda, E, 2022
)
1.02
" Patients who received continuous dosing appeared to have a higher incidence of adverse events when compared to intermittent dosing with the most common adverse events being neutropenia, fatigue, and rash."( Safety and tolerability of lenalidomide maintenance dosing in patients with multiple myeloma post-autologous stem cell transplant.
Davis, JA; Gaffney, KJ; Hashmi, H; Shockley, A; Smith, D; Weeda, E, 2022
)
1.02
" Further studies need to be conducted to understand the impact of dosing strategies of anti-MM agents in the real world."( Real-world data on lenalidomide dosing and outcomes in patients newly diagnosed with multiple myeloma: Results from the Canadian Myeloma Research Group Database.
Aslam, M; Gul, E; Jimenez-Zepeda, VH; Kardjadj, M; Kotb, R; LeBlanc, R; Louzada, M; Masih-Khan, E; McCurdy, A; Mian, H; Reece, D; Reiman, A; Sebag, M; Song, K; Stakiw, J; Venner, CP; White, D, 2023
)
1.24
" The protection displayed a dose-response pattern roughly."( [Study on the Therapeutic Effect of Lenalidomide on Hemophilic Arthropathy].
Hua, BL; Lin, ZY; Sun, JJ; Wang, YF; Wu, X; Xiao, X; Zhang, FX; Zhou, XY, 2022
)
1
" Variability in patient baseline characteristics, such as the number of prior lines of treatment, refractoriness to prior treatments, prior stem cell transplant, and timing and dosing of prior lenalidomide, makes it difficult to select the best options for patients with RRMM for whom first-line treatments have failed."( Clinical evidence for immune-based strategies in early-line multiple myeloma: current challenges in decision-making for subsequent therapy.
Iida, S; Mateos, MV; Raje, N; Reece, D, 2023
)
1.1
" Lenalidomide was dosed at 20 mg daily from days 1 to 21 of each 28-day cycle."( Adding venetoclax to lenalidomide and rituximab is safe and effective in patients with untreated mantle cell lymphoma.
Bond, D; Boonstra, P; Carty, SA; Danilov, AV; Herrera, AF; Kaminski, MS; Kandarpa, M; Karimi, YH; Kump, K; Maddocks, K; Mayer, TL; Nachar, V; Nikolovska-Coleska, Z; Phillips, TJ; Popplewell, L; Takiar, R; Wilcox, RA, 2023
)
2.14
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (3)

RoleDescription
angiogenesis inhibitorAn agent and endogenous substances that antagonize or inhibit the development of new blood vessels.
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
immunomodulatorBiologically active substance whose activity affects or plays a role in the functioning of the immune system.
[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 (4)

ClassDescription
isoindoles
dicarboximideAn imide in which the two acyl substituents on nitrogen are carboacyl groups.
piperidones
aromatic amineAn amino compound in which the amino group is linked directly to an aromatic system.
[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 (15)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
EWS/FLI fusion proteinHomo sapiens (human)Potency7.07910.001310.157742.8575AID1259253; AID1259256
activating transcription factor 6Homo sapiens (human)Potency10.68220.143427.612159.8106AID1159516
Cellular tumor antigen p53Homo sapiens (human)Potency7.49780.002319.595674.0614AID651631
[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)
Cereblon isoform 4Magnetospirillum gryphiswaldenseKi3.10000.64203.94289.6000AID1569040
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)133.00000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)133.00000.20005.677410.0000AID1473741
Bromodomain-containing protein 4Homo sapiens (human)IC50 (µMol)5.19000.00040.40329.0500AID1658259
Bile salt export pumpHomo sapiens (human)IC50 (µMol)133.00000.11007.190310.0000AID1473738
DNA damage-binding protein 1Homo sapiens (human)IC50 (µMol)1.59530.28601.72773.0000AID1794852; AID1794857; AID1814571
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
Protein cereblonHomo sapiens (human)IC50 (µMol)7.68100.28601.70663.0000AID1387868; AID1685005; AID1794852; AID1794853; AID1794857; AID1814571
Protein cereblonHomo sapiens (human)Ki8.89501.49006.580010.0000AID1658260; AID1685005
[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)
DNA-binding protein IkarosHomo sapiens (human)EC50 (µMol)0.06700.02400.04550.0670AID1387871
Protein cereblonHomo sapiens (human)EC50 (µMol)0.07700.00900.03650.0870AID1387871; AID1387872
Zinc finger protein AiolosHomo sapiens (human)EC50 (µMol)0.08700.02200.05450.0870AID1387872
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
DNA-binding protein IkarosHomo sapiens (human)DC500.09000.02670.10310.1927AID1769486
DNA-binding protein IkarosHomo sapiens (human)fEC500.45000.11100.28050.4500AID1769475
Protein cereblonHomo sapiens (human)DC505.04500.00800.48352.1000AID1769486; AID1772713
Protein cereblonHomo sapiens (human)fEC500.45000.11100.28050.4500AID1769475
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (334)

Processvia Protein(s)Taxonomy
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)
regulation of transcription by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
positive regulation of G2/M transition of mitotic cell cycleBromodomain-containing protein 4Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
chromatin organizationBromodomain-containing protein 4Homo sapiens (human)
DNA damage responseBromodomain-containing protein 4Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionBromodomain-containing protein 4Homo sapiens (human)
positive regulation of DNA-templated transcriptionBromodomain-containing protein 4Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
regulation of inflammatory responseBromodomain-containing protein 4Homo sapiens (human)
positive regulation of T-helper 17 cell lineage commitmentBromodomain-containing protein 4Homo 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)
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)
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)
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)
chromatin organizationDNA-binding protein IkarosHomo sapiens (human)
mesoderm developmentDNA-binding protein IkarosHomo sapiens (human)
lymphocyte differentiationDNA-binding protein IkarosHomo sapiens (human)
erythrocyte differentiationDNA-binding protein IkarosHomo sapiens (human)
negative regulation of DNA-templated transcriptionDNA-binding protein IkarosHomo sapiens (human)
regulation of transcription by RNA polymerase IIDNA-binding protein IkarosHomo sapiens (human)
proteasomal protein catabolic processDNA damage-binding protein 1Homo sapiens (human)
nucleotide-excision repairDNA damage-binding protein 1Homo sapiens (human)
ubiquitin-dependent protein catabolic processDNA damage-binding protein 1Homo sapiens (human)
apoptotic processDNA damage-binding protein 1Homo sapiens (human)
DNA damage responseDNA damage-binding protein 1Homo sapiens (human)
spindle assembly involved in female meiosisDNA damage-binding protein 1Homo sapiens (human)
Wnt signaling pathwayDNA damage-binding protein 1Homo sapiens (human)
protein ubiquitinationDNA damage-binding protein 1Homo sapiens (human)
viral release from host cellDNA damage-binding protein 1Homo sapiens (human)
cellular response to UVDNA damage-binding protein 1Homo sapiens (human)
ectopic germ cell programmed cell deathDNA damage-binding protein 1Homo sapiens (human)
regulation of circadian rhythmDNA damage-binding protein 1Homo sapiens (human)
negative regulation of apoptotic processDNA damage-binding protein 1Homo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processDNA damage-binding protein 1Homo sapiens (human)
epigenetic programming in the zygotic pronucleiDNA damage-binding protein 1Homo sapiens (human)
positive regulation of viral genome replicationDNA damage-binding protein 1Homo sapiens (human)
positive regulation of gluconeogenesisDNA damage-binding protein 1Homo sapiens (human)
positive regulation of protein catabolic processDNA damage-binding protein 1Homo sapiens (human)
positive regulation by virus of viral protein levels in host cellDNA damage-binding protein 1Homo sapiens (human)
rhythmic processDNA damage-binding protein 1Homo sapiens (human)
negative regulation of developmental processDNA damage-binding protein 1Homo sapiens (human)
biological process involved in interaction with symbiontDNA damage-binding protein 1Homo sapiens (human)
UV-damage excision repairDNA damage-binding protein 1Homo sapiens (human)
regulation of mitotic cell cycle phase transitionDNA damage-binding protein 1Homo sapiens (human)
negative regulation of reproductive processDNA damage-binding protein 1Homo sapiens (human)
DNA repairDNA damage-binding protein 1Homo 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 ubiquitinationProtein cereblonHomo sapiens (human)
positive regulation of Wnt signaling pathwayProtein cereblonHomo sapiens (human)
negative regulation of protein-containing complex assemblyProtein cereblonHomo sapiens (human)
positive regulation of protein-containing complex assemblyProtein cereblonHomo sapiens (human)
negative regulation of monoatomic ion transmembrane transportProtein cereblonHomo sapiens (human)
locomotory exploration behaviorProtein cereblonHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processProtein cereblonHomo sapiens (human)
mesoderm developmentZinc finger protein AiolosHomo sapiens (human)
response to bacteriumZinc finger protein AiolosHomo sapiens (human)
B cell differentiationZinc finger protein AiolosHomo sapiens (human)
T cell differentiationZinc finger protein AiolosHomo sapiens (human)
regulation of B cell proliferationZinc finger protein AiolosHomo sapiens (human)
regulation of apoptotic processZinc finger protein AiolosHomo sapiens (human)
regulation of B cell differentiationZinc finger protein AiolosHomo sapiens (human)
regulation of lymphocyte differentiationZinc finger protein AiolosHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIZinc finger protein AiolosHomo sapiens (human)
regulation of transcription by RNA polymerase IIZinc finger protein AiolosHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (102)

Processvia Protein(s)Taxonomy
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)
transcription cis-regulatory region bindingBromodomain-containing protein 4Homo sapiens (human)
p53 bindingBromodomain-containing protein 4Homo sapiens (human)
chromatin bindingBromodomain-containing protein 4Homo sapiens (human)
transcription coregulator activityBromodomain-containing protein 4Homo sapiens (human)
transcription coactivator activityBromodomain-containing protein 4Homo sapiens (human)
protein bindingBromodomain-containing protein 4Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityBromodomain-containing protein 4Homo sapiens (human)
enzyme bindingBromodomain-containing protein 4Homo sapiens (human)
lysine-acetylated histone bindingBromodomain-containing protein 4Homo sapiens (human)
RNA polymerase II C-terminal domain bindingBromodomain-containing protein 4Homo sapiens (human)
P-TEFb complex bindingBromodomain-containing protein 4Homo sapiens (human)
histone reader activityBromodomain-containing protein 4Homo 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)
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)
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 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)
DNA bindingDNA-binding protein IkarosHomo sapiens (human)
protein bindingDNA-binding protein IkarosHomo sapiens (human)
protein domain specific bindingDNA-binding protein IkarosHomo sapiens (human)
metal ion bindingDNA-binding protein IkarosHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingDNA-binding protein IkarosHomo sapiens (human)
DNA-binding transcription factor activityDNA-binding protein IkarosHomo sapiens (human)
damaged DNA bindingDNA damage-binding protein 1Homo sapiens (human)
DNA bindingDNA damage-binding protein 1Homo sapiens (human)
protein bindingDNA damage-binding protein 1Homo sapiens (human)
protein-macromolecule adaptor activityDNA damage-binding protein 1Homo sapiens (human)
protein-containing complex bindingDNA damage-binding protein 1Homo sapiens (human)
WD40-repeat domain bindingDNA damage-binding protein 1Homo sapiens (human)
cullin family protein bindingDNA damage-binding protein 1Homo sapiens (human)
ubiquitin ligase complex scaffold activityDNA damage-binding protein 1Homo 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 bindingProtein cereblonHomo sapiens (human)
transmembrane transporter bindingProtein cereblonHomo sapiens (human)
metal ion bindingProtein cereblonHomo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificZinc finger protein AiolosHomo sapiens (human)
protein bindingZinc finger protein AiolosHomo sapiens (human)
identical protein bindingZinc finger protein AiolosHomo sapiens (human)
protein homodimerization activityZinc finger protein AiolosHomo sapiens (human)
histone deacetylase bindingZinc finger protein AiolosHomo sapiens (human)
sequence-specific DNA bindingZinc finger protein AiolosHomo sapiens (human)
metal ion bindingZinc finger protein AiolosHomo sapiens (human)
protein heterodimerization activityZinc finger protein AiolosHomo sapiens (human)
promoter-specific chromatin bindingZinc finger protein AiolosHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingZinc finger protein AiolosHomo sapiens (human)
DNA-binding transcription factor activityZinc finger protein AiolosHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (55)

Processvia Protein(s)Taxonomy
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)
condensed nuclear chromosomeBromodomain-containing protein 4Homo sapiens (human)
nucleusBromodomain-containing protein 4Homo sapiens (human)
nucleoplasmBromodomain-containing protein 4Homo 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)
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)
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)
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)
nucleusDNA-binding protein IkarosHomo sapiens (human)
nucleoplasmDNA-binding protein IkarosHomo sapiens (human)
cytosolDNA-binding protein IkarosHomo sapiens (human)
pericentric heterochromatinDNA-binding protein IkarosHomo sapiens (human)
protein-containing complexDNA-binding protein IkarosHomo sapiens (human)
nucleusDNA damage-binding protein 1Homo sapiens (human)
nucleolusDNA damage-binding protein 1Homo sapiens (human)
chromosome, telomeric regionDNA damage-binding protein 1Homo sapiens (human)
extracellular spaceDNA damage-binding protein 1Homo sapiens (human)
nucleusDNA damage-binding protein 1Homo sapiens (human)
nucleoplasmDNA damage-binding protein 1Homo sapiens (human)
cytoplasmDNA damage-binding protein 1Homo sapiens (human)
Cul4A-RING E3 ubiquitin ligase complexDNA damage-binding protein 1Homo sapiens (human)
extracellular exosomeDNA damage-binding protein 1Homo sapiens (human)
Cul4B-RING E3 ubiquitin ligase complexDNA damage-binding protein 1Homo sapiens (human)
protein-containing complexDNA damage-binding protein 1Homo sapiens (human)
Cul4-RING E3 ubiquitin ligase complexDNA damage-binding protein 1Homo sapiens (human)
site of double-strand breakDNA damage-binding protein 1Homo 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)
nucleusProtein cereblonHomo sapiens (human)
cytoplasmProtein cereblonHomo sapiens (human)
cytosolProtein cereblonHomo sapiens (human)
membraneProtein cereblonHomo sapiens (human)
perinuclear region of cytoplasmProtein cereblonHomo sapiens (human)
Cul4A-RING E3 ubiquitin ligase complexProtein cereblonHomo sapiens (human)
nucleusZinc finger protein AiolosHomo sapiens (human)
nucleoplasmZinc finger protein AiolosHomo sapiens (human)
cytoplasmZinc finger protein AiolosHomo sapiens (human)
cytosolZinc finger protein AiolosHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (253)

Assay IDTitleYearJournalArticle
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.
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.
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.
AID1794857TR-FRET cereblon binding assay from Article 10.1021/acs.jmedchem.6b01921: \\A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.\\2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID1794852Fluorescence thermal melt assay from Article 10.1038/leu.2012.119: \\Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide.\\2012Leukemia, Nov, Volume: 26, Issue:11
Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide.
AID1794853Thalidomide analog bead assay from Article 10.1038/leu.2012.119: \\Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide.\\2012Leukemia, Nov, Volume: 26, Issue:11
Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide.
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.
AID1355084Cytotoxicity against human WI38 cells after 72 hrs by MTS assay
AID604331Toxicity against mouse RAW264.7 cells assessed as cell viability at 30 uM by Cell titer 96 squeous one solution cell proliferation assay untreated control2011Bioorganic & medicinal chemistry, Jul-01, Volume: 19, Issue:13
Design, synthesis and biological assessment of novel N-substituted 3-(phthalimidin-2-yl)-2,6-dioxopiperidines and 3-substituted 2,6-dioxopiperidines for TNF-α inhibitory activity.
AID327425Upregulation of CYP4F3 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1769476Drug metabolism in po dosed healthy human assessed as 5-hydroxy-lenalidomide metabolite in plasma by LC-MS/MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID327434Down-regulation of HBA2 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID726391Inhibition of IL-2 production in human T cells measured after 2 to 3 days by ELISA2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity.
AID327467Increase in SPARC gene expression in CD34- mononuclear cells from non-del(5q) myelodysplastic syndrome patient by RT-PCR after 2 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327397Inhibition of cell proliferation of CD34+ progenitor cells from myelodysplastic syndrome del(5q) patient after 14 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327410Upregulation of PPIC gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327411Upregulation of INHBA gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327405Upregulation of DEFA1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1667516Antiproliferative activity against lenalidomide-resistant human H929-R10-1 cells incubated for 5 days by CellTiter-Glo assay2020Journal of medicinal chemistry, 07-09, Volume: 63, Issue:13
Discovery of CRBN E3 Ligase Modulator CC-92480 for the Treatment of Relapsed and Refractory Multiple Myeloma.
AID327466Increase in activin A gene expression in erythroid progenitor cells from non-del(5q) myelodysplastic syndrome patient by RT-PCR after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID215445Inhibition of lipopolysaccharide stimulated TNF-alpha release in human whole blood1999Bioorganic & medicinal chemistry letters, Jun-07, Volume: 9, Issue:11
Amino-substituted thalidomide analogs: potent inhibitors of TNF-alpha production.
AID159803Inhibition of Phosphodiesterase 4 from U937 cells at 100 uM1999Bioorganic & medicinal chemistry letters, Jun-07, Volume: 9, Issue:11
Amino-substituted thalidomide analogs: potent inhibitors of TNF-alpha production.
AID1718348Down regulation of IKZF1 protein expression in human JJN-3 cells at 10 to 50 uM after 48 hrs by immunoblotting analysis2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1707351Half-life in human liver microsomes2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID327424Upregulation of ANGPT2 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1861179Antiproliferative activity against human MM1.S cells assessed as cell viability measured after 3 days by CCK-8 assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Structure-based rational design enables efficient discovery of a new selective and potent AKT PROTAC degrader.
AID327470Increase in activin A gene expression in CD34- mononuclear cells from non-del(5q) myelodysplastic syndrome patient by RT-PCR after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327404Upregulation of VSIG4 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327439Down-regulation of HBB gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1759175Inhibition of STAT3 phosphorylation in mouse BMOL-NT cells assessed as change in nuclear phosphorylated STAT3 level at 10 uM incubated for 6 hrs by immunofluorescence assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID327447Down-regulation of KLF1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1769481Metabolic stability in human liver microsome assessed as half-life at 1 uM in presence of NADPH incubated up to 60 mins by LC-MS/MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID1355085Cytotoxicity against human MCF10A cells after 72 hrs by MTS assay
AID1317723Growth inhibition of BMOL-NT cells2016European journal of medicinal chemistry, Sep-14, Volume: 120Identification of a thalidomide derivative that selectively targets tumorigenic liver progenitor cells and comparing its effects with lenalidomide and sorafenib.
AID327402Decrease in myeloid differentiation of CD34+ progenitor cells from myelodysplastic syndrome del(5q) patient assessed as CD33 expression after 14 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625288Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for jaundice2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1597308Antiproliferative activity against human Kasumi-1 cells measured after 168 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID1667542Induction of apoptosis in lenalidomide-resistant human H929-R10-1 cells assessed as caspase3 induction at 1 uM measured over 150 hrs by live cell imaging analysis2020Journal of medicinal chemistry, 07-09, Volume: 63, Issue:13
Discovery of CRBN E3 Ligase Modulator CC-92480 for the Treatment of Relapsed and Refractory Multiple Myeloma.
AID1759152Antiproliferative activity against mouse BMOL-T cells assessed as cell growth by measuring doubling time at 10 uM measured every 4 hrs for 3 days by IncuCyte zoom live cell analysis (Rvb = 14.3 to 15 hrs)2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1718341Growth inhibition of human KMM-1 cells at 10 uM after 48 hrs by PrestoBlue cell viability assay relative to control2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1387868Displacement of cy5-conjugated 2-((1E,3E,5Z)-5-(1-(6-((4-(2-((2-(2,6-Dioxopiperidin-3-yl)-1-oxoisoindolin-4-yl)oxy)acetamido)butyl)amino)-6-oxohexyl)-3,3-dimethylindolin-2-ylidene)penta-1,3-dien-1-yl)-1,3,3-trimethyl-3H-indol-1-ium from DDB1-fused human f2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID1688910Antiproliferative activity against human HCC827 cells harboring EGFRDel19 mutant assessed as reduction in cell viability incubated for 72 hrs by MTT assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Discovery of potent epidermal growth factor receptor (EGFR) degraders by proteolysis targeting chimera (PROTAC).
AID1597304Cytotoxicity against human PBMC cells by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID327455Down-regulation of ALAS2 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1569038Inhibition of CRBN-mediated CK1alpha degradation in human OPM2 cells measured after 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives.
AID327416Upregulation of CXCL1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327468Increase in activin A gene expression in CD34- mononuclear cells from non-del(5q) myelodysplastic syndrome patient by RT-PCR after 2 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327459Down-regulation of AMMECR1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1759155Inhibition of NFkB p65 phosphorylation in mouse BMOL-NT cells assessed as change in phosphorylated p65 level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID327417Upregulation of TPBG gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1597300Antiproliferative activity against human MM1S cells measured after 168 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID327418Upregulation of TPBG gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
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.
AID1769478Apparent permeability in in human Caco-2 cells at 10 uM incubated for 90 mins by LC-MS/MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID1759173Inhibition of Akt phosphorylation in mouse BMOL-NT cells assessed as change in nuclear phosphorylated Akt level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1718343Growth inhibition of human MM1.R cells at 100 uM after 48 hrs by PrestoBlue cell viability assay relative to control2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID257636Inhibitory activity in HUVEC tube formation assay at 100 uM2005Bioorganic & medicinal chemistry letters, Dec-15, Volume: 15, Issue:24
Angiogenesis inhibitors derived from thalidomide.
AID1685005Binding affinity to human CRBN-thalidomide binding domain expressed in Escherichia coli by measuring baseline corrected normalized fluorescence by MST based assay2021ACS medicinal chemistry letters, Jan-14, Volume: 12, Issue:1
Sweet and Blind Spots in E3 Ligase Ligand Space Revealed by a Thermophoresis-Based Assay.
AID327464Down-regulation of LRP11 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327426Upregulation of CYP4F3 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1387871Induction of cereblon-mediated ikaros degradation in human DF15 cells expressing ePL-tagged ikaros after 4 hrs by luminometric analysis2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID327420Upregulation of MAOA gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1769486Protac activity at CRBN/HiBiT-tagged IKZF1 in human MOLT-4 cells assessed as IKZF1 degradation by measuring luminescence by luminescence based HiBiT assay2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID1718350Down regulation of IRF4 protein expression in human MM1.R cells at 10 to 50 uM after 48 hrs by immunoblotting analysis2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID215453Inhibition of lipopolysaccharide stimulated TNF-alpha release in human PBMC at 100 uM1999Bioorganic & medicinal chemistry letters, Jun-07, Volume: 9, Issue:11
Amino-substituted thalidomide analogs: potent inhibitors of TNF-alpha production.
AID1387875Induction of cereblon-mediated ikaros degradation in human OPM2 cells at 0.1 to 10 uM after 5 hrs by immunoblot analysis2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID1814571Displacement of Cy5-O-Len probe from 6XHis-tagged CRBN/DDB1 (unknown origin) incubated in dark for 1 hr by fluorescence polarization assay
AID1707353Intrinsic clearance in human2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID327441Down-regulation of HBBP1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1317703Cytotoxicity against BMOL-NT cells assessed as induction of cell death at 100 uM2016European journal of medicinal chemistry, Sep-14, Volume: 120Identification of a thalidomide derivative that selectively targets tumorigenic liver progenitor cells and comparing its effects with lenalidomide and sorafenib.
AID327442Down-regulation of HBBP1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625289Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver disease2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID327412Upregulation of INHBA gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1769484Drug accumulation in human MOLT-4 cells assessed as intracellular drug accumulation incubated for 4 hrs by LC-MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID1321635Antiinflammatory activity in human HT-29 cells assessed as inhibition of TNF-alpha induced BCECF-AM labeled human U937 cell adhesion at 1 uM preincubated for 1 hr followed by stimulation with TNF-alpha for 3 hrs followed by further co-incubation with BCEC2016Bioorganic & medicinal chemistry letters, 10-01, Volume: 26, Issue:19
In vitro and in vivo inhibitory activity of 6-amino-2,4,5-trimethylpyridin-3-ols against inflammatory bowel disease.
AID327409Upregulation of PPIC gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718323Induction of apoptosis in human KMM-1 cells assessed as late apoptotic cells at 10 uM after 72 hrs by Annexin V/PropidiumIodide staining based flow cytometric analysis (Rvb = 4 +/- 0.5%)2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1707360Antitumor activity against human MM1.S cells xenografted in CB17-SCID mouse assessed as reduction in tumor weight at 60 mg/kg, po qd for 14 days and measured after 14 days2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID1597324Antiinflammatory activity in human PBMC cells assessed as reduction in LPS-induced TNF-alpha production preincubated for 1 hr followed by LPS addition and measured after 18 to 20 hrs by ELISA2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID625287Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatomegaly2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID726393Inhibition of TNF-alpha production in LPS-stimulated human PBMC preincubated for 1 hr before LPS challenge measured after 28 to 20 hrs by ELISA2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity.
AID1387792Induction of CRL4/CRBN ubiquitin ligase-mediated GSPT1 degradation in human DF15 cells expressing pLOC-ePL-tagged GSPT1 after 4 hrs by luminescence based beta-galactosidase enzyme fragmentation complementation assay2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
Protein Degradation via CRL4
AID327444Down-regulation of SPTA1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327449Down-regulation of GYPA gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625290Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver fatty2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID327408Upregulation of CHI3L1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718352Down regulation of IKZF1 protein expression in human MM1.R cells at 10 to 50 uM after 48 hrs by immunoblotting analysis2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID625292Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) combined score2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID327406Upregulation of DEFA1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625284Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic failure2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1688911Antiproliferative activity against human NCI-H1975 cells harboring EGFR L858/T790M mutant assessed as reduction in cell viability incubated for 72 hrs by MTT assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Discovery of potent epidermal growth factor receptor (EGFR) degraders by proteolysis targeting chimera (PROTAC).
AID1759158Inhibition of Akt phosphorylation in mouse BMOL-T cells assessed as change in phosphorylated Akt level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID215443Inhibition of lipopolysaccharide stimulated TNF-alpha release in human whole blood1999Bioorganic & medicinal chemistry letters, Jun-07, Volume: 9, Issue:11
Amino-substituted thalidomide analogs: potent inhibitors of TNF-alpha production.
AID327429Upregulation of PRG3 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1882261Immunomodulatory activity in human MM cells2022European journal of medicinal chemistry, Feb-05, Volume: 229A review on the treatment of multiple myeloma with small molecular agents in the past five years.
AID1387876Induction of cereblon-mediated aiolos degradation in human OPM2 cells at 0.1 to 10 uM after 5 hrs by immunoblot analysis2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID327436Down-regulation of HBA1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327457Down-regulation of GYPE gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718344Induction of apoptosis in human KMM-1 cells assessed as apoptotic cells at 10 uM after 72 hrs by Annexin V/PropidiumIodide staining based flow cytometric analysis relative to control2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID327437Down-regulation of TUBB2 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1769485Drug accumulation in human MOLT-4 cells assessed as intracellular bioavailability by LC-MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID327431Upregulation of SEC14L5 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625286Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID327413Upregulation of SPARC gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID604330Inhibition of LPS-induced TNFalpha production in mouse RAW264.7 cells assessed as TNFalph activity at 30 uM by ELISA relative to untreated control2011Bioorganic & medicinal chemistry, Jul-01, Volume: 19, Issue:13
Design, synthesis and biological assessment of novel N-substituted 3-(phthalimidin-2-yl)-2,6-dioxopiperidines and 3-substituted 2,6-dioxopiperidines for TNF-α inhibitory activity.
AID327427Upregulation of DSC2 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625285Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic necrosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1597299Antiproliferative activity against human Mino cells measured after 72 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID1462007Antiproliferative activity against human EC9706 cells after 48 hrs by CCK-8 assay2017Bioorganic & medicinal chemistry letters, 09-01, Volume: 27, Issue:17
Design, synthesis and biological evaluation of Lenalidomide derivatives as tumor angiogenesis inhibitor.
AID1667518Induction of CRBN-mediated pLOC-ePL-tagged aiolos (unknown origin) degradation in human DF15 cells assessed as minimum protein remaining up to 10 uM incubated for 4 hrs by luminescence based beta-galactosidase enzyme fragmentation complementation assay re2020Journal of medicinal chemistry, 07-09, Volume: 63, Issue:13
Discovery of CRBN E3 Ligase Modulator CC-92480 for the Treatment of Relapsed and Refractory Multiple Myeloma.
AID625283Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for elevated liver function tests2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1355095Antiproliferative activity against human MV411 cells after 72 hrs by MTS assay
AID1769479Efflux ratio of apparent permeability of compound across basolateral to apical side over apical to basolateral side in human Caco-2 cells at 10 uM incubated for 90 mins by LC-MS/MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
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.
AID1707350Antiproliferative activity against human MM1.S cells assessed as reduction in cell growth measured after 7 days culturing by CellTiter 96 Aqueous non-radioactive cell proliferation assay2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID1706074Down regulation of NFkappaB p65 expression in LPS/IFNgamma-induced mouse RAW264.7 cells at 10 uM incubated for 20 hrs followed by LPS/IFNgamma stimulation and measured after 4 hrs by Western blot analysis2020European journal of medicinal chemistry, Dec-15, Volume: 208Induced protein degradation of histone deacetylases 3 (HDAC3) by proteolysis targeting chimera (PROTAC).
AID1759174Inhibition of Akt phosphorylation in mouse BMOL-T cells assessed as change in nuclear phosphorylated Akt level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1597298Antiproliferative activity against human MV411 cells measured after 168 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID1916050Inhibition of TNF-alpha release in LPS stimulated human PBMC2021European journal of medicinal chemistry, Aug-05, Volume: 220Tailor-made amino acids in the design of small-molecule blockbuster drugs.
AID1769477Distribution coefficient, logD of compound at 1 mg/ml measured at pH 7.4 by chromatography based LC-UV analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID327421Upregulation of RTN1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1658260Binding affinity to CBRN (unknown origin) incubated for 60 mins by lenalidomide displacement assay2020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Ligand Design for Cereblon Based Immunomodulatory Therapy.
AID327414Upregulation of SPARC gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327440Down-regulation of HBB gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718345Induction of apoptosis in human MM1.R cells assessed as apoptotic cells at 10 uM after 72 hrs by Annexin V/PropidiumIodide staining based flow cytometric analysis relative to control2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1707355Metabolic stability in human liver microsomes assessed as parent compound remaining2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID625280Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholecystitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1387872Induction of cereblon-mediated aiolos degradation in human DF15 cells expressing ePL-tagged aiolos after 4 hrs by luminometric analysis2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID327407Upregulation of CHI3L1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1569039Inhibition of CRBN-mediated IKFZ3 degradation in human OPM2 cells at 100 uM measured after 24 hrs by Western blot analysis relative to control2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives.
AID1861177Antiproliferative activity against human Jeko-1 cells expressing CRBN-knockout assessed as cell viability measured after 7 days by CCK-8 assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Structure-based rational design enables efficient discovery of a new selective and potent AKT PROTAC degrader.
AID327448Down-regulation of KLF1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327396Inhibition of cell proliferation of human bone marrow mononuclear cells assessed as [3H]thymidine incorporation upto 500 uM after 4 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1462009Antiproliferative activity against human EC9706 cells at 150 ug/mL after 48 hrs by CCK-8 assay2017Bioorganic & medicinal chemistry letters, 09-01, Volume: 27, Issue:17
Design, synthesis and biological evaluation of Lenalidomide derivatives as tumor angiogenesis inhibitor.
AID1759159Inhibition of STAT3 phosphorylation in mouse BMOL-NT cells assessed as change in phosphorylated STAT3 level at 10 uM incubated for 6 hrs by immunofluorescence assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID327462Down-regulation of THRB gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327452Down-regulation of CXADR gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1462008Antiproliferative activity against human EC9706 cells at >= 600 ug/mL after 48 hrs by CCK-8 assay relative to control2017Bioorganic & medicinal chemistry letters, 09-01, Volume: 27, Issue:17
Design, synthesis and biological evaluation of Lenalidomide derivatives as tumor angiogenesis inhibitor.
AID327450Down-regulation of GYPA gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1387791Induction of CRL4/CRBN ubiquitin ligase-mediated aiolos degradation in human DF15 cells expressing pLOC-ePL-tagged aiolos assessed as aiolos protein remaining at 10 uM after 4 hrs by luminescence based beta-galactosidase enzyme fragmentation complementati2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
Protein Degradation via CRL4
AID604332Inhibition of LPS-induced TNFalpha production in mouse RAW264.7 cells assessed as TNFalph activity at 30 uM by ELISA relative to thalidomide2011Bioorganic & medicinal chemistry, Jul-01, Volume: 19, Issue:13
Design, synthesis and biological assessment of novel N-substituted 3-(phthalimidin-2-yl)-2,6-dioxopiperidines and 3-substituted 2,6-dioxopiperidines for TNF-α inhibitory activity.
AID327465Increase in SPARC gene expression in erythroid progenitor cells from non-del(5q) myelodysplastic syndrome patient by RT-PCR after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1317702Anticancer activity against BMOL-T cells assessed as cell growth inhibition up to 100 uM2016European journal of medicinal chemistry, Sep-14, Volume: 120Identification of a thalidomide derivative that selectively targets tumorigenic liver progenitor cells and comparing its effects with lenalidomide and sorafenib.
AID327460Down-regulation of AMMECR1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327400Decrease in erythroid differentiation of CD34+ progenitor cells from myelodysplastic syndrome del(5q) patient assessed as CD36 expression after 14 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1387793Induction of CRL4/CRBN ubiquitin ligase-mediated aiolos degradation in human DF15 cells expressing pLOC-ePL-tagged aiolos after 4 hrs by luminescence based beta-galactosidase enzyme fragmentation complementation assay2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
Protein Degradation via CRL4
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.
AID327415Upregulation of CXCL1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1387788Induction of CRL4/CRBN ubiquitin ligase-mediated GSPT1 degradation in human DF15 cells expressing pLOC-ePL-tagged GSPT1 assessed as GSPT1 protein remaining at 10 uM after 4 hrs by luminescence based beta-galactosidase enzyme fragmentation complementation 2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
Protein Degradation via CRL4
AID327456Down-regulation of ALAS2 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718317Cytotoxicity against human Mononuclear cells assessed as reduction in cll viability at 10 uM after 48 hrs by PrestoBlue cell viability assay2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1387873Induction of cereblon-mediated ikaros degradation in human DF15 cells at 0.1 to 10 uM after 5 hrs by immunoblot analysis2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID327445Down-regulation of HBG2 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1759157Inhibition of Akt phosphorylation in mouse BMOL-NT cells assessed as change in phosphorylated Akt level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1772713PROTAC activity at CRBN/Bcr-Abl in human K562 cells assessed as degrading activity by Western blotting analysis2021European journal of medicinal chemistry, Nov-05, Volume: 223Discovery of novel BCR-ABL PROTACs based on the cereblon E3 ligase design, synthesis, and biological evaluation.
AID327432Upregulation of SEC14L5 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1772712Antiproliferative activity against human K562 cells assessed as cell growth inhibition measured after 48 hrs by CCK8 assay2021European journal of medicinal chemistry, Nov-05, Volume: 223Discovery of novel BCR-ABL PROTACs based on the cereblon E3 ligase design, synthesis, and biological evaluation.
AID1759160Inhibition of STAT3 phosphorylation in mouse BMOL-T cells assessed as change in phosphorylated STAT3 level at 10 uM incubated for 6 hrs by immunofluorescence assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID327403Upregulation of VSIG4 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1759156Inhibition of NFkB p65 phosphorylation in mouse BMOL-T cells assessed as change in phosphorylated p65 level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
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.
AID1861180Antiproliferative activity against human MM1.S cells assessed as cell viability measured after 7 days by CCK-8 assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Structure-based rational design enables efficient discovery of a new selective and potent AKT PROTAC degrader.
AID327461Down-regulation of THRB gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1688912Antiproliferative activity against human A549 cells harboring EGFR assessed as reduction in cell viability incubated for 72 hrs by MTT assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Discovery of potent epidermal growth factor receptor (EGFR) degraders by proteolysis targeting chimera (PROTAC).
AID1387874Induction of cereblon-mediated aiolos degradation in human DF15 cells at 0.1 to 10 uM after 5 hrs by immunoblot analysis2018Journal of medicinal chemistry, 01-25, Volume: 61, Issue:2
A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos.
AID327422Upregulation of RTN1 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1707352Intrinsic clearance in human liver microsomes measured per gm of protein2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID1759172Inhibition of NFkB p65 nuclear translocation in mouse BMOL-T cells assessed as change in nuclear p65 level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1597306Antiproliferative activity against human RPMI8226 cells measured after 72 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID327438Down-regulation of TUBB2 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327401Decrease in erythroid differentiation of CD34+ progenitor cells from myelodysplastic syndrome del(5q) patient assessed as glycophorin A expression after 14 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718324Induction of apoptosis in human KMM-1 cells assessed as necrotic cells at 10 uM after 72 hrs by Annexin V/PropidiumIodide staining based flow cytometric analysis (Rvb = 8 +/-1%)2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID327451Down-regulation of CXADR gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1597305Antiproliferative activity against human U266 cells measured after 72 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID1597303Antiproliferative activity against human Raji cells measured after 168 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID1718322Induction of apoptosis in human KMM-1 cells assessed as early apoptotic cells at 10 uM after 72 hrs by Annexin V/PropidiumIodide staining based flow cytometric analysis (Rvb = 1 +/- 0.4%)2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1759154Antimigratory activity against mouse BMOL-T cells assessed as inhibition of cell migration at 10 uM incubated for 16 hrs by scratch wound healing assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID327419Upregulation of MAOA gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327399Growth inhibition of CD34- mononuclear cells from myelodysplastic syndrome del(5q-) patient by FISH after 7 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1599136Binding affinity to human CRBN (1 to 442 residues)/N-terminal 6His-tagged human DDB1 (1 to 1140 residues) expressed in baculovirus infected BTI-TN-5B1-4 insect cells after 30 mins by cy5 probe based fluorescence polarization assay2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
From Inhibition to Degradation: Targeting the Antiapoptotic Protein Myeloid Cell Leukemia 1 (MCL1).
AID327423Upregulation of ANGPT2 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1861178Antiproliferative activity against human Mino cells assessed as cell viability measured after 7 days by CCK-8 assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Structure-based rational design enables efficient discovery of a new selective and potent AKT PROTAC degrader.
AID1597301Antiproliferative activity against human Z138 cells measured after 72 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID327463Down-regulation of LRP11 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327398Growth inhibition of CD34+ progenitor cells from non-del(5q) myelodysplastic syndrome patient after 14 days2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
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.
AID1718336Growth inhibition of human KMM-1 cells at 10 to 100 uM after 72 hrs by PrestoBlue cell viability assay relative to control2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID327458Down-regulation of GYPE gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1597307Antiproliferative activity against human THP1 cells measured after 168 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID327428Upregulation of DSC2 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1658259Inhibition of BRD4 bromodomain 2 (unknown origin) in HEK293 cells cotransfected with eGFP by flow cytometry based mCherry reporter gene analysis2020ACS medicinal chemistry letters, Jun-11, Volume: 11, Issue:6
Ligand Design for Cereblon Based Immunomodulatory Therapy.
AID625282Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cirrhosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID327430Upregulation of PRG3 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718321Induction of apoptosis in human KMM-1 cells assessed as viable cells at 10 uM after 72 hrs by Annexin V/PropidiumIodide staining based flow cytometric analysis (Rvb = 87 +/- 1%)2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1718351Down regulation of IKZF3 protein expression in human MM1.R cells at 10 to 50 uM after 48 hrs by immunoblotting analysis2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1569040Inhibition of MANT-uracil binding to wild-type Magnetospirillum gryphiswaldense CRBN isoform 4 by FRET assay2019Journal of medicinal chemistry, 07-25, Volume: 62, Issue:14
De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide Derivatives.
AID1707354Hepatic intrinsic clearance in human2021European journal of medicinal chemistry, Jan-01, Volume: 209Design, synthesis and biological evaluation of thioether-containing lenalidomide and pomalidomide derivatives with anti-multiple myeloma activity.
AID625279Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for bilirubinemia2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1769480Kinetic solubility of compound in PBS at 200 uM incubated for 30 mins by LC-MS/MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID1759177Effect on STAT3 expression in mouse BMOL-T cells at 10 uM by AlphaLISA assay2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID625291Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver function tests abnormal2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID327469Increase in SPARC gene expression in CD34- mononuclear cells from non-del(5q) myelodysplastic syndrome patient by RT-PCR after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327433Down-regulation of HBA2 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327454Down-regulation of GYPB gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1718346Down regulation of IRF4 protein expression in human JJN-3 cells at 10 to 50 uM after 48 hrs by immunoblotting analysis2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID1769482Stability in human plasma assessed as half life at 1 uM by LC-MS/MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID327443Down-regulation of SPTA1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327446Down-regulation of HBG2 gene expression in healthy human erythroblasts after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID1759171Inhibition of NFkB p65 nuclear translocation in mouse BMOL-NT cells assessed as change in nuclear p65 level at 10 uM incubated for 6 hrs by immunofluorescent assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1718318Cytotoxicity against human naive B cells assessed as reduction in cll viability at 10 uM after 48 hrs by PrestoBlue cell viability assay2020Journal of natural products, 12-24, Volume: 83, Issue:12
Antimyeloma Potential of Caffeic Acid Phenethyl Ester and Its Analogues through Sp1 Mediated Downregulation of IKZF1-IRF4-MYC Axis.
AID726392Antiproliferative activity against human NAMALWA cells assessed as inhibition of [3H]thymidine incorporation after 72 hrs by scintillation counting2013Bioorganic & medicinal chemistry letters, Jan-01, Volume: 23, Issue:1
Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity.
AID1769483Drug accumulation in human MOLT-4 cells assessed as intracellular unbound fraction incubated for 4 hrs by LC-MS analysis2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID1597302Antiproliferative activity against human JeKo1 cells measured after 72 hrs by CellTiter96 Aqueous non-radioactive cell proliferation assay2019European journal of medicinal chemistry, Aug-15, Volume: 176Design, synthesis and biological evaluation of the thioether-containing lenalidomide analogs with anti-proliferative activities.
AID1759176Inhibition of STAT3 phosphorylation in mouse BMOL-T cells assessed as change in nuclear phosphorylated STAT3 level at 10 uM incubated for 6 hrs by immunofluorescence assay relative to control2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1688913Antiproliferative activity against human A-431 cells harboring EGFR assessed as reduction in cell viability incubated for 72 hrs by MTT assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Discovery of potent epidermal growth factor receptor (EGFR) degraders by proteolysis targeting chimera (PROTAC).
AID1759151Antiproliferative activity against mouse BMOL-NT cells assessed as cell growth by measuring doubling time at 10 uM measured every 4 hrs for 3 days by IncuCyte zoom live cell analysis (Rvb = 18 to 19 hrs)2021European journal of medicinal chemistry, May-05, Volume: 217In pursuit of a selective hepatocellular carcinoma therapeutic agent: Novel thalidomide derivatives with antiproliferative, antimigratory and STAT3 inhibitory properties.
AID1861176Antiproliferative activity against human Jeko-1 cells assessed as cell viability measured after 7 days by CCK-8 assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Structure-based rational design enables efficient discovery of a new selective and potent AKT PROTAC degrader.
AID1769475Protac activity at CRBN/HiBiT-tagged IKZF1 in human MOLT-4 cells assessed as IKZF1 degradation by measuring intracellular unbound concentration by luminescence based HiBiT assay2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition.
AID327435Down-regulation of HBA1 gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID327453Down-regulation of GYPB gene expression in erythroblasts from myelodysplastic syndrome del(5q) patient after 7 days relative to control2007Proceedings of the National Academy of Sciences of the United States of America, Jul-03, Volume: 104, Issue:27
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
AID625281Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholelithiasis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347094qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347092qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for A673 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347089qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347093qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347099qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347090qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347107qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347100qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347091qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347106qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347104qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347098qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347103qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347101qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347154Primary screen GU AMC qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1508630Primary qHTS for small molecule stabilizers of the endoplasmic reticulum resident proteome: Secreted ER Calcium Modulated Protein (SERCaMP) assay2021Cell reports, 04-27, Volume: 35, Issue:4
A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
AID1347108qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347102qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347096qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347105qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347425Rhodamine-PBP qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID1347407qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Pharmaceutical Collection2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1347424RapidFire Mass Spectrometry qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID686947qHTS for small molecule inhibitors of Yes1 kinase: Primary Screen2013Bioorganic & medicinal chemistry letters, Aug-01, Volume: 23, Issue:15
Identification of potent Yes1 kinase inhibitors using a library screening approach.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (3,462)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's1 (0.03)18.2507
2000's437 (12.62)29.6817
2010's2109 (60.92)24.3611
2020's915 (26.43)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 88.31

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 Index88.31 (24.57)
Research Supply Index8.36 (2.92)
Research Growth Index6.91 (4.65)
Search Engine Demand Index158.48 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (88.31)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials725 (20.53%)5.53%
Reviews776 (21.97%)6.00%
Case Studies544 (15.40%)4.05%
Observational33 (0.93%)0.25%
Other1,454 (41.17%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (1001)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Randomized Phase III Trial of Lenalidomide Versus Observation Alone in Patients With Asymptomatic High-Risk Smoldering Multiple Myeloma [NCT01169337]Phase 3226 participants (Actual)Interventional2011-01-24Active, not recruiting
A Randomized Phase II/III Study of Azacitidine in Combination With Lenalidomide (NSC-703813) vs. Azacitidine Alone vs. Azacitidine in Combination With Vorinostat (NSC-701852) for Higher-Risk Myelodysplastic Syndromes (MDS) and Chronic Myelomonocytic Leuke [NCT01522976]Phase 2282 participants (Actual)Interventional2012-03-01Active, not recruiting
A Phase 1b Study of PVX-410, a Multi-Peptide Cancer Vaccine, and Citarinostat (CC-96241), a Histone Deacetylase Inhibitor (HDAC) With and Without Lenalidomide for Patients With Smoldering Multiple Myeloma [NCT02886065]Phase 119 participants (Actual)Interventional2017-03-07Active, not recruiting
Pilot-trial of Methotrexate, Tafasitamab (Minjuvi®), Lenalidomide (Revlimid®) and Rituximab in Patients Ineligible for HCT-ASCT With Primary Central Nervous System Lymphoma (PCNSL) [NCT05583071]Phase 220 participants (Anticipated)Interventional2023-05-31Not yet recruiting
Phase I Study of Lenalidomide and Conventional Chemotherapy in Acute Myeloid Leukemia [NCT01132586]Phase 161 participants (Actual)Interventional2010-05-31Completed
A Phase 1 Study of Lenalidomide Maintenance Following Allogeneic Hematopoietic Cell Transplantation in Patients With Select High Risk Hematological Malignancies [NCT01254578]Phase 117 participants (Actual)Interventional2010-11-24Completed
A Phase II Study of the Bruton's Tyrosine Kinase Inhibitor, Zanubrutinib, in Combination With Lenalidomide Plus Rituximab, Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone in Patients With Newly Diagnosed Diffuse Large B-Cell Lymphoma [NCT05200312]Phase 236 participants (Anticipated)Interventional2022-02-01Recruiting
A Multicenter, Randomized, Parallel-Group, Double-blind, Placebo-controlled Study of CC-5013 Plus Dexamethasone Versus Dexamethasone Alone in Previously Treated Subjects With Multiple Myeloma [NCT00056160]Phase 3353 participants (Actual)Interventional2003-01-01Completed
CLLM1-Protocol of the German CLL-Study Group (GCLLSG) A Phase 3, Multicenter, Randomized, Double-blind, Placebo-controlled, Parallel-group Study of the Efficacy and Safety of Lenalidomide (Revlimid®) as Maintenance Therapy for High-risk Patients With Chro [NCT01556776]Phase 389 participants (Actual)Interventional2012-07-20Completed
A Phase 1 and Phase 2 Study of Lenalidomide (Revlimid) in Combination With Cyclophosphamide (Endoxan) and Prednison (REP) in Relapsed/Refractory Multiple Myeloma [NCT01352338]Phase 1/Phase 282 participants (Actual)Interventional2011-08-31Completed
A Phase 2, Open-Label Study to Evaluate the Safety and Efficacy of iR2 (Ibrutinib,Lenalidomide, Rituximab)in Untreated and Unfit Elderly Patients With Diffuse Large B-cell Lymphoma [NCT03949062]Phase 230 participants (Actual)Interventional2019-03-13Active, not recruiting
A Phase III, Open-Label, Multicenter Randomized Study Evaluating Glofitamab as a Single Agent Versus Investigator's Choice in Patients With Relapsed/Refractory Mantle Cell Lymphoma [NCT06084936]Phase 3182 participants (Anticipated)Interventional2023-10-22Recruiting
A Phase II Study of Selinexor In Addition to Lenalidomide for Consolidation and Maintenance Treatment After Autologous Hematopoietic Cell Transplant in Multiple Myeloma [NCT05820763]Phase 235 participants (Anticipated)Interventional2024-01-31Not yet recruiting
A Randomized, Controlled, Open-label, Multicenter, Inferentially Seamless Phase 2/3 Study of Ibrutinib in Combination With Rituximab Versus Physician's Choice of Lenalidomide Plus Rituximab or Bortezomib Plus Rituximab in Participants With Relapsed or Ref [NCT05564052]Phase 236 participants (Actual)Interventional2022-12-06Active, not recruiting
A Phase 3 Randomized Study Comparing Teclistamab in Combination With Daratumumab SC and Lenalidomide (Tec-DR) and Talquetamab in Combination With Daratumumab SC and Lenalidomide (Tal-DR) Versus Daratumumab SC, Lenalidomide, and Dexamethasone (DRd) in Part [NCT05552222]Phase 31,590 participants (Anticipated)Interventional2022-10-25Recruiting
A Phase 3b, Multicenter, Open-label, Daratumumab Long-term Extension Study [NCT05438043]Phase 3500 participants (Anticipated)Interventional2022-12-15Recruiting
Phase 1b Study of REGN5458 (Anti-BCMA x Anti-CD3 Bispecific Antibody) Plus Other Cancer Treatments for Patients With Relapsed/Refractory Multiple Myeloma [NCT05137054]Phase 1317 participants (Anticipated)Interventional2022-08-17Recruiting
A Multi-arm Phase 1b Study of Talquetamab With Other Anticancer Therapies in Participants With Multiple Myeloma [NCT05050097]Phase 1182 participants (Anticipated)Interventional2021-09-22Recruiting
A Phase 3 Randomized Study Comparing Bortezomib, Lenalidomide and Dexamethasone (VRd) Followed by Ciltacabtagene Autoleucel, a Chimeric Antigen Receptor T Cell (CAR-T) Therapy Directed Against BCMA Versus Bortezomib, Lenalidomide, and Dexamethasone (VRd) [NCT04923893]Phase 3650 participants (Anticipated)Interventional2021-08-19Recruiting
A Multi-arm Phase 1b Study of Teclistamab With Other Anticancer Therapies in Participants With Multiple Myeloma [NCT04722146]Phase 1140 participants (Actual)Interventional2021-03-12Active, not recruiting
Phase I Trial of Methotrexate, Rituximab, Lenalidomide, and Nivolumab (Nivo-MR2) Induction Followed by Lenalidomide and Nivolumab Maintenance in Primary CNS Lymphoma [NCT04609046]Phase 132 participants (Anticipated)Interventional2021-05-24Recruiting
A Phase 2, Multicohort Open-Label Study of JNJ-68284528, a Chimeric Antigen Receptor T Cell (CAR-T) Therapy Directed Against BCMA in Subjects With Multiple Myeloma [NCT04133636]Phase 2169 participants (Actual)Interventional2019-11-07Active, not recruiting
A Randomized Study of Daratumumab Plus Lenalidomide Versus Lenalidomide Alone as Maintenance Treatment in Patients With Newly Diagnosed Multiple Myeloma Who Are Minimal Residual Disease Positive After Frontline Autologous Stem Cell Transplant [NCT03901963]Phase 3200 participants (Actual)Interventional2019-04-26Active, not recruiting
A Phase 3 Study Comparing Daratumumab, VELCADE (Bortezomib), Lenalidomide, and Dexamethasone (D-VRd) With VELCADE, Lenalidomide, and Dexamethasone (VRd) in Subjects With Untreated Multiple Myeloma and for Whom Hematopoietic Stem Cell Transplant is Not Pla [NCT03652064]Phase 3395 participants (Actual)Interventional2018-11-06Active, not recruiting
An Open-Label, Single Arm, Phase 2a Study of Bortezomib, Lenalidomide, Dexamethasone and Elotuzumab in Newly Diagnosed Multiple Myeloma [NCT02375555]Phase 240 participants (Actual)Interventional2015-05-07Active, not recruiting
A Phase 1 Study of Lenalidomide and Ibrutinib in Combination With Rituximab in Relapsed and Refractory CLL and SLL [NCT02160015]Phase 112 participants (Actual)Interventional2014-05-20Active, not recruiting
A Phase II Study of Lenalidomide (CC-5013) in Combination With Prednisone for the Treatment of Myelofibrosis With Myeloid Metaplasia [NCT00227591]Phase 248 participants (Actual)Interventional2005-12-31Completed
Elotuzumab in Combination With Carfilzomib, Lenalidomide and Dexamethasone (E-KRd) Versus KRd Prior to and Following Auto-SCT in Newly Diagnosed Multipe Myeloma and Subsequent Maintenance With Elotuzumab and Lenalidomide Versus Single-Agent Lenalidomide- [NCT03948035]Phase 3576 participants (Anticipated)Interventional2018-08-28Active, not recruiting
KRd Consolidation in Myeloma Patients With a Positive PET-CT After Standard First Line Treatment. A Phase II Study [NCT03314636]Phase 253 participants (Actual)Interventional2018-03-16Active, not recruiting
Phase II Trial of Ixazomib and Dexamethasone Versus Ixazomib, Dexamethasone and Lenalidomide, Randomized With NFKB2 Rearrangement. (Proteasome Inhibitor NFKB2 Rearrangement Driven Trial, PINR) [NCT02765854]Phase 272 participants (Actual)Interventional2016-09-01Active, not recruiting
A Phase II Study of MOR00208 in Combination With Lenalidomide for Patients With Relapsed or Refractory Chronic Lymphocytic Leukemia(CLL)/Small Lymphocytic Lymphoma (SLL)/Prolymphocytic Leukemia (PLL), Including Those Who Have Relapsed on Ibrutinib, or Pat [NCT02005289]Phase 241 participants (Actual)Interventional2014-01-17Active, not recruiting
Assessment of Bortezomib (Alvocade ®) Efficacy and Safety in Newly Diagnosed Multiple Myeloma Patients [NCT04348006]Phase 40 participants (Actual)Interventional2020-03-01Withdrawn(stopped due to No patients were enrolled in the study)
A Randomized, Open-label Phase 3 Study of Carfilzomib, Lenalidomide, and Dexamethasone Versus Bortezomib, Lenalidomide and Dexamethasone (KRd vs. VRd) in Patients With Newly Diagnosed Multiple Myeloma (COBRA) [NCT03729804]Phase 3250 participants (Anticipated)Interventional2019-05-07Recruiting
Open-label, Single-arm, Phase 2 Study of Initial Treatment With Daratumumab (Darzalex), Carfilzomib (Kyprolis), Lenalidomide (Revlimid) and Low Dose Dexamethasone (DKRd) in Newly Diagnosed, Multiple Myeloma Requiring Systemic Chemotherapy [NCT03500445]Phase 275 participants (Anticipated)Interventional2019-02-13Recruiting
Open-label, Single-arm, Phase 2 Study of Initial Treatment With Elotuzumab, Carfilzomib (Kyprolis), Lenalidomide (Revlimid) and Low Dose Dexamethasone (E-KRd) in Newly Diagnosed, Multiple Myeloma Requiring Systemic Chemotherapy [NCT02969837]Phase 247 participants (Actual)Interventional2017-07-10Active, not recruiting
Phase 3 Randomized Trial of Carfilzomib, Lenalidomide, Dexamethasone Versus Lenalidomide Alone After Stem-cell Transplant for Multiple Myeloma [NCT02659293]Phase 3180 participants (Actual)Interventional2016-04-26Active, not recruiting
Phase II Randomized Trial of Continuation of Post-Transplant Maintenance With Single-Agent Lenalidomide vs. Consolidation/Maintenance With Ixazomib-Lenalidomide-Dexamethasone in Patients With Residual Myeloma [NCT02389517]Phase 242 participants (Actual)Interventional2015-03-02Active, not recruiting
Prospective, Multi-center Phase I/II Trial of Lenalidomide and Dose-Adjusted EPOCH-R in MYC-Associated B-Cell Lymphomas [NCT02213913]Phase 1/Phase 246 participants (Anticipated)Interventional2014-07-29Active, not recruiting
A Phase I, Multicenter, Open-label, Dose-escalation Study to Assess the Safety of Lenalidomide in Patients With Advanced Adult T-cell Leukemia-lymphoma and Peripheral T-cell Lymphoma [NCT01169298]Phase 113 participants (Actual)Interventional2010-07-01Completed
Pilot Study Immunomonitoring NK Cells in Patients With Myeloid Malignancies [NCT02525250]14 participants (Actual)Interventional2012-12-31Completed
Randomized Study Comparing Conventional Dose Treatment Using a Combination of Lenalidomide, Bortezomib and Dexamethasone to High-Dose Treatment With ASCT in the Initial Management of Myeloma in Patients up to 65 Years of Age [NCT01191060]Phase 3700 participants (Actual)Interventional2010-10-31Completed
Phase II Multicentric Trial Maintenance Therapy With 6 Monthly Revlimid® Cycles Alternated With 6 Monthly Vidaza® Cycles in First CR After Induction LIA Chemotherapy for Elderly Fit Patients With Poor Prognosis Acute Myeloid Leukemia. [NCT01301820]Phase 2120 participants (Actual)Interventional2011-01-31Completed
A Phase 2, Multicenter, Single Arm, Open Label Study of Venetoclax Plus Lenalidomide and Dexamethasone for the Treatment of Newly Diagnosed t(11;14)-Positive Multiple Myeloma in Subjects Who Are Ineligible for High-Dose Therapy [NCT03785184]Phase 20 participants (Actual)Interventional2019-04-29Withdrawn(stopped due to Strategic considerations)
Pilot Study of GC. (Gemcitabine-Rituximab-Oxaliplatin Combination) Given Every 14 Days With Maintenance Lenalidomide for the Treatment of Patients With Relapsed or Refractory Aggressive Non-Hodgkin's Lymphoma [NCT01307592]Phase 270 participants (Anticipated)Interventional2011-02-28Recruiting
GEM-CLARIDEX: Lenalidomide and Dexamethasone (Ld) Versus Clarithromycin / Lenalidomide [Revlimid®] / Dexamethasone (BiRd) as Initial Therapy in Multiple Myeloma [NCT02575144]Phase 3286 participants (Actual)Interventional2015-09-30Active, not recruiting
Phase II Study of Lenalidomide Maintenance After Salvage Therapy in Patients With Relapsed and/or Refractory Non-Hodgkin T-cell Lymphoma [NCT03730740]Phase 279 participants (Anticipated)Interventional2018-11-16Recruiting
A Phase 1b, Dose Escalation Study to Determine the Recommended Phase 2 Dose of TAK-659 in Combination With Bendamustine (±Rituximab), Gemcitabine, Lenalidomide, or Ibrutinib for the Treatment of Patients With Advanced Non-Hodgkin Lymphoma After At Least 1 [NCT02954406]Phase 143 participants (Actual)Interventional2017-03-05Terminated(stopped due to Business decision, insufficient enrollment, no safety or efficacy concerns.)
Phase II Trial Studying the Efficacy of a Triplet Combination of MLN9708, Lenalidomide and Dexamethasone as Induction Prior to, and as Consolidation After High-dose Therapy With Peripheral Stem Cell Transplantation Followed by MLN9708 Maintenance in the I [NCT01936532]Phase 242 participants (Actual)Interventional2014-11-12Completed
Open-label Phase 2 Pilot Study of Lenalidomide (Revlimid) as Adjuvant Treatment for Refractory Cutaneous T Cell Lymphoma [NCT01132989]Phase 210 participants (Anticipated)Interventional2010-05-31Recruiting
Chindamide in Combination With Lenalidomide and Dexamethasone for the Treatment of Relapsed/Refractory Multiple Myeloma, a Phase II Trial [NCT03605056]Phase 225 participants (Anticipated)Interventional2018-07-31Not yet recruiting
A Phase 2, Multicenter, Randomized, Open-label, Parallel-group Study of a Lenalidomide (Revlimid®) Regimen or a Sequential Azacitidine (Vidaza®) Plus Lenalidomide (Revlimid®) Regimen Versus an Azacitidine (Vidaza®) Regimen for Therapy of Older Subjects Wi [NCT01358734]Phase 288 participants (Actual)Interventional2012-04-27Completed
A Phase 1/2, Open-Label, Multicenter Study of ACY-1215 (Ricolinostat) in Combination With Lenalidomide and Dexamethasone for the Treatment of Relapsed or Relapsed/Refractory Multiple Myeloma [NCT01583283]Phase 138 participants (Actual)Interventional2012-07-12Completed
Multicenter, Open Label, Phase II Trial to Evaluate the Efficacy and Safety of Treatment With Lenalidomide in Kaposi Disease Associated With HIV Infection (ANRS 154/LENAKAP) [NCT01282047]Phase 212 participants (Actual)Interventional2011-10-31Terminated
A Phase 1, Open-label, Multicenter Study to Evaluate the Safety of bb2121 in Subjects With High Risk, Newly Diagnosed Multiple Myeloma (KarMMa-4) [NCT04196491]Phase 113 participants (Actual)Interventional2020-05-27Completed
Lenalidomide Therapy In Previously Untreated, Advanced Stage Follicular Lymphoma [NCT01180569]Phase 1/Phase 20 participants (Actual)Interventional2010-08-31Withdrawn
A Phase I Open-label Dose-escalation Study With Lenalidomide in Combination With a Fixed Dose of Sorafenib for the Treatment of Hepatocellular Carcinoma [NCT01348503]Phase 15 participants (Actual)Interventional2011-05-31Terminated(stopped due to Treatment was ineffective)
A Multicenter Open-Label Phase 1b/2 Study of the Bruton's Tyrosine Kinase (BTK) Inhibitor, Ibrutinib, in Combination With Lenalidomide and Rituximab in Subjects With Relapsed or Refractory Diffuse Large B-Cell Lymphoma [NCT02077166]Phase 1/Phase 2138 participants (Actual)Interventional2014-03-13Completed
Phase 1, Multicenter, Open-label, Dose-escalation Study of Sotatercept (ACE-011) in Combination With Lenalidomide or Pomalidomide and Dexamethasone in Patients With Relapsed and/or Refractory Multiple Myeloma [NCT01562405]Phase 133 participants (Actual)Interventional2012-05-31Active, not recruiting
R2 in the Treatment of Follicular Lymphoma Grade 1-3A [NCT03715309]Phase 2115 participants (Anticipated)Interventional2018-11-01Recruiting
A Phase 1b, Open-label, Multicenter Study of (BMS-936564) in Combination With Lenalidomide (Revlimid) Plus Low-dose Dexamethasone, or With Bortezomib (Velcade) Plus Dexamethasone in Subjects With Relapsed or Refractory Multiple Myeloma [NCT01359657]Phase 146 participants (Actual)Interventional2011-09-30Completed
University of Arkansas (UARK# 2017-03): A Single-Arm, Open-label Study of Anti-SLAMF7 mAb Therapy After Autologous Stem Cell Transplant in Patients With Multiple Myeloma (Total Therapy 8) [NCT03168100]Phase 20 participants (Actual)Interventional2017-10-01Withdrawn(stopped due to No study population. No subjects enrolled.)
A Platform Study Evaluating the Safety and Efficacy of Multiple Treatments in Patients With Multiple Myeloma [NCT05583617]Phase 1/Phase 2200 participants (Anticipated)Interventional2023-11-14Recruiting
Myeloma XIV: A Phase III Trial to Compare Standard and Frailty-adjusted Induction Therapy With Ixazomib, Lenalidomide and Dexamethasone (IRD) and Maintenance Lenalidomide (R) to Lenalidomide Plus Ixazomib (R+I) [NCT03720041]Phase 3740 participants (Anticipated)Interventional2020-08-04Recruiting
A Prospective Trial of Revlimid® in Transfusion Dependent Patients With Non-del (5q) Low/Intermediate-1 Risk Myelodysplastic Syndrome [NCT01178814]Phase 257 participants (Actual)Interventional2010-07-31Active, not recruiting
LCCC 1111: An Open-Label Dose-Finding Study of Lenalidomide as Reinduction/ Consolidation Followed by Lenalidomide Maintenance Therapy for Adults ≥ 60 Years of Age With Acute Myeloid Leukemia (AML) in Partial or Complete Response Following Conventional In [NCT01578954]Phase 120 participants (Actual)Interventional2012-06-28Completed
Phase I/II Study of the Combination of 5-azacitidine With Lenalidomide in Patients With High Risk Myelodysplastic Syndrome (MDS) and Acute Myelogenous Leukemia (AML) [NCT01038635]Phase 1/Phase 294 participants (Actual)Interventional2009-12-31Completed
A Randomised Phase II Study Comparing LEnalidomide Plus Rituximab, GEmcitabine and Methylprednisolone (LR-GEM) to Rituximab, Gemcitabine, Methylprednisolone and cisplatiN (R-GEM-P) in Second-line Treatment of Diffuse Large B-cell Lymphoma (DLBCL). [NCT02060656]Phase 292 participants (Anticipated)Interventional2013-09-30Active, not recruiting
Combined Intravitreal Methotrexate and R2 Regimen Followed by Lenalidomide Maintenance in Newly-diagnosed Primary Vitreoretinal Lymphoma [NCT03746223]Phase 242 participants (Anticipated)Interventional2018-11-15Recruiting
A Phase I Trial of Lenalidomide and Radiotherapy in Children With Diffuse Intrinsic Pontine Gliomas and High-grade Gliomas [NCT01222754]Phase 129 participants (Actual)Interventional2010-11-23Completed
A Multicenter, Randomized, Double-Blind, Placebo-Controlled, 3-Arm Study of the Efficacy and Safety of 2 Doses of Lenalidomide Versus Placebo in Red Blood Cell (RBC) Transfusion-Dependent Subjects With Low- or Intermediate-1-Risk Myelodysplastic Syndromes [NCT00179621]Phase 3205 participants (Actual)Interventional2005-07-31Completed
A Phase 2 Study to Evaluate Safety and Efficacy of Teclistamab in Combination With Daratumumab, Lenalidomide, and Dexamethasone With or Without Bortezomib as Induction Therapy and Teclistamab in Combination With Daratumumab and Lenalidomide as Maintenance [NCT05695508]Phase 270 participants (Anticipated)Interventional2022-12-01Recruiting
Phase II Trial for Newly Diagnosed Low-risk Multiple Myeloma Patients Comparing 6 Cycles of Isatuximab With Lenalidomide/Bortezomib/Dexamethasone (I-VRD) Compared to 3 Cycles of I-VRD Followed by One Cycle of High-dose Therapy and Both Arms Followed by Ma [NCT05665140]Phase 2/Phase 3100 participants (Anticipated)Interventional2023-02-03Recruiting
Randomized Phase II Open Label Study of Lenalidomide R-CHOP (R2CHOP) vs. RCHOP (Rituximab, Cyclophosphamide, Doxorubicin, Vincristine and Prednisone) in Patients With Newly Diagnosed Diffuse Large B Cell Lymphoma [NCT01856192]Phase 2349 participants (Actual)Interventional2013-08-27Active, not recruiting
A Phase II Randomized Trial of Lenalidomide (NSC # 703813) in Pediatric Patients With Recurrent, Refractory or Progressive Juvenile Pilocytic Astrocytomas and Optic Pathway Gliomas [NCT01553149]Phase 275 participants (Actual)Interventional2012-03-19Active, not recruiting
Clinical Study on the Efficacy and Safety of Anti-PD-1 Monoclonal Antibody Combined Lenalidomide and Azacitidine in Relapsed/Refractory Peripheral T-cell Lymphoma [NCT05182957]Phase 240 participants (Anticipated)Interventional2022-12-14Recruiting
Safety, Tolerability, and Efficacy of Once Weekly Carfilzomib in Combination With Daratumumab, Lenalidomide and Dexamethasone, in Transplant-ineligible Multiple Myeloma Patients Non-responsive to a Bortezomib Based Induction [NCT04065789]Phase 241 participants (Actual)Interventional2018-05-02Completed
A Phase I/II Study of Lenalidomide in Patients With Chronic Myelomonocytic Leukemia [NCT01368757]Phase 1/Phase 230 participants (Anticipated)Interventional2010-06-30Active, not recruiting
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
Post-marketing Phase 4 Study to Evaluate Safety, Tolerability, and Efficacy of Kyprolis® (Carfilzomib) in Indian Patients With Relapsed or Refractory Multiple Myeloma: A Prospective, Open-label, Non-comparative, Multicenter Study [NCT03934684]Phase 4100 participants (Anticipated)Interventional2019-09-16Active, not recruiting
A Multicenter, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Lenalidomide in the Treatment of Complex Regional Pain Syndrome Type 1 [NCT00109772]Phase 2184 participants (Actual)Interventional2005-02-28Terminated(stopped due to Interim analysis showed the primary outcome was not reached)
A Phase I/II Study of the Combination of Pembrolizumab and Lenalidomide, in Patients With Relapsed Non-Hodgkin and Hodgkin Lymphoma [NCT02875067]Phase 1/Phase 26 participants (Actual)Interventional2016-08-29Terminated(stopped due to Merck's decision for early termination of the data)
A Phase III Study Comparing Lenalidomide and Subcutaneous Daratumumab (R-Dara SC) vs Lenalidomide and Dexamethasone (Rd) in Frail Subjects With Previously Untreated Multiple Myeloma Who Are Ineligible for High Dose Therapy [NCT03993912]Phase 3294 participants (Anticipated)Interventional2019-10-17Active, not recruiting
Phase 3b, Randomized Trial of Continuous Revlimid® (Lenalidomide) Therapy Versus Observation Following Induction Therapy That Does Not Include Revlimid, Pomalyst® or Thalomid® in Newly Diagnosed Multiple Myeloma [NCT02155634]Phase 30 participants (Actual)Interventional2014-07-31Withdrawn(stopped due to The study was terminated due to poor feasibility and lack of interest at the participating sites.)
Safety and Feasibility of Lenalidomide in Combination With HLA-mismatched Stem-cell Microtransplantation as Post-remission Therapy in Patients With Acute Myeloid Leukemia (AML) [NCT02255162]Phase 18 participants (Actual)Interventional2015-01-31Terminated(stopped due to Slow Accrual)
"A Prospective Non-interventional Post-authorization Safety Study (PASS) of Lenalidomide in Previously Untreated Adult Multiple Myeloma Patients Who Are Not Eligible for Transplant (Transplant Noneligible [TNE])" [NCT03106324]911 participants (Actual)Observational [Patient Registry]2017-03-31Active, not recruiting
Phase I Study of the Activity and Safety of Lenalidomide and Rituximab as Non-chemotherapy Therapy for Patients With Recurrent and Refractory Chronic Lymphocytic Leukemia [NCT01185262]Phase 125 participants (Actual)Interventional2009-04-30Completed
PILOT STUDY PHASE II, Multicenter, Non-randomized, TO ASSESS THE EFFICACY AND SAFETY OF LENALIDOMIDE IN INDUCTION AND POST-INDUCTION IN PATIENTS WITH NOVO Acute Myeloid Leukemia (AML) WITH Cytogenetic Abnormality Monosomy 5 [NCT01198054]Phase 44 participants (Actual)Interventional2011-01-31Terminated
A Phase I Study of Epigenetic Immunomodulation Through the Use of Azacitidine, Lenalidomide, and Grifola Frondosa in Patients With Advanced Malignancy [NCT01200004]Phase 11 participants (Actual)Interventional2012-04-30Terminated(stopped due to Slow Accrual.)
Phase IB Study of Lenalidomide (Revlimid®) With Liposomal Doxorubicin (Doxil®) and Bevacizumab (Avastin®) for Patients With Platinum Resistant Ovarian Cancer. [NCT01202890]Phase 11 participants (Actual)Interventional2010-09-30Terminated(stopped due to Terminated due to inadequate rate of accrual.)
CC-5013 Alone or in Combination With Rituximab in Patients With Relapsed or Refractory Chronic Lymphocytic Leukemia (CLL) [NCT00096044]Phase 245 participants (Actual)Interventional2004-03-31Completed
A Multicenter, Single-arm, Open-label Phase II Study of the Safety of Lenalidomide Monotherapy and Markers for Disease Progression in Patients With IPSS Low- or Intermediate-1 Risk Myelodysplastic Syndromes (MDS) Associated With an Isolated Deletion 5q Cy [NCT01081431]Phase 291 participants (Actual)Interventional2010-03-31Active, not recruiting
A Phase II Study of the Efficacy and Safety of Lenalidomide Combined to Azacitidine in Intermediate-2 or High Risk MDS With Del 5q [NCT01088373]Phase 250 participants (Actual)Interventional2010-03-25Completed
A Phase 2, Open-label, Multicenter Study to Evaluate the Safety and Clinical Activity of Durvalumab in Combination With Rituximab, Cyclophosphamide, Doxorubicin, Vincristine, Prednisone (R-CHOP) or With Lenalidomide Plus R-CHOP (R2-CHOP) in Subjects With [NCT03003520]Phase 246 participants (Actual)Interventional2017-02-28Completed
Phase 3 Study of Teclistamab in Combination With Lenalidomide and Teclistamab Alone Versus Lenalidomide Alone in Participants With Newly Diagnosed Multiple Myeloma as Maintenance Therapy Following Autologous Stem Cell Transplantation - MajesTEC-4 [NCT05243797]Phase 31,572 participants (Anticipated)Interventional2022-09-08Recruiting
Daratumumab, Carfilzomib, Lenalidomide, and Dexamethasone in Newly-Diagnosed Multiple Myeloma: A Clinical and Correlative Phase II Study [NCT03290950]Phase 275 participants (Actual)Interventional2017-09-25Active, not recruiting
A Multicenter, Single-arm, Open-label Study of the Efficacy and Safety of Lenalidomide Monotherapy in Red Blood Cell Transfusion-dependent Subjects With Myelodysplastic Syndromes Associated With a Del(5q) Cytogenetic Abnormality. [NCT00065156]Phase 2148 participants (Actual)Interventional2003-06-01Completed
A Phase III Study of Lenalidomide Maintenance After Debulking Therapy in Patients With Advanced Cutaneous T-Cell Lymphoma [NCT01098656]Phase 321 participants (Actual)Interventional2010-07-31Terminated(stopped due to recruitment prematurely halted following company's decision to stop financial support to the study)
Am Open-Label Phase II Study of the Safety and Efficacy of Bortezomib, Lenalidomide, and Dexamethasone Combination Therapy for Patients With Relapsed or Relapsed and Refractory Multiple Myeloma [NCT00378209]Phase 265 participants (Actual)Interventional2006-08-31Completed
An Open-label, Single-Arm, Multicenter Study to Evaluate the Efficacy and Safety of Ixazomib in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed and/or Refractory Multiple Myeloma Initially Treated With an Injection of Proteasome [NCT03416374]Phase 445 participants (Actual)Interventional2018-02-18Completed
A Phase I Study of Lenalidomide in Combination With Decitabine for Patients With High Grade Myelodysplastic Syndromes [NCT00828802]Phase 115 participants (Actual)Interventional2009-03-31Completed
A Randomized Phase III Multicenter Trial Assessing Efficacy and Toxicity of a Combination of Rituximab and Lenalidomide (R2) vs Rituximab Alone as Maintenance After Chemoimmunotherapy With Rituximab-chemotherapy (R-CHT) for Relapsed/Refractory FL Patients [NCT02390869]Phase 3128 participants (Actual)Interventional2014-05-31Active, not recruiting
A Phase I/II Clinical Trial of Lenalidomide in Combination With Oral Cyclophosphamide in Patients With Previously Treated Hormone Refractory Prostate Cancer [NCT01093183]Phase 1/Phase 225 participants (Actual)Interventional2010-03-04Terminated(stopped due to study sponsors withdrew support for the study drug.)
A Phase 1/2 Study of Ixazomib as a Replacement for Bortezomib or Carfilzomib for Multiple Myeloma (MM) Patients Recently Relapsed or Refractory to Their Last Combination Regimen Containing Either Bortezomib or Carfilzomib [NCT02206425]Phase 1/Phase 245 participants (Actual)Interventional2014-09-30Completed
A PHASE II, MULTI-CENTER, OPEN LABEL STUDY OF CYCLOPHOSPHAMIDE IN MULTIPLE MYELOMA PATIENTS WITH BIOCHEMICAL PROGRESSION DURING LENALIDOMIDE-DEXAMETHASONE TREATMENT FOR RELAPSED/REFRACTORY MULTIPLE MYELOMA [NCT02206503]Phase 213 participants (Actual)Interventional2013-03-31Completed
A Trial of Single Autologous Transplant With or Without Consolidation Therapy Versus Tandem Autologous Transplant With Lenalidomide Maintenance for Patients With Multiple Myeloma (BMT CTN 0702) [NCT01109004]Phase 3758 participants (Actual)Interventional2010-05-31Completed
A Study Comparing the Efficacy and Safety of Genotype-guided R-CHOP-X Versus R-CHOP in Patients With Diffuse Large B-Cell Lymphoma [NCT05351346]Phase 31,100 participants (Anticipated)Interventional2022-06-01Recruiting
Isatuximab in Combination With Lenalidomide-Dexamethasone Compared to Lenalidomide-Dexamethasone in Elderly Patients (Aged ≥70 Years) With Newly Diagnosed Myeloma: a Randomized Phase II Study (SGZ-2019-12650) [NCT04891809]Phase 2198 participants (Anticipated)Interventional2021-10-20Recruiting
A Randomized, Phase II Trial Evaluating the Efficacy and Safety of Lenalidomide, Bortezomib and Dexamethasone (RVD) With or Without Panobinostat in Transplant Eligible, Newly Diagnosed Multiple Myeloma [NCT02720510]Phase 26 participants (Actual)Interventional2016-06-14Terminated(stopped due to A study was terminated due to low enrollment.)
A Multicenter, Phase 1/2 Study of Selinexor in Combination With Backbone Treatments or Novel Therapies in Patients With Relapsed or Refractory (RR) Diffuse Large B-Cell Lymphoma (DLBCL) [NCT04607772]Phase 1/Phase 2350 participants (Anticipated)Interventional2020-11-18Suspended(stopped due to Sponsor decision)
Phase II, Single-arm Trial of Carfilzomib, Lenalidomide, and Dexamethasone Re-induction Followed by the 2nd ASCT in Multiple Myeloma Patients Relapsed After the 1st ASCT [NCT05497102]Phase 258 participants (Anticipated)Interventional2021-11-08Recruiting
Real-world Evidence of Carfilzomib, Lenalidomide, Dexamethasone Combination Therapy in Korean Relapsed and/or Refractory Multiple Myeloma Patients [NCT05495620]300 participants (Anticipated)Observational2022-08-01Not yet recruiting
Phase 1 Study of Lenalidomide/Dexamethasone With Nivolumab and Ipilimumab in Patients With Newly Diagnosed Multiple Myeloma [NCT03283046]Phase 10 participants (Actual)Interventional2017-10-31Withdrawn(stopped due to Similar to another study.)
Clinical Phase II, Multicenter, Open-label Study Evaluating iNduction, Consolidation and Maintenance With Isatuximab (SAR650984), Carfilzomib, LEnalidomide and Dexamethasone (I-KRd) in Primary Diagnosed High-risk Multiple Myeloma paTients [NCT03104842]Phase 2246 participants (Actual)Interventional2017-08-15Active, not recruiting
A Phase Ib Study of the Safety and Pharmacokinetics of Atezolizumab (Anti-PD-L1 Antibody) Alone or in Combination With an Immunomodulatory Drug and/or Daratumumab in Patients With Multiple Myeloma (Relapsed/Refractory and Post-Autologous Stem Cell Transpl [NCT02431208]Phase 185 participants (Actual)Interventional2015-07-22Completed
"Monoclonal Antibodies for Treatment of Multiple Myeloma. Present Status and Aspects of Effector Mechanisms With Emphasis on the CD38 Antibody Daratumumab " [NCT02419118]Phase 2/Phase 34 participants (Actual)Interventional2015-01-31Completed
A Phase 3, Randomized, Double-Blind, Multicenter Study Comparing Oral Ixazomib (MLN9708) Plus Lenalidomide and Dexamethasone Versus Placebo Plus Lenalidomide and Dexamethasone in Adult Patients With Relapsed and/or Refractory Multiple Myeloma [NCT01564537]Phase 3722 participants (Actual)Interventional2012-08-01Completed
A Phase I Trial of B-cell Maturation Antigen (BCMA) Targeted EGFRt/BCMA-41BBz Chimeric Antigen Receptor (CAR) Modified T Cells With or Without Lenalidomide for the Treatment of Multiple Myeloma (MM) [NCT03070327]Phase 120 participants (Actual)Interventional2017-02-27Active, not recruiting
Toward a Risk-adapted Strategy to Cure Myeloma : An Intensive Program With Lenalidomide, Ixazomib, and Dexamethasone Plus Daratumumab as Extended Induction and Consolidation Followed by Lenalidomide Maintenance in Newly Diagnosed Standard Risk Multiple My [NCT03669445]Phase 245 participants (Anticipated)Interventional2018-12-31Recruiting
Immune Modulation by Addition of Oral Lenalidomide to Intravesical BCG (Bacille Calmette-Guerrin) for Therapy of Non-muscle-invasive Transitional Cell Bladder Cancer [NCT01373294]Phase 217 participants (Actual)Interventional2011-11-30Completed
Prospective, Single-center, Single-arm, Open-label Study of Obinutuzumab, Zanubrutinib and Lenalidomide Sequential CD19/CD22 CAR-T in Patients With Relapsed or Refractory B-Cell Non-Hodgkin Lymphoma [NCT05797948]20 participants (Anticipated)Interventional2022-07-01Enrolling by invitation
A Multicenter, Open-label, Prospective Study of Ixazomib, Lenalidomide, and Ixazomib in Combination With Lenalidomide for Maintenance Therapy in Patients With Newly Diagnosed Multiple Myeloma [NCT04217967]Phase 4180 participants (Anticipated)Interventional2020-01-03Recruiting
A Phase IB/II Trial of Lenalidomide (Revlimid®), Ixazomib and Rituximab (RIXAR) as Front-line Therapy for High Risk Indolent B Cell Lymphoma [NCT02898259]Phase 1/Phase 219 participants (Actual)Interventional2017-02-20Terminated(stopped due to Funding was eliminated.)
[NCT01036399]Phase 29 participants (Actual)Interventional2008-11-30Terminated(stopped due to The EC withdrawn the approval becuase of possible conflicts of interests between our Institute and Supporter (Celgene))
Phase 1 Clinical Trial to Evaluate Security and Dose of Expanded and Activated Autologous NK Cells Infusions in Consolidation of Multiple Myeloma Patients Treatment on Second or Later Relapse. [NCT02481934]Phase 15 participants (Actual)Interventional2013-03-31Completed
A Multi-center, Open-label Extended Access Program of Lenalidomide Plus Low-dose Dexamethasone in Chinese Subjects With Relapsed/Refactory Multiple Myeloma Who Participated in Study CC-5013-MM-021 for at Least One Year. [NCT02348528]Phase 265 participants (Actual)Interventional2012-09-11Completed
A Phase 1 Trial of Brentuximab Vedotin Plus Lenalidomide in Patients With Relapsed/ Refractory Cutaneous T-Cell Lymphomas [NCT03373305]Phase 10 participants (Actual)Interventional2019-03-31Withdrawn(stopped due to contract not executed)
An Open-Label, Multicenter Phase 1b Study Investigating the Safety of TAK-079 in Combination With Backbone Regimens for the Treatment of Patients With Newly Diagnosed Multiple Myeloma and for Whom Stem Cell Transplantation Is Not Planned as Initial Therap [NCT03984097]Phase 150 participants (Actual)Interventional2019-07-29Active, not recruiting
Single Arm Open-label Trial to Investigate the Efficacy and Safety of Lenlidomide as a Treatment for Recurrent or Refractory Crow-Fukase (POEMS) Syndrome [NCT02193698]Phase 25 participants (Actual)Interventional2014-07-31Completed
A Phase 2, Open-Label, Multicenter Study of Ixazomib Plus Lenalidomide and Dexamethasone in Adult Japanese Patients With Relapsed and/or Refractory Multiple Myeloma [NCT02917941]Phase 234 participants (Actual)Interventional2016-11-01Completed
Phase I Open-Label, Dose Escalation Study To Determine The Maximum Tolerated Dose And To Evaluate The Safety Profile of Lenalidomide (Revlimid® CC-5013) With Every Three Week Docetaxel (Taxotere®) In Subjects With Androgen Independent Prostate Cancer [NCT01378091]Phase 164 participants (Actual)Interventional2005-08-31Completed
Combination of Rituximab, Lenalidomide and Nivolumab for Relapsed/ Refractory Non-germinal Center (Non-GCB) Type Diffuse Large B Cell Lymphoma (DLBCL) Including Primary Central Nervous System Lymphoma (PCNSL): A Phase 1/2 Trial [NCT03558750]Phase 16 participants (Actual)Interventional2018-06-14Terminated(stopped due to Low accrual)
Pilot Clinical Study of DNA Vaccination Against Neuroblastoma [NCT04049864]Early Phase 112 participants (Anticipated)Interventional2019-01-09Recruiting
Phase II Trial of Daratumumab for Transplant-Eligible Multiple Myeloma Patients [NCT03477539]Phase 250 participants (Actual)Interventional2018-04-09Active, not recruiting
Phase II Study of Rituximab Plus Pembrolizumab (MK-3475) in Subjects With Relapsed Follicular Lymphoma [NCT02446457]Phase 253 participants (Actual)Interventional2015-07-31Active, not recruiting
Phase II Study of Ofatumumab in Combination With High Dose Methylprednisolone Followed by Ofatumumab and Lenalidomide Consolidative Therapy for the Treatment of Untreated CLL/SLL The HiLOG Trial [NCT01496976]Phase 245 participants (Actual)Interventional2012-03-30Active, not recruiting
A Real-world Study Evaluating the Safety and Efficacy of Tafasitamab in Combination With Lenalidomide in Patients With Relapsed or Refractory Diffuse Large B-cell Lymphoma [NCT05883709]15 participants (Anticipated)Observational2023-07-01Not yet recruiting
Isa-RVD Study: Phase II, Multi-centre, Single-Arm, Open-Label Study to Evaluate the Efficacy and Safety of the Combination Regimen Isatuximab, Lenalidomide, Bortezomib, and Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma [NCT05123131]Phase 243 participants (Anticipated)Interventional2022-04-01Recruiting
Phase I Dose Escalation Study of Velcade in Combination With Lenalidomide in Patients With Relapsed Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) After Allogeneic Stem Cell Transplantation [NCT02312102]Phase 122 participants (Actual)Interventional2015-02-28Active, not recruiting
Phase II Study of Bendamustine and Rituximab Induction Chemoimmunotherapy Followed by Maintenance Rituximab and Lenalidomide in Previously Untreated Chronic Lymphocytic Leukemia (CLL) and Small Lymphocytic Lymphoma (SLL) [NCT01754857]Phase 236 participants (Actual)Interventional2013-11-12Completed
A Phase I/II Study of Ublituximab in Combination With Lenalidomide (Revlimid®) in Patients With B-Cell Lymphoid Malignancies Who Have Relapsed or Are Refractory After CD20 Directed Antibody Therapy [NCT01744912]Phase 110 participants (Actual)Interventional2012-11-21Terminated(stopped due to Sponsor's decision)
MInimal Residual Disease Adapted Strategy: Frontline Therapy for Patients Eligible for Autologous Stem Cell Transplantation Less Than 66 Years; a Prospective Study [NCT04934475]Phase 3791 participants (Actual)Interventional2021-12-08Active, not recruiting
Optimizing Prolonged Treatment In Myeloma Using MRD Assessment (OPTIMUM) [NCT03941860]Phase 3510 participants (Anticipated)Interventional2022-02-16Active, not recruiting
A Prospective, Multicenter, Observational Study in Relapsed and/or Refractory Multiple Myeloma Patients Treated With Ixazomib Plus Lenalidomide and Dexamethasone [NCT03433001]295 participants (Actual)Observational2018-04-02Completed
A Multicenter Phase 2 Study to Evaluate Subcutaneous Daratumumab in Combination With Standard Multiple Myeloma Treatment Regimens [NCT03412565]Phase 2265 participants (Actual)Interventional2018-04-26Active, not recruiting
A Phase 3 Study Comparing Daratumumab, Lenalidomide, and Dexamethasone (DRd) vs Lenalidomide and Dexamethasone (Rd) in Subjects With Previously Untreated Multiple Myeloma Who Are Ineligible for High Dose Therapy [NCT02252172]Phase 3737 participants (Actual)Interventional2015-02-16Active, not recruiting
An Open-Label, Multicenter, Phase 1b Study of JNJ-54767414 (HuMax CD38) (Anti-CD38 Monoclonal Antibody) in Combination With Backbone Regimens for the Treatment of Subjects With Multiple Myeloma [NCT01998971]Phase 1242 participants (Actual)Interventional2014-02-18Active, not recruiting
An Open Label, International, Multicenter, Dose Escalating Phase I/II Trial Investigating the Safety of Daratumumab in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed or Relapsed and Refractory Multiple Myeloma [NCT01615029]Phase 1/Phase 245 participants (Actual)Interventional2012-06-30Active, not recruiting
A Phase II, Prospective, Single-center Study of Lenalidomide in Combination With R-DA-EPOCH in Patients With Untreated DLBCL With MYC Rearrangement [NCT04432714]Phase 1/Phase 281 participants (Anticipated)Interventional2020-06-09Recruiting
A Phase I Study of Lenalidomide in Combination With Rituximab and Ibrutinib in Relapsed and Refractory CLL and SLL [NCT02200848]Phase 15 participants (Actual)Interventional2014-04-30Terminated(stopped due to Recruitment difficulties and toxicity)
A Phase 3, Randomized, Double-Blind, Multicenter Study Comparing Oral MLN9708 Plus Lenalidomide and Dexamethasone Versus Placebo Plus Lenalidomide and Dexamethasone in Adult Patients With Newly Diagnosed Multiple Myeloma [NCT01850524]Phase 3705 participants (Actual)Interventional2013-04-29Completed
Efficacy and Safety of Lenalidomide in Combination With R-GemOx in First-line Treatment of Elderly Diffuse Large B Cell Lymphoma [NCT04432402]124 participants (Anticipated)Interventional2020-06-14Recruiting
Phase II Study of Romidepsin Plus Lenalidomide for Patients With Previously Untreated PTCL [NCT02232516]Phase 235 participants (Actual)Interventional2015-06-30Active, not recruiting
A Phase 3B Randomized Study of Lenalidomide (CC-5013) Plus Rituximab Maintenance Therapy Followed by Lenalidomide Single-Agent Maintenance Versus Rituximab Maintenance in Subjects With Relapsed/Refractory Follicular, Marginal Zone, or Mantle Cell Lymphoma [NCT01996865]Phase 3503 participants (Actual)Interventional2014-04-01Active, not recruiting
An Open Label, Multicenter, Phase 2, Pilot Study, Evaluating Early Treatment With Bispecific T-cell Redirectors (Teclistamab and Talquetamab) in the Frontline Therapy of Newly Diagnosed High-risk Multiple Myeloma [NCT05849610]Phase 230 participants (Anticipated)Interventional2023-11-30Recruiting
Relapsed Follicular Lymphoma Randomised Trial Against Standard ChemoTherapy (REFRACT): A Randomised Phase II Trial of Investigator Choice Standard Therapy Versus Sequential Novel Therapy Experimental Arms [NCT05848765]Phase 2284 participants (Anticipated)Interventional2023-09-04Recruiting
A Phase 3, Multicenter, Randomized, Double-blind, Placebo-controlled Trial Comparing the Efficacy and Safety of Tafasitamab Plus Lenalidomide in Addition to R-CHOP Versus R-CHOP in Previously Untreated, High-intermediate and High-risk Patients With Newly- [NCT04824092]Phase 3899 participants (Actual)Interventional2021-05-11Active, not recruiting
A Phase I Study of Venetoclax + Lenalidomide + Rituximab Hyaluronidase in Relapsed or Refractory (R/R) Indolent Non-Hodgkin's Lymphoma (iNHL). [NCT04447716]Phase 130 participants (Anticipated)Interventional2020-10-16Recruiting
A Phase 1 Study of Romidepsin, CC-486 (5-azacitidine), Dexamethasone, and Lenalidomide (RAdR) for Relapsed/Refractory T-cell Malignancies [NCT04447027]Phase 130 participants (Anticipated)Interventional2020-12-17Recruiting
Intergroup Randomized Phase 2 Four Arm Study In Patients ≥ 60 With Previously Untreated Mantle Cell Lymphoma Of Therapy With: Arm A = Rituximab+ Bendamustine Followed By Rituximab Consolidation (RB → R); Arm B = Rituximab + Bendamustine + Bortezomib Follo [NCT01415752]Phase 2373 participants (Actual)Interventional2012-08-09Active, not recruiting
Phase II Study of Lenalidomide/Rituximab Maintenance for Transplantation Ineligible Patients With Primary CNS Diffuse Large B-cell Lymphoma (Nickname: Lemon-C Study) [NCT04627753]Phase 230 participants (Anticipated)Interventional2020-11-02Recruiting
Phase II Trial of Acalabrutinib With Rituximab and Lenalidomide in Relapsed/Refractory B-cell Non-Hodgkin Lymphoma [NCT04094142]Phase 266 participants (Actual)Interventional2019-07-09Completed
Adoptive Transfer of NY-ESO-1 TCR Engineered Peripheral Blood Mononuclear Cells (PBMC) and Peripheral Blood Stem Cells (PBSC) After a High Dose Melphalan Conditioning Regimen, With Administration of Interleukin-2, in Patients With Multiple Myeloma [NCT03506802]Phase 10 participants (Actual)Interventional2018-07-10Withdrawn(stopped due to No Participants Enrolled)
A Phase I Study of the Combination of Lenalidomide With the Histone Deacetylase Inhibitor, Vorinostat in Hodgkin and Non Hodgkin's Lymphoma [NCT01116154]Phase 130 participants (Anticipated)Interventional2010-05-31Terminated(stopped due to Sponsor withdrew support for the study)
Double Blind Randomized Phase III Study of Lenalidomide Maintenance Versus Placebo in Responding Elderly Patients With DLBCL and Treated With R-CHOP in First Line [NCT01122472]Phase 3650 participants (Actual)Interventional2009-04-30Completed
A Single Arm Phase II Study to Investigate the Use of Lenalidomide in the Treatment of Patients With Early Stage CLL Associated With Poor Prognostic Factors [NCT01127542]Phase 21 participants (Actual)Interventional2010-05-31Terminated(stopped due to Potential safety issue of second primary malignancies in patients treated with lenalidomide.)
A Phase I, Open-Label Study To Determine The Maximum Tolerated Dose (Mtd) Of The Combination Of Lenalidomide And Cetuximab, And To Evaluate The Efficacy Of This Combination In Subjects With Wild Type K-Ras Metastatic Colorectal Carcinoma [NCT01126450]Phase 13 participants (Actual)Interventional2009-10-31Terminated(stopped due to low accrual,loss of funding and results from EU study showing drug ineffective.)
Effect of Lenalidomide (Revlimid®) in Solid Tumour Patients With Inflammatory Cancer Cachexia Syndrome on Lean Body Mass and Muscle Strength: A Multicenter, Proof-of-concept Study of Fixed Dose or CRP-response-guided Dose of Lenalidomide in Relation to Ne [NCT01127386]Phase 1/Phase 2200 participants (Actual)Interventional2009-03-31Completed
Phase II Study of Glofitamab, Poseltinib and Lenalidomide in Patients With Relapsed/Refractory Diffuse Large B Cell Lymphoma [NCT05335018]Phase 276 participants (Anticipated)Interventional2022-05-30Recruiting
A Two Stage Trial of lénalidomide (Revlimid®) : a Phase II Study of lénalidomide as Single Agent in Asymptomatic Ovarian Cancer Patients With Increasing CA 125 in Late Relapse: Followed by a Phase I of lénalidomide in Combination With Carboplatin and Lipo [NCT01111903]Phase 1/Phase 267 participants (Actual)Interventional2009-05-31Completed
EVALUATION OF CHOLECALCIFEROL (VitD3) MAINTENANCE SUPPLEMENTATION IN PATIENTS WITH MULTIPLE MYELOMA (MM) UNDERGOING TRANSPLANTATION AND IN COMBINATION WITH LENALIDOMIDE MAINTENANCE [NCT05846880]Early Phase 1100 participants (Anticipated)Interventional2023-12-31Not yet recruiting
A Phase 1/2, Dose and Schedule Evaluation Study to Investigate the Safety and Clinical Activity of Belantamab Mafodotin Administrated in Combination With Lenalidomide, Dexamethasone and Nirogacestat in Patients With Transplant Ineligible Newly Diagnosed M [NCT05573802]Phase 1/Phase 236 participants (Anticipated)Interventional2023-07-14Recruiting
A Multi-center, Open Label, Phase 1b/2 Study to Study the Efficacy and Safety of MIL62 Plus Lenalidomide in Subjects With Relapsed/Refractory Follicular Lymphoma or Marginal Zone Lymphoma [NCT04110301]Phase 1/Phase 253 participants (Anticipated)Interventional2019-11-28Recruiting
A Phase I Study of Lenalidomide in Combination With Dexamethasone in Anti-MAG Demyelinating Sensorimotor Neuropathy [NCT03701711]Phase 111 participants (Actual)Interventional2018-09-10Terminated(stopped due to Higher than expected occurrence of VTE (venous thromboembolic event))
A Phase Ib/IIa Study of Romidepsin in Combination With Lenalidomide in Adults With Relapsed or Refractory Lymphomas and Myeloma [NCT01755975]Phase 1/Phase 262 participants (Actual)Interventional2012-12-31Completed
A Phase 1b/2, Open-Label Trial to Assess the Safety and Preliminary Efficacy of Epcoritamab (GEN3013; DuoBody®-CD3xCD20) in Combination With Other Agents in Subjects With B-cell Non-Hodgkin Lymphoma (B-NHL) [NCT04663347]Phase 1/Phase 2662 participants (Anticipated)Interventional2020-11-03Recruiting
A Pilot Study of Lenalidomide Maintenance Therapy Following Autologous Stem Cell Transplantation in Patients With Relapsed/Refractory Hodgkin Lymphoma [NCT01207921]Phase 128 participants (Actual)Interventional2011-04-28Completed
Multicenter Phase I Study on the Safety, Anti-tumor Activity and Pharmacology of IPH2101, a Human Monoclonal Anti-KIR, Combined With Lenalidomide in Patients With Multiple Myeloma Experiencing a First or Second Relapse [NCT01217203]Phase 115 participants (Actual)Interventional2010-09-30Completed
A Phase II, Multi-center, Open-label, Repeat-dose Study of Lenalidomide (Revlimid ®) Plus Low-dose Dexamethasone in Patients With Refractory B Cell Lineage Acute Lymphoblastic Leukemia or in Relapse After 2 Lines of Treatment [NCT01116193]Phase 210 participants (Actual)Interventional2010-01-31Completed
Observational, Non-interventional, Multicenter Study Aimed at Collecting Retrospective/Prospective 648/96 Italian Registry Data Related to Lenalidomide (Revlimid®) Prescription to Patients With Myelodysplastic Syndromes [NCT01347944]149 participants (Actual)Observational2011-01-01Completed
A Randomized Parallel Phase 2 Study of Elotuzumab Plus Lenalidomide (Elo/Rev) for the Treatment of Serologic Relapse/Progression While on Lenalidomide Maintenance for Multiple Myeloma [NCT03411031]Phase 218 participants (Actual)Interventional2018-10-04Terminated(stopped due to Sponsor no longer providing drug)
Clinical Study to Evaluate the Safety and Efficacy of Daratumumab and Carfilzomib-based Induction/Consolidation/Maintenance Therapy in Transplant-eligible, Ultra High-risk, Newly Diagnosed Multiple Myeloma [NCT06140966]Phase 254 participants (Anticipated)Interventional2023-10-20Recruiting
Post Marketing Surveillance on Safety Evaluation of REVLIMID® (Lenalidomide) Treatment of Myelodysplastic Syndromes Associated With a Deletion 5q or Mantle Cell Lymphoma in Korea [NCT04036448]600 participants (Anticipated)Observational2019-08-18Active, not recruiting
Carfilzomib, Lenalidomide, and Dexamethasone in Newly-Diagnosed Multiple Myeloma: A Clinical and Correlative Phase I/II Dose Escalation Study [NCT02937571]Phase 1/Phase 237 participants (Actual)Interventional2016-10-31Active, not recruiting
Phase 3 Study Comparing Daratumumab, Lenalidomide, and Dexamethasone (DRd) vs Lenalidomide and Dexamethasone (Rd) in Subjects With Relapsed or Refractory Multiple Myeloma [NCT02076009]Phase 3569 participants (Actual)Interventional2014-05-23Active, not recruiting
Randomized Phase III Trial Comparing the Frequency of Major Erythroid Response (MER) to Treatment With Lenalidomide (Revlimid�) Alone and in Combination With Epoetin Alfa (Procrit�) in Subjects With Low- or Intermediate-1 Risk MDS and Symptomatic Anemia [NCT00843882]Phase 3247 participants (Actual)Interventional2009-01-29Active, not recruiting
A Randomized Phase III Trial of CC-5013 (Lenalidomide, NSC-703813) and Low Dose Dexamethasone (LLD) Versus Bortezomib (PS-341, NSC-681239), Lenalidomide and Low Dose Dexamethasone (BLLD) for Induction, in Patients With Previously Untreated Multiple Myelom [NCT00644228]Phase 3525 participants (Actual)Interventional2008-04-01Active, not recruiting
An Intergroup Phase III Randomized Controlled Trial Comparing Melphalan, Prednisone and Thalidomide (MPT) Versus Melphalan, Prednisone and Lenalidomide (Revlimid(TM))(MPR) in Newly Diagnosed Multiple Myeloma Patients Who Are Not Candidates for High-Dose T [NCT00602641]Phase 3306 participants (Actual)Interventional2008-02-29Active, not recruiting
A Phase III Randomized, Double-Blind Study of Maintenance Therapy With CC-5013 (NSC # 703813) or Placebo Following Autologous Stem Cell Transplantation for Multiple Myeloma [NCT00114101]Phase 3460 participants (Actual)Interventional2004-12-15Active, not recruiting
A Randomized Phase III Study of CC-5013 Plus Dexamethasone Versus CC-5013 Plus Low Dose Dexamethasone in Multiple Myeloma With Thalidomide Plus Dexamethasone Salvage Therapy for Non-Responders [NCT00098475]Phase 3452 participants (Actual)Interventional2004-11-03Active, not recruiting
An Open-Label Phase 2 Study of Lenalidomide (Revlimid) in Combination With Oral Dexamethasone in the Treatment of Previously Untreated, Symptomatic Patients With Chronic Lymphocytic Leukemia (CLL) [NCT01133743]Phase 231 participants (Actual)Interventional2010-05-31Completed
A Phase II Study of the Efficacy and Safety of Lenalidomide, Subcutaneous Bortezomib and Dexamethasone Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma [NCT02441686]Phase 245 participants (Anticipated)Interventional2015-12-31Active, not recruiting
An Open Label, Multicenter, Phase II Study of Belantamab Mafodotin in Combination With VRd for the Treatment of Newly Diagnosed Transplant Eligible Multiple Myeloma Patients [NCT04802356]Phase 250 participants (Anticipated)Interventional2021-04-07Recruiting
A National, Open-label, Multicenter, Randomized, Comparative Phase IIb Study of Treatment for Newly Diagnosed Multiple Myeloma Patients Older Than 65 Years With Sequential Melphalan/Prednisone/Velcade (MPV) Followed by Revlimid/Low Dose Dexamethasone (Rd) [NCT01237249]Phase 2250 participants (Actual)Interventional2011-02-28Completed
Phase 2 Trial of LDE225 and Lenalidomide Maintenance Post Autologous Stem Cell Transplant for Multiple Myeloma [NCT02086552]Phase 228 participants (Actual)Interventional2014-01-17Completed
Phase Ib of CC-5013 and Paclitaxel in Patients With Advanced Solid Tumors [NCT01155505]Phase 128 participants (Anticipated)Interventional2009-11-30Active, not recruiting
Lenalidomide Based Immunotherapy Efficacy Related Molecular Biomarker in Diffuse Large B-cell Lymphoma [NCT03715296]200 participants (Anticipated)Observational [Patient Registry]2018-10-18Recruiting
Phase 1/2 Study of Lenalidomide and Cetuximab in Patients With Advanced Solid Tumors [NCT01166035]Phase 1/Phase 220 participants (Anticipated)Interventional2010-03-31Recruiting
Efficacy and Safety Study of Rituximab, Methotrexate and Lenalidomide Chemotherapy(R2-MTX) in Newly Diagnosed Primary Central Nervous System Lymphoma:a Single Arm, Multicenter, Phase 2 Study [NCT04934579]Phase 217 participants (Actual)Interventional2020-01-01Completed
A Phase I Study of Lenalidomide in Combination With Bevacizumab, Sorafenib, Temsirolimus, or 5-fluorouracil, Leucovorin, Oxaliplatin (FOLFOX) in Patients With Advanced Cancers [NCT01183663]Phase 1180 participants (Actual)Interventional2010-08-31Completed
IFM2008: Frontline Therapy in de Novo Multiple Myeloma Patients Under 65, (a Phase 2 Multicenter Trial) [NCT01206205]Phase 231 participants (Actual)Interventional2009-08-31Completed
Lenalidomide, Rituximab, Gemcitabine, Oxaliplatin and Dexamethasone (R2-GOD) in Treatment of Relapse/Refractory DLBCL:A Phase I Study [NCT03795571]Phase 112 participants (Anticipated)Interventional2019-01-01Recruiting
A Phase 1, Open-label, Randomized, Three-period, Two-way Crossover Study in Healthy Subjects to Evaluate the Bioavailability of a Test Lenalidomide Oral Suspension Relative to the Reference Capsule Formulation and to Assess the Effect of Food on the Bioav [NCT02521714]Phase 128 participants (Anticipated)Interventional2015-08-14Completed
A Phase 3 Randomized, Open-label, Multicenter Study Assessing the Clinical Benefit of Isatuximab (SAR650984) in Combination With Bortezomib (Velcade®), Lenalidomide and Dexamethasone Versus Bortezomib, Lenalidomide and Dexamethasone in Patients With Newly [NCT03319667]Phase 3475 participants (Actual)Interventional2017-12-07Active, not recruiting
A Phase II Study of Daily Alternating Thalidomide and Lenalidomide Therapy Plus Rituximab (ThRiL) as Initial Treatment for Patients With CLL [NCT01125176]Phase 215 participants (Actual)Interventional2012-03-30Completed
Enhancement of Cetuximab-Induced Antibody-Dependent Cellular Cytotoxicity (ADCC) With Lenalidomide in Advanced Solid Tumors: A Phase I/IB Study [NCT01254617]Phase 124 participants (Actual)Interventional2011-02-10Completed
A Phase 1 Study of MLN9708 in Japanese Patients With Relapsed and/or Refractory Multiple Myeloma [NCT04272775]Phase 114 participants (Actual)Interventional2012-06-05Terminated(stopped due to Business Decision; No Safety Or Efficacy Concerns)
A Clinical Trial of Programmed Cell Death Protein 1(PD1) Antibody and Lenalidomide as a Treatment for Epstein-Barr Virus-associated Hemophagocytic Lymphohistiocytosis or Chronic Active EBV Infection(CAEBV) [NCT04084626]Phase 340 participants (Anticipated)Interventional2019-09-15Recruiting
A Multicentric, Phase II Trial of Lenalidomide, Cyclophosphamide and Dexamethasone in Patients With Primary Systemic Amyloidosis (AL) Newly Diagnosed, Not Candidates for Hematopoietic Stem Cell Transplantation [NCT01194791]Phase 230 participants (Anticipated)Interventional2010-10-31Completed
A Single Arm, Multi-center, Phase II Clinical Trial of Rituximab, Lenalidomide Combined With High-dose Methotrexate and Temozolomide (RL-MT) in the First-line Treatment for Patients With Primary Central Nervous System Lymphoma [NCT04737889]Phase 230 participants (Anticipated)Interventional2021-01-13Recruiting
Randomized Phase II Trial on Fitness- and Comorbidity- Tailored Treatment in Elderly Patients With Newly Diagnosed Primary CNS Lymphoma (FIORELLA Trial) [NCT03495960]Phase 2208 participants (Anticipated)Interventional2019-06-15Recruiting
Iceland Screens Treats or Prevents Multiple Myeloma (iStopMM): A Nationwide Phase 2 Trial of Patients With Smoldering and Active Multiple Myeloma (MM) [NCT03815279]Phase 280 participants (Anticipated)Interventional2019-06-24Enrolling by invitation
A Phase 1 Study of Lenalidomide in Combination With Vorinostat in Pediatric Patients With High Grade or Progressive Central Nervous System Tumors [NCT03050450]Phase 18 participants (Actual)Interventional2016-08-10Terminated(stopped due to Lack of feasibility to accrue patients in allotted time.)
Randomized Phase II Trial Seeking the Most Promising Drug Association With Azacitidine- in Higher Risk Myelodysplastic Syndromes [NCT01342692]Phase 2320 participants (Anticipated)Interventional2011-06-30Active, not recruiting
A Randomized, Multicenter, Phase 3 Study Comparing Carfilzomib, Lenalidomide, and Dexamethasone (CRd) vs Lenalidomide and Dexamethasone (Rd) in Subjects With Relapsed Multiple Myeloma [NCT01080391]Phase 3792 participants (Actual)Interventional2010-07-14Completed
A Phase 3 Multicenter, Randomized, Double-blind, Placebo-Controlled, First Line Maintenance Study Of Lenalidomide (Revlimid®) In Patients With Mantle-Cell Lymphoma [NCT01021423]Phase 39 participants (Actual)Interventional2010-04-01Terminated(stopped due to Terminated by sponsor due to new unpublished data that rendered the current design of the study no longer clinically relevant. There were no safety concerns.)
A Phase 3, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study To Compare The Efficacy And Safety of Lenalidomide (Revlimid®) Versus Placebo In Subjects With Transufsion-Dependent Anemia Due to IPSS Low Or Imtermidate-1 Risk My [NCT01029262]Phase 3239 participants (Actual)Interventional2010-01-26Completed
A Prospective Phase 2 Study to Assess the Minimal Residual Disease After Ixazomib Plus Lenalidomide Plus Dexamethasone (IRd) Treatment for Newly Diagnosed Transplant Eligible Myeloma Patients [NCT03376672]Phase 2120 participants (Actual)Interventional2018-05-31Active, not recruiting
Response Adapted Therapy With Bortezomib/Dexamethasone Followed by Addition of Lenalidomide in Non Responders as Initial Treatment for Patients With Multiple Myeloma [NCT01919086]Phase 230 participants (Actual)Interventional2013-08-31Active, not recruiting
"A PHASE 3 OPEN-LABEL RANDOMIZED STUDY TO COMPARE THE EFFICACY AND SAFETY OF RITUXIMAB PLUS LENALIDOMIDE (CC-5013) VERSUS RITUXIMAB PLUS CHEMOTHERAPY FOLLOWED BY RITUXIMAB IN SUBJECTS WITH PREVIOUSLY UNTREATED FOLLICULAR LYMPHOMA The RELEVANCE Trial (Ritu [NCT01650701]Phase 31,030 participants (Actual)Interventional2012-02-29Active, not recruiting
Carfilzomib, Daratumumab, Lenalidomide and Dexamethasone as First Line Treatment in Multiple Myeloma [NCT04288765]Phase 30 participants (Actual)Interventional2020-03-01Withdrawn(stopped due to incomplete recruitment)
Phase II Study of Bendamustine and Rituximab Induction Chemoimmunotherapy With Maintenance Lenalidomide and Rituximab in Relapsed/Refractory CLL/SLL [NCT00974233]Phase 234 participants (Actual)Interventional2009-10-31Completed
A Phase I Study of Safety, Tolerability and Immunological Effects of SVN53-67/M57-KLH in Patients With Multiple Myeloma Receiving Lenalidomide Maintenance Therapy [NCT02334865]Phase 118 participants (Actual)Interventional2017-04-13Active, not recruiting
Phase 1b Multicenter Dose Escalation Study of Carfilzomib With Lenalidomide and Dexamethasone for Safety and Activity in Relapsed Multiple Myeloma [NCT00603447]Phase 184 participants (Actual)Interventional2008-05-31Completed
A Phase 3 Study to Evaluate the Efficacy and Safety of Docetaxel and Prednisone With or Without Lenalidomide in Subjects With Castrate-Resistant Prostate Cancer (CRPC) [NCT00988208]Phase 31,059 participants (Actual)Interventional2009-11-11Completed
A Phase 1/2, Multicenter, Open-Label, Dose-Escalation Study to Evaluate the Safety and Efficacy of Lenalidomide in Combination With Sunitinib in Subjects With Advanced or Metastatic Renal Cell Carcinoma [NCT00975806]Phase 1/Phase 216 participants (Actual)Interventional2009-09-01Terminated(stopped due to MTD determined sub-optimal as efficacious treatment for renal cell carcinoma.)
Fludarabine/Rituximab Combined With Escalating Doses of Lenalidomide Followed by Rituximab/Lenalidomide in Untreated Chronic Lymphocytic Leukemia (CLL) - a Dose-finding Study With Concomitant Evaluation of Safety and Efficacy. [NCT00738829]Phase 1/Phase 245 participants (Actual)Interventional2008-10-31Completed
A Prospective, Multicenter, Single Arm, Phase II Clinical Trial of Clarithromycin, Lenalidomide and Dexamethasone (BiRd Regimen) in the Treatment of the First Relapsed Multiple Myeloma [NCT04063189]Phase 2100 participants (Anticipated)Interventional2017-03-21Recruiting
A Multicentre Phase II Study of the Efficacy and Safety of Lenalidomide in High-risk Myeloid Disease (High-risk MDS and AML) With a Karyotype Including Del(5q) or Monosomy 5 [NCT00761449]Phase 228 participants (Actual)Interventional2007-10-31Completed
Phase Ib / II Open-Label Stduy of APG-2575 Monotherapy or in Combination With Lenalidomide / Dexamethasone in Subjects With Relapsed or Refractory Multiple Myeloma [NCT04674514]Phase 1/Phase 248 participants (Anticipated)Interventional2021-04-13Recruiting
A Randomized Phase III Trial Assessing the Benefit of the Addition of Isatuximab to Lenalidomide / Bortezomib / Dexamethasone (RVd) Induction and Lenalidomide Maintenance in Patients With Newly Diagnosed Multiple Myeloma [NCT03617731]Phase 3662 participants (Actual)Interventional2018-10-18Active, not recruiting
Lenalidomide, Adriamycin, Dexamethasone (RAD) Versus Lenalidomide, Bortezomib, Dexamethasone (VRD) for Induction in Newly Diagnosed Multiple Myeloma Followed by Response-adapted Consolidation and Lenalidomide Maintenance - A Randomized Multicenter Phase I [NCT01685814]Phase 3406 participants (Anticipated)Interventional2012-05-31Active, not recruiting
A Phase II Trial to Evaluate the Safety and Activity of Single-agent Lenalidomide Given as Maintenance Therapy After Response to Second-line Therapy in Patients With Relapsed DLBCL, Not Eligible for High-dose Chemotherapy and ASCT [NCT00799513]Phase 247 participants (Anticipated)Interventional2009-10-31Recruiting
A Phase 3, Multicenter, Randomized, Openlabel, Parallel-Group Study of the Efficacy and Safety of Lenalidomide (Revlimid®) Versus Chlorambucil as First-Line Therapy for Previously Untreated Elderly Patients With B-Cell Chronic Lymphocytic Leukemia (The Or [NCT00910910]Phase 3450 participants (Actual)Interventional2009-10-13Completed
Safety And Efficacy Of Lenalidomide As Maintenance Therapy In Patients With Newly Diagnosed Multiple Myeloma Following A Tandem Autologous-Allogeneic Transplant [NCT01264315]Phase 253 participants (Actual)Interventional2008-09-30Completed
Open-Label, One Arm Pilot Investigation of Lenalidomide Therapy for Patients With Relapsed and/or Refractory, Peripheral T-Cell Lymphomas [NCT00704691]Early Phase 11 participants (Actual)Interventional2008-06-30Terminated(stopped due to Closed due to futility with only 1 patient accrued)
Open-Label, Multi-Center Phase I Dose-Escalation Study With Lenalidomide In Patients With Acute Myeloid Leukemia [NCT00839059]Phase 114 participants (Actual)Interventional2009-01-31Terminated(stopped due to Results of an interim analysis and a hardly ongoing enrolment in the last 10 months in all six participating centres)
"A Phase I Pilot Study of Immunotherapy Using Lenalidomide Plus Bystander Vaccine in Patients With High-Risk Myelodysplastic Syndrome (MDS)" [NCT00840931]Phase 122 participants (Actual)Interventional2009-02-02Completed
Single-arm, Multi-center Clinical Study of PD-1 Antibody, Chidamide, Lenalidomide and Gemcitabine for Peripheral T-cell Lymphoma [NCT04040491]Phase 4100 participants (Anticipated)Interventional2019-09-01Recruiting
Phase 1 Study of Venetoclax, Ibrutinib, Prednisone, Obinutuzumab, and Revlimid in Combination With Polatuzumab (ViPOR-P) in Relapsed/Refractory B-cell Lymphoma [NCT04739813]Phase 155 participants (Anticipated)Interventional2021-07-09Recruiting
A Phase 1 Study of Lenalidomide in Combination With EPOCH Chemotherapy for HTLV-Associated Adult T-Cell Leukemia-Lymphoma (ATLL) [NCT04301076]Phase 130 participants (Anticipated)Interventional2021-08-31Recruiting
Aggressive Smoldering Curative Approach Evaluating Novel Therapies (ASCENT): A Phase 2 Trial of Induction, Consolidation, and Maintenance in Subjects With High Risk Smoldering Multiple Myeloma (SMM) [NCT03289299]Phase 287 participants (Actual)Interventional2018-05-25Active, not recruiting
Phase II Study of Umbilical Cord Blood-Derived Natural Killer Cells in Conjunction With Elotuzumab, Lenalidomide and High Dose Melphalan Followed by Autologous Stem Cell Transplant for Patients With Multiple Myeloma [NCT01729091]Phase 272 participants (Anticipated)Interventional2013-06-10Active, not recruiting
A Randomized Phase II Dose Finding Study of Revlimid™ and Melphalan in Patients With Previously Untreated Multiple Myeloma [NCT00305812]Phase 251 participants (Actual)Interventional2006-03-09Completed
Selinexor Combined With Prednisone, Etoposide, and Lenalidomide as Introductive Treatment Following Immune-chemotherapy as Consolidated Therapy for Refractory Diffuse Large B-cell Lymphoma With p53 and/or c-Myc Expression [NCT05498636]Phase 1/Phase 267 participants (Anticipated)Interventional2022-10-01Not yet recruiting
A Single-arm, Phase Ⅰ/Ⅱ Study Evaluating the Safety, Tolerability, and Preliminary Efficacy of ATG-010 in Combination With Lenalidomide and Rituximab (R2) in Adult Patients With Relapsed/Refractory DLBCL and iNHL Who Are Ineligible for High-dose Chemother [NCT05265975]Phase 1/Phase 284 participants (Anticipated)Interventional2022-04-07Recruiting
Lenalidomide in Combination With Plerixafor in Patients With Previously Treated Chronic Lymphocytic Leukemia [NCT01373229]Phase 121 participants (Actual)Interventional2012-01-31Completed
A Phase I/II Trial of Romidepsin, Rituximab and Lenalidomide (R3) in Relapsed/Refractory B Cell Lymphomas Including Transformed Follicular Lymphoma [NCT02281279]Phase 1/Phase 20 participants (Actual)Interventional2016-10-31Withdrawn
A Phase 1 Open-Label Study of the Safety and Efficacy of PD 0332991 (Palbociclib) in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT02030483]Phase 19 participants (Actual)Interventional2014-02-28Terminated(stopped due to The company providing one of the study drugs withdrew its support due to low enrollment. Therefore, we had to close the study due to lack of funding.)
A Phase 1 Pharmacokinetic and Tolerability Study of Oral MLN9708 Plus Lenalidomide and Dexamethasone in Adult Asian Patients With Relapsed and/or Refractory Multiple Myeloma [NCT01645930]Phase 143 participants (Actual)Interventional2012-12-17Completed
A Phase 2 Randomized, Open-Label, Multicenter Study to Evaluate the Efficacy and Safety of XmAb13676 (Plamotamab) Combined With Tafasitamab Plus Lenalidomide Versus Tafasitamab Plus Lenalidomide in Subjects With Relapsed or Refractory Diffuse Large B-Cell [NCT05328102]Phase 23 participants (Actual)Interventional2022-04-25Terminated(stopped due to The study has been terminated early by the sponsor due to business decision.)
A Pilot Study Evaluating Lenalidomide and CC-486 in Combination With Radiotherapy For Patients With Plasmacytoma (LENAZART Study) [NCT04174196]Phase 220 participants (Anticipated)Interventional2019-11-19Recruiting
Phase I/II Study of Lenalidomide (Revlimid), All-trans Retinoic Acid (ATRA) and Dexamethasone in Patients With Relapsed/Refractory Multiple Myeloma [NCT01985477]Phase 12 participants (Actual)Interventional2013-12-31Terminated(stopped due to Terminated Phase I due to slow accrual without progression to Phase II.)
"An Intensive Program With Quadruplet Induction and Consolidation Plus Tandem Autologous Stem Cell Transplantation in Newly Diagnosed High Risk Multiple Myeloma Patients: a Phase II Study of the Intergroupe Francophone du Myélome IFM 2018-04" [NCT03606577]Phase 250 participants (Anticipated)Interventional2019-07-30Active, not recruiting
A Phase II Study of the Efficacy and Safety of Lenalidomide Plus ICE in the Treatment of Refractory and Relapsed Diffuse Large B-cell Lymphoma [NCT03367143]Phase 239 participants (Anticipated)Interventional2016-12-31Active, not recruiting
Immuno-PRISM (PRecision Intervention Smoldering Myeloma): A Randomized Phase II Platform Study of Select Immunotherapies for High-Risk Smoldering Myeloma [NCT05469893]Phase 251 participants (Anticipated)Interventional2022-08-10Recruiting
A Prospective, Single-center, Phase II Study of Venetoclax/Selinexor Plus VRD Combined With CART-ASCT-CART2 Treatment in Patients With Newly Diagosed Primary Plasma Cell Leukemia [NCT05870917]Phase 220 participants (Anticipated)Interventional2023-04-25Recruiting
The Efficacy and Safety of Zanubrutinib, Rituximab and Lenalidomide (ZR2) Versus Rituximab Combined With Low-dose CHOP (R-miniCHOP) in the Treatment of Unfit or Frail de Novo Diffuse Large B-cell Lymphoma Patients Aged Older Than or Equal to 70 Years: A M [NCT05179733]Phase 3280 participants (Anticipated)Interventional2022-03-02Recruiting
A Phase 1, Open Label, Multiple Dose, Dose Escalation and Expansion Study of Bruton Tyrosine Kinase (BTK) Inhibitor, Zanubrutinib, in Combination With Lenalidomide, With or Without Rituximab in Patients With Relapsed/Refractory Diffuse Large B-Cell Lympho [NCT04436107]Phase 167 participants (Anticipated)Interventional2020-09-11Recruiting
A Phase I/II, Open-label, Dose Escalation and Expansion Study to Evaluate Safety, Tolerability, and Clinical Activity of the Antibody-Drug Conjugate GSK2857916 Administered in Combination With Lenalidomide Plus Dexamethasone (Arm A), or Bortezomib Plus De [NCT03544281]Phase 1/Phase 2152 participants (Actual)Interventional2018-09-20Active, not recruiting
A Randomized Phase I/II Study of Optimal Induction Therapy of Bortezomib, Dexamethasone and Lenalidomide With or Without Elotuzumab (NSC-764479) for Newly Diagnosed High Risk Multiple Myeloma (HRMM) [NCT01668719]Phase 1/Phase 2142 participants (Actual)Interventional2012-11-30Active, not recruiting
A Phase I Trial of Brentuximab Vedotin in Combination With Lenalidomide in Relapsed or Refractory Diffuse Large B-cell Lymphoma [NCT02086604]Phase 137 participants (Actual)Interventional2014-09-18Completed
Phase 2 Trial of Belantamab Mafodotin Consolidation Treatment in Patients With Multiple Myeloma and MRD Positivity After Autologous Stem Cell Transplantation [NCT04876248]Phase 220 participants (Anticipated)Interventional2023-12-15Recruiting
A Phase Ib/II, Open-Label, Multicenter Study With a Non-Randomized Stage Evaluating the Safety, Pharmacokinetics, and Efficacy of Mosunetuzumab Plus Lenalidomide (+Len), and a Randomized Stage Evaluating the Safety, Tolerability, and Pharmacokinetics of S [NCT04246086]Phase 1/Phase 2187 participants (Anticipated)Interventional2020-08-12Active, not recruiting
An Open-Label, Single-Arm, Multicenter Study to Evaluate the Effectiveness and Safety of Ixazomib (NINLARO®) in Combination With Lenalidomide and Dexamethasone (IRD) in Patients With Multiple Myeloma Previously Receiving a Bortezomib-Based Induction Regim [NCT03173092]Phase 4141 participants (Actual)Interventional2017-09-20Active, not recruiting
An Expanded Access Program for Elotuzumab in Combination With Lenalidomide Plus Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT02368301]0 participants Expanded AccessNo longer available
An Open-label, Phase 1b/2 Study of Acalabrutinib Alone or in Combination Therapy in Subjects With B-cell Non-Hodgkin Lymphoma [NCT02180711]Phase 1113 participants (Actual)Interventional2014-12-29Active, not recruiting
Phase 3B, Randomized Trail of Revlimid® (Lenalidomide) Versus Placebo Maintenance Therapy Following Melphalan Prednisone Velcade (Bortezomib) Induction Therapy In Newly Diagnosed Multiple Myeloma [NCT02112175]Phase 346 participants (Actual)Interventional2014-04-30Completed
A Multicenter Clinical Study of Orelabrutinib Combined With Lenalidomide and Rituximab (OR2) in the Treatment of Recurrent and Refractory CD20+ B-cell Lymphoma [NCT05014100]Phase 255 participants (Anticipated)Interventional2021-09-01Not yet recruiting
Phase 2 Trial of Intensive Chemo-immunotherapy With Carfilzomib, Lenalidomide, Dexamethasone and Daratumumab for Relapsed/Refractory Myeloma in the Context of Salvage Autologous Hematopoietic Cell Transplantation [NCT03556332]Phase 241 participants (Actual)Interventional2018-07-02Active, not recruiting
"A Phase III Randomized Trial for Newly Diagnosed Multiple Myeloma (NDMM) Patients Considered Frail or in a Subset of Intermediate Fit Comparing Upfront Three-Drug Induction Regimens Followed by Double or Single-Agent Maintenance" [NCT05561387]Phase 3510 participants (Anticipated)Interventional2023-10-12Recruiting
A Phase Ib/II Open-Label Study of APG-2575 in Combination With Novel Therapeutic Regimens in Subjects With Relapsed or Refractory Multiple Myeloma and Immunoglobin Light Chain Amyloidosis [NCT04942067]Phase 1/Phase 2108 participants (Anticipated)Interventional2021-12-23Recruiting
A Phase 2, Open Label, Randomized Trial Evaluating Ixazomib Compared to Ixazomib-Lenalidomide Combination Maintenance Therapy for Frontline Multiple Myeloma Patients [NCT03733691]Phase 219 participants (Actual)Interventional2019-03-01Terminated(stopped due to Insufficient enrollment)
A Phase 3, Open Label, Randomized Study to Compare the Efficacy and Safety of Odronextamab (REGN1979), an Anti-CD20 x Anti-CD3 Bispecific Antibody, in Combination With Lenalidomide Versus Rituximab in Combination With Lenalidomide Therapy in Relapsed/Refr [NCT06149286]Phase 3470 participants (Anticipated)Interventional2023-12-18Recruiting
A Randomized, Double-blind, Placebo-Controlled, Active-Comparator, Multicenter, Phase 3 Study of Brentuximab Vedotin or Placebo in Combination With Lenalidomide and Rituximab in Subjects With Relapsed or Refractory Diffuse Large B-cell Lymphoma (DLBCL) [NCT04404283]Phase 3240 participants (Actual)Interventional2020-08-20Active, not recruiting
Sub-cutaneous Rituximab-miniCHOP Versus Sub-cutaneous Rituximab-miniCHOP + Lenalidomide (R2-miniCHOP) in Diffuse Large B Cell Lymphoma for Patients of 80 Years Old or More. A Multicentric Phase III Study of the LYSA Association [NCT02128061]Phase 3250 participants (Actual)Interventional2014-08-31Completed
A Phase 1, Randomized, Dose and Schedule Evaluation Study to Investigate the Safety, Pharmacokinetics, Pharmacodynamics and Clinical Activity of Belantamab Mafodotin Administered in Combination With Standard of Care in Participants With Newly Diagnosed Mu [NCT04091126]Phase 1144 participants (Anticipated)Interventional2019-12-18Recruiting
A Prospective, Single-arm, Multicenter Clinical Study of Mitoxantrone Hydrochloride Liposome Combined With Rituximab and Lenalidomide in the Treatment of Relapsed and Refractory Diffuse Large B-cell Lymphoma [NCT05575973]Phase 255 participants (Anticipated)Interventional2022-10-10Not yet recruiting
A Pilot Study Examining Selinexor's Ability to Overcome Resistance in Multiple Myeloma Patients Who Are Refractory to Lenalidomide-containing Therapy. [NCT04519476]Phase 122 participants (Anticipated)Interventional2020-11-01Recruiting
A Phase II Study of Brentuximab Vedotin and Lenalidomide in Relapsed and Refractory T-Cell Lymphomas [NCT03409432]Phase 226 participants (Actual)Interventional2018-03-16Active, not recruiting
Alternating the Administration of Ixazomib and Lenalidomide as Maintenance Therapy After Autologous Transplant for Treating Multiple Myeloma [NCT02619682]Phase 230 participants (Actual)Interventional2015-12-30Active, not recruiting
A Protocol to Assess the Safety, Efficacy, and Pharmacokinetics of Continuous Subcutaneous Administration of Low-dose Lenalidomide (STAR-LLD) for the Treatment of Multiple Myeloma (MM) [NCT06087653]Phase 1/Phase 26 participants (Anticipated)Interventional2023-10-02Recruiting
Phase III Study of Isatuximab-Carfilzomib-Lenalidomide-Dexamethasone (Isa-KRd) Versus Carfilzomib-Lenalidomide-Dexamethasone (KRd) in Newly Diagnosed Multiple Myeloma Patients Eligible for Autologous Stem Cell Transplantation (IsKia TRIAL [NCT04483739]Phase 3302 participants (Actual)Interventional2020-09-25Active, not recruiting
Phase II Study of Lenalidomide Plus Rituximab in Patients With Previously Untreated Mantle Cell Lymphoma [NCT01472562]Phase 238 participants (Actual)Interventional2011-07-29Completed
International, Multi-center, Open-label, Treatment Extension Study in Patients With Multiple Myeloma Who Are Still Benefitting From Isatuximab-based Therapy Following Completion of a Phase 1, 2, or 3 Parental Study [NCT05669989]Phase 270 participants (Anticipated)Interventional2023-04-05Recruiting
A Phase 1/2, Open-label, Multi-center Study to Assess the Safety and Tolerability of Durvalumab (Anti-PDL1 Antibody) as Monotherapy and in Combination Therapy in Subjects With Lymphoma or Chronic Lymphocitic Leukemia [NCT02733042]Phase 1/Phase 2106 participants (Actual)Interventional2016-05-11Completed
Phase II Trial of Lenalidomide in Patients With Lymphoma of the Mucosa Associated Lymphoid Tissue (MALT) Type [NCT00923663]Phase 216 participants (Anticipated)Interventional2009-07-31Recruiting
Lenalidomide, Bendamustine and Rituximab as First-line Therapy for Patients Over 65 Years With Mantle Cell Lymphoma - a Nordic Lymphoma Group Trial [NCT00963534]Phase 1/Phase 251 participants (Actual)Interventional2009-09-30Completed
A Phase I/II Trial of Lenalidomide Combined With Cyclophosphamide and Intermediate Dose Dexamethasone in Patients With Primary (AL) Systemic Amyloidosis [NCT00981708]Phase 1/Phase 237 participants (Actual)Interventional2008-02-29Completed
Phase 2, Multi-Center, Single-Arm, Open-Label Study to Evaluate the Efficacy and Safety of the Combination Regimen Isatuximab, Lenalidomide, Bortezomib, and Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma [NCT04653246]Phase 243 participants (Anticipated)Interventional2021-07-13Recruiting
A Phase I Trial Evaluating the Safety and Efficacy of Vorinostat (Zolinza ®) + RVD (Lenalidomide {Revlimid ®} + Bortezomib {Velcade ®} + Dexamethasone) for Patients With Newly Diagnosed Multiple Myeloma [NCT01038388]Phase 130 participants (Actual)Interventional2010-01-15Completed
A Phase II Trial of MRD (Melphalan, Lenalidomide and Dexamethasone) for Patients With AL Amyloidosis [NCT00679367]Phase 216 participants (Actual)Interventional2008-05-31Completed
Phase I/II Trial of Lenalidomide in Combination With Vorinostat and Dexamethasone as Therapy in Relapsed or Refractory Patients With Peripheral T-Cell Non-Hodgkin's Lymphoma (PTCL) [NCT00972842]Phase 1/Phase 220 participants (Anticipated)Interventional2009-09-30Terminated(stopped due to slow recruitment)
Phase II Study for the Determination of Efficacy and Tolerability of the Combination of Valproic Acid and Lenalidomide in the Treatment of Patients With Myelodysplastic Syndrome With Favorable Risk Profile [NCT00977132]Phase 223 participants (Actual)Interventional2009-10-31Terminated(stopped due to delayed recruitment)
Phase II Study of Revlimid®, Oral Cyclophosphamide and Prednisone (RCP) for Patients With Newly Diagnosed Multiple Myeloma [NCT00540644]Phase 270 participants (Actual)Interventional2007-10-31Completed
REVLIMID® Drug Use Examination [NCT02556905]624 participants (Actual)Observational2011-03-09Completed
Salvage Treatment With Lenalidomide and Dexamethaosne(LEN-DEX) in Patients With Relapsed/Refractory Mantle Cell Lymphoma (MCL) [NCT00786851]Phase 233 participants (Actual)Interventional2008-07-31Completed
Phase I/II Trial of Allogeneic Peripheral Blood Stem Cell Transplantation Followed by Maintenance Therapy With Lenalidomide and Sirolimus in Patients With High-Risk Multiple Myeloma [NCT01303965]Phase 1/Phase 214 participants (Actual)Interventional2011-02-07Terminated(stopped due to Slow accrual)
The Official Title is A Multi-center, Randomized, Parallel-group, Double-blind, Placebo Controlled Study of CC-5013 Plus Dexamethasone Versus Dexamethasone Alone in Previously Treated Subjects With Multiple Myeloma. [NCT00424047]Phase 3351 participants (Actual)Interventional2003-01-01Completed
Lenalidomide and Dexamethasone (Rd) Versus Clarithromycin [Biaxin®] / Lenalidomide [Revlimid®] / Dexamethasone (BiRd) as Initial Therapy in Multiple Myeloma [NCT02516696]Phase 312 participants (Actual)Interventional2016-02-29Terminated(stopped due to low enrollment)
A Randomized Phase II Trial of Rituximab Versus Lenalidomide (REVLIMID™, Cc-5013) (IND#73034) Versus Rituximab + Lenalidomide in Recurrent Follicular Non-Hodgkin Lymphoma (NHL) That is Not Rituximab-Refractory [NCT00238238]Phase 297 participants (Actual)Interventional2006-03-31Completed
Phase 2 Trial of Lenalidomide (Revlimid)-Dexamethasone + Rituximab in Recurrent Small B-Cell Non-Hodgkin Lymphomas (NHL) Resistant to Rituximab [NCT00783367]Phase 250 participants (Actual)Interventional2008-07-31Completed
A Pilot Study of Lenalidomide, Melphalan and Dexamethasone in AL Amyloidosis [NCT00890552]25 participants (Actual)Interventional2009-04-30Completed
A Phase IB Study of Escalating Doses of REVLIMID in Association With R-CHOP (R2-CHOP) in the Treatment of B-cell Lymphoma [NCT00901615]Phase 1/Phase 2108 participants (Actual)Interventional2009-01-06Completed
A Phase 3, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study of the Efficacy and Safety of Lenalidomide (Revlimid®) as Maintenance Therapy for Patients With B-Cell Chronic Lymphocytic Leukemia Following Second-Line Therapy (T [NCT00774345]Phase 3317 participants (Actual)Interventional2009-01-27Completed
Carfilzomib, Lenalidomide, and Dexamethasone in High-Risk Smoldering Multiple Myeloma: a Clinical and Correlative Pilot Study [NCT01572480]Phase 255 participants (Actual)Interventional2012-05-29Active, not recruiting
Lenalidomide in the Treatment of Mucosal Behçet's Syndrome [NCT05449548]42 participants (Anticipated)Interventional2023-04-01Recruiting
A Phase 2 Evaluation of Daratumumab-Based Treatment in Newly Diagnosed Multiple Myeloma Patients With Renal Insufficiency [NCT04352205]Phase 225 participants (Anticipated)Interventional2020-05-07Recruiting
A Phase 2 Study of Subcutaneous Daratumumab in Combination With Dose-Attenuated Bortezomib, Lenalidomide, and Dexamethasone in Elderly Newly Diagnosed Multiple Myeloma Patients [NCT04052880]Phase 238 participants (Anticipated)Interventional2019-10-24Recruiting
A Phase II Trial of Lenalidomide (Revlimid®), Cyclophosphamide and Dexamethasone in Patients With Primary Systemic Amyloidosis [NCT00564889]Phase 235 participants (Actual)Interventional2007-12-31Completed
A Prospective Single-center Study on the Efficacy and Safety of Lenalidomide Combined With Azacitidine vs Azacitidine in the Treatment of MDS-RS [NCT06004765]Phase 4138 participants (Anticipated)Interventional2023-08-31Not yet recruiting
A Phase II Trial of the Anti -PD-1 Monoclonal Antibody Pembrolizumab (MK-3475) + Lenalidomide + Dexamethasone as Post Autologous Transplant Consolidation in Patients With High-risk Multiple Myeloma [NCT02906332]Phase 212 participants (Actual)Interventional2016-12-12Terminated(stopped due to FDA Hold Due to Updated Risks)
A Randomized Phase III Study to Compare Bortezomib, Melphalan, Prednisone (VMP) With High Dose Melphalan Followed by Bortezomib, Lenalidomide, Dexamethasone (VRD) Consolidation and Lenalidomide Maintenance in Patients With Newly Diagnosed Multiple Myeloma [NCT01208766]Phase 31,503 participants (Actual)Interventional2011-01-31Active, not recruiting
Combination of Lenalidomide and Sintilimab for Patients With Relapsed/Refractory NK/T-cell Lymphoma Who Failed Pegaspargase-based Regimens: a Single Arm, Open, Phase II Study [NCT04231370]Phase 220 participants (Anticipated)Interventional2020-04-01Recruiting
A Randomized Phase II Study of Lenalidomide Maintenance Therapy in AML Patients Aged > 60 Years in CR1 or Higher and < 60 Years in CR2 or Higher [NCT00957385]Phase 224 participants (Actual)Interventional2008-06-30Completed
The Efficacy and Safety of ZR2 Versus R-CHOP-like Regimen for Elderly Patients With Newly Diagnosed Diffuse Large B Cell Lymphoma. [NCT05428670]Phase 2150 participants (Anticipated)Interventional2022-06-15Recruiting
MCLENA-1: A Phase II Clinical Trial for the Assessment of Safety, Tolerability, and Efficacy of Lenalidomide in Patients With Mild Cognitive Impairment Due to Alzheimer's Disease [NCT04032626]Phase 230 participants (Anticipated)Interventional2020-07-22Recruiting
Selinexor With Alternating Bortezomib or Lenalidomide Plus Dexamethasone in Transplant Ineligible Newly Diagnosed Multiple Myeloma Patients (SABLe): An Investigator Sponsored Trial [NCT04717700]Phase 250 participants (Anticipated)Interventional2021-08-18Recruiting
A Dose Escalation, Safety, Pharmacokinetic, Pharmacodynamic and Preliminary Efficacy Study of SAR650984 (Isatuximab) Administered Intravenously in Combination With Bortezomib - Based Regimens in Adult Patients With Newly Diagnosed Multiple Myeloma Non Eli [NCT02513186]Phase 190 participants (Actual)Interventional2015-09-30Active, not recruiting
A Phase II, Single-Arm, Open-Label, Multicentre Study to Evaluate the Safety and Efficacy of Lenalidomide Combined With MOR00208 in Patients With Relapsed or Refractory Diffuse Large B-Cell Lymphoma (R-R DLBCL) [NCT02399085]Phase 281 participants (Actual)Interventional2016-03-29Completed
A Phase 2, Multicenter, Randomized Open-Label Study To Determine the Efficacy of Lenalidomide (Revlimid®) Versus Investigator's Choice in Patients With Relapsed or Refractory Mantle Cell Lymphoma [NCT00875667]Phase 2254 participants (Actual)Interventional2009-04-30Completed
Phase 1 Study of Elotuzumab in Combination With Autologous Stem Cell Transplantation and Lenalidomide Maintenance for Multiple Myeloma [NCT02655458]Phase 115 participants (Actual)Interventional2016-01-31Completed
Rituximab, Bendamustine and Lenalidomide in Patients With Aggressive B-cell Lymphoma Not Eligible for High Dose Chemotherapy or Anthracycline-Based Therapy. A Phase I/II Trial. [NCT00987493]Phase 1/Phase 249 participants (Actual)Interventional2009-09-30Completed
Lenalidomide Maintenance Therapy in Patients With MDS or AML With Cytogenetic Abnormalities Involving Monosomy 5 or del5q After Allogeneic Hematopoietic Stem Cell Transplantation (HSCT) [NCT00720850]Phase 210 participants (Actual)Interventional2008-04-30Terminated(stopped due to Low recruitment, scientific rationale not applicable anymore to all patients and possible induction of GvHD by the study drug)
Phase II Study of Pegylated Liposomal Doxorubicin (Doxil®), Low Frequency Dexamethasone and Revlimid® (Dd-R) in Newly Diagnosed Multiple Myeloma [NCT00617591]Phase 257 participants (Actual)Interventional2008-01-31Completed
A Multicentre, Single-arm, Open-label Safety Study of Lenalidomide Plus Dexamethasone in Previously Treated Subjects With Multiple Myeloma [NCT00420849]Phase 3587 participants (Actual)Interventional2006-11-30Completed
Safety and Preliminary Efficacy of Epcoritamab (GEN3013; DuoBody®-CD3×CD20) in Japanese Subjects With Relapsed or Refractory B-Cell Non-Hodgkin Lymphoma - A Phase 1/2, Open-Label, Dose-Escalation Trial With Expansion Cohorts [NCT04542824]Phase 1/Phase 2102 participants (Anticipated)Interventional2020-08-20Active, not recruiting
A Phase II Study of Lenalidomide (REVLIMID®) in Combination With Rituximab for Patients With CD5+/CD20+ Hematologic Malignancies Who Relapse or Progress After Rituximab [NCT00609869]Phase 229 participants (Actual)Interventional2007-10-31Completed
A Randomized, Open-label, Multi-center Phase III Trial Comparing Tisagenlecleucel to Standard of Care in Adult Participants With Relapsed or Refractory Follicular Lymphoma (FL) [NCT05888493]Phase 3108 participants (Anticipated)Interventional2023-10-02Recruiting
A Phase 1 Study of ANV419 as Monotherapy, and ANV419 in Combination With Daratumumab or With Lenalidomide Plus Low-Dose Dexamethasone, in Patients With Relapsed or Refractory Multiple Myeloma (OMNIA-2) [NCT05641324]Phase 14 participants (Actual)Interventional2023-02-10Terminated(stopped due to The study was ended due to a lack of recruitment)
Phase 1b/2, Open-Label Study to Evaluate Safety and Tolerability of Epcoritamab in Combination With Anti-Neoplastic Agents in Subjects With Non-Hodgkin Lymphoma [NCT05283720]Phase 2394 participants (Anticipated)Interventional2022-06-14Recruiting
Phase I/II Study of Lenalidomide (Revlimid), Rituximab, Cyclophosphamide, Doxorubicin, Vincristine and Prednisone (R2CHOP) Chemoimmunotherapy in Patients With Newly Diagnosed Diffuse Large Cell and Follicular Grade IIIA/B B Cell Lymphoma [NCT00670358]Phase 1/Phase 2138 participants (Actual)Interventional2008-08-25Active, not recruiting
A Phase 2 Study of Teclistamab in Combination With Daratumumab or Lenalidomide in Elderly Patients With Newly Diagnosed Multiple Myeloma [NCT05572229]Phase 274 participants (Anticipated)Interventional2023-09-30Not yet recruiting
A Randomized, Controlled, Double-Blind, Phase 3 Study to Evaluate the Efficacy and Safety of Orelabrutinib Plus Lenalidomide and Rituximab (R2) Versus Placebo Plus R2 in Relapsed/Refractory Marginal Zone Lymphoma [NCT06082102]Phase 3324 participants (Anticipated)Interventional2023-10-25Not yet recruiting
Phase I Clinical Trial of Bcl2 Inhibitor Venetoclax in Combination With Lenalidomide and Dexamethasone (Ven-Rd), Daratumumab and Dexamethasone (Ven-Dd), or Daratumumab-Lenalidomide-Dexamethasone (Ven-DRd) in t(11;14) Multiple Myeloma [NCT06042725]Phase 1100 participants (Anticipated)Interventional2023-12-01Not yet recruiting
Minimal Residual Disease Guided Maintenance Therapy With Belantamab Mafodotin and Lenalidomide After Autologous Hematopoietic Cell Transplantation in Patients With Newly Diagnosed Multiple Myeloma [NCT05091372]Phase 294 participants (Anticipated)Interventional2022-12-01Recruiting
[NCT00902915]Phase 250 participants (Anticipated)Interventional2009-05-31Active, not recruiting
Randomized Phase III Trial of Consolidation Therapy With Bortezomib (Velcade®)-Lenalidomide (Revlimid®) -Dexamethasone (VRD) Versus Bortezomib (Velcade®)-Dexamethasone (VD) for Patients With Multiple Myeloma Who Have Completed a Dexamethasone Based Induct [NCT00522392]Phase 348 participants (Actual)Interventional2007-09-30Terminated(stopped due to Slow accrual)
An Open, Randomized Clinical Phase I/II Trial to Investigate Maximum Tolerated Dose, Efficacy, and Safety of Lenalidomide/Low-dose Dexamethasone in Combination With Continuous Oral Cyclophosphamide Compared to Lenalidomide/Low-dose Dexamethasone Combined [NCT01019174]Phase 1/Phase 240 participants (Actual)Interventional2009-11-30Completed
Maintenance Therapy With Lenalidomide, Dexamethasone and Clarithromycin (Biaxin) Following Autologous/Syngeneic Transplant for Multiple Myeloma [NCT00445692]Phase 232 participants (Actual)Interventional2007-01-10Completed
A Phase II Study of Thalidomide (THALOMID®), Clarithromycin (BIAXIN®), Lenalidomide(REVLIMID®), and Dexamethasone (DECADRON®) for Subjects With Newly Diagnosed Multiple Myeloma [NCT00538733]Phase 226 participants (Actual)Interventional2007-10-31Completed
Phase II Study of Bortezomib, Melphalan, Prednisone (VMP) Followed by Lenalidomide Maintenance vs. VMP Without Maintenance in Myeloma Patients Not Eligible to High-dose Chemotherapy and Autologous Stem Cell Transplantation [NCT02145598]Phase 2/Phase 385 participants (Actual)Interventional2013-08-31Terminated(stopped due to Insufficient recruitment)
A Single-arm, Multi-center, Phase II Clinical Trial of R-CHOP Combined With Lenalidomide in the First-line Treatment for Patients With Medium to High Risk/High Risk Diffuse Large B Cell Lymphoma [NCT04214626]Phase 260 participants (Actual)Interventional2020-01-02Active, not recruiting
Phase 2 Study With Minimal Residual Disease (MRD) Driven Adaptive Strategy in Treatment for Newly Diagnosed Multiple Myeloma (MM) With Upfront Daratumumab-based Therapy [NCT04140162]Phase 257 participants (Actual)Interventional2020-10-05Active, not recruiting
A Phase I Dose Escalation Study of the Combination of Lenalidomide (Revlimid®), Dexamethasone and Cyclophosphamide in Patients Refractory or Relapsing From Stable Disease With Multiple Myeloma [NCT00915408]Phase 1/Phase 232 participants (Actual)Interventional2006-09-30Completed
Phase 2 Study Assessing Feasibility and Tolerance of the Combination of Elotuzumab, Lenalidomide and Dexamethasone in Induction, Consolidation and Maintenance Treatment of Transplant-Eligible Patients Newly Diagnosed With Multiple Myeloma [NCT02843074]Phase 253 participants (Actual)Interventional2016-09-21Completed
Efficacy of Alternating Immunochemotherapy Consisting of R-CHOP + R-HAD vs R-CHOP Alone, Followed by Maintenance Therapy Consisting of Additional Lenalidomide + Rituximab vs Rituximab Alone for Older Patients With Mantle Cell Lymphoma [NCT01865110]Phase 3623 participants (Actual)Interventional2013-11-30Active, not recruiting
A Phase Ib/II Study of Escalating Doses of Revlimid in Association With R-CHOP (R2-CHOP) in the Treatment of B-cell Lymphoma [NCT01393756]Phase 280 participants (Actual)Interventional2010-12-31Completed
A Phase I/II Study of the Tolerability of Lenalidomide and Low Dose Dexamethasone in Previously Treated Multiple Myeloma Patients With Impaired Renal Function [NCT00790842]Phase 1/Phase 263 participants (Actual)Interventional2009-01-21Terminated(stopped due to Slow enrollment)
Open-Label, Single-Arm Study of the Safety and Efficacy of CC-5013 Monotherapy for Subjects With Multiple Myeloma: A Companion Study for Studies THAL-MM-003, CC-5013-MM-009, and CC-5013-MM-010 [NCT00622336]Phase 3330 participants (Actual)Interventional2003-04-01Completed
Phase II Trial of High Dose Lenalidomide in Patients With MDS and AML With Trilineage Dysplasia (AML-TLD) [NCT00867308]Phase 232 participants (Actual)Interventional2009-07-31Terminated(stopped due to Lack of efficacy)
Phase II Trial of CC-5013 (Lenalidomide, Revlimid®) in Patients With Cutaneous T-Cell Lymphoma [NCT00466921]Phase 233 participants (Actual)Interventional2005-04-19Completed
A Single-Arm, Open-Label Study of the Efficacy and Safety of Lenalidomide in Combination With Cyclosporine A in Patients With Red Blood Cell Transfusion-Dependent Myelodysplastic Syndromes [NCT00840827]Phase 26 participants (Actual)Interventional2008-12-31Terminated(stopped due to Lack of efficacy.)
A Multi-center, Single-arm, Open Label Study to Assess the Efficacy and Safety of Anti-PD-1 Antibody (Penpulimab) Plus Lenalidomide, Rituximab, Gemcitabine and Oxaliplatin (R2-GemOx) in Patients With Relapsed/ Refractory Diffuse Large B Cell Lymphoma (DLB [NCT05186558]Phase 254 participants (Anticipated)Interventional2022-05-15Not yet recruiting
A Phase 2 Study of Lenalidomide in Previously Untreated, Symptomatic Chronic Lymphocytic Leukemia (CLL) [NCT00751296]Phase 227 participants (Actual)Interventional2006-08-31Terminated(stopped due to Seven years of follow-up & final analysis done in Dec 2012.)
A Phase 2, Multicenter, Single-Arm, Open-Label Study To Determine The Efficacy And Safety Of Single-Agent Lenalidomide (Revlimid®) In Patients With Mantel Cell NHL Who Have Relapsed Or Progressed After Treatment With Bortezomib Or Are Refractory To Bortez [NCT00737529]Phase 2134 participants (Actual)Interventional2008-12-22Completed
Phase I Trial of Irinotecan Plus Lenalidomide in Adult Patients With Recurrent Glioblastoma Multiforme [NCT00671801]Phase 124 participants (Actual)Interventional2008-04-29Terminated(stopped due to Toxicity)
A Phase 1b/2, Multicenter, Open-label, Dose-escalation Study of Elotuzumab (Humanized Anti-CS1 Monoclonal IgG1 Antibody) in Combination With Lenalidomide and Dexamethasone in Subjects With Relapsed Multiple Myeloma [NCT00742560]Phase 2101 participants (Actual)Interventional2008-08-31Completed
A First-in-human, Dose-escalation Followed by Expansion Study to Assess the Safety and Preliminary Efficacy of a Bispecific Antibody OT-A201 as Monotherapy and in Combination Therapy in Patients With Selected Hematological Malignancies and Solid Tumors [NCT05828459]Phase 1150 participants (Anticipated)Interventional2023-07-10Recruiting
Desensitization of Immunomodulating Agent-Related Hypersensitivity Reactions as a Means to Provide Therapeutic Options in the Management of Plasma Cell Disorders (DeHyperPCD) [NCT03959358]Phase 210 participants (Actual)Interventional2020-07-03Completed
A Phase II Clinical Trial for Untreated Patients With Multiple Myeloma Eligible for Stem Cell Transplant: Lenalidomide (Revlimid®) Plus Low-dose Dexamethasone (Ld x 4 Cycles) Then Stem Cell Collection Followed by Randomization to Continued Ld or Stem Cell [NCT00807599]Phase 267 participants (Actual)Interventional2008-12-10Completed
A Phase II Study of Lenalidomide Revlimid(Registered Trademark) in Previously Treated Patients With Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma [NCT00439231]Phase 233 participants (Actual)Interventional2007-02-28Completed
Lenalidomide for Advanced Hepatocellular Cancer:A Phase II Trial [NCT00717756]Phase 241 participants (Actual)Interventional2009-01-31Completed
Phase 2 Study of the Initial and Post-Transplant Treatment With Carfilzomib, Lenalidomide and Low Dose Dexamethasone (CRd) in Transplant Candidates With Newly Diagnosed, Multiple Myeloma Requiring Systemic Chemotherapy [NCT01816971]Phase 276 participants (Actual)Interventional2013-01-31Active, not recruiting
A Phase 2, Open-Label Study To Evaluate The Efficacy And Safety Of Lenalidomide In Combination With Cetuximab In Pretreated Subjects With K-Ras Mutant Metastatic Colorectal Cancer [NCT01032291]Phase 251 participants (Actual)Interventional2009-12-31Terminated(stopped due to A business decision not to continue with Phase 2b based on non-safety observations during proof of concept phase.)
A Phase II Study of Lenalidomide (Revlimid®) as Second Line Therapy in Patients With Chronic Graft-Versus-Host Disease (GVHD) [NCT00675441]Phase 25 participants (Actual)Interventional2008-04-30Terminated(stopped due to Terminated due to slow accrual.)
A Phase I Study of a Combination of 5-azacitidine Followed by Lenalidomide in High-risk MDS or Relapsed/Refractory AML Patients With Cytogenetic Abnormalities Including -5 or Del(5q) [NCT00923234]Phase 10 participants Interventional2009-06-30Terminated(stopped due to The primary objective has already been answered with the number of recruited patients.)
A Safety Confirmation Study On Lenalidomide With Dexamethasone In Japanese Patients With Previously Treated Multiple Myeloma [NCT00928486]Phase 325 participants (Actual)Interventional2009-04-28Completed
A Phase II Study of Rituximab, Lenalidomide, and Ibrutinib Combined With Chemotherapy for Patients With High Risk Diffuse Large B-Cell Lymphoma [NCT02636322]Phase 260 participants (Actual)Interventional2016-03-29Completed
A Phase II Study of Lenalidomide for Adult Histiocyte Disorders [NCT02523040]Phase 212 participants (Anticipated)Interventional2015-08-31Active, not recruiting
Phase I/II Trial of Ibrutinib, Dexamethasone, and Lenalidomide as Initial Therapy for Transplant Ineligible Multiple Myeloma Patients [NCT03015792]Phase 1/Phase 218 participants (Actual)Interventional2017-03-10Terminated(stopped due to Per CS0139535 -submitter stated we can update status to Admin complete- low accrual reasoning)
Early Patient Access Single Named Patient Program for the Use of Ulocuplumab for the Treatment of Multiple Myeloma [NCT02666209]0 participants Expanded AccessNo longer available
Phase I/II Study of Lenalidomide (Revlimid), Thalidomide, and Dexamethasone in Patients With Relapsed/Refractory Multiple Myeloma [NCT00966693]Phase 1/Phase 277 participants (Actual)Interventional2009-08-25Completed
Phase I/II Study of Oral Lenalidomide and High Dose Melphalan Supported by Autologous Peripheral Blood Stem Cell Infusion for Patients With Multiple Myeloma [NCT01142232]Phase 1/Phase 260 participants (Actual)Interventional2010-08-27Completed
A Phase II Randomized Study of Lenalidomide or Lenalidomide and Rituximab as Maintenance Therapy Following Standard Chemotherapy for Patients With High/High-intermediate Risk Diffuse Large B-Cell Lymphoma [NCT00765245]Phase 244 participants (Actual)Interventional2008-10-31Completed
A Combination of Lenalidomide and Rituximab as Front Line Therapy for the Treatment of Elderly Frail Patients Evaluated in CGA With Diffuse Large B-cells Non-Hodgkin Lymphoma. A Phase II Study of the Fondazione Italiana Linfomi (FIL) [NCT02955823]Phase 268 participants (Actual)Interventional2016-09-30Completed
A Pharmacokinetic And Pharmacodynamic Study Of Oral Lenalidomide (Revlimid) In Subjects With Low- Or Intermediate-1-Risk Myelodysplastic Syndromes [NCT00910858]Phase 1/Phase 240 participants (Actual)Interventional2005-01-31Completed
Phase 2 Trial of Intracycle Sequential Ofatumumab and Lenalidomide for the Treatment of Chronic Lymphocytic Leukemia in Patients Previously Exposed to Rituximab [NCT01123356]Phase 221 participants (Actual)Interventional2010-05-31Completed
A Phase 1b Study of Brentuximab Vedotin and Lenalidomide in Patients With Relapsed/ Refractory Cutaneous T-cell Lymphoma, CD30-positive Peripheral T-cell Lymphoma, or CD30-positive Hodgkin Lymphoma [NCT03302728]Phase 16 participants (Actual)Interventional2018-08-30Completed
A Multicenter, Phase Ib/II Study That Combines Luspatercept and Lenalidomide (L2) in Lower-risk, Non-del(5q) MDS Patients [NCT04539236]Phase 1/Phase 250 participants (Anticipated)Interventional2021-11-09Recruiting
Carfilzomib - Lenalidmide - Dexamethasone (KRd) Versus Lenalidomi - Dexamethasone (Rd) in Newly Diagnosed Myeloma Patients Not Eligible for Autologous Stem Cell Transplantation: a Randomized Phas III Trial [NCT04096066]Phase 384 participants (Actual)Interventional2019-07-01Active, not recruiting
A Phase 1/2, Open-label Study of Valemetostat in Combination With Rituximab and Lenalidomide in Relapsed or Refractory Follicular Lymphoma [NCT05683171]Phase 1/Phase 260 participants (Anticipated)Interventional2023-05-19Recruiting
A Phase 1b/2, Open-Label, Multicenter Study to Evaluate the Safety and Pharmacokinetics of a Modified Tafasitamab IV Dosing Regimen Combined With Lenalidomide in Patients With Relapsed or Refractory Diffuse Large B-Cell Lymphoma [NCT05222555]Phase 1/Phase 251 participants (Anticipated)Interventional2022-07-19Recruiting
Smart Stop: A Phase II Trial of Rituximab, Lenalidomide, Acalabrutinib, Tafasitamab Prior to and With Standard Chemotherapy for Patients With Newly Diagnosed DLBCL [NCT04978584]Phase 260 participants (Anticipated)Interventional2022-03-03Recruiting
Monoclonal Antibody-Based Sequential Therapy for Deep Remission in Multiple Myeloma - MASTER Trial [NCT03224507]Phase 2123 participants (Actual)Interventional2018-03-14Completed
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
Phase 1 Trial of Flavopiridol in Combination With Lenalidomide in Patients With Relapsed or Refractory B-Cell CLL/SLL [NCT00735930]Phase 139 participants (Actual)Interventional2008-08-31Completed
The Efficacy and Safety of Lenalidomide (Revlimid®) Monotherapy in Red Blood Cell Transfusion Dependent Subjects With Myelodysplastic Syndrome Associated With Del (5q) Cytogenetic Abnormality [NCT00874978]Phase 236 participants (Anticipated)Interventional2005-01-31Completed
A Phase II Study of the Efficacy and Safety of Lenalidomide Combined to Escalating Doses of Chemotherapy in Intermediate-2-or High Risk MDS and AML With Del 5q [NCT00885508]Phase 285 participants (Actual)Interventional2009-02-28Active, not recruiting
A Phase I/II Optimal Dose Study of Lenalidomide in the Non-5q- LOW and INT-1 Risk MDS Patients [NCT00699842]Phase 1/Phase 28 participants (Actual)Interventional2008-07-31Terminated(stopped due to Administratively terminated per FDA recommendation)
Phase II Trial of Bevacizumab Combined With Lenalidomide and Dexamethasone (BEV/REV/DEX) in Relapsed or Refractory Multiple Myeloma [NCT00410605]Phase 239 participants (Actual)Interventional2006-11-30Completed
The Efficacy and Safety of Zanubrutinib, Lenalidomide and Rituximab (ZR2) Regimen in Elderly Treatment-naive Patients With Diffuse Large B-cell Lymphoma (DLBCL) [NCT04460248]Phase 240 participants (Anticipated)Interventional2020-07-22Active, not recruiting
An Open-label, Phase II Study of Cyclophosphamide, Lenalidomide and Dexamethasone (CLD) for Previously Treated Patients With AL Amyloidosis [NCT00607581]Phase 221 participants (Actual)Interventional2008-02-29Completed
A Phase I/II Study of Combination Dasatinib and Lenalidomide in Purine Analogue-Failed Chronic Lymphocytic Leukemia [NCT00829647]Phase 1/Phase 20 participants (Actual)Interventional2009-01-31Withdrawn(stopped due to Unable to enroll)
A Phase II Study of Revlimid in Combination With Rituximab as Initial Treatment for Patients With Indolent Non-Hodgkin's Lymphoma (NHL) [NCT00695786]Phase 2156 participants (Actual)Interventional2008-06-10Completed
A Phase 3 Randomized, Open-Label Multicenter Study of Zanubrutinib (BGB-3111) Plus Anti-CD20 Antibodies Versus Lenalidomide Plus Rituximab in Patients With Relapsed/Refractory Follicular or Marginal Zone Lymphoma [NCT05100862]Phase 3750 participants (Anticipated)Interventional2022-03-10Recruiting
Single-arm, Multi-center Clinical Study of PD-1 Antibody, Chidamide, Lenalidomide and Etoposide for Relapsed or Refractory Natural Killer/T Cell Lymphoma [NCT04038411]Phase 450 participants (Anticipated)Interventional2019-04-01Recruiting
A Prospective Single Center Trial of Treatment With Lenalidomide-Melphalan-Dexamethasone in Patients With AL Amyloidosis [NCT00883623]Phase 250 participants (Actual)Interventional2009-04-30Completed
Phase 1b/2, Open-Label Trial to Evaluate Safety and Preliminary Efficacy of Epcoritamab As Monotherapy or Combined With Standard-of-Care Therapies in Chinese Subjects With B-Cell Non-Hodgkin Lymphoma [NCT05201248]Phase 149 participants (Actual)Interventional2022-03-10Active, not recruiting
A Randomized, Open-label, Phase 3 Study Comparing Once-weekly vs Twice-weekly Carfilzomib in Combination With Lenalidomide and Dexamethasone in Subjects With Relapsed or Refractory Multiple Myeloma (A.R.R.O.W.2) [NCT03859427]Phase 3454 participants (Actual)Interventional2019-05-08Completed
Phase I Trial of Maintenance Lenalidomide in Patients With Acute Myeloid Leukemia or High Risk Myelodysplastic Syndrome Post Allogeneic Bone Marrow Transplantation [NCT01433965]Phase 116 participants (Actual)Interventional2012-08-08Completed
Phase II Study To Evaluate The Safety And Efficacy Of Lenalidomide For The Treatment Of Refractory Cutaneous Lupus [NCT01408199]Phase 415 participants (Actual)Interventional2010-01-31Completed
Phase 1/2 Study of VELCADE® (Bortezomib), Dexamethasone, and Revlimid® (Lenalidomide) Versus VELCADE, Dexamethasone, Cyclophosphamide, and Revlimid Versus VELCADE, Dexamethasone and Cyclophosphamide in Subjects With Previously Untreated Multiple Myeloma [NCT00507442]Phase 1/Phase 2158 participants (Actual)Interventional2007-08-31Completed
An Open-Label, Non-Comparative, Two-Cohort, Multicenter Study to Evaluate the Effectiveness and Safety of Ixazomib (NINLARO®) in Combination With Pomalidomide and Dexamethasone (IPd, Cohort A) or With Lenalidomide and Dexamethasone (IRd, Cohort B) in Pati [NCT05183139]Phase 40 participants (Actual)Interventional2022-06-30Withdrawn(stopped due to Business decision (no enrollment))
Phase I/II Double Blinded Randomized Study to Determine the Tolerability and Efficacy of 2 Different Doses of Revlimid (CC-5013, Lenalidomide) in Biochemically Relapsed Prostate Cancer Patients (M0) After Local Treatment [NCT00348595]Phase 1/Phase 277 participants (Actual)Interventional2006-07-20Completed
A Phase Ib Multiple Ascending Dose Study of BMS-833923 Alone or in Combination With Lenalidomide (Revlimid) Plus Dexamethasone or in Combination With Bortezomib (Velcade) in Subjects With Relapsed or Refractory Multiple Myeloma [NCT00884546]Phase 127 participants (Actual)Interventional2009-07-31Completed
Phase I/II Study of Lenalidomide (RevlimidTM ) and GM-CSF in Androgen Independent Prostate Cancer [NCT00939510]Phase 1/Phase 232 participants (Actual)Interventional2005-07-31Completed
Phase I Trial of AVD Plus Lenalidomide (Revlimid) in Elderly Intermediate or Advanced Stage Hodgkin Lymphoma Patients [NCT01056679]Phase 130 participants (Anticipated)Interventional2010-04-30Completed
A Phase III, Randomized, Open-label, 3-arm Study to Determine the Efficacy and Safety of Lenalidomide(REVLIMID) Plus Low-dose Dexamethasone When Given Until Progressive Disease or for 18 Four-week Cycles Versus the Combination of Melphalan, Prednisone, an [NCT00689936]Phase 31,623 participants (Actual)Interventional2008-08-21Completed
Phase I/II Study Of The Combination Of Lenalidomide With High-Dose Melphalan For Autologous Transplant in Patients With Multiple Myeloma [NCT01079936]Phase 1/Phase 261 participants (Actual)Interventional2010-03-31Completed
Phase I/II Trial of Combination of Lenalidomide (Revlimid, LEN) and Autologous Mature Dendritic Cells Pulsed With α-galactosyl Ceramide (α-GalCer; KRN7000) in Myeloma [NCT00698776]Phase 16 participants (Actual)Interventional2009-04-30Completed
[NCT00779922]Phase 239 participants (Actual)Interventional2008-11-30Completed
"Revlimid®, and Metronomic Melphalan in the Management of Higher Risk Myelodysplastic Syndromes (MDS) and CMML: A Phase 2 Study" [NCT00744536]Phase 220 participants (Actual)Interventional2008-01-31Completed
Dose-finding Study of Lenalidomide as Maintenance Therapy in Multiple Myeloma After Allogeneic Stem Cell Transplantation [NCT00778752]Phase 1/Phase 224 participants (Actual)Interventional2009-04-30Completed
A Phase II Trial of Lenalidomide as a Treatment for Neuropathy Associated With Nonmalignant Monoclonal Gammopathy of Undetermined Significance (MGUS) [NCT00665652]Phase 28 participants (Actual)Interventional2008-04-30Terminated(stopped due to The study has been suspended for slower than anticipated enrollment to date.)
A Phase 2 Study of Lenalidomide, Rituximab, Cyclophosphamide and Dexamethasone (LR-CD) for Untreated Low Grade Non-Hodgkin Lymphoma Requiring Therapy [NCT00784927]Phase 236 participants (Actual)Interventional2008-11-30Completed
Efficacy and Safety of Velcade Plus Dexamethasone (VD), VD+Cyclophosphamide or VD Plus Lenalidomide in MMY Patients Who Are Refractory or Have Relapsed After Their Primary Therapy for MMY and Have Achieved Stable Disease After 4 Cycles of VD [NCT00908232]Phase 2163 participants (Actual)Interventional2008-05-31Completed
A Single-center Clinical Trial to Evaluate the Efficacy and Safety of Colchicine Combined With Conventional Therapy in Multiple Myeloma Patients [NCT05802992]Phase 330 participants (Anticipated)Interventional2022-03-30Recruiting
Phase I/II Trial of Lenalidomide in Combination With Liposomal Doxorubicin for the Treatment of Recurrent Epithelial Ovarian, Fallopian Tube and Primary Peritoneal Cancer [NCT00903630]Phase 1/Phase 215 participants (Actual)Interventional2009-04-30Terminated(stopped due to lack of funding)
A Multicenter, Single-arm Study to Assess the Safety, Pharmacokinetics and Efficacy of Lenalidomide in Japanese Subjects With Low- or Intern=Mediate-1-risk Myelodysplastic Syndromes (MDS) Associated With a Deletion 5 (q31-33) Abnormality and Symptomatic A [NCT00812968]Phase 211 participants (Actual)Interventional2007-09-01Completed
A Phase II, Multicenter, Single-Arm, Open-Label Study to Evaluate the Safety and Efficacy of Single-Agent Lenalidomide (Revlimid®) in Subjects With Relapsed or Refractory T-Cell Non-Hodgkin's Lymphoma [NCT00655668]Phase 254 participants (Actual)Interventional2008-03-01Terminated(stopped due to Decision not to pursue as single agent in the study population.)
A Phase I Single Arm Dose Escalation Study of the Combination of Dasatinib (Sprycel®) With Lenalidomide (Revlimid®) and Dexamethasone in Subjects With Relapsed and/ or Refractory Multiple Myeloma [NCT00560391]Phase 135 participants (Actual)Interventional2008-05-31Completed
A Phase II Study Evaluating the Toxicity and Efficacy of Single Agent Lenalidomide (Revlimid®) in Chemotherapy-Naïve Androgen-Independent Prostate Cancer Patients [NCT00654186]Phase 232 participants (Actual)Interventional2008-02-29Completed
A Phase I Study of Vorinostat in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT00642954]Phase 131 participants (Actual)Interventional2008-02-13Completed
A Phase Ib/II Study of OBINUTUZUMAB Combined to LENALIDOMIDE for the Treatment of Follicular and Relapsed/Refractory Aggressive (DLBCL and MCL) B-cell Lymphoma [NCT01582776]Phase 1/Phase 2317 participants (Actual)Interventional2012-10-03Completed
A Genetic Risk-Stratified, Randomized Phase II Study of Four Fludarabine/Antibody Combinations for Patients With Symptomatic, Previously Untreated Chronic Lymphocytic Leukemia [NCT00602459]Phase 2418 participants (Actual)Interventional2008-01-15Completed
A Two-Arm, Multi-center Trial of Revlimid® and Rituximab, for First-Line Treatment in Patients With B-cell Chronic Lymphocytic Leukemia (CLL) [NCT00628238]Phase 280 participants (Anticipated)Interventional2008-02-29Recruiting
Phase I/II Trial of Azacitidine Plus Lenalidomide in the Treatment of Acute Myeloid Leukemia [NCT01016600]Phase 1/Phase 231 participants (Actual)Interventional2010-04-30Completed
Frontline Lenalidomide for AL Amyloidosis Involving Myocardium: Investigation of Organ Reversing Capacity of Lenalidomide [NCT04298372]Phase 330 participants (Anticipated)Interventional2019-02-20Recruiting
Lenalidomide Observational Study in Patients With Mantle Lymphoma in Relapse/Refraction. Spanish Programme RRMCL Results. [NCT04109872]25 participants (Anticipated)Observational2018-09-15Recruiting
Phase I/II Study Evaluating Rituximab, Lenalidomide, and Bortezomib in the First-Line or Second-Line Treatment of Patients With Mantle Cell Lymphoma [NCT00633594]Phase 1/Phase 239 participants (Actual)Interventional2008-06-30Completed
Post-Autologous Transplant Maintenance With Isatuximab and Lenalidomide in Minimal Residual Disease Positive Multiple Myeloma [NCT05344833]Phase 250 participants (Anticipated)Interventional2023-01-05Recruiting
A Phase 1/2 Multicenter, Open-label Study to Determine the Recommended Dose and Regimen of Durvalumab (MEDI4736) in Combination With Lenalidomide (LEN) With and Without Dexamethasone (DEX)in Subjects With Newly Diagnosed Multiple Myeloma (NDMM) [NCT02685826]Phase 1/Phase 256 participants (Actual)Interventional2016-04-25Completed
A Pilot Study of Lenalidomide Alternating With Ipilimumab Post Allogeneic and Autologous Stem Cell Transplantation [NCT01919619]Phase 241 participants (Actual)Interventional2013-11-04Active, not recruiting
Combination of Lenalidomide and Rituximab in Patients With Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL-SLL) as Initial Treatment or Subsequent Therapy [NCT01446133]Phase 2120 participants (Anticipated)Interventional2011-12-12Active, not recruiting
A Phase II Study of IRD (Ixazomib, Lenalidomide, & Dexamethasone) for Consolidation Therapy Post Autologous Stem Cell Transplantation Followed by Maintenance Ixazomib or Lenalidomide for Multiple Myeloma [NCT02253316]Phase 2236 participants (Actual)Interventional2015-01-20Active, not recruiting
[NCT00968331]Phase 225 participants (Actual)Interventional2009-03-31Terminated(stopped due to The EC withdrawn the approval becuase of possible conflicts of interests between our Institute and Supporter (Celgene))
Evaluation of Efficacy and Safety of Lenalidomide (Revlimid®) in Patients With POEMS Syndrome [NCT00971685]Phase 216 participants (Anticipated)Interventional2009-07-31Recruiting
A Phase 2 Study of Isatuximab in Combination With Bortezomib, Cyclophosphamide and Dexamethasone Followed by Isatuximab and Lenalidomide Maintenance in Newly Diagnosed Patients With Multiple Myeloma and Severe Renal Impairment (EAE116) [NCT05147493]Phase 251 participants (Anticipated)Interventional2022-04-30Not yet recruiting
Lenalidomide, Melphalan, Prednisone, and Thalidomide (RMPT) for Relapsed/Refractory Multiple Myeloma [NCT00961467]Phase 244 participants (Actual)Interventional2007-02-28Completed
Phase I Clinical Trial of Bendamustine, Lenalidomide and Rituximab in B-Cell Lymphoid Malignancies [NCT00864942]Phase 128 participants (Actual)Interventional2009-02-28Completed
Phase II Study of the Efficacy and Safety of Lenalidomide in Adult Subjects With Intermediate-2-or Higt Risk Myelodysplastic Syndrome(MDS) Associated With a Deletion (DEL) 5q [31] [NCT00424229]Phase 249 participants Interventional2006-10-31Recruiting
An Open, Single-arm, Multi-center Clinical Trial of Molecular Subtype-guided R-MINE+X Regimen in the Treatment of Relapsed/Refractory Diffuse Large B-cell Lymphoma (DLBCL) [NCT05784987]60 participants (Anticipated)Interventional2023-04-15Not yet recruiting
BrUOG 401: A Phase 2 Study of Mosunetuzumab With Lenalidomide Augmentation as First-line Therapy for Follicular and Marginal Zone Lymphoma [NCT04792502]Phase 252 participants (Anticipated)Interventional2022-07-14Recruiting
Maintenance Lenalidomide Therapy After Autologous Stem Cell Transplant in Patients With High Risk Relapsed/Refractory Lymphomas [NCT01575860]Phase 1/Phase 28 participants (Actual)Interventional2012-04-30Completed
A Phase Ib, Multi-center, Open-label, Dose-escalation Study of Oral LBH589 When Administered in Combination With Oral Lenalidomide & Dexamethasone in Adult Patients With Multiple Myeloma [NCT00532675]Phase 146 participants (Actual)Interventional2008-04-22Completed
Phase II Study of Lenalidomide and Radiation Therapy in Patients With Newly Diagnosed Glioblastoma Multiforme. [NCT00165477]Phase 223 participants (Actual)Interventional2005-09-30Completed
A Randomised Comparison of Daily 25 mg Versus 5 mg Lenalidomide as Maintenance Therapy After High-dose Therapy and Autologous Stem Cell Transplantation in Patients With Multiple Myeloma [NCT00891384]Phase 3194 participants (Actual)Interventional2009-04-01Completed
Pharmacogenomic Study to Predict Survival, Best Response and Toxicity in Newly Diagnosed Myeloma Patients Above the Age of 65 Treated With Either a Combination of Melphalan-prednisone-thalidomide or Lenalidomide-dexamethasone [NCT00907452]143 participants (Actual)Interventional2009-07-29Completed
S0833, Modified Total Therapy 3 (TT3) for Newly Diagnosed Patients With Multiple Myeloma (MM): A Phase II SWOG Trial for Patients Aged ≤ 65 Years [NCT01055301]Phase 20 participants (Actual)Interventional2011-07-31Withdrawn(stopped due to lack of accrual)
A Phase II Study of Lenalidomide (REVLIMID, NSC-703813) for Previously Untreated Non-M3, Deletion 5q Acute Myeloid Leukemia (AML) in Patients Age 60 or Older Who Decline Remission Induction Chemotherapy [NCT00352365]Phase 241 participants (Actual)Interventional2006-06-30Completed
A Phase III, Multicentre, Randomized, Double-Blind, Placebo-Controlled, 3-Arm Parallel Group Study To Determine The Efficacy And Safety Of Lenalidomde (Revlimid®) In Combination With Melphalan And Prednisone Versus Placebo Plus Melphalan And Prednisone In [NCT00405756]Phase 3459 participants (Actual)Interventional2007-01-31Completed
A Phase II, Multicenter, Single-Arm, Open-Label Study To Evaluate The Safety And Efficacy Of Single-Agent Lenalidomide (Revlimid®, CC-5013) in Subjects With Relapsed Or Refractory Aggressive Non-Hodgkin's Lymphoma [NCT00413036]Phase 2217 participants (Actual)Interventional2006-06-30Completed
Phase I Study of Bendamustine in Combination With Lenalidomide (CC-5013) and Dexamethasone in Patients With Refractory or Relapsed Multiple Myeloma [NCT01042704]Phase 129 participants (Actual)Interventional2008-02-29Completed
Phase I/II Trial of Maintenance Therapy With Lenalidomide and Rituximab Following High-Dose Chemotherapy and Autologous Stem Cell Transplantation for B-cell Non-Hodgkin Lymphoma [NCT01045928]Phase 15 participants (Actual)Interventional2010-01-31Terminated(stopped due to Extreme toxicity in Phase I, study did not proceed to Phase II)
A Randomized Phase 2 Study of Daratumumab-Selinexor-Velcade-Dexamethasone (Dara-SVD) for High-Risk Newly Diagnosed Multiple Myeloma [NCT06169215]Phase 282 participants (Anticipated)Interventional2024-02-20Not yet recruiting
A RANDOMIZED, 2-ARM, PHASE 3 STUDY OF ELRANATAMAB (PF-06863135) VERSUS LENALIDOMIDE IN PATIENTS WITH NEWLY DIAGNOSED MULTIPLE MYELOMA AFTER UNDERGOING AUTOLOGOUS STEM-CELL TRANSPLANTATION [NCT05317416]Phase 3760 participants (Anticipated)Interventional2022-03-25Recruiting
A Phase I/II Trial of CHOEP Chemotherapy Plus Lenalidomide as Front Line Therapy for Patients With Stage II, III and IV Peripheral T-Cell Non-Hodgkin's Lymphoma [NCT02561273]Phase 1/Phase 254 participants (Actual)Interventional2015-09-28Completed
Third-line Therapy of Multiple Myeloma With Lenalidomide in Combination With Pioglitazone, Dexamethasone and Metronomic Low-dose Chemotherapy With Treosulfan [NCT01010243]Phase 1/Phase 254 participants (Anticipated)Interventional2009-10-31Recruiting
A PHASE 2, SINGLE ARM STUDY TO DETERMINE THE SAFETY AND EFFICACY OF AZACITIDINE, AND LENALIDOMIDE IN HIGHER RISK MYELODYSPLASTIC SYNDROME [NCT01053806]Phase 26 participants (Anticipated)Interventional2011-08-31Recruiting
Phase II Trial of Lenalidomide Maintenance After High-dose Methotrexate-based Immunochemotherapy in Patients With Primary Central Nervous System Lymphoma [NCT05260619]Phase 228 participants (Anticipated)Interventional2022-04-01Recruiting
Phase 1 Dose-Ranging Study of Ezatiostat Hydrochloride (Telintra®, TLK199 Tablets)in Combination With Lenalidomide (Revlimid®)in Patients With Non-Deletion(5q) Low to Intermediate-1 Risk Myelodysplastic Syndrome (MDS) [NCT01062152]Phase 119 participants (Actual)Interventional2009-11-30Completed
Phase II Trial to Assess the Activity of Ketoconazole Plus Lenalidomide in Patients With Prostate Cancer Progressive After Androgen Deprivation [NCT00460031]Phase 234 participants (Actual)Interventional2006-09-01Completed
A Phase I Study of Lenalidomide Therapy Prior to Re-induction Chemotherapy With Mitoxantrone, Etoposide, and Cytarabine (MEC) for the Treatment of Relapsed or Refractory Acute Myeloid Leukemia (AML) [NCT01904643]Phase 117 participants (Actual)Interventional2014-02-28Terminated(stopped due to Accrual factor)
A Randomized, National, Open-label, Multicenter, Phase III Trial Studying Induction Therapy With Bortezomib/Lenalidomide/Dexamethasone (VRD-GEM), Followed by High-dose Chemotherapy With Melphalan-200 (MEL-200) Versus Busulfan-melphalan (BUMEL), and Consol [NCT01916252]Phase 3460 participants (Anticipated)Interventional2013-09-30Completed
A Phase II Single-arm, Open-label Study to Evaluate the Safety and Efficacy of Combination Lenalidomide (Revlimid®) With Rituximab in Subjets With Relapsed or Refractory Diffuse Large b Cell Non-Hodgkin's Lymphoma. [NCT01939327]Phase 234 participants (Actual)Interventional2013-09-30Completed
Rituximab, Methotrexate, Procarbazine, Vincristine, Lenalidomide (RL-MPV) Followed by BBC (BCNU, Busulfan, Cyclophosphamide) High-dose Chemotherapy With Auto-HCT and Maintenance Therapy With Nivolumab in Newly Diagnosed Primary CNS Lymphoma [NCT05425654]Phase 230 participants (Anticipated)Interventional2021-05-17Recruiting
A Non-interventional, Observational Post-marketing Registry of Multiple Myeloma Adult Patients Treated With Revlimid (Lenalidomide) in China [NCT01947309]176 participants (Actual)Observational2013-11-30Terminated(stopped due to Business Decision)
A Study of Low-dose Lenalidomide After Non-myeloablative Allogeneic Stem Cell Transplant With Bortezomib as GVHD Prophylaxis in High Risk Multiple Myeloma [NCT01954784]Phase 18 participants (Actual)Interventional2013-10-07Terminated(stopped due to Funding unavailable)
Phase II Study Evaluating the Efficacy of Lenalidomide in Association With Rituximab in Refractory or Relapse of Primary Central Nervous System Lymphoma (PCNSL) [NCT01956695]Phase 245 participants (Actual)Interventional2013-09-18Completed
MCLENA-2: A Phase II Clinical Trial for the Assessment of Biomarker Trajectory in Patients With Mild Cognitive Impairment Due to Alzheimer's Disease Treated With Lenalidomide (Amendment to IND # 142121) [NCT06177028]Phase 245 participants (Anticipated)Interventional2024-01-02Not yet recruiting
A Prospective Phase II Study of Polatuzumab, Rituximab, and Lenalidomide(Pola-R2) in Newly-diagnosed Non-fit Elderly DLBCL Patients [NCT06176729]Phase 230 participants (Anticipated)Interventional2023-10-24Recruiting
A Phase 3, Two-stage, Randomized, Multi-center, Controlled, Open-label Study Comparing Iberdomide Maintenance to Lenalidomide Maintenance Therapy After Autologous Stem Cell Transplantation (ASCT) in Participants With Newly Diagnosed Multiple Myeloma (NDMM [NCT05827016]Phase 31,216 participants (Anticipated)Interventional2023-06-22Recruiting
A Phase I Open Label Study Of Panobinostat In Combination With Lenalidomide, Bortezomib, And Dexamethasone In Patients With Relapsed And Relapsed/Refractory Multiple Myeloma [NCT01965353]Phase 121 participants (Actual)Interventional2013-10-31Completed
Phase II Study of Daratumumab Based Response Adapted Therapy for Older Adults With Newly Diagnosed Multiple Myeloma [NCT04151667]Phase 233 participants (Actual)Interventional2019-11-22Active, not recruiting
A Personalized Vaccine for the Immune Prevention of Multiple Myeloma [NCT03631043]Early Phase 130 participants (Actual)Interventional2018-12-21Active, not recruiting
A Phase I Trial of Lenalidomide, Umbralisib and Ublituximab in Patients With Relapsed or Refractory Indolent Non-Hodgkin Lymphoma or Mantle Cell Lymphoma [NCT04635683]Phase 10 participants (Actual)Interventional2022-09-30Withdrawn(stopped due to Investigational Drug removed from the market)
Phase I Trial of Daily Lenalidomide (CC-5013, Revlimid™) and Docetaxel Given Every Three Weeks in Patients With Advanced Solid Tumors [NCT00253344]Phase 133 participants (Actual)Interventional2005-06-30Completed
Phase II Trial of Combination of Ixazomib and Lenalidomide and Dexamethasone in Smoldering Multiple Myeloma [NCT02916771]Phase 255 participants (Actual)Interventional2016-10-31Active, not recruiting
A Pilot Study of Lenalidomide Maintenance Therapy in Stage IIIB/IV Non-small Cell Lung Cancer After First-line Chemotherapy [NCT02018523]Phase 17 participants (Actual)Interventional2014-06-30Terminated(stopped due to Study did not enroll enough subjects to make a statistically sound conclusion.)
Phase 2, Open-Label Randomized Study of Daratumumab, Carfilzomib, Lenalidomide, and Dexamethasone vs Carfilzomib, Lenalidomide, and Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma [NCT04268498]Phase 2306 participants (Anticipated)Interventional2020-02-11Recruiting
Phase I/II Trial of AT-101 in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed Symptomatic Multiple Myeloma [NCT02697344]Phase 110 participants (Actual)Interventional2016-04-14Completed
Randomized Phase II Trial in Early Relapsing or Refractory Follicular Lymphoma [NCT03269669]Phase 295 participants (Anticipated)Interventional2018-01-23Recruiting
A Phase II Trial of Combination of Oral Lenalidomide and Low-dose Cyclophosphamide for Patients With Antibiotics-unresponsive Extranodal Marginal Zone B-cell Lymphoma [NCT04604028]Phase 221 participants (Anticipated)Interventional2020-11-10Not yet recruiting
A Phase I/II Trial Of Very Low to Low-Doses of Continuous Azacitidine in Combination With Standard Doses of Lenalidomide and Low-Dose Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT01155583]Phase 1/Phase 245 participants (Actual)Interventional2010-06-30Completed
Lenalidomide Combined With Rituximab as Front-line Therapy in Elderly Patients Aged Over 80 Years With Diffuse Large B Cell Lymphoma [NCT04622579]Phase 220 participants (Anticipated)Interventional2020-10-23Recruiting
A Phase I Dose Escalating Study of Bortezomib and Lenalidomide in Patients With Untreated or Previously Treated, Primary and Secondary Non 5q- Del Myelodysplasia [NCT00580242]Phase 123 participants (Actual)Interventional2007-11-30Completed
Phase II Study of Cetuximab and Lenalidomide in Recurrent/Metastatic Squamous Cell Carcinoma of the Head and Neck [NCT01133665]Phase 242 participants (Actual)Interventional2010-02-28Completed
A Phase II Clinical Trial of Lenalidomide and Prednisone in Low and Intermediate-1 IPSS Risk, Non-del (5q) MDS Patients [NCT01133275]Phase 228 participants (Actual)Interventional2010-04-28Completed
A Phase II, Multicenter, Single-Arm, Open-Label Study To Evaluate The Safety And Efficacy Of Single-Agent Lenalidomide (Revlimid®, CC-5013) In Subjects With Relapsed Or Refractory Aggressive Non-Hodgkin's Lymphoma [NCT00179660]Phase 250 participants (Actual)Interventional2005-08-31Completed
A Phase II, Multicenter, Single-Arm, Open-Label Study to Evaluate the Safety and Efficacy of Lenalidomide (Revlimid ®) in Combination With Dexamethasone in Subjects With Relapsed or Refractory Diffuse Large B-Cell Lymphoma [NCT00474188]Phase 226 participants (Actual)Interventional2007-05-31Terminated(stopped due to Business decision)
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
A Phase II, Multicenter, Single-Arm, Open-Label Study to Evaluate the Safety and Efficacy of Single-Agent Lenalidomide (Revlimid®, CC-5013) in Participants With Relapsed or Refractory Indolent Non-Hodgkin's Lymphoma [NCT00179673]Phase 243 participants (Actual)Interventional2005-08-31Completed
Phase I/II Trial of Melphalan, Prednisone Plus Lenalidomide in Patients With Newly Diagnosed Multiple Myeloma Who Are Not Candidates for Stem Cell Transplant [NCT00477750]Phase 1/Phase 233 participants (Actual)Interventional2005-06-30Completed
A Phase II Study of the Efficacy and Safety of Lenalidomide, Subcutaneous Bortezomib, and Dexamethasone Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma [NCT02219178]Phase 242 participants (Actual)Interventional2014-11-30Completed
Phase II Study of Subcutaneous (SC) Bortezomib, Lenalidomide and Dexamethasone for Relapsed and/or Refractory Multiple Myeloma; Followed by SC Bortezomib Maintenance [NCT01647165]Phase 20 participants (Actual)Interventional2012-07-11Withdrawn
A Phase Ib, Multi-center, Open-label Dose Escalation and Expansion Platform Study of VAY736 as Single Agent and in Combination With Select Antineoplastic Agents in Patients With Non-Hodgkin Lymphoma (NHL) [NCT04903197]Phase 1124 participants (Anticipated)Interventional2022-01-24Recruiting
B- PRISM (Precision Intervention Smoldering Myeloma): A Phase II Trial of Combination of Daratumumab, Bortezomib, Lenalidomide and Dexamethasone in High- Risk Smoldering Multiple Myeloma [NCT04775550]Phase 260 participants (Anticipated)Interventional2021-03-08Recruiting
A Phase 3, Randomized, Controlled, Open-label, Multicenter, Safety and Efficacy Study of Dexamethasone Plus MLN9708 or Physicians Choice of Treatment Administered to Patients With Relapsed or Refractory Systemic Light Chain (AL) Amyloidosis [NCT01659658]Phase 3177 participants (Actual)Interventional2012-12-12Terminated(stopped due to Sponsor's decision)
Treatment of B-CLL With Autologous IL2 and CD40 Ligand-Expressing Tumor Cells + Lenalidomide [NCT01604031]Phase 12 participants (Actual)Interventional2013-02-28Terminated
A Phase 3, Randomized, Open Label Trial of Lenalidomide/Dexamethasone With or Without Elotuzumab in Subjects With Previously Untreated Multiple Myeloma [NCT01335399]Phase 3748 participants (Actual)Interventional2011-08-04Completed
The Safety, Tolerability and Preliminary Efficacy of PD-L1 Monoclonal Antibody Combined With Lenalidomide in Third-line Post-Treatment of Patients With Microsatellite Stable Advanced Colorectal Cancer: A Phase I Clinical Study [NCT04326296]Phase 133 participants (Anticipated)Interventional2020-05-30Not yet recruiting
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
The Combination of Lenalidomide and Dexamethasone With or Without Intensification by High-dose Melphalan in the Treatment of Multiple Myeloma [NCT01090089]Phase 3348 participants (Actual)Interventional2010-03-01Completed
A Phase 1, Multi-center, Open-label Study of the Safety and Efficacy of a Stepwise Dose-escalation Schedule of Lenalidomide Monotherapy in Subjects With Relapsed or Refractory B-cell Chronic Lymphocytic Leukemia [NCT00419250]Phase 152 participants (Actual)Interventional2006-12-01Completed
A Phase 1/2 Trial of Durvalumab (MEDI4736) When Given as a Single Agent or in Combination With Lenalidomide in Patients With Relapsed/ Refractory Peripheral T-cell Lymphoma, Including Cutaneous T-cell Lymphoma [NCT03011814]Phase 1/Phase 262 participants (Anticipated)Interventional2017-03-08Recruiting
Phase Ib/IIa Study of Combination Therapy With Carfilzomib, Romidepsin, Lenalidomide in Patients With Relapsed or Refractory B- and T-cell Lymphomas [NCT02341014]Phase 1/Phase 231 participants (Anticipated)Interventional2015-01-02Completed
Lenalidomide and Low Dose Dexamethasone Induction Therapy Followed by Low Dose Melphalan, Prednisone, Lenalidomide and Bortezomib Sequential Maintenance Therapy for Newly Diagnosed High-risk Multiple Myeloma [NCT00691704]Phase 218 participants (Actual)Interventional2008-08-31Completed
Phase II, Open-Label Study Evaluating Efficacy of Tafasitamab and Lenalinomide Associated to Rituximab in Frontline Diffuse Large B-Cell Lymphoma Patients of 80 y/o or Older [NCT04974216]Phase 271 participants (Anticipated)Interventional2021-12-20Recruiting
A Prospective Multicenter Pilot Trial to Evaluate the Efficacy of a Treatment With Fludarabine, Cyclophosphamide, Lenalidomide (FCL) for Advanced Relapsed/Refractory Chronic Lymphocytic Leukemia (CLL) Patients. [NCT00727415]Phase 1/Phase 242 participants (Actual)Interventional2008-02-29Completed
A Modular Phase I-II Trial of Lenalidomide and Paclitaxel in Men With Castration-Resistant Prostate Cancer and Lymph-Node Dominant Metastases [NCT00933426]Phase 117 participants (Actual)Interventional2009-08-31Terminated(stopped due to Terminated early due to slow accrual as Phase I dose escalation study.)
Consolidation Therapy With Lenalidomide (Revlimid®) With or Without Rituximab Followed by Maintenance Therapy With Revlimid® After Autologous/Syngeneic Transplant for Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma [NCT00833534]Phase 20 participants (Actual)Interventional2009-02-28Withdrawn(stopped due to No patient enrolled on study. Did not get patients in timely fashion. All referrals for study when assessed were not eligible.)
Lenalidomide Combined With R-CHOP in Newly Diagnosed Double-expressor Diffuse Large B-Cell Lymphoma Patients [NCT04164368]Phase 262 participants (Anticipated)Interventional2019-10-22Recruiting
Quality of Life, Symptoms and Treatment Satisfaction in Adult Patients With Relapsed and/or Refractory Multiple Myeloma, Receiving Ixazomib (Ninlaro®) in Combination With Lenalidomide and Dexamethasone in a Real World Setting: Pilot Study [NCT03903406]40 participants (Anticipated)Observational2019-05-31Not yet recruiting
Treatment With Lenalidomide, Bendamustine and Prednisone (RBP) in Patients With Relapsed or Refractory Multiple Myeloma After Autologous Stem Cell Transplantation or Conventional Chemotherapy OSHO #077 [NCT01002703]Phase 1/Phase 250 participants (Anticipated)Interventional2009-09-30Recruiting
A Phase I Trial Using Cyclophosphamide, Rituximab and Revlimid (CR2) for the Treatment of Relapsed/Refractory B-Cell Chronic Lymphocytic Leukemia (B-CLL) and SLL [NCT01005979]Phase 16 participants (Actual)Interventional2010-07-31Terminated(stopped due to Slow Accrual)
Vorinostat (SAHA) and Lenalidomide After Autologous Transplant for Patients With Multiple Myeloma [NCT00729118]Phase 119 participants (Actual)Interventional2008-09-26Completed
Phase III Randomised Trial of Immunomodulatory Therapy in High Risk Solitary Bone Plasmacytoma [NCT02544308]Phase 336 participants (Actual)Interventional2017-03-10Active, not recruiting
Tafasitamab, Lenalidomide and Venetoclax Combination Therapy for Relapsed or Refractory Mantle Cell Lymphoma (V-MIND): A Phase II Study With Safety Lead-In [NCT05910801]Phase 2100 participants (Anticipated)Interventional2023-12-29Not yet recruiting
Efficacy and Safety of Epcoritamab Monotherapy and in Combination With Lenalidomide as First-line Therapy for Anthracycline-ineligible Diffuse Large B-Cell Lymphoma Patients, an Open-label, Randomized, Multicenter, Global Phase 2 Trial [NCT05660967]Phase 2180 participants (Anticipated)Interventional2023-03-06Recruiting
A Phase 2 Study Evaluating the Efficacy of Epigenetic Modulation in Relapsed/Refractory Follicular Lymphoma and Marginal Zone Lymphoma [NCT01121757]Phase 211 participants (Actual)Interventional2010-04-30Terminated(stopped due to The study closed temporarily in February 2012 pending analysis of samples collected. In October, 2012, Celgene requested closure of the study.)
A Phase II Study of Pegylated Liposomal Doxorubicin, Bortezomib, Dexamethasone and Lenalidomide (DVD-R) for Patients With Relapsed/Refractory Multiple Myeloma [NCT01160484]Phase 240 participants (Actual)Interventional2009-09-30Completed
Study Of Pgv001 A Multi-Peptide Therapeutic Vaccine Platform For Use In The Treatment Of Malignancies In The Adjuvant Setting [NCT02721043]Phase 113 participants (Actual)Interventional2016-04-30Completed
An Open-Label, Dose-Escalation, Phase 1/2 Study of the Oral Form of Ixazomib (MLN9708), a Second-Generation Proteasome Inhibitor, Administered in Combination With Lenalidomide and Low-Dose Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma Re [NCT01217957]Phase 1/Phase 265 participants (Actual)Interventional2010-11-22Completed
Phase I/II Trial of Cediranib Alone or Cediranib and Lenalidomide in Iodine 131-Refractory Differentiated Thyroid Cancer [NCT01208051]Phase 1/Phase 2127 participants (Actual)Interventional2010-09-09Completed
Frailty Score-guided Dosing of Lenalidomide, Dexamethasone and Daratumumab Induction Therapy in Elderly, Frail Newly Diagnosed Myeloma (MMY2035) [NCT04223661]Phase 20 participants (Actual)Interventional2021-12-06Withdrawn(stopped due to lack of funding)
Lenalidomide, Bortezomib, and Dexamethasone Combination Therapy as Induction Followed by Bortezomib and Lenalidomide Maintenance in Patients With Newly Diagnosed High Risk Multiple Myeloma [NCT03641456]Phase 250 participants (Anticipated)Interventional2020-09-25Recruiting
A Phase 2/3, Multicenter, Randomized, Open-label Study to Compare the Efficacy and Safety of Lenalidomide (Revlimid ®) Versus Investigator's Choice in Patients With Relapsed or Refractory Diffuse Large B-cell Lymphoma [NCT01197560]Phase 2/Phase 3111 participants (Actual)Interventional2010-09-02Completed
Treatment of IgG4-Related Disease With Revlimid and Rituximab: The TIGR2 Trial [NCT02705638]Phase 16 participants (Actual)Interventional2016-04-30Completed
A Multicenter Phase I/II Dose Escalation Study of Lenalidomide in Combination With Melphalan and Dexamethasone in Subjects With Newly-diagnosed Light-chain (AL)-Amyloidosis [NCT00621400]Phase 1/Phase 227 participants (Actual)Interventional2008-01-31Completed
Phase I/II Investigation of Temsirolimus Plus Lenalidomide in Relapsed Non-Hodgkin Lymphomas [NCT01076543]Phase 1/Phase 2110 participants (Actual)Interventional2010-04-15Completed
Nonmyeloablative Stem Cell Transplantation With or Without Lenalidomide for Chronic Lymphocytic Leukemia (RV-CLL-PI-0294) [NCT00899431]Phase 239 participants (Actual)Interventional2009-05-06Terminated(stopped due to Terminated per PI's request at the time of continuing review)
A Phase 1-2 Study of Azacitidine in Combination With Lenalidomide for Previously Untreated Elderly Patients With Acute Myeloid Leukemia [NCT00890929]Phase 1/Phase 245 participants (Actual)Interventional2009-04-30Completed
Revlimid® Capsules Drug Use-results Surveillance Relapsed or Refractory Adult T-cell Leukemia Lymphoma [NCT03098589]80 participants (Anticipated)Observational2017-05-30Recruiting
Phase I-II Study of Carfilzomib, Lenalidomide, Dexamethasone, and Panobinostat, Ca-R-Pa-Diem, as Induction Therapy for Newly Diagnosed, Untreated, Transplant-Eligible, Multiple Myeloma Patients [NCT02802163]Phase 1/Phase 20 participants (Actual)Interventional2017-06-30Withdrawn(stopped due to study drug unavailable)
Lenalidomide in Combination With Bendamustine and Rituximab for Previously Untreated Chronic Lymphocytic Leukemia or Small Lymphocytic Lymphoma(CLL/SLL): a Phase I Study [NCT01400685]Phase 134 participants (Anticipated)Interventional2012-12-31Completed
A Phase 1/2 Study of Lenalidomide in Combination With Bendamustine (LEBEN) in Relapsed and Primary Refractory Hodgkin Lymphoma [NCT01412307]Phase 1/Phase 236 participants (Anticipated)Interventional2011-07-31Active, not recruiting
Relevance of Maintenance Therapy Using Lenalidomide (Revimid®) After Autologous Stem Cell Transplantation Patients Under the Age Of 65. (Open, Randomised, Multi-centric Trial Versus Placebo). [NCT00430365]Phase 3614 participants (Anticipated)Interventional2006-06-30Completed
Safety of a Maintenance Therapy With Lenalidomide After Reduced-intensity Allogeneic Stem Cell Transplantation for Chemosensitive Relapsed Multiple Myeloma [NCT01421927]Phase 113 participants (Actual)Interventional2011-08-31Completed
A Pilot/Feasibility Phase I Study of Bendamustine, Rituximab and Lenalidomide in Patients With Refractory/Relapsed Indolent NHL [NCT01429025]Phase 126 participants (Actual)Interventional2012-05-31Completed
A Multidose Phase I Study of Oral CC5013, a Thalidomide Derivative, in Patients With Refractory Metastatic Cancer [NCT00031941]Phase 10 participants Interventional2002-04-30Completed
Evaluation of Lenalidomide as Maintenance Therapy Post Allogeneic Hematopoietic Cell Transplantation for High-risk Multiple Myeloma [NCT00847639]Phase 230 participants (Actual)Interventional2009-02-28Completed
A Phase 1 Study of Mosunetuzumab With Polatuzumab Vedotin and Lenalidomide (M+Pola+Len) in Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL) [NCT06015880]Phase 130 participants (Anticipated)Interventional2024-09-03Recruiting
Phase III Randomized, Open-Label, Multicenter Study Evaluating Efficacy and Safety of Mosunetuzumab in Combination With Lenalidomide in Comparison to Rituximab in Combination With Lenalidomide With a Non-Randomized Single Arm US Extension of Mosunetuzumab [NCT04712097]Phase 3474 participants (Anticipated)Interventional2021-10-27Recruiting
A Phase II, Open-label Study of Lenalidomide and Dexamethasone Followed by Donor Lymphocyte Infusions in Relapsed Multiple Myeloma Following Allogeneic Stem Cell Transplant [NCT03413800]Phase 210 participants (Actual)Interventional2018-02-12Active, not recruiting
Daratumumab Versus Lenalidomide Maintenance Therapy for Multiple Myeloma: A Randomized Pilot Study Comparing Patient-Reported Health Related Quality of Life Measures With a Plant Based Nutrition Intervention Sub Study (NUTRIVENTION-4) [NCT04497961]Phase 2100 participants (Anticipated)Interventional2020-08-28Recruiting
A Phase I Study of DVd +/ CC-5013 in Relapsed Refractory Multiple Myeloma (MM) [NCT00091624]Phase 177 participants (Actual)Interventional2003-03-31Completed
UARK 2003-35, A Phase III Study of Bortezomib Versus Bortezomib in Two Doses in Combination With Revlimid™ for Patients Relapsing or Progressing on Total Therapy II (UARK 98-026) [NCT00093028]Phase 3315 participants (Actual)Interventional2004-01-31Completed
Evaluation of Ixazomib, Lenalidomide, Dexamethasone Induction and Extended Consolidation Followed by Lenalidomide Maintenance in Newly Diagnosed Multiple Myeloma Patients ≤65 Years Eligible for High Dose Therapy [NCT02897830]Phase 246 participants (Actual)Interventional2016-08-05Terminated(stopped due to negatives results)
Phase I/II Open-Label, Dose Escalation Study To Determine The Maximum Tolerated Dose And To Evaluate The Safety Profile of Lenalidomide (Revlimid®) With Topotecan In Subjects With Advanced Ovarian and Primary Peritoneal Carcinoma [NCT00179712]Phase 1/Phase 260 participants Interventional2005-04-30Completed
A Phase I/II Study Of Lenalidomide (Revlimid ) In Combination With Gemcitabine In Patients With Untreated Advanced Carcinoma Of The Pancreas [NCT00179751]Phase 1/Phase 250 participants Interventional2005-04-30Terminated
A Multicenter, Phase I Study to Determine the Maximum Tolerated Dose, Safety, Pharmacokinetics and Efficacy of Lenalidomide With and Without Dexamethasone in Japanese Subjects With Previously Treated Multiple Myeloma [NCT00555100]Phase 115 participants (Actual)Interventional2007-07-01Completed
A Phase 1/2 Open-label, Multicentre, Dose Escalation and Expansion Study to Investigate the Safety, Tolerability, and Clinical Activity of Belantamab as Monotherapy and in Combination With Other Treatments in Participants With Multiple Myeloma [NCT05714839]Phase 1124 participants (Anticipated)Interventional2023-06-14Recruiting
A Phase 3, Open-Label Study to Evaluate Safety and Efficacy of Epcoritamab in Combination With Rituximab and Lenalidomide (R2) Compared to R2 in Subjects With Relapsed or Refractory Follicular Lymphoma (EPCORE FL-1) [NCT05409066]Phase 3500 participants (Anticipated)Interventional2022-09-20Recruiting
Phase I Open-Label, Dose Escalation Study To Determine The Maximum Tolerated Dose And To Evaluate The Safety Profile Of Lenalidomide (Revlimid®, CC-5013) With Pemetrexed In Subjects With Advanced Non-Small Cell Lung Cancer [NCT00179699]Phase 140 participants Interventional2005-09-30Terminated
A Multicenter, Single-Arm, Open-Label, Study To Evaluate The Safety And Efficacy Of Single-Agent Lenalidomide (Revlimid®) In Subjects With Androgen Independent Prostate Cancer [NCT00179738]Phase 240 participants Interventional2005-04-30Terminated
A Phase II Study of Clarithromycin (Biaxin), Lenalidomide (Revlimid), and Dexamethasone (Decadron) for Newly Diagnosed Subjects With Multiple Myeloma [NCT00151203]Phase 250 participants (Anticipated)Interventional2004-12-31Completed
A Phase II Trial of CC-5013 in Patients With Primary Systemic Amyloidosis [NCT00166413]Phase 238 participants (Anticipated)Interventional2005-04-30Completed
Phase I Safety Study of the Combination of Lenalidomide and Dacarbazine (DTIC) in Patients With Metastatic Malignant Melanoma Previously Untreated With Systemic Chemotherapy (CC-5013-MEL-003) [NCT00412581]Phase 128 participants (Actual)Interventional2005-09-30Completed
A Phase I, Open-Label, Multi-Center Study of CKD-581 in Combination With Lenalidomide and Dexamethasone in Patients With Previously Treated Multiple Myeloma [NCT03150316]Phase 118 participants (Anticipated)Interventional2017-05-10Recruiting
Evaluation of Ruxolitinib and Lenalidomide Combination as a Therapy for Patients With Myelofibrosis [NCT01375140]Phase 231 participants (Actual)Interventional2011-09-22Completed
Pilot Study of Lenalidomide and Dexamethasone in Combination With MEDI-551 in Previously Untreated Multiple Myeloma. [NCT01861340]Early Phase 120 participants (Actual)Interventional2014-05-31Completed
Phase I/II Study of Venetoclax or Lenalidomide in Combination With Ublituximab and Umbralisib in Subjects With Relapsed or Refractory CLL/SLL and NHL [NCT03379051]Phase 1/Phase 278 participants (Actual)Interventional2018-03-27Terminated(stopped due to Strategic/Business Decision)
A Randomized Phase II, 2-armed Study in Transplant Ineligible (TI) Patients With Newly Diagnosed Multiple Myeloma (NDMM) Comparing Carfilzomib + Thalidomide + Dexamethasone (KTd) Versus Carfilzomib + Lenalidomide + Dexamethasone (KRd) Induction Therapy Wi [NCT02891811]Phase 2124 participants (Actual)Interventional2017-03-10Active, not recruiting
A PHASE IB STUDY OF THE BTKi CC-292 COMBINED WITH LENALIDOMIDE IN ADULTS PATIENTS WITH RELAPSED/REFRACTORY B-CELL LYMPHOMA [NCT01766583]Phase 118 participants (Actual)Interventional2013-02-28Completed
A Phase II Trial of Ibrutinib, Lenalidomide and Rituximab for Patients With Relapsed/Refractory Mantle Cell Lymphoma [NCT02460276]Phase 250 participants (Actual)Interventional2015-04-30Completed
Double-blind Randomized Controlled Clinical Trial of Low-dose Lenalidomide in the Treatment of COVID-19 Disease [NCT04361643]Phase 4120 participants (Anticipated)Interventional2020-10-27Not yet recruiting
A Phase I Dose Escalation Study of Autologous Expanded Natural Killer (NK) Cells for Immunotherapy of Relapsed Refractory Neuroblastoma With Dinutuximab +/- Lenalidomide [NCT02573896]Phase 113 participants (Actual)Interventional2019-01-14Active, not recruiting
A Phase II Clinical Trial of Lenalidomide Intensification in Patients With Serologic/Asymptomatic Progression of Multiple Myeloma While on Lenalidomide Maintenance [NCT01463670]Phase 211 participants (Actual)Interventional2011-10-28Completed
Phase I/II Study of Lenalidomide Maintenance Following BEAM (+/- Rituximab) for Chemo-Resistant or High Risk Non-Hodgkin?s Lymphoma [NCT01035463]Phase 1/Phase 274 participants (Actual)Interventional2009-11-12Completed
Phase II Study of Lenalidomide for the Treatment of Relapsed or Refractory Hodgkin's Lymphoma [NCT00478959]Phase 230 participants (Actual)Interventional2006-12-31Completed
QUIREDEX: A National, Open-Label, Multicenter, Randomized, Phase III Study of Revlimid (Lenalidomide) and Dexamethasone (ReDex) Treatment Versus Observation in Patients With Smoldering Multiple Myeloma With High Risk of Progression [NCT00480363]Phase 3120 participants (Actual)Interventional2007-05-31Completed
Phase I Study of Lenalidomide in Acute Leukemias and Chronic Lymphocytic Leukemia. [NCT00466895]Phase 137 participants (Actual)Interventional2007-04-30Completed
Multicenter, Randomized, Double-blind, Placebo-controlled Study to Compare the Efficacy and Safety of CC-5013 vs. Placebo in Subjects With Metastatic Malignant Melanoma Whose Disease Has Progressed on Treatment With DTIC, IL-2, or IFN Based Therapy [NCT00057616]Phase 3274 participants Interventional2002-10-01Completed
Open Label Multi-center Pilot Phase I/II Non-controlled Clinical Trial to Assess Safety and Efficieecy of the Lenalidomide and Dexamethasone Association in Patients With Chronic Relapse or Treatment Resistant Lymphatic Leukemia Following Treatment Contain [NCT01246557]Phase 1/Phase 220 participants (Anticipated)Interventional2009-12-31Completed
Phase I Trial of Cytarabine and Lenalidomide in Relapsed or Refractory Acute Myeloid Leukemia Patients [NCT01246622]Phase 132 participants (Actual)Interventional2011-02-07Completed
A Phase I Combination Trial of SGN-33 (Anti-huCD33 mAb; HuM195; Lintuzumab) and Lenalidomide (Revlimid®) in Patients With Myelodysplastic Syndromes (MDS) [NCT00502112]Phase 113 participants (Actual)Interventional2008-03-31Completed
A Phase II, Single Arm Study Examining the Combination of Revlimid (Lenalidomide) and Vidaza (Azacitidine) (RA-CLL) for the Treatment of Relapsed/Refractory Chronic Lymphocytic Leukemia (CLL) and Small Lymphocytic Lymphoma (SLL) [NCT01241786]Phase 25 participants (Actual)Interventional2010-07-31Terminated(stopped due to The study was closed early due to poor accrual)
Randomized, Open Label, Multicenter Phase III Study of Efficacy and Safety in Polycythemia Vera Subjects Who Are Resistant to or Intolerant of Hydroxyurea: JAK Inhibitor INC424 Tablets Versus Best Available Care (The RESPONSE Trial) [NCT01243944]Phase 3222 participants (Actual)Interventional2010-10-27Completed
A Phase II Trial to Determine the Effect of Imetelstat (GRN163L) on Patients With Previously Treated Multiple Myeloma [NCT01242930]Phase 213 participants (Actual)Interventional2010-11-30Completed
A Phase I Study of SGN-40 (Anti-huCD40 mAb), Lenalidomide (Revlimid®, cc 5013), and Dexamethasone in Patients With Multiple Myeloma (MM) [NCT00525447]Phase 136 participants (Actual)Interventional2007-08-31Completed
Phenotypic Personalized Medicine: Systematically Optimized Combination Therapy in Multiple Myeloma Using CURATE.AI [NCT03759093]Phase 2/Phase 320 participants (Anticipated)Interventional2023-09-10Recruiting
A Phase 1b Study of JNJ-54767414 (Daratumumab) in Combination With Lenalidomide and Dexamethasone (DRd) in Japanese Subjects With Previously Untreated Multiple Myeloma Who Are Ineligible for High-dose Therapy and Autologous Stem Cell Transplantation [NCT02918331]Phase 17 participants (Actual)Interventional2016-09-30Completed
A Phase I/II Study of Lenalidomide and Obinutuzumab With CHOP for Diffuse Large B Cell Lymphoma [NCT02529852]Phase 1/Phase 259 participants (Actual)Interventional2015-11-04Completed
A Phase Ib/III Study to Evaluating the Efficacy and Safety of Parsaclisib in Combination With Rituximab and Lenalidomide Versus Rituximab in Combination With Lenalidomide in Subjects With Relapsed or Refractory Follicular Lymphoma [NCT05867030]Phase 10 participants (Actual)Interventional2023-07-28Withdrawn(stopped due to Development strategy adjustment)
Prospective, Single-arm, Phase II Clinical Study of Rituximab, Lenalidomide, and Zanubrutinib Combination Regimen Followed by Immunochemotherapy in the Treatment of Elderly Patients With Newly-diagnosed Diffuse Large B-cell Lymphoma [NCT05290090]Phase 231 participants (Anticipated)Interventional2021-10-01Recruiting
A Phase I Dose Escalation Study of Lenalidomide (Revlimid) in Combination With Fludarabine-Rituximab (Rituxan) for Previously Untreated CLL/SLL [NCT00543114]Phase 19 participants (Actual)Interventional2007-10-31Terminated(stopped due to lack of efficacy and tolerability)
Phase II Study of Lenalidomide and Rituximab in Subjects With Previously Untreated Indolent Non Hodgkin's Lymphoma [NCT01316523]Phase 230 participants (Actual)Interventional2010-12-31Active, not recruiting
A Phase II Trial of the Combination of Lenalidomide and Rituximab in Patients With Relapsed/Refractory Follicular NHL (RV 0163) [NCT00848328]Phase 230 participants (Anticipated)Interventional2008-08-25Active, not recruiting
Natural Killer Cells in Allogeneic Cord Blood Transplantation [NCT01619761]Phase 113 participants (Actual)Interventional2013-05-03Active, not recruiting
Efficacy of Eltrombopag Plus Lenalidomide Combination Therapy in Patients With IPSS Low and Intermediate-risk Myelodysplastic Syndrome With Isolated del5q: a Multicenter, Randomized, Double-blind, Placebo Controlled Study - QOL-ONE Rev2MDS [NCT02928419]Phase 22 participants (Actual)Interventional2015-05-31Terminated(stopped due to "the study has been closed due to a low rate of patient enrollment (2 patients since the start of the trial)")
Lenalidomide Following Rituximab and Fludarabine in Untreated Chronic Lymphocytic Leukemia [NCT00860457]Phase 222 participants (Actual)Interventional2008-02-29Completed
Bortezomib-lenalidomide-dexamethasone Combined With Radiotherapy for Newly Diagnosed Solitary Plasmacytoma [NCT05248633]Phase 2220 participants (Anticipated)Interventional2022-04-21Recruiting
A Phase 2, Multi-Center, Randomized, Double-Blinded, Parallel Group Study of the Safety and Efficacy of Different Lenalidomide (REVLIMID®) Dose Regimens in Subjects With Relapsed or Refractory B-Cell Chronic Lymphocytic Leukemia [NCT00963105]Phase 2104 participants (Actual)Interventional2009-10-19Completed
A Phase 3 Study Comparing Daratumumab, VELCADE (Bortezomib), Lenalidomide, and Dexamethasone (D-VRd) vs VELCADE, Lenalidomide, and Dexamethasone (VRd) in Subjects With Previously Untreated Multiple Myeloma Who Are Eligible for High-dose Therapy [NCT03710603]Phase 3690 participants (Actual)Interventional2018-12-14Active, not recruiting
A Phase II Study of Acalabrutinib, Lenalidomide, and Rituximab (aR2) in Patients With Previously Untreated Follicular Lymphoma [NCT04404088]Phase 224 participants (Anticipated)Interventional2020-07-16Recruiting
Phase 2 Trial of Pembrolizumab, Lenalidomide, and Dexamethasone for Initial Therapy of Newly Diagnosed Multiple Myeloma Eligible for Stem Cell Transplantation [NCT02880228]Phase 211 participants (Actual)Interventional2016-09-16Completed
An Open-label, Pharmacokinetic Study of Lenalidomide (Revlimid) and High-dose Dexamethasone Induction Therapy in Previously Untreated Multiple Myeloma Patients With Various Degrees of Renal Dysfunction - Validation of Official Dosing Guidelines for Renal [NCT01270932]Phase 228 participants (Actual)Interventional2010-11-30Completed
A Phase I/II Study of Carfilzomib Plus Lenalidomide and Rituximab in the Treatment of Relapsed/Refractory B-Cell Non-Hodgkin Lymphoma [NCT01729104]Phase 118 participants (Actual)Interventional2013-04-25Terminated(stopped due to Low response)
A Phase II Trial of Bortezomib (NSC #681239) + Lenalidomide (Revlimid™, CC-5013) (NSC #703813) for Relapsed/Refractory Mantle Cell Lymphoma [NCT00553644]Phase 253 participants (Actual)Interventional2007-11-15Completed
An Open-Label Phase II Study of the Safety and Efficacy of Doxorubicin and Cyclophosphamide in Combination With Bortezomib, Lenalidomide, and Dexamethasone for Treatment of Patients With Newly Diagnosed Multiple Myeloma [NCT01481194]Phase 235 participants (Actual)Interventional2011-11-30Completed
Phase II Trial of CC-5013 in Patients With Advanced Renal Cell Carcinoma With Either No Prior Treatment or One Prior Treatment Regimen [NCT00096525]Phase 20 participants Interventional2004-07-31Completed
A Multicenter, Open-label Study to Determine the Safety and Efficacy of Single-agent CC-5013 in Subjects With Relapsed and Refractory Multiple Myeloma [NCT00065351]Phase 2222 participants (Actual)Interventional2003-07-01Completed
Administration of an Allogeneic Myeloma GM-CSF Vaccine in Conjunction With a Lenalidomide Containing Regimen in Myeloma Patients With Near Complete Remission [NCT01349569]Phase 219 participants (Actual)Interventional2012-01-31Completed
A Multicenter, Double-Blind, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Lenalidomide in the Treatment of Painful Lumbar Radiculopathy [NCT00120120]Phase 2/Phase 3181 participants (Actual)Interventional2005-01-31Completed
Phase II Trial of REVLIMID® (Lenalidomide) for Therapy of Radioiodine-Unresponsive Papillary & Follicular Thyroid Carcinomas [NCT00287287]Phase 225 participants (Actual)Interventional2006-02-28Completed
A Phase 3, Randomized, Double-Blind, Placebo-Controlled, Multicenter Study to Evaluate the Efficacy and Safety of Tafasitamab Plus Lenalidomide in Addition to Rituximab Versus Lenalidomide in Addition to Rituximab in Patients With Relapsed/Refractory (R/R [NCT04680052]Phase 3654 participants (Actual)Interventional2021-04-15Active, not recruiting
A Phase 3 Randomized, Open Label, Multicenter Study of Isatuximab (SAR650984) in Combination With Lenalidomide and Dexamethasone Versus Lenalidomide and Dexamethasone in Patients With High-risk Smoldering Multiple Myeloma [NCT04270409]Phase 3320 participants (Anticipated)Interventional2020-06-16Recruiting
A Multicenter, Open Label, Phase Ib/II Study of Lenalidomide, Venetoclax and Obinutuzumab in Patients With Treatment-Naïve Follicular Lymphoma [NCT03980171]Phase 1/Phase 261 participants (Anticipated)Interventional2019-08-19Recruiting
LCI-HEM-MYE-KRdD-001: Phase II Study of Daratumumab Combined With Carfilzomib, Lenalidomide and Dexamethasone in Newly Diagnosed Multiple Myeloma [NCT04113018]Phase 239 participants (Actual)Interventional2020-01-10Active, not recruiting
A Phase II Trial of Revlimid® and Rituximab, for the Treatment of Patients With Relapsed or Refractory Chronic Lymphocytic Leukemia (CLL) [NCT01199575]Phase 230 participants (Actual)Interventional2010-08-31Completed
A Phase II Trial Addressing Feasibility and Activity of Clarithromycin + Lenalidomide Combination: a Full Oral Treatment for Patients With Relapsed/Refractory Extranodal Marginal Zone Lymphoma [NCT03031483]Phase 244 participants (Actual)Interventional2017-04-03Active, not recruiting
Lenalidomide to Reverse Drug Resistance After Lenvatinib Combined With PD-1 Inhibitors in the First-line Treatment of Advanced HCC :a Prospective, Exploratory, Single-arm, Open-label, Multi-center Clinical Study [NCT05831969]Phase 223 participants (Anticipated)Interventional2023-06-05Not yet recruiting
A Prospective, Open-label, Single-arm, Multicenter Study to Evaluate the Efficacy and Safety of Obinutuzumab(GA101) in Combination With Lenalidomide in Relapsed and Refractory(R/R) Marginal Zone Lymphoma (MZL) [NCT05846750]Phase 259 participants (Anticipated)Interventional2022-11-01Recruiting
A Phase II Study of Steroid Sparing Treatment With Daratumumab and Lenalidomide in Newly Diagnosed Transplant Ineligible Patients With Multiple Myeloma [NCT04635189]Phase 228 participants (Anticipated)Interventional2021-07-27Recruiting
A Phase II Study Evaluating the Efficacy/Safety of Lenalidomide With or Without Epoetin Beta in Transfusion-dependent ESA-resistant Patients With IPSS Low- and Intermediate-1 Risk Myelodysplastic Syndromes Without Chromosome 5 Abnormality. [NCT01718379]Phase 2132 participants (Actual)Interventional2010-07-31Completed
Lenalidomide and Azacitidine for Adaptive Immunotherapy in Multiple Myeloma: Pilot Study of Autologous Lymphocyte Mobilization Following Immuno-modulatory Therapy [NCT01050790]17 participants (Actual)Interventional2010-01-31Completed
A Pilot Study of the Combination of Lenalidomide (Revlimid®) With Two Different Dose Levels of Short Term Administration of Recombinant Human Stem Cell Factor (rhSCF; Ancestim) for Myelodysplasia. [NCT00434239]Early Phase 125 participants (Actual)Interventional2007-02-28Active, not recruiting
A Phase I Trial of CC-5013 (Lenalidomide) in Pediatric Patients With Recurrent or Refractory Primary CNS Tumors [NCT00100880]Phase 145 participants (Actual)Interventional2004-11-30Completed
Phase II Study of Response Adapted Therapy Using Single Agent Lenalidomide in Older Adults With Newly Diagnosed, Standard Risk Multiple Myeloma [NCT01054144]Phase 227 participants (Actual)Interventional2010-01-14Completed
A Phase 1b Open-label Study to Evaluate the Safety and Tolerability of Intravenous Modakafusp Alfa as Part of Combination Therapy in Adult Patients With Multiple Myeloma [NCT05556616]Phase 1120 participants (Anticipated)Interventional2022-10-27Active, not recruiting
Phase II Combination Immunotherapy After ASCT for Advanced Myeloma to Study MAGE-A3 Immunizations With Hiltonol® (Poly-ICLC) Plus Transfer of Vaccine-Primed Autologous T Cells Followed by Lenalidomide Maintenance [NCT01245673]Phase 228 participants (Actual)Interventional2011-05-10Completed
An Open-Label Phase I Study of the Safety and Efficacy of Bortezomib in Combination With CC-5013 in the Treatment of Subjects With Relapsed and Relapsed/Refractory Multiple Myeloma [NCT00153933]Phase 158 participants (Actual)Interventional2004-08-31Completed
A Non-interventional, Observational Post-marketing Registry of Patients Treated With Revlimid (Lenalidomide) in Taiwan [NCT01752075]100 participants (Actual)Observational [Patient Registry]2011-01-01Completed
Sintilimab and Lenalidomide as a Treatment for CAEBV:a Multicenter, Randomized, Double-blind, Placebo-controlled Clinical Trial [NCT04518982]Phase 251 participants (Anticipated)Interventional2020-08-01Recruiting
A Phase III Non-Inferiority Randomized Controlled Trial of Fixed Duration Versus Continuous Daratumumab Among Transplant Ineligible Older Adults With Newly Diagnosed Multiple Myeloma [NCT06182774]Phase 3559 participants (Anticipated)Interventional2024-02-28Not yet recruiting
Randomized Phase II Study of the Addition of Targeted Therapeutic Agents to Tafasitamab-Based Therapy in Non-Transplant-Eligible Patients With Relapsed/Refractory Large B-Cell Lymphoma [NCT05890352]Phase 2227 participants (Anticipated)Interventional2023-09-26Recruiting
A Phase 3 Randomized, Open-Label, Multicenter Study Evaluating the Efficacy of Axicabtagene Ciloleucel Versus Standard of Care Therapy in Subjects With Relapsed/Refractory Follicular Lymphoma [NCT05371093]Phase 3230 participants (Anticipated)Interventional2022-09-12Recruiting
A Dose Escalation and Expansion Study of ABBV-383 in Combination With Anti-Cancer Regimens for the Treatment of Patients With Relapsed/Refractory Multiple Myeloma [NCT05259839]Phase 1270 participants (Anticipated)Interventional2022-10-20Recruiting
Phase II Multicenter Trial of Anti-BCMA CAR T-Cell Therapy for MM Patients With Sub-Optimal Response After Auto HCT and Maintenance Len. BMTCTN1902 [NCT05032820]Phase 240 participants (Anticipated)Interventional2022-01-05Recruiting
Phase II Multicenter Trial of Single Autologous Hematopoietic Cell Transplant Followed by Lenalidomide Maintenance for Multiple Myeloma With or Without Vaccination With Dendritic Cell/Myeloma Fusions (BMT CTN 1401) [NCT02728102]Phase 2203 participants (Actual)Interventional2016-07-31Completed
A Phase Ib/II Study Evaluating the Safety and Efficacy of Obinutuzumab in Combination With Polatuzumab Vedotin and Lenalidomide in Patients With Relapsed or Refractory Follicular Lymphoma and Rituximab in Combination With Polatuzumab Vedotin and Lenalidom [NCT02600897]Phase 1/Phase 2114 participants (Actual)Interventional2016-03-24Completed
A Phase I Study of Gemcitabine, Carboplatin and Lenalidomide (GCL) for Treatment of Patients With Advanced/Metastatic Urothelial Carcinoma (UC) and Other Solid Tumors [NCT01352962]Phase 118 participants (Actual)Interventional2011-09-26Completed
A Phase II Trial of Revlimid, Cyclophosphamide, and Dexamethasone in Patients With > Newly Diagnosed Active Multiple Myeloma [NCT00478218]Phase 253 participants (Actual)Interventional2006-07-31Completed
A Phase II Study of Dexamethasone (DECADRON®), Thalidomide (THALOMID®), and Lenalidomide (REVLIMID®) for Subjects With Relapsed or Refractory Multiple Myeloma [NCT00538824]Phase 25 participants (Actual)Interventional2007-12-31Terminated(stopped due to low enrollment)
A Phase II Trial For High-Risk Myeloma Evaluating Accelerating and Sustaining Complete Remission (AS-CR) by Applying Non-Host-Exhausting and Timely Dose-Reduced Mel-80-CFZ-TD-Pace Transplant(s) With Interspersed Mel-20-CFZ-TD-Pace With CFZ-RD and CFZ-D Ma [NCT02128230]Phase 220 participants (Actual)Interventional2014-06-10Terminated(stopped due to Low enrollment and Futility as determined by Amgen's Carfilzomib NASCR program.)
Phase Ib Dose Finding Study of Bruton's Tyrosine Kinase (BTK) Inhibitor, Ibrutinib (PCI-32765) Plus Lenalidomide / Rituximab in Relapsed or Refractory Mantle Cell Lymphoma (MCL) [NCT02446236]Phase 127 participants (Actual)Interventional2015-06-18Active, not recruiting
A Phase II Study of Lenalidomide in Patients With Chronic Lymphocytic Leukemia Older Than 65 Years of Age [NCT01011894]Phase 226 participants (Actual)Interventional2009-11-06Completed
Multicenter, Open-label, Single-arm, Phase 1b/2 Study of the Safety and Efficacy of Combination Treatment w/ Carfilzomib, Lenalidomide (Revlimid®) and Dexamethasone (CRD) in Subjects w/ Newly Diagnosed, Previously Untreated Multiple Myeloma Requiring Syst [NCT01029054]Phase 1/Phase 253 participants (Actual)Interventional2009-09-30Completed
A Pilot Study of Lenalidomide in Adult Diamond-Blackfan Anemia Patients With Red Blood Cell Transfusion-Dependent Anemia [NCT01034592]Phase 12 participants (Actual)Interventional2009-11-30Terminated(stopped due to Poor accrual related to rarity of Diamond-Blackfan anemia (DBA))
A Multicenter Prospective Phase II Study of Rituximab Combined, Lenalidomide, Dexamethasone Followed by Lenalidomide Maintenance in Patients With Newly Diagnosed Waldenström's Macroglobulinemia [NCT03697356]Phase 254 participants (Anticipated)Interventional2019-03-06Recruiting
A Phase II Trial of Panobinostat and Lenalidomide in Patients With Relapsed or Refractory Hodgkin's Lymphoma [NCT01460940]Phase 224 participants (Actual)Interventional2011-10-13Completed
A Phase 2 Study of Epcoritamab and Lenalidomide (E-Len) in Patients With Previously Untreated Follicular Lymphoma (FL) [NCT06112847]Phase 227 participants (Anticipated)Interventional2024-03-22Not yet recruiting
A Randomized, Open-Label, Phase 3 Trial to Compare the Efficacy and Safety of Idecabtagene Vicleucel With Lenalidomide Maintenance Versus Lenalidomide Maintenance Therapy Alone in Adult Participants With Newly Diagnosed Multiple Myeloma Who Have Suboptima [NCT06045806]Phase 3618 participants (Anticipated)Interventional2023-10-16Recruiting
Phase I Trial of Three-Weekly Docetaxel, Carboplatin and Oral CC-5013 in Patients With Advanced Solid Tumors [NCT00415116]Phase 114 participants (Actual)Interventional2004-08-31Completed
Combination Post-transplant Consolidation Therapy With Isatuximab, Lenalidomide, Dexamethasone (IsaRD) in Multiple Myeloma Patients With Persistent Marrow Minimal Residual Disease (Elimination of MRD After Transplant; E-MAT) [NCT05690984]Phase 231 participants (Anticipated)Interventional2023-06-26Recruiting
Lenalidomide Maintenance in Plasma Cell Myeloma [NCT02538198]Phase 2108 participants (Actual)Interventional2015-08-31Active, not recruiting
A Multicenter, Randomized, Double-Blind, Placebo-Controlled,Parallel-Group Study to Evaluate the Safety and Efficacy of CC-5013 in the Treatment of Adolescents and Adults With Moderately Severe Crohn's Disease [NCT00446433]Phase 290 participants Interventional2002-03-31Completed
Phase II Trial of Low Dose Lenalidomide and Dexamethasone in Relapsed or Refractory Multiple Myeloma (Rev-Lite) in Patients at High Risk for Myelosuppression [NCT00482261]Phase 2150 participants (Anticipated)Interventional2007-06-30Active, not recruiting
A Multi-Arm Complete Phase 1 Trial of Valproic Acid-Based 2-Agent Oral Regimens for Patients With Advanced Solid Tumor [NCT00495872]Phase 1204 participants (Actual)Interventional2007-06-30Completed
Phase II Trial of Lenalidomide in Older Patients (>/= 60 Years) With Untreated Acute Myeloid Leukemia Without Chromosome 5q Abnormalities [NCT00546897]Phase 248 participants (Actual)Interventional2007-02-28Completed
A Phase 2 Study of Lenalidomide and Low-dose Dexamethasone in Combination With Dalteparin in Previously Untreated Multiple Myeloma [NCT01518465]Phase 213 participants (Actual)Interventional2012-01-09Terminated(stopped due to Insufficient Accrual)
Combination of Lenalidomide and Obinutuzumab (GA101) in Patients With Recurrent Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL/SLL) [NCT02225275]Phase 29 participants (Actual)Interventional2016-03-31Terminated(stopped due to Due to slow accrual)
An Open Label, Phase 2 Study of Ibrutinib in Combination With Rituximab and Lenalidomide in Previously Untreated Subjects With Follicular Lymphoma and Marginal Zone Lymphoma [NCT02532257]Phase 246 participants (Actual)Interventional2016-04-11Completed
Phase II Study of the Combination of Elotuzumab With Lenalidomide as Maintenance Therapy Post Autologous Stem Cell Transplant in Patients With Multiple Myeloma [NCT02420860]Phase 2113 participants (Actual)Interventional2015-04-14Active, not recruiting
A Randomized, Double-blind, Placebo-Controlled Phase II Trial of an Allogeneic Myeloma GM-CSF Vaccine With Lenalidomide in Multiple Myeloma Patients in Complete or Near Complete Remission [NCT03376477]Phase 254 participants (Anticipated)Interventional2019-09-23Active, not recruiting
Lenalidomide and Low Dose Dexamethasone Versus Bortezomib, Lenalidomide and Low Dose Dexamethasone for Induction, in Patients With Previously Untreated Multiple Myeloma [NCT01530594]Phase 3440 participants (Actual)Interventional2009-01-31Completed
A Pilot Study of G-CSF to Disrupt the Bone Marrow Microenvironment in Bortezomib-, Carfilzomib-, or IMID-Refractory Multiple Myeloma [NCT01537861]Early Phase 17 participants (Actual)Interventional2012-06-30Terminated(stopped due to Unexpected toxicity (2 early deaths))
Lenalidomide Plus Rituximab for Recurrent/Refractory CNS and Intraocular Lymphoma [NCT01542918]Phase 114 participants (Actual)Interventional2012-12-17Completed
Lenalidomide as Second-line Treatment for Advanced Hepatocellular Carcinoma (HCC): a Phase II Clinical Trial [NCT01545804]Phase 255 participants (Actual)Interventional2011-08-31Completed
A Pilot Study of Lenalidomide and Dexamethasone in Patients With Primary Plasma Cell Leukemia [NCT01553357]Phase 223 participants (Actual)Interventional2009-03-31Completed
STUDY OF THE EFFICACY AND SAFETY OF FIRST LINE TREATMENT WITH CHOP AND LENALIDOMIDE (Rev-CHOP) IN PATIENTS AGED FROM 60 TO 80 YEARS WITH PREVIOUSLY UNTREATED ANGIOIMMUNOBLASTIC T-CELL LYMPHOMA (AITL) [NCT01553786]Phase 280 participants (Actual)Interventional2011-11-30Completed
Randomised Comparisons, in Myeloma Patients of All Ages, of Thalidomide, Lenalidomide, Carfilzomib and Bortezomib Induction Combinations, and of Lenalidomide and Combination Lenalidomide Vorinostat as Maintenance (Myeloma XI) [NCT01554852]Phase 34,420 participants (Actual)Interventional2010-05-31Active, not recruiting
A Multicentre Prospective Phase II Single Arm Trial Evaluating the Benefit of Therapy With Lenalidomide (Revlimid®) in Relapsed or Refractory Primary-cutaneous Large B-cell Lymphoma (Leg-type) After First Line Treatment by Chemotherapy Plus Rituximab for [NCT01556035]Phase 219 participants (Actual)Interventional2012-07-31Completed
A Multicentre Open Randomized Phase II Study of the Efficacy and Safety of Azacitidine Alone or in Combination With Lenalidomide in High-risk Myeloid Disease (High-risk Myelodysplastic Syndrome and Acute Myeloid Leukemia) With a Karyotype Including Del(5q [NCT01556477]Phase 272 participants (Anticipated)Interventional2012-03-31Recruiting
Phase I Study of CC-5013 (Lenalidomide NSC# 703813) in Pediatric Patients With Relapsed/Refractory Solid Tumors or Myelodysplastic Syndrome [NCT00104962]Phase 124 participants (Actual)Interventional2005-03-31Completed
A Single-Center, Open-Label, Between-Patient, Dose-Escalation Phase 1 Study of CDC-501 In Patients With Solid Tumors [NCT00046735]Phase 124 participants (Actual)Interventional2002-06-01Completed
A Phase I Trial of CC-5013 (Lenalidomide) and CCI-779 in Patients With Relapsed or Refractory Multiple Myeloma [NCT00398515]Phase 125 participants (Actual)Interventional2007-03-31Completed
A Non-interventional, Post-authorization Safety Study of Patients With Relapsed or Refractory Mantle Cell Lymphoma to Further Investigate and Characterize the Association of Lenalidomide With Tumor Flare Reaction and High Tumor Burden [NCT03647124]560 participants (Anticipated)Observational2019-03-13Recruiting
Phase II Clinical Protocol for the Treatment of Patients With Previously Untreated CLL With Four or Six Cycles of Fludarabine and Cyclophosphamide With Rituximab (FCR) Plus Lenalidomide Followed by Lenalidomide Consolidation/ Maintenance [NCT01723839]Phase 221 participants (Actual)Interventional2012-02-22Completed
A Multicenter, Open Label Study of Oral Melphalan, Prednisone, and CC-5013 (Revlimid) (MPR) as Induction Therapy in Elderly Newly Diagnosed Multiple Myeloma Patients [NCT00396045]Phase 1/Phase 254 participants Interventional2005-01-31Completed
A Phase I Trial Of A Thalidomide Analog, CC-5013, For The Treatment Of Patients With Recurrent High-Grade Gliomas [NCT00036894]Phase 10 participants Interventional2002-03-31Completed
A Phase Ib, Open-label, Randomized Study to Assess Safety and Preliminary Efficacy of Tafasitamab in Addition to R-CHOP or Tafasitamab Plus Lenalidomide in Addition to R-CHOP in Patients With Newly Diagnosed Diffuse Large B-Cell Lymphoma (DLBCL) - First-M [NCT04134936]Phase 166 participants (Actual)Interventional2019-12-11Completed
A Multicenter, Single-Arm, Open-Label Study of the Efficacy and Safety of CC-5013 Monotherapy in Subjects With Myelodysplastic Syndromes [NCT00064974]Phase 2215 participants (Actual)Interventional2003-06-30Completed
A Multicenter, Open-Label Study to Evaluate the Preliminary Safety and Efficacy of CC-5013 in the Treatment of Complex Regional Pain Syndrome (CRPS) [NCT00067743]Phase 240 participants (Actual)Interventional2003-08-31Completed
A Phase II Multicenter Study of Carfilzomib, Lenalidomide and Dexamethasone (KRd) as Induction Therapy, Followed by High-dose Therapy With Melphalan and Autologous Peripheral Blood Stem Cell Transplantation, Consolidation With KRd, and Maintenance With Le [NCT02415413]Phase 290 participants (Anticipated)Interventional2015-05-31Active, not recruiting
A Phase II Study of Continuous Versus Syncopated Dosing of CC-5013 for the Treatment of Refractory Multiple Myeloma [NCT00051116]Phase 2100 participants (Actual)Interventional2002-05-31Completed
A Phase II Open Label Study of the Safety and Efficacy of CC-5013 Treatment For Patients With Myelodysplastic Syndrome [NCT00044382]Phase 225 participants Interventional2002-02-01Completed
Multicenter, Randomized, Controlled, Double-Blind, Parallel-Group Study to Compare the Efficacy and Safety of Two CC-5013 Dose Regimens in Subjects With Metastatic Malignant Melanoma Whose Disease Has Progressed on Treatment With DTIC, IL-2 or IFN Based T [NCT00055562]Phase 2/Phase 3274 participants Interventional2003-01-31Completed
A Phase I/II Trial Investigating the Combination of Pembrolizumab (Keytruda) With Cyclophosphamide and Lenalidomide for Patients With Relapsed Multiple Myeloma [NCT03191981]Phase 1/Phase 20 participants (Actual)Interventional2017-08-01Withdrawn(stopped due to Withdrawal of pharma support prior to opening to recruitment)
Phase I Safety Study of the Combination of Lenalidomide and DTIC (Dacarbazine) in Patients With Metastatic Malignant Melanoma Previously Untreated With Systemic Chemotherapy [NCT00179608]Phase 128 participants Interventional2005-09-01Completed
A Multicenter, Single-Arm, Open-Label, Study to Evaluate the Safety and Efficacy of Single-Agent Lenalidomide (Revlimid, CC-5013) in Subjects With Recurrent Non-Small Cell Lung Cancer [NCT00179686]Phase 240 participants Interventional2005-03-31Completed
Phase I/II Open-Label, Dose Escalation Study To Determine The Maximum Tolerated Dose And To Evaluate The Safety Profile of Lenalidomide (Revlimid®) With Liposomal Doxorubicin In Subjects With Advanced Ovarian and Primary Peritoneal Carcinoma [NCT00179725]Phase 1/Phase 260 participants Interventional2005-11-30Terminated
PH Ib Study of Elotuzumab in Combination With Lenalidomide and Dexamethasone in Subjects With Multiple Myeloma and Normal Renal Function, Severe Renal Impairment, or End Stage Renal Disease Requiring Dialysis [NCT01393964]Phase 135 participants (Actual)Interventional2012-01-06Completed
A Phase 3, Randomized, Open Label Trial of Lenalidomide/Dexamethasone With or Without Elotuzumab in Subjects With Previously Untreated Multiple Myeloma [NCT01891643]Phase 323 participants (Actual)Interventional2013-09-30Terminated(stopped due to Insufficient enrollment)
A Phase I/II Study of Revlimid (Lenalidomide) in Combination With Vidaza (Azacitidine) in Patients With Advanced Myelodysplastic Syndrome (MDS) [NCT00352001]Phase 1/Phase 237 participants (Actual)Interventional2006-05-31Completed
A Phase II Study of Combination Oral CC-5013 Lenalidomide (Revlimid™), Oral Sunitinib (Sutent™) and Low Dose Oral Metronomic Cyclophosphamide for the Treatment of Stage IV Ocular Melanoma [NCT00482911]Phase 212 participants (Actual)Interventional2007-04-30Terminated(stopped due to Investigator left the institute.)
A Phase II Study of Lenalidomide as Initial Treatment of Patients With Chronic Lymphocytic Leukemia Age 65 or Older. [NCT00535873]Phase 261 participants (Actual)Interventional2007-10-31Completed
A Phase I/II Study of Fludarabine, Rituximab, and Lenalidomide in Minimally Treated and Untreated Patients With Chronic Lymphocytic Leukemia [NCT00536341]Phase 1/Phase 264 participants (Actual)Interventional2008-01-31Completed
A Phase II Multicenter Study of Lenalidomide in Relapsed or Refractory Classical Hodgkin Lymphoma [NCT00540007]Phase 280 participants (Actual)Interventional2007-09-06Completed
A PHASE 1B/2, OPEN LABEL UMBRELLA STUDY OF ELRANATAMAB (PF-06863135), A B-CELL MATURATION ANTIGEN (BCMA) CD3 BISPECIFIC ANTIBODY, IN COMBINATION WITH OTHER ANTI-CANCER TREATMENTS IN PARTICIPANTS WITH MULTIPLE MYELOMA [NCT05090566]Phase 2105 participants (Anticipated)Interventional2021-10-27Recruiting
Phase 1b/2 Study of Venetoclax, Ibrutinib, Prednisone, Obinutuzumab, and Revlimid (ViPOR) in Relapsed/Refractory B-cell Lymphoma [NCT03223610]Phase 1/Phase 2145 participants (Anticipated)Interventional2018-02-09Recruiting
Phase II Study of Lenalidomide and Rituximab for Patients With Relapsed and/or Refractory CD20+ Multiple Myeloma [NCT00567229]Phase 23 participants (Actual)Interventional2007-11-30Terminated(stopped due to Lack of Accrual)
A Multicenter Study of Ibrutinib and Lenalidomide in Combination With DA-EPOCH-R in Subjects With Relapsed or Refractory Diffuse Large B-cell Lymphoma [NCT02142049]Phase 1/Phase 235 participants (Actual)Interventional2014-05-31Completed
A Phase II Study of Lenalidomide in Patients With Relapsed or Refractory HTLV-1 Associated Adult T Cell Leukemia/Lymphoma [NCT01274533]Phase 24 participants (Actual)Interventional2010-12-31Completed
DLCL002 Protocol for Young Patients With Newly Diagnosed High Risk Aggressive B-cell Lymphoma, a Multicenter Phase II Study [NCT03837873]Phase 2118 participants (Anticipated)Interventional2019-01-21Recruiting
"A Phase II Trial of Revlimid® and On Demand Dexamethasone Dosing in Patients With Newly Diagnosed Symptomatic Multiple Myeloma" [NCT00772915]Phase 239 participants (Actual)Interventional2008-12-03Completed
A Pilot Open Label Study to Investigate the Effect of Cytotoxic Therapy and/or Radiotherapy Including Lenalidomide on Cancer Related Sleep Disturbances in Chronic Lymphocytic Leukemia (CLL) and Breast Cancer Patients Experiencing Clinically Significant Fa [NCT00792077]Phase 26 participants (Actual)Interventional2008-11-30Completed
A Phase II Study of Lenalidomide in Combination With Gemcitabine in Subjects With Untreated Advanced Carcinoma of the Pancreas [NCT00837031]Phase 272 participants (Actual)Interventional2009-02-28Completed
Phase II Subcutaneous VELCADE and Oral Cyclophosphamide-based Induction + Sequential VELCADE and Revlimid Maintenance for Newly Diagnosed Multiple Myeloma in Non-transplant Candidates: An Entirely Non-intravenous Regimen [NCT01729338]Phase 217 participants (Actual)Interventional2012-12-19Terminated
Post-Transplant Maintenance Therapy With Isatuximab Plus Lenalidomide for High-Risk Multiple Myeloma Patients [NCT05776979]Phase 261 participants (Anticipated)Interventional2023-09-30Recruiting
A Phase II Study of Obinutuzumab and Lenalidomide in Previously Untreated Subjects With Follicular Lymphoma [NCT02871219]Phase 296 participants (Actual)Interventional2016-12-06Active, not recruiting
A Phase 1b/2 Study of Selinexor (KPT-330) in Combination With Backbone Treatments for Relapsed/Refractory Multiple Myeloma and Newly Diagnosed Multiple Myeloma [NCT02343042]Phase 1/Phase 2518 participants (Actual)Interventional2015-10-31Active, not recruiting
A Phase I/II Study of Lenalidomide and Obinutuzumab (GA101) in Relapsed Indolent Non-Hodgkin's Lymphoma [NCT01995669]Phase 1/Phase 270 participants (Actual)Interventional2014-05-21Completed
A Multicenter, Single-Arm, Open-Label, Expanded Access Program for Lenalidomide With or Without Dexamethasone in Previously Treated Subjects With Multiple Myeloma [NCT00179647]Phase 31,913 participants (Actual)Interventional2005-09-30Completed
A Multicenter, Single-Arm, Open-Label Expanded Access Program for Lenalidomide Plus Dexamethasone in Previously Treated Subjects With Multiple Myeloma [NCT00478777]Phase 3150 participants (Actual)Interventional2007-03-31Completed
Lenalidomide Plus R-CHOP for CNS Relapse Prophylaxis in Diffuse Large B-cell Lymphoma [NCT04544059]Phase 287 participants (Anticipated)Interventional2020-10-01Not yet recruiting
E- PRISM: Precision Intervention Smoldering Myeloma: Phase II Trial of Combination of Elotuzumab, Lenalidomide and Dexamethasone in High-Risk Smoldering Multiple Myeloma [NCT02279394]Phase 251 participants (Actual)Interventional2014-12-11Completed
A Phase I Trial of Lenalidomide and Idelalisib in Recurrent Follicular Lymphoma [NCT01644799]Phase 18 participants (Actual)Interventional2013-07-31Completed
A Phase I Study of Lenalidomide Plus Chemotherapy With Mitoxantrone, Etoposide, and Cytarabine for the Reinduction of Patients With Acute Myelogenous Leukemia [NCT01681537]Phase 136 participants (Actual)Interventional2012-09-30Completed
An Open-Label Phase I Study of the Safety of Perifosine in Combination With Lenalidomide and Dexamethasone for Patients With Relapsed or Refractory Multiple Myeloma [NCT00415064]Phase 132 participants (Actual)Interventional2006-12-31Completed
A Multicenter Phase I/II Trial Evaluating the Safety and Efficacy of Lenalidomide (Revlimid, CC-5013) in Combination With Doxorubicin and Dexamethasone (RAD) in Patients With Relapsed or Refractory Multiple Myeloma [NCT00306813]Phase 1/Phase 253 participants (Anticipated)Interventional2004-09-30Completed
Evaluation of Lenalidomide (REVLIMID®) in Cutaneous LE: A Prospective, Non Controlled, Open Label Pilot Study to Evaluate the Off-Label Use of the FDA-approved Drug Lenalidomide (REVLIMID®) in Patients With Cutaneous Lupus Erythematosus [NCT00633945]5 participants (Actual)Interventional2007-11-30Completed
Phase 1 Study of Tasquinimod Alone and in Combination With Standard Therapy for Relapsed or Refractory Myeloma [NCT04405167]Phase 134 participants (Anticipated)Interventional2020-07-10Recruiting
A Phase 1 Dose-Escalation and Exploratory Dose Expansion Study of KRT-232 (AMG 232) in Combination With Carfilzomib, Lenalidomide, and Dexamethasone in Relapsed and/or Refractory Myeloma [NCT03031730]Phase 140 participants (Anticipated)Interventional2018-06-14Recruiting
2015-12: A Phase II Study Exploring the Use of Early and Late Consolidation/Maintenance With Anti-CD38 (Protein) Monoclonal Antibody to Improve Progression Free Survival in Patients With Newly Diagnosed Multiple Myeloma [NCT03004287]Phase 250 participants (Anticipated)Interventional2017-07-01Active, not recruiting
Randomized Phase III Trial of Bortezomib, Lenalidomide, and Dexamethasone (VRd) Versus Carfilzomib, Lenalidomide, and Dexamethasone (CRd) Followed by Limited or Indefinite Duration Lenalidomide Maintenance in Patients With Newly Diagnosed Symptomatic Mult [NCT01863550]Phase 31,087 participants (Actual)Interventional2013-12-06Active, not recruiting
An Open, Single-arm Clinical Study of Lenalidomide Combined With G-CHOP(LO-CHOP) in the Treatment of Newly Diagnosed Diffuse Large B-cell Lymphoma With Follicular Lymphoma (CDLBCL-FL). [NCT06151080]Phase 238 participants (Anticipated)Interventional2023-11-22Recruiting
Phase 1 Study of Venetoclax, Ibrutinib, Prednisone, Obinutuzumab, and Revlimid (VIPOR) for Diffuse Large B-cell Lymphoma Involving the Central Nervous System [NCT05211336]Phase 112 participants (Anticipated)Interventional2022-04-19Recruiting
A Phase II, Multi-center, Open-label, Randomized Study of Vorinostat Plus Lenalidomide and Dexamethasone or Lenalidomide Plus Dexamethasone in Multiple Myeloma Patients Who Experience Biochemical Relapse During Lenalidomide Maintenance Therapy [NCT01501370]Phase 20 participants (Actual)Interventional2012-01-31Withdrawn
Clofarabine Followed by Lenalidomide for Treatment of High Risk Myelodysplastic Syndromes and Acute Myeloid Leukemia [NCT01629082]Phase 14 participants (Actual)Interventional2012-06-06Completed
Phase II Trial With Lenalidomide-Dexamethasone Combination in the Treatment of POEMS Syndrome. [NCT01639898]Phase 251 participants (Actual)Interventional2012-07-31Completed
A Phase I Study of Lenalidomide and Anti-GD2 Mab Ch14.18 +/- Isotretinoin in Patients With Refractory/Recurrent Neuroblastoma [NCT01711554]Phase 127 participants (Actual)Interventional2013-02-04Active, not recruiting
Salvage in Patients With Myelodysplastic Syndrome After Failure of Hypomethylating Agents: Lenalidomide as a Second-line Therapy [NCT01673308]Phase 235 participants (Anticipated)Interventional2012-08-31Active, not recruiting
A Multicenter, Phase I/II Study of Sequential Epigenetic and Immune Targeting in Combination With Nab-Paclitaxel/Gemcitabine in Patients With Advanced Pancreatic Ductal Adenocarcinoma. [NCT04257448]Phase 1/Phase 275 participants (Anticipated)Interventional2020-05-25Active, not recruiting
A Phase I/II Study of Bendamustine, Lenalidomide and Low-dose Dexamethasone, (BdL) for the Treatment of Patients With Relapsed Myeloma. [NCT01686386]Phase 1/Phase 260 participants (Anticipated)Interventional2010-02-28Recruiting
Clinical Study of Azacytidine Combined With Lenalidomide As Maintenance Therapy Based on MRD Monitoring In Elderly or Unfit Patients With Acute Myeloid Leukemia [NCT04490707]Phase 360 participants (Anticipated)Interventional2020-09-01Recruiting
An Open, Multicentric Phase II Trial to Evaluate the Efficacy and Safety of Bendamustine, Lenalidomide (Revlimid®) and Dexamethasone (BRd) as 2nd-line Therapy for Patients With Relapsed or Refractory Multiple Myeloma [NCT01701076]Phase 250 participants (Actual)Interventional2012-03-31Completed
Fludarabine/Rituximab Combined With Escalating Doses of Lenalidomide in Untreated Chronic Lymphocytic Leukemia (CLL) - a Dose-finding Study With Escalating Starting Dose of Lenalidomide and Concomitant Evaluation of Safety and Efficacy [NCT01703364]Phase 1/Phase 212 participants (Actual)Interventional2012-06-30Completed
S1803, Phase III Study of Daratumumab/rHuPH20 (NSC-810307) + Lenalidomide or Lenalidomide as Post-Autologous Stem Cell Transplant Maintenance Therapy in Patients With Multiple Myeloma (MM) Using Minimal Residual Disease to Direct Therapy Duration (DRAMMAT [NCT04071457]Phase 31,100 participants (Anticipated)Interventional2019-08-13Recruiting
A Pilot Study of DKN-01 and Lenalidomide (Revlimid®)/Dexamethasone Versus Lenalidomide/Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT01711671]Phase 18 participants (Actual)Interventional2013-05-31Completed
A Phase 1, Open-Label, Two-Part, Fixed-Sequence Crossover Study to Evaluate the Effect of P-glycoprotein Inhibition on Lenalidomide Pharmacokinetics in Healthy Male Subjects [NCT01712828]Phase 131 participants (Actual)Interventional2012-10-01Completed
A Phase 1/2a Dose Escalation Study of PVX-410, a Multi-Peptide Cancer Vaccine, in Patients With Smoldering Multiple Myeloma [NCT01718899]Phase 122 participants (Actual)Interventional2012-11-30Completed
A MULTICENTER, PHASE 1B, OPEN-LABEL STUDY TO DETERMINE THE SAFETY AND ACTIVITY OF CC-292 IN COMBINATION WITH LENALIDOMIDE IN SUBJECTS WITH RELAPSED AND/OR REFRACTORY CHRONIC LYMPHOCYTIC LEUKEMIA / SMALL LYMPHOCYTIC LYMPHOMA [NCT01732861]Phase 120 participants (Actual)Interventional2012-12-28Completed
Bendamustine, Lenalidomide and Rituximab (R2-B) Combination as a Second-Line Therapy for First Relapsed-Refractory Mantle Cell Lymphomas: A Phase II Study [NCT01737177]Phase 242 participants (Actual)Interventional2012-07-31Completed
An Open Label, International, Multi-centre, Phase I/IIa Study of Lenalidomide (Revlimid) and Romidepsin (Istodax) for Relapsed /Refractory Hodgkin Lymphoma, Mature T-cell Lymphoma and Multiple Myeloma. (RId Study) [NCT01742793]Phase 112 participants (Actual)Interventional2012-10-31Terminated(stopped due to The study was terminated due to withdrawal of institutional support, Phase II never began.)
An Open-Label, Dose-Escalation, Phase 1/2 Study of the Oral Formulation of IXAZOMIB (MLN9708), Administered Twice-weekly in Combination With Lenalidomide and Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma Requiring Systemic Treatment [NCT01383928]Phase 1/Phase 264 participants (Actual)Interventional2011-10-31Completed
A Phase II Trial of Mosunetuzumab, Polatuzumab, Tafasitamab, and Lenalidomide in Patients With Relapsed B-cell NHL [NCT05615636]Phase 236 participants (Anticipated)Interventional2023-04-28Recruiting
A Phase 3 Randomized Study Comparing Daratumumab, Bortezomib, Lenalidomide and Dexamethasone (DVRd) Followed by Ciltacabtagene Autoleucel Versus Daratumumab, Bortezomib, Lenalidomide and Dexamethasone (DVRd) Followed by Autologous Stem Cell Transplant (AS [NCT05257083]Phase 3750 participants (Anticipated)Interventional2023-10-10Recruiting
MRD-Guided Sequential Therapy For Deep Response in Newly Diagnosed Multiple Myeloma - MASTER-2 Trial [NCT05231629]Phase 2300 participants (Anticipated)Interventional2023-12-13Recruiting
"Phase Ib Trial With Dose Expansion of the Brutons Tyrosine Kinase (BTK) Inhibitor, Ibrutinib, in Combination With Rituximab and Lenalidomide in Patients With Refractory/Recurrent Primary Central Nervous System Lymphoma (PCNSL) and Refractory/Recurrent Se [NCT03703167]Phase 125 participants (Actual)Interventional2019-01-22Active, not recruiting
MAGNETISMM-1 A PHASE I, OPEN LABEL STUDY TO EVALUATE THE SAFETY, PHARMACOKINETIC, PHARMACODYNAMIC AND CLINICAL ACTIVITY OF ELRANATAMAB (PF-06863135), A B-CELL MATURATION ANTIGEN (BCMA) - CD3 BISPECIFIC ANTIBODY, AS A SINGLE AGENT AND IN COMBINATION WITH I [NCT03269136]Phase 1101 participants (Actual)Interventional2017-11-29Active, not recruiting
An Expanded Access Program for Elotuzumab in Combination With Lenalidomide Plus Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma in Japan [NCT02856438]0 participants Expanded AccessNo longer available
Phase II Study of the Combination of MLN 9708 With Lenalidomide as Maintenance Therapy Post Autologous Stem Cell Transplant in Patients With Multiple Myeloma [NCT01718743]Phase 283 participants (Actual)Interventional2012-12-03Completed
Phase I Study of Lenalidomide to Augment Anti-Tumor Immunity Following Allogeneic Transplantation [NCT01750762]Phase 11 participants (Actual)Interventional2012-12-31Terminated
A Phase I Trial of Ipilimumab (Anti CTLA- 4 Antibody) in Combination With Lenalidomide (IMiD) in Patients With Advanced Malignancies [NCT01750983]Phase 1101 participants (Anticipated)Interventional2013-03-31Completed
HO11415: Phase II Study of Bendamustine and Rituximab Induction Chemoimmunotherapy Followed by Maintenance Rituximab (Rituxan®) and Lenalidomide (Revlimid®) in Relapsed and Refractory Chronic Lymphocytic Leukemia (CLL) and Small Lymphocytic Lymphoma (SLL) [NCT01754870]Phase 20 participants (Actual)Interventional2013-11-30Withdrawn(stopped due to slow accrual)
A Phase II, Single-Arm, Open-Label, Multicentre Study to Evaluate the Safety and Efficacy of Tafasitamab Combined With Lenalidomide in Patients With Relapsed or Refractory Diffuse Large B-Cell Lymphoma [NCT05552937]Phase 250 participants (Anticipated)Interventional2021-09-06Recruiting
Lenalidomide in Conjunction With Methotrexate, Leucovorin, Cytarabine and Rituximab for the Treatment of Relapsed or Refractory CD20-positive Aggressive Lymphomas: an Open-label, Multicenter Phase I/II Trial [NCT01788189]Phase 1/Phase 237 participants (Actual)Interventional2013-01-31Completed
An Open-label Phase II Study to Determine the Efficacy and Safety of Lenalidomide Plus Dexamethasone (LDex) in Patients With Newly Diagnosed POEMS Syndrome [NCT01816620]Phase 241 participants (Actual)Interventional2014-03-31Completed
A Pilot Trial of a WT1 Analog Peptide Vaccine in Patients With Multiple Myeloma Following Autologous Stem Cell Transplantation [NCT01827137]22 participants (Actual)Interventional2013-04-30Active, not recruiting
Continued, Long-Term Follow-Up and Lenalidomide Maintenance Therapy for Patients on BMT CTN 0702 Protocol (BMT CTN #07LT) [NCT02322320]Phase 3273 participants (Actual)Interventional2015-03-31Completed
An Open-Label Study to Determine the Maximum Tolerated Dose and Evaluate the Safety and Efficacy of CEP-18770 in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT01348919]Phase 1/Phase 211 participants (Actual)Interventional2011-08-03Completed
Impact of First Autologous Transplant on Minimal Residual Disease Markers in Previously Untreated Myeloma Undergoing Initial Treatment With Velcade Based Therapy [NCT01215344]Phase 236 participants (Actual)Interventional2010-11-30Completed
Multi-Center Phase Ib/II Trial of Gemcitabine, Cisplatin, Plus Lenalidomide as First-line Therapy for Patients With Metastatic Urothelial Carcinoma [NCT01342172]Phase 1/Phase 29 participants (Actual)Interventional2011-03-31Terminated(stopped due to low accrual)
Phase II Trial of High Dose Lenalidomide in Patients With Myelodysplastic Syndrome Refractory to Hypomethylating Agents [NCT01246076]Phase 224 participants (Actual)Interventional2011-06-30Completed
A Phase 2 Trial of Daily Alternating Thalidomide and Lenalidomide Plus Rituximab (ThRiL) for Patients With Previously Treated Waldenstrom Macroglobulinemia [NCT01779167]Phase 24 participants (Actual)Interventional2012-06-30Terminated(stopped due to Slow Accrual)
Phase I-II Multicenter Study to Assess the Efficacy and Safety of the Chlorambucil + Lenalidomide Combination and Lenalidomide Maintenance Therapy in Untreated Elderly Pts With CLL. EudraCT Number 2009-013415-35 [NCT01403246]Phase 1/Phase 29 participants (Actual)Interventional2011-11-30Terminated(stopped due to Slow patient enrollment and new molecules for chronic lymphoid leukemia, have importantly reduced the interest of conducting the phase II of this study.)
A Phase 2, Multicenter, Single-arm, Open-label Study to Evaluate the Safety and Efficacy of Lenalidomide in Patients With Relapsed or Recurrent Adult T-cell Leukemia-lymphoma [NCT01724177]Phase 226 participants (Actual)Interventional2012-11-12Completed
A Phase 1/2a, Open-Label, Multicentre, Dose-Escalation Study to Evaluate the Safety and Preliminary Efficacy of the Human Anti-CD 38 Antibody MOR03087 as Monotherapy and in Combination With Standard Therapy in Subjects With Relapsed/Refractory Multiple My [NCT01421186]Phase 1/Phase 291 participants (Actual)Interventional2011-07-31Completed
A Phase I Clinical Trial to Evaluate the Maximally Tolerated Dose (MTD), Dose Limiting Toxicities (DLTs) and Safety Profiles of Increasing Doses of Lenalidomide After Allo-HCT in AML and MDS Subjects With Minimal Residual Disease (MRD) Detected by the CD3 [NCT02370888]Phase 111 participants (Actual)Interventional2016-05-16Terminated(stopped due to Slow accrual)
Nelfinavir and Lenalidomide/Dexamethasone in Patients With Progressive Multiple Myeloma That Have Failed Lenalidomide-containing Therapy - A Single Arm Phase I/II Trial [NCT01555281]Phase 1/Phase 233 participants (Actual)Interventional2012-02-23Terminated(stopped due to In connection with the restructuring of SAKK, older ongoing studies were analyzed to determine whether a continuation of the trial would still add value to the data analysis, or whether they do not and could therefore be prematurely terminated.)
A Study of Immune-adjuvant Effect of Lenalidomide in Patients With Chronic Lymphocytic Leukemia and Hypogammaglobulinemia and Impaired Response to Vaccinations - RV-CL-CLL-PI-002544 [NCT01924169]Phase 23 participants (Actual)Interventional2014-11-24Terminated(stopped due to Slow Accrual)
A Phase II Randomized Study of Three Subcutaneous Bortezomib-based Consolidation Treatments for Patients Completing Induction Therapy and Stem Cell Transplantation for Newly Diagnosed Multiple Myeloma [NCT01706666]Phase 23 participants (Actual)Interventional2012-12-07Completed
Efficacy and Safety Study of BiRD (Biaxin [Clarithromycin]/Revlimid [Lenalidomide]/Dexamethasone) Combination Therapy in Relapsed/Refractory Myeloma [NCT02986451]Phase 20 participants (Actual)Interventional2016-12-31Withdrawn
Investigation of the Enhancement of the Response to Hepatitis B Vaccine by Lenalidomide (RevlimidTM, CC-5013) in Plasma Cell Dyscrasias [NCT02041325]Phase 238 participants (Actual)Interventional2005-04-30Completed
Short Course Daratumumab in Minimal Residual Disease (MRD) Positive Myeloma Patients After Induction Therapy With/Without Consolidative High Dose Chemotherapy/Autologous Stem Cell Support [NCT03490344]Phase 210 participants (Actual)Interventional2018-05-03Completed
MAGNETISMM-6: AN OPEN-LABEL, 2-ARM, MULTICENTER, RANDOMIZED PHASE 3 STUDY TO EVALUATE THE EFFICACY AND SAFETY OF ELRANATAMAB (PF-06863135) + DARATUMUMAB + LENALIDOMIDE VERSUS DARATUMUMAB + LENALIDOMIDE + DEXAMETHASONE IN TRANSPLANT-INELIGIBLE PARTICIPANTS [NCT05623020]Phase 3966 participants (Anticipated)Interventional2022-11-10Recruiting
A Phase I/IB Pilot Study of CC-486 Combined With Lenalidomide and Obinutuzumab for Relapsed/Refractory Indolent B-Cell Lymphoma [NCT04578600]Phase 18 participants (Actual)Interventional2020-10-23Active, not recruiting
Phase II Study of Lenalidomide in Combination With Rituximab (R) for the Treatment of Indolent Non Follicular Non Hodgkin Lymphoma (NHL). [NCT01830478]Phase 244 participants (Anticipated)Interventional2009-04-30Active, not recruiting
A Pilot Study of Thalidomide to Overcome Lenalidomide Resistance in Patients Suffering Biochemical Progression on Maintenance Therapy After Autologous Hematopoietic Stem Cell Transplantation for Multiple Myeloma [NCT01927718]10 participants (Actual)Interventional2014-01-31Terminated(stopped due to Poor Response Rate)
A Double-blind Placebo Controlled Immunogenicity Study of Vacc-4x + Lenalidomide Versus Vacc-4x With an Initial Open-label Dose Escalation Assessment of Lenalidomide in HIV-1-infected Subjects on Antiretroviral Therapy (ART). [NCT01704781]Phase 1/Phase 236 participants (Actual)Interventional2012-09-30Completed
Multi-center Randomized Study to Compare Efficacy and Safety of Lenalidomide Plus CHOP (L-CHOP) Versus CHOP in Patients With Previously Untreated Peripheral T-cell Lymphoma [NCT04922567]Phase 2289 participants (Anticipated)Interventional2021-04-01Recruiting
Phase I/Ib Trial of the Efficacy and Safety of Combination Therapy of Lenalidomide/Bortezomib/Dexamethasone and Panobinostat in Transplant Eligible Patients With Newly Diagnosed Multiple Myeloma (MM) [NCT01440582]Phase 177 participants (Actual)Interventional2013-02-18Completed
Exploratory Study of Ixazomib in Combination With Reduced Dose Lenalidomide Versus Ixazomib Alone for Maintenance Treatment of High Risk Multiple Myeloma [NCT05722405]Phase 4100 participants (Anticipated)Interventional2022-07-01Recruiting
A Phase I/II Study of Carfilzomib, Lenalidomide, Dexamethasone and the Anti-B-Cell Maturation Antigen (BCMA) Antibody Drug Conjugate Belantamab Mafodotin in Multiple Myeloma [NCT04822337]Phase 1/Phase 270 participants (Anticipated)Interventional2021-05-19Recruiting
Continuing Treatment for Subjects Who Have Participated in a Prior Protocol Investigating Elotuzumab [NCT02719613]Phase 267 participants (Actual)Interventional2016-07-15Active, not recruiting
Phase 2, Randomized, Open-Label Study Comparing Daratumumab, Lenalidomide, Bortezomib, and Dexamethasone (D-RVd) Versus Lenalidomide, Bortezomib, and Dexamethasone (RVd) in Subjects With Newly Diagnosed Multiple Myeloma Eligible for High-Dose Chemotherapy [NCT02874742]Phase 2224 participants (Actual)Interventional2016-08-29Completed
Phase II Study of Lenalidomide and Eltrombopag in Patients With Symptomatic Anemia in Low or Intermediate I Myelodysplastic Syndrome (MDS) [NCT01772420]Phase 252 participants (Actual)Interventional2012-10-31Completed
European Myeloma Network Sequential Phase I / Phase II Trial on RIC Allogeneic Transplantation: an Optimized Program for High Risk Relapsed Patients [NCT01460420]Phase 1/Phase 249 participants (Actual)Interventional2011-11-30Completed
A Phase 2 Clinical Trial To Evaluate Lenalidomide And Obinutuzumab For The Treatment Of Patients With Not Previously Treated Chronic Lymphocytic Leukemia [NCT02371590]Phase 20 participants (Actual)Interventional2018-02-28Withdrawn(stopped due to Study Not Activated Due to Contract Issues)
A Phase II Pilot Study to Determine Efficacy and Safety of Lenalidomide (Revlimid) for Treatment of Autistic Spectrum Disorders(ASD) With Regression and Markers of Cerebrospinal Fluid Cytokine Elevation and Elevated TNF-alpha Levels [NCT00996931]Phase 26 participants (Actual)Interventional2009-02-28Completed
Phase II Study of Lenalidomide and Darbepoetin Alfa in Myelodysplastic Syndrome (Low to Intermediate-1 Risk Category Excluding 5q Deletion) [NCT01222195]Phase 21 participants (Actual)Interventional2008-02-29Terminated(stopped due to Low accrual.)
A Phase II, Prospective, Single-center Study of Lenalidomide in Combination With CHOP in Patients With Untreated PTCL [NCT04423926]Phase 1/Phase 291 participants (Anticipated)Interventional2020-06-10Recruiting
Phase II Study of Revlimid (Lenalidomide), Melphalan, and Dexamethasone (ReMeDex) for Newly Diagnosed Multiple Myeloma Patients Not Undergoing Autologous Transplantation [NCT00843310]Phase 28 participants (Actual)Interventional2008-11-30Terminated(stopped due to Due to slow accrual)
Impact of Short Term Lenalidomide on Immune Response to Prevnar 13® in Individuals With Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic Leukemia (SLL), and Monoclonal B Cell Lymphocytosis (MBL) [NCT02309515]Phase 212 participants (Actual)Interventional2015-06-11Terminated(stopped due to Slow Accrual)
A Post-authorization, Non-interventional, Safety Study Study of Patients With Myelodysplastic Syndromes (MDS) Treated With Lenalidomide. [NCT02279654]389 participants (Actual)Observational [Patient Registry]2014-12-17Completed
A Randomized Phase 2 Trial of Lenalidomide/ Dexamethasone/ Elotuzumab +/- Cyclophosphamide Followed by Lenalidomide/ Dexamethasone/Elotuzumab Maintenance as Second-Line Therapy for Patients With Relapsed AL Amyloidosis [NCT03252600]Phase 253 participants (Anticipated)Interventional2017-08-25Active, not recruiting
Combination of Lenalidomide and Ofatumumab in Patients With Previously Treated Chronic Lymphocytic Leukemia and Small Lymphocytic Lymphoma (CLL/SLL) [NCT01002755]Phase 236 participants (Actual)Interventional2010-01-19Completed
A Phase I/II Clinical Trial of Lenalidomide in Combination With AT-101 for the Treatment of Relapsed B-Cell Chronic Lymphocytic Leukemia (B-CLL) [NCT01003769]Phase 1/Phase 25 participants (Actual)Interventional2015-07-09Terminated(stopped due to Lack of funding)
A Phase I/II, Multicenter, Open-label, Dose-escalation Study of Bendamustine in Combination With Lenalidomide and Dexamethasone in Patients With Relapsed Multiple Myeloma [NCT01049945]Phase 1/Phase 270 participants (Actual)Interventional2010-02-28Completed
A Multi-center Phase III Randomized Study Comparing Continuous Versus Fixed Duration Therapy With Daratumumab, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma [NCT03836014]Phase 3436 participants (Actual)Interventional2019-07-25Active, not recruiting
Chidamide Combination With Lenalidomide in Patients With Relapsed or Refractory Peripheral T-cell Lymphoma: an Open Label, Single Arm, Phase II Study [NCT04329130]Phase 244 participants (Anticipated)Interventional2020-03-27Recruiting
A Phase I Trial of the Combination of Lenalidomide and Blinatumomab in Patients With Relapsed or Refractory Non-Hodgkins Lymphoma (NHL) [NCT02568553]Phase 144 participants (Anticipated)Interventional2016-11-15Active, not recruiting
Induction Therapy With Bortezomib-melphalan and Prednisone (VMP) Followed by Lenalidomide and Dexamethasone (Rd) Versus Carfilzomib, Lenalidomide and Dexamethasone (KRd) Plus/Minus Daratumumab, 18 Cycles, Followed by Consolidation and Maintenance Therapy [NCT03742297]Phase 3462 participants (Actual)Interventional2018-10-22Active, not recruiting
A Study of Lenalidomide in Patients With Chronic Lymphocytic Leukemia and Residual Disease After Chemotherapy - RV-CLL-PI-0270 [NCT00632359]Phase 233 participants (Actual)Interventional2008-02-29Completed
A Phase I/II Study of Lenalidomide Maintenance After Autologous Stem Cell Transplant for Elderly Patients With Acute Myeloid Leukemia (AML) [NCT02038153]Phase 1/Phase 23 participants (Actual)Interventional2013-12-31Terminated(stopped due to Low accrual)
Multicenter, Randomized, Double-blind, Phase III Study of REVLIMID (Lenalidomide) Versus Placebo in Patients With Low Risk Myelodysplastic Syndrome (Low and Intermediate-1 IPSS) With Alteration in 5q- and Anemia Without the Need of Transfusion. [NCT01243476]Phase 361 participants (Actual)Interventional2010-01-31Completed
A Phase I/II Open Label, Single Center, Study of the Combination of ALX148, Rituximab and Lenalidomide in Patients With Indolent and Aggressive B-Cell Non-Hodgkin Lymphoma [NCT05025800]Phase 1/Phase 247 participants (Anticipated)Interventional2021-10-13Recruiting
A Phase 3, Randomized, Open-label, Parallel-controlled, Multi-center Study Comparing TJ202, Lenalidomide and Dexamethasone vs. Lenalidomide and Dexamethasone in Subjects With Relapsed or Refractory Multiple Myeloma Who Received at Least 1 Prior Line of Tr [NCT03952091]Phase 3291 participants (Actual)Interventional2019-03-27Active, not recruiting
Rituximab Plus Lenalidomide or Rituximab Monotherapy for Untreated Patients With Follicular Lymphoma in Need of Therapy. A Randomized, Open-Label, Multicenter Phase II Trial. [NCT01307605]Phase 2154 participants (Actual)Interventional2011-02-09Terminated(stopped due to It was decided by the trial team to stop the collection of follow up data in trial SAKK 35/10.)
A Phase II Study of Busulfan & Melphalan as Conditioning Regimen for ASCT in Patients Who Received Bortezomib Based Induction for Newly Diagnosed Multiple Myeloma Followed by Lenalidomide Maintenance Until Progression. [NCT01702831]Phase 278 participants (Actual)Interventional2013-10-01Completed
A Pilot Study to Establish the Safety & Efficacy of a Combination of Dexamethasone & Lenalidomide in Patients With Relapsed/Refractory Chronic Lymphocytic Leukaemia (CLL) [NCT01459211]Phase 212 participants (Actual)Interventional2012-05-31Completed
A Phase II Study Investigating Treatment of Post-Allogeneic Transplant Progression or Relapse of CLL/SLL/PLL or NHL With Lenalidomide Alone or With Rituximab [NCT01419795]Phase 23 participants (Actual)Interventional2012-05-31Terminated(stopped due to Low accrual)
Phase I Multi-site Study Evaluating the MTD, Safety and Efficacy of the Combination Venetoclax, Lenalidomide and Rituximab in Patients With Previously Untreated Mantle Cell Lymphoma [NCT03523975]Phase 130 participants (Actual)Interventional2018-12-24Active, not recruiting
Open-label, Multicenter Phase I/II Study: Salvage Therapy of Progressive and Relapsed Aggressive Non-Hodgkin-Lymphoma by Combination of Lenalidomide (Revlimid®) With Rituximab, Dexamethason, High-dose ARA-C and Cisplatinum (R²-DHAP) [NCT02983097]Phase 1/Phase 234 participants (Actual)Interventional2010-11-30Terminated(stopped due to Phase II: no scientific interests are given anymore)
Lenalidomide (Revlimid) in Patients With Previously Treated Chronic Lymphocytic Leukemia [NCT00267059]Phase 245 participants (Actual)Interventional2005-12-31Completed
Azacitidine Plus Lenalidomide Combination in Elderly Patients With Previously Treated Acute Myeloid Leukemia (AML) & High-Risk Myelodysplastic Syndromes (MDS) (VIREL2 Trial) [NCT01442714]Phase 233 participants (Actual)Interventional2011-08-31Terminated(stopped due to Lack of efficacy - Inability to meet the primary response endpoint)
[NCT02406144]Phase 3316 participants (Actual)Interventional2014-11-30Completed
A Phase II Study of Modified Lenalidomide, Bortezomib and Dexamethasone for Transplant-Ineligible Patients With Newly Diagnosed Multiple Myeloma [NCT01782963]Phase 250 participants (Actual)Interventional2013-03-31Completed
Front-line Therapy With Carfilzomib, Lenalidomide, and Dexamethasone (CRd) Induction Followed by Autologous Stem Cell Transplantation, CRd Consolidation and Lenalidomide Maintenance in Newly Diagnosed Multiple Myeloma Patients ≤65 Years Old [NCT02405364]Phase 246 participants (Actual)Interventional2014-02-28Active, not recruiting
Randomized Phase 2 Trial of Retreatment With Pomalidomide or Lenalidomide With Dexamethasone for Patients With Relapsed Myeloma [NCT01794039]Phase 29 participants (Actual)Interventional2014-03-31Completed
ARIA: A Phase 1b/2, Open-label, Multi Cohort Trial of Tazemetostat in Combination With Various Treatments in Subjects With Relapsed or Refractory Hematologic Malignancies [NCT05205252]Phase 1/Phase 20 participants (Actual)Interventional2021-12-22Withdrawn(stopped due to Epizyme Inc. has revised the Tazemetostat development strategy and made the decision to terminate the hematological malignancies basket trial.)
Lenalidomide Combined With Anti-CD20 Monoclonal Antibodies-CHOP in Untreated Diffuse Large B-Cell Lymphoma Patients With MYC and BCL2 Co-expression : An Open-lable,Multicenter,Phase II Study [NCT04842487]Phase 280 participants (Anticipated)Interventional2021-04-10Not yet recruiting
Tislelizumab in Combination With Lenalidomide in in Patients With Relapsed or Refractory Elderly Patients With Non-GCB Diffuse Large B Cell Lymphoma: a Prospective Phase Ib/II, Multicentre, Open-label, Single-arm Trial [NCT04796857]Phase 1/Phase 230 participants (Anticipated)Interventional2021-03-31Recruiting
A Phase III Study of Lenalidomide and Low-Dose Dexamethasone With or Without Pembrolizumab (MK3475) in Newly Diagnosed and Treatment Naïve Multiple Myeloma (KEYNOTE 185). [NCT02579863]Phase 3310 participants (Actual)Interventional2015-10-19Terminated(stopped due to The study was terminated early due to business reasons)
A Phase I/II Study of Tafasitamab Plus Lenalidomide in Relapsed CNS Lymphoma [NCT05351593]Phase 1/Phase 235 participants (Anticipated)Interventional2022-06-08Recruiting
Phase 1b Study of Carfilzomib Administered Once Weekly in Combination With Lenalidomide and Dexamethasone in Subjects With Multiple Myeloma [NCT02335983]Phase 1107 participants (Actual)Interventional2015-04-30Completed
Randomized Phase II Study of Bortezomib, Lenalidomide and Dexamethasone Versus Lenalidomide and Dexamethasone in Elderly Patients With Newly Diagnosed Multiple Myeloma [NCT04277845]Phase 249 participants (Actual)Interventional2020-08-06Active, not recruiting
Safety and Efficacy of Lenalidomide in the Treatment of Refractory Cutaneous Dermatomyositis [NCT05488327]10 participants (Anticipated)Interventional2022-08-01Not yet recruiting
Id and Rd Maintenance Regimens After Induction of Remission in Multiple Myeloma: a Prospective, Randomized, Controlled, Multicenter Clinical Study [NCT05477797]420 participants (Anticipated)Interventional2023-02-15Not yet recruiting
LOC-R01: Randomized Phase IB/II Study of Escalating Doses of Lenalidomide and Ibrutinib in Association With R-MPV as a Targeted Induction Treatment for Patients Aged 18 to 60 (up to 65 for Phase II) With a Newly Diagnosed Primary Central Nervous System Ly [NCT04446962]Phase 1/Phase 2118 participants (Anticipated)Interventional2020-10-30Recruiting
Phase II Study Of Durvalumab In Combination With Lenalidomide In Relapsed/Refractory EBV Associated Subtypes Of DLBCL, Primary CNS Lymphoma And Primary Testicular DLBCL - DuRIANS (Durvalumab Revlimid In Aggressive NHL Subtypes) [NCT03212807]Phase 20 participants (Actual)Interventional2017-08-31Withdrawn(stopped due to FDA Hold for Combination Studies using Imids and PD1/PDL1 Compounds)
A Phase II Study of Lenalidomide, Ixazomib, Dexamethasone, and Daratumumab in Transplant-Ineligible Patients With Newly Diagnosed Multiple Myeloma [NCT04009109]Phase 2188 participants (Anticipated)Interventional2020-10-21Recruiting
A phaseI/II Safety and Efficacy Trial of a Combination of Bendamustine, Rituximab and Lenalidomide (BRL) in Patients With Relapsed or Refractory Chronic Lymphocytic Leukemia [NCT01558167]Phase 1/Phase 222 participants (Actual)Interventional2011-02-28Completed
A Multi-center, Open-Label Phase II Study to Determine the Efficacy and Safety of Lenalidomide Plus Low-Dose Dexamethasone in Chinese Subjects With Relapsed/Refractory Multiple Myeloma [NCT01593410]Phase 2194 participants (Actual)Interventional2010-08-01Completed
MUK Nine b: OPTIMUM. A Phase II Study Evaluating Optimised Combination of Biological Therapy in Newly Diagnosed High Risk Multiple Myeloma and Plasma Cell Leukaemia. [NCT03188172]Phase 295 participants (Anticipated)Interventional2017-09-28Active, not recruiting
A Multicenter, Prospective, Observational Clinical Protocol for Chidamide in Combination With Rituximab and Lenalidomide (cR2) in Real-world Practice in Patients With Relapsed/Refractory Diffuse Large B-cell Lymphoma [NCT05690191]169 participants (Anticipated)Observational2021-06-01Enrolling by invitation
Phase I Trial of High Dose Lenalidomide in Patients With Refractory/Relapsed Acute Leukemia as a Bridge to Bone Marrow Transplant [NCT01615042]Phase 16 participants (Actual)Interventional2012-08-31Terminated(stopped due to Lack of enrollment)
Autologous Peripheral Blood Stem Cell Transplantation and Maintenance Lenalidomide After High-dose Melphalan for Multiple Myeloma [NCT01617213]Phase 21 participants (Actual)Interventional2012-04-30Terminated(stopped due to Study is no longer needed as recent data have answered the primary hypotheses for this study.)
An Open-label, Multicenter,Randomized Phase 2 Trial to Compare the Efficacy and Safety of Ibrutinib Versus Lenalidomide in Combination With MRE(Methotrexate,Rituximab,Etoposide)-Chemotherapy for Adult Patients With Recurrent/Refractory Primary Central Ner [NCT04129710]Phase 2120 participants (Anticipated)Interventional2020-01-01Recruiting
R2-MTX Regimen Combined With Lenalidomide Maintenance as Frontline Therapy for Primary Central Nervous System Lymphoma: a Multicenter Prospective Single Arm Trial. [NCT04120350]Phase 1/Phase 247 participants (Anticipated)Interventional2019-08-16Recruiting
A Phase II Trial of Revlimid® (Lenalidomide) and Low Dose Vidaza® (Azacitidine) in Patients With Low - Intermediate-1 Risk Myelodysplastic Syndromes [NCT01379274]Phase 22 participants (Actual)Interventional2011-01-31Terminated(stopped due to Loss of funding.)
Phase III Trial Comparing Dexamethasone (DEX) to the Combination of DEX + CC-5013 in Patients With Previously Untreated Multiple Myeloma [NCT00064038]Phase 3198 participants (Actual)Interventional2004-11-30Completed
Phase II Study of CC-5013 in Myelofibrosis [NCT00087672]Phase 241 participants (Actual)Interventional2004-07-31Completed
A Phase II Trial of the Immunomodulatory Drug CC-5013 for Patients With AL Amyloidosis [NCT00091260]Phase 282 participants (Actual)Interventional2004-01-31Completed
Evaluation of Lenalidomide (CC-5013) and Prednisone as a Therapy for Patients With Myelofibrosis (MF) [NCT00352794]Phase 240 participants (Actual)Interventional2006-07-07Completed
Phase II Study of Lenalidomide in Patients With Relapsed/Refractory Acute Myelogenous Leukemia or High-Risk Myelodysplastic Syndrome Associated With Chromosome 5 Abnormalities [NCT00360672]Phase 227 participants (Actual)Interventional2009-01-31Completed
A Phase II Clinical Trial of Lenalidomide for T-cell Non-Hodgkin's Lymphoma [NCT00322985]Phase 240 participants (Actual)Interventional2006-06-30Completed
A Controlled, Parallel-Group, Randomized, Open-Label Study to Evaluate Two Lenalidomide Dose Regimens When Used in Combination With Low Dose Dexamethasone for the Treatment of Subjects With Relapsed Multiple Myeloma [NCT01380106]Phase 233 participants (Actual)Interventional2010-08-31Completed
Phase II Study of Ofatumumab in Combination With High Dose Methylprednisolone Followed by Ofatumumab and Lenalidomide Consolidative Therapy for the Treatment of Relapsed or Refractory CLL/SLL The HiLOG Trial [NCT01497496]Phase 229 participants (Actual)Interventional2012-01-26Completed
A Phase 2, Multicenter, Single-arm, Open-label Study to Evaluate the Activity, Safety and Pharmacokinetics of Lenalidomide (Revlimid®) in Pediatric Subjects From 1 to = 18 Years of Age With Relapsed or Refractory Acute Myeloid Leukemia. [NCT02538965]Phase 217 participants (Actual)Interventional2015-11-19Completed
Lenalidomide Combined With Vorinostat/Gemcitabine/Busulfan/Melphalan With Autologous Stem-Cell Transplantation in Diffuse Large B-Cell Lymphoma of the ABC Subtype [NCT02589145]Phase 1/Phase 28 participants (Actual)Interventional2016-06-22Terminated(stopped due to Closed due to very slow accrual)
Isatuximab in Combination With Bortezomib and Lenalidomide With Minimal Dexamethasone in Transplant-ineligible Multiple Myeloma [NCT04939844]Phase 251 participants (Actual)Interventional2021-06-29Active, not recruiting
A Phase II, Single-Center, Open-Label Study Of Oral Panobinostat (LBH589) When Administered In Combination With Lenalidomide And Weekly Dexamethasone In Patients With Multiple Myeloma [NCT01651039]Phase 232 participants (Actual)Interventional2012-07-31Completed
A Phase I/II Study of the PD-1 Antibody Nivolumab in Combination With Lenalidomide in Relapsed/Refractory Non-Hodgkin's Lymphoma (NHL) and Hodgkin's Disease (HD) [NCT03015896]Phase 1/Phase 236 participants (Actual)Interventional2017-02-14Active, not recruiting
A Phase 2, Randomized, Open Label Trial of Lenalidomide/Dexamethasone With or Without Elotuzumab in Subjects With Previously Untreated Multiple Myeloma in Japan [NCT02272803]Phase 282 participants (Actual)Interventional2015-02-20Completed
A Pilot Study of Lenalidomide as a Chemopreventive Agent for Patients With High-Risk, Early Stage B-Chronic Lymphocytic Leukemia (CLL) [NCT01649791]8 participants (Actual)Interventional2010-01-31Terminated
A Phase I Multi-Cohort Trial of Pembrolizumab (MK-3475) in Combination With Backbone Treatments for Subjects With Multiple Myeloma [NCT02036502]Phase 177 participants (Actual)Interventional2014-02-14Terminated(stopped due to Business Reasons)
Phase 2 Multi-center Study of Anti-Programmed-Death-1 [Anti-PD-1] During Lymphopenic State After High-Dose Chemotherapy and Autologous Hematopoietic Stem Cell Transplant [HDT/ASCT] for Multiple Myeloma [NCT02331368]Phase 232 participants (Actual)Interventional2015-06-30Completed
Phase I/II Trial of the Combination of Lenalidomide (Revlimid) and Nab-paclitaxel (Abraxane) in the Treatment of Relapsed/Refractory Multiple Myeloma [NCT02075021]Phase 1/Phase 23 participants (Actual)Interventional2014-03-31Terminated(stopped due to PI left the institution)
A Phase 3, Single-Arm, Open-Label, Multicenter Study to Evaluate the Safety and Efficacy of Tafasitamab Plus Lenalidomide in Participants With Relapsed or Refractory Diffuse Large B-Cell Lymphoma [NCT05429268]Phase 381 participants (Anticipated)Interventional2022-12-23Recruiting
A Phase 1/2, Dose and Schedule Evaluation Study to Investigate the Safety and Clinical Activity of Belantamab Mafodotin Administered in Combination With Daratumumab, Lenalidomide and Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma Transpla [NCT05280275]Phase 1/Phase 236 participants (Anticipated)Interventional2022-04-13Recruiting
Phase 2 Study of Tafasitamab and Lenalidomide in Relapsed or Refractory Mantle Cell Lymphoma [NCT05788289]Phase 239 participants (Anticipated)Interventional2023-03-14Recruiting
Sequential Treatment With Azacitidine and Lenalidomide for Relapsed and Refractory Patients With Acute Myeloid Leukemia [NCT01743859]Phase 237 participants (Actual)Interventional2012-12-06Completed
A Global Multicenter Phase 1/2 Trial of EO2463, a Novel Microbial-Derived Peptide Therapeutic Vaccine, as Monotherapy, and in Combination With Lenalidomide and Rituximab, for Treatment of Patients With Indolent Non-Hodgkin's Lymphoma [NCT04669171]Phase 1/Phase 260 participants (Anticipated)Interventional2021-07-05Recruiting
A Phase 3, Double-blind, Randomized Study to Compare the Efficacy and Safety of Rituximab Plus Lenalidomide (CC-5013) Versus Rituximab Plus Placebo in Subjects With Relapsed/Refractory Indolent Lymphoma [NCT01938001]Phase 3358 participants (Actual)Interventional2013-11-21Completed
An Open Label, Single Arm, Multi-Center Exploratory Study to Evaluate the Efficacy and Safety of SVRd for the Treatment of Newly Diagnosed Multiple Myeloma Patients Presenting With Extramedullary Disease. [NCT05900882]Phase 235 participants (Anticipated)Interventional2022-07-15Recruiting
Randomized Phase 1 / 2 Trial of Belantamab Mafodotin, Lenalidomide, and Daratumumab in Relapsed or Newly Diagnosed Multiple Myeloma Patients [NCT04892264]Phase 1/Phase 25 participants (Actual)Interventional2021-06-03Active, not recruiting
A Phase 3 Open-Label Randomized Study to Compare the Efficacy and Safety of Rituximab Plus Lenalidomide (CC-5013) Versus Rituximab Plus Chemotherapy in Subjects With Previously Untreated Follicular Lymphoma [NCT01476787]Phase 3255 participants (Actual)Interventional2011-12-29Active, not recruiting
A Phase 1/2, Open-Label, Dose-Escalation, Safety and Tolerability Study of INCB052793 in Subjects With Advanced Malignancies [NCT02265510]Phase 1/Phase 283 participants (Actual)Interventional2014-09-10Terminated
Mechanism Investigation of Selinexor Combined With Lenalidomide and Rituximab in the Treatment of Diffuse Large B-cell Lymphoma [NCT05923879]30 participants (Anticipated)Observational2023-06-30Not yet recruiting
A RANDOMIZED, MULTICENTER, OPEN LABEL STUDY COMPARING TWO STANDARD TREATMENTS, BORTEZOMIB-MELPHALAN-PREDNISONE (VMP) WITH OR WITHOUT DARATUMUMAB (Dara-VMP) VS LENALIDOMIDE-DEXAMETHASONE (Rd) WITH OR WITHOUT DARATUMUMAB (Dara-Rd) IN AUTOLOGOUS STEM CELL TR [NCT03829371]Phase 4450 participants (Anticipated)Interventional2019-01-03Recruiting
A Phase I Study of Obinutuzumab, Venetoclax, and Lenalidomide in Relapsed and Refractory B-cell Non-Hodgkin Lymphoma [NCT02992522]Phase 122 participants (Actual)Interventional2017-02-21Active, not recruiting
A PHASE III, MULTICENTRE, RANDOMIZED, CONTROLLED STUDY TO DETERMINE THE EFFICACY AND SAFETY OF STANDARD SCHEDULE VERSUS A NEW ALGORITHM OF DOSE REDUCTIONS IN ELDERLY AND UNFIT NEWLY DIAGNOSED MULTIPLE MYELOMA PATIENTS RECEIVING LENALIDOMIDE PLUS STEROIDS [NCT02215980]Phase 3210 participants (Actual)Interventional2014-07-31Active, not recruiting
A 3-Arm Randomized Phase II Trial of Bendamustine-Rituximab (BR) Followed by Rituximab vs Bortezomib-BR (BVR) Followed by Rituximab vs BR Followed by Lenalidomide/Rituximab in High Risk Follicular Lymphoma [NCT01216683]Phase 2289 participants (Actual)Interventional2011-02-09Completed
A Phase II Trial of Lenalidomide (Revlimid (TM), CC-5013) (NSC #703813) Plus Rituximab in Previously Untreated Follicular Non-Hodgkin Lymphoma (NHL) [NCT01145495]Phase 266 participants (Actual)Interventional2010-06-15Completed
A PHASE 3, MULTICENTRE, RANDOMIZED, CONTROLLED STUDY TO DETERMINE THE EFFICACY AND SAFETY OF LENALIDOMIDE, MELPHALAN AND PREDNISONE (MPR) Versus MELPHALAN (200 mg/m2) FOLLOWED BY STEM CELL TRANSPLANT IN NEWLY DIAGNOSED MULTIPLE MYELOMA SUBJECTS [NCT00551928]Phase 3402 participants (Actual)Interventional2007-06-30Active, not recruiting
Phase I/II Double Blind Randomized Trial of Lenalidomide/Dexamethasone/Anakinra vs. Lenalidomide/Dexamethasone/Placebo in Patients With Early Stage Multiple Myeloma and High Plasma Cell Growth Rate [NCT02492750]Phase 114 participants (Actual)Interventional2016-04-30Completed
Immunotherapy With Ex Vivo-Expanded Cord Blood-Derived NK Cells Combined With Rituximab High-Dose Chemotherapy and Autologous Stem Cell Transplant for B-Cell Non-Hodgkin's Lymphoma [NCT03019640]Phase 222 participants (Actual)Interventional2017-10-10Completed
Obinutuzumab, High Dose Methylprednisolone (HDMP), and Lenalidomide for the Treatment of Patients With Richter's Syndrome [NCT03113695]Phase 14 participants (Actual)Interventional2017-12-20Completed
VIRel: Viral Immunotherapy in Relapsed/Refractory Multiple Myeloma - A Phase I Study to Assess the Safety and Tolerability of REOLYSIN® (Pelareorep) in Combination With Lenalidomide or Pomalidomide [NCT03015922]Phase 14 participants (Actual)Interventional2017-06-05Active, not recruiting
Allogeneic Stem Cell Transplantation vs. Conventional Therapy as Salvage Therapy for Relapsed / Progressive Patients With Multiple Myeloma After First-line Therapy [NCT05675319]Phase 3482 participants (Anticipated)Interventional2023-03-03Recruiting
Evaluating Mechanisms of Immunomodulator Sensitivity and Resistance in Multiple Myeloma [NCT05288062]Phase 2190 participants (Anticipated)Interventional2022-03-22Recruiting
Phase 2 Study of Mitoxantrone, Etoposide, and Cytarabine (MEC) Plus Lenalidomide for the Treatment of Adult Patients With Relapsed or Refractory Acute Myeloid Leukemia [NCT03118466]Phase 241 participants (Actual)Interventional2017-09-25Active, not recruiting
Phase II Study of Lenalidomide in Combination With Nivolumab In Patients With Relapsed/Refractory Multiple Myeloma [NCT03333746]Phase 21 participants (Actual)Interventional2018-03-21Terminated(stopped due to Study was discontinued due to FDA recommendations of the potential toxicities of the combination of drugs.)
A Phase Ib/IIa Clinical Study of IMM0306 fo r Injection in Combination With Lenalidomide for the Treatment of Relapsed/Refractory CD20-Positive B-Cell Non-Hodgkin's Lymphoma Cell Non-Hodgkin's Lymphoma [NCT05771883]Phase 1/Phase 2102 participants (Anticipated)Interventional2023-05-31Not yet recruiting
National, Open-label, Multicentre Phase I-II Study of Combination R-ESHAP With Lenalidomide as Salvage Therapy for Patients With Relapsed or Refractory Diffuse Large B-cell Lymphoma Candidates to Stem-cell Transplantation [NCT02340936]Phase 1/Phase 253 participants (Actual)Interventional2011-01-31Completed
A Phase II Study of Once Weekly Carfilzomib, Lenalidomide, Dexamethasone, and Isatuximab in Newly Diagnosed, Transplant-Eligible Multiple Myeloma [NCT04430894]Phase 250 participants (Actual)Interventional2020-07-10Active, not recruiting
A Phase II Trial of Daratumumab, Lenalidomide and Dexamethasone (DRd) in Combination With Selinexor for Patients With Newly Diagnosed Multiple Myeloma [NCT04782687]Phase 2100 participants (Anticipated)Interventional2021-09-10Recruiting
Rituximab,Zanubrutinib in Combination With Lenalidomide, Followed by Zanubrutinib or Lenalidomide Maintenance in Patients With Primary or Secondary CNS Lymphoma [NCT04938297]Phase 2100 participants (Anticipated)Interventional2021-05-26Recruiting
Lenalidomide and Dexamethasone for Rosai-Dorfman Disease: A Single Arm, Single Center, Prospective Phase 2 Study [NCT04924647]Phase 216 participants (Anticipated)Interventional2021-06-08Recruiting
A Phase II Study of Ibrutinib Plus Rituximab and Lenalidomide in Elderly Patients With Newly Diagnosed MCL [NCT03232307]Phase 20 participants (Actual)Interventional2019-07-01Withdrawn(stopped due to Sponsor does not wish to proceed)
GEM21menos65. A Phase III Trial for NDMM Patients Who Are Candidates for ASCT Comparing Extended VRD Plus Early Rescue Intervention vs Isatuximab-VRD vs Isatuximab-V-Iberdomide-D [NCT05558319]Phase 3480 participants (Anticipated)Interventional2022-10-31Not yet recruiting
A Phase 1a/1b Multicenter, Open Label, Dose-Finding Study to Assess the Safety, Tolerability, Pharmacokinetics and Efficacy of CWP232291 Administered Intravenously Either Alone or in Combination With Lenalidomide and Dexamethasone in Subjects With Relapse [NCT02426723]Phase 125 participants (Actual)Interventional2015-10-19Completed
A Phase 2 Clinical Trial To Evaluate The Immune Restoration Potential Of Lenalidomide For Patients With CLL-Associated Immunodeficiency [NCT02371577]Phase 20 participants (Actual)Interventional2017-02-01Withdrawn(stopped due to withdrawn)
Efficacy and Safety Study of Lenalidomide Plus Rituximab and Methotrexate Chemotherapy(R2-MTX) Versus Rituximab and Methotrexate Chemotherapy(R-MTX) in Newly Diagnosed Primary Central Nervous System Lymphoma: a Multicenter, Open-Label, Randomised Phase 2 [NCT04481815]Phase 2240 participants (Anticipated)Interventional2020-10-01Recruiting
A Phase 1b/2, Open-Label, Safety and Efficacy Study of Epcoritamab (GEN3013; DuoBody®-CD3xCD20) in Relapsed/Refractory Chronic Lymphocytic Leukemia and Richter's Syndrome [NCT04623541]Phase 1/Phase 2184 participants (Anticipated)Interventional2020-11-25Recruiting
Phase I/II Study of Lenalidomide Combined With Modified DA-EPOCH and Rituximab (EPOCH-R2) in Primary Effusion Lymphoma or KSHV-Associated Large Cell Lymphoma [NCT02911142]Phase 1/Phase 212 participants (Actual)Interventional2017-07-03Active, not recruiting
A Phase I/II Study of Lenalidomide in Patients With AIDS-Associated Kaposi's Sarcoma [NCT01057121]Phase 1/Phase 238 participants (Actual)Interventional2010-08-27Completed
Selinexor(ATG-010) Plus Bortezomib, Lenalidomide and Dexamethasone (XVRd) in High Risk Newly Diagnosed Multiple Myeloma [NCT05422027]Phase 1/Phase 242 participants (Anticipated)Interventional2022-07-25Recruiting
Administration of an Oral PDE5 Inhibitor, Tadalafil in Conjunction With Lenalidomide and Dexamethasone in Patients With Multiple Myeloma [NCT01374217]Phase 214 participants (Actual)Interventional2012-04-30Terminated(stopped due to early stopping rule)
Selinexor in Combination With Immunomodulator to Treat Relapsed/Refractory Multiple Myeloma Patients [NCT04941937]Phase 290 participants (Anticipated)Interventional2022-01-27Recruiting
A Real-world Study of the Efficacy and Safety of Obinutuzumab-based Therapy for Previously Untreated Follicular Lymphoma [NCT05899621]332 participants (Anticipated)Observational2023-06-01Recruiting
Phase I-II Single Cycle Melphalan/Total Marrow Irradiation (TMI) and Autologous Stem Cell Transplantation (ASCT) Followed by Maintenance in Patients With High-Risk Myeloma and/or Poor Response to Induction Therapy Within 12 Months of Diagnosis [NCT03100877]Phase 1/Phase 20 participants (Actual)Interventional2018-01-31Withdrawn(stopped due to Feasibility Issues)
Efficacy and Safety of Tislelizumab Combined Treatment in Refractory Natural Killer/T-cell Lymphoma [NCT05058755]62 participants (Anticipated)Interventional2021-09-17Recruiting
A Phase II Trial of Tafasitamab and Lenalidomide Followed by Tafasitamab and ICE as Salvage Therapy for Transplant Eligible Patients With Relapsed/ Refractory Large B-Cell Lymphoma [NCT05821088]Phase 237 participants (Anticipated)Interventional2023-06-29Recruiting
Effective Quadruplet Utilization After Treatment Evaluation (EQUATE): A Randomized Phase 3 Trial for Newly Diagnosed Multiple Myeloma Not Intended for Early Autologous Transplantation [NCT04566328]Phase 31,450 participants (Anticipated)Interventional2021-02-24Recruiting
Daratumumab to Enhance Therapeutic Effectiveness of Revlimid in Smoldering Myeloma (DETER-SMM) [NCT03937635]Phase 3288 participants (Anticipated)Interventional2019-09-16Recruiting
Multicenter Phase II Study to Demonstrate Clinical Benefit of Lenalidomide and Dexamethasone in Elderly Unfit Patients With Newly Diagnosed Multiple Myeloma [NCT03809780]Phase 270 participants (Actual)Interventional2019-03-11Active, not recruiting
Phase 2 Trial of Ixazomib, Lenalidomide, Dexamethasone, and Daratumumab in Patients With Newly Diagnosed Multiple Myeloma [NCT03012880]Phase 280 participants (Actual)Interventional2017-04-12Active, not recruiting
Phase I Trial of the Combination of Ibrutinib and Lenalidomide for the Treatment of Patients With MDS Who Have Failed or Refuse Standard Therapy [NCT03359460]Phase 14 participants (Actual)Interventional2017-12-01Completed
A Phase 3, Single Arm, Multi-Center Study to Assess the Efficacy and Safety of Clarithromycin(Biaxin)-Lenalidomide-Low-Dose-Dexamethasone (BiRd) Combined With B-cell Maturation Antigen (BCMA)-Directed Chimeric Antigen Receptor (CAR) T-cell Therapy in Pati [NCT04287660]Phase 320 participants (Anticipated)Interventional2017-10-19Recruiting
Lenalidomide for the Treatment of CLL Patients With High-Risk Disease [NCT01271283]Phase 20 participants (Actual)Interventional2010-12-31Withdrawn
Lenalidomide and Rituximab Treatment of Relapsed Mantle Cell Lymphoma and Diffuse Large B-Cell Non-Hodgkin's Lymphoma, Transformed Large Cell Lymphoma, and/or Grade 3 Follicular Lymphoma (Follicular Cleaved Large Cell Lymphoma or Follicular Non-Cleaved La [NCT00294632]Phase 1/Phase 254 participants (Actual)Interventional2006-02-28Completed
A Phase 2 Trial of Bevacizumab, Lenalidomide, Docetaxel, and Prednisone (ART-P) for Treatment of Metastatic Castrate-Resistant Prostate Cancer [NCT00942578]Phase 263 participants (Actual)Interventional2009-07-16Completed
A Study of Umbilical Cord Blood-Derived Natural Killer Cells in Conjunction With Lymphodepleting Chemotherapy and Lenalidomide as Immunotherapy in Patients With Hematologic Malignancies [NCT02280525]Phase 18 participants (Actual)Interventional2015-03-05Completed
A Phase 2, Multicenter, Open-label, Single Arm Study of Lenalidomide (CC-5013) in Combination With Low-dose Dexamethasone in Japanese Patients With Previously Untreated Multiple Myeloma [NCT01698801]Phase 226 participants (Actual)Interventional2012-10-01Completed
REsponse Adapted Combination Therapy Approaches for High-Risk Multiple Myeloma (REACH) [NCT05497804]Phase 275 participants (Anticipated)Interventional2022-09-22Recruiting
A Phase 3, Intergroup Multicentre, Randomized, Controlled 3 Arm Parallel Group Study to Determine the Efficacy and Safety of Lenalidomide in Combination With Dexamethasone (RD) Versus Melphalan, Prednisone and Lenalidomide (MPR) Versus Cyclophosphamide, P [NCT01093196]Phase 3660 participants (Anticipated)Interventional2009-10-31Active, not recruiting
A Phase 3, Multicentre, Randomized, Controlled Study to Determine the Efficacy and Safety of Cyclophosphamide, Lenalidomide and Dexamethasone (CRD) Versus Melphalan (200 mg/m2) Followed By Stem Cell Transplant In Newly Diagnosed Multiple Myeloma Subjects [NCT01091831]Phase 3389 participants (Actual)Interventional2009-07-31Active, not recruiting
Phase I/II Study of Lenalidomide and Gemcitabine as First-line Treatment in Patients With Locally Advanced or Metastatic Pancreatic Cancer [NCT01547260]Phase 1/Phase 234 participants (Actual)Interventional2009-10-31Completed
Phase 2 Study of Isatuximab Plus Lenalidomide and Dexamethasone in Highly Toxicity-vulnerable Subjects With Newly Diagnosed Multiple Myeloma [NCT05145400]Phase 250 participants (Anticipated)Interventional2022-02-18Recruiting
An Open-Label Phase I/II Study of the Safety and Efficacy of Bortezomib, Lenalidomide and Dexamethasone Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma [NCT00378105]Phase 1/Phase 268 participants (Actual)Interventional2006-09-30Active, not recruiting
Phase II Study of CC-5103 and Rituximab in Waldenstrom's Macroglobulinemia [NCT00142168]Phase 216 participants (Actual)Interventional2004-09-30Terminated(stopped due to Toxicity)
Phase III, Multicenter, Open Label, Randomized, Controlled Study Investigating Mosunetuzumab-Lenalidomide Versus Investigator Choices in Patients With Relapsed or Refractory Marginal Zone Lymphoma [NCT06006117]Phase 3260 participants (Anticipated)Interventional2023-09-05Recruiting
A Phase 1b/2 Study of Tafasitamab, Tafasitamab Plus Lenalidomide, Tafasitamab Plus Parsaclisib, and Tafasitamab Plus Lenalidomide in Combination With R-CHOP in Japanese Participants With Non-Hodgkin Lymphoma [NCT04661007]Phase 165 participants (Anticipated)Interventional2020-12-15Recruiting
Symphony-1: A Phase 1b/3 Double-Blind, Randomized, Active-Controlled, 3-Stage, Biomarker Adaptive Study Of Tazemetostat Or Placebo In Combination With Lenalidomide Plus Rituximab In Subjects With Relapsed/Refractory Follicular Lymphoma [NCT04224493]Phase 3540 participants (Anticipated)Interventional2019-12-19Recruiting
A Multiple-center Phase 2 Study of Acalabrutinib-Lenalidomide-Rituximab in Patients With Previous Untreated Mantle Cell Lymphoma [NCT03863184]Phase 224 participants (Anticipated)Interventional2019-10-11Recruiting
Ixazomib, Lenalidomide, and Dexamethasone for Patients With POEMS Syndrome [NCT02921893]Early Phase 121 participants (Actual)Interventional2016-10-31Active, not recruiting
A Phase II Study of Lenalidomide Following Allogeneic Stem Cell Transplant for Multiple Myeloma Patients Who Relapse or Have Disease Progression [NCT00619684]Phase 218 participants (Actual)Interventional2008-02-29Completed
Phase II Trial of Pentostatin, Cyclophosphamide, and Rituximab Followed by Consolidation With Lenalidomide for Previously Untreated B-Chronic Lymphocytic Leukemia (CLL) or Small Lymphocytic Lymphoma (SLL) [NCT00602836]Phase 245 participants (Actual)Interventional2008-02-29Completed
Phase II Trial of Rituximab Plus Lenalidomide in Patients With Lymphoma of the Mucosa Associated Lymphoid Tissue (MALT) Type [NCT01611259]Phase 250 participants (Actual)Interventional2012-05-31Completed
A Phase 1/2 Study of Sequential Idarubicin + Cytarabine, Followed by Lenalidomide, in Patients With Myelodysplastic Syndrome (RAEB-2) or With Previously Untreated Acute Myeloid Leukemia [NCT00831766]Phase 1/Phase 251 participants (Actual)Interventional2009-06-25Completed
A Phase I/Randomized Phase II Trial of Idelalisib and Lenalidomide in Patients With Relapsed/Refractory Mantle Cell Lymphoma [NCT01838434]Phase 1106 participants (Actual)Interventional2013-07-31Completed
Phase I/II Study of Lenalidomide in Combination With Anti-PD-1 Monoclonal Antibody CT-011 in Patients With Relapsed/Refractory Multiple Myeloma [NCT02077959]Phase 120 participants (Actual)Interventional2014-03-03Terminated(stopped due to Pharmaceutical Companies decision)
A Prospective Phase II Clinical Study of Orelabrutinib in Combination With Lenalidomide and Rituximab (OLR) in First-line Treatment of Mantle Cell Lymphoma [NCT05076097]Phase 229 participants (Anticipated)Interventional2021-09-30Recruiting
Lenalidomide Maintenance Therapy in Multiple Myeloma: A Phase II Clinical and Biomarker Study [NCT01675141]Phase 211 participants (Actual)Interventional2012-08-20Terminated(stopped due to Original investigator left the NIH and the primary outcome was not reached)
A Phase 1B/2 Randomized, Multicenter, Open-Label Study Of Iberdomide (CC-220) In Combination With Polatuzumab Vedotin Plus Rituximab Or Tafasitamab Or Rituximab Plus Chemotherapy For Subjects With Relapsed Or Refractory Aggressive B-Cell Lymphoma [NCT04882163]Phase 1/Phase 20 participants (Actual)Interventional2021-10-10Withdrawn(stopped due to Business objectives have changed)
Empliciti® (Elotuzumab) Post-Marketing Surveillance Study for Patients With Multiple Myeloma in Taiwan [NCT06163040]27 participants (Anticipated)Observational2023-11-30Not yet recruiting
A Phase I/II Study of Carfilzomib, Lenalidomide, Vorinostat, and Dexamethasone in Relapsed and/or Refractory Multiple Myeloma [NCT01297764]Phase 1/Phase 217 participants (Actual)Interventional2011-04-30Active, not recruiting
A Phase Ib/II Study Evaluating the Safety and Efficacy of Atezolizumab in Combination With Obinutuzumab Plus Lenalidomide in Patients With Relapsed or Refractory Follicular Lymphoma [NCT02631577]Phase 1/Phase 238 participants (Actual)Interventional2015-12-31Completed
Phase II Trial of the PD-1 Antibody Nivolumab in Combination With Lenalidomide and Low Dose Dexamethasone in Patients With High-Risk Smoldering Multiple Myeloma [NCT02903381]Phase 28 participants (Actual)Interventional2016-10-20Completed
Carfilzomib, Lenalidomide, and Dexamethasone in Newly Diagnosed Multiple Myeloma: Clinical and Correlative Phase II Study [NCT01402284]Phase 245 participants (Actual)Interventional2011-07-21Completed
Phase 1 Study of Daratumumab When Given in Combination With Bortezomib, Dexamethasone, Doxil, and Lenalidomide in Patients With Plasma Cell Leukemia [NCT03591744]Phase 10 participants (Actual)Interventional2018-10-25Withdrawn(stopped due to Budgetary constraints)
Revlimid to Augment Efficacy of Prevnar Vaccines in Patients With Relapsed or Refractory Myeloma [NCT00445484]Phase 222 participants (Actual)Interventional2007-01-31Completed
A Pilot Clinical Trial of Lenalidomide (Revlimid®) for the Treatment of Refractory Cancer Pain [NCT00684242]Phase 24 participants (Actual)Interventional2008-05-31Terminated(stopped due to Low accrual.)
Phase I/II Study of Sorafenib, Lenalidomide, and Dexamethasone in Relapsed/Refractory Multiple Myeloma [NCT00687674]Phase 113 participants (Actual)Interventional2008-08-31Terminated(stopped due to Due to study design (and toxicity), this trial closed to accrual prior to opening the phase II portion.)
A Multi-Institutional Phase I/II Study of Revlimid® (Lenalidomide), Velcade® (Bortezomib), Dexamethasone, and Doxil®, (RVDD) Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma [NCT00724568]Phase 1/Phase 274 participants (Actual)Interventional2008-05-31Completed
A Phase 1 Study of Amrubicin in Combination With Lenalidomide and Weekly Dexamethasone in Relapsed/Refractory Multiple Myeloma [NCT01355705]Phase 1/Phase 214 participants (Actual)Interventional2011-08-31Completed
A Phase Ib Multi-Cohort Trial of MK-3475 (Pembrolizumab) in Subjects With Hematologic Malignancies [NCT01953692]Phase 1197 participants (Actual)Interventional2013-11-22Completed
Single Agent Lenalidomide in Adult Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL) [NCT01401322]Phase 25 participants (Actual)Interventional2011-01-31Terminated(stopped due to low accrual)
A Phase I Study of Lenalidomide Plus Pembrolizumab in Patients With Relapsed and/or Refractory Solid Tumors With Phase II Expansion in Non-small Cell Lung Cancer [NCT02963610]Phase 1/Phase 23 participants (Actual)Interventional2017-03-29Terminated(stopped due to Study terminated prematurely due to funding/support being withdrawn by grantor)
2015-09: A Phase II Randomized, Open-label Study of Anti-SLAMF7 mAb During Maintenance Therapy Versus Standard Maintenance Therapy in Gene Expression Profiling (GEP)- Defined Low Risk Multiple Myeloma Patients With High Risk Cytogenetic Abnormalities [NCT03000634]Phase 20 participants (Actual)Interventional2017-05-31Withdrawn(stopped due to Study was not and will not be initiated due to lack of funding.)
Myeloma Cure Project: Prospective, Randomized Trial of Indefinite Revlimid Maintenance Versus Observation for Currently Event-Free Patients With Multiple Myeloma Who Have Completed 3 Years of VTD/TD or VTD or VRD Maintenance on Total Therapy 3 (TT3) Trial [NCT01621672]Phase 342 participants (Actual)Interventional2010-04-30Completed
A Randomized Phase III Trial on the Effect of Elotuzumab in VRD Induction /Consolidation and Lenalidomide Maintenance in Patients With Newly Diagnosed Myeloma [NCT02495922]Phase 3564 participants (Actual)Interventional2015-06-30Completed
Functional Cure Strategy and Clinical Study of AIDS--Study on the Reduction of HIV Viral Reservoir by Immunomodulators (IMs) [NCT05598580]Phase 448 participants (Anticipated)Interventional2022-11-20Recruiting
Phase I/II Study of Lenalidomide in Combination With Rituximab, Ifosfamide, Etoposide, and Carboplatin (RICER) [NCT01241734]Phase 1/Phase 218 participants (Actual)Interventional2010-10-31Completed
Phase II Open Label Study for the Assessment of the Efficacy and Safety of Lenalidomide & Adriamycin & Low Dose Dexamethasone (RAD) in Newly Diagnosed, Symptomatic Multiple Myeloma Patients [NCT02471820]Phase 245 participants (Actual)Interventional2014-11-30Completed
[NCT01450215]Phase 262 participants (Actual)Interventional2011-03-31Completed
A Dose Escalation Study of Ibrutinib With Lenalidomide for Relapsed and Refractory Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma [NCT01886859]Phase 127 participants (Actual)Interventional2013-04-26Completed
Phase 1/2 Study of the Combination of Glofitamab, Venetoclax and Lenalidomide in Patients With Newly Diagnosed High Risk Mantle Cell Lymphoma [NCT05861050]Phase 1/Phase 250 participants (Anticipated)Interventional2023-08-10Recruiting
Carfilzomib, Lenalidomide and Dexamethasone Versus Lenalidomide and Dexamethasone in High- Risk Smoldering Multiple Myeloma: A Randomized Phase II Study. [NCT03673826]Phase 258 participants (Actual)Interventional2018-11-19Active, not recruiting
A Phase 1 Study of Ruxolitinib, Steroids and Lenalidomide for Relapsed/Refractory Multiple Myeloma (RRMM) Patients [NCT03110822]Phase 1134 participants (Anticipated)Interventional2017-02-01Recruiting
A Phase 2 Study of Weekly 70 mg/m2 Carfilzomib for Multiple Myeloma Patients Refractory to 27 mg/m2 Carfilzomib [NCT02294357]Phase 245 participants (Anticipated)Interventional2014-12-31Terminated(stopped due to Early termination due to the difficulties to enroll subjects.)
An Open Label, Single-Arm, Phase 1b/2 Study of the Safety and Efficacy of Combination Treatment With Lenalidomide, Bortezomib, Dexamethasone and Siltuximab (CNTO 328) in Subjects With Newly Diagnosed, Previously Untreated Multiple Myeloma Requiring System [NCT01531998]Phase 1/Phase 214 participants (Actual)Interventional2012-05-31Completed
A PHASE 1b/2 STUDY OF PF-07901801, A CD47 BLOCKING AGENT, WITH TAFASITAMAB AND LENALIDOMIDE FOR PARTICIPANTS WITH RELAPSED/REFRACTORY DIFFUSE LARGE B CELL LYMPHOMA NOT ELIGIBLE FOR STEM CELL TRANSPLANTATION [NCT05626322]Phase 270 participants (Anticipated)Interventional2023-08-04Recruiting
Phase I/II, Open-Label Study of R-ICE (Rituximab-Ifosfamide-Carboplatin-Etoposide) With Lenalidomide (R2-ICE) in Patients With First-Relapse/Primary Refractory Diffuse Large B-Cell Lymphoma (DLBCL) [NCT02628405]Phase 1/Phase 263 participants (Actual)Interventional2016-05-20Active, not recruiting
A Randomized, Phase III Study Comparing Conventional Dose Treatment Using a Combination of Lenalidomide, Bortezomib, and Dexamethasone (RVD) to High-Dose Treatment With Peripheral Stem Cell Transplant in the Initial Management of Myeloma in Patients Up to [NCT01208662]Phase 3729 participants (Actual)Interventional2010-10-31Active, not recruiting
Maintenance Therapy With Lenalidomide Following Bendamustine and Rituximab Induction Therapy for Chronic Lymphocytic Leukemia [NCT01465230]Phase 22 participants (Actual)Interventional2012-03-31Terminated(stopped due to This study was unsuccessful in enrolling the target number of subjects during the funding period.)
A Phase III Multicenter, Randomized Study With Lenalidomide Maintenance vs Observation After Induction Regimen Containing Rituximab Followed by High Dose Chemotherapy and ASCT as First Line Treatment in Adult Patients With Advanced Mantle Cell Lymphoma [NCT02354313]Phase 3300 participants (Anticipated)Interventional2010-05-31Active, not recruiting
Lenalidomide Plus Melphalan as a Preparative Regimen for Autologous Stem Cell Transplantation in Relapsed Multiple Myeloma: A Phase 1 / 2 Study [NCT01054196]Phase 1/Phase 252 participants (Actual)Interventional2010-08-31Active, not recruiting
Phase 3 Randomized, Double-Blind, Placebo Controlled, Multicenter Study to Compare the Efficacy and Safety of Lenalidomide (CC-5013) Plus R-CHOP Chemotherapy (R2-CHOP) Versus Placebo Plus R-CHOP Chemotherapy in Subjects With Previously Untreated Activated [NCT02285062]Phase 3570 participants (Actual)Interventional2015-02-17Completed
Phase II Study: Therapy With Bortezomib + Lenalidomide + Dexamethasone With Lenalidomide + Dexamethasone as Post Transplant Consolidation and Maintenance for Patients With Symptomatic Multiple Myeloma Following Autologous Transplantation [NCT02353468]Phase 23 participants (Actual)Interventional2009-12-31Terminated(stopped due to Principle investigator left the institution)
A Phase 1b Study of SAR650984 (Anti-CD38 mAb) in Combination With Lenalidomide and Dexamethasone for the Treatment of Relapsed or Refractory Multiple Myeloma [NCT01749969]Phase 157 participants (Actual)Interventional2013-02-06Completed
Phase II Study of CC-5013 in Patients With Advanced Renal Cell Carcinoma (RCC) [NCT00403169]Phase 226 participants (Anticipated)Interventional2004-08-31Completed
Efficacy and Safety of Rituximab Plus Zanubrutinib and Lenalidomide for Relapsed and Refractory Diffuse Large B Cell Lymphoma, a Multicenter, Open and Prospective Clinical Trial [NCT05392257]Phase 220 participants (Anticipated)Interventional2022-05-01Recruiting
A Multicenter Phase I/II Dose Escalation Study of Lenalidomide in Relapse/Refractory Waldenstrom Macroglobulinemia [NCT02302469]Phase 1/Phase 217 participants (Actual)Interventional2009-03-31Completed
Prospective Observational Study Evaluating the Safety of Lenalidomide/Dexamethasone Treatment in Patients With Relapsed or Refractory Multiple Myeloma [NCT02692339]22 participants (Actual)Observational [Patient Registry]2016-02-25Completed
Phase 3, Randomized, Open Label Trial of Lenalidomide/Dexamethasone With or Without Elotuzumab in Relapsed or Refractory Multiple Myeloma (MM) [NCT01239797]Phase 3646 participants (Actual)Interventional2011-06-20Completed
The Efficacy and Safety of Lenalidomide With or Without Rituximab and Other Drugs in B-cell Non-Hodgkin Lymphomas: a Real-world Study. [NCT04435743]1,000 participants (Anticipated)Observational2019-11-01Recruiting
A Randomized Phase III Non-inferiority Trial Assessing Lenalidomide, Bortezomib and Dexamethasone Induction Therapy With Either Intravenous or Subcutaneous Isatuximab in Transplant-eligible Patients With Newly Diagnosed Multiple Myeloma. [NCT05804032]Phase 3514 participants (Anticipated)Interventional2023-04-14Recruiting
A Phase I/II, Randomized, Open-label Platform Study Utilizing a Master Protocol to Study Belantamab Mafodotin (GSK2857916) as Monotherapy and in Combination With Anti-Cancer Treatments in Participants With Relapsed/Refractory Multiple Myeloma (RRMM) - DRE [NCT04126200]Phase 1/Phase 2464 participants (Anticipated)Interventional2019-10-07Recruiting
Sequential Chemotherapy and Lenalidomide Followed by Rituximab and Lenalidomide Maintenance for Untreated Mantle Cell Lymphoma: A Phase II Study [NCT02633137]Phase 249 participants (Actual)Interventional2015-12-14Completed
A Phase I Study of Ibrutinib (PCI-32765) in Combination With Lenalidomide in Relapsed and Refractory B-Cell Non-Hodgkin's Lymphoma [NCT01955499]Phase 134 participants (Anticipated)Interventional2013-09-13Active, not recruiting
A Phase I Study of Rituximab, Lenalidomide, and Ibrutinib in Previously Untreated Follicular Lymphoma [NCT01829568]Phase 133 participants (Actual)Interventional2013-06-21Active, not recruiting
Phase II Study of Lenalidomide to Repair Immune Synapse Response and Humoral Immunity in Early-Stage, Asymptomatic Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL/SLL) With High-Risk Genomic Features [NCT01351896]Phase 248 participants (Anticipated)Interventional2011-09-08Active, not recruiting
A Randomized, Phase 2 Study of Biomarker Guided Treatment in DLBCL [NCT04025593]Phase 2128 participants (Anticipated)Interventional2019-07-17Recruiting
A Single Arm,Phase Ib/II Study of the Combination of Lenalidomide and Gemcitabine in Relapsed or Refractory Peripheral T-cell Lymphomas (PTCL) [NCT05105412]Phase 1/Phase 233 participants (Anticipated)Interventional2021-10-31Not yet recruiting
Carfilzomib (K) Plus Lenalidomide (R) and Dexamethasone (D) for BTK Inhibitors Relapsed-refractory or Intolerant Mantle Cell Lymphomas: a Phase II Study [NCT03891355]Phase 216 participants (Actual)Interventional2019-09-30Active, not recruiting
A Phase I, Multiple Center, Open-label, Dose Escalation and Dose Expansion Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Preliminary Efficacy of CM313 in Subjects With Relapsed or Refractory Multiple Myeloma and Lymphoma [NCT04818372]Phase 187 participants (Anticipated)Interventional2021-04-26Recruiting
Lenalidomide in Combination With Rituximab as Treatment for Patients With Relapsed Chronic Lymphocytic Leukemia - RV-CLL-PI-0292 [NCT00759603]Phase 260 participants (Actual)Interventional2008-09-30Completed
Phase 1b/2, Multicenter, Open-label Study of Oprozomib and Dexamethasone, in Combination With Lenalidomide or Oral Cyclophosphamide in Patients With Newly Diagnosed Multiple Myeloma [NCT01881789]Phase 1/Phase 222 participants (Actual)Interventional2013-10-28Terminated(stopped due to A program evaluation identified that the safety profile and pharmacokinetic (PK) characteristics of the formulation used in all oprozomib studies required further optimization and thus enrollment in OPZ003 was halted during dose-escalation.)
First-in-Human Study of the BCL-2 Inhibitor ABBV-453 in Biomarker-Selected Subjects With Relapsed or Refractory Multiple Myeloma [NCT05308654]Phase 1360 participants (Anticipated)Interventional2022-05-17Recruiting
Minimal Residual Disease Response-adapted Deferral of Transplant in Dysproteinemia - MILESTONE Trial [NCT04991103]Phase 220 participants (Anticipated)Interventional2021-09-22Recruiting
A Phase I Study of Ibrutinib (PCI-32765) in Combination With Revlimid/Dexamethasone (Rd) in Relapsed/Refractory Multiple Myeloma [NCT03702725]Phase 114 participants (Actual)Interventional2019-08-29Active, not recruiting
A MULTICENTER, RANDOMIZED, OPEN LABEL PHASE II STUDY OF CARFILZOMIB, CYCLOPHOSPHAMIDE AND DEXAMETHASONE (CCyd) as Pre Transplant INDUCTION and Post Transplant Consolidation or CARFILZOMIB, LENALIDOMIDE AND DEXAMETHASONE (CRd) as Pre Transplant INDUCTION a [NCT02203643]Phase 2477 participants (Actual)Interventional2015-02-28Active, not recruiting
Phase-II Trial to Assess the Efficacy and Safety of Lenalidomide in Addition to 5-Azacitidine and Donor Lymphocyte Infusions (DLI) for the Treatment of Patients With MDS, CMML or AML Who Relapse After Allogeneic Stem Cell Transplantation [NCT02472691]Phase 250 participants (Actual)Interventional2015-05-31Completed
Solitary Plasmacytoma of Bone: Randomized Phase III Trial to Evaluate Treatment With Adjuvant Systemic Treatment and Zoledronic Acid Versus Zoledronic Acid After Definite Radiation Therapy [NCT02516423]Phase 311 participants (Actual)Interventional2015-12-31Active, not recruiting
A Randomized Clinical Trial of Lenalidomide (CC-5013) and Dexamethasone With and Without Autologous Peripheral Blood Stem Cell Transplant in Patients With Newly Diagnosed Multiple Myeloma [NCT01731886]Phase 460 participants (Actual)Interventional2012-09-30Completed
Phase I/II Study of Lenalidomide and Ofatumumab for the Treatment of Relapsed or Refractory Non-Hodgkin's Lymphoma [NCT01060384]Phase 1/Phase 246 participants (Actual)Interventional2010-03-29Completed
An Open-label, Randomized, Single Dose, Crossover Bioequivalent Study to Compare Pharmacokinetic Property of SYP-1512 Tab and Revlimid Cap, 25mg in Healthy Male Volunteers [NCT03208218]Phase 142 participants (Actual)Interventional2016-08-17Completed
Assessment of Survival and Autonomy With Rituximab Plus Chemotherapy or Rituximab Plus Lenalidomide for Elderly Patients With Relapsed Diffuse Large B-cell Lymphoma [NCT04113226]Phase 2114 participants (Anticipated)Interventional2021-07-26Recruiting
A Randomized Phase II Study of Oral Lenalidomide (Revlimid [TM]), an Antiangiogenic and Immunomodulatory Agent, in Subjects With Stage IV Ocular Melanoma [NCT00109005]Phase 217 participants (Actual)Interventional2005-04-30Completed
Tandem High-Dose Therapy With Melphalan and Total Marrow Irradiation (TMI) With Peripheral Blood Progenitor Cell Support and Lenalidomide Maintenance in Multiple Myeloma: A Phase I/II Trial [NCT00112827]Phase 1/Phase 254 participants (Actual)Interventional2004-11-30Completed
Phase II Study of Lenalidomide Maintenance in Patients With High Risk AML in Remission [NCT02126553]Phase 229 participants (Actual)Interventional2014-11-13Completed
A Multi-Center, Phase 1/2, Open-Label Study of Selinexor (KPT- 330), Lenalidomide, and Dexamethasone in Patients With Relapsed/Refractory Multiple Myeloma [NCT02389543]Phase 1/Phase 20 participants (Actual)Interventional2015-07-31Withdrawn(stopped due to Terminated this trial and added a Lenalidomide arm to KCP-330-017)
A Phase II Trial of Revlimid® as Consolidation Treatment of Residual Disease in Patients With Chronic Lymphocytic Leukemia (CLL) [NCT01600053]Phase 211 participants (Actual)Interventional2011-11-30Terminated(stopped due to interim analysis showed that the trial will not meet the interim endpoint.)
Multicenter Open Label Phase 3 Study of Isatuximab Plus Lenalidomide and Dexamethasone With/Without Bortezomib in the Treatment of Newly Diagnosed Non Frail Transplant Ineligible Multiple Myeloma Elderly Patients (≥ 65; < 80 Years). [NCT04751877]Phase 3270 participants (Actual)Interventional2021-07-17Active, not recruiting
A Phase II Study of Sequential Carfilzomib, Clarithromycin (Biaxin®), Lenalidomide (Revlimid®), and Dexamethasone (Decadron®) [Car-BiRD] Therapy for Subjects With Newly Diagnosed Multiple Myeloma [NCT01559935]Phase 274 participants (Actual)Interventional2012-03-31Active, not recruiting
A Phase 2 Single Arm Study of Safety of Elotuzumab Administered Over Approximately 60 Minutes in Combination With Lenalidomide and Dexamethasone for Newly Diagnosed or Relapsed/Refractory Multiple Myeloma Patients [NCT02159365]Phase 284 participants (Actual)Interventional2014-03-08Completed
A Phase 3, Multicenter, Randomized, Open-label Study to Compare the Efficacy and Safety of bb2121 Versus Standard Regimens in Subjects With Relapsed and Refractory Multiple Myeloma (RRMM) (KarMMa-3) [NCT03651128]Phase 3381 participants (Actual)Interventional2019-04-16Active, not recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00056160 (4) [back to overview]Time to First Worsening of Eastern Cooperative Oncology Group (ECOG) Performance Status Scale (Best Score=0, Fully Active, Able to Carry on All Pre-disease Performance Without Restriction; Worst Score=5, Dead.)
NCT00056160 (4) [back to overview]Time to Tumor Progression (TTP)
NCT00056160 (4) [back to overview]Myeloma Response
NCT00056160 (4) [back to overview]Overall Survival
NCT00064038 (2) [back to overview]Progression-Free Survival
NCT00064038 (2) [back to overview]Toxicity
NCT00065156 (12) [back to overview]Participants With Bone Marrow Progression
NCT00065156 (12) [back to overview]Participants Who Achieved Red Blood Cell (RBC) -Transfusion Independence
NCT00065156 (12) [back to overview]Participants Who Relapsed or Maintained Their Transfusion Independence After Achieving Transfusion Independence During the Study
NCT00065156 (12) [back to overview]Participant Counts of Platelet Response
NCT00065156 (12) [back to overview]Participant Counts of Cytogenetic Response
NCT00065156 (12) [back to overview]Participants With Adverse Experiences
NCT00065156 (12) [back to overview]Participant Counts of Absolute Neutrophil Count (ANC) Response
NCT00065156 (12) [back to overview]Participants With a >= 50% Decrease From Baseline in Red Blood Cell (RBC) Transfusion Requirements Over Any Consecutive 56 Days During Study
NCT00065156 (12) [back to overview]Kaplan Meier Estimate for Duration of Transfusion Independence Response
NCT00065156 (12) [back to overview]Time to Transfusion Independence
NCT00065156 (12) [back to overview]Change in Hemoglobin Concentration From Baseline to Maximum Value During Response Period for Responders
NCT00065156 (12) [back to overview]Participants With Complete or Partial Bone Marrow Improvement
NCT00087672 (1) [back to overview]Efficacy of CC-5013 in Myelofibrosis
NCT00091260 (3) [back to overview]Number of Patients Removed From Study Treatment Due to Toxicities
NCT00091260 (3) [back to overview]Number of Patients Who Received Both CC-5013 and Dexamethasone and Had a Hematologic Response
NCT00091260 (3) [back to overview]Number of Patients With Hematologic Response With Single-agent CC-5013
NCT00096044 (6) [back to overview]Number of Participants With Adverse Events on Combination Therapy of CC-5013+Rituximab
NCT00096044 (6) [back to overview]Percentage of Patients Achieving a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) on Combination Therapy of CC-5013+Rituximab
NCT00096044 (6) [back to overview]Percentage of Patients Achieving a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) on Single Agent CC-5013 at 6 Months
NCT00096044 (6) [back to overview]Time to Progression for the Combination Therapy of CC-5013+Rituximab
NCT00096044 (6) [back to overview]Number of Participants With Adverse Events on Single Agent CC-5013
NCT00096044 (6) [back to overview]Time to Progression for Single Agent CC-5013
NCT00098475 (2) [back to overview]Proportion of Patients With Objective Response (First Phase, Step 2)
NCT00098475 (2) [back to overview]Proportion of Patients With Objective Response (First Phase, Step 1)
NCT00109005 (2) [back to overview]Clinical Responses in Patients With Metastatic Ocular Melanoma
NCT00109005 (2) [back to overview]Number of Participants With Adverse Events
NCT00109772 (17) [back to overview]Change From Baseline in the Brief Pain Inventory (BPI) Total Score at Week 12
NCT00109772 (17) [back to overview]"Change From Baseline in Mechanically Evoked (Allodynia) Numeric Rating Scale (NRS) Score at Week 12"
NCT00109772 (17) [back to overview]Change From Baseline in the Evening Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in Daily Sleep Assessment Average Score at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in the Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score Using Averaged Morning and Evening Readings at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in Activity Level Rating Using a Numeric Rating Scale (NRS) at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in Participant Assessment of CRPS Symptoms Total Score at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in the Total Score of the Short Form McGill Pain Questionnaire (SF-MPQ) at Week 12
NCT00109772 (17) [back to overview]Percentage of Participants Who Have a >= 30% Reduction (Improvement) in the Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score From Baseline to the Last Assessment
NCT00109772 (17) [back to overview]Participants With Treatment-Emergent Adverse Events in the Double-Blind Period or the Extension Period
NCT00109772 (17) [back to overview]Participants Who Had a Change to CRPS Pain Medication During the Treatment Period
NCT00109772 (17) [back to overview]Patient Global Impression of Change (PGIC) at Week 12
NCT00109772 (17) [back to overview]Difference in Allodynia Rating Between the CRPS-affected Limb and the Normal Limb at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in the Profile of Mood States (POMS) at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in the Morning Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in the Maximal Composite Sensory Nerve Conduction Velocity at Week 12
NCT00109772 (17) [back to overview]Change From Baseline in the Maximal Composite Motor Nerve Conduction Velocity at Week 12
NCT00112827 (3) [back to overview]Maximum Tolerated Dose (MTD)
NCT00112827 (3) [back to overview]Overall Survival
NCT00112827 (3) [back to overview]Number of Subjects With Response
NCT00114101 (4) [back to overview]Response to Autologous Hematopoietic Stem-cell Transplant (HSCT) at Day 100
NCT00114101 (4) [back to overview]Number of Participants With Progression, Death or Diagnosis of Second Primary Malignancy
NCT00114101 (4) [back to overview]Overall Survival
NCT00114101 (4) [back to overview]Time to Progression
NCT00142168 (4) [back to overview]Minor Response Rate
NCT00142168 (4) [back to overview]Major Response Rate
NCT00142168 (4) [back to overview]Time to Progression
NCT00142168 (4) [back to overview]Overall Response
NCT00179621 (15) [back to overview]Participants' Response Based on Bone Marrow Samples by the International MDS Working Group (IWG 2000) During Double-blind Period
NCT00179621 (15) [back to overview]Participants' Response in Absolute Neutrophil Counts as Defined by the International MDS Working Group (IWG 2000) During Double-blind Period
NCT00179621 (15) [back to overview]Participants' Response in Platelet Counts as Defined by the International MDS Working Group (IWG 2000) During Double-blind Period
NCT00179621 (15) [back to overview]Summary of Participants Who Had Adverse Events (AE) During the Double-blind Period
NCT00179621 (15) [back to overview]Participants Who Progressed to Acute Myeloid Leukemia (AML) During the Study
NCT00179621 (15) [back to overview]Change From Baseline in the Functional Assessment of Cancer Therapy-Anemia (FACT-An) Endpoints at Week 12
NCT00179621 (15) [back to overview]Change From Baseline in the Trial Outcome Index-Anemia (TOI-An) Endpoints at Week 12
NCT00179621 (15) [back to overview]Change From Baseline in the Trial Outcome Index-Fatigue (TOI-F) Endpoints at Week 12
NCT00179621 (15) [back to overview]Duration of Red Blood Cell (RBC) Transfusion Independence for Participants Who Became RBC Transfusion Independent for at Least 182 Days
NCT00179621 (15) [back to overview]Kaplan Meier Estimates of Overall Survival by Randomized Group
NCT00179621 (15) [back to overview]Maximum Change From Baseline in Hemoglobin During the Double-blind Period for Participants Who Became Red Blood Cell (RBC) Transfusion Independent for at Least 182 Days
NCT00179621 (15) [back to overview]Participant Count of Deaths During Double-blind and Open-label by Randomized Group
NCT00179621 (15) [back to overview]Participants Who Achieved Red Blood Cell (RBC) Transfusion Independence for >= 26 Weeks (182 Days)
NCT00179621 (15) [back to overview]Participants Who Achieved Red Blood Cell (RBC) Transfusion Independence for 56 Days
NCT00179621 (15) [back to overview]Participants Showing Cytogenetic Response by the International MDS Working Group (IWG 2000) During Double-blind Period as Evaluated by Central Review
NCT00179647 (1) [back to overview]Overall Incidence of Adverse Events
NCT00179660 (6) [back to overview]Duration of Response
NCT00179660 (6) [back to overview]Duration of Tumor Control
NCT00179660 (6) [back to overview]Percentage of Participants With Response
NCT00179660 (6) [back to overview]Percentage of Participants With Tumor Control
NCT00179660 (6) [back to overview]Progression-free Survival
NCT00179660 (6) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00179673 (5) [back to overview]Percentage of Participants With Response
NCT00179673 (5) [back to overview]Percentage of Participants With Tumor Control
NCT00179673 (5) [back to overview]Progression Free Survival (PFS)
NCT00179673 (5) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00179673 (5) [back to overview]The Duration of Response
NCT00227591 (1) [back to overview]Overall Response Rate
NCT00238238 (2) [back to overview]Time to Progression
NCT00238238 (2) [back to overview]Overall Response Rate
NCT00267059 (1) [back to overview]Number of Patients in Overall Response Categories
NCT00287287 (1) [back to overview]Response Rate
NCT00294632 (2) [back to overview]Maximum Tolerated Dose (MTD) of Lenalidomide in Combination With Rituximab
NCT00294632 (2) [back to overview]Objective Response Rate of Participants Treated With Lenalidomide 20 mg: Overall Response as % of Participants With Complete or Partial Response
NCT00348595 (4) [back to overview]Plasma Concentration of Revilimid at Steady State
NCT00348595 (4) [back to overview]Safety, Feasibility and Tolerance of Revlimid as Assessed by Number of Participants Experiencing Grade 3 and 4 Adverse Events.
NCT00348595 (4) [back to overview]Change in of PSA Slope
NCT00348595 (4) [back to overview]Number of Participants With Prostate-specific Antigen (PSA) Progression
NCT00352001 (8) [back to overview]Time to Transformation to Acute Myeloid Leukemia or Death
NCT00352001 (8) [back to overview]Overall Survival Among Patients With Complete Response
NCT00352001 (8) [back to overview]PHASE I: Maximum Tolerated Dose of Azacitidine
NCT00352001 (8) [back to overview]PHASE II: Determine the Number of Patients With Responses for Efficacy(Measured as Response Rate)
NCT00352001 (8) [back to overview]Time to Relapse After Achieving Complete Response
NCT00352001 (8) [back to overview]PHASE II: Determine the Number of Patients With Responses for Efficacy(Measured as Response Rate)
NCT00352001 (8) [back to overview]Number of Patients That Experience Grade 3 or 4 Treatment Related Non-hematologic Adverse Events
NCT00352001 (8) [back to overview]PHASE I: Maximum Tolerated Dose of Lenalidomide
NCT00352365 (4) [back to overview]Number of Patients With Grade 3 Through 5 Adverse Events That Are Related to Study Drug
NCT00352365 (4) [back to overview]Total Response
NCT00352365 (4) [back to overview]Cytogenetic Abnormalities
NCT00352365 (4) [back to overview]Complete Response
NCT00352794 (1) [back to overview]Number of Patients With Objective Response (Complete and Partial Response + Hematological Improvement)
NCT00360672 (1) [back to overview]Number of Participants With a Response (Complete Remissions (CR), Complete Remissions With Incomplete Platelet Recovery [CRp] and Partial Responses)
NCT00378105 (4) [back to overview]Objective Response Rate of the Drug Combination in This Patient Populations.
NCT00378105 (4) [back to overview]Estimated 18-month Overall Survival Rate
NCT00378105 (4) [back to overview]Percentage of Patients Who Remained in Response for More Than 18 Months
NCT00378105 (4) [back to overview]Estimated 18-month Progression Free Survival (PFS) Rate
NCT00378209 (5) [back to overview]Overall Survival
NCT00378209 (5) [back to overview]The Proportion of Patients Alive and Without Progressive Disease (PD) for ≥6 Months
NCT00378209 (5) [back to overview]Duration of Response
NCT00378209 (5) [back to overview]Objective Response Rate
NCT00378209 (5) [back to overview]Progression Free Survival
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Constipation Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Diarrhoea Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Dyspnoea Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Emotional Functioning Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Congitive Functioning Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Fatigue Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Financial Difficulties Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Social Functioning Scale
NCT00405756 (28) [back to overview]Summary of Participants With Treatment-Emergent Adverse Events (TEAE) During the Double-Blind Treatment Period
NCT00405756 (28) [back to overview]Number of Participants in Disease Response Categories Representing Their Best Response During the Double-blind Treatment Period
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) In Side Effects of Treatment Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) In Future Perspective Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) In Body Image Scale
NCT00405756 (28) [back to overview]Kaplan Meier Estimates for Duration of Response as Determined by the Central Adjudication Committee (CAC)
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Role Functioning Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Physical Functioning Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Disease Symptoms Scale
NCT00405756 (28) [back to overview]Kaplan Meier Estimates of Progression-free Survival (PFS) From Start of Maintenance Therapy Period Based on the Response Assessment by the Central Adjudication Committee (CAC)
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Pain Scale
NCT00405756 (28) [back to overview]Kaplan Meier Estimates for Time to Next Antimyeloma Therapy
NCT00405756 (28) [back to overview]Kaplan Meier Estimates of Overall Survival (OS)
NCT00405756 (28) [back to overview]Kaplan Meier Estimates of Time to Progression (TTP) Based on the Response Assessment by the Central Adjudication Committee (CAC)
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Global Quality of Life Scale
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Appetite Loss Scale
NCT00405756 (28) [back to overview]Kaplan Meier Estimates of Progression-free Survival (PFS) Based on the Response Assessment by the Central Adjudication Committee (CAC)
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Nausea and Vomiting Scale
NCT00405756 (28) [back to overview]Time to First Response
NCT00405756 (28) [back to overview]Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Insomnia Scale
NCT00410605 (5) [back to overview]Confirmed Anti-tumor Response Rate (Complete Response and Partial Response) to the Combination of Bevacizumab and Lenalidomide
NCT00410605 (5) [back to overview]Effect of Bev/Rev on Markers of Myeloma Activity in Myeloma Cells and Stromal Cells at Baseline
NCT00410605 (5) [back to overview]Toxicity and Tolerability of the Bevacizumab and Lenalidomide Combination
NCT00410605 (5) [back to overview]Progression Free Survival (Time to Progression)
NCT00410605 (5) [back to overview]Effect of Bev/Rev on Markers of Myeloma Activity in Myeloma Cells and Stromal Cells at 3 Months Post-baseline
NCT00413036 (4) [back to overview]Progression-free Survival as Determined by Central Review
NCT00413036 (4) [back to overview]Duration of Response as Determined by Central Review
NCT00413036 (4) [back to overview]Participants Categorized by Best Response as Determined by Central Review
NCT00413036 (4) [back to overview]Time to Progression as Determined by Central Review
NCT00420849 (24) [back to overview]Time to First Venous Thromboembolic Treatment-Emergent Adverse Event (TEAE)
NCT00420849 (24) [back to overview]Time to First Peripheral Neuropathy Treatment-Emergent Adverse Event (TEAE)
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Side Effects Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Future Perspective Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Disease Symptoms Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Body Image Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Social Functioning Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Role Functioning Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Physical Functioning Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Pain Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Nausea/Vomiting Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Insomnia Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Global Quality of Life Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Fatigue Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Emotional Functioning Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Dyspnoea Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Diarrhea Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Constipation Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Cognitive Functioning Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Appetite Loss Scale
NCT00420849 (24) [back to overview]Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Financial Problems Scale
NCT00420849 (24) [back to overview]Participants With Adverse Events of Special Interest: Venous Thromboembolic Events
NCT00420849 (24) [back to overview]Participants With Adverse Events of Special Interest: Peripheral Neuropathy
NCT00420849 (24) [back to overview]Overall Incidence of Treatment-emergent Adverse Events (TEAEs), by Severity, Seriousness, and Relationship to Treatment
NCT00424047 (12) [back to overview]Kaplan-Meier Estimate of Duration of Response
NCT00424047 (12) [back to overview]Time to First Symptomatic Skeletal-related Event (SRE) (Clinical Need for Radiation or Surgery to Bone)
NCT00424047 (12) [back to overview]Kaplan-Meier Estimate of Time to Tumor Progression (TTP) (Later Cut-off Date of 02 Mar 2008)
NCT00424047 (12) [back to overview]Kaplan-Meier Estimate of Overall Survival (OS) (Later Cut-off Date of 02 March 2008)
NCT00424047 (12) [back to overview]Kaplan-Meier Estimate of Duration of Response (Cut-off at a Later Date of 03 March 2008)
NCT00424047 (12) [back to overview]Kaplan-Meier Estimate of Time to Tumor Progression (TTP)
NCT00424047 (12) [back to overview]Number of Participants With Adverse Events (AE)
NCT00424047 (12) [back to overview]Kaplan-Meier Estimate of Overall Survival (OS)
NCT00424047 (12) [back to overview]Time to First Worsening on the Eastern Cooperative Oncology Group (ECOG) Performance Scale
NCT00424047 (12) [back to overview]Time to First Worsening on the Eastern Cooperative Oncology Group (ECOG) Performance Scale (Later Cut-off Date of 02 March 2008)
NCT00424047 (12) [back to overview]Myeloma Response Rates Based on the Reviewers Best Response Assessment (Later Cut-off Date of 02 March 2008)
NCT00424047 (12) [back to overview]Summary of Myeloma Response Rates Based on Best Response Assessment
NCT00439231 (1) [back to overview]To Establish the Overall Response Rate Measured at 24 Weeks After First Dose of Lenalidomide Using This Dosing Regimen
NCT00445484 (4) [back to overview]6B Antibody Response to Prevnar Vaccine in Peripheral Blood
NCT00445484 (4) [back to overview]14F Antibody Response to Prevnar Vaccine in Peripheral Blood
NCT00445484 (4) [back to overview]19F Antibody Response to Prevnar Vaccine in Peripheral Blood
NCT00445484 (4) [back to overview]23F Antibody Response to Prevnar Vaccine in Peripheral Blood
NCT00445692 (2) [back to overview]Time to Disease Progression
NCT00445692 (2) [back to overview]Episodes of Grade 3-4 Non Infectious, Non-dermatological or Non-neurological Toxicities, Episodes of Any Infections, Grade 3-4 Dermatological or Episodes of Grade 2-3 Peripheral Neuropathy Common Terminology Criteria for Adverse Events Version 3
NCT00460031 (5) [back to overview]Ratio of Change in Immune Response From Baseline
NCT00460031 (5) [back to overview]Change in Immune Response From Baseline
NCT00460031 (5) [back to overview]Time to Progression
NCT00460031 (5) [back to overview]Number of Patients With a Partial Response, Progressive Disease, or Stable Disease Based on Prostate-Specific Antigen (PSA) or Measurable Disease
NCT00460031 (5) [back to overview]Number of Patients With Grade 3 and 4 Toxicity as Assessed by NCI CTCAE v3.0
NCT00466921 (4) [back to overview]Response to Treatment
NCT00466921 (4) [back to overview]Number of Patients Who Experience Toxicity as Assessed by NCI CTCAE v3.0
NCT00466921 (4) [back to overview]Progression-free Survival (PFS)
NCT00466921 (4) [back to overview]Duration of Response (DOR)
NCT00477750 (5) [back to overview]Overall Survival (OS) at 3 Years
NCT00477750 (5) [back to overview]Duration of Response (DOR)
NCT00477750 (5) [back to overview]Progression-free Survival
NCT00477750 (5) [back to overview]Percentage of Participants With Toxicity, Assessed Using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0 (v3)
NCT00477750 (5) [back to overview]Patients With Overall Confirmed Response
NCT00478218 (4) [back to overview]Overall Survival (OS)
NCT00478218 (4) [back to overview]Duration of Response (DOR)
NCT00478218 (4) [back to overview]Number of Participants Who Achieved a Confirmed Response (CR), Very Good Partial Response (VGPR) or Partial Response (PR) During Treatment
NCT00478218 (4) [back to overview]Progression-free Survival (PFS)
NCT00478777 (3) [back to overview]Participants With Treatment-emergent Adverse Experiences (TEAEs)
NCT00478777 (3) [back to overview]Participant's Best Overall Response Based on the European Group for Blood and Marrow Transplantation (EBMT) Myeloma Response Criteria
NCT00478777 (3) [back to overview]Kaplan Meier Estimate for Time to Disease Progression
NCT00482911 (2) [back to overview]Response Rate (Complete and Partial Response)
NCT00482911 (2) [back to overview]Toxicity
NCT00507442 (11) [back to overview]Number of Patients With Stringent Complete Response Rate
NCT00507442 (11) [back to overview]Overall Survival
NCT00507442 (11) [back to overview]Probability of 1-year Survival
NCT00507442 (11) [back to overview]Number of Patients With Combined Complete Response and Very Good Partial Response
NCT00507442 (11) [back to overview]Time to Disease Progression
NCT00507442 (11) [back to overview]Time to Response
NCT00507442 (11) [back to overview]Progression-free Survival
NCT00507442 (11) [back to overview]Duration of Response
NCT00507442 (11) [back to overview]Number of Patients With Adverse Events (AEs)
NCT00507442 (11) [back to overview]Number of Patients With Complete Response Rate + Near Complete Response Rate
NCT00507442 (11) [back to overview]Number of Patients With Overall Response
NCT00522392 (4) [back to overview]Response Rates (Complete Response [CR] or Very Good Partial Response [VGPR])
NCT00522392 (4) [back to overview]Progression-free Survival (PFS)
NCT00522392 (4) [back to overview]Overall Survival (OS)
NCT00522392 (4) [back to overview]Change in Quality of Life (QOL) From Baseline to 6 Months Post Consolidation as Assessed by the Functional Assessment of Cancer Therapy-Neurotoxicity Trial Outcome Index (FACT-Ntx TOI)
NCT00535873 (1) [back to overview]Overall Response Rate (ORR)
NCT00536341 (4) [back to overview]Complete Response Rate
NCT00536341 (4) [back to overview]Overall Survival
NCT00536341 (4) [back to overview]Number of Adverse Events as a Measure of Safety and Tolerability
NCT00536341 (4) [back to overview]Progression-Free Survival
NCT00538733 (4) [back to overview]Effect of Drug Combination on Multiple Myeloma
NCT00538733 (4) [back to overview]Progression Free Survival
NCT00538733 (4) [back to overview]Median Time to Maximum Response
NCT00538733 (4) [back to overview]Event Free Survival
NCT00538824 (1) [back to overview]Effect of Drug Combination on Multiple Myeloma
NCT00540007 (8) [back to overview]Relapse Free Survival (RFS)
NCT00540007 (8) [back to overview]Safety and Tolerability of Lenalidomide Therapy as Measured by the Number of Participants Who Experience Each Adverse Event (Grade 3 or 4 Adverse Events Only) Refractory cHL.
NCT00540007 (8) [back to overview]Time to Progression (TTP).
NCT00540007 (8) [back to overview]Duration of Response
NCT00540007 (8) [back to overview]Cytostatic Overall Response Rate
NCT00540007 (8) [back to overview]Event Free Survival (EFS).
NCT00540007 (8) [back to overview]Overall Survival (OS)
NCT00540007 (8) [back to overview]Objective Overall Response Rate (ORR) in Relapsed or Refractory cHL.
NCT00540644 (3) [back to overview]Quality of Life Using the FACT-G Data
NCT00540644 (3) [back to overview]Response Rate (RR) After 6 Cycles of Therapy Using the Proposed International Myeloma Working Group Uniform Response Criteria
NCT00540644 (3) [back to overview]Treatment Related Adverse Events Grade 3 or Higher
NCT00546897 (12) [back to overview]Duration of CR for Complete Responders
NCT00546897 (12) [back to overview]CR With Complete Blood Counts (CRi) Rate
NCT00546897 (12) [back to overview]Response Rate (RR)
NCT00546897 (12) [back to overview]Morphologic Leukemia Free State
NCT00546897 (12) [back to overview]Morphologic Complete Remission Rate (CRm)
NCT00546897 (12) [back to overview]Safety and Tolerability (Removal From Study Due to Adverse Events)
NCT00546897 (12) [back to overview]Relapse Free Survival (RFS) for Complete Responders
NCT00546897 (12) [back to overview]Progression-free Survival
NCT00546897 (12) [back to overview]Partial Remission Rate (PR)
NCT00546897 (12) [back to overview]Overall Survival (OS)
NCT00546897 (12) [back to overview]Complete Remission Rate (CRm + CRi + CRc)
NCT00546897 (12) [back to overview]Cytogenetics CR Rate (CRc)
NCT00553644 (4) [back to overview]Number of Participants With an Overall Response Defined as Complete Response and Partial Response
NCT00553644 (4) [back to overview]Time to Progression
NCT00553644 (4) [back to overview]Overall Survival
NCT00553644 (4) [back to overview]Incidence of Adverse Events
NCT00560391 (10) [back to overview]Number of Participants in the Dose Escalation Phase Who Reached Maximum Tolerated Dose (MTD) of Dasatinib With Lenalidomide and Dexamethasone
NCT00560391 (10) [back to overview]Number of Participants With Dose-limiting Toxicity (DLT)
NCT00560391 (10) [back to overview]Number of Participants With Minimal Response
NCT00560391 (10) [back to overview]Number of Participants With Partial Response
NCT00560391 (10) [back to overview]Number of Participants Who Died, Serious Adverse Events (SAEs), Adverse Events (AEs) and AEs Leading to Study Drug Discontinuation
NCT00560391 (10) [back to overview]Number of Participants With Complete Response and Very Good Partial Response
NCT00560391 (10) [back to overview]Number of Participants With Serum Chemistry Abnormalities (Worst On-study Grade vs Baseline): Aspartate Aminotransferase (AST), Alanine Aminotransferase (ALT), Total Bilirubin (TB), and Serum Creatinine (SC)
NCT00560391 (10) [back to overview]Number of Participants With Serum Chemistry Abnormalities (Worst On-study Grade vs Baseline): High Calcium, Low Calcium, Low Magnesium, and Low Phosphorus
NCT00560391 (10) [back to overview]Recommended Phase II Dose (RP2D) of the Combination (Dasatinib + Lenalidomide + Dexamethasone)
NCT00560391 (10) [back to overview]Number of Participants With Hematology Abnormalities (Worst On-study Grade vs Baseline): Leukopenia, Neutropenia, Thrombocytopenia, and Anemia
NCT00564889 (5) [back to overview]Number of Patients With Organ Response
NCT00564889 (5) [back to overview]Number of Participants Who Achieved a Confirmed Response Defined as a Complete Response (CR), Very Good Partial Response (VGPR) or Partial Response (PR)
NCT00564889 (5) [back to overview]Number of Participants With Severe Adverse Events
NCT00564889 (5) [back to overview]Overall Survival (OS)
NCT00564889 (5) [back to overview]Progression Free Survival (PFS)
NCT00567229 (1) [back to overview]Final Response Rate After 4 Courses of Treatment
NCT00602459 (6) [back to overview]Overall Response Rate in Patients Without Del(11q22.3)
NCT00602459 (6) [back to overview]Overall Response Rates in Patients With Del(11q22.3)
NCT00602459 (6) [back to overview]PFS Rate of Patients With Del(11q22.3)
NCT00602459 (6) [back to overview]Time-to-progression in Patients With Del(11q22.3)
NCT00602459 (6) [back to overview]Time-to-progression in Patients Without Del(11q22.3)
NCT00602459 (6) [back to overview]2-Year Progression Free Survival (PFS) Rate
NCT00602641 (4) [back to overview]Change in Functional Assessment of Cancer Therapy-Neurotoxicity Trial Outcome Index (FACT-Ntx TOI) Score From Baseline to Cycle 12
NCT00602641 (4) [back to overview]Overall Survival
NCT00602641 (4) [back to overview]Very Good Partial Response (VGPR) Rate
NCT00602641 (4) [back to overview]Progression-Free Survival (PFS)
NCT00602836 (6) [back to overview]Overall Survival (OS)
NCT00602836 (6) [back to overview]Treatment Free Survival (TFS)
NCT00602836 (6) [back to overview]Number of Participants Who Convert From a CR With Minimal Residual Disease (MRD) Positive Status After PCR to a CR With MRD-negative Status After 6 Courses of Consolidation With Lenalidomide
NCT00602836 (6) [back to overview]Number of Participants Who Convert From a Nodular Partial Response (nPR), Partial Response (PR), or Stable Disease (SD) After Pentostatin, Cyclophosphamide, and Rituximab (PCR) to a Complete Response (CR) After 6 Courses of Consolidation With Lenalidomide
NCT00602836 (6) [back to overview]Number of Participants With a Response (CR, nPR, PR)
NCT00602836 (6) [back to overview]Number of Participants With Complete Response (CR)
NCT00603447 (2) [back to overview]Number of Participants With Dose-limiting Toxicities
NCT00603447 (2) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00609869 (3) [back to overview]Clinical Benefit Rate
NCT00609869 (3) [back to overview]Median Time to Treatment Failure (TTF)
NCT00609869 (3) [back to overview]Overall Response Rate (ORR)
NCT00617591 (5) [back to overview]Median Progression Free Survival (PFS) in Months
NCT00617591 (5) [back to overview]Overall Response Rate (ORR) - Percentage of Participants With Partial Response or Better With Induction Regimen
NCT00617591 (5) [back to overview]Percentage of Participants With Very Good Partial Remission (VGPR) or Better
NCT00617591 (5) [back to overview]Occurrence of Induction Toxicities
NCT00617591 (5) [back to overview]2 Year Overall Survival (OS) Rate
NCT00619684 (6) [back to overview]Number of Patients Who Experience Improvement in GVHD on Lenalidomide, Defined as the Reduction in Severity of GVHD as Defined by the National Institutes of Health (NIH) Consensus Criteria
NCT00619684 (6) [back to overview]Response Rate, Defined as the Number of Patients Achieving Complete Response (CR), Partial Response (PR), or Minor Response (MR)
NCT00619684 (6) [back to overview]Overall Survival
NCT00619684 (6) [back to overview]Adverse Events, Graded According to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0
NCT00619684 (6) [back to overview]TTP
NCT00619684 (6) [back to overview]Number of Patients Requiring Dose Interruption, Dose Reduction or Discontinuance of Lenalidomide
NCT00622336 (2) [back to overview]Number of Participants With Adverse Events (AE) During the Extension Phase
NCT00622336 (2) [back to overview]Number of Participants With Adverse Events (AE) During the Treatment Phase
NCT00632359 (3) [back to overview]Time to Progression
NCT00632359 (3) [back to overview]Response Assessments: Disease Status at the End of Lenalidomide Consolidation Per International Workshop on Chronic Lymphocytic Leukemia (IWCLL) Response Criteria
NCT00632359 (3) [back to overview]Improvement in Quality of Remission: Number of Participants With Response Versus No Response by NCI Working Group Criteria Disease Status Change at Start/End of Lenalidomide Consolidation
NCT00633594 (12) [back to overview]Incidence of Non-Serious Adverse Events as a Measure of Safety and Tolerability, Phase II
NCT00633594 (12) [back to overview]Duration of Response (DoR) of Phase I and Phase II Participants
NCT00633594 (12) [back to overview]Duration of Response (DoR) of Previously Treated and Previously Untreated Participants
NCT00633594 (12) [back to overview]Maximum Tolerated Dose of Lenalidomide Combined With Bortezomib and Rituximab in Phase I Participants
NCT00633594 (12) [back to overview]Overall Response Rate (ORR) of Phase I and Phase II Participants
NCT00633594 (12) [back to overview]Overall Response Rate (ORR) of Previously Treated and Previously Untreated Participants
NCT00633594 (12) [back to overview]Overall Survival of Phase I and Phase II Participants
NCT00633594 (12) [back to overview]Overall Survival of Previously Treated and Previously Untreated Participants
NCT00633594 (12) [back to overview]Time to Best Response of Previously Treated and Previously Untreated Participants
NCT00633594 (12) [back to overview]Progression Free Survival (PFS) of Previously Treated and Previously Untreated Participants
NCT00633594 (12) [back to overview]Time to Best Response of Phase I and Phase II Participants
NCT00633594 (12) [back to overview]Progression Free Survival (PFS) of Phase I and Phase II Participants
NCT00633945 (9) [back to overview]Skindex Function
NCT00633945 (9) [back to overview]Patient General Assessment (PtGA) for Skin
NCT00633945 (9) [back to overview]Pain in Skin
NCT00633945 (9) [back to overview]Number of Participants With Change in IFN and CD4 Levels at 6 Weeks
NCT00633945 (9) [back to overview]Fatigue
NCT00633945 (9) [back to overview]Cutaneous Lupus Area and Severity Index (CLASI)
NCT00633945 (9) [back to overview]Itch in Skin
NCT00633945 (9) [back to overview]Skindex Symptoms
NCT00633945 (9) [back to overview]Physician Global Assessment (PGA) for Skin
NCT00642954 (3) [back to overview]Number of Participants Experiencing Best Overall Response Determined by Complete Response (CR), Near Complete Response (NCR), Very Good Partial Response (VGPR), Partial Response (PR), Minimal Response (MR), Stable Disease (SD) or Progressive Disease (PD)
NCT00642954 (3) [back to overview]Number of Participants Experiencing Drug-Related Adverse Events (AEs)
NCT00642954 (3) [back to overview]Number of Participants Experiencing Dose-Limiting Toxicities (DLTs)
NCT00644228 (3) [back to overview]Response Rates ()
NCT00644228 (3) [back to overview]Progression-free Survival
NCT00644228 (3) [back to overview]Overall Survival
NCT00654186 (3) [back to overview]Number of Participants With Overall Clinical Benefit (OCB), Defined as the Sum of Complete Response (CR), Partial Response (PR), and Stable Disease (SD) Divided by the Number of Participants
NCT00654186 (3) [back to overview]Time to PSA Progression
NCT00654186 (3) [back to overview]Time to Disesase Progression as Measured by Radiographic Progression
NCT00655668 (4) [back to overview]Progression-Free Survival
NCT00655668 (4) [back to overview]Duration of Response
NCT00655668 (4) [back to overview]Participants Categorized by Best Response as Determined by Investigator
NCT00655668 (4) [back to overview]Safety
NCT00665652 (1) [back to overview]Change in Total Neuropathy Score in Subjects With MGUS Associated Neuropathy After Treatment With Lenalidomide.
NCT00670358 (3) [back to overview]Event-free > Survival at 12 Months (Phase 2, DLBCL/Mixed Dose Level 3)
NCT00670358 (3) [back to overview]Progression-free > Survival at 24 Months (Phase 2, Transformed/Composite)
NCT00670358 (3) [back to overview]Toxicity as Assessed by NCI CTCAE v3.0 (Phase I)
NCT00675441 (1) [back to overview]Number of Participants' With Treatment Response of Complete or Partial Response
NCT00679367 (3) [back to overview]Number of Participants With Hematologic Response
NCT00679367 (3) [back to overview]Number of Participants Removed From Study Due to Toxicities
NCT00679367 (3) [back to overview]Number of Organs Improved or Stable Based on Description Below:
NCT00684242 (1) [back to overview]Change in Cancer Pain Intensity Determined by Edmonton Symptom Assessment Scale (ESAS)
NCT00687674 (1) [back to overview]Number of Participants With a Grade 3 and 4 Adverse Event (Phase I)
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Constipation Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Diarrhea Domain
NCT00689936 (43) [back to overview]Kaplan Meier Estimates for Time to Second-line Anti-myeloma Treatment (AMT)
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Fatigue Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Financial Difficulties Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Insomnia Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Nausea/Vomiting Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Pain Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain
NCT00689936 (43) [back to overview]Time to First Response Based on the Review by the IRAC
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Dyspnea Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Social Functioning Domain
NCT00689936 (43) [back to overview]Percentage of Participants With an Objective Response After Second-line Anti-myeloma Treatment at the Time of Final Analysis
NCT00689936 (43) [back to overview]Percentage of Participants With a Myeloma Response by Uncertain Risk Cytogenetic Risk Category Based on IRAC Review
NCT00689936 (43) [back to overview]Percentage of Participants With a Myeloma Response by Normal Risk Cytogenetic Risk Category Based on IRAC Review
NCT00689936 (43) [back to overview]Kaplan Meier Estimates of Duration of Myeloma Response as Determined by an Investigator Assessment at Time of Final Analysis
NCT00689936 (43) [back to overview]Percentage of Participants With a Myeloma Response by Favorable Hyperdiploidy Risk Cytogenetic Risk Category Based on IRAC Review
NCT00689936 (43) [back to overview]Percentage of Participants With a Myeloma Response by Adverse Risk Cytogenetic Risk Category Based on IRAC Review.
NCT00689936 (43) [back to overview]Kaplan-Meier Estimates of Progression-free Survival (PFS) Based on the Response Assessment by the Independent Review Adjudication Committee (IRAC)
NCT00689936 (43) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Global Health Status Domain
NCT00689936 (43) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Body Image Scale
NCT00689936 (43) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Disease Symptoms Scale
NCT00689936 (43) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Future Perspective Scale
NCT00689936 (43) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Side Effects Treatment Scale
NCT00689936 (43) [back to overview]Change From Baseline in the European Quality of Life-5 Dimensions (EQ-5D) Health Utility Index Score
NCT00689936 (43) [back to overview]Number of Participants With Adverse Events (AEs) During the Active Treatment Phase
NCT00689936 (43) [back to overview]Shift From Baseline to Most Extreme Postbaseline Value in Absolute Neutrophil Count During the Active Treatment Phase
NCT00689936 (43) [back to overview]Shift From Baseline to Most Extreme Postbaseline Value in Creatinine Clearance (CrCl) During the Active Treatment Phase
NCT00689936 (43) [back to overview]Shift From Baseline to Most Extreme Postbaseline Value in Hemoglobin During the Active Treatment Phase
NCT00689936 (43) [back to overview]Kaplan Meier Estimates of Duration of Myeloma Response as Determined by the IRAC
NCT00689936 (43) [back to overview]Kaplan Meier Estimates of Overall Survival at the Time of Final Analysis (OS)
NCT00689936 (43) [back to overview]Kaplan Meier Estimates of Time to Second Line Therapy AMT at the Time of Final Analysis
NCT00689936 (43) [back to overview]Kaplan Meier Estimates of Time to Treatment Failure (TTF)
NCT00689936 (43) [back to overview]Kaplan Meier Estimates of Time to Treatment Failure (TTF) at the Time of Final Analysis
NCT00689936 (43) [back to overview]Kaplan-Meier Estimates of PFS Based on the Response Assessment by the Investigator At the Time of Final Analysis
NCT00689936 (43) [back to overview]Shift From Baseline to Most Extreme Postbaseline Value in Platelet Count During the Active Treatment Phase.
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Role Functioning Domain
NCT00689936 (43) [back to overview]Time to First Response Based on the Investigator Assessment at the Time of Final Analysis
NCT00689936 (43) [back to overview]Percentage of Participants With an Objective Response Based on IRAC Review
NCT00689936 (43) [back to overview]Percentage of Participants With an Objective Response Based on Investigator Assessment at Time of Final Analysis
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Appetite Loss Domain
NCT00689936 (43) [back to overview]Change From Baseline in the EORTC QLQ-C30 Cognitive Functioning Domain
NCT00691704 (5) [back to overview]Time to Response
NCT00691704 (5) [back to overview]Progression Free Survival
NCT00691704 (5) [back to overview]Time to Progression
NCT00691704 (5) [back to overview]Overall Survival
NCT00691704 (5) [back to overview]Duration of Response
NCT00695786 (1) [back to overview]Number of Participants With Best Overall Disease Response
NCT00717756 (1) [back to overview]Response Rate by Recist Criteria
NCT00724568 (2) [back to overview]Maximum Tolerated Dose (MTD) of Combination Therapy With VELCADE, Dexamethasone, and Doxil, (RVDD)
NCT00724568 (2) [back to overview]The Percentage of Patients That Achieved Partial or Complete Response to Treatment.
NCT00727415 (6) [back to overview]Number of Patients With Severe Infections
NCT00727415 (6) [back to overview]Overall Complete Response (CR) Rate (Phase II)
NCT00727415 (6) [back to overview]Maximum Tolerated Dose of Lenalidomide (Phase I)
NCT00727415 (6) [back to overview]Toxicity as Assessed by NCI CTCAE v3.0
NCT00727415 (6) [back to overview]Correlation Between Complete Response (CR) and Baseline Biologic Parameters (i.e., IgHV, CD38, Etc.).
NCT00727415 (6) [back to overview]Number of Patients Reaching Disease-free Survival (DSF) Overall
NCT00737529 (11) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs)
NCT00737529 (11) [back to overview]Time to Complete Response (CR+CRu) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Time to Response (TTR)
NCT00737529 (11) [back to overview]Overall Survival (OS)
NCT00737529 (11) [back to overview]Percentage of Participants With a Complete Response (CR) /Complete Response Unconfirmed (CRu) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Kaplan Meier Estimate of Duration of Complete Response (DoCR) (CR+CRu) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Kaplan Meier Estimate of Duration of Response (DoR) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Kaplan Meier Estimate of Time to Progression (TTP) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Kaplan-Meier Estimate of Progression-Free Survival (PFS) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Kaplan-Meier Estimate of Time to Treatment Failure (TTF) According to the Independent Review Committee
NCT00737529 (11) [back to overview]Percentage of Participants Who Achieved an Overall Response According to the Independent Review Committee (IRC)
NCT00742560 (12) [back to overview]Progression-free Survival (PFS)
NCT00742560 (12) [back to overview]Time to Progression (TTP)
NCT00742560 (12) [back to overview]Mean Serum Concentrations of Elotuzumab During Cycle 1
NCT00742560 (12) [back to overview]Mean Serum Concentrations of Elotuzumab During Cycle 1
NCT00742560 (12) [back to overview]Number of Participants With Infusion Reactions
NCT00742560 (12) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs)
NCT00742560 (12) [back to overview]Plasma Cell Myeloma Cytogenetic Subtype
NCT00742560 (12) [back to overview]Maximum Tolerated Dose (MTD) of Elotuzumab in Combination With Lenalidomide and Dexamethasone (Phase 1)
NCT00742560 (12) [back to overview]Duration of Response
NCT00742560 (12) [back to overview]Objective Response Rate (ORR) According to the International Myeloma Working Group Uniform Response Criteria (Phase 1)
NCT00742560 (12) [back to overview]Percentage of Participants With Treatment-emergent Anti-elotuzumab Antibody (ADA)
NCT00742560 (12) [back to overview]Objective Response Rate (ORR) According to the International Myeloma Working Group Uniform Response Criteria (Phase 2)
NCT00751296 (2) [back to overview]Percentage of Participants With Progression-free Survival (PFS) and Overall Survival (OS).
NCT00751296 (2) [back to overview]To Assess the Efficacy (Response Rate) of Oral Lenalidomide in the Treatment of Patients With Symptomatic, Previously Untreated, Chronic Lymphocytic Leukemia (CLL)
NCT00759603 (1) [back to overview]Overall Participant Response Rate: Percentage of Participants With Complete + Partial Response According to Revised National Cancer Institute-sponsored Working Group Guidelines
NCT00765245 (3) [back to overview]Number of Patients With Each Worst-Grade Toxicity
NCT00765245 (3) [back to overview]Disease-free Survival at 2 Years
NCT00765245 (3) [back to overview]Disease-free Survival at 1 Year
NCT00772915 (5) [back to overview]Progression-free Survival (PFS) Rate at 12 Months
NCT00772915 (5) [back to overview]Progression-free Survival (PFS)
NCT00772915 (5) [back to overview]Overall Survival (OS)
NCT00772915 (5) [back to overview]Number of Participants Who Experienced at Least One Grade 3 or Higher Adverse Event at Least Possibly Related to Treatment (Toxicity)
NCT00772915 (5) [back to overview]Confirmed Response Rate
NCT00774345 (3) [back to overview]Overall Survival (OS)
NCT00774345 (3) [back to overview]Progression Free Survival 2 (PFS2)
NCT00774345 (3) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00783367 (2) [back to overview]Time Until Progression After Lenalidomide-dexamethasone + Rituximab Therapy in Relapsed Small B-cell Lymphomas With Rituximab Resistance
NCT00783367 (2) [back to overview]Response Rate to Lenalidomide-dexamethasone + Rituximab Therapy in Relapsed Small B-cell Lymphoma With Rituximab Resistance
NCT00784927 (5) [back to overview]Assessment of Tumor Response
NCT00784927 (5) [back to overview]Progression-free Survival Time
NCT00784927 (5) [back to overview]Survival Time
NCT00784927 (5) [back to overview]Time to Treatment Failure
NCT00784927 (5) [back to overview]Tumor Response to Lenalidomide, Rituximab, Cyclophosphamide and Dexamethasone in the Subgroup of Patients With Lymphoplasmacytic Lymphoma (Waldenstrom's Macroglobulinemia).
NCT00790842 (6) [back to overview]Progression-free Survival
NCT00790842 (6) [back to overview]Percentage of Participants Who Experience a Response [sCR, CR, VGPR, PR]
NCT00790842 (6) [back to overview]Overall Survival Time
NCT00790842 (6) [back to overview]Worst Degree Treatment-Related Adverse Events Across All Event Types Per Patient
NCT00790842 (6) [back to overview]Number of Participants in Phase I Component With Dose Limiting Toxicities During the First Cycle of Therapy
NCT00790842 (6) [back to overview]Duration of Response
NCT00792077 (4) [back to overview]ECOG Performance Status
NCT00792077 (4) [back to overview]The Functional Assessment of Chronic Illness Therapy-Fatigue Subscale Score
NCT00792077 (4) [back to overview]Total Sleep Time as Measured by Polysomnography (PSG)
NCT00792077 (4) [back to overview]Epwort Sleep Scale
NCT00807599 (2) [back to overview]Progression Free Survival (PFS) Rate at 2 Years After Enrollment in Untreated Patients With Multiple Myeloma.
NCT00807599 (2) [back to overview]Overall Survival
NCT00812968 (19) [back to overview]Number of Participants With Adverse Events (AE)
NCT00812968 (19) [back to overview]Percentage of Bone Marrow Myeloblasts
NCT00812968 (19) [back to overview]Change From Baseline in Percentage of Bone Marrow Erythroblasts
NCT00812968 (19) [back to overview]Percentage of Bone Marrow Promyelocytes
NCT00812968 (19) [back to overview]Time to Erythroid Response
NCT00812968 (19) [back to overview]Area Under the Plasma Concentration-time Curve From Time Zero to Infinity (AUC∞) of Lenalidomide
NCT00812968 (19) [back to overview]Change From Baseline in Hemoglobin Concentration
NCT00812968 (19) [back to overview]Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUCt) of Lenalidomide
NCT00812968 (19) [back to overview]Area Under the Plasma Concentration-time Curve Over the Dosing Interval (AUCτ) of Lenalidomide
NCT00812968 (19) [back to overview]Terminal Half-life (T1/2) of Lenalidomide
NCT00812968 (19) [back to overview]Time to Maximum Plasma Concentration (Tmax) of Lenalidomide
NCT00812968 (19) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Lenalidomide
NCT00812968 (19) [back to overview]Number of Participants With a Erythroid Response
NCT00812968 (19) [back to overview]Duration of Erythroid Response
NCT00812968 (19) [back to overview]Number of Participants With a Cytogenetic Response
NCT00812968 (19) [back to overview]Apparent Terminal Elimination Rate Constant of Lenalidomide
NCT00812968 (19) [back to overview]Number of Participants With a Neutrophil Response
NCT00812968 (19) [back to overview]Apparent Total Plasma Clearance (CL/F) of Lenalidomide
NCT00812968 (19) [back to overview]Apparent Volume of Distribution (VzF) of Lenalidomide
NCT00831766 (5) [back to overview]Median Overall Survival (OS)
NCT00831766 (5) [back to overview]Rate of Lenalidomide Related Toxicity During Maintenance Therapy
NCT00831766 (5) [back to overview]Median Progression-Free Survival (PFS)
NCT00831766 (5) [back to overview]Phase I: Recommended Phase II Dose
NCT00831766 (5) [back to overview]Phase II: Complete Response Rate of Participants Treated at Maximum Tolerated Dose (MTD)
NCT00837031 (3) [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
NCT00837031 (3) [back to overview]Six-Month Overall Survival (OS) Probability, the Percentage of Patients Estimated to be Alive Six Months After Beginning Protocol Treatment
NCT00837031 (3) [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
NCT00843882 (9) [back to overview]Time to Major Erythroid Response (MER)
NCT00843882 (9) [back to overview]Proportion of Patients With Minor Erythroid Response
NCT00843882 (9) [back to overview]Proportion of Patients With Major Erythroid Response (MER) to Salvage Combination Therapy
NCT00843882 (9) [back to overview]Proportion of Patients With Major Erythroid Response (MER) Among Those With Chromosome 5q31.1 Deletion
NCT00843882 (9) [back to overview]Proportion of Patients With Major Erythroid Response (MER)
NCT00843882 (9) [back to overview]Proportion of Patients With Cytogenetic Response
NCT00843882 (9) [back to overview]Proportion of Patients With Bone Marrow Response
NCT00843882 (9) [back to overview]Pretreatment Endogenous Erythropoietin Level
NCT00843882 (9) [back to overview]Duration of Major Erythroid Response (MER)
NCT00860457 (1) [back to overview]Complete Response Rate
NCT00867308 (2) [back to overview]Response Rate
NCT00867308 (2) [back to overview]Grade 3-4 Toxicity
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Financial Problems Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Global Health Status / QoL Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Insomnia Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Nausea and Vomiting Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Pain Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Role Functioning Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Social Functioning Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Percentage of Participants Who Achieved an Overall Response According to the IRC Central Review
NCT00875667 (47) [back to overview]Percentage of Participants Who Achieved an Overall Response as Assessed by the Investigator at the Final Analysis
NCT00875667 (47) [back to overview]Percentage of Participants With a Complete Response, Unconfirmed Complete Response, Partial Response and Stable Disease According to the IRC Central Review
NCT00875667 (47) [back to overview]Percentage of Participants With a Complete Response, Unconfirmed Complete Response, Partial Response and Stable Disease at the Final Analysis
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Appetite Loss Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Cognitive Functioning Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Constipation
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Diarhoea
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Dyspnoea Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Fatigue Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Financial Problems Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Cognitive Functioning Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Insomnia Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Nausea / Vomiting Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Pain Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Social Functioning Domain
NCT00875667 (47) [back to overview]Number of Participants With Treatment Emergent Adverse Events
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Role Functioning Domain
NCT00875667 (47) [back to overview]Kaplan Meier Estimate for Duration of Response (DOR) According to the IRC Central Review
NCT00875667 (47) [back to overview]Kaplan Meier Estimate for Duration of Response as Assessed by the Investigator at the Final Analysis
NCT00875667 (47) [back to overview]Kaplan Meier Estimate for Overall Survival (OS) According to the IRC Central Review
NCT00875667 (47) [back to overview]Kaplan Meier Estimate for Overall Survival as Assessed by the Investigator at the Final Analysis
NCT00875667 (47) [back to overview]Kaplan Meier Estimate for Progression Free Survival (PFS) by Independent Review Committee (IRC) Central Review
NCT00875667 (47) [back to overview]Kaplan Meier Estimate for Progression Free Survival by Investigator's Assessment at the Final Analysis
NCT00875667 (47) [back to overview]Kaplan Meier Estimate of Time to First Response (TTFR) According to the IRC Central Review
NCT00875667 (47) [back to overview]Kaplan Meier Estimate of Time to First Response as Assessed by the Investigator at the Final Analysis
NCT00875667 (47) [back to overview]Kaplan Meier Estimate of Time to Progression According to the IRC Central Review
NCT00875667 (47) [back to overview]Kaplan Meier Estimate of Time to Progression as Assessed by the Investigator at the Final Analysis
NCT00875667 (47) [back to overview]Kaplan Meier Estimate of Time to Treatment Failure (TTF) as Assessed by the Investigator
NCT00875667 (47) [back to overview]Kaplan Meier Estimate of Time to Treatment Failure as Assessed by the Investigator at the Final Analysis
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Appetite Loss Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Global Health Status / QoL Domain
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Constipation Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Diarhoea Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Dyspnoea Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain to Treatment Discontinuation Visit
NCT00875667 (47) [back to overview]Maximum Change From Baseline in the EORTC QLQ-C30 Fatigue Domain to Treatment Discontinuation Visit
NCT00890552 (4) [back to overview]Event-free Survival (EFS)
NCT00890552 (4) [back to overview]Duration of Response
NCT00890552 (4) [back to overview]Overall Survival (OS)
NCT00890552 (4) [back to overview]Hematologic Response Rate
NCT00890929 (9) [back to overview]Time to CR
NCT00890929 (9) [back to overview]Maximum Tolerated Dose (MTD) of Lenalidomide
NCT00890929 (9) [back to overview]Remission Duration
NCT00890929 (9) [back to overview]OS of Responders
NCT00890929 (9) [back to overview]Overall Response Rate (ORR)
NCT00890929 (9) [back to overview]Overall Survival (OS)
NCT00890929 (9) [back to overview]4-week Survival Rate
NCT00890929 (9) [back to overview]Compete Remission (CR) Rate
NCT00890929 (9) [back to overview]Time to PR
NCT00899431 (1) [back to overview]Percentage of Participants With GVHD (Graft Versus Host Disease)
NCT00903630 (5) [back to overview]Phase 2 - Number of Subjects Who Are Progression-Free and Alive
NCT00903630 (5) [back to overview]Phase 1 - Maximum Tolerated Dose (MTD) of Lenalidomide When Combined With Fixed Dose Liposomal Doxorubicin in Women With Recurrent Epithelial Ovarian, Fallopian Tube, and Primary Peritoneal Cancer
NCT00903630 (5) [back to overview]Phase 1 - Number of Participants Who Experienced Dose-Limiting Toxicities (DLTs)
NCT00903630 (5) [back to overview]Phase 2 - Number of Subjects Achieving a Partial or Complete Response
NCT00903630 (5) [back to overview]Phase 2 - Number of Subjects Who Are Progression-Free and Alive
NCT00908232 (6) [back to overview]Overall Survival
NCT00908232 (6) [back to overview]Time to Progression
NCT00908232 (6) [back to overview]Progression Free Survival
NCT00908232 (6) [back to overview]Median Time to First Confirmed Response
NCT00908232 (6) [back to overview]One Year Survival
NCT00908232 (6) [back to overview]Overall Best Confirmed Response
NCT00910858 (10) [back to overview]PK Phase: Area-under-the Concentration-time Curve (AUC0-24) for Lenalidomide
NCT00910858 (10) [back to overview]Monotherapy Phase: Area-under-the Concentration-time Curve (AUC0-5) for Lenalidomide
NCT00910858 (10) [back to overview]Monotherapy Phase: Maximum Plasma Concentration of Lenalidomide (Cmax)
NCT00910858 (10) [back to overview]Percentage of Participants Overall With Erythroid Response by Baseline Erythropoietin Level
NCT00910858 (10) [back to overview]Percentage of Participants With a Erythroid Response Across All Phases
NCT00910858 (10) [back to overview]PK Phase: Maximum Plasma Concentration of Lenalidomide (Cmax)
NCT00910858 (10) [back to overview]PK Phase: Percent of Administered Lenalidomide Excreted Over 24 Hours After a Single, Oral Dose
NCT00910858 (10) [back to overview]PK Phase: Terminal Half-life (t1/2)
NCT00910858 (10) [back to overview]Monotherapy Phase: Percent of Lenalidomide Excreted Over 5 Hours Post Day 14 Dose
NCT00910858 (10) [back to overview]Time to Grade 4 Neutropenia or Thrombocytopenia
NCT00910910 (14) [back to overview]Number of Participants With Adverse Events With a Later Cut-off Date of 31 March 2014
NCT00910910 (14) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00910910 (14) [back to overview]Number of Participants and Types of Subsequent Anti-cancer Therapies Received Post Treatment
NCT00910910 (14) [back to overview]Time to Response
NCT00910910 (14) [back to overview]Percentage of Participants With the Best Overall Response Based on the International Workshop on Chronic Lymphocytic Leukemia (IWCLL) Guidelines
NCT00910910 (14) [back to overview]Percentage of Participants With a Best Overall Response Based on IWCLL Guidelines With a Later Cut-off Date of 31 March 2014
NCT00910910 (14) [back to overview]Kaplan Meier Estimate of Overall Survival
NCT00910910 (14) [back to overview]Kaplan-Meier Estimate for Duration of Response
NCT00910910 (14) [back to overview]Time to Response for a Later Cut-off Date of 31 March 2014
NCT00910910 (14) [back to overview]Kaplan-Meier Estimate for Duration of Response With a Later Cut-off Date of 31 March 2014
NCT00910910 (14) [back to overview]Kaplan-Meier Estimate of Progression Free Survival (PFS)
NCT00910910 (14) [back to overview]Kaplan Meier Estimate for Overall Survival at the Final Analysis
NCT00910910 (14) [back to overview]Kaplan-Meier Estimate of Progression Free Survival (PFS) With a Later Cut-off Date of 14 March 2014
NCT00910910 (14) [back to overview]Number of Participants Deaths During the Treatment and Survival Follow-Up Phase
NCT00928486 (3) [back to overview]Kaplan-Meier Estimates of Duration of Response (DoR)
NCT00928486 (3) [back to overview]Number of Participants Experiencing Treatment-Emergent Adverse Events (TEAE)
NCT00928486 (3) [back to overview]Myeloma Response Rate
NCT00939510 (3) [back to overview]RECIST-defined Measurable Disease
NCT00939510 (3) [back to overview]Number of Patients With Statistically Significant Change in Immune Response From Baseline to End of Study
NCT00939510 (3) [back to overview]Number of Patients With a PSA Response
NCT00942578 (10) [back to overview]Count of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0) Who Were Administered the Four-Drug Combination
NCT00942578 (10) [back to overview]Count of Participants With a Radiologic Response
NCT00942578 (10) [back to overview]Recommended Phase 2 Dose (RP2D)
NCT00942578 (10) [back to overview]Median Time to Progression (TTP)
NCT00942578 (10) [back to overview]Median Overall Survival of Patients Studied
NCT00942578 (10) [back to overview]Count of Participants With Dose-Limiting Toxicities (DLT)
NCT00942578 (10) [back to overview]Survival Based on Expression of T Cell Immunoglobulin and Mucin Domain (TIM-3) on Cluster of Differentiation 8 (CD8) + T Cells
NCT00942578 (10) [back to overview]Survival Based on Expression of Programmed Cell Death Protein 1 (PD-1) on Cluster of Differentiation 8 (CD8) + T Cells
NCT00942578 (10) [back to overview]Count of Participants With Prostatic Antigen-Specific (PSA) Declines
NCT00942578 (10) [back to overview]Count of Participants With Changes in Circulating Apoptotic Endothelial Cells (CAEC) From Baseline After Drug Administration
NCT00963105 (8) [back to overview]Overall Response Rate (ORR)
NCT00963105 (8) [back to overview]Kaplan-Meier Estimate of Progression Free Survival
NCT00963105 (8) [back to overview]Kaplan-Meier Estimate of Event-Free Survival
NCT00963105 (8) [back to overview]Kaplan-Meier Estimate of Duration of Response
NCT00963105 (8) [back to overview]Kaplan-Meier Estimate of Time to Progression
NCT00963105 (8) [back to overview]Kaplan-Meier Estimate of Overall Survival
NCT00963105 (8) [back to overview]Number of Participants With Treatment-emergent Adverse Events
NCT00963105 (8) [back to overview]Time to Response
NCT00966693 (7) [back to overview]Complete Response(CR) and Very Good Partial Response(VGPR)
NCT00966693 (7) [back to overview]Incidence of Adverse Events
NCT00966693 (7) [back to overview]Progression Free Survival
NCT00966693 (7) [back to overview]Number of Participants With Dose Limitations Toxicities of the Combination of Lenalidomide and Thalidomide and Dexamethasone (LTD) in Patients With Relapsed/Refractory Multiple Myeloma (RRMM)
NCT00966693 (7) [back to overview]Time to Best Response
NCT00966693 (7) [back to overview]Time to Progression
NCT00966693 (7) [back to overview]Time to Next Therapy
NCT00974233 (5) [back to overview]Progression Free Survival
NCT00974233 (5) [back to overview]Overall Survival
NCT00974233 (5) [back to overview]Toxicities Observed With Induction Chemotherapy and Maintenance Therapy
NCT00974233 (5) [back to overview]Progression-free Survival
NCT00974233 (5) [back to overview]Objective Response Rate (Complete + Partial Responses)
NCT00975806 (3) [back to overview]Phase 1: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) While on Both Lenalidomide and Sunitinib
NCT00975806 (3) [back to overview]Phase 1 : Tumor Response Rate According to RECIST 1.1
NCT00975806 (3) [back to overview]Phase 1: Maximum Tolerated Dose (MTD)
NCT00988208 (7) [back to overview]Progression-Free Survival (PFS)
NCT00988208 (7) [back to overview]Percentage of Participants With an Objective Response According to Response Evaluation Criteria in Solid Tumors - RECIST Version 1.1 Criteria
NCT00988208 (7) [back to overview]Percentage of Participants Who Received Post-Study Therapies
NCT00988208 (7) [back to overview]Overall Survival (OS)
NCT00988208 (7) [back to overview]Percentage of Participants With Secondary Primary Malignancies During the Course of the Trial
NCT00988208 (7) [back to overview]Number of Participants With Treatment Emergent Adverse Events (AEs)
NCT00988208 (7) [back to overview]Time to Onset of Secondary Primary Malignancies
NCT00996931 (2) [back to overview]Change in Childhood Autism Rating Scale (CARS)Value From Baseline to 6 Weeks
NCT00996931 (2) [back to overview]Change in TNF-alpha Levels
NCT01002755 (3) [back to overview]Overall Response Rate
NCT01002755 (3) [back to overview]Number of Participants With Tolerance of the Medication Combination
NCT01002755 (3) [back to overview]Progression Free Survival
NCT01011894 (1) [back to overview]Best Response
NCT01016600 (15) [back to overview]Morphologic Leukemia-free State
NCT01016600 (15) [back to overview]Partial Remission Rate (PR)
NCT01016600 (15) [back to overview]Phase I Only - Maximum Tolerated Dose (MTD) as Measured by Dose-limiting Toxicities (DLTs)
NCT01016600 (15) [back to overview]Phase II Only - Complete Remission Rate (CRm + CRi) in Participants With Untreated AML ≥60 Years of Age
NCT01016600 (15) [back to overview]Phase I Only - Maximum Tolerated Dose (MTD)
NCT01016600 (15) [back to overview]Relapse Free Survival (RFS)
NCT01016600 (15) [back to overview]Time to Progression (TTP)
NCT01016600 (15) [back to overview]Response Rate (CRm + CRc + CRi + PR)
NCT01016600 (15) [back to overview]Toxicity Profile (Grade 3/4 Toxicities)
NCT01016600 (15) [back to overview]Overall Survival
NCT01016600 (15) [back to overview]CR With Incomplete Blood Counts Rate
NCT01016600 (15) [back to overview]Cytogenetic CR (CRc) Rate
NCT01016600 (15) [back to overview]Duration of CR for Complete Responders
NCT01016600 (15) [back to overview]Event Free Survival
NCT01016600 (15) [back to overview]Morphologic Complete Remission Rate (CRm)
NCT01021423 (1) [back to overview]Participants With Treatment Emergent Adverse Events (TEAEs)
NCT01029054 (3) [back to overview]The Maximum Tolerated Dose (MTD) of Carfilzomib
NCT01029054 (3) [back to overview]The Percentage of Patients Alive Without Progression
NCT01029054 (3) [back to overview]The Percentage of Patients That Achieve a Response to Treatment
NCT01029262 (29) [back to overview]Mean Change From Baseline in the Emotional Functioning Domain Associated With the EORTC QLQ-C30 Scale at Weeks 12 and 24
NCT01029262 (29) [back to overview]Healthcare Resource Utilization (HRU): Duration of Hospitalizations Due to Adverse Events
NCT01029262 (29) [back to overview]Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline in the Emotional Functioning Domain at Weeks 12 and 24
NCT01029262 (29) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAE)
NCT01029262 (29) [back to overview]Mean Change From Baseline in the Physical Functioning Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24
NCT01029262 (29) [back to overview]Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline Within the Physical Functioning Domain at Weeks 12 and 24
NCT01029262 (29) [back to overview]Percentage of Participants Who Achieved an Erythroid Response Based on the Modified International Working Group (IWG) 2006 Criteria
NCT01029262 (29) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain at Week 12 and 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Global Health Status/Quality of Life (QOL) Domain at Week 12 and 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Fatigue Domain at Week 12 and 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain at Week 12 and 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in the EORTC QLQ-C30 Dyspnea Domain at Week 12 and 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in the Global Health Status/QoL Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in the Dyspnea Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24
NCT01029262 (29) [back to overview]Mean Change From Baseline in Fatigue Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24
NCT01029262 (29) [back to overview]Compliance Rates Using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) From Baseline to Week 48
NCT01029262 (29) [back to overview]Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline in the Dyspnea Domain at Weeks 12 and 24
NCT01029262 (29) [back to overview]Percentage of Participants Who Achieved an Erythroid Response Based on Original IWG 2006 Criteria
NCT01029262 (29) [back to overview]Percentage of Participants With a Erythroid Gene Signature Who Achieved RBC Transfusion Independence for ≥ 56 Days as Determined by an Independent Review Committee (IRC)
NCT01029262 (29) [back to overview]Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline in the Global Health Status/QOL Domain at Weeks 12 and 24
NCT01029262 (29) [back to overview]Percentage of Participants With a Clinically Meaningful Improvement in QOL (EORTC QLQ-C-30 Scale) From Baseline in Fatigue Domain at Weeks 12 and 24
NCT01029262 (29) [back to overview]Time to 56-Day RBC-Transfusion-Independent (TI) Response as Determined by the Sponsor
NCT01029262 (29) [back to overview]Percentage of Participants Who Achieved Red Blood Cell (RBC) Transfusion Independence for ≥ 56 Days as Determined by an Independent Review Committee (IRC)
NCT01029262 (29) [back to overview]Percentage of Participants Who Achieved RBC Transfusion Independence With a Duration of ≥ 24 Weeks (168 Days) as Determined by the Sponsor
NCT01029262 (29) [back to overview]Kaplan Meier Estimates of Duration of 56-day RBC Transfusion Independence Response as Determined by the Sponsor
NCT01029262 (29) [back to overview]Kaplan Meier Estimates for Progression to Acute Myeloid Leukemia (AML)
NCT01029262 (29) [back to overview]Kaplan Meier Estimate for Overall Survival (OS)
NCT01029262 (29) [back to overview]Healthcare Resource Utilization (HRU): Rate of Inpatient Hospitalizations Related to Adverse Events Per Person Year
NCT01029262 (29) [back to overview]Healthcare Resource Utilization (HRU): Number of Days of Hospitalization Due to Adverse Events Per Person-Years
NCT01032291 (3) [back to overview]Best Overall Response Assessed by an Independent Review Using Response Evaluation Criteria In Solid Tumors (RECIST 1.1) During the Proof of Concept Period Prior to Early Study Termination
NCT01032291 (3) [back to overview]Participants With Dose Limiting Toxicities (DLTs) During the First Treatment Cycle of the Safety Lead-In Period
NCT01032291 (3) [back to overview]Participants With Treatment-Emergent Adverse Events (TEAE)
NCT01034592 (7) [back to overview]Duration of Response
NCT01034592 (7) [back to overview]Hemoglobin Concentration
NCT01034592 (7) [back to overview]Red Blood Cell (RBC) Transfusion Independence
NCT01034592 (7) [back to overview]Platelet Response
NCT01034592 (7) [back to overview]Toxicity
NCT01034592 (7) [back to overview]Red Blood Cell (RBC) Transfusions
NCT01034592 (7) [back to overview]Neutrophil Response
NCT01035463 (3) [back to overview]Overall Survival
NCT01035463 (3) [back to overview]Maximum Tolerated Dose of Lenalidomide (Phase I)
NCT01035463 (3) [back to overview]Event-free Survival
NCT01038635 (3) [back to overview]Overall Response: Number of Participants With CR or CRi Response
NCT01038635 (3) [back to overview]Overall Response Rate (ORR) of Lenalidomide in Combination With 5-azacytidine (5-AZA) in Participants With Leukemia
NCT01038635 (3) [back to overview]Number of Dose Limiting Toxicities for Determining Maximum Tolerated Dose (MTD) of Lenalidomide in Combination With 5-azacytidine (5-AZA)
NCT01049945 (6) [back to overview]Progression Free Survival (Phase II)
NCT01049945 (6) [back to overview]Overall Survival (Phase II)
NCT01049945 (6) [back to overview]Event Free Survival (Phase II)
NCT01049945 (6) [back to overview]Dose Limiting Toxicity of Bendamustine Hydrochloride and Lenalidomide in Combination With Dexamethasone (Phase I)
NCT01049945 (6) [back to overview]Confirmed Response Rate (Dose Level 4) Reported as the Percentage of Patients Achieving a Confirmed Response (sCR, CR, VGPR, or PR).
NCT01049945 (6) [back to overview]Duration of Response (DOR) (Phase II)
NCT01050790 (6) [back to overview]Complete Response Rate at 6 Months
NCT01050790 (6) [back to overview]Time to Progression Post Transplant
NCT01050790 (6) [back to overview]CTA Expression Before and After Azacitidine Therapy
NCT01050790 (6) [back to overview]Feasibility to Mobilize and Infuse Autologous Lymphocytes (ALI) After Immunomodulatory Therapy and After Stem Cell Transplant Engraftment
NCT01050790 (6) [back to overview]Progression-free and Overall Survival
NCT01050790 (6) [back to overview]Toxicity as Assessed by NCI CTCAE v3.0
NCT01054144 (5) [back to overview]Combined Therapy - Median Progression Free Survival
NCT01054144 (5) [back to overview]Number of Participants With 1 Year Overall Survival (OS)
NCT01054144 (5) [back to overview]Number of Participants With Serious Adverse Events
NCT01054144 (5) [back to overview]Single Agent - Median Progressive Free Survival (PFS)
NCT01054144 (5) [back to overview]Response Rate
NCT01057121 (4) [back to overview]Tumor Response Rate
NCT01057121 (4) [back to overview]Maximum Tolerated Dose of Lenalidomide Defined as the Dose Level at Which 0/6 or 1/6 Subjects Experience Dose Limiting Toxicity (DLT) With the Next Higher Dose Having at Least 2/3 or 2/6 Subjects Encountering DLT (Phase I)
NCT01057121 (4) [back to overview]Time to Death
NCT01057121 (4) [back to overview]Time to Response
NCT01060384 (2) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Lenalidomide
NCT01060384 (2) [back to overview]Phase I and Phase II: Event Free Survival and Overall Survival
NCT01075321 (5) [back to overview]Overall Survival for All Eligible Patients
NCT01075321 (5) [back to overview]Progression-Free Survival For All Eligible Patients
NCT01075321 (5) [back to overview]Time to Treatment Failure for All Eligible Patients
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
NCT01076543 (5) [back to overview]Incidence of Dose-limiting Toxicity (DLT), Phase I Patients Only
NCT01076543 (5) [back to overview]Complete Response (Phase II)
NCT01076543 (5) [back to overview]Overall Response Rate (Phase II)
NCT01076543 (5) [back to overview]Overall Survival (OS) (Phase II)
NCT01076543 (5) [back to overview]Progression-free Survival (PFS) (Phase II)
NCT01079936 (4) [back to overview]Participants With Grade 3 =/> Adverse Events
NCT01079936 (4) [back to overview]Number of Participants With Response (CR at Day 90)
NCT01079936 (4) [back to overview]Number of Participants With Day 30 DLT (Overall Study, Phase I/Phase II)
NCT01079936 (4) [back to overview]Maximum Tolerated Dose (MTD) of Lenalidomide
NCT01080391 (7) [back to overview]Overall Response Rate
NCT01080391 (7) [back to overview]Duration of Response
NCT01080391 (7) [back to overview]Duration of Disease Control
NCT01080391 (7) [back to overview]Disease Control Rate
NCT01080391 (7) [back to overview]Quality of Life Core Module (QLQ-C30) Global Health Status/Quality of Life Scores
NCT01080391 (7) [back to overview]Progression-free Survival (PFS)
NCT01080391 (7) [back to overview]Overall Survival
NCT01088048 (15) [back to overview]Sub-study: Plasma Concentration of IDELA (Cohorts 1-4)
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 7)
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 6)
NCT01088048 (15) [back to overview]Time to Response
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 1, Cohorts 2 and 3, Cohort 5)
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]Plasma Concentration of Bendamustine
NCT01088048 (15) [back to overview]Plasma Concentration of IDELA (Cohort 4)
NCT01088048 (15) [back to overview]Duration of Exposure to IDELA
NCT01088048 (15) [back to overview]Plasma Concentration of Lenalidomide
NCT01088048 (15) [back to overview]Plasma Concentration of Everolimus
NCT01088048 (15) [back to overview]Toxicity of Administration of IDELA
NCT01093183 (4) [back to overview]Proportion of Patients Achieving CR
NCT01093183 (4) [back to overview]Overall Survival
NCT01093183 (4) [back to overview]Number of Patients Achieving Objective PSA Response (50% Decrease in PSA Levels Sustained for at Least 4 Weeks) as Defined by PSA Working Group Criteria
NCT01093183 (4) [back to overview]Maximum Tolerated Dose of Lenalidomide Administered in Combination With Oral Cyclophosphamide (Phase I)
NCT01109004 (10) [back to overview]Percentage of Participants With Overall Survival (OS)
NCT01109004 (10) [back to overview]Percentage of Participants With Disease Progression
NCT01109004 (10) [back to overview]MOS SF-36 Physical Component Summary
NCT01109004 (10) [back to overview]Percentage of Participants With Treatment-related Mortality (TRM)
NCT01109004 (10) [back to overview]Number of Participants With Treatment Response
NCT01109004 (10) [back to overview]FACT-G Total Score
NCT01109004 (10) [back to overview]MOS SF-36 Mental Component Summary
NCT01109004 (10) [back to overview]FACT-BMT Score
NCT01109004 (10) [back to overview]Percentage of Participants With Progression-free Survival (PFS)
NCT01109004 (10) [back to overview]FACT-BMT Trial Outcome Index
NCT01121757 (4) [back to overview]Overall Response
NCT01121757 (4) [back to overview]Response Predicted by Molecular Signatures Compared to True Response
NCT01121757 (4) [back to overview]Number of Participants With Grade 3 and 4 Toxicities
NCT01121757 (4) [back to overview]Overall Response
NCT01123356 (4) [back to overview]Frequency of Adverse and Severe Adverse Events
NCT01123356 (4) [back to overview]Dose Reductions Due to Adverse Events.
NCT01123356 (4) [back to overview]Frequency of Adverse Events
NCT01123356 (4) [back to overview]Overall Response Rate
NCT01125176 (5) [back to overview]Time to Response
NCT01125176 (5) [back to overview]Overall Survival
NCT01125176 (5) [back to overview]Progression Free Survival
NCT01125176 (5) [back to overview]Overall Response Rate as Defined as the Number of Patients Who Experience a Response (Complete or Partial) to Treatment at the Time of Best Response
NCT01125176 (5) [back to overview]Duration of Response
NCT01133275 (2) [back to overview]Number of Participants With Erythroid Response
NCT01133275 (2) [back to overview]Number of Participants With Grade 3 or 4 Adverse Events Possibly Related to Treatment
NCT01133665 (38) [back to overview]Number of Participants With Headache Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Hyperglycemia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Hypoalbuminemia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Hypocalcaemia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Hypokalemia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Diarrhea Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Hypophosphatemia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Aspartate Aminotransferase Increased Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Anorexia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Anemia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Alkaline Phosphatase Increased Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Alanine Aminotransferase Increased Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Acneiform Rash Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Correlate the Presence of Specific Fc RIIIa Polymorphisms With Progression-free Survival in Subjects Receiving Cetuximab and Lenalidomide for SCCHN.
NCT01133665 (38) [back to overview]Number of Participants With Back Pain Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Weight Loss Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Vomiting Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Thrombocytopenia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Pain Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Oral Mucositis Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Neutropenia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Neck Pain Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Blood Bilirubin Increased Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With White Blood Cell Decreased Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Xerostomia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With C. Diff Infection Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Constipation Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Hyponatremia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Infusion Related Reaction Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Lymphocyte Count Increased Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Lymphopenia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Maculopapular Rash Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Nausea Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Dyspnea Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Fatigue Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Febrile Neutropenia Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Fever Related to Cetuximab/Lenalidomide
NCT01133665 (38) [back to overview]Number of Participants With Peripheral Sensory Neuropathy Related to Cetuximab/Lenalidomide
NCT01142232 (3) [back to overview]Phase I: Number of Patients With Dose Limiting Toxicity
NCT01142232 (3) [back to overview]Phase II: Overall Response Rate
NCT01142232 (3) [back to overview]Phase II: Treatment-Related Adverse Events Grade 3 or Higher
NCT01145495 (3) [back to overview]Number of Participants Who Achieved a Complete Response
NCT01145495 (3) [back to overview]Toxicity of Study Treatment, Assessed by the NCI Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0
NCT01145495 (3) [back to overview]Disease Progression
NCT01155583 (5) [back to overview]Percent of Participants With Clinical Benefit and Response According to International Response Criteria
NCT01155583 (5) [back to overview]Percent of Participants With Clinical Benefit and Response According to International Response Criteria
NCT01155583 (5) [back to overview]Phase I: Highest Tolerated Low Dose (HTLD)
NCT01155583 (5) [back to overview]Overall Survival
NCT01155583 (5) [back to overview]Median Progression-free Survival (PFS)
NCT01160484 (7) [back to overview]Follow-up Time
NCT01160484 (7) [back to overview]Progression-free Survival
NCT01160484 (7) [back to overview]Duration of Response
NCT01160484 (7) [back to overview]Time to Best Response
NCT01160484 (7) [back to overview]Time to First Response
NCT01160484 (7) [back to overview]Time to Progression
NCT01160484 (7) [back to overview]International Myeloma Working Group (IMWG) Response Criteria
NCT01169337 (7) [back to overview]Proportion of Participants With Response (Phase III Secondary Endpoint)
NCT01169337 (7) [back to overview]Proportion of Patients With Grade 3 Adverse Events That Effect Vital Organ Function or Any Grade 4 or Higher Non-hematologic Adverse Events (Phase II Primary Endpoint)
NCT01169337 (7) [back to overview]1-year Progression-free Survival (PFS) Rate (Phase III Secondary Endpoint)
NCT01169337 (7) [back to overview]2-year Overall Survival (OS) Rate (Phase III Secondary Endpoint)
NCT01169337 (7) [back to overview]2-year Progression-free Rate (Phase III Secondary Endpoint)
NCT01169337 (7) [back to overview]2-year Progression-free Survival (PFS) Rate (Phase III Primary Endpoint)
NCT01169337 (7) [back to overview]Proportion of Participants With Response (Phase II Secondary Endpoint)
NCT01197560 (9) [back to overview]Stage 1: Percentage of Participants With an Overall Response According to the IWG Response Criteria Based on the Investigators Assessment at the Final Data Cut During the Core Treatment Phase
NCT01197560 (9) [back to overview]Stage 1: Percentage of Participants With a Complete Response According to the IWG Response Criteria as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase
NCT01197560 (9) [back to overview]Stage 1: Kaplan Meier Estimates of Overall Survival As Assessed by the Investigators at the Final Data Cut During The Core Treatment Phase
NCT01197560 (9) [back to overview]Number of Participants With Treatment Emergent Events (TEAEs) in the Overall Treatment Phase by Initial Treatment Assignment
NCT01197560 (9) [back to overview]Stage 1: Percentage of Participants With an Overall Response According to the International Working Group (IWG) Response Criteria for Non Hodgkin's Lymphoma (NHL), Cheson 1999 and Evaluated by the Independent Response Adjudication Committee (IRAC)
NCT01197560 (9) [back to overview]Stage 1: Percentage of Participants With a Durable Overall Response (dORR) According to the IWG Response Criteria as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase
NCT01197560 (9) [back to overview]Stage 1: Kaplan Meier Estimates of Progression-Free Survival As Assessed By The Investigators At The Final Data Cut During The Core Treatment Phase
NCT01197560 (9) [back to overview]Stage 1: Kaplan Meier Estimates of Duration of Overall Response (DoR) as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase
NCT01197560 (9) [back to overview]Stage 1: Kaplan Meier Estimates of Duration of Complete Response (DoCR) as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase
NCT01199575 (2) [back to overview]Overall Response Rate (ORR)
NCT01199575 (2) [back to overview]Adverse Events to Study Treatment
NCT01208051 (6) [back to overview]Overall Survival (Final Results After Crossover)
NCT01208051 (6) [back to overview]Percent Change in Tumor Size (Phase II)
NCT01208051 (6) [back to overview]Progression-free Survival (Final Results After Crossover)
NCT01208051 (6) [back to overview]Progression-free Survival (Phase II Futility Analysis)
NCT01208051 (6) [back to overview]Dose Limiting Toxicity
NCT01208051 (6) [back to overview]Objective Response Rate
NCT01208662 (17) [back to overview]5-Year Overall Survival (OS)
NCT01208662 (17) [back to overview]Partial Response (PR) Rate
NCT01208662 (17) [back to overview]5-Year Time to Progression (TTP)
NCT01208662 (17) [back to overview]Change From Baseline on the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACT/GOG-NTX)
NCT01208662 (17) [back to overview]Median Maintenance Treatment Duration
NCT01208662 (17) [back to overview]Median Event Free Survival (EFS)
NCT01208662 (17) [back to overview]Quality-Adjusted Life Years (QALYs)
NCT01208662 (17) [back to overview]5-year Cumulative Incidence of Second Primary Malignancy (SPM)
NCT01208662 (17) [back to overview]Median Progression-Free Survival (PFS)
NCT01208662 (17) [back to overview]Subsequent Therapy Rate
NCT01208662 (17) [back to overview]Very Good Partial Response (VGPR) Rate
NCT01208662 (17) [back to overview]Change From Baseline on the European Organization for Research and Treatment of Cancer Quality-of-Life Questionnaire Core 30 (EORTC QLQ-C30): Global Health Status/Quality of Life (QoL) Sub-scale
NCT01208662 (17) [back to overview]Median Treatment Duration
NCT01208662 (17) [back to overview]Next Generation Sequencing (NGS) Minimal Residual Disease Negative (MRD-) Rate at Maintenance Start
NCT01208662 (17) [back to overview]Median Duration of Response: Partial Response (DOR PR)
NCT01208662 (17) [back to overview]Grade 3 or Higher Treatment-Related Non-Hematologic Adverse Event (AE) Rate
NCT01208662 (17) [back to overview]Complete Response (CR) Rate
NCT01215344 (2) [back to overview]The Percent of Patients With Minimal Residual Disease (MRD) Status Changing to Negative at Day 100 (Post-AHCT), Among Patients With MRD Positive at the End of Induction (EOI).
NCT01215344 (2) [back to overview]Progression Free Survival by MRD Status at Day 100.
NCT01216683 (25) [back to overview]3-year Progression-free Survival Rate
NCT01216683 (25) [back to overview]1-year Post-induction Disease-free Survival (DFS) Rate
NCT01216683 (25) [back to overview]1-year Disease-free Survival (DFS) Rate
NCT01216683 (25) [back to overview]1-year Disease-free Survival (DFS) Rate
NCT01216683 (25) [back to overview]Complete Remission (CR) Rate
NCT01216683 (25) [back to overview]Complete Remission (CR) Rate
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy - General (FACT-G) Total Score at Baseline
NCT01216683 (25) [back to overview]Complete Remission (CR) Rate
NCT01216683 (25) [back to overview]Proportion of Patients With Grade 3 or Higher Peripheral Neuropathy
NCT01216683 (25) [back to overview]5-year Overall Survival Rate
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) Subscale Score at the End of Induction Treatment
NCT01216683 (25) [back to overview]Progression-free Survival
NCT01216683 (25) [back to overview]Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Subscale Score at Mid-induction Treatment
NCT01216683 (25) [back to overview]Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Subscale Score at End of Induction Treatment
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy - General (FACT-G) Total Score at End of Induction Treatment
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) Subscale Score at Baseline
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) Subscale Score at End of Induction Treatment
NCT01216683 (25) [back to overview]Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Subscale Score at Baseline
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) Subscale Score at Mid-induction Treatment
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) Subscale Score at Baseline
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) Subscale Score at Mid-induction Treatment
NCT01216683 (25) [back to overview]5-year Overall Survival Rate
NCT01216683 (25) [back to overview]5-year Overall Survival Rate
NCT01216683 (25) [back to overview]Functional Assessment of Cancer Therapy - General (FACT-G) Total Score at Mid-treatment
NCT01216683 (25) [back to overview]3-year Progression-free Survival Rate
NCT01217957 (18) [back to overview]Phase 2: 1 Year Survival Rate
NCT01217957 (18) [back to overview]Phase 2: Duration of Response (DOR)
NCT01217957 (18) [back to overview]Phase 2: Objective Response Rate (ORR) Following Treatment With the Combination Of Oral Ixazomib, Lenalidomide And Low-Dose Dexamethasone
NCT01217957 (18) [back to overview]Phase 2: Overall Response Rate (ORR)
NCT01217957 (18) [back to overview]Phase 2: Overall Survival (OS)
NCT01217957 (18) [back to overview]Phase 2: Progression Free Survival (PFS)
NCT01217957 (18) [back to overview]Phase 2: Time to Best Response
NCT01217957 (18) [back to overview]Phase 1: Maximum Tolerated Dose (MTD) of Ixazomib Administered Weekly in Combination With Lenalidomide and Low-Dose Dexamethasone
NCT01217957 (18) [back to overview]Phase 1: Rac: Accumulation Ratio of Ixazomib
NCT01217957 (18) [back to overview]Phase 1: Tmax: Time to Reach the Maximum Observed Plasma Concentration (Cmax) for Ixazomib
NCT01217957 (18) [back to overview]Phase 2: Time to Progression (TTP)
NCT01217957 (18) [back to overview]Phase 1: AUC(0-168): Area Under the Plasma Concentration-Time Curve From Time 0 to 168 Hours Postdose for Ixazomib
NCT01217957 (18) [back to overview]Phase 1: Cmax: Maximum Observed Plasma Concentration for Ixazomib
NCT01217957 (18) [back to overview]Phase 1: Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs) as a Measure of Safety and Tolerability
NCT01217957 (18) [back to overview]Phase 2: Percentage of Participants With Complete Response (CR) and Very Good Partial Response (VGPR)
NCT01217957 (18) [back to overview]Phase 2: Percentage of Participants With Complete Response (CR), Stringent Complete Response (sCR), Very Good Partial Response (VGPR), Near Complete Response (nCR), Partial Response (PR) and Minimal Response (MR)
NCT01217957 (18) [back to overview]Phase 2: Percentage of Participants With Grade 3 or Higher AEs, SAEs and Treatment Discontinuation
NCT01217957 (18) [back to overview]Phase 1: Recommended Phase 2 Dose of Ixazomib Given in Combination With Lenalidomide and Low-Dose Dexamethasone
NCT01222195 (1) [back to overview]Number of Patients With a Transfusion Independence Response
NCT01239797 (6) [back to overview]Median Progression Free Survival (PFS)
NCT01239797 (6) [back to overview]Objective Response Rate (ORR)
NCT01239797 (6) [back to overview]Median Progression Free Survival (PFS) - Extended Collection
NCT01239797 (6) [back to overview]Median Overall Survival (OS)
NCT01239797 (6) [back to overview]Change From Baseline of Mean Score Pain Severity (BPI-SF)
NCT01239797 (6) [back to overview]Change From Baseline of Mean Score Pain Interference (BPI-SF)
NCT01241734 (7) [back to overview]Stage II - Overall Response Rate
NCT01241734 (7) [back to overview]Stage I - Dose Limiting Toxicity Incidence Rate
NCT01241734 (7) [back to overview]Stage I - Safety (Type, Frequency, Severity and Relationship of Adverse Events to Study Treatment) and Tolerability
NCT01241734 (7) [back to overview]Stage II - 1 Year Overall Survival (OS)
NCT01241734 (7) [back to overview]Stage II - 1 Year Progression Free Survival (PFS)
NCT01241734 (7) [back to overview]Stage II - 2 Year Overall Survival (OS)
NCT01241734 (7) [back to overview]Stage II - 2 Year Progression Free Survival (PFS)
NCT01241786 (4) [back to overview]The Overall Survival of Patients Treated With the Combination of Lenalidomide and Azacitidine
NCT01241786 (4) [back to overview]Assess for Treatment Related Toxicity Following Administration of Lenalidomide/ Azacitidine.
NCT01241786 (4) [back to overview]the Rate of Response to the Combination of Azacitidine + Lenalidomide in Select Patients
NCT01241786 (4) [back to overview]The Progression Free Survival of Patients Treated With the Combination of Lenalidomide and Azacitidine
NCT01243944 (13) [back to overview]Duration of Reduction in Spleen Volume
NCT01243944 (13) [back to overview]The Percentage of Participants Who Achieved Durable Spleen Volume Reduction at Week 48
NCT01243944 (13) [back to overview]The Percentage of Participants Achieving a Durable Complete or Partial Clinicohematologic Response at Week 48
NCT01243944 (13) [back to overview]The Percentage of Participants Who Achieved a Durable Hematocrit Control at Week 48
NCT01243944 (13) [back to overview]The Percentage of Participants Who Achieved a Durable Complete Hematological Remission at Week 48
NCT01243944 (13) [back to overview]The Percentage of Participants Who Achieved Overall Clinicohematologic Response at Week 32
NCT01243944 (13) [back to overview]Estimated Duration of the Primary Response
NCT01243944 (13) [back to overview]Duration of the Absence of Phlebotomy Eligibility
NCT01243944 (13) [back to overview]Estimated Duration of the Complete Hematological Remission
NCT01243944 (13) [back to overview]Duration of The Overall Clinicohematologic Response
NCT01243944 (13) [back to overview]The Percentage of Participants Achieving Complete Hematological Remission at Week 32
NCT01243944 (13) [back to overview]The Percentage of Participants Achieving a Primary Response at Week 32
NCT01243944 (13) [back to overview]The Percentage of Participants Achieving a Durable Primary Response at Week 48
NCT01246076 (5) [back to overview]Time to Discontinuation of Treatment
NCT01246076 (5) [back to overview]Number of Participants With Confirmed Responses (Complete Remission, Partial Remission, or Hematologic Improvement) as Defined by the International Working Group Criteria
NCT01246076 (5) [back to overview]Toxicity as Measured by Number of Participants Who Experienced Related Grade 3-5 Adverse Events Based on CTCAE Version 4
NCT01246076 (5) [back to overview]Duration of Response
NCT01246076 (5) [back to overview]Overall Survival Rate
NCT01303965 (8) [back to overview]Phase I: Number of Participants With Dose Limiting Toxicity
NCT01303965 (8) [back to overview]Phase II - Time to Platelet Engraftment
NCT01303965 (8) [back to overview]Phase II - Time to Neutrophil Engraftment
NCT01303965 (8) [back to overview]Phase II - Percent of Patients With Treatment-related Deaths at 100 Days
NCT01303965 (8) [back to overview]Phase II - Percent of Patients With Acute Graft Versus Host Disease (GvHD)
NCT01303965 (8) [back to overview]Phase II - Percent of Patients With Chronic Graft Versus Host Disease (GvHD)
NCT01303965 (8) [back to overview]Phase II - Percent of Patients With Treatment-related Deaths at 1 Year
NCT01303965 (8) [back to overview]Phase II: Percent of Patients Alive and Free of Progression at 12 Months Following Transplant
NCT01335399 (5) [back to overview]Progression Free Survival (PFS) Rate at Specific Time-points
NCT01335399 (5) [back to overview]Mean Change From Baseline of Pain Severity Score and Pain Interference Score
NCT01335399 (5) [back to overview]Progression-Free Survival (PFS)
NCT01335399 (5) [back to overview]Overall Survival (OS)
NCT01335399 (5) [back to overview]Objective Response Rate (ORR)
NCT01342172 (4) [back to overview]Maximum Tolerated Dose (MTD) of Lenalidomide
NCT01342172 (4) [back to overview]Best Overall Response
NCT01342172 (4) [back to overview]Number of Grade >=3 Adverse Events
NCT01342172 (4) [back to overview]The Objective Response Rate to Treatment With Gemcitabine, Cisplatin, Plus Lenalidomide
NCT01348919 (8) [back to overview]Maximum Tolerated Dose of CEP-18770
NCT01348919 (8) [back to overview]Duration of Response (DOR) for Participants Treated With CEP-18770 at the MTD, as Assessed Using IMWG Criteria
NCT01348919 (8) [back to overview]Overall Response Rate (ORR) in Participants Treated at the (Maximum Tolerated Dose) MTD, as Assessed Using International Myeloma Working Group (IMWG) Criteria
NCT01348919 (8) [back to overview]Number of Participants With Adverse Events (AEs)
NCT01348919 (8) [back to overview]Time to Reach Cmax (Tmax) of CEP-18770
NCT01348919 (8) [back to overview]Maximum Observed Plasma Concentration (Cmax) of CEP-18770
NCT01348919 (8) [back to overview]Area Under the Plasma Concentration-Time Curve From Time 0 to t (AUC0-t) of CEP-18770
NCT01348919 (8) [back to overview]Time to Progression (TTP) for Participants Treated With CEP-18770 at the MTD, as Assessed Using IMWG Criteria
NCT01349569 (4) [back to overview]Grade 3-4 Toxicity
NCT01349569 (4) [back to overview]Response Conversion Rate
NCT01349569 (4) [back to overview]Time to Response
NCT01349569 (4) [back to overview]Tumor-specific Immunity as Assessed by Percentage of CD3+/CSFSE-low/IFN-gamma+ Cells
NCT01355705 (4) [back to overview]Time-to-next Treatment
NCT01355705 (4) [back to overview]Response Rates After Amrubicin + Lenalidomide + Dexamethasone, Per International Myeloma Working Group Uniform Response Criteria
NCT01355705 (4) [back to overview]Duration of Response (DOR)
NCT01355705 (4) [back to overview]Progression-free Survival (PFS)
NCT01358734 (6) [back to overview]Percentage of Participants Alive at One Year
NCT01358734 (6) [back to overview]Kaplan Meier Estimates for One Year Survival
NCT01358734 (6) [back to overview]Percentage of Participants With 30-Day Treatment-Related Mortality
NCT01358734 (6) [back to overview]Number of Participants With a Second Primary Malignancy
NCT01358734 (6) [back to overview]Overall Survival
NCT01358734 (6) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAE)
NCT01373229 (4) [back to overview]Progression-free Survival (PFS)
NCT01373229 (4) [back to overview]Overall Survival (OS)
NCT01373229 (4) [back to overview]Overall Response (Complete Response/Partial Response)
NCT01373229 (4) [back to overview]Maximum Tolerated Dose
NCT01373294 (2) [back to overview]Treatment Emergent Adverse Events (AEs) or Serious Adverse Events (SAEs)
NCT01373294 (2) [back to overview]Arm A: Progression Free Survival (PFS)
NCT01374217 (4) [back to overview]Duration of Response
NCT01374217 (4) [back to overview]Quality of Life Scores
NCT01374217 (4) [back to overview]Time to Progression
NCT01374217 (4) [back to overview]Response Rate
NCT01375140 (1) [back to overview]Participants With Objective Response
NCT01380106 (2) [back to overview]Serious Adverse Events
NCT01380106 (2) [back to overview]Duration Until Best Response (at Least MR or Minimal Response)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Very Good Partial Response (VGPR)
NCT01383928 (29) [back to overview]Phase 2: Time to Response
NCT01383928 (29) [back to overview]Progression Free Survival (PFS)
NCT01383928 (29) [back to overview]Time to Disease Progression (TTP)
NCT01383928 (29) [back to overview]Phase 1: AUC(0-72): Area Under the Plasma Concentration-Time Curve From Time 0 to 72 Hours Postdose for Ixazomib
NCT01383928 (29) [back to overview]Phase 1: Number of Participants With Clinically Significant Change From Baseline in Vital Signs
NCT01383928 (29) [back to overview]Phase 1: Number of Participants With Markedly Abnormal Laboratory Values Reported as Treatment Emergent Adverse Events (TEAEs)
NCT01383928 (29) [back to overview]Phase 1: Number of Participants With Treatment-Emergent Adverse Events (TEAE) Related to Neurotoxicity
NCT01383928 (29) [back to overview]Phase 1: Percentage of Participants Experiencing 1 or More Treatment-Emergent Adverse Events (TEAEs) or Serious Adverse Events (SAEs)
NCT01383928 (29) [back to overview]Phase 1: Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Ixazomib
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Complete Response (CR) and Very Good Partial Response (VGPR) After Cycles 4, 8, and 16
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Stringent Complete Response (sCR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Minimal Response (MR)
NCT01383928 (29) [back to overview]Phase 1: Recommended Phase 2 Dose (RP2D)
NCT01383928 (29) [back to overview]Kaplan-Meier Estimate of Percentage of Participants Achieving Survival at Year 1
NCT01383928 (29) [back to overview]Overall Survival
NCT01383928 (29) [back to overview]Phase 1: Maximum Tolerated Dose (MTD)
NCT01383928 (29) [back to overview]Phase 1: Percentage of Participants With Best Overall Response
NCT01383928 (29) [back to overview]Phase 1: Rac: Accumulation Ratio of Ixazomib
NCT01383928 (29) [back to overview]Phase 1: Cmax: Maximum Plasma Concentration for Ixazomib
NCT01383928 (29) [back to overview]Phase 2: Duration of Response (DOR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants Experiencing Serious Adverse Events
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Complete Response (CR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Complete Response (CR) + Very Good Partial Response (VGPR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Grade 3 or Higher Adverse Events
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Near Complete Response (nCR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Overall Response (CR+VGPR+PR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Partial Response (PR)
NCT01383928 (29) [back to overview]Phase 2: Percentage of Participants With Treatment-Emergent Adverse Events Resulting in Study Drug Discontinuation
NCT01393964 (11) [back to overview]Number of Participants With Worst Toxicity Grade Chemistry Laboratory Tests
NCT01393964 (11) [back to overview]Number of Participants With Worst Toxicity Grade Hematology Laboratory Tests
NCT01393964 (11) [back to overview]Number of Participants With Worst Toxicity Grade Renal and Liver Function Laboratory Tests
NCT01393964 (11) [back to overview]Geometric Mean Apparent Volume of Distribution (Vz) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method
NCT01393964 (11) [back to overview]Geometric Mean Maximum Observed Serum Concentration (Cmax) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method
NCT01393964 (11) [back to overview]Geometric Mean Total Body Clearance (CLT) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method
NCT01393964 (11) [back to overview]Mean Terminal-phase Elimination Half-life (T-Half) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method
NCT01393964 (11) [back to overview]Median Time to Maximal Concentration (Tmax) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method
NCT01393964 (11) [back to overview]Geometric Mean Area Under Serum Concentration-Time Curve From Time Zero to Time of Last Quantifiable Concentration AUC(0-T) and From Time Zero Extrapolated to Infinite Time AUC(INF) of Elotuzumab Following Cycle 1, Day 1 - Grouping by C-G CrCl Method
NCT01393964 (11) [back to overview]Number of Participants With Persistent Elotuzumab Anti-drug Antibodies (ADA) and Number of Participants ADA Positive at Cycle 2 Pre-dose.
NCT01393964 (11) [back to overview]Number of Participants With Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, and Who Died
NCT01401322 (1) [back to overview]Time-to-Progression (TTP)
NCT01402284 (7) [back to overview]Rate of Minimal Residual Disease (MRD) by Flow Cytometry
NCT01402284 (7) [back to overview]Percentage of Responders With Duration of Response (DOR) at 48 Months
NCT01402284 (7) [back to overview]Overall Survival (OS) Rate
NCT01402284 (7) [back to overview]Overall Response Rate
NCT01402284 (7) [back to overview]Complete Response (CR) and Minimal Residual Disease Neg (MRDneg) CR Rates at Treatment Intervals With Carfilzomib, Lenalidomide & Dexamethasone (CRd) in New Multiple Myeloma Patients After 8 Cycles of Induction, 1 Year Maintenance, and 2 Years Maintenance
NCT01402284 (7) [back to overview]Number of Participants With Serious and Non-serious Adverse Events
NCT01402284 (7) [back to overview]Progression Free Survival (PFS) at 48 Months
NCT01403246 (1) [back to overview]Number of Dose Limiting Toxic Events (DLT) of Lenalidomide Given in Combination With Chlorambucil.
NCT01419795 (6) [back to overview]Incidences of Grades II-IV Acute GVHD and Limited or Extensive Chronic GVHD
NCT01419795 (6) [back to overview]Improvement in Overall Survival of Patients Receiving Lenalidomide With or Without Rituximab in Comparison to Historical Controls Managed by Single or Multiple Chemotherapeutic Agents or Donor Lymphocyte Infusion (DLI) (Cohort 1)
NCT01419795 (6) [back to overview]Rate of Response (CR, PR, or SD) and Time to Progression
NCT01419795 (6) [back to overview]Grade III-IV Toxicity in Patients Receiving Lenalidomide With or Without Rituximab
NCT01419795 (6) [back to overview]Comparison of Rates of Overall Response and Complete Remission Between the First, Second, and Third Cohorts
NCT01419795 (6) [back to overview]Comparison of Incidences of Adverse Events Between the First, Second, and Third Cohorts
NCT01421186 (8) [back to overview]Pharmacokinetics: AUC Cycle 1+2 - Area Under the Time/Concentration Curve for MOR202
NCT01421186 (8) [back to overview]Pharmacokinetics: Cmax - Maximum Observed Serum Concentration for MOR202
NCT01421186 (8) [back to overview]Progression-free Survival
NCT01421186 (8) [back to overview]Time to Progression
NCT01421186 (8) [back to overview]Determination of Maximum Tolerated Dose and / or Recommended Dose and Dosing Regimen of MOR03087
NCT01421186 (8) [back to overview]Duration of Response
NCT01421186 (8) [back to overview]Number of Participants Who Develop Anti-MOR03087 Antibodies
NCT01421186 (8) [back to overview]Overall Response Rate
NCT01442714 (3) [back to overview]Median Duration of Response
NCT01442714 (3) [back to overview]Overall Response Rate (ORR)
NCT01442714 (3) [back to overview]Overall Survival
NCT01460940 (3) [back to overview]Progression-free Survival in Patients With Previously Treated Hodgkin's Lymphoma Receiving Combined Lenalidomide and Panobinostat
NCT01460940 (3) [back to overview]Determine the Overall Response Rate (ORR), Including Complete Responses (CR) and Partial Responses (PR)
NCT01460940 (3) [back to overview]Assess the Safety and Tolerability of Combined Lenalidomide and Panobinostat in Patients With Previously Treated Hodgkin's Lymphoma.
NCT01463670 (2) [back to overview]Number of Participants Evaluated for Toxicity
NCT01463670 (2) [back to overview]Overall Response Rate (ORR)
NCT01472562 (1) [back to overview]Overall Response Rate
NCT01496976 (4) [back to overview]Number of Participants With Complete Response (CR)
NCT01496976 (4) [back to overview]Number of Participants With Overall Survival (OS)
NCT01496976 (4) [back to overview]Number of Participants With Partial Response (PR)
NCT01496976 (4) [back to overview]Rate of Progression/Relapse Free Survival (PFS)
NCT01497496 (1) [back to overview]Objective Response Rate (ORR)
NCT01522976 (5) [back to overview]Overall Survival
NCT01522976 (5) [back to overview]Relapse-free Survival
NCT01522976 (5) [back to overview]Response Rate (Phase II)
NCT01522976 (5) [back to overview]Pre-study Cytogenetic Abnormalities
NCT01522976 (5) [back to overview]Toxicity Rate
NCT01531998 (2) [back to overview]Number of Participants With Response
NCT01531998 (2) [back to overview]Maximum Tolerated Dose (MTD) of Siltuximab
NCT01553149 (6) [back to overview]Pharmacokinetic Parameters of Lenalidomide
NCT01553149 (6) [back to overview]Overall Survival [OS]
NCT01553149 (6) [back to overview]Event-free Survival [EFS]
NCT01553149 (6) [back to overview]Number of Patients Who Demonstrate Complete or Partial Response
NCT01553149 (6) [back to overview]Number of Patients Who Demonstrate Early Progression
NCT01553149 (6) [back to overview]Number of Patients With Toxic Events After 2 Dose Reductions
NCT01559935 (4) [back to overview]Stem Cells Collection
NCT01559935 (4) [back to overview]Response to Car-BiRD Treatment.
NCT01559935 (4) [back to overview]Event Free Survival
NCT01559935 (4) [back to overview]Progression Free Survival
NCT01564537 (18) [back to overview]Duration of Response (DOR)
NCT01564537 (18) [back to overview]Overall Response Rate (ORR) as Assessed by the IRC
NCT01564537 (18) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Multiple Myeloma Module (QLQ-MY-20)
NCT01564537 (18) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) Questionnaire (EORTC-QLQ-C30)
NCT01564537 (18) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) Questionnaire (EORTC-QLQ-C30)
NCT01564537 (18) [back to overview]Time to Progression (TTP) as Assessed by the IRC
NCT01564537 (18) [back to overview]PFS in High-Risk Participants
NCT01564537 (18) [back to overview]Percentage of Participants With Complete Response (CR) and Very Good Partial Response (VGPR) as Assessed by the IRC
NCT01564537 (18) [back to overview]Overall Response Rate in Participants Defined by Polymorphism
NCT01564537 (18) [back to overview]Overall Survival (OS)
NCT01564537 (18) [back to overview]Plasma Concentration Over Time for Ixazomib
NCT01564537 (18) [back to overview]Plasma Concentration Over Time for Ixazomib
NCT01564537 (18) [back to overview]Number of Participants With Change From Baseline in Pain Response
NCT01564537 (18) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT01564537 (18) [back to overview]Overall Survival in High-Risk Participants Carrying Deletion 17 [Del(17)]
NCT01564537 (18) [back to overview]Progression Free Survival (PFS) as Assessed by the Independent Review Committee (IRC)
NCT01564537 (18) [back to overview]Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Multiple Myeloma Module (QLQ-MY-20)
NCT01564537 (18) [back to overview]OS in High-Risk Participants
NCT01572480 (6) [back to overview]Percentage of Participants With Minimal Residual Disease (MRD)Negative Complete Response (CR) Per International Myeloma Working Group (IMWG) Criteria
NCT01572480 (6) [back to overview]Overall Response Rate
NCT01572480 (6) [back to overview]Percentage of Participants That Have Minimal Residual Disease (MRD)-Negative Complete Response (CR) for a Minimum of 1 Year
NCT01572480 (6) [back to overview]Number of Participants With Serious and/or Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0)
NCT01572480 (6) [back to overview]Clinical Progression Free Survival
NCT01572480 (6) [back to overview]Biochemical Progression Free Survival
NCT01575860 (2) [back to overview]Progression Free Survival
NCT01575860 (2) [back to overview]Number of Subjects With Dose-limiting Toxicities
NCT01600053 (1) [back to overview]Recording of the Occurrence of Adverse Events
NCT01615029 (8) [back to overview]Phase 2: Time to Progression (TTP)
NCT01615029 (8) [back to overview]Phase 2: Duration of Response
NCT01615029 (8) [back to overview]Phase 1: Percentage of Participants With Overall Response Rate (ORR)
NCT01615029 (8) [back to overview]Phase 2: Overall Survival (OS)
NCT01615029 (8) [back to overview]Phase 2: Percentage of Participants With Overall Response Rate (ORR)
NCT01615029 (8) [back to overview]Phase 2: Progression-Free Survival (PFS)
NCT01615029 (8) [back to overview]Phase 2: Time to Response
NCT01615029 (8) [back to overview]Phase 1: Time to Response
NCT01621672 (1) [back to overview]Progression Free Survival (PFS)
NCT01645930 (12) [back to overview]Duration of Response (DOR)
NCT01645930 (12) [back to overview]Cmax: Maximum Observed Plasma Concentration for Ixazomib
NCT01645930 (12) [back to overview]Cmax: Maximum Observed Plasma Concentration for Ixazomib
NCT01645930 (12) [back to overview]AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to Time of the Last Quantifiable Concentration for Ixazomib
NCT01645930 (12) [back to overview]AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to Time of the Last Quantifiable Concentration for Ixazomib
NCT01645930 (12) [back to overview]Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Ixazomib
NCT01645930 (12) [back to overview]Number of Participants With Clinically Significant Vital Signs Reported as Adverse Events
NCT01645930 (12) [back to overview]Number of Participants With Clinically Significant Laboratory Abnormalities Reported as Adverse Events of ≥Grade 3 Intensity
NCT01645930 (12) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT01645930 (12) [back to overview]Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Ixazomib
NCT01645930 (12) [back to overview]Percentage of Participants With Confirmed Best Response Category
NCT01645930 (12) [back to overview]Number of Participants With Dose Limiting Toxicities (DLTs)
NCT01649791 (3) [back to overview]Incidence of Immune Mediated Flare Reaction
NCT01649791 (3) [back to overview]Median Progression-free Survival
NCT01649791 (3) [back to overview]Overall Response Rate (CR+PR)
NCT01651039 (8) [back to overview]Clinical Benefit Rate
NCT01651039 (8) [back to overview]The Best Overall Response Rate (ORR)
NCT01651039 (8) [back to overview]Response Rates
NCT01651039 (8) [back to overview]Response Rates for Len Refractory Patients
NCT01651039 (8) [back to overview]Disease Control Rate for Lens Refractory Rate
NCT01651039 (8) [back to overview]Clinical Benefit Rate for Len Refractory Patients
NCT01651039 (8) [back to overview]Overall Response Rate for Len Refractory Patients
NCT01651039 (8) [back to overview]Disease Control Rate
NCT01659658 (21) [back to overview]Plasma Concentration of Ixazomib
NCT01659658 (21) [back to overview]Time To Subsequent Anticancer Treatment
NCT01659658 (21) [back to overview]Percentage of Participants With Overall Hematologic Response
NCT01659658 (21) [back to overview]Percentage of Participants With Complete Hematologic Response
NCT01659658 (21) [back to overview]Time To Treatment Failure (TTF)
NCT01659658 (21) [back to overview]Time to Vital Organ (Heart or Kidney) Deterioration and Mortality Rate
NCT01659658 (21) [back to overview]Change From Baseline in Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACT/GOG-Ntx) Score at Week 28 of the PFS Follow-up
NCT01659658 (21) [back to overview]2-Year Vital Organ (Heart or Kidney) Deterioration and Mortality Rate
NCT01659658 (21) [back to overview]Change From Baseline in 36-item Short Form General Health Survey (SF-36) Mental Component Summary Score at Week 28 of the PFS Follow-up
NCT01659658 (21) [back to overview]Change From Baseline in Amyloidosis Symptom Scale Total Score at Week 28 of the PFS Follow-up
NCT01659658 (21) [back to overview]Duration of Hematologic Response
NCT01659658 (21) [back to overview]EuroQol 5-Dimension 3-Level (EQ-5D-3L) Visual Analogue Scale Score
NCT01659658 (21) [back to overview]Number of Participants in Each Category of the EuroQol 5-Dimensional (EQ-5D) Questionnaire Score
NCT01659658 (21) [back to overview]Hematologic Disease Progression Free Survival
NCT01659658 (21) [back to overview]Number of Hospitalizations
NCT01659658 (21) [back to overview]Number of Participants With Serious Adverse Events (SAEs)
NCT01659658 (21) [back to overview]Overall Survival
NCT01659658 (21) [back to overview]Percentage of Participants With Best Vital Organ (Cardiac and/or Kidney) Response
NCT01659658 (21) [back to overview]Change From Baseline in SF-36 Physical Component Summary Score at Week 28 of the PFS Follow-up
NCT01659658 (21) [back to overview]Vital Organ Progression Free Survival
NCT01659658 (21) [back to overview]Progression Free Survival (PFS)
NCT01668719 (5) [back to overview]Number of Participants With Gr 3 Through 5 Adverse Events That Are Related to Study Drugs
NCT01668719 (5) [back to overview]Response (Partial Response [PR] or Better) Rate
NCT01668719 (5) [back to overview]Progression-free Survival
NCT01668719 (5) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Elotuzumab in Combination With Bortezomib, Lenalidomide and Dexamethasone
NCT01668719 (5) [back to overview]Overall Survival
NCT01675141 (3) [back to overview]Number of Participants With Serious and Non-serious Adverse Events
NCT01675141 (3) [back to overview]Progression Free Survival (PFS)
NCT01675141 (3) [back to overview]Duration of Response
NCT01698801 (6) [back to overview]Time to Response
NCT01698801 (6) [back to overview]Overall Survival (OS)
NCT01698801 (6) [back to overview]Duration of Response
NCT01698801 (6) [back to overview]Overall Response Rate
NCT01698801 (6) [back to overview]Number of Participants With Adverse Events
NCT01698801 (6) [back to overview]Progression Free Survival (PFS)
NCT01704781 (7) [back to overview]Part A and B: Safety and Tolerability
NCT01704781 (7) [back to overview]Part A: To Establish Highest Tolerated Dose of Lenalidomide, CD4 Counts Over Time
NCT01704781 (7) [back to overview]Part A: To Establish Highest Tolerated Dose of Lenalidomide, Dose-Limiting Toxicity
NCT01704781 (7) [back to overview]Part B: Incidents of Delayed-type Hypersensitivity
NCT01704781 (7) [back to overview]Part B: Evaluate the Effect on HIV Viral Load
NCT01704781 (7) [back to overview]Part B: Change in CD8 Count
NCT01704781 (7) [back to overview]Part B: Change in CD4 Count
NCT01706666 (3) [back to overview]Survival Time
NCT01706666 (3) [back to overview]Proportion of Patients Experiencing a Stringent Complete Response (sCR) After 12 Cycles, 24 Months
NCT01706666 (3) [back to overview]Progression-free Survival
NCT01723839 (2) [back to overview]Overall Response Rate
NCT01723839 (2) [back to overview]Complete Response
NCT01724177 (8) [back to overview]Time to Response
NCT01724177 (8) [back to overview]Percentage of Participants Who Achieved a Complete Response, Unconfirmed Complete Response (CRu), Partial Response or Stable Disease (SD) as Assessed by the ESEC
NCT01724177 (8) [back to overview]Kaplan-Meier Estimate of Duration of Response (DOR) for Responders as Assessed by the ESEC
NCT01724177 (8) [back to overview]Kaplan-Meier Estimate for Overall Survival
NCT01724177 (8) [back to overview]Number of Participants With Treatment Emergent Adverse Events
NCT01724177 (8) [back to overview]Kaplan Meier Estimate of Progression Free Survival (PFS) as Assessed by the ESEC
NCT01724177 (8) [back to overview]Percentage of Participants Who Achieved a Complete Response, Unconfirmed Complete Response, or Partial Response as Assessed by the Efficacy-Safety Evaluation Committee (ESEC)
NCT01724177 (8) [back to overview]Kaplan-Meier Estimate of Time to Progression (TTP)
NCT01729338 (11) [back to overview]QLQ-C30 Question 30
NCT01729338 (11) [back to overview]Overall Response Rate (ORR) During Induction Therapy
NCT01729338 (11) [back to overview]Median Time to Response
NCT01729338 (11) [back to overview]QLQ-C30 Question 29
NCT01729338 (11) [back to overview]Median Progression-free Survival
NCT01729338 (11) [back to overview]Median Overall Survival
NCT01729338 (11) [back to overview]Median Duration of Response
NCT01729338 (11) [back to overview]Functionality as Assessed Using the Cancer and Leukemia Group B (CALGB) Geriatric Assessment Tool
NCT01729338 (11) [back to overview]Severe Adverse Event Rate
NCT01729338 (11) [back to overview]Maximum Depth of Response During Induction Therapy
NCT01729338 (11) [back to overview]Maximum Depth of Response During Maintenance Therapy
NCT01731886 (5) [back to overview]Overall Survival Rate (OS)
NCT01731886 (5) [back to overview]Overall Survival Rate (OS)
NCT01731886 (5) [back to overview]Progression Free Survival (PFS)
NCT01731886 (5) [back to overview]Progression Free Survival (PFS)
NCT01731886 (5) [back to overview]Complete Response Rate
NCT01743859 (7) [back to overview]Percentage of Participants With Complete Remission or Complete Remission With Incomplete Recovery Blood Counts
NCT01743859 (7) [back to overview]Progression-free Survival
NCT01743859 (7) [back to overview]Response or Remission Duration
NCT01743859 (7) [back to overview]Toxicity and SAEs Related to Treatment
NCT01743859 (7) [back to overview]Determine Biomarkers That Predict Response/Toxicity
NCT01743859 (7) [back to overview]Overall Response Rate
NCT01743859 (7) [back to overview]Overall Survival
NCT01754857 (2) [back to overview]Time to Progression
NCT01754857 (2) [back to overview]Count of Events Related to Toxicity
NCT01772420 (7) [back to overview]Number of Patients With Hematologic Improvement in Platelet Counts (HI-P)
NCT01772420 (7) [back to overview]Number of Patients With Clinically Significant Bleeding Events
NCT01772420 (7) [back to overview]Number of Patients With Hematologic Improvement in Neutrophil Counts (HI-N)
NCT01772420 (7) [back to overview]Number of Patients Demonstrating Overall Hematologic Improvement (HI)
NCT01772420 (7) [back to overview]Duration of Hematologic Improvement (HI)
NCT01772420 (7) [back to overview]Time to Attain Hematologic Improvement (HI)
NCT01772420 (7) [back to overview]Number of Patients With Hematologic Improvement in Erythrocyte Counts (HI-E)
NCT01779167 (1) [back to overview]Number of Patients Who Demonstrate a Response (Complete, Partial, Minor) to Treatment
NCT01782963 (6) [back to overview]Mean Plasma Bortezomib Concentration Following Intravenous and and Subcutaneous Injection
NCT01782963 (6) [back to overview]Median Overall Survival
NCT01782963 (6) [back to overview]Median Progression Free Survival
NCT01782963 (6) [back to overview]Median Time to Response
NCT01782963 (6) [back to overview]Number of Participants With Grade 3 or Higher Treatment Related Adverse Events
NCT01782963 (6) [back to overview]Objective Response Rate
NCT01794039 (4) [back to overview]Proportion of Confirmed Tumor Responses Defined to be a Partial Response or Better Noted as the Objective Status on Two Consecutive Evaluations
NCT01794039 (4) [back to overview]Time to Progression
NCT01794039 (4) [back to overview]Number of Participants With Adverse Events, Graded According to the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0
NCT01794039 (4) [back to overview]Overall Survival
NCT01816971 (6) [back to overview]Percentage of Participants With Progression-free Survival (PFS)
NCT01816971 (6) [back to overview]Percentage of Participants With Overall Survival (OS)
NCT01816971 (6) [back to overview]Time to Progression
NCT01816971 (6) [back to overview]Percentage of Patients Achieving sCR
NCT01816971 (6) [back to overview]Overall Response Rate, Defined as at Least a Partial Response to Therapy (> PR), at Least Very Good Partial Response (VGPR) and at Least Near Complete Response (nCR) Rate
NCT01816971 (6) [back to overview]Duration of Response
NCT01850524 (20) [back to overview]Cmax: Maximum Plasma Concentration for Ixazomib
NCT01850524 (20) [back to overview]Number of Participants With Abnormal Serum Chemistry and Hematology Laboratory Values Based on Treatment-emergent Adverse Events (TEAEs)
NCT01850524 (20) [back to overview]Number of Participants With Shifts From Baseline to Worst Value in Eastern Cooperative Oncology Group (ECOG) Performance Score
NCT01850524 (20) [back to overview]Pain Response Rate as Assessed by the Brief Pain Inventory- Short Form (BPI-SF) and Analgesic Use
NCT01850524 (20) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT01850524 (20) [back to overview]Time to Response
NCT01850524 (20) [back to overview]Overall Survival (OS)
NCT01850524 (20) [back to overview]Overall Response Rate (ORR)
NCT01850524 (20) [back to overview]OS in High-risk Population Carrying Del(17p), t(4;14), or t(14;16) Mutations
NCT01850524 (20) [back to overview]Duration of Response
NCT01850524 (20) [back to overview]Complete Response (CR) Rate
NCT01850524 (20) [back to overview]Change From Baseline in Health-Related Quality of Life (HRQOL) Measured by European Organisation for Research and Treatment of Cancer-Quality of Life Questionnaire (EORTC-QLQ)-C30 Scale Total Score
NCT01850524 (20) [back to overview]Percentage of Participants With MRD-Negative Status as Assessed by Flow Cytometry
NCT01850524 (20) [back to overview]Percentage of Participants With New or Worsening of Existing Skeletal-related Events (SREs)
NCT01850524 (20) [back to overview]PFS in High-risk Population Carrying Del(17p), t(4;14), or t(14;16) Mutations
NCT01850524 (20) [back to overview]Time to Progression (TTP)
NCT01850524 (20) [back to overview]Change From Baseline in HRQOL Measured by EORTC-QLQ-MY20 Scale
NCT01850524 (20) [back to overview]Time to Pain Progression
NCT01850524 (20) [back to overview]Progression Free Survival (PFS)-2
NCT01850524 (20) [back to overview]Progression Free Survival (PFS)
NCT01856192 (4) [back to overview]Proportion of Patients With Response
NCT01856192 (4) [back to overview]3-year Progression-free Survival Rate
NCT01856192 (4) [back to overview]Overall Survival Rate at 3 Years
NCT01856192 (4) [back to overview]Proportion of Patients With Complete Response
NCT01881789 (6) [back to overview]Duration of Response (DOR)
NCT01881789 (6) [back to overview]Number of Participants With Dose-Limiting Toxicities (DLTs)
NCT01881789 (6) [back to overview]Progression-Free Survival (PFS)
NCT01881789 (6) [back to overview]Number of Participants With Treatment-emergent Adverse Events (AEs)
NCT01881789 (6) [back to overview]Plasma Oprozomib Concentration
NCT01881789 (6) [back to overview]Overall Response Rate (ORR)
NCT01891643 (3) [back to overview]Levels of CS1 Soluble Form (sCS1) in Serum
NCT01891643 (3) [back to overview]Change From Baseline in the Levels of CS1 Soluble Form (sCS1) in Serum During Therapy and At Progression
NCT01891643 (3) [back to overview]Percent of Bone Marrow-Derived Multiple Myeloma (MM) Cells Expressing Cell Surface CS1 at Time of Progression
NCT01924169 (1) [back to overview]Number of Participants With IgG Response
NCT01927718 (1) [back to overview]Number of Participants With Response
NCT01938001 (10) [back to overview]Kaplan-Meier Estimate of Duration of Objective Response as Assessed by the IRC According to the 2007 IWGRC
NCT01938001 (10) [back to overview]Kaplan-Meier Estimate of Duration of Complete Response (DOCR) as Assessed by the IRC According to the 2007 IWGRC
NCT01938001 (10) [back to overview]Kaplan Meier Estimate of Time to Next Anti-Lymphoma Treatment (TTNLT)
NCT01938001 (10) [back to overview]Kaplan Meier Estimate of Progression Free Survival Assessed by the Independent Review Committee (IRC) According to the 2007 International Working Group Response Criteria (IWGRC)
NCT01938001 (10) [back to overview]Kaplan Meier Estimate of Event Free Survival as Assessed by the IRC According to the 2007 IWGRC
NCT01938001 (10) [back to overview]Durable Complete Response Rate (DCCR) as Assessed by the IRC According to the 2007 IWGRC
NCT01938001 (10) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs)
NCT01938001 (10) [back to overview]Percentage of Participants With an Objective Response as Assessed by the IRC According to the 2007 IWGRC
NCT01938001 (10) [back to overview]Percentage of Participants With a Best Response of Complete Response as Assessed by the IRC According to the 2007 IWGRC
NCT01938001 (10) [back to overview]Kaplan-Meier Estimate of Overall Survival (OS)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide): Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide): Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide): Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)
NCT01953692 (42) [back to overview]Overall Survival (OS)
NCT01953692 (42) [back to overview]Overall Survival (OS) in Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Overall Survival (OS) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Progression-free Survival (PFS) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)
NCT01953692 (42) [back to overview]Progression-free Survival (PFS) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Progression-free Survival (PFS) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Progression-free Survival (PFS) in Participants Pooled From the Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Progression-free Survival (PFS) in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)
NCT01953692 (42) [back to overview]Stringent Complete Remission (sCR) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)
NCT01953692 (42) [back to overview]Time to Progression (TTP) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Complete Remission Rate (CRR) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Complete Response (CR) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)
NCT01953692 (42) [back to overview]Cytogenic Complete Response in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT01953692 (42) [back to overview]Cytogenic Partial Response in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT01953692 (42) [back to overview]Duration of Response (DOR) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Duration of Response (DOR) in Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)
NCT01953692 (42) [back to overview]Duration of Response (DOR) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)
NCT01953692 (42) [back to overview]Duration of Response (DOR) in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)
NCT01953692 (42) [back to overview]Erythroid Response in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT01953692 (42) [back to overview]Marrow Complete Response (mCR) in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT01953692 (42) [back to overview]Neutrophil Response in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT01953692 (42) [back to overview]Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)
NCT01953692 (42) [back to overview]Number of Participants Who Experienced One or More Adverse Events (AEs):
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)
NCT01953692 (42) [back to overview]Objective Response Rate (ORR) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)
NCT01953692 (42) [back to overview]Platelet Response in Cohort 1: Myelodysplastic Syndrome (MDS)
NCT02036502 (8) [back to overview]Progression Free Survival (PFS) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment
NCT02036502 (8) [back to overview]Duration of Response (DOR) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment
NCT02036502 (8) [back to overview]Disease Control Rate (DCR) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment
NCT02036502 (8) [back to overview]Overall Survival (OS)
NCT02036502 (8) [back to overview]Number of Participants Experiencing Dose-Limiting Toxicities (DLTs) According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (NCI-CTCAE v.4.0)
NCT02036502 (8) [back to overview]Objective Response Rate (ORR) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment
NCT02036502 (8) [back to overview]Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)
NCT02036502 (8) [back to overview]Number of Participants Who Experienced One or More Adverse Events (AEs)
NCT02041325 (4) [back to overview]Phenotypic Changes
NCT02041325 (4) [back to overview]Safety
NCT02041325 (4) [back to overview]Quantity of Subjects With a T-cell Response
NCT02041325 (4) [back to overview]Positive for Hepatitis B Surface Antigen
NCT02076009 (9) [back to overview]Time to Disease Progression (TTP)
NCT02076009 (9) [back to overview]Time to Response
NCT02076009 (9) [back to overview]Percentage of Participants With Negative Minimal Residual Disease (MRD)
NCT02076009 (9) [back to overview]Time to Subsequent Anticancer Treatment
NCT02076009 (9) [back to overview]Duration of Response (DOR)
NCT02076009 (9) [back to overview]Overall Response Rate
NCT02076009 (9) [back to overview]Overall Survival (OS)
NCT02076009 (9) [back to overview]Percentage of Participants Who Achieved Very Good Partial Response (VGPR) or Better
NCT02076009 (9) [back to overview]Progression-free Survival (PFS)
NCT02077166 (10) [back to overview]Phase 2: Overall Survival (OS)
NCT02077166 (10) [back to overview]Phase 1b: Number of Participants With Treatment-Emergent Adverse Events (TEAEs), Serious TEAEs, and Discontinuations Due to TEAEs
NCT02077166 (10) [back to overview]Phase 2: Number of Participants With TEAEs, Serious TEAEs, and Discontinuations Due to TEAEs
NCT02077166 (10) [back to overview]Phase 1b: Recommended Phase 2 Dose of Lenalidomide in Combination With Fixed Doses of Ibrutinib and Rituximab in Participants With Relapsed or Refractory Diffuse Large B Cell Lymphoma (DLBCL)
NCT02077166 (10) [back to overview]Phase 2: CR Rate
NCT02077166 (10) [back to overview]Phase 2: Duration of Response (DOR)
NCT02077166 (10) [back to overview]Phase 2: Overall Response Rate (ORR)
NCT02077166 (10) [back to overview]Phase 2: Progression Free Survival (PFS)
NCT02077166 (10) [back to overview]Phase 1b: Complete Response (CR) Rate
NCT02077166 (10) [back to overview]Phase 1b: ORR
NCT02086552 (4) [back to overview]Overall Survival
NCT02086552 (4) [back to overview]Complete Response, Assessed Using the International Myeloma Working Group (IMWG) Uniform Response Criteria
NCT02086552 (4) [back to overview]Progression-free Survival
NCT02086552 (4) [back to overview]Progression-free Survival (1 Year Survival Rate)
NCT02126553 (4) [back to overview]Complete Response (CR) Duration
NCT02126553 (4) [back to overview]Event-free Survival (EFS)
NCT02126553 (4) [back to overview]Overall Survival (OS)
NCT02126553 (4) [back to overview]Relapse-free Survival (RFS)
NCT02128230 (1) [back to overview]The Remission Rate for Participants With High-risk Myeloma
NCT02142049 (6) [back to overview]Number of Participants With Complete Responses (CR) and Partial Responses (PR) as a Measure of Efficacy-ORR
NCT02142049 (6) [back to overview]Number of Participants With Complete Responses (CR) and Partial Responses (PR) as a Measure of Efficacy
NCT02142049 (6) [back to overview]Progression Free Survival (PFS) and Overall Survival (OS) as a Measure of Efficacy
NCT02142049 (6) [back to overview]Number of Subjects With Adverse Events as a Measure of Safety and Tolerability
NCT02142049 (6) [back to overview]Number of Participants With Dose-Limiting Toxicities as a Measure of Safety and Tolerability
NCT02142049 (6) [back to overview]Duration of Response (DOR)
NCT02159365 (2) [back to overview]Number of Participants With Any Grade and Grade 3 or Grade 4 (G3/4) Infusion Reactions Over the Entire Study Period
NCT02159365 (2) [back to overview]Number of Participants With Grade 3 or Grade 4 (G3/4) Infusion Reactions by the End of Treatment Cycle 2
NCT02225275 (3) [back to overview]Number of Participants With a Response
NCT02225275 (3) [back to overview]Time to Next Treatment
NCT02225275 (3) [back to overview]Overall Survival
NCT02252172 (4) [back to overview]Primary: Progression-free Survival (PFS)
NCT02252172 (4) [back to overview]Change From Baseline in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ)-C30 Global Health Status Score to Day 1 of Cycle 3, 6, 9 and 12
NCT02252172 (4) [back to overview]Change From Baseline in EuroQol-5 Dimensions-5 Levels (EQ-5D-5L) Utility Score to Day 1 of Cycle 3, 6, 9 and 12
NCT02252172 (4) [back to overview]Change From Baseline in EuroQol-5 Dimensions-5 Levels (EQ-5D-5L) Visual Analogue Scale (VAS) to Day 1 of Cycle 3, 6, 9 and 12
NCT02253316 (6) [back to overview]Rate of MRD-positive to MRD-negative Conversion Between the Two Maintenance Arms
NCT02253316 (6) [back to overview]Number of Participants With Improvement in Minimal Residual Disease (MRD)
NCT02253316 (6) [back to overview]MRD-negative Rate After ASCT
NCT02253316 (6) [back to overview]Compare Response Rate Between the Two Maintenance Arms
NCT02253316 (6) [back to overview]Toxicity of IRD Consolidation
NCT02253316 (6) [back to overview]Response Rate of IRD Consolidation
NCT02265510 (10) [back to overview]Phase 1a and 1b: Number of Participants With at Least One Treatment-Emergent Adverse Event (TEAE) and Serious Adverse Event (SAE)
NCT02265510 (10) [back to overview]Phase 1a, 1b, and Phase 2: Cmax: Maximum Observed Plasma Concentration for INCB052793
NCT02265510 (10) [back to overview]Phase 1A and 1B: Percentage of Participants With Response as Determined by Investigator's Assessment
NCT02265510 (10) [back to overview]Phase 1a, 1b, and Phase 2: AUC0-τ: Area Under the Plasma Concentration-time Curve Over Dosing Interval for INCB052793
NCT02265510 (10) [back to overview]Phase 1a, 1b, and Phase 2: AUC0-τ: Area Under the Plasma Concentration-time Curve Over Dosing Interval for Itacitinib
NCT02265510 (10) [back to overview]Phase 1a, 1b, and Phase 2: Cmax: Maximum Observed Plasma Concentration of Itacitinib
NCT02265510 (10) [back to overview]Phase 1a, 1b, and Phase 2: Tmax: Time to Maximum Plasma Concentration for INCB052793
NCT02265510 (10) [back to overview]Phase 1a, 1b, and Phase 2: Tmax: Time to Maximum Plasma Concentration for Itacitinib
NCT02265510 (10) [back to overview]Phase 2: Objective Response Rate (ORR) in Hematological Malignancies
NCT02265510 (10) [back to overview]Phase 2: Number of Participants With at Least One TEAE and SAE
NCT02272803 (4) [back to overview]Objective Response Rate (ORR)
NCT02272803 (4) [back to overview]Progression Free Survival (PFS)
NCT02272803 (4) [back to overview]Objective Response Rate (ORR) of Participants Treated With Elotuzumab + Lenalidomide/Dexamethasone (E-Ld)
NCT02272803 (4) [back to overview]Progression Free Survival (PFS) Rate
NCT02279394 (4) [back to overview]Overall Survival
NCT02279394 (4) [back to overview]Time to Progression
NCT02279394 (4) [back to overview]Percent of Patients Who Are Progression Free at 2 Years
NCT02279394 (4) [back to overview]Objective Response Percent
NCT02285062 (15) [back to overview]Mean Change From Baseline in the FACT-Lym Additional Concerns Subscale
NCT02285062 (15) [back to overview]K-M Estimate of Time to Next Lymphoma Therapy (TTNLT)
NCT02285062 (15) [back to overview]Percentage of Participants Who Completed the Euroqol 5-Dimension 3-Level (EQ-5D-3L) Health Related Quality of Life (HR-QoL) Questionnaire
NCT02285062 (15) [back to overview]Percentage of Participants Who Completed the Functional Assessment of Cancer Therapy Lymphoma (FACT-Lym) Questionnaire
NCT02285062 (15) [back to overview]Mean Change From Baseline in the EQ-5D-3L Visual Analogue Scale (VAS)
NCT02285062 (15) [back to overview]K-M Estimate of Overall Survival (OS)
NCT02285062 (15) [back to overview]K-M Estimate of Duration of Complete Response
NCT02285062 (15) [back to overview]Mean Change From Baseline in the FACT-Lym Physical Well-Being Subscale
NCT02285062 (15) [back to overview]Percentage of Participants Who Achieved an Objective Response
NCT02285062 (15) [back to overview]Mean Change From Baseline in the Euroqol 5-Dimension 3-Level (EQ-5D-3L) Index Score
NCT02285062 (15) [back to overview]Percentage of Participants Who Achieved a Complete Response (CR)
NCT02285062 (15) [back to overview]Mean Change From Baseline in the FACT-Lym Trial Outcome Index (TOI)
NCT02285062 (15) [back to overview]Kaplan-Meier Estimate of Progression Free Survival (PFS)
NCT02285062 (15) [back to overview]Kaplan-Meier (K-M) Estimate of Event Free Survival (EFS)
NCT02285062 (15) [back to overview]Mean Change From Baseline in the FACT-Lym Functional Well-Being Subscale
NCT02309515 (1) [back to overview]Proportion of Participants With Successful Response
NCT02322320 (5) [back to overview]Percentage of Participants With Event-free Survival (EFS)
NCT02322320 (5) [back to overview]Percentage of Participants With Secondary Primary Malignancies (SPM)
NCT02322320 (5) [back to overview]Percentage of Participants With Progression-free Survival (PFS)
NCT02322320 (5) [back to overview]Percentage of Participants With Overall Survival (OS)
NCT02322320 (5) [back to overview]Percentage of Participants With Disease Progression
NCT02331368 (3) [back to overview]The Estimated Percentage of Patients Alive Without Relapse or Progression at 2 Years
NCT02331368 (3) [back to overview]The Estimated Percentage of Patients Alive at 2 Years
NCT02331368 (3) [back to overview]The Number of Patients That Achieve Complete Response
NCT02335983 (13) [back to overview]Change From Baseline in Hemoglobin Levels
NCT02335983 (13) [back to overview]Change From Baseline in Neutrophil Count
NCT02335983 (13) [back to overview]Change From Baseline in Platelet Count
NCT02335983 (13) [back to overview]Number of Participants With Adverse Events (AEs)
NCT02335983 (13) [back to overview]Change From Baseline in Bilirubin
NCT02335983 (13) [back to overview]Time to Maximum Plasma Concentration of Carfilzomib on Day 8 of Cycle 1 by Dose Group
NCT02335983 (13) [back to overview]Progression-free Survival (PFS)
NCT02335983 (13) [back to overview]Overall Response Rate (ORR)
NCT02335983 (13) [back to overview]Maximum Plasma Concentration of Carfilzomib on Day 8 of Cycle 1 by Dose Group
NCT02335983 (13) [back to overview]Change From Baseline in Creatinine
NCT02335983 (13) [back to overview]Duration of Response (DOR)
NCT02335983 (13) [back to overview]Complete Response Rate (CRR)
NCT02335983 (13) [back to overview]Area Under the Curve From Time Zero to Time of Last Quantifiable Concentration of Carfilzomib on Day 8 of Cycle 1 by Dose Group
NCT02375555 (6) [back to overview]Median Time to Response
NCT02375555 (6) [back to overview]Grade 3 and 4 Treatment-Emergent Adverse Event (TEAE) Rate
NCT02375555 (6) [back to overview]4 Cycle Response Rate
NCT02375555 (6) [back to overview]Successful Stem Cell Mobilization (SC Mob) Rate
NCT02375555 (6) [back to overview]Objective Response Rate (ORR) at End of 8 Cycles of Induction Therapy.
NCT02375555 (6) [back to overview]4 Cycle Ever Dose Modification (DM) Rate
NCT02399085 (14) [back to overview]Time to Progression (TTP) by IRC Evaluation
NCT02399085 (14) [back to overview]Number of Participants That Experienced Treatment-emergent Adverse Events (TEAEs)
NCT02399085 (14) [back to overview]Disease Control Rate (DCR) by IRC Evaluation
NCT02399085 (14) [back to overview]DCR by INV Evaluation
NCT02399085 (14) [back to overview]Duration of Response (DoR) by IRC Evaluation
NCT02399085 (14) [back to overview]Time to Next Treatment (TTNT)
NCT02399085 (14) [back to overview]Serum Drug Levels of MOR00208
NCT02399085 (14) [back to overview]Progression-free Survival (PFS) by IRC Evaluation
NCT02399085 (14) [back to overview]PFS by INV Evaluation
NCT02399085 (14) [back to overview]Overall Survival (OS)
NCT02399085 (14) [back to overview]Number of Participants With Best Objective Response Rate (ORR)
NCT02399085 (14) [back to overview]Event-free Survival (EFS) by IRC Evaluation
NCT02399085 (14) [back to overview]DoR by Investigator (INV) Evaluation
NCT02399085 (14) [back to overview]TTP by INV Evaluation
NCT02481934 (2) [back to overview]Number of Participants With Peripheral Blood Monoclonal Protein Reduction or Stabilization
NCT02481934 (2) [back to overview]Number of Participants With Adverse Events During NKAE Treatment
NCT02492750 (2) [back to overview]Number of Participants Experiencing a Dose-limiting Toxicity (DLT)
NCT02492750 (2) [back to overview]Number of Participants Who Experienced at Least One Grade 3+ Adverse Events Deemed at Least Possibly Related to Treatment, Graded According to NCI CTCAE Version 4.0
NCT02516696 (10) [back to overview]Survival Duration Without Disease Progression
NCT02516696 (10) [back to overview]Overall Survival
NCT02516696 (10) [back to overview]Overall Response Rate
NCT02516696 (10) [back to overview]Number of Patients With Objective Response Rate (CR+PR)
NCT02516696 (10) [back to overview]Number of Patients With Complete Response Rate (CR)
NCT02516696 (10) [back to overview]Number of Days for Event-Free Survival
NCT02516696 (10) [back to overview]Number of Days for Duration of Response
NCT02516696 (10) [back to overview]Number of Days After Initiating Treatment With BiRd Regimen to Disease Progression, as Compared to Subjects on Rd Treatment Regimen.
NCT02516696 (10) [back to overview]Number of Adverse Events Experienced
NCT02516696 (10) [back to overview]Functional Assessment of Chronic Illness Therapy - Fatigue Subscale (FS; Version 4) Score of Patients Receiving BiRd vs Rd Treatment
NCT02538965 (13) [back to overview]Apparent Total Clearance (CL/F) of Lenalidomide
NCT02538965 (13) [back to overview]Apparent Volume of Distribution (Vz/F) of Lenalidomide
NCT02538965 (13) [back to overview]Area Under the Plasma Concentration Time Curve From 0 Extrapolated to Infinity (AUC-inf, AUC0∞) Of Lenalidomide
NCT02538965 (13) [back to overview]Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration of Lenalidomide (AUC-t)
NCT02538965 (13) [back to overview]Number of Participants Who Achieved a Best Response of Morphologic Complete Remission, Morphologic Complete Remission Incomplete or Partial Remission
NCT02538965 (13) [back to overview]Maximum Observed Concentration (Cmax) of Lenalidomide
NCT02538965 (13) [back to overview]Number of Participants Who Achieved a Morphologic Complete Response Within the First Four Cycles of Lenalidomide Treatment According to the Modified International Working Group (IWG) Criteria
NCT02538965 (13) [back to overview]Percentage of Participants With of Graft Versus Host Disease (GVHD)
NCT02538965 (13) [back to overview]Terminal Half-Life (t1/2) of Lenalidomide
NCT02538965 (13) [back to overview]Time to Reach Maximum Concentration (Tmax) of Lenalidomide
NCT02538965 (13) [back to overview]Number of Participants Who Experienced Treatment Emergent Adverse Events (TEAE)
NCT02538965 (13) [back to overview]Number of Participants Who Received a Haematopoietic Stem Cell Transplant (HSCT)
NCT02538965 (13) [back to overview]Number of Participants With a Morphologic CR, CRi, PR or Treatment Failure at Cycles 1, 2 and 3
NCT02561273 (7) [back to overview]Number of Participants With Adverse Events Graded According to CTC (Phase II)
NCT02561273 (7) [back to overview]Number of Participants With Adverse Events Graded According to Common Toxicity Criteria (CTC) (Phase I)
NCT02561273 (7) [back to overview]Progression-free Survival
NCT02561273 (7) [back to overview]Overall Survival
NCT02561273 (7) [back to overview]Overall Response Rate
NCT02561273 (7) [back to overview]Maximum Tolerated Dose (MTD) of Lenalidomide and CHOEP
NCT02561273 (7) [back to overview]Complete Response Rate (Phase II)
NCT02579863 (7) [back to overview]Overall Survival (OS)
NCT02579863 (7) [back to overview]Number of Participants Who Experienced One or More Adverse Events (AEs)
NCT02579863 (7) [back to overview]Number of Participants Discontinuing Study Treatment Due to an AE
NCT02579863 (7) [back to overview]Duration of Response (DOR) Evaluated According to IMWG Response Criteria by CAC Blinded Central Review
NCT02579863 (7) [back to overview]Disease Control Rate (DCR) Evaluated According to the IMWG Response Criteria by CAC Blinded Central Review
NCT02579863 (7) [back to overview]Progression Free Survival (PFS) Evaluated According to the International Myeloma Working Group (IMWG) Response Criteria 2011 by Clinical Adjudication Committee (CAC) Blinded Central Review
NCT02579863 (7) [back to overview]Overall Response Rate (ORR) Evaluated According to the IMWG Response Criteria by CAC Blinded Central Review
NCT02600897 (34) [back to overview]Percentage of Participants With Adverse Events (AEs)
NCT02600897 (34) [back to overview]Percentage of Participants With Best Response of CR or PR, Determined by the Investigator on the Basis of CT Scans Alone
NCT02600897 (34) [back to overview]Percentage of Participants With Complete Response (CR) at End of Induction (EOI), Determined by an Independent Review Committee (IRC) on the Basis of Positron Emission Tomography (PET) and Computed Tomography (CT) Scans
NCT02600897 (34) [back to overview]Percentage of Participants With CR at EOI, Determined by Investigator on the Basis of CT Scans Alone
NCT02600897 (34) [back to overview]Percentage of Participants With CR at EOI, Determined by the Investigator on the Basis of PET-CT Scans
NCT02600897 (34) [back to overview]Percentage of Participants With CR at EOI, Determined by the IRC on the Basis of CT Scans Alone
NCT02600897 (34) [back to overview]Percentage of Participants With Dose-Limiting Toxicities (DLTs)
NCT02600897 (34) [back to overview]Percentage of Participants With Objective Response (OR) at EOI, Determined by the IRC on the Basis of PET-CT Scans
NCT02600897 (34) [back to overview]Percentage of Participants With Objective Response at EOI, Determined by Investigator on the Basis of PET-CT Scans
NCT02600897 (34) [back to overview]Percentage of Participants With Objective Response at EOI, Determined by the IRC on the Basis of CT Scans Alone
NCT02600897 (34) [back to overview]Percentage of Participants With Objective Response, Determined by the Investigator on the Basis of CT Scans Alone
NCT02600897 (34) [back to overview]Number of Participants With Anti-therapeutic Antibodies (ATAs) to Polatuzumab Vedotin
NCT02600897 (34) [back to overview]Number of Participants With Human Anti-chimeric Antibodies (HACAs) to Rituximab
NCT02600897 (34) [back to overview]Number of Participants With Human Anti-human Antibodies (HAHAs) to Obinutuzumab
NCT02600897 (34) [back to overview]Observed Plasma Lenalidomide Concentration
NCT02600897 (34) [back to overview]Observed Plasma Lenalidomide Concentration
NCT02600897 (34) [back to overview]Observed Serum Obinutuzumab Concentration
NCT02600897 (34) [back to overview]Observed Serum Obinutuzumab Concentration
NCT02600897 (34) [back to overview]Observed Serum Obinutuzumab Concentration
NCT02600897 (34) [back to overview]Observed Serum Obinutuzumab Concentration
NCT02600897 (34) [back to overview]Observed Serum Rituximab Concentration
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE
NCT02600897 (34) [back to overview]Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE
NCT02600897 (34) [back to overview]Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody
NCT02600897 (34) [back to overview]Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody
NCT02600897 (34) [back to overview]Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody
NCT02600897 (34) [back to overview]Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody
NCT02600897 (34) [back to overview]Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody
NCT02619682 (3) [back to overview]Disease Free Survival
NCT02619682 (3) [back to overview]Number of Participants With Adverse Events, Graded According to CTCAE Version 4.0
NCT02619682 (3) [back to overview]Overall Survival
NCT02631577 (15) [back to overview]Number of Participants With Dose-limiting Toxicities (DLTs) During Cycle 2 of Study Treatment
NCT02631577 (15) [back to overview]Serum Concentration of Obinutuzumab (mcg/mL)
NCT02631577 (15) [back to overview]Percentage of Participants Achieving CR at EOI, as Determined by the Investigator Using Modified Lugano 2014 Criteria
NCT02631577 (15) [back to overview]Number of Participants Positive for Human Anti-human Antibodies (HAHA) to Obinutuzumab
NCT02631577 (15) [back to overview]Percentage of Participants Achieving CR at EOI, as Determined by the IRC and Investigator Using Lugano 2014 Criteria
NCT02631577 (15) [back to overview]Percentage of Participants With Adverse Events and Serious Adverse Events
NCT02631577 (15) [back to overview]Percentage of Participants With Best Response (CR or PR) During the Study as Determined by the Investigator on the Basis of CT Scans Alone
NCT02631577 (15) [back to overview]Percentage of Participants With Objective Response (CR or PR) at EOI as Determined by the IRC and Investigator on the Basis of CT Scans Alone
NCT02631577 (15) [back to overview]Percentage of Participants With Objective Response (CR or PR) at EOI as Determined by the IRC and Investigator on the Basis of PET-CT Scans
NCT02631577 (15) [back to overview]Number of Participants Positive for Anti-therapeutic Antibodies (ATAs) to Atezolizumab
NCT02631577 (15) [back to overview]Serum Concentration of Atezolizumab (mcg/mL)
NCT02631577 (15) [back to overview]Serum Concentration of Lenalidomide (ng/mL)
NCT02631577 (15) [back to overview]Serum Concentration of Obinutuzumab (mcg/mL)
NCT02631577 (15) [back to overview]Serum Concentration of Atezolizumab (mcg/mL)
NCT02631577 (15) [back to overview]Percentage of Participants Achieving Complete Response (CR) at End of Induction (EOI), as Determined by the Independent Review Committee (IRC) Using Modified Lugano 2014 Criteria
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Time to Maximum Observed Concentration (Tmax)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 15: Time to Maximum Observed Concentration (Tmax)
NCT02685826 (26) [back to overview]Participants With Treatment Emergent Adverse Events (TEAE)
NCT02685826 (26) [back to overview]Time to Response (for Cohorts A and B)
NCT02685826 (26) [back to overview]Response Improvement Rate (RIR) for Cohort C: Percentage of Participants Achieving a Response Improved From Cycle 1 Day 1 as Assessed by the Investigators Using the International Myeloma Working Group (IMWG) Uniform Response Criteria
NCT02685826 (26) [back to overview]Participants With Dose-Limiting Toxicities (DLTs) During the Dose-Determining Timeframe (Day 1 - Day 28)
NCT02685826 (26) [back to overview]Participants Who Had Either Disease Progression or Death
NCT02685826 (26) [back to overview]Participants Who Died Up To Data Cut-off Date (15 December 2017)
NCT02685826 (26) [back to overview]Participants Who Developed Anti-drug Antibody Against Durvalumab
NCT02685826 (26) [back to overview]Overall Response Rate (ORR) for Cohorts A and B: Percentage of Participants Who Achieved a Partial Response or Better According to the International Myeloma Working Group (IMWG) Uniform Response Criteria
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma PK Parameters in Cycle 1 Day 15: Area Under the Concentration-time Curve From Time Zero to the Last Measured Time Point (AUC0-last)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 15: Maximum Observed Concentration (Cmax)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Time to Maximum Observed Concentration (Tmax)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Terminal Elimination Half-life (t1/2)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Maximum Observed Concentration (Cmax)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Area Under the Concentration-time Curve From Time Zero to the Last Measured Time Point (AUC0-last)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Area Under the Concentration-time Curve From Time Zero to Infinity (AUC0-inf)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Apparent Volume of Distribution (Vz/F)
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum Pharmacokinetic (PK) Parameters in Cycle 1: Area Under the Concentration-time Curve From Time Zero to the Last Measured Time Point (AUC0-last)
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Area Under the Concentration-time Curve From Time Zero to Infinity (AUC0-inf)
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Clearance (CL)
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Maximum Observed Concentration (Cmax)
NCT02685826 (26) [back to overview]Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Apparent Clearance (CL/F)
NCT02685826 (26) [back to overview]Kaplan-Meier Estimates for Duration of Response (for Cohort A and B)
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Volume of Distribution (Vz)
NCT02685826 (26) [back to overview]Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Terminal Elimination Half-life (t1/2)
NCT02728102 (15) [back to overview]Percentage of Participants With Progression-Free Survival in Pairwise Analysis
NCT02728102 (15) [back to overview]Number of Grade ≥ 3 Toxicities
NCT02728102 (15) [back to overview]Percentage of Participants With 1-year Response Rate of CR/sCR
NCT02728102 (15) [back to overview]Percentage of Participants With Grade 2 and 3 Infections
NCT02728102 (15) [back to overview]Percentage of Participants With Treatment-related Mortality (TRM)
NCT02728102 (15) [back to overview]Percentage of Participants With Myeloma Progression of Vaccine and Non-vaccine Arms
NCT02728102 (15) [back to overview]Participants With Grade ≥ 3 Toxicities
NCT02728102 (15) [back to overview]Participants Response to Treatment
NCT02728102 (15) [back to overview]Number of Participants With Minimal Residual Disease (MRD)
NCT02728102 (15) [back to overview]Number of Grade 2 and 3 Infections
NCT02728102 (15) [back to overview]Percentage of Participants With Progression-Free Survival
NCT02728102 (15) [back to overview]Percentage of Participants With Overall Survival in Pairwise Analysis
NCT02728102 (15) [back to overview]Percentage of Participants With Overall Survival
NCT02728102 (15) [back to overview]Percentage of Participants With Myeloma Progression in Pairwise Analysis
NCT02728102 (15) [back to overview]Percentage of Participants With Grade 2 and 3 Infections in Pairwise Analysis
NCT02733042 (21) [back to overview]Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUClast) of Ibrutinib
NCT02733042 (21) [back to overview]Area Under the Plasma Concentration-time Curve From Time Zero to Infinity (AUCinf) of Durvalumab
NCT02733042 (21) [back to overview]Time to First Response
NCT02733042 (21) [back to overview]Time to Maximum Plasma Concentration (Tmax) of Durvalumab
NCT02733042 (21) [back to overview]Volume of Distribution (Vz) of Durvalumab
NCT02733042 (21) [back to overview]Time to Maximum Observed Plasma Concentration (Tmax) of Lenalidomide
NCT02733042 (21) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Ibrutinib
NCT02733042 (21) [back to overview]Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUClast) of Durvalumab
NCT02733042 (21) [back to overview]Kaplan-Meier Estimate of Progression-free Survival (PFS)
NCT02733042 (21) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Durvalumab
NCT02733042 (21) [back to overview]Kaplan-Meier Estimate of Duration of Response
NCT02733042 (21) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Lenalidomide
NCT02733042 (21) [back to overview]Clearance (CL) of Durvalumab
NCT02733042 (21) [back to overview]Number of Participants With Treatment-emergent Adverse Events
NCT02733042 (21) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs) - Extended Collection
NCT02733042 (21) [back to overview]Time to Maximum Observed Plasma Concentration (Tmax) of Ibrutinib
NCT02733042 (21) [back to overview]Terminal Elimination Phase Half-Life (t½) of Durvalumab
NCT02733042 (21) [back to overview]Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUClast) of Lenalidomide
NCT02733042 (21) [back to overview]Overall Response Rate (ORR) During Durvalumab Treatment
NCT02733042 (21) [back to overview]Part 1: Number of Participants With Dose Limiting Toxicities (DLTs)
NCT02733042 (21) [back to overview]Overall Response Rate During the Entire Study
NCT02843074 (8) [back to overview]Maintenance Feasibility Rate (MFR)
NCT02843074 (8) [back to overview]Number of Participants With Treatment-emergent Adverse Events as a Measure of Safety
NCT02843074 (8) [back to overview]Complete Response Rate (CRR) for Complete Time on Study
NCT02843074 (8) [back to overview]Overall Survival (OS)
NCT02843074 (8) [back to overview]Progression-free Survival (PFS)
NCT02843074 (8) [back to overview]Consolidation Feasibility Rate (CFR)
NCT02843074 (8) [back to overview]Induction Feasibility Rate (IFR)
NCT02843074 (8) [back to overview]Overall Response Rate (ORR) for Complete Time on Study
NCT02874742 (16) [back to overview]Percentage of Participants With Overall Stringent Complete Response (sCR)
NCT02874742 (16) [back to overview]Percentage of Participants With Overall Response Rate (ORR)
NCT02874742 (16) [back to overview]Duration of Complete Response or Better
NCT02874742 (16) [back to overview]Duration of Response
NCT02874742 (16) [back to overview]Duration of Stringent Complete Response (sCR)
NCT02874742 (16) [back to overview]Overall Survival (OS)
NCT02874742 (16) [back to overview]Percentage of Participants With Stringent Complete Response (sCR)
NCT02874742 (16) [back to overview]Progression-free Survival (PFS)
NCT02874742 (16) [back to overview]Time to Complete Response or Better
NCT02874742 (16) [back to overview]Time to Partial Response (PR) or Better
NCT02874742 (16) [back to overview]Time to Progression (TTP)
NCT02874742 (16) [back to overview]Time to Stringent Complete Response (sCR)
NCT02874742 (16) [back to overview]Time to Very Good Partial Response (VGPR) or Better
NCT02874742 (16) [back to overview]Percentage of Participants Who Achieved Very Good Partial Response (VGPR) or Better
NCT02874742 (16) [back to overview]Percentage of Participants With Complete Response (CR) or Better
NCT02874742 (16) [back to overview]Percentage of Participants With Negative Minimal Residual Disease (MRD)
NCT02880228 (5) [back to overview]Progression-free Survival
NCT02880228 (5) [back to overview]Proportion of Complete Response Plus Very Good Partial Response (VGPR)
NCT02880228 (5) [back to overview]Proportion of Successful Stem Cell Collection
NCT02880228 (5) [back to overview]Survival Time
NCT02880228 (5) [back to overview]Partial Response (PR)
NCT02898259 (7) [back to overview]Duration of Response
NCT02898259 (7) [back to overview]Maximum Tolerated Dose (MTD) of Oral Ixazomib
NCT02898259 (7) [back to overview]Overall Survival
NCT02898259 (7) [back to overview]Progression Free Survival
NCT02898259 (7) [back to overview]Time to Progression
NCT02898259 (7) [back to overview]Time to Treatment Failure
NCT02898259 (7) [back to overview]Overall Response Rate
NCT02903381 (8) [back to overview]Number of Participants With Adverse Events
NCT02903381 (8) [back to overview]Objective Response Percent
NCT02903381 (8) [back to overview]Progression Free Survival (PFS) Probability at 2-years
NCT02903381 (8) [back to overview]Progression Free Survival Rate-Without Cyclophosphamide
NCT02903381 (8) [back to overview]Time to Progression Probability at 2-years
NCT02903381 (8) [back to overview]Overall Survival Probability at 2-years
NCT02903381 (8) [back to overview]2 Year Progression Free Percent
NCT02903381 (8) [back to overview]Duration of Response Probability at 2-years
NCT02906332 (4) [back to overview]Evaluation of Stringent Complete Response, Complete Response, and Very Good Partial Response Rate (sCR + CR + VGPR Rate).
NCT02906332 (4) [back to overview]Number of Participants Serious Adverse Events
NCT02906332 (4) [back to overview]Number of Participants Who Progressed at 12 Months
NCT02906332 (4) [back to overview]Progression Free Survival (PFS)
NCT02917941 (9) [back to overview]Number of Participants With NCI CTCAE Grade 3 or Higher TEAEs Related Laboratory Parameters
NCT02917941 (9) [back to overview]Number of Participants With One or More Treatment-emergent Adverse Events (TEAEs)
NCT02917941 (9) [back to overview]Overall Response Rate (ORR)
NCT02917941 (9) [back to overview]Overall Survival (OS)
NCT02917941 (9) [back to overview]Percentage of Participants With Very Good Partial Response (VGPR) or Better Response (Complete Response (CR) + VGPR)
NCT02917941 (9) [back to overview]Duration of Response (DOR)
NCT02917941 (9) [back to overview]Time to Progression (TTP)
NCT02917941 (9) [back to overview]Progression-free Survival (PFS)
NCT02917941 (9) [back to overview]Number of Participants With NCI CTCAE Grade 3 or Higher TEAEs Related to Vital Signs
NCT02954406 (8) [back to overview]Dose Escalation Phase: Maximum Tolerated Dose (MTD) of TAK-659
NCT02954406 (8) [back to overview]Time to Progression (TTP)
NCT02954406 (8) [back to overview]Tmax: Time to Reach the Maximum Plasma Concentration for TAK-659
NCT02954406 (8) [back to overview]Cmax: Maximum Observed Plasma Concentration for TAK-659
NCT02954406 (8) [back to overview]Dose Escalation Phase: Recommended Phase 2 Dose (RP2D) of TAK-659
NCT02954406 (8) [back to overview]AUCtau: Area Under the Plasma Concentration-time Curve During Dosing Interval
NCT02954406 (8) [back to overview]Overall Response Rate (ORR)
NCT02954406 (8) [back to overview]Duration of Response (DOR)
NCT03003520 (8) [back to overview]Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for Immunohistochemistry (IHC) Programmed Death Ligand - 1 (PDL1) Total Percentage
NCT03003520 (8) [back to overview]Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for the Interferon Gamma Score (IFNG-Score) From Ribonucleic Acid (RNA)-Sequencing Data
NCT03003520 (8) [back to overview]Percentage of Participants Who Achieved a Complete Response (CR) at the End of Induction Therapy
NCT03003520 (8) [back to overview]Percentage of Participants Who Responded During Induction and Continued Into Consolidation Therapy (Database Cutoff Date: 02-Aug-2018)
NCT03003520 (8) [back to overview]Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for Immunohistochemistry (IHC) CD8 T-Cell Density
NCT03003520 (8) [back to overview]Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for Immunohistochemistry (IHC) Programmed Death Ligand - 1 (PDL1) Percentage of Tumor Cells
NCT03003520 (8) [back to overview]Participants With Treatment Emergent Adverse Events (TEAE)
NCT03003520 (8) [back to overview]Participants With Treatment Emergent Adverse Events (TEAE)
NCT03019640 (2) [back to overview]Treatment-related Mortality Within 30 Days (TRM30)
NCT03019640 (2) [back to overview]Number of Participants Who Survived
NCT03050450 (3) [back to overview]Total Number of Adverse Events
NCT03050450 (3) [back to overview]Best Response of Children With Recurrent or Refractory Central Nervous System Tumors
NCT03050450 (3) [back to overview]Number Participants With Hematologic and Non-hematologic Toxicities
NCT03118466 (6) [back to overview]Transfusion Support: Number of Red Blood Cell and Platelet Transfusions
NCT03118466 (6) [back to overview]Treatment-related Mortality
NCT03118466 (6) [back to overview]Overall Survival
NCT03118466 (6) [back to overview]Number of Patients That Achieved Platelet Recovery
NCT03118466 (6) [back to overview]Number of Patients That Achieved ANC Recovery
NCT03118466 (6) [back to overview]Complete Response Rate
NCT03224507 (8) [back to overview]Overall Survival
NCT03224507 (8) [back to overview]Percentage of Patients Achieving Complete Remission Following Complete Therapy
NCT03224507 (8) [back to overview]Serious Adverse Events (SAEs) From the KRdD Treatment
NCT03224507 (8) [back to overview]Percentage of Patients With MRD(-) Status at the Completion of Induction Therapy
NCT03224507 (8) [back to overview]Progression-free Survival
NCT03224507 (8) [back to overview]Percentage of Patients With MRD(-) Remissions at the Completion of Consolidation Therapy
NCT03224507 (8) [back to overview]Percentage of Patients With Auto-HCT That Convert From Positive to Negative MRD
NCT03224507 (8) [back to overview]Percentage of Patients That Convert From MRD(-) to MRD(+) Following Treatment Discontinuation
NCT03411031 (3) [back to overview]Overall Response
NCT03411031 (3) [back to overview]Minimum Response (MR)
NCT03411031 (3) [back to overview]Percentage of Participants With Progression Free Survival (PFS)
NCT03412565 (14) [back to overview]Percentage of Participants With Anti-Daratumumab Antibodies
NCT03412565 (14) [back to overview]D-VRd Cohort: Overall Response Rate (ORR)
NCT03412565 (14) [back to overview]D-VMP, D-Rd, and D-Kd Cohorts: Percentage of Participants With VGPR or Better Response
NCT03412565 (14) [back to overview]D-VMP, D-Rd, and D-Kd Cohorts: Percentage of Participants With Minimal Residual Disease (MRD) Negative Rate
NCT03412565 (14) [back to overview]D-VRd Cohort: Percentage of Participants With Very Good Partial Response (VGPR) or Better Response
NCT03412565 (14) [back to overview]Percentage of Participants With Infusion-Related Reactions (IRRs)
NCT03412565 (14) [back to overview]Maximum Observed Serum Concentration (Cmax) of Daratumumab
NCT03412565 (14) [back to overview]Maximum Observed Serum Concentration (Cmax) of Daratumumab
NCT03412565 (14) [back to overview]Maximum Observed Serum Concentration (Cmax) of Daratumumab
NCT03412565 (14) [back to overview]Maximum Observed Serum Concentration (Cmax) of Daratumumab
NCT03412565 (14) [back to overview]D-VMP, D-Rd, and D-Kd Cohorts: Overall Response Rate (ORR)
NCT03412565 (14) [back to overview]D-VMP, D-Rd and D-Kd Cohorts: Duration of Response (DOR)
NCT03412565 (14) [back to overview]Percentage of Participants With CR or Better Response
NCT03412565 (14) [back to overview]Percentage of Participants With Anti-rHuPH20 Antibodies
NCT03416374 (21) [back to overview]Percentage of Participants With Bone Lesions (Bone Evaluation)
NCT03416374 (21) [back to overview]Percentage of Participants Who Achieved VGPR or Better (CR + VGPR)
NCT03416374 (21) [back to overview]Percentage of Participants Continuing Treatment With Ixazomib at 12 Months From the Start of Study Treatment
NCT03416374 (21) [back to overview]Overall Survival (OS) From the Start of Study Treatment
NCT03416374 (21) [back to overview]Overall Response Rate (ORR)
NCT03416374 (21) [back to overview]Number of Participants Reporting One or More Treatment-Emergent AEs (TEAEs)
NCT03416374 (21) [back to overview]Healthcare Resource Utilization (HCRU): Number of Events With Hospitalization Per Participants-Month
NCT03416374 (21) [back to overview]Healthcare Resource Utilization (HCRU): Duration of Hospital Stay Per Participants
NCT03416374 (21) [back to overview]Duration of Therapy (DOT)
NCT03416374 (21) [back to overview]Duration of Response (DOR)
NCT03416374 (21) [back to overview]Evaluation of Modified Quality-Adjusted Life-Years (QALYs)
NCT03416374 (21) [back to overview]Number of Participants With Minimal Residual Disease (MRD) Positive or Negative in Bone Marrow in Participants Who Achieved CR
NCT03416374 (21) [back to overview]Time to Next Treatment (TTNT)
NCT03416374 (21) [back to overview]Progression-Free Survival (PFS) Rate at 12 Months From the Start of Study Treatment
NCT03416374 (21) [back to overview]PFS From the Start of Study Treatment
NCT03416374 (21) [back to overview]Percentage of Participants Who Achieve or Maintain Any Best Response
NCT03416374 (21) [back to overview]Relative Dose Intensity (RDI)
NCT03416374 (21) [back to overview]Patient-Reported Outcome HRQoL Based on EORTC Multiple Myeloma Module (EORTC QLQ-MY20) Score
NCT03416374 (21) [back to overview]Patient-Reported Outcome HRQoL Based on EORTC Multiple Myeloma Module (EORTC QLQ-MY20) Score
NCT03416374 (21) [back to overview]Patient-Reported Outcome Health-Related Quality of Life (HRQoL) Based on Global Health Status Scale of European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-C30 (EORTC QLQ-C30)
NCT03416374 (21) [back to overview]Patient-Reported Outcome Health-Related Quality of Life (HRQoL) Based on Global Health Status Scale of European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-C30 (EORTC QLQ-C30)
NCT03433001 (15) [back to overview]Number of Participants Reporting One or More Treatment-Emergent AEs (TEAEs)
NCT03433001 (15) [back to overview]Overall Response Rate (ORR)
NCT03433001 (15) [back to overview]Overall Survival (OS)
NCT03433001 (15) [back to overview]Percentage of Participants Who Achieve VGPR or Better (CR+VGPR)
NCT03433001 (15) [back to overview]Percentage of Participants With Bone Lesions (Bone Evaluation)
NCT03433001 (15) [back to overview]Time to Next Treatment (TTNT)
NCT03433001 (15) [back to overview]Patient-Reported Outcome Health-Related Quality of Life (HRQoL) Based on European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-C30 (EORTC QLQ-C30) Global Health Status Score
NCT03433001 (15) [back to overview]Relative Dose Intensity (RDI)
NCT03433001 (15) [back to overview]Percentage of Participants Who Achieve or Maintain Any Best Response
NCT03433001 (15) [back to overview]Percentage of Participants Who Continue to Receive Treatment at 12 Months and 24 Months After Start of Treatment
NCT03433001 (15) [back to overview]Progression-Free Survival (PFS)
NCT03433001 (15) [back to overview]Patient-Reported Outcome HRQoL Based on EORTC Multiple Myeloma Module (EORTC QLQ-MY20) Score
NCT03433001 (15) [back to overview]Duration of Therapy (DOT)
NCT03433001 (15) [back to overview]Rate of Minimal Residual Disease (MRD) Negativity in Bone Marrow in Participants Who Achieved CR
NCT03433001 (15) [back to overview]PFS Rate at 12 Months and 24 Months After the Start of Treatment
NCT04272775 (8) [back to overview]Number of Participants Who Achieved Complete Response (CR), Very Good Partial Response (VGPR), and Partial Response (PR)
NCT04272775 (8) [back to overview]Number of Participants Who Experienced at Least One Treatment-emergent Adverse Event (TEAE)
NCT04272775 (8) [back to overview]Number of Participants Who Experienced Dose Limiting Toxicities (DLTs)
NCT04272775 (8) [back to overview]Number of Participants With Grade 3 or Higher Laboratory Tests Abnormalities
NCT04272775 (8) [back to overview]Cmax: Maximum Observed Plasma Concentration for Ixazomib
NCT04272775 (8) [back to overview]Number of Participants With Grade 3 or Higher TEAE Related to Vital Signs
NCT04272775 (8) [back to overview]Number of Participants With Grade 3 or Higher TEAE Related to Body Weight
NCT04272775 (8) [back to overview]Number of Participants With Grade 3 or Higher TEAE Related to 12-lead Electrocardiograms (ECGs)
NCT04430894 (9) [back to overview]Overall Response Rate After 4 Cycles Induction Therapy
NCT04430894 (9) [back to overview]Minimal Residual Disease (MRD) Rate at 10^(-5) in Patients Receiving Upfront Stem Cell Transplant
NCT04430894 (9) [back to overview]Minimal Residual Disease (MRD) Rate at 10^(-5) in Patients Deferring Stem Cell Transplant
NCT04430894 (9) [back to overview]Complete Response Rate in Patients With Upfront Stem Cell Transplant
NCT04430894 (9) [back to overview]Complete Response (CR + Stringent CR) Rate
NCT04430894 (9) [back to overview]Overall Survival at 12 Months
NCT04430894 (9) [back to overview]Progression Free Survival at 12 Months
NCT04430894 (9) [back to overview]Minimal Residual Disease (MRD) Rate After 4 Cycles Induction Therapy
NCT04430894 (9) [back to overview]Complete Response Rate in Patients Deferring Stem Cell Transplant

Time to First Worsening of Eastern Cooperative Oncology Group (ECOG) Performance Status Scale (Best Score=0, Fully Active, Able to Carry on All Pre-disease Performance Without Restriction; Worst Score=5, Dead.)

The time to first worsening on the ECOG Performance Scale was calculated as the time from randomization to the date of the first worsening compared to the last ECOG evaluation obtained prior to randomization. (NCT00056160)
Timeframe: 30 weeks (mean time to first worsening of ECOG performance status for CC-5013/Dex treatment group)

InterventionWeeks (Mean)
CC-5013/Dex29.9
Placebo/Dex15.0

[back to top]

Time to Tumor Progression (TTP)

Time to progression (TTP) was calculated as the time from randomization to the first documentation of progressive disease based on the myeloma response determination criteria developed by Bladé et. al., Br J Haematol 1998; 102:1115-1123. (NCT00056160)
Timeframe: 60 weeks (median Time To Progression of CC-5013/Dex treatment group)

InterventionWeeks (Median)
CC-5013/Dex60.1
Placebo/Dex20.1

[back to top]

Myeloma Response

The overall confirmed response that was maintained for ≥6 weeks. Complete Response (CR):Disappearance of monoclonal paraprotein. Remission Response (RR):75-99% reduction in monoclonal paraprotein/90-99% reduction in 24-hr urinary light chain excretion. Partial Response (PR):50-74% reduction in monoclonal paraprotein/50-89% reduction in 24-hr urinary light chain excretion. Stable Disease (SD):Criteria for PR or PD not met. Plateau Phase:If PR, stable monoclonal paraprotein (within 25% above or below nadir)/stable soft tissue plasmacytomas. Progressive Disease (PD):Disease worsens. (NCT00056160)
Timeframe: Up to Unblinding (07 Jun 2005)

InterventionParticipants (Number)
CC-5013/Dex107
Placebo/Dex34

[back to top]

Overall Survival

Overall survival was calculated as the time from randomization to death from any cause. (NCT00056160)
Timeframe: 170 weeks (median overall survival of CC-5013/Dex treatment group)

InterventionWeeks (Median)
CC-5013/Dex170.1
Placebo/Dex136.4

[back to top]

Progression-Free Survival

"Progression is defined as a > 25% increase from baseline in myeloma protein production or other signs of disease progression such as hypercalcemia, etc.~In patients with a confirmed Partial Remission, Remission, or Complete Remission, relapse is defined as the first occurrence of any of the following: 1) a myeloma protein increase by than 100% from the lowest level recorded on study, provided the absolute magnitude of this increase is at least 1g/dL for a serum monoclonal protein or at least 500 mg/24 hrs of urine M-protein; 2) a myeloma protein increase above the response criteria for Partial Remission, with the same requirements for the absolute magnitude of the protein increase; 3)reappearance of any myeloma peak that had disappeared while on protocol treatment, provided it meets the same requirements listed above; 4) increase in the size and number of lytic bone lesions recognized on radiographs." (NCT00064038)
Timeframe: From date of initial registration to date of progression/relapse of disease or death from any cause, whichever came first, up to 5 years

Interventionpercentage of participants (Number)
Lenalidomide+Dexamethasone78
Dexamethasone52

[back to top]

Toxicity

Compare the toxicity profile of these regimens, including thrombotic complications, in these patients, based on CTCAE v. 3.0. (NCT00064038)
Timeframe: From time of initiating study treatment until discontinuation of study treatment or of open-label REVLIMID + LOW DOSE DEX, whichever comes last, up to 5 years

,,
InterventionParticipants (Number)
ALT, SGPT (serum glutamic pyruvic transaminase)AST, SGOTAlbumin, serum-low (hypoalbuminemia)AnorexiaApneaArthritis (non-septic)AspirationAtaxia (incoordination)Bilirubin (hyperbilirubinemia)CNS cerebrovascular ischemiaCalcium, serum-high (hypercalcemia)Calcium, serum-low (hypocalcemia)Cardiopulmonary arrest, cause unknown (non-fatal)CataractCognitive disturbanceColitisColitis, infectious (e.g., Clostridium difficile)ConfusionConstipationCreatinineCystitisDehydrationDermatology/Skin-Other (Specify)DiarrheaDizzinessDyspnea (shortness of breath)Edema: limbEncephalopathyFatigue (asthenia, lethargy, malaise)Febrile neutropeniaFever in absence of neutropenia, ANC lt1.0x10e9/LFractureGastritis (including bile reflux gastritis)Gastrointestinal-Other (Specify)Glomerular filtration rateGlucose, serum-high (hyperglycemia)Glucose, serum-low (hypoglycemia)Heartburn/dyspepsiaHemoglobinHemorrhage, CNSHemorrhage, GI - ColonHypertensionHypotensionHypoxiaINR (of prothrombin time)Inf (clin/microbio) w/Gr 3-4 neuts - BloodInf (clin/microbio) w/Gr 3-4 neuts - BronchusInf (clin/microbio) w/Gr 3-4 neuts - LungInf (clin/microbio) w/Gr 3-4 neuts - Up aerodigestInf (clin/microbio) w/Gr 3-4 neuts - Upper airwayInf w/normal ANC or Gr 1-2 neutrophils - BladderInf w/normal ANC or Gr 1-2 neutrophils - BronchusInf w/normal ANC or Gr 1-2 neutrophils - ColonInf w/normal ANC or Gr 1-2 neutrophils - LungInf w/normal ANC or Gr 1-2 neutrophils - SkinInf w/normal ANC or Gr 1-2 neutrophils - UTIInf w/normal ANC or Gr 1-2 neutrophils - Up airwayInf w/normal ANC or Gr 1-2 neutrophils - WoundInf w/normal ANC or Gr 1-2 neutrophils -Nerve-cranInfection with unknown ANC - Lung (pneumonia)Infection with unknown ANC - Upper airway NOSInfection-Other (Specify)InsomniaJoint-functionLeft ventricular diastolic dysfunctionLeft ventricular systolic dysfunctionLeukocytes (total WBC)LymphopeniaMood alteration - agitationMood alteration - anxietyMood alteration - depressionMucositis/stomatitis (clinical exam) - Oral cavityMuscle weakness, not d/t neuropathy - Extrem-lowerMuscle weakness, not d/t neuropathy - body/generalMusculoskeletal/Soft Tissue-Other (Specify)NauseaNeuropathy: motorNeuropathy: sensoryNeutrophils/granulocytes (ANC/AGC)Ocular/Visual-Other (Specify)Ophthalmoplegia/diplopia (double vision)Opportunistic inf associated w/gt=Gr 2 lymphopeniaPTT (Partial thromboplastin time)Pain - Abdomen NOSPain - BackPain - BladderPain - BonePain - Extremity-limbPain - Head/headachePain - JointPain - MusclePain - Pain NOSPain - StomachPain-Other (Specify)Pancreatic endocrine: glucose intolerancePerforation, GI - ColonPerforation, GI - Small bowel NOSPhosphate, serum-low (hypophosphatemia)PlateletsPneumonitis/pulmonary infiltratesPotassium, serum-low (hypokalemia)Prolonged QTc intervalProteinuriaPulmonary/Upper Respiratory-Other (Specify)Rash/desquamationRash: acne/acneiformRash: erythema multiformeRenal failureSVT and nodal arrhythmia - Atrial fibrillationSodium, serum-high (hypernatremia)Sodium, serum-low (hyponatremia)Somnolence/depressed level of consciousnessSpeech impairment (e.g., dysphasia or aphasia)Syncope (fainting)Thrombosis/embolism (vascular access-related)Thrombosis/thrombus/embolismThrombotic microangiopathyThyroid function, low (hypothyroidism)TinnitusVision-blurred visionVomitingWeight loss
Crossover to Rev+Dex00210000120302011011030120218100101200600001010000000000001000001048102013000071010011000010010002323000111200311005000101
Dexamethasone1001000102120110002010111210120110001801500301200002011312100011600055118134211460101220301110111014422000110121200005000310
Lenalidomide and Dexamethasone12121110010711100100010514001911001051061103411151120112231011041011410111101902032110200101103310001057361111000003001219111220

[back to top]

Participants With Bone Marrow Progression

"Bone marrow aspirate was assessed by a central reviewer. Progression is represented in two categories according to changes from baseline in French-American-British (FAB) classification (see Baseline Characteristics):~Baseline classification of refractory anemia (RA) or refractory anemia with ringed sideroblasts (RARS) to a during treatment (plus 30 days) classification of refractory anemia with excess blasts (RAEB).~Any baseline FAB classification to a during treatment (plus 30 days) classification of acute myeloid leukemia (AML)." (NCT00065156)
Timeframe: up to 2 years

Interventionparticipants (Number)
RA/RARS to RAEBRA/RARS/RAEB/CMML to AML
Lenalidomide116

[back to top]

Participants Who Achieved Red Blood Cell (RBC) -Transfusion Independence

"Number of participants who achieved RBC-transfusion independence, which was defined as the absence of an intravenous infusion of any RBC transfusion during any consecutive rolling 56 days during the treatment period (eg, Days 1 to 56, Days 2 to 57, Days 3 to 58, etc), and accompanied by at least a 1 g/dL increase from screening/baseline in hemoglobin." (NCT00065156)
Timeframe: Up to 2 years

Interventionparticipants (Number)
Lenalidomide59

[back to top]

Participants Who Relapsed or Maintained Their Transfusion Independence After Achieving Transfusion Independence During the Study

"Transfusion independence was defined as the absence of an intravenous infusion of any RBC transfusion during any consecutive rolling 56 days during the treatment period (e.g., Days 1 to 56, Days 2 to 57, Days 3 to 58, etc.), and accompanied by at least a 1 g/dL increase from screening/baseline in hemoglobin. Participants who relapsed required a transfusion after the period of transfusion independence. Participants who maintained transfusion independence did not require a transfusion during the remainder of the study." (NCT00065156)
Timeframe: up to 2 years

Interventionparticipants (Number)
Relapsed (had a transfusion after response)Maintained transfusion independence
Lenalidomide3524

[back to top]

Participant Counts of Platelet Response

"Major platelet response: participants with a minimum pretreatment platelet of <100,000/mm^3 in all values within 56 days of start of treatment, an absolute increase of ≥30,000/mm^3 sustained for ≥56 consecutive days. In platelet transfusion-dependent participants, a major response was stabilization of platelet counts and platelet transfusion independence.~Minor platelet response: participants with a minimum pretreatment platelet of <100,000/mm^3, a ≥ 50% increase in platelet count with a net increase >10,000/mm^3 for a consecutive 56-day period in the absence of platelet transfusions." (NCT00065156)
Timeframe: up to 2 years

Interventionparticipants (Number)
MajorMinorNone
Lenalidomide2013

[back to top]

Participant Counts of Cytogenetic Response

"Participants deemed evaluable by the central cytogenetic review had their cytogenetic response categorized as major or minor. A major cytogenetic response was defined as ≥ 20 metaphases recorded at baseline, and at least~1 post baseline evaluation with ≥ 20 metaphases analyzed with no abnormal metaphases observed. A minor cytogenetic response was defined as ≥ 20 metaphases analyzed at baseline, and at least 1 post baseline evaluation with ≥ 20 metaphases analyzed with a ≥ 50% reduction in the proportion of hematopoietic cells with cytogenetic abnormalities compared with baseline." (NCT00065156)
Timeframe: up to 2 years

Interventionparticipants (Number)
MajorMinorNone
Lenalidomide182014

[back to top]

Participants With Adverse Experiences

"Counts of study participants who had adverse events (AEs) during the study. A participant with multiple occurrences of an adverse event within a category is counted only once in that category. Adverse events were evaluated by the investigator.~The National Cancer Institute (NCI)'s Common Toxicity Criteria for AEs (NCI CTC) was used to grade AE severity. Severity grade 3= severe and undesirable AE. Severity grade 4= life-threatening or disabling AE." (NCT00065156)
Timeframe: Up to 2 Years

Interventionparticipants (Number)
At least one AEAt least one AE related to study drugAt least one NCI CTC grade 3-4 AEAt least one NCI CTC grade 3-4 AE related to drugAt least one serious AEAt least one serious AE related to study drugAE leading to dose reduction or interruptionAE leading to discontinuation of study drug
Lenalidomide148143140131894013147

[back to top]

Participant Counts of Absolute Neutrophil Count (ANC) Response

"Major neutrophil response: participants with a minimum pretreatment ANC concentration of < 1500/mm^3 in all values obtained within 56 days of start of treatment, a ≥ 100% increase or an absolute increase of~≥ 500/mm^3, whichever was greater (at least to be ≥ 500/mm^3), sustained for 56 consecutive days. Minor neutrophil response: participants with a minimum pretreatment ANC concentration of < 1500/mm^3, an increase in ANC concentration of ≥ 100% sustained for 56 consecutive days." (NCT00065156)
Timeframe: up to 2 years

Interventionparticipant (Number)
MajorMinorNone
Lenalidomide9018

[back to top]

Participants With a >= 50% Decrease From Baseline in Red Blood Cell (RBC) Transfusion Requirements Over Any Consecutive 56 Days During Study

A participant was categorized as having a transfusion reduction response if there was a ≥ 50% decrease from pretreatment transfusion requirements (before the start of the study mediation) compared to any consecutive 56 days during the study (i.e. post treatment). (NCT00065156)
Timeframe: Baseline (Day -54 to Day 0), During study (Day 1 up to 2 years)

Interventionparticipant (Number)
Lenalidomide70

[back to top]

Kaplan Meier Estimate for Duration of Transfusion Independence Response

Duration of response is measured from the first of the consecutive 56 days during which the participant was free of RBC transfusions to the date of the first RBC transfusion after this period. Duration of response was censored at the date of last visit for participants who maintained transfusion independence. (NCT00065156)
Timeframe: up to 2 years

Interventionweeks (Median)
Lenalidomide97.0

[back to top]

Time to Transfusion Independence

"Transfusion independence was defined as the absence of an intravenous infusion of any RBC transfusion during any consecutive rolling 56 days during the treatment period (e.g., Days 1 to 56, Days 2 to 57, Days 3 to 58, etc.), and accompanied by at least a 1 g/dL increase from screening/baseline in hemoglobin. Time to transfusion independence was defined as the day of the first dose of study drug to the first day of the first 56-day RBC transfusion-free period." (NCT00065156)
Timeframe: up to 2 years

Interventionweeks (Mean)
Lenalidomide6.2

[back to top]

Change in Hemoglobin Concentration From Baseline to Maximum Value During Response Period for Responders

The change from baseline in hemoglobin for participants who became RBC-transfusion independent. The maximum hemoglobin value obtained during the response period is used in the calculation of change from baseline. (NCT00065156)
Timeframe: Baseline (Day -54 to Day 0), During study (Day 1 up to 2 years)

Interventiong/dL (Mean)
Lenalidomide6.1

[back to top]

Participants With Complete or Partial Bone Marrow Improvement

Bone marrow aspirates were assessed by a central reviewer. A complete bone marrow improvement required a baseline French-American-British (FAB) classification (see Baseline Characteristics) of refractory anemia (RA), refractory anemia with ringed sideroblasts (RARS), refractory anemia with excess blasts (RAEB) or chronic myelomonocytic leukemia (CMML) and a during study assessment of no MDS. A partial bone marrow improvement reflected an improved FAB classification compared to baseline (e.g. RARS to RA) but evidence of MDS continued to exist. (NCT00065156)
Timeframe: up to 2 years

Interventionparticipants (Number)
Complete bone marrow improvementPartial bone marrow improvement
Lenalidomide2215

[back to top]

Efficacy of CC-5013 in Myelofibrosis

"Response evaluation, sustained for 2 weeks: Complete Remission (Neutrophil count between 1 to 10 x 10^9/L without peripheral blasts in blood or bone marrow); Partial Hematologic Response/Partial Remission (Increase in neutrophil by 50% + above 10^9/L for neutropenia); Hematologic Improvement (increase in Neutrophil count, hemoglobin, platelet count or reduction in blood/marrow blasts) or No Response.~If nine or < patients respond to therapy (response other than 'No Response'), therapy declared ineffective. However, if 11 or > patients respond to therapy, therapy considered efficacious." (NCT00087672)
Timeframe: 3 - 4 Months for all patients; 24 months for responders

InterventionParticipants (Number)
Complete RemissionPartial RemissionHematologic ImprovementNo Response
CC-501379129

[back to top]

Number of Patients Removed From Study Treatment Due to Toxicities

(NCT00091260)
Timeframe: 1 year

Interventionparticipants (Number)
Revlimid31

[back to top]

Number of Patients Who Received Both CC-5013 and Dexamethasone and Had a Hematologic Response

(NCT00091260)
Timeframe: 1 year

Interventionparticipants (Number)
Revlimid26

[back to top]

Number of Patients With Hematologic Response With Single-agent CC-5013

"Complete response = Absence of detectable monoclonal protein in serum or urine by immunofixation electrophoresis, less than 5% plasma cells on bone marrow biopsy without clonal dominance of kappa or lambda isotype, and normal serum free light chain assay.~Partial response= For patients with detectable and quantifiable monoclonal marrow plasmacytosis= a reduction of 50% or more in plasma cells as a percentage of nucleated bone marrow cells. For patients with a detectable monoclonal peak on serum or urine protein electrophoresis= a reduction in the peak height of 50% or more.~For patients with quantifiable urinary kappa or lambda chain concentration= a 50% reduction in daily light chain excretion in 24 hour urine.~For patients with an elevated serum free light chain assay, a reduction of 50% or more." (NCT00091260)
Timeframe: 3 months

InterventionParticipants (Count of Participants)
Revlimid5

[back to top]

Number of Participants With Adverse Events on Combination Therapy of CC-5013+Rituximab

Number of Participants with Adverse Events on Combination Therapy of CC-5013+Rituximab, Graded According to NCI CTCAE Version 3.0 (NCT00096044)
Timeframe: Up to 30 days from last date of institution of combination therapy of CC-5013+Rituximab.

InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4
Oral Lenalidomide1033

[back to top]

Percentage of Patients Achieving a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) on Combination Therapy of CC-5013+Rituximab

Percentage of patients achieving CR, PR or maintaining SD using the 1996 NCI-WF Criteria. CR: absence of lymph nodes and constitutional symptoms; no hepatomegaly or splenomegaly by physical examination; neutrophil count >1500/μL; platelet count >100,000/μL; untransfused hemoglobin concentration >11.0g/dL; lymphocyte count <4000/μL; bone marrow sample must be at least normocellular for age; with less than 30% of nucleated cells being lymphocytes and no lymphoid nodules. PR: ≥50% decrease in lymphocyte count from baseline; ≥50% reduction in lymph nodes from baseline; ≥50% reduction in the size of the liver/spleen from baseline; neutrophil count ≥1500/μL or ≥50% improvement from baseline; platelet count ≥100,000/μL or ≥50% improvement from baseline; untransfused hemoglobin concentration ≥11.0g/dL or ≥50% improvement from baseline. Patients who have not exhibited as reappearance of malignant CLL clone on flow cytometry or by PCR analysis in blood or bone marrow, are considered to have SD. (NCT00096044)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Oral Lenalidomide100

[back to top]

Percentage of Patients Achieving a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) on Single Agent CC-5013 at 6 Months

Percentage of patients achieving CR, PR or maintaining SD using the 1996 NCI-WF Criteria. CR: absence of lymph nodes and constitutional symptoms; no hepatomegaly or splenomegaly by physical examination; neutrophil count >1500/μL; platelet count >100,000/μL; untransfused hemoglobin concentration >11.0g/dL; lymphocyte count <4000/μL; bone marrow sample must be at least normocellular for age; with less than 30% of nucleated cells being lymphocytes and no lymphoid nodules. PR: ≥50% decrease in lymphocyte count from baseline; ≥50% reduction in lymph nodes from baseline; ≥50% reduction in the size of the liver/spleen from baseline; neutrophil count ≥1500/μL or ≥50% improvement from baseline; platelet count ≥100,000/μL or ≥50% improvement from baseline; untransfused hemoglobin concentration ≥11.0g/dL or ≥50% improvement from baseline. Patients have not exhibited as reappearance of malignant CLL clone on flow cytometry or by PCR analysis in blood or bone marrow, are considered to have SD. (NCT00096044)
Timeframe: at 6 Months

Interventionpercentage of participants (Number)
Oral Lenalidomide68.9

[back to top]

Time to Progression for the Combination Therapy of CC-5013+Rituximab

Progressive disease is defined as reappearance of malignant CLL clone on flow cytometry or by PCR analysis in blood or bone marrow using the 1996 NCI-WF Criteria. (NCT00096044)
Timeframe: Every month up to 6 months and every 3 months thereafter up to 5 years

Interventionmonths (Median)
Oral Lenalidomide18.7

[back to top]

Number of Participants With Adverse Events on Single Agent CC-5013

"Number of Participants with Adverse Events on Single Agent CC-5013, Graded According to NCI CTCAE Version 3.0~Please refer to the adverse event reporting for more detail." (NCT00096044)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4Grade 5
Oral Lenalidomide037332

[back to top]

Time to Progression for Single Agent CC-5013

Progressive disease is defined as reappearance of malignant CLL clone on flow cytometry or by PCR analysis in blood or bone marrow using the 1996 NCI-WF Criteria. (NCT00096044)
Timeframe: 5 years

Interventionmonths (Median)
Oral Lenalidomide23.0

[back to top]

Proportion of Patients With Objective Response (First Phase, Step 2)

"Objective response is defined as either complete response (CR) or partial response (PR). Patients who have complete disappearance of an M-protein and no evidence of myeloma in the bone marrow are considered to have CR. PR requires all the following: (1) ≥50% reduction in the level of the serum monoclonal paraprotein. (2) Reduction in 24-hour urinary light chain excretion either by ≥90% or to <200 mg. (3)For patients with non-secretory (or oligosecretory) myeloma only, a ≥50% reduction in plasma cells in a bone marrow aspirate and on trephine biopsy must be documented. (4)50% reduction in size of soft tissue plasmacytoma (by radiography or clinical examination). (5) No increase in the number or size of lytic bone lesions (development of a compression fracture does not exclude response).~As the expansion phase was a substudy terminated early with only 7 patients enrolled, the clinical results presented are mainly for the first phase only." (NCT00098475)
Timeframe: Assessed every 4 weeks for 16 weeks during Step 2

InterventionProportion of patients (Number)
Arm I (Lenalidomide, Dexamethasone)0
Arm II (Lenalidomide, Low-dose Dexamethasone)0

[back to top]

Proportion of Patients With Objective Response (First Phase, Step 1)

"Objective response is defined as either complete response (CR) or partial response (PR). Patients who have complete disappearance of an M-protein and no evidence of myeloma in the bone marrow are considered to have CR. PR requires all the following: (1) ≥50% reduction in the level of the serum monoclonal paraprotein. (2) Reduction in 24-hour urinary light chain excretion either by ≥90% or to <200 mg. (3)For patients with non-secretory (or oligosecretory) myeloma only, a ≥50% reduction in plasma cells in a bone marrow aspirate and on trephine biopsy must be documented. (4)50% reduction in size of soft tissue plasmacytoma (by radiography or clinical examination). (5) No increase in the number or size of lytic bone lesions (development of a compression fracture does not exclude response).~As the expansion phase was a substudy terminated early with only 7 patients enrolled, the clinical results presented are mainly for the first phase only." (NCT00098475)
Timeframe: Assessed every 4 weeks for 16 weeks during Step 1

InterventionProportion of patients (Number)
Arm I (Lenalidomide, Dexamethasone)0.79
Arm II (Lenalidomide, Low-dose Dexamethasone)0.683

[back to top]

Clinical Responses in Patients With Metastatic Ocular Melanoma

Clinical response is assessed by the Response Evaluation Criteria for Adverse Events in Solid Tumors (RECIST). Complete response (CR) is disappearance of all target lesions. Partial response (PR) is at least a 30% decrease in the sum of the longest diameter (LD) of target lesions. Progressive disease (PD) is at least a 20% increase in the sum of the LD of target lesions. Stable disease is neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT00109005)
Timeframe: 12 months

InterventionParticipants (Number)
Complete ResponsePartial ResponseProgressive DiseaseStable Disease
Cohort 1 & 2 -25 mg & 5 mg Lenalidomide (Revlimid)0097

[back to top]

Number of Participants With Adverse Events

Here is the number of participants with adverse events. For the detailed list of adverse events see the adverse event module. (NCT00109005)
Timeframe: 24 months

InterventionParticipants (Number)
Cohort 1 - 25 mg Lenalidomide (Revlimid)8
Cohort 2 - 5 mg Lenalidomide (Revlimid)9

[back to top]

Change From Baseline in the Brief Pain Inventory (BPI) Total Score at Week 12

Participants completed the Brief Pain Inventory which asks twelve questions that are rated on an eleven-point scale in which 0=most positive outcome and 10=the most negative outcome for a total scale of 0-120. BPI contains questions that concern the level of pain over the last week and the level of pain right now, the extent to which pain interfered with sleep, normal activities, ability to work, relationships, walking etc. Week 12 values are compared to baseline values. Negative change values indicate improvement. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-4.8
Placebo to Lenalidomide-3.7

[back to top]

"Change From Baseline in Mechanically Evoked (Allodynia) Numeric Rating Scale (NRS) Score at Week 12"

The investigator rated the degree of allodynia on both the CRPS-affected limb on an eleven-point scale where 0=no pain and 10=worst pain imaginable. This outcome compares the baseline values for the CRPS affected-limb to the values at week 12. Negative change values indicate improvement. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-0.8
Placebo to Lenalidomide-0.1

[back to top]

Change From Baseline in the Evening Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score at Week 12

Participants rated the intensity of pain in the CRPS-affected limb twice each day in a diary. The PI-NRS is an eleven point scale with 0=no pain and 10=worst pain imaginable. The evening pain ratings at baseline and Week 12 are compared. Negative changes indicate improvement in level of pain. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-0.6
Placebo to Lenalidomide-0.4

[back to top]

Change From Baseline in Daily Sleep Assessment Average Score at Week 12

Participants rated how much CRPS pain interfered with their sleep each day in a diary. The Sleep Assessment uses an eleven point scale for four questions. Questions concern ability to fall asleep, ability to stay asleep, how refreshed the participant feels upon waking and how alert the participant is during the day. All use a scale of 0-10, where the higher number is the positive response (e.g. 0=Pain completely interferes with sleep and 10=Pain does not interfere). The mean of all four responses was calculated if at least 3 of the 4 questions had a value. Week 12 values are compared to baseline values. Positive change values indicate improvement. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide0.7
Placebo to Lenalidomide0.7

[back to top]

Change From Baseline in the Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score Using Averaged Morning and Evening Readings at Week 12

Participants rated the intensity of pain in the CRPS-affected limb twice each day in a diary. Morning and evening scores are averaged. The PI-NRS is an eleven point scale with 0=no pain and 10=worst pain imaginable. Week 12 values are compared to baseline values. Negative changes indicate improvement in level of pain. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-0.6
Placebo to Lenalidomide-0.4

[back to top]

Change From Baseline in Activity Level Rating Using a Numeric Rating Scale (NRS) at Week 12

Participants rated how the activity level on a given day compares with their activity level prior to the start of treatment. A seven-point scale is used with -3=much worse and +3=much better. Positive change values indicate improvement. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide0.1
Placebo to Lenalidomide0.2

[back to top]

Change From Baseline in Participant Assessment of CRPS Symptoms Total Score at Week 12

Participants rated twelve CRPS symptoms using a four-point rating scale in which 1=the most positive outcome and 4= the most negative outcome for a total scale of 12-48. Week 12 values are compared to baseline values. Negative change values indicate improvement. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-2.4
Placebo to Lenalidomide-1.4

[back to top]

Change From Baseline in the Total Score of the Short Form McGill Pain Questionnaire (SF-MPQ) at Week 12

Short Form McGill Pain Questionnaire (SF-MPQ) is comprised of 15 pain qualities that are rated by the participant on a 4 point scale with 0=none and 3=severe. The scale for the Total Score is 0-45. Week 12 values are compared to baseline values. Negative changes indicate improvement in level of pain. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-5.2
Placebo to Lenalidomide-3.0

[back to top]

Percentage of Participants Who Have a >= 30% Reduction (Improvement) in the Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score From Baseline to the Last Assessment

Participants rated the intensity of pain in the CRPS-affected limb twice each day in a diary. The PI-NRS is an eleven point scale with 0=no pain and 10=worst pain imaginable. Responders are participants who completed 12 weeks of treatment and their week 12 PI-NRS score showed at least a 30% improvement from baseline. Participants who did not complete 12 weeks of treatment are considered non-responders. (NCT00109772)
Timeframe: Day 0, Week 12

,
Interventionpercentage of participants (Number)
RespondersNon-responders
Lenalidomide16.183.9
Placebo to Lenalidomide16.183.9

[back to top]

Participants With Treatment-Emergent Adverse Events in the Double-Blind Period or the Extension Period

"Counts of study participants who had adverse events (AEs) while treated in either the Double-blind or Extension Periods. The NCI Common Terminology Criteria for Adverse Events (CTCAE), Version 3.0 was used by the investigator to grade the severity of the AEs: 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.~AEs are also summarized by whether they were serious, related to treatment and whether the AE caused treatment to be altered.~A serious AE (SAE) was any event that resulted in death, was life-threatening, required inpatient hospitalization or prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity or congenital anomaly/birth defect or was an important medical event could have jeopardized the patient's safety or required medical or surgical intervention to prevent one of the outcomes listed above." (NCT00109772)
Timeframe: Day 1 up to week 158

,
Interventionparticipants (Number)
Adverse event (AE)Serious adverse event (SAE)AE leading to study drug discontinuationAE leading to dose reduction or interruptionTreatment-related AETreatment-related SAEGrade 2 or higher AEGrade 3 or higher AE
Lenalidomide831229106816315
Placebo to Lenalidomide85162986656818

[back to top]

Participants Who Had a Change to CRPS Pain Medication During the Treatment Period

Participants who had any change in CRPS medication during the double-blind treatment period (up to week 12) are summarized. Changes include additions, discontinuations or dosage change of CRPS medication(s). (NCT00109772)
Timeframe: Day 1 to week 12

,
Interventionparticipants (Number)
Change to CRPS medication(s)No change to CRPS medication
Lenalidomide1176
Placebo to Lenalidomide786

[back to top]

Patient Global Impression of Change (PGIC) at Week 12

The Patient Global Impression of Change asks the question: Overall, how would you rate your CRPS condition since the start of study drug? Answers are represented on a seven-point scale with -3=much worst and +3=much better. (NCT00109772)
Timeframe: Week 12

Interventionunits on a scale (Mean)
Lenalidomide0.2
Placebo to Lenalidomide0.2

[back to top]

Difference in Allodynia Rating Between the CRPS-affected Limb and the Normal Limb at Week 12

The investigator rated the degree of allodynia on both the CRPS-affected limb and the normal (or less-affected) limb on an eleven-point scale where 0=no pain and 10=worst pain imaginable. This outcome compares the values for the CRPS affected-limb to the normal limb at week 12. (NCT00109772)
Timeframe: Week 12

Interventionunits on a scale (Mean)
Lenalidomide3.7
Placebo to Lenalidomide4.1

[back to top]

Change From Baseline in the Profile of Mood States (POMS) at Week 12

Participants completed the Profile of Mood States questionnaire that asks participants to rate how each of 65 words reflected their mood in the past week on a 5-point scale with 0=not at all and 4=extremely for a total scale of 0-260. Week 12 values are compared to baseline values. Negative change values indicate improvement. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-9.6
Placebo to Lenalidomide-4.5

[back to top]

Change From Baseline in the Morning Complex Regional Pain Syndrome (CRPS) Pain Intensity Numeric Rating Scale (PI-NRS) Score at Week 12

Participants rated the intensity of pain in the CRPS-affected limb twice each day in a diary. The PI-NRS is an eleven point scale with 0=no pain and 10=worst pain imaginable. The morning pain ratings at baseline and Week 12 are compared. Negative changes indicate improvement in level of pain. (NCT00109772)
Timeframe: Day 0, week 12

Interventionunits on a scale (Mean)
Lenalidomide-0.6
Placebo to Lenalidomide-0.4

[back to top]

Change From Baseline in the Maximal Composite Sensory Nerve Conduction Velocity at Week 12

An electrophysiological evaluation using standard electrophysiological and electromyography to measure the speed and extent of nerve conduction. Week 12 values are compared to baseline values for maximal sensory nerve conduct velocity. (NCT00109772)
Timeframe: Day 0, week 12

Interventionmeters/second (Mean)
Lenalidomide1.2
Placebo to Lenalidomide0.1

[back to top]

Change From Baseline in the Maximal Composite Motor Nerve Conduction Velocity at Week 12

An electrophysiological evaluation using standard electrophysiological and electromyography to measure the speed and extent of nerve conduction. Week 12 values are compared to baseline values for maximal motor nerve conduct velocity. (NCT00109772)
Timeframe: Day 0, week 12

Interventionmeters/second (Mean)
Lenalidomide0.3
Placebo to Lenalidomide0.1

[back to top]

Maximum Tolerated Dose (MTD)

The highest dose tested (Total Marrow Irradiation) in which there is no treatment related mortality and none or only one patient experienced dose limited toxicity (DLT) attributable to the study drug(s), when at least six were fully treated at that dose and fully followed for toxicity. The MTD is one dose level below the lowest dose tested in which 2 or more patients experienced DLT attributable to the treatment or there was a treatment related death. At least 6 patients will be treated at the MTD. (NCT00112827)
Timeframe: 8 weeks from start of treatment, up to 2 years

InterventioncGy (Number)
Treatment Arm1600

[back to top]

Overall Survival

Estimated using the product-limit method of Kaplan and Meier. Event defined as death due to any cause. (NCT00112827)
Timeframe: From date of treatment until the date of death from any cause, assessed up to 14 years

Interventionmonths (Median)
Treatment Arm95.8

[back to top]

Number of Subjects With Response

Response defined as complete response or very good partial response. Complete response defined as the absence of bone marrow or blood findings of multiple myeloma on at least 2 measurements at a minimum of a 6 week interval. Thus all evidence of serum and urinary M-components must disappear on electrophoresis as well as by immunofixation studies. The follow-up bone marrow may not contain more than 5% plasma cells on aspiration or core biopsy and no evidence of increasing anemia. Skeletal X-rays must either show recalcification or no change in osteolytic lesions. Resolution of soft tissue plasmocytomas. Very good partial response defined as reduction of bone marrow or blood findings of multiple myeloma on at least 2 measurements at a minimum of a 6 week interval by greater than or equal to 90%. (NCT00112827)
Timeframe: Evaluated after each course until completion of treatment.

InterventionParticipants (Count of Participants)
Phase 1 Cohort 1 (Total TMI Dose: 1000 cGy)3
Phase 1 Cohort 2 (Total TMI Dose: 1200 cGy)3
Phase 1 Cohort 3 (Total TMI Dose: 1400 cGy)2
Phase 1 Cohort 4 & Phase 2 MTD (Total TMI Dose:1600 cGy)20
Phase 1 Cohort 5 (Total TMI Dose: 1800 cGy)4

[back to top]

Response to Autologous Hematopoietic Stem-cell Transplant (HSCT) at Day 100

"Response was defined according to International Myeloma Working Group criteria (2006)~Complete Response: Complete disappearance of M-protein from serum & urine on immunofixation, normalization of Free Light Chain (FLC) ratio & <5% plasma cells in bone marrow (BM)~Partial Response: >= 50% reduction in serum M-Component and/or Urine M-Component >= 90% reduction or <200 mg per 24 hours; or >= 50% decrease in difference between involved and uninvolved FLC levels~Marginal Response: 25-49% reduction in serum M-component & urine M-component by 50-89% which still exceeds 200mg/24hour~Progressive Disease: Defined in primary outcome measure~Stable Disease: Not meeting any of the criteria above" (NCT00114101)
Timeframe: Day 100

,
Interventionparticipants (Number)
Complete responsePartial responseMarginal responseStable diseaseProgressive diseaseUnknown
Lenalidomide Maintenance67115113800
Placebo Maintenance7910953231

[back to top]

Number of Participants With Progression, Death or Diagnosis of Second Primary Malignancy

Patients who develop progression (defined in primary outcome measure), died or develop a new primary malignancy (cancer) will summarized in this outcome. (NCT00114101)
Timeframe: Duration of study (up to 10 years)

Interventionparticipants (Number)
Lenalidomide Maintenance92
Placebo Maintenance133

[back to top]

Overall Survival

Overall Survival was measured from the date of randomization to date of death due to any cause. OS was estimated using the Kaplan Meier method. (NCT00114101)
Timeframe: Duration of study (up to 10 years)

Interventionmonths (Median)
Lenalidomide MaintenanceNA
Placebo MaintenanceNA

[back to top]

Time to Progression

"Time to progression (TTP) was defined as the date of transplant to date of progression or death due to any cause, whichever occurs first. TTP was estimated using the Kaplan Meier method.~Progression was defined per the International Myeloma Working Group definition as one more of the following:~25% increase in serum M-component (absolute increase >= 0.5g/dl)~25% increase in urine M-component (absolute increase >= 200mg/24hour~25% increase in the difference between involved and uninvolved Free Light Chain levels (absolute increase >= 10mg/dl)~25 % increase in bone marrow plasma cell percentage (absolute increase of >=10%)~Definite development of new bone lesion or soft tissue plasmacytomas~Development of hypercalcemia" (NCT00114101)
Timeframe: Duration of study (up to 10years)

Interventionmonths (Median)
Lenalidomide Maintenance39
Placebo Maintenance21

[back to top]

Minor Response Rate

A minor response is defined as having achieved >25% but less than 50% reduction in serum IgM levels. (NCT00142168)
Timeframe: 34.3 months

Interventionparticipants (Number)
Lenalidomide and Rituximab4

[back to top]

Major Response Rate

Major response rate is the number of participants who achieve at a PR or better. A PR or better will be defined as achieving a >50% reduction in serum IgM levels. (NCT00142168)
Timeframe: 34.3 months

Interventionparticipants (Number)
Lenalidomide and Rituximab4

[back to top]

Time to Progression

Time to progression is measured as the length in time in months from starting therapy until progression, defined as 25% increase in serum IgM from nadir. (NCT00142168)
Timeframe: 34.3 months

Interventionmonths (Median)
Lenalidomide and Rituximab17.1

[back to top]

Overall Response

Overall response is the total number of participants who respond to therapy. Patients achieving a complete response (CR) will be defined as having achieved resolution of all symptoms, normalization of their serum IgM levels with complete disappearance of their IgM paraprotein by immunofixation, and resolution of any adenopathy or splenomegaly during any point while in this study and normal bone marrow biopsy. Patients achieving a partial response (PR) and a minor response (MR) will be defined as achieving a > 50% and > 25% reduction in serum IgM levels, respectively, during any point while in this study. Patients with stable disease (SD) will be defined as having < 25% change in serum IgM levels, in the absence of new or increasing adenopathy or splenomegaly and/or other progressive signs or symptoms of WMduring any point while in this study. (NCT00142168)
Timeframe: 34.3 months

Interventionparticipants (Number)
Lenalidomide and Rituximab8

[back to top]

Participants' Response Based on Bone Marrow Samples by the International MDS Working Group (IWG 2000) During Double-blind Period

The IWG criteria for bone marrow improvement: a complete remission is bone marrow sampling showing less than 5% myeloblasts with normal maturation of all cell lines, with no evidence for dysplasia. A partial remission is ≥ 50% decrease in blasts over pre-treatment. Bone marrow progression is a ≥ 50% increase in blasts that exceed the top range of the pretreatment percentile range: a) <5% blasts b) 5-10% blasts c) 10-20% blasts d) 20-30% blasts. For example, a participant with <5% blasts pretreatment with an on study blast increase of 50% which is now >5% showed bone marrow progression. (NCT00179621)
Timeframe: up to 52 weeks

,,
InterventionParticipants (Number)
Complete remissionPartial remissionStable DiseaseProgression
Lenalidomide 10 mg121333
Lenalidomide 5 mg75354
Placebo00373

[back to top]

Participants' Response in Absolute Neutrophil Counts as Defined by the International MDS Working Group (IWG 2000) During Double-blind Period

A major neutrophil response is defined by the International MDS Working Group (IWG) criteria as at least a 100% increase, or an absolute increase of ≥500/mm^3 for participants with absolute neutrophil counts (ANC) of less than 1,500/mm^3 before therapy, whichever is greater. A minor response for such participants is defined as an ANC increase of at least 100%, but absolute increase <500/mm^3. (NCT00179621)
Timeframe: up to week 52

,,
InterventionParticipants (Number)
MajorMinorNone
Lenalidomide 10 mg2114
Lenalidomide 5 mg3015
Placebo109

[back to top]

Participants' Response in Platelet Counts as Defined by the International MDS Working Group (IWG 2000) During Double-blind Period

The International MDS Working Group (IWG) defines a major platelet response for participants with a pre-treatment platelet count of <100,000/mm^3 as an absolute increase of ≥30,000/mm^3 whereas a minor response is defined as a ≥50% increase in platelet count with a net increase greater than 10,000/mm^3 but less than 30,000/mm^3. (NCT00179621)
Timeframe: up to 52 weeks

,,
InterventionParticipants (Number)
MajorMinorNone
Lenalidomide 10 mg103
Lenalidomide 5 mg105
Placebo003

[back to top]

Summary of Participants Who Had Adverse Events (AE) During the Double-blind Period

"Counts of study participants who had adverse events (AEs) during the double-blind period by MedDRA System Organ Class (SOC) and preferred term. A participant with multiple occurrences of an adverse event within a category is counted only once in that category. Adverse events were evaluated by the investigator.~The National Cancer Institute (NCI)'s Common Terminology Criteria for AEs (CTCAE) was used to grade AE severity. Severity grade 3= severe and undesirable AE. Severity grade 4= life-threatening or disabling AE." (NCT00179621)
Timeframe: up to week 52

,,
Interventionparticipants (Number)
At least one AEAt least one AE related to study drugAt least one NCI CTCAE grade 3-4 AEAt least one related NCI CTCAE grade 3-4 AEAt least one serious AEAt least one serious AE related to study drugAn AE leading to discontinuation of study drugAn AE leading to dose reduction or interruptionDeaths within 30 days of last dose of study drug
Lenalidomide 10 mg6966656132136514
Lenalidomide 5 mg69686261311712442
Placebo64342913141354

[back to top]

Participants Who Progressed to Acute Myeloid Leukemia (AML) During the Study

Number of participants who progressed to acute myeloid leukemia during the study, summarized at three different timepoints: first 16 weeks of the double-blind study, week 52 of the double-blind study, and up to 36 months which includes the double-blind and open-label periods of the study. The counts are cumulative by timeframe. (NCT00179621)
Timeframe: up to 3 years

,,
InterventionParticipants (Number)
Double-Blind (first 16 weeks)Double-Blind (52 weeks)Double-Blind + Open-Label
Lenalidomide 10 mg0215
Lenalidomide 5 mg2716
Placebo2421

[back to top]

Change From Baseline in the Functional Assessment of Cancer Therapy-Anemia (FACT-An) Endpoints at Week 12

"The Functional Assessment of Cancer Therapy-Anemia (FACT-An) questionnaire (Yellen, 1997) was used to assess health-related quality of life (HRQoL).~In addition to general HRQoL, the FACT-An measures the impact of fatigue and other anemia-related symptoms on patient functioning. The overall score range for the FACT-An is 0-188. Higher scores indicate better HRQoL." (NCT00179621)
Timeframe: Baseline, Week 12

Interventionunits on a scale (Mean)
Placebo-2.5
Lenalidomide 5 mg5.9
Lenalidomide 10 mg5.8

[back to top]

Change From Baseline in the Trial Outcome Index-Anemia (TOI-An) Endpoints at Week 12

The Trial Outcome Index-Anemia (TOI-An) composed of the physical and functional subscales of the FACT-G along with the Anemia subscale was used to assess health-related quality of life (HRQoL). The overall score range for the TOI-An is 0-136. Higher scores indicate better HRQoL. (NCT00179621)
Timeframe: Baseline, Week 12

Interventionunits on a scale (Mean)
Placebo-1.1
Lenalidomide 5 mg5.6
Lenalidomide 10 mg4.9

[back to top]

Change From Baseline in the Trial Outcome Index-Fatigue (TOI-F) Endpoints at Week 12

The Trial Outcome Index-Fatigue(TOI-F) composed of the physical and functional subscales of the FACT-G along with the fatigue items from the Anemia subscale was used to assess health-related quality of life (HRQoL). The overall score range for the TOI-F is 0-108. Higher scores indicate better HRQoL. (NCT00179621)
Timeframe: Baseline, Week 12

Interventionunits on a scale (Mean)
Placebo-0.8
Lenalidomide 5 mg4.8
Lenalidomide 10 mg3.9

[back to top]

Duration of Red Blood Cell (RBC) Transfusion Independence for Participants Who Became RBC Transfusion Independent for at Least 182 Days

Mean number of weeks that participants who achieved RBC transfusion independence for at least 182 days were able to maintain RBC transfusion independence. Both double-blind and open-label periods are included. (NCT00179621)
Timeframe: up to 3 years

InterventionWeeks (Mean)
Placebo61.4
Lenalidomide 5 mg107.7
Lenalidomide 10 mg108.6

[back to top]

Kaplan Meier Estimates of Overall Survival by Randomized Group

Kaplan Meier estimate for median length of survival for study participants as they were randomized at the start of the study. (NCT00179621)
Timeframe: up to 3 years

InterventionMonths (Median)
Placebo42.4
Lenalidomide 5 mgNA
Lenalidomide 10 mg44.5

[back to top]

Maximum Change From Baseline in Hemoglobin During the Double-blind Period for Participants Who Became Red Blood Cell (RBC) Transfusion Independent for at Least 182 Days

For participants who became RBC transfusion independent for at least 182 days during the double-blind study period, the mean maximum change from baseline in hemoglobin is summarized. (NCT00179621)
Timeframe: Baseline, up to 52 weeks

Interventiong/dL (Mean)
Placebo2.0
Lenalidomide 5 mg5.5
Lenalidomide 10 mg6.0

[back to top]

Participant Count of Deaths During Double-blind and Open-label by Randomized Group

Count of participant deaths throughout the entire study and reported by the original treatment assignment. (NCT00179621)
Timeframe: up to 3 years

InterventionParticipants (Number)
Placebo35
Lenalidomide 5 mg32
Lenalidomide 10 mg34

[back to top]

Participants Who Achieved Red Blood Cell (RBC) Transfusion Independence for >= 26 Weeks (182 Days)

The count of study participants who had no RBC transfusions for 26 consecutive weeks or more during the double-blind period. (NCT00179621)
Timeframe: Up to 52 weeks

InterventionParticipants (Number)
Placebo3
Lenalidomide 5 mg20
Lenalidomide 10 mg23

[back to top]

Participants Who Achieved Red Blood Cell (RBC) Transfusion Independence for 56 Days

Count of study participants who had no RBC transfusions during any 56 or more consecutive study days during the double-blind period. (NCT00179621)
Timeframe: Up to 52 weeks

InterventionParticipants (Number)
Placebo4
Lenalidomide 5 mg24
Lenalidomide 10 mg25

[back to top]

Participants Showing Cytogenetic Response by the International MDS Working Group (IWG 2000) During Double-blind Period as Evaluated by Central Review

The IWG criteria for evaluating cytogenetic response require a minimum of 20 baseline and post-baseline analyzable metaphases using conventional cytogenetic techniques. A major cytogenetic response is defined as no detectable cytogenetic abnormality if preexisting abnormality was present whereas a minor response requires ≥50% reduction in abnormal metaphases. Progression could be concluded based on as few as 3 metaphases if there were additional abnormalities. The best response is represented. (NCT00179621)
Timeframe: up to 52 weeks

,,
InterventionParticipants (Number)
Major responseMinor responseCytogenetic progressionNot evaluable/data not available
Lenalidomide 10 mg10781
Lenalidomide 5 mg531010
Placebo00510

[back to top]

Overall Incidence of Adverse Events

Data from all subjects who received any study drug were included in the analysis. Adverse events were classified using the Medical Dictionary for Regulatory Activities (MedDRA) classification system. A subject having the same event more than once was counted only once. Adverse events were summarized by worst NCI (National Cancer Institute) CTCAE (Common Terminology Criteria for Adverse Events) VERSION 3.0 grade. Incidence was defined as the number of subjects who experienced an adverse event within their period of participation in this study. (NCT00179647)
Timeframe: Median time-on-study=18.3 weeks

InterventionParticipants (Number)
Lenalidomide1877

[back to top]

Duration of Response

The duration of response was calculated as the first response assessment demonstrating evidence of at least a partial response to the first documentation of progressive disease (as determined by computed tomography scan) or death due to NHL, whichever occurred first. For participants without documentation of progression, the duration of response was censored at the last date of tumor assessment indicating no progression. Median was based on the Kaplan-Meier estimate. (NCT00179660)
Timeframe: From enrollment through study completion. Median duration on study was 3.7 months, with a maximum of 32.5 months.

Interventionmonths (Median)
Lenalidomide10.2

[back to top]

Duration of Tumor Control

The duration of tumor control was calculated as the time from the first response assessment demonstrating at least stable disease to the first documentation of progressive disease or death due to NHL. For participants without documentation of progression, the duration of response was censored at the last date of tumor assessment indicating no progression. Median was based on the Kaplan-Meier estimate. (NCT00179660)
Timeframe: From enrollment through study completion. Median duration on study was 3.7 months, with a maximum of 32.5 months.

Interventionmonths (Median)
Lenalidomide6.0

[back to top]

Percentage of Participants With Response

"Response was defined as participants with a complete response (CR), unconfirmed complete response (Cru) or partial response (PR), assessed using the International Workshop Lymphoma Response Criteria (IWLRC) and based on best responses as determined by the investigator.~CR: Complete disappearance of all detectable clinical and radiographic evidence of disease, disappearance of any disease-related symptoms, and normalization of biochemical abnormalities.~Cru: Criteria for CR above but with 1 or more of the following:~A residual lymph node mass > 1.5 cm in greatest transverse diameter that has regressed by more than 75% in the sum of the products of diameters (SPD)~Indeterminate bone marrow (increased number or size of aggregates without cytologic or architectural atypia).~PR: 50% decrease in SPD of the 6 largest dominant nodes or nodal masses. No increase in the size of other nodes, liver, or spleen. Splenic and hepatic nodules must regress by at least 50% in the SPD." (NCT00179660)
Timeframe: From enrollment through study completion. Median duration on study was 3.7 months, with a maximum of 32.5 months.

Interventionpercentage of participants (Number)
Lenalidomide34.7

[back to top]

Percentage of Participants With Tumor Control

"Tumor control was defined as participants with a complete response, unconfirmed complete response, partial response or stable disease (SD), assessed using the International Workshop Lymphoma Response Criteria (IWLRC) and based on best responses as determined by the investigator.~SD was defined as a response less than a PR (see above) but not Progressive Disease (PD).~PD was defined as~≥ 50 % increase from nadir in the SPD of any previously identified abnormal node for partial responders or non-responders.~Appearance of any new lesion during or at the end of therapy." (NCT00179660)
Timeframe: From enrollment through study completion. Median duration on study was 3.7 months, with a maximum of 32.5 months.

Interventionpercentage of participants (Number)
Lenalidomide59.2

[back to top]

Progression-free Survival

"Progression-free survival was defined as the time from the start of study drug therapy to the first observation of disease progression or death due to any cause, whichever came first.~Participants who withdrew for any reason or received another NHL therapy including stem cell transplantation without documented progressive disease were censored on the date of their last adequate response assessment indicating no progression (or last adequate assessment prior to receiving other NHL therapy). Participants who were still active without progressive disease at the time of the data cut-off date were censored on the date of their last adequate response assessment." (NCT00179660)
Timeframe: From enrollment through study completion. Median duration on study was 3.7 months, with a maximum of 32.5 months.

Interventionmonths (Median)
Lenalidomide3.6

[back to top]

Number of Participants With Adverse Events (AEs)

"The Investigator determined the relationship between the administration of study drug and the occurrence of an AE as suspected if the temporal relationship of the adverse event to study drug administration made a causal relationship possible, and other drugs, therapeutic interventions, or underlying conditions did not provide a sufficient explanation for the observed event.~The Investigator graded the severity of AEs according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) criteria and the following scale:~Grade 1 = Mild~Grade 2 = Moderate~Grade 3 = Severe~Grade 4 = Life threatening~Grade 5 = Death~A Serious AE is defined as any AE which results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or constitutes an important medical event." (NCT00179660)
Timeframe: From the start of study drug through 30 days after the last dose of study drug. Maximum time on study drug was 15.2 months.

Interventionparticipants (Number)
Any adverse eventAdverse event related to study drugGrade 3-5 adverse eventGrade 3-5 adverse event related to study drugSerious adverse eventSerious adverse event related to study drugAE leading to discontinuation of study drugRelated AE leading to study drug discontinuationAE leading to dose reduction or interruption
Lenalidomide494236272169428

[back to top]

Percentage of Participants With Response

"Response was defined as participants with a complete response (CR), unconfirmed complete response (Cru) or partial response (PR), assessed using the International Workshop Lymphoma Response Criteria (IWLRC) and based on best responses as determined by the investigator. CR: Complete disappearance of all detectable clinical and radiographic evidence of disease, disappearance of any disease-related symptoms, and normalization of biochemical abnormalities.~Cru: Criteria for CR above but with 1 or more of the following:~A residual lymph node mass > 1.5 cm in greatest transverse diameter that has regressed by more than 75% in the sum of the products of diameters (SPD)~Indeterminate bone marrow (increased number or size of aggregates without cytologic or architectural atypia).~PR: ≥ 50% decrease in SPD of the 6 largest dominant nodes or nodal masses. No increase in the size of other nodes, liver, or spleen. Splenic and hepatic nodules must regress by at least 50% in the SPD." (NCT00179673)
Timeframe: From enrollment through study completion. Median duration on study was 4.4 months with a maximum of 32 months

Interventionpercentage of participants (Number)
Lenalidomide23.3

[back to top]

Percentage of Participants With Tumor Control

"Tumor control was defined as participants with a complete response, unconfirmed complete response, partial response or stable disease (SD), assessed using the International Workshop Lymphoma Response Criteria (IWLRC) and based on best responses as determined by the investigator.~SD was defined as a response less than a PR (see above) but not Progressive Disease (PD).~PD was defined as~≥ 50 % increase from nadir in the SPD of any previously identified abnormal node for partial responders or non-responders.~Appearance of any new lesion during or at the end of therapy." (NCT00179673)
Timeframe: From enrollment through study completion. Median duration on study was 4.4 months with a maximum of 32 months

Interventionpercentage of participants (Number)
Lenalidomide60.5

[back to top]

Progression Free Survival (PFS)

Progression-free survival was defined as the time from the start of study drug therapy to the first observation of disease progression or death due to any cause, whichever came first. Participants who withdrew for any reason or received another NHL therapy including stem cell transplantation without documented progressive disease were censored on the date of their last adequate response assessment indicating no progression (or last adequate assessment prior to receiving other NHL therapy). Participants who were still active without progressive disease at the time of the data cut-off date were censored on the date of their last adequate response assessment. (NCT00179673)
Timeframe: From enrollment through study completion. Median duration on study was 4.4 months with a maximum of 32 months

Interventionmonths (Median)
Lenalidomide4.4

[back to top]

Number of Participants With Adverse Events (AEs)

"The Investigator determined the relationship between the administration of study drug and the occurrence of an AE as suspected if the temporal relationship of the adverse event to study drug administration made a causal relationship possible, and other drugs, therapeutic interventions, or underlying conditions did not provide a sufficient explanation for the observed event.~The Investigator graded the severity of AEs according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) criteria and the following scale:~Grade 1 = Mild~Grade 2 = Moderate~Grade 3 = Severe~Grade 4 = Life threatening~Grade 5 = Death~A Serious AE is defined as any AE which results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or constitutes an important medical event." (NCT00179673)
Timeframe: From the start of study drug through 30 days after the last dose of study drug. Maximum time on study drug was 13.8 months.

Interventionparticipants (Number)
At least one Adverse Event (AE)≥ 1 AE related to study drugGrade (GR) 3-5 AEGrade 3-5 AE related to study drugSerious adverse event (SAE)SAE related to study drugAE leading to discontinuation of study drugRelated AE leading to study drug discontinuationAE leading to dose reduction or interruption
Lenalidomide4237272418109527

[back to top]

The Duration of Response

The duration of response was calculated as the first response assessment demonstrating evidence of at least a partial response to the first documentation of progressive disease (as determined by computed tomography scan) or death due to NHL, whichever occurred first. For participants without documentation of progression, the duration of response was censored at the last date of tumor assessment indicating no progression. Median was based on the Kaplan-Meier estimate. (NCT00179673)
Timeframe: From enrollment through study completion. Median duration on study was 4.4 months with a maximum of 32 months

Interventionmonths (Median)
LenalidomideNA

[back to top]

Overall Response Rate

"Response was evaluated for Anemia and Spleen:~Major anemia response: hemoglobin increase to within normal limits in the absence of transfusion. Minor anemia response: hemoglobin improvement of at least 2 grams per deciliter independent of transfusion support, or achievement of transfusion independence in transfusion-dependent patients. Major spleen response: normalization of spleen size to the range of 12-14 centimeters by ultrasound. Minor spleen response: a 50% or more decrease in excess spleen size by ultrasound. Complete remission (CR): complete resolution of disease-related symptoms, splenomegaly, normalization of peripheral blood count, white cell differential and smear, and normalization of bone marrow histology. Partial remission (PR): a major or minor response in anemia or splenomegaly. Overall Response (OR)=CR + PR, assessed among eligible, treated patients." (NCT00227591)
Timeframe: Assessed at the end of cycle 3

InterventionProportion of participants (Number)
Lenalidomide0.26

[back to top]

Time to Progression

Time to progression (TTP) is defined as the time from study entry until progression or death without progression. The median TTP with 95% CI was estimated using the Kaplan-Meier method. (NCT00238238)
Timeframe: Up to 10 years

Interventionyears (Median)
Arm II - Lenalidomide1.1
Arm III - Lenalidomide and Rituximab2

[back to top]

Overall Response Rate

Response is assessed by investigator according to International Working Group (IWG) criteria. A complete response requires disappearance of all evidence of disease. A partial response is a >/= 50% decrease in the sum of products of 6 largest dominant nodes or nodal masses as well as for splenic and hepatic nodules. No increase in size of nodes, liver or spleen and no new sites of disease. (NCT00238238)
Timeframe: Duration of treatment (12 cycles)

Interventionpercentage of participants (Number)
Arm II - Lenalidomide53.3
Arm III - Lenalidomide and Rituximab76.1

[back to top]

Number of Patients in Overall Response Categories

Overall Response defined as participant had either complete response (CR) or partial response (PR) assessed after three cycles, at six months and yearly thereafter using the NCI-Working Group Criteria: Complete Response, Complete Response with Nodules, Partial Response, or No Response. (NCT00267059)
Timeframe: Evaluated after three 28-day cycles of lenalidomide.

InterventionParticipants (Number)
Complete ResponseComplete Response With NodulesPartial ResponseNo Response
Lenalidomide311030

[back to top]

Response Rate

Percentage of patients who responded to treatment (either stable disease or complete response) based on total tumor volume measurements from CT scans. (NCT00287287)
Timeframe: 4 years

InterventionParticipants (Count of Participants)
Lenalidomide (Revlimid)17

[back to top]

Maximum Tolerated Dose (MTD) of Lenalidomide in Combination With Rituximab

MTD is defined as the highest dose level in which 1 or fewer participants experienced a dose limiting toxicity (DLT) in 6 participants treated. DLT is any grade III or IV toxicity during the first 28 days (first cycle) of therapy. (NCT00294632)
Timeframe: 28 days of cycle 1

Interventionmg (Number)
Phase I: Lenalidomide + Rituximab20

[back to top]

Objective Response Rate of Participants Treated With Lenalidomide 20 mg: Overall Response as % of Participants With Complete or Partial Response

Objective response rate defined as percentage of participants with complete or partial response after 2 cycles of therapy maintained for one month. Objective response monitored using Simon's optimal 2-stage design. (NCT00294632)
Timeframe: 56 days

Interventionpercentage of participants (Number)
Lenalidomide + Rituximab57

[back to top]

Plasma Concentration of Revilimid at Steady State

Mean plasma concentration (ng/mL) of Revilimid at steady state (Day 21 of second treatment cycle) (NCT00348595)
Timeframe: Day 21 of second treatment cycle

Interventionng/mL (Mean)
Revlimid 5mg/Day12.67
Revlimid 25mg/Day65.14

[back to top]

Safety, Feasibility and Tolerance of Revlimid as Assessed by Number of Participants Experiencing Grade 3 and 4 Adverse Events.

Number of participants experiencing Grade 3 and 4 adverse events as defined by the National Cancer Institute Common Toxicity Criteria version 3.0 (NCT00348595)
Timeframe: 6 months post-intervention

InterventionParticipants (Count of Participants)
Revlimid 5mg/Day3
Revlimid 25mg/Day10

[back to top]

Change in of PSA Slope

Mean change in PSA slope from baseline to 6 months. PSA slope was calculated using the regression of log PSA over 6 months in each patient. A negative mean change in PSA slope reflects a better outcome. (NCT00348595)
Timeframe: Change from baseline to 6 months post-intervention

Interventionlog PSA (Mean)
Revlimid 5mg/Day-0.033
Revlimid 25mg/Day-0.172

[back to top]

Number of Participants With Prostate-specific Antigen (PSA) Progression

Number of participants with greater than or equal to 25% increase in PSA at 6 months (NCT00348595)
Timeframe: 6 months post-intervention

InterventionParticipants (Count of Participants)
Revlimid 5mg/Day7
Revlimid 25mg/Day5

[back to top]

Time to Transformation to Acute Myeloid Leukemia or Death

Time (in months) patients took to evolve to myeloid leukemia or death after achieving a complete response using the RECIST criteria (NCT00352001)
Timeframe: After 7 months of treatment, until the date of first documented myeloid leukemia or death, whichever came first, assessed up to 55 months

Interventionmonths (Median)
Lenalidomide and Azacitidine13.6

[back to top]

Overall Survival Among Patients With Complete Response

Time (in months) patients who achieved a complete response using the RECIST criteria were alive on study (NCT00352001)
Timeframe: After 7 months of treatment, until the date of first documented myeloid leukemia or death, whichever came first, assessed up to 55 months

Interventionmonths (Median)
Lenalidomide and Azacitidine37

[back to top]

PHASE I: Maximum Tolerated Dose of Azacitidine

"Participants will be enrolled on the Phase I study portion in blocks of 3 to varying doses of Revlimid® (lenalidomide) and Vidaza® (azacitidine) (Table 1). To determine the MTD, a standard 3+3 design will be used. DLT will be assessed during the first cycle of therapy within each treatment group. No Maximum dose was reach but the go-forward dose agreed upon by the investigators is reported here." (NCT00352001)
Timeframe: After 1 courses (1 months)

Interventionmg/m2 subcutaneously for 5 days (Number)
Lenalidomide and Azacitidine75

[back to top]

PHASE II: Determine the Number of Patients With Responses for Efficacy(Measured as Response Rate)

"For the Phase II study portion, to determine the efficacy (measured as response rate) of the combination therapy as defined by the International Working Group (IWG) criteria (CR, complete remission; PR, partial remission; or HI, hematological improvement.~Complete response (CR) is defined as: Disappearance of the chromosomal abnormality without appearance of new ones.~Partial response (PR) is defined as: At least 50% reduction of the chromosomal abnormality.~Hematologic Improvement (HI) is defined as: red blood cell increase of >=1.5g/dL, a platelet response of >=30X10^9/L or by at least 100% for values starting <20X10^9/L, or a neutrophil response of at least 100% and absolute increase of >0.5X10^9/L" (NCT00352001)
Timeframe: After 4 courses (4 months)

Interventionparticipants (Number)
Lenalidomide and Azacitidine26

[back to top]

Time to Relapse After Achieving Complete Response

(NCT00352001)
Timeframe: After 7 months of treatment, until the date of first documented myeloid leukemia or death, whichever came first, assessed up to 55 months

Interventionmonths (Median)
Lenalidomide and Azacitidine17

[back to top]

PHASE II: Determine the Number of Patients With Responses for Efficacy(Measured as Response Rate)

"For the Phase II study portion, to determine the efficacy (measured as response rate) of the combination therapy as defined by the International Working Group (IWG) criteria (CR, complete remission; PR, partial remission; or HI, hematological improvement)~Complete response (CR) is defined as: Disappearance of the chromosomal abnormality without appearance of new ones.~Partial response (PR) is defined as: At least 50% reduction of the chromosomal abnormality.~Hematologic Improvement (HI) is defined as: red blood cell increase of >=1.5g/dL, a platelet response of >=30X10^9/L or by at least 100% for values starting <20X10^9/L, or a neutrophil response of at least 100% and absolute increase of >0.5X10^9/L" (NCT00352001)
Timeframe: After 7 courses (months)

Interventionparticipants (Number)
Lenalidomide and Azacitidine26

[back to top] [back to top]

PHASE I: Maximum Tolerated Dose of Lenalidomide

"Participants will be enrolled on the Phase I study portion in blocks of 3 to varying doses of Revlimid® (lenalidomide) and Vidaza® (azacitidine) (Table 1). To determine the MTD, a standard 3+3 design will be used. DLT will be assessed during the first cycle of therapy within each treatment group. No Maximum dose was reach but the go-forward dose agreed upon by the investigators is reported here." (NCT00352001)
Timeframe: After 1 courses (1 months)

Interventionmg orally for 21 days (Number)
Lenalidomide and Azacitidine10

[back to top] [back to top]

Total Response

Morphologic complete remission (CR): ANC >=1,000/mcl, platelet count >=100,000/mcl, <5% bone marrow blasts, no Auer rods, no evidence of extramedullary disease. Morphologic complete remission with incomplete blood count recovery (CRi): Same as CR but ANC may be <1,000/mcl and/or platelet count <100,000/mcl. Partial remission (PR): ANC >1,000/mcl, platelet count >100,000/mcl, and at least 50% decrease in the percentage of marrow aspirate blasts to 5-25%, or marrow blasts <5% with persistent Auer rods. (NCT00352365)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Induction Therapy14

[back to top]

Cytogenetic Abnormalities

Number of baseline cytogenetic abnormalities by responders (CR, CRi, and PR) and nonresponders. (NCT00352365)
Timeframe: Up to 5 years

InterventionNumber of abnormalities (Median)
Responders8
Nonresponders8

[back to top]

Complete Response

Morphologic complete remission (CR): ANC >=1,000/mcl, platelet count >=100,000/mcl, <5% bone marrow blasts, no Auer rods, no evidence of extramedullary disease. Morphologic complete remission with incomplete blood count recovery (CRi): Same as CR but ANC may be <1,000/mcl and/or platelet count <100,000/mcl. (NCT00352365)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Induction Therapy11

[back to top]

Number of Patients With Objective Response (Complete and Partial Response + Hematological Improvement)

Time to response defined as the time from start of therapy until the response criteria are fulfilled. Response duration defined as the time from response until relapse (progressive disease) or death. (NCT00352794)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Lenalidomide + Prednisone14

[back to top]

Number of Participants With a Response (Complete Remissions (CR), Complete Remissions With Incomplete Platelet Recovery [CRp] and Partial Responses)

Response for Acute Myeloid Leukemia (AML) according to 2003 International Working Group (IWG) criteria: CR required absolute neutrophil count (ANC) >1 * 10^9/L, platelet count ≥100 * 10^9/L, < 5% of blast cells in bone marrow. CRp: as above except platelet count <100 * 10^9/L. Partial remission: as CR except for presence of 5-25% marrow blasts and with a decrease of marrow blast at least 50%. Response for Myelodysplastic Syndrome (MDS) was defined based on the 2006 IWG criteria. All participants with MDS who achieved hematological CR, Partial Response (PR), marrow CR, and hematological improvement considered responders. (NCT00360672)
Timeframe: Following three 28-day cycles evaluated for response

Interventionparticipants (Number)
Complete RemissionComplete Remission Incomplete Platelet RecoveryPartial Remission
Revlimid110

[back to top]

Objective Response Rate of the Drug Combination in This Patient Populations.

Overall Response (OR) was defined as partial response (PR) or better. Response was assessed according to European Group for Blood and Marrow Transplant criteria, modified to include nCR and VGPR, from the International Uniform Response Criteria. (NCT00378105)
Timeframe: Full response assessment was conducted at the end of cycle 8 (average of168 days) and after cycle 4 (84 days) for patients proceeding to transplant.

Interventionpercentage of participants (Number)
Phase 1 Population100
Phase II Population100
Total100

[back to top]

Estimated 18-month Overall Survival Rate

Overall survival was measured from treatment initiation to death, censored at the date patients were last known to be alive for those who had not died. (NCT00378105)
Timeframe: Survival rate at 18 months

InterventionPercentage of participants (Number)
All Patients97

[back to top]

Percentage of Patients Who Remained in Response for More Than 18 Months

Duration of response was measured from first response to progression or death, censored at the date patients were last known to be alive and disease free for patients who had not progressed or died. (NCT00378105)
Timeframe: Response rate at 18 months

InterventionPercentage of participants (Number)
All Patients68

[back to top]

Estimated 18-month Progression Free Survival (PFS) Rate

"PD from European Bone Marrow Transplant (EBMT) Response Criteria Required one or more:~>25% increased in the level of serum monoclonal paraprotein, which must also be an absolute increase of at least 5 g/L and confirmed on a repeat investigation, or >25% increased in 24-hour urinary light chain excretion (must also be an absolute increase of at least 200 mg/24 h and confirmed on a repeat investigation), or >25% increased in plasma cells in a bone marrow aspirate or biopsy (must also be an absolute increase of at least 10%) Definite increase in the size of existing lytic bone lesions or soft tissue plasmacytomas.~Development of new bone lesions or soft tissue plasmacytomas (not including compression fracture).~Development of hypercalcemia (corrected serum calcium >11.5 mg/dL or 2.8 mmol/L not attributable to any other cause).~PFS was measured from treatment initiation to progression or death, censored at the date patients were last known to be alive and disease free" (NCT00378105)
Timeframe: PFS rate at 18 months

InterventionPercentage of participants (Number)
All Patients75

[back to top]

Overall Survival

defined as time from treatment initiation to death, or last known to be alive for those who had not died (NCT00378209)
Timeframe: assesed at a median follow-up of 44 months

Interventionmonth (Median)
Lenalidomide, Dexamethasone, Bortezomib Combination30

[back to top]

The Proportion of Patients Alive and Without Progressive Disease (PD) for ≥6 Months

"Response assessed by the European Group for Blood and Marrow Transplant (EBMT) criteria, modified to include nCR and VGPR from the international uniform response criteria (IMWG).~Progressive disease (PD) required one or more of the following:~>25% increased in serum monoclonal paraprotein (must also be an absolute increase of at least 5 g/L and confirmed on a repeat investigation) >25% increased in 24-hour urinary light chain excretion (must also be an absolute increase of at least 200 mg/24 h and confirmed on a repeat investigation) >25% increased in plasma cells in a bone marrow aspirate or on trephine biopsy (must also be an absolute increase of at least 10%) Definite increase in the size of existing lytic bone lesions or soft tissue plasmacytomas.~Development of new bone lesions or soft tissue plasmacytomas (not including compression fracture).~Development of hypercalcemia (corrected serum calcium >11.5 mg/dL or 2.8 mmol/L not attributable to any other cause)." (NCT00378209)
Timeframe: 6 months after therapy

Interventionpercentage of treated patients (Number)
Lenalidomide, Dexamethasone, Bortezomib Combination75

[back to top]

Duration of Response

Duration of response will be measured as the time from initiation of a response to first documentation of disease progression or death, or date last known progression-free and alive for those who have not progressed or died. (NCT00378209)
Timeframe: Assessed at a median follow-up of 44 months

Interventionmonths (Median)
Lenalidomide, Dexamethasone, Bortezomib Combination8.7

[back to top]

Objective Response Rate

"Response assessed by the European Group for Blood and Marrow Transplant (EBMT) criteria, modified to include nCR and VGPR from the international uniform response criteria (IMWG).~Objective response was defined by the achievement of at least Partial Response (PR) or better (CR-complete response, nCR-near complete response, and VGPR-very good partial response)." (NCT00378209)
Timeframe: Assessed every cycle for up to 8 cycles and best response was reported

Interventionpercentage of treated patients (Number)
Lenalidomide, Dexamethasone, Bortezomib Combination64

[back to top]

Progression Free Survival

Progression-free survival is defined as the time from registration to the disease progression or death from any cause, censored at date last known progression-free for those who have not progressed or died. (NCT00378209)
Timeframe: aassesed at a median follow-up of 44 months

Interventionmonths (Median)
Lenalidomide, Dexamethasone, Bortezomib Combination9.5

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Constipation Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the constipation scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=114,124,128)Cycle 7 - approximately Month 7 (n=96,111,112)Cycle 10 - approximately Month 10 (n=86,93,97)Cycle 13 - approximately Month 13 (n=73,73,81)Cycle 16 - approximately Month 16 (n=63,51,62)
MPp+p-4.9-2.7-1.7-3.3-2.2
MPR+p4.80.6-1.1-2.7-5.2
MPR+R-1.8-3.5-5.0-5.0-1.6

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Diarrhoea Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the diarrhea scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=115,125,124)Cycle 7 - approximately Month 7 (n=98,109,112)Cycle 10 - approximately Month 10 (n=87,92,95)Cycle 13 - approximately Month 13 (n=73,73,80)Cycle 16 - approximately Month 16 (n=63,52,61)
MPp+p3.20.9-0.00.80.5
MPR+p1.9-1.21.4-1.41.3
MPR+R2.33.41.15.510.6

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Dyspnoea Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the dyspnoea scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=117,126,126)Cycle 7 - approximately Month 7 (n=100,110,110)Cycle 10 - approximately Month 10 (n=86,93,96)Cycle 13 - approximately Month 13 (n=73,73,81)Cycle 16 - approximately Month 16 (n=62,53,62)
MPp+p-0.02.13.8-0.01.6
MPR+p-6.4-8.5-4.3-2.3-6.3
MPR+R-2.6-1.7-4.3-5.0-3.2

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Emotional Functioning Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better level of emotional functioning. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=115,125,128)Cycle 7 - approximately Month 7 (n=98,111,112)Cycle 10 - approximately Month 10 (n=86,92,97)Cycle 13 - approximately Month 13 (n=73,73,83)Cycle 16 - approximately Month 16 (n=63,52,63)
MPp+p6.85.04.76.66.9
MPR+p2.74.21.61.1-0.2
MPR+R4.88.89.08.29.9

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Congitive Functioning Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better level of cognitive functioning. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=115,125,128)Cycle 7 - approximately Month 7 (n=98,111,113)Cycle 10 - approximately Month 10 (n=87,92,97)Cycle 13 - approximately Month 13 (n=73,73,83)Cycle 16 - approximately Month 16 (n=63,52,63)
MPp+p1.30.7-2.7-1.4-4.0
MPR+p-2.00.1-4.4-3.0-3.5
MPR+R0.32.91.0-0.00.3

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Fatigue Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the fatigue scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=120,127,129)Cycle 7 - approximately Month 7 (n=100,112,110)Cycle 10 - approximately Month 10 (n=87,95,95)Cycle 13 - approximately Month 13 (n=74,74,82)Cycle 16 - approximately Month 16 (n=64,53,62)
MPp+p-5.1-5.7-6.9-7.5-4.1
MPR+p-5.5-9.5-7.5-10.7-9.7
MPR+R-3.0-7.6-7.5-7.1-10.0

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Financial Difficulties Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a problem scale like the financial problems scale = higher level of financial problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=111,123,125)Cycle 7 - approximately Month 7 (n=94,111,112)Cycle 10 - approximately Month 10 (n=84,92,97)Cycle 13 - approximately Month 13 (n=70,72,83)Cycle 16 - approximately Month 16 (n=61,52,63)
MPp+p-2.9-2.1-1.7-4.0-5.3
MPR+p-1.1-0.60.7-0.5-0.6
MPR+R2.42.16.04.81.6

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Social Functioning Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better level of social functioning. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=115,125,127)Cycle 7 - approximately Month 7 (n=98,111,112)Cycle 10 - approximately Month 10 (n=87,92,97)Cycle 13 - approximately Month 13 (n=72,73,83)Cycle 16 - approximately Month 16 (n=63,52,63)
MPp+p6.06.14.16.29.8
MPR+p0.34.44.57.56.1
MPR+R5.18.310.911.813.2

[back to top]

Summary of Participants With Treatment-Emergent Adverse Events (TEAE) During the Double-Blind Treatment Period

Data as of 11 May 2010 cutoff. Participant counts in different categories of TEAEs during the double-blind treatment period. A TEAE is as any AE occurring or worsening on or after the first treatment of any study drug, and within 30 days after the last dose of the last study drug. Severity grades according to Common Terminology Criteria for Adverse Events v3.0 (CTCAE) on a 1-5 scale: 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. Dose reduction includes reduction with or without interruption. (NCT00405756)
Timeframe: Up to 169 weeks (Double-blind therapy period plus 4 weeks)

,,
Interventionparticipants (Number)
>=1 adverse event (AE)>=1 CTCAE grade 3-4 AE>=1 CTCAE grade 5 AE>=1 serious AE (SAE)>=1 AE related to Lenaldomide/Placebo>=1 AE related to Melphalan>=1AE related to Prednisone>=1 Grade 3-4 AE related to Lenaldomide/Placebo>=1 Grade 3-4 AE related to Melphalan>=1 Grade 3-4 AE related to Prednisone>=1 Grade 5 AE related to Lenalidomide/Placebo>=1 Grade 5 AE related to Melphalan>=1 Grade 5 AE related to Prednisone>=1 SAE related to Lenalidomide/Placebo>=1 SAE related to Melphalan>=1 SAE related to Prednisone>=1 AE leading to Lenalidomide/Placebo withdrawal>=1 AE leading to Melphalan withdrawal>=1 AE leading to Prednisone withdrawal>=1 AE leading to Lenalidomide/Plac dose reduction>=1 AE leading to Melphalan dose reduction>=1 AE leading to Prednisone dose reduction>=1 AE leading to Lenalidomide/Plac dose interrupt>=1 AE leading to Melphalan dose interruption>=1 AE leading to Prednisone dose interruption
MPp+p15310775613112693686222231111151410102621551015
MPR+p15112966214513494117110292113224162419197058782139
MPR+R150137766148140871281183233138271926202071471592528

[back to top]

Number of Participants in Disease Response Categories Representing Their Best Response During the Double-blind Treatment Period

Data as of 11 May 2010 cutoff. Best response was determined by the Central Assessment Committee (CAC) based on the European Group for Blood and Marrow Transplantation (EBMT) criteria: Complete Response (CR)-absence of serum and urine monoclonal paraprotein for 6 weeks, plus no increase in size or number of bone lesions, plus other factors); Partial Response (PR)-not all CR criteria, plus >=50% reduction in serum monoclonal paraprotein plus others; Stable Disease (SD)- not PR or PD; Progressive Disease (PD)- reappearance of monoclonal paraprotein, bone lesions, other; Not Evaluable (NE). (NCT00405756)
Timeframe: Up to 165 weeks

,,
Interventionparticipants (Number)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Response not evaluable (NE)
MPp+p5727007
MPR+p5994027
MPR+R151022807

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) In Side Effects of Treatment Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale; higher score for the side effects scale = higher level of symptomatology. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=113,120,125)Cycle 7 - approximately Month 7 (n=95,108,111)Cycle 10 - approximately Month 10 (n=85,89,94)Cycle 13 - approximately Month 13 (n=72,72,81)Cycle 16 - approximately Month 16 (n=62,50,61)
MPp+p0.61.80.30.3-0.9
MPR+p0.1-1.70.0-1.0-2.9
MPR+R1.30.4-1.6-3.8-2.1

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) In Future Perspective Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale. For the future perspective scale, higher score = better perspective of the future. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=112,121,124)Cycle 7 - approximately Month 7 (n=93,108,112)Cycle 10 - approximately Month 10 (n=83,88,97)Cycle 13 - approximately Month 13 (n=71,73,81)Cycle 16 - approximately Month 16 (n=62,52,62)
MPp+p7.69.814.511.914.4
MPR+p4.37.76.66.37.7
MPR+R4.714.617.317.318.5

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) In Body Image Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale. For the body image scale, higher scores = better body image. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=110,117,119)Cycle 7 - approximately Month 7 (n=88,104,108)Cycle 10 - approximately Month 10 (n=79,83,94)Cycle 13 - approximately Month 13 (n=68,72,79)Cycle 16 - approximately Month 16 (n=59,52,61)
MPp+p4.55.23.95.12.7
MPR+p-0.32.6-4.0-0.56.4
MPR+R2.13.87.61.03.4

[back to top]

Kaplan Meier Estimates for Duration of Response as Determined by the Central Adjudication Committee (CAC)

"Data as of 11 May 2010 cutoff. Duration of myeloma response was defined as the time from the initial response date to the earlier of progressive disease (PD) as determined by the CAC or death on study. PD was based on the European Group for Blood and Marrow Transplantation/International Bone Marrow Transplant Registry/Autologous Bone Marrow Transplant Registry [EBMT/IBMTR/ABMTR] criteria.~PD criteria includes increasing monoclonal paraprotein levels, bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia." (NCT00405756)
Timeframe: Up to 149 weeks

Interventionweeks (Median)
MPR+R121.6
MPR+p56.1
MPp+p55.4

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Role Functioning Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score=better level of role functioning. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=119,127,130)Cycle 7 - approximately Month 7 (n=99,112,113)Cycle 10 - approximately Month 10 (n=86,95,95)Cycle 13 - approximately Month 13 (n=74,74,82)Cycle 16 - approximately Month 16 (n=64,53,63)
MPp+p7.46.95.65.77.1
MPR+p3.08.07.511.78.5
MPR+R1.85.79.39.712.2

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Physical Functioning Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score=better level of physical functioning. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=120,127,130)Cycle 7 - approximately Month 7 (n=100,112,112)Cycle 10 - approximately Month 10 (n=88,95,96)Cycle 13 - approximately Month 13 (n=75,74,83)Cycle 16 - approximately Month 16 (n=64,53,63)
MPp+p4.52.75.13.31.1
MPR+p3.38.18.59.77.6
MPR+R1.98.28.98.610.0

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13, 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Disease Symptoms Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. EORTC QLQ-MY20 includes four scales: disease symptoms, treatment side-effects, future perspective, and body image. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale; higher score for the disease symptoms scale = higher level of symptomatology. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=113,121,127)Cycle 7 - approximately Month 7 (n=96,109,112)Cycle 10 - approximately Month 10 (n=85,91,95)Cycle 13 - approximately Month 13 (n=72,73,82)Cycle 16 - approximately Month 16 (n=62,51,62)
MPp+p-5.4-6.0-5.4-6.3-3.3
MPR+p-8.7-9.7-7.1-8.8-5.9
MPR+R-8.9-9.0-7.9-7.2-10.5

[back to top]

Kaplan Meier Estimates of Progression-free Survival (PFS) From Start of Maintenance Therapy Period Based on the Response Assessment by the Central Adjudication Committee (CAC)

"Data as of 11 May 2010 cutoff. PFS calculated from the start of the Maintenance period to the earlier of the first documentation of progressive disease (PD) as determined by the CAC, or death on study due to any cause.~PD was based on the European Group for Blood and Marrow Transplantation/International Bone Marrow Transplant Registry/Autologous Bone Marrow Transplant Registry [EBMT/IBMTR/ABMTR] criteria.~PD criteria includes increasing monoclonal paraprotein levels, bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia." (NCT00405756)
Timeframe: Approximately week 37 (start of cycle 10) to week 165

Interventionweeks (Median)
MPR+R112.0
MPR+p32.3

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Pain Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the pain scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=120,127,129)Cycle 7 - approximately Month 7 (n=100,112,113)Cycle 10 - approximately Month 10 (n=88,95,97)Cycle 13 - approximately Month 13 (n=74,74,83)Cycle 16 - approximately Month 16 (n=64,53,63)
MPp+p-13.4-11.5-9.8-12.1-12.2
MPR+p-13.8-16.5-15.6-14.9-11.0
MPR+R-14.4-17.8-17.2-13.7-20.3

[back to top] [back to top]

Kaplan Meier Estimates of Overall Survival (OS)

Data as of 11 May 2010 cutoff. Overall survival (OS) was defined as the time between randomization and death. Participants who died, regardless of the cause of death, were considered to have had an event. Participants who were lost to follow-up prior to the end of the trial, or who were withdrawn from the trial, were censored at the time of last contact. Participants who were still being treated were censored at the last available date available, or clinical cut-off date, if it was earlier. (NCT00405756)
Timeframe: up to 177 weeks

Interventionweeks (Median)
MPR+RNA
MPR+pNA
MPp+pNA

[back to top]

Kaplan Meier Estimates of Time to Progression (TTP) Based on the Response Assessment by the Central Adjudication Committee (CAC)

"Data as of 11 May 2010 cutoff. TTP was the time between randomization and disease progression as determined by the CAC. PD was based on the European Group for Blood and Marrow Transplantation/International Bone Marrow Transplant Registry/Autologous Bone Marrow Transplant Registry [EBMT/IBMTR/ABMTR] criteria.~PD criteria includes increasing monoclonal paraprotein levels, bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia." (NCT00405756)
Timeframe: up to 165 weeks

Interventionweeks (Median)
MPR+R148.1
MPR+p62.7
MPp+p61.3

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Global Quality of Life Scale

Data as of 11 May 2010 cutoff. EORTC QLC-C30 is a 30-item questionnaire to assess the quality of life in cancer patients. EORTC QLQ-C30 includes functional scales (physical, role, cognitive, emotional, social), global health status, symptom scales (fatigue, pain, nausea/vomiting), and other (dyspnoea, appetite loss, insomnia, constipation/diarrhea, financial difficulties). Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); two used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better quality of life. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=114,121,125)Cycle 7 - approximately Month 7 (n=96,108,110)Cycle 10 - approximately Month 10 (n=84,86,96)Cycle 13 - approximately Month 13 (n=70,70,82)Cycle 16 - approximately Month 16 (n=61,50,62)
MPp+p6.14.26.25.48.1
MPR+p5.68.18.88.87.2
MPR+R2.38.012.47.610.7

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Appetite Loss Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the appetite loss scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=119,125,130)Cycle 7 - approximately Month 7 (n=99,111,111)Cycle 10 - approximately Month 10 (n=87,93,96)Cycle 13 - approximately Month 13 (n=75,72,83)Cycle 16 - approximately Month 16 (n=64,52,63)
MPp+p-5.6-5.7-8.0-4.8-6.4
MPR+p1.9-5.7-5.4-8.8-16.0
MPR+R1.7-3.7-5.0-6.2-7.8

[back to top]

Kaplan Meier Estimates of Progression-free Survival (PFS) Based on the Response Assessment by the Central Adjudication Committee (CAC)

"Data as of 11 May 2010 cutoff. PFS was calculated as the time from randomization to the earlier of the first documentation of progressive disease (PD) as determined by the CAC, or death on study due to any cause. PD was based on the European Group for Blood and Marrow Transplantation/International Bone Marrow Transplant Registry/Autologous Bone Marrow Transplant Registry [EBMT/IBMTR/ABMTR] criteria.~PD criteria includes increasing monoclonal paraprotein levels, bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia." (NCT00405756)
Timeframe: up to 165 weeks

Interventionweeks (Median)
MPR+R136.1
MPR+p62.1
MPp+p56.1

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Nausea and Vomiting Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the nausea/vomiting scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=120,127,130)Cycle 7 - approximately Month 7 (n=99,112,112)Cycle 10 - approximately Month 10 (n=87,95,97)Cycle 13 - approximately Month 13 (n=75,72,83)Cycle 16 - approximately Month 16 (n=64,52,62)
MPp+p-0.00.70.3-0.4-1.3
MPR+p-1.3-0.7-1.4-3.0-4.2
MPR+R3.30.51.90.71.0

[back to top]

Time to First Response

Data as of 11 May 2010 cutoff. Time to first response was defined as the time from start of treatment until first response as assessed by the Central Assessment Committee (CMC) based on European Group for Blood and Marrow Transplantation (EBMT) criteria. (NCT00405756)
Timeframe: Up to 66 weeks

Interventionweeks (Mean)
MPR+R10.0
MPR+p9.3
MPp+p16.2

[back to top]

Change From Baseline to Cycles 4, 7, 10, 13 and 16 in European Organization for Research and Treatment of Cancer Questionnaire for Patients With Cancer (EORTC QLQ-C30) Insomnia Scale

Data as of 11 May 2010 cutoff. EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the insomnia scale = higher level of symptomatology/problems. (NCT00405756)
Timeframe: Baseline (Day 0), Months 4, 7, 10, 13, 16

,,
Interventionunits on a scale (Mean)
Cycle 4 - approximately Month 4 (n=118,124,128)Cycle 7 - approximately Month 7 (n=100,109,111)Cycle 10 - approximately Month 10 (n=87,94,96)Cycle 13 - approximately Month 13 (n=75,73,83)Cycle 16 - approximately Month 16 (n=64,53,63)
MPp+p-5.0-5.7-1.7-6.8-3.7
MPR+p-1.6-6.4-2.50.9-0.6
MPR+R2.0-1.0-5.0-4.9-4.7

[back to top]

Confirmed Anti-tumor Response Rate (Complete Response and Partial Response) to the Combination of Bevacizumab and Lenalidomide

Patient response to the treatment were determined by the definitions for complete response, partial response, marginal response, stable disease, and progressive disease outlined by IMBTR/ABMTR (Blade criteria). Responses were analyzed by descriptive statistics and summarized in tabular format (frequency tables). Furthermore, two-sided 95% confidence intervals for the proportions of subjects with a confirmed anti-tumor response were computed using the method proposed by Chang, which takes into account the multiplicity problem associated with the two-stage testing procedure. The objective response rate was estimated by using Whitehead's bias-adjustment approach. (NCT00410605)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Bevacizumab, Dexamethasone, and Lenalidomide64

[back to top]

Effect of Bev/Rev on Markers of Myeloma Activity in Myeloma Cells and Stromal Cells at Baseline

A Wilconxon signed-rank test conducted to determine if biomarker levels differed between baseline levels and those after three full cycles of treatment for all patients. (NCT00410605)
Timeframe: Baseline

Interventionmg per liter (Mean)
IL-6 level rangeMIP-1 level rangeVEGF level rangeVEGFR1 level rangeVEGFR2 level range
Bevacizumab, Dexamethasone, and Lenalidomide17.20.0390.0840.128.85

[back to top]

Toxicity and Tolerability of the Bevacizumab and Lenalidomide Combination

Adverse events/toxicities were collected during regular clinical visits. Confidence intervals for the estimate of the true number of patients suffereing from grade 3 or 4 toxicities per common terminology criteria were calculated using the Wilson interval. Ninety-five percent confidence intervals for the proportions of patients with complications (grade 3 or higher toxicities) were constructed. (NCT00410605)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Bevacizumab, Dexamethasone, and Lenalidomide72

[back to top]

Progression Free Survival (Time to Progression)

Patient response to the treatment were determined by the definitions for complete response, partial response, marginal response, stable disease, and progressive disease outlined by IMBTR/ABMTR (Blade criteria). Progression free survival was summarized using point estimates of the median time to progression and associated 95% confidence intervals. The data was presented graphically using Kaplan-Meier plots. Exploratory analysis, including multivariate Cox regression with demographic variables and markers of myeloma activity as covariates was performed. (NCT00410605)
Timeframe: up to five years

Interventionmonths (Median)
Bevacizumab, Dexamethasone, and Lenalidomide9

[back to top]

Effect of Bev/Rev on Markers of Myeloma Activity in Myeloma Cells and Stromal Cells at 3 Months Post-baseline

A Wilconxon signed-rank test conducted to determine if biomarker levels differed between baseline levels and those after three full cycles of treatment for all patients. (NCT00410605)
Timeframe: Up to Course 4 Day 1 (3 Months Post-baseline)

Interventionmg per liter (Mean)
IL-6 level rangeMIP-1 level rangeVEGF level rangeVEGFR1 level rangeVEGFR2 level range
Bevacizumab, Dexamethasone, and Lenalidomide5.50.0470.0540.128.17

[back to top]

Progression-free Survival as Determined by Central Review

"Kaplan-Meier estimate of progression-free survival is defined as start of study drug therapy to the first observation of progressive disease or death due to any cause, whichever comes first.~Response assessed according to Cheson, Journal of Clinical Oncology, 1999. Full definition of progressive disease, refer to Cheson article.~Progressive Disease(PD): Appearance of new lesion during/end of therapy; >=50% increase from lowest measurement in SPD." (NCT00413036)
Timeframe: Up to 1459 days

InterventionMonths (Median)
Lenalidomide4.5

[back to top]

Duration of Response as Determined by Central Review

"Kaplan-Meier estimates for the duration of response were calculated for responders and defined as the time from at least a partial response (PR) to progression of disease (PD) or death due to Non-Hodgkin's lymphoma.~For response assessment criteria (per Cheson, 1999) see the primary outcome measure in this results posting." (NCT00413036)
Timeframe: Up to 1459 days

InterventionMonths (Median)
Lenalidomide18.4

[back to top]

Participants Categorized by Best Response as Determined by Central Review

"Response assessed according to Cheson, Journal of Clinical Oncology, 1999. Full definitions, refer to Cheson article.~Complete Response(CR): Complete disappearance of all detectable disease and disease-related symptoms if present before therapy; normalization of lab abnormalities assignable to NHL. If bone marrow involved before treatment, must be cleared on repeat biopsy.~Complete Response Unconfirmed(CRu): CR, with one of the following: 1)residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters(SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2)indeterminate bone marrow.~Partial Response(PR): >50% decrease in 6 largest nodes or nodal masses. Nodes selected according to Cheson.~Stable Disease(SD): Less than PR, but not progressive disease.~Progressive Disease(PD): Appearance of new lesion during/end of therapy; >=50% increase from lowest measurement in SPD." (NCT00413036)
Timeframe: Up to 1459 days

InterventionParticipants (Number)
Complete Response (CR)Complete Response Unconfirmed (CRu)Partial Response (PR)Stable Disease (SD)Progressive Disease
Lenalidomide721407178

[back to top]

Time to Progression as Determined by Central Review

"Kaplan-Meier estimate of time-to-progression is calculated as time from the start of study drug therapy to the first observation of disease progression.~Response assessed according to Cheson, Journal of Clinical Oncology, 1999. Full definition of progressive disease, refer to Cheson article.~Progressive Disease(PD): Appearance of new lesion during/end of therapy; >=50% increase from lowest measurement in SPD." (NCT00413036)
Timeframe: Up to 1459 days

InterventionMonths (Median)
Lenalidomide4.5

[back to top]

Time to First Venous Thromboembolic Treatment-Emergent Adverse Event (TEAE)

Time between first dose and when a TEAE for venous thromboembolic event was reported. The mean is the univariate mean without adjusting for censoring. The treatment duration was used for censored participants. (NCT00420849)
Timeframe: up to 124 weeks

Interventionweeks (Mean)
Lenalidomide Plus Dexamethasone26.5

[back to top]

Time to First Peripheral Neuropathy Treatment-Emergent Adverse Event (TEAE)

Time between first dose and when a TEAE for peripheral neuropathy was reported. The mean is the univariate mean without adjusting for censoring. The treatment duration was used for censored participants. (NCT00420849)
Timeframe: up to 124 weeks

Interventionweeks (Mean)
Lenalidomide Plus Dexamethasone25.6

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Side Effects Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale; higher score for the side effects scale = higher level of symptomatology. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia4.9
Lenalidomide - Subpopulation From UK + Ireland4.7
Lenalidomide - Subpopulation From Spain2.0

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Future Perspective Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale. For the future perspective scale, higher score = better perspective of the future. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia5.8
Lenalidomide - Subpopulation From UK + Ireland3.4
Lenalidomide - Subpopulation From Spain4.4

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Disease Symptoms Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. EORTC QLQ-MY20 includes four scales: disease symptoms, treatment side-effects, future perspective, and body image. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale; higher score for the disease symptoms scale = higher level of symptomatology. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-2.4
Lenalidomide - Subpopulation From UK + Ireland-1.2
Lenalidomide - Subpopulation From Spain-3.9

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Body Image Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. Questions used 4-point scale (1 'Not at All' to 4 'Very Much'). Scores are averaged, and transformed to 0-100 scale. For the body image scale, higher scores = better body image. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-4.3
Lenalidomide - Subpopulation From UK + Ireland-2.0
Lenalidomide - Subpopulation From Spain-5.3

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Social Functioning Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better level of social functioning. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-5.2
Lenalidomide - Subpopulation From UK + Ireland-5.3
Lenalidomide - Subpopulation From Spain-3.1

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Role Functioning Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score=better level of role functioning. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-2.4
Lenalidomide - Subpopulation From UK + Ireland-1.5
Lenalidomide - Subpopulation From Spain-2.6

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Physical Functioning Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. EORTC QLQ-C30 includes functional scales (physical, role, cognitive, emotional, and social), global health status, symptom scales (fatigue, pain, nausea/vomiting), and other (dyspnoea, appetite loss, insomnia, constipation/diarrhea, and financial difficulties). Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score=better level of physical functioning. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-3.6
Lenalidomide - Subpopulation From UK + Ireland-2.9
Lenalidomide - Subpopulation From Spain-1.8

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Pain Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the pain scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-3.9
Lenalidomide - Subpopulation From UK + Ireland-5.2
Lenalidomide - Subpopulation From Spain-6.6

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Nausea/Vomiting Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the nausea/vomiting scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia1.0
Lenalidomide - Subpopulation From UK + Ireland0.1
Lenalidomide - Subpopulation From Spain-2.2

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Insomnia Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the insomnia scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-3.8
Lenalidomide - Subpopulation From UK + Ireland2.2
Lenalidomide - Subpopulation From Spain-1.8

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Global Quality of Life Scale

EORTQ QLC-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better quality of life. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia1.1
Lenalidomide - Subpopulation From UK + Ireland-1.8
Lenalidomide - Subpopulation From Spain-2.2

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Fatigue Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the fatigue scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia2.6
Lenalidomide - Subpopulation From UK + Ireland5.3
Lenalidomide - Subpopulation From Spain1.0

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Emotional Functioning Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better level of emotional functioning. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-0.2
Lenalidomide - Subpopulation From UK + Ireland-4.0
Lenalidomide - Subpopulation From Spain-0.7

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Dyspnoea Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the dyspnoea scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia2.7
Lenalidomide - Subpopulation From UK + Ireland2.7
Lenalidomide - Subpopulation From Spain-0.9

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Diarrhea Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the diarrhea scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia8.6
Lenalidomide - Subpopulation From UK + Ireland8.1
Lenalidomide - Subpopulation From Spain9.0

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Constipation Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the constipation scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia1.1
Lenalidomide - Subpopulation From UK + Ireland7.9
Lenalidomide - Subpopulation From Spain2.6

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Cognitive Functioning Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score = better level of cognitive functioning. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia-1.9
Lenalidomide - Subpopulation From UK + Ireland-4.9
Lenalidomide - Subpopulation From Spain3.5

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Appetite Loss Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a symptom scale like the appetite loss scale = higher level of symptomatology/problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia0.4
Lenalidomide - Subpopulation From UK + Ireland3.3
Lenalidomide - Subpopulation From Spain-4.5

[back to top]

Change From Baseline to Week 24 in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Financial Problems Scale

EORTC QLQ-C30 is a 30-item questionnaire to assess the overall quality of life in cancer patients. Most questions used 4-point scale (1 'Not at All' to 4 'Very Much'); 2 questions used 7-point scale (1 'Very Poor' to 7 'Excellent'). Scores are averaged, and transformed to 0-100 scale; higher score for a problem scale like the financial problems scale = higher level of financial problems. (NCT00420849)
Timeframe: Baseline (Day 0), Week 24

Interventionunits on a scale (Mean)
Lenalidomide - Subpopulation From Austria + Australia2.0
Lenalidomide - Subpopulation From UK + Ireland0.8
Lenalidomide - Subpopulation From Spain0.9

[back to top]

Participants With Adverse Events of Special Interest: Venous Thromboembolic Events

Number of participants with at least one venous thromboembolic treatment-emergent adverse event (TEAE), and number of participants reporting AEs coded to preferred terms that comprise the search terms for venous thromboembolic events in MedDRA version 9.0 are listed. A participant with multiple occurrences of a TEAE was counted only once for that category. (NCT00420849)
Timeframe: up to 124 weeks

Interventionparticipants (Number)
At least one venous thromboembolic eventDeep vein thrombosisPulmonary embolismThrombophlebitisVenous thrombosis limb
Lenalidomide Plus Dexamethasone60382371

[back to top]

Participants With Adverse Events of Special Interest: Peripheral Neuropathy

Number of participants with at least one peripheral neuropathy treatment-emergent adverse event (TEAE), and number of participants reporting AEs coded to preferred terms that comprise the search terms for peripheral neuropathy in MedDRA version 9.0 are listed. A participant with multiple occurrences of a TEAE was counted only once for that category. (NCT00420849)
Timeframe: up to 124 weeks

Interventionparticipants (Number)
At least one TEAE of peripheral neuropathyNeuropathy peripheralPeripheral sensory neuropathyNeuralgiaPeripheral motor neuropathyPolyneuropathySensory disturbance
Lenalidomide Plus Dexamethasone8446335221

[back to top]

Overall Incidence of Treatment-emergent Adverse Events (TEAEs), by Severity, Seriousness, and Relationship to Treatment

"Counts of study participants who had treatment-emergent adverse events (TEAEs) defined as any reported AE that started on or after the first day of study drug dosing. A participant with multiple occurrences of an adverse event within a category is counted only once in that category.~National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE version 3.0) was used by investigators to assess TEAEs. Severity scale ranges from 0 (none) to 5 (death). Grade 3=severe AE; Grade 4=life threatening or disabling AE; Grade 5=death." (NCT00420849)
Timeframe: up to 123 weeks

Interventionparticipants (Number)
At least one treatment-emergent AE (TEAE)At least one TEAE related to study drugAt least one TEAE with severity grade of 3 or 4At least one serious AE (SAE)
Lenalidomide Plus Dexamethasone586519471340

[back to top]

Kaplan-Meier Estimate of Duration of Response

Duration of response was calculated for responders and defined as the time from the first observation of a response (e.g., the first time that the appropriate decrease in M-protein level was observed for confirmed responders) to the first documented progression or relapse. Response duration was censored at the last adequate assessment showing evidence of no progression. (NCT00424047)
Timeframe: Up to data cut off of 03 August 2005; up to 24 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone67.6
Placebo Plus Dexamethasone33.3

[back to top] [back to top]

Kaplan-Meier Estimate of Time to Tumor Progression (TTP) (Later Cut-off Date of 02 Mar 2008)

Time to progression was calculated as the time from randomization to the first occurrence of disease progression, as determined by a detailed review of all the myeloma response assessment data using the Bladé criteria (Bladé, 1998). Disease progression was also based on bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia. (NCT00424047)
Timeframe: From randomization up to cut-off date of 02 March 2008; up to 51 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone52.4
Placebo Plus Dexamethasone20.1

[back to top]

Kaplan-Meier Estimate of Overall Survival (OS) (Later Cut-off Date of 02 March 2008)

OS was calculated as the time from randomization to death from any cause. OS was censored at the last date that the participant was known to be alive for participants who were alive at the time of analysis and for participants who were lost to follow-up before death was documented. (NCT00424047)
Timeframe: Randomization to data cut off of 02 March 2008; up to 51 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone161.9
Placebo Plus Dexamethasone133.3

[back to top]

Kaplan-Meier Estimate of Duration of Response (Cut-off at a Later Date of 03 March 2008)

Duration of response was calculated for responders and defined as the time from the first observation of a response (e.g., the first time that the appropriate decrease in M-protein level was observed for confirmed responders) to the first documented progression or relapse. Response duration was censored at the last adequate assessment showing evidence of no progression. (NCT00424047)
Timeframe: Up to data cut off of 03 Mar 2008; up to 51 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone68.1
Placebo Plus Dexamethasone33.3

[back to top]

Kaplan-Meier Estimate of Time to Tumor Progression (TTP)

Time to progression was calculated as the time from randomization to the first occurrence of disease progression, as determined by a detailed review of all the myeloma response assessment data using the Bladé criteria (Bladé, 1998). Disease progression was also based on bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia. (NCT00424047)
Timeframe: From randomization up to cut-off date of 03 August 2005; up to 24 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone52.1
Placebo Plus Dexamethasone20.1

[back to top]

Number of Participants With Adverse Events (AE)

"An AE is any sign, symptom, illness, or diagnosis that appears or worsens during the course of the study. Treatment-emergent AEs (TEAEs) are any AE occurring or worsening on or after the first treatment of the study drug and within 30 days after the last cycle end date of study drug. A serious AE = any AE which results in death; is life-threatening; requires or prolongs existing inpatient hospitalization; results in persistent or significant disability is a congenital anomaly/birth defect; constitutes an important medical event.~The severity of AEs were graded according to the National Cancer Institute (NCI) Common Terminology Criteria for AEs (CTCAE, Version 2.0): Grade 1 = Mild (no limitation in activity or intervention required); Grade 2 = Moderate (some limitation in activity; no/minimal medical intervention required); Grade 3 = Severe (marked limitation in activity; medical intervention required, hospitalization possible); Grade 4 = Life-threatening; Grade 5 = Death." (NCT00424047)
Timeframe: From first dose of study drug through to 30 days after the last dose, until the data cut-off date of 25 June 2013; up to 90 months

,
Interventionparticipants (Number)
≥ 1 Adverse Event≥ 1 Serious Adverse Event≥ 1 AE leading to study drug discontinuation≥ 1 AE leading to dose reduction or interruption≥ 1 Drug-Related Adverse Event≥ 1 Drug-Related Serious Adverse Event≥Death within ≤ 30 days of last dose of study drug≥ 1 Grade 1 or Higher Adverse Event≥ 1 Grade 2 or Higher Adverse Event≥ 1 Grade 3 or Higher Adverse Event≥ 1 Grade 4 or Higher Adverse Event
Lenalidomide Plus Dexamethasone17610546137160541717616814652
Placebo Plus Dexamethasone1757931100151302017516711937

[back to top]

Kaplan-Meier Estimate of Overall Survival (OS)

OS was calculated as the time from randomization to death from any cause. OS was censored at the last date that the participant was known to be alive for participants who were alive at the time of analysis and for participants who were lost to follow-up before death was documented. (NCT00424047)
Timeframe: Randomization to data cut off of 03 August 2005; up to 24 months

Interventionweeks (Median)
Lenalidomide Plus DexamethasoneNA
Placebo Plus DexamethasoneNA

[back to top]

Time to First Worsening on the Eastern Cooperative Oncology Group (ECOG) Performance Scale

The time to first worsening of the ECOG performance status was calculated as the time from randomization to the date of the first worsening compared with the last ECOG evaluation obtained prior to randomization. Data were censored at the last date that the participant was known to be unchanged or improved from before randomization for the participants who had not had worsened at the time of the analysis and for the patients who were lost to follow-up before worsening in the ECOG performance status was documented. (NCT00424047)
Timeframe: Randomization to cut off date of 03 August 2005; up to 24 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone10.1
Placebo Plus Dexamethasone12.3

[back to top]

Time to First Worsening on the Eastern Cooperative Oncology Group (ECOG) Performance Scale (Later Cut-off Date of 02 March 2008)

The time to first worsening of the ECOG performance status was calculated as the time from randomization to the date of the first worsening compared with the last ECOG evaluation obtained prior to randomization. Data were censored at the last date that the participant was known to be unchanged or improved from before randomization for the participants who had not had worsened at the time of the analysis and for the patients who were lost to follow-up before worsening in the ECOG performance status was documented. (NCT00424047)
Timeframe: Randomization to cut off date of 02 March 2008; up to 51 months

Interventionweeks (Median)
Lenalidomide Plus Dexamethasone10.1
Placebo Plus Dexamethasone12.3

[back to top]

Myeloma Response Rates Based on the Reviewers Best Response Assessment (Later Cut-off Date of 02 March 2008)

Complete Response (CR): Disappearance of monoclonal paraprotein and maintained for ≥ 6 weeks . Remission Response (RR):75-99% reduction in the level of the serum monoclonal paraprotein compared to baseline; 90-99% reduction in 24-hr urinary light chain excretion. Partial Response (PR): 50-74% reduction in the level of monoclonal paraprotein compared to baseline; 50-89% reduction in 24-hr urinary light chain excretion. Stable Disease (SD): Criteria for PR or PD have not been met. Plateau Phase: If PR, stable monoclonal paraprotein values (within 25% above or below nadir)/stable soft tissue plasmacytomas maintained for at least 3 months. Progressive Disease (PD): Reappearance of serum or urinary monoclonal paraprotein on immunofixation or electrophoresis on two consecutive occasions at least one week apart. Increase of percentage of plasma cells in bone marrow aspirate or biopsy to ≥ 5%. Development of at least one new lytic bone lesion or soft tissue plasmacytoma. (NCT00424047)
Timeframe: Randomization to data cut-off of 02 Mar 2008; up to 51 months

,
Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Not Evaluable (NE) those without response data
Lenalidomide Plus Dexamethasone17.042.628.43.48.5
Placebo Plus Dexamethasone4.019.456.614.35.7

[back to top]

Summary of Myeloma Response Rates Based on Best Response Assessment

Complete Response (CR): Disappearance of monoclonal paraprotein and maintained for ≥ 6 weeks . Remission Response (RR):75-99% reduction in the level of the serum monoclonal paraprotein compared to baseline; 90-99% reduction in 24-hr urinary light chain excretion. Partial Response (PR): 50-74% reduction in the level of monoclonal paraprotein compared to baseline; 50-89% reduction in 24-hr urinary light chain excretion. Stable Disease (SD): Criteria for PR or PD have not been met. Plateau Phase: If PR, stable monoclonal paraprotein values (within 25% above or below nadir)/stable soft tissue plasmacytomas maintained for at least 3 months. Progressive Disease (PD): Reappearance of serum or urinary monoclonal paraprotein on immunofixation or electrophoresis on two consecutive occasions at least one week apart. Increase of percentage of plasma cells in bone marrow aspirate or biopsy to ≥ 5%. Development of at least one new lytic bone lesion or soft tissue plasmacytoma. (NCT00424047)
Timeframe: Randomization to 03 August 2005; up to 24 months

,
Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Not Evaluable (NE) those without response data
Lenalidomide Plus Dexamethasone15.343.829.02.89.1
Placebo Plus Dexamethasone4.019.456.614.35.7

[back to top]

To Establish the Overall Response Rate Measured at 24 Weeks After First Dose of Lenalidomide Using This Dosing Regimen

To establish the overall response rate based on peripheral blood measures (absolute neutrophil count, platelets, and/or hemoglobin), lymphadenopathy, hepatomegaly, splenomegaly or constitutional symptoms; and bone marrow biopsy measured at 24 weeks after first dose of lenalidomide using this dosing regimen (NCT00439231)
Timeframe: 24 weeks of lenalidomide therapy

Interventionparticipants (Number)
Complete responsePartial responseNo response
CLL Subject Response Rate After Lenalidomide Therapy0528

[back to top]

6B Antibody Response to Prevnar Vaccine in Peripheral Blood

Serum IgG levels against the PVC serotype were measured by ELISA (NCT00445484)
Timeframe: basline and 8 weeks after second vaccination

Interventionfold change (Mean)
Vaccine Started 14 Days Prior to Lenalidomide3.69
Vaccine Started 45 Days After Lenalidomide7.58

[back to top]

14F Antibody Response to Prevnar Vaccine in Peripheral Blood

Serum IgG levels against the PVC serotype were measured by ELISA (NCT00445484)
Timeframe: basline and 8 weeks after second vaccination

Interventionfold change (Mean)
Vaccine Started 14 Days Prior to Lenalidomide9.42
Vaccine Started 45 Days After Lenalidomide11.95

[back to top]

19F Antibody Response to Prevnar Vaccine in Peripheral Blood

Serum IgG levels against the PVC serotype were measured by ELISA (NCT00445484)
Timeframe: basline and 8 weeks after second vaccination

Interventionfold change (Mean)
Vaccine Started 14 Days Prior to Lenalidomide2.025
Vaccine Started 45 Days After Lenalidomide2.12

[back to top]

23F Antibody Response to Prevnar Vaccine in Peripheral Blood

Serum IgG levels against the PVC serotype were measured by ELISA (NCT00445484)
Timeframe: basline and 8 weeks after second vaccination

Interventionfold change (Mean)
Vaccine Started 14 Days Prior to Lenalidomide4.1
Vaccine Started 45 Days After Lenalidomide2.42

[back to top]

Time to Disease Progression

International Myeloma Working Group Uniform Response Criteria was used (NCT00445692)
Timeframe: Up to 10.25 years

Interventionmonths (Median)
Treatment (Clarithromycin, Dexamethasone, Lenalidomide)30.5

[back to top]

Episodes of Grade 3-4 Non Infectious, Non-dermatological or Non-neurological Toxicities, Episodes of Any Infections, Grade 3-4 Dermatological or Episodes of Grade 2-3 Peripheral Neuropathy Common Terminology Criteria for Adverse Events Version 3

(NCT00445692)
Timeframe: First year of therapy

Interventionepisodes (Number)
NeutropeniaThrombocytopeniaDeep venous thrombus/Pulmonary EmbolismAnemiaPneumoniaUpper respiratory infectionsSinusitis/acute otitis mediaEpiglottic appendagitisCellulitisClostridium difficile colitisVaginitisPeripheral neuropathyDermal leukocytic vasculitisSecondary cancer Acute Myeloid LeukemiaRe-occurrence of skin cancer
Treatment (Clarithromycin, Dexamethasone, Lenalidomide)61114123131310111

[back to top]

Ratio of Change in Immune Response From Baseline

The pattern of immune response by assessing T cell and dendritic cell markers, specifically by measuring the ratio of BDCA-2 to BDCA-1 cells (NCT00460031)
Timeframe: Week 8

Interventionratio (Mean)
Ketoconazole Plus Lenalidomide-0.39

[back to top]

Change in Immune Response From Baseline

The pattern of immune response by assessing T cell and dendritic cell markers, specifically by measuring the levels of CD4+ FoxP3+ Regulatory T cells (NCT00460031)
Timeframe: Week 8

Interventioncells/ul (Mean)
Ketoconazole Plus Lenalidomide0.18

[back to top]

Time to Progression

Patients will be evaluated for clinical benefit monthly with PSA values.Patients who are benefiting from treatment are eligible for additional cycles of treatment. Thereafter, therapy will continue until criteria for progressive disease are met. Response is based on the RECIST criteria from the National Cancer institute. Complete Response (CR) disappearance of all target lesions; Partial Response (PR) >= 30% decrease in the sum of the longest diameter of target lesions from baseline; Progressive Disease (PD) >= increase in the sum of the longest diameter of target lesions from baseline; Stable Disease (SD) neither sufficient for partial response nor sufficient increase for progressive disease. (NCT00460031)
Timeframe: One year (12 months) after start of treatment

InterventionMonths (Median)
Ketoconazole Plus Lenalidomide3

[back to top]

Number of Patients With a Partial Response, Progressive Disease, or Stable Disease Based on Prostate-Specific Antigen (PSA) or Measurable Disease

Patients will be evaluated for clinical benefit monthly with PSA values.Patients who are benefiting from treatment are eligible for additional cycles of treatment. Thereafter, therapy will continue until criteria for progressive disease are met. Response is based on the RECIST criteria from the National Cancer institute. Complete Response (CR) disappearance of all target lesions; Partial Response (PR) >= 30% decrease in the sum of the longest diameter of target lesions from baseline; Progressive Disease (PD) >= increase in the sum of the longest diameter of target lesions from baseline; Stable Disease (SD) neither sufficient for partial response nor sufficient increase for progressive disease. (NCT00460031)
Timeframe: 28 days

Interventionparticipants (Number)
Partial ResponseProgressive DiseaseStable Disease
Ketoconazole Plus Lenalidomide779

[back to top]

Number of Patients With Grade 3 and 4 Toxicity as Assessed by NCI CTCAE v3.0

Patients will be evaluated for toxicity every 2 weeks during the first cycle. Thereafter, evaluations will be done every 28 days or more frequently if clinically indicated. (NCT00460031)
Timeframe: Up to 30 days after discontinuation of treatment

Interventionparticipants (Number)
Ketoconazole Plus Lenalidomide19

[back to top]

Response to Treatment

"In general response to treatment is defined as either complete response (CR) or partial response (PR) assessed using Composite Assessment (CA) of index lesion disease severity and is defined as the following:~CR =CA ratio=0/no evidence of new disease (abnormal or pathologically positive lymph nodes, cutaneous or other tumor manifestations, visceral disease) present over 4 weeks. Patients with Sézary Syndrome must have no evidence of circulating Sézary cells (< 5% Sézary cells=not significant). Skin biopsy is required for documentation of CR. Confirmatory CT scans are required, if baseline CTs were abnormal.~PR= CA ratio ≥0.5/no new clinically abnormal lymph nodes/no progression of existing clinically abnormal lymph nodes (<25%)/no new cutaneous tumors/no new pathologically positive lymph nodes or visceral disease in an area previously documented as-ve for at least 4 weeks. In patients with circulating Sézary cells at least a 50% reduction of malignant lymphocytes is required." (NCT00466921)
Timeframe: After cycle 4 of treatment (1 cycle =28 days)

Interventionparticipants (Number)
CRPR
Lenalidomide09

[back to top]

Number of Patients Who Experience Toxicity as Assessed by NCI CTCAE v3.0

"Toxicity is defined as the number of patients who patients who experienced an adverse event that was determined to be at least possibly related to study drug and determined to be a grade 3 or higher in severity as assessed by the Common Terminology Criteria for Adverse Events v3.0 (CTCAE) where generally:~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" (NCT00466921)
Timeframe: From treatment initiation until up to 30 days post treatment with possible 4 cycles of initial treatment (1 cycle =28 days) and up to 2 further years of treatment permitted if meeting response criteria

Interventionpatients (Number)
Fatigue (grade 3)Infection (grade 3)Leukopenia (grade 3)
Lenalidomide711

[back to top]

Progression-free Survival (PFS)

"PFS is defined from the time of treatment initiation until documentation of progressive disease or death from any cause.~Progressive disease is defined as (PD) ≥25% increase in CA ratio, ≥25% increase in no. or area of clinically abnormal lymph nodes/new tumors/new pathologically positive lymph nodes/visceral disease/an increase >25% in no. of Sézary cells." (NCT00466921)
Timeframe: From time of treatment initiation until progression or death from any cause (up to a possible maximum of approximately 6 years)

Interventionmonths (Median)
Lenalidomide8

[back to top]

Duration of Response (DOR)

DOR is defined as time of initial documentation of response to the time of documentation of progression in patients who achieve either a complete response (CR) and partial response (PR) (NCT00466921)
Timeframe: From time of initial response until progressive disease (up to approximately 1 year)

Interventionmonths (Median)
Lenalidomide10

[back to top]

Overall Survival (OS) at 3 Years

OS was defined as the time from registration to death due to any cause. Patients who were alive were censored at date of last follow-up. The overall survival at 3 years (a percentage) is reported below. (NCT00477750)
Timeframe: registration to death (up to 3 years)

Interventionpercentage of patients (Number)
Treatment (Lenalidomide, Melphalan, Prednisone)58

[back to top]

Duration of Response (DOR)

Duration of response was calculated from documentation of first response to date of progression in the subset of patients who responded. Patients without progression were censored at the date of last tumor evaluation. (NCT00477750)
Timeframe: from first response to progression or death (up to 3 years)

Interventionmonths (Median)
Treatment (Lenalidomide, Melphalan, Prednisone)16.3

[back to top]

Progression-free Survival

"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 was defined as any one or more of the following:~An increase of 25% from lowest confirmed response in:~Serum M-component (absolute increase >= 0.5g/dl)~Urine M-component (absolute increase >= 200mg/24hour~Difference between involved and uninvolved Free Light Chain levels (absolute increase >= 10mg/dl~Bone marrow plasma cell percentage (absolute increase of >=10%)" (NCT00477750)
Timeframe: registration to progressive disease (up to 3 years)

Interventionmonths (Median)
Treatment (Lenalidomide, Melphalan, Prednisone)21.4

[back to top]

Percentage of Participants With Toxicity, Assessed Using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0 (v3)

The overall toxicity rates (percentages) for grade 3 or higher adverse events considered at least possibly related to treatment are reported below. (NCT00477750)
Timeframe: Every cycle during treatment up to 3 years

Interventionpercentage of patients (Number)
Grade 3Grade 4Grade 5
Treatment (Lenalidomide, Melphalan, Prednisone)33670

[back to top]

Patients With Overall Confirmed Response

"Response that was confirmed on 2 consecutive evaluations.>~Complete Response (CR): Complete disappearance of M-protein from serum and urine on immunofixations, normalization of Free Light Chain (FLC) ratio and <=5% plasma cells in bone marrow>~Very Good Partial Response (VGPR): >=90% reduction in serum M-spike, Urine M-spike <100mg per 24 hours>~Partial Response (PR): >=50% reduction in serum M-spike, Urine M-spike >=90% reduction or < 200mg per 24 hours, or >=50% decrease in difference between involved and uninvolved FLC levels or 50% decrease in bone marrow plasma cells" (NCT00477750)
Timeframe: Every cycle during treatment

Interventionparticipants (Number)
CRVGPRPR
Treatment (Lenalidomide, Melphalan, Prednisone)3510

[back to top]

Overall Survival (OS)

OS was defined as the time from registration to death of any cause. Participants were followed for a maximum of 5 years from randomization. The median OS with 95%CI was estimated using the Kaplan Meier method. (NCT00478218)
Timeframe: up to 5 years

Interventionmonths (Median)
LCD (Cyclophosphamide 300 mg/m^2)NA
LCD (Cyclophosphamide 300 mg)NA

[back to top]

Duration of Response (DOR)

Duration of response was calculated from the documentation (date) of first response (CR, VGPR, or PR) until the date of progression or last follow-up in the subset of patients who responded. The median DOR with 95%CI was estimated using the Kaplan Meier method. (NCT00478218)
Timeframe: up to 5 years

Interventionmonths (Median)
LCD (Cyclophosphamide 300 mg/m^2)26.1
LCD (Cyclophosphamide 300 mg)NA

[back to top]

Number of Participants Who Achieved a Confirmed Response (CR), Very Good Partial Response (VGPR) or Partial Response (PR) During Treatment

"Response that was confirmed on 2 consecutive evaluations during treatment~Complete Response(CR): Complete disappearance of M-protein from serum & urine on immunofixation, normalization of Free Light Chain (FLC) ratio & <5% plasma cells in bone marrow (BM)~Very Good Partial Response(VGPR): >=90% reduction in serum M-component; Urine M-Component <100 mg per 24 hours; <=5% plasma cells in BM~Partial Response PR): >= 50% reduction in serum M-Component and/or Urine M-Component >= 90% reduction or <200 mg per 24 hours; or >= 50% decrease in difference between involved and uninvolved FLC levels" (NCT00478218)
Timeframe: Duration of Treatment (up to 5 years)

Interventionparticipants (Number)
LCD (Cyclophosphamide 300 mg/m^2)28
LCD (Cyclophosphamide 300 mg)16

[back to top]

Progression-free Survival (PFS)

"PFS was defined as the time from registration to progression or death due to any cause. The median PFS with 95%CI was estimated using the Kaplan Meier method.> Progression was defined as any one or more of the following:> An increase of 25% from lowest confirmed response in:>~Serum M-component (absolute increase >= 0.5g/dl)>~Urine M-component (absolute increase >= 200mg/24hour>~Difference between involved and uninvolved Free Light Chain levels (absolute increase >= 10mg/dl>~Bone marrow plasma cell percentage (absolute increase of >=10%)" (NCT00478218)
Timeframe: up to 5 years

Interventionmonths (Median)
LCD (Cyclophosphamide 300 mg/m^2)27
LCD (Cyclophosphamide 300 mg)NA

[back to top]

Participants With Treatment-emergent Adverse Experiences (TEAEs)

"Counts of study participants who had treatment-emergent adverse events (TEAEs) defined as any reported AE that started on or after the first day of study drug dosing. A participant with multiple occurrences of an adverse event within a category is counted only once in that category.~National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE version 3.0) was used by investigators to assess TEAEs. Severity scale ranges from 0 (none) to 5 (death). Grade 3=severe AE; Grade 4=life threatening or disabling AE; Grade 5=death." (NCT00478777)
Timeframe: up to 8 months

Interventionparticipants (Number)
>=1 TEAE>=1 TEAE related to study drug>=1 NCI CTCAE grade 3 or 4 TEAE>=1 NCI CTCAE grade 3 or 4 TEAE related to drug>=1 serious AE (SAE)>=1 study drug related SAEDiscontinued due to TEAEDiscontinued due to TEAE related to study drugTEAE leading to dose reductionTEAE leading to dose interruptionDeaths
Lenalidomide Plus Dexamethasone1391191058379483221356479

[back to top]

Participant's Best Overall Response Based on the European Group for Blood and Marrow Transplantation (EBMT) Myeloma Response Criteria

"Best overall response was calculated as the best assessment from all cycles (including treatment discontinuation visit) and follow-up. The response rate was summarized as complete response (CR), partial response (PR), stable disease (SD), progression (PD), response not evaluable, and derived categories (PR+CR) and (PR+CR+SD).~CR is negative immunofixation on both serum and urine maintained for 6 weeks straight. PR is a 50% decrease in serum paraprotein maintained for 6 weeks straight. SD is serum paraprotein values within 25% of baseline." (NCT00478777)
Timeframe: Up to 827 days

Interventionparticipants (Number)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Not evaluable (NE)CR + PRCR + PR + SD
Lenalidomide Plus Dexamethasone6973038103133

[back to top]

Kaplan Meier Estimate for Time to Disease Progression

"Time to disease progression (TTP) was based on the European Group for Blood and Marrow Transplantation (EBMT) myeloma response determination criteria developed by Bladé (Bladé, 1998). TTP is a Kaplan Meier estimate of the time from randomization to the first documentation of progressive disease.~Progressive disease based on increasing monoclonal paraprotein levels require a confirmatory value one week apart. Disease progression can also be based on bone marrow findings, worsening lytic bone disease, progressively enlarging extramedullary plasmacytomas, or hypercalcemia." (NCT00478777)
Timeframe: up to 827 days

Interventiondays (Median)
Lenalidomide Plus Dexamethasone214.0

[back to top]

Response Rate (Complete and Partial Response)

Response was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST). Complete response is the disappearance of all target lesions. Partial response is at least a 30% decrease in the sum of the longest diameter (LD) of target lesions taking as reference the baseline sum LD. (NCT00482911)
Timeframe: 2 years

,
InterventionParticipants (Number)
Complete ResponsePartial Response
Cohort 1-lenalidomide & Cyclophosphamide00
Cohort 2-sunitinib & Cyclophosphamide00

[back to top]

Toxicity

Here is the number of participants with adverse events. For a detailed list of adverse events see the adverse event module. (NCT00482911)
Timeframe: 16 months

InterventionParticipants (Number)
Cohort 1-lenalidomide & Cyclophosphamide3
Cohort 2-sunitinib & Cyclophosphamide8

[back to top]

Number of Patients With Stringent Complete Response Rate

Stringent Complete Response is defined as complete response plus normal free light chain (kappa/lambda) ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence. (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression

Interventionparticipants (Number)
V-DR7
VDCR6
V-DC3
VDC-mod5

[back to top]

Overall Survival

Overall survival is defined as time from the date of randomization to the date of death (NCT00507442)
Timeframe: Up to 48 weeks or until death

Interventiondays (Median)
V-DRNA
VDCRNA
V-DCNA
VDC-modNA

[back to top]

Probability of 1-year Survival

(NCT00507442)
Timeframe: survival probability at 1 year after randomization

Interventionpercentage of patients (Number)
V-DR100
VDCR91.6
V-DC100
VDC-mod100

[back to top]

Number of Patients With Combined Complete Response and Very Good Partial Response

"Complete response requires negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow.~Very good partial response requires serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 h" (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression

Interventionparticipants (Number)
V-DR21
VDCR23
V-DC13
VDC-mod9

[back to top]

Time to Disease Progression

"Time to disease progression is defined as time from the date of randomization to the date of first documented progressive disease.~Disease progression requires any one or more of the following: serum m-protein increase >= 25% from nadir(absolute increase >= 0.5 g/dL); Urine m-protein increase >= 25% from nadir(absolute increase >= 200 mg/24 hr), bone marrow plasma cell percentage increase >= 25% from nadir(absolute increase >= 10%), new bone lesion or soft tissue plasmacytomas." (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression

Interventiondays (Median)
V-DRNA
VDCRNA
V-DCNA
VDC-modNA

[back to top]

Time to Response

Time to response is defined as time from date of randomization to the date of the first documentation of a confirmed response. confirmed response is a response that has been observed on at least two consecutive assessments. (NCT00507442)
Timeframe: Up to 48 weeks or until disease response

Interventiondays (Median)
V-DR49
VDCR50
V-DC55
VDC-mod49

[back to top]

Progression-free Survival

"Progression-free survival is defined as time from the date of randomization to the date of the first documented progressive disease or death.~Disease progression requires any one or more of the following: serum m-protein increase >= 25% from nadir(absolute increase >= 0.5 g/dL); Urine m-protein increase >= 25% from nadir(absolute increase >= 200 mg/24 hr), bone marrow plasma cell percentage increase >= 25% from nadir(absolute increase >= 10%), new bone lesion or soft tissue plasmacytomas." (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression/death

Interventiondays (Median)
V-DRNA
VDCR631
V-DCNA
VDC-modNA

[back to top]

Duration of Response

"Duration of response is the time from date of first documented confirmed response to date of first documented progressive disease. A confirmed response is a response that has been observed on at least two consecutive assessments.~Disease progression requires any one or more of the following: serum m-protein increase >= 25% from nadir(absolute increase >= 0.5 g/dL); Urine m-protein increase >= 25% from nadir(absolute increase >= 200 mg/24 hr), bone marrow plasma cell percentage increase >= 25% from nadir(absolute increase >= 10%), new bone lesion or soft tissue plasmacytomas." (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression

Interventiondays (Median)
V-DRNA
VDCRNA
V-DCNA
VDC-modNA

[back to top]

Number of Patients With Adverse Events (AEs)

Evaluate the safety and tolerability of the combination therapy (NCT00507442)
Timeframe: From first dose of study drug through the 30 day post-treatment AE assessment visit

Interventionparticipants (Number)
V-DR42
VDCR65
V-DC33
VDC-mod17

[back to top]

Number of Patients With Complete Response Rate + Near Complete Response Rate

"Complete response requires negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow.~Near Complete response requires positive immunofixation on the serum and/or urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow." (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression

Interventionparticipants (Number)
V-DR17
VDCR14
V-DC10
VDC-mod8

[back to top]

Number of Patients With Overall Response

"Overall Response includes complete response and partial response.~Complete response requires negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow.~Partial response requires at least 50% reduction of serum M-protein and reduction in 24-h urinary M-protein by at least 90% or to < 200 mg per 24 hour." (NCT00507442)
Timeframe: Up to 48 weeks or until disease progression

Interventionparticipants (Number)
V-DR35
VDCR35
V-DC24
VDC-mod17

[back to top]

Response Rates (Complete Response [CR] or Very Good Partial Response [VGPR])

"CR:~Patients with complete disappearance of an M-protein and no evidence of myeloma in the bone marrow are considered to have CR. To be considered CR, patients must meet all of the following criteria:~Negative immunofixation on the serum and urine at two consecutive times~Disappearance of any soft tissue plasmacytomas~≤5% plasma cells in bone marrow~If serum and urine M protein are unmeasurable and the immunoglobulin free light chain (FLC) parameter is being used, patients must have a normal ratio of 0.26-1.65 at two consecutive times~VGPR:~Serum and urine M-component detectable by immunofixation but not on electrophoresis OR~>=90% reduction in serum M-component plus urine M-component <100 mg per 24 hours (by SPEP and UPEP)~If the serum and urine M protein are unmeasurable and the immunoglobulin FLC parameter is being used, a >90% decrease in the difference between involved and uninvolved FLC levels is required in place of the M protein criteria" (NCT00522392)
Timeframe: Assessed at the end of each cycle, every 3 months if patient is < 2 years from study entry, every 6 months if patient is 2-5 years from study entry, every 12 months if patient is 6-10 years from study entry, up to 10 years

InterventionProportion of patients (Number)
Arm A (VRD)0.625
Arm B (VD)0.188

[back to top]

Progression-free Survival (PFS)

"Progression-free survival was defined as the time from randomization to the earliest documentation of disease progression (PD) or death. If a patient died without evidence of PD, the patient was considered an event if death occurred within 3 months of the last disease assessment. Patients who died outside of the specified interval or patients who were alive without evidence of PD were censored at the date of last disease assessment.~The PFS results are based on data as of August 2012, while overall survival (OS) was updated in April 2014. Given the early termination and limited sample size, data management efforts to update PFS were not pursued." (NCT00522392)
Timeframe: Assessed every 3 months if patient is < 2 years from study entry, every 6 months if patient is 2-5 years from study entry, every 12 months if patient is 6-10 years from study entry, up to 10 years

InterventionMonths (Median)
Arm A (VRD)NA
Arm B (VD)17.4

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from randomization to death or date of last known alive. The OS results are based on data as of April 2014. (NCT00522392)
Timeframe: Assessed every 3 months if patient is < 2 years from study entry, every 6 months if patient is 2-5 years from study entry, every 12 months if patient is 6-10 years from study entry, up to 10 years

InterventionMonths (Median)
Arm A (VRD)64.0
Arm B (VD)NA

[back to top]

Change in Quality of Life (QOL) From Baseline to 6 Months Post Consolidation as Assessed by the Functional Assessment of Cancer Therapy-Neurotoxicity Trial Outcome Index (FACT-Ntx TOI)

The combined score on the FACT-Ntx TOI is of interest. The FACT-Ntx TOI has 25 items and the score ranges from 0 (worst possible quality of life) -100 (best possible quality of life). The primary QOL endpoint is defined as the change in the FACT-Ntx TOI score from registration to 6 months post consolidation treatment. (NCT00522392)
Timeframe: Baseline and 6 months post consolidation treatment

Interventionunits on a scale (Mean)
Arm A (VRD)-7.9
Arm B (VD)-2.6

[back to top]

Overall Response Rate (ORR)

ORR defined as number of participants with best response of Complete Response (CR) or Partial Response (PR) out of total number of participants. CR is defined as absence of lymphadenopathy, hepatomegaly or splenomegaly on physical exam. Normal Complete Blood Count (CBC) with polymorphonuclear leukocytes >1500/µL, platelets >100,000/µL, hemoglobin >11.0 g/dL (untransfused); lymphocyte count <5,000/µL; Bone marrow aspirate and biopsy must be normocellular for age with <30% of nucleated cells being lymphocytes. Lymphoid nodules must be absent. PR requires a 50% decrease in peripheral lymphocyte count from , 50% reduction in lymphadenopathy, and/or 50% reduction in splenomegaly/hepatomegaly for a period of at least two months from completion of therapy. These patients must have one of the following: Polymorphonuclear leukocytes 1,500/µL or 50% improvement ; Platelets >100,000/µL or 50% improvement ; Hemoglobin >11.0 g/dL (untransfused) or 50% improvement from pre-treatment value. (NCT00535873)
Timeframe: From 3 cycles (90 days) up to 6 cycles (approximately 180 days)

Interventionparticipants (Number)
Lenalidomide39

[back to top]

Complete Response Rate

An improvement in complete response to at least 60% following treatment, assessed using CT scans, clinical/lab examinations, and bone marrow aspirations, as defined by National Cancer Institute Working Group Response Criteria. (NCT00536341)
Timeframe: At 12 weeks during treatment and 2 months post-treatment until disease progression, projected 8 months

Interventionparticipants (Number)
Dose Level 14
Dose Level 29

[back to top]

Overall Survival

Defined as the time from Day 1 of treatment administration to date of death from any cause, estimated using Kaplan-Meier methods. (NCT00536341)
Timeframe: Every 3 months until treatment discontinuation, expected average of 6 months and then every 6 months thereafter up to 5 years

Interventionmonths (Median)
All PatientsNA

[back to top]

Number of Adverse Events as a Measure of Safety and Tolerability

Recorded from first treatment until 30 days after last treatment and assessed using Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (NCT00536341)
Timeframe: 63 months

,
Interventionparticipants (Number)
FatigueNeutropeniaAnemiaLeukopeniaThrombocytopeniaRashNauseaConstipationAnorexiaFeverHyperhidrosisArthralgiaEdema LimbsPruritusBack painHeadacheChillsInsomniaVomitingDysgeusiaAbdominal PainAllergic ReactionDiarrheaCoughDizzinessDyspneaHypotensionMyalgia
Dose Level 18787867343313031222124322012
Dose Level 24041343430272515121313141215111311111111978881098

[back to top]

Progression-Free Survival

Measured from first treatment to disease progression and assessed using Kaplan-Meier methods. (NCT00536341)
Timeframe: Every 3 months during treatment until disease progression and every 6 months thereafter, up to 5 years

Interventionmonths (Median)
All Patients24.64

[back to top]

Effect of Drug Combination on Multiple Myeloma

Objective response rate, defined according to the International Myeloma Working Group (IMWG) criteria as greater then or equal to a Partial Response (PR). The best response was recorded. The IMWG criteria can be found here: imwg.myeloma.org/international-myeloma-working-group-imwg-uniform-response-criteria-for-multiple-myeloma/ (NCT00538733)
Timeframe: This was collected from patients for their duration on study treatment. Only the best response was recorded. Best responses were reported at any point of the study, from start of treatment up until removal of study, which occurred up to 57.4 cycles

Interventionparticipants (Number)
sCR (stringent complete response)VGPR (very good partial response)PR (partial response)SD (stable disease)
T-BiRD Therapy (All Patients)2995

[back to top]

Progression Free Survival

"Progression determined using International Myeloma Working Group criteria, as defined below.~An increase of > 25% from lowest response value one or more of the following:~Serum M-component and/or (the absolute increase must be > 0.5 g/dL)*~Urine M-component and/or (the absolute increase must be > 200 mg/24 h)~Only in patients without measurable serum and urine M-protein levels; the difference between involved and uninvolved FLC levels. The absolute increase must be > 10 mg/dL~Bone marrow plasma cell percentage; the absolute percentage must be > 10%~Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas~Development of hypercalcaemia (corrected serum calcium > 11.5 mg/dL or 2.65 mmol/L) that can be attributed solely to the plasma cell proliferative disorder *if starting serum M protein is greater then 5 g/dL, absolute increase of 1g/dL is sufficient to determine relapse." (NCT00538733)
Timeframe: From start of treatment, to the date of first progression

Interventionmonths (Median)
T-BiRD Therapy (All Patients)35.6

[back to top]

Median Time to Maximum Response

Median Time to maximum response, reported in cycles of treatment. One cycle = 28 days. (NCT00538733)
Timeframe: from baseline to cycle with maximum response

Interventioncycles (Median)
T-BiRD Therapy (All Patients)4

[back to top]

Event Free Survival

(NCT00538733)
Timeframe: from baseline to the time of first event that lead to removal from study (defined as progression, death, withdrawal of consent, or removal for toxicity)

Interventionmonths (Median)
T-BiRD Therapy (All Patients)21.5

[back to top]

Effect of Drug Combination on Multiple Myeloma

The maximum response for all patients that were treated on study. Maximum response was assessed using the International myeloma working group (IMWG) guidelines for response. http://imwg.myeloma.org/international-myeloma-working-group-imwg-uniform-response-criteria-for-multiple-myeloma/ (NCT00538824)
Timeframe: The best response for all patients at any point were assessed for patients that were treated on study, from start of treatment up to 20 weeks

Interventionparticipants (Number)
partial response (PR)minimal response (MR)progression of disease
DexTR (All Patients)212

[back to top]

Relapse Free Survival (RFS)

(NCT00540007)
Timeframe: Through 3.5 years from study entry or until disease progression

Interventionmonths (Median)
Cohort 1 - Lenalidomide Daily on Days 1-213.78
Cohort 2 - Lenalidomide Daily on Days 1-283.93

[back to top]

Safety and Tolerability of Lenalidomide Therapy as Measured by the Number of Participants Who Experience Each Adverse Event (Grade 3 or 4 Adverse Events Only) Refractory cHL.

"Adverse events were graded according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 3.0~The higher the grade the worse the adverse event was considered" (NCT00540007)
Timeframe: 30 days following the completion of treatment

,
Interventionparticipants (Number)
NeutropeniaLeukopeniaAnemiaLymphopeniaThrombocytopeniaFatigueASTALTBilirubinSensory neuropathyDehydrationInfection without neutropeniaInfection with neutropeniaEdemaDyspneaPleural effusionAlkaline phosphataseAbdominal painLow potassiumLow sodiumLow albuminLow calciumHigh calciumLow phosphorusHearing lossThrombosis/embolismRashFebrile neutropeniaPneumoniaHigh potassiumHyperuricemiaConfusionDizzinessSpeech impairmentChest painExtremity painMuscle painSecondary malignancy - MDS
Cohort 1 - Lenalidomide Daily on Days 1-2118111097332222211111132111100010000000000
Cohort 220134108211200300000032000011122111121211

[back to top]

Time to Progression (TTP).

-Time to progression (TTP) is defined as the time from study entry until documented lymphoma progression or death as a result of lymphoma. (NCT00540007)
Timeframe: Through 3.5 years from study entry or until disease progression

Interventionmonths (Median)
Cohort 1 - Lenalidomide Daily on Days 1-213.68
Cohort 24.08

[back to top]

Duration of Response

-Duration of response: defined as the interval from the date of response (CR or PR) is documented to the date of progression, taking as reference the smallest measurements recorded since the treatment started (NCT00540007)
Timeframe: Through 3.5 years from study entry or until disease progression

Interventionmonths (Median)
Cohort 1 - Lenalidomide Daily on Days 1-213.68
Cohort 2 - Lenalidomide Daily on Days 1-284.08

[back to top]

Cytostatic Overall Response Rate

"Cytostatic overall response rate = CR + PR + SD greater than or equal to 6 months~Definitions per 2007 Cheson Lymphoma Response Criteria" (NCT00540007)
Timeframe: From 6 months through 3.5 years after study entry

InterventionParticipants (Count of Participants)
Cohort 1 - Lenalidomide Daily on Days 1-2115
Cohort 2 - Lenalidomide Daily on Days 1-2818

[back to top]

Event Free Survival (EFS).

-Event-free survival (time to treatment failure) is measured from the time from study entry to any treatment failure including disease progression, or discontinuation of treatment for any reason (eg, disease progression, toxicity, patient preference, initiation of new treatment without documented progression, or death). (NCT00540007)
Timeframe: Through 3.5 years from study entry or until disease progression

Interventionmonths (Median)
Cohort 1 - Lenalidomide Daily on Days 1-213.78
Cohort 2 - Lenalidomide Daily on Days 1-283.93

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from entry onto the clinical trial until death as a result of any cause. (NCT00540007)
Timeframe: Through 3.5 years from study entry or until disease progression

Interventionmonths (Median)
Cohort 1 - Lenalidomide Daily on Days 1-2117.434
Cohort 2 - Lenalidomide Daily on Days 1-2823.717

[back to top]

Objective Overall Response Rate (ORR) in Relapsed or Refractory cHL.

"Overall response rate = CR + PR~Definitions per 2007 Cheson Lymphoma Response Criteria" (NCT00540007)
Timeframe: Through 3.5 years from study entry or until disease progression

Interventionpercentage of participants (Number)
Cohort 1 - Lenalidomide Daily on Days 1-2121.0
Cohort 2 - Lenalidomide Daily on Days 1-2828.6

[back to top]

Quality of Life Using the FACT-G Data

"Change from baseline FACT-G scores. The quality of life questionnaire (FACT-G) was given at various timepoints during the study. The values for change from baseline to endpoint are provided.~Physical Well-Being (PWB; sum of 7 items, point range 0-28); Social/Family Well-Being (SWB, sum of 7-items, point range 0-28); Emotional Well-Being (EWB; sum of 6-items, point range 0-24); Functional Well-Being (FWB; sum of 7-items, point range 0-28) ; Fact-G score=sum of PWB, SWB, EWB, FWB, point range 0-108. Note: The higher the score, the better the outcome" (NCT00540644)
Timeframe: baseline and after last cycle (up to 6 cycles)

,
Interventionscores on a scale (Mean)
Physical Well-Being Change from BaselineSocial/Family Well-Being Change from BaselineEmotional Well-Beling Change from BaselineFunctional Well-Being Change from BaselineFACT-G Change from Baseline
Extension - Revlimid, Cyclophosphamide, Prednisone-2.81-0.230.60-1.17-3.61
Original Study - Revlimid, Cyclophosphamide, Prednisone1.57-0.032.523.387.44

[back to top]

Response Rate (RR) After 6 Cycles of Therapy Using the Proposed International Myeloma Working Group Uniform Response Criteria

Evaluate the response rate of patients receiving therapy. Patients are considered as having a response if their overall response is Partial Response or better using the proposed International Myeloma Working Group uniform response criteria. The percentage of patients achieving this and the exact 95% confidence interval will be calculated. (NCT00540644)
Timeframe: After 6 cycles

Interventionpercentage of participants (Number)
Original Study - Revlimid, Cyclophosphamide, Prednisone86.7
Extension - Revlimid, Cyclophosphamide, Prednisone71.4

[back to top] [back to top]

Duration of CR for Complete Responders

Duration of remission: Defined as the interval from the date complete remission is documented to the date of recurrence (NCT00546897)
Timeframe: 2 years

Interventionmonths (Median)
Cohort 210

[back to top]

CR With Complete Blood Counts (CRi) Rate

CRi = Defined as CR with the exception of neutropenia <1000/uL or thrombocytopenia <100,000/ul. (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

Interventionparticipants (Number)
Cohort 10
Cohort 24

[back to top]

Response Rate (RR)

"RR = as patients obtaining any response (CRm + CRc +CRi + PR).~CRm = Defined as morphologic leukemia-free state, including <5% blasts in BM aspirate with marrow spicules and a count of > 200 nucleated cells and no blasts with Auer rods, no persistent extramedullary disease, ANC > 1000/uL, platelet count > 100,000/uL. Patient must be independent of transfusions for a minimum of 1 week before each marrow assessment. There is no duration requirement for this designation.~CRc = Cytogenetic complete remission (CRc): Only patients with an identified cytogenetic abnormality may receive this designation. Defines as a morphologic complete remission plus reversion to a normal karyotype (no clonal abnormalities detected in a minimum of 20 mitotic cells).~Morphologic complete remission with incomplete blood count recovery (CRi): Defined as CR with the exception of neutropenia <1000/uL or thrombocytopenia <100,000/ul.~Partial remission (PR): Requires" (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

,
Interventionparticipants (Number)
CRmCRcCRiPR
Cohort 10101
Cohort 23340

[back to top]

Morphologic Leukemia Free State

Morphologic leukemia-free state: Defined as < 5% blasts on the BM aspirate with spicules and a count of > 200 nucleated cells and no blasts with Auer rods, and no persistent extramedullary disease. (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

Interventionparticipants (Number)
Cohort 11
Cohort 210

[back to top]

Morphologic Complete Remission Rate (CRm)

CRm = Defined as morphologic leukemia-free state, including <5% blasts in BM aspirate with marrow spicules and a count of > 200 nucleated cells and no blasts with Auer rods, no persistent extramedullary disease, ANC > 1000/uL, platelet count >100,000/uL. Patient must be independent of transfusions for a minimum of 1 week before each marrow assessment. There is no duration requirement for this designation. (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

Interventionparticipants (Number)
Cohort 10
Cohort 23

[back to top]

Safety and Tolerability (Removal From Study Due to Adverse Events)

Toxicity will be scored using CTCAE Version 3.0 for toxicity and adverse event reporting (NCT00546897)
Timeframe: 4 weeks after last dose of study drug [median duration of therapy was 65 days (range, 3-413 days)]

Interventionparticipants (Number)
Cohort 11
Cohort 28

[back to top]

Relapse Free Survival (RFS) for Complete Responders

This is determined only for patients achieving a complete remission. Defined as the interval from the date of first documentation of a leukemia free state to date of recurrence or death due to any cause. (NCT00546897)
Timeframe: 2 years

Interventionmonths (Median)
Cohort 210

[back to top]

Progression-free Survival

Progression-free survival (PFS) denotes the chances of staying free of disease progression for a group of individuals suffering from a cancer after a particular treatment. It is the percentage of individuals in the group whose disease is likely to remain stable (and not show signs of progression) after a specified duration of time. Progression-free survival rates are an indication of how effective a particular treatment is. (NCT00546897)
Timeframe: 2 years

Interventionmonths (Median)
Cohort 22

[back to top]

Partial Remission Rate (PR)

Partial remission (PR): Requires that the criteria for complete remission be met with the following exceptions: decrease of >50% in the percentage of blasts to 5-25% in the BM aspirate. A value of < 5% blasts in BM with Auer rods is also considered a partial remission. (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

Interventionparticipants (Number)
Cohort 11
Cohort 20

[back to top]

Overall Survival (OS)

Overall survival: Defined as the date of first dose of study drug to the date of death from any cause. (NCT00546897)
Timeframe: 2 years

Interventionmonths (Median)
Cohort 24

[back to top]

Complete Remission Rate (CRm + CRi + CRc)

"CRm = Defined as morphologic leukemia-free state, including <5% blasts in BM aspirate with marrow spicules and a count of > 200 nucleated cells and no blasts with Auer rods, no persistent extramedullary disease, ANC > 1000/uL, platelet count >100,000/uL. Patient must be independent of transfusions for a minimum of 1 week before each marrow assessment. There is no duration requirement for this designation.~CRi = Defined as CR with the exception of neutropenia <1000/uL or thrombocytopenia <100,000/ul.~Cytogenetic complete remission (CRc): Only patients with an identified cytogenetic abnormality may receive this designation. Defines as a morphologic complete remission plus reversion to a normal karyotype (no clonal abnormalities detected in a minimum of 20 mitotic cells)." (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

,
Interventionparticipants (Number)
CRmCRiCRc
Cohort 1001
Cohort 2343

[back to top]

Cytogenetics CR Rate (CRc)

Cytogenetic complete remission (CRc): Only patients with an identified cytogenetic abnormality may receive this designation. Defines as a morphologic complete remission plus reversion to a normal karyotype (no clonal abnormalities detected in a minimum of 20 mitotic cells). (NCT00546897)
Timeframe: After 2 cycles of low dose lenalidomide (approximately Day 113 for Cohort 1 and approximately Day 104 for Cohort 2)

Interventionparticipants (Number)
Cohort 11
Cohort 23

[back to top]

Number of Participants With an Overall Response Defined as Complete Response and Partial Response

"Response is assessed by investigator according to International Working Group (IWG) criteria.~A complete response requires disappearance of all evidence of disease. A partial response is a >/= 50% decrease in the sum of products of 6 largest dominant nodes or nodal masses as well as for splenic and hepatic nodules. No increase in size of nodes, liver or spleen and no new sites of disease." (NCT00553644)
Timeframe: Duration of treatment (assessed up to 6 years)

Interventionparticipants (Number)
Treatment (Antiangiogenesis Therapy, Enzyme Inhibitor Therapy)21

[back to top]

Time to Progression

Time to progression (TTP) is defined as the time from study entry until progression or death due to any cause. The median TTP with 95% CI was estimated using the Kaplan-Meier method. (NCT00553644)
Timeframe: Assessed up to 6 years

Interventionyears (Median)
Treatment (Antiangiogenesis Therapy, Enzyme Inhibitor Therapy)0.58

[back to top]

Overall Survival

Overall survival (OS) is defined as the time from study entry until death. The median OS with 95% CI was estimated using the Kaplan-Meier method.. (NCT00553644)
Timeframe: Assessed up to 6 years

Interventionyears (Median)
Treatment (Antiangiogenesis Therapy, Enzyme Inhibitor Therapy)2.17

[back to top]

Incidence of Adverse Events

Number of participants who experienced a maximum grade 3, 4 or 5 adverse event. The grading scales found in the revised NCI CTCAE version 4.0 was utilized for adverse event reporting (NCT00553644)
Timeframe: Duration of Treatment (up to 6 years)

Interventionparticipants (Number)
Grade 2Grade 3Grade 4Grade 5
Treatment (Antiangiogenesis Therapy, Enzyme Inhibitor Therapy)429171

[back to top]

Number of Participants in the Dose Escalation Phase Who Reached Maximum Tolerated Dose (MTD) of Dasatinib With Lenalidomide and Dexamethasone

The MTD is considered the last dose level combination tested just below the maximum administered dose (MAD) level combination and for which DLTs were observed in less than 33% of participants during the escalation and expansion phase. Please refer to outcome 2 for the complete definition of DLT. If the MTD was not reached at the highest dose administered as defined by protocol, the highest dose (dasatinib 140 mg QD + lenalidomide 25 mg QD) administered was selected for the dose expansion phase of the study. (NCT00560391)
Timeframe: From the date of first dose to end of treatment (median duration of dasatinib treatment=5.2 months [range 0 to 33 months]).

Interventionparticipants (Number)
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg0
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg0
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg0

[back to top]

Number of Participants With Dose-limiting Toxicity (DLT)

DLTs: At least possibly drug-related AEs occurring during the first cycle of treatment and are:GR4 neutropenia >5 days/neutropenic fever;platelet count <10000mm^3 on >1 occasion;GR4 fatigue,or 2-point decline in ECOG performance status;>=GR3 nausea,diarrhea,and vomiting despite medical intervention;Any other clinically significant non-hematologic toxicity of >=GR3 considered not related to underlying MM;Any GR3/4 laboratory abnormality requiring hospitalization;dose interruption of either dasatinib and/or lenalidomide for >15 days due to any toxicity related to treatment with the combination. (NCT00560391)
Timeframe: From the date of first dose until at least 30 days after the last dose of study drug (median duration of dasatinib treatment=5.2 months [range 0 to 33 months]).

Interventionparticipants (Number)
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg1
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg1
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg0

[back to top]

Number of Participants With Minimal Response

Response criteria was based on The International Uniform Response Criteria for Multiple Myeloma (with a slight modification). Minimal Response was achieved when there was 25% to 49% reduction of serum M-Protein, 50% to 89% reduction in 24 hour urinary M-protein which still exceeded 200 mg/24 hour. If the serum and urine M-protein were unmeasurable, 25% to 49% reduction in plasma cells was required. In addition, if present at baseline, a 25% to 49% reduction in the size of soft tissue plasmacytomas was also required. (NCT00560391)
Timeframe: Baseline, at the end of the treatment period (median duration of dasatinib treatment=5.2 months [range 0 to 33 months]).

Interventionparticipants (Number)
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg0
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg0
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg0

[back to top]

Number of Participants With Partial Response

Partial response was achieved when there was ≥50% reduction of serum M-protein (Mpr)and reduction in 24 hour urinary Mpr by ≥90% or to <200 mg/24 hr. If the serum and urine Mpr were unmeasurable, a ≥50% decrease in the difference between involved and uninvolved free light chain (FLC) levels was required in place of the Mpr criteria. If serum, urine Mpr, and serum FLC assay were unmeasurable, ≥50% reduction in plasma cells was required in place of Mpr, provided baseline bone marrow plasma cell percentage was ≥30%; a ≥50% reduction in the size of soft tissue plasmacytomas was also required. (NCT00560391)
Timeframe: Baseline, at the end of the treatment period (median duration of dasatinib treatment=5.2 months [range 0 to 33 months]).

Interventionparticipants (Number)
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg1
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg1
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg2
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg4
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg3

[back to top]

Number of Participants Who Died, Serious Adverse Events (SAEs), Adverse Events (AEs) and AEs Leading to Study Drug Discontinuation

AE: New untoward medical occurrence or worsening of a preexisting medical condition that does not have causal relationship with this treatment. SAE: Untoward medical event that at any dose: results in death, persistent or significant disability/incapacity, drug dependency/abuse; life-threatening, an important medical event, a congenital anomaly/birth defect; requires inpatient hospitalization/prolongs existing hospitalization. Grade 1= Mild; Grade 2= Moderate; Grade 3= Severe; Grade 4 = Life-threatening or disabling. (NCT00560391)
Timeframe: Baseline (pretreatment), from the date of first dose until at least 30 days after the last dose of study drug (median duration of dasatinib treatment=5.2 months [range 0 to 33 months]).

,,,,
Interventionparticipants (Number)
All deathsDeaths within 30 days of last doseAll SAEsDrug-related SAEsAEs leading to discontinuationDrug-related AEs leading to discontinuationAll AEsDrug-related AEs
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg00211033
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg10511066
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg3294871717
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg31312266
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg21211033

[back to top]

Number of Participants With Complete Response and Very Good Partial Response

Response criteria were based on The International Uniform Response Criteria for Multiple Myeloma (with a slight modification). Complete response was achieved when there was negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow. Very good partial response was achieved when serum and urine M-component was detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-component plus urine M-component < 100 mg per 24 hour. (NCT00560391)
Timeframe: Baseline, At the end of the treatment period (median duration of dasatinib treatment=5.2 months [range 0 to 33 months]).

,,,,
Interventionparticipants (Number)
Complete responseVery good partial responseNo responseNot reportedResponse Undetermined
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg00100
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg01100
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg031100
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg00410
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg00200

[back to top]

Number of Participants With Serum Chemistry Abnormalities (Worst On-study Grade vs Baseline): Aspartate Aminotransferase (AST), Alanine Aminotransferase (ALT), Total Bilirubin (TB), and Serum Creatinine (SC)

Grading as per NCI CTCAE Version 3.0 criteria. GR1=Mild; GR2=Moderate; GR3=Severe; GR4=Life-threatening or disabling. Aspartate aminotransferase (AST) and alanine aminotransferase(ALT): GR1=>ULN-2.5*ULN (upper limit of normal); GR2=>2.5-5.0*ULN; GR3=>5.0-20.0*ULN; GR4:>20.0*ULN; TB:GR1=>ULN-1.5*ULN, GR2=>1.5-3.0*ULN, GR3=>3-10*ULN, GR4=>10*ULN; SC: GR1=>ULN-1.5*ULN, GR2=>1.5-3.0*ULN, GR3=>3.0-6.0*ULN, GR4=>6.0*ULN. BL=Baseline; PBL=post baseline. (NCT00560391)
Timeframe: Baseline (pretreatment); Cycles 1 and 2 (within 24 hours of Days 1, 4, 8, 11, 15, 18, 21, and 25); beyond Cycle 2 (within 24 hours of Days 1 and 15,off treatment visit ) (median duration of dasatinib treatment=5.2 mo [range 0 to 33 mo])

,,,,
Interventionparticipants (Number)
AST; GR 0 at BL, GR 0 PBLAST; GR 0 at BL, GR 1 to 2 PBLAST; GR 0 at BL, GR 3 to 4 PBLAST; GR 1 to 2 at BL, GR 0 PBLAST; GR 1 to 2 at BL, GR 1 to 2 PBLAST; GR 1 to 2 at BL, GR 3 to 4 PBLHigh ALT; GR 0 at BL, GR 0 PBLALT; GR 0 at BL, GR 1 to 2 PBLALT; GR 0 at BL, GR 3 to 4 PBLALT; GR 1 to 2 at BL, GR 0 PBLALT; GR 1 to 2 at BL, GR 1 to 2 PBLALT; GR 1 to 2 at BL, GR 3 to 4 PBLTotal Bilirubin; GR 0 at BL, GR 0 PBLTotal Bilirubin; GR 0 at BL, GR 1 to 2 PBLTotal Bilirubin; GR 0 at BL, GR 3 to 4 PBLSC; GR 0 at BL, GR 0 PBLSC; GR 0 at BL, GR 1 to 2 PBLSC; GR 0 at BL, GR 3 to 4 PBLSC; GR 1 to 2 at BL, GR 0 PBLSC; GR 1 to 2 at BL, GR 1 to 2 PBLSC; GR 1 to 2 at BL, GR 3 to 4 PBL
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg200010101010210120000
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg500010230010420600000
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg58003159002113401231001
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg420000240000510420000
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg020010101010110110010

[back to top]

Number of Participants With Serum Chemistry Abnormalities (Worst On-study Grade vs Baseline): High Calcium, Low Calcium, Low Magnesium, and Low Phosphorus

Grading as per NCI CTCAE Version 3.0 criteria. GR1=Mild; GR2=Moderate; GR3=Severe; GR4=Life-threatening or disabling. Calcium (low): GR1: ULN - 11.5 mg/dL, GR2: >11.5 - 12.5 mg/dL, GR3: >12.5 - 13.5 mg/dL, GR4: >13.5 mg/dL. Magnesium (Low): GR1: NCT00560391)
Timeframe: Baseline (pretreatment); Cycles 1 and 2 (within 24 hours of Days 1, 4, 8, 11, 15, 18, 21, and 25); beyond Cycle 2 (within 24 hours of Days 1 and 15,off treatment visit ) (median duration of dasatinib treatment=5.2 mo [range 0 to 33 mo])

,,,,
Interventionparticipants (Number)
High Calcium; GR 0 at BL, GR 0PBLHigh Calcium; GR 0 at BL, GR 1 to 2 PBLHigh Calcium; GR 0 at BL, GR 3 to 4 PBLHigh Calcium; GR 1 to 2 at BL, GR 0 PBLHigh Calcium; GR 1 to 2 at BL, GR 1 to 2 PBLHigh Calcium; GR 1 to 2 at BL, GR 3 to 4 PBLLow Calcium; GR 0 at BL, GR 0 PBLLow Calcium; GR 0 at BL, GR 1 to 2 PBLLow Calcium; GR 0 at BL, GR 3 to 4 PBLLow Calcium; GR 1 to 2 at BL, GR 0 PBLLow Calcium; GR 1 to 2 at BL, GR 1 to 2 PBLLow Calcium; GR 1 to 2 at BL, GR 3 to 4 PBLLow Magnesium; GR 0 at BL, GR 0 PBLLow Magnesium; GR 0 at BL, GR 1 to 2 PBLLow Magnesium; GR 0 at BL, GR 3 to 4 PBLLow Magnesium; GR 1 to 2 at BL, GR 0 PBLLow Magnesium; GR 1 to 2 at BL, GR 1 to 2 PBLLow Magnesium; GR 1 to 2 at BL, GR 3 to 4 PBLLow Magnesium; GR Not reported at BLLow Phosphorus; GR 0 at BL, GR 0 PBLLow Phosphorus; GR 0 at BL, GR 1 to 2 PBLLow Phosphorus; GR 0 at BL, GR 3 to 4 PBLLow Phosphorus; GR 1 to 2 at BL, GR 0 PBLLow Phosphorus; GR 1 to 2 at BL, GR 1 to 2 PBLLow Phosphorus; GR 1 to 2 at BL, GR 3 to 4 PBLLow Phosphorus; GR 3 to 4 at BL, GR 0 PBLLow Phosphorus; GR 3 to 4 at BL, GR 1 to 2 PBLLow Phosphorus; GR 3 to 4 at BL, GR 3 to 4 PBL
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg3000001200003000000200000001
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg4000202400002300100321000000
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg151001011220117610300826001000
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg4100012300102300100131000000
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg2000100200101200000030000000

[back to top] [back to top]

Number of Participants With Hematology Abnormalities (Worst On-study Grade vs Baseline): Leukopenia, Neutropenia, Thrombocytopenia, and Anemia

As per NCI CTCAE Version 3.0 criteria. Grade (GR)1=Mild; GR2=Moderate; GR3=Severe; GR4=Life-threatening or disabling. White blood cell (WBC):GR1=NCT00560391)
Timeframe: Baseline (pretreatment); Cycles 1 and 2 (within 24 hours of Days 1, 4, 8, 11, 15, 18, 21, and 25); beyond Cycle 2 (within 24 hours of Days 1 and 15,off treatment visit ) (median duration of dasatinib treatment=5.2 mo [range 0 to 33 mo])

,,,,
Interventionparticipants (Number)
Leukopenia; GR 0 at BL, GR 0 PBLLeukopenia; GR 0 at BL, GR 1 to 2 PBLLeukopenia; GR 0 at BL, GR 3 to 4 PBLLeukopenia; GR 1 to 2 at BL, GR 0 PBLLeukopenia; GR 1 to 2 at BL, GR 1 to 2 PBLLeukopenia; GR 1 to 2 at BL, GR 3 to 4 PBLNeutropenia; GR 0 at BL, GR 0 PBLNeutropenia; GR 0 at BL, GR 1 to 2 PBLNeutropenia; GR 0 at BL, GR 3 to 4 PBLNeutropenia; GR 1 to 2 at BL, GR 0Neutropenia; GR 1 to 2 at BL, GR 1 to 2 PBLNeutropenia; GR 1 to 2 at BL, GR 3 to 4 PBLThrombocytopenia; GR 0 at BL, GR 0 PBLThrombocytopenia; GR 0 at BL, GR 1 to 2 PBLThrombocytopenia; GR 0 at BL, GR 3 to 4 PBLThrombocytopenia; GR 1 to 2 at BL, GR 0 PBLThrombocytopenia; GR 1 to 2 at BL, GR 1 to 2 PBLThrombocytopenia; GR 1 to 2 at BL, GR 3 to 4 PBLAnemia; GR 0 at BL, GR 0 PBLAnemia; GR 0 at BL, GR 1 to 2 PBLAnemia; GR 0 at BL, GR 3 to 4 PBLAnemia; GR 1 to 2 at BL, GR 0 PBLAnemia; GR 1 to 2 at BL, GR 1 to 2 PBLAnemia; GR 1 to 2 at BL, GR 3 to 4 PBLAnemia; GR 3 to 4 at BL, GR 0 PBLAnemia; GR 3 to 4 at BL, GR 1 to 2 PBLAnemia; GR 3 to 4 at BL, GR 3 to 4 PBLAnemia; GR Not reported at BL
Dasatinib 100 mg + Lenalidomide 20 mg + Dexamethasone 40 mg0300000210000200100100110000
Dasatinib 100 mg + Lenalidomide 25 mg + Dexamethasone 40 mg1200030300031300110300110001
Dasatinib 140 mg + Lenalidomide 25 mg + Dexamethasone 40 mg2630151380142520350610550000
Dasatinib 70 mg + Lenalidomide 15 mg + Dexamethasone 40 mg0120030230010300120000410010
Dasatinib 70 mg + Lenalidomide 20 mg + Dexamethasone 40 mg2100001110001100010100110000

[back to top]

Number of Patients With Organ Response

"Organ response was evaluated on the basis of improvement of one or more affected organ; only one parameter was required to satisfy the criteria. Response needed to be maintained for a minimum of 3 months to be considered valid.~Renal response required a 50% reduction in 24-hour urine protein excretion (at least 0.5 g/d) with stable creatinine. Cardiac response required one of >= 2-mm reduction in the interventricular septal (IVS) thickness by echocardiogram, or improvement of ejection fraction by >= 20%, or improvement by 2 NYHA classes without an increase in diuretic use. Hepatic response required either >= 50% decrease in (or normalization of) an initially elevated alkaline phosphatase level or reduction in the size of the liver by at least 2 cm by radiographic determination. Gastrointestinal tract improvement was defined as normalization of a low serum carotene level, or reduction of diarrhea to < 50% of previous movements/day, or decrease in fecal fat excretion by 50%." (NCT00564889)
Timeframe: Duration of study (up to 3 years)

Interventionparticipants (Number)
Len/Cyc/Dex11

[back to top]

Number of Participants Who Achieved a Confirmed Response Defined as a Complete Response (CR), Very Good Partial Response (VGPR) or Partial Response (PR)

"Response that was confirmed on 2 consecutive evaluations during treatment.~Complete Response(CR): Complete disappearance of M-protein from serum and urine on immunofixation, normalization of Free Light Chain (FLC) ratio and <5% plasma cells in bone marrow.~Very Good Partial Response(VGPR): >=90% reduction in serum M-component; Urine M-Component <=100 mg per 24 hours.~Partial Response(PR): >=50% reduction in serum M-component and/or Urine M-Component >=90% reduction or <200 mg per 24 hours; or >=50% decrease in difference between involved and uninvolved FLC levels." (NCT00564889)
Timeframe: Duration on study (up to 3 years)

Interventionparticipants (Number)
Len/Cyc/Dex21

[back to top]

Number of Participants With Severe Adverse Events

Severe adverse events were defined as grade 3 or higher, at least possibly related to study drugs. Adverse events were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 3. (NCT00564889)
Timeframe: Duration of study (up to 3 years)

Interventionparticipants (Number)
Len/Cyc/Dex26

[back to top]

Overall Survival (OS)

Overall survival (OS) was defined as the time from registration to death of any cause. Surviving patients were censored at the date of last follow-up. The median OS with 95% CI was estimated using the Kaplan Meier method. (NCT00564889)
Timeframe: Duration of study (up to 3 years)

Interventionmonths (Median)
Len/Cyc/Dex37.8

[back to top]

Progression Free Survival (PFS)

Progression free survival (PFS) was defined as the time from registration to hematologic progression or death of any cause. Progression free and alive patients were censored at the date of last follow-up. The median PFS with 95% CI was estimated using the Kaplan Meier method. (NCT00564889)
Timeframe: Duration of study (up to 3 years)

Interventionmonths (Median)
Len/Cyc/Dex28.3

[back to top]

Final Response Rate After 4 Courses of Treatment

(NCT00567229)
Timeframe: 2 years

Interventionparticipants (Number)
Stable Disease (SD)Progression of Disease (POD)
Lenalidomide and Rituximab21

[back to top]

Overall Response Rate in Patients Without Del(11q22.3)

Percentage of non-del(11q22.3) participants with a complete response (CR) or partial response (PR). CR: no lymphadenopathy, hepatomegaly, splenomegaly or constitutional symptoms; normal complete blood count; confirmed by bone marrow (BM) aspirate & biopsy PR: 50% decrease in peripheral blood lymphocytes, lymphadenopathy, liver/spleen size, presence/absence of constitutional symptoms; plus >= 1 of the following: >= 1500/uL polymorphonuclear leukocytes, > 100,000/uL platelets, > 11.0 g/dL hemoglobin or 50% improvement for these parameters without transfusions. (NCT00602459)
Timeframe: Up to 15 years

Interventionpercentage of participants (Number)
Arm A (Rituximab, Fludarabine Phosphate)75
Arm B (Rituximab, Fludarabine Phosphate, Lenalidomide)69
Arm C (Rituximab, Fludarabine Phosphate, Cyclophosphamide)71

[back to top]

Overall Response Rates in Patients With Del(11q22.3)

Percentage of del(11q22.3) participants with a complete response (CR) or partial response (PR). CR: no lymphadenopathy, hepatomegaly, splenomegaly or constitutional symptoms; normal complete blood count; confirmed by bone marrow (BM) aspirate & biopsy PR: 50% decrease in peripheral blood lymphocytes, lymphadenopathy, liver/spleen size, presence/absence of constitutional symptoms; plus >= 1 of the following: >= 1500/uL polymorphonuclear leukocytes, > 100,000/uL platelets, > 11.0 g/dL hemoglobin or 50% improvement for these parameters without transfusions. (NCT00602459)
Timeframe: Up to 15 years

Interventionpercentage of participants (Number)
Arm C (Rituximab, Fludarabine Phosphate, Cyclophosphamide)59
Arm D (Rituximab, Fludarabine, Cyclophosphamide, Lenalidomide)74

[back to top]

PFS Rate of Patients With Del(11q22.3)

Proportion of del (11q22.3) participants who were alive and progression free at 2 years. (NCT00602459)
Timeframe: 2 years

Interventionproportion of participants (Number)
Arm C2, FCR in Del(11q22.3)0.56
Arm D, FCR+L in Del(11q22.3)0.65

[back to top]

Time-to-progression in Patients With Del(11q22.3)

Time to progression (TTP) in del(11q22.3) participants was defined as the registration date to date of progression or death due to any cause, whichever occurs first. TTP was estimated using the Kaplan Meier method. (NCT00602459)
Timeframe: Up to 15 years

Interventionmonths (Median)
Arm C2, FCR in Del(11q22.3)35.5
Arm D, FCR+L in Del(11q22.3)44.6

[back to top]

Time-to-progression in Patients Without Del(11q22.3)

Time to progression (TTP) was defined as the registration date to date of progression or death due to any cause, whichever occurs first. TTP was estimated using the Kaplan Meier method. Progressive disease (PD) required at least one of the following: >= 50% increase in the absolute number of lymphocytes, appearance of new palpable lymph nodes, >= 50% increase in the product of at least two lymphnodes, >= 50% increase in the enlargement of the liver and/or spleen. (NCT00602459)
Timeframe: Up to 15 years

Interventionmonths (Median)
Arm A, FR in Non-del(11q22.3)43.5
Arm B, FR+L in Non-del(11q22.3)66.0
Arm C1, FCR in Non-del(11q22.3)78.0

[back to top]

2-Year Progression Free Survival (PFS) Rate

Proportion of participants who were alive and progression free at 2 years. (NCT00602459)
Timeframe: 2 years

Interventionproportion of participants (Number)
Arm A, FR in Non-del(11q22.3)0.64
Arm B, FR+L in Non-del(11q22.3)0.71
Arm C1, FCR in Non-del(11q22.3)0.74

[back to top]

Change in Functional Assessment of Cancer Therapy-Neurotoxicity Trial Outcome Index (FACT-Ntx TOI) Score From Baseline to Cycle 12

A combined scale was used to assess the quality of life (QOL) comprising of the well established and validated functional well-being (FWB) and physical well-being (PWB) components of FACT-G version 4 (14 questions), which will address the physical and functional well-being of multiple myeloma patients plus the FACT-neurotoxicity (NTX, 11 questions), which will evaluate symptoms of neurotoxicity. This pooled scale is referred to as the FACT Ntx TOI. The FACT-Ntx TOI has 25 items and the score ranges from 0 (worst possible outcome) to 100 (best possible outcome). (NCT00602641)
Timeframe: Administered at registration, the beginning of cycle 7 d1, the end of cycle 12 d28, then at the end of cycle 18, 24, and 38 d28. For patients who discontinue treatment early, assessed at time of discontinuation and at the next quarterly follow-up visit.

Interventionunits on a scale (Mean)
Arm I (MPT-T)-2.8
Arm II (mPR-R)3.3

[back to top]

Overall Survival

Overall survival was defined as time from randomization to death from any cause. (NCT00602641)
Timeframe: Assessed every 3 months for 2 years, then every 6 months for 3 years, then annually for 10 years from the date of randomization.

Interventionmonths (Median)
Arm I (MPT-T)52.6
Arm II (mPR-R)47.7

[back to top]

Very Good Partial Response (VGPR) Rate

Response evaluation was based on the International Myeloma Working Group (IMWG) response criteria. VGPR rate was defined as patients achieving at least VGPR which include patients who achieving complete response (CR) and VGPR. CR refers to patients who have complete disappearance of an M-protein and no evidence of myeloma in the bone marrow. VGPR refers to patients who meet the following criteria: Serum and urine M-component detectable by immunofixation but not on electrophoresis; Or 90% or greater reduction in serum M-component plus urine M-component <100 mg per 24 hours; If the serum and urine M protein are unmeasurable and the immunoglobulin free light chain parameter is being used to measure response, a ≥ 90% decrease in the difference between involved and uninvolved free light chain (FLC) levels is required in place of the M protein criteria. (NCT00602641)
Timeframe: Assessed every cycle (1 cycle=28 days) for the first 12 cycles, and then every 2 cycles while on treatment. Post treatment assessed every 3 months < 2 years from study entry, every 6 months if 2-5 years, every 12 months if 6-10 years from study entry.

Interventionproportion of participants (Number)
Arm I (MPT-T)0.247
Arm II (mPR-R)0.316

[back to top]

Progression-Free Survival (PFS)

PFS is defined as the time from randomization to the earlier of progression or death of any cause. (NCT00602641)
Timeframe: Assessed every 3 months for 2 years, then every 6 months for 3 years, then annually for 10 years from the date of randomization.

Interventionmonths (Median)
Arm I (MPT-T)21.0
Arm II (mPR-R)18.7

[back to top]

Overall Survival (OS)

Overall Survival (OS) was defined as the time from registration to death of any cause. Participants were followed for a maximum of 5 years from registration. The median OS with 95% CI was estimated using the Kaplan Meier method. (NCT00602836)
Timeframe: time from registration to death (up to 5 years)

Interventionmonths (Median)
PCR-LenalidomideNA

[back to top]

Treatment Free Survival (TFS)

Treatment Free Survival (TFS) was defined as the time from registration to the earliest date documentation of subsequent treatment or death, whichever came first. Participants were followed for a maximum of 5 years from registration. The median TFS with 95% CI was estimated using the Kaplan Meier method. (NCT00602836)
Timeframe: time from registration to progression (up to 5 years)

Interventionmonths (Median)
PCR-LenalidomideNA

[back to top]

Number of Participants Who Convert From a CR With Minimal Residual Disease (MRD) Positive Status After PCR to a CR With MRD-negative Status After 6 Courses of Consolidation With Lenalidomide

MRD refers to small number of leukemic cells that remain in the participant during treatment or after treatment when the participant has achieved CR. For all participants who achieved CR, the follow-up bone marrow sample was tested for malignant B cells to determine if there was any MRD. (NCT00602836)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
PCR-Lenalidomide4

[back to top]

Number of Participants Who Convert From a Nodular Partial Response (nPR), Partial Response (PR), or Stable Disease (SD) After Pentostatin, Cyclophosphamide, and Rituximab (PCR) to a Complete Response (CR) After 6 Courses of Consolidation With Lenalidomide

"According to the NCIWG criteria, response is defined as follows:~nPR: Meets all criteria for CR, as described above, except the presence of residual clonal nodules in the bone marrow PR: 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 SD: participant who does not meet any of the criteria described above" (NCT00602836)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
PCR-Lenalidomide5

[back to top]

Number of Participants With a Response (CR, nPR, PR)

Response criteria described in above outcomes. (NCT00602836)
Timeframe: During treatment (up to 5 years)

InterventionParticipants (Count of Participants)
PCR-Lenalidomide38

[back to top]

Number of Participants With Complete Response (CR)

"A complete response, as defined by the National Cancer Institute Working Group (NCIWG), requires all of the following for a period of at least 2 months:~- CR: no lymphadenopathy, hepatomegaly, splenomegaly or constitutional symptoms; normal complete blood count; confirmed by bone marrow (BM) aspirate & biopsy" (NCT00602836)
Timeframe: 12 months

Interventionparticipants (Number)
PCR-Lenalidomide14

[back to top]

Number of Participants With Dose-limiting Toxicities

"Dose-limiting toxicity was defined as any of the following events assessed as related to carfilzomib, lenalidomide, or dexamethasone: Nonhematologic~≥ Grade 2 neuropathy with pain~≥ Grade 3 nonhematologic toxicity (excluding nausea, vomiting, diarrhea, hyperglycemia due to dexamethasone, and rash due to lenalidomide)~≥ Grade 3 nausea, vomiting, or diarrhea uncontrolled by maximal supportive therapy~≥ Grade 4 fatigue persisting > 7 days~Treatment delay for toxicity > 21 days~Hematologic~Grade 4 neutropenia (absolute neutrophil count [ANC] < 500/mm³) > 7 days~Febrile neutropenia (ANC < 1,000/mm³ with fever ≥ 38.3ºC)~Grade 4 thrombocytopenia (platelets < 25,000/mm³) for > 7 days despite holding treatment, or Grade 3 or 4 thrombocytopenia associated with bleeding~Treatment delay for toxicity > 21 days.~The maximum-tolerated dose was defined as the dose level below which a drug-related DLT was observed in ≥ 33% of participants in a cohort." (NCT00603447)
Timeframe: Cycle 1, 28 days

Interventionparticipants (Number)
1: CFZ 15 mg/m² + LEN 10 mg0
2: CFZ 15 mg/m² + LEN 15 mg0
3: CFZ 15 mg/m² + LEN 20 mg0
4: CFZ 20 mg/m² + LEN 20 mg0
5: CFZ 20 mg/m² + LEN 25 mg0
6: CFZ 20/27 mg/m² + LEN 25 mg1

[back to top]

Number of Participants With Adverse Events (AEs)

"Treatment-related are those AEs with possible or probable relationship to carfilzomib, lenalidomide or dexamethasone as assessed by the Investigator. The severity of each adverse event was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 3.0, per the following: Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death.~Serious adverse events were defined as AEs meeting one of the following: death, life-threatening, required or prolonged in-patient hospitalization, resulted in persistent or significant disability/incapacity, a congenital anomaly/birth defect in the offspring of an exposed participant, important medical events that may jeopardize the participant and may require medical or surgical intervention to prevent one of the outcomes listed above, or pregnancy or suspected pregnancy." (NCT00603447)
Timeframe: From the first dose of study drug until 30 days after the last dose; 1 to 52 months, with an average of 12 months.

,,,,,,
Interventionparticipants (Number)
Any adverse eventTreatment-related adverse eventGrade 3 or higher adverse eventTreatment-related Grade 3 or higher adverse eventSerious adverse eventAE leading to discontinuation of any study drugAE leading to discontinuation of carfilzomibDeaths within 30 days of last dose of study drug
1: CFZ 15 mg/m² + LEN 10 mg64513100
2: CFZ 15 mg/m² + LEN 15 mg65643100
3: CFZ 15 mg/m² + LEN 20 mg88874310
4: CFZ 20 mg/m² + LEN 20 mg64644420
5: CFZ 20 mg/m² + LEN 25 mg66663210
6: CFZ 20/27 mg/m² + LEN 25 mg88886310
7: CFZ 20/27 mg/m² + LEN 25 mg44434138221893

[back to top]

Clinical Benefit Rate

The ORR rate plus Stable Disease (SD). NCI-WG: SD is the absence of progressive disease (PD) and failure to achieve at least a PR; PD: at least one of the criteria of Group A or Group B has to be met. (NCT00609869)
Timeframe: Up to 6 years

Interventionpercentage of participants (Number)
Experimental: Lenalidomide and Rituximab83.3

[back to top]

Median Time to Treatment Failure (TTF)

The time from start of therapy to death, Progressive Disease (PD) or initiation of next therapy. PD: at least one of the NCI-WG criteria of Group A or Group B has to be met. (NCT00609869)
Timeframe: Up to 6 years

Interventionmonths (Median)
Experimental: Lenalidomide and Rituximab17.7

[back to top]

Overall Response Rate (ORR)

The sum of Complete Remission (CR) plus Partial Remission (PR) rates. Duration of overall response is measured from the time measurement criteria are met for CR or PR (whichever is first recorded) until the first date that recurrent or progressive disease is objectively documented, and must be confirmed greater than 8 weeks after first meeting CR or PR criteria. Response and progression for Chronic Lymphocytic Leukemia (CLL) were evaluated using 2008 updated National Cancer Institute-Sponsored Working Group Guidelines (NCI-WG) for Chronic Lymphocytic Leukemia. CR: all of the criteria must be met, and patients have the lack disease-related constitutional symptoms: PR: at least two of the criteria of Group A plus one of the criteria of group B have to be met. Group A Parameters: Lymphadenopathy; Hepatomegaly; Splenomegaly; Blood; Lymphocytes; Marrow. Group B Parameters: Platelet count; Hemoglobin; Neutrophils. (NCT00609869)
Timeframe: Up to 6 years

Interventionpercentage of participants (Number)
Experimental: Lenalidomide and Rituximab61

[back to top]

Median Progression Free Survival (PFS) in Months

PFS: Time from study entry to progression/relapse or death from study entry to death of any cause, assessed using International Myeloma Working Group Response Definitions. Progressive Disease (PD): One of the following criteria must be met: a. Increase of 25% or greater in serum M protein (absolute increase greater or equal to 0.5g/dl); b. Increase of 25% or greater in urine M protein (absolute increase greater than 200 mg/24h); c. Increase of 25% or greater in the difference between the involved and uninvolved free light chain (absolute increase greater than 10 mg/dl); d. Increase of 25% or greater in bone marrow plasma cell percentage (absolute percent greater than 5% in case the patient was in CR and 10% otherwise); i.e. Definite development of new bone lesions or soft tissue plasmacytomas, or increase in the size of existing plasmacytomas by greater or equal to 25%. Development of hypercalcemia (serum calcium > 11.5 mg/dl) attributable only to the plasma cell dyscrasia. (NCT00617591)
Timeframe: 24 Months

Interventionmonths (Median)
Induction and Maintenance Therapy28

[back to top]

Overall Response Rate (ORR) - Percentage of Participants With Partial Response or Better With Induction Regimen

ORR assessed using International Myeloma Working Group Response Definitions. Partial Remission (PR): A greater than 50% reduction in the serum paraprotein, and if present, a greater than 90% reduction in the urine M protein excretion. Patients must also have a decrease by 50% in the size of soft tissue plasmacytoma. If serum and urine M protein are not measurable, a 50% or greater decreased in the difference of the involved and uninvolved free light chain. Very Good Partial Remission (VGPR): Detectable serum and urine M component on immunofixation but not on electrophoresis or 90% or greater reduction in serum M protein with less than 100 mg/24 h of urinary M protein. Complete Remission (CR): The presence of less than 5% bone marrow plasmacytosis and the disappearance of all evidence of serum and urine M-components on electrophoresis as well as by immunofixation. In addition, soft tissue plasmacytoma must have disappeared. (NCT00617591)
Timeframe: 24 Months

Interventionpercentage of participants (Number)
Induction and Maintenance Therapy77.2

[back to top]

Percentage of Participants With Very Good Partial Remission (VGPR) or Better

Quality of response: % Complete Response (CR) + Very Good Partial Remission (VGPR) to induction Dd-R as assessed using International Myeloma Working Group Response Definitions. Very Good Partial Remission (VGPR): Detectable serum and urine M component on immunofixation but not on electrophoresis or 90% or greater reduction in serum M protein with less than 100 mg/24 h of urinary M protein. Complete Remission (CR): The presence of less than 5% bone marrow plasmacytosis and the disappearance of all evidence of serum and urine M-components on electrophoresis as well as by immunofixation. In addition, soft tissue plasmacytoma must have disappeared. (NCT00617591)
Timeframe: 24 Months

Interventionpercentage of participants (Number)
Induction and Maintenance Therapy42.1

[back to top]

Occurrence of Induction Toxicities

"Tolerability of full dose Revlimid® with full dose Doxil® in combination with reduced schedule dexamethasone was to be assessed during Cycle 1 and at the start of Cycle 2 using, whenever possible, the National Cancer Institute Common Terminology for Adverse Events (NCI CTCAE) v3.0.~Due to increased neutropenia and fatigue, toxicities were reviewed after the first 29 participants were enrolled." (NCT00617591)
Timeframe: 24 Months

Interventionpercentage of participants (Number)
Percentage with Dose Reductions RequiredPercentage Receiving < 4 Cycles of TherapyPercentage Who Discontinued After Only 1 CyclePercentage with Grade 3/4 NeutropeniaPercentage with Grade 3/4 Fatigue
Induction at Initial Full Dose242013.794820

[back to top]

2 Year Overall Survival (OS) Rate

Percentage of participants with Overall Survival in response to Dd-R in newly diagnosed multiple myeloma patients with active disease. Overall survival is time from study entry to death of any cause. (NCT00617591)
Timeframe: 24 Months

Interventionpercentage of participants (Number)
Induction and Maintenance Therapy79.6

[back to top]

Number of Patients Who Experience Improvement in GVHD on Lenalidomide, Defined as the Reduction in Severity of GVHD as Defined by the National Institutes of Health (NIH) Consensus Criteria

(NCT00619684)
Timeframe: Up to 9 years

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide)0

[back to top]

Response Rate, Defined as the Number of Patients Achieving Complete Response (CR), Partial Response (PR), or Minor Response (MR)

"CR: No Monoclonal Protein (MP) in the blood AND no serum/urine MP by Immunofixation (IF < 0) AND < 5% plasma cells in bone marrow aspirate.~VGPR: More than 90% decrease of MP and urine M protein < 100 mg/d OR serum protein electrophoresis (SPEP)/urine protein electrophoresis(UPEP) negative but serum immunofixation (IFs) or IFu urine immunofixation (IFu) ) still positive.~PR: Over 50% decrease of serum MP AND > 90% reduction in 24h urinary light chain excretion or M proteinuria < 200mg/d MR: Between 25 and 49% decrease of MP in the blood AND 50-89% reduction in 24h urinary light chain excretion (monoclonal proteinuria>200 mg/d)" (NCT00619684)
Timeframe: Up to 9 years

InterventionParticipants (Count of Participants)
Complete Response (CR)Very Good Partial Response (VGPR)Partial Response (PR)Minimal Response (MR)
Treatment (Lenalidomide)5231

[back to top]

Overall Survival

Kaplan-Meier estimate of survival (NCT00619684)
Timeframe: At 1 and 2 years after starting treatment with lenalidomide

Interventionpercentage of participants (Number)
Percent Overall Survival at 1 yearPercent Overall Survival at 2 years
Treatment (Lenalidomide)7158

[back to top]

Adverse Events, Graded According to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0

Grade 1-2 adverse events occurring in >10% of participants. Grade 3 or higher adverse events occurring in one or more participants. (NCT00619684)
Timeframe: Up to 30 days after completion of study treatment

Interventionpercentage of participants (Number)
Grade 1-2 constipationGrade 1-2 diarrheaGrade 1-2 fatigueGrade 1-2 myalgiaGrade 1-2 nauseaGrade 1-2 neuropathyGrade 1-2 thrombocytopeniaGrade 3 pneumoniaGrade 3 H1N1 influenzaGrade 3 fever, hypoxia and neuropathyGrade 3 myalgiaGrade 3 neuropathyGrade 3 neutropenia
Treatment (Lenalidomide)28171717111111171166644

[back to top]

TTP

"Time to Progression (TTP): Time from start of therapy to meeting the definition of Progressive Disease (PD).~PD: 25% increase compared to the lowest value of:~Serum MP (absolute increase at least ≥ 0.5 g/dl)~Or: Urine MP (absolute increase at least > 200 mg/24h)~Or: for patients without measurable MP, Serum Free Light Chain test: the difference between involved and uninvolved FLC levels (absolute increase at least >100 mg/L)" (NCT00619684)
Timeframe: Up to 9 years

InterventionMonths (Median)
Treatment (Lenalidomide)8.5

[back to top]

Number of Patients Requiring Dose Interruption, Dose Reduction or Discontinuance of Lenalidomide

Dose interruption, dose reduction or discontinuation of lenalidomide due to toxicity, GVHD or disease progression (NCT00619684)
Timeframe: Up to 9 years

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide)13

[back to top]

Number of Participants With Adverse Events (AE) During the Extension Phase

An AE is any sign, symptom, illness, or diagnosis (either observed or volunteered) that appears or worsens during the course of the study Serious adverse event (SAE) = any AE which results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect; constitutes an important medical event. A treatment emergent AE is defined as any AE occurring or worsening on or after the first dose of study drug and within 30 days after the last dose of study drug. Safety and severity was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 2.0; Severity of AEs were graded (including second primary malignancies) as Grade 1- Mild; Grade 2- Moderate; Grade 3- Severe; Grade 4- Life-threatening; Grade 5-Fatal; (NCT00622336)
Timeframe: From 22 Oct 2009 to November 2013; AEs/SAEs were recorded from informed consent to 30 days post treatment discontinuation visit.

Interventionparticipants (Number)
≥ 1 Adverse Event (AE)Adverse Event related to study drugAdverse Event NCI CTC (Version2.0) Grade 3 or 4≥ 1 Serious Adverse Event (SAE)≥ 1 AE leading to discontinuation of study drug≥ 1 AE leading to dose reduction/interruption
Lenalidomide 25mg (CC-5013)1315513

[back to top]

Number of Participants With Adverse Events (AE) During the Treatment Phase

An AE is any sign, symptom, illness, or diagnosis (either observed or volunteered) that appears or worsens during the course of the study Serious adverse event (SAE) = any AE which results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect; constitutes an important medical event. A treatment emergent AE is defined as any AE occurring or worsening on or after the first dose of study drug and within 30 days after the last dose of study drug. Safety and severity was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 2.0; Severity of AEs were graded (including second primary malignancies) as Grade 1- Mild; Grade 2- Moderate; Grade 3- Severe; Grade 4- Life-threatening; Grade 5-Fatal; (NCT00622336)
Timeframe: Until data cut-off of 22 Oct 2009; AEs/SAEs were recorded from informed consent to 30 days post treatment discontinuation visit. Maximum exposure to Lenalidomide treatment was 1260 days.

Interventionparticipants (Number)
Adverse Event (AE)Adverse Event related to study drugAdverse Event NCI CTC (Version2.0) Grade 3 or 4Serious Adverse Event (SAE)AE leading to discontinuation of study drugAE leading to dose reduction/interruption
Lenalidomide 25mg (CC-5013)32726825617752210

[back to top]

Time to Progression

Time from the start of study drug therapy to the first documentation of disease progression. Progressive disease characterized by at least one of the following: >50% increase in the sum of products of at least 2 lymph nodes on 2 consecutive examinations. At least one node larger than 2 cm. Or, appearance of new enlarged lymph nodes. >50% increase in size of liver and/or spleen as determined by physical examination or appearance of splenomegaly which was not previously present. >50% increase in number of circulating lymphocytes with absolute count of at least 10,000. (NCT00632359)
Timeframe: From baseline to 12 months

InterventionMonths (Mean)
Lenalidomide8

[back to top]

Response Assessments: Disease Status at the End of Lenalidomide Consolidation Per International Workshop on Chronic Lymphocytic Leukemia (IWCLL) Response Criteria

Complete remission (CR), requiring absence of peripheral blood clonal lymphocytes by immunophenotyping, absence of lymphadenopathy, absence of hepatomegaly or splenomegaly, absence of constitutional symptoms and satisfactory blood counts; positive or negative minimal residual disease (MRD); Partial remission (PR), defined as ≥ 50% fall in lymphocyte count, ≥ 50% reduction in lymphadenopathy or ≥ 50% reduction in liver or spleen, together with improvement in peripheral blood counts; Nodular Partial Remission (nPR) is CR with bone marrow nodules identified histologically. Progressive disease (PD), defined as ≥ 50% rise in lymphocyte count to > 5 x109/L, ≥ 50% increase in lymphadenopathy, ≥ 50% increase in liver or spleen size, Richter's transformation, or new cytopenias due to CLL; Stable disease, defined as not meeting criteria for CR, PR or PD. (NCT00632359)
Timeframe: 12 Months, Disease status assessed at the End of Lenalidomide Consolidation

Interventionparticipants (Number)
CR, MRD Positive or NegativePRnPRPD
Assessment at End of Consolidation13871

[back to top]

Improvement in Quality of Remission: Number of Participants With Response Versus No Response by NCI Working Group Criteria Disease Status Change at Start/End of Lenalidomide Consolidation

To evaluate ability of lenalidomide to improve quality of remission (e.g. from partial remission (PR) to complete remission (CR)), disease status assessed at start & end of Lenalidomide consolidation. NCI Working Group Response Criteria: Participants who began therapy in PR, improvement of status to nodular partial remission (nPR) or CR; Participants in nPR (otherwise CR, bone marrow nodules identified histologically), improvement of status to CR. Participants in CR, resolution of measurable disease in blood &/or bone marrow per immuno flow cytometry or PCR testing. Progressive Disease (PD): One or more of following: >50% increase in sum of products of =/>2 lymph nodes on 2 consecutive examinations; One+ node must be >2 cm or appearance of new enlarged lymph nodes; >50% increase in liver &/or spleen as determined by physical examination/appearance of splenomegaly not previously present; >50% increase in circulating lymphocytes with absolute count >10,000. (NCT00632359)
Timeframe: Baseline to 12 Months, Disease status assessed at Start/End of Lenalidomide Consolidation

Interventionparticipants (Number)
ResponseNo Response
Lenalidomide1021

[back to top]

Incidence of Non-Serious Adverse Events as a Measure of Safety and Tolerability, Phase II

"A count of affected participants with non-serious adverse events (regardless of relationship to study treatments) occurring in >= 15% of treated patients enrolled in the Phase II section of the study.~Lenalidomide DL-1 dose (10 mg orally, once daily (PO QD)) Day 1-14 followed by 7 days of rest, Rituximab 375 mg/m2 IV Days 1, 8, and 15 of Cycle 1; Cycles 2-6: 375 mg/m2 IV Day 1, Bortezomib 1.3 mg/m2 subcutaneous Days 1, 4, 8, and 11 for Cycles 1-6" (NCT00633594)
Timeframe: Collected from day of first dose to 30 days after the last dose of study medication, a maximum of 18 weeks and 30 days after last study treatment

Interventionparticipants (Number)
RashFatigueThrombocytopeniaLeukopeniaNauseaDiarrheaEdemaHyperglycemiaPeripheral NeuropathyNeutropeniaHypoalbuminemiaConstipationHypocalcemiaPain in ExtremityAnemiaCoughFeverDehydrationPruritusDyspneaHyponatremiaInsomniaAbdominal PainDizzinessHypokalemiaWeight LossAnorexiaErythemaHypomagnesemiaAllergic ReactionChillsHyperhidrosisMyalgiaHeadacheMucositisHypoglycemia
Phase II - Lenalidomide 10mg PO QD19181613121111111010109998887777666665555554444

[back to top]

Duration of Response (DoR) of Phase I and Phase II Participants

"Measured from the documented beginning of response (CR or PR) to the time of relapse. This is measured in responders per Non-Hodgkin's Lymphoma Revised Response Criteria for Malignant Lymphoma (Cheson et al. 2007.) CR: complete disappearance of detectable clinical evidence of disease and disease-related symptoms; PR: 50% or greater decrease in sum of product of diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses, no increase in size of other nodes, liver or spleen, no new disease sites, patients with CR and persistent morphologic bone marrow involvement.~Duration of Response will be examined using time-to-event analysis methods. Kaplan-Meier figures will be generated and the log-rank test will be used to examine differences existing between various levels of stratification." (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years or until documented disease progression

Interventionmonths (Median)
Phase I Participants (10 mg/15 mg Lenalidomide)25.72
Phase II Participants (10 mg Lenalidomide)17.81

[back to top]

Duration of Response (DoR) of Previously Treated and Previously Untreated Participants

Measured from the documented beginning of response (CR or PR) to the time of relapse. This is measured in responders per Non-Hodgkin's Lymphoma Revised Response Criteria for Malignant Lymphoma (Cheson et al. 2007.) CR: complete disappearance of detectable clinical evidence of disease and disease-related symptoms; PR: 50% or greater decrease in sum of product of diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses, no increase in size of other nodes, liver or spleen, no new disease sites, patients with CR and persistent morphologic bone marrow involvement. (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years or until documented disease progression

Interventionmonths (Median)
Previously Treated Participants17.94
Previously Untreated Participants21.09

[back to top]

Maximum Tolerated Dose of Lenalidomide Combined With Bortezomib and Rituximab in Phase I Participants

"Determination of the maximum tolerated dose (MTD) of lenalidomide combined with bortezomib and rituximab, defined as the highest dose at which ≤1 of 6 patients experiences a dose-limiting toxicity according to the NCI CTCAE v. 4.03.~MTD of Lenalidomide was tested, included with 1.3 mg/m2 subcutaneous (D1, 4, 8, 11) bortezomib, 375 mg/m2 (D1, 8, 15 of Cycle 1, D1 on subsequent cycles) rituximab.~Three dose limiting toxicities were reported in two patients (grade 4 neutropenia and grade 3 neuropathy, grade 3 rash)" (NCT00633594)
Timeframe: Collected from day of first dose to the end of the first treatment cycle, up to 21 days

Interventionmg lenalidomide, orally, daily, day 1-14 (Number)
Phase I Participants (10 mg/15 mg Lenalidomide)10

[back to top]

Overall Response Rate (ORR) of Phase I and Phase II Participants

Response to treatment (Complete Response (CR) or Partial Response (PR)) determined using Non-Hodgkin's Lymphoma Revised Response Criteria for Malignant Lymphoma (Cheson et al. 2007.) CR: complete disappearance of all detectable clinical evidence of disease and disease-related symptoms; PR: 50% or greater decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or extranodal masses, no increase in the size of other nodes, liver or spleen, no new sites of disease, patients who achieve CR but have persistent morphologic bone marrow involvement; Stable Disease (SD): failing to attain PR or CR, but not fulfilling criteria for progressive disease; Progressive Disease (PD)/Relapse: appearance of new lesions more than 1.5 cm in any axis, 50% or greater increase from nadir SPD of any previously involved sites, 50% or greater increase in the longest diameter of any single previously identified node or extranodal mass more than 1 cm in short axis. (NCT00633594)
Timeframe: Every 6 weeks until treatment discontinuation then every 3 months thereafter, projected average 24 months

InterventionParticipants (Count of Participants)
Phase I Participants (10 mg/15 mg Lenalidomide)12
Phase II Participants (10 mg Lenalidomide)21

[back to top]

Overall Response Rate (ORR) of Previously Treated and Previously Untreated Participants

Response to treatment (Complete Response (CR) or Partial Response (PR)) determined using Non-Hodgkin's Lymphoma Revised Response Criteria for Malignant Lymphoma (Cheson et al. 2007.) CR: complete disappearance of all detectable clinical evidence of disease and disease-related symptoms; PR: 50% or greater decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or extranodal masses, no increase in the size of other nodes, liver or spleen, no new sites of disease, patients who achieve CR but have persistent morphologic bone marrow involvement; Stable Disease (SD): failing to attain PR or CR, but not fulfilling criteria for progressive disease; Progressive Disease (PD)/Relapse: appearance of new lesions more than 1.5 cm in any axis, 50% or greater increase from nadir SPD of any previously involved sites, 50% or greater increase in the longest diameter of any single previously identified node or extranodal mass more than 1 cm in short axis. (NCT00633594)
Timeframe: Every 6 weeks until treatment discontinuation then every 3 months thereafter, projected average 24 months

InterventionParticipants (Count of Participants)
Previously Treated Participants8
Previously Untreated Participants25

[back to top]

Overall Survival of Phase I and Phase II Participants

"Defined as the date of study entry to the date of death.~Overall Survival will be examined using time-to-event analysis methods. Kaplan-Meier figures will be generated and the log-rank test will be used to examine differences existing between various levels of stratification" (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years, then every 6 months after documented disease progression

Interventionmonths (Median)
Phase I Participants (10 mg/15 mg Lenalidomide)51.45
Phase II Participants (10 mg Lenalidomide)35.35

[back to top]

Overall Survival of Previously Treated and Previously Untreated Participants

"Defined as the date of study entry to the date of death.~Overall Survival will be examined using time-to-event analysis methods. Kaplan-Meier figures will be generated and the log-rank test will be used to examine differences existing between various levels of stratification" (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years, then every 6 months after documented disease progression

Interventionmonths (Median)
Previously Treated Participants28.4189
Previously Untreated Participants71.2608

[back to top]

Time to Best Response of Previously Treated and Previously Untreated Participants

Measured from the time of study entry to the documented beginning of response (CR or PR). This is measured in responders per Non-Hodgkin's Lymphoma Revised Response Criteria for Malignant Lymphoma (Cheson et al. 2007.) CR: complete disappearance of detectable clinical evidence of disease and disease-related symptoms; PR: 50% or greater decrease in sum of product of diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses, no increase in size of other nodes, liver or spleen, no new disease sites, patients with CR and persistent morphologic bone marrow involvement. (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years

Interventionmonths (Median)
Previously Treated Participants2.04
Previously Untreated Participants2.37

[back to top]

Progression Free Survival (PFS) of Previously Treated and Previously Untreated Participants

"Defined as the time from entry onto study until lymphoma progression or death from any cause.~Progression Free Survival will be examined using time-to-event analysis methods. Kaplan-Meier figures will be generated and the log-rank test will be used to examine differences existing between various levels of stratification." (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years, then every 6 months after documented disease progression

Interventionmonths (Median)
Previously Treated Participants12.4517
Previously Untreated Participants25.2649

[back to top]

Time to Best Response of Phase I and Phase II Participants

"Measured from the time of study entry to the documented beginning of response (CR or PR). This is measured in responders per Non-Hodgkin's Lymphoma Revised Response Criteria for Malignant Lymphoma (Cheson et al. 2007.) CR: complete disappearance of detectable clinical evidence of disease and disease-related symptoms; PR: 50% or greater decrease in sum of product of diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses, no increase in size of other nodes, liver or spleen, no new disease sites, patients with CR and persistent morphologic bone marrow involvement.~Time to Best Response will be examined using time-to-event analysis methods. Kaplan-Meier figures will be generated and the log-rank test will be used to examine differences existing between various levels of stratification." (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years

Interventiondays (Median)
Phase I Participants (10 mg/15 mg Lenalidomide)63.50
Phase II Participants (10 mg Lenalidomide)71.50

[back to top]

Progression Free Survival (PFS) of Phase I and Phase II Participants

"Defined as the time from entry onto study until lymphoma progression or death from any cause.~Progression Free Survival will be examined using time-to-event analysis methods. Kaplan-Meier figures will be generated and the log-rank test will be used to examine differences existing between various levels of stratification." (NCT00633594)
Timeframe: Every 3 months (+/- 2 weeks) after discontinuation of study treatment for 2 years, then every 6 months after documented disease progression

Interventionmonths (Median)
Phase I Participants (10 mg/15 mg Lenalidomide)27.70
Phase II Participants (10 mg Lenalidomide)19.35

[back to top]

Skindex Function

"Skindex function scores are scored 1-5 and then normalized to 100, with a higher score corresponding to a worse impression. The absence of function impact on QoL is scored as 20 (this is represented as a 1 on the 1-5 scale. The worst impact of function on QoL is 100. This is represented as a 5 on the 1-5 scale." (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide41.2630.4229.24.2

[back to top]

Patient General Assessment (PtGA) for Skin

General skin scores were recorded by the patient on a 10 cm visual analogue scale at each visit by the patient. At each visit on scale 0-10, 0 corresponding to worst skin condition imaginable and 10 to perfect health. (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide5.63.64.20.8

[back to top]

Pain in Skin

Pain in skin was recorded on a 10 cm visual analogue scale at each visit by the patient. At each visit on scale 0-10, 0 corresponding to no pain and 10 to pain as bad as you can imagine. (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide4.522.50.5

[back to top]

Number of Participants With Change in IFN and CD4 Levels at 6 Weeks

CXCL10, an interferon-inducible chemokine, and immunophenotyping by immunostaining. Measurement of interferon-inducible genes from peripheral blood mononuclear cells before and after treatment. (NCT00633945)
Timeframe: 6 weeks

,
InterventionParticipants (Count of Participants)
IFN score decreaseCD4 decrease
Lenalidomide Nonresponder11
Lenalidomide Responders44

[back to top]

Fatigue

Fatigue scores were recorded on a 10 cm visual analogue scale at each visit by the patient. At each visit on scale 0-10, 0 corresponding to no fatigure and 10 to fatigue as bad as you can imagine. (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide7.253.45.253

[back to top]

Cutaneous Lupus Area and Severity Index (CLASI)

The Cutaneous Lupus Area and Severity Index (CLASI); range of disease activity is 0-70. Lower scores reflect less activity. (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
0 weeks6 weeks12 weeks52 weeks
Lenalidomide21.410.88.65.3

[back to top]

Itch in Skin

Itch scores were recorded on a 10 cm visual analogue scale at each visit by the patient. At each visit on scale 0-10, 0 corresponding to no itch and 10 to itch as bad as you can imagine. (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide4.01.22.250

[back to top]

Skindex Symptoms

"Skindex symptoms scores are scored 1-5 and then normalized to 100, with a higher score corresponding to a worse impression. The absence of symptom impact on QoL is scored as 20 (this is represented as a 1 on the 1-5 scale. The worst impact of symptoms on QoL is 100. This is represented as a 5 on the 1-5 scale." (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide58.541.8236.412.5

[back to top]

Physician Global Assessment (PGA) for Skin

General skin scores were recorded on a 10 cm visual analogue scale at each visit. At each visit on scale 0-10, 0 corresponding to worst skin condition imaginable and 10 to perfect health. (NCT00633945)
Timeframe: Weeks 0 through 52

Interventionunits on a scale (Mean)
week 0week 6week 12week 52
Lenalidomide633.82

[back to top]

Number of Participants Experiencing Best Overall Response Determined by Complete Response (CR), Near Complete Response (NCR), Very Good Partial Response (VGPR), Partial Response (PR), Minimal Response (MR), Stable Disease (SD) or Progressive Disease (PD)

Best overall tumor response consisted of CR (negative M-protein immunofixation [IF], <5% bone marrow [BM] plasma cells, no soft tissue plasmacytomas [STP]), NCR (positive M-protein IF, <5% BM plasma cells, no STP), VGPR (≥90% M-protein decrease, <5% BM plasma cells, no STP), PR (≥50% M-protein, BM plasma cells, STP decrease), MR (25-49% M-protein, BM plasma cells, STP decrease), SD (<25% decrease-25% increase in M-protein, BM plasma cells; <25% decrease-increase in definite STP) or PD (≥25% M-protein, BM plasma cells increase; new STP, lesions). Per protocol participants experiencing best overall response by CR, NCR, VGPR, PR, MR, SD or PD were analyzed as a single prespecified analysis at the time of protocol-specified final statistical analysis with a 3-Sep-2012 data cut-off. The number of participants experiencing best overall response by CR, NCR, VGPR, PR, MR, SD or PD is reported here for all participants who got ≥1 dose of study drug and had a post baseline efficacy assessment. (NCT00642954)
Timeframe: Up to ~55 months (through pre-specified final statistical analysis cut-off date of 3-Sep-2012)

,,,,
InterventionParticipants (Count of Participants)
CRNCRVGPRPRMRSDPD
Level 1: Vorinostat 300 mg + Lenalidomide 10 mg0011011
Level 2: Vorinostat 400 mg + Lenalidomide 10 mg0101011
Level 3: Vorinostat 400 mg + Lenalidomide 15 mg0000111
Level 4: Vorinostat 400 mg + Lenalidomide 20 mg0011001
Level 5: Vorinostat 400 mg + Lenalidomide 25 mg1025251

[back to top] [back to top]

Number of Participants Experiencing Dose-Limiting Toxicities (DLTs)

DLTs consisted of hematologic or non-hemtologic toxicities. Hematologic DLT was any grade (gr) 4 neutropenia lasting ≥7 days, gr 4 thrombocytopenia or gr 5 hematologic toxicity. Non-hematologic DLT was any gr 3, 4 or 5 non-hematologic toxicity with the exception of (1) gr 3 nausea, vomiting, diarrhea or dehydration not compliant with medical care, lasting <48 hours (2) gr 3 acidosis/alkalosis returning to ≤ gr 2 by 48 hours of medical care (3) gr 3 liver function test elevation without symptoms, lasting ≤5 days (4) Gr 3 amylase elevation without symptoms (5) Gr 3 hypocalcemia, hypokalemia, hypomagnesemia, hyponatremia or hyphosphatemia responding to medical care (6) Gr 3 hypercholeresterolemia or hypertriglyceridemia. A drug-related AE causing dose modification of study drug is also considered a DLT. Per protocol DLTs were analyzed in the first 28-day treatment cycle. The number of participants experiencing DLTs is reported here for all participants who got ≥1 dose of study drug. (NCT00642954)
Timeframe: Cycle 1 (Up to 28 days)

InterventionParticipants (Count of Participants)
Level 1: Vorinostat 300 mg + Lenalidomide 10 mg0
Level 2: Vorinostat 400 mg + Lenalidomide 10 mg0
Level 3: Vorinostat 400 mg + Lenalidomide 15 mg0
Level 4: Vorinostat 400 mg + Lenalidomide 20 mg0
Level 5: Vorinostat 400 mg + Lenalidomide 25 mg1

[back to top]

Response Rates ()

The response rate was calculated as the number of patients with documented confirmed partial response (PR) or better, which includes confirmed/unconfirmed stringent complete response (sCR), confirmed/unconfirmed complete response (CR), confirmed/unconfirmed very good partial response (VGPR), or confirmed partial response (PR), as best response divided by the total number of evaluable patients, in each arm. Patients with measurable disease, as defined in the protocol, are evaluable. Response rates were compared between the two treatment arms using a stratified Cochran-Mantel-Haenszel test. Response designations were based on the International Uniform Response Criteria for Multiple Myeloma. Due to the complexity of these criteria, the details of these criteria have been omitted. (NCT00644228)
Timeframe: Up to 6 years

InterventionParticipants (Count of Participants)
Arm I (Dexamethasone and Lenalidomide)153
Arm II (Dexamethasone, Lenalidomide, Bortezomib)176

[back to top]

Progression-free Survival

Unstratified median progression-free survival in months. (NCT00644228)
Timeframe: From date of registration to date of first documentation of progression or symptomatic deterioration, or death due to any cause, assessed up to 6 years

InterventionMonths (Median)
Arm I (Dexamethasone and Lenalidomide)30
Arm II (Dexamethasone, Lenalidomide, Bortezomib)43

[back to top]

Overall Survival

Unstratified median overall survival in months. (NCT00644228)
Timeframe: Up to 6 years

InterventionMonths (Median)
Arm I (Dexamethasone and Lenalidomide)64
Arm II (Dexamethasone, Lenalidomide, Bortezomib)75

[back to top]

Number of Participants With Overall Clinical Benefit (OCB), Defined as the Sum of Complete Response (CR), Partial Response (PR), and Stable Disease (SD) Divided by the Number of Participants

"The OCB was assessed using Recist 1.0 as defined in the protocol. A CR was defined as the disappearance of all lesions. A PR was defined as > or equal to a 30% decrease in the sum of the longest diameter of measureable lesions, SD was defined < a 30% decrease in the sum of the longest diameter of measureable lesions and < a 20% increase in the sum of the longest diameter of measureable lesions. For a CR, PR or SD, there are no new lesions.~Prostate-Specific Antigen (PSA) was also evaluated. A PSA CR was a PSA < or equal to 4 ng/dl. A PSA PR was a PSA that decreased by > or equal to 50%. Stable PSA was defined as a PSA that increased >25% and decreased < 50%." (NCT00654186)
Timeframe: 24 months for acrual

Interventionpercentage of patients (Number)
Revlimid Oral for 21days74

[back to top]

Time to PSA Progression

As defined in the protocol PSA progression was an increase of at least 25% (NCT00654186)
Timeframe: 24 months for acrual

Interventionmonths (Median)
Revlimid Oral for 21days3

[back to top]

Time to Disesase Progression as Measured by Radiographic Progression

Progressive disease (PD) was determined, as outlined in the protocol, by using Recist 1.0. PD is defined as greater than or equal to a 20% increase in the sum of all measureable lesions or the apprearance of two new bone lesions or the appearnce of one new soft tissue lesion. (NCT00654186)
Timeframe: 24 months

Interventionmonths (Median)
Revlimid Oral for 21days4

[back to top]

Progression-Free Survival

Kaplan-Meier estimate of progression-free survival is defined as the start of study drug therapy to the first observation of disease progression or death due to any cause. (NCT00655668)
Timeframe: Up to 24 months

InterventionMonths (Median)
Single Agent Lenalidomide2.53

[back to top]

Duration of Response

Kaplan-Meier Estimate of duration of response calculated as the time from first computed tomography (CT) Scan or magnetic resonance imaging (MRI) that demonstrates at least a partial response to the first documentation of disease progression, including death due to Non-Hodgkin's Lymphoma. (NCT00655668)
Timeframe: Up to 24 months

InterventionMonths (Median)
Single Agent Lenalidomide3.55

[back to top]

Participants Categorized by Best Response as Determined by Investigator

"Participant response assessed by investigator; criteria by B. Cheson in Journal of Clinical Oncology, 1999 (see article for more detail):~Complete Response(CR): Complete disappearance of all detectable disease~Complete Response Unconfirmed(CRu): CR, but indeterminate bone marrow~Partial Response(PR): >50% decrease in six largest nodes/nodal masses~Stable Disease(SD): Less than PR, but not progressive disease~Relapsed Disease: In CR/CRu Patients, new lesions seen or increased by >=50% in previous sites~Progressive Disease(PD): >=50% increase from low in PR/Non-Responders" (NCT00655668)
Timeframe: Up to 24 months

InterventionParticipants (Number)
Complete Response (CR)Complete Response Unconfirmed (CRu)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)No Response AssessmentOtherTumor Control (CR+CRu+PR+SD)
Single Agent Lenalidomide42616169128

[back to top]

Safety

Summary of Treatment-Emergent Events in Safety Population (participants with at least one dose of study drug). Events assessed using National Cancer Institute, Common Terminology Criteria for Adverse Events (NCI CTCAE, Version 3: Following is the scale: Grade 1=Mild Adverse Event (AE), Grade 2=Moderate AE, Grade 3=Severe and Undesirable AE, Grade 4=Life-threatening or Disabling AE, and Grade 5=Death Related to AE.) (NCT00655668)
Timeframe: Up to 24 months

InterventionParticipants (Number)
At least 1 adverse event (AE)At least 1 AE related to drugAt least 1 NCI CTCAE Grade 3-4 AEAt least 1 NCI CTCAE Gr 3-4 AE related to drugAt least 1 serious adverse event (SAE)At least 1 SAE related to drugAt least 1 AE leading to drug withdrawal (WD)At least 1 AE leading to drug interruption/WD
Single Agent Lenalidomide5340341929162119

[back to top]

Change in Total Neuropathy Score in Subjects With MGUS Associated Neuropathy After Treatment With Lenalidomide.

"Minimum value of the Total Neuropathy Score is 0, maximum value is 40. A higher score represents a worsening." (NCT00665652)
Timeframe: Baseline, 12 months

Interventionunits on a scale (Mean)
Lenalidomide0.17

[back to top]

Event-free > Survival at 12 Months (Phase 2, DLBCL/Mixed Dose Level 3)

Other Phase II Cohorts were not evaluable for event-free survival analysis. (NCT00670358)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Ph II, DLBCL/Mixed44

[back to top]

Progression-free > Survival at 24 Months (Phase 2, Transformed/Composite)

"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~Other Phase II Cohorts were not evaluable for progression-free survival analysis." (NCT00670358)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Ph II, Transformed/Composite27

[back to top]

Toxicity as Assessed by NCI CTCAE v3.0 (Phase I)

(NCT00670358)
Timeframe: 5 years

,,
InterventionParticipants (Count of Participants)
Grade 3+ Adverse EventGrade 4+ Adverse EventGrade 3+ Hem Adverse EventGrade 4+ Hem Adverse EventGrade 3+ Non-Hem Adverse Event
Ph 1, DL 132321
Ph1, DL 231311
Ph1, DL 354542

[back to top]

Number of Participants' With Treatment Response of Complete or Partial Response

Treatment responses defined as complete (CR) or partial organ response (PR) of chronic Graft-Versus-Host Disease (GVHD) to lenalidomide. Complete organ response (CR) indicates resolution of all reversible manifestations related to chronic GVHD in a specific organ. Partial organ response (PR) requires at least 50% improvement in scale used to measure disease manifestations related to chronic GVHD. Tools for response evaluation were skin assessment and functional assessment including minute walk and grip strength. (NCT00675441)
Timeframe: Response assessed after completing 28 day cycle, repeated with each cycle for 6 cycles, approximately 180 days.

Interventionparticipants (Number)
Lenalidomide2

[back to top]

Number of Participants With Hematologic Response

"Complete hematologic response: Absence of detectable monoclonal protein in serum or urine by immunofixation electrophoresis, bone marrow biopsy with less than 5% plasma cells without clonal dominance of kappa or lambda isotype, and normal serum free light chain assay.~Partial hematologic response: Amyloid patients have highly individualized measures of disease burden. For patients with detectable and quantifiable monoclonal marrow plasmacytosis, a reduction of 50% or more in plasma cells as a percentage of nucleated bone marrow cells. For patients with a detectable monoclonal peak on serum or urine protein electrophoresis, a reduction in the peak height of 50% or more. For patients with quantifiable urinary kappa or lambda chain concentration, a 50% reduction in daily light chain excretion (concentration x 24 hour urine volume). For patients with an elevated serum free light chain assay, reduction of 50% or more." (NCT00679367)
Timeframe: one year

Interventionparticipants (Number)
Melphalan Revlimid and Dexamethasone7

[back to top]

Number of Participants Removed From Study Due to Toxicities

Number of study participants removed from study treatment due to toxicities (NCT00679367)
Timeframe: One year

InterventionParticipants (Count of Participants)
Melphalan Revlimid and Dexamethasone6

[back to top]

Number of Organs Improved or Stable Based on Description Below:

"Renal response - > 50% decrease in daily 24 hour proteinuria, without worsening renal insufficiency.~Hepatic response - decrease of 2 centimeters or more of the liver span and/or decrease of the alkaline phosphatase by 50% if elevated at baseline.~Cardiac response - decrease of 2 millimeters or more in mean left ventricular wall thickness in patients with baseline wall thickness > 11 mm or a decrease in New York Heart Association heart failure class.~Autonomic nervous system response - resolution of orthostatic vital signs and symptoms, and resolution of symptoms of gastric atony or of functional ileus.~Gastrointestinal response - a greater than one grade improvement in diarrhea due to biopsy proven amyloid.~Peripheral nervous system response - resolution of clinical signs of peripheral neuropathy." (NCT00679367)
Timeframe: one year

Interventionnumber of organs stable or improved (Number)
Melphalan Revlimid and Dexamethasone10

[back to top]

Change in Cancer Pain Intensity Determined by Edmonton Symptom Assessment Scale (ESAS)

"Changes in cancer pain from baseline to day 15 using ESAS to measure participant responses to 10 common symptoms (pain, fatigue, nausea, depression, anxiety, drowsiness, shortness of breath, appetite, sleep problems, and feeling of well-being). Intensity of symptoms rated on a 0 to 10 scale from 0 no symptom to 10 worst possible symptom." (NCT00684242)
Timeframe: From baseline to Day 15

Interventionunits on a scale (Number)
Participant 1Participant 2
Lenalidomide0-2

[back to top]

Number of Participants With a Grade 3 and 4 Adverse Event (Phase I)

"Adverse events were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 3.~Description of Grades:~Grade 1: Mild Grade 2: Moderate Grade 3: Severe Grade 4: Life-threatening Grade 5: Death" (NCT00687674)
Timeframe: up to 3 years

Interventionparticipants (Number)
Sorafenib + Lenalidomide + Dexamethasone11

[back to top]

Change From Baseline in the EORTC QLQ-C30 Constipation Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Constipation Scale is scored between 0 and 100, with a high score indicating a higher level of constipation. Negative change from Baseline values indicate improvement in constipation and positive values indicate worsening of constipation. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd186.30.0-5.1-5.2-5.9-7.5
Lenalidomide and Low-Dose Dexamethasone (Rd)8.31.8-2.4-2.4-4.5-7.9
Melphalan + Prednisone + Thalidomide (MPT)18.413.96.83.70.0-2.2

[back to top]

Change From Baseline in the EORTC QLQ-C30 Diarrhea Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Diarrhea Scale is scored between 0 and 100, with a high score indicating a higher level of diarrhea. Negative change from Baseline values indicate improvement in diarrhea and positive values indicate worsening of diarrhea. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd182.33.46.09.110.96.4
Lenalidomide and Low-Dose Dexamethasone (Rd)3.83.78.211.814.810.8
Melphalan + Prednisone + Thalidomide (MPT)-0.6-2.4-2.2-2.5-1.7-0.5

[back to top]

Kaplan Meier Estimates for Time to Second-line Anti-myeloma Treatment (AMT)

Time to second-line anti-myeloma therapy was defined as time from randomization to the start of another non-protocol anti-myeloma therapy. (NCT00689936)
Timeframe: From date of randomization until the data cut-off of 24 May 2013; median follow-up for all participants was 23.0 months

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)39.1
Lenalidomide and Dexamethasone Rd1828.5
Melphalan + Prednisone + Thalidomide (MPT)26.7

[back to top]

Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Emotional Functioning Scale is scored between 0 and 100, with a high score indicating better functioning/support. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd180.13.95.84.93.13.7
Lenalidomide and Low-Dose Dexamethasone (Rd)0.63.84.64.65.82.6
Melphalan + Prednisone + Thalidomide (MPT)1.02.15.55.15.1-0.0

[back to top]

Change From Baseline in the EORTC QLQ-C30 Fatigue Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Fatigue Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate improvement in symptoms and positive values indicate worsening symptoms. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd184.4-3.4-5.9-2.30.1-1.6
Lenalidomide and Low-Dose Dexamethasone (Rd)2.6-2.5-3.7-4.3-3.10.3
Melphalan + Prednisone + Thalidomide (MPT)2.8-1.8-4.5-3.9-4.32.7

[back to top]

Change From Baseline in the EORTC QLQ-C30 Financial Difficulties Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Financial Difficulties Scale is scored between 0 and 100, with a high score indicating a higher level of financial difficulties. Negative change from Baseline values indicate improvement in financial difficulties and positive values indicate worsening of financial difficulties. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-0.3-0.4-0.31.61.80.5
Lenalidomide and Low-Dose Dexamethasone (Rd)2.11.91.40.42.01.9
Melphalan + Prednisone + Thalidomide (MPT)0.51.90.71.10.45.0

[back to top]

Change From Baseline in the EORTC QLQ-C30 Insomnia Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Insomnia Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate improvement in symptoms and positive values indicate worsening symptoms. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd183.2-1.3-1.91.11.4-1.6
Lenalidomide and Low-Dose Dexamethasone (Rd)2.10.2-1.2-1.0-0.5-5.2
Melphalan + Prednisone + Thalidomide (MPT)-10.5-8.9-11.6-9.6-6.0-4.5

[back to top]

Change From Baseline in the EORTC QLQ-C30 Nausea/Vomiting Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Nausea/Vomiting Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate improvement in symptoms and positive values indicate worsening symptoms. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-0.5-2.5-4.0-3.6-2.7-4.2
Lenalidomide and Low-Dose Dexamethasone (Rd)1.8-1.1-1.3-2.2-2.30.4
Melphalan + Prednisone + Thalidomide (MPT)4.0-1.2-3.9-3.9-3.91.0

[back to top]

Change From Baseline in the EORTC QLQ-C30 Pain Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Pain Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate improvement in symptoms and positive values indicate worsening symptoms. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-4.4-13.1-16.1-14.7-12.4-7.9
Lenalidomide and Low-Dose Dexamethasone (Rd)-5.4-13.4-14.4-14.0-14.4-8.0
Melphalan + Prednisone + Thalidomide (MPT)-7.8-12.1-13.4-14.3-14.7-6.0

[back to top]

Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Physical Functioning Scale is scored between 0 and 100, with a high score indicating better functioning/support. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-1.44.77.67.46.83.0
Lenalidomide and Low-Dose Dexamethasone (Rd)-1.73.44.75.06.9-0.1
Melphalan + Prednisone + Thalidomide (MPT)-0.92.25.36.98.3-0.1

[back to top]

Time to First Response Based on the Review by the IRAC

The time to first myeloma response was defined as the time from randomization to the time when the response criteria for at least a PR was first met based on the IMWG criteria. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)1.8
Lenalidomide and Dexamethasone Rd181.8
Melphalan + Prednisone + Thalidomide (MPT)2.8

[back to top]

Change From Baseline in the EORTC QLQ-C30 Dyspnea Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Dyspnoea Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate improvement in symptoms and positive values indicate worsening symptoms. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd183.6-1.9-2.9-1.62.90.8
Lenalidomide and Low-Dose Dexamethasone (Rd)0.9-0.8-2.3-3.5-1.8-1.0
Melphalan + Prednisone + Thalidomide (MPT)4.22.00.1-1.60.47.8

[back to top]

Change From Baseline in the EORTC QLQ-C30 Social Functioning Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Social Functioning Scale is scored between 0 and 100, with a high score indicating better functioning/support. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-2.22.05.23.83.22.7
Lenalidomide and Low-Dose Dexamethasone (Rd)-4.30.74.02.94.2-1.2
Melphalan + Prednisone + Thalidomide (MPT)-1.42.43.45.86.0-3.5

[back to top]

Percentage of Participants With an Objective Response After Second-line Anti-myeloma Treatment at the Time of Final Analysis

Objective response according to IMWG Uniform Response Criteria was defined as a best overall response including a complete response (CR), very good partial response (VGPR) or partial response (PR) based on the IRAC Review. A CR is defined s: negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPR is serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is: ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment; data cut-off date of 21 January 2016; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionpercentage of participants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)46.2
Lenalidomide and Dexamethasone Rd1853.1
Melphalan + Prednisone + Thalidomide (MPT)45.7

[back to top]

Percentage of Participants With a Myeloma Response by Uncertain Risk Cytogenetic Risk Category Based on IRAC Review

Participants were placed in adverse and non-adverse cytogenetic risk categories at baseline and response rates evaluated. Adverse Risk: t(4;14), t(14;16), del(13q) or monosomy 13, del(17p), 1q gain Favorable Hyperdiploidy: : t(11;14), gains of 5/9/15; Normal: a normal result, gains other than 5/9/15, IgH deletion Uncertain risk: probes used for analysis cannot place participant in any of the other risk categories. Objective response = best overall response including CR, VGPR or PR based on the IRAC Review; A CR is negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPRis serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionpercentage of participants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)60.5
Lenalidomide and Dexamethasone Rd1876.8
Melphalan + Prednisone + Thalidomide (MPT)57.5

[back to top]

Percentage of Participants With a Myeloma Response by Normal Risk Cytogenetic Risk Category Based on IRAC Review

Participants were placed in adverse and non-adverse cytogenetic risk categories at baseline and response rates evaluated. Adverse Risk: t(4;14), t(14;16), del(13q) or monosomy 13, del(17p), 1q gain Favorable Hyperdiploidy: : t(11;14), gains of 5/9/15; Normal: a normal result, gains other than 5/9/15, IgH deletion Uncertain risk: probes used for analysis cannot place participant in any of the other risk categories. Objective response = best overall response including CR, VGPR or PR based on the IRAC Review; A CR is negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPRis serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionpercentage of particpants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)80.4
Lenalidomide and Dexamethasone Rd1874.8
Melphalan + Prednisone + Thalidomide (MPT)61.0

[back to top]

Kaplan Meier Estimates of Duration of Myeloma Response as Determined by an Investigator Assessment at Time of Final Analysis

Duration of response was defined as the duration from the time when the response criteria were first met for CR or VGPR or PR based on IMWG criteria until the first date the response criteria were met for progressive disease or until the participant died from any cause, whichever occurred first. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment; data cut-off date of 21 January 2016; median follow-up for responders was 19.9 months

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)31.5
Lenalidomide and Dexamethasone Rd1821.5
Melphalan + Prednisone + Thalidomide (MPT)22.1

[back to top]

Percentage of Participants With a Myeloma Response by Favorable Hyperdiploidy Risk Cytogenetic Risk Category Based on IRAC Review

Participants were placed in adverse and non-adverse cytogenetic risk categories at baseline and response rates evaluated. Adverse Risk: t(4;14), t(14;16), del(13q) or monosomy 13, del(17p), 1q gain Favorable Hyperdiploidy: : t(11;14), gains of 5/9/15; Normal: a normal result, gains other than 5/9/15, IgH deletion Uncertain risk: probes used for analysis cannot place participant in any of the other risk categories. Objective response = best overall response including CR, VGPR or PR based on the IRAC Review; A CR is negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPRis serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionpercentage of participants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)80.4
Lenalidomide and Dexamethasone Rd1881.6
Melphalan + Prednisone + Thalidomide (MPT)70.6

[back to top]

Percentage of Participants With a Myeloma Response by Adverse Risk Cytogenetic Risk Category Based on IRAC Review.

Participants were placed in adverse and non-adverse cytogenetic risk categories at baseline and response rates evaluated. Adverse Risk: t(4;14), t(14;16), del(13q) or monosomy 13, del(17p), 1q gain Favorable Hyperdiploidy: : t(11;14), gains of 5/9/15; Normal: a normal result, gains other than 5/9/15, IgH deletion Uncertain risk: probes used for analysis cannot place participant in any of the other risk categories. Objective response = best overall response including CR, VGPR or PR based on the IRAC Review; A CR is negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPRis serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

InterventionPercentage of participants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)70.0
Lenalidomide and Dexamethasone Rd1869.7
Melphalan + Prednisone + Thalidomide (MPT)58.2

[back to top]

Kaplan-Meier Estimates of Progression-free Survival (PFS) Based on the Response Assessment by the Independent Review Adjudication Committee (IRAC)

PFS was calculated as the time from randomization to the first documented PD or death due to any cause during the study, which ever occurred first based on the International Myeloma Working Group Uniform Response criteria (IMWG). Those who withdrew for any reason or received another anti-myeloma therapy without documented PD were censored on the date of their last response assessment, prior to receiving any other anti-myeloma therapy. Censoring rules for PFS: - No baseline assessments and no progression or death documented within the 2 scheduled assessments; Death within the lst two assessments without any adequate response assessment; Progression documented between scheduled assessments; Death between adequate assessments; no progression; study discontinuations for reasons other than PD or death; new anti-myeloma started prior to PD; death or PD after an extended lost to follow-up time period (2 or more missed scheduled assessment's). (NCT00689936)
Timeframe: From date of randomization until the data cut-off date of 24 May 2013. Median follow-up time for all participants was 17.1 months.

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)25.5
Lenalidomide and Dexamethasone Rd1820.7
Melphalan + Prednisone + Thalidomide (MPT)21.2

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Cancer (EORTC QLQ-C30) Global Health Status Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Global Health Status/QOL scale is scored between 0 and 100, with a high score indicating better Global Health Status/QOL. Negative change from Baseline values indicate deterioration in QOL or functioning and positive values indicate improvement. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Study discontinuation
Lenalidomide and Dexamethasone Rd18-1.34.75.43.25.75.0
Lenalidomide and Low-Dose Dexamethasone (Rd)0.44.85.94.86.4-0.1
Melphalan + Prednisone + Thalidomide (MPT)1.04.36.16.54.80.3

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Body Image Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. EORTC QLQ-MY20 includes four scales: disease symptoms, treatment side-effects, future perspective, and body image. Questions used a 4-point scale (from 1 'Not at All' to 4 'Very Much'). Scores were averaged, and transformed to a 0-100 scale; for the body image scale, a higher score indicates a better body image. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-1.50.81.5-0.4-0.31.8
Lenalidomide and Low-Dose Dexamethasone (Rd)-4.5-1.7-1.4-1.4-2.3-5.6
Melphalan + Prednisone + Thalidomide (MPT)-1.6-3.0-2.8-2.6-1.1-5.6

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Disease Symptoms Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. EORTC QLQ-MY20 includes four scales: disease symptoms, treatment side-effects, future perspective, and body image. Questions used a 4-point scale (from 1 'Not at All' to 4 'Very Much'). Scores were averaged, and transformed to a 0-100 scale; a higher score indicates more severe disease symptom(s). (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-4.1-10.0-9.9-8.7-6.2-4.5
Lenalidomide and Low-Dose Dexamethasone (Rd)-4.0-9.1-8.8-7.8-8.7-3.5
Melphalan + Prednisone + Thalidomide (MPT)-4.4-7.0-7.9-6.5-7.9-3.7

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Future Perspective Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. EORTC QLQ-MY20 includes four scales: disease symptoms, treatment side-effects, future perspective, and body image. Questions used a 4-point scale (from 1 'Not at All' to 4 'Very Much'). Scores were averaged, and transformed to a 0-100 scale; for the future perspective scale, a higher score indicates a better perspective of the future. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd183.99.212.312.111.78.8
Lenalidomide and Low-Dose Dexamethasone (Rd)4.78.59.810.812.75.8
Melphalan + Prednisone + Thalidomide (MPT)3.36.38.010.09.53.2

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Patients With Multiple Myeloma (EORTC QLQ-MY20) Side Effects Treatment Scale

EORTC QLQ-MY20 is a validated questionnaire to assess the overall quality of life in patients with multiple myeloma. EORTC QLQ-MY20 includes four scales: disease symptoms, treatment side-effects, future perspective, and body image. Questions used a 4-point scale (from 1 'Not at All' to 4 'Very Much'). Scores were averaged, and transformed to a 0-100 scale; a higher score represents a more severe overall side effect of treatment. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd184.01.2-0.41.22.3-1.0
Lenalidomide and Low-Dose Dexamethasone (Rd)2.51.01.71.92.20.6
Melphalan + Prednisone + Thalidomide (MPT)5.63.52.94.74.33.8

[back to top]

Change From Baseline in the European Quality of Life-5 Dimensions (EQ-5D) Health Utility Index Score

EQ-5D is a self-administered questionnaire that assesses health-related quality of life. The EQ-5D descriptive health profile comprises five dimensions of health (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression). Each dimension has 3 levels of response: No problem (1), some problems (2), and extreme problems (3). A unique EQ-5D health state is defined by combining one level from each of the five dimensions into a single utility index score. EQ-5D index values range from -0.59 to 1.00 where higher EQ-5D scores represent better health status. A positive change from baseline score indicates improvement in health status and better health state. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-0.00.10.10.10.10.0
Lenalidomide and Low-Dose Dexamethasone (Rd)0.00.10.10.10.10.0
Melphalan + Prednisone + Thalidomide (MPT)0.00.10.10.10.10.0

[back to top]

Number of Participants With Adverse Events (AEs) During the Active Treatment Phase

A TEAE is any AE occurring or worsening on or after the first treatment of any study drug, and within 30 days after the last dose of the last study drug. Severity grades according to Common Terminology Criteria for Adverse Events v3.0 (CTCAE) on a 1-5 scale: 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. A serious AE is any AE occurring at any dose that: • Results in death; • Is life-threatening; • Requires or prolongs existing inpatient hospitalization; • Results in persistent or significant disability/incapacity; • Is a congenital anomaly/birth defect; • Constitutes an important medical event. (NCT00689936)
Timeframe: From first dose of study drug through 28 days following the discontinuation visit from active treatment phase; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

,,
InterventionParticipants (Number)
≥ 1 adverse event (AE)≥ 1 grade (Gr) 3 or 4 AE≥ 1 grade (Gr) 5 AE≥ 1 serious adverse event (SAE)≥ 1 AE related to Lenalidomide/Dex/Mel/Pred/Thal≥ 1 AE related to Lenalidomide≥ 1 AE related to dexamethasone≥ 1 AE related to melphalan≥ 1 AE related to prednisone≥ 1 AE related to thalidomide≥1 AE related to Lenalidomide/Dex or Mel/Pred/Thal≥ 1 Gr 3 or 4 AE related to Len/Dex/Mel/Pred/Thal≥ 1 grade 3 or 4 AE related to Lenalidomide≥ 1 grade 3 or 4 AE related to dexamethasone≥ 1 grade 3 or 4 AE related to melphalan≥ 1 grade 3 or 4 AE related to prednisone≥ 1 grade 3 or 4 AE related to Thalidomide≥1Gr 3 or 4 AE related to Len/Dex or Mel/Pred/Thal≥ 1 Grade 5 AE related to Len/Dex/Mel/Pred/Thal≥ 1 Grade 5 AE related to Lenalidomide≥ 1 Grade 5 AE related to Dexamethasone≥ 1 Grade 5 AE related to melphalan≥ 1 Grade 5 AE related to prednisone≥ 1 Grade 5 AE related to Thalidomide≥1 Grade 5 AE related to Len/Dex or Mel/Pred/Thal≥1 SAE related to Len/Dex/Mel/Pred/Thal≥1 SAE related to Lenalidomide≥1 SAE related to dexamethasone≥1 SAE related to melphalan≥1 SAE related to prednisone≥1 SAE related to thalidomide≥1 SAE related to Len/Dex or Mel/Pred/Thal≥1AE leading to Len/Dex/Mel/Pred/Thal Withdrawal≥1 AE leading to Lenalidomide withdrawal≥1 AE leading to dexamethasone withdrawal≥1 AE leading to melphalan withdrawal≥1 AE leading to prednisone withdrawal≥1 AE leading to Thalidomide withdrawal≥1AE leading to Len/DexOR Mel/Pred/Thal Withdrawal≥1AE leading to Len/Dex/Mel/Pred/Thal reduction≥1 AE leading to Lenalidomide reduction≥1 AE leading to dexamethasone reduction≥1 AE leading to melphalan reduction≥1 AE leading to prednisone reduction≥1 AE leading to thalidomide reduction≥1AE leading to Len/Dex or Mel/Pred/Thal reduction≥1 AE leading to Rd or MPT interruption≥1 AE leading to Lenalidomide interruption≥1 AE leading to dexamethasone interruption≥1 AE leading to melphalan interruption≥1 AE leading to prednisone interruption≥1 AE leading to Thalidomide interruption≥1 AE leading to Len and Dex or MPT interruption
Lenalidomide and Dexamethasone Rd1853643336308501481410000269326290177000104119700051581309700064109931040008421415511800020321301280000241
Lenalidomide and Low-Dose Dexamethasone (Rd)5294535035950648242900026937334222900013117121600011195165130000951571091520009627920317000030368353319000290
Melphalan + Prednisone + Thalidomide (MPT)53948038270527004413264931454230030711831649100065521420075629427153008378146713480019947254241900328324388249

[back to top]

Shift From Baseline to Most Extreme Postbaseline Value in Absolute Neutrophil Count During the Active Treatment Phase

Neutrophil counts was assessed for participants from baseline grade to most extreme severity grade using the NCI CTCAE v 3.0 grading scale. (NCT00689936)
Timeframe: Randomization to end of treatment or the data cut off of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

,,
Interventionparticipants (Number)
Normal Baseline Grade to Normal Postbaseline GradeNormal Baseline Grade to Grade 1 postbaselineNormal Baseline Grade to Grade 2 postbaselineNormal Baseline Grade to Grade 3 postbaselineNormal Baseline Grade to Grade 4 postbaselineGrade 1 Baseline to Normal postbaselineGrade1 Baseline to Grade 1 postbaselineGrade 1 Baseline to Grade 2 postbaselineGrade 1 Baseline to Grade 3 postbaselineGrade 1 Baseline to Grade 4 postbaselineGrade 2 Baseline to normal postbaselineGrade 2 Baseline to Grade 1 postbaselineGrade 2 Baseline to Grade 2 postbaselineGrade 2 Baseline to Grade 3 postbaselineGrade 2 Baseline to Grade 4 postbaselineGrade 3 Baseline to Normal postbaselineGrade 3 Baseline to Grade 1 postbaselineGrade 3 Baseline to Grade 2 postbaselineGrade 3 Baseline to Grade 3 postbaselineGrade3 Baseline to Grade 4 postbaselineGrade 4 Baseline to Normal postbaseline GradeGrade 4 Baseline to Grade 1 postbaseline GradeGrade 4 Baseline to Grade 2 postbaselineGrade 4 Baseline Grade to Grade 3 postbaselineGrade 4 Baseline to Grade 4 postbaseline
Lenalidomide and Dexamethasone Rd181338510971306111530401111850012200000
Lenalidomide and Low-Dose Dexamethasone (Rd)103961217021781725911141890022001000
Melphalan + Prednisone + Thalidomide (MPT)3779128141452211202101721100000100000

[back to top]

Shift From Baseline to Most Extreme Postbaseline Value in Creatinine Clearance (CrCl) During the Active Treatment Phase

Renal function was assessed for participants from baseline to the most extreme value in creatinine clearance calculated using the Cockcroft-Gault estimation. (NCT00689936)
Timeframe: Randomization to end of treatment or the data cut off of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

,,
Interventionparticipants (Number)
CrCl< 30 mL/min to CrCl< 30 mL/minCrCl < 30 mL/min to CrCl ≥ 30 but < 50 mL/minCrCl < 30 mL/min to CrCl ≥ 50 but < 80 mL/minCrCl< 30 mL/min to ≥ 80 mL/minCrCl≥ 30 but < 50 mL/min to < 30 mL/minCrCl ≥ 30 but < 50 mL/min to CrCl ≥ 30 but < 50 mLCrCl ≥ 30 but < 50 mL/min to CrCl ≥ 50 but < 80 mLCrCl ≥ 30 but < 50 mL/min to ≥ 80 mL/minCrCl ≥ 50 but < 80 mL to CrCl< 30 mL/minCrCl ≥ 50 but < 80 mL to CrCl ≥ 30 but < 50 mL/minCrCl ≥ 50 but < 80 mL to CrCl ≥ 50 but < 80 mL/minCrCl ≥ 50 but < 80 mL to ≥ 80 mL/minCrCl ≥ 80 mL/min to CrCl< 30 mL/minCrCl ≥ 80 mL/min to CrCl ≥ 30 but < 50 mL/minCrCl ≥ 80 mL/min to CrCl ≥ 50 but < 80 mL/minCrCl ≥ 80 mL/min to CrCl ≥ 80 mL/min
Lenalidomide and Dexamethasone Rd18171482241551201130991010114
Lenalidomide and Low-Dose Dexamethasone (Rd)15187213767904112107006109
Melphalan + Prednisone + Thalidomide (MPT)1919500416520410297009121

[back to top]

Shift From Baseline to Most Extreme Postbaseline Value in Hemoglobin During the Active Treatment Phase

Hemoglobin was assessed for participants from baseline grade to most extreme severity grade using the NCI CTCAE v 3.0 grading scale. (NCT00689936)
Timeframe: Randomization to end of treatment or the data cut off of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

,,
Interventionparticipants (Number)
Normal Baseline Grade to Normal Postbaseline GradeNormal Baseline Grade to Grade 1 postbaselineNormal Baseline Grade to Grade 2 postbaselineNormal Baseline Grade to Grade 3 postbaselineNormal Baseline Grade to Grade 4 postbaselineGrade 1 Baseline to Normal postbaselineGrade 1 Baseline to Grade 1 postbaselineGrade1 Baseline to Grade 2 postbaselineGrade 1 Baseline to Grade 3 postbaselineGrade 1 Baseline to Grade 4 postbaselineGrade 2 Baseline to normal postbaselineGrade 2 Baseline to Grade 1 postbaselineGrade 2 Baseline to Grade 2 postbaselineGrade 2 Baseline to Grade 3 postbaselineGrade 2 Baseline to Grade 4 postbaselineGrade 3 Baseline to Normal postbaselineGrade 3 Baseline to Grade 1 postbaselineGrade 3 Baseline to Grade 2 postbaselineGrade 3 Baseline to Grade 3 postbaselineGrade 3 Baseline to Grade 4 postbaselineGrade 4 Baseline to Normal postbaselineGrade 4 Baseline to Grade 1 postbaselineGrade 4 Baseline to Grade 2 postbaselineGrade 4 Baseline to Grade 3 postbaselineGrade 4 Baseline to Grade 4 postbaseline
Lenalidomide and Dexamethasone Rd18103081001261231750121354190148300011
Lenalidomide and Low-Dose Dexamethasone (Rd)639800010612825208125484001210500001
Melphalan + Prednisone + Thalidomide (MPT)92541001101232040141334711001010200102

[back to top]

Kaplan Meier Estimates of Duration of Myeloma Response as Determined by the IRAC

Duration of response was defined as the duration from the time when the response criteria were first met for CR or VGPR or PR based on IMWG criteria until the first date the response criteria were met for progressive disease or until the participant died from any cause, whichever occurred first. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median follow-up for responders was 20.1 months

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)35.0
Lenalidomide and Dexamethasone Rd1822.1
Melphalan + Prednisone + Thalidomide (MPT)22.3

[back to top]

Kaplan Meier Estimates of Overall Survival at the Time of Final Analysis (OS)

Overall survival was defined as the time between randomization and death. Participants, who died, regardless of the cause of death, were considered to have had an event. All participants who were lost to follow-up prior to the end of the trial or who were withdrawn from the trial were censored at the time of last contact. Participants who were still being treated were censored at the last available date the participant was known to be alive. (NCT00689936)
Timeframe: From date of randomization to date of data cut-off date of 21 January 2016; median follow-up for all participants was 48.3 months

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)59.1
Lenalidomide and Dexamethasone Rd1862.3
Melphalan + Prednisone + Thalidomide (MPT)49.1

[back to top]

Kaplan Meier Estimates of Time to Second Line Therapy AMT at the Time of Final Analysis

Time to second-line anti-myeloma therapy is defined as time from randomization to the start of another non-protocol anti-myeloma therapy. Those who do not receive another anti-myeloma therapy were censored at the last assessment or follow-up visit known to have received no new therapy. (NCT00689936)
Timeframe: From date of randomization until the data cut-off of date 21 January 2016; median follow-up for all participants was 23.0 months

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)36.7
Lenalidomide and Dexamethasone Rd1828.5
Melphalan + Prednisone + Thalidomide (MPT)26.7

[back to top]

Kaplan Meier Estimates of Time to Treatment Failure (TTF)

TTF is defined as the time between the randomization and discontinuation of study treatment for any reason, including disease progression (determined by IRAC based on the IMWG response criteria), treatment toxicity, start of another anti-myeloma therapy (AMT) or death. (NCT00689936)
Timeframe: From date of randomization until the data cut-off of 24 May 2013; median follow-up for all participants was 16.1 months.

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)16.9
Lenalidomide and Dexamethasone Rd1817.2
Melphalan + Prednisone + Thalidomide (MPT)14.1

[back to top]

Kaplan Meier Estimates of Time to Treatment Failure (TTF) at the Time of Final Analysis

TTF is defined as the time between the randomization and discontinuation of study treatment for any reason, including disease progression (determined by the investigators assessment based on the IMWG response criteria), treatment toxicity, start of another anti-myeloma therapy (AMT) or death. (NCT00689936)
Timeframe: From date of randomization until the data cut-off date of 21 January 2016; median follow up for all participants was 16.1 months.

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)16.9
Lenalidomide and Dexamethasone Rd1817.2
Melphalan + Prednisone + Thalidomide (MPT)14.1

[back to top]

Kaplan-Meier Estimates of PFS Based on the Response Assessment by the Investigator At the Time of Final Analysis

PFS was calculated as the time from randomization to the first documented PD or death due to any cause during the study, which ever occurred first based on the International Myeloma Working Group Uniform Response criteria (IMWG). Those who withdrew for any reason or received another anti-myeloma therapy without documented PD were censored on the date of their last response assessment, prior to receiving any other anti-myeloma therapy. Censoring rules for PFS: - No baseline assessments and no progression or death documented within the 2 scheduled assessments; Death within the lst two assessments without any adequate response assessment; Progression documented between scheduled assessments; Death between adequate assessments; no progression; study discontinuations for reasons other than PD or death; new anti-myeloma started prior to PD; death or PD after an extended lost to follow-up time period (2 or more missed scheduled assessment's). (NCT00689936)
Timeframe: From date of randomization to date of data cut-off date of 21 January 2016; median follow-up for all participants was 17.7 months

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)26.0
Lenalidomide and Dexamethasone Rd1821.0
Melphalan + Prednisone + Thalidomide (MPT)21.9

[back to top]

Shift From Baseline to Most Extreme Postbaseline Value in Platelet Count During the Active Treatment Phase.

Improvement in platelets was assessed for participants from baseline grade to most extreme severity grade using the NCI CTCAE v 3.0 grading scale. (NCT00689936)
Timeframe: Randomization to end of treatment or the data cut off of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

,,
Interventionparticipants (Number)
Normal Baseline Grade to Normal Postbaseline GradeNormal Baseline Grade to Grade 1 postbaselineNormal Baseline Grade to Grade 2 postbaselineNormal Baseline Grade to Grade 3 postbaselineNormal Baseline Grade to Grade 4 postbaselineGrade1 Baseline to Normal postbaseline GradeGrade 1 Baseline to Grade 1 postbaselineGrade 1 Baseline to Grade 2 postbaselineGrade 1 Baseline to Grade 3 postbaselineGrade 1 Baseline to Grade 4 postbaselineGrade 2 Baseline to normal postbaseline GradeGrade 2 Baseline to Grade 1 postbaselineGrade 2 Baseline to Grade 2 postbaselineGrade 2 Baseline to Grade 3 postbaselineGrade 2 Baseline to Grade 4 postbaselineGrade 3 Baseline to Normal postbaseline GradeGrade 3 Baseline to Grade 1 postbaselineGrade 3 Baseline to Grade 2 postbaselineGrade 3 Baseline to Grade 3 postbaselineGrade 3 Baseline to Grade 4 postbaseline
Lenalidomide and Dexamethasone Rd1819721130125338191210132000001
Lenalidomide and Low-Dose Dexamethasone (Rd)19721624154134151020033100002
Melphalan + Prednisone + Thalidomide (MPT)16520827311165171010212200110

[back to top]

Change From Baseline in the EORTC QLQ-C30 Role Functioning Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Role Functioning Scale is scored between 0 and 100, with a high score indicating better functioning/support. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-4.66.38.69.49.13.8
Lenalidomide and Low-Dose Dexamethasone (Rd)-2.72.46.37.88.0-0.3
Melphalan + Prednisone + Thalidomide (MPT)-2.44.18.211.814.5-1.0

[back to top]

Time to First Response Based on the Investigator Assessment at the Time of Final Analysis

The time to first myeloma response was defined as the time from randomization to the time when the response criteria for at least a PR was first met based on the IMWG criteria assessed by the investigator. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 21 January 2016; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm.

Interventionmonths (Median)
Lenalidomide and Low-Dose Dexamethasone (Rd)1.8
Lenalidomide and Dexamethasone Rd181.8
Melphalan + Prednisone + Thalidomide (MPT)2.8

[back to top]

Percentage of Participants With an Objective Response Based on IRAC Review

Objective response according to IMWG Uniform Response Criteria was defined as a best overall response including a complete response (CR), very good partial response (VGPR) or partial response (PR) based on the IRAC Review. A CR is defined as: negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPR is serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is: ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 24 May 2013; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionpercentage of participants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)75.1
Lenalidomide and Dexamethasone Rd1873.4
Melphalan + Prednisone + Thalidomide (MPT)62.3

[back to top]

Percentage of Participants With an Objective Response Based on Investigator Assessment at Time of Final Analysis

Objective response according to IMWG Uniform Response Criteria was defined as a best overall response including a complete response (CR), very good partial response (VGPR) or partial response (PR) based on the IRAC Review. A CR is defined s: negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in BM; A VGPR is serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein and urine M-protein level <100 mg/24 hours; A PR is: ≥50% reduction of serum M-Protein and reduction in urinary M-protein by ≥90% or to <200 mg/24 hours. If present at baseline a ≥50% reduction in size of soft tissue plasmacytomas. (NCT00689936)
Timeframe: Disease response was assessed every 28 days until end of treatment or the data cut-off date of 21 January 2016; median duration of treatment was 80.2 weeks in the Rd arm; 72 weeks in the Rd18 arm and 67.1 weeks in the MPT arm

Interventionpercentage of participants (Number)
Lenalidomide and Low-Dose Dexamethasone (Rd)80.7
Lenalidomide and Dexamethasone Rd1878.6
Melphalan + Prednisone + Thalidomide (MPT)67.5

[back to top]

Change From Baseline in the EORTC QLQ-C30 Appetite Loss Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Appetite Loss Scale is scored between 0 and 100, with a high score indicating a higher level of appetite loss. Negative change from Baseline values indicate improvement in appetite and positive values indicate worsening of appetite. (NCT00689936)
Timeframe: Cycle 1 Day 1 (Baseline), then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd182.9-3.3-8.6-6.4-5.1-7.5
Lenalidomide and Low-Dose Dexamethasone (Rd)1.3-5.9-9.8-7.3-8.1-1.0
Melphalan + Prednisone + Thalidomide (MPT)1.0-6.2-13.5-10.5-12.2-2.6

[back to top]

Change From Baseline in the EORTC QLQ-C30 Cognitive Functioning Domain

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used in clinical research to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Cognitive Functioning Scale is scored between 0 and 100, with a high score indicating better functioning/support. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00689936)
Timeframe: Cycle 1 Day 1, (Baseline) then Months 1, 3, 6, 12, 18 and Discontinuation visit

,,
Interventionunits on a scale (Mean)
Month 1Month 3Month 6Month 12Month 18Discontinuation Visit
Lenalidomide and Dexamethasone Rd18-1.71.80.9-1.2-2.8-2.6
Lenalidomide and Low-Dose Dexamethasone (Rd)-1.2-0.7-0.9-1.6-2.2-4.9
Melphalan + Prednisone + Thalidomide (MPT)-1.8-1.5-0.3-0.6-0.7-7.1

[back to top]

Time to Response

Time to response will be measured in the population of subjects with a confirmed response as the time from the date of initiation of treatment to the date of the first documentation of a confirmed response. Full restaging was performed at 6, 12, 18, 24, 36, 48, 60, 72 months). Time to response will be estimated and plotted with the Kaplan-Meier method. The median will be calculated with 95% confidence intervals. (NCT00691704)
Timeframe: 6 months

Interventionmonths (Mean)
High-risk Multiple Myeloma2

[back to top]

Progression Free Survival

Progression free survival (PFS) is defined for all subjects as the time from the date of initiation of treatment to the date of first documentation of relapse, progression, or death due to any cause. Full restaging was performed at 6, 12, 18, 24, 36, 48, 60, 72 months). PFS survival will be estimated and plotted with the Kaplan-Meier method. The median will be calculated with 95% confidence intervals. (NCT00691704)
Timeframe: 2 years

Interventionparticipants (Number)
PFS at 2 yearsPFS at 1 year
High-risk Multiple Myeloma39

[back to top]

Time to Progression

Time to progression (TTP) is defined for all patients as the time from initiation of treatment to disease progression with deaths owing to causes other than progression not counted as events, but censored (Durie et al., 2006). (NCT00691704)
Timeframe: 6 years

Interventionmonths (Mean)
High-risk Multiple Myeloma11

[back to top]

Overall Survival

Overall survival is defined as the time from the date of initiation of treatment to the date of death due to any cause. Overall response rate will be estimated with its 80% confidence interval, and the numbers of subjects achieving a sustained complete response (sCR), complete response (CR), very good partial response (VGPR), partial response (PR), or stable disease (SD) will be tabulated. Overall survival will be estimated and plotted with the Kaplan-Meier method. The median will be calculated with 95% confidence intervals. (NCT00691704)
Timeframe: 6 years

Interventionmonths (Mean)
High-risk Multiple Myeloma36

[back to top]

Duration of Response

The duration of response, defined only among the responders, is measured from start of achieving Partial Response (PR) [first observation of PR before confirmation] to the time of disease progression, with deaths owing to causes other than progression not counted as events, but censored (Durie et al., 2006). Duration of response will be estimated and plotted with the Kaplan-Meier method. The median will be calculated with 95% confidence intervals. (NCT00691704)
Timeframe: 6 years

Interventionmonths (Mean)
High-risk Multiple Myeloma11

[back to top]

Number of Participants With Best Overall Disease Response

Will be monitored simultaneously for each of the subgroups separately using the Bayesian approach of Thall, Simon, Estey. Summary statistics will be provided for continuous variables. Frequency tables will be used to summarize categorical variables. Logistic regression will be will be utilized to assess the effect of patient prognostic factors on the response rate. (NCT00695786)
Timeframe: At the end of 3 courses (84 days)

,,,
InterventionParticipants (Count of Participants)
Best Overall Response - CRBest Overall Response - CRu ResponseBest Overall Response - PRBest Overall Response - SDBest Overall Response - PDBest Overall Response - Inevaluable
Follicular Lymphoma56136103
Marginal Zone Lymphoma1724314
Other Histology001001
Small Lymphocytic Lymphoma8521361

[back to top]

Response Rate by Recist Criteria

"radiographic response defined as partial response defined by RECIST:At least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LD~It is noted that while on average the time frame for scans was 4 months, there were two patients who at 32 and 36 months had not progressed." (NCT00717756)
Timeframe: on average about every 2 months until progression, on average about 4 months.

Interventionparticipants (Number)
Lenalidomide6

[back to top]

Maximum Tolerated Dose (MTD) of Combination Therapy With VELCADE, Dexamethasone, and Doxil, (RVDD)

"Dose Level 1:~15 mg Revlimid daily on days 1-14 followed by 7-day rest every 21 days 1.3 mg/m2 Velcade daily on days 1, 4, 8 and 11 20 mg dexamethasone daily on Days 1, 2, 4, 5, 8, 9, 11, 12* and 20 mg/m2 Doxil daily on day 4~Dose Level 2:~20 mg Revlimid daily on days 1-14 followed by 7-day rest every 21 days 1.3 mg/m2 Velcade daily on days 1, 4, 8 and 11 20 mg dexamethasone daily on Days 1, 2, 4, 5, 8, 9, 11, 12* and 20 mg/m2 Doxil daily on day 4~Dose Level 3:~25 mg Revlimid daily on days 1-14 followed by 7-day rest every 21 days 1.3 mg/m2 Velcade daily on days 1, 4, 8 and 11 20 mg dexamethasone daily on Days 1, 2, 4, 5, 8, 9, 11, 12* and 20 mg/m2 Doxil daily on day 4~Dose Level 4:~25 mg Revlimid daily on days 1-14 followed by 7-day rest every 21 days 1.3 mg/m2 Velcade daily on days 1, 4, 8 and 11 20 mg dexamethasone daily on Days 1, 2, 4, 5, 8, 9, 11, 12* and 30 mg/m2 Doxil daily on day 4" (NCT00724568)
Timeframe: 1 month post treatment

Interventionmg (Number)
RevlimidVELCADEDexamethasoneDoxil
Combination Drug Therapy251.32030

[back to top]

The Percentage of Patients That Achieved Partial or Complete Response to Treatment.

"Partial Response:~50% reduction in the level of serum monoclonal protein for at least two determinations six weeks apart.~If present, reduction in 24-hour urinary light chain excretion by either, greater than or equal to 90%, or to <200 mg for at least two determinations six weeks apart.~50% reduction in the size of soft tissue plasmacytomas (by clinical or radiographic examination) for at least six weeks.~No increase in size or number of lytic bone lesions (development of compression fracture does not exclude response).~Complete Response:~Disappearance of the original monoclonal protein from the blood and urine on at least two determinations for a minimum of six weeks.~<5% plasma cells in the bone marrow on at least two determinations for a minimum of six weeks.~No increase in the size or number of lytic bone lesions." (NCT00724568)
Timeframe: 24 weeks (8, 21-day cycles)

Interventionpercentage of patients (Number)
Combination Drug Therapy96

[back to top]

Number of Patients With Severe Infections

Severe infection requiring more than 2 weeks of antibiotic therapy. (NCT00727415)
Timeframe: At 24 months from study entry (end of follow-up)

Interventionparticipants with severe infecitons (Number)
Phase I-II Lenalidomide2

[back to top]

Overall Complete Response (CR) Rate (Phase II)

Response will be assessed by clinical examination, peripheral blood, bone marrow aspirate and biopsy, radiographic evaluation. Response will be evaluated at three different levels: clinical, cytometric and molecular. (NCT00727415)
Timeframe: After 6 months from study entry (end of treatment).

Interventionpercentage of patients in CR (Number)
Phase I-II Lenalidomide22.5

[back to top]

Maximum Tolerated Dose of Lenalidomide (Phase I)

Maximum tolerated dose of lenalidomide given in combination with fludarabine. (NCT00727415)
Timeframe: The MTD of Lenalinomide will be evaluated during the two courses given with the escalated dose of Lenalinomide defined by the respective dose level.

Interventionnumber of patients without DLT (Number)
Dose level 1 - 5 mgDose level 2 - 10 mg
Phase I-II Lenalidomide61

[back to top]

Toxicity as Assessed by NCI CTCAE v3.0

Data from all subjects who receive any study drug will be included in the safety analyses. (NCT00727415)
Timeframe: At 24 months from study entry (end of follow-up)

InterventionParticipants who died during the study (Number)
Phase I-II Lenalidomide14

[back to top]

Correlation Between Complete Response (CR) and Baseline Biologic Parameters (i.e., IgHV, CD38, Etc.).

(NCT00727415)
Timeframe: After 6 months from study entry (end of treatment).

Interventionpercentage of participants (Number)
CR according to IgHV mutatedCR according to CD19+/CD38+, <30%CR according to CD19+/CD38+, >30%CR according to deletion 11q and 17p, absent
Phase I-II Lenalidomide28.0033.3316.6731.82

[back to top]

Number of Patients Reaching Disease-free Survival (DSF) Overall

Response will be assessed by clinical examination, peripheral blood, bone marrow aspirate and biopsy, radiographic evaluation. Response will be evaluated at three different levels: clinical, cytometric and molecular. (NCT00727415)
Timeframe: After 6 months from study entry (end of treatment)

Interventionpercentage of participants on DFS (Number)
Phase I-II Lenalidomide35.33

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs)

Adverse events were assessed using National Cancer Institute, Common Terminology Criteria for Adverse Events (NCI CTCAE), Version 3: according to the following scale: Grade 1 = Mild Adverse Event (AE), Grade 2 = Moderate AE, Grade 3 = Severe and Undesirable AE, Grade 4 = Life-threatening or Disabling AE, and Grade 5 = Death; Serious AEs (SAEs) are those that resulted in death, were life-threatening, required or prolonged inpatient hospitalization, resulted in persistent or significant disability/incapacity, congenital anomaly, or resulted in an important medical event that may have jeopardized the patient or required medical or surgical intervention to prevent one of the outcomes listed above. after the first dose of study drug and within 28 days after the last dose. A TEAE is defined as any AE occurring or worsening on or after the first dose of study drug and within 28 days after the last dose of study drug. (NCT00737529)
Timeframe: From the first dose of lenalidomide through 28 days after the last dose during the follow-up phase; median (minimum, maximum) duration of treatment was 94.0 (1.0, 1950 days)

Interventionparticipants (Number)
Any TEAEAny TEAE Related to Investigational Product (IP)Any TEAE Grade 3-5 AEAny TEAE Grade 3 AEAny TEAE Grade 4 AEAny TEAE Grade 5 AEAny Grade 3-5 AE Related to IPAny Grade 3 AE Related to IPAny Grade 4 AE Related to IPAny Grade 5 AE Related to IPAny TEAE Serious Adverse Event (SAE)Any SAE Related to IPAny TEAE Leading to Stopping of IPAny Treatment Related AE Leading to Stopping IPAny AE Leading to Dose ReductionAny AE Leading to IP InterruptionAny Treatment Related AE Leading to Dose ReductionTreatment Related AE Leading to IP Interruption
Lenalidomide132118106101571890884127030281655815266

[back to top]

Time to Complete Response (CR+CRu) According to the Independent Review Committee

Time to Complete Response (CR+CRu) was defined as the time from the first dose of study drug to the date of the first occurrence of at least CRu and was calculated only for participants with CR or CRu. (NCT00737529)
Timeframe: From Day 1 of study drug to first documented CR/CRu or better; up to data cut-off date of 06 April 2016; median duration of treatment was 94.5 days

Interventionmonths (Median)
Lenalidomide3.9

[back to top]

Time to Response (TTR)

Time to Response was defined as the time from first dose of study drug to the date of the first response (having at least a PR) and was calculated only for responding participants. (NCT00737529)
Timeframe: From Day 1 of study drug to time of first documented PR or better; up to data cut-off date of 06 April 2016; median duration of treatment was 94.5 days

Interventionmonths (Median)
Lenalidomide3.5

[back to top]

Overall Survival (OS)

Kaplan Meier estimate of overall survival was calculated from the time the first dose of study drug to death from any cause. Participants who had not died were censored at the last date the participant was known to be alive. (NCT00737529)
Timeframe: From Day 1 of study drug to first documented date of progressive disease or death; up to the final data cut-off date of 30 March 2017; median duration of follow-up for surviving participants was 62.94 months

Interventionmonths (Median)
Lenalidomide19.50

[back to top]

Percentage of Participants With a Complete Response (CR) /Complete Response Unconfirmed (CRu) According to the Independent Review Committee

The percentage of participants whose best response was CR or CRu. Participants who had discontinued before CR/CRu was observed, or changed to other anti-lymphoma treatments before a CR/CRu response had been observed, were considered as non-responders. CR is defined as the disappearance of all clinical and radiographic evidence of disease; CRu is defined as a CR, with a 1) residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters (SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2) indeterminate bone marrow. (NCT00737529)
Timeframe: From Day 1 of study drug to progression or early treatment discontinuation; up to data cut-off date of 06 April 2016; Median duration of treatment was 94.5 days

Interventionpercentage of participants (Number)
Lenalidomide9.0

[back to top]

Kaplan Meier Estimate of Duration of Complete Response (DoCR) (CR+CRu) According to the Independent Review Committee

Kaplan Meier estimates for the duration of CR/CRu was calculated from the date of the first occurrence of CR/CRu to the date of documented disease progression or death (without documented progression) for participants who obtained a CR/CRu; participants who had not progressed (or died) were censored at the last valid assessment. (NCT00737529)
Timeframe: From Day 1 of study drug to progression or early discontinuation; up to data cut-off date of 06 April 2016; median time in follow-up was 16.34 months

Interventionmonths (Median)
Lenalidomide24.43

[back to top]

Kaplan Meier Estimate of Duration of Response (DoR) According to the Independent Review Committee

Kaplan Meier estimate for the duration of response (DoR) was calculated from the date of the first occurrence of initial response for responders (demonstrating evidence of at least a PR) to the date of first documented disease progression (any new lesion or increase by ≥ 50% of previously involved sites from nadir) or death (without documented progression) for participants who responded; participants who had not progressed (or died) were censored at the last valid assessment. (NCT00737529)
Timeframe: From Day 1 of study drug to progression or early treatment discontinuation; up to data cut-off date of 06 April 2016; Median duration of treatment was 94.5 days.

Interventionmonths (Median)
Lenalidomide16.64

[back to top]

Kaplan Meier Estimate of Time to Progression (TTP) According to the Independent Review Committee

Kaplan Meier estimate of time to progression was calculated as time from the start of the study drug therapy to the first observation of disease progression. Participants who died without progression were censored at the date of death; otherwise, the censoring rules presented above for PFS applied to the analysis of TTP. Progressive Disease(PD): Appearance of new lesion or increase by ≥50% from previously involved sites from nadir (NCT00737529)
Timeframe: From Day 1 of study drug to first documented time of progression; up to data cut-off date of 06 April 2016; median time in follow-up was 16.34 months

Interventionmonths (Median)
Lenalidomide5.46

[back to top]

Kaplan-Meier Estimate of Progression-Free Survival (PFS) According to the Independent Review Committee

Kaplan Meier estimates of PFS was defined as the start of study drug therapy to the first observation of disease progression or death due to any cause, whichever comes first. If a participant had not progressed or died, PFS was censored at the time of last adequate assessment when the participant was known not to have progressed. For participants who received other anti-lymphoma therapy with no evidence of progression, PFS was censored at time of last adequate tumor assessment with no evidence of progression prior to the start of new anti-lymphoma treatment. (NCT00737529)
Timeframe: From Day 1 of study drug to first documented date of disease progression; up to data cut-off date of 06 April 2016; median time in follow-up was 16.34 months

Interventionmonths (Median)
Lenalidomide4.01

[back to top]

Kaplan-Meier Estimate of Time to Treatment Failure (TTF) According to the Independent Review Committee

Time to treatment failure (TTF) was calculated from the start of study drug therapy to early discontinuation from treatment due to any cause, including disease progression, toxicity, or death and was based on site-reported data. (NCT00737529)
Timeframe: From Day 1 of study drug to first documented time of treatment failure; up to data cut-off date of 06 April 2016; median duration of treatment was 94.5 days

Interventionmonths (Median)
Lenalidomide3.75

[back to top]

Percentage of Participants Who Achieved an Overall Response According to the Independent Review Committee (IRC)

Overall Response Rate (ORR) was defined as the percentage of participants whose best response was Complete Response, Complete Response unconfirmed or Partial Response. Participants who had discontinued before any response has been observed, or changed to other anti-lymphoma treatments before response had been observed, were considered as non-responders. Tumor Response was assessed by a modification of the International Lymphoma Workshop Response Criteria, IWRC, Cheson, 1999); CR is defined as the disappearance of all clinical and radiographic evidence of disease; CRu is defined as a CR, with a 1) residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters (SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2) indeterminate bone marrow; PR = is defined ≥50% decrease in 6 largest nodes or nodal masses. (NCT00737529)
Timeframe: From Day 1 of study treatment to progession or early treatment discontinuation; up to data cut-off date of 06 April 2016; median duration of treatment was 94.5 days.

Interventionpercentage of participants (Number)
Lenalidomide29.9

[back to top]

Progression-free Survival (PFS)

PFS is defined as the time from first dose (phase 1) or time from randomization (phase 2) to disease progression or death. The distribution of PFS was estimated for each treatment group using Kaplan-Meier methodology. Point estimates and 95% CIs for the median for the PFS distribution are provided. (NCT00742560)
Timeframe: From first dose of elotuzumab (phase 1) or randomization (phase 2) until 60 days following the last infusion (or before initiation of new therapy), up to 101 months

Interventionmonths (Median)
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)6.08
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)22.23
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)NA
Total (Phase 1)32.92
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)32.49
Elotuzumab 10 mg/kg Administered as an IV Infusion in Combinat25.00
Total (Phase 2)28.62

[back to top]

Time to Progression (TTP)

TTP is defined as the time from first dose (phase 1) or time from randomization (phase 2) to disease progression. The distribution of TTP was estimated for each treatment group using Kaplan-Meier methodology. Point estimates and 95% CIs for the median for the TTP distribution are provided. (NCT00742560)
Timeframe: From first dose of elotuzumab (phase 1) or randomization (phase 2) until 60 days following the last infusion (or before initiation of new therapy), up to 101 months

Interventionmonths (Median)
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)6.08
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)11.53
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)52.93
Total (Phase 1)52.93
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)32.49
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 2)19.94
Total (Phase 2)28.16

[back to top]

Mean Serum Concentrations of Elotuzumab During Cycle 1

Blood samples were collected during Phase 1, Cycle 1, prior to elotuzumab infusion (time 0 hours) and 30 minutes (0.5 hours) and 4 hours post-infusion (Day 1), 30 minutes (0.5 hours) post-infusion (Day 8 and Day 15), or 30 minutes (0.5 hours), 2 hours, and 4 hours post-infusion (Day 15). Blood samples were collected during Phase 2, Cycle 1, prior to elotuzumab infusion (time 0 hours) and 30 minutes (0.5 hours), 2 hours, and 4 hours post-infusion (Day 1), 30 minutes (0.5 hours) and 2 hours post-infusion (Day 8 and Day 15), or 30 minutes (0.5 hours), 2 hours, and 4 hours post-infusion (Day 15). The samples were analyzed for the concentration of elotuzumab using validated analytical methods. Mean serum concentrations on Cycle 1, Days 1, 8, 15, and 22 (measured in μg/mL) are reported overall (across Phase 1 and Phase 2) by dose. (NCT00742560)
Timeframe: Cycle 1: Days 1 (pre-infusion and 0.5, 2 and 4 hours post-infusion), 8 (pre-infusion and 0.5 and 2 hours post-infusion), 15 (pre-infusion and 0.5 hours and 2 hours post-infusion), and 22 (pre-infusion and 0.5, 2, and 4 hours post-infusion)

Interventionμg/mL (Mean)
Day 1: 0 hoursDay 1: 0.5 hoursDay 1: 4 hoursDay 8: 0 hoursDay 8: 0.5 hoursDay 15: 0 hoursDay 15: 0.5 hoursDay 22: 0 hoursDay 22: 0.5 hoursDay 22: 2 hoursDay 22: 4 hours
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone0.0078.4885.5632.44133.3749.84140.0961.93168.61268.53128.94

[back to top]

Mean Serum Concentrations of Elotuzumab During Cycle 1

Blood samples were collected during Phase 1, Cycle 1, prior to elotuzumab infusion (time 0 hours) and 30 minutes (0.5 hours) and 4 hours post-infusion (Day 1), 30 minutes (0.5 hours) post-infusion (Day 8 and Day 15), or 30 minutes (0.5 hours), 2 hours, and 4 hours post-infusion (Day 15). Blood samples were collected during Phase 2, Cycle 1, prior to elotuzumab infusion (time 0 hours) and 30 minutes (0.5 hours), 2 hours, and 4 hours post-infusion (Day 1), 30 minutes (0.5 hours) and 2 hours post-infusion (Day 8 and Day 15), or 30 minutes (0.5 hours), 2 hours, and 4 hours post-infusion (Day 15). The samples were analyzed for the concentration of elotuzumab using validated analytical methods. Mean serum concentrations on Cycle 1, Days 1, 8, 15, and 22 (measured in μg/mL) are reported overall (across Phase 1 and Phase 2) by dose. (NCT00742560)
Timeframe: Cycle 1: Days 1 (pre-infusion and 0.5, 2 and 4 hours post-infusion), 8 (pre-infusion and 0.5 and 2 hours post-infusion), 15 (pre-infusion and 0.5 hours and 2 hours post-infusion), and 22 (pre-infusion and 0.5, 2, and 4 hours post-infusion)

,
Interventionμg/mL (Mean)
Day 1: 0 hoursDay 1: 0.5 hoursDay 1: 2 hoursDay 1: 4 hoursDay 8: 0 hoursDay 8: 0.5 hoursDay 8: 2 hoursDay 15: 0 hoursDay 15: 0.5 hoursDay 22: 0 hoursDay 22: 0.5 hoursDay 22: 2 hoursDay 22: 4 hours
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone0.00217.90213.31251.3492.47281.53268.35111.11282.29135.92310.03298.85538.88
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone0.00434.20388.58525.98168.55593.80520.97298.82661.91308.02699.70704.48981.16

[back to top]

Number of Participants With Infusion Reactions

During Phase 1, a list of 118 pre-defined MedDRA preferred terms that had been adjudicated to be clinically relevant to infusion reactions by a safety committee was used to search for TEAEs that could potentially be associated with an infusion reaction following elotuzumab administration. Examples of these terms included angioedema, bronchospasm, chills, flushing, pyrexia, rash and urticaria. During Phase 2, the method for capturing TEAEs associated with an infusion reaction was modified to include investigators' designation of AEs judged as clinically relevant infusion reactions. The number of participants infusion reactions are provided overall and by highest toxicity grade (CTCAE v 3.0). (NCT00742560)
Timeframe: Cycles 1 and 2: Days 1, 8, 15, and 22 (day of infusion of elotuzumab) and Days 2, 9, 16, and 23 (day following infusion); and Cycles 3 and greater: Days 1 and 15 (day of infusion) and Days 2 and 16 (day after infusion) (up to 95 months)

,,,,
Interventionparticipants (Number)
Any reactionGrade 5Grade 4Grade 3Grade 2Grade 1
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)300012
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)500113
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)20012512
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 2)300012
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)200002

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs)

An adverse event (AE) is defined as any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. The investigator assessed the relationship of each event to the use of study drug as either definitely related, probably related, possibly related or unrelated. A serious adverse event (SAE) is an event that results in death, is life-threatening, requires or prolongs hospitalization, results in a congenital anomaly, persistent or significant disability/incapacity or is an important medical event that, based on medical judgment, may jeopardize the subject and may require medical or surgical intervention to prevent any of the outcomes listed above. Treatment-emergent events (TEAEs/TESAEs) are defined as any event that began or worsened in severity after the first dose of study drug. For more details on adverse events please see the Adverse Event section. (NCT00742560)
Timeframe: Treatment-emergent adverse events (TEAEs) and serious adverse events (TESAEs) were collected from first dose of study drug until 60 days after the last dose of study drug (up to 95 months)

,,,,
Interventionparticipants (Number)
Any TEAEAny TESAETEAEs ≥ Grade 3TEAEs related to study drugTESAEs related to study drug
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)33330
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)362132292
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)221219162
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 2)372125265
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)30230

[back to top]

Plasma Cell Myeloma Cytogenetic Subtype

Plasma cell myeloma cytogenetic subtype was assessed at the screening visit using standard karyotyping and/or fluorescence in situ hybridization. The number of participants in each cytogenetic risk category are provided: High Risk (International Staging System [ISS] stage II or III and t(4;14) or del(17p) abnormality); Standard Risk (not high or low risk); and Low Risk (ISS stage I or II and absence of t(4;14), del(17p) and 1q21 abnormalities AND age < 55). (NCT00742560)
Timeframe: Screening (up to 14 days prior to dosing)

,,,,
Interventionparticipants (Number)
High RiskStandard RiskLow RiskNot Reported
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)0300
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)13023
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)01732
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 2)32437
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)1200

[back to top]

Maximum Tolerated Dose (MTD) of Elotuzumab in Combination With Lenalidomide and Dexamethasone (Phase 1)

MTD was determined by testing increasing doses up to 20 mg/kg once daily dose escalation cohorts 1 to 3 with 3 patients each. MTD reflects highest dose of drug that did not cause an unacceptable side effect (dose limiting toxicity [DLT]) in more than 30% of patients; e.g., hematologic toxicities like Common Toxicity Criteria for Adverse Events (CTCAE) Grade 4 neutropenia in specific conditions, platelets < 10,000 cells/mm^3 that do not recover to 25,000 cells/mm^3; and specific non-hematologic/biochemical toxicities CTCAE Grade 3 or 4 (except fatigue and Grade 3 infections); CTCAE version 3.0 were used. (NCT00742560)
Timeframe: 4 weeks

Interventionmg/kg (Number)
Phase 1 Elotuzumab + Lenalidomide and Dexamethasone20

[back to top]

Duration of Response

Duration of response is defined as the time from the initial objective response to disease progression or death, whichever occurs first. The distribution of duration of response was estimated for each treatment group using Kaplan-Meier methodology. Point estimates and 95% CIs for the median for the duration of response distribution are provided. (NCT00742560)
Timeframe: From first dose of elotuzumab (phase 1) or randomization (phase 2) until 60 days following the last infusion (or before initiation of new therapy), up to 101 months

Interventionmonths (Median)
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)4.47
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)9.92
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)NA
Total (Phase 1)NA
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)34.83
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 2)29.01
Total (Phase 2)29.24

[back to top]

Objective Response Rate (ORR) According to the International Myeloma Working Group Uniform Response Criteria (Phase 1)

ORR: Percentage of participants with confirmed complete response (CR; negative immunofixation on the serum and urine, disappearance of any soft tissue plasmacytomas, and ≤5% plasma cells in bone marrow), partial response (PR; ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to ≤200 mg per 24 hour; if serum and urine M-protein are unmeasurable, a ≥50% decrease in the difference between involved and uninvolved free light chain (FLC] levels is required in place of the M-protein criteria; if serum and urine M-protein are unmeasurable, and serum FLC is also unmeasurable, a ≥50% reduction in plasma cells is required in place of M-protein, provided baseline bone marrow plasma cell percentage was ≥30%; and, if present at baseline, a ≥50% reduction in the size of soft tissue plasmacytomas), very good PR (VGPR; normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence), or stringent CR (sCR; CR plus VGPR). (NCT00742560)
Timeframe: From first dose of elotuzumab until 60 days following the last infusion (or before initiation of new therapy), up to 100.5 months

Interventionpercentage of participants (Number)
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone (Phase 1)100
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 1)100
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 1)77.3
Total (Phase 1)82.1

[back to top]

Percentage of Participants With Treatment-emergent Anti-elotuzumab Antibody (ADA)

Treatment-emergent (post-dose) positive elotuzumab-specific ADA is differentiated from pre-existing (positive at the predose time point) positive elotuzumab-specific ADA. The percentage of participants with confirmed treatment-emergent ADA overall by dose is provided. (NCT00742560)
Timeframe: From screening through 60-day follow up period (up to 101 months)

Interventionpercentage of participants (Number)
Elotuzumab 5 mg/kg + Lenalidomide and Dexamethasone0
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone6
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone5

[back to top]

Objective Response Rate (ORR) According to the International Myeloma Working Group Uniform Response Criteria (Phase 2)

ORR: Percentage of participants with confirmed complete response (CR; negative immunofixation on the serum and urine, disappearance of any soft tissue plasmacytomas, and ≤5% plasma cells in bone marrow), partial response (PR; ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to ≤200 mg per 24 hour; if serum and urine M-protein are unmeasurable, a ≥50% decrease in the difference between involved and uninvolved free light chain (FLC] levels is required in place of the M-protein criteria; if serum and urine M-protein are unmeasurable, and serum FLC is also unmeasurable, a ≥50% reduction in plasma cells is required in place of M-protein, provided baseline bone marrow plasma cell percentage was ≥30%; and, if present at baseline, a ≥50% reduction in the size of soft tissue plasmacytomas), very good PR (VGPR; normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence), or stringent CR (sCR; CR plus VGPR). (NCT00742560)
Timeframe: From date of randomization until 60 days following the last infusion (or before initiation of new therapy), up to 101 months

Interventionpercentage of participants (Number)
Elotuzumab 10 mg/kg + Lenalidomide and Dexamethasone (Phase 2)91.7
Elotuzumab 20 mg/kg + Lenalidomide and Dexamethasone (Phase 2)75.7
Total (Phase 2)83.6

[back to top]

Percentage of Participants With Progression-free Survival (PFS) and Overall Survival (OS).

Assess the time to disease progression and overall survival. (Progressive disease is defined as at least one of the following: more than or equal to 50% increase in the sum of the products of the greatest diameters of at least 2 lymph nodes on 2 consecutive determinations 2 weeks apart (at least one node must be ≥ 2 cm) or new palpable lymph nodes, more than or equal to 50% increase in the size of the liver and/or spleen as determined by measurement below the costal margin or appearance of palpable hepatomegaly or splenomegaly not previously present, more than or equal to 50% increase in the absolute number of circulating lymphocytes to at least 5.0 x109/L, OR transformation to a more aggressive histology (e.g. Richter's syndrome or prolymphocytic leukemia with >55% prolymphocytes)). (NCT00751296)
Timeframe: Patients will be treated with lenalidomide until disease progression or 2 cycles past CR (no maximum of cycles). Participants were followed upto 53.2 months for the final data analysis.

Interventionpercentage of participants (Number)
Overall survivalProgression free survival
Lenalidiomide85.364.6

[back to top]

To Assess the Efficacy (Response Rate) of Oral Lenalidomide in the Treatment of Patients With Symptomatic, Previously Untreated, Chronic Lymphocytic Leukemia (CLL)

"The primary endpoint was objective response to lenalidomide (Complete response +Partial response) evaluated as per the revised 1996 National Cancer Institute Working Group Guidelines.~Complete response: absence of lymphadenopathy and organomegaly by physical exam and radiology, absence of constitutional symptoms, normal CBC. Bone marrow to be done 2 months after the above criteria are met, must be normocellular, with <30% lymphocytes.~Partial Response: ≥ 50% decrease in the peripheral blood lymphocytes from pre-treatment value, ≥ 50% reduction in lymphadenopathy and organomegaly by physical exam or on CT scan. one or more of the following: neutrophils ≥ 1.5 x109/L, platelets > 100 x109/L or 50% improvement over baseline, hemoglobin > 110 g/L or 50% improvement over baseline (without transfusion)." (NCT00751296)
Timeframe: Patients will be treated with lenalidomide until disease progression or 2 cycles past CR (no maximum of cycles). Participants were followed upto 53.2 months for the final data analysis.

Interventionparticipants (Number)
Complete responsePartial responseStable Disease
Lenalidiomide5136

[back to top]

Overall Participant Response Rate: Percentage of Participants With Complete + Partial Response According to Revised National Cancer Institute-sponsored Working Group Guidelines

Complete response: Absence lymphadenopathy, hepatomegaly or splenomegaly & constitutional symptoms; Normal complete blood count (CBC) exhibited by polymorphonuclear leukocytes>1500/µL, platelets>100,000/µL, hemoglobin>11.0 g/dL (untransfused); lymphocyte count <5,000/µL; Bone marrow aspirate & biopsy normocellular for age with <30% nucleated cells lymphocytes; Absence Lymphoid nodules. Fulfillment CR criteria after induction with exception of treatment related persistent cytopenia & bone marrow lymphoid nodules both considered partial response; Partial response: Requires 50% decrease in peripheral lymphocytes from pre-treatment, 50% reduction in lymphadenopathy, &/or 50% reduction in splenomegaly/hepatomegaly for 2+ months from therapy completion. Additionally one following from pre-treatment: Polymorphonuclear leukocytes 1,500/µL or 50% improvement; Platelets>100,000/µL or 50% improvement; Hemoglobin>11.0 g/dL (untransfused) or 50% improvement. (NCT00759603)
Timeframe: Responses assessed after 12 cycles, up to 48 weeks with interim assessments performed after 3, 6 and 12 cycles.

InterventionPercentage of Participants (Number)
Lenalidomide + Rituximab66

[back to top]

Number of Patients With Each Worst-Grade Toxicity

Count of patients according to the worst-grade toxicity experienced by each, where worst-grade toxicity is per NCI common toxicity criteria: grade 1, mild; grade 2, moderate; grade 3, severe; grade 4, life-threatening; grade 5, death. Toxicities present at baseline and continuing without change in grade are excluded when considering worst-grade toxicity. (NCT00765245)
Timeframe: 30 days after completing treatment, for up to 13 months

,
Interventionparticipants (Number)
Number of patients with WGT=1Number of patients with WGT=2Number of patients with WGT=3Number of patients with WGT=4Number of patients with WGT=5
Arm I: Lenalidomide26650
Arm II: Lenalidomide and Rituximab IV08850

[back to top]

Disease-free Survival at 2 Years

Disease-free survival is the estimated probable duration of life from on-study date to date of death from any cause, using the Kaplan-Meier method where death is an event, with censoring for non-expired patients at last known date alive. (NCT00765245)
Timeframe: From on-treatment date to disease recurrence, up to 2 years

Interventionyears (Median)
Arm I: Lenalidomide0.818
Arm II: Lenalidomide and Rituximab IV0.757

[back to top]

Disease-free Survival at 1 Year

Disease-free survival is the time from on-treatment to first relapse or death (whichever comes first). Those who are alive and without relapse are censored at the last date known alive. (NCT00765245)
Timeframe: From on-treatment date to disease recurrence, up to 1 year

Interventionyears (Mean)
Arm I: Lenalidomide0.818
Arm II: Lenalidomide and Rituximab IV0.90

[back to top]

Progression-free Survival (PFS) Rate at 12 Months

"PFS rate at 12 months is defined as the percentage of participants who are alive and progression-free at 12 months. Progression is exclusively defined as a patient with progressive disease while receiving treatment with lenalidomide in combination with dexamethasone. Progression was defined as any one or more of the following: An increase of 25% from lowest confirmed response in:~Serum M-component (absolute increase >= 0.5g/dl)~Urine M-component (absolute increase >= 200mg/24hour~Difference between involved and uninvolved Free Light Chain levels (absolute increase >= 10mg/dl~Bone marrow plasma cell percentage (absolute increase of >=10%)" (NCT00772915)
Timeframe: 12 months from registration

Interventionpercentage of participants (Number)
Lenalidomide With On-Demand Dexamethasone79

[back to top]

Progression-free Survival (PFS)

"PFS was defined as the time from registration to progression or death due to any cause. The median PFS with 95%CI was estimated using the Kaplan Meier method.~Progression was defined as any one or more of the following:An increase of 25% from lowest confirmed response in:~Serum M-component (absolute increase >= 0.5g/dl)~Urine M-component (absolute increase >= 200mg/24hour~Difference between involved and uninvolved Free Light Chain levels (absolute increase >= 10mg/dl~Bone marrow plasma cell percentage (absolute increase of >=10%)" (NCT00772915)
Timeframe: Time from registration to progression or death (up to 3 years)

Interventionmonths (Median)
Lenalidomide With On-Demand Dexamethasone27

[back to top]

Overall Survival (OS)

OS was defined as the time from registration to death of any cause. Participants were followed for a maximum of 3 years from randomization. The median OS with 95% CI was estimated using the Kaplan Meier method (NCT00772915)
Timeframe: Time from registration to death (up to 3 years)

Interventionmonths (Median)
Lenalidomide With On-Demand Dexamethasone61.1

[back to top] [back to top]

Confirmed Response Rate

"Confirmed response rate is defined as the percentage of participants who achieved a response that was confirmed on 2 consecutive evaluations during treatment~Complete Response(CR): Complete disappearance of M-protein from serum & urine on immunofixation, normalization of Free Light Chain (FLC) ratio & <5% plasma cells in bone marrow (BM)~Very Good Partial Response(VGPR): >=90% reduction in serum M-component; Urine M-Component <100 mg per 24 hours; <=5% plasma cells in BM~Partial Response PR): >= 50% reduction in serum M-Component and/or Urine M-Component >= 90% reduction or <200 mg per 24 hours; or >= 50% decrease in difference between involved and uninvolved FLC levels" (NCT00772915)
Timeframe: Up to 18 cycles from registration

Interventionpercentage of participants (Number)
Lenalidomide With On-Demand Dexamethasone61

[back to top]

Overall Survival (OS)

Overall Survival (OS) is defined as the time from randomization to death from any cause. OS will be censored at the last date that the participant was known to be alive for participants who were alive at the time of analysis and for participants who were lost to follow-up before death was documented. (NCT00774345)
Timeframe: Up to approximately 11 years

InterventionMonths (Median)
Lenalidomide95.09
Placebo73.28

[back to top]

Progression Free Survival 2 (PFS2)

Progression Free Survival (PFS2) assessed by investigator is defined as the time from randomization to the second objective disease progression, or death from any cause, whichever occurs first. (NCT00774345)
Timeframe: Up to 6 years

InterventionMonths (Median)
LenalidomideNA
Placebo35.9

[back to top]

Number of Participants With Adverse Events (AEs)

Number of participants with adverse events (AEs) that measure type, frequency and severity of AEs graded by National Cancer Institute Common Terminology Criteria (NCI CTCAE V 3.0) including any grade adverse events (AEs), Grade 3-4 AEs, AEs related to study drug, grade 3-4 AEs related to study drug. (NCT00774345)
Timeframe: From first dose to 30 days post last dose (up to 9 years)

,
InterventionParticipants (Number)
Adverse Events (AEs)Grade 3-4 AEsAEs related to Study drugsGrade 3-4 AEs related to Study drugs
Lenalidomide155136143117
Placebo149739841

[back to top]

Time Until Progression After Lenalidomide-dexamethasone + Rituximab Therapy in Relapsed Small B-cell Lymphomas With Rituximab Resistance

Progression free survival time in months (NCT00783367)
Timeframe: 9 years from enrollment of first subject

Interventionmonths (Median)
Cohort 122.2
Cohort 222.4

[back to top]

Response Rate to Lenalidomide-dexamethasone + Rituximab Therapy in Relapsed Small B-cell Lymphoma With Rituximab Resistance

Response rate is defined as a complete response or partial response using anatomic criteria of the International Workshop Response Critieria (Cheson, 1999). (NCT00783367)
Timeframe: 3 months

InterventionParticipants (Count of Participants)
Cohort 114
Cohort 213

[back to top]

Assessment of Tumor Response

"The proportion of responses was determined by counting the number of complete responses and partial responses and dividing by the number of evaluable patients.~Complete Response (CR): Disappearance of all evidence of disease and disease-related symptoms. The spleen and/or liver, if considered enlarged before therapy on the basis of a physical examination or CT scan, should not be palpable on physical examination and should be considered normal size by imaging studies, and nodules related to lymphoma should disappear. If the bone marrow was involved by lymphoma before treatment, the infiltrate must have cleared on repeat bone marrow biopsy.~Partial Response (PR): At least a 50% decrease in sum of the product of the diameters (SPD) of up to six of the largest dominant nodes or nodal masses. No increase should be observed in the size of other nodes, liver, or spleen. No new sites of disease should be observed." (NCT00784927)
Timeframe: Up to 1 year from registration.

Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Treatment30.354.5

[back to top]

Progression-free Survival Time

"Progression-free survival time is defined as the time from registration to the earliest date of documentation of disease progression. If a patient dies without a documentation of disease progression the patient will be considered to have had disease progression at the time of their death.~Progressive disease is defined as having one of the following:~The appearance of any new lesion more than 1.5 cm in any axis during or at the end of therapy, even if other lesions are decreasing in size.~At least a 50% increase from nadir in the sum of the product of the dimension (SPD) of any previously involved nodes.~At least a 50% increase in the longest diameter of any single previously identified node more than 1 cm in its short axis." (NCT00784927)
Timeframe: Up to 1 year from registration.

Interventionmonths (Median)
Treatment38.3

[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. (NCT00784927)
Timeframe: Up to 1 year from registration.

Interventionmonths (Median)
TreatmentNA

[back to top]

Time to Treatment Failure

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. (NCT00784927)
Timeframe: Up to 1 year from registration.

Interventionmonths (Median)
TreatmentNA

[back to top]

Tumor Response to Lenalidomide, Rituximab, Cyclophosphamide and Dexamethasone in the Subgroup of Patients With Lymphoplasmacytic Lymphoma (Waldenstrom's Macroglobulinemia).

"The proportion of responses in Waldenstrom's macroglobulinemia was determined by counting the number of complete responses and partial responses and dividing by the number of evaluable patients.~Complete Response (CR): Disappearance of all evidence of disease and disease-related symptoms. The spleen and/or liver, if considered enlarged before therapy on the basis of a physical examination or CT scan, should not be palpable on physical examination and should be considered normal size by imaging studies, and nodules related to lymphoma should disappear. If the bone marrow was involved by lymphoma before treatment, the infiltrate must have cleared on repeat bone marrow biopsy.~Partial Response (PR): At least a 50% decrease in sum of the product of the diameters (SPD) of up to six of the largest dominant nodes or nodal masses. No increase should be observed in the size of other nodes, liver, or spleen. No new sites of disease should be observed." (NCT00784927)
Timeframe: Up to 1 year from registration.

Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Treatment6.766.7

[back to top]

Progression-free Survival

Progression-free survival is the time from registration to disease progression or death. Patients alive without disease progression were censored at the time of the last disease assessment. (NCT00790842)
Timeframe: 56 months

InterventionMonths (Median)
Group A=30-60 CrCl (mL/Min)12.6
Group B=CrCL<30 mL/Min Not on Dialysis11.4
Group C=CrCL<30 mL/Min and on DialysisNA

[back to top]

Percentage of Participants Who Experience a Response [sCR, CR, VGPR, PR]

Per International Myeloma Working Group criteria, complete response (CR): negative immunofixation of serum and urine, normalization of free light chain (FLC) ratio if at study entry FLC was only measurable non-bone parameter, <5% plasma cells in bone marrow, disappearance of any soft tissue plasma cytomas; stringent complete response (sCR): all of above, + normal serum FLC ratio in all patients and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence; partial response (PR): >=50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by >=90% or to < 200 mg per 24 hours, >=50% decrease in difference between involved and uninvolved FLC levels or a 50% decrease in level of involved FLC with 50% decrease in ratio, >=50% reduction in bone marrow plasma cells, if baseline percentage was >=30%, >=50% reduction in size of soft tissue plasmacytoma; very good partial response (VGPR): PR + improvements in serum and urine M-components (NCT00790842)
Timeframe: 56 months

Interventionpercentage of participants (Number)
Group A CrCl 30-60 mL/Min60.0
Group B CrCL<30 mL/Min, Not on Dialysis60.0
Group C CrCL<30 mL/Min and on Dialysis20.0

[back to top]

Overall Survival Time

Overall survival is the time from registration to death from any cause. Patients alive at the time of analysis were censored at the date last known alive. (NCT00790842)
Timeframe: 56 months

Interventionmonths (Median)
Group A=30-60 CrCl (mL/Min)20.8
Group B=CrCL<30 mL/Min Not on Dialysis20.0
Group C=CrCL<30 mL/Min and on DialysisNA

[back to top] [back to top]

Number of Participants in Phase I Component With Dose Limiting Toxicities During the First Cycle of Therapy

"Dose Limiting Toxicity (DLT) was defined as any of the following events determined by the investigator to be possibly, probably, or definitely related to lenalidomide within the first cycle of therapy irrespective of whether the adverse events resolved:~Grade 3 or higher neutropenia with fever ≥38.5 degrees C~Grade 4 neutropenia ≥7 days~Grade 4 or higher thrombocytopenia~Other non-hematologic Grade 4 or higher adverse event not present prior to starting therapy or not due to underlying cause" (NCT00790842)
Timeframe: First cycle of therapy (28 days)

InterventionParticipants (Count of Participants)
Group A Lenalidomide 10 mg/Day0
Group A Lenalidomide 15 mg/Day0
Group A Lenalidomide 25 mg/Day0
Group B Lenalidomide 15 mg/2 Days0
Group B Lenalidomide 25 mg/2 Days0
Group B Lenalidomide 15 mg/Day0
Group B Lenalidomide 25 mg/Day0
Group C Lenalidomide 15 mg 3x/Week0
Group C Lenalidomide 10 mg/Day0
Group C Lenalidomide 15 mg/Day0
Group C Lenalidomide 25 mg/Day0

[back to top]

Duration of Response

Duration of response was defined as time between the onset of response and disease progression in months, among patients treated at the recommended phase II dose who experienced a response to treatment. Per criteria of the International Myeloma Working Group, progressive disease was defined as one of the following: increase of 25% from best confirmed response in serum M-component, urine M-component, free light chain (FLC), bone marrow plasma cell percentage, or development of new or increase in size of bone lesions or soft tissue plasma cytomas. Development of hypercalcemia that can be attributed solely to the myeloma also constituted progression. (NCT00790842)
Timeframe: 56 months

InterventionMonths (Median)
Group A=30-60 CrCl (mL/Min)21.8
Group B=CrCL<30 mL/Min Not on Dialysis8.4
Group C=CrCL<30 mL/Min and on Dialysis25.4

[back to top]

ECOG Performance Status

This criteria is used to assess how a patient's disease is progressing and to assess how the disease affects the daily living abilities of the patient. The scores on this instrument range from 0-5 (0=fully active and 5=dead), with higher scores indicating poorer performance. It was measured as a change in performance status between before and after 1st cycle of treatment with Lenalidomide. (NCT00792077)
Timeframe: before and after first cycle of Lenalidomide treatment, up to 8 weeks

Interventionscore on a scale (Median)
Lenalidomide0

[back to top]

The Functional Assessment of Chronic Illness Therapy-Fatigue Subscale Score

The FACIT-F subscale is a validated measure of fatigue. In the FACIT-F, the patient rates the intensity of their fatigue and its related symptoms on a scale of 0 to 4. The total score can range between 0 and 52, with higher scores denoting less fatigue. The score 0= worst fatigue possible, 52 indicates no fatigue. The scores reported is the median (IQR) change at 8 weeks compared to baseline. (NCT00792077)
Timeframe: before and after 1st cycle of Lenalidomide treatment, up to 8 weeks

Interventionscore on a scale (Median)
Lenalidomide4

[back to top]

Total Sleep Time as Measured by Polysomnography (PSG)

Polysomnography (PSG) was done to measure the total sleep time in minutes. It was measured as a change in total sleep time between the before and after 1st cycle of treatment with Lenalidomide. (NCT00792077)
Timeframe: before and after 1st cycle of Lenalidomide treatment, up to 8 weeks

InterventionMinutes (Median)
Lenalidomide-25.5

[back to top]

Epwort Sleep Scale

The Epworth sleep scale is an 8-item questionnaire designed to asses general level of daytime sleepiness, and scores on this instrument range form 0-24, with higher scores indicating greater sleepiness. It was measured as a change in total score between the before and after 1st cycle of treatment with Lenalidomide. (NCT00792077)
Timeframe: before and after 1st cycle of Lenalidomide treatment, up to 8 weeks

Interventionscore on a scale (Median)
Lenalidomide-1

[back to top]

Progression Free Survival (PFS) Rate at 2 Years After Enrollment in Untreated Patients With Multiple Myeloma.

(NCT00807599)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Continue Lenalidomide and Dexamethasone84.6
Stem Cell Transplant x 1 or x 283.3

[back to top]

Overall Survival

(NCT00807599)
Timeframe: up to 4 years

Interventionpercentage of participants alive (Number)
Continue Lenalidomide and Dexamethasone95.7
Stem Cell Transplant x 1 or x 290.6

[back to top]

Number of Participants With Adverse Events (AE)

"An AE that resulted in any of the following outcomes was defined as a serious adverse event (SAE):~Death;~Life-threatening event;~Any inpatient hospitalization or prolongation of existing hospitalization;~Persistent or significant disability or incapacity;~Congenital anomaly or birth defect;~Any other important medical event.~The investigator determined the relationship of an AE to study drug based on the timing of the AE relative to drug administration and whether or not other drugs, therapeutic interventions, or underlying conditions could provide a sufficient explanation for the event.~The severity of an AE was evaluated by the investigator according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) (Version 3.0) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death." (NCT00812968)
Timeframe: After the first study dose until 28 days after completion of/discontinuation from the study (maximum time on study was 155 weeks).

Interventionparticipants (Number)
Any adverse event (AE)AE related to study drugGrade 3 or 4 AEGrade 3 or 4 AE related to study drugSerious AE (SAE)SAE related to study drugAE leading to discontinuation of study drugRelated AE leading to discontinuationAE leading to a dose reduction or interruptionRelated AE leading to dose reduction/interruptionDeaths
Lenalidomide1111111131001090

[back to top]

Percentage of Bone Marrow Myeloblasts

Bone marrow morphology was assessed by the central hematologic reviewers based on the locally-prepared bone marrow smear slide and clot section. (NCT00812968)
Timeframe: Baseline, at the end of Cycle 3 (Day 85) and Cycle 6 (Day 169).

InterventionPercentage of myeloblasts (Median)
Baseline (N=11)End of Cycle 3 (N=10)End of Cycle 6 (N=10)
Lenalidomide3.771.471.79

[back to top]

Change From Baseline in Percentage of Bone Marrow Erythroblasts

Bone marrow morphology was assessed by the central hematologic reviewers based on the locally-prepared bone marrow smear slide and clot section. (NCT00812968)
Timeframe: Baseline, at the end of Cycle 3 (Day 85) and Cycle 6 (Day 169).

InterventionPercentage of Bone Marrow Erythroblasts (Median)
Change from Baseline at the end of Cycle 3Change from Baseline at the end of Cycle 6
Lenalidomide36.521.5

[back to top]

Percentage of Bone Marrow Promyelocytes

Bone marrow morphology was assessed by the central hematologic reviewers based on the locally-prepared bone marrow smear slide and clot section. (NCT00812968)
Timeframe: Baseline, at the end of Cycle 3 (Day 85) and Cycle 6 (Day 169).

InterventionPercentage of promyelocytes (Median)
Baseline (N=11)End of Cycle 3 (N=10)End of Cycle 6 (N=10)
Lenalidomide555

[back to top]

Time to Erythroid Response

Time to erythroid response was calculated as the time from the first dose of study drug to the start of the first major or minor erythroid response. Similarly, time to major erythroid response was calculated as the time from the first dose of study drug to the start of the first major erythroid response. (NCT00812968)
Timeframe: From the first dose of study drug through Week 156

Interventionweeks (Median)
Time to erythroid response (major or minor)Time to major erythroid response
Lenalidomide2.16.3

[back to top]

Area Under the Plasma Concentration-time Curve From Time Zero to Infinity (AUC∞) of Lenalidomide

Area under the plasma concentration-time curve from time zero to infinity (AUC∞) of lenalidomide after a single dose on Day 1. (NCT00812968)
Timeframe: Day 1 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12 and 24 hours post-dose.

Interventionng*h/mL (Geometric Mean)
Lenalidomide878.0

[back to top]

Change From Baseline in Hemoglobin Concentration

Change in hemoglobin concentration from Baseline to the maximum observed value during the major erythroid response period for major erythroid responders. (NCT00812968)
Timeframe: Baseline and from Day1 until the maximum observed value (up to 155 weeks)

Interventiong/dL (Median)
Baseline concentrationMaximum concentration during studyChange from Baseline to maximum value
Lenalidomide7.013.16.0

[back to top]

Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUCt) of Lenalidomide

Area under the plasma concentration-time curve from time zero to the last measurable concentration (AUCt) of lenalidomide after a single dose on Day 1 and multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

Interventionng*h/mL (Geometric Mean)
Lenalidomide: Day 1718.4
Lenalidomide: Day 4803.5

[back to top]

Area Under the Plasma Concentration-time Curve Over the Dosing Interval (AUCτ) of Lenalidomide

Area under the plasma concentration-time curve over the dosing interval (AUCτ) of lenalidomide after a single dose on Day 1 and multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, 12 and 24 hours post-dose.

Interventionng*h/mL (Geometric Mean)
Lenalidomide: Day 1866.5
Lenalidomide: Day 4877.9

[back to top]

Terminal Half-life (T1/2) of Lenalidomide

The apparent terminal half-life is the time required for plasma concentration to decrease by 50% after pseudo-equilibrium of distribution has been reached, and calculated as the natural logarithm of 2 (0.693) / Apparent terminal rate constant (λz). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

Interventionhours (Geometric Mean)
Lenalidomide: Day 13.26
Lenalidomide: Day 43.57

[back to top]

Time to Maximum Plasma Concentration (Tmax) of Lenalidomide

Time to maximum observed plasma concentration of lenalidomide after a single dose on Day 1 and multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

Interventionhours (Median)
Lenalidomide: Day 12.52
Lenalidomide: Day 42.93

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Lenalidomide

Maximum observed plasma concentration of lenalidomide after a single dose on Day and after multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

Interventionng/mL (Geometric Mean)
Lenalidomide: Day 1136
Lenalidomide: Day 4149

[back to top]

Number of Participants With a Erythroid Response

"Erythroid response was determined using the International Working Group (IWG) 2000 criteria, categorized as a major response or minor response.~A major response in patients with transfusion-dependent anemia (receiving ≥ 4.5 units of red blood cell (RBC) transfusion during 56 consecutive days at Baseline) is defined as RBC transfusion independence accompanied by a ≥1.0 g/dL increase from Baseline in hemoglobin sustained for 56 days consecutively during the treatment period. In patients with transfusion-independent anemia with hemoglobin < 10 g/dL at Baseline a major response is defined as a > 2.0 g/dL increase from Baseline in hemoglobin sustained for consecutive 56 days.~Minor response in patients with transfusion-dependent anemia defined as ≥ 50% decrease from Baseline in transfusion requirements sustained for consecutive 56 days, and in transfusion-independent patients as 1.0 to 2.0 g/dL increase from Baseline in hemoglobin sustained for consecutive 56 days." (NCT00812968)
Timeframe: Response was assessed every 28 days through Week 156.

Interventionparticipants (Number)
Erythroid responders (major or minor)Major erythroid responders
Lenalidomide1111

[back to top]

Duration of Erythroid Response

Duration of erythroid response was calculated as the time from the start of the first major or minor erythroid response to the end of the response. Similarly, duration of major erythroid response was calculated as the time from the start of the first major erythroid response to the end of the response. Response duration was censored at the last adequate assessment for patients who maintained response. (NCT00812968)
Timeframe: From the first dose of study drug through Week 156

Interventionweeks (Median)
Duration of erythroid response (major or minor)Duration of major erythroid response
Lenalidomide76.672.1

[back to top]

Number of Participants With a Cytogenetic Response

"Cytogenetic (chromosome structure) abnormalities were assessed by a central cytogenetic reviewer based on prints and cytogenetic reports of the bone marrow sample from the central laboratory. Cytogenetic response was determined using the IWG (2000) criteria and categorized as either a major response or minor response. Twenty metaphases were analyzed for the determination of cytogenetic response.~A major response was defined as no detectable cytogenetic abnormality, if an abnormality was present at Baseline, sustained for consecutive 56 days during the treatment period. A minor response was defined as ≥ 50% reduction from Baseline in abnormal metaphases sustained for consecutive 56 days during the treatment period." (NCT00812968)
Timeframe: Response was assessed every 12 weeks through Week 156

Interventionparticipants (Number)
Major responseMinor response
Lenalidomide15

[back to top]

Apparent Terminal Elimination Rate Constant of Lenalidomide

Apparent terminal elimination rate constant of lenalidomide determined after a single dose on Day 1 and multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

Intervention1/h (Geometric Mean)
Lenalidomide: Day 10.213
Lenalidomide: Day 40.194

[back to top]

Number of Participants With a Neutrophil Response

"Neutrophil response was determined using the IWG (2000) criteria. A major response for participants with a Baseline neutrophil count < 1,500/mm^3 is defined as a ≥ 100% increase or a ≥ 500/mm^3 increase, whichever is greater, sustained for consecutive 56 days during the treatment period.~A minor response for participants with a Baseline neutrophil count < 1,500/mm^3 is defined as a ≥ 100% increase, but an absolute increase < 500/mm^3, sustained for consecutive 56 days during the treatment period." (NCT00812968)
Timeframe: Response was assessed every 28 days through Week 156

Interventionparticipants (Number)
Major responseMinor response
Lenalidomide10

[back to top]

Apparent Total Plasma Clearance (CL/F) of Lenalidomide

Apparent total plasma clearance (CL/F) of lenalidomide after a single dose on Day 1 and multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

InterventionmL/minute (Geometric Mean)
Lenalidomide: Day 1189.8
Lenalidomide: Day 4189.9

[back to top]

Apparent Volume of Distribution (VzF) of Lenalidomide

Apparent volume of distribution of lenalidomide after a single dose on Day 1 and multiple doses (Day 4). (NCT00812968)
Timeframe: Days 1 and 4 at predose and 0.5, 1, 1.5, 2, 3, 4, 6, 9, and 12 hours post-dose.

Interventionliters (Geometric Mean)
Lenalidomide: Day 153.6
Lenalidomide: Day 458.6

[back to top]

Median Overall Survival (OS)

Overall Survival (OS), defined for those patients who have achieved CR or CRi as the time from study entry to disease progression, relapse or death due to any cause, whichever is earlier, to be analyzed similarly. Descriptive analysis was planned for this measure. (NCT00831766)
Timeframe: Up to 24 Months

Interventionmonths (Median)
All Participants Treated at MTD11.22

[back to top] [back to top]

Median Progression-Free Survival (PFS)

Progression-free survival (PFS), defined as the time from study entry to disease progression, relapse, or death due to any cause, whichever is earlier, will be summarized with the Kaplan-Meier curve. (NCT00831766)
Timeframe: 24 months

Interventionmonths (Median)
All Participants Treated at MTD7.55

[back to top] [back to top]

Phase II: Complete Response Rate of Participants Treated at Maximum Tolerated Dose (MTD)

Percentage of participants achieving CR/CRi. Complete Response (CR) plus Complete Response with Incomplete Count Recovery (CRi) rates. Response rates (CR + CRi) of lenalidomide following idarubicin and cytarabine induction therapy in older patients with previously untreated AML. A CR designation requires that the patient achieve the morphologic leukemia-free state and have an absolute neutrophil count of more than 1,000/μL and platelets of 100,000/μL. CRi: After chemotherapy, patients fulfill all of the criteria for CR except for residual neutropenia (1,000/μL) or thrombocytopenia (100,000/μL). (NCT00831766)
Timeframe: 24 months

Interventionpercentage of participants (Number)
All Participants Treated at MTD54

[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

The length of time, in months, that patients were alive from their first date of protocol treatment until worsening of their disease (NCT00837031)
Timeframe: 18 months

Interventionmonths (Median)
Lenalidomide/Gemcitabine2.3

[back to top]

Six-Month Overall Survival (OS) Probability, the Percentage of Patients Estimated to be Alive Six Months After Beginning Protocol Treatment

The percentage of patients who were alive 6 months after beginning treatment (NCT00837031)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Lenalidomide/Gemcitabine37

[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

Length of time, in months, that patients were alive from their first date of protocol treatment until death (NCT00837031)
Timeframe: 18 months

Interventionmonths (Median)
Lenalidomide/Gemcitabine4.7

[back to top]

Time to Major Erythroid Response (MER)

"Time to major erythroid response (MER) is defined in responders as the time from randomization to the documented date of MER.~For transfusion independent patients, the date of MER is the first date of the elevation in hemoglobin level of more than 2 g/dL that has been sustained for at least 8 weeks. For transfusion dependent patients, the date of MER is the beginning date of the time interval of transfusion independence that has been sustained for at least eight weeks." (NCT00843882)
Timeframe: Assessed every cycle during treatment and then 3 and 6 months after last protocol treatment

Interventionmonths (Median)
Arm A (Lenalidomide; Randomization)3.6
Arm B (Lenalidomide + Epoetin Alfa; Randomization)3.7

[back to top]

Proportion of Patients With Minor Erythroid Response

The definition of minor erythroid response: the mean hemoglobin is sustained 1.0 to 2.0 g/dL above the baseline value for a minimum of 8 weeks; or a 50% or greater decrease in 8-week red blood cell transfusion requirements compared to baseline. (NCT00843882)
Timeframe: Assessed every cycle during treatment and after completion of 16 weeks of treatment

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Randomization)0.208
Arm B (Lenalidomide + Epoetin Alfa; Randomization)0.182

[back to top]

Proportion of Patients With Major Erythroid Response (MER) to Salvage Combination Therapy

Major erythroid response is defined as transfusion-independence for ≥ 8 consecutive weeks for patients who were red blood cell transfusion-dependent at baseline AND a ≥ 1 g/dL hemoglobin rise compared to mean pre-transfusion baseline value; or a > 2 g/dL rise in hemoglobin without transfusion for non-transfusion dependent patients. (NCT00843882)
Timeframe: Assessed after completion of 16 weeks of treatment

Interventionproportion of participants (Number)
Arm B (Lenalidomide + Epoetin Alfa; Crossover)0.25

[back to top]

Proportion of Patients With Major Erythroid Response (MER) Among Those With Chromosome 5q31.1 Deletion

Major erythroid response is defined as transfusion-independence for ≥ 8 consecutive weeks for patients who were red blood cell transfusion-dependent at baseline AND a ≥ 1 g/dL hemoglobin rise compared to mean pre-transfusion baseline value; or a > 2 g/dL rise in hemoglobin without transfusion for non-transfusion dependent patients. (NCT00843882)
Timeframe: Assessed after completion of 16 weeks of treatment

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Chromosome 5q31.1 Deletion)0.632

[back to top]

Proportion of Patients With Major Erythroid Response (MER)

Major erythroid response is defined as transfusion-independence for ≥ 8 consecutive weeks for patients who were red blood cell transfusion-dependent at baseline AND a ≥ 1 g/dL hemoglobin rise compared to mean pre-transfusion baseline value; or a > 2 g/dL rise in hemoglobin without transfusion for non-transfusion dependent patients. (NCT00843882)
Timeframe: Assessed after completion of 16 weeks of treatment

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Randomization)0.115
Arm B (Lenalidomide + Epoetin Alfa; Randomization)0.283

[back to top]

Proportion of Patients With Cytogenetic Response

"Evaluation of cytogenetic response requires 20 analyzable metaphases when using conventional techniques. Analysis of data will require 20 metaphases before and after treatment, which must be done on bone marrow only (peripheral blood is not a substitute). Fluorescent in situ hybridization (FISH) may be used as a supplement to follow a specifically defined cytogenetic abnormality, but it is not a substitute for conventional cytogenetic studies. Cytogenetic response is defined as follows: Complete response: Restoration of a normal karyotype in patients with a documented pre-existing clonal (>2 metaphases abnormal) chromosome abnormalities.~Partial response: > 50% reduction in the percentage of bone marrow metaphases with only clonal abnormality." (NCT00843882)
Timeframe: Assessed at baseline and after completion of 16 weeks of treatment

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Randomization)0.07
Arm B (Lenalidomide + Epoetin Alfa; Randomization)0.10

[back to top]

Proportion of Patients With Bone Marrow Response

"Bone marrow response includes complete remission (CR) and partial remission (PR).~Complete remission (CR): Bone marrow showing < 5% myeloblasts with normal maturation of all cell lines, with no evidence for dysplasia. When erythroid precursors constitute < 50% of bone marrow nucleated cells, the percent of blasts is based on all nucleated cells; when there are ≥ 50% erythroid cells, the percent blasts should be based on the non-erythroid cells.~Partial remission (PR): All of the CR criteria (if abnormal prior to treatment), except blasts decreased by 50% over pre-treatment, or a less advanced Myelodysplastic Syndromes (MDS) World Health Organization (WHO) classification than pretreatment. Cellularity and morphology are not relevant." (NCT00843882)
Timeframe: Assessed at 16 weeks

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Randomization)0.01
Arm B (Lenalidomide + Epoetin Alfa; Randomization)0.03

[back to top]

Pretreatment Endogenous Erythropoietin Level

"Pretreatment endogenous erythropoietin level was assessed at baseline. The association between pretreatment endogenous erythropoietin level and major erythroid response was evaluated among patients who received lenalidomide alone.~Major erythroid response is defined as transfusion-independence for ≥ 8 consecutive weeks for patients who were red blood cell transfusion-dependent at baseline AND a ≥ 1 g/dL hemoglobin rise compared to mean pre-transfusion baseline value; or a > 2 g/dL rise in hemoglobin without transfusion for non-transfusion dependent patients." (NCT00843882)
Timeframe: Assessed at baseline and after completion of 16 weeks of treatment

InterventionmU/mL (Median)
Arm A Patients With Major Erythroid Response514.5
Arm A Patients Without Major Erythroid Response150

[back to top]

Duration of Major Erythroid Response (MER)

Duration of major erythroid response (MER) is defined as the time interval between the documented date of MER and the earliest date of resumption of red blood cell transfusions ≥ 2 units in an 8-week period, a reduction in hemoglobin concentration ≥ 2 g/dL in the absence of acute infection, gastrointestinal bleeding and hemolysis, or death. (NCT00843882)
Timeframe: Assessed every cycle during treatment and then 3 and 6 months after last protocol treatment

Interventionmonths (Median)
Arm A (Lenalidomide; Randomization)13
Arm B (Lenalidomide + Epoetin Alfa; Randomization)23.8

[back to top]

Complete Response Rate

Response assessments were made per the NCI working group criteria for CLL (Hallek et al, Blood, 2008). Complete response rate is defined as an achievement of all of the following: Peripheral blood lymphocytes (evaluated by blood and differential count) below 4 × 109/L (4000/μL), absence of significant lymphadenopathy (lymph nodes must be < 1.5 cm), absence of splenomegaly and hepatomegaly, absence of constitutional symptoms, normal blood counts, and bone marrow sample must be at least normocellular for age, with less than 30% of nucleated cells being lymphocytes. Lymphoid nodules should be absent. (NCT00860457)
Timeframe: 3 years

Interventionpercentage of patients (Number)
Chemotherapy36

[back to top]

Response Rate

Number of participants with a complete or partial response according to International Working Group 2006 criteria. (NCT00867308)
Timeframe: 15 weeks

,
InterventionParticipants (Count of Participants)
Complete responsePartial response
Lenalidomide 15 mg00
Lenalidomide 50 mg02

[back to top]

Grade 3-4 Toxicity

Number of participants who experienced at least one grade 3-4 non-hematological toxicity by CTCAE 3.0 that was attributed to lenalidomide. (NCT00867308)
Timeframe: Up to 8 months

InterventionParticipants (Count of Participants)
Lenalidomide 15 mg6
Lenalidomide 50 mg12

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Financial Problems Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Financial Problems Domain Scale was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-10.9
Investigators Choice-2.3

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Global Health Status / QoL Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Global Health Status / QoL Domain to Treatment Scale was scored between 0 and 100, with a higher score representing a higher quality of life. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide4.6
Investigators Choice5.6

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Insomnia Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Insomnia Scale was scored between 0 and 100, with a high score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-12.8
Investigators Choice-7.6

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Nausea and Vomiting Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Nausea and Vomiting Scale was scored between 0 and 100, with a high score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-2.3
Investigators Choice-0.6

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Pain Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Pain Scale was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and time of discontinuation from treatment visit.Up to final data cut-0ff date of 07 March 2014

Interventionunits on a scale (Mean)
Lenalidomide-5.8
Investigators Choice-3.5

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Physical Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide3.4
Investigators Choice-1.8

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Role Functioning Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Role Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide3.1
Investigators Choice5.0

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Social Functioning Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Social Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide5.1
Investigators Choice3.8

[back to top]

Percentage of Participants Who Achieved an Overall Response According to the IRC Central Review

Overall Response Rate (ORR) was defined as the percentage of participants whose best response was Complete Response (CR), Complete Response unconfirmed (CRu) or Partial Response (PR). Participants who discontinued before any response had been observed or changed to other anti-lymphoma treatments before response had been observed, were considered as non-responders. Tumor Response was assessed by a modification of the International Lymphoma Workshop Response Criteria, IWRC, Cheson, 1999; CR is defined as the disappearance of all clinical and radiographic evidence of disease; CRu is defined as a CR, with a 1) residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters (SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2) indeterminate bone marrow; PR = is defined ≥50% decrease in 6 largest nodes or nodal masses. (NCT00875667)
Timeframe: From date of randomization to the data cut-off date of 07 March 2014; median treatment duration was 24.3 weeks for the lenalidomide arm and 13.1 weeks for the investigators choice arm

InterventionPercentage of Participants (Number)
Lenalidomide40.0
Investigators Choice10.7

[back to top]

Percentage of Participants Who Achieved an Overall Response as Assessed by the Investigator at the Final Analysis

Overall Response Rate (ORR) was defined as the percentage of participants whose best response was Complete Response, Complete Response unconfirmed or Partial Response. Participants who had discontinued before any response has been observed or changed to other anti-lymphoma treatments before response had been observed, were considered as non-responders. Tumor Response was assessed by a modification of the International Lymphoma Workshop Response Criteria, IWRC, Cheson, 1999; CR is defined as the disappearance of all clinical and radiographic evidence of disease; CRu is defined as a CR, with a 1) residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters (SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2) indeterminate bone marrow; PR = is defined ≥50% decrease in 6 largest nodes or nodal masses. (NCT00875667)
Timeframe: From date of randomization to the study discontinuation date of 09 October 2018; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm

InterventionPercentage of Participants (Number)
Lenalidomide45.9
Investigators Choice22.6

[back to top]

Percentage of Participants With a Complete Response, Unconfirmed Complete Response, Partial Response and Stable Disease According to the IRC Central Review

Tumor control rate was defined as the percentage of participants with a complete response (CR), unconfirmed complete response (CRu), partial response (PR) and stable disease (SD). Tumor Response was assessed by a modification of the International Lymphoma Workshop Response Criteria, IWRC, Cheson, 1999); CR is defined as the disappearance of all clinical and radiographic evidence of disease; CRu is defined as a CR, with a 1) residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters (SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2) indeterminate bone marrow; PR = is defined ≥50% decrease in 6 largest nodes or nodal masses. Stable disease (SD) is defined as less than a PR (see above) but is not progressive disease or relapsed disease. (NCT00875667)
Timeframe: From date of randomization to the data cut-off date of 07 March 2014; median treatment duration was 24.3 weeks for the lenalidomide arm and 13.1 weeks for the investigators choice arm

InterventionPercentage of Participants (Number)
Lenalidomide69.4
Investigators Choice63.1

[back to top]

Percentage of Participants With a Complete Response, Unconfirmed Complete Response, Partial Response and Stable Disease at the Final Analysis

Tumor control rate was defined as the percentage of participants with a complete response (CR), unconfirmed complete response (CRu), partial response (PR) and stable disease (SD). Tumor Response was assessed by a modification of the International Lymphoma Workshop Response Criteria, IWRC, Cheson, 1999); CR is defined as the disappearance of all clinical and radiographic evidence of disease; CRu is defined as a CR, with a 1) residual lymph node mass >1.5 cm that has decreased by 75% in the sum of the product of the diameters (SPD). Individual nodes previously confluent decreased by more than 75% in the SPD compared with original mass; 2) indeterminate bone marrow; PR = is defined ≥50% decrease in 6 largest nodes or nodal masses. Stable disease (SD) is defined as less than a PR (see above) but is not progressive disease or relapsed disease. (NCT00875667)
Timeframe: From date of randomization to the discontinuation date of 09 October 2018; median treatment duration was 24.3 weeks for the lenalidomide arm and 13.1 weeks for the investigators choice arm

InterventionPercentage of participants (Number)
Lenalidomide70.0
Investigators Choice65.5

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Appetite Loss Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Appetite Loss Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice16.2-0.85.1-12.5-11.15.4
Lenalidomide18.12.51.9-2.3-4.34.8

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Cognitive Functioning Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Cognitive Functioning Domain ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice83.6-2.31.34.25.6-2.3
Lenalidomide84.60.0-1.9-3.2-2.5-5.1

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Constipation

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Constipation Domain was scored between 0 and 100, with a high score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice8.6-0.81.30.00.00.8
Lenalidomide12.56.34.23.5-0.710.2

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Diarhoea

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Diarhoea Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice12.6-3.11.3-4.20.00.0
Lenalidomide15.7-3.5-4.22.4-2.11.6

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Dyspnoea Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Dyspnoea Domain was scored between 0 and 100, with a high score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice21.2-0.80.04.25.60.8
Lenalidomide26.5-1.6-1.4-2.91.46.0

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Fatigue Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Fatigue Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice39.22.13.4-6.9-7.42.6
Lenalidomide40.20.1-3.2-1.0-3.95.2

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Financial Problems Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Financial Problems Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice10.8-0.8-3.8-4.2-5.61.6
Lenalidomide19.5-7.0-7.0-2.9-9.2-4.3

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Cognitive Functioning Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Cognitive Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide3.2
Investigators Choice2.9

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Insomnia Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Insomnia Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice25.7-4.7-6.4-16.7-16.70.8
Lenalidomide29.4-7.6-5.2-1.8-7.1-3.2

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Nausea / Vomiting Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Nausea and Vomiting Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice3.80.45.82.12.86.6
Lenalidomide4.92.52.65.3-0.70.5

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Pain Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Pain Domain was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineCycle 3 Day 1Cycle 5 Day 1Cycle 7 Day 1Cycle 9 Day 1Treatment Discontinuation
Investigators Choice13.7-1.2-2.60.0-2.83.5
Lenalidomide22.6-2.2-0.23.2-3.24.6

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Physical Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice78.9-3.7-2.14.211.1-5.1
Lenalidomide71.8-0.51.62.42.8-5.6

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Social Functioning Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Social Functioning Domain ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice78.4-1.2-4.52.10.0-2.7
Lenalidomide74.9-1.01.6-1.54.3-5.1

[back to top]

Number of Participants With Treatment Emergent Adverse Events

Adverse events were assessed using National Cancer Institute, Common Terminology Criteria for Adverse Events (NCI CTCAE), Version 3: according to the following scale: Grade 1 = Mild Adverse Event (AE), Grade 2 = Moderate AE, Grade 3 = Severe and Undesirable AE, Grade 4 = Life-threatening or Disabling AE, and Grade 5 = Death; Serious AEs (SAEs) are those that resulted in death, were life-threatening, required or prolonged inpatient hospitalization, resulted in persistent or significant disability/incapacity, congenital anomaly, or resulted in an important medical event that may have jeopardized the patient or required medical or surgical intervention to prevent one of the outcomes listed above. after the first dose of study drug and within 28 days after the last dose. A Treatment Emergent Adverse event (TEAE) is defined as any AE occurring or worsening on or after the first dose of study drug and within 28 days after the last dose of study drug. (NCT00875667)
Timeframe: From the date of the first dose of study drug to 28 days after the last dose, up to the study discontinuation date of 09 October 2018; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigators choice arm

,
InterventionParticipants (Count of Participants)
Any TEAEAny TEAE Grade 3 AEAny TEAE Grade 4 AEAny TEAE Grade 5 AEAny TEAE Related to the IPAny Grade 3 AE Related to IPAny Grade 4 AE Related to IPAny Grade 5 AE Related to IPAny Serious Adverse Event (SAE)Any SAE Related to IPAny TEAE Leading to Stopping of IPAny Treatment Related AE Leading to Stopping IPTEAE Leading to Dose Reduction/InterruptionRelated AE Leading to Dose Reduct/InterruptionTEAE Leading to Dose ReductionRelated AE Leading to Dose ReductionTEAE Leading to Dose InterruptionRelated AE Leading to Dose Interruption
Investigators Choice694929251361902212147332913102825
Lenalidomide159126561514110646075383118114103726911098

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Emotional Domain ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice78.51.31.3-3.11.4-1.5
Lenalidomide73.73.43.68.14.5-1.3

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Role Functioning Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Role Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice73.93.5-6.40.013.9-4.3
Lenalidomide71.5-4.81.40.31.8-9.1

[back to top]

Kaplan Meier Estimate for Duration of Response (DOR) According to the IRC Central Review

Duration of response was defined as the time from when the first response of CR, CRu, or PR was first achieved until documented tumor progression, or until the participant died from any cause, whichever occurred first. Participants who did not progress or die at the time of analysis were censored at the last assessment date that the participant was known to be progression-free. Participants who received a new treatment without documented progression were censored at the last assessment date that the participant was known to be progression-free. (NCT00875667)
Timeframe: From date of randomization to the data cut-off date of 07 March 2014; median study duration was 70.7 weeks for the lenalidomide arm and 69.3 weeks for the investigators choice arm

InterventionWeeks (Median)
Lenalidomide69.6
Investigators Choice45.1

[back to top]

Kaplan Meier Estimate for Duration of Response as Assessed by the Investigator at the Final Analysis

Duration of response was defined as the time from when the first response of CR, CRu, or PR was first achieved until documented tumor progression, or until the participant died from any cause, whichever occurred first. Participants who did not progress or die at the time of analysis were censored at the last assessment date that the participant was known to be progression-free. Participants who received a new treatment without documented progression were censored at the last assessment date that the participant was known to be progression-free. (NCT00875667)
Timeframe: From date of randomization to the study discontinuation date of 09 October 2018; median study duration was 103.9 weeks for lenalidomide and 87.0 weeks for the investigator choice arm

InterventionWeeks (Median)
Lenalidomide70.1
Investigators Choice91.7

[back to top]

Kaplan Meier Estimate for Overall Survival (OS) According to the IRC Central Review

Overall survival was defined as the time from randomization until death from any cause. Participants alive or lost to follow-up at the time of analysis were censored at the last date they were known to be alive. (NCT00875667)
Timeframe: From date of randomization to the data cut-off date of 07 March 2014; overall median follow-up was 93.9 weeks

Interventionweeks (Median)
Lenalidomide121.0
Investigators Choice91.7

[back to top]

Kaplan Meier Estimate for Overall Survival as Assessed by the Investigator at the Final Analysis

Overall survival was defined as the time from randomization until death from any cause. Participants alive or lost to follow-up at the time of analysis were censored at the last date they were known to be alive. (NCT00875667)
Timeframe: From randomization to progression of disease or death; up to the study discontinuation date of 09 October 2018; overall median follow-up time was 285 weeks

Interventionweeks (Median)
Lenalidomide120.6
Investigators Choice91.7

[back to top]

Kaplan Meier Estimate for Progression Free Survival (PFS) by Independent Review Committee (IRC) Central Review

PFS was defined as time of randomization to the first observation of disease progression or death due to any cause, whichever was first. If a participant had not progressed or died, PFS was censored at the time of last assessment when the participant was known not to have progressed. For participants who received other anti-lymphoma therapy with no evidence of progression, PFS was censored at time of last tumor assessment with no evidence of progression prior to the start of new anti-lymphoma treatment. (NCT00875667)
Timeframe: From randomization to progression of disease or death; up to data cut off date of 07 March 2014; overall median follow-up time was 93.9 weeks

Interventionweeks (Median)
Lenalidomide37.6
Investigators Choice22.7

[back to top]

Kaplan Meier Estimate for Progression Free Survival by Investigator's Assessment at the Final Analysis

Kaplan Meier estimates of PFS were defined as the time from randomization to the first observation of disease progression or death due to any cause, whichever was first. If a participant had not progressed or died, PFS was censored at the time of last completed assessment when the participant was known not to have progressed. For participants who received other anti-lymphoma therapy with no evidence of progression, PFS was censored at time of last tumor assessment with no evidence of progression prior to the start of new anti-lymphoma treatment. (NCT00875667)
Timeframe: From randomization to progression of disease or death; up to study discontinuation of 09 October 2018; overall median follow-up time was 285 weeks

Interventionweeks (Median)
Lenalidomide37.3
Investigators Choice23.6

[back to top]

Kaplan Meier Estimate of Time to First Response (TTFR) According to the IRC Central Review

Time to Response was defined as the time from first dose of study drug to the date of the first response (having at least a PR) and was calculated only for responding participants. ). Participants with progression at the time of analysis were censored at the first assessment date that the participant was known to have progressed. Participants with SD at the time of analysis were censored at the last assessment date that the participant was known to be progression-free. (NCT00875667)
Timeframe: From randomization of study drug to time of first documented PR or better response; up to data cut-off date of 07 March 2014; median treatment duration was 24.3 weeks for the lenalidomide arm and 13.1 weeks for the investigators choice arm

InterventionWeeks (Median)
Lenalidomide18.7
Investigators ChoiceNA

[back to top]

Kaplan Meier Estimate of Time to First Response as Assessed by the Investigator at the Final Analysis

Time to first response was defined as the time from first dose of study drug to the date of the first response (having at least a PR). Participants with progression at the time of analysis were censored at the first assessment date that the participant was known to have progressed. Participants with SD at the time of analysis were censored at the last assessment date that the subject was known to be progression-free. (NCT00875667)
Timeframe: From date of randomization to the study discontinuation date of 09 October 2018; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm

InterventionWeeks (Median)
Lenalidomide23.9
Investigators Choice40.0

[back to top]

Kaplan Meier Estimate of Time to Progression According to the IRC Central Review

Time to progression (TTP) was defined as the time from randomization until objective tumor progression. Time to progression did not include deaths. Participants without progression at the time of analysis were censored at the last assessment date that the participant was known to be progression-free. Participants who received a new anti-lymphoma treatment without documented progression were censored at the last assessment date that the participant was known to be progression-free. (NCT00875667)
Timeframe: From date of randomization to the data cut-off date of 07 March 2014; median study duration was 70.7 weeks for the lenalidomide arm and 69.3 weeks for the investigators choice arm

InterventionWeeks (Median)
Lenalidomide39.3
Investigators Choice24.7

[back to top]

Kaplan Meier Estimate of Time to Progression as Assessed by the Investigator at the Final Analysis

Time to progression (TTP) was defined as the time from randomization until objective tumor progression. Time to progression did not include deaths. Participants without progression at the time of analysis were censored at the last assessment date that the participant was known to be progression-free. Participants who received a new anti-lymphoma treatment without documented progression were censored at the last assessment date that the participant was known to be progression-free. (NCT00875667)
Timeframe: From date of randomization to the study discontinuation date of 09 October 2018; median study duration was 103.9 weeks for lenalidomide and 87.0 weeks for the investigator choice arm

InterventionWeeks (Median)
Lenalidomide39.3
Investigators Choice24.7

[back to top]

Kaplan Meier Estimate of Time to Treatment Failure (TTF) as Assessed by the Investigator

Time to treatment failure was defined as the time from the first dose of study drug to discontinuation of treatment for any reason, including disease progression assessed by the investigator, treatment toxicity, or death. Participants who were on-treatment or completed the treatment according to the protocol were censored at the last date of drug intake. (NCT00875667)
Timeframe: From the date of the first treatment to the data cut-off date of 07 March 2014; median treatment duration was 24.3 weeks for the lenalidomide arm and 13.1 weeks for the investigators choice arm

Interventionweeks (Median)
Lenalidomide24.4
Investigators Choice17.9

[back to top]

Kaplan Meier Estimate of Time to Treatment Failure as Assessed by the Investigator at the Final Analysis

Time to treatment failure was defined as the time from the first dose of study drug to discontinuation of treatment for any reason, including disease progression assessed by the investigator, treatment toxicity, or death. Participants who were on-treatment or completed the treatment according to the protocol were censored at the last date of drug intake. (NCT00875667)
Timeframe: From date of first dose of treatment to the study discontinuation date of 09 October 2018; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm

Interventionweeks (Median)
Lenalidomide24.4
Investigators Choice17.9

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Appetite Loss Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Appetite Loss Domain to Treatment Scale was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-4.8
Investigators Choice-4.1

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Global Health Status / QoL Domain

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Global Health Status / QoL Domain was scored between 0 and 100, with a higher score representing a higher quality of life. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

,
Interventionunits on a scale (Mean)
BaselineChange from Baseline to Cycle 3 Day 1Change from Baseline to Cycle 5 Day 1Change from Baseline to Cycle 7 Day 1Change from Baseline to Cycle 9 Day 1Change from Baseline to Treatment Discontinuation
Investigators Choice58.42.33.27.38.3-1.0
Lenalidomide59.0-3.4-0.71.04.3-5.8

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Constipation Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Constipation Scale was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-0.3
Investigators Choice-3.5

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Diarhoea Domain to Treatment Discontinuation Visit

"The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact).~The EORTC QLQ-C30 Diarhoea Scale was scored between 0 and 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement." (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and time of discontinuation from treatment visit.Up to final data cut-0ff date of 07 March 2014

Interventionunits on a scale (Least Squares Mean)
Lenalidomide-7.2
Investigators Choice-5.8

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Dyspnoea Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Dyspnoea Domain to Treatment Scale was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-7.3
Investigators Choice-5.8

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Emotional Functioning Scale ranges from 0 to 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide6.9
Investigators Choice3.7

[back to top]

Maximum Change From Baseline in the EORTC QLQ-C30 Fatigue Domain to Treatment Discontinuation Visit

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Fatigue Scale was scored between 0 and 100, with a higher score representing worse symptomatic expression. (NCT00875667)
Timeframe: Baseline known as screening visit, after cycle 2 (C3D1), after Cycle 4 (C5D1), after Cycle 6, (C7D1), after Cycle 8, (C9D1) and at discontinuation; median treatment duration was 24.3 weeks for lenalidomide and 13.1 weeks for the investigator choice arm.

Interventionunits on a scale (Mean)
Lenalidomide-4.9
Investigators Choice-2.9

[back to top]

Event-free Survival (EFS)

Assessed as the median value for EFS 12 months after starting MDR treatment (NCT00890552)
Timeframe: 12 months

Interventionmonths (Median)
Lenalidomide, Melphalan and Dexamethasone (MDR)3.15

[back to top]

Duration of Response

Assessed as the median value for the time from first partial response until progression; death; or last follow-up. (NCT00890552)
Timeframe: 32 months

Interventionmonths (Median)
Lenalidomide, Melphalan and Dexamethasone (MDR)9.1

[back to top]

Overall Survival (OS)

Participants alive 12 months after starting MDR treatment. (NCT00890552)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Lenalidomide, Melphalan and Dexamethasone (MDR)58

[back to top]

Hematologic Response Rate

At the end of each treatment cycle (4 weeks), hematologic response rate as assessed. Hematologic response was considered to be amyloid complete response (normal FLC ratio and negative serum and urine immunofixation); very good partial response (difference between involved and uninvolved FLCs [dFLC] < 40 mg/L); or partial response (dFLC decrease > 50%). (NCT00890552)
Timeframe: 8 weeks

Interventionparticipants (Number)
Complete Response (CR)Very good Partial Response (VGPR)Partial Response (PR)No Response (NR)Response not evaluable
Lenalidomide, Melphalan and Dexamethasone (MDR)24891

[back to top]

Time to CR

CR includes subjects with CR but incomplete recovery of blood counts (CRi). Responses were assessed according to the ELN guidelines. (NCT00890929)
Timeframe: 18 weeks

Interventionweeks (Median)
Azacitidine Followed by Lenalidomide12

[back to top]

Maximum Tolerated Dose (MTD) of Lenalidomide

The maximum tolerated dose (MTD) of lenalidomide was determined in study phase 1, for use in study Phase 2 (not conducted). The outcome is reported as the dose of lenalidomide that represents the MTD. (NCT00890929)
Timeframe: 15 months

Interventionmg/day lenalidomide (oral) (Number)
Azacitidine Followed by Lenalidomide50

[back to top]

Remission Duration

Responses and remission were assessed according to the ELN guidelines. (NCT00890929)
Timeframe: 26 months

Interventionweeks (Median)
Azacitidine Followed by Lenalidomide6

[back to top]

OS of Responders

OS from the start of treatment of responders (per ELN guidelines) was assessed at a median follow up of 88 weeks from the end of treatment (range, 1-120), and was censored at 1 April 2012. (NCT00890929)
Timeframe: 88 weeks (median)

Interventionweeks (Median)
Azacitidine Followed by Lenalidomide69

[back to top]

Overall Response Rate (ORR)

ORR includes subjects with CR, CRi, and partial response (PR). Responses were assessed according to the ELN guidelines. (NCT00890929)
Timeframe: 26 months

Interventionpercentage of subjects (Number)
Azacitidine Followed by Lenalidomide41

[back to top]

Overall Survival (OS)

OS from the start of treatment was assessed at a median follow up of 88 weeks from the end of treatment (range, 1-120), and was censored at 1 April 2012. (NCT00890929)
Timeframe: 88 weeks (median)

Interventionweeks (Median)
Azacitidine Followed by Lenalidomide20

[back to top]

4-week Survival Rate

"Early death was assessed as death within 28 days of the start of treatment" (NCT00890929)
Timeframe: 28 days

Interventionpercentage of subjects remaining alive (Number)
Azacitidine Followed by Lenalidomide83

[back to top]

Compete Remission (CR) Rate

Compete Remission (CR) includes subjects with CR but incomplete recovery of blood counts (CRi). CR was assessed according to the European LeukemiaNet (ELN) guidelines, and is defined as the absence of clonal lymphocytes in the peripheral blood. (NCT00890929)
Timeframe: 12 months

Interventionpercentage of subjects (Number)
Azacitidine Followed by Lenalidomide28

[back to top]

Time to PR

Responses were assessed according to the ELN guidelines. (NCT00890929)
Timeframe: 36 weeks

Interventionweeks (Median)
Azacitidine Followed by Lenalidomide6

[back to top]

Percentage of Participants With GVHD (Graft Versus Host Disease)

Acute grade 2 to 4 Graft versus host disease( GVHD )for patients who were able to be analyzed by measuring the T cell counts for increased CD3+ before and after lenalidomide. (NCT00899431)
Timeframe: Up to 6 months after allotransplant

InterventionParticipants (Count of Participants)
Group 10
Group 23
Group 31

[back to top]

Phase 2 - Number of Subjects Who Are Progression-Free and Alive

"Progression is defined as:~At least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, or the appearance of one or more new lesion(s)." (NCT00903630)
Timeframe: 3 months after starting treatment

Interventionparticipants (Number)
Lenalidomide With Liposomal Doxorubicin8

[back to top]

Phase 1 - Maximum Tolerated Dose (MTD) of Lenalidomide When Combined With Fixed Dose Liposomal Doxorubicin in Women With Recurrent Epithelial Ovarian, Fallopian Tube, and Primary Peritoneal Cancer

The maximum tolerated dose (MTD) reflects the highest dose of Lenalidomide when combined with fixed dose Liposomal Doxorubicin at which no more than one out of 6 participants experiences a dose limiting toxicity (DLT). (NCT00903630)
Timeframe: 1 cycle (28 days)

Interventionmilligrams (mg) (Number)
Lenalidomide With Liposomal Doxorubicin10

[back to top]

Phase 1 - Number of Participants Who Experienced Dose-Limiting Toxicities (DLTs)

DLT is defined as the inability to complete cycle 1 and/or begin cycle 2 within 7 days of the planned start due to a grade 4 or greater hemtologic toxicity or a grade 3 or greater non-hematologic toxicity. Grading was based on Common Toxicity Criteria (CTC) Version 4. (NCT00903630)
Timeframe: within 5 weeks of starting treatment

Interventionparticipants (Number)
Phase 1 - Dose Level 10
Phase I - Dose Level 22

[back to top]

Phase 2 - Number of Subjects Achieving a Partial or Complete Response

"Partial response is defined as:~At least a 30% decrease in the sum of the longest diameters of target lesions, taking as reference the baseline sum longest diameter. To be assigned a status of partial response, changes in tumor measurements must be confirmed by repeat assessments performed no less than four weeks after the criteria for response are first met.~Complete response is defined as:~The disappearance of all target lesions. To be assigned a status of complete response, changes in tumor measurements must be confirmed by repeat assessments performed no less than four weeks after the criteria for response are first met." (NCT00903630)
Timeframe: 3 months after starting treatment

Interventionparticipants (Number)
Phase 21

[back to top]

Phase 2 - Number of Subjects Who Are Progression-Free and Alive

"Progression is defined as:~At least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurement, or the appearance of one or more new lesion(s)." (NCT00903630)
Timeframe: 6 months after starting treatment

Interventionparticipants (Number)
Lenalidomide With Liposomal Doxorubicin5

[back to top]

Overall Survival

Is defined as the time interval from start of treatment to the date of death due to any cause. In the absence of confirmation of death (including subjects lost to follow-up), survival time will be censored at the last date the subject is known to be alive (NCT00908232)
Timeframe: At each visit from baseline to end of treatment. After treatment, monthly visit until progression or relapse or until the start of alternative MMY therapy. Further follow up by monthly phone call until the last patient was treated and followed for 1 year

Interventiondays (Median)
Complete to Partial Response: Bortezomib + DexamethasoneNA
Stable Disease After 4 Cycles: VD, VDC, VDLNA

[back to top]

Time to Progression

Is calculated as the time from start of treatment to the date of the first observation of disease progression or relapse from CR. Deaths owing to causes other than progression not counted, but censored. Subjects who withdraw from the study or die will be censored at the time of last disease assessment. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0). (NCT00908232)
Timeframe: At Day 1 of each treatment cycle, at the End of Treatment visit until there is evidence of Progressive Disease or relapse from Complete Response (CR), Median Follow-up of 16.9 months

Interventiondays (Median)
Complete to Partial Response: Bortezomib + Dexamethasone366.0
Stable Disease After 4 Cycles: VD, VDC, VDL214.0

[back to top]

Progression Free Survival

"Time from start of treatment to date of disease progression, relapse from CR or death. Estimated using the kaplan-meier method. 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, or similar definition as accurate and appropriate." (NCT00908232)
Timeframe: At Day 1 of each treatment cycle, at the End of Treatment visit until there is evidence of Progressive Disease or relapse from Complete Response (CR). Median Follow-Up of 16.9 months

Interventiondays (Median)
Complete to Partial Response: Bortezomib + Dexamethasone311.0
Stable Disease After 4 Cycles: VD, VDC, VDL214.0

[back to top]

Median Time to First Confirmed Response

Time from start of treatment to the date of the first documentation of a confirmed response. Estimated using the Kaplan-Meier method. Response was assessed using the International Myeloma Working Group (IMWG) Uniform Response Criteria and validated by an Independent Monitoring Committee. (NCT00908232)
Timeframe: At Day 1 of each treatment cycle, at the End of Treatment visit until there is evidence of Progressive Disease or relapse from Complete Response (CR), up to 168 days

Interventiondays (Median)
Complete to Partial Response: Bortezomib + Dexamethasone43.0
Stable Disease After 4 Cycles: VD, VDC, VDLNA

[back to top]

One Year Survival

Percent Probability of Survival at 1 year from the start of treatment, estimated using Kaplan-Meier analysis. (NCT00908232)
Timeframe: At each visit from baseline to end of treatment. After treatment, monthly visit until progression or relapse or until the start of alternative MMY therapy, up to 1 year

Interventionpercent probability (Number)
Complete to Partial Response: Bortezomib + Dexamethasone80
Stable Disease After 4 Cycles: VD, VDC, VDL89

[back to top]

Overall Best Confirmed Response

Overall Best Confirmed Response is the best Overall Response Rate with borezomib-dexamathasone (+/-cyclophosphamide or lenalidomide) recorded between baseline and end of treatment. Response was assessed using the International Myeloma working Group (IMWG) Uniform Response Criteria and validated by an Independent Monitoring Committee. (NCT00908232)
Timeframe: Prior to treatment at day 1 of each cycle and at the end of treatment (day 21 of cycle 8), up to 168 days

Interventionnumber of participants (Number)
Complete to Partial Response: Bortezomib + Dexamethasone101
Stable Disease After 4 Cycles: VD, VDC, VDL6

[back to top]

PK Phase: Area-under-the Concentration-time Curve (AUC0-24) for Lenalidomide

Area under the plasma concentration-time curve from Time 0 to 24 hours post-dose for lenalidomide after a single dose, calculated using the log-linear trapezoidal method. (NCT00910858)
Timeframe: On Day -7 blood samples were taken at predose (0 hour), 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 10, 12, and 24 hours post-dose.

Interventionng*h/mL (Geometric Mean)
10 mg Lenalidomide817

[back to top]

Monotherapy Phase: Area-under-the Concentration-time Curve (AUC0-5) for Lenalidomide

Area under the plasma concentration-time curve from Time 0 to 5 hours postdose for lenalidomide (its R- and S- enantiomers and the enantiomers combined) after multiple dosing for 14 days, calculated using the log-linear trapezoidal method. (NCT00910858)
Timeframe: On Day 14 blood samples were taken at predose (0 hour), 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, and 5 hours postdose.

Interventionng*h/mL (Geometric Mean)
Total LenalidomideS-LenalidomideR-Lenalidomide
10 mg Lenalidomide563315248

[back to top]

Monotherapy Phase: Maximum Plasma Concentration of Lenalidomide (Cmax)

The Maximum observed plasma concentration (Cmax) of lenalidomide (its R- and S- enantiomers and the enantiomers combined) after multiple dosing for 14 days. (NCT00910858)
Timeframe: On Day 14 blood samples were taken at predose (0 hour), 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, and 5 hours postdose.

Interventionng/mL (Geometric Mean)
Total LenalidomideS-LenalidomideR-Lenalidomide
10 mg Lenalidomide18510480.7

[back to top]

Percentage of Participants Overall With Erythroid Response by Baseline Erythropoietin Level

To evaluate the predictive value of pretreatment serum erythropoietin (EPO) concentration for erythroid response to lenalidomide, the percentage of erythroid responders versus non-responders were stratified by Baseline EPO levels (≤ 500 mIU/mL versus > 500 mIU/mL). Response includes participants with either a major or minor response. (NCT00910858)
Timeframe: Assessed every 28 days until study discontinuation (up to 1218 days)

,,
Interventionpercentage of participants (Number)
Baseline EPO ≤ 500 mIU/mLBaseline EPO > 500 mIU/mL
Non-responders47.839.1
Overall53.838.5
Responders62.537.5

[back to top]

Percentage of Participants With a Erythroid Response Across All Phases

Erythroid response was categorized as either a major response or a minor response. A major response was defined as red blood cell (RBC) transfusion independence during any consecutive 56-day period and an increase in hemoglobin of at least 1.5 g/dL. A minor response was defined as a ≥ 50% or ≥ 4 unit decrease in RBC transfusions from pretreatment requirements (the number of RBC transfusions required over an 8-week period before the start of study drug treatment). (NCT00910858)
Timeframe: Assessed every 28 days until study discontinuation (up to 1218 days).

,,
Interventionpercentage of participants (Number)
Major responseMinor response
10 mg Del 5q85.70
10 mg Non-del 5q17.65.9
15 mg Non-del 5q40.00

[back to top]

PK Phase: Maximum Plasma Concentration of Lenalidomide (Cmax)

The maximum observed plasma concentration (Cmax) of lenalidomide (its R- and S- enantiomers and the enantiomers combined) after a single dose on day -7. (NCT00910858)
Timeframe: On Day -7 blood samples were taken at predose (0 hour), 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 10, 12, and 24 hours postdose.

Interventionng/mL (Geometric Mean)
Total LenalidomideS-LenalidomideR-Lenalidomide
10 mg Lenalidomide17910178.3

[back to top]

PK Phase: Percent of Administered Lenalidomide Excreted Over 24 Hours After a Single, Oral Dose

"Percent of the administered dose of lenalidomide excreted unchanged in urine over 24 hours postdose after a single dose on Day -7, calculated as:~(amount excreted unchanged in urine over 24 hours postdose / Dose) * 100.~The dose was 10 mg for total lenalidomide and 5 mg for the enantiomers." (NCT00910858)
Timeframe: On Day -7 at predose and over the intervals of 0-5, 5-8, 8-12, and 12-24 hours postdose.

Interventionpercent of administered dose (Geometric Mean)
Total LenalidomideS-LenalidomideR-Lenalidomide
10 mg Lenalidomide65.167.962.2

[back to top]

PK Phase: Terminal Half-life (t1/2)

The apparent terminal half-life is the time required for plasma concentration to decrease by 50% after pseudo-equilibrium of distribution has been reached, and calculated as the natural logarithm of 2 (0.693) / Apparent terminal rate constant (λz). (NCT00910858)
Timeframe: On Day -7 blood samples were taken at predose (0 hour), 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 10, 12, and 24 hours postdose.

Interventionhours (Geometric Mean)
Total LenalidomideS-LenalidomideR-Lenalidomide
10 mg Lenalidomide3.724.143.58

[back to top]

Monotherapy Phase: Percent of Lenalidomide Excreted Over 5 Hours Post Day 14 Dose

"Percent of the administered lenalidomide dose excreted unchanged in urine over 5 hours postdose after multiple dosing for 14 days, calculated as:~(amount excreted unchanged in urine over the first 5 hours postdose / Dose) * 100.~The dose was 10 mg for total lenalidomide and 5 mg for the enantiomers." (NCT00910858)
Timeframe: On Day 14, at predose and over the interval of 0-5 hours postdose.

Interventionpercent of administered dose (Geometric Mean)
Total LenalidomideS-LenalidomideR-Lenalidomide
10 mg Lenalidomide34.035.432.5

[back to top]

Time to Grade 4 Neutropenia or Thrombocytopenia

Time to the first event of grade 4 neutropenia or thrombocytopenia, graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 3.0, was calculated as date of first event - date of first dose + 1. (NCT00910858)
Timeframe: From the date of first dose until 30 days after the last dose (up to 1218 days)

,
Interventiondays (Median)
Grade 4 NeutropeniaGrade 4 Thrombocytopenia
Del 5q28.029.0
Non-del 5q69.053.0

[back to top]

Number of Participants With Adverse Events With a Later Cut-off Date of 31 March 2014

AEs = any noxious, unintended, or untoward medical occurrence that may appear or worsen during the course of a study. It may be a new intercurrent illness, a worsening concomitant illness, an injury, or any concomitant impairment of the participant's health, regardless of cause. Serious AE (SAE) = any AE which results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. The severity of AEs were graded based upon the participants symptoms according to the Common Terminology Criteria for Adverse Events (CTCAE, Version 4.0); AEs were evaluated for severity according to the following scale: Grade 1 = Mild - transient or mild discomfort; no medical intervention required; Grade 2 = Moderate - mild to moderate limitation in activity; Grade 3 = Severe; Grade 4 = Life threatening; Grade 5 = Death (NCT00910910)
Timeframe: From randomization to the data cut-off date of 31 March 2014; Up to 53 months; maximum duration of exposure for Lenalidomide was 1140 days and 406 days for Chlorambucil

,
Interventionparticipants (Number)
≥ 1 TEAE≥ 1 TEAE related to study drug≥ 1 NCI CTC Grade 3-4 TEAEGrade 3-4 adverse event related to any study drug≥ 1 NCI CTC Grade 5 TEAE≥ Grade 5 adverse event related to any study drug≥ 1 Serious TEAE≥ 1 Serious TEAE related to any study drug≥1 TEAE leading to stopping either study drug≥1 Related TEAE leading to stopping either drug
Chlorambucil2021551319011190534223
Lenalidomide2161941881572161481077046

[back to top]

Number of Participants With Adverse Events (AEs)

AEs = any noxious, unintended, or untoward medical occurrence that may appear or worsen during the course of a study. It may be a new intercurrent illness, a worsening concomitant illness, an injury, or any concomitant impairment of the participant's health, regardless of cause. Serious AE (SAE) = any AE which results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. The severity of AEs were graded based upon the participants symptoms according to the Common Terminology Criteria for Adverse Events (CTCAE, Version 4.0); AEs were evaluated for severity according to the following scale: Grade 1 = Mild - transient or mild discomfort; no medical intervention required; Grade 2 = Moderate - mild to moderate limitation in activity; Grade 3 = Severe; Grade 4 = Life threatening; Grade 5 = Death (NCT00910910)
Timeframe: From randomization up to data cut-off of 18 Feb 2013; Up to approximately 39 months; maximum duration of exposure for Lenalidomide was 1086 days and 406 days for Chlorambucil

,
Interventionparticipants (Number)
≥ 1 TEAE≥ 1 TEAE related to study drug≥ 1 NCI CTC Grade 3-4 TEAEGrade 3-4 adverse event related to any study drug≥ 1 NCI CTC Grade 5 TEAE≥ Grade 5 adverse event related to any study drug≥ 1 Serious TEAE≥ 1 Serious TEAE related to any study drug≥1 TEAE leading to stopping either study drug≥1 Related TEAE leading to stopping either drug
Chlorambucil186139117829176463419
Lenalidomide202183173143216129956139

[back to top]

Number of Participants and Types of Subsequent Anti-cancer Therapies Received Post Treatment

Subsequent anti-cancer therapies administered to participants following the discontinuation of study drug (either Lenalidomide or Chlorambucil) (NCT00910910)
Timeframe: Up to the last patient last visit date of 19 May 2018; median follow-up for all participants was 46.7 months

,
Interventionparticipants (Number)
Participants receiving additional CLL therapyParticipants receiving alkylating agentsParticipants receiving antineoplastic aentsParticipants receiving antimetabolitesParticipants receiving corticosteroidsParticipants receiving plant alkaloidsParticipants receiving cytotoxic antibioticsParticipants receiving immunosuppressantsParticipants receiving therapeutic productsParticipants receiving other unspecified productsAntihistamine For Systemic UseDrugs for Peptic ulcer and Gastric ReflexImmunoglobulinsOther Analgesics and AntipyreticsSpecific Antirheumatic AgentsAntiemetics and AntinauseantsCorticosteriods for Systemic UseImmunostimulants
Chlorambucil12010686241611323210210111
Lenalidomide125107933427221034011111000

[back to top]

Time to Response

Time to response was calculated as the time from randomization to the first nPR, PR, CRi or CR based on IWCLL guidelines (NCT00910910)
Timeframe: Up to data cut-off of 18 Feb 2013; up to approximately 39 months

Interventionweeks (Median)
Lenalidomide8.6
Chlorambucil8.1

[back to top]

Percentage of Participants With the Best Overall Response Based on the International Workshop on Chronic Lymphocytic Leukemia (IWCLL) Guidelines

"A best overall response rate is a CR, CRi, nPR or PR and is defined as:~Complete Remission (CR):~No lymphadenopathy~No hepatomegaly or splenomegaly~Absence of constitutional symptoms~Polymorphonuclear leukocytes ≥ 1500/ul~No circulating clonal B-lymphocytes~Platelets > 100,000/ul~Hemoglobin > 11.0 g/dl~Normocellular <30% lymphocytes, no B-lymphoid nodules;~Incomplete Clinical Response (CRi):~• CR without bone marrow biopsy confirmation.~Nodular Partial Response (nPR):~• CR with the presence of residual clonal nodules.~Partial Response (PR) requires:~≥ 50% decrease in peripheral blood lymphocyte count~≥ 50% reduction in lymphadenopathy~≥ 50% reduction in size of liver and/or spleen~1 or more of the following:~Polymorphonuclear leukocytes ≥ 1500/ul~Platelets >100,000/ul" (NCT00910910)
Timeframe: Up to data cut-off date of 18 Feb 2013; approximately 39 months

Interventionpercentage of participants (Number)
Lenalidomide51.9
Chlorambucil62.3

[back to top]

Percentage of Participants With a Best Overall Response Based on IWCLL Guidelines With a Later Cut-off Date of 31 March 2014

"A best overall response rate is a CR, CRi, nPR or PR and is defined as:~Complete Remission (CR):~No lymphadenopathy~No hepatomegaly or splenomegaly~Absence of constitutional symptoms~Polymorphonuclear leukocytes ≥ 1500/ul~No circulating clonal B-lymphocytes~Platelets > 100,000/ul~Hemoglobin > 11.0 g/dl~Normocellular <30% lymphocytes, no B-lymphoid nodules;~Incomplete Clinical Response (CRi):~• CR without bone marrow biopsy confirmation.~Nodular Partial Response:~• CR with the presence of residual clonal nodules.~Partial Response requires:~≥ 50% decrease in peripheral blood lymphocyte count~≥ 50% reduction in lymphadenopathy~≥ 50% reduction in size of liver and/or spleen~1 or more of the following:~Polymorphonuclear leukocytes ≥ 1500/ul~Platelets >100,000/ul" (NCT00910910)
Timeframe: Up to data cut-off of 31 March 2014; approximately 53 months

Interventionpercentage of participants with response (Number)
Lenalidomide60.9
Chlorambucil70.2

[back to top]

Kaplan Meier Estimate of Overall Survival

Overall Survival is defined as the time between randomization and death from any cause. (NCT00910910)
Timeframe: Up to data cut off of 31 March 2014; median follow-up for all participants was 18.8 months

InterventionMonths (Median)
LenalidomideNA
Chlorambucil44.0

[back to top]

Kaplan-Meier Estimate for Duration of Response

Duration of response was defined as the time from first nPR, PR, CRi, or CR to PD. Duration of response was censored at the last date that the patient was known to be progression-free for: 1) participants who had not progressed at the time of analysis; 2) participants who had withdrawn consent or were lost to follow-up prior to documentation of progression (NCT00910910)
Timeframe: Up to data cut-off of 18 Feb 2013; up to approximately 39 months

Interventionweeks (Median)
LenalidomideNA
Chlorambucil105.3

[back to top]

Time to Response for a Later Cut-off Date of 31 March 2014

Time to response was calculated as the time from randomization to the first nPR, PR, CRi or CR based on IWCLL guidelines (NCT00910910)
Timeframe: Up to data cut-off of 31 March 2014; up to approximately 53 months

Interventionweeks (Median)
Lenalidomide10.4
Chlorambucil8.1

[back to top]

Kaplan-Meier Estimate for Duration of Response With a Later Cut-off Date of 31 March 2014

Duration of response was defined as the time from first nPR, PR, CRi, or CR to PD. Duration of response was censored at the last date that the patient was known to be progression-free for: 1) patients who had not progressed at the time of analysis; 2) patients who had withdrawn consent or were lost to follow-up prior to documentation of progression (NCT00910910)
Timeframe: Up to data cut-off of 31 March 2014; up to approximately 53 months

Interventionweeks (Median)
LenalidomideNA
Chlorambucil87.1

[back to top]

Kaplan-Meier Estimate of Progression Free Survival (PFS)

Progression-free survival was defined as the time from randomization to the first documented progression confirmed per investigator's assessment or death due to any cause on study, whichever occurred first. The progression date was assigned to the earliest time when any progression was observed without prior missing assessments. If withdrawal of consent or lost to follow-up occurred before documented progression or death, then these observations were censored at the date when the last complete tumor assessments determined a lack of progression (NCT00910910)
Timeframe: From first dose of study drug to date of data cut-off of 18 Feb 2013; up to approximately 39 months

Interventionmonths (Median)
Lenalidomide30.8
Chlorambucil23.0

[back to top]

Kaplan Meier Estimate for Overall Survival at the Final Analysis

Overall Survival is defined as the time between randomization and death from any cause. Overall survival was censored at the last date that the subject was known to be alive for participants who were alive as of the data cutoff date and for participants who were lost to follow-up before death was documented. (NCT00910910)
Timeframe: Up to the last patient last visit date of 19 May 2018; median follow-up for all participants was 46.7 months

InterventionMonths (Median)
Lenalidomide74.3
Chlorambucil70.5

[back to top]

Kaplan-Meier Estimate of Progression Free Survival (PFS) With a Later Cut-off Date of 14 March 2014

Progression-free survival was defined as the time from randomization to the first documented progression confirmed per investigator's assessment or death due to any cause on study, whichever occurred first. Progressive disease included lymphadenopathy, an appearance of any new lesion such as enlarged lymph nodes (> 1.5 cm), splenomegaly, hepatomegaly or other organ infiltrates, an increase by 50% or more in greatest determined diameter of any previous site or an increase by 50% or more in the sum of the product of diameters of multiple nodes. The progression date was assigned to the earliest time when any progression was observed without prior missing assessments. If withdrawal of consent or lost to follow-up occurred before documented progression or death, then these observations were censored at the date when the last complete tumor assessments determined a lack of progression. (NCT00910910)
Timeframe: From randomization to data cut off date of 31 March 2014; median follow up time for all participants was 12.6 months

Interventionmonths (Median)
Lenalidomide30.8
Chlorambucil21.4

[back to top]

Number of Participants Deaths During the Treatment and Survival Follow-Up Phase

The number of study participants deaths during the treatment and follow-up phase (NCT00910910)
Timeframe: From the first dose of study drug up to the last patient last visit date of 19 May 2018; median follow-up for all participants was 46.7 months

InterventionParticipants (Number)
Lenalidomide101
Chlorambucil95

[back to top]

Kaplan-Meier Estimates of Duration of Response (DoR)

Duration of response was defined as the time from the first observation of a response (CR, RR or PR) to the first documented disease progression or relapse. For participants who did not progress during the study, duration of response was censored at the last adequate response assessment showing evidence of no disease progression. Disease progression is defined as an increase in M-protein serum monoclonal paraprotein and/or urine paraprotein or evidence of bone marrow plasmacytosis and plasma cells, an appearance of new or existing soft tissue plasmacytomas, an appearance of new or existing lytic bone lesions and/or hypercalcemia >11.5mg/dL (NCT00928486)
Timeframe: From the time of the first dose of study drug to study completion; the median duration on study was 42.1 weeks

Interventionweeks (Median)
Lenalidomide and DexamethasoneNA

[back to top]

Number of Participants Experiencing Treatment-Emergent Adverse Events (TEAE)

A TEAE was defined as any AE that started on or after the first dose of study drug, and within End of Study (EOS) (28 days after the last dose of study drug received). A serious AE (SAE) = any AE which results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability; is a congenital anomaly/birth defect; or constitutes an important medical event. The intensity of AEs were graded 1 to 5 according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0. For all other AEs not described in the CTCAE criteria, the intensity was assessed by the investigator as mild grade (Grade 1), moderate (grade 2), severe (grade 3), life-threatening (grade 4) or death (grade 5) (NCT00928486)
Timeframe: Day 1 of study drug through 28 days after the last dose of study drug; maximum treatment duration was 60.3 weeks

Interventionparticipants (Number)
Any Adverse Event (AE)Drug-Related AEGrade 3-4 Adverse EventDrug-Related Grade 3-4 AESerious Adverse Event (SAE)Drug-Related SAEAE Leading to Study Drug DiscontinuationRelated AE Leading to Len/Dex DiscontinuationAE leading to dose reduction or interruptionRelated AE leading to dose reduction/interruption
Lenalidomide and Dexamethasone25252221129421716

[back to top]

Myeloma Response Rate

Overall myeloma response rate was determined by the investigator using the Myeloma Response Determination Criteria adapted from Bladé criteria. A responder is any patient who showed at least a partial response. Overall myeloma response rate is defined as the percentage of participants who achieved a Complete Response (CR), plus a Remission Response (RR), plus a Partial Response (PR). A CR is the disappearance of monoclonal paraprotein and maintained for ≥ 6 weeks. RR is a 75-99% reduction in the level of the serum monoclonal paraprotein compared to baseline; 90-99% reduction in 24-hr urinary light chain excretion. PR is a 50-74% reduction in the level of monoclonal paraprotein compared to baseline; 50-89% reduction in 24-hr urinary light chain excretion. (NCT00928486)
Timeframe: From the time of the first dose of study drug to study completion; median duration on study was 42.1 weeks

Interventionpercentage of participants (Number)
Lenalidomide and Dexamethasone62.5

[back to top]

RECIST-defined Measurable Disease

Patients who have a response of Complete Response (CR), Partial Response (PR), or Stable Disease (SD) by RECIST criteria. To be assigned a status of PR or CR, changes in tumor measurements must be confirmed by repeat assessments that should be performed no less than 4 weeks after the criteria for response are first met. In the case of SD, follow-up measurements must have met the SD criteria at least once after study entry at a minimum interval of 6-8 weeks (NCT00939510)
Timeframe: every 8 weeks and at end of treatment

Interventionparticipants (Number)
Number of patients with stable disease (SD)Number of patients with partial response (PR)Number of patients with progressive disease
Lenalidomide (Revlimid) and Sargramostim (GM-CSF)425

[back to top]

Number of Patients With Statistically Significant Change in Immune Response From Baseline to End of Study

The change in mean T cell immunohistochemical markers and dendritic cells over time will be evaluated using analysis of variance methods for repeated measures with additional main factors included in the analysis for subset comparisons. The pattern of immune response will be evaluated based upon overall clinical response using these same techniques. (NCT00939510)
Timeframe: every 28 days for first 3 cycles, end of study

Interventionparticipants (Number)
Lenalidomide (Revlimid) and Sargramostim (GM-CSF)0

[back to top]

Number of Patients With a PSA Response

Number of patients with a PSA Response defined as a PSA decline greater or equal to 50% compared with baseline value. (NCT00939510)
Timeframe: reevaluated for response every eight weeks

Interventionparticipants (Number)
PSA responsePSA no response
Lenalidomide (Revlimid) and Sargramostim (GM-CSF)427

[back to top]

Count of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0) Who Were Administered the Four-Drug Combination

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. (NCT00942578)
Timeframe: Date treatment consent signed to date off study, approximately 93 months and 22 days.

InterventionParticipants (Count of Participants)
15 mg Dose Lenalidomide14
20 mg Dose Lenalidomide3
25 mg Dose Lenalidomide46

[back to top]

Count of Participants With a Radiologic Response

Radiologic response was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST) v1.0 criteria. 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, taking as reference the baseline sum diameters. Progressive disease (PD) 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). Stable disease is neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. (NCT00942578)
Timeframe: median time of potential follow-up of 47.5 months

,,
InterventionParticipants (Count of Participants)
Confirmed radiologic PRComplete ResponseStable Disease
15 mg Lenalidomide301
20 mg Lenalidomide220
25 mg Lenalidomide2003

[back to top] [back to top]

Median Time to Progression (TTP)

TTP is evaluated from the on-study date until the date of progression or last follow-up after progression. (NCT00942578)
Timeframe: median time of potential follow-up of 47.5 months

InterventionMonths (Median)
Single Arm - 4 Drug Combination18.2

[back to top]

Median Overall Survival of Patients Studied

OS is evaluated from the on-study date until the date of death or last follow-up. (NCT00942578)
Timeframe: median time of potential follow-up of 47.5 months

InterventionMonths (Median)
Single Arm - 4 Drug Combination24.6

[back to top]

Count of Participants With Dose-Limiting Toxicities (DLT)

DLT is defined as a ≥grade 3 non-hematological toxicity related to lenalidomide. (NCT00942578)
Timeframe: First 28 days of treatment.

InterventionParticipants (Count of Participants)
Single Arm - 4 Drug Combination0

[back to top]

Survival Based on Expression of T Cell Immunoglobulin and Mucin Domain (TIM-3) on Cluster of Differentiation 8 (CD8) + T Cells

Expression of TIM-3 on CD8 + T cells was evaluated by flow cytometry. (NCT00942578)
Timeframe: 46.5 months

,,
InterventionMonths (Median)
Low expressionHigh expression
15 mg Lenalidomide23.6NA
20 mg LenalidomideNANA
25 mg Lenalidomide20.212.7

[back to top]

Survival Based on Expression of Programmed Cell Death Protein 1 (PD-1) on Cluster of Differentiation 8 (CD8) + T Cells

Expression of PD-1 on CD8 + T cells was evaluated by flow cytometry. High and low expression are based on the median values. Patients with a low expression of PD-1 proteins had better survival than those with a high expression. (NCT00942578)
Timeframe: median time of potential follow-up of 47.5 months

,,
InterventionMonths (Median)
Low expressionHigh expression
15 mg Dose Lenalidomide21.422.4
20 mg Dose Lenalidomide17.826.1
25 mg Dose Lenalidomide28.914.8

[back to top]

Count of Participants With Prostatic Antigen-Specific (PSA) Declines

PSA decline is defined as a ≥50% decline in measurable disease. Measurable lesions are defined as those that can be accurately measured in at least one dimension (longest diameter to be recorded) as ≥20 mm by chest x-ray, as ≥10 mm with computed tomography, or ≥10 mm with calipers by clinical exam. (NCT00942578)
Timeframe: median time of potential follow-up of 47.5 months

,,
InterventionParticipants (Count of Participants)
Participants with PSA>30%Participants with PSA>50%Participants with PSA>90%Not Evaluable
15 mg Lenalidomide131251
20 mg Lenalidomide3310
25 mg Lenalidomide4140271

[back to top]

Count of Participants With Changes in Circulating Apoptotic Endothelial Cells (CAEC) From Baseline After Drug Administration

The definition of an increase is any increase (any number greater than zero) in the percent CAEC among total peripheral blood mononuclear cells comparing each patient's percent CAEC among total peripheral blood mononuclear cells at baseline to each patient's percent CAEC among total peripheral blood mononuclear cells at cycle 3 day 1. The definition of a decrease is any decrease (any number less than zero) in the percent CAEC among total peripheral blood mononuclear cells comparing each patient's percent CAEC among total peripheral blood mononuclear cells at baseline to each patient's percent CAEC among total peripheral blood mononuclear cells at cycle 3 day 1. (NCT00942578)
Timeframe: After drug administration, an average of 3 months

InterventionParticipants (Count of Participants)
Increase in CAEC after 3 monthsdecrease in CAEC after 3 months
Single Arm - 4 Drug Combination3020

[back to top]

Overall Response Rate (ORR)

ORR was defined as the percentage of patients with a complete response (CR), CR with incomplete bone marrow (BM) recovery (CRi) or partial response (PR) during treatment. Response was assessed according to the 2008 International Workshop on Chronic Lymphocytic Leukemia (iwCLL) guidelines. Per the guidelines, a CR required peripheral blood lymphocytes below 4 x 10^9/L, absence of lymphadenopathy, no hepatomegaly or splenomegaly, absence of disease and blood counts neutrophils >1.5 x 10^9/L, platelets >100 x 10^9/L, hemoglobin (hgb) >11g/dL) and BM at least normocellular for age. CRi = CR with incomplete BM recovery. PR = required at least 2 months from end of treatment, a ≥50% decrease in peripheral blood lymphocyte count from the pre-treatment value and either a ≥ 50% reduction in lymphadenopathy or ≥50% reduction of liver enlargement or ≥50% reduction of spleen enlargement plus neutrophils >1.5 x 10^9/ or ≥50% increase, platelets >100 x 10^9/L or ≥50% increase, hgb 11 g/dL. (NCT00963105)
Timeframe: Response was assessed after 3 cycles of therapy (Week 12) and every 4 weeks thereafter until disease progression. Maximum time on study was 91 months.

Interventionpercentage of participants (Number)
Lenalidomide 5 mg47.1
Lenalidomide 10 mg37.1
Lenalidomide 15 mg40.0

[back to top]

Kaplan-Meier Estimate of Progression Free Survival

Progression-free survival (PFS) was calculated as the time from randomization to the first documented progression or death due to any cause during or after the treatment period, whichever occurred first. The progression date was assigned to the earliest time when any progression is observed without prior missing assessments. If withdrawal of consent or loss to follow-up occurred before documented progression or death, then these observations were censored at the date when the last complete tumor assessments determined a lack of progression. (NCT00963105)
Timeframe: From randomization until the end of the study; maximum time on study was 91 months.

Interventionweeks (Median)
Lenalidomide 5 mg31.4
Lenalidomide 10 mg45.1
Lenalidomide 15 mg66.3

[back to top]

Kaplan-Meier Estimate of Event-Free Survival

Event-free survival (EFS) is the interval between the start of treatment to the first sign of disease progression, or treatment for relapse or death (whichever occurred first). If withdrawal of consent or loss to follow-up occurred before documented progression or death, then these observations were censored at the date when the last complete tumor assessments determined a lack of progression. (NCT00963105)
Timeframe: From randomization until the end of the study; maximum time on study was 91 months.

Interventionweeks (Median)
Lenalidomide 5 mg25.6
Lenalidomide 10 mg31.9
Lenalidomide 15 mg24.1

[back to top]

Kaplan-Meier Estimate of Duration of Response

Duration of response (DOR) was defined as the time from the first visit where PR, CRi, or CR was documented to progressive disease (PD). Duration of response was censored at the last date that the participant was known to be progression-free for participants who had not progressed at the time of analysis or who withdrew consent or were lost to follow-up prior to documentation of progression. (NCT00963105)
Timeframe: Response was assessed after 3 cycles of therapy (Week 12) and every 4 weeks thereafter until disease progression. Maximum time on study was 91 months.

Interventionweeks (Median)
Lenalidomide 5 mg101.1
Lenalidomide 10 mg35.1
Lenalidomide 15 mg88.8

[back to top]

Kaplan-Meier Estimate of Time to Progression

Time to progression (TTP) was defined as the time from randomization to the first documented progression. For participants who did not progress during the study, TTP was censored at the last adequate response assessment showing evidence of no disease progression. (NCT00963105)
Timeframe: From randomization until the end of the study; maximum time on study was 91 months.

Interventionweeks (Median)
Lenalidomide 5 mg96.3
Lenalidomide 10 mg47.6
Lenalidomide 15 mg66.3

[back to top]

Kaplan-Meier Estimate of Overall Survival

Overall survival (OS) was defined as the time from randomization to death from any cause. Overall survival was censored at the last date that the participant was known to be alive for participants who were alive at the time of analysis and for participants who had withdrawn consent or were lost to follow-up before death was documented. (NCT00963105)
Timeframe: From randomization until the end of the study; maximum time on study was 91 months.

Interventionweeks (Median)
Lenalidomide 5 mg161.0
Lenalidomide 10 mg106.7
Lenalidomide 15 mg154.6

[back to top]

Number of Participants With Treatment-emergent Adverse Events

Adverse events (AEs) were graded for severity by the investigator according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 3.0 with the exceptions of hematologic toxicities and tumor lysis syndrome, according to the following scale: Grade 1 = Mild Grade 2 = Moderate Grade 3 = Severe Grade 4 = Life Threatening or disabling AE Grade 5 = Death The investigator determined the relationship of each AE to study drug based on the timing of the AE and whether other medications, therapeutic interventions, or underlying conditions could provide a sufficient explanation for the observed event. (NCT00963105)
Timeframe: From first dose of study drug to 30 days after the last dose; the maximum duration of treatment was 251, 265, and 267 weeks in the 5 mg, 10 mg, and 15 mg treatment groups respectively.

,,
InterventionParticipants (Count of Participants)
Any adverse eventsTreatment-related adverse events (TRAE)Grade 3/4 adverse eventsTreatment-related Grade 3/4 adverse eventsGrade 5 adverse eventsTreatment-related Grade 5 adverse eventsSerious adverse eventsTreatment-related serious adverse eventsAE leading to discontinuation of study drugTRAE leading to discontinuation of study drugAE leading to study drug dose reduction onlyAE leading to study drug dose interruption onlyAE leading to study drug interruption & reduction
Lenalidomide 10 mg34343232422413171462426
Lenalidomide 15 mg35323430302720161352519
Lenalidomide 5 mg34333331422415211682518

[back to top]

Time to Response

Time to response (TTR) was calculated as the time from randomization to the first documented date of response (PR, CRi or CR) based on iwCLL guidelines for participants with an objective response during the treatment period. (NCT00963105)
Timeframe: Response was assessed after 3 cycles of therapy (Week 12) and every 4 weeks thereafter until disease progression. Maximum time on study was 91 months.

Interventionweeks (Median)
Lenalidomide 5 mg16.9
Lenalidomide 10 mg12.6
Lenalidomide 15 mg12.7

[back to top]

Complete Response(CR) and Very Good Partial Response(VGPR)

To determine the best overall response (CR+VGPR+PR) of the lenalidomide, thalidomide, dexamethasone combination based on IMWG criteria at nadir. (NCT00966693)
Timeframe: Evaluated each 28-day cycle and nadir of criteria is considered best overall response (median time to best response for this study was 2 cycles (range for best overall response was 1-21 cycles).

,,,
InterventionParticipants (Count of Participants)
Partial RemissionProgressive DiseaseVery Good Partial RemissionMinimal Residual DiseaseStringent Complete RemissionNon EvaluableStable Disease
RTD (Cohort 1, Phase 1)2000010
RTD (Cohort 2, Phase 1)0230000
RTD (Cohort 2, Phase 2)13689377
RTD (Cohort 3, Phase 1)2211030

[back to top]

Incidence of Adverse Events

Linear regression was utilized to assess the effect of patient prognostic factors on the toxicity rate. (NCT00966693)
Timeframe: Up to 9 years

InterventionParticipants (Count of Participants)
RTD (Cohort 1, Phase 1)0
RTD (Cohort 2, Phase 1)0
RTD (Cohort 3, Phase 1)2
RTD (Cohort 2, Phase 2)10

[back to top]

Progression Free Survival

Estimated using the method of Kaplan and Meier. Comparison of time-to-event endpoints by important subgroups was made using the log-rank test. Cox proportional hazard regression will be employed for multivariate analysis on time-to-event outcomes. (NCT00966693)
Timeframe: Up to 9 years

InterventionMonths (Mean)
RTD (Cohort 1, Phase 1)32.74
RTD (Cohort 2, Phase 1)9.04
RTD (Cohort 3, Phase 1)14.61
RTD (Cohort 2, Phase 2)10.02

[back to top]

Number of Participants With Dose Limitations Toxicities of the Combination of Lenalidomide and Thalidomide and Dexamethasone (LTD) in Patients With Relapsed/Refractory Multiple Myeloma (RRMM)

To determine the dose limitations toxicities of the combination of lenalidomide and thalidomide and dexamethasone (LTD) in patients with relapsed/refractory multiple myeloma (RRMM). (NCT00966693)
Timeframe: After one 28-day cycle

InterventionParticipants (Count of Participants)
RTD (Cohort 1, Phase 1)1
RTD (Cohort 2, Phase 1)0
RTD (Cohort 3, Phase 1)3
RTD (Cohort 2, Phase 2)0

[back to top]

Time to Best Response

Estimated using the method of Kaplan and Meier. Comparison of time-to-event endpoints by important subgroups was made using the log-rank test. Cox proportional hazard regression will be employed for multivariate analysis on time-to-event outcomes. (NCT00966693)
Timeframe: Up to 9 years

Interventionmonths (Mean)
RTD (Cohort 1, Phase 1)19
RTD (Cohort 2, Phase 1)5
RTD (Cohort 3, Phase 1)5.16
RTD (Cohort 2, Phase 2)3.3

[back to top]

Time to Progression

Time to Progression was estimated using Kaplan Meier analysis. (NCT00966693)
Timeframe: Up to 9 years

InterventionMonths (Mean)
RTD (Cohort 1, Phase 1)32.74
RTD (Cohort 2, Phase 1)9.04
RTD (Cohort 3, Phase 1)14.61
RTD (Cohort 2, Phase 2)10.02

[back to top] [back to top]

Progression Free Survival

The primary endpoint of this study was progression-free survival (PFS), defined as the number of days from the day of first study drug administration to the day the patient experienced disease progression or death from any cause. Response and progression in cases of small lymphocytic lymphoma(SLL) were evaluated using the International Working Group Criteria for response in NHL (Cheson, et al 1996). Response and progression in cases of chronic lymphocytic leukemia (CLL) were evaluated using the NCI-sponsored CLL Working Group guidelines for CLL (Cheson, et al 2007). (NCT00974233)
Timeframe: 42 months (6 months induction therapy, 12 months maintenance, 24 months long-term follow-up)

Interventionmonths (Median)
Induction/Maintenance Chemotherapy18.3

[back to top]

Overall Survival

Overall survival (OS) is defined as the time from the day of first study drug administration until death from any cause. (NCT00974233)
Timeframe: 42 months (6 months induction therapy, 12 months maintenance, 24 months long-term follow-up)

Interventionmonths (Median)
Overall Study Population42.8

[back to top]

Toxicities Observed With Induction Chemotherapy and Maintenance Therapy

Toxicities were reported using the Common Terminology Criteria for Adverse Events, version 3.0. (NCT00974233)
Timeframe: 42 months (6 months induction therapy, 12 months maintenance, 24 months long-term follow-up)

,
Interventionparticipants (Number)
Grade 3 leukopeniaGrade 4 leukopeniaGrade 3 neutropeniaGrade 4 neutropeniaGrade 3 anemiaGrade 3 thrombocytopeniaGrade 4 thrombocytopeniaGrade 3 febrile neutropeniaGrade 3 infectionGrade 4 infectionGrade 5 infectionGrade 3 fatigueGrade 4 fatigueGrade 3 nausea/emesisGrade 3 serum transaminase levels (Gr 3/Gr 4)
Induction Chemoimmunotherapy55614152410112034
Maintenance617201014100100

[back to top]

Progression-free Survival

Progression-free survival (PFS) is defined as the time from the day of first study drug administration until progression of CLL/SLL or death from any cause. PFS is reported as the proportion of participants with PFS up to 42 months. (NCT00974233)
Timeframe: 42 months (6 months induction therapy, 12 months maintenance, 24 months long-term follow-up)

InterventionProportion of participants (Median)
1-year progression-free survival2-year progression-free survival3-year progression-free survival
Induction/Maintenance Chemotherapy0.620.350.23

[back to top]

Objective Response Rate (Complete + Partial Responses)

Response and progression in cases of SLL were evaluated using the International Working Group Criteria for response in NHL (Cheson, et al 1996). Response and progression in cases of CLL were evaluated using the NCI-sponsored CLL Working Group guidelines for CLL (Cheson, et al 2007). Complete response defined as resolution enlarged lymph nodes, spleen and liver; normalization of blood counts (neutrophils, hemoglobin, platelets); no residual CLL/SLL detectable in the bone marrow. Partial response defined as 50% or more reduction in size of enlarged lymph nodes, liver or spleen; 50% or more improvement of blood counts; 50% or more improvement in the blood lymphocyte count. Progressive disease defined as 50% or more increase in the combined measurements of at least 2 lymph nodes as measured on CT scans or the appearance of new enlarged lymph nodes; 50% of more increase in the size of the spleen or liver; 50% or more increase in blood lymphocyte count. (NCT00974233)
Timeframe: 42 months (6 months induction therapy, 12 months maintenance, 24 months long-term follow-up)

Interventionparticipants (Number)
Complete Response (CR)7
Partial Response (PR)12
Stable Disease9
Overall Response Rate19

[back to top]

Phase 1: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) While on Both Lenalidomide and Sunitinib

Adverse event (AE) = any noxious, unintended, or untoward medical occurrence occurring at any dose that may appear or worsen in a participant during the course of a study, including new intercurrent illness, worsening concomitant illness, injury, or any concomitant impairment of participants health, including laboratory test values, regardless of etiology. Serious adverse event (SAE) = any AE which: results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. TEAE = any AE occurring or worsening on or after the first treatment with any study drug. Related = suspected by investigator to be related to study treatment. National Cancer Institute [NCI] Common Toxicity Criteria for Adverse Events [CTCAE], Version 4.0, grades: 1 = mild, 2 = moderate, 3 = severe, 4 = life threatening, 5 = death (NCT00975806)
Timeframe: First day of study drug to within 28 days after the last dose of the last study drug; The duration of exposure to lenalidomide and sunitinib was 7.0 to 327 and 7.0 to 328 days respectively

,,
Interventionparticipants (Number)
Participants with at least 1 TEAEParticipants with at least 1 serious TEAE≥ 1 TEAE leading to stopping lenalidomide≥ 1 TEAE leading to stopping sunitinib≥ 1 TEAE -> dose reduction/interrruption of Len≥ 1 TEAE dose reduction/interrruption of SunitinibParticipants with ≥1 TEAE related to lenalidomideParticipants with ≥1 TEAE related to Sunitinib≥ 1 NCI CTC Gr 3 or higher TEAE≥ 1 NCI CTC Gr 3 or higher related to lenalidomide≥ 1 NCI CTC Gr 3 or higher related to Sunitinib≥ 1 serious TEAE related to lenalidomide≥ 1 serious TEAE related to sunitinib
Cohort A: Lenalidomide 10mg and Sunitinib 37.5 mg5322555554421
Cohort F: Lenalidomide 10mg and Sunitinib 37.5mg7422657765522
Cohort G: Lenalidomide 15mg and Sunitinib 37.5mg4222434444402

[back to top]

Phase 1 : Tumor Response Rate According to RECIST 1.1

"Tumor response was evaluated every 3 cycles beginning with Cycle 3 Day 1 and at treatment discontinuation. Response was evaluated using the Response Criteria Evaluation in Solid Tumors (RECIST 1.1) criteria:~Treatment response includes both complete response and partial response~Complete response-disappearance of all lesions~Partial response-30% decrease in the sum of diameters of target lesions from baseline~Stable disease-neither shrinkage nor increase of lesions~Progressive Disease-20% increase in the sum of diameters of target lesions from nadir" (NCT00975806)
Timeframe: Every 3 cycles; up to month 25

,,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable Disease
Cohort A: Lenalidomide 10mg and Sunitinib 37.5 mg011
Cohort F: Lenalidomide 10mg and Sunitinib 37.5mg003
Cohort G: Lenalidomide 15mg and Sunitinib 37.5mg003

[back to top]

Phase 1: Maximum Tolerated Dose (MTD)

"The MTD of lenalidomide in combination with sunitinib was defined as the highest dose level at which no more than 1 out of 6 participants experienced a dose limiting toxicity (DLT). Dose limiting toxicities were:~• Inability to deliver Lenalidomide in Cycle 1 due to a drug-related toxicity resulting in:~Grade (GR) 3 or 4 non-hematological toxicity lasting for ≥ 14 days~Febrile neutropenia~Gr 4 neutropenia lasting for ≥ 7 days~Gr 4 thrombocytopenia The occurrence of one of the above drug-related toxicities resulting in a clinical and/or laboratory assessment being done within 7 days following the initial finding to examine the participants for resolution of the toxicity. Lack of resolution of the toxicities was considered a DLT.~If ≤ 7 doses of lenalidomide or Sunitinib were missed in Cycle 1 due to non-drug related event, the participant data was to be included in the evaluation of dose escalation." (NCT00975806)
Timeframe: Within 21 days of first dose of treatment

Interventionmg (Number)
Cohort F: Lenalidomide 10mg and Sunitinib 37.5mg10

[back to top]

Progression-Free Survival (PFS)

PFS was the time from randomization to disease progression, or death, whatever occurred first. Progression criteria was met by analysis of target and non-target lesions as defined by Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 criteria. Progressive Disease (PD) is defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters while on study or the appearance of one or more new lesions; an increase of at least 5mm as a total sum. Lymph nodes identified as target lesions (≥ 15 mm diameter in short axis) will be followed and reported by changes in diameter of short axis; or the unequivocal progression of a non-target lesion defined as an increase in the overall disease burden based on the change in non-measurable disease that is comparable in scope to the increase required to declare PD for measurable disease; Two or more new bone lesions as detected by bone scan (NCT00988208)
Timeframe: From randomization until disease progression or death from any cause; up to the cut-off date of 13 Jan 2012; maximum time on study was approximately 26 months

InterventionWeeks (Median)
Docetaxel/Prednisone/Placebo (DP)46
Docetaxel/Prednisone/Lenalidomide (DPL)45

[back to top]

Percentage of Participants With an Objective Response According to Response Evaluation Criteria in Solid Tumors - RECIST Version 1.1 Criteria

Objective response (OR) is defined as having complete response (CR) or partial response (PR) as best overall response based on RECIST Criteria 1.1 and defines a CR = Disappearance of all target lesions except lymph nodes (LN); LN must have a decrease in the short axis to <10mm; PR = 30% decrease in sum of diameters of target lesions taking as reference the baseline sum diameters; Progressed Disease (PD) = 20% increase in sum of diameters of target lesions taking as a reference the smallest sum of diameters and an absolute increase of ≥5 mm; the appearance of ≥1 new lesions; Stable Disease (SD)= Neither shrinkage to qualify for PR nor increase to qualify for PD taking the smallest sum diameters on study as reference. For non-target lesions a CR = Disappearance of all non-target lesions and all LN must be non-pathological in size <10 mm; Non-CR/Non PD: persistence of one or more non-target lesions; PD = unequivocal progression of existing non-target lesions or appearance of new ones (NCT00988208)
Timeframe: From day 1 to data cut-off 13 January 2012; maximum time on study was approximately 26 months

Interventionpercentage of participants (Number)
Docetaxel/Prednisone/Placebo (DP)24.3
Docetaxel/Prednisone/Lenalidomide (DPL)22.1

[back to top]

Percentage of Participants Who Received Post-Study Therapies

Percentage of Participants Who Received Post-Study Therapies for advanced Prostate Cancer. (NCT00988208)
Timeframe: The date when the first consent form was signed to the last date of AE data collection;up to 5 years; up to the date of the final data analysis date of 20 April 2017

InterventionPercentage of Participants (Number)
Docetaxel/Prednisone/Placebo (DP)70.8
Docetaxel/Prednisone/Lenalidomide (DPL)69.0

[back to top]

Overall Survival (OS)

Overall survival (OS) was the time from the date of randomization to the date of death from any cause. If no death was reported for a participant before the cut-off date for OS analysis, OS was censored at the last date at which the participant was alive. The median OS was calculated based on Kaplan-Meier estimates and corresponding 95% confidence interval (CI) was calculated using the method provided by Brookmeyer and Crowley. (NCT00988208)
Timeframe: From randomization until death from any cause up to the cut-off date of 13 January 2012; up to approximately 26 months

Interventionweeks (Median)
Docetaxel/Prednisone/Placebo (DP)NA
Docetaxel/Prednisone/Lenalidomide (DPL)77

[back to top]

Percentage of Participants With Secondary Primary Malignancies During the Course of the Trial

Second primary malignancies were monitored as events of interest and reported as serious adverse events throughout the course of the trial. (NCT00988208)
Timeframe: The date when the first consent form was signed to the last date of AE data collection; up to the date of the final data analysis date of 30 November 2016; 7 years and 19 days

,
Interventionpercentage of participants (Number)
Invasive Secondary Primary MalignanciesNon-invasive Secondary Primary Malignancies
Docetaxel/Prednisone/Lenalidomide (DPL)1.71.0
Docetaxel/Prednisone/Placebo (DP)1.30.4

[back to top]

Number of Participants With Treatment Emergent Adverse Events (AEs)

A TEAE is defined as any AE occurring or worsening on or after the first dose of study drug and within 28 days after the last dose of study drug. A TESAE is defined as any serious adverse event (SAE) occurring or worsening on or after the first dose of study drug and within 28 days after the last dose of study drug. Safety and severity was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 4.0; Severity of AEs were graded (including second primary malignancies) as Grade 1- Mild; Grade 2- Moderate; Grade 3- Severe; Grade 4- Life-threatening; Grade 5-Fatal; (NCT00988208)
Timeframe: From the time from of first dose of study drug administration to 28 days after the last dose of study drug and up to the data cut off date of 13 January 2012; the maximum duration of study drug was 93 weeks for DP and 90.6 weeks for DPL

,
Interventionparticipants (Number)
Any TEAEAny TEAE related to lenalidomide or placeboAny TEAE related to docetaxel/prednisoneAny severity grade 3-4 TEAEAny serious AE (SAE)Any SAE related to lenalidomide or placeboAny SAE related to docetaxel/prednisoneAny AE causing discontinuation of lenalidomide/PBOAny AE causing withdrawal of docetaxel/prednisoneAny TEAE leading to death
Docetaxel/Prednisone/Lenalidomide (DPL)51741248138127916718215016924
Docetaxel/Prednisone/Placebo (DP)51237947530317162868212716

[back to top]

Time to Onset of Secondary Primary Malignancies

Time of Onset of Secondary Primary Malignancies was considered an event of interest (NCT00988208)
Timeframe: The date when the first consent form was signed to the last date of AE data collection; up to the date of the final data analysis date of 30 November 2016; 7 years and 19 days

Interventionmonths (Median)
Docetaxel/Prednisone/Placebo (DP)29.7
Docetaxel/Prednisone/Lenalidomide (DPL)19.7

[back to top]

Change in Childhood Autism Rating Scale (CARS)Value From Baseline to 6 Weeks

Change in CARS value from baseline to 6 weeks. Total CARS scores range from a fifteen to 60, with a minimum score of thirty serving as the cutoff for a diagnosis of autism on the mild end of the autism spectrum. (NCT00996931)
Timeframe: Baseline and 6 weeks

Interventionmean change in units on scale (Mean)
Lenalidomide-2.08

[back to top]

Change in TNF-alpha Levels

Change in CSF-TNF-α from baseline to 12 weeks. (NCT00996931)
Timeframe: Baseline and 12 weeks

Interventionmean % change (Mean)
Lenalidomide57

[back to top]

Overall Response Rate

A Simon's two-stage minmax design will be used. Includes complete remission (CR) and partial remission (PR). Complete Response Requires the absence of disease signs and symptoms, and normalization of Peripheral blood and bone marrow. Partial Response it at lease a 50% reduction in disease signs and symptoms and normalization of peripheral blood. (NCT01002755)
Timeframe: Up to 8 years

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide, Ofatumumab)24

[back to top]

Number of Participants With Tolerance of the Medication Combination

Incidence of grade 3 and 4 non-hematological toxicity in more than 50 percent of the participants. Will be monitored based on the Bayesian model (beta-binomial). (NCT01002755)
Timeframe: Up to 8 years

Interventionparticipants (Number)
Treatment (Lenalidomide, Ofatumumab)0

[back to top]

Progression Free Survival

The time from the start of therapy to death, disease progression, or the initiation of the next therapy. Disease progression is the loss of response or transformation to a more aggressive histology. (NCT01002755)
Timeframe: Up to 8 years

Interventionmonths (Median)
Treatment (Lenalidomide, Ofatumumab)16

[back to top]

Best Response

The major criteria for determination of response to therapy in patients with CLL include physical examination and examination of the peripheral blood and bone marrow. Complete Response (CR): Absence of lymphadenopathy, hepatomegaly or splenomegaly by physical examination and appropriate radiographic techniques (if abnormal pre-treatment), No constitutional symptoms, ANC >/= 1,500/ul, platelets> 100,000/ul, Hgb>11gm/dl (untransfused); Partial Response (PR): >50% decrease in peripheral blood lymphocyte count from the pre-treatment baseline value, reduction in lymphadenopathy, reduction in size of the liver and/or the spleen; Progressive Disease (PD): Characterized by at least one of the following: > 50% increase in the sum of the products of at least 2 lymph nodes on at least 2 consecutive exams at least 2 weeks apart. At least 1 node must be >/= to 2cm in size, Appearance of new palpable LN, > 50% increase in size of liver or spleen determined by measurement below the respective costal (NCT01011894)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Stable DiseaseProgressive DiseaseNot Evaluable
Participants Receiving Lenalidomide2033

[back to top]

Morphologic Leukemia-free State

Defined as < 5% blasts on the BM aspirate with spicules and a count of >200 nucleated cells and no blasts with Auer rods, and no persistent extramedullary disease. (NCT01016600)
Timeframe: Median number of cycles completed [3 cycles (12 weeks) full range (1 (4 weeks)-17 (68 weeks))]

Interventionparticipants (Number)
Cohort 12
Cohort 21
Cohort 32
Phase II2

[back to top]

Partial Remission Rate (PR)

Requires that the criteria for complete remission be met with the following exceptions: decrease of >50% in the percentage of blasts to 5-25% in the BM aspirate. A value of < 5% blasts in BM with Auer rods is also considered a partial remission. (NCT01016600)
Timeframe: Completion of treatment (median follow-up was 8 weeks) (range 4-68 weeks)

Interventionparticipants (Number)
Cohort 12
Cohort 20
Cohort 32
Phase II5

[back to top]

Phase I Only - Maximum Tolerated Dose (MTD) as Measured by Dose-limiting Toxicities (DLTs)

"The maximum tolerated dose (MTD) is defined as the dose level immediately below the dose level at which 2 patients of a cohort (of 2 to 6 patients) experience dose-limiting toxicity during the first cycle.~Hematologic DLT is as a persistent bone marrow aplasia with ≤ 10 % cellularity, which persists for > 60 days from the start of a chemotherapy cycle.~Non-hematologic DLT is defined as any Grade 3 or Grade 4 non-hematologic toxicity that occurs during the first cycle with the specific exceptions of nausea, vomiting, anorexia, weight loss, infections or electrolyte abnormalities attributable to any other cause. Grade 3 triglycerides will be considered a DLT only for patients who have Grade 3 in spite of appropriate lipid lowering drug therapy." (NCT01016600)
Timeframe: Completion of the phase I portion of study (approximately 1 year and 4 months)

Interventiondose-limiting toxicities (Number)
Cohort 11
Cohort 20
Cohort 30

[back to top]

Phase II Only - Complete Remission Rate (CRm + CRi) in Participants With Untreated AML ≥60 Years of Age

"Morphologic complete remission (CRm): Defined as morphologic leukemia-free state, including <5% blasts in BM aspirate with marrow spicules and a count of > 200 nucleated cells and no blasts with Auer rods, no persistent extramedullary disease, ANC > 1000/uL, platelet count > 100,000/uL. Patient must be independent of transfusions for a minimum of 1 week before each marrow assessment.~Morphologic complete remission with incomplete blood count recovery (CRi): Defined as CR with the exception of neutropenia <1000/uL or thrombocytopenia <100,000/ul." (NCT01016600)
Timeframe: Completion of treatment (median follow-up was 8 weeks) (range 4-68 weeks)

Interventionpercentage of participants (Number)
Phase II22

[back to top]

Phase I Only - Maximum Tolerated Dose (MTD)

"The maximum tolerated dose (MTD) is defined as the dose level immediately below the dose level at which 2 patients of a cohort (of 2 to 6 patients) experience dose-limiting toxicity during the first cycle.~Hematologic DLT is as a persistent bone marrow aplasia with ≤ 10 % cellularity, which persists for > 60 days from the start of a chemotherapy cycle.~Non-hematologic DLT is defined as any Grade 3 or Grade 4 non-hematologic toxicity that occurs during the first cycle with the specific exceptions of nausea, vomiting, anorexia, weight loss, infections or electrolyte abnormalities attributable to any other cause. Grade 3 triglycerides will be considered a DLT only for patients who have Grade 3 in spite of appropriate lipid lowering drug therapy." (NCT01016600)
Timeframe: Completion of the phase I portion of study (approximately 1 year and 4 months)

Interventionmg/m^2 (Number)
Phase I Cohort75

[back to top]

Relapse Free Survival (RFS)

This is determined only for patients achieving a complete remission. Defined as the interval from the date of first documentation of a leukemia free state to date of recurrence or death due to any cause. (NCT01016600)
Timeframe: Until death - median follow-up 4.6 months (full range (0.3-31.4 months))

Interventionmonths (Median)
Cohort 112.0
Cohort 21.4
Cohort 34.9
Phase II12.2

[back to top]

Time to Progression (TTP)

Defined as the interval from the date of the first dose of study drug to the date of progressive disease. (NCT01016600)
Timeframe: Until progressive disease - median follow-up 4.6 months (full range (0.3-31.4 months))

Interventionmonths (Median)
Cohort 15.7
Cohort 23.4
Cohort 37.8
Phase II3.7

[back to top]

Response Rate (CRm + CRc + CRi + PR)

"Response rate (CRm + CRc + CRi + PR)~CRm = morphologic complete remission~CRc = cytogenetic complete remission~CRi = morphologic complete remission with incomplete blood count recovery~PR = partial remission" (NCT01016600)
Timeframe: Median number of cycles completed [3 cycles (12 weeks) full range (1 (4 weeks)-17 (68 weeks))]

,,,
Interventionparticipants (Number)
CRmCRcCRiPR
Cohort 11112
Cohort 20010
Cohort 31012
Phase II1015

[back to top]

Toxicity Profile (Grade 3/4 Toxicities)

AML ≥18 years or untreated AML ≥60 years (NCT01016600)
Timeframe: 30 days after completion of treatment (median follow-up was 12 weeks (range 8-72 weeks))

,,,
Interventionparticipants (Number)
AnemiaFebrile neutropeniaOtitis mastoditis - worseningDiarrheaNauseaVomitingDental carriesFatigueSepsisBronchial infectionBacteremiaLung infectionSkin infectionActivated partial thromboplastin time prolongedBlood bilirubin increasedCreatinine increasedNeutrophil count decreasedPlatelet count decreasedWhite blood cell count decreasedLymphocyte count decreasedDehydrationHypernatremiaHypocalcemiaHypophosphatemiaBack painGeneralized muscle weaknessPain in extremityLeukemia vasculitis left calfAcute kidney injuryDyspneaEpistaxisProductive coughPulmonary edemaRash maculo-papularSkin ulcerationHypotensionConstipationEdema limbsCatheter related infectionAlanine aminotransferase increasedINR increasedHyperglycemiaHematuriaCoughWheezingCardiac arrestPerianal hemorrhageCyst infectionIncarcerated herniaHypoalbuminemiaHypokalemiaRespiratory failurePruritusHypertensionAtrial fibrillationChest pain cardiacLeft ventricular systolic dysfunctionSupraventricular tachycardiaDysphagiaEsophagitisLower gastrointestinal hemorrhagePainOtitis mediaScrotal infectionUpper respiratory infectionUrinary tract infectionWound infectionFallFractureWeight lossAnorexiaHyponatremiaNeck painPolyarthropathySyncopePleuritic pain
Cohort 13411121111231121747521141211131111130000000000000000000000000000000000000000
Cohort 20100000000002000021100000000020000001111111110000000000000000000000000000000
Cohort 30302000420122021234100130100110101010010000001111121110000000000000000000000
Phase II48001204100920302610230060210020001010111051000000131001111111111111111131122

[back to top]

Overall Survival

Defined as the date of first dose of study drug to the date of death from any cause. (NCT01016600)
Timeframe: Until death - median follow-up 4.6 months (full range (0.3-31.4 months))

Interventionmonths (Median)
Cohort 14.2
Cohort 24.5
Cohort 38.9
Phase II4.3

[back to top]

CR With Incomplete Blood Counts Rate

Defined as CR with the exception of neutropenia <1000/uL or thrombocytopenia <100,000/ul. (NCT01016600)
Timeframe: Completion of treatment (median follow-up was 8 weeks) (range 4-68 weeks)

Interventionparticipants (Number)
Cohort 11
Cohort 21
Cohort 31
Phase II1

[back to top]

Cytogenetic CR (CRc) Rate

Only patients with an identified cytogenetic abnormality may receive this designation. Defines as a morphologic complete remission plus reversion to a normal karyotype (no clonal abnormalities detected in a minimum of 20 mitotic cells). (NCT01016600)
Timeframe: Completion of treatment (median follow-up was 8 weeks) (range 4-68 weeks)

Interventionparticipants (Number)
Cohort 11
Cohort 20
Cohort 30
Phase II0

[back to top]

Duration of CR for Complete Responders

(NCT01016600)
Timeframe: Completion of treatment (median follow-up was 8 weeks) (range 4-68 weeks)

Interventionmonths (Median)
Cohort 110.9
Cohort 21.4
Cohort 34.95
Phase II12.15

[back to top]

Event Free Survival

Defined as the interval from the date of first dose of study drug to date of treatment failure, recurrence, or death due to any cause. (NCT01016600)
Timeframe: Until death - median follow-up 4.6 months (full range (0.3-31.4 months))

Interventionmonths (Median)
Cohort 13.8
Cohort 23.4
Cohort 37.8
Phase II2.9

[back to top]

Morphologic Complete Remission Rate (CRm)

Defined as morphologic leukemia-free state, including <5% blasts in BM aspirate with marrow spicules and a count of > 200 nucleated cells and no blasts with Auer rods, no persistent extramedullary disease, ANC > 1000/uL, platelet count > 100,000/uL. Patient must be independent of transfusions for a minimum of 1 week before each marrow assessment. There is no duration requirement for this designation. (NCT01016600)
Timeframe: Completion of treatment (median follow-up was 8 weeks) (range 4-68 weeks)

Interventionparticipants (Number)
Cohort 11
Cohort 20
Cohort 31
Phase II1

[back to top]

Participants With Treatment Emergent Adverse Events (TEAEs)

"Participants with treatment-emergent adverse events (TEAEs) during the treatment period plus 30 days. A participant with multiple occurrences of an adverse event within a category is counted only once in that category. Adverse events were evaluated by the investigator.~The National Cancer Institute (NCI)'s Common Toxicity Criteria for AEs (NCI CTC) was used to grade AE severity. Severity grade 3= severe and undesirable AE. Severity grade 4= life-threatening or disabling AE." (NCT01021423)
Timeframe: up to 9 months

,
Interventionparticipants (Number)
At least one TEAEAt least one TEAE related to study drugAt least one NCI CTC grade 3-4 TEAEAt least one NCI CTC grade 3-4 related to drugAt least one serious TEAEAt least one serious TEAE related to drugTEAE leading to discontinuation of drugRelated TEAE leading to discontinuation of drugTEAE leading to dose reduction or interruptionRelated TEAE - dose reduction or interruption
Lenalidomide4311111111
Placebo4211000011

[back to top]

The Maximum Tolerated Dose (MTD) of Carfilzomib

Determine the MTD of Carfilzomib when combined with Lenalidomide and Dexamethasone. The estimated time to determine the MTD is 6 months. (NCT01029054)
Timeframe: 6 Months

Interventionmg/m^2 (Number)
Carfilzomib, Lenalidomide w/Dexamethasone36

[back to top]

The Percentage of Patients Alive Without Progression

"The Progression Free Survival (PFS) rate will be determined at 12 and 24 months post treatment.~Progressive Disease (PD) is defined as an increase of greater than or equal to 25% from lowest response level in serum M-component and/ or urine M-component and/ or the difference between involved or uninvolved SFLC levels and/ or bone marrow % plasma cells. PD may also be the development of new bone lesions or soft tissue plasmacytomas or the increase in size of existing lesions. PD may also be the development of hypercalcemia." (NCT01029054)
Timeframe: 12 Months and 24 Months Post Treatment

Interventionpercentage of patients (Number)
Percentage of Patients Without Progression at 12 mPercentage of Patients Without Progression at 24 m
Carfilzomib, Lenalidomide w/Dexamethasone9792

[back to top]

The Percentage of Patients That Achieve a Response to Treatment

"The percentage of patients that achieve at least a sCR (Stringent Complete Response), at least a VGPR (Very Good Partial Response) and at least a PR (Partial Response) will be determined.~sCR is defined as:~Negative immunofixation on the serum and urine and~Disappearance of any soft tissue plasmacytomas and~< 5% plasma cells in bone marrow and~Normal SFLC ratio and~Absence of clonal cells in bone marrow~VGPR is defined as:~Serum and urine M-protein detectable by immunofixation but not on electrophoresis or~≥ 90% reduction in serum M-component with urine M-component < 100 mg per 24 hours~PR is defined as:~≥ 50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥ 90% or to < 200 mg per 24 hours~If present at baseline, a ≥ 50% reduction in the size of soft tissue plasmacytomas is also required" (NCT01029054)
Timeframe: 4 Months After Treatment Start

Interventionpercentage of patients (Number)
% of patients that achieve at least a sCR% of patients that achieve at least a VGPR% of patients that achieve at least a PR
Carfilzomib, Lenalidomide w/Dexamethasone428198

[back to top]

Mean Change From Baseline in the Emotional Functioning Domain Associated With the EORTC QLQ-C30 Scale at Weeks 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Emotional Functioning Scale is scored between 0 and 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Least Squares Mean)
Week 12Week 24
Lenalidomide-1.876-1.129
Placebo1.458-6.746

[back to top]

Healthcare Resource Utilization (HRU): Duration of Hospitalizations Due to Adverse Events

Hospitalizations due to adverse events exclude those for transfusions, elective procedures or protocol-driven procedures. HRU was defined as any consumption of healthcare resources directly or indirectly related to the treatment of the patient. (NCT01029262)
Timeframe: From first dose of study drug until 28 days after the last dose, as of the data cut-off date of 17 March 2014; median (minimum, maximum) duration of treatment was 168 (14, 449) and 164 (7, 1158) days in each treatment group respectively.

InterventionDays (Median)
Placebo9.0
Lenalidomide11.0

[back to top]

Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline in the Emotional Functioning Domain at Weeks 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Emotional Functioning Domain was scored between 0 and 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionpercentage of participants (Number)
Week 12Week 24
Lenalidomide20.521.7
Placebo25.017.0

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAE)

"A TEAE was defined as an AE that begins or worsens in intensity of frequency on or after the first dose of study drug through 28 days after last dose of study drug.~A serious adverse event (SAE) is any:~Death;~Life-threatening event;~Any inpatient hospitalization or prolongation of existing hospitalization;~Persistent or significant disability or incapacity;~Congenital anomaly or birth defect;~Any other important medical event~The investigator determined the relationship of an AE to study drug based on the timing of the AE relative to drug administration and whether or not other drugs, therapeutic interventions, or underlying conditions could provide a sufficient explanation for the event. The severity of an AE was evaluated by the investigator according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) (Version 3.0) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death." (NCT01029262)
Timeframe: From the first dose of study drug through 28 days after discontinuation from the study treatment; up to the final data cut-off date of 03 July 2018; maximum exposure was 2100 days in the lenalidomide arm and 529 days in the placebo arm.

,
InterventionParticipants (Count of Participants)
At least 1 TEAE≥ 1 Treatment Related AE (TEAE)≥ 1 Treatment related TEAE Causing Discontinuation≥ 1 TEAE Leading to Dose Reduction≥ 1 TEAE Leading to Dose Interruption≥ 1 TEAE Leading to Dose Interruption & Reduction≥ 1 TEAE Leading to Discontinuation of Study Drug≥ 1 Serious TEAE≥1 Treatment-Related Serious TEAE≥1 Serious TEAE Leading to Dose Reduction≥1 serious TEAE leading to dose interruption≥1 SAE Causing Dose Interruption & reduction≥1 Serious TEAE Leading to Stopping of Study Drug≥1 Grade (GR) 3-4 TEAE≥ 1 GR 3-4 Related TEAE≥ 1 GR 3-4 Leading to Dose Reduction≥ 1 GR 3-4 TEAE Leading to Dose Interruption≥ 1 GR 3-4 TEAE dose Interruption &reduction≥ 1 GR 3-4 TEAE Leading to Stopping of Study Med≥ 1 GR 5 TEAE≥1 GR Treatment Related 5 TEAE
Lenalidomide16014440108968516225121324139127880644163
Placebo744231115916304143516194621

[back to top]

Mean Change From Baseline in the Physical Functioning Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). The EORTC QLQ-C30 Physical Functioning was scored between 0 and 100, with a high score indicating better Global Health Status/QOL. Negative change from Baseline values indicate deterioration in Global Health Status/QOL and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Least Squares Mean)
Week 12Week 24
Lenalidomide-2.919-1.484
Placebo0.732-5.451

[back to top]

Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline Within the Physical Functioning Domain at Weeks 12 and 24

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). Subscale scores are transformed to a 0 to 100 scale, with higher scores on functional scales indicating better function and higher score on symptom scales indicating worse symptoms. A change of at least 10 points on the standardized domain scores was required for it to be considered clinically meaningful. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionpercentage of participants (Number)
Week 12Week 24
Lenalidomide16.424.1
Placebo26.812.8

[back to top]

Percentage of Participants Who Achieved an Erythroid Response Based on the Modified International Working Group (IWG) 2006 Criteria

"A participant was considered as having achieved an erythroid response if the participant either:~- had a hemoglobin (Hgb) increase ≥1.5 g/dL compared to baseline and confirmed by another central laboratory hemoglobin value at 4 to 8 weeks after the first Hgb measurement that also increased ≥1.5 g/dL. All Hgb values during this time interval must have had a ≥ 1.5 g/dL increase (ie, no central laboratory Hgb increase during this timeframe could be less <1.5 g/dL) OR - had a 50% reduction in the number of the RBC transfusion units over any consecutive 56 days period compared to the baseline transfusion burden.~The baseline transfusion burden is the number of units over 112 days by the randomization divided by 2. Only transfusions given for a pre-transfusion Hgb value of 9 g/dL or less were used in this response assessment." (NCT01029262)
Timeframe: From first dose of study drug until 28 days after the last dose, as of the data cut-off date of 17 March 2014; median (minimum, maximum) duration of treatment was 168 (14, 449) and 164 (7, 1158) days in each treatment group respectively.

Interventionpercentage of participants (Number)
Placebo30.4
Lenalidomide38.8

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Physical Functioning Domain at Week 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Physical Functioning Scale was scored between 0 and 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline and Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Mean)
Week 12Week 24
Lenalidomide-2.1-0.4
Placebo-1.4-5.7

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Global Health Status/Quality of Life (QOL) Domain at Week 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Global Health Status/QOL scale was scored between 0 and 100, with a high score indicating better Global Health Status/QOL. Negative change from Baseline values indicate deterioration in Global Health Status/QOL and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline and Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Mean)
Week 12Week 24
Lenalidomide-1.4-2.4
Placebo-2.1-4.1

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Fatigue Domain at Week 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Fatigue Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate reduction in fatigue (i.e. improvement in symptom) and positive values indicate increases in fatigue (i.e. worsening of symptom). (NCT01029262)
Timeframe: Baseline and Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Mean)
Week 12Week 24
Lenalidomide2.4-1.5
Placebo0.67.6

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Emotional Functioning Domain at Week 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Emotional Functioning Domain was scored between 0 and 100, with a high score indicating better functioning. Negative change from Baseline values indicate deterioration in functioning and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline and Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Mean)
Week 12Week 24
Lenalidomide-1.40.8
Placebo1.2-7.1

[back to top]

Mean Change From Baseline in the EORTC QLQ-C30 Dyspnea Domain at Week 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Dyspnea scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate decreased dyspnea (i.e. improvement in symptom) and positive values indicate increased dyspnea (i.e. worsening of symptom). (NCT01029262)
Timeframe: Baseline and Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Mean)
Week 12Week 24
Lenalidomide2.21.2
Placebo0.64.3

[back to top]

Mean Change From Baseline in the Global Health Status/QoL Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). The EORTC QLQ-C30 Global Health Status/QOL scale was scored between 0 and 100, with a high score indicating better Global Health Status/QOL. Negative change from Baseline values indicate deterioration in Global Health Status/QOL and positive values indicate improvement. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Least Squares Mean)
Week 12Week 24
Lenalidomide-2.690-2.441
Placebo-1.201-4.502

[back to top]

Mean Change From Baseline in the Dyspnea Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Dyspnea scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate decreased dyspnea (i.e. improvement in symptom) and positive values indicate increased dyspnea (i.e. worsening of symptom). (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Least Squares Mean)
Week 12Week 24
Lenalidomide3.374-0.206
Placebo1.6965.998

[back to top]

Mean Change From Baseline in Fatigue Domain Associated With the EORTC QLQ-C-30 Scale at Week 12 and Week 24

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). The EORTC QLQ-C30 Fatigue Scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate reduction in fatigue (i.e. improvement in symptom) and positive values indicate increases in fatigue (i.e. worsening of symptom). (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionunits on a scale (Least Squares Mean)
Week 12Week 24
Lenalidomide3.4970.196
Placebo-0.4647.376

[back to top]

Compliance Rates Using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) From Baseline to Week 48

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). Subscale scores are transformed to a 0 to 100 scale, with higher scores on functional scales indicating better function and higher score on symptom scales indicating worse symptoms. A participant was considered compliant at a visit if at least 15 out of the QLQ-C30 items in the questionnaire were checked. (NCT01029262)
Timeframe: Baseline, Week 12, (±3 days), Week 24, (±3 days), Week 36, (±3 days), and Week 48 (±3 days); up to data cut-off of 17 Mar 2014

,
Interventionpercentage of participants (Number)
BaselineWeek 12Week 24Week 36Week 48
Lenalidomide9083.885.880.571.9
Placebo88.678.580.610050

[back to top]

Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline in the Dyspnea Domain at Weeks 12 and 24

The European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) questionnaire (EORTC QLQ-C30) is a 30-question tool used to assess the overall quality of life in cancer patients. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact). The EORTC QLQ-C30 Dyspnea scale is scored between 0 and 100, with a high score indicating a higher level of symptoms. Negative change from Baseline values indicate decreased dyspnea (i.e. improvement in symptom) and positive values indicate increased dyspnea (i.e. worsening of symptom). Improvement means at least 10 points better compared to baseline. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionpercentage of participants (Number)
Week 12Week 24
Lenalidomide21.320.5
Placebo19.612.8

[back to top]

Percentage of Participants Who Achieved an Erythroid Response Based on Original IWG 2006 Criteria

"A participant was considered as having achieved an erythroid response when:~- a Hgb increase ≥1.5 g/dL compared to baseline and confirmed by another central laboratory hemoglobin value at 4 to 8 weeks after the first Hgb measurement that had also increased ≥1.5 g/dL for at least 8 weeks. All Hgb values during this time interval must have had a ≥ 1.5 g/dL increase (ie, no central laboratory Hgb increase during this timeframe can be less than a 1.5 g/dL) OR - had an absolute reduction of 4 RBC transfusion units over any consecutive 56 days period compared to the baseline transfusion burden.~The baseline transfusion burden is the number of units over the 112 days prior to randomization divided by 2. Only transfusions given for a pre-transfusion Hgb value of 9.5 g/dL or less may be used in this response assessment." (NCT01029262)
Timeframe: From first dose of study drug until 28 days after the last dose, as of the data cut-off date of 17 March 2014; median (minimum, maximum) duration of treatment was 168 (14, 449) and 164 (7, 1158) days in each treatment group respectively.

Interventionpercentage of participants (Number)
Placebo20.3
Lenalidomide35.6

[back to top]

Percentage of Participants With a Erythroid Gene Signature Who Achieved RBC Transfusion Independence for ≥ 56 Days as Determined by an Independent Review Committee (IRC)

"The percentage of participants who achieved the 56-day RBC TI response was defined as the absence of any RBC transfusions during any consecutive rolling 56-day interval within the double-blind treatment phase (ie, Days 2 (Day 1 is the first study drug day) to 57, Days 3 to 58, etcetera). A participant who achieved at least a 56-day RBC-transfusion-independent response was considered a 56-day RBC-TI responder." (NCT01029262)
Timeframe: From first dose of study drug until 28 days after the last dose, as of the data cut-off date of 17 March 2014; median (minimum, maximum) duration of treatment was 168 (14, 449) and 164 (7, 1158) days in each treatment group respectively.

Interventionpercentage of participants (Number)
Placebo0.0
Lenalidomide7.1

[back to top]

Percentage of Participants With a Clinically Meaningful Improvement in HRQOL Associated With the EORTC QLQ-C-30 Scale From Baseline in the Global Health Status/QOL Domain at Weeks 12 and 24

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). Subscale scores are transformed to a 0 to 100 scale, with higher scores on functional scales indicating better function and higher score on symptom scales indicating worse symptoms. A change of at least 10 points on the standardized domain scores was required for it to be considered clinically meaningful. (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionpercentage of participants (Number)
Week 12Week 24
Lenalidomide22.126.5
Placebo19.614.9

[back to top]

Percentage of Participants With a Clinically Meaningful Improvement in QOL (EORTC QLQ-C-30 Scale) From Baseline in Fatigue Domain at Weeks 12 and 24

The European Organization for Research and Treatment of Cancer QOL Questionnaire for Patients with Cancer (EORTC QLQ-C30) was a 30-item oncology-specific questionnaire. The questionnaire was developed to assess the quality of life of cancer patients. It contains 30 questions, 24 of which form 9 multi-item scales representing various aspects of HRQOL: 1 global scale, 5 functional scales (Physical, Role, Emotional, Cognitive and Social), and 3 symptom scales (Fatigue, Pain, and Nausea). The remaining 6 items are intended to be mono-item scales describing relevant cancer-oriented symptoms (dyspnea, insomnia, appetite, constipation, diarrhea, financial difficulties). Subscale scores are transformed to a 0 to 100 scale, with higher scores on functional scales indicating better function and higher score on symptom scales indicating worse symptoms. Improvement means at least 10 points better compared to baseline (NCT01029262)
Timeframe: Baseline, Week 12, ±3 days and Week 24, ±3 days

,
Interventionpercentage of participants (Number)
Week 12Week 24
Lenalidomide39.338.6
Placebo30.429.8

[back to top] [back to top]

Percentage of Participants Who Achieved Red Blood Cell (RBC) Transfusion Independence for ≥ 56 Days as Determined by an Independent Review Committee (IRC)

"The percentage of participants who achieved the 56-day RBC transfusion independent (TI) response was defined as the absence of any RBC transfusions during any consecutive rolling 56-day interval within the double-blind treatment phase (ie, Days 2 (Day 1 is the first study drug day) to 57, Days 3 to 58, etcetera). The double-blind treatment phase was defined as the period between the 1st dosing up until 28 days after the last study drug dose" (NCT01029262)
Timeframe: From first dose of study drug until 28 days after the last dose, as of the data cut-off date of 17 March 2014; median (minimum, maximum) duration of treatment was 168 (14, 449) and 164 (7, 1158) days in each treatment group respectively.

Interventionpercentage of participants (Number)
Placebo2.5
Lenalidomide26.9

[back to top] [back to top] [back to top]

Kaplan Meier Estimates for Progression to Acute Myeloid Leukemia (AML)

Progression to AML is part of the natural course of MDS and is a manifestation of disease progression. The time to progress to AML was calculated from the day of randomization to the first day when AML was diagnosed. Participants who died without AML were censored at the date of death. The participants who were lost to follow-up were censored at the last known day when participants did not have AML. Participants who did not progress to AML at the last follow-up contact were censored at the day of the last follow-up contact. (NCT01029262)
Timeframe: From randomization to final data cut-off date of 03 Jul 2018; median follow up time for progression to AML was 2.3 years (range = 0 to 5.0 years) in the placebo arm and 2.6 years (range = 0 to 6.4 years) in the lenalidomide arm.

Interventionyears (Median)
PlaceboNA
LenalidomideNA

[back to top]

Kaplan Meier Estimate for Overall Survival (OS)

Overall survival was assessed using the time between randomization and the date of death or date of censoring. Participants who were alive at a data cutoff date and participants who were lost to follow-up were censored at the last date when participants were known to be alive. (NCT01029262)
Timeframe: From randomization to final data cut-off date of 03 July 2018; maximum survival follow up was 6.4 years

Interventionyears (Median)
Placebo3.0
Lenalidomide3.8

[back to top] [back to top]

Healthcare Resource Utilization (HRU): Number of Days of Hospitalization Due to Adverse Events Per Person-Years

Hospitalizations due to adverse events exclude those for transfusions, elective procedures or protocol-driven procedures. HRU was defined as any consumption of healthcare resources directly or indirectly related to the treatment of the patient (NCT01029262)
Timeframe: From first dose of study drug until 28 days after the last dose, as of the data cut-off date of 17 March 2014; median (minimum, maximum) duration of treatment was 168 (14, 449) and 164 (7, 1158) days in each treatment group respectively.

InterventionDays per person-years (Number)
Placebo6.37
Lenalidomide8.92

[back to top]

Best Overall Response Assessed by an Independent Review Using Response Evaluation Criteria In Solid Tumors (RECIST 1.1) During the Proof of Concept Period Prior to Early Study Termination

"Tumor response was evaluated every 2 cycles beginning with Cycle 3 Day 1 and at treatment discontinuation. Response and progression were evaluated using the RECIST 1.1 criteria (Eisenhauer, 2009).~Complete response-disappearance of all lesions~Partial response-30% decrease in the sum of diameters of target lesions from baseline~Stable disease-neither shrinkage nor increase of lesions.~Progressive Disease-20% increase in the sum of diameters of target lesions from nadir.~Participants with evidence of objective tumor response have the response confirmed with repeat assessments performed at the next scheduled scan." (NCT01032291)
Timeframe: Week 9 up to week 24

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseResponse Not Evaluable
Lenalidomide (Proof of Concept)003135
Lenalidomide Plus Cetuximab (Proof of Concept)005133

[back to top]

Participants With Dose Limiting Toxicities (DLTs) During the First Treatment Cycle of the Safety Lead-In Period

"The number of participants with DLTs determines the maximum tolerated dose of the combination therapy used in the Proof of Concept (POC) period:~If <2 of the initial 6 participants experience a DLT, then the POC will start with lenalidomide at 25 mg.~If ≥2 of the initial 6 participants experienced a DLT, then 6 more subjects were to be enrolled at 20 mg lenalidomide.~If <2 of the additional 6 subjects experienced a DLT, then the lenalidomide starting dose for the POC was to be 20 mg.~If ≥2 of the additional 6 subjects experienced a DLT, then 6 more subjects were to be enrolled at 15 mg lenalidomide.~If <2 of the additional 6 subjects experienced a DLT, then the POC was to start with lenalidomide at 15 mg.~If ≥2 of the additional 6 subjects experienced a DLT, the dosing for the study was to be reassessed by Celgene Corporation and the investigators." (NCT01032291)
Timeframe: Up to Day 28 (Cycle 1)

Interventionparticipants (Number)
Lenalidomide Plus Cetuximab (Safety Lead-in)1

[back to top]

Participants With Treatment-Emergent Adverse Events (TEAE)

TEAEs are any adverse event occurring or worsening on or after the first treatment of any study drug and within 28 days after the last dose of the last study drug received. Relation to study drug was determined by the investigator. Severity of AE is graded according to National Cancer Institute Common Toxicity Criteria for Adverse Events (NCI CTCAE) Version 4.0. Severity is a 5-point scale: 3= severe or medically significant but not life-threatening 4=life-threatening, urgent intervention required 5=death related to AE. (NCT01032291)
Timeframe: up to week 28

,,
Interventionparticipants (Number)
>=1 TEAESerious TEAETEAE leading to discontinuing lenalidomideTEAE leading to discontinuing cetuximabTEAE leading to reduction/interruption of lenalidoTEAE leading to reduction/interruption of cetuximaTEAE related to lenalidomideTEAE related to cetuximabTEAE NCI CTC grade 3 or higherTEAE NCI CTC grade 3+ related to lenalidomideTEAE NCI CTC grade 3+ related to cetuximabSerious TEAE related to lenalidomideSerious TEAE related to cetuximab
Lenalidomide (Proof of Concept)2097NA6NA9NA134NA0NA
Lenalidomide Plus Cetuximab (Proof of Concept)21956971319124722
Lenalidomide Plus Cetuximab (Safety Lead-In)8533215843222

[back to top]

Duration of Response

The response duration was measured from the last of the consecutive 56 days during which the subject was free of red blood cells (RBC) transfusions to the date of the first RBC transfusion after the 56-day RBC-transfusion-free period. (NCT01034592)
Timeframe: 6 months

Interventiondays (Median)
Lenalidomide0

[back to top]

Hemoglobin Concentration

The effect on hemoglobin concentration was assessed as the change from baseline, measured in g/dL. (NCT01034592)
Timeframe: 6 months

Interventiong/dL (Median)
Lenalidomide0.4

[back to top]

Red Blood Cell (RBC) Transfusion Independence

"Red blood cell (RBC) transfusion independence is reported as the number of subjects who achieve a continuous absence of the intravenous infusion of any RBC transfusion during any consecutive rolling 56 days during the treatment period." (NCT01034592)
Timeframe: 6 months

Interventionparticipants (Number)
Lenalidomide0

[back to top]

Platelet Response

The effect on platelet levels as assessed as the change in platelet count from baseline. (NCT01034592)
Timeframe: 6 months

Intervention1000/uL (Median)
Lenalidomide25.5

[back to top]

Toxicity

Toxicity was assessed as the number of adverse events related to lenalidomide. (NCT01034592)
Timeframe: 6 months

InterventionRelated Adverse Events (Number)
Lenalidomide1

[back to top]

Red Blood Cell (RBC) Transfusions

The effect on red blood cell (RBC) transfusions was assessed as the number of participants that achieved a greater than 50% decrease in RBC transfusion requirements. (NCT01034592)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Lenalidomide0

[back to top]

Neutrophil Response

The effect on neutrophil levels was assessed as the change in neutrophil count from baseline. (NCT01034592)
Timeframe: 6 months

Intervention1000/uL (Median)
Lenalidomide0.50

[back to top]

Overall Survival

The Kaplan-Meier method will be used to estimate the overall survival distribution. This outcome only reports data as it pertains to overall survival at one year. All-cause mortality includes survival for follow up for all subjects on the study. (NCT01035463)
Timeframe: 1 year

Interventionpercentage of participants (Number)
All Phase I Participants100
All Phase II Participants95

[back to top]

Maximum Tolerated Dose of Lenalidomide (Phase I)

The Maximum Tolerated Dose (MTD) is defined to be the dose cohort below which 3 out of 6 subjects experience dose limiting toxicities during cycle 1. Dose limiting toxicities graded using the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 (NCT01035463)
Timeframe: Cycle 1, 28 days

Interventionmilligrams PO daily (Number)
Treatment (Stem Cell Transplantation)10

[back to top]

Event-free Survival

The Kaplan-Meier method will be used to estimate the event-free survival distribution. (NCT01035463)
Timeframe: 1 year

Interventionpercentage of participants (Number)
All Phase I Participants84
All Phase II Participants87

[back to top]

Overall Response: Number of Participants With CR or CRi Response

Response defined as complete remission (CR) or complete remission with incomplete platelet recovery (CRi) for AML or any response for myelodysplastic syndrome (MDS) using international working group (IWG)-06 criteria. Complete response (CR) requires normalization of peripheral counts (absolute neutrophil count 10^9/L or more, platelet count 100 x 10^9/L or more), and a bone marrow with 5% or less marrow blasts. A hematologic improvement (HI) is defined as a CR with a platelet count above 30 x 10^9/L, without the need for transfusion of Platelets. (NCT01038635)
Timeframe: 6 months

Interventionparticipants (Number)
Complete Remission (CR)Incomplete Count Recover (CRi)
Overall Study: 5-AZA + LEN MTD1516

[back to top]

Overall Response Rate (ORR) of Lenalidomide in Combination With 5-azacytidine (5-AZA) in Participants With Leukemia

Response defined as complete remission (CR) or complete remission with incomplete platelet recovery (CRi) for AML or any response for myelodysplastic syndrome (MDS) using international working group (IWG)-06 criteria. Complete response (CR) requires normalization of peripheral counts (absolute neutrophil count 10^9/L or more, platelet count 100 x 10^9/L or more), and a bone marrow with 5% or less marrow blasts. A hematologic improvement (HI) is defined as a CR with a platelet count above 30 x 10^9/L, without the need for transfusion of Platelets. (NCT01038635)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Phase I: 5-AZA + LEN MTD14
Phase II: 5-AZA + LEN45

[back to top]

Number of Dose Limiting Toxicities for Determining Maximum Tolerated Dose (MTD) of Lenalidomide in Combination With 5-azacytidine (5-AZA)

DLT determined only during first course of therapy, at least 28 days from treatment of last participant before a new dose level initiated. All severe (Grade 3-4) non-hematological toxicities that are drug related considered for DLT determination. If 1 participant develops grade III-IV non-hematological toxicity, 3 more will be accrued at that particular dose level. If 2 or more participants develop grade III-IV non-hematologic toxicity, the doses of the combination at which this occurs will be considered too toxic. A total of 10 patients will be treated at the maximally tolerated dose (MTD) of the combination (the dose level below that considered to be too toxic) to confirm its tolerability. (NCT01038635)
Timeframe: 3-8 week cycles, up to 24 weeks

Interventionparticipants (Number)
5-AZA + 5 Days LEN 10 mg0
5-AZA + 5 Days LEN 15 mg0
5-AZA + 5 Days LEN 20 mg0
5-AZA + 5 Days LEN 25 mg0
5-AZA + 5 Days LEN 50 mg0
5-AZA + 5 Days LEN 75 mg0
5-AZA + 10 Days LEN 75 mg 75 mg0

[back to top]

Progression Free Survival (Phase II)

The progression-free survival time is defined as the time from registration to disease progression (PD=Increase of > 25% from lowest response value in Serum/Urine M-component) while receiving bendamustine, lenalidomide, and dexamethasone or death due to any cause, whichever comes first. The distribution of progression-free survival will be estimated using the method of Kaplan-Meier. Treatment response was assessed using the International Myeloma Working Group uniform criteria. (NCT01049945)
Timeframe: Up to 2 years from study completion

Interventionmonths (Median)
Maximum Tolerated Dose, Dose Level 411.8

[back to top]

Overall Survival (Phase II)

The overall 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. The overall survival rate at 6 months is defined as the percentage of participants who are alive at 6 months. (NCT01049945)
Timeframe: at 6 months

Interventionpercentage of participants (Number)
Maximum Tolerated Dose, Dose Level 487

[back to top]

Event Free Survival (Phase II)

The event-free survival time is defined as the time from registration to disease progression (PD=Increase of > 25% from lowest response value in Serum/Urine M-component) while receiving bendamustine, lenalidomide, and dexamethasone, death due to any cause, or subsequent treatment for multiple myeloma. The distribution of event-free survival will be estimated using the method of Kaplan-Meier. Treatment response was assessed using the International Myeloma Working Group uniform criteria. (NCT01049945)
Timeframe: Up to 2 years from study completion

Interventionmonths (Median)
Maximum Tolerated Dose, Dose Level 45.6

[back to top]

Dose Limiting Toxicity of Bendamustine Hydrochloride and Lenalidomide in Combination With Dexamethasone (Phase I)

"The Maximum Tolerated Dose (MTD) is the dose level below that at which a dose limiting toxicity (DLT) is observed in ≥ 33% (i.e., ≥ 2 of 6) subjects in a cohort. A dose limiting toxicity is defined as one of the following adverse events in the Common Terminology Criteria for Adverse Events (CTCAE) v 3.0 deemed at least possibly related to treatment:~Grade 2 neuropathy with pain~Any grade 3 Non-Hematologic toxicity~Any grade Non-Hematologic event requiring a dose reduction in cycle 1 or delaying the next cycle by >14 days.~Grade 4 neutropenia~Febrile neutropenia~Grade 4 thrombocytopenia~Grade 3 thrombocytopenia associated with bleeding~Any Hematologic event requiring a dose reduction in cycle 1 or a delay in the next cycle of treatment by >14 days.~We are reporting the results of this endpoint as the number of DLTs per dose level." (NCT01049945)
Timeframe: One cycle of treatment

InterventionDose Limiting Toxic Events (Number)
Phase I, Dose Level 10
Phase I, Dose Level 21
Phase I, Dose Level 30
Phase I, Dose Level 40
Phase I, Dose Level 52

[back to top]

Confirmed Response Rate (Dose Level 4) Reported as the Percentage of Patients Achieving a Confirmed Response (sCR, CR, VGPR, or PR).

"Complete response (CR)~- Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and <5% plasma cells in bone marrow.~Stringent complete response (sCR) - A CR plus normal FLC ratio and no clonal cells in bone marrow~Near complete response (nCR) A CR, with the persistence of original monoclonal protein~Very good partial response (VGPR)~- Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-component plus urine M-component <100 mg per 24 h~Partial response (PR)~≥50% reduction of serum M-protein and reduction in 24-h urinary M-protein by ≥90% or to <200 mg per 24 h.~a ≥50% decrease in the difference between involved and uninvolved FLC levels~or a ≥50% reduction in plasma cells is required in place of M-protein, if ≥30% at baseline." (NCT01049945)
Timeframe: Up to 6 cycles of treatment

Interventionpercentage of participants (Number)
Maximum Tolerated Dose, Dose Level 444

[back to top]

Duration of Response (DOR) (Phase II)

DOR is the time from the date the patient's objective status is first noted to be PR or better to the earliest date of progression (PD=Increase of > 25% from lowest response value in Serum/Urine M-component) is documented. Treatment response was assessed using the International Myeloma Working Group uniform criteria. Complete response (CR)=Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and <5% plasma cells in bone marrow. Stringent complete response (sCR)=A CR plus normal FLC ratio and no clonal cells in bone marrow. Near complete response (nCR)=A CR, with the persistence of original monoclonal protein. Very good partial response (VGPR) =Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-component plus urine M-component <100 mg per 24 h, Partial response (PR)=≥50% reduction of serum M-protein and reduction in 24-h urinary M-protein by ≥90% or to <200 mg per 24 h. (NCT01049945)
Timeframe: Up to 2 years from study completion

Interventionmonths (Median)
Maximum Tolerated Dose, Dose Level 424.4

[back to top]

Complete Response Rate at 6 Months

16 of 17 patients proceeded to transplant. 6 month CR rate post transplant was 8/16 (50%). (NCT01050790)
Timeframe: 6 months

Interventionpercentage of participants (Number)
5-azacytidine + Lenalidomide -> Auto Stem Cell Transplant50

[back to top]

Time to Progression Post Transplant

Time to progression post transplant: For patients not in Complete Response (CR), progressive disease requires one or more: >25% increase in the level of the serum monoclonal paraprotein(absolute increase of at least 0.5 g/dL); > 25% increase in 24-hour urinary light chain excretion(absolute increase of at least 200m/24 hours). Increase plasma cells in a bone marrow aspirate( absolute increase of at least 10%). Definite increase in the size of existing bone lesions or soft tissue plasmacytomas. Development of new bone lesions or soft tissue plasmacytomas. Development of hypercalcemia (corrected serum Ca > 11.5 mg/dL or > 2.65 mmol/L) not attributable to any other cause. All relapse categories require two consecutive assessments made any time before classification as relapse or progressive disease. (NCT01050790)
Timeframe: 28 months

Interventionmonths (Median)
5-azacytidine + Lenalidomide -> Auto Stem Cell Transplant14.9

[back to top]

CTA Expression Before and After Azacitidine Therapy

Six patients tested have demonstrated CTA up-regulation in either unfractionated bone marrow (n = 4) or CD138+ cells (n = 2). CTA (CTAG1B)-specific T cell response has been observed in all three patients tested and persists following SCT. (NCT01050790)
Timeframe: 3 months

Interventionparticipants (Number)
CTA up-regulationCD138+ cellsunfractionated bone marrowCTA (CTAG1B)-specific T cell response
5-azacytidine + Lenalidomide -> Auto Stem Cell Transplant6243

[back to top]

Feasibility to Mobilize and Infuse Autologous Lymphocytes (ALI) After Immunomodulatory Therapy and After Stem Cell Transplant Engraftment

Time frame is post 2nd and 3rd cycles of rev/aza and after stem cell transplant engraftment. (NCT01050790)
Timeframe: 6 months

Interventionparticipants (Number)
mobilize lymphocyteslymphocytes infused post transplantnot able to mobilize stem cells/no transplant
5-azacytidine + Lenalidomide -> Auto Stem Cell Transplant17161

[back to top]

Progression-free and Overall Survival

"Survival and event-free survival curves (any event of fatality, relapse, acute or chronic GVHD) with Kaplan-Meier curves. The incidence curve for relapse - accounting for the competing risk of fatality - is plotted with step-wise curves. The R statistical software (version 2.15) was used for all time-to-event analyses, with the survival package used for survival curves, and the cmprsk package used for all competing risk curves.~Results: The one-year survival rate is 93.3% (SE = 0.4%), and the two-year survival rate is 86.1% (SE = 0.9%)." (NCT01050790)
Timeframe: 1 year to 2 years

Interventionpercentage of participants (Number)
one- year survival ratetwo-year survival rateone-year relapse ratetwo-year relapse rateone-year event free survival ratetwo-year event free survival rate
5-azacytidine + Lenalidomide -> Auto Stem Cell Transplant93.386.112.519.287.567.3

[back to top]

Toxicity as Assessed by NCI CTCAE v3.0

Time frame includes after stem cell transplant engraftment. Toxicity post ALI infusion: 1 patient grade 1 hypertension 90 min post infusion. Toxicity post Rev maintenance: 1 patient not tolerated, 1 patient dose decreased due to counts. (NCT01050790)
Timeframe: 6 months

Interventionparticipants (Number)
Toxicity post ALI infusionToxicity post Rev maintenance
5-azacytidine + Lenalidomide -> Auto Stem Cell Transplant12

[back to top]

Combined Therapy - Median Progression Free Survival

Progression free survival (PFS) of older adults with mildly symptomatic multiple myeloma treated on this response adapted approach (i.e. time from start of lenalidomide to failure of lenalidomide and low dose dexamethasone) (NCT01054144)
Timeframe: up to 36 months

Interventionmonths (Median)
Response Adapted Therapy36

[back to top]

Number of Participants With 1 Year Overall Survival (OS)

The 1 year overall survival of older adults with mildly symptomatic multiple myeloma treated on this response adapted approach (NCT01054144)
Timeframe: 1 Year

Interventionparticipants (Number)
Response Adapted Therapy27

[back to top]

Number of Participants With Serious Adverse Events

Number of participants with serious adverse events (NCT01054144)
Timeframe: Day 1 through Off Study Date, an average of 48 months

Interventionparticipants (Number)
Response Adapted Therapy20

[back to top]

Single Agent - Median Progressive Free Survival (PFS)

The progression free survival of patients treated with single agent lenalidomide (NCT01054144)
Timeframe: First measure at 8 weeks

Interventionmonths (Median)
Single Agent29

[back to top]

Response Rate

Response rate in older adults with mildly symptomatic multiple myeloma to single agent lenalidomide, lenalidomide prednisone and lenalidomide low dose dexamethasone in patients with suboptimal responses to lenalidomide monotherapy. The study used the uniform response assessment of the International Myeloma Working Group with the addition of MR (minimal response) (Durie et al, 2006; Kumar et al, 2016). MR was defined as a 25-49% decrease in serum M spike, and a 50-89% improvement in urine M spike. For patients without a measurable serum or urine M spike, a 25-49% decrease in the difference between the involved and uninvolved free light chains was required. The response in this trial is defined as complete remission (CR), stringent complete remission (SRC), very good partial remission (VGPR) and partial remission (PR) and minimal response (MR). (NCT01054144)
Timeframe: Every 8 weeks up to 12 months

,
InterventionParticipants (Count of Participants)
Complete Response & Stringent Complete ResponseVery Good Partial Response (VGPR)Partial Response (PR)Minimal Response (MR)Stable DiseaseOverall Response >/= PR
Response Adapted Therapy112504
Single Agent Lenalidomide43124319

[back to top]

Tumor Response Rate

"Percentage of patients who achieve a partial or complete response Complete response was defined as the absence of any detectable residual disease, including tumor-associated edema, that persisted for at least 4 weeks.~Partial response was defined as no new lesions (skin or oral), or new visceral sites of involvement (or the appearance or worsening of tumor-associated edema or effusions), and a 50% or greater decrease in the number of all previously existing lesions that lasted for at least 4 weeks, or complete flattening of at least 50% of all previously raised lesions, or a 50% or greater decrease in the sum of the products of the largest perpendicular diameters of the marker lesions." (NCT01057121)
Timeframe: Up to 30 days after completion of study treatment

Interventionpercent of participants who responded (Number)
Phase I - 10 mg/Day (Lenalidomide)0
Phase I - 15 mg/Day (Lenalidomide)0
Phase I: 20 mg/Day Lenalidomide50
Phase I: 25 mg/Day Lenalidomide80
Phase II: 25 mg/Day Lenalidomide53

[back to top] [back to top]

Time to Death

Percentage of patients who died (NCT01057121)
Timeframe: Up to 30 days after completion of study treatment

Interventionpercentage of participants who died (Number)
Phase I - 10 mg/Day Lenalidomide0
Phase I - 15 mg/Day Lenalidomide0
Phase I: 20 mg/Day Lenalidomide0
Phase I: 25 mg/Day Lenalidomide0
Phase II: 25 mg/Day Lenalidomide0

[back to top]

Time to Response

"time from enrollment to first response (complete or partial) as defined below: Complete response is defined as the absence of any detectable residual disease, including tumor-associated edema, persisting for at least 4 weeks.~Partial response is defined as no new lesions (skin or oral), or new visceral sites of involvement (or the appearance or worsening of tumor-associated edema or effusions), and a 50% or greater decrease in the number of all previously existing lesions lasting for at least 4 weeks, or complete flattening of at least 50% of all previously raised lesions, or a 50% or greater decrease in the sum of the products of the largest perpendicular diameters of the marker lesions." (NCT01057121)
Timeframe: Up to 30 days after completion of study treatment

Interventionweeks (Median)
Phase I: 20 mg/Day Treatment (Lenalidomide)8
Phase I: 25 mg/Day Treatment (Lenalidomide)20.5
Phase II: 25 m/Day Treatment (Lenalidomide)13

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Lenalidomide

The maximum tolerated dose (MTD) will be defined as the next lowest dose cohort below where ≥ 2/3 or ≥ 3/6 patients experience dose limiting toxicities in cycle 1. (NCT01060384)
Timeframe: 7 months

Interventionmilligrams (Number)
Phase 110

[back to top]

Phase I and Phase II: Event Free Survival and Overall Survival

"Event free survival is defined as the time from start of treatment to disease progression or death from any cause. Overall survival (OS) is defined as the time from start of treatment to death from any cause. A response-evaluable subject will be considered anyone who completes at least 2 cycles of therapy with documented response or documented progression of disease after at least one complete cycle of therapy but, prior to 2 complete cycles of therapy.~A response-evaluable subject will be considered anyone who completes at least 2 cycles of therapy with documented response or documented progression of disease after at least one complete cycle of therapy but, prior to 2 complete cycles of therapy. A non-evaluable subject will be one who receives less than one complete cycle of therapy (ie. 4 infusions of ofatumumab and 21 days of lenalidomide). A non-evaluable subject will also be one that has no documented response prior to treatment withdrawal." (NCT01060384)
Timeframe: 2 years from start of treatment

Interventionpercent of participants (Number)
Estimated 1 year Event Free SurvivalEstimated 1 year Overall SurvivalEstimated 2 year Event Free SurvivalEstimated 2 year Overall Survival
Phase II: Lemalidomide at MTD and Ofatumumab38532044

[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]

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]

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]

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]

Incidence of Dose-limiting Toxicity (DLT), Phase I Patients Only

Incidence of dose-limiting toxicity (DLT) defined as any grade 3 or 4 adverse events as graded by the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (Phase I) (NCT01076543)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
Phase I, All Histologies, Lenalidomide 15 mg1
Phase I, All Histologies, Lenalidomide 20 mg0
Phase I, All Histologies, Lenalidomide 25 mg3
Phase II, Diffuse Large B-Cell Subtype0
Phase II, Follicular Subtype0
Phase II, Lymphoma NOS0

[back to top]

Complete Response (Phase II)

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by CT or MRI: Complete Response (CR), Disappearance of all target lesions; (NCT01076543)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Phase I, All Histologies, Lenalidomide 15 mg0
Phase I, All Histologies, Lenalidomide 20 mg0
Phase I, All Histologies, Lenalidomide 25 mg0
Phase II, Diffuse Large B-Cell Subtype5
Phase II, Follicular Subtype5
Phase II, Lymphoma NOS7

[back to top]

Overall Response Rate (Phase II)

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and 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 lesions; Overall Response (OR) = CR + PR. (NCT01076543)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Phase I, All Histologies, Lenalidomide 15 mg0
Phase I, All Histologies, Lenalidomide 20 mg0
Phase I, All Histologies, Lenalidomide 25 mg0
Phase II, Diffuse Large B-Cell Subtype10
Phase II, Follicular Subtype7
Phase II, Lymphoma NOS25

[back to top]

Overall Survival (OS) (Phase II)

Kaplan-Meier curves will be generated for OS stratified by histology; median OS times will be determined and 90% confidence intervals derived as described in Brookmeyer and Crowley. (NCT01076543)
Timeframe: Time from study entry until death from any cause, assessed up to 6 years

InterventionMonths (Median)
Phase II, Diffuse Large B-Cell Subtype9.1
Phase II, Follicular Subtype35.8
Phase II, Lymphoma NOS25.5

[back to top]

Progression-free Survival (PFS) (Phase II)

Kaplan-Meier curves will be generated for PFS stratified by histology; median PFS times will be determined and 90% confidence intervals derived as described in Brookmeyer and Crowley. (NCT01076543)
Timeframe: Time from study entry until disease progression or death from any cause, assessed up to 5 years

InterventionMonths (Median)
Phase II, Diffuse Large B-Cell Subtype7.0
Phase II, Follicular Subtype27.7
Phase II, Lymphoma NOS7.0

[back to top]

Participants With Grade 3 =/> Adverse Events

Number of participants experiencing adverse events above a Grade 3 according to the Common Terminology Criteria for Adverse Events (CTCAE) version 2. (NCT01079936)
Timeframe: Day 90 after stem cell transplant

Interventionparticipants (Number)
25 Mg Lenalidomide3
50 mg Lenalidomide5
75 mg Lenalidomide15
100 mg Lenalidomide17

[back to top]

Number of Participants With Response (CR at Day 90)

Response is defined as the event that the participant is alive with complete response (CR) at day 90 (+/-30 days). CR defined as: A) Absence of monoclonal protein in urine and serum when analyzed by immunofixation electrophoresis. B) The bone marrow should be normal by morphological examination with <5% plasma cells. There should be < 1% aneuploid light chain restricted population by flow cytometry for DNA/cIg. C) While healing of bone lesions not required, no new lytic lesion should appear. Further compression fracture of spine will be not considered as progressive disease. (NCT01079936)
Timeframe: Day 90 after stem cell transplant

Interventionparticipants (Number)
25 Mg Lenalidomide0
50 mg Lenalidomide0
75 mg Lenalidomide4
100 mg Lenalidomide4

[back to top]

Number of Participants With Day 30 DLT (Overall Study, Phase I/Phase II)

Dose limiting toxicity (DLT) was defined as regimen-related death, graft failure, grade 3 or 4 atrial fibrillation, grade 4 deep venous thrombosis, or pulmonary embolism before day 30 after auto-HCT. (NCT01079936)
Timeframe: Day 30 following transplant

Interventionparticipants (Number)
25 Mg Lenalidomide0
50 mg Lenalidomide0
75 mg Lenalidomide2
100 mg Lenalidomide0

[back to top]

Maximum Tolerated Dose (MTD) of Lenalidomide

There were 4 doses of lenalidomide in the dose escalation phase: 25 mg, 50 mg, 75 mg, and 100 mg. The first 12 patients were treated at these dose levels (3 patients per level) and safety assessed at each level. The MTD dose level was to be the level at which participants at each lenalidomide dose level had no dose limiting toxicity (DLT). DLT defined as as regimen-related death, graft failure, grade 3 or 4 atrial fibrillation, grade 4 deep venous thrombosis, or pulmonary embolism before day 30 after auto-HCT. Each participant received a fixed dose of Melphalan plus one of the four doses 25, 50, 75 or 100 mg of Lenalidomide orally for each of 7 days, -8 to -2 pre transplant. (NCT01079936)
Timeframe: Assessed at 21-28 Day Cycle

Interventionmg/day (Number)
Lenalidomide + High-Dose Melphalan100

[back to top]

Overall Response Rate

Overall response rate is defined as the percentage of participants who achieved either a confirmed stringent complete response (sCR), complete response (CR), very good partial response (VGPR), or partial response (PR) as their best response based on the Independent Review Committee (IRC) assessed response outcome. Response was determined using the International Myeloma Working Group - Uniform Response Criteria (IMWG-URC). (NCT01080391)
Timeframe: From randomization through the data cutoff date of 16 June 2014. Median follow-up time was approximately 31 months.

Interventionpercentage of participants (Number)
Lenalidomide and Dexamethasone (Rd)66.7
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)87.1

[back to top]

Duration of Response

Duration of response (DOR) was calculated for participants who achieved a best response of stringent complete response (sCR), complete response (CR), very good partial response (VGPR), or partial response (PR). Duration of response was defined as the time in months from the initial start of response (PR or better) to the earlier of documented progressive disease (PD) or death due to any cause. Participants who had not progressed or died were censored according to the censoring rules defined previously for PFS. (NCT01080391)
Timeframe: From randomization through the data cutoff date of 16 June 2014. Longest follow-up time was approximately 42 months.

Interventionmonths (Median)
Lenalidomide and Dexamethasone (Rd)21.2
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)28.6

[back to top]

Duration of Disease Control

Duration of disease control (DDC) was calculated for participants who achieved disease control. DDC was defined as the time in months from randomization to the earlier of documented progressive disease (PD) or death due to any cause. Participants who had not progressed or died were censored according to the censoring rules defined previously for PFS. (NCT01080391)
Timeframe: From randomization through the data cutoff date of 16 June 2014. Longest follow-up time was approximately 46 months.

Interventionmonths (Median)
Lenalidomide and Dexamethasone (Rd)18.9
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)28.7

[back to top]

Disease Control Rate

Disease control rate was defined as the percentage of participants who achieved a best response of stringent complete response (sCR), complete response (CR), very good partial response (VGPR), partial response (PR), minimal response (MR), or stable disease (SD) lasting ≥ 8 weeks according to International Myeloma Working Group - Uniform Response Criteria (IMWG-URC) (MR was determined using European Group for Blood and Marrow Transplantation criteria). (NCT01080391)
Timeframe: From randomization through the data cutoff date of 16 June 2014. Median follow-up time was approximately 31 months.

Interventionpercentage of participants (Number)
Lenalidomide and Dexamethasone (Rd)87.1
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)92.7

[back to top]

Quality of Life Core Module (QLQ-C30) Global Health Status/Quality of Life Scores

Health-related quality of life was assessed with the use of the European Organization for Research and Treatment of Cancer Quality of Life Core Module (QLQ-C30) questionnaire, a validated instrument in multiple myeloma patients. Scores range from 0 to 100, with higher scores indicating better health related quality of life. (NCT01080391)
Timeframe: Cycle 1 Day 1 (Baseline), Day 1 of Cycles 3, 6, 12, 18

,
Interventionscores on a scale (Mean)
Cycle 1 Day 1 (Baseline)Cycle 3, Day 1Cycle 6, Day 1Cycle 12, Day 1Cycle 18, Day 1
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)58.359.962.562.764.3
Lenalidomide and Dexamethasone (Rd)58.156.858.957.359.9

[back to top]

Progression-free Survival (PFS)

Kaplan-Meier estimate of median time from randomization to progressive disease (PD) or all-cause death. PD was assessed using International Myeloma Working Group-Uniform Response Criteria (IMWG-URC). One or more conditions were required to meet PD: 2 consecutive rising serum or urine M-protein from central lab; documented new bone lesion(s) or soft tissue plasmacytoma(s) or increased size of existing bone lesion(s) or plasmacytoma(s); or confirmed hypercalcemia due solely to plasma cell proliferative disorder (local lab greater than 11.5 mg/dL on 2 separate occasions). Censoring conditions (censoring dates) were: no post-baseline disease assessment (DA) (randomization date); started non-protocol systemic anticancer treatment before PD or death (last DA date before such treatment); died or had PD after more than 1 missed DA (last DA date without PD before the first missed visit); or were alive and without documentation of PD, including lost to follow-up without PD (last DA date). (NCT01080391)
Timeframe: From randomization through the data cutoff date of 16 June 2014. Median follow-up time was approximately 31 months.

Interventionmonths (Median)
Lenalidomide and Dexamethasone (Rd)17.6
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)26.3

[back to top]

Overall Survival

Overall survival (OS) was defined as the duration from randomization to death due to any cause. Participants who were still alive were censored at the date when the participant was last known to be alive or the data cutoff date, whichever occurred earlier. (NCT01080391)
Timeframe: From randomization through the data cutoff date of 28 April 2017 for the final analysis of overall survival; median follow up time was 67.1 months in each treatment group.

Interventionmonths (Median)
Lenalidomide and Dexamethasone (Rd)40.4
Carfilzomib, Lenalidomide, and Dexamethasone (CRd)48.3

[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 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]

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]

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]

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]

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]

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]

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]

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 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]

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]

Proportion of Patients Achieving CR

(NCT01093183)
Timeframe: At 4 months

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide and Cyclophosphamide)0

[back to top]

Overall Survival

(NCT01093183)
Timeframe: Up to 4 years

Interventionmonths (Median)
Treatment (Lenalidomide and Cyclophosphamide)20

[back to top]

Number of Patients Achieving Objective PSA Response (50% Decrease in PSA Levels Sustained for at Least 4 Weeks) as Defined by PSA Working Group Criteria

(NCT01093183)
Timeframe: 4 weeks

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide and Cyclophosphamide)7

[back to top]

Maximum Tolerated Dose of Lenalidomide Administered in Combination With Oral Cyclophosphamide (Phase I)

Defined to be the dose cohort below which 2 of 3 or 3 of 6 patients experience dose-limiting toxicities in course 1 or the highest dose cohort of 25 mg. (NCT01093183)
Timeframe: 28 days

Interventionmg (Number)
Treatment (Lenalidomide and Cyclophosphamide)25

[back to top]

Percentage of Participants With Overall Survival (OS)

Overall survival is defined as survival of death from any cause. To account for loss to follow-up of a few participants, the Kaplan-Meier estimator was used to estimate overall survival at 38 months post-randomization. (NCT01109004)
Timeframe: 38 months post-randomization

Interventionpercentage of participants (Number)
Tandem Auto Transplant81.8
RVD Consolidation85.4
Lenalidomide Maintenance83.7

[back to top]

Percentage of Participants With Disease Progression

"Disease Progression is defined as progression of multiple myeloma, including one or more of the following:~A reappearance of serum monoclonal paraprotein, with a level of at least 0.5 g/dL~24-hour urine protein electrophoresis with at least 200 mg paraprotein/24 hours~Abnormal free light chain levels of >10 mg/dl, only in patients without measurable paraprotein in the serum and urine~At least 10% plasma cells in a bone marrow aspirate or on trephine biopsy~Definite increase in the size of existing bone lesions or soft tissue plasmacytomas~Development of new bone lesions or soft tissue plasmacytomas~Development of hypercalcemia (corrected serum Ca >11.5 mg/dL or >2.8 mmol/L) not attributable to any other cause~To account for loss to follow-up of a few participants, the cumulative incidence of TRM at 38 months post-randomization was estimated using the Aalen-Johansen estimator, treating death prior to disease progression as a competing risk." (NCT01109004)
Timeframe: 38 months post-randomization

Interventionpercentage of participants (Number)
Tandem Auto Transplant39.8
RVD Consolidation41.0
Lenalidomide Maintenance45.6

[back to top]

MOS SF-36 Physical Component Summary

The Medical Outcome Study (MOS) SF-36 Physical Component Summary is a subscale of the SF-36 intended to measure physical well-being. It is scored on a scale of 0-100, with higher scores indicating higher levels of well-being. (NCT01109004)
Timeframe: Up to 3 years post-randomization

,,
Interventionscore on a scale (Mean)
Baseline1 Year2 Years3 Years
Lenalidomide Maintenance38424343
RVD Consolidation39434442
Tandem Auto Transplant37434442

[back to top] [back to top]

Number of Participants With Treatment Response

"The number of participants with very good partial response (VGPR) or better [complete response (CR), near CR (nCR), and stringent CR (sCR)] according to the International Uniform Response Criteria will be calculated. The Worse than VGPR group includes PR, stable disease, and progressive disease.~sCR requires, in addition to CR: Normal free light chain ratio (FLC), Absence of clonal cells in bone marrow CR requires, in addition to nCR: Absence of the original monoclonal paraprotein (PPN), Disappearance of soft tissue plasmacytomas nCR is defined as: < 5% plasma cells in a bone marrow aspirate, No increase in lytic bone lesions VGPR requires: Serum or urine PPN not detectable on electrophoresis OR >=90% reduction in serum PPN plus urine PPN <100 mg/24hrs, >= 50% reduction in the level of serum monoclonal PPN or reduction in 24 hour urinary monoclonal PPN either >= 90% or to <200 mg/24 hours in light chain disease, >= 50% reduction in the size of soft tissue plasmacytomas" (NCT01109004)
Timeframe: 1 and 2 years post-randomization

InterventionParticipants (Count of Participants)
1 Year725726851 Year725726861 Year725726872 Years725726852 Years725726872 Years72572686
Worse than VGPRCR or sCRVGPR or nCR
Tandem Auto Transplant97
RVD Consolidation122
Lenalidomide Maintenance98
Tandem Auto Transplant56
RVD Consolidation50
Lenalidomide Maintenance60
Tandem Auto Transplant39
Lenalidomide Maintenance50
Tandem Auto Transplant98
RVD Consolidation117
Lenalidomide Maintenance93
RVD Consolidation37
Lenalidomide Maintenance41
Tandem Auto Transplant20
RVD Consolidation21
Lenalidomide Maintenance26

[back to top]

FACT-G Total Score

The Functional Assessment of Cancer Therapy-General (FACT-G) is a quality of life instrument that assesses the effects of cancer therapy on a patient's physical, social/family, emotional, and functional well-being. The assessment has 27 questions, each scored on a Likert scale from 0-4. The overall score is computed by adding scores of the questions and falls in the range 0-108, with higher scores indicating higher levels of overall well-being. (NCT01109004)
Timeframe: Up to 3 years post-randomization

,,
Interventionscore on a scale (Mean)
Baseline1 Year2 Years3 Years
Lenalidomide Maintenance77838585
RVD Consolidation79848484
Tandem Auto Transplant79848485

[back to top]

MOS SF-36 Mental Component Summary

The Medical Outcome Study (MOS) SF-36 Mental Component Summary is a subscale of the SF-36 intended to measure mental well-being. It is scored on a scale of 0-100, with higher scores indicating higher levels of well-being. (NCT01109004)
Timeframe: Up to 3 years post-randomization

,,
Interventionscore on a scale (Mean)
Baseline1 Year2 Years3 Years
Lenalidomide Maintenance48505051
RVD Consolidation48515050
Tandem Auto Transplant49505051

[back to top]

FACT-BMT Score

The Functional Assessment of Cancer Therapy-Bone Marrow Transplant scale (FACT-BMT) is a quality of life instrument that assesses the effects of bone marrow transplantation (BMT) on a patient's physical, social/family, emotional, and functional well-being while taking into consideration BMT-specific concerns. The assessment has 37 questions, each scored on a Likert scale from 0-4. The overall score is computed by adding scores of the questions and falls in the range 0-148, with higher scores indicating higher levels of overall well-being. (NCT01109004)
Timeframe: Up to 3 years post-randomization

,,
Interventionscore on a scale (Mean)
Baseline1 Year2 Years3 Years
Lenalidomide Maintenance105113115115
RVD Consolidation107115115114
Tandem Auto Transplant107113114115

[back to top]

Percentage of Participants With Progression-free Survival (PFS)

Progression-free survival is defined as survival without disease progression or initiation of non-protocol anti-myeloma therapy. To account for loss to follow-up of a few participants, the Kaplan-Meier estimator was used to estimate progression-free survival at 38 months post-randomization. (NCT01109004)
Timeframe: 38 months post-randomization

Interventionpercentage of participants (Number)
Tandem Auto Transplant58.5
RVD Consolidation57.8
Lenalidomide Maintenance53.9

[back to top]

FACT-BMT Trial Outcome Index

The Functional Assessment of Cancer Therapy (FACT) Trial Outcome Index is a quality of life instrument that assesses the impact of bone marrow transplantation (BMT) on a patient's physical and functional well-being while taking into consideration BMT-specific concerns. The assessment has 24 questions, each scored on a Likert scale from 0-4. The overall score is computed by adding scores of the questions and falls in the range 0-96, with higher scores indicating higher levels of overall well-being. (NCT01109004)
Timeframe: Up to 3 years post-randomization

,,
Interventionscore on a scale (Mean)
Baseline1 Year2 Years3 Years
Lenalidomide Maintenance63717373
RVD Consolidation65727371
Tandem Auto Transplant64707373

[back to top]

Overall Response

"Number of patients with a complete or partial response using Cheson criteria for lymphoma.~A complete response is defined as a complete disappearance of all detectable clinical and radiographic evidence of disease and disappearance of all disease-related symptoms if present before therapy. A partial response is defined as 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. Patients who have been on study drug for at least 2 months will be considered evaluable for response as long as they have had repeat imaging to assess response or clear progression based on physical exam." (NCT01121757)
Timeframe: Response will be assessed after at least 4 months on first study drug.

Interventionparticipants (Number)
1a-Azacitidine Followed by Lenalidomide1
1b-Lenalidomide Followed by Azacitidine0

[back to top]

Response Predicted by Molecular Signatures Compared to True Response

"The predicted response (response to therapy vs. no response to therapy) using gene sequencing will be compared to the overall true response (reported in Primary Outcome 2)." (NCT01121757)
Timeframe: approximately one year

,
Interventionparticipants (Number)
Predicted: Response; Actual: ResponsePredicted: No Response; Actual: No ResponsePredicted: Response; Actual: No Response
1a-Azacitidine Followed by Lenalidomide123
1b-Lenalidomide Followed by Azacitidine030

[back to top]

Number of Participants With Grade 3 and 4 Toxicities

Evaluate the safety of lenalidomide, azacitidine and the combination of azacitidine + lenalidomide in patients with lymphoma; grading the adverse events using Common Toxicity Criteria for Adverse Events (CTCAE) version 4.0 (NCT01121757)
Timeframe: While taking the study drug and 30 days after the last dose

,
Interventionparticipants (Number)
Grade 3Grade 4
1a-Azacitidine Followed by Lenalidomide51
1b-Lenalidomide Followed by Azacitidine31

[back to top]

Overall Response

"Number of patients with a complete or partial response using Cheson criteria for lymphoma.~A complete response is defined as a complete disappearance of all detectable clinical and radiographic evidence of disease and disappearance of all disease-related symptoms if present before therapy. A partial response is defined as 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. Patients who have been on study drug for at least 2 months will be considered evaluable for response as long as they have had repeat imaging to assess response or clear progression based on physical exam." (NCT01121757)
Timeframe: Response will be assessed after at least 4 months on second drug.

Interventionparticipants (Number)
1a-Azacitidine Followed by Lenalidomide2
1b-Lenalidomide Followed by Azacitidine0

[back to top]

Frequency of Adverse and Severe Adverse Events

Frequency of adverse and severe adverse events (NCT01123356)
Timeframe: 30 weeks

Interventionparticipants (Number)
Number of SAEsNeutropeniagrade 3/4 neutropeniathrombocytopeniagrade 3/4 thrombocytopeniatumor flare reactiongrade 3 tumor flare reaction
Oratumumab and Lenalidomide4191015491

[back to top]

Dose Reductions Due to Adverse Events.

Number of dose reductions due to toxicity. (NCT01123356)
Timeframe: 30 weeks

Interventiondose reductions (Number)
Oratumumab and Lenalidomide17

[back to top]

Frequency of Adverse Events

Number of adverse events occuring in greater than 20% of subjects (NCT01123356)
Timeframe: 30 weeks

Interventionevents (Number)
Oratumumab and Lenalidomide15

[back to top]

Overall Response Rate

"Obtain early assessment of the efficacy of the intracycle sequential administration of ofatumumab and lenalidomide in the treatment of chronic lymphocytic leukemia (CLL) after prior use of rituximab. Response was categorized according to the IW-CLL criteria which includes the following: Complete remission (CR), CR with incomplete marrow recovery (CRi)Partial remission (PR), Progressive disease (PD), Stable disease (SD).~Overall response rate was defined as those who experienced a response of CR, CRi or PR." (NCT01123356)
Timeframe: 30 Weeks

Interventionpercentage of participants (Number)
Oratumumab and Lenalidomide53

[back to top]

Time to Response

Measured from time of study drug administration to initial response (partial or complete), measured in months. (NCT01125176)
Timeframe: From date of study drug initiation to date of initial response, assessed up to 12 months.

Interventionmonths (Median)
All Subjects10

[back to top]

Overall Survival

Measured from time of study drug administration to death, measured in months. (NCT01125176)
Timeframe: From date of study drug initiation to date of death, assessed through study completion up to 105 months.

Interventionmonths (Median)
All Subjects97

[back to top]

Progression Free Survival

Measured from time of study drug administration to progression or death, measured in months. (NCT01125176)
Timeframe: From date of study drug initiation until date of progression or death, whichever occurs first, assessed through study completion up to 100 months.

Interventionmonths (Median)
All Subjects48

[back to top]

Overall Response Rate as Defined as the Number of Patients Who Experience a Response (Complete or Partial) to Treatment at the Time of Best Response

Response will be evaluated in this study using the International Workshop on CLL (IWCLL) update of the 1996 NCI-Working Group criteria for CLL, which includes assessment of the following: clonal lymphocytes in the peripheral blood by flow cytometry, blood tests for neutrophil count, hemoglobin, and platelet count, CT examination for lymphadenopathy, hepatomegaly, splenomegaly, constitutional symptoms, bone marrow assessment via biopsy/aspirate, and evaluation for disease transformation. (NCT01125176)
Timeframe: From date of study drug initiation until date of best response, assessed up to 6 years.

InterventionParticipants (Count of Participants)
All Subjects12

[back to top]

Duration of Response

Measured from end of treatment to progression or death, measured in months. (NCT01125176)
Timeframe: From date of end of treatment to progression or death, whichever occurs first, assessed through study completion up to 100 months.

Interventionmonths (Median)
All Subjects70

[back to top]

Number of Participants With Erythroid Response

The rate of erythroid response to treatment with the lenalidomide/prednisone combination in non-del (5q) low and int-1 risk Myelodysplastic Syndrome (MDS) with symptomatic anemia. Hematological improvement erythroid response (HI-E) according to International Working Group (IWG) 2006 criteria. (NCT01133275)
Timeframe: Up to 7 months

Interventionparticipants (Number)
Lenalidomide and Prednisone Therapy5

[back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top]

Correlate the Presence of Specific Fc RIIIa Polymorphisms With Progression-free Survival in Subjects Receiving Cetuximab and Lenalidomide for SCCHN.

Progression-free survival (PFS) was defined as time from date of the first treatment dose administered to the earlier of disease progression or death from any cause. 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. (NCT01133665)
Timeframe: 24 months

Interventionmonths (Median)
Cetuximab and Lenalidomide1.8

[back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top] [back to top]

Phase I: Number of Patients With Dose Limiting Toxicity

The number of patients who had a DLT during the dose finding portion (Phase I) of the trial for the safety of lenalidomide when used in combination with high dose melphalan in the setting of autologous stem cell transplantation in patients with multiple myeloma. (NCT01142232)
Timeframe: up to 1 month

InterventionParticipants (Count of Participants)
Phase I, Dose Level 10
Phase I, Dose Level 20
Phase I, Dose Level 30
Phase I, Dose Level 40

[back to top]

Phase II: Overall Response Rate

Evaluate the overall response rate of patients receiving therapy. Patients are considered as having a response if their overall response is Partial Response or better (CR+sCR+VGPR+PR). The percentage of patients achieving this and the exact 95% confidence interval will be calculated. Responses will be defined using the response criteria determined by the International Working Group for Multiple Myeloma (CR= Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and 5% plasma cells in bone marrow; sCR=CR as defined above plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunoflurorescence; VGPR=Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-component plus urine M-component<100 mg per 24 h; PR=>=50% reduction of serum M-protein and reduction in 24-h urinary M-protein by >=90% or to <200mg per 24 h). (NCT01142232)
Timeframe: up to 5 years

Interventionpercentage of participants (Number)
Phase II87.5

[back to top] [back to top]

Number of Participants Who Achieved a Complete Response

Response is assessed by investigator according to International Working Group (IWG) criteria. Complete response requires disappearance of all evidence of disease. (NCT01145495)
Timeframe: At 12 months

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide, Rituximab)47

[back to top]

Toxicity of Study Treatment, Assessed by the NCI Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0

Data will be summarized using frequency tables. (NCT01145495)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Grade 1-2 FatigueGrade 1-2 DiarrheaGrade 1-2 RashGrade 1-2 Febrile neutropeniaGrade 3-4 NeutropeniaGrade 3-4 LymphopeniaGrade 3-4 ThrombocytopeniaGrade 3-4 InfectionGrade 3-4 Rash
Treatment (Lenalidomide, Rituximab)5124211146175

[back to top]

Disease Progression

Kaplan-Meier method will be used. 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. (NCT01145495)
Timeframe: Up to 5 years

Interventionproportion of participants (Number)
2 Years PFS3 Years PFS4 Years PFS5 Years PFS
Treatment (Lenalidomide, Rituximab)0.860.810.740.72

[back to top]

Percent of Participants With Clinical Benefit and Response According to International Response Criteria

Percent of participants with response according to international response criteria (>= PR) and percent of participants with clinical benefit response (>= minor response according to adapted EBMT criteria). Determined with serum and 24 hour urine protein electrophoresis, and as appropriate, supplemented by immunofixation, serum free light chain assay, and bone marrow examination. Response before high dose melphalan and autologous stem cell transplant will also be confirmed by two separate blood and 24 hour urine tests between the last dose of combination therapy and the first dose of the mobilizing agent. (NCT01155583)
Timeframe: at 6 months

,
Interventionpercentage of participants (Number)
international response criteria (>= PR)clinical benefit response (>= minor response)
Overall Study - (Azacitidine/Lenalidomide/Dexamethasone)2032
Participants Who Received HTLD/HTLD-CKD2332

[back to top]

Percent of Participants With Clinical Benefit and Response According to International Response Criteria

Percent of participants with response according to international response criteria (>= PR) and percent of participants with clinical benefit response (>= minor response according to adapted EBMT criteria). Determined with serum and 24 hour urine protein electrophoresis, and as appropriate, supplemented by immunofixation, serum free light chain assay, and bone marrow examination. Response before high dose melphalan and autologous stem cell transplant will also be confirmed by two separate blood and 24 hour urine tests between the last dose of combination therapy and the first dose of the mobilizing agent. (NCT01155583)
Timeframe: at 12 months

,
Interventionpercentage of participants (Number)
International response criteria (>= PR)clinical benefit response (>= minor response)
Overall Study - (Azacitidine/Lenalidomide/Dexamethasone)2232
Participants Who Received HTLD/HTLD-CKD2332

[back to top]

Phase I: Highest Tolerated Low Dose (HTLD)

Azacitidine given at low but increasing doses up to 50mg/m2 twice a week. Maximum tolerated dose reported. (NCT01155583)
Timeframe: During the first 28-day cycle

Interventionmg/m^2 (Number)
Arm A - Azacitidine/Lenalidomide/Dexamethasone50
Arm B - CKD Azacitidine/Lenalidomide/Dexamethasone50

[back to top]

Overall Survival

Overall survival will be measured from study entry to death from any cause - median months survival will be reported (NCT01155583)
Timeframe: up to 3 years

Interventionmonths (Median)
Overall Study - (Azacitidine/Lenalidomide/Dexamethasone)18.6

[back to top]

Median Progression-free Survival (PFS)

Median PFS, measured in months from study entry to progression as defined by international response criteria or death of any cause, whichever comes first (NCT01155583)
Timeframe: Up to 3 years

Interventionmonths (Median)
Overall Study - (Azacitidine/Lenalidomide/Dexamethasone)3.1

[back to top]

Follow-up Time

time that patients were monitored for disease progression and overall survival (NCT01160484)
Timeframe: Follow-up visits for disease progression and overall survival every 3 months after study discontinuation. After progression, follow-up visits for survival status every 6 months or until alternate therapy needs to be started or death intervenes

Interventionmonths (Median)
DVD-R Single Arm11

[back to top]

Progression-free Survival

(NCT01160484)
Timeframe: Time from the start of treatment to progressive disease or until death

Interventionmonths (Median)
DVD-R Single Arm9

[back to top]

Duration of Response

(NCT01160484)
Timeframe: First evidence of PR or better (for overall response) and MR or better (for clinical benefit response) to start of disease progression or death.

Interventionmonths (Median)
DVD-R Single Arm12

[back to top]

Time to Best Response

(NCT01160484)
Timeframe: Up to 7.5 months (eight 28-day cycles)

Interventionmonths (Median)
DVD-R Single Arm2

[back to top]

Time to First Response

(NCT01160484)
Timeframe: Up to 7.5 months (eight 28-day cycles)

Interventionmonths (Median)
DVD-R Single Arm1

[back to top]

Time to Progression

(NCT01160484)
Timeframe: Time from the start of treatment to progressive disease

Interventionmonths (Median)
DVD-R Single Arm9

[back to top]

International Myeloma Working Group (IMWG) Response Criteria

The investigator will evaluate each patient for response to therapy according to criteria augmented from those developed by Bladé et al., 1998 presented below (Table 7-1). Assessment of disease response will be performed prior to drug administration on Day 1 of Cycles 2 8 and at the End of Study Treatment visit. If a patient is determined to have complete response (CR), very good partial response (VGPR), partial response (PR), or minor response (MR), then assessment of disease response is to be performed 4 weeks later to confirm the response. (NCT01160484)
Timeframe: Up to 7.5 months (eight 28-day cycles)

Interventionparticipants (Number)
CRVGPRPRObjective Response (CR+VGPR+PR)MRClinical Benefit (CR+VGPR+PR+MR)Estable diseaseProgressive disease
DVD-R Single Arm84719143342

[back to top]

Proportion of Participants With Response (Phase III Secondary Endpoint)

"Response is defined as complete response (CR), very good partial response (VGPR) or partial response (PR).~CR: Negative immunofixation on the serum and urine, disappearance of any soft tissue plasmacytomas and <=5% plasma cells in bone marrow VGPR: Serum and urine M-component detectable by immunofixation but not on electrophoresis or ≥ 90% reduction in serum M-component plus urine M-component < 100 mg per 24 hours~PR:~≥ 50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥ 90% or to < 200 mg per 24 hours~If followed by free light chain (FLC) only, a ≥ 50% decrease in the difference between involved and uninvolved FLC levels~If unmeasurable disease by serum M-protein, urine M-protein, and serum FLC at baseline, a ≥50% reduction in plasma cells provided baseline bone marrow percentage was ≥ 30%~If present at baseline, a ≥ 50% reduction in the size of soft tissue plasmacytomas" (NCT01169337)
Timeframe: Assessed every 3 months if patient is < 2 years from study entry, every 6 months if patient is 2-5 years from study entry, then annually for years 6-10

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase III)0.50
Arm B (Observation; Phase III)0

[back to top]

Proportion of Patients With Grade 3 Adverse Events That Effect Vital Organ Function or Any Grade 4 or Higher Non-hematologic Adverse Events (Phase II Primary Endpoint)

Proportion of patients with grade 3 adverse events that effect vital organ function (such as cardiac, hepatic or thromboembolic) or any grade 4 or higher non-hematologic adverse events (NCT01169337)
Timeframe: Assessed every 4 weeks while on treatment up to 24 weeks

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase II)0.056

[back to top]

1-year Progression-free Survival (PFS) Rate (Phase III Secondary Endpoint)

"PFS is defined as time from randomization to progression or death, whichever occurs first. Patients are considered to have progression if both of the following criteria are met. Kaplan-Meier method was used to estimate 1-year PFS rate.~Any of the following:~Increase in serum M-protein to ≥ 25% above the lowest response level with an absolute increase of at least 0.5g/dl to qualify as progression~Increase in urine M-protein to ≥ 25% above the lowest response level for 24-hour excretion with an absolute increase of at least 200mg/24 hours of urine M-protein to qualify as progression~Increase in bone marrow plasma cell percentage to ≥ 25% from lowest response value (the absolute % increase must be ≥ 10%)~Any of the following felt related to the underlying clonal plasma cell proliferative disorder:~Hypercalcemia (> 11 mg/dL)~Decrease in hemoglobin of ≥ 2 gms/dL~Serum creatinine level ≥ 2mg/dL~Development of myeloma bone lesions or soft tissue plasmacytoma" (NCT01169337)
Timeframe: Assessed every 3 months for one year

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase III)0.98
Arm B (Observation; Phase III)0.89

[back to top]

2-year Overall Survival (OS) Rate (Phase III Secondary Endpoint)

Overall survival is defined as the time from randomization to death or date last known alive among all randomized patients in the phase III part of the study. Kaplan-Meier method was used to estimate the 2-year OS rate. (NCT01169337)
Timeframe: Assessed every 3 months for 2 years

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase III)0.98
Arm B (Observation; Phase III)1.00

[back to top]

2-year Progression-free Rate (Phase III Secondary Endpoint)

"TTP is defined as the time from randomization to progression. Patients are considered to have progression if both of the following criteria are met. Kaplan-Meier method was used to estimate 2-year progression-free rate.~Any of the following:~Increase in serum M-protein to ≥ 25% above the lowest response level with an absolute increase of at least 0.5g/dl to qualify as progression~Increase in urine M-protein to ≥ 25% above the lowest response level for 24-hour excretion with an absolute increase of at least 200mg/24 hours of urine M-protein to qualify as progression~Increase in bone marrow plasma cell percentage to ≥ 25% from lowest response value (the absolute % increase must be ≥ 10%)~Any one or more of the following felt related to the underlying clonal plasma cell proliferative disorder:~Hypercalcemia (> 11 mg/dL)~Decrease in hemoglobin of ≥ 2 gms/dL~Serum creatinine level ≥ 2mg/dL~Development of myeloma bone lesions or soft tissue plasmacytoma" (NCT01169337)
Timeframe: Assessed every 3 months for 2 years

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase III)94.3
Arm B (Observation; Phase III)75.8

[back to top]

2-year Progression-free Survival (PFS) Rate (Phase III Primary Endpoint)

"PFS is defined as time from randomization to progression or death, whichever occurs first. Patients are considered to have progression if both of the following criteria are met. Kaplan-Meier method was used to estimate 2-year PFS rate.~Any of the following:~Increase in serum M-protein to ≥ 25% above the lowest response level with an absolute increase of at least 0.5g/dl to qualify as progression~Increase in urine M-protein to ≥ 25% above the lowest response level for 24-hour excretion with an absolute increase of at least 200mg/24 hours of urine M-protein to qualify as progression~Increase in bone marrow plasma cell percentage to ≥ 25% from lowest response value (the absolute % increase must be ≥ 10%)~Any of the following felt related to the underlying clonal plasma cell proliferative disorder:~Hypercalcemia (> 11 mg/dL)~Decrease in hemoglobin of ≥ 2 gms/dL~Serum creatinine level ≥ 2mg/dL~Development of myeloma bone lesions or soft tissue plasmacytoma" (NCT01169337)
Timeframe: Assessed every 3 months for 2 years

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase III)0.93
Arm B (Observation; Phase III)0.76

[back to top]

Proportion of Participants With Response (Phase II Secondary Endpoint)

"Response is defined as complete response (CR), very good partial response (VGPR) or partial response (PR).~CR: Negative immunofixation on the serum and urine, disappearance of any soft tissue plasmacytomas and <=5% plasma cells in bone marrow VGPR: Serum and urine M-component detectable by immunofixation but not on electrophoresis or ≥ 90% reduction in serum M-component plus urine M-component < 100 mg per 24 hours~PR:~≥ 50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥ 90% or to < 200 mg per 24 hours~If followed by free light chain (FLC) only, a ≥ 50% decrease in the difference between involved and uninvolved FLC levels~If unmeasurable disease by serum M-protein, urine M-protein, and serum FLC at baseline, a ≥50% reduction in plasma cells provided baseline bone marrow percentage was ≥ 30%~If present at baseline, a ≥ 50% reduction in the size of soft tissue plasmacytomas" (NCT01169337)
Timeframe: Assessed every 3 months if patient is < 2 years from study entry, every 6 months if patient is 2-5 years from study entry, then annually for years 6-10

Interventionproportion of participants (Number)
Arm A (Lenalidomide; Phase II)0.477

[back to top]

Stage 1: Percentage of Participants With an Overall Response According to the IWG Response Criteria Based on the Investigators Assessment at the Final Data Cut During the Core Treatment Phase

Response was defined as having a CR, CRu or PR, based on IWG 1999 Response Criteria for NHL as evaluated by the investigators. CR = complete disappearance of disease and disease related symptoms. All lymph nodes and nodal masses regressed on computed tomography to normal size (≤ 1.5 cm in their greatest transverse diameter for nodes > 1.5 cm prior to therapy and ≤ 1.0 cm in their short axis for nodes 1.1-1.5 cm in their long axis and > 1.0 cm in their short axis prior to therapy). Spleen and/or liver not palpable on physical exam, normal size by imaging, and absence of nodules related to lymphoma. If BM was involved prior to therapy, infiltrate must have cleared on repeat biopsy. PR = ≥ 50% decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses. No increase in the other nodes, liver, or spleen. Splenic and hepatic nodules regressed by ≥ 50% in their SPD or, for single nodules, in the greatest transverse diameter. No new disease. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

InterventionPercentage of participants (Number)
Lenalidomide29.4
Investigators Choice (IC)13.7

[back to top]

Stage 1: Percentage of Participants With a Complete Response According to the IWG Response Criteria as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase

A complete response was defined as participants with a complete response (CR), or unconfirmed complete response (CRu) based on IWG 1999 Response Criteria for NHL as assessed by the investigator. A CR is a complete disappearance of all disease with the exception of nodes. No new lesions. Previously enlarged organs must have regressed and not be palpable. Bone marrow must be negative if positive at baseline. Normalization of markers. CR Unconfirmed (CRu) does not qualify for CR above, due to a residual nodal mass or an indeterminate BM. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

Interventionpercentage of participants (Number)
Lenalidomide13.7
Investigators Choice (IC)3.9

[back to top]

Stage 1: Kaplan Meier Estimates of Overall Survival As Assessed by the Investigators at the Final Data Cut During The Core Treatment Phase

Overall survival was defined as time from randomization until death of any cause. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

InterventionWeeks (Median)
Lenalidomide31.0
Investigators Choice (IC)24.6

[back to top]

Number of Participants With Treatment Emergent Events (TEAEs) in the Overall Treatment Phase by Initial Treatment Assignment

"A TEAE was defined as an AE that begins or worsens in intensity of frequency on or after the first dose of study drug through 28 days after last dose of study drug.~A serious adverse event (SAE) is any:~Death;~Life-threatening event;~Any inpatient hospitalization or prolongation of existing hospitalization;~Persistent or significant disability or incapacity;~Congenital anomaly or birth defect;~Any other important medical event The investigator determined the relationship of an AE to study drug based on the timing of the AE relative to drug administration and whether or not other drugs, therapeutic interventions, or underlying conditions could provide a sufficient explanation for the event.The severity of an AE was evaluated by the investigator according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) (Version 4.03) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death" (NCT01197560)
Timeframe: From first dose of study drug to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

,
InterventionParticipants (Count of Participants)
Any TEAEsAny Treatment Related TEAEAny TEAE Grade ≥ 3Any TEAE Grade ≥ 4Any TEAE Grade 5Any TEAE Grade 3 or 4Any Treatment Related TEAE Grade ≥ 3Any Treatment Related TEAEs Grade ≥ 4Any Treatment Related TEAE Grade 5Any Treatment Related TEAE Grade 3 or 4Any Serious Adverse Events (SAEs)Any Treated Related SAEsAny AE leading to stopping of study drugAny drug related AE leading to halt of study drugAny AE leading to dose interruption/reductAny drug related AE leading to interruption/reduct
Investigators Choice (IC)554553361852392123942211743430
Lenalidomide54494329942301503031141153227

[back to top]

Stage 1: Percentage of Participants With an Overall Response According to the International Working Group (IWG) Response Criteria for Non Hodgkin's Lymphoma (NHL), Cheson 1999 and Evaluated by the Independent Response Adjudication Committee (IRAC)

An overall response is a complete response (CR), unconfirmed complete response (CRu) or partial response (PR) and was evaluated by the IRAC. A CR = complete disappearance of disease and related symptoms. Lymph nodes and nodal masses regressed on computed tomography to normal size (≤ 1.5 cm in their greatest transverse diameter for nodes > 1.5 cm prior to therapy and ≤ 1.0 cm in their short axis for nodes 1.1-1.5 cm in their long axis and > 1.0 cm in their short axis prior to therapy). Spleen and/or liver not palpable on exam, normal size by imaging, and absence of nodules related to lymphoma. If bone marrow was involved prior to therapy, infiltrate must have cleared on repeat biopsy. PR = ≥ 50% decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses. No increase in other nodes, liver, or spleen. Splenic and hepatic nodules regressed by ≥ 50% in their SPD or for single nodules, in the greatest transverse diameter;no new disease. (NCT01197560)
Timeframe: From the date of randomization to the data cut-off of 4 July 2013; when all patients reached the scheduled 16-week assessment or had progressed/died before the scheduled 16-week assessment); the median study duration was 27.0 and 19.7 weeks, respectively.

,
Interventionpercentage of participants (Number)
ORR for All ParticipantsGCB SubtypeNon-GCB
Investigators Choice (IC)11.812.011.5
Lenalidomide27.526.128.6

[back to top]

Stage 1: Percentage of Participants With a Durable Overall Response (dORR) According to the IWG Response Criteria as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase

Durable overall response rate was defined as the percentage of participants who maintained a response for at least 16 weeks after initial response. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

Interventionpercentage of participants (Median)
Lenalidomide23.5
Investigators Choice (Control Arm)9.8

[back to top]

Stage 1: Kaplan Meier Estimates of Progression-Free Survival As Assessed By The Investigators At The Final Data Cut During The Core Treatment Phase

Progression-free survival was defined as the time from randomization to the first documented disease progression or death due to any cause. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

InterventionWeeks (Median)
Lenalidomide9.6
Investigators Choice (IC)7.1

[back to top]

Stage 1: Kaplan Meier Estimates of Duration of Overall Response (DoR) as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase

Duration of overall response was calculated as the time of initial response (CR+CRu+PR) until documented disease progression determinted by computerized scan CT scan or MRI or death due to lymphoma, whichever occurred earlier, for participants who responded. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

InterventionWeeks (Median)
Lenalidomide64.7
Investigators Choice (Control Arm)63.1

[back to top]

Stage 1: Kaplan Meier Estimates of Duration of Complete Response (DoCR) as Assessed by the Investigators at the Final Data Cut During the Core Treatment Phase

Duration of complete response was defined as the time from the first documented complete response (CR + CRu) until the first disease progression or death for participants who had a CR. (NCT01197560)
Timeframe: From the date of randomization to the final data cut-off date of 18 May 2018; median study duration was 27.0 and 19.7 weeks, respectively.

InterventionWeeks (Median)
Lenalidomide66.4
Investigators Choice (Control Arm)179.3

[back to top]

Overall Response Rate (ORR)

(NCT01199575)
Timeframe: nine months

InterventionParticipants (Count of Participants)
Revlimid + Rituximab25

[back to top]

Adverse Events to Study Treatment

(NCT01199575)
Timeframe: one year

InterventionParticipants (Count of Participants)
Revlimid + Rituximab25

[back to top]

Overall Survival (Final Results After Crossover)

Time from randomization to death from any cause. Patients who have not died are censored as of the date last known alive. (NCT01208051)
Timeframe: 24 months

Interventionpercentage of participants (Number)
Dose Level 0NA
Dose Level +160.0
Dose Level +2100
Phase II Arm A (Cediranib Maleate)64.8
Phase II Arm B (Cediranib Maleate Plus Lenalidomide)75.3

[back to top]

Percent Change in Tumor Size (Phase II)

The percent change in tumor size from baseline to the end of cycle 2 (two months). The post-treatment total sum of lengths for a patient with a new lesion at cycle 2 will be scored as 1.2*max(pre-sum, post-sum) to ensure that the appearance of new lesions corresponds to a disease progression per Response Evaluation Criteria in Solid Tumors criteria. In the event of any early deaths prior to cycle 2, a nonparametric rank sum test will be used with deaths ranked at the extreme end of the distribution. (NCT01208051)
Timeframe: From baseline to 2 months

Interventionpercent change (Median)
Phase II Arm A (Cediranib Maleate)-12.5
Phase II Arm B (Cediranib Maleate Plus Lenalidomide)-4.2

[back to top]

Progression-free Survival (Final Results After Crossover)

Time from study enrollment until disease progression or death from any cause. Surviving patients without progression are censored as of the date of the last negative examination. 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. (NCT01208051)
Timeframe: Assessed up to 3 years

Interventionmonths (Median)
Dose Level 0NA
Dose Level +114.8
Dose Level +223.6
Phase II Arm A (Cediranib Maleate)14.8
Phase II Arm B (Cediranib Maleate Plus Lenalidomide)11.3

[back to top]

Progression-free Survival (Phase II Futility Analysis)

Time from enrollment on study to disease progression or death from any cause. Surviving patients without progression are censored as of the date of the last negative examination. 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. This analysis corresponds to the planned futility analysis after 40 events. (NCT01208051)
Timeframe: Assessed up to 3 years

Interventionmonths (Median)
Phase II Arm A (Cediranib Maleate)20.9
Phase II Arm B (Cediranib Maleate Plus Lenalidomide Thru April 10, 2015)10.6

[back to top]

Dose Limiting Toxicity

"Dose limiting toxicity was defined as any of the following occurring during the first cycle (28 days) of therapy:~Hematological toxicities:~Any grade 4 neutropenia (ANC < 500) lasting more than 5 days~Any grade 4 neutropenia with concomitant fever (temperature > 38.5)~Any grade 4 neutropenia and sepsis or other severe infection~Any grade 4 thrombocytopenia~Any other grade 3-4 non-hematological adverse drug reactions, except untreated nausea/vomiting, or hypersensitivity reactions.~Grade 4 hypertension~Grade 4 proteinuria Delay in the administration of a subsequent dose of cediranib and lenalidomide exceeding 2 weeks, due to an adverse drug reaction" (NCT01208051)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
Phase I Dose Level 00
Phase I Dose Level +11
Phase I Dose Level +22

[back to top]

Objective Response Rate

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and 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 lesions; Overall Response (OR) = CR + PR. (NCT01208051)
Timeframe: Assessed up to 3 years

InterventionParticipants (Count of Participants)
Dose Level 00
Dose Level +12
Dose Level +24
Phase II Arm A (Cediranib Maleate)17
Phase II Arm B (Cediranib Maleate Plus Lenalidomide)30

[back to top]

5-Year Overall Survival (OS)

OS was estimated using the KM method and defined as time from randomization to death due to any cause. Patients alive were censored at date last known alive. The 5-year OS endpoint is a probability. (NCT01208662)
Timeframe: In long-term follow-up, survival follow-up every 2 months until death. Median (maximum) OS follow-up was 76 and 129 months.The probability estimate is at 5 years.

Interventionpercent probability (Number)
RVD Alone79.2
RVD Plus ASCT80.7

[back to top]

Partial Response (PR) Rate

The PR rate is the percentage of participants achieving PR or better on treatment and was evaluated based on the IMWG criteria. PR was defined as > 50% reduction of serum M-protein and reduction in 24-h urinary M-protein by > 90% or to < 200mg per 24 hours. If the serum and urine M-protein are unmeasurable, a > 50% decrease in the difference between involved and uninvolved free light chain levels is required in place of the M-protein criteria. If serum and urine M-protein are unmeasurable, and serum free light assay is also unmeasurable, >50% reduction in plasma cells is required in place of M-protein, provided baseline bone marrow plasma cell percentage was > 30%. In addition to the above listed criteria, if present at baseline, a > 50% reduction in the size of soft tissue plasmacytomas was also required. Exact (Clopper-Pearson) confidence limits of 98.2857% represents Bonferroni adjustment for 7 tests (1-0.05/7) per statistical analysis plan for key secondary outcomes. (NCT01208662)
Timeframe: Disease was assessed every cycle up to maintenance and every 3 cycles on maintenance. Median treatment duration (months) from randomization 28.2 (RVD Alone) and 36.1 (RVD plus ASCT). Maximum treatment duration was 133.5 months in this study cohort.

Interventionpercentage of participants (Number)
RVD Alone95
RVD Plus ASCT97.5

[back to top]

5-Year Time to Progression (TTP)

TTP was estimated using the KM method and defined as time from randomization to time of documented IMWG disease progression or censoring time (time of last disease evaluation for those alive, time to death among those who died). Patients initiating non-protocol therapy prior to progression or death were censored at the date of non-protocol therapy in the TTP analysis. The 5-year TTP endpoint is a probability. (NCT01208662)
Timeframe: On treatment, disease was assessed every cycle up to maintenance and every 3 cycles on maintenance. In long-term follow-up, disease was assessed every 2 months until PD or death. Median TTP follow-up was 70 months. The probability estimate is at 5 years.

Interventionpercent probability (Number)
RVD Alone41.6
RVD Plus ASCT58.4

[back to top]

Change From Baseline on the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACT/GOG-NTX)

The FACT/GOG-NTX assessment consists of 11 questions that are all used to construct 1 subscale to summarize symptoms of peripheral neuropathy, including sensory, motor, and auditory problems and cold sensitivity. (https://www.facit.org/measures/FACT-GOG-NTX) Each question has 4 possible responses from 0 (Not at all) to 4 (Very Much) which are reverse-scored and summed. This sum is then scaled by the number of questions answered by multiplying by 11 and then dividing by the number of questions that are non-blank, in order to keep all final scores proportional to one another even if some questions are left blank. The aggregate score ranges from 0 to 44, with higher scores indicating less neurotoxicity and lower scores indicating more neurotoxicity. Change from baseline is the difference (NCT01208662)
Timeframe: Cycle 1 (Baseline), Cycle 2, Pre-Mobilization, Cycle 5 Arm A / Post Auto-HSCT Arm B, Cycle 8 Arm A /Cycle 5 Arm B, Maintenance Day 1, 2 years from baseline, 3 years from baseline

,
Interventionunits on a scale (Mean)
Cycle 2Pre-MobilizationCycle 5 Arm A / Post Auto-HSCT Arm BCycle 8 Arm A /Cycle 5 Arm BMaintenance Day 12 years from baseline3 years from baseline
RVD Alone-1.8-3.9-4.4-4.7-3.7-4.9-4.6
RVD Plus ASCT-1.3-3.5-4.4-2.5-2.3-2.6-3.2

[back to top]

Median Maintenance Treatment Duration

Maintenance treatment duration estimated using the KM method is defined as the time from start of maintenance to the time off maintenance (event) or censored at date of last maintenance treatment. (NCT01208662)
Timeframe: Up to 128 months

Interventionmonths (Median)
RVD Alone36.4
RVD Plus ASCT41.5

[back to top]

Median Event Free Survival (EFS)

EFS was estimated using the KM method and defined as the time from randomization to the earliest of IMWG disease progression, death, or initiation of non-protocol therapy (events); patients were censored date of last disease evaluation. (NCT01208662)
Timeframe: Assessed up to approximately 130 months.

Interventionmonths (Median)
RVD Alone32.0
RVD Plus ASCT47.3

[back to top]

Quality-Adjusted Life Years (QALYs)

QALYs were estimated with a model beginning at initiation of first-line therapy by arm. A lifetime horizon, as well as subsequent lines of therapy, was examined using open-source Amua 0.3.0 software. Base case analysis was performed using 10,000 first-order Monte Carlo simulations. Conditional probabilities were extracted from Kaplan-Meier curves from pivotal clinical trials using WebPlotDigitizer. Costs were estimated from RED BOOK and DFCI charge reporting in US Dollars ($) after inflation adjustment to 2022 and 3% discounting, and QALYs effects were measured using EQ-5D data from Hatswell et al. (NCT01208662)
Timeframe: Maximum observation of survival for this study cohort was 129.4 months.

Interventionyears gained (Mean)
RVD Alone5.67
RVD Plus ASCT6.10

[back to top]

5-year Cumulative Incidence of Second Primary Malignancy (SPM)

Second primary malignancy (SPM) is defined as the development of another new, unrelated cancer, regardless of treatment for previous malignancy attribution. The cumulative incidence of SPMs was estimated with death as a competing risk. SPMs were collected using an SAE form or MEDWATCH 3500A form, with intensity determined by using the NCI CTCAE version 4 as a guideline. (NCT01208662)
Timeframe: 5 years

Interventionpercentage of patients (Number)
RVD Alone4.9
RVD Plus ASCT6.5

[back to top]

Median Progression-Free Survival (PFS)

PFS was estimated using the Kaplan-Meier (KM) method and defined as time from randomization to the earlier of disease progression (PD) as determined by central review or death from any cause (events). Patients who started non-protocol therapy (NPT) were censored at the date of NPT initiation if available or date treatment ended if date of NPT was missing. Deaths occurring beyond 1 year from the date last known progression-free are not counted as events and censored at date of last disease evaluation. Patients who had not started NPT, progressed, or died were censored at the date of last disease evaluation. PD was based upon the International Myeloma Working Group (IMWG) uniform response criteria. [Kumar S, et al Lancet Oncol 2016;17(8):e328-e346]. (NCT01208662)
Timeframe: On treatment, disease was assessed every cycle up to maintenance and every 3 cycles on maintenance. In long-term follow-up, disease was assessed every 2 months until PD or death. Median (maximum) PFS follow-up was 70 and 129 months.

Interventionmonths (Median)
RVD Alone46.2
RVD Plus ASCT67.5

[back to top]

Subsequent Therapy Rate

Subsequent therapy rate was the percentage of participants who discontinued treatment and initiated subsequent non-protocol therapy. (NCT01208662)
Timeframe: Up to 129 months

InterventionParticipants (Count of Participants)
RVD Alone222
RVD Plus ASCT192

[back to top]

Very Good Partial Response (VGPR) Rate

The VGPR rate is the percentage of participants achieving VGPR or better on treatment and was evaluated based on IMWG criteria. VGPR was defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100mg per 24 hours. (NCT01208662)
Timeframe: Disease was assessed every cycle up to maintenance and every 3 cycles on maintenance. Median treatment duration (months) from randomization 28.2 (RVD Alone) and 36.1 (RVD plus ASCT). Maximum treatment duration was 134 months in this study cohort.

Interventionpercentage of patients (Number)
RVD Alone79.6
RVD Plus ASCT82.7

[back to top]

Change From Baseline on the European Organization for Research and Treatment of Cancer Quality-of-Life Questionnaire Core 30 (EORTC QLQ-C30): Global Health Status/Quality of Life (QoL) Sub-scale

The EORTC QLQ-C30 assessment consists of 30 questions that are used to construct 15 distinct sub-scales (five function scales, nine symptom scales, and a global health status/QoL scale). The global health status/QoL scale is comprised of two questions each with a range of 6 (1=Very Poor to 7=Excellent). Scores on all sub-scales range from 0 to 100 after linear transformation of the raw scores, with higher scores representing better global health status and quality of life. (NCT01208662)
Timeframe: Cycle 1 (Baseline), Cycle 2, Pre-Mobilization, Cycle 5 Arm A / Post Auto-HSCT Arm B, Cycle 8 Arm A /Cycle 5 Arm B, Maintenance Day 1, 2 years from baseline, 3 years from baseline

,
Interventionunits on a scale (Mean)
Cycle 2Pre-MobilizationCycle 5 Arm A / Post Auto-HSCT Arm BCycle 8 Arm A /Cycle 5 Arm BMaintenance Day 12 years from baseline3 years from baseline
RVD Alone0.10.43.01.25.35.13.5
RVD Plus ASCT4.34.4-11.18.39.911.112.7

[back to top]

Median Treatment Duration

Treatment duration estimated using the KM method is defined as the time from registration (C1) to the time off treatment (event) or censored at date of last treatment. (NCT01208662)
Timeframe: Up to 134 months

Interventionmonths (Median)
RVD Alone28.2
RVD Plus ASCT36.8

[back to top] [back to top]

Median Duration of Response: Partial Response (DOR PR)

DOR PR was estimated using the KM method and defined as the time from documented best response as CR to documented disease progression per IMWG criteria. Patients who have not progressed or died were censored at the date last known progression-free. (NCT01208662)
Timeframe: On treatment, disease was assessed every cycle up to maintenance and every 3 cycles on maintenance. In long-term follow-up, disease was assessed every 2 months until PD or death. Median (maximum) DOR PR follow-up was 62.9 and 122.9 months.

Interventionmonths (Median)
RVD Alone38.9
RVD Plus ASCT56.4

[back to top] [back to top]

Complete Response (CR) Rate

The CR rate is the percentage of participants achieving CR or better on treatment and was evaluated based on IMWG criteria. CR was defined as the negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and <5% plasma cells in bone marrow. Confirmation with repeat bone marrow biopsy was not needed. (NCT01208662)
Timeframe: Disease was assessed every cycle up to maintenance and every 3 cycles on maintenance. Median treatment duration (months) from randomization 28.2 (RVD Alone) and 36.1 (RVD plus ASCT). Maximum treatment duration was 134 months in this study cohort.

Interventionpercentage of patients (Number)
RVD Alone42.0
RVD Plus ASCT46.8

[back to top]

The Percent of Patients With Minimal Residual Disease (MRD) Status Changing to Negative at Day 100 (Post-AHCT), Among Patients With MRD Positive at the End of Induction (EOI).

Patients were treated with induction therapy (VRD) followed by autologous hematopoietic cell transplant (AHCT). MRD status of a patient with at least partial response was evaluated at the end of induction (EOI) and day 100 (post-AHCT). MRD of a patient is measured by seven-color flow cytometry. (NCT01215344)
Timeframe: 6-months post ASCT

Interventionpercentage of participants (Number)
VELCADE, Lenalidomide, Dexamethasone (VRD)30

[back to top]

Progression Free Survival by MRD Status at Day 100.

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 (NCT01215344)
Timeframe: up to 7 years

Interventionyears (Median)
MRD Negative at Day 1002.64
MRD Status Positive at Day 100 (Post-AHCT)NA

[back to top]

3-year Progression-free Survival Rate

"Progression-free survival is defined as the time from registration of induction treatment to progression, relapse or death, whichever occurs first. Patients alive without documented progression are censored at last disease assessment. 3-year progression-free survival rate is the proportion of patients who were progression-free and alive at 3 years estimated using the method of Kaplan-Meier.~Progression/relapse is defined as appearance of any new lesion more than 1.5 cm in any axis during or at the end of therapy, >=50% increase from nadir in the SPD of any previously involved nodes or extranodal masses or the size of other lesions, or >=50% increase in the longest diameter of any single previously identified node or extranodal mass more than 1 cm in its short axis." (NCT01216683)
Timeframe: Assessed every cycle during treatment and every 6 months between 2 and 5 years from study entry

Interventionproportion of participants (Number)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)0.77
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)0.82
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)0.76

[back to top]

1-year Post-induction Disease-free Survival (DFS) Rate

"1-year post induction disease-free survival rate is defined as the proportion of patients achieving complete remission during induction treatment and are alive and maintaining complete remission at 1 year after induction completion.~The purpose of this analysis is to compare the 1-year post-induction disease-free survival (DFS) rate with rituximab plus lenalidomide to rituximab alone as continuation therapy following induction treatment of bendamustine+rituximab, therefore, patients with induction treatment of bendamustine + rituximab + bortezomib were not included in this analysis." (NCT01216683)
Timeframe: Assessed at 1 year post-induction, approximately 1.5 years

Interventionproportion of participants (Number)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)0.85
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)0.67

[back to top]

1-year Disease-free Survival (DFS) Rate

"1-year post induction disease-free survival rate is defined as the proportion of patients achieving complete remission during induction treatment and are alive and maintaining complete remission at 1 year after induction completion.~This analysis was conducted among 250 evaluable patients with Cumulative Illness Rating Scale (CIRS) data available. The proportion of patients disease-free and alive at 1 year post induction treatment was compared between patients with CIRS <10 and patients with CIRS >=10. Higher CIRS scores indicate higher severity with max score of 56 points." (NCT01216683)
Timeframe: Assessed at 1 year post-induction, approximately 1.5 years

Interventionproportion of participants (Number)
CIRS <100.62
CIRS >=100.65

[back to top]

1-year Disease-free Survival (DFS) Rate

"1-year post induction disease-free survival rate is defined as the proportion of patients achieving complete remission during induction treatment and are alive and maintaining complete remission at 1 year after induction completion.~This analysis was conducted among 203 evaluable patients in the continuation treatment portion of the study. The 1-year post induction disease-free survival rate was compared between patients with FLIPI of 3-5 and patients with FLIPI of 0-2/unknown.~The FLIPI was developed in order to predict prognosis of patients with newly diagnosed follicular lymphoma (FL). The five FLIPI risk factors were: age > 60 years, Ann Arbor stage III-IV, hemoglobin level < 12 gm/dL, >4 nodal areas, and serum LDH level above normal. The FLIPI score was calculated by summing the number of risk factors. The higher the FLIPI score, the worse the prognosis." (NCT01216683)
Timeframe: Assessed at 1 year post-induction, approximately 1.5 years

Interventionproportion of participants (Number)
FLIPI 0-2/Unknown0.84
FLIPI 3-50.74

[back to top]

Complete Remission (CR) Rate

"Complete remission is defined as complete disappearance of all detectable clinical evidence of disease and disease-related symptoms if present prior to therapy.~This analysis was conducted among 222 evaluable patients. The proportion of patients with complete remission was compared between patients with Follicular Lymphoma International Prognostic Index (FLIPI) of 3-5 and patients with FLIPI of 0-2/unknown.~The FLIPI was developed in order to predict prognosis of patients with newly diagnosed follicular lymphoma (FL). The five FLIPI risk factors were: age > 60 years, Ann Arbor stage III-IV, hemoglobin level < 12 gm/dL, >4 nodal areas, and serum LDH level above normal. The FLIPI score was calculated by summing the number of risk factors. The higher the FLIPI score, the worse the prognosis." (NCT01216683)
Timeframe: Assessed every 4 weeks during induction treatment and every 8 weeks during continuation treatment, up to 2 years

Interventionproportion of participants (Number)
FLIPI 0-2/Unknown0.71
FLIPI 3-50.64

[back to top]

Complete Remission (CR) Rate

"Complete remission is defined as complete disappearance of all detectable clinical evidence of disease and disease-related symptoms if present prior to therapy.~This analysis was conducted among 250 evaluable patients with Cumulative Illness Rating Scale (CIRS) data available. The proportion of patients with complete remission was compared between patients with CIRS <10 and patients with CIRS >=10. Higher CIRS scores indicate higher severity with max score of 56 points." (NCT01216683)
Timeframe: Assessed every 4 weeks during induction treatment and every 8 weeks during continuation treatment, up to 2 years

Interventionproportion of participants (Number)
CIRS <100.70
CIRS >=100.70

[back to top]

Functional Assessment of Cancer Therapy - General (FACT-G) Total Score at Baseline

The Functional Assessment of Cancer Therapy - General (FACT-G) is a 27-item questionnaire that has four areas of measurements (physical well-being, social/family well-being, emotional well-being and functional well-being) with a scale of 0-4. The FACT-G total score ranges between 0 and 108. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at baseline

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)83.9
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)84.7
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)86.0

[back to top]

Complete Remission (CR) Rate

"Complete remission is defined as complete disappearance of all detectable clinical evidence of disease and disease-related symptoms if present prior to therapy.~This analysis was conducted among 222 evaluable patients for the primary analysis. The purpose of this analysis is to compare the complete remission rate of rituximab + bendamustine vs. bortezomib + rituximab + bendamustine as induction therapy, therefore, the proportion of patients with complete remission was compared between Arm B (bortezomib + rituximab + bendamustine) and Arms A and C combined (rituximab + bendamustine)." (NCT01216683)
Timeframe: Assessed every 4 weeks during induction treatment and every 8 weeks during continuation treatment, up to 2 years

Interventionproportion of participants (Number)
Arm A and Arm C (Induction With Bendamustine + Rituximab)0.62
Arm B (Induction With Bendamustine + Rituximab + Bortezomib)0.75

[back to top]

Proportion of Patients With Grade 3 or Higher Peripheral Neuropathy

Peripheral neuropathy was assessed using NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Proportion of patients with grade 3 or higher peripheral neuropathy was compared between patients with subcutaneous bortezomib and patients with intravenous bortezomib. (NCT01216683)
Timeframe: Assessed every cycle during treatment and for 30 days after discontinuation of treatment, up to 15 years

Interventionproportion of participants (Number)
Subcutaneous Bortezomib0.06
Intravenous Bortezomib0.12

[back to top]

5-year Overall Survival Rate

Overall survival is defined as the time from randomization to death or date last known alive. 5-year overall survival rate is the proportion of patients who were alive at 5 years estimated using the method of Kaplan-Meier. (NCT01216683)
Timeframe: Assessed every cycle during treatment and every 6 months between 2 and 5 years from study entry

Interventionproportion of participants (Number)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)0.87
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)0.86
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)0.83

[back to top]

Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) Subscale Score at the End of Induction Treatment

The Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) subscale is comprised of 11 items that assess neurotoxicity with a scale of 0-4. The FACT-GOG-NTX subscale score ranges between 0 and 44. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at end of induction treatment (cycle 6), approximately 6 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)39.8
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)36.1
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)39.0

[back to top]

Progression-free Survival

"Progression-free survival is defined as the time from registration of induction treatment to progression, relapse or death, whichever occurs first. Patients alive without documented progression are censored at last disease assessment. Progression/relapse is defined as appearance of any new lesion more than 1.5 cm in any axis during or at the end of therapy, >=50% increase from nadir in the SPD of any previously involved nodes or extranodal masses or the size of other lesions, or >=50% increase in the longest diameter of any single previously identified node or extranodal mass more than 1 cm in its short axis.~The five FLIPI risk factors were: age > 60 years, Ann Arbor stage III-IV, hemoglobin level < 12 gm/dL, >4 nodal areas, and serum LDH level above normal. The FLIPI score was calculated by summing the number of risk factors. The higher the FLIPI score, the worse the prognosis." (NCT01216683)
Timeframe: Assessed every cycle during treatment and every 6 months between 2 and 5 years from study entry

Interventionyears (Median)
FLIPI 0-2/Unknown6.1
FLIPI 3-56.2

[back to top]

Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Subscale Score at Mid-induction Treatment

"The Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) is comprised of 13 items that assess fatigue and its impact with a scale of 0-4. The FACIT-Fatigue subscale score ranges between 0 and 52. The higher the score, the better the quality of life.~FACIT-Fatigue subscale score at cycle 3 is considered as mid-treatment score. If FACIT-Fatigue subscale score at cycle 3 is not available, the score at cycle 4 will be used as the mid-treatment score." (NCT01216683)
Timeframe: Assessed at cycle 3 or cycle 4, approximately 3 or 4 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)39.2
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)34.5
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)36.3

[back to top]

Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Subscale Score at End of Induction Treatment

The Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) is comprised of 13 items that assess fatigue and its impact with a scale of 0-4. The FACIT-Fatigue subscale score ranges between 0 and 52. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at cycle 6, approximately 6 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)39.9
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)39.1
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)36.3

[back to top]

Functional Assessment of Cancer Therapy - General (FACT-G) Total Score at End of Induction Treatment

The Functional Assessment of Cancer Therapy - General (FACT-G) is a 27-item questionnaire that has four areas of measurements (physical well-being, social/family well-being, emotional well-being and functional well-being) with a scale of 0-4. The FACT-G total score ranges between 0 and 108. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at cycle 6, approximately 6 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)86.0
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)86.5
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)84.9

[back to top]

Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) Subscale Score at Baseline

The Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) is a 15-item questionnaire that evaluates disease-related symptoms and concerns specific to lymphoma with a scale of 0-4. The FACT-Lym subscale score ranges between 0 and 60. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at baseline

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)43.8
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)44.3
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)44.9

[back to top]

Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) Subscale Score at End of Induction Treatment

The Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) is a 15-item questionnaire that evaluates disease-related symptoms and concerns specific to lymphoma with a scale of 0-4. The FACT-Lym subscale score ranges between 0 and 60. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at cycle 6, approximately 6 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)48.9
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)48.4
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)47.2

[back to top]

Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Subscale Score at Baseline

The Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) is comprised of 13 items that assess fatigue and its impact with a scale of 0-4. The FACIT-Fatigue subscale score ranges between 0 and 52. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at baseline

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)38.6
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)37.9
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)38.1

[back to top]

Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) Subscale Score at Mid-induction Treatment

"The Functional Assessment of Cancer Therapy - Lymphoma (FACT-Lym) is a 15-item questionnaire that evaluates disease-related symptoms and concerns specific to lymphoma with a scale of 0-4. The FACT-Lym subscale score ranges between 0 and 60. The higher the score, the better the quality of life.~FACT-Lym subscale score at cycle 3 is considered as mid-treatment score. If FACT-Lym subscale score at cycle 3 is not available, the score at cycle 4 will be used as the mid-treatment score." (NCT01216683)
Timeframe: Assessed at cycle 3 or cycle 4, approximately 3 or 4 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)47.9
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)46.8
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)46.5

[back to top]

Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) Subscale Score at Baseline

The Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) subscale is comprised of 11 items that assess neurotoxicity with a scale of 0-4. The FACT-GOG-NTX subscale score ranges between 0 and 44. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at baseline

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)39.9
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)38.7
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)38.8

[back to top]

Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) Subscale Score at Mid-induction Treatment

The Functional Assessment of Cancer Therapy/Gynecologic Oncology Group - Neurotoxicity (FACT-GOG-NTX) subscale is comprised of 11 items that assess neurotoxicity with a scale of 0-4. The FACT-GOG-NTX subscale score ranges between 0 and 44. The higher the score, the better the quality of life. (NCT01216683)
Timeframe: Assessed at cycle 3, approximately 3 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)40.0
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)38.6
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)39.1

[back to top]

5-year Overall Survival Rate

"Overall survival is defined as the time from randomization to death or date last known alive. 5-year overall survival rate is the proportion of patients who were alive at 5 years estimated using the method of Kaplan-Meier.~The FLIPI was developed in order to predict prognosis of patients with newly diagnosed follicular lymphoma (FL). The five FLIPI risk factors were: age > 60 years, Ann Arbor stage III-IV, hemoglobin level < 12 gm/dL, >4 nodal areas, and serum LDH level above normal. The FLIPI score was calculated by summing the number of risk factors. The higher the FLIPI score, the worse the prognosis." (NCT01216683)
Timeframe: Assessed every cycle during treatment and every 6 months between 2 and 5 years from study entry

Interventionproportion of participants (Number)
FLIPI 0-2/Unknown0.90
FLIPI 3-50.81

[back to top]

5-year Overall Survival Rate

"Overall survival is defined as the time from randomization to death or date last known alive. 5-year overall survival rate is the proportion of patients who were alive at 5 years estimated using the method of Kaplan-Meier.~The 5-year overall survival rate is reported by Cumulative Illness Rating Scale (CIRS) score (<10 vs. >=10). Higher CIRS scores indicate higher severity with max score of 56 points." (NCT01216683)
Timeframe: Assessed every cycle during treatment and every 6 months between 2 and 5 years from study entry

Interventionproportion of participants (Number)
CIRS <100.87
CIRS >=100.80

[back to top]

Functional Assessment of Cancer Therapy - General (FACT-G) Total Score at Mid-treatment

"The Functional Assessment of Cancer Therapy - General (FACT-G) is a 27-item questionnaire that has four areas of measurements (physical well-being, social/family well-being, emotional well-being and functional well-being) with a scale of 0-4. The FACT-G total score ranges between 0 and 108. The higher the score, the better the quality of life.~FACT-G total score at cycle 3 is considered as mid-treatment score. If FACT-G total score at cycle 3 is not available, the score at cycle 4 will be used as the mid-treatment score." (NCT01216683)
Timeframe: Assessed at cycle 3 or cycle 4, approximately 3 or 4 months

Interventionscore on a scale (Mean)
Arm A Then Arm D (Induction With Bendamustine + Rituximab; Continuation With Rituximab)85.5
Arm B Then Arm E (Induction With Bendamustine + Rituximab + Bortezomib; Continuation With Rituximab)84.2
Arm C Then Arm F (Induction With Bendamustine+Rituximab; Continuation With Lenalidomide + Rituximab)85.2

[back to top]

3-year Progression-free Survival Rate

"Progression-free survival is defined as the time from registration of induction treatment to progression, relapse or death, whichever occurs first. Patients alive without documented progression are censored at last disease assessment. 3-year progression-free survival rate is the proportion of patients who were progression-free and alive at 3 years estimated using the method of Kaplan-Meier.~Progression/relapse is defined as appearance of any new lesion more than 1.5 cm in any axis during or at the end of therapy, >=50% increase from nadir in the SPD of any previously involved nodes or extranodal masses or the size of other lesions, or >=50% increase in the longest diameter of any single previously identified node or extranodal mass more than 1 cm in its short axis.~The 3-year progression-free survival rate is reported by Cumulative Illness Rating Scale (CIRS) score (<10 vs. >=10). Higher CIRS scores indicate higher severity with max score of 56 points." (NCT01216683)
Timeframe: Assessed every cycle during treatment and every 6 months between 2 and 5 years from study entry

Interventionproportion of participants (Number)
CIRS <100.83
CIRS >=100.68

[back to top]

Phase 2: 1 Year Survival Rate

1-year survival rate is defined as the percentage of participants still alive at year after the first dose of stud drug. (NCT01217957)
Timeframe: 1 year after first dose of study drug

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone92
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + Dexamethasone92

[back to top]

Phase 2: Duration of Response (DOR)

DOR was measured as the time in months from the date of first documentation of a confirmed response (CR + PR+ VGPR) to the date of the first documented disease progression (PD). Response was assessed by the investigator using International Myeloma Working Group (IMWG) Criteria. CR=negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow. VGPR=Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. (NCT01217957)
Timeframe: Up to 787 days

Interventionmonths (Median)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + DexamethasoneNA
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + DexamethasoneNA

[back to top]

Phase 2: Objective Response Rate (ORR) Following Treatment With the Combination Of Oral Ixazomib, Lenalidomide And Low-Dose Dexamethasone

ORR was defined as the percentage of participants with Complete (CR) + Very Good Partial Response (VGPR) assessed by the investigatory using International Myeloma Working Group (IMWG) Criteria. CR=Negative immunofixation on the serum and urine and; disappearance of any soft tissue plasmacytomas and; < 5% plasma cells in bone marrow. VGPR=Serum and urine M-protein detectable by immunofixation but not on electrophoresis or; 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. (NCT01217957)
Timeframe: Until occurrence of progressive disease or unacceptable toxicity (Up to 787 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone59
Phase 2: Ixazomib 4.0mg/2.23 + Lenalidomide + Dexamethasone62

[back to top]

Phase 2: Overall Response Rate (ORR)

ORR was defined as the percentage of participants with CR, VGPR and Partial Response (PR) assessed by the investigator using IMWG criteria. CR=Negative immunofixation on the serum and urine + Disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow. PR=50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by 90% or to < 200 mg per 24 hours. VGPR= Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. (NCT01217957)
Timeframe: Up to 787 days

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone88
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + Dexamethasone88

[back to top]

Phase 2: Overall Survival (OS)

OS was measured as the time in months from the first dose of study treatment to the date of death + 1 day. (NCT01217957)
Timeframe: From the first dose of study treatment to the date of death (up to 787 days)

Interventionparticipants (Median)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + DexamethasoneNA
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + DexamethasoneNA

[back to top]

Phase 2: Progression Free Survival (PFS)

PFS was measured as the time in months from the first dose of study treatment to the date of the first documented PD or death. (NCT01217957)
Timeframe: Up to 787 days

Interventionmonths (Median)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone14.98
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + DexamethasoneNA

[back to top]

Phase 2: Time to Best Response

Time to Best Response was measured as the time in months from the first dose of study treatment to the date of first documented documentation of a confirmed response of partial response (PR) or better. (NCT01217957)
Timeframe: Up to 787 days

Interventionmonths (Median)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone2.96
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + Dexamethasone3.01

[back to top]

Phase 1: Maximum Tolerated Dose (MTD) of Ixazomib Administered Weekly in Combination With Lenalidomide and Low-Dose Dexamethasone

MTD of ixazomib will be determined by assessing adverse events and serious adverse events, clinical laboratory values, neurotoxicity grading, and vital sign measurements. (NCT01217957)
Timeframe: Until occurrence of progressive disease or unacceptable toxicity (Up to 336 days)

Interventionmg/m^2 (Number)
Phase 1: Ixazomib + Lenalidomide + Dexamethasone2.97

[back to top]

Phase 1: Rac: Accumulation Ratio of Ixazomib

The accumulation ratio (Rac) was estimated as the ratio of AUC(0-168) on Day 15 to the AUC(0-168) on Day 1. AUC(0-168) is the area under the plasma concentration-time curve from time 0 to 168 hours postdose for ixazomib. (NCT01217957)
Timeframe: Cycle 1, Day 15

InterventionRatio (Geometric Mean)
Phase 1: Ixazomib 1.68 mg/m^2 + Lenalidomide + DexamethasoneNA
Phase 1: Ixazomib 2.23 mg/m^2 + Lenalidomide + Dexamethasone1.849
Phase 1: Ixazomib 2.97 mg/m^2 + Lenalidomide + Dexamethasone2.051

[back to top]

Phase 1: Tmax: Time to Reach the Maximum Observed Plasma Concentration (Cmax) for Ixazomib

Tmax: Time to reach the first maximum observed plasma concentration (Cmax), equal to time (hours) to Cmax, obtained directly from the plasma concentration-time curve. (NCT01217957)
Timeframe: Cycle 1, Days 1 and 15

,,,
Interventionhours (Median)
Day 1 (n=1, 3, 4, 1)Day 15 (n=2, 3, 4, 1)
Phase 1: Ixazomib 1.68 mg/m^2 + Lenalidomide + Dexamethasone1.0204.165
Phase 1: Ixazomib 2.23 mg/m^2 + Lenalidomide + Dexamethasone1.5201.000
Phase 1: Ixazomib 2.97 mg/m^2 + Lenalidomide + Dexamethasone1.0601.015
Phase 1: Ixazomib 3.95 mg/m^2 + Lenalidomide + Dexamethasone0.2502.000

[back to top]

Phase 2: Time to Progression (TTP)

TTP was measured as the time in months from the first dose of study treatment to the date of the first documented progressive disease (PD). (NCT01217957)
Timeframe: From the first dose of study treatment to the date of first documented progressive disease (Up to 787 days)

Interventionmonths (Median)
Phase 2: Ixazomib 4.0 mg + Lenalidomide + DexamethasoneNA
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + DexamethasoneNA

[back to top]

Phase 1: AUC(0-168): Area Under the Plasma Concentration-Time Curve From Time 0 to 168 Hours Postdose for Ixazomib

AUC(0-168) is a measure of the area under the plasma concentration-time curve from time 0 to 168 hours postdose for Ixazomib. (NCT01217957)
Timeframe: Cycle 1, Days 1 and 15

,,,
Interventionhr*ng/mL (Geometric Mean)
Day 1 (n=1, 3, 4, 1)Day 15 (n=2, 3, 3, 1)
Phase 1: Ixazomib 1.68 mg/m^2 + Lenalidomide + DexamethasoneNA834.608
Phase 1: Ixazomib 2.23 mg/m^2 + Lenalidomide + Dexamethasone587.6671083.998
Phase 1: Ixazomib 2.97 mg/m^2 + Lenalidomide + Dexamethasone923.4841831.324
Phase 1: Ixazomib 3.95 mg/m^2 + Lenalidomide + DexamethasoneNANA

[back to top]

Phase 1: Cmax: Maximum Observed Plasma Concentration for Ixazomib

Cmax: Maximum Observed Plasma Concentration (Cmax) is the peak plasma concentration of ixazomib obtained directly from the plasma concentration-time curve. (NCT01217957)
Timeframe: Cycle 1, Days 1 and 15

,,,
Interventionng/mL (Geometric Mean)
Day 1 (n=1, 3, 4, 1)Day 15 (n=2, 3, 4, 1)
Phase 1: Ixazomib 1.68 mg/m^2 + Lenalidomide + DexamethasoneNA11.999
Phase 1: Ixazomib 2.23 mg/m^2 + Lenalidomide + Dexamethasone22.30331.368
Phase 1: Ixazomib 2.97 mg/m^2 + Lenalidomide + Dexamethasone94.77953.517
Phase 1: Ixazomib 3.95 mg/m^2 + Lenalidomide + DexamethasoneNANA

[back to top]

Phase 1: Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs) as a Measure of Safety and Tolerability

An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. A serious adverse event (SAE) is an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; or congenital anomaly; or a medically important event. (NCT01217957)
Timeframe: Until occurrence of progressive disease or unacceptable toxicity (Up to 336 days)

,,,
Interventionparticipants (Number)
Any AESAE
Phase 1: Ixazomib 1.68 mg/m^2 + Lenalidomide + Dexamethasone32
Phase 1: Ixazomib 2.23 mg/m^2 + Lenalidomide + Dexamethasone33
Phase 1: Ixazomib 2.97 mg/m^2 + Lenalidomide + Dexamethasone61
Phase 1: Ixazomib 3.95 mg/m^2 + Lenalidomide + Dexamethasone32

[back to top]

Phase 2: Percentage of Participants With Complete Response (CR) and Very Good Partial Response (VGPR)

Response was assessed by the investigator using International Myeloma Working Group (IMWG) Criteria. CR is defined as negative immunofixation on the serum and urine and; disappearance of any soft tissue plasmacytomas and; < 5% plasma cells in bone marrow. VGPR is defined as Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. (NCT01217957)
Timeframe: After Cycles 3, 6 and 9 (Up to 787 days)

,
Interventionpercentage of participants (Number)
After 3 cyclesAfter 6 cyclesAfter 9 cycles
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone354757
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + Dexamethasone374858

[back to top]

Phase 2: Percentage of Participants With Complete Response (CR), Stringent Complete Response (sCR), Very Good Partial Response (VGPR), Near Complete Response (nCR), Partial Response (PR) and Minimal Response (MR)

Response was assessed by the investigator using International Myeloma Working Group (IMWG) Criteria. CR=Negative immunofixation on the serum and urine + Disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow. sCR= CR + Normal free light chain (FLC) ratio and Absence of clonal cells in bone marrow. PR=≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to < 200 mg per 24 hours. VGPR= Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. nCR=Positive immunofixation analysis of serum or urine as the only evidence of disease. Disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow. MR=25% to 49% reduction in serum paraprotein and 50% to 89% reduction in urine light chain excretion for 6 weeks. (NCT01217957)
Timeframe: Cycles 3, 6, 9 and 12 (Up to 787 days)

,
Interventionpercentage of participants (Number)
CRsCRVGPRnCRPRMR
Phase 2: Ixazomib 4.0 mg + Lenalidomide + Dexamethasone206392676
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + Dexamethasone2310382656

[back to top]

Phase 2: Percentage of Participants With Grade 3 or Higher AEs, SAEs and Treatment Discontinuation

An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. A serious adverse event (SAE) is an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; or congenital anomaly; or a medically important event. (NCT01217957)
Timeframe: Until occurrence of progressive disease or unacceptable toxicity (Up to 787 days)

,
Interventionpercentage of participants (Number)
Grade 3 or Higher AEsSAEsAEs Resulting in Treatment Discontinuation
Phase 2 :Ixazomib 4.0 mg + Lenalidomide + Dexamethasone76408
Phase 2: Ixazomib 4.0 mg/2.23 + Lenalidomide + Dexamethasone75438

[back to top] [back to top]

Number of Patients With a Transfusion Independence Response

Response defined as transfusion independence (no red blood cell transfusions) for at least 8 weeks, anytime during the six 28-day cycles of therapy. (NCT01222195)
Timeframe: Over six 28-day cycles (approximately 168 days)

Interventionparticipants (Number)
Lenalidomide + Darbepoetin Alfa0

[back to top]

Median Progression Free Survival (PFS)

Primary definition of Progression-free survival (PFS) defined as the time from randomization to the date of first documented tumor progression or death due to any cause. Participants were censored at the last adequate assessment prior to the start of any subsequent systemic-therapy or at the last adequate assessment prior to 2 missing assessments (> 10 weeks). Participants who died more than 10 weeks after the randomization date and had no on-treatment assessment were censored at the randomization date. Clinical deterioration was not considered progression. The primary analysis of PFS was based on the primary definition using the Independent Review Committee (IRC) tumor assessment using the European Group for Blood and Bone Marrow Transplant (EBMT) criteria. Tumor assessments were made every 4 weeks (±1 week) relative to the first dose of study medication. (NCT01239797)
Timeframe: From randomization up to 326 events (up to approximately 38 months)

InterventionMonths (Median)
Lenalidomide + Dexamethasone + Elotuzumab19.35
Lenalidomide + Dexamethasone14.85

[back to top]

Objective Response Rate (ORR)

Objective response rate (ORR) defined as the percentage of participants with a best response on-study of partial response (PR) or better (stringent CR [sCR], complete response [CR], very good partial response [VGPR], and partial response [PR]) based on the Independent Review Committee (IRC) assessment of best response using the European Group for Blood and Bone Marrow Transplant (EBMT) assessment criteria. Participants were censored at the last adequate assessment prior to the start of any subsequent systemic-therapy or at the last adequate assessment prior to 2 missing assessments (> 10 weeks). Participants who died more than 10 weeks after the randomization date and had no on-treatment assessment were censored at the randomization date. Clinical deterioration was not considered progression. Assessments were made every 4 weeks. (NCT01239797)
Timeframe: From randomization up to approximately 38 months

InterventionPercentage of participants (Number)
Lenalidomide + Dexamethasone + Elotuzumab78.5
Lenalidomide + Dexamethasone65.5

[back to top]

Median Progression Free Survival (PFS) - Extended Collection

"The time from randomization to the date of first documented tumor progression or death due to any cause. Participants were censored at the last adequate assessment prior to the start of any subsequent systemic-therapy or at the last adequate assessment prior to 2 missing assessments (> 10 weeks). Participants who died more than 10 weeks after the randomization date and had no on-treatment assessment were censored at the randomization date. Clinical deterioration was not considered progression. Tumor assessments were made every 4 weeks (±1 week) relative to the first dose of study medication based on Independent Review Committee (IRC) tumor assessment using the European Group for Blood and Bone Marrow Transplant (EBMT) criteria.~Note: This outcome measure represents an updated version of the primary endpoint to include additional data collection that has occurred after the primary completion date. (Assessments were made until 06-Jul-2018)" (NCT01239797)
Timeframe: From randomization up to to the date of first documented tumor progression or death (up to approximately 85 months)

InterventionMonths (Median)
Lenalidomide + Dexamethasone + Elotuzumab19.42
Lenalidomide + Dexamethasone14.92

[back to top]

Median Overall Survival (OS)

"Overall survival is defined as the time from randomization to the date of death from any cause. If a subject has not died, their survival time will be censored at the date of last contact (last known alive date). A subject will be censored at the date of randomization if they were randomized but had no follow-up. (Based on Kaplan Meier estimates)" (NCT01239797)
Timeframe: Randomization to the date of death from any cause (up to approximately 9 years)

InterventionMonths (Median)
Lenalidomide + Dexamethasone + Elotuzumab48.30
Lenalidomide + Dexamethasone39.62

[back to top]

Change From Baseline of Mean Score Pain Severity (BPI-SF)

"The change from baseline of the mean score of pain severity at the end of treatment using the Brief Pain Inventory-Short Form (BPI-SF). The BPI-SF is a self administered questionnaire developed to assess the severity of pain (the sensory dimension) as well as the degree to which pain interferes with function (the reactive dimension). The BPI-SF uses 0 (No pain, No interference) to 10 (Pain as bad as you can imagine, Highest imaginable interference) numeric rating scale." (NCT01239797)
Timeframe: From baseline up to approximately 38 months

InterventionScore on a scale (Mean)
Lenalidomide + Dexamethasone + Elotuzumab0.52
Lenalidomide + Dexamethasone-0.04

[back to top]

Change From Baseline of Mean Score Pain Interference (BPI-SF)

"The change from baseline of the mean score of pain interference at the end of treatment using the Brief Pain Inventory-Short Form (BPI-SF). The BPI-SF is a self administered questionnaire developed to assess the severity of pain (the sensory dimension) as well as the degree to which pain interferes with function (the reactive dimension). The BPI-SF uses 0 (No pain, No interference) to 10 (Pain as bad as you can imagine, Highest imaginable interference) numeric rating scale." (NCT01239797)
Timeframe: From baseline up to approximately 38 months

InterventionScore on a scale (Mean)
Lenalidomide + Dexamethasone + Elotuzumab0.95
Lenalidomide + Dexamethasone0.48

[back to top]

Stage II - Overall Response Rate

Rate of overall response by incidence of Complete Response, Partial Response, and Stable Disease per Modified International Working Group (IWG) response criteria for patients enrolled in Stage II (NCT01241734)
Timeframe: After 2 Cycles of Treatment (28 Day Cycles)

InterventionParticipants (Count of Participants)
Stage I, Cohort 10
Stage I, Cohort 20
Stage I, Cohort 30
Stage I, Cohort 40
Stage II2

[back to top]

Stage I - Dose Limiting Toxicity Incidence Rate

Incidence of DLT in patients enrolled in stage I for determination of MTD (NCT01241734)
Timeframe: Cycle 1 of Treatment (28 Days)

InterventionParticipants (Count of Participants)
Stage I, Cohort 10
Stage I, Cohort 20
Stage I, Cohort 30
Stage I, Cohort 40
Stage II0

[back to top]

Stage I - Safety (Type, Frequency, Severity and Relationship of Adverse Events to Study Treatment) and Tolerability

Determine the maximum tolerated dose (MTD) of lenalidomide when given in combination with rituximab, Ifosfamide, etoposide, and carboplatin (RICE) for the treatment of DLBCL patients in first relapse by incidence of dose-limiting toxicity (DLT) for Stage I Patients Only (NCT01241734)
Timeframe: Cycle 1 of Treatment (28 Days)

Interventionmg (Number)
Revlimid (Lenalidomide) in Combination25

[back to top]

Stage II - 1 Year Overall Survival (OS)

1 Year Overall Survival (OS) of patients enrolled in stage II (NCT01241734)
Timeframe: Up to 1 Year

InterventionParticipants (Count of Participants)
Stage I, Cohort 10
Stage I, Cohort 20
Stage I, Cohort 30
Stage I, Cohort 40
Stage II2

[back to top]

Stage II - 1 Year Progression Free Survival (PFS)

Rate of Progression Free Survival (PFS) at 1 Year of patients enrolled in stage II (NCT01241734)
Timeframe: Up to 1 Year

InterventionParticipants (Count of Participants)
Stage I, Cohort 10
Stage I, Cohort 20
Stage I, Cohort 30
Stage I, Cohort 40
Stage II2

[back to top]

Stage II - 2 Year Overall Survival (OS)

2 Year Overall Survival (OS) of patients enrolled in stage II (NCT01241734)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Stage I, Cohort 10
Stage I, Cohort 20
Stage I, Cohort 30
Stage I, Cohort 40
Stage II2

[back to top]

Stage II - 2 Year Progression Free Survival (PFS)

Rate of Progression Free Survival (PFS) at 1 Year of patients enrolled in stage II (NCT01241734)
Timeframe: Up to 2 Years

InterventionParticipants (Count of Participants)
Stage I, Cohort 10
Stage I, Cohort 20
Stage I, Cohort 30
Stage I, Cohort 40
Stage II2

[back to top]

The Overall Survival of Patients Treated With the Combination of Lenalidomide and Azacitidine

(NCT01241786)
Timeframe: 9 Months

Interventionparticipants (Number)
Revlamid + Vidaza5

[back to top] [back to top]

the Rate of Response to the Combination of Azacitidine + Lenalidomide in Select Patients

the rate of response to the combination of azacitidine + lenalidomide in select patients with relapsed/refractory CLL and small lymphocytic lymphoma (SLL). (NCT01241786)
Timeframe: 9 Months

Interventionparticipants (Number)
Study Terminated0

[back to top]

The Progression Free Survival of Patients Treated With the Combination of Lenalidomide and Azacitidine

(NCT01241786)
Timeframe: 9 Months

InterventionMonths (Median)
Revlamid + Vidaza1

[back to top]

Duration of Reduction in Spleen Volume

Duration of spleen volume reduction is defined as the time from the first occurrence of a >=35% reduction from baseline in spleen volume until the date of the first documented progression. (NCT01243944)
Timeframe: 256 Weeks

Interventionprobability (Number)
16 weeks32 weeks48 weeks64 weeks80 weeks96 weeks112 weeks128 weeks144 weeks160 weeks176 weeks192 weeks208 weeks224 weeks240 weeks256 weeks
Ruxolitinib1.001.001.001.001.000.980.950.950.950.930.930.930.870.72NANA

[back to top]

The Percentage of Participants Who Achieved Durable Spleen Volume Reduction at Week 48

Durable Spleen Volume Reduction was defined as a participant who achieved at least 35% reduction from baseline in spleen volume at Week 32 and maintained that response 48 weeks after randomization. (NCT01243944)
Timeframe: 48 Weeks

Interventionpercentage of participants (Number)
Ruxolitinib37.3
Best Available Therapy0.9

[back to top]

The Percentage of Participants Achieving a Durable Complete or Partial Clinicohematologic Response at Week 48

Durable Complete or Partial Clinicohematologic Response was defined as any participant who achieved complete or partial clinicohematologic response per the European LeukemiaNet modified criteria for response in polycythemia vera at Week 32 and maintained that response 48 weeks after randomization. (NCT01243944)
Timeframe: 48 Weeks

,
Interventionpercentage of participants (Number)
Complete response ratePartial response rate
Best Available Therapy0.90.9
Ruxolitinib7.350.9

[back to top]

The Percentage of Participants Who Achieved a Durable Hematocrit Control at Week 48

Durable Hematocrit Control was defined as any participant who achieved phlebotomy eligibility independence from Week 8 to Week 32 and maintained hematocrit control up to 48 weeks after randomization. (NCT01243944)
Timeframe: 48 Weeks

Interventionpercentage of participants (Number)
Ruxolitinib54.5
Best Available Therapy1.8

[back to top]

The Percentage of Participants Who Achieved a Durable Complete Hematological Remission at Week 48

Durable Complete Hematological Remission was defined as any participant who achieved Complete Hematological Remission at Week 32 and maintained their response up to 48 weeks after randomization. (NCT01243944)
Timeframe: 48 Weeks

Interventionpercentage of participants (Number)
Ruxolitinib20.9
Best Available Therapy0.9

[back to top]

The Percentage of Participants Who Achieved Overall Clinicohematologic Response at Week 32

Overall Clinicohematologic Response is defined as any participant who achieved a complete or partial clinicohematologic response per the European LeukemiaNet modified criteria for response in polycythemia vera (PV). A Complete Response (CR) is defined as: hematocrit control, spleen volume reduction at least 35% from baseline, platelet count less than or equal to 400 x 10(9)/L, and white blood cell count less than or equal to 10 x 10(9)/L. A Partial Response (PR) is defined as hematocrit control or response in all 3 of the other criteria. (NCT01243944)
Timeframe: 32 Weeks

,
Interventionpercentage of participants (Number)
Complete response ratePartial response rate
Best Available Therapy0.918.8
Ruxolitinib8.254.5

[back to top]

Estimated Duration of the Primary Response

"Duration of the primary response is defined as the time from the first occurrence when both components of the primary endpoint are met until the date of the first documented disease progression (end of response).~Kaplan-Meier estimates are provided for duration of primary response." (NCT01243944)
Timeframe: Through study completion, analysis was conducted when all participants had completed the Week 80 visit or discontinued the study

Interventionprobability (Number)
16 weeks32 weeks48 weeks64 weeks80 weeks96 weeks112 weeks128 weeks144 weeks160 weeks176 weeks192 weeks208 weeks224 weeks240 weeks256 weeks
Ruxolitinib1.001.000.920.920.920.880.840.840.840.790.790.740.740.74NANA

[back to top]

Duration of the Absence of Phlebotomy Eligibility

Duration of the absence of phlebotomy eligibility is defined as the time from the first occurrence of absence of phlebotomy eligibility until the date of the first documented progression. (NCT01243944)
Timeframe: 256 Weeks

Interventionprobability (Number)
16 weeks32 weeks48 weeks64 weeks80 weeks96 weeks112 weeks128 weeks144 weeks160 weeks176 weeks192 weeks208 weeks224 weeks240 weeks256 weeks
Ruxolitinib1.001.000.970.920.910.910.870.840.840.820.790.770.730.730.730.73

[back to top]

Estimated Duration of the Complete Hematological Remission

"Duration of the complete hematological remission is defined as the time from the first occurrence of complete hematological remission until the date of the first documented progression (end of response).~Kaplan-Meier estimates are provided for duration of complete hematological remission." (NCT01243944)
Timeframe: Through study completion, analysis was conducted when all participants had completed the Week 80 visit or discontinued the study

Interventionprobability (Number)
16 weeks32 weeks48 weeks64 weeks80 weeks96 weeks112 weeks128 weeks144 weeks160 weeks176 weeks192 weeks208 weeks224 weeks240 weeks256 weeks
Ruxolitinib1.001.000.880.830.740.740.690.690.650.650.550.550.550.55NANA

[back to top]

Duration of The Overall Clinicohematologic Response

Duration of the overall clinicohematologic response was defined as the time from the first occurrence of complete response (CR) or partial response (PR) until the date of the first documented disease progression. (NCT01243944)
Timeframe: 256 Weeks

Interventionprobability (Number)
16 weeks32 weeks48 weeks64 weeks80 weeks96 weeks112 weeks128 weeks144 weeks160 weeks176 weeks192 weeks208 weeks224 weeks240 weeks256 weeks
Ruxolitinib1.000.990.960.910.880.880.850.820.820.800.750.700.670.670.670.67

[back to top]

The Percentage of Participants Achieving Complete Hematological Remission at Week 32

Complete Hematological Remission at Week 32 was defined as any participant who achieved hematocrit control with a platelet count less than or equal to 400 X 10^9/L and a white blood cell count less than or equal to 10 X 10^9/L. (NCT01243944)
Timeframe: 32 Weeks

Interventionpercentage of participants (Number)
Ruxolitinib23.6
Best Available Therapy8.0

[back to top]

The Percentage of Participants Achieving a Primary Response at Week 32

Primary response was defined as having achieved hematocrit control (the absence of phlebotomy eligibility beginning at the Week 8 visit and continuing through Week 32) and Spleen Volume Reduction (a greater than or equal to 35% reduction from baseline in spleen volume at Week 32). (NCT01243944)
Timeframe: 32 Weeks

Interventionpercentage of participants (Number)
Ruxolitinib22.7
Best Available Therapy0.9

[back to top]

The Percentage of Participants Achieving a Durable Primary Response at Week 48

Durable Primary Response was defined as any participant who achieved the primary outcome measure and who maintained their response up to 48 weeks after randomization. (NCT01243944)
Timeframe: 48 Weeks

Interventionpercentage of participants (Number)
Ruxolitinib20.0
Best Available Therapy0.9

[back to top]

Time to Discontinuation of Treatment

(NCT01246076)
Timeframe: Up to 56 weeks (14 cycles)

Interventioncycles (Median)
Lenalidomide2

[back to top]

Number of Participants With Confirmed Responses (Complete Remission, Partial Remission, or Hematologic Improvement) as Defined by the International Working Group Criteria

"Complete remission (CR): ≤5% myeloblasts bone marrow blasts, normal maturation in all cell lines (dysplasia will be noted), ≥11 g/dl peripheral blood hemoglobin, ≥100x10^9cells/μL peripheral blood platelets, ≥1000 cells/ μL peripheral blood absolute neutrophil count (ANC), and 0% peripheral blood blasts.~Marrow complete remission (MCR): ≤5% myeloblasts and decreased by ≥50% compared to pre-treatment bone marrow blasts, bone marrow morphology not relevant, and peripheral blood (if hematological improvement they will be noted in addition to marrow CR).~Partial remission (PR): previously had ≥5% myeloblasts and now have ≥5% myeloblasts but decreased by ≥50% compared to pre-treatment, bone marrow morphology not relevant, ≥11 g/dl peripheral blood hemoglobin, ≥100x109cells/μL peripheral blood platelets, ≥1000 cells/ μL peripheral blood ANC, and 0% peripheral blood blasts" (NCT01246076)
Timeframe: Up to 56 weeks (14 cycles of treatment)

Interventionparticipants (Number)
CRMCRPRHI
Lenalidomide0802

[back to top] [back to top]

Duration of Response

(NCT01246076)
Timeframe: Until 6 months after end of treatment

Interventiondays (Median)
Lenalidomide70

[back to top]

Overall Survival Rate

-Overall survival rate is the percentage of participants who were alive 6 months after end of treatment. (NCT01246076)
Timeframe: 6 months after end of treatment (up to 82 weeks from start of treatment)

Interventionpercentage of participants (Number)
Lenalidomide29

[back to top]

Phase I: Number of Participants With Dose Limiting Toxicity

The number of patients who had a DLT during the dose finding/confirming portion (Phase I) of the trial for the safety of the combination of sirolimus, tacrolimus and lenalidomid. Patients will be monitored for 28 days (a cycle) to determine whether a DLT is experienced for the specific dose level. (NCT01303965)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
Phase I Dose Finding0

[back to top]

Phase II - Time to Platelet Engraftment

Time to platelet engraftment will be analyzed by the Kaplan-Meier method. The time to engraftment of platelets is defined as the time from day 0 to the first of three consecutive Complete Blood Counts (CBCs) obtained on different days after transplantation during which the platelet count is at least 20 x109/l. The CBCs obtained should be at least seven days after the most recent platelet transfusion. Only patients who achieved engraftment of platelets will be included in the analysis. The median and 95% confidence intervals will be provided. (NCT01303965)
Timeframe: Transplant (Day 0) through 1 year post-transplant

Interventiondays (Median)
Phase II19

[back to top]

Phase II - Time to Neutrophil Engraftment

Time to neutrophil engraftment will be analyzed by the Kaplan-Meier method. The time to engraftment of neutrophils is defined as the time from day 0 to the date of the first of three consecutive days after transplantation during which the absolute neutrophils count (ANC) is at least 0.5 x109/l. Patients surviving at least 14 days after transplant will be evaluable for this endpoint. Patients who did not have neutrophil engraftment before death will be censored at the date of death. The median and 95% confidence intervals will be provided. (NCT01303965)
Timeframe: Transplant (Day 0) through 1 year post transplant

Interventiondays (Median)
Phase II11

[back to top] [back to top]

Phase II - Percent of Patients With Acute Graft Versus Host Disease (GvHD)

"Percent of patients and the 95% Binomial Confidence interval who had any stage I-IV acute GvHD based on the modified Keystone Grading Scale for skin, liver and gastrointestinal symptoms for patients in Phase II. Zero means no acute GvHD was reported, and higher stages are worse outcomes (range of 0-4).~For skin: 0=no rash; 1=erthematous macular rash over <25% body surface; 2=over 25-50% of body surface; 4=bullae, exfoliation ulcerative dermatitis.~For liver (bilirubin (mg/dL)): 0= <2.0; 1= 2-<2.9; 3= 3-<5.9; 4= >=15 . For gut changes (diarrhea[ml/day]): 0=none; 1= >500-1000; 2= >1000-1500; 3= >1500; 4=severe abdominal pain with or without ileus.~Overall grade 0: Skin=0; liver=0; gut changes=0. Overall grade 1: Skin with 1 or 2; liver=0; gut changes=0. Overall grade 2: Skin with 1, 2, or 3; liver=1; gut changes=1. Overall grade 3: Skin with 2 or 3; liver with 2 or 3; gut changes with 2 or 3. Overall grade 4: Patients with grade 4 toxicity in any organ system." (NCT01303965)
Timeframe: Day 0 through 1 year post transplantation

Interventionpercentage of participants (Number)
Phase II36.4

[back to top]

Phase II - Percent of Patients With Chronic Graft Versus Host Disease (GvHD)

Percent of patients and the 95% Binomial Confidence interval who had any chronic GvHD reported based on Filipovich et al. consensus document (BB&MT 2005) and Akpek et al. chronic GvHD grading system (Blood 2003) for patients in Phase II. (NCT01303965)
Timeframe: Transplant (Day 0) through 1 year post-transplant

Interventionpercentage of participants (Number)
Phase II18.2

[back to top] [back to top]

Phase II: Percent of Patients Alive and Free of Progression at 12 Months Following Transplant

Percent of patients and the 95% Binomial Confidence interval who were alive and free of progression at 12 months following transplant for the patients in Phase II. Progression will be based on International Myeloma Working Group criteria where patients may meet any one of the following criteria - increase of 25% or more in serum or urine M-protein from baseline, Serum M-protein and/or the absolute increase must be >=0.5 g/dl, Urine M-protein and/or absolute increase must be >=200 mg/24 hours, development of new bone lesions or soft tissue plasmacyomas or definite increase in the size of existing bone lesions or soft tissue plasmacyomas, or development of hypercalcemia (corrected serum Ca++>11.5 mg/dl) that can be attributed solely to plasma cell proliferative disease. (NCT01303965)
Timeframe: Transplant (Day 0) through 1 year post-transplant

Interventionpercentage of participants (Number)
Phase II18.2

[back to top]

Progression Free Survival (PFS) Rate at Specific Time-points

PFS rate is defined as the percentage of participants experiencing PFS at the defined time-points. (NCT01335399)
Timeframe: From randomization to the specified time-point (up to 5 years)

,
InterventionPercent of participants (Number)
1 year2 year3 year4 year5 year
E-Ld Cohort0.770.590.460.360.26
Ld Cohort0.760.550.410.330.25

[back to top]

Mean Change From Baseline of Pain Severity Score and Pain Interference Score

"Pain severity (sensory dimension) and pain interference (reactive dimension, assessing the degree to which pain interferes with function) are measured using the Brief Pain Inventory- Short Form (BPI-SF).~BPI-SF numeric rating scale goes from 0 (No pain) to 10 (Pain as bad as you can imagine)." (NCT01335399)
Timeframe: From Baseline to End of Treatment (approximately 8 years)

,
InterventionRating score (Mean)
Pain SeverityPain Interference
E-Ld Cohort0.020.33
Ld Cohort-0.25-0.18

[back to top]

Progression-Free Survival (PFS)

"PFS is defined as the time from randomization to the date of the first documented tumor progression (as determined by the Independent Review Committee (IRC)) or death due to any cause.~The IRC conducted a blinded, independent review of the tumor assessments based on the European Group for Blood and Bone Marrow Transplant (EBMT) criteria.~Censoring rules applied:~Participants receiving subsequent systemic anti-myeloma therapy prior to documented progression were censored at the date of the last adequate tumor assessment prior to new therapy.~Participants who had an event (progression or death) > 10 weeks after their last tumor assessment were censored at their last adequate tumor assessment prior to the event.~Participants without progression or death (and not receiving subsequent therapy prior to progression) were censored at their last adequate tumor assessment.~Participants without any post-baseline tumor assessments were censored on the date of randomization" (NCT01335399)
Timeframe: From randomization to date of first documented tumor progression or death due to any cause (up to 8 years)

InterventionMonths (Median)
E-Ld Cohort31.38
Ld Cohort29.47

[back to top]

Overall Survival (OS)

"Survival is defined as the time from randomization to the date of death. A participant who did not die had his or her survival duration censored at the date of last contact ('last known date alive)." (NCT01335399)
Timeframe: From randomization to the date of death (up to 8 years)

InterventionMonths (Median)
E-Ld Cohort60.42
Ld Cohort57.56

[back to top]

Objective Response Rate (ORR)

ORR is defined as the percentage of participants with objective response among all randomized subjects. Participants with an objective response are those participants experiencing a partial response (PR) or better, based on Independent Review Committee (IRC) assessment, as per EBMT criteria. (NCT01335399)
Timeframe: From randomization to primary completion date (approximately 8 years)

InterventionPercent of Participants (Number)
E-Ld Cohort82.9
Ld Cohort79.4

[back to top]

Maximum Tolerated Dose (MTD) of Lenalidomide

MTD was determined by testing planned increasing doses up to 25 mg daily dose on days 1-14, starting at 10mg. MTD reflects the highest dose of drug that did not cause a Dose-Limiting Toxicity (DLT) in > 33% of participants. DLTs were defined as any lenalidomide-related Common Terminology Criteria for Adverse Events Version 4.0 (CTCAE 4.0) Grade 3 or 4 adverse events (NCT01342172)
Timeframe: after 1 cycle (each cycle is 21 days)

Interventionmg (Number)
Lenalidomide10

[back to top]

Best Overall Response

"Best Overall Response to evaluate lenalidomide as maintenance treatment in patients achieving an objective response of either complete response or partial response following completion of 6 cycles of combination therapy.~Complete Response (CR) - CR of target lesions and no new lesions Partial Response (PR) -PR of target lesions and no new lesions Stable Disease (SD) - SD of target lesions and no new lesions Progression Disease (PD) - any status of target lesions and new lesions" (NCT01342172)
Timeframe: 168 days

InterventionParticipants (Count of Participants)
CRPR
Lenalidomide12

[back to top]

Number of Grade >=3 Adverse Events

Number of grade >=3 adverse events to assess the safety of combination therapy with gemcitabine, cisplatin plus lenalidomide as determined by the frequency and severity of adverse events as per the NCI Common Terminology for Adverse Events (CTCAE) version 4.0. (NCT01342172)
Timeframe: Day 1 and Day 8 of each treatment cycle; 21 days after the last dose of Lenalidomide

Interventionevents (Number)
Grade 3Grade 4
Lenalidomide287

[back to top]

The Objective Response Rate to Treatment With Gemcitabine, Cisplatin, Plus Lenalidomide

"The objective response rate as determined by Response Evaluation Criteria in Solid Tumors (RECIST).~Complete Response (CR) Disappearance of all target lesions for a period of at least one month.~Partial Response (PR) At least a 30% decrease in the sum of the longest diameter of measures lesions (target lesions), taking as reference the baseline sum of the longest diameter.~Stable Disease (NR/SD) Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum of the longest diameter since the treatment started.~Progressive Disease (PD) A 20% or greater increase in the sum of the longest diameter of measured lesions (target lesions), taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions" (NCT01342172)
Timeframe: After 2 cycles (a cycle is 21 days)

InterventionParticipants (Count of Participants)
CRPRSDPDnot evaluable
Lenalidomide12321

[back to top]

Maximum Tolerated Dose of CEP-18770

MTD was based on the assessment of dose-limiting toxicity (DLT) during cycle 1 only and was defined as the highest dose at which fewer than one-third of participants in a cohort experience DLT. A DLT was defined as any of the following drug-related toxicities occurring during Cycle 1: Hematologic adverse events (AEs) (Grade 4 hematologic AEs, Grade 3 hematologic AEs with sequelae); Grade 3 nonhematologic AEs; Neuropathy (Grade 2 neuropathy, Grade 1 neuropathy with pain, worsening grade of neuropathy or new symptoms of pain associated with neuropathy); Any other toxicity that, in the judgment of the principal investigator, was a DLT; If a participant cannot receive 75% of the planned dose for any of the 3 agents (missing >1 dose of CEP-18770, or >5 doses of lenalidomide, or >1 dose of dexamethasone [either consecutively or separately]), due to a drug-related AE, the event was considered a DLT, even if the grade of toxicity was lower than specified DLT determination as described above. (NCT01348919)
Timeframe: Cycle 1 (28 days)

Interventionmg/m^2 (Number)
Overall Population1.8

[back to top]

Duration of Response (DOR) for Participants Treated With CEP-18770 at the MTD, as Assessed Using IMWG Criteria

DOR was defined as the time interval from the date of first response (sCR, CR, VGPR, or PR) to the date of disease progression. sCR, CR, VGPR, and PR as defined in outcome measure 1. Disease progression was defined as any 1 or more of the following: Increase of 25% or more from lowest response level in any 1 or more of the following: - serum M-component (absolute increase must be ≥0.5 grams [g]/deciliter [dL]), - urine M-component (absolute increase must be ≥200 mg/24 hours), bone marrow plasma cell percentage ≥10%; definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; and development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that could be attributed solely to the plasma cell proliferation disorder. (NCT01348919)
Timeframe: From the date of first response to the date of disease progression (up to approximately 1.5 years)

Interventionmonths (Median)
CEP-18770 Dose BNA

[back to top]

Overall Response Rate (ORR) in Participants Treated at the (Maximum Tolerated Dose) MTD, as Assessed Using International Myeloma Working Group (IMWG) Criteria

The ORR is defined as percentage of participants who achieve a best response of stringent complete response (sCR), complete response (CR), very good partial response (VGPR), or partial response (PR) during the study. sCR: negative immunofixation in serum and urine; disappearance of any soft tissue plasmacytomas; < 5% plasma cells in bone marrow; normal free light chain (FLC) ratio; and absence of clonal cells in bone marrow. CR: negative immunofixation in serum and urine; disappearance of any soft tissue plasmacytomas; and <5% plasma cells in bone marrow. VGPR: serum and urine M-protein detectable by immunofixation but not on electrophoresis; 90% or greater reduction in serum M-protein level and urine M-protein level less than 100 milligrams (mg)/24 hours. PR: ≥50% reduction in serum M-protein level; ≥90% reduction in 24-hour urinary M-protein level or reduction to less than 200 mg per 24 hours; and ≥50% reduction in the size of any soft tissue plasmacytomas present at baseline. (NCT01348919)
Timeframe: From the first administration of CEP-18770 up to approximately 1.5 years

Interventionpercentage of participants (Number)
CEP-18770 Dose B40

[back to top]

Number of Participants With Adverse Events (AEs)

An AE was defined as any untoward medical occurrence in a participant who received study drug without regard to possibility of causal relationship. SAEs were defined as death, a life-threatening AE, inpatient hospitalization or prolongation of existing hospitalization, persistent or significant disability or incapacity, a congenital anomaly or birth defect, or an important medical event that jeopardized participant and required medical intervention to prevent 1 of the outcomes listed in this definition. A summary of other non-serious AEs and all serious AEs, regardless of causality is located in Reported AE section. (NCT01348919)
Timeframe: From the first administration of CEP-18770 up to approximately 1.5 years

InterventionParticipants (Count of Participants)
CEP-18770 Dose A3
CEP-18770 Dose B5
CEP-18770 Dose C3

[back to top]

Time to Reach Cmax (Tmax) of CEP-18770

(NCT01348919)
Timeframe: Before and immediately after end of infusion (EOI) and at approximately 2, 4, and 8 hours after the EOI on Days 1 and 15 of Cycle 1

,,
Interventionhours (Median)
Day 1Day 15
CEP-18770 Dose A0.070.07
CEP-18770 Dose B0.080.08
CEP-18770 Dose C0.100.08

[back to top]

Maximum Observed Plasma Concentration (Cmax) of CEP-18770

(NCT01348919)
Timeframe: Before and immediately after end of infusion (EOI) and at approximately 2, 4, and 8 hours after the EOI on Days 1 and 15 of Cycle 1

,,
Interventionnanograms (ng)/ milliliter (mL) (Mean)
Day 1Day 15
CEP-18770 Dose A579.49639.59
CEP-18770 Dose B663.00628.07
CEP-18770 Dose C876.97977.09

[back to top]

Area Under the Plasma Concentration-Time Curve From Time 0 to t (AUC0-t) of CEP-18770

(NCT01348919)
Timeframe: Before and immediately after end of infusion (EOI) and at approximately 2, 4, and 8 hours after the EOI on Days 1 and 15 of Cycle 1

,,
Interventionng*hours/mL (Mean)
Day 1Day 15
CEP-18770 Dose A6731595
CEP-18770 Dose B13712071
CEP-18770 Dose C15272266

[back to top]

Time to Progression (TTP) for Participants Treated With CEP-18770 at the MTD, as Assessed Using IMWG Criteria

TTP was defined as the time interval from the date of first dose to the date of disease progression. Disease progression was defined as any 1 or more of the following: Increase of 25% or more from lowest response level in any 1 or more of the following: - serum M-component (absolute increase must be ≥0.5 g/dL), - urine M-component (absolute increase must be ≥200 mg/24 hours), bone marrow plasma cell percentage ≥10%; definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; and development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that could be attributed solely to the plasma cell proliferation disorder. (NCT01348919)
Timeframe: From the date of first dose of study drug to the date of disease progression (up to approximately 1.5 years)

Interventionmonths (Median)
CEP-18770 Dose BNA

[back to top]

Grade 3-4 Toxicity

Number of participants who experienced grade 3-4 toxicity as per CTCAE 4.0. (NCT01349569)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Myeloma Vaccine, Prevnar-13 Vaccine, & Lenalidomide0

[back to top]

Response Conversion Rate

Number of participants who converted from near complete remission (nCR) to complete remission (CR) as measured by the International Myeloma Working Group Uniform Response Criteria. Near complete remission is defined as negative serum and urine electrophoresis, < 5% plasma cells in the bone marrow, and positive serum and/or urine immunofixation. Complete response is defined as negative serum and urine immunofixation and a bone marrow aspirate with < 5% plasma cells. (NCT01349569)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Myeloma Vaccine, Prevnar-13 Vaccine, & Lenalidomide8

[back to top]

Time to Response

"Median time for conversion of response from near complete remission (nCR) to complete remission (CR) as measured by the International Myeloma Working Group Uniform Response Criteria. Near complete remission is defined as negative serum and urine electrophoresis, < 5% plasma cells in the bone marrow, and positive serum and/or urine immunofixation. Complete response is defined as negative serum and urine immunofixation and a bone marrow aspirate with < 5% plasma cells.~as measured by immunofixation converting from positive to negative." (NCT01349569)
Timeframe: Up to 4 years

Interventionmonths (Median)
Myeloma Vaccine, Prevnar-13 Vaccine, & Lenalidomide11.7

[back to top]

Tumor-specific Immunity as Assessed by Percentage of CD3+/CSFSE-low/IFN-gamma+ Cells

Immunity is measured by the percentage of CD3+/CSFSE-low/IFN-gamma+ cells. A positive result for a given participant is defined as greater than two standard deviations above that participant's baseline. The data are presented as three groups because the responses were analyzed separately, but all participants were part of the single study arm as represented by the remainder of the record. GVAX-specific immune response and Prevnar-specific immune response was assessed in the same patient by using GVAX and Prevnar-specific co-markers. (NCT01349569)
Timeframe: Baseline, Cycle 3 Day 14, end of study (up to 1 year)

,,
Interventionpercentage of cells (Mean)
Complete Response (CR) patients at BaselineCR patients at Cycle 3 Day 14CR patients at end of StudyProgressive Disease (PD) patients at BaselinePD patients at Cycle 3 Day 14PD patients at end of study
GVAX Vaccine0.3613.313.70.455.70.21
Pre-vaccine (Baseline)1.27.36.30.431.70.4
Prevnar Vaccine04.543.6001.7

[back to top]

Time-to-next Treatment

(NCT01355705)
Timeframe: 9 months

Interventiondays (Median)
Amrubicin + Lenalidomide + Dexamethasone92

[back to top]

Response Rates After Amrubicin + Lenalidomide + Dexamethasone, Per International Myeloma Working Group Uniform Response Criteria

"Modified International Myeloma Working Group Uniform Response Criteria:~Complete (CR)=~Negative for monoclonal protein (MP) in urine (U) and serum (S) +~No tissue plasmacytomas (PC) +~<5% plasma cells (PCs) in marrow (M)~Stringent CR (sCR)= CR with normal light chain ratio+ no PCs in M~Near CR (nCR)= CR, except MP persists in U and S~Partial (PR)= S MP ≤50%, + U MP ≤90% or <200 mg/24 hours (hr)~Very Good PR (VGPR)= in S MP ≤90%, + U MP <100 mg/24 hr~Minimal (MR)=~S MP ≤51-75%, +~If light chain is excreted, reduced 50-89%/24 hr that is also >200 mg/24 hr, +~No increase in lytic bone lesions~Progressive disease (PD)= any of:~S MP ≥125% and/or ≥+0.5 g/dL,~U MP ≥125% and/or ≥+200 mg/24 hr~New or increased bone lesions/PC~S calcium >11.5 mg/dL (attributed to increased PCs)~PD after CR/sCR=~Reappearance of S or U MP~≥5% clonal PCs in M~New PC, lytic bone lesions, hypercalcemia~Stable Disease (SD)= Not CR, VGPR, MR, PR, or PD" (NCT01355705)
Timeframe: 12 weeks

Interventionpercentage of participants (Number)
Complete Response (CR) rateVery Good Partial Response (VGPR) RateTotal CR + VGPRPartial Response (PR) RateOverall Response Rate (ORR = CR + VGPR + PR)
Amrubicin + Lenalidomide + Dexamethasone07.67.615.423

[back to top]

Duration of Response (DOR)

(NCT01355705)
Timeframe: 140 days

Interventiondays (Median)
Amrubicin + Lenalidomide + Dexamethasone133

[back to top]

Progression-free Survival (PFS)

"Progression-free survival (PFS) is alive and free from progression, per the modified International Myeloma Working Group Uniform Response Criteria, defined as any of:~Serum monoclonal protein ≥ 125% baseline and/or ≥ +0.5 g/dL from baseline,~Urine monoclonal protein ≥ 125% baseline and/or ≥ +200 mg/24 hour from baseline~New or increased bone lesions or plasmacytomas~Serum calcium > 11.5 mg/dL (attributed to increased plasma cells)" (NCT01355705)
Timeframe: 9 months

Interventiondays (Median)
Amrubicin + Lenalidomide + Dexamethasone96

[back to top]

Percentage of Participants Alive at One Year

Defined as the percentage of participants who survived at one year (NCT01358734)
Timeframe: Up to 12 months

InterventionPercentage of participants (Number)
Lenalidomide21.4
Azacitidine Plus Lenalidomide43.9
Azacitidine52.3

[back to top]

Kaplan Meier Estimates for One Year Survival

One-year survival rate was defined as all deaths within one year from the date of randomization. All others censored at the at year 1 or date of discontinuation (NCT01358734)
Timeframe: Up to 24 months

Interventionmonths (Median)
Lenalidomide3.00
Azacitidine Plus Lenalidomide9.61
Azacitidine13.67

[back to top] [back to top]

Number of Participants With a Second Primary Malignancy

Second primary malignancies were monitored as events of interest and reported as serious adverse events regardless of the treatment arm the participant was enrolled in. (NCT01358734)
Timeframe: From randomization of the last participant up to a minimum of 4 years following discontinuation

InterventionParticipants (Number)
Lenalidomide0
Azacitidine Plus Lenalidomide0
Azacitidine3

[back to top]

Overall Survival

Overall Survival reported at the end of the study are for those participants who were alive at the end of the study (NCT01358734)
Timeframe: From date of randomization until the date of the first documented date of progression or date of death of any cause; the overall median follow-up for survivng participants was 4.1 months (range 0.2 to 54.8 months)

Interventionmonths (Median)
Lenalidomide0.2
Azacitidine Plus Lenalidomide7.1
Azacitidine4.1

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAE)

TEAEs were defined as those events that started on or after the first day of study drug up until 28 days after the last dose of study drug; Serious AE (SAE) = any AE which results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability; is a congenital anomaly/birth defect; constitutes an important medical event. Severity of AEs were graded based upon the participants symptoms according to the Common Terminology Criteria for Adverse Events (CTCAE, Version 4.0); and according to the scale: Grade (Gr) 1 = Mild - transient or mild discomfort; no medical intervention required; Grade 2 = Moderate - mild to moderate limitation in activity; Grade 3 = Severe; Grade 4 = Life threatening; Grade 5 = Death (NCT01358734)
Timeframe: From the first dose of study drug up to 28 days after the last dose of study drug; up to 15 May 2018

,,
Interventionparticipants (Number)
Any TEAE≥1 TEAE related to study drug≥1 TEAE CTCAE Grade 3 or 4 TEAE≥1 TEAE CTCAE Gr 3 or 4 TEAE related to study drug≥1 TEAE CTCAE Grade 5≥1 Serious TEAE≥1 Serious TEAE related to study drug≥1TEAE leading to discontinuation of study drug≥1TEAE leading to dose reduction of study drug≥1TEAE leading to dose interruption of study drug
Azacitidine3230291852574210
Azacitidine Plus Lenalidomide3835342510291612821
Lenalidomide1413141151310608

[back to top]

Progression-free Survival (PFS)

(NCT01373229)
Timeframe: time from day 1 of treatment to disease progression, death, or 2 years, whichever comes first

Interventionmonths (Mean)
Lenalidomide + Plerixafor+ Rituximab11

[back to top]

Overall Survival (OS)

(NCT01373229)
Timeframe: the time from day 1 of treatment to death or 2 years, whichever comes first

Interventionmonths (Mean)
Lenalidomide + Plerixafor+ Rituximab5.5

[back to top]

Overall Response (Complete Response/Partial Response)

"NCI 96 Response Criteria~CR (Complete Response):~Lymphadenopathy = none > 1.5cm Hepatomegaly = none Splenomegaly = none Blood lymphocytes = <4000 per microliter Marrow = normocellular, <30% lymphocytes, no B-lymphoid nodules. Platelet count = >100,000 per microliter Hemoglobin = >11.0 grams per deciliter Neutrophils = >1500 per microliter~PR (Partial Response):~Lymphadenopathy = Decrease >/= 50% Hepatomegaly = Decrease >/= 50% Splenomegaly = Decrease >/= 50% Blood lymphocytes = Decrease >/= 50% from baseline Marrow = 50% reduction in marrow infiltrate or B-lymphoid nodules. Platelet count = >100,000 per microliter or increase >/= 50% over baseline Hemoglobin = >11.0 grams per deciliter or increase >/= 50% over baseline Neutrophils = >1500 per microliter or increase >/= 50% over baseline" (NCT01373229)
Timeframe: at the end of 4 months of combination treatment and at 2 months after completion of therapy

InterventionParticipants (Count of Participants)
Lenalidomide + Plerixafor+ Rituximab0

[back to top]

Maximum Tolerated Dose

(NCT01373229)
Timeframe: 4-16 months

Interventionmg/kg (Number)
Lenalidomide + Plerixafor+ Rituximab0.24

[back to top]

Treatment Emergent Adverse Events (AEs) or Serious Adverse Events (SAEs)

Number of participants with treatment emergent AEs or SAEs per category. SAEs will be specifically labeled as such. Participants were assessed at monthly intervals (corresponding to Revlimid™ refill points for adverse events), classified by Common Terminology Criteria for Adverse Events (CTCAE) version 4.0, and these were tabulated. (NCT01373294)
Timeframe: Duration of study treatment and follow-up - average of 12 months

,
Interventionparticipants (Number)
HematuriaUrinary frequencyUrinary urgencyUrinary tract painBladder spasmFatigueEdema faceEdema limbsFeverPainNauseaErythrodermaPruritusRash acneiformRash maculo-papularUrinary tract infectionPharyngitisPlatelet count decreasedSAE: Grade 3 Myocardial infarctionHeadachePeripheral sensory neuropathy
A: Combination Arm633217111121111211111
B: Control Arm101000000000000000000

[back to top]

Arm A: Progression Free Survival (PFS)

The 1-year progression free/ recurrence free/ bladder-intact survival was tabulated for the experimental arm for a median follow-up period of 369 days. The progression free/ recurrence free/ bladder-intact survival is defined as the time from start of study treatment to first documentation of objective tumor progression, recurrence, bladder resection or irradiation or to death due to any cause, whichever comes first. PFS data was not collected for participants in the Arm B: Control because too few participants were enrolled in Arm B to conduct the planned per Arm comparison (NCT01373294)
Timeframe: 1 year

Interventionparticipants (Number)
A: Combination Arm8

[back to top]

Duration of Response

Median length of response in months. (NCT01374217)
Timeframe: Up to 6 months

Interventionmonths (Median)
Tadalafil2.3

[back to top]

Quality of Life Scores

Median change in symptom scores. Scale is the EORTC QLQ-C30. There are three domains: symptom scale (score range 7-14); past week (score range 21-82); and global health status (score range 2-14). Higher or increasing scores mean worse outcomes; lower or decreasing scores mean better outcomes. (NCT01374217)
Timeframe: 3 months (M3) and 6 months (M6)

Interventionchange in score on a scale (Median)
Symptoms (M3)Past week (M3)Global health status (M3)Symptoms (M6)Past week (M6)Global health status (M6)
Tadalafil05-1.516-1

[back to top]

Time to Progression

Median time to progression of disease in days. (NCT01374217)
Timeframe: Up to 71 days

Interventiondays (Median)
Tadalafil48

[back to top]

Response Rate

Percentage of participants who responded to the addition of tadalafil. Response is defined as a complete remission (CR), very good partial remission (VGPR), partial remission (PR), or stable disease (SD) by International Uniform Response criteria. (NCT01374217)
Timeframe: Up to 6 months

InterventionParticipants (Count of Participants)
Tadalafil5

[back to top]

Participants With Objective Response

To determine the efficacy of the combination of Ruxolitinib + Lenalidomide in patients with Myelofibrosis (MF). Objective response rate equals Complete and Partial Response, and Clinical Improvement as defined by International Working Group for Myelofibrosis Research and Treatment (IWG-MRT). Objective response rate (ORR), defined as a clinical improvement (CI), partial remission (PR), and complete remission (CR) according to the International Working Group (IWG) Criteria. Complete remission (CR): bone marrow blasts <5%, hemoglobin >/= 10, absolute neutrophil count (ANC) >/= 1000, platelets >/= 100, <2% immature myeloid cell, spleen and liver not palpable. Partial Response (PR): CR plus one or more of the following: ANC >/= 1000, decreased platelets by 50%, hemoglobin >/= 8.5 but < 10, <2% immature myeloid cells. Clinical improvement (CI): hemoglobin increase of 2g/dl, transfusion independence or reduction splenomegaly and/or hepatomegaly >/= 50%, >/=50% reduction in MPN-SAF TSS (NCT01375140)
Timeframe: 3 cycles (28 days each) up to 3 months

InterventionParticipants (Count of Participants)
Ruxolitinib + Lenalidomide7

[back to top]

Serious Adverse Events

"Type, frequency, severity and timing of adverse events and their relationship to combination therapy with lenalidomide plus dexamethasone.~SAE Grade 3 indicates a severe or medically significant but not immediately life threatening; hospitalization or prolongation of hospitalization; disabling; limiting self care ADL SAE Grade 4 indicates a life-threatening consequences; urgent intervention indicated.~SAE Grade 5 Death related to AE." (NCT01380106)
Timeframe: Data was collected for each subject for the duration of the participation in the study, ranging between 75 to 475 days.

,
InterventionEvents (Number)
SAE Grade 3SAE Grade 4SAE Grade 5
Lenalidomide 15mg2310
Lenalidomide 25mg2341

[back to top]

Duration Until Best Response (at Least MR or Minimal Response)

Number of days between the first day of the first cycle to best M-protein response, at least Minimal Response or higher (Partial Response, Very Good Partial Response, near Complete Response, Complete Response). (NCT01380106)
Timeframe: Data was collected for each subject for the duration of the participation in the study, ranging between 75 to 475 days.

InterventionDays (Mean)
Lenalidomide 25mg280.4
Lenalidomide 15mg145.5

[back to top]

Phase 2: Percentage of Participants With Very Good Partial Response (VGPR)

VGPR as per IMWG criteria is serum and urine M-protein detectable by immunofixation but not on electrophoresis or >=90% reduction in serum M-protein plus urine M-protein level <100 mg per 24 hours. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg37

[back to top]

Phase 2: Time to Response

Time to first response is defined as the time from the date of first dose of study treatment to the date of the first documentation of a confirmed response (PR or better) in a participant who responded + 1 day. PR as per IMWG criteria is 50% reduction of serum M-protein and reduction in 24-h urinary M-protein by 90% or to <200 mg per 24 hours. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionmonths (Median)
Phase 2: Ixazomib 3 mg0.72

[back to top]

Progression Free Survival (PFS)

PFS was defined as the time from the date of first dose of study treatment to the date of first documentation of progressive disease or to death due to any cause, whichever occurred first plus 1. PD: >=25% increase from lowest value in:serum/urine M-component; difference between involved,uninvolved FLC levels; bone marrow plasma cell percent; new bone lesions/soft tissue plasmacytomas development/existing bone lesions/soft tissue plasmacytomas size rise; hypercalcaemia development. SD: not meeting criteria for CR, VGPR, PR, or PD. Participants who received ASCT or an alternate anticancer therapy were censored at the last response assessment that was SD or better before initiation of therapy. Participants without a response assessment were censored at the date of first dose. PFS was analyzed using standard survival analysis techniques based on Kaplan-Meier estimates. (NCT01383928)
Timeframe: Baseline up to a follow-up of 62.1 months

Interventionmonths (Median)
Ixazomib 3 mg29.7

[back to top]

Time to Disease Progression (TTP)

Time to progression was defined as the time from the date of first dose of study treatment to the date of first documentation of PD + 1 day. Participants that did not experience PD will be censored at the last response assessment that is SD or better. PD: >=25% increase from lowest value in:serum/urine M-component; difference between involved, uninvolved FLC levels; bone marrow plasma cell percent; new bone lesions/soft tissue plasmacytomas development/existing bone lesions/soft tissue plasmacytomas size rise; hypercalcaemia development. SD: not meeting criteria for CR, VGPR, PR, or PD. Participants that received Autologous Stem Cell Transplantation (ASCT) or an alternate cancer therapy were also be censored at the last response assessment that is, SD or better prior to initiation of therapy. Participants without response assessment will be censored at the date of first dose. TTP was analyzed using standard survival analysis techniques based on Kaplan-Meier estimates. (NCT01383928)
Timeframe: Baseline up to a follow-up of 62.1 months

Interventionmonths (Median)
Ixazomib 3 mg29.7

[back to top]

Phase 1: AUC(0-72): Area Under the Plasma Concentration-Time Curve From Time 0 to 72 Hours Postdose for Ixazomib

AUC(0-72) is a measure of the area under the plasma concentration time-curve from time zero to 72 hours post-dose for ixazomib. (NCT01383928)
Timeframe: Cycle 1, Days 1 and 11

,
Interventionhour*nanogram per milliliter (hr*ng/mL) (Geometric Mean)
Day 1Day 11
Phase 1: Ixazomib 3 mg315.4501105.44
Phase 1: Ixazomib 3.7 mg284.5761023.52

[back to top]

Phase 1: Number of Participants With Clinically Significant Change From Baseline in Vital Signs

Vital signs included body temperature, blood pressure and heart rate. (NCT01383928)
Timeframe: Baseline up to 30 days after last dose of study drug (Up to Cycle 83 - approximately 2067 days)

,
InterventionParticipants (Count of Participants)
PyrexiaBradycardiaOrthostatic hypotensionHypotension
Phase 1: Ixazomib 3 mg1101
Phase 1: Ixazomib 3.7 mg3011

[back to top]

Phase 1: Number of Participants With Markedly Abnormal Laboratory Values Reported as Treatment Emergent Adverse Events (TEAEs)

Laboratory tests included chemistry, hematology and urinalysis. Abnormal laboratory value was assessed as an AE if the value lead to discontinuation or delay in treatment, dose modification, therapeutic intervention, or was considered by the investigator to be a clinically significant change from baseline. A treatment-emergent adverse event (TEAE) is defined as an adverse event with an onset that occurs after receiving study drug. (NCT01383928)
Timeframe: Baseline up to 30 days after last dose of study drug (Up to Cycle 83 - approximately 2067 days)

,
InterventionParticipants (Count of Participants)
Alanine aminotransferase increasedAspartate aminotransferase increasedBlood creatinine increasedShift to the leftPlatelet count decreasedBlood bicarbonate decreasedAnaemiaNeutropeniaThrombocytopeniaEosinophiliaHypokalaemiaHyperkalaemiaHyperglycaemiaHyponatraemiaHypomagnesaemiaHyperchloraemiaIron deficiency anaemia
Phase 1: Ixazomib 3 mg00201111101140011
Phase 1: Ixazomib 3.7 mg32010000110001200

[back to top] [back to top]

Phase 1: Percentage of Participants Experiencing 1 or More Treatment-Emergent Adverse Events (TEAEs) or Serious Adverse Events (SAEs)

An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. A SAE is an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; or congenital anomaly; or a medically important event. A TEAE is defined as an adverse event with an onset that occurs after receiving study drug. (NCT01383928)
Timeframe: Baseline up to 30 days after last dose of study drug (Up to Cycle 83 - approximately 2067 days)

,
Interventionpercentage of participants (Number)
Adverse EventSerious Adverse Event
Phase 1: Ixazomib 3 mg10071
Phase 1: Ixazomib 3.7 mg10029

[back to top]

Phase 1: Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Ixazomib

Tmax: Time to reach the first maximum plasma concentration (Cmax), equal to time (hours) to Cmax of ixazomib after administration, obtained directly from the plasma concentration-time curve. (NCT01383928)
Timeframe: Cycle 1, Days 1 and 11

,
Interventionhours (Median)
Day 1Day 11
Phase 1: Ixazomib 3 mg1.0351.030
Phase 1: Ixazomib 3.7 mg1.0000.984

[back to top]

Phase 2: Percentage of Participants With Complete Response (CR) and Very Good Partial Response (VGPR) After Cycles 4, 8, and 16

CR as per IMWG criteria is defined as negative immunofixation on the serum and urine and disappearance of soft tissue plasmacytomas and <5% plasma cells in bone marrow. VGPR as per IMWG criteria is defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hours. VGPR were applicable only to participants who had measurable disease defined by at least 1 of the following 3 measurements: Serum M-protein; Urine M-protein; Serum FLC assay. (NCT01383928)
Timeframe: Cycles 4, 8, and 16

Interventionpercentage of participants (Number)
After 4 cyclesAfter 8 cyclesAfter 16 cycles
Phase 2: Ixazomib 3 mg496492

[back to top]

Phase 2: Percentage of Participants With Stringent Complete Response (sCR)

sCR as per IMWG criteria is CR plus normal FLC ratio and absence of clonal cells in bone marrow. CR is negative immunofixation on serum and urine and disappearance of soft tissue plasmacytomas and <5% plasma cells in bone marrow. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg22

[back to top]

Phase 2: Percentage of Participants With Minimal Response (MR)

MR as per IMWG criteria is 25%-49% reduction in serum paraprotein and 50%-89% reduction in urine light chain excretion for 6 weeks. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg6

[back to top] [back to top]

Kaplan-Meier Estimate of Percentage of Participants Achieving Survival at Year 1

The Kaplan-Meier estimate reports the percentage of participants surviving at Year 1. (NCT01383928)
Timeframe: 1 year after the first dose of study treatment

Interventionpercentage of participants (Number)
Ixazomib 3 mg94

[back to top]

Overall Survival

Overall survival was measured as the time from the date of first dose of study treatment to the time of death plus 1 day. For participants who did not die, survival was censored at the date of last contact. Overall Survival was analyzed using standard survival analysis techniques based on Kaplan-Meier estimates. (NCT01383928)
Timeframe: Baseline up to a follow-up of 62.1 months

Interventionmonths (Median)
Ixazomib 3 mgNA

[back to top]

Phase 1: Maximum Tolerated Dose (MTD)

MTD was highest dose of ixazomib given with combination drugs, at which <=1 of 6 participants experienced dose-limiting toxicity (DLT) during Cycle 1 of Phase 1. DLT defined as any of following considered possibly related to therapy: Grade 4 neutropenia (absolute neutrophil count [ANC] <500 cell per cubic millimeter [cells/mm^3]) for >7 days; Grade 3 neutropenia with fever or infection; Grade 4 thrombocytopenia for >7 days; Grade 3 thrombocytopenia with clinically significant bleeding; platelet count <10,000/mm^3; Grade 2 peripheral neuropathy with pain or >=Grade 3 peripheral neuropathy; >=Grade 3 nausea/emesis, diarrhea controlled by supportive therapy; any >=Grade 3 nonhematologic toxicity except Grade 3 arthralgia/myalgia; or <1 week Grade 3 fatigue; delay in initiation of the subsequent therapy cycle by >14 days; <=80% lenalidomide doses administered due to other >=Grade 2 combination study drug-related nonhematologic toxicities requiring therapy discontinuation. (NCT01383928)
Timeframe: Cycle 1 (21 days)

Interventionmg (Number)
Phase 1: All Participants3.7

[back to top]

Phase 1: Percentage of Participants With Best Overall Response

CR as per International Myeloma Working Group (IMWG) uniform criteria is defined as negative immunofixation on the serum and urine and disappearance of soft tissue plasmacytomas and <5% plasma cells in bone marrow. Partial response(PR):>=50% reduction of serum M-protein,urinary M-protein by >=90%/to <200 mg/24 hr reduction.Near CR(nCR):positive immunofixation of serum/urine;soft tissue plasmacytomas disappearance;<=5% plasma cells in bone marrow. VGPR as per IMWG criteria is defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hours. VGPR was applicable only to participants who had measurable disease defined by at least 1 of the following 3 measurements: Serum M-protein greater than or equal to (>=)1 g/dL; Urine M-protein >=200 mg/24 hours; Serum FLC assay level >=10 mg/dL, provided serum FLC ratio was abnormal. (NCT01383928)
Timeframe: Baseline until end of study treatment (Up to treatment Cycle 83 - approximately 2037 days)

Interventionpercentage of participants (Number)
Phase 1: Ixazomib 3 mg92

[back to top]

Phase 1: Rac: Accumulation Ratio of Ixazomib

The accumulation ratio (Rac) was estimated as the ratio of AUC (0-72) on Day 11 to the AUC (0-72) on Day 1. AUC (0-72) is the area under the plasma concentration-time curve from time zero to 72 hours post-dose for ixazomib. (NCT01383928)
Timeframe: Cycle 1, Days 1 and 11

Interventionratio (Geometric Mean)
Phase 1: Ixazomib 3 mg3.785
Phase 1: Ixazomib 3.7 mg3.967

[back to top]

Phase 1: Cmax: Maximum Plasma Concentration for Ixazomib

Maximum observed plasma concentration (Cmax) is the peak plasma concentration of ixazomib, obtained directly from the plasma concentration-time curve. (NCT01383928)
Timeframe: Cycle 1, Days 1 and 11

,
Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Day 1Day 11
Phase 1: Ixazomib 3 mg33.51558.674
Phase 1: Ixazomib 3.7 mg46.94651.832

[back to top]

Phase 2: Duration of Response (DOR)

DOR was measured as the time from the date of first documentation of a confirmed response to the date of first documented PD. PD is defined as >=25% increase from lowest value in: serum/urine M-component; difference between involved, uninvolved FLC levels; bone marrow plasma cell percent; development of new bone lesions or soft tissue plasmacytomas development or increase in the size of existing bone lesions or soft tissue plasmacytomas; hypercalcaemia development. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionmonths (Median)
Phase 2: Ixazomib 3 mg29.0

[back to top]

Phase 2: Percentage of Participants Experiencing Serious Adverse Events

A SAE is an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; or congenital anomaly; or a medically important event. (NCT01383928)
Timeframe: Baseline up to 30 days after the last dose of study drug (approximately 1905 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg46

[back to top]

Phase 2: Percentage of Participants With Complete Response (CR)

CR as per IMWG criteria is negative immunofixation on serum and urine and disappearance of soft tissue plasmacytomas and <5% plasma cells in bone marrow. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg29

[back to top]

Phase 2: Percentage of Participants With Complete Response (CR) + Very Good Partial Response (VGPR)

CR as per International Myeloma Working Group (IMWG) uniform criteria is defined as negative immunofixation on the serum and urine and disappearance of soft tissue plasmacytomas and <5% plasma cells in bone marrow. VGPR as per IMWG criteria is defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hours. VGPR was applicable only to participants who had measurable disease defined by at least 1 of the following 3 measurements: Serum M-protein greater than or equal to (>=)1 g/dL; Urine M-protein >=200 mg/24 hours; Serum FLC assay level >=10 mg/dL, provided serum FLC ratio was abnormal. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg65

[back to top]

Phase 2: Percentage of Participants With Grade 3 or Higher Adverse Events

An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. As per Common Terminology Criteria for Adverse Events v4.0 (CTCAE), Grade 3 = AE with severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self-care activities of daily living. Grade 4 = AE with life-threatening consequences; urgent intervention indicated and Grade 5 = Death related to AE. (NCT01383928)
Timeframe: Baseline up to 30 days after the last dose of study drug (approximately 1905 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg74

[back to top]

Phase 2: Percentage of Participants With Near Complete Response (nCR)

nCR as per IMWG criteria is positive immunofixation analysis of serum or urine as the only evidence of disease; appearance of any soft tissue plasmacytomas and <=5% plasma cells in bone marrow. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg10

[back to top]

Phase 2: Percentage of Participants With Overall Response (CR+VGPR+PR)

Overall response is defined as CR, VGPR or PR based on IMWG Response Criteria for malignant lymphoma. CR: disappearance of all detectable clinical evidence of disease and disease-related symptoms if present before therapy. VGPR: serum, urine M-protein detectable by immunofixation but not on electrophoresis or >=90% reduction in serum M-protein+urine M-protein level <100 mg/24h. Partial response (PR) is a minimum of 50% decrease in sum of the product of the diameters of up to 6 of the largest dominant nodes or nodal masses and no increase in the size of other nodes. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg94

[back to top]

Phase 2: Percentage of Participants With Partial Response (PR)

PR as per IMWG criteria is 50% reduction of serum M-protein and reduction in 24-hr urinary M-protein by 90% or to <200 mg per 24 hours. (NCT01383928)
Timeframe: Baseline until end of treatment (Up to treatment Cycle 74 - approximately 1875 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg65

[back to top]

Phase 2: Percentage of Participants With Treatment-Emergent Adverse Events Resulting in Study Drug Discontinuation

An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. A TEAE is defined as an adverse event with an onset that occurs after receiving study drug. (NCT01383928)
Timeframe: Baseline up to 30 days after the last dose of study drug (approximately 1905 days)

Interventionpercentage of participants (Number)
Phase 2: Ixazomib 3 mg14

[back to top]

Number of Participants With Worst Toxicity Grade Chemistry Laboratory Tests

Sodium high (H) Gr 1:>ULN - 150; Gr 2: >150 - 155; Gr 3: >155 - 160; Gr 4: >160 mmol/L; Sodium low(L) Gr 1:ULN - 5.5; Gr 2: >5.5 - 6.0; Gr 3: > 6.0 - 7.0; Gr 4: >7.0 mmol/L; Potassium (L) Gr 1: ULN - 11.5, Gr2:>11.5 - 12.5, Gr3: 12.5 - 13.5, Gr4: >13.5. (NCT01393964)
Timeframe: From first dose (Day 1) to last dose plus 60 days (Assessed up to July 2016, approximately 54 months)

,,
Interventionparticipants (Number)
Sodium High Any GradeSodium High Grade 3-4Sodium Low Any GradeSodium Low Grade 3-4Potassium High Any GradePotassium High Grade 3-4Potassium Low Any GradePotassium Low Grade 3-4Bicarbonate Any GradeBicarbonate Grade 3-4Calcium High Any GradeCalcium High Grade 3-4Calcium Low Any GradeCalcium Low Grade 3-4Glucose High Any GradeGlucose High Grade 3-4Glucose Low Any GradeGlucose Low Grade 3-4
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants205043616031827310
Elotuzumab + LD in Normal Renal Function (NRF) Participants202120205010407310
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants306120528000619400

[back to top]

Number of Participants With Worst Toxicity Grade Hematology Laboratory Tests

National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE), version 3.0 was used to measure toxicity scale. Lower Limits of Normal (LLN). Hemoglobin Gr 1:NCT01393964)
Timeframe: From first dose (Day 1) to last dose plus 60 days (Assessed up to July 2016, approximately 54 months)

,,
Interventionparticipants (Number)
Hemoglobin Any GradeHemoglobin Grade 3-4Platelet Count Any GradePlatelet Count Grade 3-4Leukocytes Any GradeLeukocytes Grade 3-4Lymphocytes (Abs) Any GradeLymphocytes (Abs) Grade 3-4Neutrophil Count (Abs) Any GradeNeutrophil Count (Abs) Grade 3-4
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants9682629562
Elotuzumab + LD in Normal Renal Function (NRF) Participants8070838663
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants9282819951

[back to top]

Number of Participants With Worst Toxicity Grade Renal and Liver Function Laboratory Tests

NCI CTCAE, version 3.0 was used to measure toxicity scale. Lower Limits of Normal (LLN). Upper Limits of Normal (ULN). Alanine transaminase (ALT); Aspartate aminotransferase (AST); Alkaline phosphatase (ALP). ALT Grade (Gr)1:>1.0 to 2.5*ULN; Gr 2: >2.5 to 5.0*ULN; Gr 3: >5.0 to 20.0*ULN; Gr 4: >20.0*ULN. AST Gr 1: >1.0 to 2.5*ULN; Gr 2: >2.5 to 5.0*ULN; Gr 3: >5.0 to 20.0*ULN; Gr 4: >20.0*ULN. Total bilirubin Gr 1: >1.0 to 1.5*ULN; Gr 2: >1.5 to 3.0*ULN; Gr 3: >3.0 to 10..0*ULN; Gr 4: >10.0.0*ULN. ALP (U/L) Gr1:>1.0 to 2.5*ULN, Gr2:>2.5 to 5.0*ULN, Gr3:>5.0 to 20.0*ULN, Gr4:>20.0*ULN. Albumin (low) Gr 1:1 - 1.5*baseline (BL)to >ULN - 1.5*ULN; Gr 2: >1.5 - 3.0*BL to > 1.5 - 3.0*ULN; Gr 3: >3.0*BL to > 3.0 - 6.0*ULN; Gr 4: >6.0*ULN. (NCT01393964)
Timeframe: From first dose (Day 1) to last dose plus 60 days (Assessed up to July 2016, approximately 54 months)

,,
Interventionparticipants (Number)
ALP Any GradeALP Grade 3-4AST Any GradeAST Grade 3-4ALT Any GradeALT Grade 3-4Bilirubin Any GradeBilirubin Grade 3-4Creatinine Any GradeCreatinine Grade 3-4Albumin Any GradeAlbumin Grade 3-4
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants705050209970
Elotuzumab + LD in Normal Renal Function (NRF) Participants003020100020
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants302050109270

[back to top]

Geometric Mean Apparent Volume of Distribution (Vz) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method

The quantification of elotuzumab in human serum was performed using validated ELISA. Cycle 1, day 1 sample times for all participants: 0 hour (h) pre-dose, end of infusion, 30 minutes (min) post end of infusion, 2 h, 4 h , and 24 h post end of infusion. Trough samples were obtained in subsequent cycles and at 30 day and 60 day follow-up visits at end of treatment. ESRD had 2 additional samples: immediately prior to and immediately after dialysis. Vz was measured in mL per kilogram body weight (mL/kg). PK parameter renal function group assignment criteria differed slightly from the criteria for safety and efficacy analyses (specifically for the SRI group). PK criteria: All participants with at least one pretreatment value ≥ 90 mL/min were assigned to the NRF group. All those with at least one pretreatment value < 30 mL/min were assigned to the SRI group. All those with a screening diagnosis of ESRD, were assigned to the ESRD group. (NCT01393964)
Timeframe: Day 1 of Cycle 1 to 28 days post dose

InterventionmL/kg (Geometric Mean)
Elotuzumab + LD in Normal Renal Function (NRF) Participants59.4
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants54.6
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants61.2

[back to top]

Geometric Mean Maximum Observed Serum Concentration (Cmax) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method

The quantification of elotuzumab in human serum was performed using a validated Enzyme-linked immunoassay (ELISA). Cycle 1, day 1 sample times for all participants: 0 hour (h) pre-dose, end of infusion, 30 minutes (min) post end of infusion, 2 h, 4 h , and 24 h post end of infusion. Trough samples were obtained in subsequent cycles and at 30 day and 60 day follow-up visits at end of treatment. ESRD had 2 additional samples: immediately prior to and immediately after dialysis. Cmax was measured in micrograms per milliliter (µg/mL). Pharmacokinetic (PK) parameter renal function group assignment criteria differed slightly from the criteria for safety and efficacy analyses (specifically for the SRI group). PK criteria: All participants with at least one pretreatment value ≥ 90 mL/min were assigned to the NRF group. All those with at least one pretreatment value < 30 mL/min were assigned to the SRI group. All those with a screening diagnosis of ESRD, were assigned to the ESRD group. (NCT01393964)
Timeframe: Day 1 of Cycle 1 to 28 days post dose

Interventionµg/mL (Geometric Mean)
Elotuzumab + LD in Normal Renal Function (NRF) Participants217
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants226
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants218

[back to top]

Geometric Mean Total Body Clearance (CLT) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method

The quantification of elotuzumab in human serum was performed using validated ELISA. Cycle 1, day 1 sample times for all participants: 0 hour (h) pre-dose, end of infusion, 30 minutes (min) post end of infusion, 2 h, 4 h , and 24 h post end of infusion. Trough samples were obtained in subsequent cycles and at 30 day and 60 day follow-up visits at end of treatment. ESRD had 2 additional samples: immediately prior to and immediately after dialysis. CLT was measured in mL per hour per kilogram body weight (mL/h/kg). PK parameter renal function group assignment criteria differed slightly from the criteria for safety and efficacy analyses (specifically for the SRI group). PK criteria: All participants with at least one pretreatment value ≥ 90 mL/min were assigned to the NRF group. All those with at least one pretreatment value < 30 mL/min were assigned to the SRI group. All those with a screening diagnosis of ESRD, were assigned to the ESRD group. (NCT01393964)
Timeframe: Day 1 of Cycle 1 to 28 days post dose

InterventionmL/h/kg (Geometric Mean)
Elotuzumab + LD in Normal Renal Function (NRF) Participants0.215
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants0.166
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants0.195

[back to top]

Mean Terminal-phase Elimination Half-life (T-Half) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method

The quantification of elotuzumab in human serum was performed using validated ELISA. Cycle 1, day 1 sample times: 0 hour (h) pre-dose, end of infusion, 30 minutes (min) post end of infusion, 2 h, 4 h , and 24 h post end of infusion. Trough samples were obtained in subsequent cycles and at 30 day and 60 day follow-up visits at end of treatment. ESRD had 2 additional samples: immediately prior to and immediately after dialysis. T-Half was measured in hours (h). PK parameter renal function group assignment criteria differed slightly from the criteria for safety and efficacy analyses (specifically for the SRI group). PK criteria: All participants with at least one pretreatment value ≥ 90 mL/min were assigned to the NRF group. All those with at least one pretreatment value < 30 mL/min were assigned to the SRI group. All those with a screening diagnosis of ESRD, were assigned to the ESRD group. (NCT01393964)
Timeframe: Day 1 of Cycle 1 to 28 days post dose

Interventionh (Mean)
Elotuzumab + LD in Normal Renal Function (NRF) Participants204
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants237
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants218

[back to top]

Median Time to Maximal Concentration (Tmax) of Elotuzumab Following Cycle 1, Day 1 Dose Administration - Grouping by Cockcroft-Gault Creatinine Clearance Method

The quantification of elotuzumab in human serum was performed using validated ELISA. Cycle 1, day 1 sample times for all participants: 0 hour (h) pre-dose, end of infusion, 30 minutes (min) post end of infusion, 2 h, 4 h , and 24 h post end of infusion. Trough samples were obtained in subsequent cycles and at 30 day and 60 day follow-up visits at end of treatment. ESRD had 2 additional samples: immediately prior to and immediately after dialysis. Tmax was measured in hours (h). PK parameter renal function group assignment criteria differed slightly from the criteria for safety and efficacy analyses (specifically for the SRI group). PK criteria: All participants with at least one pretreatment value ≥ 90 mL/min were assigned to the NRF group. All those with at least one pretreatment value < 30 mL/min were assigned to the SRI group. All those with a screening diagnosis of ESRD, were assigned to the ESRD group. (NCT01393964)
Timeframe: Day 1 of Cycle 1 to 28 days post dose

Interventionh (Median)
Elotuzumab + LD in Normal Renal Function (NRF) Participants3.23
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants3.87
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants3.33

[back to top]

Geometric Mean Area Under Serum Concentration-Time Curve From Time Zero to Time of Last Quantifiable Concentration AUC(0-T) and From Time Zero Extrapolated to Infinite Time AUC(INF) of Elotuzumab Following Cycle 1, Day 1 - Grouping by C-G CrCl Method

The quantification of elotuzumab in human serum was performed using validated ELISA. Cycle 1, day 1 sample times for all participants: 0 hour (h) pre-dose, end of infusion, 30 minutes (min) post end of infusion, 2 h, 4 h , and 24 h post end of infusion. Trough samples were obtained in subsequent cycles and at 30 day and 60 day follow-up visits at end of treatment. ESRD participants had 2 additional sample times: immediately prior to and immediately after dialysis. AUC was measured in µg*h/mL. PK parameter renal function group assignment criteria differed slightly from the criteria for safety and efficacy analyses (specifically for the SRI group). PK criteria: All participants with at least one pretreatment value ≥ 90 mL/min were assigned to the NRF group. All those with at least one pretreatment value < 30 mL/min were assigned to the SRI group. All those with a screening diagnosis of ESRD, were assigned to the ESRD group (NCT01393964)
Timeframe: Day 1 of Cycle 1 to 28 days post dose

,,
Interventionµg*h/mL (Geometric Mean)
AUC (0-T)AUC (INF)
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants4593751227
Elotuzumab + LD in Normal Renal Function (NRF) Participants3955946401
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants5008060255

[back to top]

Number of Participants With Persistent Elotuzumab Anti-drug Antibodies (ADA) and Number of Participants ADA Positive at Cycle 2 Pre-dose.

Serum samples were evaluated for the presence of ADAs using a validated bridging electrochemiluminescence (ECL) immunoassay. Samples in: Cycle 1, Day 1 0 h (pre-dose), Cycle 2, Day 1(Study Day 29), 0 h (pre-dose; 672 h post-dose), Cycle 3, Day 1, 0 h and in cycle thereafter, at end of study/discontinuation, and at 30 and 60 day follow up visits post treatment. ADA Positive Participant: baseline negative with at least one ADA positive sample at any time after initiation of treatment or baseline positive with at least one ADA positive sample at any time after initiation of treatment with a titer 9-fold greater than the baseline; Persistent Positive: ADA positive at 2 or more sequential timepoints at least 12 weeks apart; Last Sample Positive: Not persistent positive and ADA Positive Sample in the last sampling timepoint; Other Positive: not persistent positive with ADA negative sample in the last sampling; ADA Negative: no ADA positive sample after the initiation of treatment. (NCT01393964)
Timeframe: From first dose (Day 1) to last dose plus 60 days, up to Primary Endpoint (June 2014), approximately 2 years

,,
Interventionparticipants (Number)
Baseline ADA PositiveOn-Study ADA PositivePositive at Cycle 2 pre-dosePersistent PositiveLast Sample PositiveOther PositiveOn-Study ADA Negative
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants0110105
Elotuzumab + LD in Normal Renal Function (NRF) Participants1220114
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants0100013

[back to top]

Number of Participants With Serious Adverse Events (SAEs), Adverse Events (AEs) Leading to Discontinuation, and Who Died

AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. SAE=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. (NCT01393964)
Timeframe: From first dose (Day 1) to last dose plus 60 days (Assessed up to July 2016, approximately 54 months)

,,
Interventionparticipants (Number)
DeathsAny SAEAEs Leading to Discontinuation
Elotuzumab + LD in End Stage Renal Disease (ESRD) Participants071
Elotuzumab + LD in Normal Renal Function (NRF) Participants031
Elotuzumab + LD in Severe Renal Impairment (SRI) Participants054

[back to top]

Time-to-Progression (TTP)

(NCT01401322)
Timeframe: 12 weeks

InterventionDays (Median)
Lenalidomide 50 mg/Day x 28 Days20

[back to top]

Rate of Minimal Residual Disease (MRD) by Flow Cytometry

Response is assessed by the International Myeloma Working Group Criteria. Complete response is negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow. MRD is defined by M-spike, plasma cell burden, and abnormal free light chains. Immunophenotyping is performed by multi-parametric flow cytometry. (NCT01402284)
Timeframe: Day 100

Interventionpercentage of participants (Number)
Carfilzomib, Lenalidomide, and Dexamethasone44.4

[back to top]

Percentage of Responders With Duration of Response (DOR) at 48 Months

Response is assessed by the International Myeloma Working Group Criteria. DOR is measured from the time measurement criteria are met for a partial response or better until first date that recurrent or progressive disease is objectively documented. Partial response is ≥50% reduction of serum M-protein and reduction in 24-h urinary M-protein by ≥90% or to <200mg per 24h. Progressive disease requires any one or more of the following: increase of ≥25% from lowest response value in the following on 2 consecutive measurements: serum M-component and/or (the absolute increase must be ≥0.5g/dl). Urine M-component and/or (the absolute increase must be ≥200mg/24h. Only in patients without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain (FLC) levels. The absolute increase must be >10mg/dl. Bone marrow plasma cell percentage: the absolute % must be ≥10%. (NCT01402284)
Timeframe: 48 months

Interventionpercentage of participants (Number)
Carfilzomib, Lenalidomide, and Dexamethasone81.1

[back to top]

Overall Survival (OS) Rate

OS is defined as the time of start of treatment to death from any cause. (NCT01402284)
Timeframe: up to 6 months

Interventionpercentage of participants (Number)
Carfilzomib, Lenalidomide, and Dexamethasone89.5

[back to top]

Overall Response Rate

Response is assessed by the International Myeloma Working Group Criteria. Patients who attained a partial response or better (BoR) response by the end of induction. Partial response is ≥50% reduction of serum M-protein and reduction in 24h urinary M-protein. (NCT01402284)
Timeframe: 48.3 months

Interventionpercentage of participants (Number)
Carfilzomib, Lenalidomide, and Dexamethasone97.8

[back to top]

Complete Response (CR) and Minimal Residual Disease Neg (MRDneg) CR Rates at Treatment Intervals With Carfilzomib, Lenalidomide & Dexamethasone (CRd) in New Multiple Myeloma Patients After 8 Cycles of Induction, 1 Year Maintenance, and 2 Years Maintenance

Response is assessed by the International Myeloma Working Group Criteria. MRD is defined by M-spike, plasma cell burden, and abnormal free light chains (FLC). Complete response is negative immunofixation on serum, and urine and disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow. Stringent complete response (sCR) is normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence. Near complete response (nCR) is the absence of myeloma protein on electrophoresis, independent of immunofixation status. Very good partial response (VGPR) is serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100mg per 24h. Overall response rate (ORR) is patients who attained a partial response (PR) (≥50% reduction of serum M-protein and reduction in 24h urinary M-protein) or better (BoR) response. (NCT01402284)
Timeframe: up to 2 years

Interventionpercentage of participants (Number)
sCR/CR after 8 cyclesMRDnegCR after 8 cyclesnCR after 8 cycles≥VGPR after 8 cyclesORR after 8 cyclessCR after 8 cyclessCR/CR after 1 yearsCR after 1 yearCR after 1 yearMRDnegCR after 1 year'nCR after 1 year≥VGPR after 1 yearORR after 1 yearsCR/CR after 2 yearssCR after 2 yearsCR after 2 yearsMRDnegCR after 2 years*nCR after 2 years≥VGPR after 2 yearsORR after 2 years
Carfilzomib, Lenalidomide, and Dexamethasone46.744.4208997.846.762.260.02.253.511.18997.864.462.22.246.211.191.197.8

[back to top]

Number of Participants With Serious and Non-serious Adverse Events

Here is the number 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. (NCT01402284)
Timeframe: 4 years and 9 months and 2 days

InterventionParticipants (Count of Participants)
Carfilzomib, Lenalidomide, and Dexamethasone45

[back to top]

Progression Free Survival (PFS) at 48 Months

PFS is defined as time of start of treatment to time of progression or death, whichever occurs first. Response is assessed by the International Myeloma Working Group Criteria. Progressive disease requires any one or more of the following: increase of ≥25% from lowest response value in the following on 2 consecutive measurements: serum M-component and/or (the absolute increase must be ≥0.5g/dl). Urine M-component and/or (the absolute increase must be ≥200mg/24h. Only in patients without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain (FLC) levels. The absolute increase must be >10mg/dl. Bone marrow plasma cell percentage: the absolute % must be ≥10%. Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in size of existing bone lesions or soft tissue plasmacytomas. Development of hypercalcemia that can be attributed solely to the plasma cell proliferative disorder. (NCT01402284)
Timeframe: 48 months

Interventionpercentage of participants (Number)
Carfilzomib, Lenalidomide, and Dexamethasone79.2

[back to top]

Number of Dose Limiting Toxic Events (DLT) of Lenalidomide Given in Combination With Chlorambucil.

(NCT01403246)
Timeframe: At maximum 8 months from induction therapy start

InterventionToxic events (Number)
Study Group0

[back to top]

Incidences of Grades II-IV Acute GVHD and Limited or Extensive Chronic GVHD

(NCT01419795)
Timeframe: Assessed up to 30 days after completion of study treatment

InterventionParticipants (Count of Participants)
Arm I (Lenalidomide, Rituximab)0

[back to top]

Improvement in Overall Survival of Patients Receiving Lenalidomide With or Without Rituximab in Comparison to Historical Controls Managed by Single or Multiple Chemotherapeutic Agents or Donor Lymphocyte Infusion (DLI) (Cohort 1)

Estimated using the Kaplan-Meier method in all cohorts. (NCT01419795)
Timeframe: 12 months

Interventionsurvival probability (Number)
Arm I (Lenalidomide, Rituximab)0.33

[back to top]

Rate of Response (CR, PR, or SD) and Time to Progression

Estimated using the Kaplan-Meier method in all cohorts. Assessed at day 100. (NCT01419795)
Timeframe: Assessed up to 18 months

Interventionprogression free survival probability (Number)
Arm I (Lenalidomide, Rituximab)0.67

[back to top]

Grade III-IV Toxicity in Patients Receiving Lenalidomide With or Without Rituximab

(NCT01419795)
Timeframe: Assessed up to 30 days after completion of study treatment

InterventionParticipants (Count of Participants)
Arm I (Lenalidomide, Rituximab)2

[back to top]

Comparison of Rates of Overall Response and Complete Remission Between the First, Second, and Third Cohorts

(NCT01419795)
Timeframe: Assessed up to 18 months

InterventionParticipants (Count of Participants)
Arm I (Lenalidomide, Rituximab)0

[back to top]

Comparison of Incidences of Adverse Events Between the First, Second, and Third Cohorts

(NCT01419795)
Timeframe: Assessed up to 30 days after completion of study treatment

InterventionNumber of adverse events (Number)
Arm I (Lenalidomide, Rituximab)7

[back to top]

Pharmacokinetics: AUC Cycle 1+2 - Area Under the Time/Concentration Curve for MOR202

PK analysis for MOR202 4, 8 and 16 mg/kg IV once weekly dose groups only, since serum concentrations of MOR202 were substantially affected by target mediated drug disposition effects for remaining dose groups (NCT01421186)
Timeframe: 56 days

Interventionmg*days/L (Mean)
4 mg/kg QW3307.57
8 mg/kg QW7970.15
16 mg/kg QW18178.57

[back to top]

Pharmacokinetics: Cmax - Maximum Observed Serum Concentration for MOR202

PK analysis for MOR202 4, 8 and 16 mg/kg IV once weekly dose groups only, since serum concentrations of MOR202 were substantially affected by target mediated drug disposition effects for remaining dose groups (NCT01421186)
Timeframe: up to 7 days after last MOR202 dose

Interventionµg/mL (Mean)
4 mg/kg QW137.86
8 mg/kg QW311.67
16 mg/kg QW681.53

[back to top]

Progression-free Survival

Progression-free survival (Kaplan Meier estimates) (NCT01421186)
Timeframe: patients were observed up to 36 months

Interventionmonths (Median)
Part A: MOR03087 Biweekly Dose Escalation1.1
Part B: MOR03087 Weekly Dose Escalation2.1
Part C: MOR03087 Plus Dexamethasone8.4
Part D: MOR03087 Plus Pomalidomide + Dexamethasone15.9
Part E: MOR03087 Plus Lenalidomide + Dexamethasone26.7

[back to top]

Time to Progression

Time to Progression (Kaplan Meier estimate) (NCT01421186)
Timeframe: patients were observed for up to 36 months

Interventionmonths (Median)
Part A: MOR03087 Biweekly Dose Escalation1.1
Part B: MOR03087 Weekly Dose Escalation2.1
Part C: MOR03087 Plus Dexamethasone8.4
Part D: MOR03087 Plus Pomalidomide + Dexamethasone15.9
Part E: MOR03087 Plus Lenalidomide + Dexamethasone33.2

[back to top] [back to top]

Duration of Response

Duration of response (Kaplan Meier estimates) (NCT01421186)
Timeframe: patients were observed up to 36 months

Interventionmonths (Median)
Part C: MOR03087 Plus Dexamethasone16.7
Part D: MOR03087 Plus Pomalidomide + Dexamethasone21.2
Part E: MOR03087 Plus Lenalidomide + Dexamethasone32.2

[back to top]

Number of Participants Who Develop Anti-MOR03087 Antibodies

Number of participants who develop anti-MOR03087 antibodies, a measure of immunogenicity (NCT01421186)
Timeframe: during treatment period, maximum 3 years after 1st dose

InterventionParticipants (Count of Participants)
Part A: MOR03087 Biweekly Dose Escalation0
Part B: MOR03087 Weekly Dose Escalation0
Part C: MOR03087 Plus Dexamethasone0
Part D: MOR03087 Plus Pomalidomide + Dexamethasone0
Part E: MOR03087 Plus Lenalidomide + Dexamethasone0

[back to top]

Overall Response Rate

number (#) of patients responding (# stringent complete response + # complete response + # very good partial response + # partial response) (NCT01421186)
Timeframe: maximum 3 years after 1st dose

InterventionParticipants (Count of Participants)
Part A: MOR03087 Biweekly Dose Escalation0
Part B: MOR03087 Weekly Dose Escalation0
Part C: MOR03087 Plus Dexamethasone5
Part D: MOR03087 Plus Pomalidomide + Dexamethasone10
Part E: MOR03087 Plus Lenalidomide + Dexamethasone11

[back to top]

Median Duration of Response

The median duration of response was defined by the median duration of response for participants with Complete Response (CR); CR with incomplete count recovery (CRi); or Partial Response (PR). (NCT01442714)
Timeframe: 203 days

Interventiondays (Median)
Azacitidine Plus Lenalidomide118

[back to top]

Overall Response Rate (ORR)

Overall response rate was defined as the sum of Complete Response (CR) + CR with incomplete count recovery (CRi) + Partial Response (PR). (NCT01442714)
Timeframe: 203 days

InterventionParticipants (Count of Participants)
Azacitidine Plus Lenalidomide5

[back to top]

Overall Survival

Survival was measured from the 1st day of azacitidine treatment to death from any cause. (NCT01442714)
Timeframe: 462 Days

Interventionmonths (Median)
Azacitidine Plus Lenalidomide5

[back to top]

Progression-free Survival in Patients With Previously Treated Hodgkin's Lymphoma Receiving Combined Lenalidomide and Panobinostat

Determined from the date of start of therapy to death from any cause or censored at the last date the patient is known to be alive (NCT01460940)
Timeframe: 3-5 years

Interventionmonths (Median)
Lenalidomide and Panobinostat3.8

[back to top]

Determine the Overall Response Rate (ORR), Including Complete Responses (CR) and Partial Responses (PR)

Overall response rate (CR + PR) will be determined using the International response criteria with combined panobinostat and lenalidomide in patients with relapsed or refractory Hodgkin's lymphoma. (NCT01460940)
Timeframe: up to 24 months

Interventionpercentage of patients (Number)
Lenalidomide and Panobinostat16.7

[back to top]

Assess the Safety and Tolerability of Combined Lenalidomide and Panobinostat in Patients With Previously Treated Hodgkin's Lymphoma.

Safety and tolerability will be assessed for patients using the NIH-NCI Common Terminology Criteria (CTCAE) version 4.0 (NCT01460940)
Timeframe: up to 24 months

Interventionpercentage of patients (Number)
neutropeniathrombocytopeniafebrile neutropeniahypophosphatemia
Lenalidomide and Panobinostat58.341.725.025.0

[back to top]

Number of Participants Evaluated for Toxicity

All toxicities with a maximum grade and status of ≥ grade 2 will be graded using the Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0. (NCT01463670)
Timeframe: every 28 days before initiation of a new cycle through study completion until disease progression up to 5 years

InterventionParticipants (Count of Participants)
Lenalidomide11

[back to top]

Overall Response Rate (ORR)

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. (NCT01463670)
Timeframe: 6 months

Interventionparticipants (Number)
Partial ResponseMinor ResponseStable DiseaseProgression of Disease
Lenalidomide1171

[back to top]

Overall Response Rate

The primary endpoint of overall response rate will be estimated and a 95% confidence interval will be estimated via binomial proportions. (NCT01472562)
Timeframe: 30 months

Interventionpercentage of patients (Number)
All Patients92

[back to top]

Number of Participants With Complete Response (CR)

Number of participants with complete response, the disappearance of all signs of cancer in response to treatment. (NCT01496976)
Timeframe: 3 Months

InterventionParticipants (Count of Participants)
Immunotherapy1

[back to top]

Number of Participants With Overall Survival (OS)

Overall survival will be summarized with the Kaplan-Meier curve. (NCT01496976)
Timeframe: 36 Months

Interventionparticipants (Number)
Immunotherapy24

[back to top]

Number of Participants With Partial Response (PR)

The primary endpoint for this trial is the combined complete and partial response rate to the protocol therapy at 3 months, which is also the end of Cycle 3. The objective response (CR+PR) rate will be summarized using both a point estimate and its exact confidence interval based on the binomial distribution. (NCT01496976)
Timeframe: 3 Months

Interventionparticipants (Number)
Immunotherapy33

[back to top]

Rate of Progression/Relapse Free Survival (PFS)

Progression-free survival (PFS), defined as the time from study entry to disease progression, relapse or death due to any cause, whichever is earlier. (NCT01496976)
Timeframe: Up to 56 months

Interventionmonths (Median)
Immunotherapy54.4

[back to top]

Objective Response Rate (ORR)

ORR at 3 months. Assessment for response will be made following the National Cancer Institute Working Group (NCIWG) 2008 Chronic Lymphocytic Leukemia (CLL) criteria for response. Objective response = Complete Response (disappearance of all target lesions) + Partial Response (>=30% decrease in the sum of the longest diameter of target lesions) per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) (NCT01497496)
Timeframe: 3 Months

Interventionpercentage of participants (Number)
Immunotherapy48.3

[back to top]

Overall Survival

OS is calculated for all patients from the date of initial registration to date of death due to any cause. The follow- up for patients last known to be alive is censored at the date of last contact. OS will be estimated for each of the three arms using the Kaplan-Meier method. (NCT01522976)
Timeframe: Up to 5 years

InterventionDays (Median)
Arm 1: Azacitidine/Lenalidomide588
Arm 2: Azacitidine449
Arm 3: Azacitidine/Vorinostat527

[back to top]

Relapse-free Survival

RFS is calculated for patients who have achieved a response. RFS will be measured from the date of response to the date of first documentation of relapse from response (as defined in the primary objective), or death due to any cause. The follow-up for patients last known to be alive and without report of relapse is censored at the date of last contact. RFS will be estimated for each of the three arms using the Kaplan-Meier method. (NCT01522976)
Timeframe: Up to 5 years

InterventionDays (Median)
Arm 1: Azacitidine/Lenalidomide435
Arm 2: Azacitidine311
Arm 3: Azacitidine/Vorinostat455

[back to top]

Response Rate (Phase II)

A response is any of complete hematological remission, partial remission, or hematologic improvement. (NCT01522976)
Timeframe: Up to 5 years

Interventionpercentage of patients having a response (Number)
Arm 1: Azacitidine/Lenalidomide49
Arm 2: Azacitidine38
Arm 3: Azacitidine/Vorinostat27

[back to top]

Pre-study Cytogenetic Abnormalities

Cytogenetic risk group is used to identify cytogenetic abnormalities. (NCT01522976)
Timeframe: Up to 5 years

,,
Interventionparticipants (Number)
Good/very goodIntermediatePoorVery poorMissing
Arm 1: Azacitidine/Lenalidomide351382314
Arm 2: Azacitidine2916102314
Arm 3: Azacitidine/Vorinostat341881913

[back to top]

Toxicity Rate

Adverse events that are possibly, probably or definitely related to study drug are reported. (NCT01522976)
Timeframe: Up to 5 years

,,
InterventionParticipants (Number)
Abdominal infectionAbdominal painAcute kidney injuryAdult respiratory distress syndromeAlanine aminotransferase increasedAnemiaAnorectal infectionAnorexiaApneaAscitesAspartate aminotransferase increasedAtaxiaBack painBlood and lymphatic system disorders - OtherBlood bilirubin increasedBronchial infectionCD4 lymphocytes decreasedCardiac arrestCatheter related infectionCecal infectionCholecystitisColitisConfusionConstipationCreatinine increasedDehydrationDeliriumDiarrheaDizzinessDyspneaEpistaxisEsophageal painEsophagitisFallFatigueFebrile neutropeniaFeverFlushingGastric hemorrhageGastrointestinal painGeneral disorders and admin site conditions-OtherGeneralized muscle weaknessHallucinationsHeadacheHeart failureHematomaHematuriaHyperglycemiaHypernatremiaHypertensionHyperuricemiaHypoalbuminemiaHypokalemiaHypomagnesemiaHyponatremiaHypophosphatemiaHypotensionHypoxiaInfections and infestations - Other, specifyIntracranial hemorrhageInvestigations - Other, specifyLeukocytosisLower gastrointestinal hemorrhageLung infectionLymphocyte count decreasedLymphocyte count increasedMucosal infectionMucositis oralNauseaNeutrophil count decreasedPainPapulopustular rashPericardial effusionPlatelet count decreasedPneumonitisPruritusPulmonary edemaPurpuraRash maculo-papularRenal and urinary disorders - Other, specifyRespiratory failureSepsisSinus bradycardiaSkin infectionSoft tissue infectionSudden death NOSSyncopeThromboembolic eventTooth infectionUpper gastrointestinal hemorrhageUpper respiratory infectionUrinary tract infectionVascular disorders - Other, specifyVomitingWeight lossWhite blood cell decreased
Arm 1: Azacitidine/Lenalidomide1101048040100100011100002250514001091600011300001101034151404110041400116911161010113004131021000101148
Arm 2: Azacitidine00000370000000200002000010100020000611000001000001100000100020010191002480004600003002011100001101034
Arm 3: Azacitidine/Vorinostat031054413113101310011111103130311011413211103111122021010423150002231102630006410101113020041110111044

[back to top]

Number of Participants With Response

Overall response defined as number of participants with International Myeloma Working Group Uniform Response Criteria: Complete Response (CR): Negative immunofixation serum & urine, Disappearance soft tissue plasmacytomas & =/<5% plasma cells in bone marrow; Stringent Complete Remission: CR + Normal Normal free light chain (FLC) ratio & Absence clonal cells in bone marrow by Immunohistochemistry/ immunofluorescence; Very Good Partial Response (VGPR): Serum & urine M-protein detectable by immunofixation but not on electrophoresis or 90%> reduction in serum M-protein +urine M-protein level <100mg per 24 hour; Partial Remission (PR): =/>50% reduction serum M-protein & reduction in 24-hour urinaryMprotein by >90% or to < 200mg per 24 hour, =/>50% reduction of serum M-protein & reduction in 24-hour urinary Mprotein by >90%/or <200mg, and if present at baseline, a >50% reduction in size of soft tissue plasmacytomas; Stable Disease: Not CR, VGPR, PR Or Progressive disease (NCT01531998)
Timeframe: Evaluated after eight cycles of 21 days.

Interventionparticipants (Number)
Complete Remission (CR)Stringent Complete RemissionVery good partial remission (VGPR)Partial Remission (PR)Stable Disease
Siltuximab + Bortezomib + Lenalidomide20260

[back to top]

Maximum Tolerated Dose (MTD) of Siltuximab

Maximum tolerated dose (MTD) defined as follows: At first dose level, if greater than 1 out of 3 patients or greater than 1 out of 6 patients experience dose limiting toxicity (DLT), the dose level exceeds the maximum tolerated dose (MTD). Dose limiting toxicity (DLT) defined as toxicities graded in severity according to the guidelines outlined in the NCI-Common Toxicity Criteria for Adverse Effects (CTCAE) version 4.0. (NCT01531998)
Timeframe: 21 days

Interventionmg/kg (Number)
Siltuximab + Bortezomib + Lenalidomide8.3

[back to top]

Pharmacokinetic Parameters of Lenalidomide

Concentration of lenalidomide obtained from any day between day 5 and 21 of the first cycle of chemotherapy in nanograms per mL. (NCT01553149)
Timeframe: Between days 5-21 of course 1 and each dose reduction

Interventionnanograms per mL (Median)
Arm I (Low-dose Lenalidomide)15.1
Arm II (High-dose Lenalidomide)178.8

[back to top]

Overall Survival [OS]

Estimated 3-year overall survival is calculated as the time from study enrollment to death from any cause or last follow-up whichever occurs first. Kaplan-Meier method is used for estimation. Patients alive at last contact are censored at that time. (NCT01553149)
Timeframe: Up to 3 years after study enrollment

InterventionPercent Probability (Number)
Arm I (Low-dose Lenalidomide)94.59
Arm II (High-dose Lenalidomide)91.74

[back to top]

Event-free Survival [EFS]

Estimated 3-year EFS where EFS is calculated as the time from study enrollment to disease progression, disease relapse, occurrence of a second malignant neoplasm, death from any cause or last follow-up whichever occurs first. Kaplan-Meier method is used for estimation. Patients without an event are censored at last contact. (NCT01553149)
Timeframe: Up to 3 years after study enrollment

InterventionPercent Probability (Number)
Arm I (Low-dose Lenalidomide)37.84
Arm II (High-dose Lenalidomide)39.41

[back to top]

Number of Patients Who Demonstrate Complete or Partial Response

"Number of patients who demonstrate a complete or partial response as defined below:~Complete Response - Complete disappearance of all known disease for at least 4 weeks;~Partial Response - A reduction of at least 50% in the size of all measurable tumor as quantitated by the sum of the products of the largest diameters of measurable lesions when compared with that measurement at the time of study enrollment and maintained for at least 4 weeks." (NCT01553149)
Timeframe: 26 cycles of chemotherapy - up to 3 years after enrollment

InterventionParticipants (Count of Participants)
Arm I (Low-dose Lenalidomide)4
Arm II (High-dose Lenalidomide)4

[back to top]

Number of Patients Who Demonstrate Early Progression

Number of patients with disease progression during the first six months of protocol therapy. Disease progression is defined as ≥ 25% increase in the sum of the products of the largest diameters of the measurable lesions or the appearance of one or more new lesions when compared with the measurements of lesions at the time of enrollment. (NCT01553149)
Timeframe: Up to 180 days after enrollment

InterventionParticipants (Count of Participants)
Arm I (Low-dose Lenalidomide)6
Arm II (High-dose Lenalidomide)4

[back to top]

Number of Patients With Toxic Events After 2 Dose Reductions

Number of patients who have an additional significant toxicity coded using Common Terminology Criteria for Adverse Events Version 5.0 after experiencing two dose reductions from their assigned treatment dose. (NCT01553149)
Timeframe: While receiving protocol therapy up to 3 years after study enrollment

InterventionParticipants (Count of Participants)
Arm I (Low-dose Lenalidomide)2
Arm II (High-dose Lenalidomide)16

[back to top]

Stem Cells Collection

At the end of the Car Phase, all participants underwent stem cell collection. (NCT01559935)
Timeframe: At the end of the Car Phase, prior to the start of the BiRD Phase, on average after 162 days.

InterventionNumber of stem cells collected per Kg (Mean)
Car-BiRD Therapy12854635

[back to top]

Response to Car-BiRD Treatment.

"The best response for all patients who had at least one dose of drug was measured.~Response categories:~Stringent Complete Response (sCR), Complete Remission(CR), Very Good Partial Remission(VGPR), Partial Remission (PR), Progressive Disease (PD), Stable Disease (SD).~The response is evaluated based on the IMWG criteria." (NCT01559935)
Timeframe: From baseline to best response, up to 116 weeks.

InterventionParticipants (Count of Participants)
sCR/CRVGPRPRSDPDNot Evaluable
Car-BiRD Therapy283111101

[back to top]

Event Free Survival

an event is defined by coming off protocol for any reason, including progression of disease, lack of disease response, regimen intolerability, withdrawal of consent or death. (NCT01559935)
Timeframe: From date of study enrollment until the date of removal of study due to progression of disease, toxicity or withdrawal of consent, up to 1222 days.

InterventionDays (Median)
Car-BiRD Therapy401.5

[back to top]

Progression Free Survival

Progression was defined using the IMWG criteria. (NCT01559935)
Timeframe: From start of study drug until first incidence of progression, up to 1222 days.

Interventionmonths (Median)
Car-BiRD Therapy18.3

[back to top]

Duration of Response (DOR)

DOR was measured as the time in months from the date of first documentation of a confirmed response of PR or better (CR [including sCR] + PR+ VGPR) to the date of the first documented disease progression (PD) among participants who responded to the treatment. Response was assessed by the investigator using International Myeloma Working Group (IMWG) Criteria. (NCT01564537)
Timeframe: Day 1 of each cycle (every 4 weeks) until disease progression up to approximately 38 months

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone26.0
Placebo + Lenalidomide + Dexamethasone21.7

[back to top]

Overall Response Rate (ORR) as Assessed by the IRC

ORR was defined as the percentage of participants with Complete Response (CR) including stringent complete response (sCR), very good partial response (VGPR) and Partial Response (PR) assessed by the IRC using IMWG criteria. Percentages are rounded off to single decimal. (NCT01564537)
Timeframe: Day 1 of each cycle (every 4 weeks) until disease progression up to approximately 27 months(approximate median follow-up 15 months)

Interventionpercentage of participants (Number)
Ixazomib+ Lenalidomide + Dexamethasone78.3
Placebo + Lenalidomide + Dexamethasone71.5

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Multiple Myeloma Module (QLQ-MY-20)

The EORTC-QLQ-MY-20 is a patient-completed, 20-question quality of life questionnaire that has 4 independent subscales, 2 functional subscales (body image, future perspective), and 2 symptoms scales (disease symptoms and side-effects of treatment). The participant answers questions about their health during the past week using a 4-point scale where 1=Not at All to 4=Very Much. A negative change from Baseline indicates improvement. Scores are linearly transformed to a 0-100 scale. Higher scores on the symptom scales (e.g. Disease Symptoms, Side Effects of Treatment) represent higher levels of symptomatology or problems. High scores for Body Image and Future Perspective represent better quality of life or functioning. (NCT01564537)
Timeframe: Baseline, EOT and follow-up (up to approximately 97 months)

Interventionscore on a scale (Mean)
Disease Symptoms: BaselineDisease Symptoms: EOTSide Effects of Treatment: BaselineSide Effects of Treatment: EOTSide Effects of Treatment: Last Follow-upBody Image: BaselineBody Image: EOTBody Image: Last Follow-upFuture Perspective: BaselineFuture Perspective: EOTFuture Perspective: Last Follow-up
Placebo + Lenalidomide + Dexamethasone30.41-2.5817.974.4337.0479.48-5.38-33.360.26-2.75-11.11

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) Questionnaire (EORTC-QLQ-C30)

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer participants. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact).The EORTC-QLQ-C30 Global Health Status/QOL Scale is scored between 0 and 100, where higher scores indicate better Global Health Status/QOL. Negative changes from baseline indicate deterioration in QOL or functioning and positive changes indicate improvement. Scores are linearly transformed to a 0-100 scale. High scores for the global and functional domains indicate higher quality of life or functioning. Higher scores on the symptom scales represent higher levels of symptomatology or problems. (NCT01564537)
Timeframe: Baseline, EOT and follow-up (up to approximately 97 months)

Interventionscore on a scale (Mean)
Global Health Index: BaselineGlobal Health Index: End of TreatmentGlobal Health Index: Last Follow-upPhysical Functioning: BaselinePhysical Functioning: EOTPhysical Functioning: Last Follow-upRole Functioning: BaselineRole Functioning: EOTRole Functioning: Last Follow-upEmotional Functioning: BaselineEmotional Functioning: EOTEmotional Functioning: Last Follow-upCognitive Functioning: BaselineCognitive Functioning: EOTCognitive Functioning: Last Follow-upSocial Functioning: BaselineSocial Functioning: EOTSocial Functioning: Last Follow-upFatigue: BaselineFatigue: EOTFatigue: Last Follow-upPain: BaselinePain: EOTPain: Last Follow-upNausea and Vomiting: BaselineNausea and Vomiting: EOTNausea and Vomiting: Last Follow-upDyspnea: BaselineDyspnea: EOTDyspnea: Last Follow-upInsomnia: BaselineInsomnia: EOTInsomnia: Last Follow-upAppetite Loss: BaselineAppetite Loss: EOTAppetite Loss: Last Follow-upConstipation: BaselineConstipation: EOTConstipation: Last Follow-upDiarrhea: BaselineDiarrhea: EOTDiarrhea: Last Follow-upFinancial Difficulties: BaselineFinancial Difficulties: EOTFinancial Difficulties: Last Follow-up
Placebo + Lenalidomide + Dexamethasone56.4-6.016.767.3-6.20.064.4-8.6-16.775.3-6.1-25.081.6-5.8-50.075.3-7.90.039.56.722.238.53.80.06.00.633.323.72.30.030.5-0.533.315.36.50.013.52.233.38.110.80.018.61.3-33.3

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Core Quality of Life (QOL) Questionnaire (EORTC-QLQ-C30)

The EORTC-QLQ-C30 is a 30-question tool used to assess the overall quality of life in cancer participants. It consists of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, Role, Cognitive, Emotional, Social), and 9 symptom scales/items (Fatigue, Nausea and Vomiting, Pain, Dyspnea, Sleep Disturbance, Appetite Loss, Constipation, Diarrhea, Financial Impact).The EORTC-QLQ-C30 Global Health Status/QOL Scale is scored between 0 and 100, where higher scores indicate better Global Health Status/QOL. Negative changes from baseline indicate deterioration in QOL or functioning and positive changes indicate improvement. Scores are linearly transformed to a 0-100 scale. High scores for the global and functional domains indicate higher quality of life or functioning. Higher scores on the symptom scales represent higher levels of symptomatology or problems. (NCT01564537)
Timeframe: Baseline, EOT and follow-up (up to approximately 97 months)

Interventionscore on a scale (Mean)
Global Health Index: BaselineGlobal Health Index: End of TreatmentPhysical Functioning: BaselinePhysical Functioning: EOTRole Functioning: BaselineRole Functioning: EOTEmotional Functioning: BaselineEmotional Functioning: EOTCognitive Functioning: BaselineCognitive Functioning: EOTSocial Functioning: BaselineSocial Functioning: EOTFatigue: BaselineFatigue: EOTPain: BaselinePain: EOTNausea and Vomiting: BaselineNausea and Vomiting: EOTDyspnea: BaselineDyspnea: EOTInsomnia: BaselineInsomnia: EOTAppetite Loss: BaselineAppetite Loss: EOTConstipation: BaselineConstipation: EOTDiarrhea: BaselineDiarrhea: EOTFinancial Difficulties: BaselineFinancial Difficulties: EOT
Ixazomib+ Lenalidomide + Dexamethasone58.4-6.070.0-4.768.4-8.675.1-2.181.9-7.677.9-6.938.46.038.02.75.03.421.25.727.40.916.94.712.2-1.36.317.216.70.5

[back to top]

Time to Progression (TTP) as Assessed by the IRC

TTP was measured as the time in months from the first dose of study treatment to the date of the first documented progressive disease (PD) as assessed by the IRC using IMWG criteria. (NCT01564537)
Timeframe: Day 1 of each cycle (every 4 weeks) until disease progression up to approximately 27 months (approximate median follow-up 15 months)

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone22.4
Placebo + Lenalidomide + Dexamethasone17.6

[back to top]

PFS in High-Risk Participants

Progression Free Survival (PFS) is defined as the time from the date of randomization to the date of first documentation of disease progression or death due to any cause, whichever occurs first. Response was assessed by independent review committee (IRC) using IMWG response criteria. High-risk participants are defined as participants carrying cytogenic abnormalities: del(17), translocation t(4;14), or t(14;16) as reported by the central laboratory combined with those cases that lacked a central laboratory result but with known del (17), t(4;14), or t(14;16) by local laboratory. Cytogenetic abnormalities of del(13) and +1q are not included in the analysis. (NCT01564537)
Timeframe: From date of randomization until disease progression or death up to approximately 38 months (approximate median follow-up 15 months)

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone18.7
Placebo + Lenalidomide + Dexamethasone9.3

[back to top]

Percentage of Participants With Complete Response (CR) and Very Good Partial Response (VGPR) as Assessed by the IRC

Response was assessed by the IRC using International Myeloma Working Group (IMWG) Criteria. CR is defined as negative immunofixation on the serum and urine and; disappearance of any soft tissue plasmacytomas and; < 5% plasma cells in bone marrow. VGPR is defined as Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. Percentages are rounded off to single decimal. (NCT01564537)
Timeframe: Day 1 of each cycle (every 4 weeks) until disease progression up to approximately 27 months (approximate median follow-up 15 months)

Interventionpercentage of participants (Number)
Ixazomib + Lenalidomide + Dexamethasone48.1
Placebo + Lenalidomide + Dexamethasone39.0

[back to top]

Overall Response Rate in Participants Defined by Polymorphism

Data is reported for percentage of participants defined by polymorphism defined by polymorphisms in proteasome genes, such as polymorphism P11A in PSMB1 gene. Percentages are rounded off to single decimal. (NCT01564537)
Timeframe: Day 1 of each cycle (every 4 weeks) until disease progression up to approximately 27 months (approximate median follow-up 15 months)

Interventionpercentage of participants (Number)
Ixazomib+ Lenalidomide + Dexamethasone80.3
Placebo + Lenalidomide + Dexamethasone75.7

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from the date of randomization to the date of death. Participants without documentation of death at the time of the analysis were censored at the date when they were last known to be alive. (NCT01564537)
Timeframe: From date of randomization until death (up to approximately 97 months)

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone53.6
Placebo + Lenalidomide + Dexamethasone51.6

[back to top]

Plasma Concentration Over Time for Ixazomib

(NCT01564537)
Timeframe: Pre-dose and post-dose at multiple timepoints up to Cycle 10 Day 1 (each cycle length = 28 days)

Interventionμg/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 1, 1 Hour Post-DoseCycle 1 Day 1, 4 Hours Post-DoseCycle 1 Day 14, Pre-DoseCycle 2 Day 1, Pre-DoseCycle 2 Day 14, Pre-DoseCycle 3 Day 1, Pre-DoseCycle 4 Day 1, Pre-DoseCycle 5 Day 1, Pre-DoseCycle 6 Day 1, Pre-DoseCycle 7 Day 1, Pre-DoseCycle 8 Day 1, Pre-DoseCycle 9 Day 1, Pre-DoseCycle 10 Day 1, Pre-Dose
Ixazomib+ Lenalidomide + Dexamethasone4.7936.315.66.832.47.122.482.412.422.572.712.372.512.82

[back to top]

Plasma Concentration Over Time for Ixazomib

(NCT01564537)
Timeframe: Pre-dose and post-dose at multiple timepoints up to Cycle 10 Day 1 (each cycle length = 28 days)

Interventionμg/mL (Mean)
Cycle 1 Day 1, 1 Hour Post-DoseCycle 1 Day 1, 4 Hours Post-DoseCycle 1 Day 14, Pre-DoseCycle 2 Day 1, Pre-DoseCycle 2 Day 14, Pre-DoseCycle 3 Day 1, Pre-DoseCycle 4 Day 1, Pre-DoseCycle 5 Day 1, Pre-DoseCycle 6 Day 1, Pre-DoseCycle 7 Day 1, Pre-DoseCycle 8 Day 1, Pre-DoseCycle 9 Day 1, Pre-DoseCycle 10 Day 1, Pre-Dose
Placebo + Lenalidomide + Dexamethasone0000000000000

[back to top]

Number of Participants With Change From Baseline in Pain Response

"Pain response was defined as 30% reduction from Baseline in Brief Pain Inventory-Short Form (BPI-SF) worst pain score over the last 24 hours without an increase in analgesic (oral morphine equivalents) use at 2 consecutive evaluations. The BPI-SF contains 15 items designed to capture the pain severity (worst, least, average, and now [current pain]), pain location, medication to relieve the pain, and the interference of pain with various daily activities including general activity, mood, walking activity, normal work, relations with other people, sleep, and enjoyment of life. The pain severity items are rated on a 0 to 10 scale where: 0=no pain and 10=pain as bad as you can imagine and averaged for a total score of 0 (best) to 10 (Worst)." (NCT01564537)
Timeframe: Baseline and end of treatment (EOT) (up to approximately 38 months)

,
InterventionParticipants (Count of Participants)
BaselineEOT
Ixazomib+ Lenalidomide + Dexamethasone345145
Placebo + Lenalidomide + Dexamethasone351153

[back to top]

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

Eastern Cooperative Oncology Group (ECOG) performance score, laboratory values, vital sign measurements and reported adverse events (AEs) were collected and assessed to evaluate the safety of therapy throughout the study. An AE is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. A serious adverse event (SAE) is an AE resulting in any of the following outcomes or deemed significant for any other reason: death; initial or prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; or congenital anomaly; or a medically important event. (NCT01564537)
Timeframe: From the date of signing of the informed consent form through 30 days after the last dose of study drug up to approximately 115 months

,
InterventionParticipants (Count of Participants)
TEAEsSAEs
Ixazomib+ Lenalidomide + Dexamethasone359205
Placebo + Lenalidomide + Dexamethasone357201

[back to top]

Overall Survival in High-Risk Participants Carrying Deletion 17 [Del(17)]

Overall survival is defined as the time from the date of randomization to the date of death. The high-risk participants whose myeloma carried del(17) subgroup was defined as the cases reported as positive for del(17) by the central laboratory combined with those cases that lacked a central laboratory result but with known del (17) by local laboratory. Participants without documentation of death at the time of the analysis were censored at the date when they were last known to be alive. Data is only reported high-risk participants with Del(17). (NCT01564537)
Timeframe: From the time of screening until disease progression and thereafter every 12 weeks until death or study termination (up to approximately 97 months)

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone42.2
Placebo + Lenalidomide + Dexamethasone29.4

[back to top]

Progression Free Survival (PFS) as Assessed by the Independent Review Committee (IRC)

Progression Free Survival (PFS) is defined as the time from the date of randomization to the date of first documentation of disease progression (PD) or death due to any cause, whichever occurs first. Response including PD was assessed by independent review committee (IRC) using the International Myeloma Working Group (IMWG) response criteria. PD requires 1 of the following: Increase of ≥ 25% from nadir in: Serum M-component (absolute increase ≥ 0.5 g/dl); Urine M-component (absolute increase ≥ 200 mg/24 hours); In patients without measurable serum and urine M-protein levels the difference between involved and uninvolved free light chain (FLC) levels (absolute increase > 10 mg/dl); Development of new or increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium > 11.5 mg/dl) attributed solely to plasma cell proliferative disease. Status evaluated every 4 weeks until disease progression (PD) was confirmed. (NCT01564537)
Timeframe: From date of randomization until disease progression or death up to approximately 27 months (approximate median follow-up 15 months)

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone20.6
Placebo + Lenalidomide + Dexamethasone14.7

[back to top]

Change From Baseline in the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Multiple Myeloma Module (QLQ-MY-20)

The EORTC-QLQ-MY-20 is a patient-completed, 20-question quality of life questionnaire that has 4 independent subscales, 2 functional subscales (body image, future perspective), and 2 symptoms scales (disease symptoms and side-effects of treatment). The participant answers questions about their health during the past week using a 4-point scale where 1=Not at All to 4=Very Much. A negative change from Baseline indicates improvement. Scores are linearly transformed to a 0-100 scale. Higher scores on the symptom scales (e.g. Disease Symptoms, Side Effects of Treatment) represent higher levels of symptomatology or problems. High scores for Body Image and Future Perspective represent better quality of life or functioning. (NCT01564537)
Timeframe: Baseline, EOT and follow-up (up to approximately 97 months)

Interventionscore on a scale (Mean)
Disease Symptoms: BaselineDisease Symptoms: EOTDisease Symptoms: Last Follow-upSide Effects of Treatment: BaselineSide Effects of Treatment: EOTBody Image: BaselineBody Image: EOTFuture Perspective: BaselineFuture Perspective: EOT
Ixazomib+ Lenalidomide + Dexamethasone29.71-2.351.1117.234.5278.00-0.2756.992.76

[back to top]

OS in High-Risk Participants

Overall survival (OS) is defined as the time from the date of randomization to the date of death. High-risk participants are defined as participants carrying cytogenic abnormalities: del(17), translocation t(4;14), or t(14;16) as reported by the central laboratory combined with those cases that lacked a central laboratory result but with known del (17), t(4;14), or t(14;16) by local laboratory. Cytogenetic abnormalities of del(13) and +1q are not included in the analysis. Participants without documentation of death at the time of the analysis were censored at the date when they were last known to be alive. Data is only reported for high-risk participants. (NCT01564537)
Timeframe: From the time of screening until disease progression and thereafter every 12 weeks until death or study termination (up to approximately 97 months)

Interventionmonths (Median)
Ixazomib+ Lenalidomide + Dexamethasone46.9
Placebo + Lenalidomide + Dexamethasone30.9

[back to top]

Percentage of Participants With Minimal Residual Disease (MRD)Negative Complete Response (CR) Per International Myeloma Working Group (IMWG) Criteria

MRDnegative Complete Response (CR) per the IMWHG criteria is defined as absence of phenotypically aberrant clonal plasma cells by flow cytometry on bone marrow aspirates using the eight-color two tube approach with a minimum sensitivity of 1 in 10^5 nucleated cells or higher. (NCT01572480)
Timeframe: 8 months

Interventionpercentage of participants (Number)
All Participants70.4

[back to top]

Overall Response Rate

Overall response is defined as the percentage of participants who have a partial response (PR), very good partial response (VGPR), complete response (CR), or stringent complete response (sCR) defined by the International Myeloma Working Group Criteria for Multiple Myeloma out of all evaluable participants. PR is ≥50% reduction if serum M-protein and reduction in 24-hour(h) urinary M-protein by ≥90% or to <200mg per 24h. If the serum and urine M-protein are unmeasurable, a ≥50% decrease in the difference between involved and uninvolved free light chain (FLC) levels is required in place of the M-protein criteria. VGPR is serum and urine M-protein detectable by immunofixation but not on electrophoresis. CR is negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and ≥5% plasma cells in bone marrow. sCR is complete response plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence. (NCT01572480)
Timeframe: time measurement criteria are met for best response until the first date that recurrent or progressive disease is met, up to 5 years

Interventionpercentage of participants (Number)
Overall Response RatePartial ResponseVery Good Partial ResponseComplete ResponseStringent Complete Response
All Participants1005.518.5075.9

[back to top]

Percentage of Participants That Have Minimal Residual Disease (MRD)-Negative Complete Response (CR) for a Minimum of 1 Year

Percentage of participants that have Minimal Residual Disease (MRD)-negative Complete Response (CR) for a minimum of 1 year estimated along with a 95% two-sided confidence interval. MRDnegative Complete Response (CR) is defined as absence of phenotypically aberrant clonal plasma cells by flow cytometry on bone marrow aspirates using the eight-color two tube approach with a minimum sensitivity of 1 in 10^5 nucleated cells or higher. (NCT01572480)
Timeframe: 1 year

Interventionpercentage of participants (Number)
All Participants63

[back to top]

Number of Participants With Serious and/or 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/or 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. (NCT01572480)
Timeframe: Date treatment consent signed to date off study, approximately 117 months and 1 day.

InterventionParticipants (Count of Participants)
All Participants54

[back to top]

Clinical Progression Free Survival

Clinical progression was assessed by the International Myeloma Working Group Criteria for Multiple Myeloma. Progression is any one of the following: Increase of ≥25% from lowest response value in the following on 2 consecutive measurements: Serum M-component and/or (the absolute increase must be ≥0.5g/dl). The serum M-component increases of ≥1 gm/dl are sufficient to define relapse if starting M-component is ≥0.5g/dl. Urine M-component and/or (the absolute must be ≥200mg/24h. Only in participants without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain (FLC) levels. The absolute increase must be >10mg/dl. Bone marrow plasma cell percentage: the absolute % must be ≥10%. Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in size of existing bone lesions or soft tissue plasmacytomas. Development of hypercalcemia that can be attributed solely to the plasma cell proliferative disorder. (NCT01572480)
Timeframe: time from enrollment in the study until development of overt clinical multiple myeloma (end organ damage or myeloma-defining event) or death, up to 5 years

Interventionpercentage of participants (Number)
All Participants90.7

[back to top]

Biochemical Progression Free Survival

Biochemical progression free survival is defined as the time from enrollment in the study until development of overt clinical multiple myeloma (end organ damage or myeloma-defining event) or death, and progressive disease by International Myeloma Working Group (IMWG) criteria. Progression is any one of the following: Increase of ≥25% from lowest response value in the following on 2 consecutive measurements: Serum M-component and/or (the absolute increase must be ≥0.5g/dl). The serum M-component increases of ≥1 gm/dl are sufficient to define relapse if starting M-component is ≥0.5g/dl. Urine M-component and/or (the absolute must be ≥200mg/24h. Only in participants without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain (FLC) levels. The absolute increase must be >10mg/dl. Bone marrow plasma cell percentage: the absolute % must be ≥10%. (NCT01572480)
Timeframe: time from enrollment in the study until development of overt clinical multiple myeloma (end organ damage or myeloma-defining event) or death, and progressive disease by IMWG criteria, up to 5 years

Interventionpercentage of participants (Number)
All Participants76.6

[back to top]

Progression Free Survival

Progression free survival (PFS) is defined as days from start of high dose chemotherapy to first documented progression of disease, death due to any cause or last patient contact. (NCT01575860)
Timeframe: 12 months from start of lenalidomide maintenance

InterventionParticipants (Count of Participants)
Participants without Disease ProgressionParticipants with Disease Progression
Maintenance Lenalidomide in Lymphoma42

[back to top]

Number of Subjects With Dose-limiting Toxicities

Dose-limiting toxicity (DLT) is defined as any grade 3 toxicity or higher that occurs during the first 28 days of therapy and is possibly, probably, or definitely related to lenalidomide maintenance. (NCT01575860)
Timeframe: 28 days (Cycle 1)

InterventionParticipants (Count of Participants)
Maintenance Lenalidomide in Lymphoma0

[back to top]

Recording of the Occurrence of Adverse Events

(NCT01600053)
Timeframe: From date of first dose until the date of first documented progression or date of death from any cause, whichever came first

Interventionparticipants (Number)
Lenalidomide11

[back to top]

Phase 2: Time to Progression (TTP)

TTP was defined as the number of days from the date of first infusion (Day 1) to the date of first record of disease progression. Disease progression (IMWG criteria): increase of >=25 percent (%) from lowest response level in Serum M-component and/or (the absolute increase must be >=0.5 g/dL) Urine M-component and/or (the absolute increase must be >=200 mg/24 hour; only in participants without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain levels. The absolute increase must be >10 mg/dL; Bone marrow plasma cell percentage: the absolute % must be >=10 %; Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL or 2.65 mmol/L) that can be attributed solely to the plasma cell proliferative disorder. Median TTP was estimated by using the Kaplan-Meier method. (NCT01615029)
Timeframe: Up to 3 years

InterventionMonths (Median)
Phase 2: 16 mg/kg Daratumumab + Lenalidomide and DexamethasoneNA

[back to top]

Phase 2: Duration of Response

Duration of response was calculated from the date of initial documentation of a response (PR or better) to the date of first documented evidence of progressive disease, as defined in the International Myeloma Working Group (IMWG) criteria. (NCT01615029)
Timeframe: Up to 3 years

InterventionMonths (Median)
Phase 2: 16 mg/kg Daratumumab + Lenalidomide and DexamethasoneNA

[back to top]

Phase 1: Percentage of Participants With Overall Response Rate (ORR)

ORR is defined as percentage of participants who achieved stringent complete response (sCR), complete response (CR), very good partial response (VGPR) or partial response (PR). International Myeloma Working Group (IMWG) criteria- CR: Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and less than (<) 5 percentage (%) plasma cells in bone marrow; sCR: CR+Normal free light chain ratio and absence of clonal cells in bone marrow by immunohistochemistry or immuno fluorescence; PR: greater than equal to (>=) 50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by >= 90 percentage (%) or to <200 mg/24 hours; VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hour. (NCT01615029)
Timeframe: Up to 3 years

InterventionPercentage of participants (Number)
Phase 1: 2 mg/kg Daratumumab + Lenalidomide and Dexamethasone100.0
Phase 1: 4 mg/kg Daratumumab + Lenalidomide and Dexamethasone100.0
Phase 1: 8 mg/kg Daratumumab + Lenalidomide and Dexamethasone75.0
Phase 1: 16 mg/kg Daratumumab + Lenalidomide and Dexamethasone66.7

[back to top]

Phase 2: Overall Survival (OS)

Overall Survival (OS) was defined as the number of days from administration of the first infusion (Day 1) to date of death. Median Overall Survival was estimated by using the Kaplan Meier method. (NCT01615029)
Timeframe: Up to 3 years

InterventionMonths (Median)
Phase 2: 16 mg/kg Daratumumab + Lenalidomide and DexamethasoneNA

[back to top]

Phase 2: Percentage of Participants With Overall Response Rate (ORR)

ORR is defined as percentage of participants who achieved stringent complete response (sCR), complete response (CR), very good partial response (VGPR) or partial response (PR). IMWG criteria- CR: Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and less than (<) 5 percentage (%) plasma cells in bone marrow; sCR: CR+Normal free light chain ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence; PR: greater than eqaul to (>=) 50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by >= 90 percentage (%) or to <200 mg/24 hours; VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hour. (NCT01615029)
Timeframe: Up to 3 years

InterventionPercentage of participants (Number)
Phase 2: 16 mg/kg Daratumumab + Lenalidomide and Dexamethasone81.3

[back to top]

Phase 2: Progression-Free Survival (PFS)

Progression free survival (PFS) was defined as the time between the date of first dose of daratumumab and either disease progression or death, whichever occurs first. (NCT01615029)
Timeframe: Up to 3 years

InterventionMonths (Median)
Phase 2: 16 mg/kg Daratumumab + Lenalidomide and DexamethasoneNA

[back to top]

Phase 2: Time to Response

Time to first response was defined as the time from the date of first dose of daratumumab to the date of initial documentation of a response (PR or better). Time to best response was defined as the time between the date of first dose of daratumumab and the date of the initial evaluation of the best response (PR or better) to treatment. (NCT01615029)
Timeframe: Up to 3 years

InterventionMonths (Mean)
Time to first responseTime to best response
Phase 2: 16 mg/kg Daratumumab + Lenalidomide and Dexamethasone1.555.60

[back to top]

Phase 1: Time to Response

Time to first response was defined as the time from the date of first dose of daratumumab to the date of initial documentation of a response (PR or better). Time to best response was defined as the time between the date of first dose of daratumumab and the date of the initial evaluation of the best response (PR or better) to treatment. (NCT01615029)
Timeframe: Up to 3 years

,,,
InterventionMonths (Mean)
Time to first responseTime to best response
Phase 1: 16 mg/kg Daratumumab + Lenalidomide and Dexamethasone2.255.47
Phase 1: 2 mg/kg Daratumumab + Lenalidomide and Dexamethasone0.833.12
Phase 1: 4 mg/kg Daratumumab + Lenalidomide and Dexamethasone1.1412.54
Phase 1: 8 mg/kg Daratumumab + Lenalidomide and Dexamethasone1.279.75

[back to top]

Progression Free Survival (PFS)

"Progression was defined as any one or more of the following:~Serum M protein increase ≥ 25% from baseline (or an increase of ≥ 1 g/dL if serum M protein was ≥ 5 g/dL at baseline), with an absolute increase of ≥ 0.5 g/dL; or~Urine M protein increase ≥ 25% from baseline, with an absolute increase of ≥ 200 mg/24 hrs; or~If patient had serum M protein < 1 g/dL, urine M protein < 200 mg/24 hrs, and an involved serum free light chain level ≥ 10 mg/dL at baseline: ≥ 25% increase in the difference between involved and uninvolved serum free light chain level, with an absolute increase of ≥ 10 mg/dL; or~Bone marrow plasma cell percentage increase ≥ 25% from baseline, with the absolute plasma cell % ≥ 10%; or~New bone lesions or soft tissue plasmacytomas, or definite increase in size of existing bone lesions or soft tissue plasmacytomas; or~Development of hypercalcemia (corrected serum calcium > 11.5 mg/dL or 2.65 mmol/L) that can be attributed solely to multiple myeloma." (NCT01621672)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Revlimid88
Observation71

[back to top]

Duration of Response (DOR)

DOR was defined as the length of time between the date of first documented response (PR, VGPR, or CR) and the date of first documented progressive disease (PD). According to IMWG criteria: CR: negative immunofixation on serum and urine, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in bone marrow; PR: ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to <200 mg per 24 hours. VGPR: serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein plus urine M-protein level <100 mg per 24 hour. (NCT01645930)
Timeframe: From date of documentation of a confirmed response to date of progressive disease, (approximately 20 months)

Interventionmonths (Median)
Ixazomib+Lenalidomide+Dexamethasone12.9

[back to top]

Cmax: Maximum Observed Plasma Concentration for Ixazomib

(NCT01645930)
Timeframe: Cycle 1, Day 15 pre-dose and multiple time-points (up to 336 hours) post-dose

Interventionng/mL (Mean)
Ixazomib+Lenalidomide+Dexamethasone57.57

[back to top]

Cmax: Maximum Observed Plasma Concentration for Ixazomib

(NCT01645930)
Timeframe: Cycle 1, Day 1 pre-dose and multiple time-points (up to 168 hours) post-dose

Interventionng/mL (Mean)
Ixazomib+Lenalidomide+Dexamethasone37.57

[back to top]

AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to Time of the Last Quantifiable Concentration for Ixazomib

(NCT01645930)
Timeframe: Cycle 1, Day 15 pre-dose and multiple time-points (up to 336 hours) post-dose

Interventionhr*ng/mL (Mean)
Ixazomib+Lenalidomide+Dexamethasone1746.0

[back to top]

AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to Time of the Last Quantifiable Concentration for Ixazomib

(NCT01645930)
Timeframe: Cycle 1, Day 1 pre-dose and multiple time-points (up to 168 hours) post-dose

Interventionhr*ng/mL (Mean)
Ixazomib+Lenalidomide+Dexamethasone685.9

[back to top]

Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Ixazomib

(NCT01645930)
Timeframe: Cycle 1, Day 1 pre-dose and multiple time-points (up to 168 hours) post-dose

Interventionhours (Median)
Ixazomib+Lenalidomide+Dexamethasone1.5

[back to top]

Number of Participants With Clinically Significant Vital Signs Reported as Adverse Events

The number of participants who meet markedly abnormal criteria for vital signs, included diastolic and systolic blood pressure, heart rate, oral temperature, respiratory rate, and body weight. (NCT01645930)
Timeframe: From the first dose of study drug through 30 days after the last dose of study drug (up to 577 days)

Interventionparticipants (Number)
Grade 1 or 2 HypertensionGrade 2 Hypotension
Ixazomib+Lenalidomide+Dexamethasone41

[back to top]

Number of Participants With Clinically Significant Laboratory Abnormalities Reported as Adverse Events of ≥Grade 3 Intensity

Clinically significant laboratory abnormalities were defined as any test results which were observed beyond the clinically acceptable limits as per the discretion of investigator. Clinical laboratory tests included chemistry, hematology and urinalysis tests. (NCT01645930)
Timeframe: From the first dose of study drug through 30 days after the last dose of study drug (up to 577 days)

Interventionparticipants (Number)
Alanine Aminotransferase IncreasedAspartate Aminotransferase IncreasedBlood Creatinine IncreasedHaemoglobin DecreasedNeutrophil Count DecreasedPlatelet Count DecreasedAnaemiaFebrile NeutropeniaNeutropeniaThrombocytopeniaHyperglycaemiaHypocalcaemiaHypokalaemiaHypomagnesaemiaHyponatraemiaHypophosphataemia
Ixazomib+Lenalidomide+Dexamethasone21112461128135112

[back to top]

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

An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. A SAE is defined as any untoward medical occurrence that at any dose: results in death, is life-threatening, requires inpatient hospitalization or causes prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, or was a medically important event. (NCT01645930)
Timeframe: From the first dose of study drug through 30 days after the last dose of study drug (up to 577 days)

Interventionparticipants (Number)
AEsSAEs
Ixazomib+Lenalidomide+Dexamethasone4318

[back to top]

Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for Ixazomib

(NCT01645930)
Timeframe: Cycle 1, Day 15 pre-dose and multiple time-points (up to 336 hours) post-dose

Interventionhours (Median)
Ixazomib+Lenalidomide+Dexamethasone2.0

[back to top]

Percentage of Participants With Confirmed Best Response Category

Percentage of participants who achieve or maintain any best response category during the treatment period were reported. Best response includes complete response (CR), very good partial response (VGPR), and partial response (PR). Response was assessed according to IMWG criteria. CR: negative immunofixation on serum and urine, disappearance of any soft tissue plasmacytomas and ≤5% plasma cells in bone marrow; PR: ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to <200 mg per 24 hours. VGPR: serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein plus urine M-protein level <100 mg per 24 hour. (NCT01645930)
Timeframe: From Cycle 1, Day 1 to Cycle 3, Day 1 until disease progression (approximately 20 months)

Interventionpercentage of participants (Number)
Ixazomib+Lenalidomide+Dexamethasone53.5

[back to top]

Number of Participants With Dose Limiting Toxicities (DLTs)

DLT was defined as any of the following AEs that were considered by investigator to be possibly related to therapy: 1. Grade 4 neutropenia lasting at least 7 consecutive days; 2. Grade 3 neutropenia with fever and/or infection; 3. Grade 4 thrombocytopenia at least 7 consecutive days; 4. Grade 3 thrombocytopenia with clinically significant bleeding; 5. Platelet count <10,000/mm^3; 6. Grade 2 peripheral neuropathy with pain or ≥Grade 3 peripheral neuropathy; 7. Grade 3 or greater nausea and / or emesis despite the use of optimal anti-emetic prophylaxis; 8. Grade 3 or greater diarrhea that occurred despite maximal supportive therapy; 9. Any other Grade 3 or greater nonhematologic toxicity with the following exceptions: Grade 3 arthralgia/myalgia, <1 week Grade 3 fatigue; 10. A delay of >2 weeks in the subsequent cycle of treatment; 11. Other combination study drug-related nonhematologic toxicities ≥Grade 2 that, in the opinion of the investigator, required discontinuation of study drug. (NCT01645930)
Timeframe: Cycle 1 (up to Day 28)

Interventionparticipants (Number)
Ixazomib+Lenalidomide+Dexamethasone2

[back to top]

Incidence of Immune Mediated Flare Reaction

Number of participants with Tumour flare. (NCT01649791)
Timeframe: 24 months

Interventionparticipants (Number)
Treatment (Lenalidomide as Chemoprevention)4

[back to top]

Median Progression-free Survival

(NCT01649791)
Timeframe: 24 months

Interventionmonths (Median)
Treatment (Lenalidomide as Chemoprevention)43.7

[back to top]

Overall Response Rate (CR+PR)

(NCT01649791)
Timeframe: 24 months

Interventionpercentage of participants (Number)
Treatment (Lenalidomide as Chemoprevention)100

[back to top]

Clinical Benefit Rate

The number of response rates in participants that have achieved MR, PR, VGPR, CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone20

[back to top]

The Best Overall Response Rate (ORR)

The primary endpoint will be the best overall response rate (ORR). 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. (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone11

[back to top]

Response Rates

"Response Rates evaluated using the International Uniform Response Criteria the International Myeloma Working Group (2003).~CR-Negative immunofixation on the serum and urine and Disappearance of any soft tissue plasmacytomas and 5% plasma cells in bone marrow VGPR-Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90 or greater reduction in serum M-component plus urine M-component <100 mg per 24 h PR-50% reduction of serum M-protein and reduction in 24-h urinary M-protein by 90% or to <200 mg per 24 h MR-≥ 25% but < 49% reduction of serum M protein and reduction in 24 hour urine M protein by 50 - 89%, which still exceeds 200 mg/24hrs. In addition; if present at baseline, 25-49% reduction in the size of soft tissue plasmacytomas also required No increase in size or number of lytic bone lesions.~SD-Not meeting criteria for CR, VGPR, PR or progressive disease PD-Laboratory or Biochemical Relapse increase of 25% from baseline" (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Complete Response (CR)Very Good Partial Response (VGPR)Partial Response (PR)Minimal Response (MR)Stable Disease (SD)Progressive Disease (PD)
Panobinostat, Lenalidomide and Dexamethasone245961

[back to top]

Response Rates for Len Refractory Patients

"Response Rates evaluated using the International Uniform Response Criteria the International Myeloma Working Group (2003).~CR-Negative immunofixation on the serum and urine and Disappearance of any soft tissue plasmacytomas and 5% plasma cells in bone marrow VGPR-Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90 or greater reduction in serum M-component plus urine M-component <100 mg per 24 h PR-50% reduction of serum M-protein and reduction in 24-h urinary M-protein by 90% or to <200 mg per 24 h MR-≥ 25% but < 49% reduction of serum M protein and reduction in 24 hour urine M protein by 50 - 89%, which still exceeds 200 mg/24hrs. In addition; if present at baseline, 25-49% reduction in the size of soft tissue plasmacytomas also required No increase in size or number of lytic bone lesions.~SD-Not meeting criteria for CR, VGPR, PR or progressive disease PD-Laboratory or Biochemical Relapse increase of 25% from baseline" (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Complete ResponseVery Good Partial ResponsePartial ResponseMinimal ResponseStable DiseaseProgressive Disease
Panobinostat, Lenalidomide and Dexamethasone143761

[back to top]

Disease Control Rate for Lens Refractory Rate

The number of response rates in Len refractory participants with SD, MR, PR, VGPR, or CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone21

[back to top]

Clinical Benefit Rate for Len Refractory Patients

The number of response rates in Lens Refractory participants that have achieved MR, PR, VGPR, CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone15

[back to top]

Overall Response Rate for Len Refractory Patients

The primary endpoint will be the best overall response rate (ORR) (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone8

[back to top]

Disease Control Rate

The number of response rates participants with SD, MR, PR, VGPR, or CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone26

[back to top]

Plasma Concentration of Ixazomib

As prespecified in the protocol, data for this outcome measure was planned to be collected for ixazomib arm group only. (NCT01659658)
Timeframe: Cycle 1, Day 1: 1, 4 hours postdose, Day 14: 144 hours postdose; Cycle 2, Day 1: predose, Day 14: 144 hours postdose; Cycles 3 to 10, Day 1: predose (cycle length=28 days)

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Cycle 1 Day 1: 1 Hour Post-doseCycle 1 Day 14: 4 Hours Post-doseCycle 1 Day 14: 144 Hours Post-doseCycle 2 Day 1: Pre-doseCycle 2 Day 14: 144 Hours Post-doseCycle 3 Day 1: Pre-doseCycle 4 Day 1 Pre-doseCycle 5 Day 1 Pre-doseCycle 6 Day 1: Pre-doseCycle 7 Day 1: Pre-doseCycle 8 Day 1: Pre-doseCycle 9 Day 1: Pre-doseCycle 10 Day 1: Pre-dose
Arm A: Ixazomib + Dexamethasone16.51810.6523.8752.0004.7262.1872.2762.2642.2352.2992.0382.1432.232

[back to top]

Time To Subsequent Anticancer Treatment

Time to subsequent anticancer therapy was defined as the time from randomization to the first date of subsequent anticancer therapy. Participants without subsequent anticancer therapy were censored at the date of death or last known to be alive. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until subsequent anticancer treatment (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone26.48
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide12.45

[back to top]

Percentage of Participants With Overall Hematologic Response

Overall hematologic response was defined as the percentage of participants with complete response (CR), very good partial response (VGPR) and partial response (PR) based on central laboratory results and the 2010 International Society of Amyloidosis (ISA) Consensus Criteria as assessed by an adjudication committee. CR: Complete disappearance of M-protein from serum and urine on immunofixation, and normalization of free light chain (FLC) ratio. VGPR: differential free light chain (difference between involved and uninvolved FLC levels; dFLC) < 40 mg/L. PR: ≥50% reduction in dFLC. Percentages were rounded off to the nearest decimal. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionpercentage of participants (Number)
Arm A: Ixazomib + Dexamethasone53
Arm B: Dexamethasone + Melphalan58
Arm B: Dexamethasone + Cyclophosphamide30
Arm B: Dexamethasone + Thalidomide50
Arm B: Dexamethasone + Lenalidomide51

[back to top]

Percentage of Participants With Complete Hematologic Response

Complete hematologic response was defined as the percentage of participants with CR based on central laboratory results and the 2010 ISA Consensus Criteria as assessed by the investigator. CR: Complete disappearance of M-protein from serum and urine on immunofixation, and normalization of FLC ratio. Percentages were rounded off to the nearest decimal. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionpercentage of participants (Number)
Arm A: Ixazomib + Dexamethasone30
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide17

[back to top]

Time To Treatment Failure (TTF)

TTF was defined as the time from randomization to the date of first documented treatment failure. Treatment failure was defined as: 1) death due to any cause; 2) hematologic progression or major organ progression according to central laboratory results and ISA criteria as evaluated by the investigator; 3) clinically morbid organ disease requiring additional therapy; or 4) withdrawn for any reason. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone10.32
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide5.32

[back to top]

Time to Vital Organ (Heart or Kidney) Deterioration and Mortality Rate

Time to vital organ deterioration or death was assessed by the investigator and defined as the time from randomization to vital organ (heart or kidney) deterioration or death, whichever occurs first. Cardiac deterioration is defined as the need for hospitalization for heart failure. Kidney deterioration is defined as progression to ESRD with the need for maintenance dialysis or renal transplantation. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From randomization to time of vital organ deterioration or death (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone38.67
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide26.09

[back to top]

Change From Baseline in Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACT/GOG-Ntx) Score at Week 28 of the PFS Follow-up

The FACT/GOG-Ntx is a participant completed questionnaire that comprises 11 individual items evaluating symptoms of neurotoxicity on a 5-point scale where: 0=not at all (best) to 4=very much for a total possible score of 0 to 44. Symptom scores are inverted so that higher scores of FACT/GOG-Ntx indicate higher quality of life or functioning. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: Baseline, Week 28 of the PFS Follow-up

Interventionscore on a scale (Mean)
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide0.0

[back to top]

2-Year Vital Organ (Heart or Kidney) Deterioration and Mortality Rate

Cardiac (Heart) deterioration was defined as the need for hospitalization for heart failure. Kidney deterioration was defined as progression to end-stage renal disease (ESRD) with the need for maintenance dialysis or renal transplantation. Vital organ deterioration was evaluated by an adjudication committee. Percentages were rounded off to the nearest decimal. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: Up to 2 years

Interventionpercentage of participants (Number)
Arm A: Ixazomib + Dexamethasone47
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide54

[back to top]

Change From Baseline in 36-item Short Form General Health Survey (SF-36) Mental Component Summary Score at Week 28 of the PFS Follow-up

SF-36 Version 2 is a multipurpose, participant completed, short-form health survey with 36 questions that consists of an 8-scale profile of functional health and well-being scores as well as psychometrically-based physical and mental health summary measures. Physical component summary (PCS) is mostly contributed by physical function (PF), role physical (RP), bodily pain (BP), and general health (GH). Mental component summary (MCS) is mostly contributed by mental health (MH), role emotional (RE), social function (SF), and vitality (VT). Each component on the SF-36 item health survey is scored from 0 (best) to 100 (worst). Total score ranges from 0-100, where higher scores are associated with less disability and better quality of life. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: Baseline, Week 28 of the PFS Follow-up

Interventionscore on a scale (Mean)
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide2.0

[back to top]

Change From Baseline in Amyloidosis Symptom Scale Total Score at Week 28 of the PFS Follow-up

The amyloidosis symptom scale questionnaire is a participant completed questionnaire that evaluates symptom severity of 3 symptoms: Swelling, Shortness of Breath and Dizziness, each rated on an 11-point scale where: 0=no symptoms to 10=very severe symptoms. Higher scores indicate worsening of symptoms. Total Score is the sum of all responses from the amyloidosis symptom scale ranging from 0 to 30. Higher scores represent higher levels of symptomatology or problems and a negative change from baseline indicates improvement. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: Baseline, Week 28 of the PFS Follow-up

Interventionscore on a scale (Mean)
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide-16.0

[back to top]

Duration of Hematologic Response

Duration of hematologic response (DOR) was defined as the time from the date of first documentation of a hematologic response to the date of first documented hematologic disease progression as determined by the investigator. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From time of first documented response to disease progression (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + DexamethasoneNA
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide21.19

[back to top]

EuroQol 5-Dimension 3-Level (EQ-5D-3L) Visual Analogue Scale Score

The EQ visual analogue scale (VAS) records the participant's self-rated health on a 20 centimeter vertical VAS that ranges from 0 (worst imaginable health state) to 100 (best imaginable health state). Baseline is defined as the value collected at the time closest to, but prior to, the start of study drug administration. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: At Week 28 of the OS follow-up

Interventionscore on a scale (Mean)
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide23.0

[back to top]

Number of Participants in Each Category of the EuroQol 5-Dimensional (EQ-5D) Questionnaire Score

The European Quality of Life (EuroQOL) 5-Dimensional (EQ-5D) is a patient completed questionnaire consisting of 2 pages: the EQ-5D descriptive system and the EQ visual analogue scale (EQ VAS). The descriptive system comprises 5 dimensions (mobility, self-care, usual activities, pain/discomfort, anxiety/depression). Each dimension has 3 possible choices: no problems to extreme problems. Higher scores=worsening of the quality of life. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: At Week 28 of the OS follow-up

,
InterventionParticipants (Count of Participants)
Mobility: No Problems in Walking AboutMobility: Some Problem in Walking AboutMobility: Confined to BedSelf-Care: No Problems With Self- CareSelf-Care: Some Problems Washing or DressingSelf-Care: Unable to Wash or DressUsual Activities: No Problems With Performing Usual ActivitiesUsual Activities: Some Problem With Performing Usual ActivitiesUsual Activities: Unable to Performing Usual ActivitiesPain/Discomfort: No Pain or DiscomfortPain/Discomfort: Moderate Pain or DiscomfortPain/Discomfort: Extreme Pain or DiscomfortAnxiety/Depression: Not Anxious or DepressedAnxiety/Depression: Moderately Anxious or DepressedAnxiety/Depression: Extremely Anxious or Depressed
Arm A: Ixazomib + Dexamethasone000000000000000
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide010010010010001

[back to top]

Hematologic Disease Progression Free Survival

Hematologic disease PFS was defined as the time from the date of randomization to the date of first documentation of hematologic PD according to central laboratory results and ISA criteria as evaluated by an adjudication committee, or death due to any cause, whichever occurred first. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone29.50
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide27.73

[back to top]

Number of Hospitalizations

A hospitalization was defined as at least one overnight stay in an intensive care unit and/or non-intensive care unit. If a single hospitalization included both an intensive care unit stay and a non-intensive care unit stay, the hospitalization was counted only once (as an intensive care unit stay). The mean number of hospitalizations is reported in this outcome measure. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionhospitalizations (Mean)
Arm A: Ixazomib + Dexamethasone1.8
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide1.4

[back to top]

Number of Participants With Serious Adverse Events (SAEs)

A SAE was defined as any untoward medical occurrence that at any dose resulted in death, was life-threatening, required inpatient hospitalization or prolongation of an existing hospitalization, resulted in persistent or significant disability or incapacity, was a congenital anomaly/birth defect or medically important event. (NCT01659658)
Timeframe: From first dose of study drug through 30 days after administration of the last dose of study drug (up to 115 months)

InterventionParticipants (Count of Participants)
Arm A: Ixazomib + Dexamethasone44
Arm B: Dexamethasone + Melphalan11
Arm B: Dexamethasone + Cyclophosphamide2
Arm B: Dexamethasone + Thalidomide0
Arm B: Dexamethasone + Lenalidomide17

[back to top]

Overall Survival

Overall survival was defined as the time from the date of randomization to the date of death. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone69.55
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide43.17

[back to top]

Percentage of Participants With Best Vital Organ (Cardiac and/or Kidney) Response

Vital organ (heart and kidney) response rate was defined as the percentage of participants who achieved vital organ response according to central laboratory results and ISA criteria as evaluated by an adjudication committee. A vital organ response was defined as response of 1 or 2 of the involved vital organs with no change from Baseline in the rest of involved vital organs. Percentages were rounded off to the nearest decimal. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionpercentage of participants (Number)
Arm A: Ixazomib + Dexamethasone19
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide12

[back to top]

Change From Baseline in SF-36 Physical Component Summary Score at Week 28 of the PFS Follow-up

SF-36 Version 2 is a multipurpose, participant completed, short-form health survey with 36 questions that consists of an 8-scale profile of functional health and well-being scores as well as psychometrically-based physical and mental health summary measures. Physical component summary (PCS) is mostly contributed by physical function (PF), role physical (RP), bodily pain (BP), and general health (GH). Mental component summary (MCS) is mostly contributed by mental health (MH), role emotional (RE), social function (SF), and vitality (VT). Each component on the SF-36 item health survey is scored from 0 (best) to 100 (worst). Total score ranges from 0-100, where higher scores are associated with less disability and better quality of life. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: Baseline, Week 28 of the PFS Follow-up

Interventionscore on a scale (Mean)
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide10.3

[back to top]

Vital Organ Progression Free Survival

Vital organ PFS is defined as the time from the date of randomization to the date of first documentation of progression of vital organ (heart or kidney) according to central laboratory results and ISA criteria as evaluated by an adjudication committee, or death due to any cause, whichever occurs first. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone15.77
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide11.01

[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 hematologic disease progression, or organ (cardiac or renal) progression, or death due to any cause, whichever occurred first according to central laboratory results and ISA criteria as evaluated by the investigator. As prespecified in the protocol, data was planned to be collected and analyzed in a combined way for non-ixazomib arm groups in this outcome measure. (NCT01659658)
Timeframe: From first dose of study drug until discontinuation of study drug due to disease progression or unacceptable toxicity, or death whichever occurs first (up to 115 months)

Interventionmonths (Median)
Arm A: Ixazomib + Dexamethasone11.86
Arm B: Dexamethasone + Melphalan/Cyclophosphamide/Thalidomide/Lenalidomide7.62

[back to top] [back to top]

Response (Partial Response [PR] or Better) Rate

Percentage of participants with PR or better to treatment per the International Uniform Response Criteria for Multiple Myeloma stringent Complete Response- CR criteria + normal serum free light chain (FLC) ratio + absence of clonal cells in bone marrow (BM) by immunohistochemistry or immunofluorescence CR- Negative immunofixation (IFX) on serum & urine M proteins + <5% plasma cells in BM + disappearance of soft tissue plasmocytomas (STP) very good PR- PR criteria + serum & urine M proteins detectable by IFX but not on electrophoresis or >=90% reduction (RED) in serum M protein & urine M protein <100 g/24 hrs PR- >=50% RED in size of STP, if present at baseline & >=50% RED in plasma cells, if >=30% plasma cells in BM at baseline: & RED in serum M protein of >=50% & in urine M protein of >= 90% or to 200 mg/24hr OR if serum M protein <1 g/dL, urine M protein <200 mg/24 hrs, & involved serum FLC level >= 10 mg/dl at baseline: >= 50% RED in (involved-uninvolved) serum FLC levels (NCT01668719)
Timeframe: Up to 6 years post registration

InterventionPercentage of participants (Number)
Arm I (Bortezomib, Lenalidomide, Dexamethasone)88
Arm II (Bortezomib, Lenalidomide, Dexamethasone, Elotuzumab)83

[back to top]

Progression-free Survival

"From date of registration to date of first documentation of progression or death due to any cause.~Per the International Uniform Response Criteria for Multiple Myeloma, progression is defined as >=1 of Serum M protein increase >= 25% from lowest response level, with an absolute increase of >= 0.5g/dL; Urine M protein increase >= 25% from lowest response level, with an absolute increase of >= 200 mg/24 hrs; If participant had serum M protein <1 g/dL, urine M protein <200 mg/24 hrs, and an involved serum free light chain level >= 10mg/dL at baseline: >= 25% increase in the difference between involved and uninvolved serum free light chain level with an absolute increase of >= 10 mg/dL; Bone marrow plasma cell % increase =25% from baseline with the absolute plasma cell % >=10%; New bone lesions or soft tissue plasmocytomas, or definite increase in size of existing bone lesions or soft tissue plasmocytomas; Development of hypercalcemia that can be attributed solely to multiple myeloma" (NCT01668719)
Timeframe: Up to 6 years post registration

Interventionmonths (Median)
Arm I (Bortezomib, Lenalidomide, Dexamethasone)33.6
Arm II (Bortezomib, Lenalidomide, Dexamethasone, Elotuzumab)31.5

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Elotuzumab in Combination With Bortezomib, Lenalidomide and Dexamethasone

"Assess safety of elotuzumab in combination with bortezomib, lenalidomide and dexamethasone and select the optimal dose of elotuzumab for the Phase II portion from 10mg/kg on each cycle, to 5mg/kg on each cycle MTD reflects the highest dose that had a dose-limiting toxicity (DLT) rate of ≤ 1/6 participants. DLTs were defined as treatment regimen related: grade ≥ 3 non-hematologic toxicity; grade 3 nausea or diarrhea despite anti-emetic and anti-diarrheal therapy; grade 3 hyperglycemia if symptomatic or glucose level > 300mg/ml despite insulin and/or oral diabetic therapy; grade 4 neutropenia ≥ 7 days or grade 3/4 neutropenia with fever (≥ 38.5 oC); grade 4 thrombocytopenia ≥ 7 days or associated with hemorrhage; delay of treatment with any agent by > 2 weeks due to drug related toxicity.~DLT were graded using the NCI CTCAE version 4.0~Note:~i) the second, lower dose level was not tested as the first dose level was deemed safe ii) 6 participants were evaluable at phase I analysis" (NCT01668719)
Timeframe: time from first participants randomized until at least 6 patients were evaluable for DLTs. DLTs were assessed only during Cycle 1 (21 days)

Interventionmg/kg (Phase II dosing for elotuzumab) (Number)
Phase I Dose Level 1 (Elotuzumab, 10mg, and Bortezomib, Lenalidomide and Dexamethasone)10

[back to top]

Overall Survival

From date of registration to date of death due to any cause (NCT01668719)
Timeframe: Up to 6 years post registration

Interventionmonths (Median)
Arm I (Bortezomib, Lenalidomide, Dexamethasone)NA
Arm II (Bortezomib, Lenalidomide, Dexamethasone, Elotuzumab)68

[back to top]

Number of Participants With Serious and Non-serious Adverse Events

Here is the number of 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. (NCT01675141)
Timeframe: 37 months and 12 days

InterventionParticipants (Count of Participants)
Lenalidomide Maintenance Therapy for Multiple Myeloma11

[back to top]

Progression Free Survival (PFS)

PFS is defined as the time from study entry until progression or death. Progression is assessed by the International Myeloma Workshop Consensus Panel Criteria. Progressive disease requires any one or more of the following: increase of ≥25% from baseline or lowest response value in Serum M component, Urine M component, free light chain or bone marrow plasma cell percentage. Lowest response value does not need to be a confirmed value. Serum M-component absolute increase must be ≥0.5 g/dl. The serum M-component increases of ≥1 gm/dl are sufficient to define relapse if starting M-component is ≥5 g/dl. Urine M-component absolute increase must be ≥200mg/24h. Only in patients without measureable serum and urine M-protein levels: the absolute increase in difference between involved and uninvolved free light chain levels must be >10mg/dl. (NCT01675141)
Timeframe: participants were followed for the duration of their treatment, an average of 2 years

Interventionmonths (Median)
Lenalidomide Maintenance Therapy for Multiple MyelomaNA

[back to top]

Duration of Response

Duration of response is defined as time from response to disease progression or death. Progression is assessed by the International Myeloma Workshop Consensus Panel Criteria. Progressive disease requires any one or more of the following: increase of ≥25% from baseline or lowest response value in Serum M component, Urine M component, free light chain or bone marrow plasma cell percentage. Lowest response value does not need to be a confirmed value. Serum M-component absolute increase must be ≥0.5 g/dl. The serum M-component increases of ≥1 gm/dl are sufficient to define relapse if starting M-component is ≥5 g/dl. Urine M-component absolute increase must be ≥200mg/24h. Only in patients without measureable serum and urine M-protein levels: the absolute increase in difference between involved and uninvolved free light chain levels must be >10mg/dl. (NCT01675141)
Timeframe: participants were followed for the duration of their treatment, an average of 2 years

InterventionMonths (Median)
Lenalidomide Maintenance Therapy for Multiple MyelomaNA

[back to top]

Time to Response

"Time to response was calculated for the responders as the time from the first dose date to the initial documented response (CR, VGPR or PR).~CR: Negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow; VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥ 90% reduction in serum M-protein and urine M-protein level < 100 mg/24 hours; PR: ≥ 50% reduction of serum M-Protein and reduction in urinary M-protein by ≥ 90% or to < 200 mg/24 hours. If present at baseline a ≥ 50% reduction in size of soft tissue plasmacytomas is also required." (NCT01698801)
Timeframe: From the first dose of study drug treatment until the data cut-off date of 15 July 2014. Median follow-up time was 61.6 weeks.

Interventionmonths (Median)
Lenalidomide Plus Dexamethasone1.97

[back to top]

Overall Survival (OS)

The time from the start of study treatment to death due to any cause. OS was censored at the last date that the participant was known to be alive for participants who were alive at the time of analysis and for participants who were lost to follow-up before death was documented. (NCT01698801)
Timeframe: From the first dose of study drug treatment until the data cut-off date of 15 July 2014. Median follow up is 14.2 months

Interventionmonths (Median)
Lenalidomide Plus Dexamethasone17.71

[back to top]

Duration of Response

Duration of response was calculated for the responders as the time from the initial documented response (CR or VGPR or PR) to the first documented progression or death due to any cause, whichever occurred first. Duration of response for participants last known to be alive with no progression after a CR, VGPR, or PR were censored at the date of last adequate response assessment. (NCT01698801)
Timeframe: From the first dose of study drug treatment until the data cut-off date of 15 July2014. Median follow up time was 61.6 weeks.

Interventionmonths (Median)
Lenalidomide Plus DexamethasoneNA

[back to top]

Overall Response Rate

"Number of Complete Responses (CR) plus Very Good Partial Response (VGPR) plus Partial Response (PR) based on the International Myeloma Working Group criteria (IMWG). Any participant who achieved a CR, VGPR, or PR while on study treatment was defined as a responder.~CR: Negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow; VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥ 90% reduction in serum M-protein and urine M-protein level < 100 mg/24 hours; PR: ≥ 50% reduction of serum M-Protein and reduction in urinary M-protein by ≥ 90% or to < 200 mg/24 hours. In addition to the above, if present at baseline a ≥ 50% reduction in the size of soft tissue plasmacytomas is also required." (NCT01698801)
Timeframe: From first dose until the data cut-off date of 15 July 2014. Median time on follow-up was 61.6 weeks.

Interventionpercentage of participants (Number)
Lenalidomide Plus Dexamethasone87.5

[back to top]

Number of Participants With Adverse Events

An adverse event is any noxious, unintended, or untoward medical occurrence that may appear or worsen in a participant during the course of a study. A serious AE is any AE occurring at any dose that: • Results in death; • Is life-threatening; • Requires or prolongs existing inpatient hospitalization; • Results in persistent or significant disability/incapacity; • Is a congenital anomaly/birth defect; • Constitutes an important medical event. The Investigator assessed the relationship of each AE to study drug and graded the severity according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE, Version 4.0): Grade 1 = Mild (no limitation in activity or intervention required); Grade 2 = Moderate (some limitation in activity; no/minimal medical intervention required);-Grade 3 = Severe (marked limitation in activity; medical intervention required, hospitalization possible); Grade 4 = Life-threatening; Grade 5 = Death. (NCT01698801)
Timeframe: From first dose of study drug treatment through to 28 days after the last dose, until the data cut-off date of 15 July 2014; median treatment duration was 60 weeks

Interventionparticipants (Number)
Any adverse eventTEAE related to study drugTEAE related to LenalidomideTEAE related to DexamethasoneGrade 3-4 adverse eventGrade 3-4 adverse event related to any study drugGrade 3-4 adverse event related to LenalidomideGrade 3-4 adverse event related to DexamethasoneSerious TEAESerious TEAE related to any study drugSerious TEAE related to LenalidomideSerious TEAE related to DexamethasoneTEAE leading to discontinuation of either drugTEAE leading to discontinuation of lenalidomideTEAE leading to discontinuation of dexamethasoneRelated TEAE discontinuation of either drugRelated TEAE discontinuation of lenalidomideRelated TEAE discontinuation of dexamethasone
Lenalidomide Plus Dexamethasone26252520181515411883442440

[back to top]

Progression Free Survival (PFS)

PFS was calculated as the time from the first dose date to the first documented progression based on IWG criteria or death due to any cause, whichever occurred first. If progression or death was not documented at the time of data cutoff date, these observations were censored at the last adequate assessment date showing evidence of no progression or death. (NCT01698801)
Timeframe: From the first dose of study drug treatment until the data cut-off date of 15 July 2014. Median follow-up for PFS assessments was 61.6 weeks.

Interventionmonths (Median)
Lenalidomide Plus DexamethasoneNA

[back to top]

Part A and B: Safety and Tolerability

(NCT01704781)
Timeframe: Part A: 31 days and Part B: 26 weeks

,,,,
Interventionparticipants (Number)
With any TEAEWith TESAEWith any AE resultiung in withdrawalDeaths
Part A: Lenalidomide 10 mg2000
Part A: Lenalidomide 5 mg3000
Part A: Lenalidopmide 25 mg6000
Part B: Lenalidomide10100
Part B: Lenalidomide Placebo9000

[back to top]

Part A: To Establish Highest Tolerated Dose of Lenalidomide, CD4 Counts Over Time

(NCT01704781)
Timeframe: 31 days

,,
Intervention10^6 cells/mL (Mean)
BaselineWeek 1Week 4Week 5
Part A: Lenalidomide 10 mg436.3317.0399.3449.0
Part A: Lenalidomide 5 mg437.3393.0442.0432.7
Part A: Lenalidopmide 25 mg454.2342.2450.7452.3

[back to top]

Part A: To Establish Highest Tolerated Dose of Lenalidomide, Dose-Limiting Toxicity

Number of participants in each of the three groups that experienced any dose-limiting toxicity. (NCT01704781)
Timeframe: 31 days

InterventionParticipants (Count of Participants)
Part A: Lenalidomide 5 mg0
Part A: Lenalidomide 10 mg0
Part A: Lenalidopmide 25 mg0

[back to top]

Part B: Incidents of Delayed-type Hypersensitivity

Delayed-type hypersensitivity measured by induration and erythema. (NCT01704781)
Timeframe: 26 weeks

,
InterventionParticipants (Count of Participants)
Week 1, IndurationWeek 1, ErythemaWeek 26, IndurationWeek 26, Erythema
Part B: Lenalidomide0056
Part B: Lenalidomide Placebo0126

[back to top]

Part B: Evaluate the Effect on HIV Viral Load

Results BLQ (<20 HIV copies/mL) have been replaced with BLQ/2 = 10 HIV copies/mL while 'not detected' results have been replaced with 0 HIV copies/mL. (NCT01704781)
Timeframe: 26 weeks

,
InterventionCopies/mL (Mean)
BaselineWeek 1Week 4Week 12Week 13Week 21Week 26
Part B: Lenalidomide0.85.16.45.57.41.73.5
Part B: Lenalidomide Placebo2.50.00.94.31.74.22.5

[back to top]

Part B: Change in CD8 Count

Change in CD8 count from baseline to week 26. (NCT01704781)
Timeframe: 26 weeks

,
Intervention10^6 cells/L (Mean)
BaselineWeek 1Week 4Week 12Week 13
Part B: Lenalidomide734.2653.8744.9823.0811.2
Part B: Lenalidomide Placebo655.3784.5794.5760.3663.6

[back to top]

Part B: Change in CD4 Count

Change in CD4 count from baseline to Week 26. (NCT01704781)
Timeframe: Week 26

,
Intervention10^6 cells/L (Mean)
ITTPP
Part B: Lenalidomide90.9106.3
Part B: Lenalidomide Placebo42.249.1

[back to top]

Survival Time

The distribution of survival time will be estimated by arm using the method of Kaplan-Meier. (NCT01706666)
Timeframe: From registration to death due to any cause, assessed up to 3 years

InterventionMonths to death (Number)
Arm ANA
Arm BNA
Arm CNA

[back to top]

Proportion of Patients Experiencing a Stringent Complete Response (sCR) After 12 Cycles, 24 Months

Estimated by the number of sCRs divided by the total number of evaluable patients in each arm. Exact binomial confidence intervals for the true sCR rate will be calculated by arm. Stringent complete response (sCR) is defined as a complete response plus normal serum free light chain ratio and the absence of clonal cells in bone marrow by flow cytometry. (NCT01706666)
Timeframe: 24 months

Interventionpercentage of participants (Number)
Arm A100
Arm B0
Arm C100

[back to top]

Progression-free Survival

The distribution of progression-free survival will be estimated by arm using the method of Kaplan-Meier. (NCT01706666)
Timeframe: From registration to the earliest date of documentation of disease progression or death due to any cause, assessed up to 3 years

InterventionMonths to Progression (Number)
Arm ANA
Arm B9.2
Arm CNA

[back to top]

Overall Response Rate

Analysis of the other Secondary Endpoints: The overall response rate will be estimated by the number of patients with complete and partial responses divided by the total number of evaluable patients. (NCT01723839)
Timeframe: 28 day cycle, up to 6 cycles

InterventionPercentage of Participants (Number)
FCR With Lenalidomide95

[back to top]

Complete Response

Analysis of the Primary Endpoint: The complete responses will be estimated by the number of patients with CR divided by the total number of evaluable patients. (NCT01723839)
Timeframe: 28 day cycle, up to 4 cycles

InterventionPercentage of Participants (Number)
FCR With Lenalidomide45

[back to top]

Time to Response

Time to Response was defined as the time from the first dose of study treatment to the initial documented response (CR or CRu, or PR) (NCT01724177)
Timeframe: From day 1 of study treatment to first documented response; up to data cut-off date of 19 May 2017; maximum study duration was 134.1 weeks

Interventionweeks (Median)
Lenalidomide8.10

[back to top]

Percentage of Participants Who Achieved a Complete Response, Unconfirmed Complete Response (CRu), Partial Response or Stable Disease (SD) as Assessed by the ESEC

The tumor control rate was measured for those with a response of Complete Remission, + CRu, + PR + Stable Disease (SD) in the EE population based on the best response. (NCT01724177)
Timeframe: From day 1 of study treatment to first documented response; up to data cut-off date of 19 May 2017; maximum study duration was 134.1 weeks

Interventionpercentage of participants (Number)
Lenalidomide73.1

[back to top]

Kaplan-Meier Estimate of Duration of Response (DOR) for Responders as Assessed by the ESEC

The response duration in participants with an objective response was measured from the date of the first Complete Response or Complete Response unconfirmed or Partial Response to the first date of Relapsed Disease or Progressive Disease (PD). For participants who did not progress during the study, DOR was censored at the last adequate response assessment not showing evidence of PD. (NCT01724177)
Timeframe: From day 1 of study treatment to first documented response; up to data cut-off date of 19 May 2017; Maximum study duration was 134.1 Weeks

Interventionweeks (Number)
Lenalidomide24.10

[back to top]

Kaplan-Meier Estimate for Overall Survival

Overall Survival was defined as the time from the start of study treatment to the death due to any cause. For participants who were still alive at the time of the data cutoff, survival data were censored at the latest available date the participant was known to be alive. (NCT01724177)
Timeframe: From Day 1 of study treatment to disease progression or death; up to final data cut-off date of 19 May 2017; maximum surivival time was 197.9 weeks

Interventionweeks (Median)
Lenalidomide88.10

[back to top]

Number of Participants With Treatment Emergent Adverse Events

Treatment Emergent Adverse Event (TEAE) was defined as any AE occurring on or after the start of study treatment and within 28 days after the last dose. Severity was assessed using National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 (NCI CTCAE v4.0): Grade 1= Mild Grade 2= Moderate Grade 3= Severe Grade 4= Life-threatening and Grade 5= Death related to AE. Serious AEs (SAEs) were those that resulted in death, were life-threatening, required or prolonged inpatient hospitalization, resulted in persistent or significant disability/incapacity, congenital anomaly, or resulted in an important medical event that may have jeopardized the patient or required medical or surgical intervention to prevent one of the outcomes listed above. (NCT01724177)
Timeframe: From the date of the first dose of study drug up to 28 days after the last dose of study drug; up to data cutoff date of 19 May 2017; maximum treatment duration was 130.1 weeks

Interventionparticipants (Number)
≥ 1 TEAE≥ 1 TEAE Related to Lenalidomide≥ 1 NCI CTCAE Grade (GR) 3 or Greater TEAE≥ 1 NCI CTCAE ≥ GR 3 TEAE Related to Lenalidomide≥ 1 Serious TEAE≥ 1 Serious TEAE Related to Lenalidomide≥ 1 TEAE Leading to Discontinuation≥ 1 Related TEAE Leading to Discontinuation≥ 1 TEAE Leading to Dose Reduction/Interruption≥ 1 related TEAE Leading to Decrease/Interruption≥ 1 TEAE Resulting in Death
Lenalidomide262625251198817170

[back to top]

Kaplan Meier Estimate of Progression Free Survival (PFS) as Assessed by the ESEC

PFS was defined as the time from the first dose of study treatment to progressive disease (PD) or death due to any cause on study or within 28 days after study discontinuation, whichever occurred earlier. (NCT01724177)
Timeframe: From day 1 of study treatment to the date of disease progression; up to data cut date of date of 19 May 2017; maximum study duration was 134.1 weeks

Interventionweeks (Median)
Lenalidomide16.30

[back to top]

Percentage of Participants Who Achieved a Complete Response, Unconfirmed Complete Response, or Partial Response as Assessed by the Efficacy-Safety Evaluation Committee (ESEC)

"ORR is a Complete Response (CR) + Complete Response unconfirmed (CRu) + Partial Response (PR). A CR requires that target lesions have regressed to normal; nodal non-target lesions have regressed to normal; extranodal non-target lesions have disappeared; hepatomegaly/splenomegaly has disappeared; skin findings are GR 0; peripheral blood is normal; Bone marrow (BM) infiltration is negative and no new lesions. A CRu requires the sum of the product diameters (SPD) of target lesions have decreased by at least 75% from baseline; nodal non-target lesions have regressed to normal size; extranodal non-target lesions have disappeared; hepatomegaly/splenomegaly has disappeared; skin findings are Grade 0; peripheral blood is normal; BM infiltration is negative and no new lesions. A PR requires the SPD of target lesions has decreased by at least 50% from baseline; all nodal non-target lesions have regressed to normal or show no increase in size; all extranodal non-target lesions have disappeared" (NCT01724177)
Timeframe: From day 1 of study treatment to date of first documented CR, CRU or PR; Up to data cut-off date of 19 May 2017; maximum study duration was 134.1 weeks

Interventionpercentage of participants (Number)
Lenalidomide42.3

[back to top]

Kaplan-Meier Estimate of Time to Progression (TTP)

Time to progression was calculated as the time from the first dosing of study treatment to the first documented PD and assessed by the ESEC (NCT01724177)
Timeframe: From day 1 of study treatment to the date of disease progression; up to data cut date of 19 May 2017; maximum study duration was 134.1 weeks

Interventionweeks (Median)
Lenalidomide16.30

[back to top]

QLQ-C30 Question 30

"Measured as mean quality of life in study subjects, quantitatively scored using the standardized and validated European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ). Question 30: How would you rate your overall quality of life during the past week?~7 point scores are standardized to a scale of 0-100, where 100 means highest possible quality." (NCT01729338)
Timeframe: baseline, 3 months, 5 months

Interventionunits on a scale (Mean)
Baseline3 months5 months
Velcade, Cyclophosphamide, Revlimid51.766.766.7

[back to top]

Overall Response Rate (ORR) During Induction Therapy

"Defined as percentage of patients achieving a partial response or better by International Myeloma Working Group (IMWG) standard criteria:~IMWG: Complete Response (CR): negative immunofixation on the serum and urine, no soft tissue plasmacytomas and <5% plasma cells in the bone marrow; stringent CR: CR+normal free light chain ratio, no clonal cells in bone marrow by immunohistochemistry or immunofluorescence; Very Good Partial Response: serum and urine M-protein detected by immunofixation but not electrophoresis, >90% in serum M-protein+urine, M-protein level <100 mg/24hour; Partial Response (PR): ≥50% decrease of serum and M-protein, 24 hour urinary M-protein decrease by ≥90% or <200 mg/24hour." (NCT01729338)
Timeframe: Up to 8 months

Interventionpercentage of participants (Number)
Velcade, Cyclophosphamide, Revlimid64

[back to top]

Median Time to Response

Defined as median time to achievement of response (partial remission or better by International Myeloma Working Group standard criteria), in study subjects during 8 months of induction chemotherapy. (NCT01729338)
Timeframe: Up to 8 months

Interventionmonths (Median)
Velcade, Cyclophosphamide, Revlimid2

[back to top]

QLQ-C30 Question 29

Measured as mean quality of life in study subjects, quantitatively scored using the standardized and validated European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ). Question 29: how would you rate your overall health during the past week? 7 point scores are standardized to a scale of 0-100, where 100 means highest possible quality. (NCT01729338)
Timeframe: baseline, 3 months, 5 months

Interventionunits on a scale (Mean)
Baseline3 months5 months
Velcade, Cyclophosphamide, Revlimid58.36568.3

[back to top]

Median Progression-free Survival

Defined as median time elapsed in study subjects between initiation of study therapy and either disease progression or death, regardless of cause of death. (NCT01729338)
Timeframe: 4 years

Interventionmonths (Median)
Velcade, Cyclophosphamide, Revlimid24.2

[back to top]

Median Overall Survival

Defined as median time elapsed in study subjects between initiation of study therapy and death, regardless of cause. (NCT01729338)
Timeframe: Up to 3 years

Interventionmonths (Median)
Velcade, Cyclophosphamide, Revlimid29.7

[back to top]

Median Duration of Response

Defined as median time elapsed in study subjects between achievement of response and disease progression. (NCT01729338)
Timeframe: From date of first confirmed response until date of disease progression or up to 3 years

Interventionmonths (Median)
Velcade, Cyclophosphamide, Revlimid26.2

[back to top]

Functionality as Assessed Using the Cancer and Leukemia Group B (CALGB) Geriatric Assessment Tool

"Mean functionality in study subjects, quantitatively scored using the standardized and validated Cancer and Leukemia Group B (CALGB) Geriatric Assessment Tool, which comprehensively assesses aspects of a patient's functionality such as symptoms (e.g., pain, anxiety), social support (e.g., someone to turn to for help), and ability to carry out routine activities (e.g., grocery shopping) or physical exertion (e.g., climbing stairs) was assessed at baseline.~The score is obtained by inserting patient-specific data into the online calculator found at http://www.mycarg.org/Chemo_Toxicity_Calculator, which is based on the risk score described in Hurria A, Togawa K, Mohile SG, et al. Predicting chemotherapy toxicity in older adults with cancer: a prospective multicenter study. J Clin Oncol. 2011;29(25):3457-3465.~The risk score ranges from 0-19, with 0-5 indicating low risk, 6-9 intermediate risk, and 10-19 high risk for severe (grade >2) toxicity." (NCT01729338)
Timeframe: baseline

InterventionScores on a scale (Mean)
Velcade, Cyclophosphamide, Revlimid10

[back to top]

Severe Adverse Event Rate

Defined as number of patients experiencing any grade or severe (≥ grade 3 by common toxicity criteria for adverse events (CTCAE) v4.0 criteria) adverse events at any time during the study (NCT01729338)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Velcade, Cyclophosphamide, Revlimid64

[back to top]

Maximum Depth of Response During Induction Therapy

Maximum depth of response is defined as: ranging from partial response to complete response by International Myeloma Working Group (IMWG). Described as number of patients achieving each level of response. IMWG: CR: negative immunofixation on the serum and urine, no soft tissue plasmacytomas and <5% plasma cells in the bone marrow; sCR: CR+normal free light chain ratio, no clonal cells in bone marrow by immunohistochemistry or immunofluorescence; VGPR: serum and urine M-protein detected by immunofixation but not electrophoresis, >90% in serum M-protein+urine, M-protein level <100 mg/24hour; PR: ≥50% decrease of serum and M-protein, 24 hour urinary M-protein decrease by ≥90% or <200 mg/24hour. Stable disease mean that the patient has not achieved CR, VGPR, PR or progressive disease. (NCT01729338)
Timeframe: Up to 8 months

Interventionpercentage of participants (Number)
Stable DiseasePartial ResponseVery good partial response (VGPR)Complete Response/Stringent complete responseProgressive Disease/no response
Velcade, Cyclophosphamide, Revlimid217292914

[back to top]

Maximum Depth of Response During Maintenance Therapy

"Defined as maximum depth of response (ranging from partial response to complete response by International Myeloma Working Group (IMWG) criteria at any point during post-induction maintenance therapy.~IMWG: Complete Response (CR): negative immunofixation on the serum and urine, no soft tissue plasmacytomas and <5% plasma cells in the bone marrow; stringent CR: CR+normal free light chain ratio, no clonal cells in bone marrow by immunohistochemistry or immunofluorescence; Very Good Partial Response: serum and urine M-protein detected by immunofixation but not electrophoresis, >90% in serum M-protein+urine, M-protein level <100 mg/24hour; Partial Response (PR): ≥50% decrease of serum and M-protein, 24 hour urinary M-protein decrease by ≥90% or <200 mg/24hour. Stable disease mean that the patient has not achieved CR, VGPR, PR or progressive disease." (NCT01729338)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Stable DiseasePartial ResponseVery good partial responseComplete Response/Stringent complete responseProgressive Disease/no response
Velcade, Cyclophosphamide, Revlimid13050380

[back to top]

Overall Survival Rate (OS)

To compare overall survival in subjects receiving autologous peripheral blood stem cell transplant after undergoing induction therapy with lenalidomide and dexamethasone versus in those receiving only lenalidomide and dexamethasone, followed by lenalidomide maintenance in both arms. Only patients who achieved at least a partial response (PR) following 4 cycles of induction were included in the analysis. (NCT01731886)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Arm A: Low-dose Dexamethasone + Stem Cell Transplantation100
Arm B: Low-dose Dexamethasone94.7

[back to top]

Overall Survival Rate (OS)

To compare overall survival in subjects receiving autologous peripheral blood stem cell transplant after undergoing induction therapy with lenalidomide and dexamethasone versus in those receiving only lenalidomide and dexamethasone, followed by lenalidomide maintenance in both arms. Only patients who achieved at least a partial response (PR) following 4 cycles of induction were included in the analysis. (NCT01731886)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Arm A: Low-dose Dexamethasone + Stem Cell Transplantation79.8
Arm B: Low-dose Dexamethasone78.9

[back to top]

Progression Free Survival (PFS)

PFS is the length of time during and after the treatment of a disease, such as cancer, that a patient lives with the disease but it does not get worse. (NCT01731886)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Arm A: Low-dose Dexamethasone + Stem Cell Transplantation52.0
Arm B: Low-dose Dexamethasone47.4

[back to top]

Progression Free Survival (PFS)

PFS is the length of time during and after the treatment of a disease, such as cancer, that a patient lives with the disease but it does not get worse. (NCT01731886)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Arm A: Low-dose Dexamethasone + Stem Cell Transplantation36.0
Arm B: Low-dose Dexamethasone31.6

[back to top]

Complete Response Rate

The primary objective of this study is to determine the complete response rate of lenalidomide and low-dose dexamethasone versus that of lenalidomide and low-dose dexamethasone followed by autologous peripheral blood stem cell transplant in patients with newly diagnosed multiple myeloma (will include unconfirmed complete response (CR), CR and stringent complete response (sCR)). (NCT01731886)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Arm A: Low-dose Dexamethasone + Stem Cell Transplantation7
Arm B: Low-dose Dexamethasone7

[back to top]

Percentage of Participants With Complete Remission or Complete Remission With Incomplete Recovery Blood Counts

Change in baseline to end of study. To be assessed by standard criteria based on bone marrow examination. 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 (NCT01743859)
Timeframe: Interim assessment after 18 patients (estimated 2 years) and full assessment after 37 patients (estimated 3-4 years)

Interventionpercentage of participants (Number)
Azacitidine + Lenalidomide + Off Therapy11

[back to top]

Progression-free Survival

Change in baseline to end of study. To be assessed by standard criteria based on bone marrow examination (NCT01743859)
Timeframe: Depending on outcomes, will initiate this assessment after 2 years and will continue until completion of study, estimated at 4 years

Interventiondays (Median)
Azacitidine + Lenalidomide + Off Therapy112

[back to top]

Response or Remission Duration

Change in baseline to end of study. To be assessed by standard criteria based on bone marrow examination (NCT01743859)
Timeframe: Depending on outcomes, will initiate this assessment after 2 years and will continue until completion of study, estimated at 4 years

Interventiondays (Median)
Azacitidine + Lenalidomide + Off Therapy125

[back to top] [back to top]

Determine Biomarkers That Predict Response/Toxicity

Change in baseline to end of study. Planned assessments of methylation changes and other biomarkers. Computational biology modeling used to identify biomarkers and predict response. (NCT01743859)
Timeframe: Three years after initiating study

Interventionpatients w/response predictor mutations (Number)
Azacitidine + Lenalidomide + Off Therapy9

[back to top]

Overall Response Rate

Change in baseline to end of study. To be assessed by standard criteria based on bone marrow examination (NCT01743859)
Timeframe: Planned assessment after enrollment of all 37 patients (estimated 3-4 years)

Interventionpercentage of participants (Number)
Azacitidine + Lenalidomide + Off Therapy52

[back to top]

Overall Survival

Change in baseline to end of study (NCT01743859)
Timeframe: Depending on outcomes, will begin assessment at 2 years and will continue until completion of study, estimated to be at four years

Interventiondays (Median)
Azacitidine + Lenalidomide + Off Therapy166

[back to top]

Time to Progression

The primary objective is progression-free survival (PFS). Tumor measurements and disease assessments will be performed at the time of screening, following cycles 3 and 6 of induction chemotherapy, every 4 cycles during the maintenance portion of treatment, and at the end of treatment (EOT). Subjects with clinical evidence of progression prior to a planned disease assessment will be evaluated at the time of clinically suspected progression. Follow-up visits for disease assessment will occur every 3 months after the EOT visit until PD, initiation of alternate anti-neoplastic therapy, decision by the subject to withdraw from the study, or death. The follow-up period will begin after the EOT visit, and all subjects will be followed for at least 2 years after completion of therapy or until death or progression and until the last patient has been followed for at least 1 year following completion of therapy. (NCT01754857)
Timeframe: At least 24 months following completion of therapy, an average of 5 years

Interventionyears (Median)
Bendamustine, Rituximab, Lenalidomide4.76

[back to top] [back to top]

Number of Patients With Hematologic Improvement in Platelet Counts (HI-P)

The Number of Patients with Hematologic Improvement in Platelet Counts (HI-P) was assessed based on the MDS 2006 IWG criteria. Patients demonstrating an absolute increase of ≥ 30 × 10^9/L (for those patients starting with > 20 × 10^9/L platelets) or an increase from < 20 × 10^9/L to > 20 × 10^9/L along with an increase of at least 100%, were deemed to have demonstrated HI-P improvement. (NCT01772420)
Timeframe: Periodic evaluation (weekly up to a month, followed by 4x28 day cycles = 16weeks) with additional cycles and titrations depending upon treatment response; up to 2 years

InterventionParticipants (Count of Participants)
Arm A (Platelets > 50,000 k/uL) LEN Initiation0
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination3
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy6

[back to top]

Number of Patients With Clinically Significant Bleeding Events

Number of Patients With Clinically Significant Bleeding Events (NCT01772420)
Timeframe: Treatment initiation through study completion, up to 2 years

InterventionParticipants (Count of Participants)
Arm A (Platelets > 50,000 k/uL) LEN Initiation0
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination2
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy0

[back to top]

Number of Patients With Hematologic Improvement in Neutrophil Counts (HI-N)

The Number of Patients with Hematologic Improvement in Neutrophil Counts (HI-N) was assessed based on the MDS 2006 IWG criteria. Patients demonstrating an increase of at least 100% and an absolute increase > 0.5 × 10^9/L were determined to have shown an improvement in HI-N. (NCT01772420)
Timeframe: Periodic evaluation (weekly up to a month, followed by 4x28 day cycles = 16weeks) with additional cycles and titrations depending upon treatment response; up to 2 years

InterventionParticipants (Count of Participants)
Arm A (Platelets > 50,000 k/uL) LEN Initiation0
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination1
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy2

[back to top]

Number of Patients Demonstrating Overall Hematologic Improvement (HI)

The number of patients demonstrating overall Hematologic Improvement (HI) was assessed based on the MDS 2006 IWG criteria. The IWG criteria for HI define specific responses of cytopenia in the 3 hematopoietic lineages: erythroid (HI-E), platelet (HI-P), and neutrophil (HI-N) as demonstrated in corresponding outcome measures. Responses must have sustained for at minimum of 8 weeks for the participant to be included in the tally. (NCT01772420)
Timeframe: Periodic evaluation (weekly up to a month, followed by 4x28 day cycles = 16weeks) with additional cycles and titrations depending upon treatment response; up to 2 years

InterventionParticipants (Count of Participants)
Arm A (Platelets > 50,000 k/uL) LEN Initiation6
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination5
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy7

[back to top]

Duration of Hematologic Improvement (HI)

Duration to hematologic improvement as determined by median duration of HI response. (NCT01772420)
Timeframe: Time to progression/relapse following hematologic improvement, at completion of final cycle and treatment discontinuation; up to 6 years

Interventionnumber of weeks (Median)
Arm A (Platelets > 50,000 k/uL) LEN Initiation41
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination88
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy40

[back to top]

Time to Attain Hematologic Improvement (HI)

Time to hematologic improvement as determined by median time required to achieve HI response. (NCT01772420)
Timeframe: Periodic evaluation (weekly up to a month, followed by 4x28 day cycles = 16weeks) with additional cycles and titrations depending upon treatment response; up to 2 years

Interventionnumber of weeks (Median)
Arm A (Platelets > 50,000 k/uL) LEN Initiation12
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination8
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy8

[back to top]

Number of Patients With Hematologic Improvement in Erythrocyte Counts (HI-E)

The Number of Patients with Hematologic Improvement in Erythrocyte Counts (HI-E) was assessed based on the MDS 2006 IWG criteria. Patients demonstrating an Hgb increase by ≥ 1.5 g/dL were deemed to have improvement in HI-E. Only transfusions given for a Hgb of ≤ 9.0 g/dL pretreatment were counted in the RBC transfusion response. (NCT01772420)
Timeframe: Periodic evaluation (weekly up to a month, followed by 4x28 day cycles = 16weeks) with additional cycles and titrations depending upon treatment response; up to 2 years

InterventionParticipants (Count of Participants)
Arm A (Platelets > 50,000 k/uL) LEN Initiation6
Arm B (Platelets < 50,000 k/uL) ELT Initiation With Potential LEN Combination3
Arm C (Platelets < 50,000 k/uL) ELT Monotherapy4

[back to top]

Number of Patients Who Demonstrate a Response (Complete, Partial, Minor) to Treatment

"Response criteria for subjects with WM is based upon the Consensus Panel Recommendations from the Third International Workshop on Waldenstrom Macroglobulinemia.~Overall response rate (CR + PR + MR) measured at time of best response." (NCT01779167)
Timeframe: Approximately 24 months

Interventionparticipants (Number)
All Patients0

[back to top]

Mean Plasma Bortezomib Concentration Following Intravenous and and Subcutaneous Injection

The pharmacokinetic profile of intravenous and subcutaneous bortezomib administration in combination with lenalidomide and dexamethasone. (NCT01782963)
Timeframe: Day 1 (5 min, 30min, 5 hours post dose), Days 8, 15, Day 22 (pre dose and 5 min, 30 min, 5 hrs post dose), cycle 2 day 1 pre dose

,
Interventionng/mL (Mean)
Day 1, 5 minutes (min) post doseDay 1, 30 min post doseDay 1, 5 hours post doseDay 8 pre-doseDay 15 pre-doseDay 22 pre-doseDay 22, 5 min post-doseDay 22, 30 min post-doseDay 22, 5 hours post-doseCycle 2 Day 1, pre-dose
Intravenous98.848.731.820.440.660.83114.314.92.850.53
Subcutaneous1320.51.960.330.490.6610.2921.694.160.49

[back to top]

Median Overall Survival

The median overall survival as measured from the start of treatment until the time of death due to any cause. (NCT01782963)
Timeframe: From the start of treatment until death or until 5 years after the time of disease progression

Interventionyears (Median)
Treatment ArmNA

[back to top]

Median Progression Free Survival

"The median amount of time as measured from the start of treatment until either death or progression.~Progressive disease requires 1 or more of the following:~>=25% increase from lowest response level in serum M-protein (>=0.5 g/dL absolute increase) and/or urine M-component (>=200 mg/24hr absolute increase)~>=25% increase in the difference between involved and uninvolved FLC levels (absolute increase >10 mg/dL). Only for use in patients without measurable serum and M-protein levels.~>=25% increase in bone marrow plasma cell percentage (absolute percentage >=10%)~New or increase in existing bone lesions or soft tissue plasmacytomas~Hypercalcemia (serum calcium >11.5 mg/dL) due solely to the plasma cell proliferative disorder." (NCT01782963)
Timeframe: From the start of treatment until death or progression or until 3 years after the last participant is enrolled

InterventionMonths (Median)
Treatment Arm35.1

[back to top]

Median Time to Response

"Median amount of time from the start of treatment until first documented response as defined by the International Myeloma Working Group uniform response criteria~Stringent CR: Same as CR plus normal free light chain ratio and absence of clonal cells plasma cells in bone marrow (BM) CR: Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and <5% plasma cells in BM VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hours PR: ≥50% reduction of serum M-protein and reduction in 24-h urinary M-protein by ≥90% or to <200 mg per 24 hours. If the serum and urinary M-protein are not measurable, additional criteria are used to assess PR that will not fit in the space provided here. If present at baseline, a ≥50% reduction in the size of plasmacytomas is also required" (NCT01782963)
Timeframe: From the start of treatment until the time of first documented response, median duration of 1.1 months

InterventionMonths (Median)
Treatment Arm1.1

[back to top] [back to top]

Objective Response Rate

"Participants are considered to have achieved an objective response if they meet the International Myeloma Working Group uniform response criteria for any of the following:~Stringent CR: Same as CR plus normal free light chain ratio and absence of clonal cells plasma cells in bone marrow (BM)~CR: Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and <5% plasma cells in BM~VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level <100 mg per 24 hours~PR: ≥50% reduction of serum M-protein and reduction in 24-h urinary M-protein by ≥90% or to <200 mg per 24 hours. If the serum and urinary M-protein are not measurable, additional criteria are used to assess PR that will not fit in the space provided here. If present at baseline, a ≥50% reduction in the size of plasmacytomas is also required" (NCT01782963)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Stringent Complete ResponseComplete Response (CR)Very Good Partial Response (VGPR)Partial Response (PR)
Treatment Arm6161110

[back to top]

Proportion of Confirmed Tumor Responses Defined to be a Partial Response or Better Noted as the Objective Status on Two Consecutive Evaluations

The proportion of successes will be estimated in each arm independently by the number of successes divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated according to the approach of Duffy and Santner. A confirmed tumor response is defined to be a partial response or better noted as the objective status on two consecutive evaluations while receiving lenalidomide and dexmethasone (Arm A) or pomalidomide and dexamethasone (Arm B). All patients meeting the eligibility criteria who have signed a consent form and have begun treatment will be evaluable for response.The International Myeloma Working Group (IMWG) uniform response criteria (Rajkumar et al, 2011) will be used to assess response to therapy. (NCT01794039)
Timeframe: Up to 2 years

Interventionproportion of participants (Number)
Arm A (Lenalidomide, Dexamethasone).4
Arm B (Pomalidomide, Dexamethasone).25

[back to top]

Time to Progression

The distribution of time to progression will be estimated using the method of Kaplan-Meier. The International Myeloma Working Group (IMWG) uniform response criteria (Rajkumar et al, 2011) will be used to assess response to therapy. (NCT01794039)
Timeframe: Time from registration to the earliest date with documentation of disease progression, assessed up to 2 years

InterventionMonths (Median)
Arms A and B10

[back to top]

Number of Participants With Adverse Events, Graded According to the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0

Recorded and reported for each patient, and frequency tables will be reviewed to determine adverse event patterns. These results are reported in the Adverse Events section of this CT.gov report. (NCT01794039)
Timeframe: Up to 30 days after last day of study drug treatment

InterventionParticipants (Count of Participants)
Arm A (Lenalidomide, Dexamethasone)5
Arm B (Pomalidomide, Dexamethasone)4

[back to top]

Overall Survival

The distribution of survival time will be estimated using the method of Kaplan-Meier. Due to an early closer from slow accrual the data from both arms was combined in survival analysis. (NCT01794039)
Timeframe: Time from registration to death due to any cause, assessed up to 2 years

InterventionMonths (Median)
Arms A and B13.4

[back to top]

Percentage of Participants With Progression-free Survival (PFS)

Progression-free Survival rate was estimated at months 12, 24, 36, 48, and 60, by the product-limit method of Kaplan and Meier. (NCT01816971)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
PFS rate at 12 months (%)PFS rate at 24 months(%)PFS rate at 36 months(%)PFS rate at 48 months(%)PFS rate at 60 months(%)
Treatment (Dexamethasone, Carfilzomib, Lenalidomide)9793807672

[back to top]

Percentage of Participants With Overall Survival (OS)

Overall survival rate was estimated at months 12, 24, 36, 48, and 60, by the product-limit method of Kaplan and Meier. (NCT01816971)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
OS rate at 12 months (%)OS rate at 24 months (%)OS rate at 36 months (%)OS rate at 48 months (%)OS rate at 60 months (%)
Treatment (Dexamethasone, Carfilzomib, Lenalidomide)10097969384

[back to top]

Time to Progression

Estimated using the product-limit method of Kaplan and Meier. (NCT01816971)
Timeframe: Up to 5 years

Interventionmonth (Median)
Treatment (Dexamethasone, Carfilzomib, Lenalidomide)NA

[back to top]

Percentage of Patients Achieving sCR

The percentage of stringent complete response (CR) (sCR) will be reported along with 95% confidence intervals, adjusted for the two-stage nature of the trial design. (NCT01816971)
Timeframe: Day 224

Interventionpercentage of participants (Number)
Treatment (Dexamethasone, Carfilzomib, Lenalidomide)76

[back to top]

Overall Response Rate, Defined as at Least a Partial Response to Therapy (> PR), at Least Very Good Partial Response (VGPR) and at Least Near Complete Response (nCR) Rate

Reported along with its exact 95% binomial confidence interval. (NCT01816971)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Treatment (Dexamethasone, Carfilzomib, Lenalidomide)97

[back to top]

Duration of Response

Reported along with its exact 95% binomial confidence interval. Estimated using the product-limit method of Kaplan and Meier. (NCT01816971)
Timeframe: Up to 5 years

Interventionmonth (Median)
Treatment (Dexamethasone, Carfilzomib, Lenalidomide)NA

[back to top]

Cmax: Maximum Plasma Concentration for Ixazomib

(NCT01850524)
Timeframe: Cycle 1 Day 1: Post-dose at multiple timepoints up to 4 hours; Pre-dose at Cycle 1 Day 14, Cycles 2-3 Day 1 and Day 14, Cycles 4-11 Day 1 (Each cycle length = 28 days)

Interventionnanograms per milliliter (Mean)
Cycle 1 Day 1: 1 Hour Post-doseCycle 1 Day 1: 4 Hours Post-doseCycle 1 Day 14: Pre-doseCycle 2 Day 1: Pre-doseCycle 2 Day 14: Pre-doseCycle 3 Day 1: Pre-doseCycle 3 Day 14: Pre-doseCycle 4 Day 1: Pre-doseCycle 5 Day 1: Pre-doseCycle 6 Day 1: Pre-doseCycle 7 Day 1: Pre-doseCycle 8 Day 1: Pre-doseCycle 9 Day 1: Pre-doseCycle 10 Day 1: Pre-doseCycle 11 Day 1: Pre-doseCycle 12 Day 1: Pre-dose
Ixazomib44.74516.2537.8672.6648.5212.7638.4903.2843.5942.6032.5982.5392.5932.5362.6672.686

[back to top]

Number of Participants With Abnormal Serum Chemistry and Hematology Laboratory Values Based on Treatment-emergent Adverse Events (TEAEs)

The laboratory values assessment included serum chemistry and hematology. The Serum chemistry assessment included blood urea nitrogen (BUN), creatinine, bilirubin (total), urate, lactate dehydrogenase, phosphate, albumin, alkaline phosphatase (ALP), aspartate aminotransferase (AST), alanine aminotransferase (ALT), glucose, sodium, potassium, calcium, chloride, carbon dioxide (CO2), magnesium, thyroid stimulating hormone (TSH). Hematology assessment included hemoglobin, hematocrit, platelet (count), leukocytes with differential neutrophils (ANC). Participants with abnormal serum chemistry laboratory values reported as TEAEs are reported. TEAEs were defined as events that occurred after administration of the first dose of any agent in the study drug regimen and through 30 days after the last dose of any agent in the study drug regimen. (NCT01850524)
Timeframe: From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)

,,,
InterventionParticipants (Count of Participants)
HypokalaemiaBlood creatinine increasedHypophosphataemiaHypomagnesaemiaHyponatraemiaHyperglycaemiaHypocalcaemiaHyperkalaemiaAlanine aminotransferase increasedIron deficiencyHypercalcaemiaCreatinine renal clearance decreasedHypoalbuminaemiaAspartate aminotransferase increasedHyperuricaemiaAnaemiaThrombocytopeniaNeutropeniaNeutrophil count decreasedPlatelet count decreasedLymphopeniaFebrile neutropeniaLeukopeniaInternational normalised ratio increasedPancytopeniaIron deficiency anaemiaWhite blood cell count decreasedLymphocyte count decreased
Ixazomib + LenDex (Exposure ≥19 Cycles)3916915764310754352582439181542242420
Ixazomib+ LenDex (Exposure Up to 18 Cycles)336961076711212025334155677643112
Placebo + LenDex (Exposure ≥19 Cycles)3312311816123421713152144813422401133
Placebo + LenDex (Exposure Up to 18 Cycles)1692874133126251257153611605312151

[back to top]

Number of Participants With Shifts From Baseline to Worst Value in Eastern Cooperative Oncology Group (ECOG) Performance Score

Eastern Cooperative Oncology Group (ECOG) scale score ranged from 0 to 5, where 0 indicated normal activity and 5 indicated death. The data is reported for those categories where at least 1 participant had worst post-baseline value for each ECOG score. (NCT01850524)
Timeframe: Up to approximately 9 years

,
InterventionParticipants (Count of Participants)
Baseline Score 0, Post-Baseline Score 0Baseline Score 0, Post-Baseline Score 1Baseline Score 0, Post-Baseline Score 2Baseline Score 0, Post-Baseline Score 3Baseline Score 1, Post-Baseline Score 0Baseline Score 1, Post-Baseline Score 1Baseline Score 1, Post-Baseline Score 2Baseline Score 1, Post-Baseline Score 3Baseline Score 1, Post-Baseline Score 4Baseline Score 2, Post-Baseline Score 1Baseline Score 2, Post-Baseline Score 2Baseline Score 2, Post-Baseline Score 3Baseline Score 2, Post-Baseline Score 4
Ixazomib + LenDex2352231011045794835113
Placebo + LenDex235720219672186122872

[back to top]

Pain Response Rate as Assessed by the Brief Pain Inventory- Short Form (BPI-SF) and Analgesic Use

Pain response rate was defined as percentage of participants with pain response. Pain response was defined as the occurrence of at least a 30% reduction from baseline in BPI-SF worst pain score over the last 24 hours without an increase in analgesic use for 2 consecutive measurements > 28 days apart, were reported. Brief Pain Inventory - Short Form (m-BPI-SF) is a participant rated 11-point Likert rating scale ranged from 0 (no pain) to 10 (worst pain imaginable). Percentages are rounded off to the nearest single decimal. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionpercentage of participants (Number)
Placebo + LenDex51.3
Ixazomib + LenDex50.5

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

An AE was any untoward medical occurrence in a participant administered a medicinal investigational drug. The untoward medical occurrence does not necessarily have to have a causal relationship with treatment. An SAE is any untoward medical occurrence that results in death; is life-threatening; requires inpatient hospitalization or prolongation of present hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect or is a medically important event that may not be immediately life-threatening or result in death or hospitalization, but may jeopardize the participant or may require intervention to prevent one of other outcomes listed in definition above, or involves suspected transmission via a medicinal product of an infectious agent. (NCT01850524)
Timeframe: From the date of randomization through 30 days after the last dose of study drug up to end of study (up to approximately 9 years)

,,,
InterventionParticipants (Count of Participants)
TEAEsSAEs
Ixazomib + LenDex (Exposure ≥19 Cycles)191125
Ixazomib+ LenDex (Exposure Up to 18 Cycles)163119
Placebo + LenDex (Exposure ≥19 Cycles)189119
Placebo + LenDex (Exposure Up to 18 Cycles)160105

[back to top]

Time to Response

Time to response was defined as the time from the date of randomization to the first documentation of PR or better, as measured by IMWG criteria. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionmonths (Median)
Placebo + LenDex1.87
Ixazomib + LenDex1.02

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of randomization to the date of death. Participants without documented death at the time of analysis are censored at the date last known to be alive. (NCT01850524)
Timeframe: From the date of randomization to death due to any cause (Up to approximately 9 years)

Interventionmonths (Median)
Placebo + LenDexNA
Ixazomib + LenDexNA

[back to top]

Overall Response Rate (ORR)

ORR was defined as the percentage of participants who achieved CR + partial response (PR) + very good partial response (VGPR) (including sCR) or better relative to the ITT population during treatment period. CR was defined as negative immunofixation of serum and urine along with the disappearance of any soft tissue plasmacytomas and <5 % PC's in bone marrow. PR was defined as ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% along with ≥50% reduction in the size of soft tissue plasmacytomas. VGPR was defined as ≥90% in serum M-component plus urine M-component <100 mg/24. sCR is defined as stringent complete response. Percentages are rounded off to nearest whole numbers. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionpercentage of participants (Number)
Placebo + LenDex80
Ixazomib + LenDex82

[back to top]

OS in High-risk Population Carrying Del(17p), t(4;14), or t(14;16) Mutations

OS was defined as the time from the date of randomization to the date of death, as assessed in high-risk population carrying del(17p), t(4;14), or t(14;16) mutations. High risk category includes t(4;14), t(14;16), or del(17) abnormalities. (NCT01850524)
Timeframe: From the date of randomization to death due to any cause (Up to approximately 9 years)

Interventionmonths (Median)
Placebo + LenDex43.1
Ixazomib + LenDex39.0

[back to top]

Duration of Response

Duration of response was measured as the time from the date of first documentation of PR or better to the date of first documented progression (PD) for responders, as measured by IMWG criteria. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionmonths (Median)
Placebo + LenDex37.5
Ixazomib + LenDex50.6

[back to top]

Complete Response (CR) Rate

CR rate was defined as the percentage of participants who achieve CR assessed by an IRC relative to the intent-to-treat (ITT) population during the treatment period. Percentage of participants with CR, as assessed by IMWG disease assessment criteria were reported. CR was defined as negative immunofixation of serum and urine along with the disappearance of any soft tissue plasmacytomas and <5 % plasma cells (PC's) in bone marrow. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionpercentage of participants (Number)
Placebo + LenDex14
Ixazomib + LenDex26

[back to top] [back to top]

Percentage of Participants With MRD-Negative Status as Assessed by Flow Cytometry

The absence of minimal residual disease (MRD negativity) was tested in all participants who achieve a CR and maintained it until Cycle 18, using bone marrow aspirates. (NCT01850524)
Timeframe: Up to Cycle 18 (cycle length = 28 days)

Interventionpercentage of participants (Number)
Placebo + LenDex50
Ixazomib + LenDex59

[back to top] [back to top]

PFS in High-risk Population Carrying Del(17p), t(4;14), or t(14;16) Mutations

PFS was defined as the time from the date of randomization to the date of first documentation of progressive disease based on central laboratory results and IMWG criteria as evaluated by an independent review committee (IRC) or death due to any cause, whichever occurs first, as assessed in high-risk population carrying del(17p), t(4;14), or t(14;16) mutations. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionmonths (Median)
Placebo + LenDex17.5
Ixazomib + LenDex22.4

[back to top]

Time to Progression (TTP)

Time to progression was defined as the time from randomization to the date of first documented disease progression. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionmonths (Median)
Placebo + LenDex26.8
Ixazomib + LenDex45.8

[back to top]

Change From Baseline in HRQOL Measured by EORTC-QLQ-MY20 Scale

EORTC QLQ-MY20 was a validated questionnaire to assess the overall quality of life in participants with multiple myeloma. The scale has 20 questions. Subscale and individual items include future perspective items 18-20, body image item 17, disease symptoms items 1-6, side effects of treatment items 7-16. Raw scores are averaged, and transformed to 0-100 scale, where higher score is better quality of life. Positive change indicates improvement. (NCT01850524)
Timeframe: Baseline to approximately 9 years

,
Interventionscore on a scale (Mean)
Disease Symptoms: BaselineDisease Symptoms: End of TreatmentSide-Effects: BaselineSide-Effects: End of TreatmentBody Image: BaselineBody Image: End of TreatmentFuture Perspective: BaselineFuture Perspective: End of Treatment
Ixazomib + LenDex29.2-5.317.63.381.2-2.355.06.0
Placebo + LenDex30.3-3.118.01.781.7-7.857.34.4

[back to top]

Time to Pain Progression

Time to pain progression was assessed as the time from randomization to the date of initial progression classification. Pain progression was defined as the occurrence of 1 of the following and confirmed by 2 consecutive evaluations (To qualify as progression, the participant must have a BPI-SF worst pain score > 4 during pain progression): 1) a ≥ 2 point and 30% increase from Baseline in BPI-SF worst pain score without an increase in analgesic use, or 2) a 25% or more increase in analgesic use from Baseline without a decrease in BPI-SF worst pain score from Baseline. Brief Pain Inventory - Short Form (m-BPI-SF) is a participant rated 11-point Likert rating scale ranged from 0 (no pain) to 10 (worst pain imaginable). (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionmonths (Median)
Placebo + LenDex47.1
Ixazomib + LenDexNA

[back to top]

Progression Free Survival (PFS)-2

PFS2 was defined as the time from the date of randomization to the date of documentation of disease progression on the subsequent line of anticancer therapy, as assessed by the investigator in accordance with IMWG criteria, or death due to any cause, whichever occurs first. (NCT01850524)
Timeframe: Up to approximately 9 years

Interventionmonths (Median)
Placebo + LenDex52.2
Ixazomib + LenDex63.2

[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 according to International Myeloma Working Group (IMWG) criteria whichever occurs first. PD required one of the following: Increase of >=25% from nadir in: Serum M-component and/or (the absolute increase must be >=0.5 g/dL); Urine M-component and/or (the absolute increase must be >=200 mg/24 hours); in participants without measurable serum and urine M-protein levels: the difference between involved and uninvolved free light chain (FLC) levels (absolute increase must be > 10 mg/dL); Bone marrow plasma cell percentage: the absolute % must be >10%; development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; hypercalcemia (corrected serum calcium > 11.5 mg/dL or 2.85 mmol/L). (NCT01850524)
Timeframe: Up to approximately 79 months

Interventionmonths (Median)
Placebo + LenDex21.8
Ixazomib + LenDex35.3

[back to top]

Proportion of Patients With Response

"Response is defined as complete response (CR) or partial response (PR) CR: Complete disappearance of all detectable clinical evidence of disease and disease-related symptoms~PR:~>=50% decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or extranodal masses~No increase in the size of other nodes, liver or spleen~Bone marrow assessment is irrelevant for determination of a PR if the sample was positive prior to treatment. However, if positive, the cell type should be specified~No new sites of disease~The post-treatment PET should be positive at any previously involved sites~For variably FDG-avid lymphomas/FDG-avidity unknown, without a pretreatment PET scan, or if a pretreatment PET scan was negative, CT scan criteria should be used~Patients with a CR and persistent morphologic bone marrow involvement~Patients with bone marrow involved before therapy and a clinical CR but no bone marrow assessment after treatment" (NCT01856192)
Timeframe: Assessed every 3 months for 2 years, every 6 months for year 3, and then annually for years 4-10

Interventionproportion of participants (Number)
Arm A (R2CHOP)0.97
Arm B (RCHOP)0.92

[back to top]

3-year Progression-free Survival Rate

"Progression-free survival is defined as the time from randomization to disease progression, new primary of the same type or death, whichever occurs first. Progressive disease is defined as:~Appearance of any new lesion more than 1.5 cm in any axis during or at the end of therapy, even if other lesions are decreasing in size.~>= 50% increase from nadir in the sum of the product of the diameters of any previously involved nodes or extranodal masses, or in a single involved node or extranodal mass, or the size of other lesions.~>= 50% increase in the longest diameter of any single previously identified node or extranodal mass > 1 cm in its short axis.~Lesions should be PET positive unless the lesion is too small to be detected with current PET systems.~Measurable extranodal disease should be assessed in a manner similar to that for nodal disease.~3-year progression-free survival rate was calculated using Kaplan-Meier method." (NCT01856192)
Timeframe: Assessed every 3 months for 2 years, every 6 months for year 3, and then annually for years 4-10, 3-year PFS rate reported

Interventionproportion of participants (Number)
Arm A (R2CHOP)0.727
Arm B (RCHOP)0.615

[back to top]

Overall Survival Rate at 3 Years

Overall survival is defined as time from randomization to death or date last known alive. Overall survival rate at 3 years was calculated using the method of Kaplan Meier. (NCT01856192)
Timeframe: Assessed every 3 months for 2 years, every 6 months for year 3, and then annually for years 4-10

Interventionproportion of participants (Number)
Arm A (R2CHOP)0.826
Arm B (RCHOP)0.751

[back to top]

Proportion of Patients With Complete Response

Complete response is defined as complete disappearance of all detectable clinical evidence of disease, and disease-related symptoms if present prior to therapy. (NCT01856192)
Timeframe: Assessed every 3 months for 2 years, every 6 months for year 3, and then annually for years 4-10

Interventionproportion of participants (Number)
Arm A (R2CHOP)0.73
Arm B (RCHOP)0.68

[back to top]

Duration of Response (DOR)

"Duration of response was defined as the time from first evidence of partial response (PR) or better to confirmation of disease progression or death due to any cause.~Median DOR was estimated using Kaplan-Meier methods. Participants who started a new anticancer therapy before documentation of disease progression or death, or who were alive without documentation of disease progression before the data cut-off date or who died or had disease progression immediately after more than 1 consecutively missed disease assessment visit were censored at the date of last disease assessment." (NCT01881789)
Timeframe: Disease response was assessed every 4 weeks for 24 cycles then every 8 weeks until end of treatment; median time on follow-up at the analysis cut-off date of 18 July 2016 was 14.1, 3.5, 2.6, 16.0, 3.6, and 11.2 months in each group, respectively.

Interventionmonths (Median)
Oprozomib 150 mg 5/14 + Lenalidomide + DexamethasoneNA
Oprozomib 180 mg 5/14 + Lenalidomide + DexamethasoneNA
Oprozomib 210 mg 5/14 + Lenalidomide + DexamethasoneNA
Oprozomib 210 mg 2/7 + Lenalidomide + DexamethasoneNA
Oprozomib 240 mg 2/7 + Lenalidomide + DexamethasoneNA
Oprozomib 210 mg 2/7 + Cyclophosphamide + DexamethasoneNA

[back to top]

Number of Participants With Dose-Limiting Toxicities (DLTs)

"DLTs were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 4.03. A DLT is defined as any of the following treatment-related events:~Any ≥ Grade 3 nonhematologic toxicity with the following conditions:~≥ Grade 3 nausea, vomiting, diarrhea, or constipation only if for > 7 days despite optimal supportive care~Asymptomatic Grade 3 hypophosphatemia, Grade 3 hyperglycemia or toxicity solely due to dexamethasone, ≥ Grade 3 rash attributed to lenalidomide, and Grade 3 fatigue for < 14 days were not considered DLTs~Grade 4 neutropenia: absolute neutrophil count (ANC) < 0.5 × 10^9/L lasting ≥ 7 days, despite myeloid growth factor support~Febrile neutropenia~Grade 4 thrombocytopenia for ≥ 7 days or < 7 days with ≥ Grade 2 clinically significant bleeding or < 10,000 platelets requiring platelet transfusion, or Grade ≥ 3 with clinically significant bleeding or requiring platelet transfusion." (NCT01881789)
Timeframe: Cycle 1, 28 days

InterventionParticipants (Count of Participants)
Oprozomib 150 mg 5/14 + Lenalidomide + Dexamethasone2
Oprozomib 180 mg 5/14 + Lenalidomide + Dexamethasone2
Oprozomib 210 mg 5/14 + Lenalidomide + Dexamethasone2
Oprozomib 210 mg 2/7 + Lenalidomide + Dexamethasone0
Oprozomib 240 mg 2/7 + Lenalidomide + Dexamethasone1
Oprozomib 210 mg 2/7 + Cyclophosphamide + Dexamethasone0

[back to top]

Progression-Free Survival (PFS)

Progression-free survival is defined as the time from the start of treatment to disease progression or death (due to any cause), whichever occurred first. Median PFS was estimated using Kaplan-Meier methods. Participants who started a new anticancer therapy before documentation of disease progression or death, or who were alive without documentation of disease progression before the data cut-off date or who died or had disease progression immediately after more than 1 consecutively missed disease assessment visit were censored at the date of last disease assessment. (NCT01881789)
Timeframe: From first dose of study drug through the data cut-off date of 18 July 2016; median time on follow-up was 14.1, 3.5, 2.6, 16.0, 3.6, and 11.2 months in each group, respectively

Interventionmonths (Median)
Oprozomib 150 mg 5/14 + Lenalidomide + DexamethasoneNA
Oprozomib 180 mg 5/14 + Lenalidomide + DexamethasoneNA
Oprozomib 210 mg 5/14 + Lenalidomide + DexamethasoneNA
Oprozomib 210 mg 2/7 + Lenalidomide + DexamethasoneNA
Oprozomib 240 mg 2/7 + Lenalidomide + DexamethasoneNA
Oprozomib 210 mg 2/7 + Cyclophosphamide + DexamethasoneNA

[back to top]

Number of Participants With Treatment-emergent Adverse Events (AEs)

"Adverse events (AEs) were graded using NCI-CTCAE (version 4.03) and according to the following: Grade 1 = mild AE, Grade 2 = Moderate AE, Grade 3 = a severe AE, Grade 4 = life-threatening AE, and Grade 5 = death due to AE.~A serious AE is an event that met 1 or more of the following criteria:~Death~Life-threatening experience~Required inpatient hospitalization or prolongation of an existing hospitalization~Resulted in persistent or significant disability/incapacity~A congenital anomaly birth defect in the offspring of an exposed female subject or offspring of a female partner of a male subject~Important medical events that, based upon appropriate medical judgment, jeopardized the participant and may have required medical or surgical intervention to prevent an outcome listed above.~Treatment-related AEs are those considered related to at least 1 study drug by the investigator." (NCT01881789)
Timeframe: From first dose of any study treatment to 30 days after last dose; median duration of treatment was 29.1, 12.4, 11.3, 66.6, 7.8, and 46.4 weeks in each treatment group, respectively.

,,,,,
InterventionParticipants (Count of Participants)
Any treatment-emergent adverse event (TEAE)TEAE ≥ grade 3Serious adverse eventsTEAEs leading to discontinuation of study drugFatal adverse eventsTreatment-related adverse events (TRAE)TRAE ≥ grade 3Treatment-related serious adverse eventsTRAEs leading to discontinuation of study drug
Oprozomib 150 mg 5/14 + Lenalidomide + Dexamethasone332103321
Oprozomib 180 mg 5/14 + Lenalidomide + Dexamethasone764107641
Oprozomib 210 mg 2/7 + Cyclophosphamide + Dexamethasone321003210
Oprozomib 210 mg 2/7 + Lenalidomide + Dexamethasone332103211
Oprozomib 210 mg 5/14 + Lenalidomide + Dexamethasone333203222
Oprozomib 240 mg 2/7 + Lenalidomide + Dexamethasone221102211

[back to top]

Plasma Oprozomib Concentration

"Plasma samples for oprozomib concentration assays were only collected for participants in the 5/14 dosing schedule groups.~Oprozomib plasma concentrations were determined using liquid chromatography with tandem mass spectrometry." (NCT01881789)
Timeframe: Cycle 1 day 1 at 1 to 2.5 hours and 2.75 to 5 hours after end of infusion (EOI) and cycle 3 day 1 at predose and 1 to 2.5 hours and 2.75 to 5 hours after EOI.

,,
Interventionng/mL (Mean)
Cycle 1 day 1, 1 - 2.5 hours post EOICycle 1 day 1, 2.75 - 5 hours post EOICycle 3 day 1, predoseCycle 3 day 1, 1 - 2.5 hours post EOICycle 3 day 1, 2.75 - 5 hours post EOI
Oprozomib 150 mg 5/14 + Lenalidomide + Dexamethasone4972990.01112.00
Oprozomib 180 mg 5/14 + Lenalidomide + Dexamethasone63215.8142175255
Oprozomib 210 mg 5/14 + Lenalidomide + Dexamethasone69778.00.017869.1

[back to top]

Overall Response Rate (ORR)

"ORR is defined as the percentage of participants with a best overall response of partial response (PR), very good PR (VGPR), complete response (CR), or stringent CR (sCR) based on the International Myeloma Working Group Uniform Response Criteria.~PR: ≥ 50% reduction of serum M-protein and ≥ 90% reduction in urine M-protein or to < 200 mg/24 hrs, or a ≥ 50% decrease in dFLC. A ≥ 50% decrease in the size of soft tissue plasmacytomas present at baseline.~VGPR: Serum and urine M-protein detectable by immunofixation but not electrophoresis or ≥ 90% decrease in serum M-protein with urine M-protein <100 mg/24 hrs. If disease measurable only by SFLC, ≥ 90% decrease in the difference between involved and uninvolved FLC levels (dFLC).~CR: No immunofixation on serum and urine, disappearance of soft tissue plasmacytomas and <5% plasma cells in BM. Normal serum free light chain (SFLC) ratio if disease measurable only by SFLC.~sCR: As for CR, and absence of clonal plasma cells in bone marrow (BM). (NCT01881789)
Timeframe: Disease response was assessed every 4 weeks for 24 cycles then every 8 weeks until end of treatment; median duration of treatment at the analysis cutoff date of 18 July 2016 was 29.1, 12.4, 11.3, 66.6, 7.8 and 46.4 weeks in each group, respectively

Interventionpercentage of participants (Number)
Oprozomib 150 mg 5/14 + Lenalidomide + Dexamethasone66.7
Oprozomib 180 mg 5/14 + Lenalidomide + Dexamethasone57.1
Oprozomib 210 mg 5/14 + Lenalidomide + Dexamethasone66.7
Oprozomib 210 mg 2/7 + Lenalidomide + Dexamethasone100.0
Oprozomib 240 mg 2/7 + Lenalidomide + Dexamethasone50.0
Oprozomib 210 mg 2/7 + Cyclophosphamide + Dexamethasone100.0

[back to top]

Levels of CS1 Soluble Form (sCS1) in Serum

Expression levels of the free form of soluble CS1 were analyzed at different timepoints from serum samples derived from peripheral blood collection (NCT01891643)
Timeframe: At baseline (screening), during main study therapy (cycle 3 day 1, up to 64 days) and at time of progression (up to approximately 31 months)

,
Interventionug/L (Median)
BaselineCycle 3 day 1 of main study therapyTime of progression
E-Ld Cohort3.600.150.39
Ld Cohort13.270.7249.85

[back to top]

Change From Baseline in the Levels of CS1 Soluble Form (sCS1) in Serum During Therapy and At Progression

Expression levels of the free form of soluble CS1 were analyzed at different timepoints from serum samples derived from peripheral blood collection (NCT01891643)
Timeframe: From baseline (screening) to cycle 3 day 1 of the main study therapy (up to 64 days) and from baseline (screening) to time of progression (up to approximately 31 months)

,
Interventionug/L (Median)
From baseline to Cycle 3 day 1 of main study therapyFrom baseline to time of progression
E-Ld Cohort-3.51-0.85
Ld Cohort-4.08-17.41

[back to top]

Percent of Bone Marrow-Derived Multiple Myeloma (MM) Cells Expressing Cell Surface CS1 at Time of Progression

"CS1 (CD2 subset-1, also known as CRACC, SLAMF7, CD319) expression levels in multiple myeloma cells were analyzed from bone-marrow aspirates collected at time of progression.~The following conditions were considered to describe multiple myeloma cells expressing CS1 (CS1+/CD38++/CD138+/CD56+/CD19-/CD45DIM)" (NCT01891643)
Timeframe: Time of progression (up to approximately 54 months from pre-treatment screening)

InterventionPercent of cells expressing CS1 (Median)
E-Ld Cohort46.59
Ld Cohort84.62

[back to top]

Number of Participants With IgG Response

IgG response defined as having improvement in IgG level by at least 25% at 6 months, compared to baseline. (NCT01924169)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Lenalidomide0

[back to top]

Number of Participants With Response

Primary endpoint is response rate (RR) measured by the proportion of patients receiving the combination, whose disease stabilizes, or returns to at least its previous response level prior to progression, assessed at 3-months after starting the combination.1.Stringent Complete Remission (sCR): Follows criteria for CR, plus:Normal FLC ratio, Absence of clonal cells in the BM; Complete Remission (CR) All of the following criteria are met:Negative SIFE and UIFE:Disappearance of any soft tissue plasmacytomas:< 5% plasma cells in the BM. 2.Very Good Partial Response (VGPR):One or more of the following must be present:Serum and urine M-protein detectable by immunofixation but not on electrophoresis:≥ 90% reduction in serum M-protein and urine M-protein level < 100 mg/24 hours.Partial Response (PR) Both of the following must be present:≥ 50% reduction in SPEP:Reduction in 24-hour UPEP by ≥ 90% or to < 200 mg/24 hours.3.Stable Disease (SD)Does not meet the criteria for CR, VGPR, PR, or PD.4.Pr (NCT01927718)
Timeframe: 3 months

InterventionParticipants (Count of Participants)
Thalidomide + Lenalidomide2

[back to top]

Kaplan-Meier Estimate of Duration of Objective Response as Assessed by the IRC According to the 2007 IWGRC

Duration of response (DOR) was defined as the time from initial response (at least PR) until documented progressive disease (PD) or death. Participants who had not progressed at the time of analysis were censored at the last assessment date that the participant was known to be progression free. Participants who received a new treatment without documented progression were censored at the last assessment date that the participants was known to be progression free. (NCT01938001)
Timeframe: From randomization up to data cut-off date of 22 June 2018; overall median follow-up time for all participants was 28.30 months (range: 0.1 to 51.3 months).

Interventionmonths (Median)
Rituximab + Lenalidomide (R^2)36.6
Rituximab + Placebo21.7

[back to top]

Kaplan-Meier Estimate of Duration of Complete Response (DOCR) as Assessed by the IRC According to the 2007 IWGRC

DOCR was defined as the time from initial CR until documented PD or death. Participants who had not progressed at the time of analysis were censored at the last assessment date that the participant was known to be progression free. Participants who received a new treatment without documented progression were censored at the last assessment date that the participants was known to be progression free. (NCT01938001)
Timeframe: From randomization up to data cut-off date of 22 June 2018; overall median follow-up time for all participants was 28.30 months (range: 0.1 to 51.3 months).

Interventionmonths (Median)
Rituximab + Lenalidomide (R^2)NA
Rituximab + PlaceboNA

[back to top] [back to top]

Kaplan Meier Estimate of Progression Free Survival Assessed by the Independent Review Committee (IRC) According to the 2007 International Working Group Response Criteria (IWGRC)

Progression-free survival (PFS) was defined as the time from date of randomization into the study to the first observation of documented disease progression or death due to any cause, whichever occurred first. PFS was based on the data from the IRC review using the modified 2007 International Working Group Response Criteria (IWGRC) using FDA censoring rules. (NCT01938001)
Timeframe: From randomization of study drug up to disease progression or death, which occurred first; up to the data cut-off date of 22 June 2018; overall median follow-up time for all participants was 28.30 months (range: 0.1 to 51.3 months).

Interventionmonths (Median)
Rituximab + Lenalidomide (R^2)39.4
Rituximab + Placebo14.1

[back to top]

Kaplan Meier Estimate of Event Free Survival as Assessed by the IRC According to the 2007 IWGRC

Event-free survival (EFS) was defined as the time from date of randomization to date of first documented progression, relapse, institution of new anti-lymphoma treatment (chemotherapy, radiotherapy or immunotherapy) or death from any cause. Responding participants and those who were lost to follow up were censored at their last tumor assessment date. (NCT01938001)
Timeframe: From date of randomization to data cut-off date of 22 June 2018; overall median follow-up time for all participants was 28.30 months (range: 0.1 to 51.3 months).

Interventionmonths (Median)
Rituximab + Lenalidomide (R^2)27.6
Rituximab + Placebo13.9

[back to top]

Durable Complete Response Rate (DCCR) as Assessed by the IRC According to the 2007 IWGRC

DCCR was defined as the percentage of participants with a best response of complete response (CR) that lasted no less than one year (≥ 48 weeks) during the study prior to administration of new anti-lymphoma therapy. A CR is defined as a complete disappearance of any disease-related symptoms and normalization of biochemical abnormalities. (NCT01938001)
Timeframe: From first dose of investigational product (IP) to data cut-off date of 22 June 2018; the median treatment duration was 11.19 months in the rituximab/lenalidomiade arm and 11.04 months in the rituximab/placebo arm

InterventionPercentage of Participants (Number)
Rituximab + Lenalidomide (R^2)25.3
Rituximab + Placebo11.1

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs)

TEAEs include AEs that started or worsened between the date of the first dose and 28 days after the date of the last dose. A serious adverse event (SAE) is any: • Death; • Life-threatening event; • Any inpatient hospitalization or prolongation of existing hospitalization; • Persistent or significant disability or incapacity; • Congenital anomaly or birth defect; • Any other important medical event. The investigator determined the relationship of an AE to study drug based on the timing of the AE relative to drug administration and whether or not other drugs, therapeutic interventions, or underlying conditions could provide a sufficient explanation for the event. The severity of an AE was evaluated by the investigator according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) (Version 4.03) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death (NCT01938001)
Timeframe: From first dose to 28 days post last dose (Average of 55.71 months and a maximum up to 95.2 months)

,
InterventionParticipants (Count of Participants)
Any TEAEAny TEAE Related to Lenalidomide/Placebo (LEN/PBO)Any TEAE Related to Rituximab (RIT)Any Serious TEAEAny Serious TEAE Related to LEN/PBOAny Serious TEAE Related to RITAny CTCAE Grade (GR) 3/4 TEAEAny CTCAE GR 3/4 TEAE Related to LEN/PBOAny CTCAE GR 3/4 TEAE Related to RITAny GR 5 TEAEAny TEAE Leading to Dose Reduction LEN/PBOAny TEAE Leading to Dose Interruption LEN/PBOAny TEAE Leading to Dose Interruption RITAny TEAE Leading to Discontinuation of LEN/PBOAny TEAE Leading to Discontinuation of RIT
Rituximab + Lenalidomide (R^2)1741591344523131211015724611359156
Rituximab + Placebo173118105258458382026473892

[back to top]

Percentage of Participants With an Objective Response as Assessed by the IRC According to the 2007 IWGRC

Percentage of participants with an objective response is defined as having a response of at least a PR during the study without administration of new anti-lymphoma therapy. A complete response = a complete disappearance of all detectable clinical and radiographic evidence of disease, disappearance of any disease-related symptoms, and normalization of biochemical abnormalities; a partial response (PR) = 50% decrease in SPD of the 6 largest dominant nodes or nodal masses. No increase in the size of other nodes, liver, or spleen. Splenic and hepatic nodules must regress by at least 50% in the SPD. (NCT01938001)
Timeframe: From date of first dose to data cut-off date of 22 June 2018; the median treatment duration was 11.19 months in the rituximab/lenalidomide arm and 11.04 months in the rituximab/placebo arm

InterventionPercentage of Participants (Number)
Rituximab + Lenalidomide (R^2)77.5
Rituximab + Placebo53.3

[back to top]

Percentage of Participants With a Best Response of Complete Response as Assessed by the IRC According to the 2007 IWGRC

Percentage of participants with a best response of at CR during the study without administration of new anti-lymphoma therapy. A CR = Complete disappearance of all detectable clinical and radiographic evidence of disease, disappearance of any disease-related symptoms, and normalization of biochemical abnormalities. (NCT01938001)
Timeframe: From date of first dose up to data cut-off date of 22 June 2018; the median treatment duration was 11.19 months in the rituximab/lenalidomide arm and 11.04 months in the rituximab/placebo arm

InterventionPercentage of Participants (Number)
Rituximab + Lenalidomide (R^2)33.7
Rituximab + Placebo18.3

[back to top]

Kaplan-Meier Estimate of Overall Survival (OS)

Overall survival was defined as the time from randomization to death from any cause. Overall survival was censored at the last date that the participant was known to be alive for participants who were alive at the time of analysis and for participants who were lost to follow-up before death was documented. (NCT01938001)
Timeframe: From date of randomization to death due to any cause (Average of 55.71 months and a maximum up to 95.2 months)

InterventionMonths (Median)
Rituximab + Lenalidomide (R^2)NA
Rituximab + PlaceboNA

[back to top]

Objective Response Rate (ORR) in Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. Per protocol, pooled Cohort 5 was not planned to be evaluated statistically compared to a fixed efficacy target. The percentage of participants with CR and PR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)38.9

[back to top]

Objective Response Rate (ORR) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. The pooled Cohort 4 sub-cohorts were evaluated statistically by comparing the ORR for pembrolizumab to a fixed efficacy target of 25% using a binomial exact test. The percentage of participants with CR and PR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)22.1

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for participants with an indeterminate (missing) PD-L1 status. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)72.2

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)

ORR was evaluated for each of Cohort 4 sub-cohorts: 4A (primary mediastinal B-cell lymphoma), 4B (grey zone, splenic marginal zone, and mantle cell lymphomas), 4C (follicular lymphoma), and 4D (diffuse large B-Cell lymphoma). ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for participants with an indeterminate (missing) PD-L1 status. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)66.7
Cohort 4C: R/R Follicular Lymphoma (FL)50.0
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)36.4

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide): Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and for nodal masses. PR was >50% decrease in the sum of product diameters for ≤6 target dominant masses for lymph nodes, spleen nodules, and liver nodules and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for participants with an indeterminate (missing) PD-L1 status. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)33.3

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Indeterminate Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for participants with an indeterminate (missing) PD-L1 status. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)47.4

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 negative (PD-L1 of <1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)50.0

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)

ORR was evaluated for each of the Cohort 4 sub-cohorts: 4A (primary mediastinal B-cell lymphoma), 4B (grey zone, splenic marginal zone, and mantle cell lymphomas), 4C (follicular lymphoma), and 4D (diffuse large B-Cell lymphoma). ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 negative (PD-L1 of <1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)75.0
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas0.0
Cohort 4C: R/R Follicular Lymphoma (FL)7.1
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)0.0

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide): Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and for nodal masses. PR was >50% decrease in the sum of product diameters for ≤6 target dominant masses for lymph nodes, spleen nodules, and liver nodules and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 negative (PD-L1 of <1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)25.0

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Negative Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 negative (PD-L1 of <1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)11.4

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 positive (PD-L1 of ≥1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)54.5

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)

ORR was evaluated for each of the Cohort 4 sub-cohorts: 4A (primary mediastinal B-cell lymphoma), 4B (grey zone, splenic marginal zone, and mantle cell lymphomas), 4C (follicular lymphoma), and 4D (diffuse large B-Cell lymphoma). ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 positive (PD-L1 of ≥1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)27.3
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas100.0
Cohort 4C: R/R Follicular Lymphoma (FL)0.0
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)6.7

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide): Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and for nodal masses. PR was >50% decrease in the sum of product diameters for ≤6 target dominant masses for lymph nodes, spleen nodules, and liver nodules and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 positive (PD-L1 of ≥1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)75.0

[back to top]

Objective Response Rate (ORR) in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)

ORR was evaluated for each of the Cohort 4 sub-cohorts: 4A (primary mediastinal B-cell lymphoma), 4B (grey zone, splenic marginal zone, and mantle cell lymphomas), 4C (follicular lymphoma), and 4D (diffuse large B-Cell lymphoma). ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. Per protocol, Cohorts 4A, 4B, 4C, and 4D were not planned to be compared to an efficacy target. The percentage of participants with CR and PR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)47.6
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas50.0
Cohort 4C: R/R Follicular Lymphoma (FL)10.0
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)12.2

[back to top]

Overall Survival (OS)

OS was defined as the time from first dose of study treatment to death due to any cause. OS was calculated from product-limit (Kaplan-Meier) method for censored data. Participants without documented death at the time of the final analysis were censored at the date of the last follow-up. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 1: Myelodysplastic Syndrome (MDS)6.0
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)20.2
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)NA
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)37.1
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas23.8
Cohort 4C: R/R Follicular Lymphoma (FL)NA
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)4.9

[back to top]

Overall Survival (OS) in Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

OS was defined as the time from first dose of study treatment to death due to any cause. OS was calculated from product-limit (Kaplan-Meier) method for censored data. Participants without documented death at the time of the final analysis were censored at the date of the last follow-up. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)23.0

[back to top]

Overall Survival (OS) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

OS was defined as the time from first dose of study treatment to death due to any cause. OS was calculated from product-limit (Kaplan-Meier) method for censored data. Participants without documented death at the time of the final analysis were censored at the date of the last follow-up. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)12.0

[back to top]

Progression-free Survival (PFS) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)

PFS was defined as the time from first dose of study treatment to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. PD was assessed according to the International Myeloma Working Group (IMWG) 2006 response criteria. PD was defined as ≥1 of the following: increase of ≥25% from baseline of serum or urine M-component or >10 mg/dl difference between involved and uninvolved free light chain levels; development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; or hypercalcemia. PFS was calculated from the Kaplan-Meier method for censored data. PFS as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)2.7

[back to top]

Progression-free Survival (PFS) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

PFS was defined as the time from first dose of study treatment to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. PD was assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. PD was the appearance of any new lesion, >50% increase in the sum of product diameters (SPD) of ≥1 lymph node, or a >50% increase in the longest diameter of a previous lymph node, lesions positron emission tomography (PET) positive if 18F-fluorodeoxyglucose (FDG)-avid lymphoma or PET positive prior to therapy for lymph nodes; >50% increase from nadir in the SPD of previous lesions in the liver and spleen; or new or recurrent involvement of the bone marrow. PFS was calculated from the Kaplan-Meier method for censored data. PFS as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)8.7

[back to top]

Progression-free Survival (PFS) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

PFS was defined as the time from first dose of study treatment to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. PD was assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. PD was the appearance of any new lesion, >50% increase in the sum of product diameters (SPD) of ≥1 lymph node, or a >50% increase in the longest diameter of a previous lymph node, lesions positron emission tomography (PET) positive if 18F-fluorodeoxyglucose (FDG)-avid lymphoma or PET positive prior to therapy for lymph nodes; >50% increase from nadir in the SPD of previous lesions in the liver and spleen; or new or recurrent involvement of the bone marrow. PFS was calculated from the Kaplan-Meier method for censored data. PFS as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)1.7

[back to top]

Progression-free Survival (PFS) in Participants Pooled From the Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

PFS was defined as the time from first dose of study treatment to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. PD was assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. PD was the appearance of any new lesion, >50% increase in the sum of product diameters (SPD) of ≥1 lymph node, or a >50% increase in the longest diameter of a previous lymph node, lesions positron emission tomography (PET) positive if 18F-fluorodeoxyglucose (FDG)-avid lymphoma or PET positive prior to therapy for lymph nodes; >50% increase from nadir in the SPD of previous lesions in the liver and spleen; or new or recurrent involvement of the bone marrow. PFS was calculated from the Kaplan-Meier method for censored data. PFS as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)5.5

[back to top]

Progression-free Survival (PFS) in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)

PFS was defined as the time from first dose of study treatment to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. PD was assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. PD was the appearance of any new lesion, >50% increase in the sum of product diameters (SPD) of ≥1 lymph node, or a >50% increase in the longest diameter of a previous lymph node, lesions positron emission tomography (PET) positive if 18F-fluorodeoxyglucose (FDG)-avid lymphoma or PET positive prior to therapy for lymph nodes; >50% increase from nadir in the SPD of previous lesions in the liver and spleen; or new or recurrent involvement of the bone marrow. PFS was calculated from the Kaplan-Meier method for censored data. PFS as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)19.0
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas7.3
Cohort 4C: R/R Follicular Lymphoma (FL)1.7
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)1.4

[back to top]

Stringent Complete Remission (sCR) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)

sCR was assessed according to the International Myeloma Working Group (IMWG) 2006 response criteria. sCR was defined as complete response [CR] plus normal serum free light-chain ratio and absence of clonal cells in bone marrow. CR criteria are negative immunofixation of serum and urine and disappearance of soft tissue plasmacytomas and ≤5% plasma cells in the bone marrow. The percentage of participants with sCR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of partiicpants (Number)
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)0.0

[back to top]

Time to Progression (TTP) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)

TTP was defined as the time from first dose of study treatment to disease progression. Progressive disease was assessed according to the International Myeloma Working Group (IMWG) 2006 response criteria. PD was defined as ≥1 of the following: increase of ≥25% from baseline of serum or urine M-component or >10 mg/dl difference between involved and uninvolved free light chain levels; development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; bone marrow plasma cell percentage absolute % must be ≥10%; or hypercalcemia. The TTP as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)2.7

[back to top]

Objective Response Rate (ORR) in Programmed Cell Death Ligand 1 (PD-L1) Positive Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. PD-L1 was assessed by immunohistochemistry (IHC). The percentage of participants with CR and PR as assessed by the investigator is presented for PD-L1 positive (PD-L1 of ≥1%) participants. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)18.8

[back to top]

Complete Remission Rate (CRR) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

CRR was defined as the percentage of participants with complete remission according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. Complete remission was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. Cohort 3 was evaluated statistically by comparing the complete remission for pembrolizumab to a fixed efficacy target of 10% using a binomial exact test. The percentage of participants with complete remission as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)22.6

[back to top]

Complete Response (CR) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)

CR was assessed according to the International Myeloma Working Group (IMWG) 2006 response criteria. CR was defined as negative immunofixation of serum and urine and disappearance of soft tissue plasmacytomas and <5% plasmacytomas in the bone marrow. The percentage of participants with CR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)0.0

[back to top]

Cytogenic Complete Response in Cohort 1: Myelodysplastic Syndrome (MDS)

Cytogenic complete response was evaluated by detection of chromosomal abnormalities by cytogenic techniques and assessed according to the International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. Cytogenic complete response was defined as the disappearance of the chromosomal abnormality detected pre-treatment without the appearance of new chromosomal abnormalities. The percentage of participants with cytogenic complete response as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)11.1

[back to top]

Cytogenic Partial Response in Cohort 1: Myelodysplastic Syndrome (MDS)

Cytogenic partial response was evaluated by detection of chromosomal abnormalities by cytogenic techniques and assessed according to the International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. Cytogenic partial response was defined as ≥50% reduction of the chromosomal abnormality detected pre-treatment. The percentage of participants with cytogenic partial response as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)11.1

[back to top]

Duration of Response (DOR) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

For participants who demonstrated a confirmed Complete Response (CR) or partial response (PR), DOR was defined as the time from CR or PR to documented disease progression or death. CR and PR were assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. DOR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)25.0

[back to top]

Duration of Response (DOR) in Participants Pooled From Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide) Relapsed/Refractory (R/R) Diffuse Large B-Cell Lymphoma (DLBCL)

For participants who demonstrated a confirmed Complete Response (CR) or partial response (PR), DOR was defined as the time from CR or PR to documented disease progression or death. CR and PR were assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. DOR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Pooled Cohort 5 (Pembrolizumab + 20 or 25 mg Doses of Lenalidomide)NA

[back to top]

Duration of Response (DOR) in Participants Pooled From the Cohort 4 Non-Hodgkin Lymphoma (NHL) Sub-Cohorts (Cohorts 4A+4B+4C+4D)

For participants who demonstrated a confirmed Complete Response (CR) or partial response (PR), DOR was defined as the time from CR or PR to documented disease progression or death. CR and PR were assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. DOR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Pooled Cohort 4 Sub-cohorts (Cohorts 4A+4B+4C+4D)NA

[back to top]

Duration of Response (DOR) in the Cohort 4 Non-Hodgkin Lymphoma (NHL) Individual Sub-Cohorts (Cohorts 4A, 4B, 4C, and 4D)

DOR was evaluated for each of the Cohort 4 sub-cohorts: 4A (primary mediastinal B-cell lymphoma), 4B (grey zone, splenic marginal zone, and mantle cell lymphomas), 4C (follicular lymphoma), and 4D (diffuse large B-Cell lymphoma). For participants who demonstrated a confirmed Complete Response (CR) or partial response (PR), DOR was defined as the time from CR or PR to documented disease progression or death. CR and PR were assessed according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. DOR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionMonths (Median)
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)NA
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell LymphomasNA
Cohort 4C: R/R Follicular Lymphoma (FL)NA
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)13.6

[back to top]

Erythroid Response in Cohort 1: Myelodysplastic Syndrome (MDS)

Erythroid response was measured as an evaluation of hematologic improvement and was assessed according to the International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. Erythroid response baseline was based on an average of at least 2 pre-treatment measurements taken ≥1 week apart and not influenced by transfusions. Responses were considered significant if they lasted for ≥8 weeks. Criteria for an erythroid response include: hemoglobin (Hgb) increase by ≥1.5 grams/deciliter (g/dl) from pretreatment or reduction of ≥4 transfusions/8 weeks compared with the pre-treatment transfusion number in the previous 8 weeks. Only transfusions given for a Hgb of ≤9.0 g/dl pretreatment counted for response evaluation. The percentage of participants with an erythroid response as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)7.1

[back to top]

Marrow Complete Response (mCR) in Cohort 1: Myelodysplastic Syndrome (MDS)

mCR was defined as ≤5% myeloblasts in the bone marrow with a decrease in myeloblasts ≥50% over pretreatment according to the modified International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. The percentage of participants with mCR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)18.5

[back to top]

Neutrophil Response in Cohort 1: Myelodysplastic Syndrome (MDS)

Neutrophil response was measured as an evaluation of hematologic improvement and was assessed according to the International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. Neutrophil response baseline was based on an average of at least 2 pre-treatment measurements taken ≥1 week apart and not influenced by transfusions. Responses were considered significant if they lasted for ≥8 weeks. Criterion for a neutrophil response was a ≥100% increase in neutrophil count from pre-treatment and an absolute increase of >0.5 x 10^9/Liter. The percentage of participants with a neutrophil response as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)3.6

[back to top]

Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)

An adverse event was defined as any untoward medical occurrence in a participant administered study treatment and did not necessarily have a causal relationship with this treatment. An adverse event could be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that was temporally associated with the use of the study treatment was also an adverse event. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionParticipants (Count of Participants)
Cohort 1: Myelodysplastic Syndrome (MDS)6
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)2
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)3
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)1
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas1
Cohort 4C: R/R Follicular Lymphoma (FL)3
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)2
Cohort 5: R/R DLBCL Pembrolizumab+Lenalidomide 20 mg (RP2D)0
Cohort 5: R/R DLBCL Pembrolizumab+Lenalidomide 25 mg1

[back to top]

Number of Participants Who Experienced One or More Adverse Events (AEs):

An adverse event was defined as any untoward medical occurrence in a participant administered study treatment and did not necessarily have a causal relationship with this treatment. An adverse event could be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that was temporally associated with the use of the study treatment was also an adverse event. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionParticipants (Count of Participants)
Cohort 1: Myelodysplastic Syndrome (MDS)27
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)28
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)31
Cohort 4A: R/R Primary Mediastinal B-cell Lymphoma (PMBCL)21
Cohort 4B: Other Non-Hodgkin Lymphoma: Grey Zone, Splenic Marginal Zone, and Mantle Cell Lymphomas4
Cohort 4C: R/R Follicular Lymphoma (FL)21
Cohort 4D: R/R Diffuse Large B-Cell Lymphoma (DLBCL)38
Cohort 5: R/R DLBCL Pembrolizumab+Lenalidomide 20 mg (RP2D)13
Cohort 5: R/R DLBCL Pembrolizumab+Lenalidomide 25 mg5

[back to top]

Objective Response Rate (ORR) in Cohort 1: Myelodysplastic Syndrome (MDS)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. CR was demonstrated by ≤5% myeloblasts with normal maturation of all cell lines in the bone marrow (persistent dysplasia will be noted) and normal findings for hemoglobin, platelet count, neutrophil count, and absence of blasts in the blood. PR was all CR criteria if abnormal before treatment except bone marrow blasts decreased by ≥50% over pre-treatment but still >5%. Cellularity and morphology are not relevant. Cohort 1 was evaluated statistically by comparing the ORR for pembrolizumab to a fixed efficacy target of 10% using a binomial exact test. The percentage of participants with CR and PR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)0.0

[back to top]

Objective Response Rate (ORR) in Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)

ORR was defined as the percentage of the participants with either a stringent complete response (sCR), complete response (CR), very good partial response (VGPR), or partial response (PR) according to the International Myeloma Working Group (IMWG) 2006 response criteria. CR=negative immunofixation of serum and urine+disappearance of soft tissue plasmacytomas+≤5% plasma cells in the bone marrow (BM); sCR=stringent complete response, CR as above+normal serum free light-chain ratio and absence of clonal cells in BM; VGPR=serum+urine M-protein (M-p) by immunofixation but not on electrophoresis OR ≥ 90% reduction in serum M-p+urine M-p <100 mg/24 hr; PR=≥50% reduction of serum M-p+reduction in 24-hour urine M-p by ≥90% or to <200 mg/24 hours. Cohort 2 was evaluated statistically by comparing the ORR for pembrolizumab to a fixed efficacy target of 25% using a binomial exact test. The percentage of participants with CR, sCR, PR, VGPR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 2: Relapsed Refractory/Refractory (rR/R) Multiple Myeloma (MM)0.0

[back to top]

Objective Response Rate (ORR) in Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)

ORR was defined as the percentage of participants with response (complete response [CR] or partial response [PR]) according to the revised response criteria for malignant lymphoma per Cheson et al. 2007. CR was demonstrated by disappearance of all evidence of disease in the bone marrow, spleen, liver, and lymph nodes. PR was >50% decrease in the sum of product diameters (SPD) for ≤6 target masses for lymph nodes and >50% decrease in SPD for a single nodule in greatest transverse diameter for spleen and liver, and no size increase in the lymph nodes, spleen, or liver. The percentage of participants who experience ORR as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 3: Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL)64.5

[back to top]

Platelet Response in Cohort 1: Myelodysplastic Syndrome (MDS)

Platelet response was measured as an evaluation of hematologic improvement and was assessed according to the International Working Group (IWG) response criteria in myelodysplasia per Cheson et al. 2006. Platelet response baseline was based on an average of at least 2 pre-treatment measurements taken ≥1 week apart and not influenced by transfusions. Responses were considered significant if they lasted for ≥8 weeks. Criterion for a platelet response was an absolute increase of ≥30 x 10^9/Liter platelet count for participants with a pre-treatment count of ≥20 x 10^9/Liter and for participants with a pre-treatment count of <20 x 10^9/Liter there must have been an absolute increase to ≥20 x 10^9/Liter and a ≥100% increase in pre-treatment level. The percentage of participants with a platelet response as assessed by the investigator is presented. (NCT01953692)
Timeframe: Up to approximately 78.5 months

InterventionPercentage of participants (Number)
Cohort 1: Myelodysplastic Syndrome (MDS)7.1

[back to top]

Progression Free Survival (PFS) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment

PFS was defined as the time from randomization to the first documented disease progression (PD) or death due to any cause. PD= ≥1 of the following: ≥25% increase from baseline in serum/urine M-protein; hypercalcemia; increase in FLC ratio; ≥10% bone marrow plasma cells; new or increase in the size of existing bone lesions or tissue plasmacytomas. The median PFS was calculated from the Kaplan-Meier method for censored data. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionMonths (Median)
Pooled rrMM Cohort5.6
Pooled rMM Cohort14.3

[back to top]

Duration of Response (DOR) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment

DOR was the time from first evidence of response (stringent complete response [sCR]+complete response [CR], very good partial response [VGPR], partial response [PR]) until disease progression (PD) or death. CR=negative immunofixation of serum and urine+no tissue plasmacytomas+≤5% plasmacytomas in bone marrow; sCR=stringent complete response, CR+normal free light chain (FLC) ratio+no clonal cells in bone marrow; VGPR=serum+urine M-protein detectable by immunofixation but not electrophoresis OR ≥90% reduction in serum M-protein+urine M-protein <100 mg/24 hour(hr); PR = ≥50% reduction of serum M-protein+reduction in 24hr urinary M-protein by ≥90% or to <200 mg/24 hrs. PD=≥1 of the following: ≥25% increase from baseline serum/urine M-protein; hypercalcemia; increase between involved/uninvolved FLC levels; ≥10% bone marrow plasma cells; new or increase in the size of existing bone lesions or tissue plasmacytomas. DOR was calculated using the Kaplan-Meier method for censored data. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionMonths (Median)
Pooled rrMM Cohort16.4
Pooled rMM Cohort14.1

[back to top]

Disease Control Rate (DCR) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment

DCR was the percentage of participants who achieved stringent complete response (sCR), complete response (CR), very good partial response (VGPR), partial response (PR), or stable disease (SD) ≥12 weeks prior to evidence of disease progression (PD). CR=negative immunofixation of serum and urine+no tissue plasmacytomas+ ≤5% plasmacytomas in bone marrow; sCR=stringent complete response, CR+normal free light chain (FLC) ratio+no clonal cells in bone marrow; VGPR=serum+urine M-protein detectable by immunofixation but not on electrophoresis OR ≥90% reduction in serum M-protein+urine M-protein <100 mg/24 hour(hr); PR=≥50% reduction of serum M-protein+reduction in 24 hr urinary M-protein by ≥90% or to <200 mg/24 hrs; SD=not meeting the criteria for CR, VGPR, PR, or PD. PD=≥1 of the following: ≥25% increase from baseline in serum/urine M-protein; hypercalcemia; increase in FLC ratio; ≥10% bone marrow plasma cells; new or increase in the size of existing bone lesions or tissue plasmacytomas. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionPercentage of participants (Number)
Pooled rrMM Cohort84.1
Pooled rMM Cohort100.0

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to death due to any cause. OS was calculated from product-limit (Kaplan-Meier) method for censored data. Participants without documented death at the time of the final analysis were censored at the date of the last follow-up. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionMonths (Median)
Pooled rrMM Cohort29.0
Pooled rMM Cohort22.5

[back to top]

Number of Participants Experiencing Dose-Limiting Toxicities (DLTs) According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (NCI-CTCAE v.4.0)

DLTs were assessed during Cycle 1 (28 days) and were defined as the occurrence of any of the following judged by the investigator to be possibly, probably or definitely related to study drug: Grade (Gr) 4 non-hematologic toxicity; Gr 4 hematologic toxicity lasting ≥7 days; Gr 3 non-hematologic toxicity lasting >3 days; Gr 3 non-hematologic laboratory value if: medical intervention was required, abnormality led to hospitalization, was renal or liver function related, or persisted for >1 week; Gr 3/4 febrile neutropenia; thrombocytopenia <25,000 cells/mm^3 (associated with bleeding requiring platelet transfusion or life-threatening bleeding); Gr 5 toxicity; or delay of >1 week in initiating Cycle 2 or unable to complete 80% of treatment during the first course of therapy due to drug-related toxicity. DLTs for the rMM cohort were any drug-related adverse events that cannot be managed by dose modification. DLTs are reported for the maximum tolerated dose. (NCT02036502)
Timeframe: Up to 28 days in Cycle 1

InterventionParticipants (Count of Participants)
Part 1:Pembro 2mg/kg+Len 25mg+Dex 40 mg3
Part 1:Pembro 2mg/kg+Len 10 mg+Dex 40 mg0
Part 2:Pembro 200 mg+Len 25 mg+Dex 40 mg0
Part 2:Pembro 200 mg+Len 25 mg+Dex 20 mg0
Part 2:Pembro 200 mg+Len 10 mg+Dex 40 mg0

[back to top]

Objective Response Rate (ORR) Evaluated According to the International Myeloma Working Group (IMWG) 2006 Response Criteria by Confirmed Investigator Assessment

ORR was the percentage of the participants who achieved at least a partial response (stringent complete response [sCR]+complete response [CR]+very good partial response [VGPR]+partial response [PR]). CR=negative immunofixation of serum and urine+no tissue plasmacytomas+≤5% plasmacytomas in bone marrow; sCR=stringent complete response, CR+normal free light chain (FLC) ratio+no clonal cells in bone marrow; VGPR=serum+urine M-protein detectable by immunofixation but not electrophoresis OR ≥90% reduction in serum M-protein+urine M-protein <100 mg/24 hour(hr); PR = ≥50% reduction of serum M-protein+reduction in 24hr urinary M-protein by ≥90% or to <200 mg/24 hrs. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionPercentage of participants (Number)
Pooled rrMM Cohort30.2
Pooled rMM Cohort70.0

[back to top]

Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)

An adverse event was defined as any untoward medical occurrence in a participant administered study treatment and did not necessarily have a causal relationship with this treatment. An adverse event could be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that was temporally associated with the use of the study treatment was also an adverse event. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionParticipants (Count of Participants)
Part 1:Pembro 2mg/kg+Len 25mg+Dex 40 mg0
Part 1:Pembro 2mg/kg+Len 10 mg+Dex 40 mg1
Part 2:Pembro 200 mg+Len 25 mg+Dex 40 mg0
Part 2:Pembro 200 mg+Len 25 mg+Dex 20 mg0
Part 2:Pembro 200 mg+Len 10 mg+Dex 40 mg0
Part 3:Pembro 200 mg+Len 25 mg+Dex 40 mg4
Part 3:Pembro 200 mg+Len 25 mg+Dex 20 mg0
Part 3:Pembro 200 mg+Carf 56 mg/m^2 +Dex 20 mg6

[back to top]

Number of Participants Who Experienced One or More Adverse Events (AEs)

An adverse event was defined as any untoward medical occurrence in a participant administered study treatment and did not necessarily have a causal relationship with this treatment. An adverse event could be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that was temporally associated with the use of the study treatment was also an adverse event. (NCT02036502)
Timeframe: Up to approximately 72.7 months

InterventionParticipants (Count of Participants)
Part 1:Pembro 2mg/kg+Len 25mg+Dex 40 mg6
Part 1:Pembro 2mg/kg+Len 10 mg+Dex 40 mg3
Part 2:Pembro 200 mg+Len 25 mg+Dex 40 mg6
Part 2:Pembro 200 mg+Len 25 mg+Dex 20 mg1
Part 2:Pembro 200 mg+Len 10 mg+Dex 40 mg1
Part 3:Pembro 200 mg+Len 25 mg+Dex 40 mg45
Part 3:Pembro 200 mg+Len 25 mg+Dex 20 mg1
Part 3:Pembro 200 mg+Carf 56 mg/m^2 +Dex 20 mg10

[back to top]

Phenotypic Changes

Phenotypic changes in peripheral blood cells following CC-5013 (lenalidomide) administration especially in regards to CD3, CD4, CD8 T cells, and NK and NKT cells. (NCT02041325)
Timeframe: 6 weeks

,
Interventioncells/cmm (Median)
Total T cells CD3+Helper T cells CD3+/CD4+Cytotoxic T cells CD3+/CD8+Natural Killer cells CD56+/CD16+
Lenalidomide851.5591296249.5
Placebo715525183228

[back to top]

Safety

Number of participants with adverse events as a measure of safety and tolerability (NCT02041325)
Timeframe: 6 weeks

Interventionparticipants (Number)
Lenalidomide2
Placebo2

[back to top]

Quantity of Subjects With a T-cell Response

Participants who displayed a T cell responses against HbSAg following vaccination (NCT02041325)
Timeframe: 6 weeks

Interventionparticipants (Number)
Lenalidomide0
Placebo0

[back to top]

Positive for Hepatitis B Surface Antigen

The number of participants who test positive for the antibody titer against hepatitis B surface antigen (HbSAg). (NCT02041325)
Timeframe: 6 weeks

Interventionparticipants (Number)
Lenalidomide4
Placebo3

[back to top]

Time to Disease Progression (TTP)

TTP was defined as time from date of randomization to date of first documented evidence of progressive disease (PD). PD was defined as meeting any one of following criteria: Increase of >=25% in level of serum M-protein from lowest response value and absolute increase must be >=0.5 g/dL; Increase of >=25% in 24-hour urinary light chain excretion (urine M-protein) from lowest response value and absolute increase must be >=200 mg/24hours; Only in participants without measurable serum and urine M-protein levels: increase of >=25% in difference between involved and uninvolved free light chain (FLC) levels from lowest response value and absolute increase must be >10 milligram per deciliter (mg/dL); Definite increase in size of existing bone lesions or soft tissue plasmacytomas; Definite development of new bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to plasma cell (PC) proliferative disorder. (NCT02076009)
Timeframe: From randomization to disease progression (up to 21 months)

Interventionmonths (Median)
Lenalidomide, Low-dose Dexamethasone (Rd)18.43
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)NA

[back to top]

Time to Response

Time to response was defined as the time between the date of randomization and the first efficacy evaluation that the participant met all criteria for partial response (PR) or better. (NCT02076009)
Timeframe: From randomization up to first documented CR or PR (up to 21 months)

Interventionmonths (Median)
Lenalidomide, Low-dose Dexamethasone (Rd)1.3
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)1.0

[back to top]

Percentage of Participants With Negative Minimal Residual Disease (MRD)

Minimal residual disease was assessed for all participants who achieved a complete response (CR) or stringent complete response (sCR). CR: Negative immunofixation on the serum and urine, disappearance of any soft tissue plasmacytomas, and <5% PCs in bone marrow; sCR: CR and normal FLC ratio, absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4 color flow cytometry. The MRD negativity rate was defined as the percentage of participants who had negative MRD assessment at any time point after the first dose of study drugs by evaluation of bone marrow aspirates or whole blood at 10^ minus (-) 4, 10^-5, 10^-6 threshold. (NCT02076009)
Timeframe: From randomization to the date of first documented evidence of PD (up to 87.5 months)

,
Interventionpercentage of participants (Number)
MRD negative rate (10^-4)MRD negative rate (10^-5)MRD negative rate (10^-6)
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)40.633.213.3
Lenalidomide, Low-dose Dexamethasone (Rd)10.26.71.8

[back to top]

Time to Subsequent Anticancer Treatment

Time to subsequent anticancer treatment was defined as the time from randomization to the start of subsequent anticancer treatment or death due to progressive disease (PD), whichever occurs first. (NCT02076009)
Timeframe: From randomization to date of start of subsequent anticancer treatment or death due to PD, whichever occured first (up to 87.5 months)

Interventionmonths (Median)
Lenalidomide, Low-dose Dexamethasone (Rd)23.1
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)69.3

[back to top]

Duration of Response (DOR)

DOR was defined for participants with confirmed response (PR or better) as time between first documentation of response and disease progression/death due to PD, whichever occurs first. PD was defined as meeting any one of following criteria: Increase of >=25% in level of serum M-protein from lowest response value and absolute increase must be >=0.5g/dL; Increase of >=25% in 24-hour urinary light chain excretion (urine M-protein) from lowest response value and absolute increase must be >=200mg/24hours; Only in participants without measurable serum and urine M-protein levels: increase of >=25% in difference between involved and uninvolved FLC levels from lowest response value and absolute increase must be >10mg/dL; Definite increase in size of existing bone lesions/soft tissue plasmacytomas; Definite development of new bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5mg/dL) that can be attributed solely to PC proliferative disorder. (NCT02076009)
Timeframe: From randomization to the date of first documented evidence of PD (up to 21 months)

Interventionmonths (Median)
Lenalidomide, Low-dose Dexamethasone (Rd)17.4
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)NA

[back to top]

Overall Response Rate

Overall response rate was defined as the percentage of participants who achieved a partial response (PR) or better according to the International Myeloma Working Group (IMWG) criteria, during or after study treatment. IMWG criteria for PR: >=50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by >=90% or to <200 mg/24 hours, if the serum and urine M-protein are not measurable, a decrease of >=50% in the difference between involved and uninvolved FLC levels is required in place of the M-protein criteria, in addition to the above criteria, if present at baseline, a >=50% reduction in the size of soft tissue plasmacytomas is also required. (NCT02076009)
Timeframe: From randomization to disease progression (up to 21 months)

Interventionpercentage of participants (Number)
Lenalidomide, Low-dose Dexamethasone (Rd)76.4
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)92.9

[back to top]

Overall Survival (OS)

Overall survival was measured from the date of randomization to the date of the participant's death. (NCT02076009)
Timeframe: From randomization to date of death due to any cause (up to 87.5 months)

Interventionmonths (Median)
Lenalidomide, Low-dose Dexamethasone (Rd)51.84
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)67.58

[back to top]

Percentage of Participants Who Achieved Very Good Partial Response (VGPR) or Better

VGPR or better is defined as the percentage of participants who achieved VGPR, complete response (CR) and stringent complete response (sCR) according to the International Myeloma Working Group criteria (IMWG). IMWG criteria for VGPR: Serum and urine M-component detectable by immunofixation but not on electrophoresis, or >=90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours, if the serum and urine M-protein are not measurable, a decrease of >90% in the difference between involved and uninvolved FLC levels is required in place of the M-protein criteria. In addition to the above criteria, if present at baseline, a >=50% reduction in the size of soft tissue plasmacytomas is also required; CR: Negative immunofixation on the serum and urine, disappearance of any soft tissue plasmacytomas, and <5% PCs in bone marrow; sCR: CR and normal FLC ratio, absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4 color flow cytometry. (NCT02076009)
Timeframe: From randomization to disease progression (up to 21 months)

Interventionpercentage of participants (Number)
Lenalidomide, Low-dose Dexamethasone (Rd)44.2
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)75.8

[back to top]

Progression-free Survival (PFS)

PFS: duration from date of randomization to either progressive disease (PD)/death, whichever occurred first. PD: defined as meeting any 1 of following criteria: Increase of greater than equal to (>=)25 percent(%) in level of serum M-protein from lowest response value and absolute increase must be >=0.5 gram per deciliter (g/dL); Increase of >=25% in 24-hours(h) urinary light chain excretion (urine M-protein) from lowest response value and absolute increase must be >=200 mg/24h; Only in participants without measurable serum and urine M-protein levels: increase of >=25% in difference between involved and uninvolved free light chain (FLC) levels from lowest response value and absolute increase must be >10 mg/dL; Definite increase in size of existing bone lesions or soft tissue plasmacytomas; Definite development of new bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) attributed solely to plasma cell (PC) proliferative disorder. (NCT02076009)
Timeframe: From randomization to either disease progression or death whichever occurs first (up to 21 months)

Interventionmonths (Median)
Lenalidomide, Low-dose Dexamethasone (Rd)18.43
Daratumumab, Lenalidomide, Low-dose Dexamethasone (DRd)NA

[back to top]

Phase 2: Overall Survival (OS)

OS is defined as the time from the date of the first dose of study drug to the date of death due to any cause. For participants not known to have died at or prior to the database lock date, OS data was censored at the date last known alive. Participants who withdrew consent prior to study closure were censored on the date of the consent withdrawal. 2-sided 95% CI was estimated by Kaplan-Meier method. (NCT02077166)
Timeframe: Estimated median time on study in Phase 2 was 35.0 months.

Interventionmonths (Median)
Phase 2: Enrolled at Lenalidomide Dose 20 mg14.7
Phase 2: Enrolled at Lenalidomide Dose 25 mg11.6
Phase 2 Total: Enrolled at Lenalidomide Dose 20 or 25 mg14.2

[back to top]

Phase 1b: Number of Participants With Treatment-Emergent Adverse Events (TEAEs), Serious TEAEs, and Discontinuations Due to TEAEs

An adverse event (AE) is any untoward medical occurrence, which does not necessarily have a causal relationship with treatment. A serious AE is any untoward medical occurrence that at any dose: results in death; is life-threatening; requires in-patient hospitalization > 24 hours or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; is an important medical event. AEs that started or worsened during the treatment-emergent period and all possibly related or related AEs were considered TEAEs. Related events were those that were considered possibly related or related to study drug per investigator's judgment. Events were graded per the national Cancer Institute's Common Terminology Criteria for Adverse Events, version 4.03: Grade 1=mild; grade 2=moderate; grade 3=severe; grade 4=life-threatening; grade 5=death. (NCT02077166)
Timeframe: From first dose of study drug up to 30 days after last dose of study drug. Phase 1b median duration of ibrutinib exposure was 4.4 months; median duration of lenalidomide exposure was 4.4 months; median total number of doses of rituximab received was 4.0.

,,,,
InterventionParticipants (Count of Participants)
Any TEAEAny Grade >=3 TEAEAny Study Drug-Related TEAEAny Grade >=3 Study Drug-Related TEAEAny Ibrutinib-Related TEAEAny Grade >=3 Ibrutinib-Related TEAEAny Lenalidomide-Related TEAEAny Grade >=3 Lenalidomide-Related TEAEAny Rituximab-Related TEAEAny Grade >= 3 Rituximab-Related TEAEAny TEAE Leading to Dose Reduction of Any Study DrugAny TEAE Leading to Dose Reduction of IbrutinibAny TEAE Leading to Dose Reduction of LenalidomideAny TEAE Leading to Dose Delay of Any Study DrugAny TEAE Leading to Dose Delay of IbrutinibAny TEAE Leading to Dose Delay of LenalidomideAny TEAE Leading to Dose Delay of RituximabAny TEAE Leading to Discontinuation of Any Study DrugAny TEAE Leading to Discontinuation of Ibrutinib DoseAny TEAE Leading to Discontinuation of Lenalidomide DoseAny TEAE Leading to Discontinuation of Rituximab DoseAny Serious TEAEAny Grade >=3 Serious TEAEAny Treatment-Related Serious TEAEAny Ibrutinib-Related Serious TEAEAny Lenalidomide-Related Serious TEAEAny Rituximab-Related Serious TEAEAny Fatal TEAE
Phase 1b: Enrolled at Lenalidomide Dose 10 mg (Dose Level -1)7653535342000443122226622222
Phase 1b: Enrolled at Lenalidomide Dose 15 mg (Dose Level 1)12111210121012961432666444434400000
Phase 1b: Enrolled at Lenalidomide Dose 15 mg (Dose Level 1+)9998979877443777633337644422
Phase 1b: Enrolled at Lenalidomide Dose 20 mg (Dose Level 2)9987878765434887422225422211
Phase 1b: Enrolled at Lenalidomide Dose 25 mg (Dose Level 3)8787868673534888411116633301

[back to top]

Phase 2: Number of Participants With TEAEs, Serious TEAEs, and Discontinuations Due to TEAEs

An adverse event (AE) is any untoward medical occurrence, which does not necessarily have a causal relationship with treatment. A serious AE is any untoward medical occurrence that at any dose: results in death; is life-threatening; requires in-patient hospitalization > 24 hours or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; is an important medical event. AEs that started or worsened during the treatment-emergent period and all possibly related or related AEs were considered TEAEs. Related events were those that were considered possibly related or related to study drug per investigator's judgment. Events were graded per the national Cancer Institute's Common Terminology Criteria for Adverse Events, version 4.03: Grade 1=mild; grade 2=moderate; grade 3=severe; grade 4=life-threatening; grade 5=death. (NCT02077166)
Timeframe: From first dose of study drug up to 30 days after last dose of study drug. Phase 2 median duration of ibrutinib exposure was 4.9 months; median duration of lenalidomide exposure was 4.7 months; median total number of doses of rituximab received was 5.0.

,,
InterventionParticipants (Count of Participants)
Any TEAEAny Grade >=3 TEAEAny Study Drug-Related TEAEAny Grade >=3 Study Drug-Related TEAEAny Ibrutinib-Related TEAEAny Grade >=3 Ibrutinib-Related TEAEAny Lenalidomide-Related TEAEAny Grade >=3 Lenalidomide-Related TEAEAny Rituximab-Related TEAEAny Grade >= 3 Rituximab-Related TEAEAny TEAE Leading to Dose Reduction of Any Study DrugAny TEAE Leading to Dose Reduction of IbrutinibAny TEAE Leading to Dose Reduction of LenalidomideAny TEAE Leading to Dose Delay of Any Study DrugAny TEAE Leading to Dose Delay of IbrutinibAny TEAE Leading to Dose Delay of LenalidomideAny TEAE Leading to Dose Delay of RituximabAny TEAE Leading to Discontinuation of Any Study DrugAny TEAE Leading to Discontinuation of Ibrutinib DoseAny TEAE Leading to Discontinuation of Lenalidomide DoseAny TEAE Leading to Discontinuation of Rituximab DoseAny Serious TEAEAny Grade >=3 Serious TEAEAny Treatment-Related Serious TEAEAny Ibrutinib-Related Serious TEAEAny Lenalidomide-Related Serious TEAEAny Rituximab-Related Serious TEAEAny Fatal TEAE
Phase 2 Total: Enrolled at Lenalidomide Dose 20 or 25 mg8981856682608365623137213569666316181618105750282626912
Phase 2: Enrolled at Lenalidomide Dose 20 mg55515241503651403718231221403837101111116322916141648
Phase 2: Enrolled at Lenalidomide Dose 25 mg343033253224322525131491429282667574252112121054

[back to top] [back to top]

Phase 2: CR Rate

The CR rate was defined as the percentage of participants who achieve a CR, according to the Revised International Working Group Response Criteria for Malignant Lymphoma or Lugano Classification (see Cheson, 2014 for detailed criteria) in response-evaluable population, as assessed by the Investigator. (NCT02077166)
Timeframe: Estimated median time on study in Phase 2 was 35.0 months.

Interventionpercentage of participants (Number)
Phase 2: Enrolled at Lenalidomide Dose 20 mg32.1
Phase 2: Enrolled at Lenalidomide Dose 25 mg21.9
Phase 2 Total: Enrolled at Lenalidomide Dose 20 or 25 mg28.2

[back to top]

Phase 2: Duration of Response (DOR)

DOR is defined as the time from the date of the first documented response (CR or PR) to the first documented evidence of disease progression (PD) according to the Revised International Working Group Response Criteria for Malignant Lymphoma or Lugano Classification (Cheson 2014) or death from any cause. For participants who had achieved an overall response but did not die or progress at the time of analysis, DOR was censored on the date of the last adequate post-baseline disease assessment, or on the date of the first occurrence of response (CR or PR) if there was no disease assessment afterwards. 2-sided 95% CI is estimated by Kaplan-Meier method. (NCT02077166)
Timeframe: Estimated median time on study in Phase 2 was 35.0 months.

Interventionmonths (Median)
Phase 2: Enrolled at Lenalidomide Dose 20 mg38.3
Phase 2: Enrolled at Lenalidomide Dose 25 mg28.6
Phase 2 Total: Enrolled at Lenalidomide Dose 20 or 25 mg38.3

[back to top]

Phase 2: Overall Response Rate (ORR)

The ORR was defined as the percentage of participants who achieve either a partial response (PR) or complete response (CR), according to the Revised International Working Group Response Criteria for Malignant Lymphoma or Lugano Classification (Cheson 2014), as assessed by the investigator in response-evaluable population. The 95% confidence interval (CI) was calculated using the exact method. (NCT02077166)
Timeframe: Estimated median time on study in Phase 2 was 35.0 months.

Interventionpercentage of participants (Number)
Phase 2: Enrolled at Lenalidomide Dose 20 mg52.8
Phase 2: Enrolled at Lenalidomide Dose 25 mg43.8
Phase 2 Total: Enrolled at Lenalidomide Dose 20 or 25 mg49.4

[back to top]

Phase 2: Progression Free Survival (PFS)

PFS is defined as the time from the date of the first dose of study drug to confirmed PD according to the Revised International Working Group Response Criteria for Malignant Lymphoma or Lugano Classification (Cheson 2014) or death from any cause, whichever occurred first. For participants without disease progression or death, PFS data was censored at the date of the last tumor assessment. 2 sided 95% CI is estimated by Kaplan-Meier method. (NCT02077166)
Timeframe: Estimated median time on study in Phase 2 was 35.0 months.

Interventionmonths (Median)
Phase 2: Enrolled at Lenalidomide Dose 20 mg5.4
Phase 2: Enrolled at Lenalidomide Dose 25 mg4.7
Phase 2 Total: Enrolled at Lenalidomide Dose 20 or 25 mg5.4

[back to top]

Phase 1b: Complete Response (CR) Rate

The CR rate was defined as the percentage of participants who achieve a CR, according to the Revised International Working Group Response Criteria for Malignant Lymphoma (Cheson 2007), as assessed by the Investigator in response-evaluable population, where CR=disappearance of all evidence of disease, as assessed by the Investigator. (NCT02077166)
Timeframe: Estimated median time on Phase 1b study was 59.6 months.

Interventionpercentage of participants (Number)
Phase 1b: Enrolled at Lenalidomide Dose 15 mg (Dose Level 1)33.3
Phase 1b: Enrolled at Lenalidomide Dose 10 mg (Dose Level -1)0
Phase 1b: Enrolled at Lenalidomide Dose 15 mg (Dose Level 1+)42.9
Phase 1b: Enrolled at Lenalidomide Dose 20 mg (Dose Level 2)11.1
Phase 1b: Enrolled at Lenalidomide Dose 25 mg (Dose Level 3)50.0
Phase 1b Total: Enrolled at Lenalidomide Dose 10 to 25 mg (All Dose Levels)27.5

[back to top]

Phase 1b: ORR

The ORR was defined as the percentage of participants who achieve either a PR or CR, according to the Revised International Working Group Response Criteria for Malignant Lymphoma (Cheson 2007), as assessed by the Investigator in response-evaluable population, where CR=disappearance of all evidence of disease and PR=regression of measurable disease and no new sites. The 95% CI was calculated using the exact method. (NCT02077166)
Timeframe: Estimated median time on study in Phase 1b was 59.6 months.

Interventionpercentage of participants (Number)
Phase 1b: Enrolled at Lenalidomide Dose 15 mg (Dose Level 1)44.4
Phase 1b: Enrolled at Lenalidomide Dose 10 mg (Dose Level -1)0
Phase 1b: Enrolled at Lenalidomide Dose 15 mg (Dose Level 1+)71.4
Phase 1b: Enrolled at Lenalidomide Dose 20 mg (Dose Level 2)22.2
Phase 1b: Enrolled at Lenalidomide Dose 25 mg (Dose Level 3)75.0
Phase 1b Total: Enrolled at Lenalidomide Dose 10 to 25 mg (All Dose Levels)42.5

[back to top]

Overall Survival

Overall Survival (OS) is defined as the time from Stem Cell Transplant (SCT) to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT02086552)
Timeframe: Time from SCT to death due to any cause, assessed up to 3 years

Interventionyears (Median)
Treatment (Sonidegib, Lenalidomide)NA

[back to top]

Complete Response, Assessed Using the International Myeloma Working Group (IMWG) Uniform Response Criteria

"A comfirmed Complete Response (CR) is defined as a CR noted as the objective status on 2 consecutive evaluations. To be categorized as a CR, patients must exhibit the following:>~Negative immunofixation of serum and urine c, and>~Disappearance of any soft tissue plasmacytoma, and>~<5% plasma cells in Bone Marrow, and>~If the only measurable disease is the serum free light chain (FLC), a normal FLC ratio.>~We are reporting the proportion of patients achieving a CR or higher divided by the total number of evaluable patients.> Exact binomial 95% confidence intervals for the true success proportion will be calculated." (NCT02086552)
Timeframe: Up to 2 years

Interventionproportion of patients (Number)
Treatment (Sonidegib, Lenalidomide)0.48

[back to top]

Progression-free Survival

The progression-free survival time is defined as the time from SCT to progression or death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. Here, we are reporting the proportion of patients that have not had a PFS event two years after SCT. (NCT02086552)
Timeframe: Time from SCT to progression or death due to any cause, assessed at 2 years

Interventionproportion of participants (Number)
Treatment (Sonidegib, Lenalidomide)0.76

[back to top]

Progression-free Survival (1 Year Survival Rate)

The progression-free survival time is defined as the time from SCT to progression or death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. Here, we are reporting the proportion of patients that have not had a PFS event one year after SCT. (NCT02086552)
Timeframe: Time from SCT to progression or death due to any cause, assessed at 1 year

Interventionproportion of participants (Number)
Treatment (Sonidegib, Lenalidomide)0.88

[back to top]

Complete Response (CR) Duration

Complete Response is defined as disappearance of all clinical and/or radiologic evidence of disease, including extramedullary leukemia. Neutrophil count >/= 1.0 x 10^9/L and platelet count>/= 100 x 10^9, and bone marrow differential showing NCT02126553)
Timeframe: Up to 7 years

InterventionMonths (Median)
Treatment (Lenalidomide)NA

[back to top]

Event-free Survival (EFS)

Time from date of treatment start until the date of first objective documentation of disease-relapse, death or discontinuation due to adverse events. (NCT02126553)
Timeframe: Up to 7 Years

InterventionMonths (Median)
Treatment (Lenalidomide)12

[back to top]

Overall Survival (OS)

Time from date of treatment start until date of death due to any cause or last Follow-up. Survival will be measured by the estimated median survival computed by Kaplan-Meier (K-M) analysis, which is the time point at which the cumulative survival drops below 50%, if present. If not present then the median Overall Survival is not reached and not available (NA) as there are an insufficient number of participants with events. In either case ranges are provided for observed survival intervals used in the K-M analysis. (NCT02126553)
Timeframe: Up to 7 Years

InterventionMonths (Median)
Treatment (Lenalidomide)NA

[back to top]

Relapse-free Survival (RFS)

Time from date of treatment start until the date of first objective documentation of disease-relapse. (NCT02126553)
Timeframe: Up to 7 Years

InterventionMonths (Median)
Treatment (Lenalidomide)21.5

[back to top]

The Remission Rate for Participants With High-risk Myeloma

Toward improving the clinical outcomes of research subjects with high-risk MM (HR-MM) in the context of the immediately preceding TT5 trial 2008-02 and TT3 trials 2003-33 and 2006-66, TT5B will attempt to accelerate and sustain, at 2 years from starting therapy, the proportion of subjects in complete remission (AS-CR-2) by reducing host-imposed toxicity and thus facilitating timely completion of highly synergistic 8-drug combination therapy, including the next generation proteasome inhibitor, Carfilzomib (CFZ). This will result in avoiding MM re-growth that, we postulate, ensued in TT3 during recovery phases from severe de-conditioning. Furthermore, we speculate that the incidence of positive minimal residual disease (MRD) will be reduced with the addition of one cycle of consolidation therapy. (NCT02128230)
Timeframe: from baseline to either death or study completion for each subject (up to approximately 48 months)

InterventionParticipants (Number)
Total Therapy 5B0

[back to top]

Number of Participants With Complete Responses (CR) and Partial Responses (PR) as a Measure of Efficacy-ORR

Part 2 - Overall Response rate will be defined as the proportion of subjects who achieve either a Complete Response or a Partial Response according to the international Working Group Response Criteria for NHL as assessed by investigator. (NCT02142049)
Timeframe: 1 year after last subjects received the first dose

InterventionParticipants (Count of Participants)
All Subjects Treated at RP2D16
ABC Subjects Treated at RP2D9

[back to top]

Number of Participants With Complete Responses (CR) and Partial Responses (PR) as a Measure of Efficacy

Part-1: Overall Response rate (ORR) will defined as the proportion of subjects who achieve either a CR or a PR according to the international Working Group Response Criteria for NHL as assessed by investigator. (NCT02142049)
Timeframe: 1 year after last subjects received the first dose

InterventionParticipants (Count of Participants)
Part 1: Dose Level 11
Part 1: Dose Level 22
Part 1: Dose Level 30
Part 1: Dose Level 43
Part 1: All Treated6

[back to top]

Progression Free Survival (PFS) and Overall Survival (OS) as a Measure of Efficacy

Part 2: PFS will be measured as time from first study drug administration to disease progression or death from any cause. OS will be measured from the time of first study drug administration until the date of death using Kaplan-Meier methodology. (NCT02142049)
Timeframe: From initial dose date until the date of first documented progression or death from any cause, whichever came first, assessed up to approximately 1 year after the last subject received the first dose, up to 36 months at the most.

,
InterventionMonths (Median)
Progression Free Survival (PFS)Overall Survival (OS)
ABC Subjects Treated at RP2D4.8615.84
All Subjects Treated at RP2D4.8615.84

[back to top]

Number of Subjects With Adverse Events as a Measure of Safety and Tolerability

Part 2: The frequency (number and percentage) of treatment-emergent adverse events will be reported. (NCT02142049)
Timeframe: 1 year after last subjects received the first dose

InterventionParticipants (Count of Participants)
All Subjects Treated at RP2D26
ABC Subjects Treated at RP2D14

[back to top]

Number of Participants With Dose-Limiting Toxicities as a Measure of Safety and Tolerability

Part-1: To determine the maximum tolerated dose (MTD) of the combination of ibrutinib and lenalidomide with dose adjusted EPOCH-R (NCT02142049)
Timeframe: 1 year after last subjects received the first dose

InterventionParticipants (Count of Participants)
Part 1: Dose Level 10
Part 1: Dose Level 20
Part 1: Dose Level 30
Part 1: Dose Level 41
Part 1: All Treated1

[back to top]

Duration of Response (DOR)

Part 2: DOR will be measured from the time by which the measurement criteria are met for CR or PR until the first date by which recurrent or progressive disease is objectively documented. (NCT02142049)
Timeframe: From initial response date until the date of first documented progression or death from any cause, whichever came first, assessed up to approximately 1 year after the last subject received the first dose.

InterventionMonths (Median)
All Subjects Treated at RP2D Who Achieved Overall Response3.94
ABC Subjects Treated at RP2D Who Achieved Overall Response4.09

[back to top]

Number of Participants With Any Grade and Grade 3 or Grade 4 (G3/4) Infusion Reactions Over the Entire Study Period

An infusion reaction in this study is defined as any relevant sign or symptom occurring during or after elotuzumab infusion and considered by the investigator as an infusion reaction. Grade (Gr) 1=Mild, Gr 2=Moderate, Gr 3=Severe, Gr 4=Potentially Life-threatening or disabling, Gr 5=Death. (NCT02159365)
Timeframe: Date of first dose up to 60 days post last dose (approximately 4 years)

InterventionParticipants (Count of Participants)
# of participants with any grade infusion reaction# of participants with G3/4 infusion reaction
E-Ld31

[back to top]

Number of Participants With Grade 3 or Grade 4 (G3/4) Infusion Reactions by the End of Treatment Cycle 2

Infusion reaction was defined as any relevant sign or symptom occurring during or after elotuzumab infusion and considered by the investigator as an infusion reaction. Grade (Gr) 1=Mild, Gr 2=Moderate, Gr 3=Severe, Gr 4=Potentially Life-threatening or disabling, Gr 5=Death. (NCT02159365)
Timeframe: From Day 1 to End of cycle 2 treatment (approximately 56 days)

InterventionParticipants (Number)
E-Ld0

[back to top]

Number of Participants With a Response

"Response is Complete Response or Partial Response. CR is absence of Lymphadenopathy, Hepatomegaly or Splenomegaly, lymphocytes < 4000/ul, normocellular, <30% lymphocytes, no B-lymphoid nodules, Platelets > 100,000/ul, hemoglobin >11.0 g/dl and Neutrophils >1500/ul.~PR is >/= 50% decrease in lymphadenopathy, hepatomegaly, splenomegaly and Blood Lymphocytes from baseline, 50% reduction in marrow infiltrate or B-lymphoid nodules. Platelet count > 100,000/ul, Hemoglobin > 11 g/dl and Neutrophils >1500/ul or increase >/= 50% of all over base." (NCT02225275)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Treatment (Lenalidomide, Obinutuzumab)5

[back to top] [back to top]

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. Survival will be measured by the estimated median survival computed by Kaplan-Meier (K-M) analysis, which is the time point at which the cumulative survival drops below 50%, if present. If not present then the median Overall Survival is not reached and not available (NA) as there are an insufficient number of participants with events. In either case ranges are provided for observed survival intervals used in the K-M analysis. (NCT02225275)
Timeframe: Up to 5 years, 2 months

InterventionMonths (Median)
Treatment (Lenalidomide, Obinutuzumab)NA

[back to top]

Primary: Progression-free Survival (PFS)

PFS is defined as time from date of randomization to either progressive disease (PD) or death, whichever occurs first based on computerized algorithm as per International Myeloma Working Group (IMWG) criteria. PD is defined as an increase of 25 percent (%) from the lowest response value in one of the following: serum and urine M-component (absolute increase must be greater than or equal to [>=] 0.5 gram per deciliter [g/dL] and >=200 milligram [mg]/24 hours respectively); Only in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved free light chain (FLC) levels (absolute increase must be greater than [>]10 mg/dL); Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to Plasma cell (PC) proliferative disorder. (NCT02252172)
Timeframe: From randomization to disease progression, death, subsequent anti-myeloma therapy, withdrawal of consent to study participation or clinical cut-off (CCO) whichever occurs first (up to 3.5 years)

InterventionMonths (Median)
Lenalidomide + Dexamethasone (Rd)31.87
Daratumumab + Lenalidomide + Dexamethasone (DRd)NA

[back to top]

Change From Baseline in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ)-C30 Global Health Status Score to Day 1 of Cycle 3, 6, 9 and 12

"EORTC QLQ-C30 is 30 items self-reporting questionnaire, with 1 week recall period, resulting in 5 functional scales (physical functioning, role functioning, emotional functioning, cognitive functioning, and social functioning), 1 Global Health Status (GHS) scale, 3 symptom scales (fatigue, nausea and vomiting, and pain), and 6 single symptom items (dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties). Questionnaire includes 28 items with 4-point Likert type responses from 1-not at all to 4-very much to assess functioning and symptoms; 2 items with 7-point Likert scales (1= poor and 7= excellent) for global health and overall health related QoL. Scores are transformed to 0 to 100 scale, with higher scores representing better GHS, better functioning, and more symptoms. Negative change from baseline values shows deterioration in quality of life or functioning and reduction in symptom and positive values indicate improvement and worsening of symptoms." (NCT02252172)
Timeframe: Baseline and Day 1 of Cycle 3, 6, 9 and 12 (each Cycle of 28 days)

,
InterventionScore on scale (Least Squares Mean)
Global health status score: Cycle 3 Day 1Global health status score: Cycle 6 Day 1Global health status score: Cycle 9 Day 1Global health status score: Cycle 12 Day 1
Daratumumab + Lenalidomide + Dexamethasone (DRd)4.56.48.28.4
Lenalidomide + Dexamethasone (Rd)1.55.675.4

[back to top]

Change From Baseline in EuroQol-5 Dimensions-5 Levels (EQ-5D-5L) Utility Score to Day 1 of Cycle 3, 6, 9 and 12

EQ-5D-5L is standardized, participant-reported questionnaire to assess health-related quality of life. EQ-5D-5L includes 2 components: EQ-5D-5L health state profile (descriptive system) and EQ-5D-5L VAS. EQ-5D-5L descriptive system provides a profile of participant's health state 5 dimensions: mobility, self-care, usual activities, pain/discomfort and anxiety/depression. Each dimension has 5 response options (no problems, slight problems, moderate problems, severe problems and extreme problems) that reflect increasing levels of difficulty. The participant was asked to indicate his/her current health state by selecting the most appropriate level in each of the 5 dimensions. Responses to the 5-dimension scores were combined and converted into single preference-weighted health utility index score 0 (0.0- worst health state) to 1 (1.0- better health state) representing the general health status of individual (but allows for values less than 0 by United kingdom [UK] scoring algorithm). (NCT02252172)
Timeframe: Baseline and Day 1 of Cycle 3, 6, 9 and 12 (each Cycle of 28 days)

,
InterventionScore on scale (Least Squares Mean)
Change at Cycle 3 Day 1Change at Cycle 6 Day 1Change at Cycle 9 Day 1Change at Cycle 12 Day 1
Daratumumab + Lenalidomide + Dexamethasone (DRd)0.1070.1360.1240.141
Lenalidomide + Dexamethasone (Rd)0.080.1160.1240.113

[back to top]

Change From Baseline in EuroQol-5 Dimensions-5 Levels (EQ-5D-5L) Visual Analogue Scale (VAS) to Day 1 of Cycle 3, 6, 9 and 12

EQ-5D-5L is a standardized, participant-rated questionnaire to assess health-related quality of life. The EQ-5D-5L includes 2 components: the EQ-5D-5L health state profile (descriptive system) and the EQ-5D-5L Visual Analog Scale. The Visual Analogue Scale is designed to rate the participant's current health state on a scale from 0 to 100, where 0 represents the worst imaginable health state and 100 represents the best imaginable health state. (NCT02252172)
Timeframe: Baseline and Day 1 of Cycle 3, 6, 9 and 12 (each Cycle of 28 days)

,
InterventionScore on scale (Least Squares Mean)
Change at Cycle 3 Day 1Change at Cycle 6 Day 1Change at Cycle 9 Day 1Change at Cycle 12 Day 1
Daratumumab + Lenalidomide + Dexamethasone (DRd)4.9810.210.1
Lenalidomide + Dexamethasone (Rd)2.55.77.74.9

[back to top]

Rate of MRD-positive to MRD-negative Conversion Between the Two Maintenance Arms

(NCT02253316)
Timeframe: Cycle 13 Day 1 of maintenance treatment (Approximately Day 364 of maintenance treatment)

InterventionParticipants (Count of Participants)
Maintenance Arm 1: Ixazomib5
Maintenance Arm 2: Lenalidomide11

[back to top]

Number of Participants With Improvement in Minimal Residual Disease (MRD)

For the purposes of this study, a patient will be considered as having minimal residual disease if a positive result (1x10E06) is obtained using the Adaptive Clonoseq MRD testing. (NCT02253316)
Timeframe: After 4 cycles of IRD consolidation treatment (approximately Day 112 of consolidation treatment)

InterventionParticipants (Count of Participants)
Consolidation: Ixazomib, Lenalidomide, & Dexamethasone19

[back to top]

MRD-negative Rate After ASCT

For the purposes of this study, a patient will be considered as having minimal residual disease if a positive result (1x10E06) is obtained using the Adaptive Clonoseq MRD testing (NCT02253316)
Timeframe: Prior to beginning consolidation treatment (Day -28 to Day 0)

InterventionParticipants (Count of Participants)
Consolidation: Ixazomib, Lenalidomide, & Dexamethasone51

[back to top]

Compare Response Rate Between the Two Maintenance Arms

"Response will be determined by the International Myeloma Working Group (IMWG) Uniform Response Criteria. Response includes stringent complete response (sCR) and complete response (CR).~sCR requires all of the following:~CR as defined below~Normal free light chain ratio (0.26-1.65)~Absence of clonal cells in the bone marrow by immunohistochemistry or immunofluorescence~CR requires all of the following:~Disappearance of monoclonal protein by both protein electrophoresis and immunofixation studies from the blood and urine~If serum and urine monoclonal protein are unmeasurable, Normal free light chain ratio (0.26-1.65)~<5% plasma cells in the bone marrow~Disappearance of soft tissue plasmacytoma" (NCT02253316)
Timeframe: Through completion of maintenance treatment (estimated to be day 1095 of maintenance treatment)

InterventionParticipants (Count of Participants)
Maintenance Arm 1: Ixazomib50
Maintenance Arm 2: Lenalidomide80

[back to top]

Toxicity of IRD Consolidation

For the purposes of this study, toxicity will be defined as inability to receive 4 cycles of IRD consolidation due to toxicity. (NCT02253316)
Timeframe: After 4 cycles of IRD consolidation treatment (approximately Day 112 of consolidation treatment)

InterventionParticipants (Count of Participants)
Consolidation: Ixazomib, Lenalidomide, & Dexamethasone7

[back to top]

Response Rate of IRD Consolidation

For the purposes of this study, response rate is defined as the improvement in complete response rate. Response will be determined by the International Myeloma Working Group (IMWG) Uniform Response Criteria. (NCT02253316)
Timeframe: After 4 cycles of IRD consolidation treatment (approximately Day 112 of consolidation treatment)

InterventionParticipants (Count of Participants)
Consolidation: Ixazomib, Lenalidomide, & Dexamethasone49

[back to top]

Phase 1a and 1b: Number of Participants With at Least One Treatment-Emergent Adverse Event (TEAE) and Serious Adverse Event (SAE)

An AE is any untoward medical occurrence in a subject administered a medicinal investigational drug. The untoward medical occurrence does not necessarily have to have a causal relationship with treatment. An SAE is any untoward medical occurrence that results in death; is life-threatening; requires inpatient hospitalization or prolongation of present hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect or is a medically important event that may not be immediately life-threatening or result in death or hospitalization. A TEAE was defined as any AE either reported for the first time or worsening of a pre-existing event after first dose of study drug and within 30 days of the last dose of study drug. (NCT02265510)
Timeframe: From first dose of study drug up to 30 days after last dose of study drug (Up to approximately 3.4 years)

,,,,,,,,,,,
InterventionParticipants (Count of Participants)
TEAESAE
Phase 1a TGA - INCB052793 100 mg62
Phase 1a TGA - INCB052793 15 mg30
Phase 1a TGA - INCB052793 25 mg31
Phase 1a TGA - INCB052793 35 mg60
Phase 1a TGA - INCB052793 50 mg42
Phase 1a TGA - INCB052793 75 mg33
Phase 1a TGB - INCB052793 25 mg31
Phase 1a TGB - INCB052793 35 mg42
Phase 1a TGB - INCB052793 50 mg42
Phase 1b Cohort B - INCB052793 25 mg + Dexamethasone 40 mg75
Phase 1b Cohort F - INCB052793 25 mg + Azacytidine 75 mg/m^254
Phase 1b Cohort F - INCB052793 35 mg + Azacytidine 75 mg/m^21614

[back to top]

Phase 1a, 1b, and Phase 2: Cmax: Maximum Observed Plasma Concentration for INCB052793

Cmax is defined as the maximum observed plasma concentration measured at steady state (Day 15). For PK analyses subjects in TGA and TGB are combined by dosage group because only 3 subjects were enrolled in each dose group in TGB and 4 subject for first dose in TGB 50 mg. (NCT02265510)
Timeframe: Cycle 1, Day 15: predose and 0.5, 1, 2, 4, 6 hours postdose in Phase 1a; 0 (predose), 0.08, 0.5, 1, 2, 4, 6 and 8 hours postdose in Phase 1b and Phase 2

InterventionnM (Mean)
Phase 1a TGA - INCB052793 15 mg522
Phase 1a TGA - INCB052793 25 mg1110
Phase 1a TGA - INCB052793 35 mg1120
Phase 1a TGA - INCB052793 50 mg2050
Phase 1a TGA - INCB052793 75 mg1840
Phase 1a TGA - INCB052793 100 mg2890
Phase 1b Cohort B - INCB052793 25 mg + Dexamethasone 40 mg928
Phase 1b Cohort F - INCB052793 25 mg + Azacytidine 75 mg/m^21270
Phase 1b Cohort F - INCB052793 35 mg + Azacytidine 75 mg/m^21480
Phase 2 Cohort I - INCB052793 35 mg + Azacytidine 75 mg/m^21610

[back to top]

Phase 1A and 1B: Percentage of Participants With Response as Determined by Investigator's Assessment

Response rate is defined as the percentage of participants who achieved best overall response (BOR) as determined by IWG response criteria of investigator's assessment. A participant was considered an objective responder based on the following- Solid tumors: participant had a best overall response (BOR) of CR or PR, Lymphoma: participant had a BOR of complete radiologic response/complete metabolic response or partial remission/partial metabolic response, AML: participant had a BOR of CR, CRi, morphological leukemia-free state (MLFS), or PR, MDS: participant had a BOR of CR, PR, or marrow CR, MDS/myeloproliferative neoplasm (MPN): participant had a BOR of CR, PR, or marrow response, MM: participant had a BOR of stringent CR, CR, very good PR, PR, or MR. Subjects are combined by tumor type for this analysis. (NCT02265510)
Timeframe: Baseline through end of study (Up to approximately 4.5 years)

InterventionParticipants (Count of Participants)
Phase 1a TGA - INCB052793 in Solid Tumors0
Phase 1a TGB - INCB052793 in Lymphoma0
Phase 1a TGB - INCB052793 in MDS/MPN1
Phase 1a TGB - INCB052793 in MM0
Phase 1b Cohort B - INCB052793 + Dexamethasone in MM2
Phase 1b Cohort F-INCB052793 +Azacytidine in AML4
Phase 1b Cohort F-INCB052793 +Azacytidine in MDS3
Phase 1b Cohort F-INCB052793 +Azacytidine in MDS/MPN2

[back to top]

Phase 1a, 1b, and Phase 2: AUC0-τ: Area Under the Plasma Concentration-time Curve Over Dosing Interval for INCB052793

AUC0-τ is the area under the plasma concentration-time curve from time = 0 to the last measurable concentration at time = t measured at steady state (Day 15). For PK analyses subjects in TGA and TGB are combined by dosage group because only 3 subjects were enrolled in each dose group in TGB and 4 subject for first dose in TGB 50 mg. (NCT02265510)
Timeframe: Cycle 1, Day 15: predose and 0.5, 1, 2, 4, 6 hours postdose in Phase 1a; 0 (predose), 0.08, 0.5, 1, 2, 4, 6 and 8 hours postdose in Phase 1b and Phase 2

InterventionnM*hr (Mean)
Phase 1a TGA - INCB052793 15 mg4750
Phase 1a TGA - INCB052793 25 mg9170
Phase 1a TGA - INCB052793 35 mg8430
Phase 1a TGA - INCB052793 50 mg14700
Phase 1a TGA - INCB052793 75 mg18300
Phase 1a TGA - INCB052793 100 mg27800
Phase 1b Cohort B - INCB052793 25 mg + Dexamethasone 40 mg6390
Phase 1b Cohort F - INCB052793 25 mg + Azacytidine 75 mg/m^29580
Phase 1b Cohort F - INCB052793 35 mg + Azacytidine 75 mg/m^210200
Phase 2 Cohort I - INCB052793 35 mg + Azacytidine 75 mg/m^29380

[back to top]

Phase 1a, 1b, and Phase 2: AUC0-τ: Area Under the Plasma Concentration-time Curve Over Dosing Interval for Itacitinib

AUC0-τ is the area under the plasma concentration-time curve from time = 0 to the last measurable concentration at time = t measured at steady state (Day 15). (NCT02265510)
Timeframe: Cycle 1, Day 15: predose and 0.5, 1, 2, 4, 6 hours postdose in Phase 1a; 0 (predose), 0.08, 0.5, 1, 2, 4, 6 and 8 hours postdose in Phase 1b and Phase 2

InterventionnM*hr (Mean)
Phase 2 Cohort J - Itacitinib 300 mg + Azacitidine 75 mg/m^29870

[back to top]

Phase 1a, 1b, and Phase 2: Cmax: Maximum Observed Plasma Concentration of Itacitinib

Cmax is defined as the maximum observed plasma concentration measured at steady state (Day 15). (NCT02265510)
Timeframe: Cycle 1, Day 15: predose and 0.5, 1, 2, 4, 6 hours postdose in Phase 1a; 0 (predose), 0.08, 0.5, 1, 2, 4, 6 and 8 hours postdose in Phase 1b and Phase 2

InterventionnM (Mean)
Phase 2 Cohort J - Itacitinib 300 mg + Azacitidine 75 mg/m^21310

[back to top]

Phase 1a, 1b, and Phase 2: Tmax: Time to Maximum Plasma Concentration for INCB052793

Tmax is the time to maximum (peak) observed plasma drug concentration. Summary of Steady-State, Day 15, was evaluated by dosing regimen. For PK analyses subjects in TGA and TGB are combined by dosage group because only 3 subjects were enrolled in each dose group in TGB and 4 subject for first dose in TGB 50 mg. (NCT02265510)
Timeframe: Cycle 1, Day 15: predose and 0.5, 1, 2, 4, 6 hours postdose in Phase 1a; 0 (predose), 0.08, 0.5, 1, 2, 4, 6 and 8 hours postdose in Phase 1b and Phase 2

Interventionhours (hr) (Median)
Phase 1a TGA - INCB052793 15 mg1.1
Phase 1a TGA - INCB052793 25 mg0.76
Phase 1a TGA - INCB052793 35 mg2.0
Phase 1a TGA - INCB052793 50 mg1.1
Phase 1a TGA - INCB052793 75 mg2.2
Phase 1a TGA - INCB052793 100 mg2.0
Phase 1b Cohort B - INCB052793 25 mg + Dexamethasone 40 mg1.0
Phase 1b Cohort F - INCB052793 25 mg + Azacytidine 75 mg/m^21.1
Phase 1b Cohort F - INCB052793 35 mg + Azacytidine 75 mg/m^21.1
Phase 2 Cohort I - INCB052793 35 mg + Azacytidine 75 mg/m^20.51

[back to top]

Phase 1a, 1b, and Phase 2: Tmax: Time to Maximum Plasma Concentration for Itacitinib

Tmax is the time to maximum (peak) observed plasma drug concentration. (NCT02265510)
Timeframe: Cycle 1, Day 15: predose and 0.5, 1, 2, 4, 6 hours postdose in Phase 1a; 0 (predose), 0.08, 0.5, 1, 2, 4, 6 and 8 hours postdose in Phase 1b and Phase 2

Interventionhr (Median)
Phase 2 Cohort J - Itacitinib 300 mg + Azacitidine 75 mg/m^23.5

[back to top]

Phase 2: Objective Response Rate (ORR) in Hematological Malignancies

ORR is defined as the proportion of participants who achieved complete response (CR), CR with incomplete hematologic recovery (CRi), partial response (PR), or hematologic improvement (HI), using the IWG response criteria. (NCT02265510)
Timeframe: Baseline through end of study (Up to approximately 4.5 years)

InterventionParticipants (Count of Participants)
Phase 2 Cohort I-INCB052793 +Azacytidine (AML)0
Phase 2 Cohort I-INCB052793 +Azacytidine (MDS)1
Phase 2 Cohort J-Itacitinib +Azacitidine (AML)1
Phase 2 Cohort J-Itacitinib +Azacitidine (MDS)0

[back to top]

Phase 2: Number of Participants With at Least One TEAE and SAE

An AE is any untoward medical occurrence in a subject administered a medicinal investigational drug. The untoward medical occurrence does not necessarily have to have a causal relationship with treatment. An SAE is any untoward medical occurrence that results in death; is life-threatening; requires inpatient hospitalization or prolongation of present hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect or is a medically important event that may not be immediately life-threatening or result in death or hospitalization. A TEAE was defined as any AE either reported for the first time or worsening of a pre-existing event after first dose of study drug and within 30 days of the last dose of study drug. (NCT02265510)
Timeframe: From first dose of study drug up to 30 days after last dose of study drug (Up to approximately 1.3 years)

,
InterventionParticipants (Count of Participants)
TEAESAE
Phase 2 Cohort I - INCB052793 35 mg + Azacytidine 75 mg/m^297
Phase 2 Cohort J - Itacitinib 300 mg + Azacitidine 75 mg/m^2108

[back to top]

Objective Response Rate (ORR)

"ORR is the percentage of randomized participants who achieve a stringent complete response (sCR), complete response (CR), very good partial response (VGPR) or partial response (PR) as determined by investigator using the International Myeloma Working Group (IMWG) response criteria.~SCR: CR and normal free light chain (FLC) ratio and no clonal cells in bone marrow; CR: Negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow; VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥ 90% reduction in serum M-protein and urine M-protein level < 100 mg/24 hours; PR: ≥ 50% reduction of serum M-Protein and reduction in urinary M-protein by ≥ 90% or to < 200 mg/24 hours. In addition to the above, if present at baseline a ≥ 50% reduction in the size of soft tissue plasmacytomas is also required." (NCT02272803)
Timeframe: From first dose until documented response, up to approximately 72 months

InterventionPercentage of participants (Number)
Arm A: Lenalidomide + Dexamethasone + Elotuzumab (BMS-901608)92.5
Arm B: Lenalidomide + Dexamethasone73.8

[back to top]

Progression Free Survival (PFS)

PFS is defined as the time from randomization to the date of the first documented tumor progression, as determined by the investigator using the International Myeloma Working Group (IMWG) response criteria, or to death due to any cause, provided death does not occur more than 10 weeks (2 or more assessment visits) after the last tumor assessment. Clinical deterioration will not be considered progression. (NCT02272803)
Timeframe: From randomization to the date of first documented tumor progression or death due to any cause, up to approximately 72 months

InterventionMonths (Median)
Arm A: Lenalidomide + Dexamethasone + Elotuzumab (BMS-901608)NA
Arm B: Lenalidomide + Dexamethasone43.33

[back to top]

Objective Response Rate (ORR) of Participants Treated With Elotuzumab + Lenalidomide/Dexamethasone (E-Ld)

"ORR is the proportion of randomized participants who achieve a stringent complete response (sCR), complete response (CR), very good partial response (VGPR) or PR as determined by investigator using the International Myeloma Working Group (IMWG) response criteria.~SCR: CR and normal free light chain (FLC) ratio and no clonal cells in bone marrow; CR: Negative serum and urine on immunofixation, disappearance of any soft tissue plasmacytomas and ≤ 5% plasma cells in bone marrow; VGPR: Serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥ 90% reduction in serum M-protein and urine M-protein level < 100 mg/24 hours; PR: ≥ 50% reduction of serum M-Protein and reduction in urinary M-protein by ≥ 90% or to < 200 mg/24 hours. In addition to the above, if present at baseline a ≥ 50% reduction in the size of soft tissue plasmacytomas is also required." (NCT02272803)
Timeframe: From first dose until documented response (assessed up to February 2017, approximately 24 months)

InterventionPercentage of participants (Number)
Arm A: Lenalidomide + Dexamethasone + Elotuzumab (BMS-901608)87.5

[back to top]

Progression Free Survival (PFS) Rate

PFS rate is defined as the percentage of participants who have neither progressed nor died at the specified timepoints (NCT02272803)
Timeframe: From randomization up to the specified timepoints, up to 3 years

,
InterventionPercentage of Participants (Number)
1 year2 years3 years
Arm A: Lenalidomide + Dexamethasone + Elotuzumab (BMS-901608)0.870.670.61
Arm B: Lenalidomide + Dexamethasone0.890.600.56

[back to top]

Overall Survival

Time from initiation of therapy to death (NCT02279394)
Timeframe: From start of treatment up to +/- 60 months

Interventionmonths (Median)
Elo / Len / Dex50.5
Elo / Len60.2

[back to top]

Time to Progression

Time from initiation of therapy to progression defined by the IMWG criteria. (NCT02279394)
Timeframe: From start of treatment up to +/- 60 months

Interventionmonths (Median)
Elo / Len / Dex56.1
Elo / Len56.2

[back to top]

Percent of Patients Who Are Progression Free at 2 Years

Percent of patients who are alive and without documented progression after at least 2-years of follow-up. All patients who receive study treatment are assessed including those who have died or lost to follow-up prior to 2-years. Progression was defined as an increase in SPEP [25% and an absolute increase of 0.5g/d] or UPEP [25% and an absolute increase of 200mg/24hours] on 2 successive evaluations as determined by the IMWG response criteria or documented progression by the FreeLite progressive disease criteria in the absence of serum or urine involvement. (NCT02279394)
Timeframe: 2 Years

Interventionpercentage of participants (Number)
Elo / Len / Dex45.0
Elo / Len36.4

[back to top]

Objective Response Percent

Percent of patients with objective response defined as partial response or better based on the International Myeloma Working Group Response (IMWG) criteria (NCT02279394)
Timeframe: 2 Years from start of treatment

Interventionpercentage of participants (Number)
Elo / Len / Dex82.5
Elo / Len72.7

[back to top]

Mean Change From Baseline in the FACT-Lym Additional Concerns Subscale

"The FACT-Lym questionnaire is a validated instrument for assessing the impact of lymphoma on HRQL and contains 42 questions covering HRQL and common lymphoma symptoms and treatment side-effects. It begins with the Functional Assessment of Cancer Therapy - General (FACT-G), which contains 27 questions covering four core subscales: Physical Wellbeing (7 items), Social/Family Wellbeing (7), Emotional Wellbeing (6), and Functional Wellbeing (7). The FACT-Lym also includes an Additional Concerns subscale (15 questions) used to assess NHL-related symptoms such as pain, itching, night sweats,trouble sleeping, fatigue and trouble concentrating and concerns regarding lumps and swelling, fevers, infections, weight, appetite, emotional stability and treatment. All questions are answered on a 5-point scale ranging from not at all (0) to very much (4). The Additional Concerns subscale ranges from 0 to 60, where higher scores reflect better HRQoL." (NCT02285062)
Timeframe: Baseline and Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionUnits on a Scale (Mean)
MidcycleC6 D1EoT = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)3.85.86.68.3
Placebo Plus R-CHOP4.15.24.56.5

[back to top] [back to top] [back to top]

Percentage of Participants Who Completed the Functional Assessment of Cancer Therapy Lymphoma (FACT-Lym) Questionnaire

The completion rate for FACT-Lym assessments was judged by looking at the number of completed FACT-Lym assessments at each time point. The FACT-Lym was considered completed if at least 1 calculable score was present. Completion rates were calculated as the number and percentage of participants out of the total number of patients in the ITT population and summarized by visit/cycle and treatment group. The FACT-Lym is a health related quality of life (HRQoL) questionnaire targeted to the management of chronic illness, predominantly within oncology and is considered an extension of the FACT-General questionnaire. (NCT02285062)
Timeframe: Screening, Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionPercentage of Participants (Number)
ScreeningMidcycle = After Cycle 3, but before Cycle 4End of Treatment (EoT) = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)98.687.076.167.7
Placebo Plus R-CHOP98.286.379.669.5

[back to top]

Mean Change From Baseline in the EQ-5D-3L Visual Analogue Scale (VAS)

"The EQ-5D-3L questionnaire includes a visual analogue scale which records the respondent's self-rated health on a vertical, 0-100 scale where 100 = Best imaginable health state and 0 = Worst imaginable health state. Higher scores again indicate better HRQoL and positive change scores indicate that post screening values were higher than those observed at screening. The EQ-5D-3L is a generic, self-reported preference-based measure of health across five dimensions: mobility, self-care, pain, usual activities, and anxiety/depression. Each dimension has three levels of 'severity' corresponding to no problems, some problems and extreme problems." (NCT02285062)
Timeframe: Baseline and Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionUnits on a Scale (Mean)
MidcycleC6 D1EoT = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)4.06.08.012.0
Placebo Plus R-CHOP3.09.06.09

[back to top]

K-M Estimate of Overall Survival (OS)

Overall survival was assessed by the Independent Response Adjudication Committee (IRAC) and defined as time from randomization until death due to any cause. Participants who withdrew consent were censored at the time of withdrawal. Participants who were still alive before the clinical data cutoff date and participants who were lost to follow-up were censored at date last known alive. (NCT02285062)
Timeframe: From randomization until death due to any cause (up to approximately 86 months)

InterventionMonths (Median)
Lenalidomide Plus R-CHOP (R2-CHOP)NA
Placebo Plus R-CHOPNA

[back to top]

K-M Estimate of Duration of Complete Response

Duration of complete response was calculated for complete responders only and was defined as the time from documented initial complete response prior to initiation of subsequent systemic antilymphoma therapy until documented disease progression or death, whichever occurred earlier. Participants who had not progressed or died at the time of the analysis were censored at the date of last response assessment demonstrating no disease progression. Participants who changed treatment without evidence of disease progression were censored at the last assessment showing no progression prior to treatment change. (NCT02285062)
Timeframe: From randomization date up to the data cut off date of 15 March 2019; median follow-up was 24.5 months.

InterventionMonths (Median)
Lenalidomide Plus R-CHOP (R2-CHOP)NA
Placebo Plus R-CHOPNA

[back to top]

Mean Change From Baseline in the FACT-Lym Physical Well-Being Subscale

"The FACT-Lym questionnaire is a validated instrument for assessing the impact of lymphoma on HRQL and contains 42 questions covering HRQL and common lymphoma symptoms and treatment side-effects. It begins with the Functional Assessment of Cancer Therapy - General (FACT-G), which contains 27 questions covering four core subscales: Physical Wellbeing (7 items), Social/Family Wellbeing (7), Emotional Wellbeing (6), and Functional Wellbeing (7). The FACT-Lym also includes an Additional Concerns subscale (15 questions) used to assess NHL-related symptoms and concerns. All questions are answered on a 5-point scale ranging from not at all (0) to very much (4). The physical well-being subscale ranges from 0 to 28, where higher scores reflect better HRQoL." (NCT02285062)
Timeframe: Baseline and Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionUnits on a Scale (Mean)
MidcycleC6 D1EoT = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)-0.7-0.01.52.8
Placebo Plus R-CHOP0.20.90.72.6

[back to top]

Percentage of Participants Who Achieved an Objective Response

An objective response = percentage of participants who achieved a complete response or partial response after initiation of the treatment and prior to initiation of subsequent systemic anti-lymphoma therapy. A CR = complete metabolic response; Target nodes/nodal masses regressed on computed tomography to (≤ 1.5 cm in their greatest transverse diameter for nodes > 1.5 cm prior to therapy. Regressed to normal size by imaging, and absence of nodules related to lymphoma. If bone marrow was involved prior to therapy, no evidence of fluorodeoxyglucose avid disease in marrow. PR = ≥ 50% decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or nodal masses. No increase in other nodes, liver, or spleen. Splenic nodules regressed by ≥ 50% in their SPD or for single nodules, in the greatest transverse diameter; no new lesions. Participants who did not have any adequate response assessments during this period were not considered as responders. (NCT02285062)
Timeframe: From randomization date up to the data cut off date of 15 March 2019; median total treatment duration was 18.10 weeks for both treatment arms; range = 1.6 to 29.0 weeks for R2-CHOP arm and 0.3 to 22.9 weeks for placebo-R-CHOP arm

InterventionPercentage of Participants (Number)
Lenalidomide Plus R-CHOP (R2-CHOP)90.9
Placebo Plus R-CHOP90.9

[back to top]

Mean Change From Baseline in the Euroqol 5-Dimension 3-Level (EQ-5D-3L) Index Score

"The EQ-5D-3L is a generic, self-reported preference-based measure of health across five dimensions: mobility, self-care, pain, usual activities, and anxiety/depression. Each dimension has three levels of 'severity' corresponding to no problems, some problems and extreme problems. The instrument is scored as a single summary index using one of the available EQ-5D-3L value sets; in this study the UK scoring weights 9 were used. The UK index ranges from -0.594 to 1, where 0 equates to death and 1 equates to full health (-0.594 is considered 'worse than death')." (NCT02285062)
Timeframe: Baseline and Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionUnits on a Scale (Mean)
MidcycleC6 D1EoT = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)0.080.100.100.15
Placebo Plus R-CHOP0.080.140.060.09

[back to top]

Percentage of Participants Who Achieved a Complete Response (CR)

The percentage of participants who achieved a CR after initiation of the study treatment and prior to initiation of subsequent systemic antilymphoma therapy as assessed by the IRAC. A CR = complete metabolic response; target nodes/nodal masses regressed on computed tomography to (≤ 1.5 cm in their greatest transverse diameter for nodes > 1.5 cm prior to therapy. Regressed to normal size by imaging, and absence of nodules related to lymphoma. If bone marrow was involved prior to therapy, no evidence of fluorodeoxyglucose avid disease in marrow per International Working Group (IWG) 2014 for Non-Hodgkin's Lymphoma (NHL). Participants who did not have any adequate response assessments during this period were not considered as responders. (NCT02285062)
Timeframe: From randomization date up to the data cut off date of 15 March 2019; median follow-up was 24.5 months

InterventionPercentage of Participants (Number)
Lenalidomide Plus R-CHOP (R2-CHOP)69.1
Placebo Plus R-CHOP64.9

[back to top]

Mean Change From Baseline in the FACT-Lym Trial Outcome Index (TOI)

"The FACT-Lym questionnaire is a validated instrument for assessing the impact of lymphoma on HRQL and contains 42 questions covering HRQL and common lymphoma symptoms and treatment side-effects. It begins with the Functional Assessment of Cancer Therapy - General (FACT-G), which contains 27 questions covering four core subscales: Physical Wellbeing (7 items), Social/Family Wellbeing (7), Emotional Wellbeing (6), and Functional Wellbeing (7). The FACT-Lym also includes an Additional Concerns subscale (15 questions) used to assess NHL-related symptoms and concerns. All questions are answered on a 5-point scale ranging from not at all (0) to very much (4). The FACT-Lym TOI is calculated by summing the Physical Well-being, Functional Well-being and Additional Concerns scores and has a range of 0 to 116. Higher scores reflect better HRQoL or fewer symptoms." (NCT02285062)
Timeframe: Baseline and Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionUnits on a Scale (Mean)
MidcycleC6 D1EoT = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)2.65.99.113.5
Placebo Plus R-CHOP4.67.55.812.2

[back to top]

Kaplan-Meier Estimate of Progression Free Survival (PFS)

Progression free survival was defined as the time (months) from the date of randomization to the date of disease progression or death (any cause), whichever occurred earlier and was assessed by the Independent Response Adjudication Committee (IRAC). Relapse from complete response (CR) was considered as disease progression throughout the study. Disease progression was determined based on the Revised Response Criteria for Malignant Lymphoma. The PFS analysis was based on the censoring rules using the Food and Drug Administration (FDA) Guidance. Participants who did not experience disease progression and who did not die before the clinical data cutoff date were censored at the date of last adequate response assessment. (NCT02285062)
Timeframe: From the date of randomization up to the data cut off date of 15 March 2019; median follow-up of 24.5 months

Interventionmonths (Median)
Lenalidomide Plus R-CHOP (R2-CHOP)NA
Placebo Plus R-CHOPNA

[back to top]

Kaplan-Meier (K-M) Estimate of Event Free Survival (EFS)

EFS was defined as the time (months) from randomization until occurrence of one of the following events, whichever occurred first: • Disease progression • Initiation of subsequent systemic anti-lymphoma therapy • Death due to any cause The assessment of EFS was conducted by the IRAC using the International Working Group (IWG) criteria for NHL. Pre-specified optional therapies such as the extra 2 doses of single agent rituximab after Cycle 6 or consolidation radiotherapy did not count as an EFS event (initiation of subsequent systemic anti-lymphoma therapy) if the decision to treat and the location to be treated was determined prior to randomization. Participants who did not experience any of the events defined in the categories above before the clinical data cutoff date were censored at date last known alive. (NCT02285062)
Timeframe: From the date of randomization up to the data cut off date of 15 March 2019; median follow-up was 24.5 months

InterventionMonths (Median)
Lenalidomide Plus R-CHOP (R2-CHOP)NA
Placebo Plus R-CHOPNA

[back to top]

Mean Change From Baseline in the FACT-Lym Functional Well-Being Subscale

"The FACT-Lym questionnaire is a validated instrument for assessing the impact of lymphoma on HRQL and contains 42 questions covering HRQL and common lymphoma symptoms and treatment side-effects. It begins with the Functional Assessment of Cancer Therapy - General (FACT-G), which contains 27 questions covering four core subscales: Physical Wellbeing (7 items), Social/Family Wellbeing (7), Emotional Wellbeing (6), and Functional Wellbeing (7). The FACT-Lym also includes an Additional Concerns subscale (15 questions) used to assess NHL-related symptoms and concerns. All questions are answered on a 5-point scale ranging from not at all (0) to very much (4). The functional well-being subscale ranges from 0 to 28, where higher scores reflect better HRQoL." (NCT02285062)
Timeframe: Baseline and Midcycle = after Cycle 3 but before Cycle 4, Cycle 6 Day 1 (C6D1), End of Treatment (C6,D21), and Follow-Up Period up to Week 34

,
InterventionUnits on a Scale (Mean)
MidcycleC6 D1EoT = 3-4 weeks after C6Follow-Up Period: Week 34
Lenalidomide Plus R-CHOP (R2-CHOP)-0.50.01.02.3
Placebo Plus R-CHOP0.51.40.73.1

[back to top]

Proportion of Participants With Successful Response

"Rate of response to pneumococcal 13-valent conjugate vaccine defined as a four-fold rise from baseline to 28 days after immunization (day 43) for >= 2 of the 3 serotypes studied by OPA of antibodies from sera.~The proportion of successes will be estimated in each arm 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 in each arm based on the normal approximation." (NCT02309515)
Timeframe: Day 43

Interventionproportion of participants (Number)
Arm I (Lenalidomide, Pneumococcal 13-valent Conjugate Vaccine)0.2
Arm II (Pneumococcal 13-valent Conjugate Vaccine)0.0

[back to top]

Percentage of Participants With Event-free Survival (EFS)

This analysis includes all randomized subjects from the BMT CTN 0702 protocol classified according to their randomized treatment assignment (intention-to-treat). Event-free survival is defined as survival without disease progression, second primary malignancy, and death. To account for loss to follow-up, the Kaplan-Meier estimator was used to estimate event-free survival during the 5 year post-randomization follow-up period. (NCT02322320)
Timeframe: 5 years post-randomization in BMT CTN 0702

Interventionpercentage of participants (Number)
Tandem Auto Transplant44.2
RVD Consolidation42.1
Lenalidomide Maintenance42.4

[back to top]

Percentage of Participants With Secondary Primary Malignancies (SPM)

"This analysis includes all randomized subjects from the BMT CTN 0702 protocol classified according to their randomized treatment assignment (intention-to-treat). SPM is defined as development of any second malignancy, excluding non-melanoma skin cancers. To account for loss to follow-up, the Aalen-Johansen estimator was used to estimate the cumulative incidence of SPM during the 5 year post-randomization follow-up period.~The development of any SPMs excludes non-melanoma skin cancers. Death without SPMs will be considered a competing risk for this event. The cumulative incidence of SPMs will be compared between treatment arms." (NCT02322320)
Timeframe: 5 years post-randomization in BMT CTN 0702

Interventionpercentage of participants (Number)
Tandem Auto Transplant10.8
RVD Consolidation7.9
Lenalidomide Maintenance7.2

[back to top]

Percentage of Participants With Progression-free Survival (PFS)

This analysis includes all randomized subjects from the BMT CTN 0702 protocol classified according to their randomized treatment assignment (intention-to-treat). Progression-free survival is defined as survival without disease progression or initiation of non-protocol anti-myeloma therapy. To account for loss to follow-up, the Kaplan-Meier estimator was used to estimate progression-free survival during the 5 year post-randomization follow-up period. (NCT02322320)
Timeframe: 5 years post-randomization in BMT CTN 0702

Interventionpercentage of participants (Number)
Tandem Auto Transplant47.7
RVD Consolidation44.1
Lenalidomide Maintenance45.0

[back to top]

Percentage of Participants With Overall Survival (OS)

This analysis includes all randomized subjects from the BMT CTN 0702 protocol classified according to their randomized treatment assignment (intention-to-treat). Overall survival is defined as survival of death from any cause. To account for loss to follow-up, the Kaplan-Meier estimator was used to estimate overall survival during the 5 year post-randomization follow-up period. (NCT02322320)
Timeframe: 5 years post-randomization in BMT CTN 0702

Interventionpercentage of participants (Number)
Tandem Auto Transplant74.7
RVD Consolidation75.4
Lenalidomide Maintenance76.4

[back to top]

Percentage of Participants With Disease Progression

"This analysis includes all randomized subjects from BMT CTN 0702, classified by their treatment assignment (intention-to-treat). Disease progression is defined as progression of multiple myeloma, including one or more of the following:~Reappearance of serum monoclonal paraprotein at a level >= 0.5 g/dL~24-hour urine protein electrophoresis of at least 200mg paraprotein/24 hours~Abnormal free light chain levels of >10 mg/dl, only in patients without measurable paraprotein in serum and urine~At least 10% plasma cells in a bone marrow aspirate or trephine biopsy~Definite increase in the size of existing bone lesions or soft tissue plasmacytomas~Development of new bone lesions or soft tissue plasmacytomas~Development of hypercalcemia (corrected serum Ca >11.5 mg/dL or >2.8 mmol/L) not attributable to other causes~To account for loss to follow-up, the Aalen-Johansen estimator was used to estimate the cumulative incidence of progression during the follow-up period." (NCT02322320)
Timeframe: 5 years post-randomization in BMT CTN 0702

Interventionpercentage of participants (Number)
Tandem Auto Transplant48.8
RVD Consolidation54.3
Lenalidomide Maintenance54.2

[back to top]

The Estimated Percentage of Patients Alive Without Relapse or Progression at 2 Years

"Progression is defined as an increase of ≥ 25% from the lowest response value in any one or more of the following:~Serum M-component with an absolute increase ≥ 0.5 g/dL; and/or Urine M-component with an absolute increase ≥ 200 mg/24 hours; and/or Only in patients without measurable serum and urine M-protein levels: the difference between involved and uninvolved FLC levels (absolute increase must be >100mg/l) Bone marrow plasma cell percentage (absolute % must be ≥10% Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas Development of hypercalcemia (corrected serum calcium >11.5mg/100ml) that can be attributed solely to the plasma cell proliferative disorder" (NCT02331368)
Timeframe: 2 Years

Interventionpercentage of patients (Number)
Anti-PD-1 (MK-3475)83

[back to top]

The Estimated Percentage of Patients Alive at 2 Years

(NCT02331368)
Timeframe: 2 Years

Interventionpercentage of patients (Number)
Anti-PD-1 (MK-3475)100

[back to top]

The Number of Patients That Achieve Complete Response

The primary efficacy endpoint is complete response rate. The complete response rate was estimated by the observed proportion of complete responders at day 180. Complete response (CR) was defined as negative immunofixation of serum and urine and disappearance of any soft tissue plasmacytomas, and <5% plasma cells in bone marrow and negative bone marrow flow cytometry. (NCT02331368)
Timeframe: 180 days post-transplant

InterventionParticipants (Count of Participants)
Anti-PD-1 (MK-3475)7

[back to top]

Change From Baseline in Hemoglobin Levels

(NCT02335983)
Timeframe: Baseline and Cycle 1, days 8 and 15 and Cycle 2 days 1, 8, and 15

,,,,,
Interventiong/L (Mean)
BaselineChange from Baseline to Cycle 1 Day 8Change from Baseline to Cycle 1 Day 15Change from Baseline to Cycle 2 Day 1Change from Baseline to Cycle 2 Day 8Change from Baseline to Cycle 2 Day 15
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²111.50-1.25-1.75-3.75-0.130.00
NDMM Dose-expansion: Carfilzomib 56 mg/m²113.21-1.82-5.642.194.192.97
NDMM Dose-expansion: Carfilzomib 70 mg/m²120.00-5.56-6.50-3.29-2.86-5.14
RRMM Dose-evaluation: Carfilzomib 56 mg/m²116.90-1.50-6.90-5.70-6.00-8.50
RRMM Dose-evaluation: Carfilzomib 70 mg/m²120.67-3.33-9.50-9.25-8.17-11.33
RRMM Dose-expansion: Carfilzomib 70 mg/m²123.24-0.91-5.10-7.38-5.59-8.45

[back to top]

Change From Baseline in Neutrophil Count

(NCT02335983)
Timeframe: Baseline and Cycle 1, days 8 and 15 and Cycle 2 days 1, 8, and 15

,,,,,
Intervention10^9 cells/L (Mean)
BaselineChange from Baseline to Cycle 1 Day 8Change from Baseline to Cycle 1 Day 15Change from Baseline to Cycle 2 Day 1Change from Baseline to Cycle 2 Day 8Change from Baseline to Cycle 2 Day 15
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²3.040.591.080.020.570.33
NDMM Dose-expansion: Carfilzomib 56 mg/m²3.260.370.090.180.410.42
NDMM Dose-expansion: Carfilzomib 70 mg/m²3.24-0.190.430.090.460.56
RRMM Dose-evaluation: Carfilzomib 56 mg/m²2.530.020.280.360.18-0.30
RRMM Dose-evaluation: Carfilzomib 70 mg/m²3.400.03-0.09-0.33-0.67-0.54
RRMM Dose-expansion: Carfilzomib 70 mg/m²2.720.970.810.160.63-0.06

[back to top]

Change From Baseline in Platelet Count

(NCT02335983)
Timeframe: Baseline and Cycle 1, days 8 and 15 and Cycle 2 days 1, 8, and 15

,,,,,
Intervention10^9 cells/L (Mean)
BaselineChange from Baseline to Cycle 1 Day 8Change from Baseline to Cycle 1 Day 15Change from Baseline to Cycle 2 Day 1Change from Baseline to Cycle 2 Day 8Change from Baseline to Cycle 2 Day 15
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²214.002.38-23.25141.75-28.00-21.25
NDMM Dose-expansion: Carfilzomib 56 mg/m²233.03-21.42-53.39111.06-52.78-44.19
NDMM Dose-expansion: Carfilzomib 70 mg/m²242.89-51.11-101.88152.00-106.29-76.14
RRMM Dose-evaluation: Carfilzomib 56 mg/m²151.60-12.90-58.6055.00-44.10-44.40
RRMM Dose-evaluation: Carfilzomib 70 mg/m²202.50-49.50-66.8368.67-74.75-63.25
RRMM Dose-expansion: Carfilzomib 70 mg/m²172.38-17.36-66.3944.84-71.06-60.13

[back to top]

Number of Participants With Adverse Events (AEs)

"Safety and tolerability were evaluated according to the type, incidence, and severity of adverse events.~An AE is defined as any untoward medical occurrence in a clinical trial subject. The event does not necessarily have a causal relationship with study treatment.~A serious adverse event is defined as an AE that meets at least 1 of the following serious criteria:~fatal~life threatening~requires in-patient hospitalization or prolongation of existing hospitalization~results in persistent or significant disability/incapacity~congenital anomaly/birth defect~other medically important serious event~The severity of each AE was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.03 where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening, and Grade 5 = Death." (NCT02335983)
Timeframe: From the first dose of any study drug up to 30 days after the last dose of any study drug; median (range) duration of treatment with carfilzomib was 32 (1.1, 76.7) weeks in RRMM participants and 26 (1.0, 94.4) weeks in NDMM participants.

,,,,,
InterventionParticipants (Count of Participants)
Any adverse event (AE)AE Grade ≥ 3Serious adverse eventsAEs leading to discontinuation of carfilzomibAEs leading to discontinuation of lenalidomideAEs leading to discontinuation of dexamethasoneFatal adverse eventsTreatment-related adverse events (TRAE)Treatment-related AEs Grade ≥ 3Treatment-related serious adverse eventsTRAEs leading to discontinuation of carfilzomibTRAEs leading to discontinuation of lenalidomideTRAEs leading to discontinuation of dexamethasoneTreatment-related fatal adverse events
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²95300009510000
NDMM Dose-expansion: Carfilzomib 56 mg/m²3321111220321881220
NDMM Dose-expansion: Carfilzomib 70 mg/m²96322209532220
RRMM Dose-evaluation: Carfilzomib 56 mg/m²107411109611110
RRMM Dose-evaluation: Carfilzomib 70 mg/m²1295222011821110
RRMM Dose-expansion: Carfilzomib 70 mg/m²3423105552301865552

[back to top]

Change From Baseline in Bilirubin

(NCT02335983)
Timeframe: Baseline and Cycle 2 day 1

,,,,,
Interventionµmol/L (Mean)
BaselineChange from Baseline to Cycle 2 Day 1
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²10.263.42
NDMM Dose-expansion: Carfilzomib 56 mg/m²7.161.21
NDMM Dose-expansion: Carfilzomib 70 mg/m²6.840.49
RRMM Dose-evaluation: Carfilzomib 56 mg/m²8.550.86
RRMM Dose-evaluation: Carfilzomib 70 mg/m²9.981.00
RRMM Dose-expansion: Carfilzomib 70 mg/m²9.101.34

[back to top]

Time to Maximum Plasma Concentration of Carfilzomib on Day 8 of Cycle 1 by Dose Group

(NCT02335983)
Timeframe: Day 8 of Cycle 1 at predose, 15 minutes after the start of infusion, at the end of infusion, and 15 and 60 minutes after the end of infusion

Interventionhours (Median)
Carfilzomib 56 mg/m²0.28
Carfilzomib 70 mg/m²0.27

[back to top]

Progression-free Survival (PFS)

"PFS is defined as the time from the first day of study treatment to the earlier of disease progression or death due to any cause.~Disease progression was determined by the investigator according to IMWG-URC. Progressive Disease (PD): Increase of 25% from lowest value in serum M-component (absolute increase ≥ 0.5 g/dL), urine M-component (absolute increase ≥ 200 mg per 24 hours) and/or the difference between involved and uninvolved FLC levels (absolute increase >10 mg/dL) in patients without measurable serum and urine M-protein levels, and/or any new or increase in size of bone lesions or soft tissue plasmacytomas, or development of hypercalcemia.~PFS was analyzed using Kaplan-Meier methods. Participants who were alive with no documented PD, started new anticancer therapy before documentation of PD or death, had death or PD immediately after > 1 consecutively missed assessment visit, were lost to follow-up or withdrew consent were censored at the date of last disease assessment." (NCT02335983)
Timeframe: From first dose of study drug until the end of follow-up; Median time on follow-up was 10.6 months in RRMM participants and 6.9 months in NDMM participants.

Interventionmonths (Median)
RRMM Dose-evaluation: Carfilzomib 56 mg/m²NA
RRMM Dose-evaluation: Carfilzomib 70 mg/m²NA
RRMM Dose-expansion: Carfilzomib 70 mg/m²NA
RRMM: Combined Carfilzomib 70 mg/m²NA
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²NA
NDMM Dose-expansion: Carfilzomib 70 mg/m²NA
NDMM: Combined Carfilzomib 70 mg/m²NA
NDMM Dose-expansion: Carfilzomib 56 mg/m²NA

[back to top]

Overall Response Rate (ORR)

"Response was determined by the investigator based on the International Myeloma Working Group Uniform Response Criteria (IMWG URC). ORR was defined as the percentage of participants who achieved a best response of stringent complete response (sCR), complete response (CR), very good partial response (VGPR) or partial response (PR). Responses must have been confirmed in 2 consecutive assessments at any time prior to initiation of any new therapy.~Disease assessments included serum protein electrophoresis (SPEP) with immunofixation, urine protein electrophoresis (UPEP; 24-hour assessment) with immunofixation, serum free light chain (SFLC), quantitative immunoglobulins, bone marrow aspirates to determine percent plasma cell involvement, and skeletal imaging for plasmacytoma evaluation." (NCT02335983)
Timeframe: Response assessments were performed on day 1 of each treatment cycle from cycle 2 and then every 8 weeks during follow-up until disease progression. Median time on follow-up was 10.6 months in RRMM participants and 6.9 months in NDMM participants.

Interventionpercentage of participants (Number)
RRMM Dose-evaluation: Carfilzomib 56 mg/m²90.0
RRMM Dose-evaluation: Carfilzomib 70 mg/m²91.7
RRMM Dose-expansion: Carfilzomib 70 mg/m²88.2
RRMM: Combined Carfilzomib 70 mg/m²89.1
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²88.9
NDMM Dose-expansion: Carfilzomib 70 mg/m²77.8
NDMM: Combined Carfilzomib 70 mg/m²83.3
NDMM Dose-expansion: Carfilzomib 56 mg/m²97.0

[back to top]

Maximum Plasma Concentration of Carfilzomib on Day 8 of Cycle 1 by Dose Group

(NCT02335983)
Timeframe: Day 8 of Cycle 1 at predose, 15 minutes after the start of infusion, at the end of infusion, and 15 and 60 minutes after the end of infusion

Interventionng/mL (Mean)
Carfilzomib 56 mg/m²11700
Carfilzomib 70 mg/m²12400

[back to top]

Change From Baseline in Creatinine

(NCT02335983)
Timeframe: Baseline and Cycle 1, days 8 and 15 and Cycle 2 days 1, 8, and 15

,,,,,
Interventionµmol/L (Mean)
BaselineChange from Baseline to Cycle 1 Day 8Change from Baseline to Cycle 1 Day 15Change from Baseline to Cycle 2 Day 1Change from Baseline to Cycle 2 Day 8Change from Baseline to Cycle 2 Day 15
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²75.80-1.223.65-10.39-12.27-12.38
NDMM Dose-expansion: Carfilzomib 56 mg/m²80.60-0.220.42-4.31-4.32-1.99
NDMM Dose-expansion: Carfilzomib 70 mg/m²95.37-8.5129.39-17.17-20.08-21.85
RRMM Dose-evaluation: Carfilzomib 56 mg/m²80.18-2.65-4.42-3.80-6.45-6.28
RRMM Dose-evaluation: Carfilzomib 70 mg/m²82.88-5.30-4.60-7.74-11.09-13.26
RRMM Dose-expansion: Carfilzomib 70 mg/m²79.092.09-1.85-1.441.854.88

[back to top]

Duration of Response (DOR)

Duration of response (DOR) was calculated for participants who achieved a confirmed PR or better based on Investigator assessment and according to the IMWG URC. Duration of overall response is defined as the time from first documentation of response to disease progression or death due to any cause. DOR was analyzed using Kaplan-Meier methods. Participants who were alive with no documented PD, started new anticancer therapy before documentation of PD or death, had death or PD immediately after > 1 consecutively missed disease assessment visit, were lost to follow-up, or withdrew consent were censored at the date of last disease assessment. (NCT02335983)
Timeframe: From first dose of study drug until the end of follow-up; Median time on follow-up was 10.6 months in RRMM participants and 6.9 months in NDMM participants.

Interventionmonths (Median)
RRMM Dose-evaluation: Carfilzomib 56 mg/m²NA
RRMM Dose-evaluation: Carfilzomib 70 mg/m²NA
RRMM Dose-expansion: Carfilzomib 70 mg/m²NA
RRMM: Combined Carfilzomib 70 mg/m²NA
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²NA
NDMM Dose-expansion: Carfilzomib 70 mg/m²NA
NDMM: Combined Carfilzomib 70 mg/m²NA
NDMM Dose-expansion: Carfilzomib 56 mg/m²NA

[back to top]

Complete Response Rate (CRR)

"Complete Response Rate (CRR) is defined as the percentage of participants who achieved a best overall response of either stringent complete response (sCR) or complete response (CR) in accordance with International Myeloma Working Group-Uniform Response Criteria (IMWG-URC).~sCR: As for CR, and absence of clonal plasma cells in bone marrow (BM) CR: Negative serum and urine immunofixation, disappearance of any soft tissue plasmacytomas, < 5% plasma cells in BM, and normal SFLC ratio in participants with measurable disease only by SFLC." (NCT02335983)
Timeframe: Response assessments were performed on day 1 of each treatment cycle from cycle 2 and then every 8 weeks during follow-up until disease progression. Median time on follow-up was 10.6 months in RRMM participants and 6.9 months in NDMM participants.

Interventionpercentage of participants (Number)
RRMM Dose-evaluation: Carfilzomib 56 mg/m²20.0
RRMM Dose-evaluation: Carfilzomib 70 mg/m²16.7
RRMM Dose-expansion: Carfilzomib 70 mg/m²26.5
RRMM: Combined Carfilzomib 70 mg/m²23.9
NDMM Dose-evaluation: Carfilzomib 56/70 mg/m²22.2
NDMM Dose-expansion: Carfilzomib 70 mg/m²11.1
NDMM: Combined Carfilzomib 70 mg/m²16.7
NDMM Dose-expansion: Carfilzomib 56 mg/m²33.3

[back to top]

Area Under the Curve From Time Zero to Time of Last Quantifiable Concentration of Carfilzomib on Day 8 of Cycle 1 by Dose Group

(NCT02335983)
Timeframe: Day 8 of Cycle 1 at predose, 15 minutes after the start of infusion, at the end of infusion, and 15 and 60 minutes after the end of infusion

Interventionhr*ng/mL (Mean)
Carfilzomib 56 mg/m²4150
Carfilzomib 70 mg/m²9130

[back to top]

Median Time to Response

Time to response is defined as the time from the first dose of study drug to the first documentation of response partial response (PR) or better. Disease response was assessed using International Myeloma Working Group (IMWG) response criteria: Partial Response (PR) defined as 50% reduction of serum M-protein and reduction in 24h urinary M-protein by ≥90% or to <200mg/24h; Very Good Partial Response (VGPR) defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein plus urine M-protein level <100mg/24h; Complete Response (CR) defined as negative immunofixation on serum and urine and, disappearance of any soft tissue plasmacytomas and, <5% plasma cells in BM; Stringent Complete Response (sCR) defined as normal FLC ratio and absence of clonal cells in BM by immunohistochemistry or 2-4 color flow cytometry. (NCT02375555)
Timeframe: Participants were followed up to 92 days.

Interventiondays (Median)
Elotuzumab, Lenalidomide, Bortezomib, and Dexamethasone22.0

[back to top]

Grade 3 and 4 Treatment-Emergent Adverse Event (TEAE) Rate

Grade 3 and 4 TEA rate was defined as the proportion of participants who experienced a grade 3 or 4 adverse event of any attribution based on NCI Common Toxicity Criteria for Adverse Events Version 4 (CTCAEv4) on treatment. (NCT02375555)
Timeframe: The adverse event observation period defined as the time on treatment (+30d) is 278 weeks.

Interventionproportion of participants (Number)
Elotuzumab, Lenalidomide, Bortezomib, and Dexamethasone0.70

[back to top]

4 Cycle Response Rate

Disease response was defined as the proportion of patients who achieved a response of partial response or better using International Myeloma Working Group (IMWG) response criteria. Partial Response (PR) defined as 50% reduction of serum M-protein and reduction in 24h urinary M-protein by ≥90% or to <200mg/24h; Very Good Partial Response (VGPR) defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein plus urine M-protein level <100mg/24h; Complete Response (CR) defined as negative immunofixation on serum and urine and, disappearance of any soft tissue plasmacytomas and, <5% plasma cells in BM; Stringent Complete Response (sCR) defined as normal FLC ratio and absence of clonal cells in BM by immunohistochemistry or 2-4 color flow cytometry. The Clopper and Pearson method was used to estimate the 95% CIs for the response rate at Week 12. (NCT02375555)
Timeframe: Participants were followed up to 12 weeks.

Interventionproportion of participants (Number)
Elotuzumab, Lenalidomide, Bortezomib, and Dexamethasone0.971

[back to top]

Successful Stem Cell Mobilization (SC Mob) Rate

Successful SC Mob defined as the proportion of participants with the ability to collect a total of at least 2*10^6 CD34+ cells/kg. (NCT02375555)
Timeframe: The most distant time of stem cell mobilization from time of registration is 21.4 weeks with a median of 15.14 weeks.

Interventionproportion of participants (Number)
Elotuzumab, Lenalidomide, Bortezomib, and Dexamethasone0.968

[back to top]

Objective Response Rate (ORR) at End of 8 Cycles of Induction Therapy.

ORR was defined as the proportion of patients who achieved a disease response of partial response (PR) or better using IMWG response criteria. PR defined as 50% reduction of serum M-protein and reduction in 24h urinary M-protein by ≥90% or to <200mg/24h; Very Good Partial Response (VGPR) defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or ≥90% reduction in serum M-protein plus urine M-protein level <100mg/24h; Complete Response (CR) defined as negative immunofixation on serum and urine and, disappearance of any soft tissue plasmacytomas and, <5% plasma cells in BM; Stringent Complete Response (sCR) defined as normal FLC ratio and absence of clonal cells in BM by immunohistochemistry or 2-4 color flow cytometry. (NCT02375555)
Timeframe: Participants were followed up to 24 weeks.

Interventionproportion of participants (Number)
All Patients Treated With E-RVD.0.950

[back to top]

4 Cycle Ever Dose Modification (DM) Rate

The 4 cycle ever DM rate is the proportion of participants who started therapy and required dose modification of any study drug during the first four cycles of E-RVD. (NCT02375555)
Timeframe: Participants were followed up to 12 weeks.

Interventionproportion of participants (Number)
Elotuzumab, Lenalidomide, Bortezomib, and Dexamethasone.40

[back to top]

Time to Progression (TTP) by IRC Evaluation

TTP is defined as the time from the first administration of any study drug until documented DLBCL progression or death as a result of lymphoma. (NCT02399085)
Timeframe: Approximately 2.5 years after first participant enrolled

InterventionMonths (Median)
Tafasitamab (MOR00208) + Lenalidomide (LEN)16.2

[back to top]

Number of Participants That Experienced Treatment-emergent Adverse Events (TEAEs)

TEAEs are defined as any adverse event reported in the following time interval (including the lower and upper limits): date of first administration of study treatment; date of last administration of study treatment + 30 days, or if they are considered to be related to the study drug. (NCT02399085)
Timeframe: Approximately 6.5 years after first participant enrolled

InterventionParticipants (Count of Participants)
Tafasitamab (MOR00208) + Lenalidomide (LEN)81

[back to top]

Disease Control Rate (DCR) by IRC Evaluation

DCR = CR + PR + SD (Stable disease); IRC Evaluation DCR was defined as the number of participants having CR, PR or SD based on the best objective response achieved at any time during the study. (NCT02399085)
Timeframe: Approximately 2.5 years after first participant enrolled

InterventionParticipants (Count of Participants)
Tafasitamab (MOR00208) + Lenalidomide (LEN)59

[back to top]

DCR by INV Evaluation

DCR = CR + PR + SD (Stable disease); IRC Evaluation DCR was defined as the number of participants having CR, PR or SD based on the best objective response achieved at any time during the study. (NCT02399085)
Timeframe: Approximately 2.5 years after first participant enrolled

InterventionParticipants (Count of Participants)
Tafasitamab (MOR00208) + Lenalidomide (LEN)60

[back to top]

Duration of Response (DoR) by IRC Evaluation

DoR [months] = (date of assessment of tumor progression or death - date of assessment of first documented response of (CR or PR) + 1)/30.4375. (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionMonths (Median)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Treatment (MOR00208, Lenalidomide)43.9NA

[back to top] [back to top]

Serum Drug Levels of MOR00208

"The pharmacokinetics (PK) profile of MOR00208 was investigated by quantifying serum drug levels at baseline and after repeated IV administrations for up to 24 treatment cycles using sparse sampling.~MOR00208 PK sample was taken pre-dose and 1 hour ± 10 min after the end of MOR00208 infusion for Cycle 1 to Cycle 23. MOR00208 PK sample (pre-dose only) were taken in odd numbered additional treatment cycles only (e.g., treatment Cycles 13, 15,17, 19, 21, 23)." (NCT02399085)
Timeframe: Cycle 1 Days 1, 4, 15 predose and 1 hr post-dose; Cycle 2 Days 1, 15 predose and 1 hr post-dose; Cycle 3 Days 1, 15 predose and 1 hr post-dose; Cycles 4, 5, 6, 7, 9, 11,13, 15, 17, 19, 21, 23 Day 1 predose; End of Treatment

Interventionng/mL (Mean)
Cycle 1 Day 1 (Predose)Cycle 1 Day 1 (1 hour post dose)Cycle 1 Day 4 (pre-dose)Cycle 1 Day 4 (1 hour post-dose)Cycle 1 Day 15 (pre dose)Cycle 1 Day 15 (1 hour post-dose)Cycle 2 Day 1 (predose)Cycle 2 Day 1 (1 hour post-dose)Cycle 2 Day 15 (Pre Dose)Cycle 2 Day 15 (1 hour post-dose) )Cycle 3 Day 1 (predose)Cycle 3 Day 1 (1 hour post-dose)Cycle 3 Day 15 (predose)Cycle 3 Day 15 (1 hour post-dose)Cycle 4 Day 1 (predose) )Cycle 5 Day 1 (pre dose)Cycle 6 Day 1 (pre dose)Cycle 7 Day 1 (pre dose)Cycle 9 Day 1 (pre dose)Cycle 11 Day 1 (pre dose)Cycle 13 Day 1 (pre dose)Cycle 15 Day 1 (pre dose)Cycle 17 Day 1 (pre dose)Cycle 19 Day 1 (pre dose)Cycle 21 Day 1 (pre dose)Cycle 23 Day 1 (pre dose)End of Treatment
Tafasitamab (MOR00208) + Lenalidomide (LEN)6.7249075.9126306.8363626.2157722.3396262.1181870.8439788.2217846.9442940.2208520.6466135.8223909.4455635.0216328.4142134.4115132.3114661.5108640.4126472.0100853.5159676.5175855.1197045.0197228.0224253.3141240.7

[back to top]

Progression-free Survival (PFS) by IRC Evaluation

PFS time was defined as the time (in months) from the date of the first administration of any study drug to the date of tumor progression or death from any cause. (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionMonths (Median)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Tafasitamab (MOR00208) + Lenalidomide (LEN)11.611.6

[back to top]

PFS by INV Evaluation

PFS time was defined as the time (in months) from the date of the first administration of any study drug to the date of tumor progression or death from any cause. (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionMonths (Median)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Tafasitamab (MOR00208) + Lenalidomide (LEN)9.19.1

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of the first administration of any study drug until death from any cause (documented by the date of death). (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionMonths (Median)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Tafasitamab (MOR00208) + Lenalidomide (LEN)33.533.5

[back to top]

Number of Participants With Best Objective Response Rate (ORR)

"ORR = complete response [CR] + partial response [PR]; Independent Radiology/Clinical Review Committee (IRC) Evaluation.~ORR after MOR00208 and Lenalidomide combination therapy assessed by the IRC evaluation.~ORR was defined as the number of participants of the total number of participants treated with MOR00208 + LEN with CR or PR as best response achieved at any time during the study." (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionParticipants (Count of Participants)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Treatment (MOR00208, Lenalidomide)4646

[back to top]

Event-free Survival (EFS) by IRC Evaluation

EFS is defined as the time (in months) from the date of the first administration of any study drug to the date of tumour progression, first initiation of a new non-study anti-neoplastic therapy or death from any cause whichever comes first. (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionMonths (Median)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Tafasitamab (MOR00208) + Lenalidomide (LEN)8.79.1

[back to top]

DoR by Investigator (INV) Evaluation

DoR [months] = (date of assessment of tumour progression or death - date of assessment of first documented response of (CR or PR) + 1)/30.4375. (NCT02399085)
Timeframe: Approximately 4.5 years after first participant enrolled; Approximately 6.5 years after first participant enrolled

InterventionMonths (Median)
Approximately 4.5 years after first participant enrolledApproximately 6.5 years after first participant enrolled
Tafasitamab (MOR00208) + Lenalidomide (LEN)43.943.4

[back to top]

TTP by INV Evaluation

TTP is defined as the time from the first administration of any study drug until documented DLBCL progression or death as a result of lymphoma. (NCT02399085)
Timeframe: Approximately 2.5 years after first participant enrolled

InterventionMonths (Median)
Tafasitamab (MOR00208) + Lenalidomide (LEN)14.1

[back to top]

Number of Participants With Peripheral Blood Monoclonal Protein Reduction or Stabilization

Efficacy will be assessed monthly during NKAE treatment (4 months) by peripheral blood monoclonal protein monitoring. During follow-up, efficacy will be evaluated monthly the first 6 months. After that, quarterly until one year of follow-up. (NCT02481934)
Timeframe: 16 months

Interventionparticipants (Number)
NKAE Cells Infusion + Chemotherapy5

[back to top]

Number of Participants With Adverse Events During NKAE Treatment

Toxicity will be assessed by adverse events count during NKAE treatment monitoring peripheral blood absolute neutrophil count (cells/μl). Toxicity will be evaluated monthly during NKAE treatment (4 months). During follow-up, it will be assessed monthly the first 6 months. After that, quarterly until one year of follow-up, based on Common Toxicity Criteria for Adverse Events of the National Cancer Institute (CTCAE) to v.4.03. (NCT02481934)
Timeframe: 16 months

Interventionparticipants (Number)
NKAE Cells Infusion + Chemotherapy2

[back to top]

Number of Participants Experiencing a Dose-limiting Toxicity (DLT)

Number of participants experiencing a dose-limiting toxicity (DLT) is reported below. Dose-limiting toxicity is graded by the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. (NCT02492750)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
Phase I - Dose Level 10
Phase I - Dose Level 20
Phase I - Dose Level 30

[back to top] [back to top]

Survival Duration Without Disease Progression

Calculate rate of progression-free survival for subjects following treatment BiRd regimen compared to Rd treatment regimen. Progression is determined by the International Myeloma Working Group Criteria. (NCT02516696)
Timeframe: Until disease progression or death from any cause, for a maximum of approximately 5 years

Interventiondays (Median)
BiRD Treatment Regimen661
Rd Treatment Regimen1694

[back to top]

Overall Survival

Survival following treatment to the date of death of subjects on BiRd regimen as compared to Rd. (NCT02516696)
Timeframe: 4 years

Interventiondays (Median)
BiRD Treatment RegimenNA
Rd Treatment Regimen1014

[back to top]

Overall Response Rate

Capture the number of subjects who demonstrate a complete or partial response to treatment with BiRD regimen, as compared to Rd. Complete and partial responses are defined by the International Myeloma Working Group Criteria. (NCT02516696)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
BiRD Treatment Regimen7
Rd Treatment Regimen4

[back to top]

Number of Patients With Objective Response Rate (CR+PR)

(NCT02516696)
Timeframe: up to 3 years

InterventionParticipants (Count of Participants)
BiRD Treatment Regimen7
Rd Treatment Regimen4

[back to top]

Number of Patients With Complete Response Rate (CR)

Complete response is defined by the International Myeloma Working Group Criteria. (NCT02516696)
Timeframe: up to 3 years

InterventionParticipants (Count of Participants)
BiRD Treatment Regimen2
Rd Treatment Regimen0

[back to top]

Number of Days for Event-Free Survival

Descriptively presented for each treatment group and no formal statistical comparison will be made between treatment arms (NCT02516696)
Timeframe: approximately 5 years

Interventiondays (Median)
BiRD Treatment Regimen336
Rd Treatment Regimen821

[back to top]

Number of Days for Duration of Response

Descriptively presented for each treatment group and no formal statistical comparison will be made between treatment arms (NCT02516696)
Timeframe: up to 3 years

Interventiondays (Median)
BiRD Treatment Regimen308
Rd Treatment Regimen803

[back to top]

Number of Days After Initiating Treatment With BiRd Regimen to Disease Progression, as Compared to Subjects on Rd Treatment Regimen.

Progression is determined by the International Myeloma Working Group Criteria. (NCT02516696)
Timeframe: Until disease progression for a maximum of approximately 5 years

Interventiondays (Median)
BiRD Treatment Regimen661
Rd Treatment Regimen1694

[back to top]

Number of Adverse Events Experienced

Capture the number of adverse events experienced with BiRd regimen as compared to Rd regimen (NCT02516696)
Timeframe: 2 years

Interventionadverse events (Number)
BiRD Treatment Regimen79
Rd Treatment Regimen67

[back to top]

Functional Assessment of Chronic Illness Therapy - Fatigue Subscale (FS; Version 4) Score of Patients Receiving BiRd vs Rd Treatment

"The Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-F) is a 40-item measure that assesses self-reported fatigue and its impact upon daily activities and function. The FACIT- Fatigue Subscore (FS) is comprised of 13 items, within the total 40-item FACIT-F, that assess fatigue and its impact. This analysis is only based on the FS score. Items are scored on a 5 point Likert-type scale. Item scores can range from 0 (not at all) to 4 (very much), and the total, summed score from 0 to 52; lower scores indicate greater fatigue. The recall period for each item is the past 7 days. Data is descriptively presented for each treatment group and no formal statistical comparison will be made between treatment arms." (NCT02516696)
Timeframe: up to 3 years

InterventionScore on a scale (Mean)
BiRD Treatment Regimen32.75
Rd Treatment Regimen37.24

[back to top]

Apparent Total Clearance (CL/F) of Lenalidomide

Apparent volume of distribution, calculated as [(CL/F)/λz]. (NCT02538965)
Timeframe: Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

Interventionml/min (Geometric Mean)
Lenalidomide172.09

[back to top]

Apparent Volume of Distribution (Vz/F) of Lenalidomide

Apparent volume of distribution, calculated as [(CL/F)/λz]. (NCT02538965)
Timeframe: Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

InterventionLiters (Geometric Mean)
Lenalidomide34.42

[back to top]

Area Under the Plasma Concentration Time Curve From 0 Extrapolated to Infinity (AUC-inf, AUC0∞) Of Lenalidomide

Area under the plasma concentration-time curve from time 0 extrapolated to infinity, calculated as [AUCt + Ct/ λz]. Ct is the last quantifiable concentration. No AUC extrapolation was performed with unreliable λz. (NCT02538965)
Timeframe: Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

Interventionng*h/mL (Geometric Mean)
Lenalidomide5656.67

[back to top]

Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration of Lenalidomide (AUC-t)

Area under the plasma concentration-time curve from time 0 to the time of the last quantifiable concentration, calculated by linear trapezoidal method when concentrations were increasing and the logarithmic trapezoidal method when concentrations were decreasing. (NCT02538965)
Timeframe: Pharmacokinetic (PK) sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

Interventionng*h/mL (Geometric Mean)
Lenalidomide5378.83

[back to top]

Number of Participants Who Achieved a Best Response of Morphologic Complete Remission, Morphologic Complete Remission Incomplete or Partial Remission

"Overall response rate was defined as the number of participants with best response of CR, CRi or PR.~A CR was defined as:~ANC ≥ 1000/μL and platelets ≥ 100,000 without transfusions and/or exogenous growth factor support (no transfusion or exogenous growth factor within 7 days of assessment);~BM < 5% blasts evidence of trilineage hematopoiesis;~No evidence of extramedullary disease.~A CRi was defined as:~ANC < 1000/μL and platelets < 100,000/μL or > 100,000/μL without platelet recovery (requiring transfusion within 7 days of assessment);~BM with < 5% blasts and evidence of trilineage hematopoiesis;~No evidence of extramedullary disease.~A PR was defined as:~ANC of ≥ 1000/μL and platelets ≥ 100,000 without transfusions and/or exogenous growth factor support (no transfusion or exogenous growth factor within 7 days of assessment);~BM with 5% to 25% blasts and at least a 50% decrease in BM blast percent from baseline;~No evidence of extramedullary disease." (NCT02538965)
Timeframe: Response was assessed at the completion of the 21-day treatment period of cycles 1, 2, 3.

InterventionParticipants (Count of Participants)
Lenalidomide1

[back to top]

Maximum Observed Concentration (Cmax) of Lenalidomide

Maximum observed plasma concentration, obtained directly from the observed concentration versus time data. (NCT02538965)
Timeframe: PK sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

Interventionng/mL (Geometric Mean)
Lenalidomide1252.08

[back to top]

Number of Participants Who Achieved a Morphologic Complete Response Within the First Four Cycles of Lenalidomide Treatment According to the Modified International Working Group (IWG) Criteria

"The morphological complete response rate was defined as the total number of participants with morphological CR observed within the first 4 cycles of lenalidomide (regardless of whether the CR/CRi was observed at the end of Cycle 1, 2, 3 or 4) over the total number of participants evaluable for this endpoint. According to Modified IWG criteria, morphologic CR was defined as:~Absolute neutrophil count (ANC) ≥ 1000/μL and platelets ≥ 100,000 without transfusions and/or exogenous growth factor support (i.e., no transfusion or exogenous growth factor within 7 days of assessment;~Bone marrow < 5% blasts evidence of trilineage hematopoiesis;~No evidence of extramedullary disease.~Morphologic CRi was defined as:~ANC < 1000/μL and platelets < 100,000/μL or > 100,000/μL without platelet recovery (requiring transfusion within 7 days of assessment);~BM with < 5% blasts and evidence of trilineage hematopoiesis;~No evidence of extramedullary disease." (NCT02538965)
Timeframe: From day of the first dose of IP to end of cycle 4; Response was assessed at the completion of the 21-day treatment period of cycles 1, 2, 3, and 4 and at treatment discontinuation.

InterventionParticipants (Count of Participants)
Lenalidomide1

[back to top]

Percentage of Participants With of Graft Versus Host Disease (GVHD)

Acute graft versus host disease generally occurs after allogeneic hematopoietic stem cell transplantation. It is a reaction of donor immune cells against host tissues. The 3 main tissues that acute GVHD affects are the skin, liver, and gastrointestinal tract. Chronic GVHD is scored per the National Institute of Health consensus conference grading system. Clinical manifestations of chronic GVHD include skin involvement resembling lichen planus or the cutaneous manifestations of scleroderma; dry oral mucosa with ulcerations and sclerosis of the gastrointestinal tract; and a rising serum bilirubin concentration. (NCT02538965)
Timeframe: From the first dose of study drug to 28 days after the last dose of study drug; up to data cut off date of 31 December 2017; maximum treatment was 12 weeks

InterventionPercentage of Participants (Number)
Lenalidomide0

[back to top]

Terminal Half-Life (t1/2) of Lenalidomide

Terminal phase half-life in plasma, calculated as [(ln 2)/λz]. Terminal half-life was only calculated when a reliable estimate for λz could be obtained. (NCT02538965)
Timeframe: Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

Interventionhours (Geometric Mean)
Lenalidomide2.311

[back to top]

Time to Reach Maximum Concentration (Tmax) of Lenalidomide

Time to cmax was obtained directly from the observed concentration versus time data. (NCT02538965)
Timeframe: Pk sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.

InterventionHours (Median)
Lenalidomide2.000

[back to top]

Number of Participants Who Experienced Treatment Emergent Adverse Events (TEAE)

A TEAE was defined as any adverse event (AE) occurring or worsening on or after the first treatment of lenalidomide and within 28 days after the last dose. A serious AE = any AE which results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. The severity of AEs was graded based on National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), Version 4.3 and based on the following scale: Grade 1 = Mild Grade 2 = Moderate Grade 3 = Severe Grade 4 = Life threatening Grade 5 = Death. (NCT02538965)
Timeframe: From the first dose of study drug until 28 days after the last dose of study drug; up to data cut off date of 31 December 2017; maximum duration of treatment was 12 weeks

InterventionParticipants (Count of Participants)
Any TEAEAny TEAE Related to LenalidomideAny Grade 3/4 TEAEAny Grade 3/4 TEAE Related to LenalidomideAny Grade 5 TEAEAny Serious TEAEAny Serious TEAE Related to LenalidomideAny Serious TEAE Leading to Dose DiscontinuationAny TEAE Leading to Dose DiscontinuationAny TEAE Leading to Dose ReductionAny TEAE Leading to Dose InterruptionAny TEAE Leading to Death
Lenalidomide17151711113533471

[back to top]

Number of Participants Who Received a Haematopoietic Stem Cell Transplant (HSCT)

The number of participants who had undergone a haematopoietic stem cell transplant was calculated over the total number of participants in the ITT population. Percentages were also calculated based on whether the transplantation was the first, second, or subsequent transplant post IP administration. (NCT02538965)
Timeframe: From first dose of study drug up to 5 years post HSCT

InterventionParticipants (Count of Participants)
Any Transplant After Treatment StartFirst Transplant After Treatment StartSecond Transplant After Treatment StartAny Subsequent Transplant After Treatment Start
Lenalidomide2200

[back to top]

Number of Participants With a Morphologic CR, CRi, PR or Treatment Failure at Cycles 1, 2 and 3

"Disease assessment outcome at the end of Cycles 1-3 based on Cheson criteria:~Morphologic CR =~ANC ≥ 1000/μL and platelet ≥ 100,000 without transfusions and/or exogenous growth factor support (no transfusion or exogenous growth factor within 7 days of assessment)~BM < 5% blasts evidence of trilineage hematopoiesis~No evidence of extramedullary disease Morphologic CRi =~1. ANC< 1000/μL and Platelets < 100,000/μL or > 100,000/μL without platelet recovery (requiring transfusion within 7 days of assessment) 2. BM with < 5% blasts and evidence of trilineage hematopoiesis 3. No evidence of extramedullary disease PR =~ANC ≥ 1000/μL and platelets ≥ 100,000 without transfusions and/or exogenous growth factor support~BM with < 5%-25% blasts and at least a 50% decrease in BM blast percent from baseline~No evidence of extramedullary disease Treatment Failure = resistant disease; survival ≥ 7 days post-therapy; failed to achieve CR, CRi, or PR but stable with persistent AML" (NCT02538965)
Timeframe: Response was assessed at the completion of the 21-day treatment period of cycles 1, 2, 3.

InterventionParticipants (Count of Participants)
Cycle 1 Morphologic CRCycle 1 Morphologic CRiCycle 1 PRCycle 1 Treatment FailureCycle 2 Morphologic CRCycle 2 Morphologic CRiCycle 2 PRCycle 2 Treatment FailureCycle 3 Morphologic CRCycle 3 Morphologic CRiCycle 3 PRCycle 3 Treatment Failure
Lenalidomide0011301050100

[back to top]

Number of Participants With Adverse Events Graded According to CTC (Phase II)

"The toxicity profile will be further assessed based on phase II patients. Overall toxicity incidence of maximum tolerated dose level of the Intent to treat (ITT) group of subjects is summarized.~39 subjects were dosed with 10 mg dose of Lenalidamide as the ITT group." (NCT02561273)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
grade 3,4 neutropeniagrade 3, 4 leukopeniagrade 3, 4 anemiagrade 3, 4 thrombocytopeniagrade 3, 4 lymphopeniagrade 3, 4 febrile neutropeniagrade 3, 4 diarrheagrade 3, 4 Peripheral sensory neuropathygrade 3, 4 fatiguegrade 3, 4 nauseagrade 3, 4 anorexiagrade 3, 4 vomitinggrade 3, 4 mucositis oralgrade 3, 4 Rash maculo-papulargrade 3, 4 hypotensiongrade 3, 4 back pain
Treatment (Combination Chemotherapy, Lenalidomide)2725171718153211111111

[back to top]

Number of Participants With Adverse Events Graded According to Common Toxicity Criteria (CTC) (Phase I)

Non-hematologic toxicities will be evaluated via the ordinal CTC standard toxicity grading. Hematologic toxicity measures of thrombocytopenia, neutropenia, and leukopenia will be assessed using continuous variables as the outcome measures (primarily nadir) as well as categorization via CTC standard toxicity grading. Overall toxicity incidence as well as toxicity profiles by dose level and patient will be explored and summarized. (NCT02561273)
Timeframe: Up to 6 cycles of treatment (approximately 5 months)

,
InterventionParticipants (Count of Participants)
grade 3,4 neutropeniagrade 3, 4 anemiagrade 3, 4 thrombocytopeniagrade 3,4 neutropenia fevergrade 3, 4 diarrheagrade 3, 4 hyperglycemiagrade 3, 4 hypokalemiagrade 3, 4 hypotensiongrade 3, 4 hyperbilirubinemiagrade 3, 4 mucositisgrade 3,4 nauseagrade 3,4 vomiting
10 mg Lenalidomide Participants532400000000
15 mg Lenalidomide Participants433022111221

[back to top]

Progression-free Survival

The distribution of PFS will be estimated using the method of Kaplan-Meier. The PFS rate at 2 years will be estimated. A 2-year PFS rate of 60% will be considered of interest. (NCT02561273)
Timeframe: Time from registration to progression or death due to any cause, assessed up to 2 years

Interventionpercentage of participants (Number)
Treatment (Combination Chemotherapy, Lenalidomide)55

[back to top]

Overall Survival

The distribution of overall survival will be estimated using the method of Kaplan-Meier. (NCT02561273)
Timeframe: Time from registration to death due to any cause, assessed up to 1 year

Interventionpercentage of participants (Number)
Treatment (Combination Chemotherapy, Lenalidomide)89

[back to top]

Overall Response Rate

The ORR will be estimated by the total number of patients who achieve a PR or CR at the end of six cycles of treatment divided by the total number of evaluable patients. Exact binomial 95% confidence intervals for the true ORR will be calculated. (NCT02561273)
Timeframe: Up to the completion of course 6 (18 weeks)

Interventionpercentage of participants (Number)
Treatment (Combination Chemotherapy, Lenalidomide)69

[back to top]

Maximum Tolerated Dose (MTD) of Lenalidomide and CHOEP

MTD is defined as the dose level below the lowest dose that induces dose-limiting toxicity in at least one-third of patients (at least 2 of a maximum of 6 new patients) (Phase I) within the first cycle of study treatment. (NCT02561273)
Timeframe: 21 days

Interventionmilligrams (Number)
Treatment (Combination Chemotherapy, Lenalidomide)10

[back to top]

Complete Response Rate (Phase II)

A success is defined to be an objective status of CR after completion of 6 cycles of treatment. The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients. A 95% confidence interval for the true overall CR rate will be calculated according to the method of Duffy and Santner. (NCT02561273)
Timeframe: Up to the completion of course 6 (18 weeks)

Interventionpercentage of participants (Number)
Treatment (Combination Chemotherapy, Lenalidomide)49

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to death due to any cause. OS was calculated from the product-limit (Kaplan-Meier) method for censored data. Participants without documented death at the time of the final analysis were censored at the date of the last follow-up. This is an event-driven (events of death) outcome measure. At the time of data cut-off, there were an insufficient number of events from the censored data to be able to estimate certain parameters (e.g. medians). The database cutoff date was August 3, 2020. (NCT02579863)
Timeframe: Up to approximately 55 months

InterventionMonths (Median)
Pembrolizumab + Lenalidomide + DexamethasoneNA
Lenolidomide + DexamethasoneNA

[back to top]

Number of Participants Who Experienced One or More Adverse Events (AEs)

An AE was defined as any unfavorable and unintended sign including an abnormal laboratory finding, symptom or disease associated with the use of a medical treatment or procedure, regardless of whether it was considered related to the medical treatment or procedure, that occurred during the course of the study. The database cutoff date was August 3, 2020. (NCT02579863)
Timeframe: Up to approximately 55 months

InterventionParticipants (Count of Participants)
Pembrolizumab + Lenalidomide + Dexamethasone152
Lenolidomide + Dexamethasone141

[back to top]

Number of Participants Discontinuing Study Treatment Due to an AE

An AE was defined as any unfavorable and unintended sign including an abnormal laboratory finding, symptom or disease associated with the use of a medical treatment or procedure, regardless of whether it was considered related to the medical treatment or procedure, that occurred during the course of the study. The database cutoff date was August 3, 2020. (NCT02579863)
Timeframe: Up to approximately 55 months

InterventionParticipants (Count of Participants)
Pembrolizumab + Lenalidomide + Dexamethasone44
Lenolidomide + Dexamethasone26

[back to top]

Duration of Response (DOR) Evaluated According to IMWG Response Criteria by CAC Blinded Central Review

Response duration was defined as the time from first documented evidence of at least a partial response (sCR+CR+VGPR+PR]), until confirmed disease progression or death. DOR was calculated from product-limit (Kaplan-Meier) method for censored data. This is an event-driven (events of disease progression and death) outcome measure. At the time of data cut-off, there were an insufficient number of events from the censored data to be able to estimate certain parameters (e.g. medians). Full Range is the minimum and maximum of the observed duration of response. The data cutoff date was July 9, 2018. (NCT02579863)
Timeframe: Up to approximately 30 months

InterventionMonths (Median)
Pembrolizumab + Lenalidomide + DexamethasoneNA
Lenolidomide + DexamethasoneNA

[back to top]

Disease Control Rate (DCR) Evaluated According to the IMWG Response Criteria by CAC Blinded Central Review

Disease control rate was defined as the percentage of participants who achieved confirmed sCR, CR, VGPR, PR, or have demonstrated SD for at least 12 weeks prior to any evidence of progression. CR = negative immunofixation of serum and urine AND disappearance of any soft tissue plasmacytomas AND <5% plasmacytomas in the bone marrow; VGPR = serum and urine M-component detectable by immunofixation but not on electrophoresis OR ≥ 90% reduction in serum M-component plus urine M-component <100 mg/24 hr; PR = ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to <200 mg/24 hours; SD = not meeting the criteria for CR, VGPR, PR, or PD; PD = development of new bone lesions or soft tissue plasmacytomas or on a definite increase in the size of existing bone lesions or soft tissue plasmacytomas. Data cutoff date was July 9, 2018. (NCT02579863)
Timeframe: Up to approximately 30 months

InterventionPercentage of participants (Number)
Pembrolizumab + Lenalidomide + Dexamethasone89.1
Lenolidomide + Dexamethasone91.6

[back to top]

Progression Free Survival (PFS) Evaluated According to the International Myeloma Working Group (IMWG) Response Criteria 2011 by Clinical Adjudication Committee (CAC) Blinded Central Review

PFS was defined as the time from randomization to the first documented disease progression (events of new bone lesions, soft tissue plasmacytomas or an increase in existing lesions, or death due to any cause). The median PFS was calculated from the product-limit (Kaplan-Meier) method for censored data. Due to the small number of events, the tail of the estimated survival distribution was close to the median for both arms. The higher variability of the tail estimates resulted in observing the median estimate in the experimental arm but not in the standard of care arm even when number of events in 2 arms were similar. The database cutoff date was July 9, 2018. (NCT02579863)
Timeframe: Up to approximately 30 months

InterventionMonths (Median)
Pembrolizumab + Lenalidomide + Dexamethasone19.6
Lenolidomide + DexamethasoneNA

[back to top]

Overall Response Rate (ORR) Evaluated According to the IMWG Response Criteria by CAC Blinded Central Review

ORR was based on participants who achieved at least a partial response (stringent complete response [sCR]+complete response [CR]+very good partial response [VGPR]+partial response [PR]) according to the IMWG. CR = negative immunofixation of serum and urine AND disappearance of any soft tissue plasmacytomas AND <5% plasmacytomas in the bone marrow; sCR=stringent complete response, CR as above PLUS normal serum free light-chain (FLC) assay ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence; VGPR = serum and urine M-component detectable by immunofixation but not on electrophoresis OR ≥ 90% reduction in serum M-component plus urine M-component <100 mg/24 hr; PR = ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by ≥90% or to <200 mg/24 hours. The data cutoff date was July 9, 2018. (NCT02579863)
Timeframe: Up to approximately 30 months

InterventionPercentage of participants (Number)
Pembrolizumab + Lenalidomide + Dexamethasone74.4
Lenolidomide + Dexamethasone68.8

[back to top]

Percentage of Participants With Adverse Events (AEs)

An AE is any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with the treatment. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a pharmaceutical product, whether or not considered related to the pharmaceutical product. Preexisting conditions which worsen during a study are also considered as adverse events. Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: From study start up to end of study (Up to a maximum of 69 months)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCL100.0
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCL100.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL100.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL97.4

[back to top]

Percentage of Participants With Best Response of CR or PR, Determined by the Investigator on the Basis of CT Scans Alone

BOR=CR/PR per CT per MLRC. Per MLRC, CR based on CT was defined as a complete radiologic response in lymph nodes and ELS with target nodes/nodal masses regressing to ≤ 1.5 cm in LDi and no ELS of disease organ enlargement regressing to normal; no new lesions; bone marrow normal by morphology, if indeterminate, IHC negative. PR per CT only was defined as partial remission in lymph nodes and ELS with ≥50% decrease in SPD of up to 6 target measurable lymph nodes and extranodal sites, absent/normal/regressed but with no increase in non-measured lesions, spleen regressing by ≥50% in length beyond normal it, no new sites of lesions. Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: Up to every 6 months until disease progression, unacceptable toxicity or study completion (up to approximately 69 months)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL50.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL90.0
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL79.5

[back to top]

Percentage of Participants With Complete Response (CR) at End of Induction (EOI), Determined by an Independent Review Committee (IRC) on the Basis of Positron Emission Tomography (PET) and Computed Tomography (CT) Scans

CR at EOI was assessed by IRC according to Modified Lugano Response Criteria (MLRC). Per MLRC, CR based on PET-CT was defined as complete metabolic response (MR) in lymph nodes and extralymphatic sites (ELS) with a score of 1, 2, or 3 with or without residual mass, on 5-point scale (5PS) where 1=no uptake above background; 2=uptake ≤ mediastinum; 3=uptake > mediastinum but ≤ liver; 4=uptake moderately > liver; 5=uptake markedly higher than liver and/or new lesions no evidence of fluorodeoxyglucose (FDG)-avid disease in bone marrow. Bone marrow is normal by morphology; if indeterminate, immunohistochemistry (IHC) negative. Analysis was done 6-8 weeks after Cycle 6, Day 1 (cycle=28 days). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL66.7
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL0.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL60.0
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL38.5

[back to top]

Percentage of Participants With CR at EOI, Determined by Investigator on the Basis of CT Scans Alone

CR at EOI was determined by Investigator according to the MLRC. Per MLRC, CR based on CT was defined as complete radiologic response in lymph nodes and ELS with target nodes/nodal masses regressing to ≤ 1.5 cm in LDi and no ELS of disease organ enlargement regressing to normal; no new lesions; normal bone marrow by morphology, if indeterminate, IHC negative. Analysis was done 6-8 weeks after Cycle 6, Day 1 (cycle=28 days). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL16.7
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL0.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL29.7
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL28.1

[back to top]

Percentage of Participants With CR at EOI, Determined by the Investigator on the Basis of PET-CT Scans

CR at EOI was assessed by Investigator according to MLRC. Per MLRC, CR based on PET-CT was defined as complete MR in lymph nodes and ELS with a score of 1, 2, or 3 with or without residual mass, on 5PS where 1=no uptake above background; 2=uptake ≤ mediastinum; 3=uptake > mediastinum but ≤ liver; 4=uptake moderately > liver; 5=uptake markedly higher than liver and/or new lesions no evidence of FDG-avid disease in bone marrow. Bone marrow is normal by morphology; if indeterminate, IHC negative. Analysis was done 6-8 weeks after Cycle 6, Day 1 (cycle=28 days). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL66.7
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL0.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL60.0
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL33.3

[back to top]

Percentage of Participants With CR at EOI, Determined by the IRC on the Basis of CT Scans Alone

CR at EOI was determined by IRC according to the MLRC. Per MLRC, CR based on CT was defined as complete radiologic response in lymph nodes and ELS with target nodes/nodal masses regressing to ≤ 1.5 cm in longest transverse diameter (LDi) and no ELS of disease organ enlargement regressing to normal; no new lesions; normal bone marrow by morphology, if indeterminate, IHC negative. Analysis was done 6-8 weeks after Cycle 6, Day 1 (cycle=28 days). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL16.7
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL0.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL31.4
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL12.5

[back to top]

Percentage of Participants With Dose-Limiting Toxicities (DLTs)

A DLT was defined as any one of the following toxicities occurring during the first cycle of treatment and assessed by the investigator as related to study treatment: Any adverse event of any grade that lead to a delay of > 14 days in the start of the next treatment cycle, Grade 3 or 4 non-hematologic adverse events with few exceptions; increase in hepatic transaminase > 3 x baseline and an increase in direct bilirubin > 2 x upper limits of normal (ULN), without any findings of cholestasis or jaundice, or signs of hepatic dysfunction, and in the absence of other contributory factors; hematologic adverse events that met a few protocol specified criteria. DLTs were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events, Version 4.0 (NCI CTCAE v4.0). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: Day 1 of Cycle 1 to Day 1 of Cycle 2 (1 cycle = 28 days) in dose-escalation phase

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FL0.0
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FL50.0
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FL0.0
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL0.0
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCL0.0
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCL0.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL0.0

[back to top]

Percentage of Participants With Objective Response (OR) at EOI, Determined by the IRC on the Basis of PET-CT Scans

OR was defined as percentage of participants with CR or PR as assessed by the IRC according to MLRC. Per MLRC CR based on PET-CT is complete MR in lymph nodes & ELS with score of 1, 2, or 3 with or without residual mass on 5PS, where 1=no uptake above background; 2=uptake ≤ mediastinum; 3=uptake> mediastinum but ≤ liver; 4=uptake moderately > liver; 5=uptake markedly higher than liver and/or new lesions;no new lesions & no evidence of FDG-avid disease in bone marrow.Bone marrow is normal by morphology; if indeterminate, IHC negative. Partial response (PR) based on PET-CT was defined as partial MR in lymph nodes & ELS with score of 4 or 5 with reduced uptake compared with baseline & residual masses of any size at interim, residual uptake higher than uptake in normal bone marrow but reduced compared with baseline (diffuse uptake compatible with reactive changes from chemotherapy allowed). Percentages have been rounded off up to the second decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (cycle=28 days) (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL10.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL72.50
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL46.20

[back to top]

Percentage of Participants With Objective Response at EOI, Determined by Investigator on the Basis of PET-CT Scans

OR was defined percentage of participants with CR or PR assessed by the investigator according to MLRC. Per MLRC CR based on PET-CT is complete MR in lymph nodes & ELS with score of 1, 2, 3 with or without residual mass on 5PS, where 1=no uptake above background 2=uptake ≤ mediastinum 3=uptake> mediastinum but ≤ liver 4=uptake moderately > liver 5=uptake markedly higher than liver and/or new lesions;no new lesions & no evidence of FDG-avid disease in bone marrow.Bone marrow is normal by morphology; if indeterminate, IHC negative. PR based on PET-CT was defined as partial MR in lymph nodes & ELS with score of 4 or 5 with reduced uptake compared with baseline & residual masses of any size at interim, residual uptake higher than uptake in normal bone marrow but reduced compared with baseline(diffuse uptake compatible with reactive changes from chemotherapy allowed). Percentages are rounded off. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL10.0
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL80.0
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL46.20

[back to top]

Percentage of Participants With Objective Response at EOI, Determined by the IRC on the Basis of CT Scans Alone

OR was defined as the percentage of participants with CR or PR, as assessed by the IRC based on MLRC. Per MLRC, CR based on CT was defined as complete radiologic response in lymph nodes and ELS with target nodes/nodal masses regressing to ≤ 1.5 cm in LDi and no ELS of disease organ enlargement regressing to normal; no new lesions; bone marrow normal by morphology, if indeterminate, IHC negative. PR per CT only was defined as partial remission in lymph nodes and ELS with ≥50% decrease in the sum of the products of greatest diameters (SPD) of up to 6 target measurable lymph nodes and extranodal sites, absent/normal/regressed but with no increase in non-measured lesions, spleen regressing by ≥50% in length beyond normal it, no new sites of lesions. The analysis was done 6-8 weeks after Cycle 6, Day 1 (each cycle is 28 days). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL12.5
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL91.4
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL53.1

[back to top]

Percentage of Participants With Objective Response, Determined by the Investigator on the Basis of CT Scans Alone

OR was defined as the percentage of participants with CR or PR, as assessed by the investigator based on MLRC. Per MLRC, CR based on CT was defined as complete radiologic response in lymph nodes and ELS with target nodes/nodal masses regressing to ≤ 1.5 cm in LDi and no ELS of disease organ enlargement regressing to normal; no new lesions; bone marrow normal by morphology, if indeterminate, IHC negative. PR per CT only was defined as partial remission in lymph nodes and ELS with ≥50% decrease in the sum of the products of greatest diameters (SPD) of up to 6 target measurable lymph nodes and extranodal sites, absent/normal/regressed but with no increase in non-measured lesions, spleen regressing by ≥50% in length beyond normal it, no new sites of lesions. Analysis was done 6-8 weeks after Cycle 6, Day 1 (cycle=28 days). Percentages have been rounded off to the first decimal point. (NCT02600897)
Timeframe: 6 to 8 weeks after Day 1 of Cycle 6 (up to approximately 28 weeks)

Interventionpercentage of participants (Number)
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL100.0
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL11.1
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL89.2
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL59.4

[back to top]

Number of Participants With Anti-therapeutic Antibodies (ATAs) to Polatuzumab Vedotin

The number of participants with positive results for ATAs, also called ADAs against polatuzumab vedotin at baseline and at any of the post-baseline assessment time-points were reported. Number of participants positive for Treatment Emergent ADA = the number of post-baseline evaluable participants determined to have treatment induced ADA or treatment-enhanced ADA during the study period. Treatment-induced ADA = negative or missing baseline ADA result(s) and at least one positive post-baseline ADA result. Treatment-enhanced ADA = a participant with positive ADA result at baseline who has one or more post-baseline titer results that are at least 0.60 t.u. greater than the baseline titer result. (NCT02600897)
Timeframe: Baseline up to approx. 2 years after the last dose of polatuzumab vedotin (up to approximately 30 months)

,,,,,,,,
InterventionParticipants (Count of Participants)
Baseline prevalence of ADAsPost baseline incidence of ADAs
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCL00
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCL00
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL00
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL10
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL10

[back to top]

Number of Participants With Human Anti-chimeric Antibodies (HACAs) to Rituximab

The number of participants with positive results for HACAs, also called ADAs against rituximab at baseline and at any of the post-baseline assessment time-points were reported. Number of participants positive for Treatment Emergent ADA = the number of post-baseline evaluable participants determined to have treatment induced ADA or treatment-enhanced ADA during the study period. Treatment-induced ADA = negative or missing baseline ADA result(s) and at least one positive post-baseline ADA result. Treatment-enhanced ADA = a participant with positive ADA result at baseline who has one or more post-baseline titer results that are at least 0.60 t.u. greater than the baseline titer result. (NCT02600897)
Timeframe: Baseline up to approximately 2 years after last dose (up to approximately 69 months)

,,,
InterventionParticipants (Count of Participants)
Baseline prevalence of ADAsPost baseline incidence of ADAs
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCL00
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCL00
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL00
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCL00

[back to top]

Number of Participants With Human Anti-human Antibodies (HAHAs) to Obinutuzumab

The number of participants with positive results for HAHAs, also called anti-drug antibodies (ADAs) against obinutuzumab at baseline and at any of the post-baseline assessment time-points were reported. Number of participants positive for Treatment Emergent ADA = the number of post-baseline evaluable participants determined to have treatment induced ADA or treatment-enhanced ADA during the study period. Treatment-induced ADA = negative or missing baseline ADA result(s) and at least one positive post-baseline ADA result. Treatment-enhanced ADA = a participant with positive ADA result at baseline who has one or more post-baseline titer results that are at least 0.60 titer unit (t.u.) greater than the baseline titer result. (NCT02600897)
Timeframe: Baseline up to approximately 2 years after last dose (up to approximately 69 months)

,,,,
InterventionParticipants (Count of Participants)
Baseline prevalence of ADAsPost baseline incidence of ADAs
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL00
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FL00
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FL30

[back to top]

Observed Plasma Lenalidomide Concentration

(NCT02600897)
Timeframe: Day 1 Cycle 1: predose and 2 hours (hr) post-dose; Day 15 Cycle 1: predose, 0.5hr, 1hr, 2hr, 4hr, 8hr post-dose; Day 1 Cycle 6: 2hr post-dose; unscheduled visits: 2hr post-dose (1 cycle = 28 days) (up to approximately 69 months)

Interventionng/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 2hInduction Cycle 1 Day 15 / PredoseInduction Cycle 1 Day 15 / 30 minInduction Cycle 1 Day 15 / 1hInduction Cycle 1 Day 15 / 2hInduction Cycle 1 Day 15 / 4hInduction Cycle 1 Day 15 / 8hInduction Cycle 6 Day 1 / 2hUnscheduled / 2h
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA3069.99124236305214103237199

[back to top]

Observed Plasma Lenalidomide Concentration

(NCT02600897)
Timeframe: Day 1 Cycle 1: predose and 2 hours (hr) post-dose; Day 15 Cycle 1: predose, 0.5hr, 1hr, 2hr, 4hr, 8hr post-dose; Day 1 Cycle 6: 2hr post-dose; unscheduled visits: 2hr post-dose (1 cycle = 28 days) (up to approximately 69 months)

,,,,,,,
Interventionng/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 2hInduction Cycle 1 Day 15 / PredoseInduction Cycle 1 Day 15 / 30 minInduction Cycle 1 Day 15 / 1hInduction Cycle 1 Day 15 / 2hInduction Cycle 1 Day 15 / 4hInduction Cycle 1 Day 15 / 8hInduction Cycle 6 Day 1 / 2h
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FLNA2018.7417918920211656.8110
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA3055.2020227220015249.8227
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FLNA1440.7291.9540.893.365.729.276.1
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCLNA25.25.4312.569.911879.944.036.2
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCLNA2372.6441.722423213457.0258
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA1978.3373.3187328245105255
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FLNA1185.9764.061.411796.435.0124
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA27710.494.5245242205103153

[back to top]

Observed Serum Obinutuzumab Concentration

1 cycle = 28 days (NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4 & 6: predose & 30 mins postdose; EOI: predose; Day 1 of Maintenance Months 1,7, 13, 19;Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseInduction Cycle 6 Day 1 / 30 minsEOI / PredoseMaintenance Month 1 / PredoseMaintenance Month 7 / PredoseDay 120 Post Last Dose
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FLNA39445183035410325573010823112829.1

[back to top]

Observed Serum Obinutuzumab Concentration

1 cycle = 28 days (NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4 & 6: predose & 30 mins postdose; EOI: predose; Day 1 of Maintenance Months 1,7, 13, 19;Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseInduction Cycle 6 Day 1 / 30 minsMaintenance Month 7 / PredoseMaintenance Month 13 / PredoseMaintenance Month 19 / Predose1 Year Post Last Dose
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FLNA3513866953446533271.182292692040.377

[back to top]

Observed Serum Obinutuzumab Concentration

1 cycle = 28 days (NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4 & 6: predose & 30 mins postdose; EOI: predose; Day 1 of Maintenance Months 1,7, 13, 19;Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

,
Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseInduction Cycle 6 Day 1 / 30 minsMaintenance Month 1 / PredoseMaintenance Month 7 / PredoseMaintenance Month 13 / PredoseMaintenance Month 19 / PredoseStudy Drug Discontinuation
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA33348166740574238475123012513415417.3
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FLNA35837274932164450480438121214235446.4

[back to top]

Observed Serum Obinutuzumab Concentration

1 cycle = 28 days (NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4 & 6: predose & 30 mins postdose; EOI: predose; Day 1 of Maintenance Months 1,7, 13, 19;Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseInduction Cycle 6 Day 1 / 30 minsMaintenance Month 1 / PredoseMaintenance Month 7 / PredoseMaintenance Month 13 / PredoseMaintenance Month 19 / PredoseStudy Drug DiscontinuationDay 120 Post Last Dose1 Year Post Last DoseUnscheduled / Predose
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA18231258827054725554317613515016587.544.50.340561

[back to top]

Observed Serum Rituximab Concentration

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4, 6: predose and 30 mins post-dose (1 cycle = 28 days) (up to approximately 69 months)

,,,
Interventionμg/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseInduction Cycle 6 Day 1 / 30 mins
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCLNA15125.613320.415915.3135
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCLNA20333.317274.622479.5250
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA17531.722253.122074.7233
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA17426.419458.322868.9256

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1; Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

,,
Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseStudy Drug Discontinuation
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FLNA58050.217.34.965555.3432.97.700.180
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA43227.76.732.502957.685319.700.180
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FLNA66080.024.76.5562311.541619.90.635

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1; Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseUnscheduled / Predose
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA52256.616.95.3245111.364511.89.78

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1; Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseStudy Drug DiscontinuationUnscheduled / Predose
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA33311.95.951.884818.994929.821.7123.1

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Antibody-conjugated MMAE (acMMAE)

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1; Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

,,,
Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / Predose
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FLNA47611.42.500.9615088.475199.37
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCLNA56858.922.37.445374.193713.06
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCLNA10652.018.35.585688.2550711.5
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA51343.918.66.1271611.773715.8

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1 and Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

,,
Interventionng/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseStudy Drug Discontinuation
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FLNA0.2281.150.4570.02800.1510.04310.08170.03670.0180
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FLNA0.1602.050.4590.03150.1380.1170.1560.09290.0180
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA0.2331.640.4350.03340.1360.02830.07520.02670.0180

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1 and Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

Interventionng/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseUnscheduled / Predose
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA0.2972.560.8430.07130.2100.07760.2130.08520.0180

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1 and Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

Interventionng/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / PredoseStudy Drug DiscontinuationUnscheduled / Predose
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA0.3471.120.2940.02680.1020.03660.1020.03420.01800.180

[back to top]

Plasma Concentration of Polatuzumab Vedotin Analyte: Unconjugated MMAE

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose and 30 mins post-dose; Days 8 and 15 of Cycle 1 and Day 1 of Cycle 6: predose, study drug discontinuation; unscheduled visit: predose (1 cycle = 28 days) (up to approximately 69 months)

,,,
Interventionng/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 1 Day 1 / 30 minsInduction Cycle 1 Day 8Induction Cycle 1 Day 15Induction Cycle 2 Day 1 / PredoseInduction Cycle 2 Day 1 / 30 minsInduction Cycle 4 Day 1 / PredoseInduction Cycle 4 Day 1 / 30 minsInduction Cycle 6 Day 1 / Predose
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FLNA0.4202.400.3870.02440.1510.06720.1040.0821
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCLNA0.1561.890.4850.05010.1860.01800.01800.0180
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCLNA0.1671.360.4560.03100.1230.03440.1330.0517
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA0.2983.650.6830.05270.1570.07290.2080.0405

[back to top]

Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose (1 cycle = 28 days), Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

Interventionμg/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 2 Day 1 / PredoseInduction Cycle 4 Day 1 / Predose1 Year Post Last Dose
Dose-escalation Phase: 1.4 mg Pola + 15 mg L + 1000 mg G in FLNA0.2002.570.0250

[back to top]

Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose (1 cycle = 28 days), Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

,,
Interventionμg/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 2 Day 1 / PredoseInduction Cycle 4 Day 1 / PredoseStudy Drug Discontinuation
Dose-escalation Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA0.6222.120.0903
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 1000 mg G in FLNA2.014.450.170
Dose-escalation Phase: 1.8 mg Pola + 10 mg L + 375 mg R in DLBCLNA1.570.9001.50

[back to top]

Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose (1 cycle = 28 days), Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

,
Interventionμg/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 2 Day 1 / PredoseInduction Cycle 4 Day 1 / PredoseStudy Drug DiscontinuationDay 120 Post Last Dose
Dose-escalation Phase: 1.8 mg Pola + 15 mg L + 375 mg R in DLBCLNA1.612.966.160.208
Dose-escalation Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA1.473.103.250.107

[back to top]

Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose (1 cycle = 28 days), Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

,
Interventionμg/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 2 Day 1 / PredoseInduction Cycle 4 Day 1 / PredoseStudy Drug DiscontinuationDay 120 Post Last Dose1 Year Post Last Dose
Dose-escalation Phase: 1.4 mg Pola + 10 mg L + 1000 mg G in FLNA1.471.830.1060.06610.0250
Expansion Phase: 1.4 mg Pola + 20 mg L + 1000 mg G in FLNA0.3392.310.1750.03650.0357

[back to top]

Serum Concentration of Polatuzumab Vedotin Analyte: Total Antibody

(NCT02600897)
Timeframe: Day 1 of Cycles 1, 2, 4: predose (1 cycle = 28 days), Day 120 post last dose; one year post last dose; study drug discontinuation; unscheduled visit: predose (up to approximately 69 months)

Interventionμg/mL (Geometric Mean)
Induction Cycle 1 Day 1 / PredoseInduction Cycle 2 Day 1 / PredoseInduction Cycle 4 Day 1 / PredoseStudy Drug DiscontinuationDay 120 Post Last Dose1 Year Post Last DoseUnscheduled / Predose
Expansion Phase: 1.8 mg Pola + 20 mg L + 375 mg R in DLBCLNA1.353.420.4550.06660.02502.59

[back to top]

Disease Free Survival

(NCT02619682)
Timeframe: Median of 4.6 years from start of study therapy

InterventionParticipants (Count of Participants)
Post Autologous Transplant Maintenance11

[back to top]

Number of Participants With Adverse Events, Graded According to CTCAE Version 4.0

Adverse events Toxicity during the 24 months plus one month of post autologous transplant maintenance therapy (NCT02619682)
Timeframe: 2 years plus one month

InterventionParticipants (Count of Participants)
Post Autologous Transplant Maintenance30

[back to top]

Overall Survival

all MM patients who got post autologous transplant study therapy (NCT02619682)
Timeframe: Median 4.6 years from start of therapy

InterventionParticipants (Count of Participants)
Post Autologous Transplant Maintenance21

[back to top]

Number of Participants With Dose-limiting Toxicities (DLTs) During Cycle 2 of Study Treatment

Does limiting toxicity (DLT) is defined as any one of the following events occurring during Cycle 2 of treatment and assessed by the investigator as related to study treatment: - Adverse event of any grade that leads to a delay of more than 14 days at the start of the next treatment cycle; - Hematologic adverse events (neutropenia, thrombocytopenia); - Non-hematologic adverse event, except IRRs, diarrhea, nausea or vomiting (NCT02631577)
Timeframe: Day 1 - Day 28 of second cycle

InterventionNumber of Participants (Number)
Atezolizumab-G-lena 15mg0
Atezolizumab-G-lena 20mg0

[back to top]

Serum Concentration of Obinutuzumab (mcg/mL)

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; Maint M = Maintenance Month; TRTC = Study Drug Completion or Early Discontinuation; PK FU = Pharmacokinetics and Immunogenicity Follow-up; YR = Year (NCT02631577)
Timeframe: Baseline up to approximately 59 months

Interventionmcg/mL (Mean)
Ind C1 D1 - PredoseInd C1 D1 - 30 min. PostdoseInd C2 D1 - PredoseInd C2 D1 - 30 min. PostdoseInd C4 D1 - PredoseInd C4 D1 - 30 min. PostdoseInd C6 D1 - PredoseInd C6 D1 - 30 min. PostdoseMaint M1 - PredoseMaint M7 - PredoseMaint M13 - PredoseMaint M19 - PredoseTRTCOD120FUO1YFU
Atezolizumab-G-lena 20mg0.63437539275330165727265220111210411114846.257.9

[back to top]

Percentage of Participants Achieving CR at EOI, as Determined by the Investigator Using Modified Lugano 2014 Criteria

CR was evaluated through use of PET-CT scans, using the Modified Lugano 2014 criteria. Response was determined by the Investigator. (NCT02631577)
Timeframe: 6 months (up to CCOD of 23 October 2018)

InterventionPercentage of Participants (Number)
Atezolizumab-G-lena 20mg75

[back to top]

Number of Participants Positive for Human Anti-human Antibodies (HAHA) to Obinutuzumab

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; Maint M = Maintenance Month; TRTC = Study Drug Completion or Early Discontinuation; PK FU = Pharmacokinetics and Immunogenicity Follow-up; YR = Year. All baseline and post-baseline samples from participants were negative for HAHAs to obinutuzumab and the results are shown below. (NCT02631577)
Timeframe: Baseline up to approximately 59 months

,
InterventionParticipants (Count of Participants)
Baseline - NegativeInd C6 D1 - NegativeStudy drug completion or early discontinuation - NegativeOB, PK, IMMUNO FU 120D - NegativeOB, PK, IMMUNO FU 1YR - Negative
Atezolizumab-G-lena 15mg43012
Atezolizumab-G-lena 20mg342715114

[back to top]

Percentage of Participants Achieving CR at EOI, as Determined by the IRC and Investigator Using Lugano 2014 Criteria

CR was evaluated through use of CT scans, using the Lugano 2014 criteria. Response was determined by the IRC and by the Investigator. (NCT02631577)
Timeframe: 6 months (up to CCOD of 23 October 2018)

InterventionPercentage of Participants (Number)
Determined by the IRC with CT or MRIDetermined by Investigator with CT or MRI
Atezolizumab-G-lena 20mg31.350

[back to top]

Percentage of Participants With Adverse Events and Serious Adverse Events

An adverse event is any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with the treatment. An adverse event can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a pharmaceutical product, whether or not considered related to the pharmaceutical product. Preexisting conditions which worsen during a study are also considered as adverse events. (NCT02631577)
Timeframe: Baseline up to approximately 59 months

,
InterventionParticipants (Count of Participants)
Adverse EventsSerious Adverse Events
Atezolizumab-G-lena 15mg42
Atezolizumab-G-lena 20mg3416

[back to top]

Percentage of Participants With Best Response (CR or PR) During the Study as Determined by the Investigator on the Basis of CT Scans Alone

Best Response was evaluated through use of CT scans alone, using the Lugano 2014. Response was determined by the Investigator. (NCT02631577)
Timeframe: 30 months

InterventionPercentage (Number)
Best Response (CR,PR)CRPR
Atezolizumab-G-lena 20mg87.568.818.8

[back to top]

Percentage of Participants With Objective Response (CR or PR) at EOI as Determined by the IRC and Investigator on the Basis of CT Scans Alone

Objective response was evaluated through use of CT scans alone, using the Lugano 2014. Response was determined by the IRC and by the Investigator. (NCT02631577)
Timeframe: 6 months (up to CCOD of 23 October 2018)

InterventionPercentage of Participants (Number)
Determined by IRC, based on Lugano 2014 - CTDetermined by Inv., based on Lugano 2014 - CT
Atezolizumab-G-lena 20mg81.387.5

[back to top]

Percentage of Participants With Objective Response (CR or PR) at EOI as Determined by the IRC and Investigator on the Basis of PET-CT Scans

Objective response was evaluated through use of PET-CT scans, using the Lugano 2014 or modified Lugano 2014 criteria. Response was determined by the IRC and by the Investigator. (NCT02631577)
Timeframe: 6 months (up to CCOD of 23 October 2018)

InterventionPercentage of Participants (Number)
Determined by IRC, based on Lugano 2014 - PETDetermined by Inv., based on Lugano 2014 - PETDetermined by IRC, Modified Lugano 2014 - PET-CTDetermined by Inv, Modified Lugano 2014 - PET-CT
Atezolizumab-G-lena 20mg81.384.478.184.4

[back to top]

Number of Participants Positive for Anti-therapeutic Antibodies (ATAs) to Atezolizumab

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; Maint M = Maintenance Month; TRTC = Study Drug Completion or Early Discontinuation; PK FU = Pharmacokinetics and Immunogenicity Follow-up; YR = Year. All baseline and post-baseline samples were negative for ATAs to atezolizumab and the results are shown below. (NCT02631577)
Timeframe: Baseline up to approximately 59 months

,
InterventionParticipants (Count of Participants)
Ind C2 D1 - NegativeInd C2 D15 - NegativeInd C4 D1 - NegativeInd C6 D1 - NegativeMaint M1 - NegativeMaint M4 - NegativeMaint M7 - NegativeMaint M13 - NegativeMaint M19 - NegativeStudy drug completion or early discontinuation - NegativeATEZO, PK, IMMUNO FU 120D - NegativeATEZO, PK, IMMUNO FU 1YR - Negative
Atezolizumab-G-lena 15mg423333212212
Atezolizumab-G-lena 20mg31322928272521201215113

[back to top]

Serum Concentration of Atezolizumab (mcg/mL)

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; Maint M = Maintenance Month; TRTC = Study Drug Completion or Early Discontinuation; PK FU = Pharmacokinetics and Immunogenicity Follow-up; YR = Year (NCT02631577)
Timeframe: Baseline up to approximately 59 months

Interventionmicrograms per milliliter (mcg/mL) (Mean)
Ind C2 D1 - PredoseInd C2 D1 - 30 min. PostdoseInd C2 D15 - PredoseInd C4 D1 - PredoseInd C4 D1 - 30 min. PostdoseInd C6 D1 - PredoseMaint M1 - PredoseMaint M1 - Day 2, 30 min. PostdoseMaint M4 - PredoseMaint M7 - PredoseMaint M13 - PredoseMaint M19 - PredoseTRTCPK FU 120D
Atezolizumab-G-lena 20mg0.18427990.022647727917266629232230932711638.4

[back to top]

Serum Concentration of Lenalidomide (ng/mL)

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; HR = Hour (NCT02631577)
Timeframe: Baseline up to approximately 59 months

,
Interventionnanograms/milliliter (ng/mL) (Mean)
Ind C1 D15 - PredoseInd C1 D15 - 2.0 hr. PostdoseInd C2 D15 - PredoseInd C2 D15 - 30 min. PostdoseInd C2 D15 -1.0 hr. PostdoseInd C2 D15 - 2.0 hr. PostdoseInd C2 D15 - 4.0 hr. PostdoseInd C2 D15 - 8.0 hr. PostdoseInd C6 D15 - PredoseInd C6 D15 - 2.0 hr. Postdose
Atezolizumab-G-lena 15mg12.62543.5724122414695.845.95.72200
Atezolizumab-G-lena 20mg13.029415.029335426215068.49.17214

[back to top]

Serum Concentration of Obinutuzumab (mcg/mL)

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; Maint M = Maintenance Month; TRTC = Study Drug Completion or Early Discontinuation; PK FU = Pharmacokinetics and Immunogenicity Follow-up; YR = Year (NCT02631577)
Timeframe: Baseline up to approximately 59 months

Interventionmcg/mL (Mean)
Ind C1 D1 - 30 min. PostdoseInd C2 D1 - PredoseInd C2 D1 - 30 min. PostdoseInd C4 D1 - PredoseInd C4 D1 - 30 min. PostdoseInd C6 D1 - PredoseInd C6 D1 - 30 min. PostdoseMaint M1 - PredoseMaint M7 - PredoseMaint M13 - PredoseMaint M19 - PredoseOD120FUO1YFU
Atezolizumab-G-lena 15mg36428860617550923050311466.177.994.023.346.9

[back to top]

Serum Concentration of Atezolizumab (mcg/mL)

The following abbreviations apply in the table: Ind C = Induction Cycle; D = Day; Maint M = Maintenance Month; TRTC = Study Drug Completion or Early Discontinuation; PK FU = Pharmacokinetics and Immunogenicity Follow-up; YR = Year (NCT02631577)
Timeframe: Baseline up to approximately 59 months

Interventionmicrograms per milliliter (mcg/mL) (Mean)
Ind C2 D1 - 30 min. PostdoseInd C2 D15 - PredoseInd C4 D1 - PredoseInd C4 D1 - 30 min. PostdoseInd C6 D1 - PredoseMaint M1 - PredoseMaint M1 - Day 2, 30 min. PostdoseMaint M4 - PredoseMaint M7 - PredoseMaint M13 - PredoseMaint M19 - PredosePK FU 120D
Atezolizumab-G-lena 15mg34573.812834019479.065317420920335146.0

[back to top]

Percentage of Participants Achieving Complete Response (CR) at End of Induction (EOI), as Determined by the Independent Review Committee (IRC) Using Modified Lugano 2014 Criteria

Complete response (CR) was evaluated through use of PET-CT scans, using the Modified Lugano 2014 criteria. Response was determined by the IRC. (NCT02631577)
Timeframe: 6 months (up to clinical cut-off date (CCOD) of 23 October 2018)

InterventionPercentage of Participants (Number)
Atezolizumab-G-lena 20mg71.9

[back to top]

Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Time to Maximum Observed Concentration (Tmax)

Time to maximum observed concentration of Durvalumab (DURVA) after multiple doses on day 1 obtained from the observed concentration versus time data (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

Interventionday (Median)
Cohort A: High Risk, TNE0.051
Cohort B: >=65 Years Old, TNE0.106
Cohort C: High Risk, Post-transplant0.180

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 15: Time to Maximum Observed Concentration (Tmax)

Time to maximum observed concentration of Lenalidomide (LEN) after multiple doses on day 15 obtained from the observed concentration versus time data (NCT02685826)
Timeframe: Cycle 1 Day 15: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionhour (Median)
Cohort A: High Risk, TNE2.000
Cohort B: >=65 Years Old, TNE1.000
Cohort C: High Risk, Post-transplant1.042

[back to top]

Participants With Treatment Emergent Adverse Events (TEAE)

An AE is any noxious, unintended, or untoward medical occurrence that may appear or worsen during the course of a study. A TEAE includes AEs between the first dose date of either study drug and 90 days after the last dose of study drug. A serious AE is any AE occurring at any dose that: • Results in death; • Is life-threatening; • Requires or prolongs existing inpatient hospitalization; • Results in persistent or significant disability/incapacity; • Is a congenital anomaly/birth defect; • Constitutes an important medical event. The Investigator assessed the relationship of each AE to study drug and graded the severity according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE, Version 4.03): - Grade 1 = Mild - Grade 2 = Moderate (some limitation in activity; no/minimal medical intervention required) - Grade 3 = Severe (limitation in activity; medical intervention required) - Grade 4 = Life-threatening - Grade 5 = Death (NCT02685826)
Timeframe: Day 1 up to Week 84 (the longer of 90 days after discontinuing treatment with DURVA, or 28 days after the last dose of LEN or dex)

,,
InterventionParticipants (Count of Participants)
>= 1 TEAE>=1 treatment-related TEAE>=1 related to DURVA>=1 related to LEN>=1 related to dex>=1 TEAE of severity grade 3 or 4>=1 severity 3/4, related to study drug>=1 severity 3/4, related to DURVA>=1 severity 3/4, related to LEN>=1 severity 3/4, related to dex>=1 Grade 5 TEAE (Death)>=1 Grade 5 TEAE related to study drug>=1 Grade 5 TEAE related to DURVA>=1 Grade 5 TEAE related to LEN>=1 Grade 5 TEAE related to dex>=1 Serious TEAE>=1 Serious TEAE related to study drug>=1 Serious TEAE related to DURVA>=1 Serious TEAE related to LEN>=1 Serious TEAE related to dex>=1 TEAE leading to discontinuation of study drug>= TEAE leading to discontinuation of DURVA>= TEAE leading to discontinuation of LEN>= TEAE leading to discontinuation of dex>=1 leading to delay/reduction/interruption drug>=1 leading to dose delay of DURVA>=1 leading to infusion interruption of DURVA>=1 leading to dose reduction of LEN>=1 leading to interruption of LEN>=1 leading to dose reduction of dex>=1 leading to interruption of dex
Cohort A: High Risk, TNE2422162216191371151000012856532321410021319
Cohort B: >=65 Years Old, TNE9848688482000006413100007302533
Cohort C: High Risk, Post-transplant18171515NA7644NA1110NA4442NA110NA64135NANA

[back to top]

Time to Response (for Cohorts A and B)

Time to response (for responders only, per IMWG Uniform Response Criteria) is calculated as the time from the first date of dosing of study medication to the first date of documented response (PR or better). (NCT02685826)
Timeframe: Day 1 of each cycle starting with Cycle 2 up to Cycle 17 plus one week for the end of treatment visit (Day 29 up to Week 73)

Interventionweeks (Median)
Cohort A: High Risk, TNE4.20
Cohort B: >=65 Years Old, TNE4.10

[back to top]

Response Improvement Rate (RIR) for Cohort C: Percentage of Participants Achieving a Response Improved From Cycle 1 Day 1 as Assessed by the Investigators Using the International Myeloma Working Group (IMWG) Uniform Response Criteria

Response Improvement Rate is defined as the percentage of participants who achieved a response from treatment as compared to the pre-autologous stem cell transplantation [ASCT] diseases measurement used as baseline for response assessment. IMWG response categories could be stable disease (SD), partial response (PR), very good partial response (VGPR), complete response (CR), or stringent complete response (sCR), as long as it represented an improvement compared to prior to transplant. (NCT02685826)
Timeframe: Baseline (Cycle 1 Day 1); Treatment: Day 1 of each cycle starting with Cycle 2 up to Cycle 15 plus one week for the end of treatment visit (Day 29 up to Week 61)

Interventionpercentage of participants (Number)
Cohort C: High Risk, Post-transplant0

[back to top]

Participants With Dose-Limiting Toxicities (DLTs) During the Dose-Determining Timeframe (Day 1 - Day 28)

A Dose Review Team (DRT) evaluated DLTs to determine the recommended dose (RD) of Durvalumab to use in the Expansion Period. A DLT was defined as: a. Grade 4 neutropenia for >= 5 days. b. Grade 3 neutropenia associated with fever (≥ 38.5°C / 101.3°F) of any duration. c. Grade 4 thrombocytopenia or Grade 3 thrombocytopenia with bleeding, or platelets transfusion. d. Grade 4 hematologic toxicity that does not resolve to baseline level <=72 hours. e. Grade 4 anemia, unexplained by underlying disease. f. Any nonhematologic toxicity Grade ≥ 3 except for alopecia and nausea. g. Treatment interruption >= 2 weeks due to AE. If ≤ 1 of the 6 initial participants in each cohort experience a DLT during cycle 1, the RD was Durvalumab 1500 mg; If >=2 of the 6 initial participants in any cohort experience a DLT during cycle 1, the maximum tolerated dose (MTD) was exceeded and the dose level of Durvalumab was de-escalated to 750 mg (NCT02685826)
Timeframe: First treatment cycle: Day 1 to Day 28

InterventionParticipants (Count of Participants)
Cohort A: High Risk, TNE1
Cohort B: >=65 Years Old, TNE1
Cohort C: High Risk, Post-transplant0

[back to top]

Participants Who Had Either Disease Progression or Death

This outcome was originally defined as a Kaplan-Meier estimate of progression-free survival (PFS) which estimated the time between first date of dosing of study medication and disease progression, as determined by the investigator using the IMWG Uniform Response Criteria, or death during study treatment, whichever occurred earlier. However due to the early study termination and limited follow-up time, the majority of participants were censored for PFS analysis. Data reported instead represent the number of participants who died during study treatment or had disease progression within 90 days of the last dose of durvalumab. (NCT02685826)
Timeframe: Day 1 up to Week 84

InterventionParticipants (Count of Participants)
Cohort A: High Risk, TNE4
Cohort B: >=65 Years Old, TNE1
Cohort C: High Risk, Post-transplant3

[back to top]

Participants Who Died Up To Data Cut-off Date (15 December 2017)

This outcome was originally defined as a Kaplan-Meier estimate of overall survival (OS) and was defined as the time between first date of dosing of study medication and death due to any cause. However due to the early study termination and limited follow-up time, the majority of participants were censored for OS analysis. Data reported instead represent the number of participants who died due to any cause from Day 1 up to data cut-off. (NCT02685826)
Timeframe: Day 1 up to Week 87

InterventionParticipants (Count of Participants)
Cohort A: High Risk, TNE2
Cohort B: >=65 Years Old, TNE0
Cohort C: High Risk, Post-transplant1

[back to top]

Participants Who Developed Anti-drug Antibody Against Durvalumab

The number of participants who develop antidrug antibody against durvalumab at any of the sampling timepoints during the study. (NCT02685826)
Timeframe: Pre-dose samples on Day 1 of cycles 1, 2, 4, 6, 10, and 14 (study days 1, 29, 85, 141, 253, 393)

InterventionParticipants (Count of Participants)
Cohort A: High Risk, TNE0
Cohort B: >=65 Years Old, TNE1
Cohort C: High Risk, Post-transplant0

[back to top]

Overall Response Rate (ORR) for Cohorts A and B: Percentage of Participants Who Achieved a Partial Response or Better According to the International Myeloma Working Group (IMWG) Uniform Response Criteria

Tumor response, including progressive disease, was assessed by the investigators and captured the best assessment of response during the treatment period. ORR was defined as partial response (PR) or better which includes PR, very good partial response (VGPR), complete response (CR), or stringent complete response (sCR). A PR required ≥ 50% reduction of serum M-Protein and reduction in 24-hour urinary M-protein by ≥ 90% or to < 200 mg per 24 hours. If present at baseline, a ≥ 50% reduction in the size of soft tissue plasmacytomas was also required. sCR required - a negative immunofixation of serum and urine and - disappearance of any soft tissue plasmacytomas and - ≤ 5% plasma cells in bone marrow and normal free light-chain (FLC) ratio and - absence of clonal plasma cells by immunohistochemistry or 2- to 4-color flow cytometry. (NCT02685826)
Timeframe: Day 1 of each cycle starting with Cycle 2 up to Cycle 17 plus one week for the end of treatment visit (Day 29 up to Week 73)

Interventionpercentage of participants (Number)
Cohort A: High Risk, TNE66.7
Cohort B: >=65 Years Old, TNE50.0

[back to top]

Lenalidomide (LEN) Plasma PK Parameters in Cycle 1 Day 15: Area Under the Concentration-time Curve From Time Zero to the Last Measured Time Point (AUC0-last)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 15: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionhour*ng/mL (Geometric Mean)
Cohort A: High Risk, TNE1911.739
Cohort B: >=65 Years Old, TNE1754.349
Cohort C: High Risk, Post-transplant629.151

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 15: Maximum Observed Concentration (Cmax)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 15: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionng/mL (Geometric Mean)
Cohort A: High Risk, TNE409.764
Cohort B: >=65 Years Old, TNE452.850
Cohort C: High Risk, Post-transplant171.235

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Time to Maximum Observed Concentration (Tmax)

Time to maximum observed concentration of Lenalidomide (LEN) after multiple doses on day 1 obtained from the observed concentration versus time data (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionhour (Median)
Cohort A: High Risk, TNE1.050
Cohort B: >=65 Years Old, TNE1.925
Cohort C: High Risk, Post-transplant1.150

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Terminal Elimination Half-life (t1/2)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionhour (Geometric Mean)
Cohort A: High Risk, TNE3.477
Cohort B: >=65 Years Old, TNE3.051
Cohort C: High Risk, Post-transplant2.883

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Maximum Observed Concentration (Cmax)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionng/mL (Geometric Mean)
Cohort A: High Risk, TNE395.774
Cohort B: >=65 Years Old, TNE354.018
Cohort C: High Risk, Post-transplant161.372

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Area Under the Concentration-time Curve From Time Zero to the Last Measured Time Point (AUC0-last)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionhour*ng/mL (Geometric Mean)
Cohort A: High Risk, TNE1468.102
Cohort B: >=65 Years Old, TNE1442.549
Cohort C: High Risk, Post-transplant591.085

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Area Under the Concentration-time Curve From Time Zero to Infinity (AUC0-inf)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionhour*ng/mL (Geometric Mean)
Cohort A: High Risk, TNE2534.684
Cohort B: >=65 Years Old, TNE2011.889
Cohort C: High Risk, Post-transplant768.104

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Apparent Volume of Distribution (Vz/F)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

Interventionliters (Geometric Mean)
Cohort A: High Risk, TNE44.329
Cohort B: >=65 Years Old, TNE54.689
Cohort C: High Risk, Post-transplant54.157

[back to top]

Durvalumab (DURVA) Serum Pharmacokinetic (PK) Parameters in Cycle 1: Area Under the Concentration-time Curve From Time Zero to the Last Measured Time Point (AUC0-last)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

Interventionday*µg/L (Geometric Mean)
Cohort A: High Risk, TNE3535033.014
Cohort B: >=65 Years Old, TNE3582905.960
Cohort C: High Risk, Post-transplant4026520.655

[back to top]

Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Area Under the Concentration-time Curve From Time Zero to Infinity (AUC0-inf)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

Interventionday*µg/L (Geometric Mean)
Cohort A: High Risk, TNE4944601.671
Cohort B: >=65 Years Old, TNE5532568.144
Cohort C: High Risk, Post-transplant6531541.670

[back to top]

Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Clearance (CL)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

InterventionL/day (Geometric Mean)
Cohort A: High Risk, TNE0.303
Cohort B: >=65 Years Old, TNE0.271
Cohort C: High Risk, Post-transplant0.230

[back to top]

Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Maximum Observed Concentration (Cmax)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

Interventionµg/L (Geometric Mean)
Cohort A: High Risk, TNE449280.231
Cohort B: >=65 Years Old, TNE452827.419
Cohort C: High Risk, Post-transplant482602.748

[back to top]

Lenalidomide (LEN) Plasma Pharmacokinetic (PK) Parameters in Cycle 1 Day 1: Apparent Clearance (CL/F)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: Cycle 1 Day 1: pre-dose, 0.5, 1, 2, 4, and 8 hours post LEN dose

InterventionL/hour (Geometric Mean)
Cohort A: High Risk, TNE8.838
Cohort B: >=65 Years Old, TNE12.426
Cohort C: High Risk, Post-transplant13.019

[back to top]

Kaplan-Meier Estimates for Duration of Response (for Cohort A and B)

Duration of response (for responders only) was defined as the time from earliest date of documented response (PR or better) to the earliest date of disease progression (DP) as determined by the investigator per IMWG Uniform Response criteria or death during study treatment, whichever occurred first. (NCT02685826)
Timeframe: Day 1 of each cycle starting with Cycle 2 up to Cycle 17 plus one week for the end of treatment visit (Day 29 up to Week 73)

Interventionmonths (Median)
Cohort A: High Risk, TNE10.3
Cohort B: >=65 Years Old, TNENA

[back to top]

Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Volume of Distribution (Vz)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

Interventionliters (Geometric Mean)
Cohort A: High Risk, TNE4.244
Cohort B: >=65 Years Old, TNE4.582
Cohort C: High Risk, Post-transplant4.563

[back to top]

Durvalumab (DURVA) Serum PK Parameters in Cycle 1: Terminal Elimination Half-life (t1/2)

Geometric mean was obtained by computing the arithmetic mean of the logarithm-transformed values of concentration/PK parameters and then using the exponentiation to return the computation to the original scales. Geometric CV% was calculated as follows: CV% = 100*SQRT(EXP(σ2)-1), where σ2 denotes the variance of the log-transformed values. (NCT02685826)
Timeframe: pre-infusion (-60 to -5 minutes prior to dose), end of infusion (EOI), 4 hours, 168 hours (Day 8), 336 hours (Day 15) and 504 hours (Day 22) after administration of DURVA on Day 1

Interventionday (Geometric Mean)
Cohort A: High Risk, TNE10.984
Cohort B: >=65 Years Old, TNE13.376
Cohort C: High Risk, Post-transplant15.844

[back to top]

Percentage of Participants With Progression-Free Survival in Pairwise Analysis

This is the pairwise comparison for percentage of participants with Progression-Free Survival. Death or disease progression will be considered as events for this endpoint. The time to event will be calculated as time from randomization to disease progression, death, initiation of non-protocol anti-myeloma therapy, loss to follow-up or the end of the study, whichever comes first. The Kaplan-Meier estimator will be constructed for each treatment arm. Progression-free survival was compared between the vaccine arm, lenalidomide/GM-CSF arm and lenalidomide alone arm. (NCT02728102)
Timeframe: 2 year

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF79.3
Lenalidomide and GM-CSF91.3
Maintenance Lenalidomide84.9

[back to top]

Number of Grade ≥ 3 Toxicities

Toxicities are evaluated using NCI CTCAE version 4.0 at pre-maintenance initiation and during maintenance therapy monthly for the first 4 cycles and then at cycles 6, 9, 15, 21, 24, which correspond to Day 1, 29, 57, 85, 141, 225, 393, 561, and 645 post maintenance initiation. All Grade ≥ 3 toxicities will be tabulated for treatment arms. Toxicities are categorized by organ system according to the CTCAE. Toxicities that involve multiple questions per organ system are combined in one category. (NCT02728102)
Timeframe: 2 years

,,
InterventionToxicities (Number)
Auditory DisordersBlood and Lymphatic DisordersCardiovascular DisordersGI DisordersGeneral DisordersHepatobiliary/Pancreas DisordersImmune System DisordersInvestigationsMetabolism and Nutrition DisordersMusculoskeletal and Connective Tissue DisordersNervous System DisordersRenal DisordersRespiratory, Thoracic and Mediastinal DisordersSkin and Subcutaneous Tissue DisordersVascular DisordersAbnormal Liver Symptoms
Lenalidomide and GM-CSF040342100115015111
Lenalidomide, Vaccine, and GM-CSF1814155422711415460
Maintenance Lenalidomide03822210132122730

[back to top]

Percentage of Participants With 1-year Response Rate of CR/sCR

The primary objective of this randomized trial is to compare the proportion of patients alive and in complete response (CR or sCR) at one year post transplant between patients receiving DC/myeloma vaccine/GM-CSF with lenalidomide maintenance therapy to those receiving lenalidomide maintenance therapy with or without GM-CSF. Complete Response (CR) is defined to require all the followings: Absence of the original monoclonal paraprotein in serum and urine by routine electrophoresis and by immunofixation; Less than 5% plasma cells in a bone marrow aspirate and also on trephine bone biopsy, if biopsy is performed; No increase in size or number of lytic bone lesions on radiological investigations; Disappearance of soft tissue plasmacytomas. Stringent Complete Response (sCR) is defined to require all the followings in addition to CR: Normal free light chain ratio (FLC); Absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence. (NCT02728102)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF52.9
Lenalidomide With or Without GM-CSF50.0

[back to top]

Percentage of Participants With Grade 2 and 3 Infections

Grade 2 and 3 infections, as defined by the BMT CTN Technical MOP, are reported on the study. The cumulative incidence of infections post randomization, treating death as a competing risk, were compared between the vaccine and the combined non-vaccine groups using the Gray's test. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF26.6
Lenalidomide With or Without GM-CSF23.2

[back to top] [back to top]

Percentage of Participants With Myeloma Progression of Vaccine and Non-vaccine Arms

The event for this endpoint is defined as disease progression from CR/sCR or progressive disease for participants not in CR/sCR, or initiation of off protocol antimyeloma therapy. The cumulative incidence of myeloma progression will be compared between the vaccine arm and the combined non-vaccine arms using Gray's test and treating death (without documentation of disease progression) as a competing risk. Participants alive without disease progression at last observation will be censored at the date of last contact. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF20.7
Lenalidomide With or Without GM-CSF11.8

[back to top]

Participants With Grade ≥ 3 Toxicities

Toxicities are evaluated using NCI CTCAE version 4.0 at pre-maintenance initiation and during maintenance therapy monthly for the first 4 cycles and then at cycles 6, 9, 15, 21, 24, which correspond to Day 1, 29, 57, 85, 141, 225, 393, 561, and 645 post maintenance initiation. The number of participants experiencing Grade ≥ 3 toxicity are displayed for the vaccine and non-vaccine arms separately. The proportion of participants experiencing Grade ≥ 3 toxicity are compared between the vaccine and non-vaccine arms combined. (NCT02728102)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Lenalidomide, Vaccine, and GM-CSF53
Lenalidomide With or Without GM-CSF49

[back to top]

Participants Response to Treatment

A participant's disease status is evaluated based on the International Uniform Response Criteria per protocol. Before disease progression (PD), all disease classifications including stringent complete response (sCR), complete response (CR), very good partial remission (VGPR), partial response (PR), stable disease (SD) are relative to participant's disease status at study entry. Disease status is 'Not Evaluable' when disease assessment is not required, or disease status is missing. (NCT02728102)
Timeframe: 6 months, 1 year, and 2 years post-transplant and at Cycles 3(Day 57), 6(Day 141), 9(Day 225), 12(Day 309), 15 (Day 393), 18(Day 477), 21(Day 561) and 24(Day 654) of maintenance therapy

InterventionParticipants (Count of Participants)
Disease Status at 6 Months72284442Disease Status at 6 Months72284444Disease Status at 6 Months72284445Disease Status at 12 Months72284444Disease Status at 12 Months72284442Disease Status at 12 Months72284445Disease Status at 24 Months72284442Disease Status at 24 Months72284444Disease Status at 24 Months72284445Disease Status at Cycle 3 (Day 57 Assessment)72284442Disease Status at Cycle 3 (Day 57 Assessment)72284445Disease Status at Cycle 3 (Day 57 Assessment)72284444Disease Status at Cycle 6 (Day 141 Assessment)72284445Disease Status at Cycle 6 (Day 141 Assessment)72284444Disease Status at Cycle 6 (Day 141 Assessment)72284442Disease Status at Cycle 9 (Day 225 Assessment)72284442Disease Status at Cycle 9 (Day 225 Assessment)72284445Disease Status at Cycle 9 (Day 225 Assessment)72284444Disease Status at Cycle 12 (Day 309 Assessment)72284442Disease Status at Cycle 12 (Day 309 Assessment)72284444Disease Status at Cycle 12 (Day 309 Assessment)72284445Disease Status at Cycle 15 (Day 393 Assessment)72284444Disease Status at Cycle 15 (Day 393 Assessment)72284442Disease Status at Cycle 15 (Day 393 Assessment)72284445Disease Status at Cycle 18 (Day 477 Assessment)72284445Disease Status at Cycle 18 (Day 477 Assessment)72284442Disease Status at Cycle 18 (Day 477 Assessment)72284444Disease Status at Cycle 21 (Day 561 Assessment)72284445Disease Status at Cycle 21 (Day 561 Assessment)72284444Disease Status at Cycle 21 (Day 561 Assessment)72284442Disease Status at Cycle 24 (Day 654 Assessment)72284445Disease Status at Cycle 24 (Day 654 Assessment)72284444Disease Status at Cycle 24 (Day 654 Assessment)72284442
Stringent Complete Response (sCR)Complete Response (CR)Very Good Partial Remission (VGPR)Stable Disease (SD)Not EvaluablePartial Response (PR)Progression (PD)Dead
Maintenance Lenalidomide8
Lenalidomide and GM-CSF12
Lenalidomide, Vaccine, and GM-CSF22
Maintenance Lenalidomide9
Maintenance Lenalidomide3
Maintenance Lenalidomide2
Maintenance Lenalidomide0
Maintenance Lenalidomide1
Maintenance Lenalidomide10
Lenalidomide, Vaccine, and GM-CSF8
Lenalidomide and GM-CSF9
Lenalidomide, Vaccine, and GM-CSF19
Lenalidomide and GM-CSF5
Maintenance Lenalidomide14
Lenalidomide, Vaccine, and GM-CSF35
Lenalidomide and GM-CSF15
Lenalidomide, Vaccine, and GM-CSF5
Lenalidomide and GM-CSF6
Lenalidomide, Vaccine, and GM-CSF13
Lenalidomide, Vaccine, and GM-CSF18
Lenalidomide, Vaccine, and GM-CSF28
Lenalidomide and GM-CSF14
Lenalidomide, Vaccine, and GM-CSF1
Lenalidomide, Vaccine, and GM-CSF16
Lenalidomide and GM-CSF8
Lenalidomide, Vaccine, and GM-CSF17
Lenalidomide and GM-CSF7
Maintenance Lenalidomide15
Lenalidomide, Vaccine, and GM-CSF24
Lenalidomide and GM-CSF13
Lenalidomide, Vaccine, and GM-CSF6
Lenalidomide and GM-CSF0
Lenalidomide, Vaccine, and GM-CSF2
Lenalidomide and GM-CSF2
Lenalidomide, Vaccine, and GM-CSF7
Lenalidomide and GM-CSF11
Lenalidomide, Vaccine, and GM-CSF4
Lenalidomide, Vaccine, and GM-CSF3
Lenalidomide, Vaccine, and GM-CSF0
Maintenance Lenalidomide4
Maintenance Lenalidomide13
Lenalidomide, Vaccine, and GM-CSF9
Maintenance Lenalidomide7
Lenalidomide, Vaccine, and GM-CSF20
Maintenance Lenalidomide11
Lenalidomide and GM-CSF17
Maintenance Lenalidomide6
Lenalidomide, Vaccine, and GM-CSF11
Lenalidomide, Vaccine, and GM-CSF15
Lenalidomide, Vaccine, and GM-CSF14
Lenalidomide and GM-CSF16
Lenalidomide and GM-CSF3
Lenalidomide and GM-CSF1
Lenalidomide, Vaccine, and GM-CSF12
Lenalidomide and GM-CSF4

[back to top]

Number of Participants With Minimal Residual Disease (MRD)

Minimal residual disease (MRD) is defined as the presence of malignant plasma cells detected by multicolor flow cytometry among patients who are in complete remission. Multichannel flow cytometry will be used to establish MRD based on the presence of malignant plasma cells that are CD45 (-/dim), CD38+, CD138+, CD19-, CD56+ kappa or lambda restricted. The number of patients with MRD negative (MRD-) are described using frequencies at pre-randomization and 9th cycle post-randomization and compared between the vaccine arm with the no-vaccine arms combined. (NCT02728102)
Timeframe: Pre-randomization, Post-randomization at Cycle 9

,
InterventionParticipants (Count of Participants)
Pre-randomizationPost-randomization at Cycle 9
Lenalidomide With or Without GM-CSF3141
Lenalidomide, Vaccine, and GM-CSF3538

[back to top]

Number of Grade 2 and 3 Infections

Grade 2 and 3 infections, as defined by the BMT CTN Technical MOP, occurring after randomization will be reported. The incidence of definite and probable viral, fungal and bacterial infections will be tabulated for each patient. (NCT02728102)
Timeframe: 2years

,,
Interventioninfections (Number)
BacterialViralFungalOther
Lenalidomide and GM-CSF3600
Lenalidomide, Vaccine, and GM-CSF101401
Maintenance Lenalidomide61401

[back to top]

Percentage of Participants With Progression-Free Survival

Death or disease progression will be considered as events for this endpoint. The time to event will be calculated as time from randomization to disease progression, death, initiation of non-protocol anti-myeloma therapy, loss to follow-up or the end of the study, whichever comes first. The Kaplan-Meier estimator will be constructed for each treatment arm. Progression-free survival was compared between the vaccine and the combined non-vaccine arms using the log-rank test. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF79.3
Lenalidomide With or Without GM-CSF88.2

[back to top]

Percentage of Participants With Overall Survival in Pairwise Analysis

This is the pairwise comparison for percentage of participants with Overall Survival. Death from any cause is considered as events for this endpoint. The time to event is calculated as time from randomization to death, loss to follow-up or the end of the study, whichever comes first. Patients alive at the time of last observation are considered censored. The Kaplan-Meier estimator will be constructed for each treatment arm. Overall survival are compared between the vaccine arm, lenalidomide/GM-CSF arm and lenalidomide alone arm from time of randomization. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF97
Lenalidomide and GM-CSF100
Maintenance Lenalidomide96.9

[back to top]

Percentage of Participants With Overall Survival

Death from any cause is considered as events for this endpoint. The time to event is calculated as time from randomization to death, loss to follow-up or the end of the study, whichever comes first. Patients alive at the time of last observation are considered censored. The Kaplan-Meier estimator will be constructed for each treatment arm. Overall survival are compared between the vaccine and the combined non-vaccine arms from time of randomization. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF97
Lenalidomide With or Without GM-CSF98.5

[back to top]

Percentage of Participants With Myeloma Progression in Pairwise Analysis

This is the pairwise comparison for percentage of participants with Myeloma Progression. The event for this endpoint is defined as disease progression from CR/sCR or progressive disease for participants not in CR/sCR, or initiation of off protocol antimyeloma therapy. The cumulative incidence of myeloma progression will be compared between the vaccine arm, lenalidomide/GM-CSF arm and lenalidomide alone arm using Gray's test and treating death (without documentation of disease progression) as a competing risk. Participants alive without disease progression at last observation will be censored at the date of last contact. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF20.7
Lenalidomide and GM-CSF8.7
Maintenance Lenalidomide15.1

[back to top]

Percentage of Participants With Grade 2 and 3 Infections in Pairwise Analysis

This is the pairwise comparison for percentage of participants with Grade 2 and 3 infections. Grade 2 and 3 infections, as defined by the BMT CTN Technical MOP, are reported on the study. The cumulative incidence of infections post randomization, treating death as a competing risk, were compared between the vaccine arm, lenalidomide/GM-CSF arm and lenalidomide alone arm using the Gray's test. (NCT02728102)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Lenalidomide, Vaccine, and GM-CSF26.6
Lenalidomide and GM-CSF14.2
Maintenance Lenalidomide32.6

[back to top]

Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUClast) of Ibrutinib

(NCT02733042)
Timeframe: Cycle 1 Day 1 at predose and 1, 2, 4, and 24 hours post-dose

Interventionh*ng/mL (Geometric Least Squares Mean)
Arm B: Ibrutinib 420 mg586.396
Arm B: Ibrutinib 560 mg436.855

[back to top]

Area Under the Plasma Concentration-time Curve From Time Zero to Infinity (AUCinf) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

Interventiondays*μg/L (Geometric Mean)
Arm A: Durvalumab + Lenalidomide ± Rituximab4867431.378
Arm B: Durvalumab + Ibrutinib5818262.846
Arm C: Durvalumab + Bendamustine ± Rituximab4762968.345
Arm D: Durvalumab Monotherapy5593532.553

[back to top]

Time to First Response

Time to response was calculated as the time from first dose of study drug to the first response date (CR or PR for lymphoma participants and CR, CRi, nPR, PR, or PRL for CLL participants). (NCT02733042)
Timeframe: From first dose of any study drug to the end of follow-up, up to the data cutoff date of March 6, 2019; median (minimum, maximum) time on study was 16.7 (0.9, 32.9) months.

Interventionweeks (Median)
Part 1, Arm A: DUR 1500 mg + LEN 20 mg70.85
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²12.60
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²18.20
Part 1, Arm B: DUR 1500 mg + IBR 420 mg11.85
Part 1, Arm B: DUR 1500 mg + IBR 560 mg13.40
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²13.00
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²13.10
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mg12.10
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mg12.10
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²12.35
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²12.00
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²12.10
Part 2, Arm D HL: DUR 1500 mg13.10

[back to top]

Time to Maximum Plasma Concentration (Tmax) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

Interventiondays (Median)
Arm A: Durvalumab + Lenalidomide ± Rituximab0.0510
Arm B: Durvalumab + Ibrutinib0.0479
Arm C: Durvalumab + Bendamustine ± Rituximab0.0510
Arm D: Durvalumab Monotherapy0.0420

[back to top]

Volume of Distribution (Vz) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

Interventionliters (Geometric Least Squares Mean)
Arm A: Durvalumab + Lenalidomide ± Rituximab5.155
Arm B: Durvalumab + Ibrutinib6.451
Arm C: Durvalumab + Bendamustine ± Rituximab7.418
Arm D: Durvalumab Monotherapy5.957

[back to top]

Time to Maximum Observed Plasma Concentration (Tmax) of Lenalidomide

(NCT02733042)
Timeframe: Cycle 1 Day 1 at predose and 1, 2, 4, and 24 hours post-dose, and Cycle 1 Day 15 at pre-dose, 1, 2, and 4 hours post-dose.

,
Interventionhours (Median)
Cycle 1 Day 1Cycle 1 Day 15
Arm A: Lenalidomide 10 mg1.95003.0333
Arm A: Lenalidomide 20 mg1.16671.000

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Ibrutinib

(NCT02733042)
Timeframe: Cycle 1 Day 1 at predose and 1, 2, 4, and 24 hours post-dose, and Cycle 1 Day 15 at pre-dose, 1, 2, and 4 hours post-dose.

,
Interventionng/mL (Geometric Least Squares Mean)
Cycle 1 Day 1Cycle 1 Day 15
Arm B: Ibrutinib 420 mg129.70486.840
Arm B: Ibrutinib 560 mg67.72872.436

[back to top]

Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUClast) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

Interventiondays*μg/L (Geometric Mean)
Arm A: Durvalumab + Lenalidomide ± Rituximab3120149.759
Arm B: Durvalumab + Ibrutinib3225869.344
Arm C: Durvalumab + Bendamustine ± Rituximab2670168.397
Arm D: Durvalumab Monotherapy3053060.746

[back to top]

Kaplan-Meier Estimate of Progression-free Survival (PFS)

Progression-free survival was calculated as the time from first dose of study drug to the first documented progression or death (from any cause) during the entire efficacy evaluation period. For participants with no progression or death, PFS was censored at the last assessment date the participant was known to be progression-free. (NCT02733042)
Timeframe: From first dose of any study drug to the end of follow-up, up to the data cutoff date of March 6, 2019; median (minimum, maximum) time on study was 16.7 (0.9, 32.9) months.

Interventionmonths (Median)
Part 1, Arm A: DUR 1500 mg + LEN 20 mg8.41
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²NA
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²NA
Part 1, Arm B: DUR 1500 mg + IBR 420 mgNA
Part 1, Arm B: DUR 1500 mg + IBR 560 mg28.71
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²9.69
Part 1, Arm C: DUR 1500 mg + BEN 70 mg/m²1.25
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²3.82
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 90 mg/m²2.48
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mgNA
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mgNA
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²14.65
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²2.06
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²NA
Part 2, Arm D FL: DUR 1500 mg1.68
Part 2, Arm D DLBCL: DUR 1500 mg1.17
Part 2, Arm D CLL/SLL: DUR 1500 mg2.76
Part 2, Arm D MCL: DUR 1500 mg2.33
Part 2, Arm D HL: DUR 1500 mg2.66

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

Interventionμg/L (Geometric Mean)
Arm A: Durvalumab + Lenalidomide ± Rituximab420264.066
Arm B: Durvalumab + Ibrutinib361906.229
Arm C: Durvalumab + Bendamustine ± Rituximab331572.478
Arm D: Durvalumab Monotherapy392663.668

[back to top]

Kaplan-Meier Estimate of Duration of Response

Duration of response is defined for responders only as the time from the first documented response (CR or PR for lymphoma participants or CR, CRi, nPR, PR, or PRL for CLL participants) to disease progression or death (from any cause). For participants with response but no progression, or death, duration of response was censored at the last date that the participant was known to be progression-free. (NCT02733042)
Timeframe: From first dose of any study drug to the end of follow-up, up to the data cutoff date of March 6, 2019; median (minimum, maximum) time on study was 16.7 (0.9, 32.9) months.

Interventionweeks (Median)
Part 1, Arm A: DUR 1500 mg + LEN 20 mg10.14
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²NA
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²NA
Part 1, Arm B: DUR 1500 mg + IBR 420 mgNA
Part 1, Arm B: DUR 1500 mg + IBR 560 mgNA
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²29.29
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²NA
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mgNA
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mgNA
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²NA
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²24.14
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²NA
Part 2, Arm D HL: DUR 1500 mg11.14

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Lenalidomide

(NCT02733042)
Timeframe: Cycle 1 Day 1 at predose and 1, 2, 4, and 24 hours post-dose, and Cycle 1 Day 15 at pre-dose, 1, 2, and 4 hours post-dose.

,
Interventionng/mL (Geometric Least Squares Mean)
Cycle 1 Day 1Cycle 1 Day 15
Arm A: Lenalidomide 10 mg141.881107.635
Arm A: Lenalidomide 20 mg309.917174.090

[back to top]

Clearance (CL) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

InterventionL/day (Geometric Least Squares Mean)
Arm A: Durvalumab + Lenalidomide ± Rituximab0.3082
Arm B: Durvalumab + Ibrutinib0.2578
Arm C: Durvalumab + Bendamustine ± Rituximab0.3149
Arm D: Durvalumab Monotherapy0.2682

[back to top]

Number of Participants With Treatment-emergent Adverse Events

Treatment-emergent adverse events (TEAEs) are defined as adverse events (AEs) occurring or worsening on or after the first dose of any study treatment (durvalumab, lenalidomide, ibrutinib, bendamustine or rituximab) and within 90 days after last dose of durvalumab or 28 days after the last dose of other study drugs, whichever was later, as well as those serious adverse events made known to the investigator at any time thereafter that were suspected of being related to study treatment. The intensity of AEs was graded according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.03. For all other AEs not described in the CTCAE criteria, the intensity was assessed by the investigator as mild (Grade 1), moderate (Grade 2), severe (Grade 3), life-threatening (Grade 4), or death (Grade 5). (NCT02733042)
Timeframe: From first dose of any study drug to 90 days after last dose of durvalumab or 28 days after last dose of other study drugs, up to the data cut-off date of 6 March 2019. Maximum time on treatment was 55.4 weeks for DUR and 130 weeks for other study drugs.

,,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Any TEAETEAE Related to Any Study DrugCTCAE Grade 3-4 TEAECTCAE Grade 3-4 TEAE Related to Any Study DrugCTCAE Grade 5 TEAECTCAE Grade 5 TEAE Related to Any Study DrugSerious TEAESerious TEAE Related to Any Study DrugTEAE Leading to Discontinuation of Any Study DrugTEAE Leading to Dose Modifications of Study Drug
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²8877004333
Part 1, Arm A: DUR 1500 mg + LEN 20 mg3311001111
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²3333002203
Part 1, Arm B: DUR 1500 mg + IBR 420 mg3321002113
Part 1, Arm B: DUR 1500 mg + IBR 560 mg4431002004
Part 1, Arm C: DUR 1500 mg + BEN 70 mg/m²1010001001
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²3322002103
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²4432001004
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 90 mg/m²5542003104
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²9997005314
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mg101087006208
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mg109106117329
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²5553102123
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²101065005327
Part 2, Arm D CLL/SLL: DUR 1500 mg2010000000
Part 2, Arm D DLBCL: DUR 1500 mg9372407100
Part 2, Arm D FL: DUR 1500 mg5443104312
Part 2, Arm D HL: DUR 1500 mg5231002003
Part 2, Arm D MCL: DUR 1500 mg5341003013

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs) - Extended Collection

TEAEs defined as AEs occurring or worsening on or after first dose of any study treatment (durvalumab, lenalidomide, ibrutinib, bendamustine or rituximab) and within 90 days after last dose of durvalumab or 28 days after the last dose of other study drugs, whichever was later, as well as those serious adverse events made known to the investigator at any time thereafter that were suspected of being related to study treatment. Intensity of AEs graded according to the NCI CTCAE V. 4.03. For all other AEs not described in the CTCAE criteria, the intensity was assessed by investigator as mild (Grade 1), moderate (Grade 2), severe (Grade 3), life-threatening (Grade 4), or death (Grade 5). This outcome measure represents an updated version of the primary endpoint to include additional data collection that has occurred after the primary completion date (assessments made until August 21, 2022). (NCT02733042)
Timeframe: From first dose of any study drug to 90 days after last dose of durvalumab or 28 days after last dose of other study drugs, up to the study completion date of August 21, 2022 (up to approximately 75 months).

,,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Any TEAETEAE Related to Any Study DrugCTCAE Grade 3-4 TEAECTCAE Grade 3-4 TEAE Related to Any Study DrugCTCAE Grade 5 TEAECTCAE Grade 5 TEAE Related to Any Study DrugSerious TEAESerious TEAE Related to Any Study DrugTEAE Leading to Discontinuation of Any Study DrugTEAE Leading to Dose Modifications of Study Drug
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²8877004335
Part 1, Arm A: DUR 1500 mg + LEN 20 mg3311001111
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²3333002203
Part 1, Arm B: DUR 1500 mg + IBR 420 mg3321002113
Part 1, Arm B: DUR 1500 mg + IBR 560 mg4441003004
Part 1, Arm C: DUR 1500 mg + BEN 70 mg/m²1010001001
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²3322002103
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²4432001004
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 90 mg/m²5542003104
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²9997005314
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mg101098006309
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mg109106117329
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²5553102123
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²101065005327
Part 2, Arm D CLL/SLL: DUR 1500 mg2010000000
Part 2, Arm D DLBCL: DUR 1500 mg9372407100
Part 2, Arm D FL: DUR 1500 mg5443104312
Part 2, Arm D HL: DUR 1500 mg5231002003
Part 2, Arm D MCL: DUR 1500 mg5341003013

[back to top]

Time to Maximum Observed Plasma Concentration (Tmax) of Ibrutinib

(NCT02733042)
Timeframe: Cycle 1 Day 1 at predose and 1, 2, 4, and 24 hours post-dose, and Cycle 1 Day 15 at pre-dose, 1, 2, and 4 hours post-dose.

,
Interventionhours (Median)
Cycle 1 Day 1Cycle 1 Day 15
Arm B: Ibrutinib 420 mg2.0001.8833
Arm B: Ibrutinib 560 mg1.93332.000

[back to top]

Terminal Elimination Phase Half-Life (t½) of Durvalumab

(NCT02733042)
Timeframe: Cycle 1, Day 1 (pre-dose and at end of infusion), and 4, 24, 48, 168 (Day 8), 336 (Day 15), and 508 (Day 22) hours after the end of infusion.

Interventiondays (Geometric Least Squares Mean)
Arm A: Durvalumab + Lenalidomide ± Rituximab11.596
Arm B: Durvalumab + Ibrutinib17.344
Arm C: Durvalumab + Bendamustine ± Rituximab16.327
Arm D: Durvalumab Monotherapy15.399

[back to top]

Area Under the Plasma Concentration-time Curve From Time Zero to the Last Measurable Concentration (AUClast) of Lenalidomide

(NCT02733042)
Timeframe: Cycle 1 Day 1 at predose and 1, 2, 4, and 24 hours post-dose

Interventionh*ng/mL (Geometric Least Squares Mean)
Arm A: Lenalidomide 10 mg789.297
Arm A: Lenalidomide 20 mg805.299

[back to top]

Overall Response Rate (ORR) During Durvalumab Treatment

For lymphoma participants, response evaluation was based on International Working Group (IWG) response criteria for malignant lymphoma (the Lugano Classification). Overall response rate is defined as the percent of participants with best response of complete response (CR) or partial response (PR). For chronic lymphocytic leukemia participants, response evaluation was based on International Workshop on Chronic Lymphocytic Leukemia (IWCLL) guidelines for diagnosis and treatment of CLL. The ORR is defined as the percent of participants with best response of CR, complete response with incomplete marrow recovery (CRi), nodular partial response (nPR), PR, or partial response with lymphocytosis (PRL). (NCT02733042)
Timeframe: Up to 13 cycles (12 months)

Interventionpercentage of participants (Number)
Part 1, Arm A: DUR 1500 mg + LEN 20 mg33.3
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²66.7
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²80.0
Part 1, Arm B: DUR 1500 mg + IBR 420 mg66.7
Part 1, Arm B: DUR 1500 mg + IBR 560 mg75.0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²33.3
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²50.0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 90 mg/m²0
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mg88.9
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mg60.0
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²88.9
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²30.0
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²50.0
Part 2, Arm D FL: DUR 1500 mg0
Part 2, Arm D DLBCL: DUR 1500 mg0
Part 2, Arm D CLL/SLL: DUR 1500 mg0
Part 2, Arm D MCL: DUR 1500 mg0
Part 2, Arm D HL: DUR 1500 mg20.0

[back to top]

Part 1: Number of Participants With Dose Limiting Toxicities (DLTs)

Dose limiting toxicities were evaluated during the DLT evaluation period for participants in the dose finding cohorts. The severity grading was determined according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 4.03. A DLT is defined as below: Hematologic DLT • Grade 4 neutropenia observed for greater than 5 days duration • Grade 3 neutropenia associated with fever (≥ 38.5 °C) of any duration • Grade 4 thrombocytopenia or Grade 3 thrombocytopenia with bleeding, or any requirement for platelets transfusion • Grade 4 anemia, unexplained by underlying disease • Any other grade 4 hematologic toxicity that does not resolve to participant's pretreatment baseline level within 72 hours. Non-Hematologic DLT • Any non-hematological toxicity ≥ Grade 3 except for alopecia and nausea controlled by medical management • Any treatment interruption greater than 2 weeks due to adverse event. (NCT02733042)
Timeframe: Cycle 1 (28 days)

InterventionParticipants (Count of Participants)
Part 1, Arm A: DUR 1500 mg + LEN 20 mg0
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²3
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²1
Part 1, Arm B: DUR 1500 mg + IBR 420 mg0
Part 1, Arm B: DUR 1500 mg + IBR 560 mg0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 90 mg/m²1

[back to top]

Overall Response Rate During the Entire Study

For lymphoma participants, response evaluation was based on International Working Group (IWG) response criteria for malignant lymphoma (the Lugano Classification) (Cheson, 2014). Overall response rate is defined as the percent of participants with best response of complete response (CR) or partial response (PR). For chronic lymphocytic leukemia participants, response evaluation was based on International Workshop on Chronic Lymphocytic Leukemia (IWCLL) guidelines for diagnosis and treatment of CLL. The ORR is defined as the percentage of participants with best response of CR, complete response with incomplete marrow recovery (CRi), nodular partial response (nPR), PR, or partial response with lymphocytosis (PRL). (NCT02733042)
Timeframe: From first dose of any study drug to the end of follow-up, up to the data cutoff date of March 6, 2019; median (minimum, maximum) time on study was 16.7 (0.9, 32.9) months.

Interventionpercentage of participants (Number)
Part 1, Arm A: DUR 1500 mg + LEN 20 mg66.7
Part 1, Arm A: DUR 1500 mg + LEN 20 mg + RIT 375 mg/m²66.7
Part 1, Arm A: DUR 1500 mg + LEN 10 mg + RIT 375 mg/m²80.0
Part 1, Arm B: DUR 1500 mg + IBR 420 mg66.7
Part 1, Arm B: DUR 1500 mg + IBR 560 mg75.0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m²33.3
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²50.0
Part 1, Arm C: DUR 1500 mg + RIT 375 mg/m² + BEN 90 mg/m²0
Part 2, Arm B CLL/SLL: DUR 1500 mg + IBR 420 mg100.0
Part 2, Arm B MCL: DUR 1500 mg + IBR 560 mg70.0
Part 2, Arm C FL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²88.9
Part 2 Arm C DLBCL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²30.0
Part 2, Arm C CLL/SLL: DUR 1500 mg + RIT 375 mg/m² + BEN 70 mg/m²50.0
Part 2, Arm D FL: DUR 1500 mg0
Part 2, Arm D DLBCL: DUR 1500 mg0
Part 2, Arm D CLL/SLL: DUR 1500 mg0
Part 2, Arm D MCL: DUR 1500 mg0
Part 2, Arm D HL: DUR 1500 mg20.0

[back to top]

Maintenance Feasibility Rate (MFR)

defined as the percentage of patients starting induction treatment with ERd successfully completing treatment to end of maintenance (NCT02843074)
Timeframe: approximately 159 weeks (16 weeks of induction treatment, 6 weeks allowance for planning and scheduling mobilization and ASCT, 17 weeks pause in treatment after ASCT, 16 weeks of consolidation treatment, 104 weeks/2 years of maintenance treatment)

InterventionParticipants (Count of Participants)
ERd Therapy19

[back to top]

Number of Participants With Treatment-emergent Adverse Events as a Measure of Safety

Safety data and abnormal lab values and abnormal vital signs were collected and assessed using National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE V4.03). (NCT02843074)
Timeframe: weekly for 8 weeks, then every 2 weeks till the start of mobilization for Induction and 70-120 days post-ASCT, every 2 weeks during 16 weeks of Consolidation, and monthly for 24 months of Maintenance, up to 30 days after last dose of study drugs.

InterventionParticipants (Count of Participants)
ERd Therapy52

[back to top]

Complete Response Rate (CRR) for Complete Time on Study

Defined as the percentage of patients who achieve a complete response (CR) or near complete response (nCR) to treatment at each stage of the study (induction, ASCT, consolidation, end of study) per International Myeloma Working Group (IMWG) and European Group for Blood and Marrow Transplantation (EBMT) criteria.CR=bone marrow contains ≤5% plasma cells; negative immunofixation on serum and urine; disappearance of soft tissue plasmacytomas. nCR = the absence of myeloma protein on electrophoresis, with positive immunofixation, stable bone disease, and a normal serum calcium concentration. Patient must have completed at least 1 cycle to be included in analysis. (NCT02843074)
Timeframe: every 4 weeks until end of treatment visit, and every 3 months thereafter up to 3 years

InterventionParticipants (Count of Participants)
ERd Therapy25

[back to top]

Overall Survival (OS)

Defined as the time from start of induction treatment to 3 years post first study treatment or death from any cause, whichever comes first. Patient must have completed at least 1 cycle to be included in analysis. (NCT02843074)
Timeframe: every 4 weeks until end of treatment visit, and up to 3 years thereafter

Interventionmonths (Median)
ERd TherapyNA

[back to top]

Progression-free Survival (PFS)

The time from start of induction treatment to documented progressive disease (PD) or death from any cause up to 3 years post first study treatment. PD is defined as an increase of ≥ 25% from the nadir in at least one of the following criteria: serum M-protein (absolute increase must be ≥0.5 g/dL); urine M-protein (absolute increase must be ≥200 mg/24h) ; only in patients with non-measurable serum and urine M-protein levels: difference in involved and uninvolved FLC levels (absolute increase must be >10 mg/dL); only in patients with non-measurable serum and urine M-protein levels and non-measurable disease by FLC levels, bone marrow plasma cell % (absolute % must be ≥10%) OR Definite development of new bone lesions or soft tissue plasmacytomas OR definite increase in the size of existing bone lesions or soft tissue plasmacytomas OR Development of hypercalcemia (corrected serum Ca >11.5mg/dL) for patients without hypercalcemia at baseline. Must have completed 1 cycle to be included. (NCT02843074)
Timeframe: every 4 weeks until end of treatment visit, and every 3 months thereafter up to 3 years

Interventionmonths (Median)
ERd Therapy29.7002

[back to top]

Consolidation Feasibility Rate (CFR)

defined as the percentage of patients starting induction treatment with ERd successfully completing treatment to end of consolidation (NCT02843074)
Timeframe: approximately 55 weeks (16 weeks of induction treatment, 6 weeks allowance for planning and scheduling mobilization and ASCT, 17 weeks pause in treatment after ASCT, 16 weeks of consolidation treatment).

InterventionParticipants (Count of Participants)
ERd Therapy34

[back to top]

Induction Feasibility Rate (IFR)

Defined as the percentage of patients who successfully complete four 28-day cycles of induction therapy with elotuzumab, lenalidomide and dexamethasone (ERd) and start autologous stem cell transplantation (ASCT). (NCT02843074)
Timeframe: approximately 22 weeks (16 weeks of treatment and 6 weeks allowance for planning and scheduling mobilization and ASCT).

InterventionParticipants (Count of Participants)
ERd Therapy33

[back to top]

Overall Response Rate (ORR) for Complete Time on Study

Defined as percentage of patients receiving at least 1 cycle with confirmed complete response, very good partial response, or partial response (CR, VGPR, or PR) to treatment at each stage of the study (induction, ASCT, consolidation, end of study) per IMWG and EBMT criteria. CR=bone marrow contains ≤5% plasma cells; negative fixation on serum and urine; the disappearance of soft tissue plasmacytomas. VGPR=serum and urine M-protein detectable by immunofixation but not by electrophoresis or ≥90% reduction from baseline serum and urine M-protein level <100mg for 24h and In case of presence of soft tissue plasmacytoma(s) at baseline, the disappearance of any soft tissue plasmacytomas. PR=≥50% reduction from baseline in serum M-protein and ≥90% reduction from baseline in 24h urinary M-protein or urine M-protein <200 mg/24 hours and In case of presence of soft tissue plasmacytoma(s) at baseline, a reduction in the sum of products of the two longest perpendicular diameters (SPD) by >50%. (NCT02843074)
Timeframe: every 4 weeks until end of treatment visit, and every 3 months thereafter up to 3 years

InterventionParticipants (Count of Participants)
ERd Therapy47

[back to top]

Percentage of Participants With Overall Stringent Complete Response (sCR)

Overall sCR rate is defined as the percentage of participants who achieved sCR, according to the IMWG criteria. CR is defined as negative immunofixation on the serum and urine, and disappearance of any soft tissue plasmacytomas, and < 5 % PCs in bone marrow. sCR is defined as in addition to CR a normal FLC ratio, and absence of clonal PCs by immunohistochemistry or immunofluorescence or 2 to 4-color flow cytometry. (NCT02874742)
Timeframe: From randomization to end of following: induction treatment, ASCT, post-ASCT consolidation (after Cycle 6) and at the end of maintenance treatment of 24 months (overall duration up to 34 months)

,,
InterventionPercentage of participants (Number)
At the end of induction prior to ASCTAt the end of ASCT prior to consolidationAt the end of post-ASCT consolidationAt the end of Maintenance Treatment (up to 24 Months)
Randomized: Daratumumab+RVd (D-RVd)12.121.242.467.0
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)7.214.432.048.0
Safety Run-in: D-RVd043.856.393.8

[back to top]

Percentage of Participants With Overall Response Rate (ORR)

ORR- percentage of participants who achieved partial response (PR) or better (PR, Very Good Partial Response [VGPR], CR or sCR) based on computerized algorithm as per IMWG criteria. PR -greater than or equal to (>=) 50% reduction of serum M-protein and reduction in 24-hour urinary M-protein by >=90% or to <200 mg//24 hours. If serum and urine M-protein are not measurable, a decrease of >=50% in the difference between involved and uninvolved FLC levels is required. A >=50% reduction in the size of soft tissue plasmacytomas is also required; VGPR-serum and urine M-component detectable by immunofixation but not on electrophoresis, or >= 90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours; CR-negative immunofixation on the serum and urine, and disappearance of any soft tissue plasmacytomas, and <5% PCs in bone marrow. sCR- in addition to CR a normal FLC ratio, and absence of clonal PCs by immunohistochemistry or immunofluorescence or 2 to 4-color flow cytometry. (NCT02874742)
Timeframe: From randomization to end of following: induction treatment, ASCT, post-ASCT consolidation (after Cycle 6) and at the end of maintenance treatment of 24 months (overall duration up to 34 months)

,,
InterventionPercentage of participants (Number)
At the end of induction prior to ASCTAt the end of ASCT prior to consolidationAt the end of post-ASCT consolidationAt the End of Maintenance Treatment (up to 24 Months)
Randomized: Daratumumab+RVd (D-RVd)98.099.099.099.0
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)91.891.891.891.8
Safety Run-in: D-RVd100100100100.0

[back to top]

Duration of Complete Response or Better

Duration of CR or better is the duration from the date of initial documentation of a CR or sCR response, according to the IMWG criteria, to the date of first documented evidence of progressive disease (PR), or relapse from CR. PD is defined as an increase of 25 % from the lowest response value in one of the following: serum and urine M-component (absolute increase must be greater than or equal to [>=] 0.5 gram per deciliter [g/dL] and >=200 milligrams [mg]/24 hours respectively); Only in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved FLC levels (absolute increase must be > 10 mg/dL); Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to plasma cells (PCs) proliferative disorder. (NCT02874742)
Timeframe: From randomization to the date of first documented evidence of progressive disease or relapse from CR (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)NA
Randomized: Daratumumab+RVd (D-RVd)NA
Safety Run-in: D-RVdNA

[back to top]

Duration of Response

Duration of response is defined as the duration from the date of initial documentation of a response (PR or better) according to the IMWG criteria to the date of first documented evidence of progressive disease according to the IMWG criteria. PD is defined as an increase of 25 % from the lowest response value in one of the following: serum and urine M-component (absolute increase must be >= 0.5 g/dL and >=200 mg/24 hours respectively); Only in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved FLC levels (absolute increase must be > 10 mg/dL); Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to PC proliferative disorder. (NCT02874742)
Timeframe: From the date of initial documentation of a response (PR or better) to the date of first documented evidence of progressive (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)NA
Randomized: Daratumumab+RVd (D-RVd)NA
Safety Run-in: D-RVdNA

[back to top]

Duration of Stringent Complete Response (sCR)

Duration of sCR is the duration from the date of initial documentation of a sCR response, according to the IMWG criteria, to the date of first documented evidence of progressive disease, or relapse from sCR. PD is defined as an increase of 25 % from the lowest response value in one of the following: serum and urine M component (absolute increase must be >= 0.5 g/dL and >=200 mg/24 hours respectively); Only in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved FLC levels (absolute increase must be > 10 mg/dL); Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to PC proliferative disorder. (NCT02874742)
Timeframe: From randomization to the date of first documented evidence of progressive disease or relapse from sCR (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)NA
Randomized: Daratumumab+RVd (D-RVd)NA
Safety Run-in: D-RVdNA

[back to top]

Overall Survival (OS)

OS is measured from the date of randomization to the date of the participant's death. (NCT02874742)
Timeframe: From randomization to the date of initial documentation of participant's death (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)NA
Randomized: Daratumumab+RVd (D-RVd)NA
Safety Run-in: D-RVdNA

[back to top]

Percentage of Participants With Stringent Complete Response (sCR)

Percentage of participants who had achieved sCR as determined by the validated computer algorithm according to the International Myeloma Working Group (IMWG) criteria, by the end of post-autologous stem cell transplantation (post-ASCT) consolidation treatment were reported. Complete response (CR) is defined as negative immunofixation on the serum and urine, and disappearance of any soft tissue plasmacytomas, and less than (<) 5 percent (%) PCs in bone marrow. sCR is defined as in addition to CR a normal FLC ratio, and absence of clonal plasma cells (PCs) by immunohistochemistry or immunofluorescence or 2 to 4-color flow cytometry. (NCT02874742)
Timeframe: From randomization to post-ASCT consolidation (after Cycle 6) before maintenance treatment (up to 10 months)

InterventionPercentage of participants (Number)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)32.0
Randomized: Daratumumab+RVd (D-RVd)42.4

[back to top]

Progression-free Survival (PFS)

PFS is defined as the duration from the date of randomization to the date of first documented evidence of progressive disease or death, whichever comes first. PD is defined as an increase of 25 % from the lowest response value in one of the following: serum and urine M-component (absolute increase must be >= 0.5 g/dL and >=200 mg/24 hours respectively); Only in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved FLC levels (absolute increase must be > 10 mg/dL); Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; Development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to PC proliferative disorder. (NCT02874742)
Timeframe: From randomization to the date of first documented evidence of progressive disease or death (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)NA
Randomized: Daratumumab+RVd (D-RVd)NA
Safety Run-in: D-RVdNA

[back to top]

Time to Complete Response or Better

Time to CR or better is the duration from the date of randomization to the date of initial documentation of CR or better, which was confirmed by a repeated measurement as required by the IMWG criteria. (NCT02874742)
Timeframe: From randomization to the date of initial documentation of CR (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)9.6
Randomized: Daratumumab+RVd (D-RVd)8.9
Safety Run-in: D-RVd7.7

[back to top]

Time to Partial Response (PR) or Better

Time to PR or better is the duration from the date of randomization to the date of initial documentation of PR or better, which was confirmed by a repeated measurement as required by the IMWG criteria. (NCT02874742)
Timeframe: From randomization to the date of initial documentation of PR or better (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)0.8
Randomized: Daratumumab+RVd (D-RVd)0.8
Safety Run-in: D-RVd0.8

[back to top]

Time to Progression (TTP)

TTP is defined as the duration from the date of randomization to the date of first documented evidence of progressive disease according to the IMWG criteria. (NCT02874742)
Timeframe: From randomization to the date of first documented evidence of progressive disease (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)NA
Randomized: Daratumumab+RVd (D-RVd)NA
Safety Run-in: D-RVdNA

[back to top]

Time to Stringent Complete Response (sCR)

Time to sCR is the duration from the date of randomization to the date of initial documentation of sCR, which was confirmed by a repeated measurement as required by the IMWG criteria. (NCT02874742)
Timeframe: From randomization to the date of initial documentation of sCR (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)14.3
Randomized: Daratumumab+RVd (D-RVd)10.2
Safety Run-in: D-RVd8.4

[back to top]

Time to Very Good Partial Response (VGPR) or Better

Time to VGPR or better is the duration from the date of randomization to the date of initial documentation of VGPR or better, which was confirmed by a repeated measurement as required by the IMWG criteria. (NCT02874742)
Timeframe: From randomization to the date of initial documentation of VGPR or better (up to 5 years)

InterventionMonths (Median)
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)3.0
Randomized: Daratumumab+RVd (D-RVd)2.2
Safety Run-in: D-RVd2.1

[back to top]

Percentage of Participants Who Achieved Very Good Partial Response (VGPR) or Better

VGPR or better rate is defined as the percentage of participants who achieved VGPR or better, according to the IMWG criteria. VGPR is defined as serum and urine M-component detectable by immunofixation but not on electrophoresis, or >= 90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours. (NCT02874742)
Timeframe: From randomization to end of following: induction treatment, ASCT, post-ASCT consolidation (after Cycle 6) and at the end of maintenance period of 24 months (overall duration up to 34 months)

,,
InterventionPercentage of participants (Number)
At the end of induction prior to ASCTAt the end of ASCT prior to consolidationAt the end of post-ASCT consolidationAt the End of Maintenance Period (up to 24 Months)
Randomized: Daratumumab+RVd (D-RVd)71.786.990.996.0
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)56.766.073.277.6
Safety Run-in: D-RVd68.8100100100.0

[back to top]

Percentage of Participants With Complete Response (CR) or Better

CR or better rate is defined as the percentage of participants who achieve CR or sCR, according to the IMWG criteria. CR is negative immunofixation on the serum and urine, and disappearance of any soft tissue plasmacytomas, and < 5% PCs in bone marrow. sCR is defined as in addition to CR a normal FLC ratio, and absence of clonal plasma cells (PCs) by immunohistochemistry or immunofluorescence or 2 to 4-color flow cytometry. For 2 participants (1 in each randomized treatment group), data were updated by the study sites which resulted in their inclusion to the response-evaluable analysis set after the primary analysis. (NCT02874742)
Timeframe: From randomization to end of following: induction treatment, ASCT, post-ASCT consolidation (after Cycle 6) and at the end of maintenance period of 24 months (overall duration up to 34 months)

,,
InterventionPercentage of participants (Number)
At the end of induction prior to ASCTAt the end of ASCT prior to consolidationAt the end of post-ASCT consolidationAt the end of maintenance period (up to 24 Months)
Randomized: Daratumumab+RVd (D-RVd)19.227.351.583.0
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)13.419.642.360.2
Safety Run-in: D-RVd12.556.368.893.8

[back to top]

Percentage of Participants With Negative Minimal Residual Disease (MRD)

Minimal residual disease negative rate is defined as the percentage of participants who achieve MRD negative status by the respective time point. Minimal residual disease was evaluated in participants who achieved CR or sCR (including participants with VGPR or better and suspected daratumumab interference) using next-generation sequencing which utilizes multiple myeloma cell DNA from bone marrow aspirates at a threshold of less than (<) 10^5. (NCT02874742)
Timeframe: From randomization to end of following: induction treatment, post-ASCT consolidation (after Cycle 6) (up to 4.5 months), and at the end of maintenance period of 24 months (overall duration up to 34 months)

,,
InterventionPercentage of participants (Number)
MRD from randomization to prior to ASCT (10^5)Post ASCT consolidation (10^5)At the End of Maintenance Period (up to 24 Months) (10^5)
Randomized: Daratumumab+RVd (D-RVd)22.150.064.4
Randomized: Lenalidomide+Bortezomib+Dexamethasone (RVd)7.820.430.1
Safety Run-in: D-RVd18.850.081.3

[back to top]

Progression-free Survival

Progression-free survival is defined as the time from registration to the earliest date of documentation of disease progression or death due to any cause. Patients who receive subsequent treatment for myeloma before disease progression will be censored on the date of their last disease assessment prior to initiation of the subsequent treatment. Transplant will not be considered subsequent treatment. The distribution of progression-free survival will be estimated using the method of Kaplan-Meier. (NCT02880228)
Timeframe: From registration to the earliest date of documentation of disease progression or death due to any cause, assessed up to 3 years

Interventionmonths (Median)
Treatment (Lenalidomide, Dexamethasone, Pembrolizumab)NA

[back to top]

Proportion of Complete Response Plus Very Good Partial Response (VGPR)

"The International Myeloma Working Group response criteria was used to assess response to therapy. The proportion of VGPR response at any time during treatment with pembrolizumab added to lenalidomide and dexamethasone will be estimated by the number of patients achieving a VGPR, CR, or sCR at any time divided by the total number of evaluable patients. A very good partial response (VGPR) is defined as as a demonstration of:~Serum and urine M-component detectable by immunofixation but not on electrophoresis c or~greater than 90% reduction in serum m-component and urine m-component <100 mg/24 h~If the only measurable disease is FLC, a ≥90% reduction in the difference between involved and involved FLC levels~The proportion of successes will be estimated by the number of patients demonstrating a VGPR or better divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated according to the approach of Duffy and Santner." (NCT02880228)
Timeframe: Up to 112 days

Interventionproportion of participants (Number)
Treatment (Lenalidomide, Dexamethasone, Pembrolizumab)0

[back to top]

Proportion of Successful Stem Cell Collection

The proportion of successful stem cell collection following initial therapy with the combination of pembrolizumab, lenalidomide and dexamethasone in patients with newly diagnosed MM will be estimated by the number of patients with a successful stem cell collection divided by the total number of evaluable patients. Exact binomial 95% confidence intervals for the true successful proportion will be calculated. (NCT02880228)
Timeframe: Up to 112 days

Interventionproportion of participants (Number)
Treatment (Lenalidomide, Dexamethasone, Pembrolizumab)0.4

[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. (NCT02880228)
Timeframe: From time of registration to death due to any cause, assessed up to 3 years

Interventionmonths (Median)
Treatment (Lenalidomide, Dexamethasone, Pembrolizumab)NA

[back to top]

Partial Response (PR)

"The PR response after 4 cycles of induction treatment with pembrolizumab added to lenalidomide and dexamethasone will be estimated by the number of patients who achieve a PR, VGPR, CR, or sCR after 4 cycles divided by the total number of evaluable patients. A PR is defined by the following criteria:>~If present at baseline, ≥50% reduction of serum M-protein and reduction in 24-hour urinary M-protein or to <200 mg/24hrs>~If the only measurable disease is FLC, a ≥50% reduction in the difference between involved and involved FLC levels>~If the only measurable disease is BM, a ≥50% reduction in BM PC's (provided the baseline PC's was ≥30%)>~If present at baseline, ≥50% reduction in the size of soft tissue plasmacytomas>~Exact binomial 95% confidence intervals for the true success rate will be calculated." (NCT02880228)
Timeframe: Up to 112 days

Interventionproportion of participants (Number)
Treatment (Lenalidomide, Dexamethasone, Pembrolizumab)0

[back to top]

Duration of Response

"Duration of overall response: The duration of overall response is measured from the time measurement criteria are met for CR or PR (whichever is first recorded) until the first date that recurrent or progressive disease is objectively documented (taking as reference for progressive disease the smallest measurements recorded since the treatment started).~The duration of overall CR is measured from the time measurement criteria are first met for CR until the first date that progressive disease is objectively documented." (NCT02898259)
Timeframe: Up to 15 months after beginning treatment

Interventionmonths (Median)
DL1 (Ixazomib 2.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)2.7
DL2 (Ixazomib 3.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)8.9
DL3/MTD (Ixazomib 4.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)5.5
Expansion Cohort I: Follicular Lymphoma at MTD8.7

[back to top]

Maximum Tolerated Dose (MTD) of Oral Ixazomib

"To determine the MTD of the combination of oral ixazomib and lenalidomide plus rituximab in patients with previously untreated low-grade B cell lymphoma having high tumor burden by GELF criteria or FLIPI 3-5.~MTD will be determined using the first 12 participants 15 months after beginning treatment" (NCT02898259)
Timeframe: 15 months after beginning treatment

Interventionmg (Number)
Lenalidomide + Ixazomib + Rituximab4

[back to top]

Overall Survival

Overall survival is defined as the date of study entry to the date of death. (NCT02898259)
Timeframe: Up to 15 months after beginning treatment

InterventionParticipants (Count of Participants)
DL1 (Ixazomib 2.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)3
DL2 (Ixazomib 3.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)3
DL3/MTD (Ixazomib 4.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)6
Expansion Cohort I: Follicular Lymphoma at MTD5
Expansion Cohort 2: Non-Follicular Lymphoma at MTD0

[back to top]

Progression Free Survival

"A number of participants who survived without disease progression~PFS is defined as the duration of time from start of treatment to time of progression or death, whichever occurs 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." (NCT02898259)
Timeframe: Up to 15 months after beginning treatment

InterventionParticipants (Count of Participants)
DL1 (Ixazomib 2.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)3
DL2 (Ixazomib 3.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)2
DL3/MTD (Ixazomib 4.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)5
Expansion Cohort I: Follicular Lymphoma at MTD5
Expansion Cohort 2: Non-Follicular Lymphoma at MTD0

[back to top]

Time to Progression

Duration of time from start of treatment to time of progression. (NCT02898259)
Timeframe: Up to 15 months after beginning treatment

Interventionmonths (Median)
DL1 (Ixazomib 2.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)0
DL2 (Ixazomib 3.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)2.5
DL3/MTD (Ixazomib 4.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)9.0
Expansion Cohort I: Follicular Lymphoma at MTD4.6

[back to top]

Time to Treatment Failure

Time to treatment failure (event-free survival) is defined as the time from study entry to first event of disease progression, discontinuation of treatment for any reason, initiation of new treatment, or death. (NCT02898259)
Timeframe: Up to 15 months after beginning treatment

Interventionmonths (Median)
DL1 (Ixazomib 2.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)10
DL2 (Ixazomib 3.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)0
DL3/MTD (Ixazomib 4.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)6.1
Expansion Cohort I: Follicular Lymphoma at MTD5.8

[back to top]

Overall Response Rate

These criteria are based on the Revised Response Criteria for Malignant Lymphoma and include the following categories: Complete Response (CR)(Complete disappearance of all detectable clinical evidence of disease, and disease-related symptoms if present prior to therapy), Partial Response (PR) (A ≥ 50% decrease in sum of the product of the diameters (SPD) of up to 6 of the largest dominant nodes or extranodal masses), Stable Disease (SD) (Failing to attain the criteria needed for a PR or CR, but not fulfilling those for progressive disease), Relapse and Progression (PD) (For determination of relapsed and progressive disease, lymph nodes should be considered abnormal if the long axis is more than 1.5 cm, regardless of the short axis). (NCT02898259)
Timeframe: Up to 15 months after beginning treatment

,,,,
InterventionParticipants (Count of Participants)
Complete responseStable DiseasePartial ResponseRelapse and Progression
DL1 (Ixazomib 2.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)1200
DL2 (Ixazomib 3.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)2001
DL3/MTD (Ixazomib 4.0 mg + Lenalidomide 20 mg + Rituximab 375mg/m2)2311
Expansion Cohort 2: Non-Follicular Lymphoma at MTD0000
Expansion Cohort I: Follicular Lymphoma at MTD5000

[back to top]

Number of Participants With Adverse Events

For toxicity reporting, all adverse events and laboratory abnormalities will be graded and analyzed using CTCAE version 4 as appropriate. (NCT02903381)
Timeframe: Baseline to 2 Years

InterventionParticipants (Count of Participants)
Nivolumab, Lenalidomide, Dexamethasone8

[back to top]

Objective Response Percent

The percent of patients with objective response defined as achieving a partial response or better according to the modified International Myeloma Working Group (IMWG) criteria (NCT02903381)
Timeframe: 2 Years

Interventionpercentage of participants (Number)
Nivolumab, Lenalidomide, Dexamethasone87.5

[back to top]

Progression Free Survival (PFS) Probability at 2-years

Kaplan-Meier method, percent of patients alive and progression-free at 2-years (NCT02903381)
Timeframe: Baseline to disease progression or death from any cause, censored at date last known progression free for those who have not progressed or died, up to 24 months post initiation of therapy.

Interventionprobability (Number)
Nivolumab, Lenalidomide, Dexamethasone0.75

[back to top]

Progression Free Survival Rate-Without Cyclophosphamide

It is expected that approximately 20% of the patients will receive cyclophosphamide (CTX) for mobilization and this may influence the PFS. Therefore, in a secondary analysis the 2 year PFS rate will be evaluated among those patients who did not receive CTX. (NCT02903381)
Timeframe: 2 Years

Interventionpercentage of participants (Number)
Nivolumab, Lenalidomide, Dexamethasone71.4

[back to top]

Time to Progression Probability at 2-years

Time to progression (TTP) is defined as the time from protocol therapy initiation until documented progression, censored at date last known progression-free for those who have not progressed, up to 24 months post initiation of therapy. (NCT02903381)
Timeframe: Baseline to documented progression, up to 24 months post initiation of therapy.

Interventionprobability (Number)
Nivolumab, Lenalidomide, Dexamethasone0.75

[back to top]

Overall Survival Probability at 2-years

Kaplan-Meier method, percent alive at 2-years (NCT02903381)
Timeframe: Baseline to death or date last known alive, up to 24 months post initiation of therapy.

Interventionprobability (Number)
Nivolumab, Lenalidomide, Dexamethasone1.0

[back to top]

2 Year Progression Free Percent

The primary endpoint will be the 2-year progression-free percent and will be reported with corresponding 90% confidence interval. All patients who have received one dose of study treatment will be included for the analysis, including those who die or are lost to follow-up before 2 years. Progression is defined as ≥ 25% increase and an absolute increase of ≥ 0.5g/dL from their nadir in their serum or urine m-spike or FLC with no CRAB features attributable to MM progression. (NCT02903381)
Timeframe: 2 Year

Interventionpercentage of participants (Number)
Nivolumab, Lenalidomide, Dexamethasone62.5

[back to top]

Duration of Response Probability at 2-years

Kaplan-Meier method, duration of response probability in patients with partial response or better. Events defined as confirmed progression or death from any cause (NCT02903381)
Timeframe: time from objective response to disease progression or death, or date last known progression-free and alive for those who have not progressed or died, up to 24 months post initiation of therapy.

Interventionprobability (Number)
Nivolumab, Lenalidomide, Dexamethasone0.71

[back to top]

Evaluation of Stringent Complete Response, Complete Response, and Very Good Partial Response Rate (sCR + CR + VGPR Rate).

Assessed by the investigator per International Myeloma Working Group criteria(IMWG) uniform response criteria. Result reflects number of participants whose best overall response qualified as sCR, CR, or VGPR in 2 year follow up period. (NCT02906332)
Timeframe: Every 3 weeks (day 1 of every 21-day treatment cycle +/- 7 days) through 12 weeks.

InterventionParticipants (Count of Participants)
Pembrolizumab + Lenalidomide11

[back to top]

Number of Participants Serious Adverse Events

Safety will be assessed by quantifying the toxicities and grades experienced by subjects who have received pembrolizumab (MK-3475), lenalidomide and dexamethasone, including serious adverse events (SAEs). Result reflects count of participants who experienced an SAE. (NCT02906332)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Pembrolizumab + Lenalidomide1

[back to top]

Number of Participants Who Progressed at 12 Months

Assessed at 12 months; Subjects without documented PD or death will be censored at the last disease assessment date. Those who died without documented PD will be censored at the time of death. Result reflects count of participants who had progressed at 12 months. (NCT02906332)
Timeframe: Time from Day 0 (transplant) and date of enrollment to study completion (through 12 weeks) by investigator assessment.

InterventionParticipants (Count of Participants)
Pembrolizumab + Lenalidomide10

[back to top]

Progression Free Survival (PFS)

PFS will be assessed from the date of ASCT, with day 0 defined as date of stem cell infusion (if tandem transplant the 2nd of 2 transplants will be used) until the date of progression, defined as the date at which the patient starts the next line of therapy or the date of death. (NCT02906332)
Timeframe: Up to 3 years

Interventionmonths (Median)
Pembrolizumab + Lenalidomide27.6

[back to top] [back to top]

Number of Participants With One or More Treatment-emergent Adverse Events (TEAEs)

An adverse event (AE) is any unfavorable or unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal (investigational) product whether or not it was related to the medicinal product. A TEAE is defined as any AE that occurred after administration of the first dose of any study drug through 30 days after the last dose of any study drug. (NCT02917941)
Timeframe: Up to approximately 33 months

InterventionParticipants (Count of Participants)
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mg34

[back to top]

Overall Response Rate (ORR)

ORR:percentage of participants with CR including stringent complete response(sCR),VGPR,partial response(PR) per independent review committee(IRC)by IMWG criteria.CR: serum,urine -ve immunofixation; disappearance of soft tissue plasmacytomas,<5%plasma cells in bone marrow(CR in those with only measurable disease by serum FLC levels=normal FLC ratio 0.26 to 1.65+CR criteria).sCR:CR+normal FLC ratio,absence of clonal plasma cells(immunohistochemistry)or 2-to 4-color flow cytometry.VGPR:serum,urine M-protein detectable by immunofixation,not by electrophoresis or ≥90%reduction,<100mg/24hrs(VGPR in those with only measurable disease by serum FLC levels,requires>90%decrease in involved-uninvolved FLC level difference).PR: ≥50%reduction of serum M protein+reduction in 24-hr urinary M protein by≥90%/ to<200mg/24-hr or ≥50%decrease in difference between involved-uninvolved FLC levels/≥50%reduction in bone marrow plasma cells,if≥30%at baseline/≥50%soft tissue plasmacytoma size reduction. (NCT02917941)
Timeframe: Up to approximately 33 months

Interventionpercentage of participants (Number)
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mg84.4

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of first study drug administration to the date of death. (NCT02917941)
Timeframe: Up to approximately 33 months

Interventionmonths (Median)
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mgNA

[back to top]

Percentage of Participants With Very Good Partial Response (VGPR) or Better Response (Complete Response (CR) + VGPR)

Response was assessed using International Myeloma Working Group (IMWG) Criteria. CR was defined as negative immunofixation in serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow. VGPR was defined as serum and urine M-protein detectable by immunofixation but not on electrophoresis or 90% or greater reduction in serum M-protein plus urine M-protein level < 100 mg per 24 hours. (NCT02917941)
Timeframe: Up to approximately 33 months

Interventionpercentage of participants (Number)
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mg50.0

[back to top]

Duration of Response (DOR)

DOR was measured as time in months from date of first documentation of response, VGPR or better, and ORR (CR+PR (including sCR and VGPR) or better, to date of first documented PD among participants who responded to treatment. Response was assessed by investigator using IMWG Criteria. CR:negative immunofixation in serum and urine and; disappearance of any soft tissue plasmacytomas and;<5% plasma cells in bone marrow. sCR:CR plus normal FLC ratio, absence of clonal plasma cells by immunohistochemistry or 2- to 4-color flow cytometry. VGPR:serum and urine M-protein detectable by immunofixation but not on electrophoresis/≥90% reduction in serum M-protein plus urine M-protein level <100 mg/24 hours. PR:≥50% reduction of serum M protein+reduction in 24-hour urinary M protein by ≥90%/ to <200 mg/24-hour or ≥50% decrease in difference between involved and uninvolved FLC levels/≥50% reduction in bone marrow plasma cells, if ≥30% at baseline/≥50% reduction in size of soft tissue plasmacytomas. (NCT02917941)
Timeframe: Up to approximately 33 months

Interventionmonths (Median)
VGPR or betterORR
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mgNA21.65

[back to top]

Time to Progression (TTP)

TTP was measured as the time in months from the first dose of study treatment to the date of the first documented PD as assessed using IMWG criteria. PD required one of the following: increase of ≥ 25% from nadir in: serum M-component (absolute increase ≥ 0.5 g/dL); urine M-component (absolute increase ≥ 200 mg/24 hours); in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved FLC levels (absolute increase > 10 mg/dL); development of new or increase in the size of existing bone lesions or soft tissue plasmacytomas; development of hypercalcemia (corrected serum calcium > 11.5 mg/dL) attributed solely to plasma cell proliferative disease. (NCT02917941)
Timeframe: Up to approximately 33 months

Interventionmonths (Median)
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mg22.57

[back to top]

Progression-free Survival (PFS)

PFS was defined as the time in months from the date of first study drug administration to the date of first documentation of PD or death from any cause, whichever occurred first. PD required one of the following: increase of ≥ 25% from nadir in: serum M-component (absolute increase ≥ 0.5 g/dl); urine M-component (absolute increase ≥ 200 mg/24 hours); in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved free light chain (FLC) levels (absolute increase > 10 mg/dl); development of new or increase in the size of existing bone lesions or soft tissue plasmacytomas; development of hypercalcemia (corrected serum calcium > 11.5 mg/dl) attributed solely to plasma cell proliferative disease. (NCT02917941)
Timeframe: Up to approximately 33 months

Interventionmonths (Median)
Ixazomib 4 mg + Lenalidomide 25 mg + Dexamethasone 40 mg22.05

[back to top] [back to top]

Dose Escalation Phase: Maximum Tolerated Dose (MTD) of TAK-659

MTD was defined as the maximum dose that is determined to be safe and tolerable in different cohorts. Each cohort (A, B, C, D and E) received different escalating doses of TAK-659 in combination with other drugs. For each cohort the maximum tolerated dose of TAK-659 in combination with the other drug/s from the selected dose range is reported. (NCT02954406)
Timeframe: Cycle 1 (Cohorts A, B and C - each cycle was of 21 days and Cohorts D and E - each cycle was of 28 days)

Interventionmg (Number)
Dose Escalation Phase Cohort A: TAK-659 60-100 mg + Bendamustine 90 mg/m^2NA
Dose Escalation Phase Cohort B: TAK-659 60-100 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^2NA
Dose Escalation Phase Cohort C: TAK-659 60 mg + Gemcitabine 1000 mg/m^2NA
Dose Escalation Phase Cohort D: TAK-659 40-60 mg + Lenalidomide 25 mgNA
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mgNA

[back to top]

Time to Progression (TTP)

TTP was defined as the time from the date of first drug administration to the date of first documented PD. PD was defined as any new lesion or increase by >50% of previously involved sites from nadir. (NCT02954406)
Timeframe: Up to 123 weeks

Interventionmonths (Median)
Dose Escalation Phase Cohort A: TAK-659 60 mg + Bendamustine 90 mg/m^24.2
Dose Escalation Phase Cohort A: TAK-659 80 mg + Bendamustine 90 mg/m^22.7
Dose Escalation Phase Cohort A: TAK-659 100 mg + Bendamustine 90 mg/m^29.6
Dose Escalation Phase Cohort B: TAK-659 60 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^22.6
Dose Escalation Phase Cohort B: TAK-659 80 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^24.2
Dose Escalation Phase Cohort B: TAK-659 100 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^21.3
Dose Escalation Phase Cohort C: TAK-659 60 mg + Gemcitabine 1000 mg/m^21.4
Dose Escalation Phase Cohort D: TAK-659 40 mg + Lenalidomide 25 mg3.4
Dose Escalation Phase Cohort D: TAK-659 60 mg + Lenalidomide 25 mgNA
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mg2.7

[back to top]

Tmax: Time to Reach the Maximum Plasma Concentration for TAK-659

(NCT02954406)
Timeframe: Days 1 and 15: Pre-dose and at multiple time points (up to 24 hours) post-dose in Cycle 1 (Cohorts A, B and C - each cycle was of 21 days and Cohorts D and E - each cycle was of 28 days)

,,,,,,,,,
Interventionhours (h) (Median)
Cycle 1 Day 1Cycle 1 Day 15
Dose Escalation Phase Cohort A: TAK-659 100 mg + Bendamustine 90 mg/m^24.003.83
Dose Escalation Phase Cohort A: TAK-659 60 mg + Bendamustine 90 mg/m^24.004.00
Dose Escalation Phase Cohort A: TAK-659 80 mg + Bendamustine 90 mg/m^22.031.09
Dose Escalation Phase Cohort B: TAK-659 100 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^22.002.47
Dose Escalation Phase Cohort B: TAK-659 60 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^21.101.17
Dose Escalation Phase Cohort B: TAK-659 80 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^22.001.93
Dose Escalation Phase Cohort C: TAK-659 60 mg + Gemcitabine 1000 mg/m^24.132.76
Dose Escalation Phase Cohort D: TAK-659 40 mg + Lenalidomide 25 mg0.61.9
Dose Escalation Phase Cohort D: TAK-659 60 mg + Lenalidomide 25 mg2.122.08
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mg1.001.01

[back to top]

Cmax: Maximum Observed Plasma Concentration for TAK-659

(NCT02954406)
Timeframe: Days 1 and 15: Pre-dose and at multiple time points (up to 24 hours) post-dose in Cycle 1 (Cohorts A, B and C - each cycle was of 21 days and Cohorts D and E - each cycle was of 28 days)

,,,,,,,,,
Interventionnanograms(ng)/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 15
Dose Escalation Phase Cohort A: TAK-659 100 mg + Bendamustine 90 mg/m^2118.10187.73
Dose Escalation Phase Cohort A: TAK-659 60 mg + Bendamustine 90 mg/m^283.61126.49
Dose Escalation Phase Cohort A: TAK-659 80 mg + Bendamustine 90 mg/m^289.74145.03
Dose Escalation Phase Cohort B: TAK-659 100 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^2180.10223.53
Dose Escalation Phase Cohort B: TAK-659 60 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^2128.58158.28
Dose Escalation Phase Cohort B: TAK-659 80 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^2142.84222.96
Dose Escalation Phase Cohort C: TAK-659 60 mg + Gemcitabine 1000 mg/m^2141.27303.34
Dose Escalation Phase Cohort D: TAK-659 40 mg + Lenalidomide 25 mg139.00188.00
Dose Escalation Phase Cohort D: TAK-659 60 mg + Lenalidomide 25 mg65.03202.90
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mg120.23306.59

[back to top] [back to top]

AUCtau: Area Under the Plasma Concentration-time Curve During Dosing Interval

(NCT02954406)
Timeframe: Days 1 and 15: Pre-dose and at multiple time points (up to 24 hours) post-dose in Cycle 1 (Cohorts A, B and C - each cycle was of 21 days and Cohorts D and E - each cycle was of 28 days)

,,,,,,,,,
Interventionh×ng/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 15
Dose Escalation Phase Cohort A: TAK-659 100 mg + Bendamustine 90 mg/m^21532.343050.97
Dose Escalation Phase Cohort A: TAK-659 60 mg + Bendamustine 90 mg/m^21094.511845.21
Dose Escalation Phase Cohort A: TAK-659 80 mg + Bendamustine 90 mg/m^21011.491675.91
Dose Escalation Phase Cohort B: TAK-659 100 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^21444.892662.16
Dose Escalation Phase Cohort B: TAK-659 60 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^2852.361896.76
Dose Escalation Phase Cohort B: TAK-659 80 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^21455.922788.61
Dose Escalation Phase Cohort C: TAK-659 60 mg + Gemcitabine 1000 mg/m^2387.543251.29
Dose Escalation Phase Cohort D: TAK-659 40 mg + Lenalidomide 25 mg1456.032536.48
Dose Escalation Phase Cohort D: TAK-659 60 mg + Lenalidomide 25 mg758.762578.30
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mg968.824218.90

[back to top]

Overall Response Rate (ORR)

ORR was defined as the percentage of participants in the response-evaluable population who achieved either complete response (CR), or partial response (PR). CR was defined as the disappearance of all evidence of disease, and PR was defined as regression of measurable disease and no new sites. (NCT02954406)
Timeframe: Up to 123 weeks

Interventionpercentage of participants (Number)
Dose Escalation Phase Cohort A: TAK-659 60 mg + Bendamustine 90 mg/m^233.3
Dose Escalation Phase Cohort A: TAK-659 80 mg + Bendamustine 90 mg/m^275.0
Dose Escalation Phase Cohort A: TAK-659 100 mg + Bendamustine 90 mg/m^250.0
Dose Escalation Phase Cohort B: TAK-659 60 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^240.0
Dose Escalation Phase Cohort B: TAK-659 80 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^280.0
Dose Escalation Phase Cohort B: TAK-659 100 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^20.0
Dose Escalation Phase Cohort C: TAK-659 60 mg + Gemcitabine 1000 mg/m^20.0
Dose Escalation Phase Cohort D: TAK-659 40 mg + Lenalidomide 25 mg100.0
Dose Escalation Phase Cohort D: TAK-659 60 mg + Lenalidomide 25 mg33.3
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mg50.0

[back to top]

Duration of Response (DOR)

DOR was defined as the time from the date of first documented response to the date of first documented PD. PD was defined as any new lesion or increase by > 50% of previously involved sites from nadir. (NCT02954406)
Timeframe: Up to 123 weeks

Interventionmonths (Median)
Dose Escalation Phase Cohort A: TAK-659 60 mg + Bendamustine 90 mg/m^22.3
Dose Escalation Phase Cohort A: TAK-659 80 mg + Bendamustine 90 mg/m^22.8
Dose Escalation Phase Cohort A: TAK-659 100 mg + Bendamustine 90 mg/m^2NA
Dose Escalation Phase Cohort B: TAK-659 60 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^2NA
Dose Escalation Phase Cohort B: TAK-659 80 mg + Bendamustine 90 mg/m^2 + Rituximab 375 mg/m^23.9
Dose Escalation Phase Cohort D: TAK-659 40 mg + Lenalidomide 25 mg1.8
Dose Escalation Phase Cohort D: TAK-659 60 mg + Lenalidomide 25 mgNA
Dose Escalation Phase Cohort E: TAK-659 60 mg + Ibrutinib 560 mgNA

[back to top]

Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for Immunohistochemistry (IHC) Programmed Death Ligand - 1 (PDL1) Total Percentage

Data in the analysis include all participants who received durvalumab, irrespective of the study arm in which they enrolled, because the selected biomarkers have been associated with response in other anti-PD1 or anti-PDL1 studies, such as durvalumab. IHC analysis was performed on the baseline tumor biopsy to quantify CD8 T-cell density. Participants with 'high' values, i.e. above the threshold defined as the median value of 774 cells/mm^2 found in commercial DLBCL samples using matched analytical methods, were predicted to be responders to treatment with durvalumab. The definition of a complete response was that used in the primary outcome. (NCT03003520)
Timeframe: Biomarker biopsies: Days -28 to Day -1. Clinical response: From first dose of study drug to end of Induction therapy (Day 1 up to Week 26 - maximum duration of Induction Period).

Interventionpercentage of participants (Number)
High Group for PDL1 % of Total Cells75
Low Group for PDL1 % of Total Cells20

[back to top]

Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for the Interferon Gamma Score (IFNG-Score) From Ribonucleic Acid (RNA)-Sequencing Data

Data in the analysis include all participants who received durvalumab, irrespective of the study arm in which they enrolled, because the selected biomarkers have been associated with response in other anti-PD1 or anti-PDL1 studies, such as durvalumab. IHC analysis was performed on the baseline tumor biopsy to quantify CD8 T-cell density. Participants with 'high' values, i.e. above the threshold defined as the median value of 774 cells/mm^2 found in commercial DLBCL samples using matched analytical methods, were predicted to be responders to treatment with durvalumab. The definition of a complete response was that used in the primary outcome. (NCT03003520)
Timeframe: Biomarker biopsies: Days -28 to Day -1. Clinical response: From first dose of study drug to end of Induction therapy (Day 1 up to Week 26 - maximum duration of Induction Period).

Interventionpercentage of participants (Number)
High Group for RNA IFN Gamma Score43
Low Group for RNA IFN Gamma Score60

[back to top]

Percentage of Participants Who Achieved a Complete Response (CR) at the End of Induction Therapy

The primary efficacy analysis evaluated the complete response rate (CRR) at the end of the induction therapy in the efficacy evaluable population in a comparative manner against historical control. The response to treatment was assessed according to the 2014 International Working Group (IWG) Response Criteria for Non-Hodgkin's Lymphoma (NHL) (Cheson, 2014). CR was defined as a complete metabolic response and radiographic evidence showing target nodes/nodal masses regressed to ≤ 1.5 cm in longest diameter, no new lesions, regression of lymph nodes to normal size, absence of splenomegaly, and absence of bone marrow involvement. Clopper-Pearson two-sided 95% confidence interval is reported. Null hypothesis for the primary endpoint was rejected if the lower limit of the confidence interval for the complete response rate at the completion of the induction therapy in the efficacy evaluable population is above 55%. (NCT03003520)
Timeframe: From first dose of study drug to end of Induction therapy (Day 1 up to Week 26 - maximum duration of Induction Period).

Interventionpercentage of participants (Number)
DUR + R-CHOP54.1
DUR + R2-CHOP66.7

[back to top]

Percentage of Participants Who Responded During Induction and Continued Into Consolidation Therapy (Database Cutoff Date: 02-Aug-2018)

The percentage of participants who achieved a partial response (PR) or complete response (CR) at the end of Induction and continued into consolidation period in the efficacy evaluable population in a comparative manner against historical control. The response to treatment was assessed according to the 2014 International Working Group (IWG) Response Criteria for Non-Hodgkin's Lymphoma (NHL) (Cheson, 2014). CR was defined in outcome #1. PR was defined as a partial metabolic response and radiographic evidence showing ≥ 50% decrease in sum of perpendicular diameters (SPD) of up to 6 target measurable nodes and extranodal sites, no new lesions, spleen must have regressed > 50% in length beyond normal, and residual bone marrow involvement improved from baseline. Clopper-Pearson two-sided 95% confidence interval is reported. Null hypothesis was rejected if the lower limit of the confidence interval for the rate of subjects who continue consolidation therapy out of all subjects. (NCT03003520)
Timeframe: From first dose of study drug to completion of at least one cycle in the Consolidation Period (Day 1 up to Week 52)

Interventionpercentage of participants (Number)
DUR + R-CHOP67.6
DUR + R2-CHOP66.7

[back to top]

Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for Immunohistochemistry (IHC) CD8 T-Cell Density

Data in the analysis include all participants who received durvalumab, irrespective of the study arm in which they enrolled, because the selected biomarkers have been associated with response in other anti-PD1 or anti-PDL1 studies, such as durvalumab. IHC analysis was performed on the baseline tumor biopsy to quantify CD8 T-cell density. Participants with 'high' values, i.e. above the threshold defined as the median value of 774 cells/mm^2 found in commercial DLBCL samples using matched analytical methods, were predicted to be responders to treatment with durvalumab. The definition of a complete response was that used in the primary outcome. (NCT03003520)
Timeframe: Biomarker biopsies: Days -28 to Day -1. Clinical response: From first dose of study drug to end of Induction therapy (Day 1 up to Week 26 - maximum duration of Induction Period).

Interventionpercentage of participants (Number)
High Group for CD8 Density67
Low Group for CD8 Density64

[back to top]

Percentage of Participants Who Achieved a Clinical Response in the Biomarker Subpopulation for Immunohistochemistry (IHC) Programmed Death Ligand - 1 (PDL1) Percentage of Tumor Cells

Data in the analysis include all participants who received durvalumab, irrespective of the study arm in which they enrolled, because the selected biomarkers have been associated with response in other anti-PD1 or anti-PDL1 studies, such as durvalumab. IHC analysis was performed on the baseline tumor biopsy to quantify CD8 T-cell density. Participants with 'high' values, i.e. above the threshold defined as the median value of 774 cells/mm^2 found in commercial DLBCL samples using matched analytical methods, were predicted to be responders to treatment with durvalumab. The definition of a complete response was that used in the primary outcome. (NCT03003520)
Timeframe: Biomarker biopsies: Days -28 to Day -1. Clinical response: From first dose of study drug to end of Induction therapy (Day 1 up to Week 26 - maximum duration of Induction Period).

Interventionpercentage of participants (Number)
High Group for PDL1 % of Tumor Cells64
Low Group for PDL1 % of Tumor Cells56

[back to top]

Participants With Treatment Emergent Adverse Events (TEAE)

An AE is any noxious, unintended, or untoward medical occurrence that may appear or worsen during a study. A serious AE is any AE occurring at any dose that: • Results in death; • Is life-threatening; • Requires or prolongs existing inpatient hospitalization; • Results in persistent or significant disability/incapacity; • Is a congenital anomaly/birth defect; • Constitutes an important medical event. The Investigator assessed the relationship of each AE to study drug and graded the severity according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE, Version 4.03): - Grade 1 = Mild (no limitation in activity or intervention); - Grade 2 = Moderate (some limitation in activity; no/minimal medical intervention required); - Grade 3 = Severe (marked limitation in activity; medical intervention required, hospitalization possible); - Grade 4 = Life-threatening; - Grade 5 = Death. Relation to IP is determined by the investigator. (NCT03003520)
Timeframe: From the date of the first dose of study drug to within 90 days after the last dose of durvalumab or 28 days after the last dose of any investigational product (IP) whichever is greater. (Up to approximately 72 weeks)

InterventionParticipants (Count of Participants)
>= 1 Treatment-emergent adverse event (TEAE)>=1 TEAE related to durvalumab>=1 TEAE related to R-CHOP>=1 TEAE related to durvalumab or any other IP>=1 TEAE severity grade 3-4>=1 TEAE severity grade 3-4 related to durvalumab>=1 TEAE severity grade 3-4 related to R-CHOP>=1 TEAE severity grade 3-4 related to any IP>=1 TEAE severity grade 5>=1 TEAE severity grade 5 related to any IP>=1 serious TEAE>=1 serious TEAE related to durvalumab>=1 serious TEAE related to R-CHOP>=1 serious TEAE related to any IP>=1 leading to discontinuation of durvalumab>=1 leading to discontinuation of R-CHOP>=1 leading to discontinuation of any IP>=1 leading to interruption of durvalumab>=1 leading to interruption of R-CHOP>=1 leading to interruption of any IP>=1 leading to infusion interruption of durvalumab>=1 leading to dose reduction of vincristine
DUR + R-CHOP433340413718273130231010141341315121824

[back to top]

Participants With Treatment Emergent Adverse Events (TEAE)

An AE is any noxious, unintended, or untoward medical occurrence that may appear or worsen during a study. A serious AE is any AE occurring at any dose that: • Results in death; • Is life-threatening; • Requires or prolongs existing inpatient hospitalization; • Results in persistent or significant disability/incapacity; • Is a congenital anomaly/birth defect; • Constitutes an important medical event. The Investigator assessed the relationship of each AE to study drug and graded the severity according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE, Version 4.03): - Grade 1 = Mild (no limitation in activity or intervention); - Grade 2 = Moderate (some limitation in activity; no/minimal medical intervention required); - Grade 3 = Severe (marked limitation in activity; medical intervention required, hospitalization possible); - Grade 4 = Life-threatening; - Grade 5 = Death. Relation to IP is determined by the investigator. (NCT03003520)
Timeframe: From the date of the first dose of study drug to within 90 days after the last dose of durvalumab or 28 days after the last dose of any investigational product (IP) whichever is greater. (Up to approximately 72 weeks)

InterventionParticipants (Count of Participants)
>= 1 Treatment-emergent adverse event (TEAE)>=1 TEAE related to durvalumab>=1 TEAE related to R-CHOP>=1 TEAE related to lenalidomide>=1 TEAE related to durvalumab or any other IP>=1 TEAE severity grade 3-4>=1 TEAE severity grade 3-4 related to durvalumab>=1 TEAE severity grade 3-4 related to R-CHOP>=1 TEAE severity grade 3-4 related to lenalidomid>=1 TEAE severity grade 3-4 related to any IP>=1 TEAE severity grade 5>=1 TEAE severity grade 5 related to any IP>=1 serious TEAE>=1 serious TEAE related to durvalumab>=1 serious TEAE related to R-CHOP>=1 serious TEAE related to lenalidomide>=1 serious TEAE related to any IP>=1 leading to discontinuation of durvalumab>=1 leading to discontinuation of R-CHOP>=1 leading to discontinuation of lenalidomide>=1 leading to discontinuation of any IP>=1 leading to interruption of durvalumab>=1 leading to interruption of R-CHOP>=1 leading to interruption of lenalidomide>=1 leading to interruption of any IP>=1 leading to infusion interruption of durvalumab>=1 leading to dose reduction of vincristine>=1 leading to dose reduction of lenalidomide
DUR + R2-CHOP3333331323001111100002123011

[back to top] [back to top]

Number of Participants Who Survived

Number of Participants Who Survived at day 180. (NCT03019640)
Timeframe: From the time of transplant, assessed up to day 180

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, NK Infusion, Stem Cell Transplant)16

[back to top]

Total Number of Adverse Events

Collect and grade all of the adverse events to evaluate for safety. This data was collected for the first 2 cycles for each participant. (NCT03050450)
Timeframe: Two 28 day cycles

Interventiontotal adverse events (Number)
Grade 1 adverse eventsGrace 2 adverse eventsGrade 3 adverse eventsGrade 4 adverse eventsGrade 5 adverse events
Dose Level 168321221

[back to top]

Best Response of Children With Recurrent or Refractory Central Nervous System Tumors

Best response by MRIs per definitions in the protocol (complete response, partial response, stable disease, progressive disease). MRI's were obtained every 2 cycles and the best response was reported. (NCT03050450)
Timeframe: Every 2 cycles up to 24 cycles

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Dose Level 10112

[back to top]

Number Participants With Hematologic and Non-hematologic Toxicities

Number participants with grades 3 to 5 hematologic and non-hematologic toxicities. All toxicities are for end of cycle 2. (NCT03050450)
Timeframe: Two 28 day cycles

InterventionParticipants (Count of Participants)
Grade 3 & 4 Blood/Bone MarrowGrade 3 & 4 GastrointestinalGrade 3 & 4 Metabolic/LaboratoryGrade 3 & 4 Musculoskelatal/Soft TissueGrade 3 & 4 NeurologyGrade 3 & 4 PainGrade 3 & 4 VascularGrade 3 & 4 & 5 Other
Dose Level 102511006

[back to top]

Transfusion Support: Number of Red Blood Cell and Platelet Transfusions

Number of red blood cell and platelet transfusions received within the first 50 days of treatment (NCT03118466)
Timeframe: 50 days

InterventionNumber of Transfusions (Median)
Platelet TransfusionsRed Blood Cell Transfusions
Lenalidomide and MEC Chemotherapy79

[back to top] [back to top]

Overall Survival

Overall survival is defined as time from diagnosis of disease until date of death or censored on the last known date alive if patients are still alive. (NCT03118466)
Timeframe: Up to 3 years

InterventionMonths (Median)
Lenalidomide and MEC Chemotherapy16

[back to top]

Number of Patients That Achieved Platelet Recovery

The number of patients that achieved a stable platelet count > 20,000/mm3 for 3 days within 45 days of starting treatment (NCT03118466)
Timeframe: up to 45 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy27

[back to top]

Number of Patients That Achieved ANC Recovery

The number of patients that achieved a neutrophil count of > 500/mm3 for 3 days within 45 of starting treatment (NCT03118466)
Timeframe: up to 45 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy25

[back to top]

Complete Response Rate

"Proportion of patients who have achieve CR or CRp after treatment.~Morphologic Complete Remission (CR): Defined as morphologic leukemia-free state, including <5% blasts in Bone Marrow aspirate with marrow spicules, no persistent extramedullary disease, ANC >1000/mm3 and platelet count >100,000/mm3.~Morphologic Complete Remission without platelet recovery (CRp): Defined as CR with the exception of platelet count < 100,000/mm3 (CRp)." (NCT03118466)
Timeframe: up to 45 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy19

[back to top]

Overall Survival

Overall survival is defined as the time from date of study enrollment until death from any cause. (NCT03224507)
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 58 months.

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT108

[back to top]

Percentage of Patients Achieving Complete Remission Following Complete Therapy

The primary endpoint of MRD(-) rate will be estimated along with two-sided 95% confidence interval using Clopper-Pearson exact method. Simon's optimal two-stage design will be utilized in determining the rate of MRD(-) cases. MRD assessment will be done with ClonoSEQ to identify myeloma-specific sequences. Complete therapy incorporates induction and consolidation therapy. (NCT03224507)
Timeframe: Baseline up to 15 months

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT106

[back to top]

Serious Adverse Events (SAEs) From the KRdD Treatment

The Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 will be used for this assessment. SAEs include events that are Grade 3 and above; non-serious events are Grades 1-2. (NCT03224507)
Timeframe: Baseline until the progression of disease or MRD(-) status up to an estimated 15 months.

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT14

[back to top]

Percentage of Patients With MRD(-) Status at the Completion of Induction Therapy

The primary endpoint of MRD(-) rate will be estimated along with two-sided 95% confidence interval using Clopper-Pearson exact method. Simon's optimal two-stage design will be utilized in determining the rate of MRD(-) cases. MRD assessment will be done with ClonoSEQ to identify myeloma-specific sequences. (NCT03224507)
Timeframe: Baseline until MRD(-) status estimated at 6 months or until disease progression

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT45

[back to top]

Progression-free Survival

Progression-free survival is defined as the interval from the start of therapy to the earliest occurrence of the following: disease progression, initiation of anti-myeloma therapy that is not an accepted maintenance therapy of lenalidomide or death from any cause. Kaplan-Meier methods will used. (NCT03224507)
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 58 months.

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT90

[back to top]

Percentage of Patients With MRD(-) Remissions at the Completion of Consolidation Therapy

The primary endpoint of MRD(-) rate, or percentage of patients with MRD(-) remissions, will be estimated along with two-sided 95% confidence interval using Clopper-Pearson exact method. Simon's optimal two-stage design will be utilized in determining the rate of MRD(-) cases. MRD assessment will be done with ClonoSEQ to identify myeloma-specific sequences. (NCT03224507)
Timeframe: Baseline until MRD(-) is reached estimated to be up to 15 months.

Interventionpercentage of patients achieving MRD (-) (Number)
KRdD Followed by Auto-HCT81.4

[back to top]

Percentage of Patients With Auto-HCT That Convert From Positive to Negative MRD

The primary endpoint of MRD(-) rate will be estimated along with two-sided 95% confidence interval using Clopper-Pearson exact method. Simon's optimal two-stage design will be utilized in determining the rate of MRD(-) cases. MRD assessment will be done with ClonoSEQ to identify myeloma-specific sequences. (NCT03224507)
Timeframe: From baseline up to an estimated 9 months

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT32

[back to top]

Percentage of Patients That Convert From MRD(-) to MRD(+) Following Treatment Discontinuation

The primary endpoint of MRD(-) rate will be estimated along with two-sided 95% confidence interval using Clopper-Pearson exact method. Simon's optimal two-stage design will be utilized in determining the rate of MRD(-) cases. MRD assessment will be done with ClonoSEQ to identify myeloma-specific sequences. (NCT03224507)
Timeframe: Baseline to 2 years

InterventionParticipants (Count of Participants)
KRdD Followed by Auto-HCT23

[back to top]

Overall Response

Overall response with elotuzumab and lenalidomide for each study arm. Overall Response is defined as best Overall Response, as Complete Response or Partial Response. Response will be assessed per the uniform response criteria of the International Myeloma Working Group(IMWG). Myeloma participants enrolled in this clinical study will be assessed for disease response after every cycle. Complete Response= Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and <5% plasma cells in bone marrow aspirates; Partial Response= ≥50% reduction of serum M-protein plus reduction in 24 h urinary M-protein by ≥90% or to <200 mg per 24 h; (NCT03411031)
Timeframe: Up to 60 days post last study treatment

InterventionParticipants (Count of Participants)
A: Elotuzumab + Lenalidomide at 25 mg1
B: Elotuzumab + Lenalidomide at 10 mg2

[back to top]

Minimum Response (MR)

Minimum response (MR) or better with elotuzumab and lenalidomide for each study arm. The Consensus on Uniform Reporting of Response will be used to evaluate response. Myeloma participants enrolled in this clinical study will be assessed for disease response after every cycle. (NCT03411031)
Timeframe: Up to 60 days post last study treatment

InterventionParticipants (Count of Participants)
A: Elotuzumab + Lenalidomide at 25 mg2
B: Elotuzumab + Lenalidomide at 10 mg3

[back to top]

Percentage of Participants With Progression Free Survival (PFS)

Progression free survival (PFS) is defined as the time of randomization to date of death from any cause, date of relapse/progression, or the last follow-up date, whichever comes first. The Kaplan-Meier method will be used to estimate PFS for each Study Arm. The method of Brookmeyer and Crowley will be used to construct 95% confidence interval. (NCT03411031)
Timeframe: An average of 8 months

Interventionpercentage of participants (Number)
A: Elotuzumab + Lenalidomide at 25 mg11.1
B: Elotuzumab + Lenalidomide at 10 mg22.2

[back to top]

Percentage of Participants With Anti-Daratumumab Antibodies

Percentage of participants with antibodies to daratumumab were reported. (NCT03412565)
Timeframe: For D-VRD Arm: Baseline up to 2 years 3 months; For D-VMP, D-Rd and D-Kd Arms: Baseline up to 2 years 7 months

Interventionpercentage of participants (Number)
Daratumumab (D)+Bortezomib+Lenalidomide+Dexamethasone (D-VRd)0
D + Bortezomib + Melphalan + Prednisone (D-VMP)0
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)0
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)0

[back to top]

D-VRd Cohort: Overall Response Rate (ORR)

ORR was defined as the percentage of participants who achieved a PR or better, IMWG criteria, during the study or during follow up. IMWG criteria for PR >= 50% reduction of serum M-protein and reduction in 24 hour urinary M-protein by >=90% or to <200 mg/24 hours, if the serum and urine M-protein are not measurable, a decrease of >=50% in the difference between involved and uninvolved FLC levels is required in place of the M-protein criteria, If serum and urine M-protein are not measurable, and serum free light assay is also not measurable, >=50% reduction in bone marrow PCs is required in place of M-protein, provided baseline bone marrow plasma cell percentage was >=30%, in addition to the above criteria, if present at baseline, a >=50% reduction in the size of soft tissue plasmacytomas is also required. (NCT03412565)
Timeframe: Up to 2 years and 3 months

Interventionpercentage of participants (Number)
Daratumumab (D)+Bortezomib+Lenalidomide+Dexamethasone (D-VRd)97.0

[back to top]

D-VMP, D-Rd, and D-Kd Cohorts: Percentage of Participants With VGPR or Better Response

VGPR or better rate was defined as the percentage of participants who achieved VGPR or CR (including sCR) according to the IMWG criteria during or after the study treatment. VGPR: Serum and urine component detectable by immunofixation but not on electrophoresis, or >= 90% reduction in serum M-protein plus urine M-protein level <100 mg per 24 hour; CR: negative immunofixation on the serum and urine, Disappearance of any soft tissue plasmacytomas and <5% PCs in bone marrow; sCR: CR in addition to having a normal FLC ratio and an absence of clonal cells in bone marrow by immunohistochemistry, immunofluorescence, 2-4 color flow cytometry. (NCT03412565)
Timeframe: From baseline up to 2 years 7 months

Interventionpercentage of participants (Number)
D + Bortezomib + Melphalan + Prednisone (D-VMP)77.6
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)80.0
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)77.3

[back to top]

D-VMP, D-Rd, and D-Kd Cohorts: Percentage of Participants With Minimal Residual Disease (MRD) Negative Rate

MRD negativity rate was defined as the percentage of participants who were considered MRD negative after MRD testing at any timepoint after the first dose by bone marrow aspirate. MRD negativity rate was assessed by next-generation sequencing at a threshold of <10^5. (NCT03412565)
Timeframe: Up to 2 years and 3 months

Interventionpercentage of participants (Number)
D + Bortezomib + Melphalan + Prednisone (D-VMP)25.4
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)21.5
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)27.3

[back to top]

D-VRd Cohort: Percentage of Participants With Very Good Partial Response (VGPR) or Better Response

VGPR or better rate was defined as the percentage of participants who achieved VGPR or complete response (CR) (including stringent complete response [sCR]) according to the IMWG criteria during or after the study treatment. VGPR: Serum and urine component detectable by immunofixation but not on electrophoresis, or >= 90% reduction in serum M-protein plus urine M-protein level <100 mg/24 hour; CR: negative immunofixation on the serum and urine, Disappearance of any soft tissue plasmacytomas and <5% PCs in bone marrow; sCR: CR in addition to having a normal FLC ratio and an absence of clonal cells in bone marrow by immunohistochemistry, immunofluorescence, 2-4 color flow cytometry. (NCT03412565)
Timeframe: Up to 2 years and 3 months

Interventionpercentage of participants (Number)
Daratumumab (D)+Bortezomib+Lenalidomide+Dexamethasone (D-VRd)71.6

[back to top] [back to top]

Maximum Observed Serum Concentration (Cmax) of Daratumumab

Cmax was defined as maximum serum concentration observed following daratumumab administration. Each cycle for: D-VRd cohort is of 21 days, D-VMP cohort is of 42 days and D-Rd and D-Kd cohorts is of 28 days. Each cohort have a treatment period of 84 days. (NCT03412565)
Timeframe: D-VRd: Day 4 of Cycles 1 and 4 and post treatment at Week 8; D-VMP: Day 4 of Cycles 1 and 2 and post treatment at Week 8; D-Rd and D-Kd: Day 4 of Cycles 1 and 3 and post treatment at Week 8

Interventionmicrograms per milliliter (mcg/mL) (Mean)
Cycle 1 Day 4Cycle 3 Day 4
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)108648

[back to top]

Maximum Observed Serum Concentration (Cmax) of Daratumumab

Cmax was defined as maximum serum concentration observed following daratumumab administration. Each cycle for: D-VRd cohort is of 21 days, D-VMP cohort is of 42 days and D-Rd and D-Kd cohorts is of 28 days. Each cohort have a treatment period of 84 days. (NCT03412565)
Timeframe: D-VRd: Day 4 of Cycles 1 and 4 and post treatment at Week 8; D-VMP: Day 4 of Cycles 1 and 2 and post treatment at Week 8; D-Rd and D-Kd: Day 4 of Cycles 1 and 3 and post treatment at Week 8

Interventionmicrograms per milliliter (mcg/mL) (Mean)
Cycle 1 Day 4Cycle 2 Day 4Post-treatment Phase Week 8
D + Bortezomib + Melphalan + Prednisone (D-VMP)98.6612162

[back to top]

Maximum Observed Serum Concentration (Cmax) of Daratumumab

Cmax was defined as maximum serum concentration observed following daratumumab administration. Each cycle for: D-VRd cohort is of 21 days, D-VMP cohort is of 42 days and D-Rd and D-Kd cohorts is of 28 days. Each cohort have a treatment period of 84 days. (NCT03412565)
Timeframe: D-VRd: Day 4 of Cycles 1 and 4 and post treatment at Week 8; D-VMP: Day 4 of Cycles 1 and 2 and post treatment at Week 8; D-Rd and D-Kd: Day 4 of Cycles 1 and 3 and post treatment at Week 8

Interventionmicrograms per milliliter (mcg/mL) (Mean)
Cycle 1 Day 4Cycle 3 Day 4Post-treatment Phase Week 8
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)13786949.3

[back to top]

Maximum Observed Serum Concentration (Cmax) of Daratumumab

Cmax was defined as maximum serum concentration observed following daratumumab administration. Each cycle for: D-VRd cohort is of 21 days, D-VMP cohort is of 42 days and D-Rd and D-Kd cohorts is of 28 days. Each cohort have a treatment period of 84 days. (NCT03412565)
Timeframe: D-VRd: Day 4 of Cycles 1 and 4 and post treatment at Week 8; D-VMP: Day 4 of Cycles 1 and 2 and post treatment at Week 8; D-Rd and D-Kd: Day 4 of Cycles 1 and 3 and post treatment at Week 8

Interventionmicrograms per milliliter (mcg/mL) (Mean)
Cycle 1 Day 4Cycle 4 Day 4Post-treatment Phase Week 8
Daratumumab (D)+Bortezomib+Lenalidomide+Dexamethasone (D-VRd)100746263

[back to top]

D-VMP, D-Rd, and D-Kd Cohorts: Overall Response Rate (ORR)

ORR was defined as the percentage of participants who achieved partial response (PR) or better according to international myeloma working group (IMWG) criteria. IMWG criteria for PR: greater than or equal to (>=) 50 percent (%) reduction of serum M-protein and reduction in 24-hour urinary M-protein by >=90% or to less than (<) 200 milligrams (mg) per 24 hours, If the serum and urine M-protein are not measurable, a decrease of >=50% in the difference between involved and uninvolved free light chain (FLC) levels is required in place of the M-protein criteria, If serum and urine M-protein are not measurable, and serum free light assay is also not measurable, >=50% reduction in bone marrow plasma cells (PCs) is required in place of M-protein, provided baseline bone marrow plasma cell percentage was >=30%. In addition to the above criteria, if present at baseline, a >=50% reduction in the size of soft tissue plasmacytomas is also required. (NCT03412565)
Timeframe: Up to 2 years 3 months

Interventionpercentage of participants (Number)
D + Bortezomib + Melphalan + Prednisone (D-VMP)88.1
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)90.8
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)84.8

[back to top]

D-VMP, D-Rd and D-Kd Cohorts: Duration of Response (DOR)

DOR was defined as the time from the date of initial documented response (PR or better response) to the date of first documented evidence of progressive disease (PD) or death due to PD. PD is defined as an increase of 25% from the lowest response value in one of the following: serum and urine M-component (absolute increase must be >=0.5 gram per deciliter [g/dL] and >=200 mg/24 hours respectively); only in participants without measurable serum and urine M-protein levels the difference between involved and uninvolved FLC levels (absolute increase must be >10 mg/dL); definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; development of hypercalcemia (corrected serum calcium >11.5 mg/dL) that can be attributed solely to PC proliferative disorder. (NCT03412565)
Timeframe: From baseline up to 2 years 7 months

Interventionmonths (Median)
D + Bortezomib + Melphalan + Prednisone (D-VMP)NA
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)NA
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)NA

[back to top]

Percentage of Participants With CR or Better Response

CR or better rate was defined as the percentage of participants with a CR or better response (that is, CR and sCR) as per IMWG criteria. CR: as negative immunofixation on the serum and urine and disappearance of soft tissue plasmacytomas and less than (<) 5 percent plasma cells in bone marrow; sCR: CR plus normal FLC ratio and absence of clonal PCs by immunohistochemistry, immunofluorescence or 2- to 4-color flow cytometry. (NCT03412565)
Timeframe: For D-VRD Arm: Baseline up to 2 years 3 months; For D-VMP, D-Rd and D-Kd Arms: Baseline up to 2 years 7 months

Interventionpercentage of participants (Number)
Daratumumab (D)+Bortezomib+Lenalidomide+Dexamethasone (D-VRd)16.4
D + Bortezomib + Melphalan + Prednisone (D-VMP)55.2
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)50.8
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)42.4

[back to top]

Percentage of Participants With Anti-rHuPH20 Antibodies

Percentage of participants with antibodies to rHuPH20 were reported. (NCT03412565)
Timeframe: For D-VRD Arm: Baseline up to 2 years 3 months; For D-VMP, D-Rd and D-Kd Arms: Baseline up to 2 years 7 months

Interventionpercentage of participants (Number)
Daratumumab (D)+Bortezomib+Lenalidomide+Dexamethasone (D-VRd)6.1
D + Bortezomib + Melphalan + Prednisone (D-VMP)3.1
Daratumumab + Lenalidomide + Dexamethasone (D-Rd)4.8
Daratumumab + Carfilzomib + Dexamethasone (D-Kd)4.7

[back to top]

Percentage of Participants With Bone Lesions (Bone Evaluation)

(NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionPercentage of Participants (Number)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy100.0
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy59.1
[Overall]; Combination Therapy + Ixazomib Therapy64.0

[back to top]

Percentage of Participants Who Achieved VGPR or Better (CR + VGPR)

VGPR or better (CR + VGPR) were assessed by IMWG Criteria. Per IMWG criteria, PR (partial response): ≥50% reduction of serum M protein+reduction in 24-hour urinary M protein by ≥90%/ to <200 mg/24-hour or ≥50% decrease in difference between involved and uninvolved free light chain (FLC) levels/ ≥50% reduction in bone marrow plasma cells, if ≥30% at baseline/ ≥50% reduction in size of soft tissue plasmacytomas. VGPR (very good PR): serum+urine M-protein detectable by immunofixation but not on electrophoresis/ ≥90% reduction in serum M-protein+urine Mprotein level <100 mg/24-hour. CR (complete response): negative immunofixation on serum+urine+disappearance of soft tissue plasmacytomas+<5% plasma cells in bone marrow. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionPercentage of Participants (Number)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy33.3
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy43.6
[Overall]; Combination Therapy + Ixazomib Therapy42.2

[back to top]

Percentage of Participants Continuing Treatment With Ixazomib at 12 Months From the Start of Study Treatment

(NCT03416374)
Timeframe: 12 months

InterventionPercentage of Participants (Number)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy50.0
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy35.9
[Overall]; Combination Therapy + Ixazomib Therapy37.8

[back to top]

Overall Survival (OS) From the Start of Study Treatment

OS was defined as the period from the first dose of treatment in Treatment Period I to the time when death (regardless of the cause of death) was confirmed. Participants who were still alive were censored at the last confirmed date of survival or the date of data cut-off, whichever was earlier. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionMonths (Median)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone TherapyNA
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone TherapyNA
[Overall]; Combination Therapy + Ixazomib TherapyNA

[back to top]

Overall Response Rate (ORR)

ORR is defined as the percentage of participants who achieve a best response of PR or better including stringent complete response (sCR), VGPR and PR assessed with IMWG Criteria, after the start of the study treatment. Per IMWG criteria, PR (partial response): ≥50% reduction of serum M protein+reduction in 24-hour urinary M protein by ≥90%/ to <200 mg/24-hour or ≥50% decrease in difference between involved and uninvolved free light chain (FLC) levels/ ≥50% reduction in bone marrow plasma cells, if ≥30% at baseline/ ≥50% reduction in size of soft tissue plasmacytomas. VGPR (very good PR): serum+urine M-protein detectable by immunofixation but not on electrophoresis/ ≥90% reduction in serum M-protein+urine Mprotein level <100 mg/24-hour. CR (complete response): negative immunofixation on serum+urine+disappearance of soft tissue plasmacytomas+<5% plasma cells in bone marrow. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionPercentage of Participants (Number)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy83.3
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy71.8
[Overall]; Combination Therapy + Ixazomib Therapy73.3

[back to top]

Number of Participants Reporting One or More Treatment-Emergent AEs (TEAEs)

An adverse event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. A TEAE is defined as an adverse event with an onset that occurs after receiving study drug. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionParticipants (Count of Participants)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy6
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy35
[Overall]; Combination Therapy + Ixazomib Therapy41

[back to top]

Healthcare Resource Utilization (HCRU): Number of Events With Hospitalization Per Participants-Month

HCRU was calculated from Exposure-adjusted rate of hospitalization events (per participants-months) and the duration of hospitalization among participants in Treatment Period I and Treatment Period II. Number of events with hospitalization per participants-month in Treatment Period I and Treatment Period II was reported. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionNumber of Events per Participants-Month (Number)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy1.9
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy2.9
[Overall]; Combination Therapy + Ixazomib Therapy2.7

[back to top]

Healthcare Resource Utilization (HCRU): Duration of Hospital Stay Per Participants

HCRU was calculated from Exposure-adjusted rate of hospitalization events (per participants-months) and the duration of hospitalization among participants in Treatment Period I and Treatment Period II. Duration of hospital stay per participants in Treatment Period I and Treatment Period II was reported. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionDays (Mean)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy14.8
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy19.4
[Overall]; Combination Therapy + Ixazomib Therapy18.8

[back to top]

Duration of Therapy (DOT)

DOT is defined as the treatment duration of study drug at study treatment Period I. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionMonths (Median)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy14.69
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy12.43
[Overall]; Combination Therapy + Ixazomib Therapy12.43

[back to top]

Duration of Response (DOR)

DOR is defined as the time from the date of first documentation of response ≥PR to the date of first documentation of PD or death due to any cause. PR and PD will be assessed with IMWG Criteria. Per IMWG criteria, PR (partial response): ≥50% reduction of serum M protein+reduction in 24-hour urinary M protein by ≥90%/ to <200 mg/24-hour or ≥50% decrease in difference between involved and uninvolved free light chain (FLC) levels/ ≥50% reduction in bone marrow plasma cells, if ≥30% at baseline/ ≥50% reduction in size of soft tissue plasmacytomas. VGPR (very good PR): serum+urine M-protein detectable by immunofixation but not on electrophoresis/ ≥90% reduction in serum M-protein+urine Mprotein level <100 mg/24-hour. CR (complete response): negative immunofixation on serum+urine+disappearance of soft tissue plasmacytomas+<5% plasma cells in bone marrow. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionMonths (Median)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy15.31
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy28.03
[Overall]; Combination Therapy + Ixazomib Therapy28.03

[back to top]

Evaluation of Modified Quality-Adjusted Life-Years (QALYs)

Modified QALYs was calculated from the score of EORTC QLQ-C30. The health-related quality of life scale score of EORTC QLQ-C30 was converted into a utility value ranging from 0 (dead) to 1 (perfect health), and used to adjust the value of survival years; this value was assessed as the modified QALY. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionQuality-Adjusted Life-Years (Median)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy0.467
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy0.534
[Overall]; Combination Therapy + Ixazomib Therapy0.518

[back to top]

Number of Participants With Minimal Residual Disease (MRD) Positive or Negative in Bone Marrow in Participants Who Achieved CR

MRD was measured by the flow cytometry method using bone marrow aspiration. Reported data were numbers of participants with MRD positive and negative in bone marrow in participants who achieved CR. MRD positive was categorized into three sensitivity levels with the numbers of cells counted (10^-4 to - Max; 10^-5 to 10^-4; 10^-6 to 10^-5). MRD negativity is defined as absence of MRD and MRD positivity is defined as presence of MRD. If a participant is MRD-positive at their first evaluation and MRD-negative after re-examination, the participant will be considered to be MRD-negative. CR will be assessed by IMWG Criteria. (NCT03416374)
Timeframe: Up to 39 months as a maximum

,,
InterventionParticipants (Count of Participants)
Sensitivity Level; 10^-4=< - MaxSensitivity Level; 10^-5=< - <10^-4Sensitivity Level; 10^-6=< - <10^-5Negative
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy1215
[Overall]; Combination Therapy + Ixazomib Therapy1325
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy0110

[back to top] [back to top]

Progression-Free Survival (PFS) Rate at 12 Months From the Start of Study Treatment

PFS rate was defined as the percentage of participants who were alive and have not had disease progression at 12 months after the date of first dose of treatment in Treatment Period I. PFS was assessed by International Myeloma Working Group (IMWG) Criteria (2014 version). Per IMWG criteria, progressive disease (PD): serum M-component increase ≥0.5 g/dl or urine M-component increase ≥200 mg/24-hour/ difference between involved and uninvolved free light chain (FLC) levels increase >10 mg/dl or bone marrow plasma cell ≥10%/ development of new/ increase in size of existing bone lesions or soft tissue plasmacytoma or development of hypercalcemia. (NCT03416374)
Timeframe: Up to 12 months

InterventionPercentage of Participants (Number)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy50.0
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy48.7
[Overall]; Combination Therapy + Ixazomib Therapy48.9

[back to top]

PFS From the Start of Study Treatment

PFS was defined as the period from the first dose of treatment in Treatment Period I to the time of confirmed PD or confirmed death (regardless of the cause of death), whichever is earlier. PFS was assessed by IMWG Criteria. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionMonths (Median)
[VRd]; Bortezomib + Lenalidomide + Dexamethasone TherapyNA
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy28.96
[Overall]; Combination Therapy + Ixazomib Therapy28.96

[back to top]

Percentage of Participants Who Achieve or Maintain Any Best Response

Best response is defined as the cumulative numbers of participants who achieve each level of best response including PR, VGPR and CR assessed with IMWG Criteria, after each cycle of treatment. Per IMWG criteria, PR (partial response): ≥50% reduction of serum M protein+reduction in 24-hour urinary M protein by ≥90%/ to <200 mg/24-hour or ≥50% decrease in difference between involved and uninvolved free light chain (FLC) levels/ ≥50% reduction in bone marrow plasma cells, if ≥30% at baseline/ ≥50% reduction in size of soft tissue plasmacytomas. VGPR (very good PR): serum+urine M-protein detectable by immunofixation but not on electrophoresis/ ≥90% reduction in serum M-protein+urine Mprotein level <100 mg/24-hour. CR (complete response): negative immunofixation on serum+urine+disappearance of soft tissue plasmacytomas+<5% plasma cells in bone marrow. (NCT03416374)
Timeframe: Up to 39 months as a maximum

,,
InterventionPercentage of Participants (Number)
CRVGPRPR
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy23.120.528.2
[Overall]; Combination Therapy + Ixazomib Therapy24.417.831.1
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy33.3050.0

[back to top]

Relative Dose Intensity (RDI)

RDI for each study drug is defined as 100*(Total amount of dose taken)/(Total prescribed dose of treated cycles), where total prescribed dose equals [dose prescribed at enrollment* number of prescribed doses per cycle* the number of treated cycles]. (NCT03416374)
Timeframe: Up to 39 months as a maximum

InterventionPercent (Median)
Bortezomib, IxazomibCarfilzomib, IxazomibBortezomib/Carfilzomib, IxazomibLenalidomideDexamethasone
[Overall]; Combination Therapy + Ixazomib Therapy71.9387.7083.2447.1848.61

[back to top]

Patient-Reported Outcome HRQoL Based on EORTC Multiple Myeloma Module (EORTC QLQ-MY20) Score

EORTC QLQ-MY20 has 20 items across 4 independent subscales, 2 functional subscales (body image, future perspective), and 2 symptoms scales (disease symptoms, and side effects of treatment). Scores are averaged, and transformed to 0-100 scale. For the functional scales, high scores represent improvement. For the symptom scales, higher scores represent worsening. (NCT03416374)
Timeframe: Baseline and End of Treatment (Up to 23 cycles for VRd Group, Up to 32 cycles for KRd and Overall Group, each cycle was of 28 days)

InterventionScore on a Scale (Mean)
Disease Symptoms: BaselineDisease Symptoms: Cycle 23Side-Effects of Treatment: BaselineSide-Effects of Treatment: Cycle 23Body Image: BaselineBody Image: Cycle 23Future Perspective: BaselineFuture Perspective: Cycle 23
[VRd]; Bortezomib + Lenalidomide + Dexamethasone Therapy24.07016.673.7044.44066.6711.11

[back to top]

Patient-Reported Outcome HRQoL Based on EORTC Multiple Myeloma Module (EORTC QLQ-MY20) Score

EORTC QLQ-MY20 has 20 items across 4 independent subscales, 2 functional subscales (body image, future perspective), and 2 symptoms scales (disease symptoms, and side effects of treatment). Scores are averaged, and transformed to 0-100 scale. For the functional scales, high scores represent improvement. For the symptom scales, higher scores represent worsening. (NCT03416374)
Timeframe: Baseline and End of Treatment (Up to 23 cycles for VRd Group, Up to 32 cycles for KRd and Overall Group, each cycle was of 28 days)

,
InterventionScore on a Scale (Mean)
Disease Symptoms: BaselineDisease Symptoms: Cycle 23Disease Symptoms: Cycle 32Side-Effects of Treatment: BaselineSide-Effects of Treatment: Cycle 23Side-Effects of Treatment: Cycle 32Body Image: BaselineBody Image: Cycle 23Body Image: Cycle 32Future Perspective: BaselineFuture Perspective: Cycle 23Future Perspective: Cycle 32
[KRd]; Carfilzomib + Lenalidomide + Dexamethasone Therapy19.234.632.7816.6016.0518.5223.0816.6716.6744.1635.1927.78
[Overall]; Combination Therapy + Ixazomib Therapy19.883.972.7816.6114.2918.5225.9314.2916.6747.1631.7527.78

[back to top] [back to top] [back to top]

Number of Participants Reporting One or More Treatment-Emergent AEs (TEAEs)

An adverse event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. A TEAE is defined as an adverse event with an onset that occurs after receiving study drug. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionParticipants (Count of Participants)
Ixazomib + Lenalidomide + Dexamethasone249

[back to top]

Overall Response Rate (ORR)

ORR is defined as the percentage of participants who achieve a best response of PR or better including stringent complete response (sCR), VGPR and PR assessed with IMWG Criteria, after the start of the study treatment. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionPercentage of Participants (Number)
Ixazomib + Lenalidomide + Dexamethasone53.9

[back to top]

Overall Survival (OS)

OS is defined as the period from the start of IRd therapy in standard medical care to the time when death (regardless of the cause of death) is confirmed. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionMonths (Median)
Ixazomib + Lenalidomide + Dexamethasone20.23

[back to top]

Percentage of Participants Who Achieve VGPR or Better (CR+VGPR)

The percentage of participants of CR + VGPR is defined as the rate of participants who achieve a best response of VGPR or better (sCR, CR, or VGPR) according to the IMWG Criteria after the start of the IRd therapy. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionPercentage of Participants (Number)
Ixazomib + Lenalidomide + Dexamethasone31.5

[back to top]

Percentage of Participants With Bone Lesions (Bone Evaluation)

(NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionPercentage of Participants (Number)
Ixazomib + Lenalidomide + Dexamethasone21.5

[back to top] [back to top] [back to top]

Relative Dose Intensity (RDI)

RDI is defined as 100*(Total amount of dose taken)/(Total prescribed dose of treated cycles), where total prescribed dose equals [dose prescribed at enrollment* number of prescribed doses per cycle* the number of treated cycles]. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionPercent (Mean)
IxazomibLenalidomideDexamethasone
Ixazomib + Lenalidomide + Dexamethasone66.4944.7241.07

[back to top]

Percentage of Participants Who Achieve or Maintain Any Best Response

Best response is defined as the cumulative numbers of participants who achieve each level of best response including partial response (PR), very good PR (VGPR) and complete response (CR) assessed with IMWG Criteria after each cycle of treatment. Per IMWG criteria, PR: ≥50% reduction of serum M protein+reduction in 24-hour urinary M protein by ≥90%/ to <200 mg/24-hour or ≥50% decrease in difference between involved and uninvolved free light chain (FLC) levels/ ≥50% reduction in bone marrow plasma cells, if ≥30% at baseline/ ≥50% reduction in size of soft tissue plasmacytomas. VGPR: serum+urine M-protein detectable by immunofixation but not on electrophoresis/ ≥90% reduction in serum M-protein+urine M-protein level <100 mg/24-hour. CR: negative immunofixation on serum+urine +disappearance of soft tissue plasmacytomas+<5% plasma cells in bone marrow. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionPercentage of Participants (Number)
CRVGPRPR
Ixazomib + Lenalidomide + Dexamethasone20.08.522.0

[back to top]

Percentage of Participants Who Continue to Receive Treatment at 12 Months and 24 Months After Start of Treatment

(NCT03433001)
Timeframe: 12 months and 24 months

InterventionPercentage of Participants (Number)
Month 12Month 24
Ixazomib + Lenalidomide + Dexamethasone40.021.7

[back to top]

Progression-Free Survival (PFS)

PFS was defined as the period from the start of ixazomib, lenalidomide, dexamethasone (IRd) therapy in standard medical care to the time of confirmed progressive disease (PD) or confirmed death (regardless of the cause of death), whichever was earlier. PFS was assessed by International Myeloma Working Group (IMWG) Criteria (2014 version). Per IMWG criteria, PD: serum M-component increase ≥ 0.5 g/dl or urine M-component increase ≥ 200 mg/24-hour/ difference between involved and uninvolved free light chain (FLC) levels increase >10 mg/dl or bone marrow plasma cell ≥ 10%/ development of new/ increase in size of existing bone lesions or soft tissue plasmacytoma or development of hypercalcemia. (NCT03433001)
Timeframe: Up to 36 Months as a maximum

InterventionMonths (Median)
Ixazomib + Lenalidomide + Dexamethasone4.79

[back to top]

Patient-Reported Outcome HRQoL Based on EORTC Multiple Myeloma Module (EORTC QLQ-MY20) Score

EORTC QLQ-MY20 has 20 items across 4 independent subscales, 2 functional subscales (body image, future perspective), and 2 symptoms scales (disease symptoms, and side effects of treatment). Scores are averaged, and transformed to 0-100 scale. For the future perspective scale, higher score = better perspective of the future. For the body image scale, higher scores = better body image. Higher score for the disease symptoms scale = higher level of symptomatology. (NCT03433001)
Timeframe: Baseline and End of Treatment (Up to 37 cycles, each cycle was of 28 days)

InterventionScore on a Scale (Mean)
Disease Symptoms: BaselineDisease Symptoms: End of TreatmentSide-Effects of Treatment: BaselineSide-Effects of Treatment: End of TreatmentBody Image: BaselineBody Image: End of TreatmentFuture Perspective: BaselineFuture Perspective: End of Treatment
Ixazomib + Lenalidomide + Dexamethasone19.3311.1117.8016.6728.1816.6742.4933.33

[back to top]

Duration of Therapy (DOT)

DOT is defined as the treatment duration of IRd therapy. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionDays (Mean)
Ixazomib + Lenalidomide + Dexamethasone353.3

[back to top]

Rate of Minimal Residual Disease (MRD) Negativity in Bone Marrow in Participants Who Achieved CR

Rate of MRD will be calculated by the percentage of participants who are MRD-negative. (NCT03433001)
Timeframe: Up to 36 months as a maximum

InterventionPercentage of Participants (Number)
10^-4=< - Max10^-5=< - <10^-410^-6=< - <10^-5Negative
Ixazomib + Lenalidomide + Dexamethasone26.716.76.750.0

[back to top]

PFS Rate at 12 Months and 24 Months After the Start of Treatment

PFS rate was defined as the percentage of participants who were alive and have not had disease progression at 12 months and 24 months after the date of start of study treatment. PFS was assessed by IMWG Criteria (2014 version). Per IMWG criteria, PD: serum M-component increase ≥ 0.5 g/dl or urine M-component increase ≥ 200 mg/24-hour/ difference between involved and uninvolved FLC levels increase >10 mg/dl or bone marrow plasma cell ≥ 10%/ development of new/ increase in size of existing bone lesions or soft tissue plasmacytoma or development of hypercalcemia. (NCT03433001)
Timeframe: 12 months and 24 months

InterventionPercentage of Participants (Number)
Month 12Month 24
Ixazomib + Lenalidomide + Dexamethasone5741

[back to top]

Number of Participants Who Achieved Complete Response (CR), Very Good Partial Response (VGPR), and Partial Response (PR)

Number of participants who achieved CR, PR, VGPR were assessed in accordance to International Myeloma Working Group Uniform Response Criteria for Multiple Myeloma. CR: No immunofixation on serum and urine, disappearance of soft tissue plasmacytomas and <5% plasma cells in bone marrow. Normal serum free light chain (SFLC) ratio if disease measurable only by SFLC. VGPR: Serum and urine M-protein detectable by immunofixation but not electrophoresis or greater than or equal to (>=) 90% decrease in serum M-protein with urine M-protein <100 milligram per 24 hours (mg/24 hrs). If disease measurable only by SFLC, >= 90% decrease in the difference between involved and uninvolved FLC levels (dFLC). PR: >= 50% reduction of serum M-protein and >= 90% reduction in urine M-protein or to <200 mg/24 hrs, or a >= 50% decrease in dFLC. A >=50% decrease in the size of soft tissue plasmacytomas present at baseline (NCT04272775)
Timeframe: Baseline up to 29 days after last dose of study drug (up to Cycle 87 Day 44) (Each Cycle length=28 days)

InterventionParticipants (Count of Participants)
Cohort 1: Ixazomib 4.0 mg0
Cohort 2: Ixazomib 4.0 mg + Lenalidomide and Dexamethasone2

[back to top]

Number of Participants Who Experienced at Least One Treatment-emergent Adverse Event (TEAE)

(NCT04272775)
Timeframe: Baseline up to 29 days after last dose of study drug (up to Cycle 87 Day 44) (Each Cycle length=28 days)

InterventionParticipants (Count of Participants)
Cohort 1: Ixazomib 4.0 mg7
Cohort 2: Ixazomib 4.0 mg + Lenalidomide and Dexamethasone7

[back to top]

Number of Participants Who Experienced Dose Limiting Toxicities (DLTs)

DLT:Any following adverse events (AEs) possibly related to ixazomib assessed by Common Terminology Criteria for AEs (CTCAE) version 4.03; Grade 4 neutropenia/thrombocytopenia lasting >7 consecutive days; Grade 3/greater neutropenia with fever/infections; Grade 3/greater thrombocytopenia with clinically significant bleeding; platelet count less than (<)10,000 per cubic meter(/mm^3); Grade 2 peripheral neuropathy with pain/Grade 3 or greater peripheral neuropathy; Grade 3/greater nonhematologic toxicities with exceptions of arthralgia/myalgia, fatigue lasting <7 days manageable nausea/emesis with antiemetic prophylaxis, diarrhea that is controlled with supportive care; treatment delay of >14 days at start of Cycle 2 due to failure of hematologic/nonhematologic recovery; Other Grade 2/greater ixazomib related nonhematologic toxicities required permanent discontinuation of ixazomib;Inability to receive at least 80% of planned lenalidomide doses due to the AEs related to ixazomib. (NCT04272775)
Timeframe: From Cycle 1 Day 1 until Cycle 2 Day 1 (Cycle length is equal to [=] 28 days)

InterventionParticipants (Count of Participants)
Cohort 1: Ixazomib 4.0 mg1
Cohort 2: Ixazomib 4.0 mg + Lenalidomide and Dexamethasone1

[back to top]

Number of Participants With Grade 3 or Higher Laboratory Tests Abnormalities

Laboratory tests abnormalities were graded using the CTCAE version 4.03. as: Grade 1 (Mild, asymptomatic or mild symptoms); Grade 2 (Moderate, minimal, local or noninvasive intervention indicated); Grade 3 (Severe or medically significant but not immediately life-threatening, hospitalization or prolongation of hospitalization indicated); Grade 4 (Life-threatening consequences, urgent intervention indicated); Grade 5 (Death related to AE). (NCT04272775)
Timeframe: Baseline up to 29 days after last dose of study drug (up to Cycle 87 Day 44) (Each Cycle length=28 days)

,
InterventionParticipants (Count of Participants)
Grade 4: Neutrophils lowGrade 4: Leukocytes lowGrade 4: Lymphocytes lowGrade 4: Platelets lowGrade 4: Sodium lowGrade 4: Potassium lowGrade 3: Neutrophils lowGrade 3: Hemoglobin lowGrade 3: Leukocytes lowGrade 3: Lymphocytes lowGrade 3: Platelets lowGrade 3: Albumin lowGrade 3: Corrected calcium highGrade 3: Sodium lowGrade 3: Potassium lowGrade 3: Phosphate low
Cohort 1: Ixazomib 4.0 mg0033004233010111
Cohort 2: Ixazomib 4.0 mg + Lenalidomide and Dexamethasone2102112223211001

[back to top]

Cmax: Maximum Observed Plasma Concentration for Ixazomib

(NCT04272775)
Timeframe: Days 1 and 15 of Cycle 1: pre-dose and at multiple time points (15, 30, 60, and 90 minutes and 2, 4, 8, 24, 48, 96, and 168 hours) post-dose (Cycle length=28 days)

,
Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Day 1Day 15
Cohort 1: Ixazomib 4.0 mg65.368.8
Cohort 2: Ixazomib 4.0 mg + Lenalidomide and Dexamethasone32.934.5

[back to top] [back to top] [back to top] [back to top]

Overall Response Rate After 4 Cycles Induction Therapy

"Overall response rate will be the percentage of patients who achieve at least partial response (PR) per International Myeloma Working Group (IMWG) Uniform Response criteria after 4 cycles of of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI).~The IMWG response criteria define a PR as:~> 50% reduction of serum M-protein and reduction in 24 hours urinary M-protein by >90% or to < 200 mg/24 h, OR If the serum and urine M-protein are unmeasurable, a > 50% decrease in the difference between involved and uninvolved FLC levels in place of the M-protein criteria, OR If serum and urine M-protein are not measurable, and serum free light assay is also not measurable, > 50% reduction in plasma cells is required in place of M-protein, provided baseline bone marrow plasma cell percentage was > 30%.~In addition to the above listed criteria, if present at baseline, a > 50% reduction in the size of soft tissue plasmacytomas is also required for a partial response." (NCT04430894)
Timeframe: 112 days (4 cycles)

InterventionParticipants (Count of Participants)
Induction47

[back to top]

Minimal Residual Disease (MRD) Rate at 10^(-5) in Patients Receiving Upfront Stem Cell Transplant

MRD is the small number myeloma cells surviving in the bone marrow after a clinical response has been measured and the patient is in remission. MRD negativity will be determined by deep sequencing of genomic deoxyribonucleic acid from bone marrow samples using rearranged Ig locus-specific primers (Adaptive, Seattle, WA). The MRD rate is the percentage of patients who are MRD negative at 10^(-5), meaning that no MRD was detected in a sample of 100,000 cells. MRD status was assessed after 4 cycles of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI) induction, stem cell transplant, and 2 cycles of KRDI consolidation therapy (6 cycles total.) (NCT04430894)
Timeframe: 168 Days (6 cycles, upfront transplant)

Interventionpercentage of participants (Number)
Upfront Stem Cell Transplant (4 Cycles Induction Therapy, SCT, and 2 Cycles Consolidation Therapy)20

[back to top]

Minimal Residual Disease (MRD) Rate at 10^(-5) in Patients Deferring Stem Cell Transplant

MRD is the small number myeloma cells surviving in the bone marrow after a clinical response has been measured and the patient is in remission. MRD negativity will be determined by deep sequencing of genomic deoxyribonucleic acid from bone marrow samples using rearranged Ig locus-specific primers (Adaptive, Seattle, WA). The MRD rate is the percentage of patients who are MRD negative at 10^(-5), meaning that no MRD was detected in a sample of 100,000 cells. MRD status was assessed after 4 cycles of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI) induction, stem cell collection, and 4 additional cycles of KRDI (8 cycles total). (NCT04430894)
Timeframe: 224 Days (8 cycles transplant-deferred)

InterventionParticipants (Count of Participants)
Deferred Stem Cell Transplant (4 Cycles, Stem Cell Collection, 4 Additional Cycles)26

[back to top]

Complete Response Rate in Patients With Upfront Stem Cell Transplant

"Percentage of patients who achieved a complete response (CR) or stringent CR (sCR) per International Myeloma Working Group (IMWG) Uniform Response criteria after 4 cycles of of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI), stem cell transplant, and 2 cycles of KRDI consolidation (6 cycles total). IMWG criteria define a CR as Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow and an sCR as CR plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence." (NCT04430894)
Timeframe: 168 Days

Interventionpercentage of participants (Number)
Upfront Stem Cell Transplant (4 Cycles Induction Therapy, SCT, and 2 Cycles Consolidation Therapy)80

[back to top]

Complete Response (CR + Stringent CR) Rate

"Percentage of patients who achieved a complete response (CR) or stringent CR (sCR) per International Myeloma Working Group (IMWG) Uniform Response criteria after 4 cycles of of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI). IMWG criteria define a CR as Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow and an sCR as CR plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence." (NCT04430894)
Timeframe: 112 Days

InterventionParticipants (Count of Participants)
Induction19

[back to top]

Overall Survival at 12 Months

Overall survival (OS) is defined as the time between the start of treatment and death due to any cause. OS at 12 months is the percentage of patients who were alive 12 months after initiating treatment with of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI). (NCT04430894)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Induction97.9

[back to top]

Progression Free Survival at 12 Months

Progression free survival (PFS) is defined as the time between the start of treatment and disease progression or death due to any cause. PFS at 12 months is the percentage of patients who were alive and without disease progression 12 months after initiating treatment with of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI). (NCT04430894)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Induction97.9

[back to top]

Minimal Residual Disease (MRD) Rate After 4 Cycles Induction Therapy

MRD is the small number myeloma cells surviving in the bone marrow after a clinical response has been measured and the patient is in remission. MRD negativity will be determined by deep sequencing of genomic deoxyribonucleic acid from bone marrow samples using rearranged Ig locus-specific primers (Adaptive, Seattle, WA). The MRD rate after 4 cycles of of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI) is the percentage of patients who are MRD negative at 10^(-5) and 10^(-6), meaning that no MRD was detected in samples of 100,000 cells and 1,000,000 cells, respectively. (NCT04430894)
Timeframe: 112 Days (4 Cycles)

InterventionParticipants (Count of Participants)
MRD negative at 10^(-5)MRD negative at 10^(-6)
Induction129

[back to top]

Complete Response Rate in Patients Deferring Stem Cell Transplant

"Percentage of patients who achieved a complete response (CR) or stringent CR (sCR) per International Myeloma Working Group (IMWG) Uniform Response criteria after 4 cycles of of Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone (KRDI), stem cell collection, and 4 additional cycles of KRDI after deferring stem cell transplant (8 cycles total). IMWG criteria define a CR as Negative immunofixation on the serum and urine and disappearance of any soft tissue plasmacytomas and < 5% plasma cells in bone marrow and an sCR as CR plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence." (NCT04430894)
Timeframe: 224 Days

Interventionpercentage of participants (Number)
Deferred Stem Cell Transplant (4 Cycles, Stem Cell Collection, 4 Additional Cycles)67

[back to top]