Page last updated: 2024-11-04

temozolomide

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 CID5394
CHEMBL ID810
CHEBI ID72564
SCHEMBL ID3739
MeSH IDM0136562

Synonyms (149)

Synonym
AC-758
BIDD:GT0204
3-methyl-4-oxo-3,4-dihydro-imidazo[5,1-d][1,2,3,5]tetrazine-8-carboxylic acid amide(temozolomide)
3-methyl-4-oxo-3,4-dihydro-imidazo[5,1-d][1,2,3,5]tetrazine-8-carboxylic acid amide
bdbm50034562
AKOS005557098
HMS3269P05
HMS3393O12
AB00639915-08
AB00639915-09
temozolomide [usan:inn:ban]
temozolodida
yf1k15m17y ,
temozolomidum
unii-yf1k15m17y
3-methyl-4-oxo-imidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide
3-methyl-4-oxo-3,4-dihydroimidazo[5,1-d][1,2,3,5]tetraazine-8-carboxamide
{imidazo[5,1-d]-1,2,3,5-tetrazine-8-carboxamide,} 3, 4-dihydro-3-methyl-4-oxo-
NCI60_003316
sch-52365
mls002701861 ,
nsc362856
temozolomide ,
nsc-362856
temodar (tn) (schering)
85622-93-1
imidazo[5,2,3,5-tetrazine-8-carboxamide, 3,4-dihydro-3-methyl-4-oxo-
methazolastone
temozolomidum [latin]
ccrg 81045
temodal
mb 39831
imidazo(5,1-d)(1,2,3,5)tetrazine-8-carboxamide, 3,4-dihydro-3-methyl-4-oxo-
m&b 39831
3,4-dihydro-3-methyl-4-oxoimidazo(5,1-d)-as-tetrazine-8-carboxamide
temozolodida [spanish]
nsc 362856
3-methyl-4-oxo-3,4-dihydroimidazo(5,1-d)(1,2,3,5)tetrazine-8-carboxamide
c6h6n6o2
sch 52365
brn 5547136
ccrg-81045
m&b-39831
m & b 39831
temodar
ccris 8996
8-carbamoyl-3-methylimidazo(5,1-d)-1,2,3,5-tetrazin-4(3h)-one
3,4-dihydro-3-methyl-4-oxoimidazo(5,1-d)-1,2,3,5-tetrazine-8-carboxamide
temozolomide (jan/usan/inn)
D06067
temodar (tn)
temodal (tn)
MLS001424028
tmz ,
temozolamide
smr000466338
MLS000759447
DB00853
NCGC00167429-02
NCGC00167429-01
3-methyl-4-oxo-3,4-dihydroimidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide ,
HMS2090B08
HMS2051O12
CHEMBL810 ,
mk-7365
chebi:72564 ,
m-39831
3-methyl-4-oxoimidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide
STK623541
3-methyl-4-oxo-8-imidazo[5,1-d][1,2,3,5]tetrazinecarboxamide
A841386
3-methyl-4-oxidanylidene-imidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide
HMS3264I14
4-methyl-5-oxo-2,3,4,6,8-pentazabicyclo[4.3.0]nona-2,7,9-triene-9-carboxamide
cas-85622-93-1
tox21_112433
dtxsid5043714 ,
dtxcid3023714
3,4-dihydro-3-methyl-4-oxoimidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide
T2744
nsc-759883
pharmakon1600-01502289
nsc759883
temozolomida
HMS2232N13
CCG-100870
HY-17364
FT-0674845
BCP0726000154
FT-0630936
NCGC00167429-05
AM20110227
CS-0943
S1237
3-methyl-4-oxo-3h,4h-imidazo[4,3-d][1,2,3,5]tetrazine-8-carboxamide
gtpl7301
HMS3372K13
temozolomide [ema epar]
temozolomide [usp-rs]
temozolomide [usp monograph]
temozolomide [mart.]
temozolomide [ep monograph]
temozolomide [mi]
temozolomide [who-dd]
temozolomide [inn]
temozolomide [jan]
3,4-dihydro-3-methyl-4-oxoimidazo[5,1-d]-as-tetrazine-8-carboxamide
temozolomide [orange book]
temozolomide [vandf]
temozolomide [usan]
DL-190
AB00639915-06
BPEGJWRSRHCHSN-UHFFFAOYSA-N
3-methyl-4-oxo-3,4-dihydro-imidazo[5,1-d][1,2,3,5]tetrazine-8-carboxylic acid amide (temozolomide)
3-methyl-8-aminocarbonyl-imidazo[5,1-d]-1,2,3,5-tetrazin-4(3h)-one
NC00120
SCHEMBL3739
tox21_112433_1
NCGC00167429-04
KS-1216
Q-201786
3,4-dihydro-3-methyl-4-oxoimidazo[5,1-d]-1,2,3,5-tetrazine-8-carboxamide
AB00639915_11
AB00639915_10
8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4(3h)-one
mfcd00866492
SR-01000759347-5
sr-01000759347
SR-01000759347-4
temozolomide, united states pharmacopeia (usp) reference standard
temozolomide, vetranal(tm), analytical standard
HMS3654N05
temozolomide, >=98% (hplc)
temozolomide, pharmaceutical secondary standard; certified reference material
HMS3713H16
3-methyl-4-oxo-3,4-dihydroimidazo-[5,1-d][1,2,3,5]tetrazine-8-carboxamide
SW197500-4
BCP03692
3-methyl-4-oxo-3,4-dihydroimidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide.
HMS3677D06
HMS3413D06
Q425088
BRD-K32107296-001-04-5
EN300-122324
temozolomide 100 microg/ml in acetonitrile
BP-25388
BT164447
temozolomide- bio-x
Z1201620684

Research Excerpts

Overview

Temozolomide (TMZ) is a chemotherapeutic agent that is used in the treatment of glioblastoma and other malignant gliomas. The occurrence of drug resistance limits its antitumor activity.

ExcerptReferenceRelevance
"Temozolomide is a chemotherapeutic agent that is used in the treatment of glioblastoma and other malignant gliomas. "( Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
Banerjee, M; Bernales, S; Chakravarty, S; Dixit, V; Guerrero, J; Gupta, A; Hung, DT; Jangir, R; McCullagh, E; Melkani, P; Middya, S; Olivares, F; Patil, U; Pham, SM; Protter, AA; Rai, R; Raja, M; Riquelme, E; Shrivastava, R; Surya, A; Tripathi, S; Wong, DH; Yadav, S, 2016
)
3.32
"Temozolomide (TMZ) is a first-line chemotherapeutic agent used in GBM therapy, but the occurrence of drug resistance limits its antitumor activity."( Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
Chang, JT; Chang, KF; Hsiao, CY; Huang, XF; Huang, YC; Lo, WS; Tsai, NM, 2020
)
1.51
"Temozolomide is an oral alkylating agent incorporated in the treatment of glioblastoma multiforme (GBM) that can lead to lymphopenia. "( Temozolomide is a risk factor for invasive pulmonary aspergillosis: A case report and literature review.
Brault, C; Chouaki, T; Maizel, J; Nyga, R; Zerbib, Y, 2021
)
3.51
"Temozolomide (TMZ) is a recommended treatment option, but its effects are difficult to predict, and the alternatives are limited."( Efficacy of temozolomide combined with capecitabine (CAPTEM) on refractory prolactinomas as assessed using an ex vivo 3D spheroid assay.
Fukuoka, H; Inoshita, N; Ishida, A; Ogawa, W; Shichi, H; Yamada, S, 2022
)
1.82
"Temozolomide is an alkylating agent most commonly used with a few other second line options."( Nanomedicine in the treatment of Glioblastoma.
Bukhari, SS; Kishwar Jafri, SK; Shamim, MS, 2021
)
1.34
"Temozolomide (TMZ) is a prodrug of 5-(3-methyltriazene-1-yl)imidazole-4-carboxamide (MTIC, short-lived) and used as a first-line therapy drug for glioblastoma multiforme (GBM). "( Visible Light and Glutathione Dually Responsive Delivery of a Polymer-Conjugated Temozolomide Intermediate for Glioblastoma Chemotherapy.
Du, K; Feng, F; Sun, J; Xia, Q, 2021
)
2.29
"Temozolomide is an oral alkylating agent used as first line treatment of glioblastoma multiforme (GBM). "( Drug-induced hypersensitivity syndrome following temozolimide for glioblastoma multiforme and the role of desensitization therapy.
Ambur, A; Ambur, L; Khan, L; Nathoo, R, 2022
)
2.16
"Temozolomide (TMZ) is a commonly used drug for GBM management."( Extracellular vesicles carry miR-27a-3p to promote drug resistance of glioblastoma to temozolomide by targeting BTG2.
Chen, L; Deng, Q; Guo, S; Hao, P; Hu, S; Li, Z, 2022
)
1.67
"Temozolomide (TMZ) is a standard-of-care chemotherapeutic drug for the treatment of glioblastoma (GBM), but TMZ-acquired resistance limits its therapeutic effect. "( Efficacy of Temozolomide-Conjugated Gold Nanoparticle Photothermal Therapy of Drug-Resistant Glioblastoma and Its Mechanism Study.
Chu, L; Liu, X; Sha, C; Sun, K; Sun, Y; Wang, A; Wang, S; Xu, L; Yang, X; Yu, Y; Zhou, L, 2022
)
2.54
"Temozolomide (TMZ) is a first-line chemotherapeutic agent for the treatment of glioma. "( The RNA-binding protein fragile-X mental retardation autosomal 1 (FXR1) modulates glioma cells sensitivity to temozolomide by regulating ferroptosis.
Duan, S; Gong, F; Li, Q; Wei, Y, 2022
)
2.38
"Temozolomide (TMZ) is a DNA alkylating agent that appears to have a radiosensitizing effect when used in combination with RT and may be worthwhile in meningioma treatment."( Concomitant Temozolomide plus radiotherapy for high-grade and recurrent meningioma: a retrospective chart review.
Belanger, K; Damek, D; Lillehei, KO; Ormond, DR; Ung, TH, 2022
)
1.82
"Temozolomide (TMZ) is a chemotherapeutic agent used in glioblastoma and melanoma treatment."( DNA polymerase eta protects human cells against DNA damage induced by the tumor chemotherapeutic temozolomide.
Bastos, AU; de Souza, I; Hoch, NC; Latancia, MT; Leandro, GS; Menck, CFM; Moreno, NC; Ribeiro, VC; Rocha, CRR; Vieira, WKM, 2022
)
1.66
"Temozolomide (TMZ) is an imidazotetrazine prodrug used to treat glioblastoma multiforme. "( Temozolomide: An Overview of Biological Properties, Drug Delivery Nanosystems, and Analytical Methods.
Carvalho, SG; Chorilli, M; Di Filippo, LD; Dutra, JAP; Luiz, MT; Tavares Junior, AG, 2022
)
3.61
"Temozolomide (TMZ) is a widely used chemotherapeutic drug for glioma."( FOXM1-mediated NUF2 expression confers temozolomide resistance to human glioma cells by regulating autophagy via the PI3K/AKT/mTOR signaling pathway.
Guo, L; Wu, Z, 2022
)
1.71
"Temozolomide (TMZ) is a chemotherapeutic agent that has been the first-line standard of care for the aggressive brain cancer glioblastoma (GBM) since 2005. "( Temozolomide-induced guanine mutations create exploitable vulnerabilities of guanine-rich DNA and RNA regions in drug-resistant gliomas.
Alamillo-Ferrer, C; Cheng, SY; Drewry, DH; Erdogdu, B; Goenka, A; Goldlust, SA; Haddad, BR; Hogg, JR; Hu, B; Jin, L; Pertea, M; Pickett, JE; Razaghi, R; Riggins, RB; Sadowski, N; Song, X; Tiek, DM; Timp, W; Wells, CI; Zuercher, WJ, 2022
)
3.61
"Temozolomide (TMZ) is a chemotherapeutic drug for the treatment of GBM."( Temozolomide increases heat shock proteins in extracellular vesicles released from glioblastoma cells.
Adıgüzel, Z; Kıyga, E; Önay Uçar, E, 2022
)
2.89
"Temozolomide (TMZ) is a first-line chemotherapeutic agent for glioblastoma, but the emergence of drug resistance limits its anti-tumor activity."( GBP3 promotes glioblastoma resistance to temozolomide by enhancing DNA damage repair.
Chen, CC; Chen, Y; Grigore, FN; Jin, J; Lan, Q; Li, M; Li, S; Ma, J; Wang, J; Wang, Q; Wu, G; Xu, H; Zhu, H, 2022
)
1.71
"Temozolomide (TMZ) is an auxiliary alkylating agent that is extensively used in conjunction with surgical resection and forms the mainstay of clinical treatment strategies for gliomas."( Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review).
Du, J; Li, S; Peng, C; Peng, F; Xie, X, 2022
)
1.8
"Temozolomide (TMZ) is a first-line clinical chemotherapeutic drug."( Piperlongumine-inhibited TRIM14 signaling sensitizes glioblastoma cells to temozolomide treatment.
Chen, KC; Chen, PH; Ho, KH; Kuo, YY; Liu, AJ; Shih, CM, 2022
)
1.67
"Temozolomide (TMZ) is a conventional chemotherapeutic drug for glioma, however, its clinical application and efficacy is severely restricted by its drug resistance properties. "( The nanoprodrug of polytemozolomide combines with MGMT siRNA to enhance the effect of temozolomide in glioma.
Li, L; Liu, H; Ma, H; Qian, F; Quan, A; Ren, Y; Wang, L; Xu, H; Yu, R; Zhang, Y, 2023
)
2.66
"Temozolomide is an oral alkylating agent that is used as the first line treatment for glioblastoma multiform, and in recurrent anaplastic astrocytoma, as well as having demonstrable activity in patients with metastatic melanoma. "( Is Autophagy Inhibition in Combination with Temozolomide a Therapeutically Viable Strategy?
Elshazly, AM; Gewirtz, DA, 2023
)
2.61
"Temozolomide (TMZ) is a first line agent used in the clinic for glioblastoma and it has been useful in increasing patient survival rates."( Efficient delivery of Temozolomide using ultrasmall large-pore silica nanoparticles for glioblastoma.
Ahmed-Cox, A; Akhter, DT; Cao, Y; Fletcher, NL; Janjua, TI; Kavallaris, M; Moniruzzaman, M; Popat, A; Raza, A; Thurecht, KJ, 2023
)
1.95
"Temozolomide (TMZ) is a chemotherapy agent used to treat primary central nervous system tumors."( Metastatic Melanoma: A Preclinical Model Standardization and Development of a Chitosan-Coated Nanoemulsion Containing Temozolomide to Treat Brain Metastasis.
Azambuja, JH; Braganhol, E; de Cássia Sant'ana, R; de Souza, PO; Debom, GN; Fachel, FNS; Gelsleichter, NE; Lenz, GS; Michels, LR; Roliano, GG; Teixeira, FC; Teixeira, HF; Visioli, F, 2023
)
1.84
"Temozolomide (TMZ) is a DNA alkylating agent that can cross the blood-brain barrier."( Expert opinion on translational research for advanced glioblastoma treatment.
Cui, X; Kang, C; Wang, Q; Wang, Y; Zhou, J, 2023
)
1.63
"Temozolomide (TMZ) is a common chemotherapy drug used to treatment of glioblastoma, but drug resistance against this drug is an important barrier to successful treatment of this cancer."( Combination of SIX4-siRNA and temozolomide inhibits the growth and migration of A-172 glioblastoma cancer cells.
Baghbanzadeh, A; Baradaran, B; Barpour, N; Doustvandi, MA; Javadrashid, D; Mohammadpour, ZJ; Mohammadzadeh, R, 2023
)
1.92
"Temozolomide (TMZ) is a standard chemotherapeutic for GBM, but TMZ treatment benefits are compromised by chemoresistance."( Identification of the E2F1-RAD51AP1 axis as a key factor in MGMT-methylated GBM TMZ resistance.
Cui, X; Kang, C; Liu, X; Tong, F; Wang, G; Wang, Q; Wang, Y; Zhao, J; Zhou, J, 2023
)
1.63
"Temozolomide (TMZ) is a common alkylating chemotherapeutic agent used to treat brain tumors such as glioblastoma multiforme (GBM) and anaplastic astrocytoma. "( LncRNA-associated competing endogenous RNA network analysis uncovered key lncRNAs involved in temozolomide resistance and tumor recurrence of glioblastoma.
Mallick, B; Nayak, R, 2023
)
2.57
"Temozolomide (TMZ) is a standard treatment for glioblastoma (GBM) patients. "( Hypoxanthine phosphoribosyl transferase 1 metabolizes temozolomide to activate AMPK for driving chemoresistance of glioblastomas.
Agnihotri, S; Cao, Y; Chen, D; Ding, F; Ge, X; Ge, Z; Huang, G; Ji, J; Lin, F; Lu, Z; Qian, X; Shi, Z; Wang, Q; Wang, X; Yin, J; You, Y; Zhang, J; Zhao, N; Zhou, Q, 2023
)
2.6
"Temozolomide (TMZ) is a commonly used chemotherapeutic agent for glioblastoma (GBM), but acquired drug resistance prevents its therapeutic efficacy. "( NFYB increases chemosensitivity in glioblastoma by promoting HDAC5-mediated transcriptional inhibition of SHMT2.
Huang, H; Liu, P; Xie, Y; Zhang, Y, 2023
)
2.35
"Temozolomide (TMZ) is an important first-line treatment for glioblastoma (GBM), but there are limitations to TMZ response in terms of durability and dependence on the promoter methylation status of the DNA repair gene O"( Epigenetic Activation of TUSC3 Sensitizes Glioblastoma to Temozolomide Independent of MGMT Promoter Methylation Status.
Berglund, AE; Etame, AB; Macaulay, RJ; Wu, Q, 2023
)
2.6
"Temozolomide (TMZ) is a first-line chemotherapeutic agent used against glioblastoma multiforme (GBM), but this disease exhibits recurrence and high lethality. "( miR-140 targeting CTSB signaling suppresses the mesenchymal transition and enhances temozolomide cytotoxicity in glioblastoma multiforme.
Chen, KC; Chen, PH; Cheng, CH; Chou, CM; Ho, KH; Lin, CW; Liu, AJ; Shih, CM, 2019
)
2.18
"Temozolomide is an alkylating agent used as the first line of treatment for glioblastoma. "( Temozolomide has anti-tumor effects through the phosphorylation of cPLA
Hara, H; Iwama, T; Nakamura, S; Noda, Y; Ohno, Y; Saio, M; Shimazawa, M; Tsuji, S; Yamada, T, 2019
)
3.4
"Temozolomide (TMZ) is a chemotherapy agent used to treat Grade IV astrocytoma, also known as glioblastoma (GBM). "( Simvastatin increases temozolomide-induced cell death by targeting the fusion of autophagosomes and lysosomes.
Aghaei, M; Akbari, M; Albokashy, M; Alizadeh, J; Butterfield, Y; Cole, LK; Eftekharpour, E; Ghavami, S; Hatch, GM; Hombach-Klonisch, S; Islam, MI; Kardami, E; Klonisch, T; Koleini, N; Marzban, H; Samiei, E; Shojaei, S; Thliveris, J; Vosoughi, AR; Xu, F, 2020
)
2.32
"Temozolomide (TMZ) is a first-line alkylating agent for glioblastoma multiforme (GBM). "( IGF-1-enhanced miR-513a-5p signaling desensitizes glioma cells to temozolomide by targeting the NEDD4L-inhibited Wnt/β-catenin pathway.
Chen, KC; Chen, PH; Cheng, CH; Chou, CM; Ho, KH; Lee, CC; Shih, CM, 2019
)
2.19
"Temozolomide is an oral alkylating agent used for treating several cancers including glioblastoma and melanoma. "( Biomarker-guided implementation of the old drug temozolomide as a novel treatment option for patients with metastatic colorectal cancer.
Benelli, M; de Braud, F; Di Donato, S; Pietrantonio, F; Randon, G; Romagnoli, D, 2020
)
2.26
"Temozolomide is a first line anti-tumor drug used for the treatment of patients with Glioblastoma multiforme (GBM). "( Microarray expression profiles and bioinformatics analysis of mRNAs, lncRNAs, and circRNAs in the secondary temozolomide-resistant glioblastoma.
Gao, Y; Guo, R; Li, H; Yang, B; Zhao, C, 2020
)
2.21
"Temozolomide is an alkylating agent which is used in glioblastoma treatment. "( Effects of temozolomide on U87MG glioblastoma cell expression of CXCR4, MMP2, MMP9, VEGF, anti-proliferatory cytotoxic and apoptotic properties.
Elieh Ali Komi, D; Kiani, A; Mirabdaly, S; Moini, A; Shakiba, Y, 2020
)
2.39
"Temozolomide (TMZ) is a frequently used chemotherapy for glioma; however, chemoresistance is a major problem limiting its effectiveness. "( A
An, W; Attenello, F; Bi, J; Chen, B; Chen, J; Guo, D; He, A; Lu, W; Qian, Y; Shi, J; Shi, T; Wang, W; Zhang, L; Zhao, Z, 2020
)
2
"Temozolomide (TMZ) is a drug of choice in glioblastoma treatment. "( Revealing the epigenetic effect of temozolomide on glioblastoma cell lines in therapeutic conditions.
Barciszewska, AM; Barciszewski, J; Belter, A, 2020
)
2.28
"Temozolomide (TMZ) is an effective drug for prolonging the overall survival time of patients, while drug-resistance is an important clinical problem at present."( A steroidal saponin form Paris vietnamensis (Takht.) reverses temozolomide resistance in glioblastoma cells via inducing apoptosis through ROS/PI3K/Akt pathway.
Fang, F; Ji, Y; Li, H; Lu, Y; Qiu, P; Tang, H; Zhang, S, 2020
)
1.52
"Temozolomide is a drug approved for treating glioblastomas, which has 100% oral bioavailability but gets degraded at physiological pH thus having very short half-life and only 20-30% brain bioavailability. "( Design of temozolomide-loaded proliposomes and lipid crystal nanoparticles with industrial feasible approaches: comparative assessment of drug loading, entrapment efficiency, and stability at plasma pH.
Dubey, SK; Gorantla, S; Khosa, A; Rapalli, VK; Saha, RN; Singhvi, G; Waghule, T, 2021
)
2.47
"Temozolomide (TMZ) is a DNA alkylating agent and is currently a first line chemotherapeutic treatment for GBM."( Inhibition of Nrf2 might enhance the anti-tumor effect of temozolomide in glioma cells via inhibition of Ras/Raf/MEK signaling pathway.
Lu, Z; Sun, W; Yu, J; Zhang, W, 2021
)
1.59
"Temozolomide is a conventional chemotherapy drug for adjuvant treatment of patients with high-risk gliomas, including grade II to grade IV."( Blockade of Cell Volume Regulatory Protein NKCC1 Increases TMZ-Induced Glioma Apoptosis and Reduces Astrogliosis.
Amankulor, NM; Begum, G; Castro, MG; Ding, D; Dodelson, J; Fiesler, VM; Gayden, J; Guan, X; Hasan, MN; Hu, B; Jia, W; Kohanbash, G; Luo, L; Sun, B; Sun, D, 2020
)
1.28
"Temozolomide/etoposide is a feasible candidate as salvage therapy for relapsed osteosarcoma."( Temozolomide and etoposide combination for the treatment of relapsed osteosarcoma.
Adachi, S; Akazawa, R; Arakawa, Y; Hiramatsu, H; Kato, I; Moritake, H; Okamoto, T; Saida, S; Sakamoto, A; Sumiyoshi, S; Takita, J; Umeda, K, 2020
)
2.72
"Temozolomide is a first line anti-tumor drug used for the treatment of patients with Glioblastoma multiforme (GBM). "( MicroRNA-128-3p Enhances the Chemosensitivity of Temozolomide in Glioblastoma by Targeting c-Met and EMT.
Guan, F; Guo, R; Li, H; Li, M; Liu, X; Ma, S; Wu, J; Yang, B; Zhao, C, 2020
)
2.26
"Temozolomide (TMZ) is a DNA-alkylating agent used for chemo-radiotherapy of glioblastoma, which is also a target cancer for boron neutron capture therapy (BNCT). "( The combined effect of neutron irradiation and temozolomide on glioblastoma cell lines with different MGMT and P53 status.
Ikawa, T; Kinashi, Y; Takahashi, S, 2020
)
2.26
"Temozolomide (TMZ) is a life prolonging DNA alkylating agent active against glioblastomas (GBM) in which the O6-methylguanine-DNA methyltransferase (MGMT) gene is silenced by promoter methylation. "( PARP inhibition suppresses the emergence of temozolomide resistance in a model system.
Bering, EA; Black, S; Blough, MD; Cairncross, JG; Liao, J; Maxwell, L; Meode, M; Pedersen, H; Tan, M; Willms, J; Yuan, AL, 2020
)
2.26
"Temozolomide (TMZ) is a chemotherapeutic used for the treatment of glioblastoma. "( A co-formulation of interferons type I and II enhances temozolomide response in glioblastoma with unmethylated MGMT promoter status.
Bello-Rivero, I; Leenstra, S; van der Kaaij, M; Vázquez-Blomquist, D; Villarreal, A, 2020
)
2.25
"Temozolomide (TMZ) is an alkylating chemotherapy agent used in the clinical treatment of glioblastoma multiforme (GBM) patients. "( Piperine synergistically enhances the effect of temozolomide against temozolomide-resistant human glioma cell lines.
Jeong, S; Jung, S; Oh, JW; Park, GS; Shin, J, 2020
)
2.26
"Temozolomide (TMZ) is an oral alkylating agent used for the treatment of glioblastoma and is now becoming a chemotherapeutic option in patients diagnosed with high-risk low-grade gliomas. "( MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas.
Bao, Z; Galán-Ganga, M; Jiang, T; Kim, H; Kroon, P; Liu, H; Mu, Q; Nam, DH; Oldrini, B; Rabadan, R; Rodriguez-Perales, S; Sa, JK; Squatrito, M; Vaquero-Siguero, N; Verhaak, RGW; Wang, J; Wang, Z; Zhang, Y; Zhao, J, 2020
)
2
"Temozolomide is an oral DNA-alkylating agent capable of crossing the blood-brain barrier and used as chemotherapy primarily to treat glioblastoma and other brain cancers."( Central diabetes insipidus induced by temozolomide: A report of two cases.
Capes, A; Duck, L; Duprez, T; Labriola, L; Mahiat, C; Whenham, N, 2021
)
1.61
"Temozolomide (TMZ) is an oral chemotherapy drug constituting the backbone of chemotherapy regimens utilized as first-line treatment of GBM."( LncRNA NEAT1 promotes malignant phenotypes and TMZ resistance in glioblastoma stem cells by regulating let-7g-5p/MAP3K1 axis.
Bi, CL; Fang, JS; Lan, S; Liu, JF; Yang, ZY; Zhang, MY, 2020
)
1.28
"Temozolomide (TMZ) is an oral alkylating agent principally indicated for neurological malignancies including glioblastoma (GBM) and astrocytoma. "( Temozolomide-induced aplastic anaemia: Case report and review of the literature.
Gilbar, PJ; Mangos, HM; Pokharel, K, 2021
)
3.51
"Temozolomide (TMZ) is a first-line chemotherapy drug for the treatment of malignant glioma and resistance to it poses a major challenge. "( Regulation of temozolomide resistance in glioma cells via the RIP2/NF-κB/MGMT pathway.
Hu, YH; Jiao, BH; Wang, CY; Wu, JL, 2021
)
2.42
"Temozolomide (TMZ) is an effective chemotherapy drug for glioblastoma, but the resistance to TMZ has come to represent a major clinical problem, and its underlying mechanism has yet to be elucidated."( Exosomal connexin 43 regulates the resistance of glioma cells to temozolomide.
Bi, QC; Gan, LJ; Han, XJ; Hong, T; Jiang, LP; Lan, XM; Liu, LH; Tan, RJ; Wei, MJ; Yang, ZJ; Zhang, LL, 2021
)
1.58
"Temozolomide (TMZ) is a broad spectrum alkylating agent found effective in the treatment of glioblastoma multiforme, refractory anaplastic astrocytoma, and metastatic melanoma. "( UV spectroscopic method for estimation of temozolomide: Application in stability studies in simulated plasma pH, degradation rate kinetics, formulation design, and selection of dissolution media.
Narayan Saha, R; Singhvi, G; Waghule, T, 2021
)
2.33
"Temozolomide (TMZ) is an alkylating agent widely used for glioma treatment."( miR-23b-5p promotes the chemosensitivity of temozolomide via negatively regulating TLR4 in glioma.
Cui, B; Gao, K; Qiao, Y; Wang, T, 2021
)
1.6
"Temozolomide (TEM) is an active treatment in metastatic neuroendocrine tumors (NETs). "( Should temozolomide be used on the basis of O
Bagnardi, V; Curigliano, G; Fazio, N; Fumagalli, C; Gervaso, L; Guerini Rocco, E; Laffi, A; Peveri, G; Pisa, E; Rubino, M; Spada, F; Trillo Aliaga, P, 2021
)
2.52
"Temozolomide (TMZ) is a first-choice alkylating agent inducted as a gold standard therapy for glioblastoma multiforme (GBM) and astrocytoma. "( Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance.
Kumar, A; Shrivastava, A; Srivastava, C; Tomar, MS, 2021
)
2.35
"Temozolomide (TMZ) is an alkylating agent frequently used in well-differentiated metastatic pancreatic neuroendocrine tumors (PNETs) with very variable responses. "( Short article: Evaluation of O6-methylguanine-DNA methyltransferase as a predicting factor of response to temozolomide-based chemotherapy in well-differentiated metastatic pancreatic neuroendocrine tumors.
Caroli-Bosc, FX; Cauchin, E; Dumars, C; Foubert, F; Girot, P; Matysiak-Budnik, T; Mosnier, JF; Muzellec, L; Regenet, N; Senellart, H; Touchefeu, Y, 2017
)
2.11
"Temozolomide (TMZ) is a promising chemotherapeutic agent to treat Glioblastoma multiforme (GBM). "( MiR-181b modulates chemosensitivity of glioblastoma multiforme cells to temozolomide by targeting the epidermal growth factor receptor.
Chen, Y; Li, R; Liu, N; Pan, M; Shi, Z; Wang, X; Yan, W; You, Y; Zhang, J, 2017
)
2.13
"Temozolomide (TMZ) is an alkylating agent that has been widely used to treat GBM; resistance to this drug is often found."( Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low-frequency electromagnetic field (100Hz, 100G).
Abadi, MFS; Ahmadi, M; Akbarnejad, Z; Dini, L; Eskandary, H; Farsinejad, A; Nematollahi-Mahani, SN; Vergallo, C, 2017
)
1.52
"Temozolomide (TMZ) is an effective drug for malignant glioma, however, the intracellular and molecular mechanisms behind this anti-cancer effect have yet to be fully understood."( Down-Regulation of AQP4 Expression via p38 MAPK Signaling in Temozolomide-Induced Glioma Cells Growth Inhibition and Invasion Impairment.
Chen, Y; Gao, F; Hou, J; Jiang, R; Kang, L; Li, Y; Liu, H; Liu, X; Yang, M; Yi, Y, 2017
)
1.42
"Temozolomide (TMZ) is an alkylating chemotherapeutic agent widely used in anti-glioma treatment. "( Genomic profiling of long non-coding RNA and mRNA expression associated with acquired temozolomide resistance in glioblastoma cells.
Fu, Z; Guo, H; Lian, C; Liu, B; Liu, Y; Xu, N; Yang, Z; Zeng, H, 2017
)
2.12
"Temozolomide (TMZ) is an oral alkylating agent that has shown promise in treating aggressive pituitary adenomas and carcinomas that are resistant to other therapies."( Temozolomide therapy for resistant prolactin-secreting pituitary adenomas and carcinomas: a systematic review.
Ahmad, MM; Aldabas, BS; Aljoaib, NN; Almalki, MH; Alotaibi, MJ; Alshahrani, F; Wahedi, TS, 2017
)
2.62
"Temozolomide (TMZ) is an alkylating agent previously used in conjunction with doxorubicin (DOX) to treat dogs with relapsed lymphoma. "( Temozolomide alone or in combination with doxorubicin as a rescue agent in 37 cases of canine multicentric lymphoma.
Baines, SJ; Blackwood, L; Elliott, JW; Treggiari, E, 2018
)
3.37
"Temozolomide (TMZ) is an integral part of upfront treatment of high-grade gliomas. "( Rechallenge temozolomide in glioma: A case series from India.
Chandrasekaran, A; Epari, S; Gupta, T; Jalali, R; Kothari, R; Pande, N; Patil, VM; Tonse, R,
)
1.95
"Temozolomide is an alkylating chemotherapeutic agent used in malignant neuroendocrine neoplasia, melanoma, brain metastases and an essential component of adjuvant therapy in the treatment of glioblastoma multiforme and anaplastic astrocytoma. "( 65 YEARS OF THE DOUBLE HELIX: Treatment of pituitary tumors with temozolomide: an update.
Kovacs, K; Ortiz, LD; Rotondo, F; Syro, LV, 2018
)
2.16
"Temozolomide is an oral alkylating cytotoxic drug and like any other alkylating agent can induce side effects."( Severe hematologic temozolomide-related toxicity and lifethreatening infections.
Nikitovic, M; Stepanovic, A,
)
1.18
"Temozolomide (TMZ) is an antiangiogenic agent."( Opposite Interplay Between the Canonical WNT/β-Catenin Pathway and PPAR Gamma: A Potential Therapeutic Target in Gliomas.
Guillevin, R; Lecarpentier, Y; Vallée, A; Vallée, JN, 2018
)
1.2
"Temozolomide (TMZ) is a standard chemotherapeutic drug used in the treatment of glioblastoma multiforme (GBM); however, resistance to this drug is common. "( Association between SOX9 and CA9 in glioma, and its effects on chemosensitivity to TMZ.
Cheng, Y; Jin, W; Liu, H; Liu, N; Tu, Y; Wang, X; Wang, Z; Xu, X; Yang, H; Zhang, P; Zhang, Y, 2018
)
1.92
"Temozolomide (TMZ) is an alkylating agent used for glioblastoma multiforme (GBM) treatment. "( Synergistic effect of temozolomide and thymoquinone on human glioblastoma multiforme cell line (U87MG).
Khazaei, M; Khazaei, MR; Moradi, MT; Pazhouhi, M; Sariri, R,
)
1.89
"Temozolomide (TMZ) is a first-line chemotherapeutic drug for malignant gliomas. "( Honokiol enhances temozolomide-induced apoptotic insults to malignant glioma cells via an intrinsic mitochondrion-dependent pathway.
Chen, RM; Chio, CC; Liu, SH; Mohanraj, M; Tai, YT; Yang, ST, 2018
)
2.26
"Temozolomide (TMZ) is a drug used to treat GBM, while the survival period of GBM patients with positive treatment remains less than 15 months."( Study on Therapeutic Action and Mechanism of TMZ Combined with RITA Against Glioblastoma.
Cao, Z; Li, L; Wu, Q; Xiao, W; Xie, Q; Zhang, B; Zhao, W; Zhu, L, 2018
)
1.2
"Temozolomide (TMZ) is an alkylating agent commonly used as a first‑line treatment for high‑grade glioblastoma. "( Synergistic anticancer effect of acteoside and temozolomide-based glioblastoma chemotherapy.
Choi, DE; Hwang, TW; Jang, TW; Kim, DB; Kim, DH; Kim, GH; Kim, JJ; Moon, M; Park, JH; Yoon, KA, 2019
)
2.21
"Temozolomide (TMZ) is a widely used chemotherapeutic agent for glioblastoma multiforme (GBM). "( Momelotinib sensitizes glioblastoma cells to temozolomide by enhancement of autophagy via JAK2/STAT3 inhibition.
Li, A; Liu, T; Xin, Y; Xu, Y, 2019
)
2.22
"Temozolomide is a first-line treatment for newly diagnosed glioblastoma. "( Evidence-Based Practice: Temozolomide Beyond Glioblastoma.
Chua, J; Leung, D; Nafziger, E, 2019
)
2.26
"Temozolomide (TMZ) is an alkylating agent used in the treatment of high-grade malignant glioma, notably glioblastoma multiforme, the most aggressive form of brain cancer. "( Are There Thresholds in Glioblastoma Cell Death Responses Triggered by Temozolomide?
He, Y; Kaina, B, 2019
)
2.19
"Temozolomide (TMZ) is an important agent against GBM."( Inhibition of cyclin E1 overcomes temozolomide resistance in glioblastoma by Mcl-1 degradation.
Chen, Z; Liang, H; Sun, L, 2019
)
1.51
"Temozolomide (TMZ) is a frequently used primary chemotherapeutic agent for LGGs."( Comprehensive Analysis Reveals a 4-Gene Signature in Predicting Response to Temozolomide in Low-Grade Glioma Patients.
Chen, Y; He, Z; Wang, Q,
)
1.08
"Temozolomide (TMZ) is an alkylating agent widely used to treat cancer, resistance to this drug is often found."( Impact of extremely low-frequency electromagnetic field (100 Hz, 100 G) exposure on human glioblastoma U87 cells during Temozolomide administration.
Ahmadi-Zeidabadi, M; Akbarnejad, Z; Eskandary, H; Esmaeeli, M; Masoumi-Ardakani, Y; Mohammadipoor-Ghasemabad, L, 2019
)
1.44
"Temozolomide is an oral chemotherapy used to treat aggressive pituitary tumours since 2006. "( How and when to use temozolomide to treat aggressive pituitary tumours.
Whitelaw, BC, 2019
)
2.28
"Temozolomide (TMZ) is an alkylating agent shown to prolong survival in patients with high grade glioma and is routinely used to treat melanoma brain metastases. "( Myeloablative temozolomide enhances CD8⁺ T-cell responses to vaccine and is required for efficacy against brain tumors in mice.
Archer, GE; Bigner, DD; Choi, BD; Cui, X; Flores, C; Herndon, JE; Johnson, LA; Mitchell, DA; Sampson, JH; Sanchez-Perez, LA; Schmittling, RJ; Snyder, D, 2013
)
2.19
"Temozolomide is a novel oral chemotherapy drug that penetrates into the brain and purportedly has a low incidence of adverse events."( Temozolomide for high grade glioma.
Garside, R; Grant, R; Hart, MG; Rogers, G; Stein, K, 2013
)
2.55
"Temozolomide (TMZ) is an alkylating agent that is the first-line chemotherapy for glioblastoma."( Phase 1/1b study of lonafarnib and temozolomide in patients with recurrent or temozolomide refractory glioblastoma.
Colman, H; Conrad, C; Gilbert, MR; Groves, M; Hsu, S; Kang, S; Levin, V; Liu, D; Liu, V; Puduvalli, V; Yuan, Y; Yung, WK; Yust-Katz, S, 2013
)
1.39
"Temozolomide is an oral alkylating agent with proven efficacy in recurrent high-grade glioma. "( Effect of temozolomide on male gametes: an epigenetic risk to the offspring?
Bashamboo, A; Berthaut, I; Deluen, F; Dessolle, L; McElreavey, K; Montjean, D; Morcel, K; Poirot, C; Ravel, C, 2013
)
2.23
"Temozolomide (TMZ) is a DNA methylating agent used to treat brain cancer. "( Early Chk1 phosphorylation is driven by temozolomide-induced, DNA double strand break- and mismatch repair-independent DNA damage.
Gaensler, K; Ito, M; Mukherjee, J; Ohba, S; Pieper, RO; Ronen, SM, 2013
)
2.1
"Temozolomide (TMZ) is an oral alkylating agent used for the treatment of recurrent or newly diagnosed malignant gliomas with significant survival benefit. "( Temozolomide-related hematologic toxicity.
De Sanctis, V; Enrici, RM; Minniti, G; Scaringi, C, 2013
)
3.28
"Temozolomide (TMZ) is an alkylating agent primarily used to treat tumors of the central nervous system. "( Central diabetes insipidus: a previously unreported side effect of temozolomide.
Faje, AT; Klibanski, A; Makimura, H; Miller, KK; Nachtigall, L; Wexler, D, 2013
)
2.07
"Temozolomide (TMZ) is an oral alkylating agent used in the treatment of central nervous system neoplasms and metastatic melanoma. "( A phase I, dose-escalation study of cyclical weekly oral temozolomide and weekly PEG-interferon alpha-2b in patients with refractory or advanced solid tumours.
Beelen, AP; Coker, SA; Crosby, NA; Dandamudi, UB; Ernstoff, MS; Fisher, JL; Lewis, LD; Obrocea, M, 2013
)
2.08
"Temozolomide (TMZ) is an alkylating agent currently used as first-line therapy for gliomas treatment due to its DNA-damaging effect. "( O6-methylguanine DNA methyltransferase as a promising target for the treatment of temozolomide-resistant gliomas.
Cao, H; Chen, L; Fan, CH; Jiang, G; Liu, WL; Wen, C, 2013
)
2.06
"Temozolomide (TMZ) is an alkylating agent used in chemoradiotherapy and adjuvant chemotherapy regimens for treatment of newly diagnosed or recurrent glioblastoma. "( Temozolomide-induced liver damage. A case report.
Becker, F; Fietkau, R; Hecht, M; Schmidtner, J; Semrau, S, 2014
)
3.29
"Temozolomide (TMZ) is a promising chemotherapeutic agent for treating glioblastomas. "( miR-125b inhibitor enhance the chemosensitivity of glioblastoma stem cells to temozolomide by targeting Bak1.
Chen, J; Fu, X; Jiang, D; Shi, L; Wan, Y; Wang, Z, 2014
)
2.07
"Temozolomide (TMZ) is a first-line chemotherapeutic agent but the efficacy is limited by intrinsic and acquired resistance in GBM."( Triptolide synergistically enhances temozolomide-induced apoptosis and potentiates inhibition of NF-κB signaling in glioma initiating cells.
Chen, YS; Chen, ZP; Guan, S; Guo, CC; Li, WP; Li, WY; Mou, YG; Sai, K; Wang, J; Yang, QY, 2014
)
1.4
"Temozolomide (TMZ) is an alkylating agent used for the treatment of glioblastoma multiforme (GBM), the main form of human brain tumours in adults. "( Bak and Mcl-1 are essential for Temozolomide induced cell death in human glioma.
Gratas, C; Oliver, L; Rabé, M; Séry, Q; Vallette, FM, 2014
)
2.13
"Temozolomide (TMZ) is an alkylating agent that is widely used in chemotherapy for cancer. "( Progression of O⁶-methylguanine-DNA methyltransferase and temozolomide resistance in cancer research.
Cheng, Q; Jiang, AJ; Jiang, G; Li, LT; Xin, Y; Zheng, JN, 2014
)
2.09
"Temozolomide (TMZ) is an oral alkylating agent which is widely used in the treatment of GBM following surgery."( miR-128 and miR-149 enhance the chemosensitivity of temozolomide by Rap1B-mediated cytoskeletal remodeling in glioblastoma.
Cui, Y; Lei, Q; Li, G; She, X; Wang, Z; Wu, M; Xiang, J; Xu, G; Yu, Z, 2014
)
1.37
"Temozolomide (TMZ) is an alkylating agent used for the treatment of glioblastoma multiforme (GBM), the most common and deadliest of malignant primary brain tumors."( Glioblastoma multiforme and hepatitis B: do the right thing(s).
Begini, P; Delle Fave, G; Gallina, S; Marignani, M; Minniti, G; Purchiaroni, F, 2014
)
1.12
"Temozolomide is an oral alkylating agent with schedule-dependent antitumor activity against high-grade malignancies including high-grade glioma. "( Temozolomide retreatment in a recurrent prolactin-secreting pituitary adenoma: Hormonal and radiographic response.
Laterra, JJ; Salvatori, R; Strowd, RE, 2016
)
3.32
"Temozolomide is an increasingly described treatment option for refractory pituitary adenomas and carcinomas. "( Temozolomide retreatment in a recurrent prolactin-secreting pituitary adenoma: Hormonal and radiographic response.
Laterra, JJ; Salvatori, R; Strowd, RE, 2016
)
3.32
"Temozolomide (TMZ) is an oral alkylating drug with good tolerability, approved for treatment of malignant gliomas."( Long-term outcome and MGMT as a predictive marker in 24 patients with atypical pituitary adenomas and pituitary carcinomas given treatment with temozolomide.
Andersen, M; Bengtsson, D; Berinder, K; Burman, P; Feldt Rasmussen, U; Hoybye, C; Johannsson, G; Maiter, D; Petersson, M; Ragnarsson, O; Rasmussen, ÅK; Schrøder, HD; van der Lely, AJ, 2015
)
1.34
"Temozolomide rechallenge is a treatment option for MGMT promoter-methylated recurrent glioblastoma. "( MGMT Promoter Methylation Is a Strong Prognostic Biomarker for Benefit from Dose-Intensified Temozolomide Rechallenge in Progressive Glioblastoma: The DIRECTOR Trial.
Bähr, O; Felsberg, J; Goldbrunner, R; Hau, P; Herrlinger, U; Homicsko, K; Hüsing, J; Kästner, B; Ketter, R; Kollias, S; Marosi, C; Meixensberger, J; Nikkhah, G; Pichler, J; Platten, M; Reifenberger, G; Sabel, MC; Schlegel, U; Schnell, O; Steinbach, JP; Stupp, R; Tabatabai, G; Tonn, JC; Vajkoczy, P; Weller, M; Wick, A; Wick, W; Wirsching, HG, 2015
)
2.08
"Temozolomide (TMZ) is an alkylating agent used for the treatment of glioblastoma. "( The synergistic effect of combination temozolomide and chloroquine treatment is dependent on autophagy formation and p53 status in glioma cells.
Hong, SH; Hong, YK; Joe, YA; Kim, HK; Kim, HS; Lee, NH; Lee, SW; Yi, HY, 2015
)
2.13
"Temozolomide (TMZ) is an effective agent for clinical glioma treatment, but the innate and acquired resistance of glioma always limits its application. "( p38 MAPK-dependent Nrf2 induction enhances the resistance of glioma cells against TMZ.
Gu, Y; Liu, J; Ma, L; Qi, J; Yu, W; Zhang, X, 2015
)
1.86
"Temozolomide is a chemotherapeutic drug that is mainly used in the treatment of primary glioblastoma multiforme and recurrent high-grade glioma. "( Synthesis of [3-N-(11) C-methyl]temozolomide via in situ activation of 3-N-hydroxymethyl temozolomide and alkylation with [(11) C]methyl iodide.
Eriksson, J; Froklage, FE; Hendrikse, NH; Reijneveld, JC; Schuit, RC; Van Kooij, R; Windhorst, AD, 2015
)
2.14
"Temozolomide (TMZ) is an alkylating drug used usually in glioma treatment by inducing the apoptosis in glioma cell. "( Temozolomide may induce cell cycle arrest by interacting with URG4/URGCP in SH-SY5Y neuroblastoma cells.
Avcı, ÇB; Çıtışlı, V; Dodurga, Y; Eroğlu, C; Şatıroğlu-Tufan, NL; Seçme, M, 2015
)
3.3
"Temozolomide (TMZ) is an oral, potentially less toxic derivative of dacarbazine, which has shown activity both as a single agent and in combination with other drugs."( Temozolomide in Advanced Neuroendocrine Neoplasms: Pharmacological and Clinical Aspects.
Barberis, M; Berruti, A; Fazio, N; Fumagalli, C; Galdy, S; Kaltsas, G; Koumarianou, A; Kulke, MH; Oberg, K; Spada, F; Strosberg, JR, 2015
)
2.58
"Temozolomide (TMZ) is an oral alkylating agent with established effects on the central nervous system of glioblastoma (GBM) patients. "( Do glioma patients derive any therapeutic benefit from taking a higher cumulative dose of temozolomide regimens?: a meta-analysis.
Du, S; Liao, G; Ren, C; Sun, H; Xie, X; Yuan, YW, 2015
)
2.08
"Temozolomide is a novel cytotoxic agent currently used as first-line chemotherapy for glioblastoma multiforme (GBM). "( TAZ promotes temozolomide resistance by upregulating MCL-1 in human glioma cells.
Li, A; Li, Z; Lu, H; Luo, R; Tian, T; Zhang, M, 2015
)
2.23
"Temozolomide (TMZ) is a promising chemotherapeutic agent for treating glioblastomas. "( PI3K inhibitor combined with miR-125b inhibitor sensitize TMZ-induced anti-glioma stem cancer effects through inactivation of Wnt/β-catenin signaling pathway.
Fei, X; Shi, L; Wang, Z; You, Y, 2015
)
1.86
"Temozolomide (TMZ) is an oral alkylating chemotherapeutic agent that prolongs the survival of patients with glioblastoma (GBM). "( A New Epigenetic Mechanism of Temozolomide Action in Glioma Cells.
Barciszewska, AM; Gurda, D; Głodowicz, P; Naskręt-Barciszewska, MZ; Nowak, S, 2015
)
2.15
"Temozolomide (TMZ) is an anticancer drug used for the treatment of adult brain tumour and skin cancer. "( Electrochemical monitoring of the interaction between Temozolamide and nucleic acids by using disposable pencil graphite electrodes.
Altay, C; Congur, G; Eksin, E; Erdem, A, 2015
)
1.86
"Temozolomide is a standard chemotherapy agent for malignant gliomas, but the efficacy is still not satisfactory. "( Temozolomide reverses doxorubicin resistance by inhibiting P-glycoprotein in malignant glioma cells.
Kanamori, M; Saito, R; Shibahara, I; Sonoda, Y; Sugiyama, S; Tominaga, T; Zhang, R, 2016
)
3.32
"Temozolomide (TMZ) is an oral DNA-alkylating agent used for treating patients with glioblastoma. "( Bone morphogenetic protein 7 sensitizes O6-methylguanine methyltransferase expressing-glioblastoma stem cells to clinically relevant dose of temozolomide.
Bui, Y; Cloughesy, TF; Hong, I; Lai, A; Liau, LM; McBride, WH; Menjivar, JC; Nelson, SF; Stream, A; Tso, CL; Tso, JL; Yamada, K; Yang, S; Yong, WH; Zhang, Y, 2015
)
2.06
"Temozolomide (TMZ) is an oral alkylating agent that has been used over the past 8 years to treat aggressive pituitary tumors resistant to conventional therapy. "( Temozolomide for aggressive ACTH pituitary tumors: failure of a second course of treatment.
Aller, J; Campderá, M; Estrada, J; Lilienfeld, H; Magallón, R; Martín, P; Palacios, N; Saucedo, G, 2016
)
3.32
"Temozolomide is an additional effective therapeutic option for the treatment of aggressive pituitary tumors."( Temozolomide therapy in patients with aggressive pituitary adenomas or carcinomas.
Bogazzi, F; Cannavo, S; Ceccato, F; Curtò, L; De Marinis, L; Iacovazzo, D; Lombardi, G; Losa, M; Mantovani, G; Mazza, E; Minniti, G; Nizzoli, M; Reni, M; Scaroni, C, 2016
)
2.6
"Temozolomide is an alkylating agent used along with concurrent radiation therapy in the treatment of glioblastoma. "( Temozolomide-induced biliary ductopenia: a case report.
Balakrishnan, A; Jaglal, M; Ledford, R, 2016
)
3.32
"Temozolomide (TMZ) is a standard agent used in the treatment of various types of cancers, including lung carcinoma, but TMZ resistance is common and accounts for many treatment failures. "( miR-487b-5p Regulates Temozolomide Resistance of Lung Cancer Cells Through LAMP2-Medicated Autophagy.
Bao, L; Chen, Y; Feng, J; Han, S; Lv, L; Wang, X; Zhao, H, 2016
)
2.19
"Temozolomide is an oral chemotherapy with a favorable side-effect profile that has shown activity against pituitary adenomas."( Is there a role for early chemotherapy in the management of pituitary adenomas?
DeAngelis, LM; Lin, AL; Sum, MW, 2016
)
1.16
"Temozolomide (TMZ) is an alkylating agent used to treat glioblastoma. "( Sulfasalazine intensifies temozolomide cytotoxicity in human glioblastoma cells.
Castilho, RF; De Melo, DR; Facchini, G; Ignarro, RS; Lopes-Cendes, I; Rogerio, F; Vieira, AS, 2016
)
2.18
"Temozolomide (TMZ) is an alkylating agent that has become the mainstay treatment of the most malignant brain cancer, glioblastoma multiforme (GBM). "( Zinc enhances temozolomide cytotoxicity in glioblastoma multiforme model systems.
Assoulin, M; Constantini, S; Daniels, D; Fisher, T; Freedman, S; Guez, D; Last, D; Mardor, Y; Mehrian-Shai, R; Moshe, I; Pismenyuk, T; Reichardt, JK; Simon, AJ; Toren, A; Yalon, M, 2016
)
2.24
"Temozolomide (TMZ) is a DNA-methylating agent."( Current and Future Drug Treatments for Glioblastomas.
Hirose, Y; Ohba, S, 2016
)
1.16
"Temozolomide (TMZ) is an oral imidazotetrazine methylating agent which is used for the treatment of glioblastoma multiforme (GBM). "( Acute temozolomide induced liver injury: mixed type hepatocellular and cholestatic toxicity.
Agan, AF; Altınok, AY; Aygun, C; Balaban, Y; Çakır, A,
)
2.05
"Temozolomide (TMZ) is an alkylating agent used for the treatment of aggressive forms of brain tumor based on its antitumor actions. "( Temozolomide inhibits cellular growth and motility via targeting ERK signaling in glioma C6 cells.
Gao, S; Liang, J; Wang, W; Wang, Y, 2016
)
3.32
"Temozolomide is a commonly used chemotherapy drug and frequently causes lymphocytopenia."( Cutaneous invasive aspergillosis in a patient with glioblastoma receiving long-term temozolomide and corticosteroid therapy.
Hatakeyama, S; Ikeda, T; Morisawa, Y; Norizuki, M; Okabe, T; Onishi, T; Sasahara, T; Suzuki, J; Toshima, M; Yokota, H, 2017
)
1.4
"Temozolomide (TMZ) is an alkylating agent used for treatment of brain neoplasms and levetiracetam (LEV) is a commonly used antiepileptic. "( Acute liver injury induced by levetiracetam and temozolomide co-treatment.
Abu Rmeileh, A; Benson, AA; Chen, S; Cohen, J; Daher, S; Khoury, T; Mizrahi, M; Yaari, S, 2017
)
2.15
"Temozolomide (TMZ) is a novel cytotoxic agent used as first-line chemotherapy for GBM, however, some individual cells can't be isolated for surgical resection and show treatment-resistance, thus inducing poor prognosis."( MALAT1 is a prognostic factor in glioblastoma multiforme and induces chemoresistance to temozolomide through suppressing miR-203 and promoting thymidylate synthase expression.
Chen, C; Chen, W; Ge, XS; He, J; Kong, KK; Li, FC; Li, H; Li, JL; Li, P; Wang, F; Xu, XK, 2017
)
1.4
"Temozolomide (TMZ) is an oral imidazotetrazine methylating agent which is used for the treatment of glioblastoma multiforme (GBM). "( Acute temozolomide induced liver injury : Mixed type hepatocellular and cholestatic toxicity.
Agan, AF; Altınok, AY; Aygun, C; Balaban, Y; Çakır, A,
)
2.05
"Temozolomide is an alkylating agent which crosses the blood brain barrier and has demonstrated antitumor activity against a broad range of tumor types, including malignant glioma, melanoma, non small cell lung cancer and carcinoma of the ovary and colon."( Phase I trial of weekly docetaxel and daily temozolomide in patients with metastatic disease.
Bukowski, RM; Dreicer, R; Elson, P; Mekhail, T; Olencki, T; Roman, S; Tamaskar, I, 2008
)
1.33
"The temozolomide is a promising orally cytotoxic agent used in malignant glioma. "( The safety of the temozolomide in patients with malignant glioma.
Dario, A; Tomei, G, 2006
)
1.23
"Temozolomide is a relatively new chemotherapeutic agent frequently associated with selective CD4+ T-lymphocytopenia. "( Disseminated salmonellosis in a patient treated with temozolomide.
Adachi, J; Georgescu, G; Isola, IM; Rolston, K; Youssef, S, 2008
)
2.04
"Temozolomide is a novel oral chemotherapeutic drug that penetrates into the brain and has a low incidence of adverse effects."( Temozolomide for high grade glioma.
Garside, R; Grant, R; Hart, MG; Rogers, G; Somerville, M; Stein, K, 2008
)
2.51
"Temozolomide is an effective therapy in GBM for prolonging survival and delaying progression as part of primary therapy without impacting on QoL and with a low incidence of early adverse events. "( Temozolomide for high grade glioma.
Garside, R; Grant, R; Hart, MG; Rogers, G; Somerville, M; Stein, K, 2008
)
3.23
"Temozolomide is an alkylating agent approved for treatment of glioblastoma in association with radiotherapy."( [Alveolo-interstitial pneumonia due to Temozolamide].
Autret-Leca, E; Beau Salinas, F; Carré, P; de Luca, K; Diot, P; Guilleminault, L; Narciso, B, 2008
)
1.79
"Temozolomide (TMZ) is a cytotoxic agent of the imidazotetrazine class, chemically related to dacarbazine. "( Tuberculosis in a patient on temozolomide: a case report.
de Barros e Silva, MJ; de Paiva, TF; Fanelli, MF; Gimenes, DL; Rinck, JA, 2009
)
2.09
"Temozolomide is an effective alkylating agent that is increasingly used for the treatment of pediatric brain tumors. "( Treatment-related myelodysplastic syndrome after temozolomide use in a child: first report.
Auger, N; Bhangoo, R; Da Costa, L; Dufour, C; Grill, J; Jullien, M, 2008
)
2.04
"Temozolomide is a newly developed chemotherapeutic agent in central nervous system tumors."( Efficacy of temozolomide for recurrent embryonal brain tumors in children.
Chang, KP; Hsu, TR; Wang, CH; Wong, TT, 2009
)
1.45
"Temozolomide is a novel cytotoxic agent for malignant gliomas. "( Carboplatin and oral cyclophosphamide combination after temozolomide failure in malignant gliomas.
Benekli, M; Buyukberber, S; Coskun, U; Kaya, AO; Ozturk, B; Uner, A; Yamac, D; Yaman, E; Yildiz, R,
)
1.82
"Temozolomide (TMZ) is an oral anticancer agent approved for the treatment of newly diagnosed glioblastoma in combination with radiotherapy. "( Recent approaches to improve the antitumor efficacy of temozolomide.
Graziani, G; Tentori, L, 2009
)
2.04
"Temozolomide (TMZ) is an alkylating agent used in the management of gliomas. "( Long-term use of temozolomide: could you use temozolomide safely for life in gliomas?
Bell, D; Khasraw, M; Wheeler, H, 2009
)
2.14
"Temozolomide is a new drug which has shown promise in treating malignant gliomas and other difficult-to-treat tumors."( Temozolomide with radiation therapy in high grade brain gliomas: pharmaceuticals considerations and efficacy; a review article.
Beli, I; Chaldeopoulos, D; Fotineas, A; Koukourakis, GV; Kouloulias, V; Kouvaris, J; Maravelis, G; Pantelakos, P; Papadimitriou, C; Zacharias, G, 2009
)
2.52
"Temozolomide is an oral alkylating cytotoxic agent of second generation, used in the treatment of high-grade gliomas."( [Prescription guidebook for temozolomide usage in brain tumors].
Borget, I; Brignone, M; Cartalat-Carel, S; Chinot, O; Hassani, Y; Taillandier, L; Taillibert, S; Tilleul, P, 2009
)
1.37
"Temozolomide is a well-tolerated oral chemotherapy in patients with malignant DET, including those who have already received treatment. "( Temozolomide: a safe and effective treatment for malignant digestive endocrine tumors.
Couvelard, A; Faivre, S; Hammel, P; Hentic, O; Larroque, B; Lévy, P; Maire, F; Raymond, E; Ruszniewski, P; Yapur, L; Zappa, M, 2009
)
3.24
"Temozolomide (TMZ) is an oral alkylating agent with significant activity against glioblastoma multiforme (GBM) and melanoma. "( Non-Hodgkin lymphoma following temozolomide.
Das, P; Dwary, A; Goyal, S; Gupta, D; Mohanti, BK; Muzumder, S; Sharma, A; Thulkar, S, 2009
)
2.08
"Temozolomide (TMZ) is an oral alkylating agent that is regarded as a tolerable and effective drug. "( Temozolomide in malignant gliomas: current use and future targets.
Bressler, LR; Seery, TE; Villano, JL, 2009
)
3.24
"Temozolomide (TMZ) is a methylating agent used in chemotherapy against glioblastoma. "( DNA ligase IV as a new molecular target for temozolomide.
Kondo, N; McKinnon, PJ; Mori, E; Nakase, H; Ohnishi, K; Ohnishi, T; Sakaki, T; Takahashi, A, 2009
)
2.06
"Temozolomide (TMZ) is an alkylating cytostaticum."( Temozolomide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy.
Andersen, M; Hagen, C; Hansen, S; Schroeder, HD, 2009
)
2.52
"Temozolomide (TMZ) is a second-generation alkylating agent that has recently shown some efficacy in stage IV melanoma. "( Temozolomide and cisplatin combination in naive patients with metastatic cutaneous melanoma: results of a phase II multicenter trial.
Bedane, C; Guillet, G; Guillot, B; Mourey, L; Sassolas, B; Tourani, JM; Wierzbicka-Hainaut, E, 2010
)
3.25
"Temozolomide is an easily administered, well-tolerated chemotherapeutic option in advanced or recurrent uterine leiomyosarcomas with a reasonable response rate. "( Temozolomide in advanced and recurrent uterine leiomyosarcoma and correlation with o6-methylguanine DNA methyltransferase expression: a case series.
Atkins, KA; Ferriss, JS; Jazaeri, AA; Lachance, JA; Modesitt, SC, 2010
)
3.25
"Temozolomide (TMZ) is a pro-drug releasing a DNA alkylating agent that is the most effective drug to treat glial tumors when combined with radiation. "( Temozolomide delivery to tumor cells by a multifunctional nano vehicle based on poly(β-L-malic acid).
Black, KL; Ding, H; Holler, E; Hu, J; Inoue, S; Konda, B; Ljubimova, JY; Patil, R; Portilla-Arias, J; Shin, PK; Wawrowsky, KA, 2010
)
3.25
"Temozolomide (TMZ) is a recently introduced alkylating agent that has yielded significant benefits and become a key agent in the treatment of high-grade gliomas."( Gene expression profiling predicts response to temozolomide in malignant gliomas.
Fukushima, T; Katayama, Y; Naruse, N; Ogino, A; Ohta, T; Okamoto, Y; Sano, E; Tsumoto, K; Watanabe, T; Yachi, K; Yoshino, A, 2010
)
1.34
"Temozolomide is an oral alkylating agent with established antitumor activity in patients with primary brain tumors and melanoma. "( Dose-dense temozolomide regimens: antitumor activity, toxicity, and immunomodulatory effects.
Hwu, WJ; Neyns, B; Reardon, DA; Tosoni, A, 2010
)
2.19
"Temozolomide (TMZ) is a monofunctional methylating agent which is spontaneously activated in aqueous solution into the dacarbazine metabolite 5-(3-methyl-1-triazeno)imidazole-4-carboxamide. "( The use of temozolomide for the treatment of malignant tumors: clinical evidence and molecular mechanisms of action.
Bei, R; Marzocchella, L; Turriziani, M, 2010
)
2.19
"Temozolomide (TMZ) is an oral alkylating agent with proven antitumoral activity in preclinical and clinical studies in adults with high-grade glioma (HGG). "( Feasibility study of 21-day-on/7-day-off temozolomide in children with brain tumors.
Barone, G; Lazzareschi, I; Riccardi, R; Ridola, V; Rizzo, D; Ruggiero, A, 2011
)
2.08
"Temozolomide is an active agent in metastatic pancreatic endocrine carcinomas. "( First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas.
Chen, DT; Choi, J; Coppola, D; Fine, RL; Helm, J; Kvols, L; Nasir, A; Strosberg, JR, 2011
)
2.07
"Temozolomide (TMZ) is an alkylating agent licensed for treatment of high-grade glioma (HGG). "( Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma.
Beall, S; Brada, M; Collins, VP; Erridge, S; Gabe, R; Gattamaneni, R; Hopkins, K; Lee, SM; Levy, D; Rampling, R; Saran, F; Stenning, S; Thompson, LC, 2010
)
3.25
"Temozolomide is an oral alkylating agent approved for the treatment of glioblastoma and anaplastic astrocytoma, and is currently under clinical investigation for the treatment of brain metastases from a variety of cancers. "( Urticarial hypersensitivity reaction caused by temozolomide.
Hsu, MY; Ibrahimi, OA; Kesari, S; Pothiawala, S; Yang, C, 2010
)
2.06
"Temozolomide (TMZ) is an oral alkylating agent used for the treatment of high-grade gliomas. "( Acquisition of temozolomide chemoresistance in gliomas leads to remodeling of mitochondrial electron transport chain.
Bailey, SM; Darley-Usmar, VM; Diers, A; Gillespie, GY; Griguer, CE; Landar, A; Markert, JM; McClugage, SG; Nozell, SE; Oliva, CR; Sarkaria, JN, 2010
)
2.16
"Temozolomide is an oral alkylating agent approved for glioblastoma multiforme treatment that has only recently shown promise in treating some pituitary tumors."( Temozolomide for corticotroph pituitary adenomas refractory to standard therapy.
Delashaw, JB; Dillard, TH; Fleseriu, M; Gultekin, SH; Neuwelt, EA; Yedinak, CG, 2011
)
2.53
"Temozolomide (TMZ) is an oral alkylating agent with demonstrated efficacy as second-line therapy for patients with recurrent anaplastic astrocytoma and glioblastoma multiforme (GBM). "( The outcomes of concomitant radiation plus temozolomide followed by adjuvant temozolomide for newly diagnosed high grade gliomas: the preliminary results of single center prospective study.
Abo Hamar, AH; El-Shorbagy, D; Galal, S; Shawky, H; Zakaria, F, 2009
)
2.06
"Temozolomide (TMZ) is an alkylating chemotherapeutic agent that has recently been used in some cases as a new therapeutic tool for pituitary carcinomas and aggressive pituitary adenomas. "( Temozolomide-induced shrinkage of a pituitary carcinoma causing Cushing's disease--report of a case and literature review.
Altavilla, G; Cannavò, S; Curtò, L; Ferraù, F; Granata, F; Hofland, LJ; Longo, M; Pitini, V; Torre, ML; Trimarchi, F, 2010
)
3.25
"Temozolomide is a second-generation alkylating chemotherapeutic agent, introduced to therapy of primary brain tumors in the 1990s."( Temozolomide: Expanding its role in brain cancer.
Adair, J; Kiem, HP; Mrugala, MM, 2010
)
2.52
"Temozolomide is an alkylating agent recently employed in older patients with newly diagnosed glioblastoma."( Treatment of glioblastoma in elderly patients: an overview of current treatments and future perspective.
Lanzetta, G; Minniti, G,
)
0.85
"Temozolomide is an alkylating chemotherapeutic agent used routinely in the management of high grade gliomas."( MGMT immunoexpression in adamantinomatous craniopharyngiomas.
Altuntaş, Y; Aydın, Y; Karaman, O; Müslüman, AM; Ozderya, A; Ozkayalar, H; Oztürk, FY; Tanık, C; Zuhur, SS, 2011
)
1.09
"Temozolomide (TMZ) is a DNA-alkylating agent used for the treatment of glioma, astrocytoma, and melanoma."( A novel approach to overcome temozolomide resistance in glioma and melanoma: Inactivation of MGMT by gene therapy.
Jiang, G; Liu, YQ; Pei, DS; Wei, ZP; Xin, Y; Zheng, JN, 2011
)
1.38
"Temozolomide (TMZ) is an oral alkylating agent that has been widely used in the treatment of refractory glioma, although inherent and acquired resistance to this drug is common. "( Enhancement of temozolomide-induced apoptosis by valproic acid in human glioma cell lines through redox regulation.
Chang, YJ; Chen, CH; Chung, KT; Ku, MS; Yang, JT, 2011
)
2.16
"Temozolomide is an effective treatment for seizure control in patients with brain tumors."( Seizure control: a secondary benefit of chemotherapeutic temozolomide in brain cancer patients.
Hu, A; Kesari, S; Kim, RY; Lee, JW; Nguyen, A; Xu, Z, 2011
)
1.34
"Temozolomide (TMZ) is a widely used oral alkylating agent that has been associated with the development of severe hematologic adverse events (HAEs). "( Hematologic adverse events associated with temozolomide.
Abdur, S; Bressler, LR; Letarte, N; Villano, JL; Yu, JM, 2012
)
2.08
"Temozolomide (TMZ), is a new alkylating agent with promising antitumour efficacy for malignant gliomas, and the effect of TMZ on GSCs invasion has not been known."( Temozolomide decreases invasion of glioma stem cells by down-regulating TGF-β2.
Jing, Z; Qiu, B; Wang, Y; Wu, A; Zhang, D, 2011
)
2.53
"Temozolomide (TMZ) is an oral derivative of dacarbazine that induces DNA damage by methylating nucleotide bases. "( Multicenter phase II trial of temozolomide in mycosis fungoides/sezary syndrome: correlation with O⁶-methylguanine-DNA methyltransferase and mismatch repair proteins.
Baron, J; Dolan, ME; Foss, F; Guitart, J; Kuzel, TM; Pezen, DS; Querfeld, C; Rademaker, A; Rosen, ST; Yarosh, DB, 2011
)
2.1
"Temozolomide (TMZ) is an oral alkylating agent with activity in high and LGG."( Using different schedules of Temozolomide to treat low grade gliomas: systematic review of their efficacy and toxicity.
Athanasiou, T; Lashkari, HP; Moreno, L; Saso, S; Zacharoulis, S, 2011
)
1.38
"Temozolomide (TMZ) is an alkylating agent that has yielded significant benefits and is a current standard agent in the treatment of malignant gliomas. "( Gene expression profiles predicting the response to IFN-β and a combination of temozolomide and IFN-β in malignant gliomas.
Fukushima, T; Katayama, Y; Ogino, A; Ohta, T; Okamoto, Y; Sano, E; Tashiro, S; Tsumoto, K; Watanabe, T; Yachi, K; Yoshino, A, 2011
)
2.04
"Temozolomide is an active drug against gliomas in adults. "( Temozolomide in relapsed pediatric brain tumors: 14 cases from a single center.
Akyüz, C; Büyükpamukçu, M; Demir, HA; Kutluk, T; Varan, A; Yalçin, B, 2012
)
3.26
"Temozolomide (TMZ) is a novel cytotoxic alkylating agent for chemotherapy of malignant gliomas. "( Chemoresistance to temozolomide in human glioma cell line U251 is associated with increased activity of O6-methylguanine-DNA methyltransferase and can be overcome by metronomic temozolomide regimen.
Dong, XT; Li, JM; Li, Y; Pan, Q; Wang, HM; Wang, W; Yang, XJ, 2012
)
2.15
"Temozolomide (TMZ) is an alkylating agent used for treating gliomas. "( Acquisition of chemoresistance in gliomas is associated with increased mitochondrial coupling and decreased ROS production.
Gillespie, GY; Griguer, CE; Moellering, DR; Oliva, CR, 2011
)
1.81
"Temozolomide (TMZ) is an alkylating, antineoplastic agent which is being used to treat cases of refractory anaplastic astrocytoma, newly-diagnosed glioblastoma multiforme and metastatic melanoma. "( Temozolomide-related infections: review of the literature.
Aksoy, S; Altundag, K; Ararat, E; Kizilarslanoglu, MC; Sahin, I; Yildirim, NO,
)
3.02
"Temozolomide is an oral chemotherapy agent, which has been used as a salvage therapy to treat aggressive pituitary adenomas and carcinomas, including prolactinomas, unresponsive to all conventional treatment."( Temozolomide in the management of dopamine agonist-resistant prolactinomas.
Aylwin, SJ; Barazi, S; Buchanan, CR; Dworakowska, D; Gilbert, JA; Hampton, T; King, AP; Landau, DB; Lipscomb, D; Riordan-Eva, P; Thomas, NW; Whitelaw, BC, 2012
)
2.54
"Temozolomide is an oral alkylating agent with clinical activity against glioblastoma multiforme (GM). "( Temozolomide-associated bronchiolitis obliterans organizing pneumonia successfully treated with high-dose corticosteroid.
Ban, HJ; Chi, SY; Kang, HW; Kim, KS; Kim, TO; Kim, YC; Kim, YI; Kwon, YS; Lim, SC; Oh, IJ, 2012
)
3.26
"Temozolomide (TMZ) is an alkylating chemotherapeutic agent that prolongs the survival of patients with glioblastoma. "( Distinct molecular mechanisms of acquired resistance to temozolomide in glioblastoma cells.
Florea, AM; Happold, C; Reifenberger, G; Roth, P; Schmidt, N; Silginer, M; Weller, M; Wick, W, 2012
)
2.07
"Temozolomide is an alkylating agent used in the treatment of gliomas and, more recently, aggressive pituitary adenomas and carcinomas. "( Temozolomide in aggressive pituitary adenomas and carcinomas.
Fadul, CE; Horvath, E; Kovacs, K; Ortiz, LD; Rotondo, F; Scheithauer, BW; Syro, LV; Uribe, H, 2012
)
3.26
"Temozolomide is an effective agent in HGGs with favorable outcome and low toxicity profile even in advanced age."( Long term experience in high grade glial tumors with temozolomide.
Akmansu, M; Baykara, M; Benekli, M; Buyukberber, S; Coskun, U; Demirci, U; Uner, A; Yamac, D; Yaman, E,
)
1.82
"Temozolomide (TMZ) is an oral alkylating agent widely used in the treatment of refractory glioma. "( The synergistic therapeutic effect of temozolomide and hyperbaric oxygen on glioma U251 cell lines is accompanied by alterations in vascular endothelial growth factor and multidrug resistance-associated protein-1 levels.
Cao, K; Li, QY; Lu, PS; Lu, XY; Yuan, ZC, 2012
)
2.09
"Temozolomide is an attractive candidate treatment in neuroblastoma with methylated MGMT, especially in central nervous system relapsed cases."( Efficacy of temozolomide in a central nervous system relapse of neuroblastoma with O 6 -methylguanine methyltransferase (MGMT) promoter methylation.
Marutsuka, K; Moritake, H; Nunoi, H; Shimonodan, H; Takeshima, H; Yamada, A; Yokogami, K, 2013
)
1.49
"Temozolomide (TMZ) is an antineoplastic alkylating agent with activity against serious and aggressive types of brain tumours. "( In situ electrochemical evaluation of anticancer drug temozolomide and its metabolites-DNA interaction.
Lopes, IC; Oliveira, SC; Oliveira-Brett, AM, 2013
)
2.08
"Temozolomide (TMZ) is a prodrug for an alkylating agent used for the treatment of malignant brain tumors. "( An efficient and practical radiosynthesis of [11C]temozolomide.
Carlin, SM; Hooker, JM; Moseley, CK; Neelamegam, R, 2012
)
2.07
"Temozolomide is an alkylating agent approved for treating malignant gliomas."( Impact of temozolomide on gonadal function in patients with primary malignant brain tumors.
Blackwood, R; Brown, M; Harmon, M; Lesser, G; Lovato, J; Strowd, RE; Yalcinkaya, T, 2013
)
1.51
"Temozolomide (TMZ) is an alkylating agent and was a first-line chemotherapeutic agent for malignant gliomas. "( DNA mismatch repair protein (MSH6) correlated with the responses of atypical pituitary adenomas and pituitary carcinomas to temozolomide: the national cooperative study by the Japan Society for Hypothalamic and Pituitary Tumors.
Amano, K; Arita, K; Asano, K; Fujio, S; Fukuhara, N; Hirohata, T; Hizuka, N; Ikeda, H; Ishii, Y; Isozaki, O; Iwai, Y; Kawamata, T; Matsuno, A; Nishioka, H; Ogawa, Y; Osamura, RY; Sakata, K; Shimatsu, A; Tahara, S; Takano, K; Takano, S; Teramoto, A; Tominaga, A; Yamada, S, 2013
)
2.04
"Temozolomide is an imidazotetrazine with a mechanism of action and efficacy similar to dacarbazine (DTIC). "( Temozolomide: a novel oral alkylating agent.
Danson, SJ; Middleton, MR, 2001
)
3.2
"Temozolomide (TZM) is a novel methylating agent currently under investigation for treatment of recurrent high-grade gliomas. "( Poly(ADP-ribose) polymerase inhibitor increases growth inhibition and reduces G(2)/M cell accumulation induced by temozolomide in malignant glioma cells.
Graziani, G; Navarra, P; Portarena, I; Scerrati, M; Tentori, L; Torino, F, 2002
)
1.97
"Temozolomide is a novel second-generation oral alkylating agent with demonstrated efficacy and safety in patients with recurrent glioblastoma multiforme (GBM) and anaplastic astrocytoma (AA). "( A phase II study of temozolomide in patients with newly diagnosed supratentorial malignant glioma before radiation therapy.
Friedman, HS; Gilbert, MR; Kuttesch, JF; Olson, JJ; Prados, MD; Reaman, GH; Zaknoen, SL, 2002
)
2.08
"Temozolomide is a new cytotoxic alkylating agent that has recently been approved in Portugal for the treatment of recurrent high-grade glioma. "( Temozolomide in second-line treatment after prior nitrosurea-based chemotherapy in glioblastoma multiforme: experience from a Portuguese institution.
Albano, J; Cernuda, M; Garcia, I; Lima, L; Oliveira, C; Portela, I; Teixeira, MM, 2002
)
3.2
"Temozolomide is an alkylating cytostatic drug that finds increasing application in the treatment of melanoma, anaplastic astrocytoma and glioblastoma multiforme. "( Temozolomide induces apoptosis and senescence in glioma cells cultured as multicellular spheroids.
Arnold, H; Damasceno, R; Günther, W; Pawlak, E; Terzis, AJ, 2003
)
3.2
"Temozolomide is a prodrug that undergoes spontaneous chemical degradation at physiologic pH to form the highly reactive alkylating agent, methyl-triazenyl imidazole carboxamide (MTIC). "( Plasma and cerebrospinal fluid pharmacokinetics of intravenous temozolomide in non-human primates.
Balis, FM; Godwin, K; McCully, C; Patel, M, 2003
)
2
"Temozolomide (TMZ) is a chemotherapeutic agent used in the treatment of high-grade brain tumors. "( Formation of DNA adducts and induction of lacI mutations in Big Blue Rat-2 cells treated with temozolomide: implications for the treatment of low-grade adult and pediatric brain tumors.
Berger, MS; Bodell, WJ; Gaikwad, NW; Miller, D, 2003
)
1.98
"Temozolomide is an imidazotetrazine with a mechanism of action similar to dacarbazine and equivalent activity in melanoma. "( Randomized phase II study of temozolomide given every 8 hours or daily with either interferon alfa-2b or thalidomide in metastatic malignant melanoma.
Arance, A; Ashcroft, L; Clamp, A; Danson, S; Hodgetts, J; Lomax, L; Lorigan, P; Middleton, MR; Ranson, M; Thatcher, N, 2003
)
2.05
"Temozolomide (TMZ) is an oral alkylating agent with a good safety profile and proven efficacy in the treatment of malignant glioma. "( Phase I study of temozolamide (TMZ) combined with procarbazine (PCB) in patients with gliomas.
Foster, T; Newlands, ES; Zaknoen, S, 2003
)
1.76
"Temozolomide (TMZ) is a lipophilic methylating agent used in the therapy of malignant melanoma and other tumors."( Inhibition of angiogenesis by non-toxic doses of temozolomide.
Kurzen, H; Möhler, T; Näher, H; Schmitt, S, 2003
)
1.29
"Temozolomide is an oral alkylating agent that readily crosses the blood-brain barrier and has activity in patients with advanced melanoma. "( A phase I (tumour site-specific) study of carboplatin and temozolomide in patients with advanced melanoma.
Boxall, J; Marples, M; Meyer, T; Napier, MP; Rustin, GJ; Strauss, SJ, 2003
)
2.01
"Temozolomide (TMZ) is a DNA methylating agent that has shown promising antitumor activity in recent clinical trials against high grade gliomas, metastatic melanoma, and brain lymphoma. "( Systemic administration of GPI 15427, a novel poly(ADP-ribose) polymerase-1 inhibitor, increases the antitumor activity of temozolomide against intracranial melanoma, glioma, lymphoma.
D'Amati, G; Graziani, G; Kalish, V; Leonetti, C; Portarena, I; Scarsella, M; Tentori, L; Vergati, M; Xu, W; Zhang, J; Zupi, G, 2003
)
1.97
"Temozolomide (TMZ) is a DNA alkylating agent currently used as adjuvant treatment for anaplastic astrocytomas. "( Inhibition of DNA repair for sensitizing resistant glioma cells to temozolomide.
Bedwell, J; Germano, IM; Kanzawa, T; Kondo, S; Kondo, Y, 2003
)
2
"Temozolomide is an oral alkylating chemotherapy agent licensed for the treatment of recurrent high-grade gliomas, anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM)."( Temozolomide in radio-chemotherapy combined treatment for newly-diagnosed glioblastoma multiforme: phase II clinical trial.
Campanella, C; Costa, A; Fedele, F; Frati, A; Frati, L; Gagliardi, FM; Innocenzi, G; Lanzetta, G; Minniti, G; Nappa, M; Rozzi, A; Salvati, M; Vecchione, A,
)
2.3
"Temozolomide is an alkylating agent that mediates its cytotoxic effects via O(6)-methylguanine (O(6)-meG) adducts in DNA and their recognition and processing by the postreplication mismatch repair system (MMR). "( Dual repair modulation reverses Temozolomide resistance in vitro.
Barvaux, VA; Brown, R; Margison, GP; McElhinney, RS; McMurry, TB; Ranson, M, 2004
)
2.05
"Temozolomide appears to be an ideal, first-line, single-agent, with a safe profile and demonstrated HQL benefits in patients with high-grade gliomas."( Temozolomide as first-line agent in treating high-grade gliomas: phase II study.
Andreuccetti, M; Benvenuti, L; Buxton, N; Caprio, A; Carnesecchi, S; Chibbaro, S; Faggionato, F; Gagliardi, R; Galli, C; Pulerà, F; Serino, D,
)
3.02
"Temozolomide is an oral chemotherapeutic agent with efficacy against malignant gliomas and a favorable safety profile."( Phase II study of temozolomide without radiotherapy in newly diagnosed glioblastoma multiforme in an elderly populations.
Barrie, M; Braguer, D; Chinot, OL; Dufour, H; Figarella-Branger, D; Frauger, E; Grisoli, F; Hoang-Xuan, K; Martin, PM; Moktari, K; Palmari, J; Peragut, JC, 2004
)
1.38
"Temozolomide (Temodal) is an oral imidazotetrazine. "( Prolonged schedule of temozolomide (Temodal) plus liposomal doxorubicin (Caelyx) in advanced solid cancers.
Awada, A; de Valeriola, D; Dubuisson, M; Gil, T; Klastersky, J; Moerman, C; Piccart, MJ; Sales, F; Vereecken, P, 2004
)
2.08
"Temozolomide is a well-tolerated oral alkylating agent with activity in the CNS. "( Temozolomide for the treatment of brain metastases associated with metastatic melanoma: a phase II study.
Agarwala, SS; Atkins, M; Buzaid, A; Czarnetski, B; Dreno, B; Gore, M; Kirkwood, JM; Rankin, EM; Skarlos, D; Thatcher, N, 2004
)
3.21
"Temozolomide is a well-tolerated alkylating agent, that is able to permeate the blood-brain barrier (BBB), and has additive cytotoxicity when given with radiotherapy (RT). "( Salvage chemotherapy with temozolomide in primary CNS lymphomas: preliminary results of a phase II trial.
Abbadessa, A; Bernardi, D; Bordonaro, R; Candela, M; Dell'Oro, S; Ferreri, AJ; Franceschi, E; Latte, G; Mason, W; Pace, A; Perry, J; Reni, M; Stelitano, C; Villa, E; Zaja, F, 2004
)
2.07
"Temozolomide is an oral alkylating agent that can cross the blood-brain barrier and in phase II and III trials, patients with advanced metastatic melanoma achieved overall response rates of 13 to 21%."( The effect of temozolomide-based chemotherapy in patients with cerebral metastases from melanoma.
Bafaloukos, D; Briassoulis, E; Christodoulou, C; Fountzilas, G; Gogas, H; Hatzichristou, H; Kalofonos, HP; Linardou, H; Panagiotou, P; Tsoutsos, D, 2004
)
1.41
"Temozolomide (TMZ) is a new oral alkylating agent which has proven to be as active as dacarbazine (DTIC) in the treatment of melanoma, but with a lower toxicity. "( Temozolomide and interferon-alpha in metastatic melanoma: a phase II study of the Italian Melanoma Intergroup.
Amaducci, L; Biasco, G; Guida, M; Leoni, M; Michiara, M; Poletti, P; Ravaioli, A; Ridolfi, R; Romanini, A; Sileni, VC, 2004
)
3.21
"Temozolomide is an oral alkylating agent shown to have modest efficacy in the treatment of glioblastoma multiforme. "( Transcriptional targeting of adenovirally delivered tumor necrosis factor alpha by temozolomide in experimental glioblastoma.
Gillespie, GY; Kufe, DW; Weichselbaum, RR; Yamini, B; Yu, X, 2004
)
1.99
"Temozolomide is an alkylating agent that mediates its cytotoxic effects via O(6)-methylguanine (O(6)-meG) adducts in DNA. "( Sensitization of a human ovarian cancer cell line to temozolomide by simultaneous attenuation of the Bcl-2 antiapoptotic protein and DNA repair by O6-alkylguanine-DNA alkyltransferase.
Barvaux, VA; Gillum, AM; Lorigan, P; Margison, GP; McElhinney, RS; McMurry, TB; Ranson, M, 2004
)
2.02
"Temozolomide (TMZ) is a methylating agent with promising antitumor efficacy for the treatment of melanomas and intermediate-grade gliomas. "( The piperidine nitroxide Tempol potentiates the cytotoxic effects of temozolomide in human glioblastoma cells.
Cereda, E; Gariboldi, MB; Monti, E; Ravizza, R, 2004
)
2
"Temozolomide seems to be a promising drug in the chemotherapy of malignant gliomas and can be applied as a second line chemotherapy, as well."( Temozolomide chemotherapy of patients with recurrent anaplastic astrocytomas and glioblastomas.
Afra, D; Sipos, L; Vitanovics, D, 2004
)
2.49
"Temozolomide (TMZ) is an oral alkylating agent that produces methyl adducts at the 0.6 position of guanine. "( Temozolomide and cisplatin versus temozolomide in patients with advanced melanoma: a randomized phase II study of the Hellenic Cooperative Oncology Group.
Bafaloukos, D; Briassoulis, E; Chalkidou, S; Christodoulou, C; Efstathiou, E; Fountzilas, G; Gogas, H; Iconomou, T; Kalofonos, H; Kouroussis, C; Linardou, E; Panagiotou, P; Polyzos, A; Tsoutsos, D, 2005
)
3.21
"Temozolomide (TMZ) is a promising new drug that seems to be effective in patients with recurrent disease."( Adjuvant chemotherapy in the treatment of high grade gliomas.
Brandes, AA; Lonardi, S; Tosoni, A, 2005
)
1.05
"Temozolomide (TMZ) is an oral alkylating agent; it produces DNA methyl adducts, which are removed by the DNA repair enzyme AGAT. "( Temozolomide and cisplatin in avdanced malignant melanoma.
Ascierto, PA; Comella, G; Daponte, A; Gravina, A; Melucci, M; Ottaiano, A; Palmieris, G; Scala, S; Simeone, E,
)
3.02
"Temozolomide (TMZ) is an alkylating agent that was approved for anaplastic astrocytoma and glioblastoma. "( Optimal role of temozolomide in the treatment of malignant gliomas.
Hegi, ME; Stupp, R; van den Bent, MJ, 2005
)
2.12
"Temozolomide (TMZ) is an oral alkylating agent with demonstrated efficacy as therapy for glioblastoma multiforme (GBM) and anaplastic astrocytoma. "( Economic evaluation of temozolomide in the treatment of recurrent glioblastoma multiforme.
Hallinen, T; Kivioja, A; Martikainen, JA; Vihinen, P, 2005
)
2.08
"Temozolomide is an oral alkylating agent that has equivalent activity to dacarbazine, but it has the advantage of CNS penetration."( A phase II study of biochemotherapy for advanced melanoma incorporating temozolomide, decrescendo interleukin-2 and GM-CSF.
Anderson, C; Baron, A; Gibbs, P; Gonzalez, R; Lewis, KD; O'Day, S; Richards, J; Russ, P; Weber, J; Zeng, C, 2005
)
1.28
"Temozolomide is a DNA-methylating agent used in the treatment of malignant gliomas. "( Poly(ADP-ribose) polymerase-1 inhibition reverses temozolomide resistance in a DNA mismatch repair-deficient malignant glioma xenograft.
Ali-Osman, F; Bigner, DD; Cheng, CL; Dolan, ME; Friedman, HS; Johnson, SP; Keir, ST; Li, H; Modrich, P; Quinn, JA; Salzman, AL; Szabo, C, 2005
)
2.02
"Temozolomide is a well-tolerated agent that results in objective responses and stabilisation of disease."( Phase II study of two-weekly temozolomide in patients with high-grade gliomas.
Ashley, D; Cher, L; Dowling, A; Jennens, R; Rosenthal, MA; Wong, S; Woods, AM, 2006
)
1.35
"Temozolomide appears to be an ideal, first-line, single-agent, with a safe profile and demonstrated HQL benefits in patients with high-grade gliomas."( Surgery, radiotherapy and temozolomide in treating high-grade gliomas.
Barbarisi, M; Moraci, A; Moraci, M; Parlato, C, 2006
)
1.36
"Temozolomide (TMZ) is a methylating agent which prolongs survival when administered during and after radiotherapy in the first-line treatment of glioblastoma and which also has significant activity in recurrent disease. "( O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells.
Hermisson, M; Kaina, B; Klumpp, A; Nagel, G; Roos, W; Weller, M; Wick, W; Wischhusen, J, 2006
)
2.02
"Temozolomide (TMZ) is an alkylating agent earlier approved for recurrent anaplastic astrocytoma and approved for the treatment of newly diagnosed glioblastoma in the USA and Europe in 2005. "( Temozolomide: a milestone in the pharmacotherapy of brain tumors.
Steinbach, JP; Weller, M; Wick, W, 2005
)
3.21
"Temozolomide is an oral alkylating agent that crosses the blood-brain barrier, and has preclinical activity in breast cancer. "( Temozolomide in metastatic breast cancer (MBC): a phase II trial of the National Cancer Institute of Canada - Clinical Trials Group (NCIC-CTG).
Bordeleau, L; Charpentier, D; Crump, M; Eisenhauer, E; Matthews, S; Trudeau, ME; Yelle, L, 2006
)
3.22
"Temozolomide is a rapidly absorbed chemotherapeutic agent, achieving significant central nervous system penetration. "( Alternating chemo-immunotherapy with temozolomide and low-dose interleukin-2 in patients with metastatic melanoma.
Hansson, J; Månsson-Brahme, E; Masucci, GV; Nilsson, B; Ragnarsson-Olding, B; Wagenius, G, 2006
)
2.05
"Temozolomide is a novel oral alkylating agent, active against metastatic melanoma. "( Phase II multicentre study of temozolomide in combination with interferon alpha-2b in metastatic malignant melanoma.
Crespo, C; del Muro, XG; Filipovich, E; García, M; Germà-Lluch, JR; López, JJ; Pérez, X; Rifà, J; Tres, A; Valladares, M, 2006
)
2.07
"Temozolomide is a recently introduced alkylating agent that has yielded a significant benefit in the treatment of high-grade gliomas."( Genetic alterations associated with acquired temozolomide resistance in SNB-19, a human glioma cell line.
Auger, N; Dutrillaux, B; Idbaih, A; Legrier, ME; Poupon, MF; Sanson, M; Thillet, J; Wanherdrick, K, 2006
)
1.31
"Temozolomide (TMZ) is a DNA methylating agent that has shown promising antitumor activity against high grade glioma. "( Potentiation of antiglioma effect with combined temozolomide and interferon-beta.
Hong, YK; Joe, YA; Kim, TG; Park, JA, 2006
)
2.03
"Temozolomide is an alkylating agent with activity in the treatment of melanoma metastatic to the brain. "( A phase I/II study of lomustine and temozolomide in patients with cerebral metastases from malignant melanoma.
Bate, SC; Beirne, DA; Eisen, TG; Gibbens, IM; Gore, ME; Hughes, SA; Larkin, JM; Patel, PM; Thomas, K, 2007
)
2.06
"Temozolomide (TMZ) is a methylating agent that spontaneously decomposes into the active metabolite of dacarbazine, the most effective agent for the systemic treatment of melanoma."( Combined effect of temozolomide and hyperthermia on human melanoma cell growth and O6-methylguanine-DNA methyltransferase activity.
Bonmassar, E; Caporali, S; Caporaso, P; D'Atri, S; Falcinelli, S; Pagani, E; Pepponi, R; Turriziani, M, 2007
)
1.39
"Temozolomide is an oral alkylating agent that is considered to be a well-tolerated chemotherapeutic agent."( [Temozolomide, an oral chemotherapeutic agent with potential severe toxicity].
Gijtenbeek, JM; Kappelle, AC; Soetekouw, PM; van der Maazen, RW; van Herpen, CM, 2007
)
1.97
"Temozolomide is a novel second-generation alkylating agent that has shown efficacy for the treatment of high-grade gliomas."( [Glioma therapy up-date].
Dalmau, J; de la Fuente, BP; Rosenfeld, M, 2007
)
1.06
"Temozolomide is an alkylating agent used frequently in the management of gliomas. "( Prolonged and severe myelosuppression in two patients after low-dose temozolomide treatment- case study and review of literature.
Brown, MP; Selva-Nayagam, S; Singhal, N, 2007
)
2.02
"Temozolomide is an oral alkylating agent recently approved for the treatment of glioblastoma multiforme. "( Temozolomide-associated organizing pneumonitis.
Aubrey, MC; Lim, KG; Limper, AH; Maldonado, F, 2007
)
3.23
"Temozolomide is an oral cytotoxic agent that has demonstrated its interest in high grade glioma tumors. "( Temozolomide treatment of an adult with a relapsing medulloblastoma.
Bay, JO; Durando, X; Gilliot, O; Irthum, B; Thivat, E; Verrelle, P; Vincent, C, 2007
)
3.23
"Temozolomide is an effective chemotherapeutic agent for patients with glioblastoma multiforme, but it induces significant lymphopenia."( Immunological responses in a patient with glioblastoma multiforme treated with sequential courses of temozolomide and immunotherapy: case study.
Aldape, K; Archer, GE; Bigner, DD; Crutcher, L; Dey, M; Gilbert, M; Hassenbusch, SJ; Heimberger, AB; Hussain, SF; Mitchell, DA; Sampson, JH; Sawaya, R; Schmittling, B; Sun, W, 2008
)
1.28
"Temozolomide is a methylating agent that is typically administered once daily."( Multi-institutional phase II study of temozolomide administered twice daily in the treatment of recurrent high-grade gliomas.
Alavi, J; Balmaceda, C; Chen, J; Cheung, YK; Fine, RL; Fisher, PG; Pannullo, S; Peereboom, D; Sisti, M, 2008
)
1.34
"Temozolomide is an oral alkylating agent used in the treatment of metastatic melanoma. "( Temozolomide-induced desquamative skin rash in a patient with metastatic melanoma.
Neff, WJ; Nystrom, KK; Pick, AM, 2008
)
3.23
"Temozolomide is a proautophagic (type II programmed cell death) drug and can thus circumvent part of the glioblastoma resistance to apoptosis."( [The sodium pump could constitute a new target to combat glioblastomas].
Kiss, R; Lefranc, F; Mijatovic, T, 2008
)
1.07
"Temozolomide (SCH 52365) is an imidazotetrazine derivative which exhibits broad spectrum activity against murine tumors and is structurally related to dacarbazine (DTIC). "( A phase II study of temozolomide in advanced untreated pancreatic cancer.
Feld, R; Hedley, D; Moore, MJ; Oza, A; Siu, LL, 1998
)
2.07
"Temozolomide (TMZ) is a new imidazotetrazine derivative with early clinical activity in glioma and melanoma. "( Phase I trial of temozolomide (NSC 362856) in patients with advanced cancer.
Ames, MM; Buckner, JC; Burch, PA; Dhodapkar, M; Pitot, HC; Reid, JM; Rubin, J; Suman, VJ, 1997
)
2.08
"Temozolomide (TMZ) is an oral imidazotetrazinone that is spontaneously converted to 5-(3-methyltriazen-1-yl)imidazole-4-carboxamide (MTIC) at physiological pH. "( A Phase I and pharmacokinetic study of temozolomide and cisplatin in patients with advanced solid malignancies.
Agarwala, SS; Baker, SD; Barrington, R; Britten, CD; Diab, SG; Eckardt, JR; Eckhardt, SG; Fraass, U; Hammond, LA; Johnson, T; Rowinsky, EK; Statkevich, P; Villalona-Calero, M; Von Hoff, DD, 1999
)
2.02
"Temozolomide is an imidazotetrazine alkylating agent which undergoes chemical conversion at physiological pH to the active species 5-(3-methyltriazene-1-yl)imidazole-4-carboxamide (MTIC) but is stable at acid pH. "( Effect of gastric pH on the relative oral bioavailability and pharmacokinetics of temozolomide.
Beale, P; Brada, M; Cutler, DL; Judson, I; Marco, A; Moore, S; Reidenberg, P; Statkevich, P, 1999
)
1.97
"Temozolomide (TMZ) is a new cytotoxic triazene compound of clinical interest that is able to generate methyl adducts at the O(6)-guanine of DNA, which can be repaired by O(6)-alkylguanine-DNA alkyltransferase (OGAT). "( O(6)-benzylguanine enhances the in vitro immunotoxic activity of temozolomide on natural or antigen-dependent immunity.
Alvino, E; Bonmassar, E; D'Atri, S; Lacal, PM; Nunziata, C; Pagani, E; Pepponi, R, 1999
)
1.98
"Temozolomide is a novel, oral, second-generation alkylating agent. "( Temozolomide in early stages of newly diagnosed malignant glioma and neoplastic meningitis.
Friedman, HS, 2000
)
3.19
"Temozolomide (TMZ) is a new, orally administered, second-generation imidazotetrazine prodrug with essentially 100% oral bioavailability that has demonstrated meaningful efficacy and an acceptable safety profile in the treatment of patients with recurrent glioblastoma multiforme. "( Future directions in the treatment of malignant gliomas with temozolomide.
Prados, MD, 2000
)
1.99
"Temozolomide is a novel methylating agent with proven efficacy against malignant gliomas (MGs) after systemic administration but with dose-limiting myelotoxicity."( Temozolomide delivered by intracerebral microinfusion is safe and efficacious against malignant gliomas in rats.
Archer, GE; Bigner, DD; Friedman, AH; Friedman, HS; Heimberger, AB; Hulette, C; McLendon, RE; Sampson, JH, 2000
)
2.47
"Temozolomide (TMZ) is a DNA-methylating agent that has recently been introduced into Phase II and III trials for the treatment of gliomas. "( p53 effects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells.
Berger, MS; Hirose, Y; Pieper, RO, 2001
)
1.99
"Temozolomide (TMZ) is a methylating agent of the imidotetrazine class, whose cytotoxic product is O(6)-methylguanine DNA adducts, which initiate a futile recycling of the mismatch repair pathway causing DNA strand breaks and apoptotic cell death in mismatch repair proficient cells. "( Temozolomide: the effect of once- and twice-a-day dosing on tumor tissue levels of the DNA repair protein O(6)-alkylguanine-DNA-alkyltransferase.
Gerson, SL; Haaga, J; Liu, L; Majka, S; Spiro, TP; Willson, JK, 2001
)
3.2
"Temozolomide is a novel oral alkylating agent that has been approved for the treatment of patients with refractory malignant glioma. "( New approaches for temozolomide therapy: use in newly diagnosed glioma.
Newlands, E; Stupp, R, 2001
)
2.08
"Temozolomide is an effective agent in the treatment of recurrent malignant gliomas. "( A phase II study of extended low-dose temozolomide in recurrent malignant gliomas.
Abrey, LE; Bazylewicz, KA; Khan, RB; Malkin, MG; Raizer, JJ, 2002
)
2.03
"Temozolomide is a novel oral alkylating agent that is effective against melanoma. "( Temozolomide in combination with docetaxel in patients with advanced melanoma: a phase II study of the Hellenic Cooperative Oncology Group.
Bafaloukos, D; Briassoulis, E; Fountzilas, G; Georgoulias, V; Gogas, H; Kalofonos, Ch; Karabelis, A; Kosmidis, P; Samantas, E; Skarlos, D, 2002
)
3.2
"Temozolomide is a novel oral alkylating agent with demonstrated efficacy as second-line therapy for patients with recurrent anaplastic astrocytoma and glioblastoma multiforme (GBM). "( Promising survival for patients with newly diagnosed glioblastoma multiforme treated with concomitant radiation plus temozolomide followed by adjuvant temozolomide.
de Tribolet, N; Dietrich, PY; Janzer, R; Leyvraz, S; Maeder, P; Maillard, I; Meuli, R; Miralbell, R; Mirimanoff, RO; Ostermann Kraljevic, S; Pica, A; Pizzolato, G; Porchet, F; Regli, L; Stupp, R, 2002
)
1.97
"Temozolomide (TZM) is a DNA-methylating agent that has recently been introduced into various clinical trials for treatment of solid or hematologic neoplasias, including brain lymphomas. "( Combined treatment with temozolomide and poly(ADP-ribose) polymerase inhibitor enhances survival of mice bearing hematologic malignancy at the central nervous system site.
Bonmassar, E; d'Amati, G; Graziani, G; Leonetti, C; Portarena, I; Scarsella, M; Tentori, L; Zupi, G, 2002
)
2.06
"Temozolomide (TMZ) is a newly approved alkylating agent for the treatment of malignant gliomas. "( Biochemical changes associated with a multidrug-resistant phenotype of a human glioma cell line with temozolomide-acquired resistance.
Berman, E; Gallo, JM; Ma, J; Murphy, M; O'Dwyer, PJ; Reed, K, 2002
)
1.97
"Temozolomide is a new anticancer agent which in the early clinical investigation has shown promising antitumor activity. "( Influence of O6-methylguanine on DNA damage and cytotoxicity of temozolomide in L1210 mouse leukemia sensitive and resistant to chloroethylnitrosoureas.
Bonfanti, M; Catapano, CV; Citti, L; D'Incalci, M; Taverna, P, 1992
)
1.96

Effects

Temozolomide has an established role in treating pituitary tumours which have demonstrated metastases. It may be regarded as the preferred treatment for recurrent anaplastic gliomas after radiotherapy.

Temozolomide (TMZ) has recently been recommended as a novel approach in the management of aggressive pituitary tumors. The drug has an acceptable tolerance in elderly patients with GBM and KPS less than 70.

ExcerptReferenceRelevance
"Temozolomide has an established role in treating pituitary tumours which have demonstrated metastases or which are refractory and progressing, despite all conventional treatment (so-called salvage treatment)."( How and when to use temozolomide to treat aggressive pituitary tumours.
Whitelaw, BC, 2019
)
1.56
"Temozolomide treatment has a wide range of efficacy in patients with pituitary carcinoma or locally aggressive pituitary adenoma. "( Salvage therapy with temozolomide in patients with aggressive or metastatic pituitary adenomas: experience in six cases.
Cangi, MG; Gill, AJ; Losa, M; Mazza, E; McCormack, A; Mortini, P; Motta, M; Reni, M; Talarico, A; Terreni, MR, 2010
)
2.12
"Temozolomide has an acceptable tolerance in elderly patients with GBM and KPS less than 70. "( Temozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trial.
Barrie, M; Beauchesne, P; Campello, C; Cartalat-Carel, S; Catry-Thomas, I; Chinot, O; Delattre, JY; Ducray, F; Gállego Pérez-Larraya, J; Guillamo, JS; Honnorat, J; Huchet, A; Matta, M; Mokhtari, K; Monjour, A; Taillandier, L; Tanguy, ML, 2011
)
3.25
"Temozolomide has an acceptable safety profile and may be regarded as the preferred treatment for recurrent anaplastic gliomas after radiotherapy."( [Favourable result for temozolomide in recurrent high-grade glioma].
Enting, RH; Kros, JM; Sillevis Smitt, PA; Taal, W; van den Bent, MJ; van der Rijt, CD; van Heuvel, I, 2005
)
1.36
"Temozolomide has an evolving role in the treatment of high grade gliomas. "( An Australian experience with temozolomide for the treatment of recurrent high grade gliomas.
Ashley, DL; Cher, L; Harris, MT; Rosenthal, MA, 2001
)
2.04
"Temozolomide (TMZ) has been widely used in GBM therapy with noticeable side effects."( Cold Plasma Discharge Tube Enhances Antitumoral Efficacy of Temozolomide.
Keidar, M; Keir, ST; Lin, L; Peters, KB; Sherman, JH; Yan, D; Yao, X, 2022
)
1.68
"Once temozolomide has failed, there is no recommended treatment option for pituitary carcinomas and aggressive pituitary tumors. "( Immunotherapy in aggressive pituitary tumors and carcinomas: a systematic review.
Ilie, MD; Jouanneau, E; Raverot, G; Vasiljevic, A, 2022
)
1.24
"Temozolomide (TMZ) has anti-proliferative and cytotoxic effects and is indicated for glioblastoma multiforme and recurrent mesenchymal astrocytoma."( Progress in research and development of temozolomide brain-targeted preparations: a review.
Chen, J; Fan, W; Fu, Z; Wu, X; Xu, Y; Yang, J, 2023
)
1.9
"Temozolomide (TMZ) has been used as standard-of-care for glioblastoma multiforme (GBM), but the resistance to TMZ develops quickly and frequently. "( Involvement of cell shape and lipid metabolism in glioblastoma resistance to temozolomide.
An, YJ; Choo, M; Kim, DH; Kim, HS; Ku, JL; Lee, SK; Mai, VH; Park, CK; Park, S, 2023
)
2.58
"Temozolomide has been used in patients with aggressive Nelson's with no consistent results."( Management of Nelson's Syndrome.
Fountas, A; Karavitaki, N, 2022
)
1.44
"Temozolomide (TMZ) has been determined to be the chemotherapeutic drug with efficacy for glioblastoma (GBM). "( Ultrasound-excited temozolomide sonosensitization induces necroptosis in glioblastoma.
Song, S; Tong, X; Wang, F; Wang, Y; Wen, B; Wu, H; Wu, Q; Xu, L; Yan, H; Zhou, Y, 2023
)
2.68
"Temozolomide (TMZ) has been used in the last 15 years in patients with aggressive pituitary tumors."( Knowing when to discontinue Temozolomide therapy in responding aggressive pituitary tumors and carcinomas: a systematic review and Padua (Italy) case series.
Barbot, M; Bergo, E; Caccese, M; Ceccato, F; Cerretti, G; Lombardi, G; Occhi, G; Padovan, M; Scaroni, C, 2023
)
1.93
"Temozolomide (TMZ) has been used as an initial therapy for gliomas."( LncRNA GSCAR promotes glioma stem cell maintenance via stabilizing SOX2 expression.
Chen, M; Chen, Y; Jiang, X; Jin, Z; Li, Y; Liu, B; Pu, J; Shi, Y; Yan, D; Yang, C; Yuan, Y; Zhai, H; Zhang, C; Zhang, Y, 2023
)
1.63
"Temozolomide has been mainly used for the treatment of malignant gliomas over a decade."( [Medical Treatments for Malignant Brain Tumor].
Kitamura, Y; Toda, M, 2023
)
1.63
"Temozolomide has shown a significant anti-proliferative activity on adrenocortical cancer (ACC) cells in vitro."( Activity and safety of temozolomide in advanced adrenocortical carcinoma patients.
Badalamenti, G; Basile, V; Berruti, A; Cerri, S; Cosentini, D; Dalla Volta, A; Ferrari, VD; Grisanti, S; Incorvaia, L; Laganà, M; Luppi, G; Musso, E; Perotti, P; Rapa, I; Russo, A; Sigala, S; Spallanzani, A; Terzolo, M; Volante, M, 2019
)
2.27
"Temozolomide (TMZ) has been the standard-of-care chemotherapy for glioblastoma (GBM) patients for more than a decade. "( Image-based metric of invasiveness predicts response to adjuvant temozolomide for primary glioblastoma.
Bendok, BR; Doyle, T; Hawkins-Daarud, A; Hu, LS; Jackson, PR; Johnston, SK; Massey, SC; Mrugala, MM; Porter, AB; Sarkaria, JN; Singleton, KW; Swanson, KR; Vora, S; White, H; Whitmire, P, 2020
)
2.24
"Temozolomide (TMZ) has been used as a first-line drug for the treatment of GBM for over a decade, but its treatment benefits are limited by acquired resistance."( The effect of polysaccharides from Cibotium barometz on enhancing temozolomide-induced glutathione exhausted in human glioblastoma U87 cells, as revealed by
Li, FF; Shi, Y; Wang, N; Wang, SQ; Wang, X; You, YL, 2020
)
1.52
"Temozolomide (TEM) has been reported to be active alone or in combination with capecitabine (CAP) in patients with neuroendocrine neoplasms (NENs). "( Temozolomide alone or in combination with capecitabine in patients with advanced neuroendocrine neoplasms: an Italian multicenter real-world analysis.
Antonuzzo, L; Barberis, M; Campana, D; Faviana, P; Fazio, N; Fumagalli, C; Gelsomino, F; Maisonneuve, P; Marconcini, R; Messerini, L; Puliafito, I; Rossi, G; Spada, F, 2021
)
3.51
"Temozolomide (TMZ) has been used to treat glioma."( Circ-VPS18 Knockdown Enhances TMZ Sensitivity and Inhibits Glioma Progression by MiR-370/RUNX1 Axis.
Jia, X; Li, W; Liu, Q; Ma, Q; Wang, X; Yan, P, 2021
)
1.34
"Temozolomide (TMZ) has been widely used as a first-line treatment for GBM."( Intranasal Delivery of Temozolomide-Conjugated Gold Nanoparticles Functionalized with Anti-EphA3 for Glioblastoma Targeting.
Li, N; Li, Y; Lv, Y; Sha, C; Sun, K; Tang, S; Wang, A; Wang, L; Yan, X; Yu, Y, 2021
)
1.65
"Temozolomide (TMZ) has been shown to determine radiological and biochemical response in malignant PCC/PGLs."( Temozolomide treatment of a malignant pheochromocytoma and an unresectable MAX-related paraganglioma.
Bertorelle, R; Ferrara, AM; Iacobone, M; Lombardi, G; Meringolo, D; Nardin, M; Opocher, G; Pambuku, A; Schiavi, F; Zagonel, V; Zovato, S, 2018
)
2.64
"Temozolomide (TMZ) has been considered to be one of the most effective chemotherapeutic agents to prolong the survival of patients with glioblastoma."( Differential Characterization of Temozolomide-Resistant Human Glioma Cells.
Chen, CC; Huang, BR; Lai, SW; Lin, C; Lin, HY; Liu, YS; Lu, DY; Tsai, CF, 2018
)
1.48
"Temozolomide has several effects on the immune system that are dependent on mode of delivery and the dosing strategy, which may have unpredicted effects on immunotherapy."( Temozolomide for immunomodulation in the treatment of glioblastoma.
Dastmalchi, F; Karachi, A; Mitchell, DA; Rahman, M, 2018
)
2.64
"Temozolomide (TMZ) has been reported to be useful as an adjunctive treatment for some patients."( Early Recognition and Initiation of Temozolomide Chemotherapy for Refractory, Invasive Pituitary Macroprolactinoma with Long-Term Sustained Remission.
Barkhoudarian, G; Eisenberg, A; Kelly, DF; Ogunbameru, R; Palejwala, SK; Wei, H, 2018
)
1.48
"Temozolomide has been available to oncologists for over 30 years. "( Role of Temozolomide in the Treatment of Cancers Involving the Central Nervous System.
Grossman, SA; Schreck, KC, 2018
)
2.36
"Temozolomide (TMZ) has been successfully used to treat a variety of malignancies, such as glioblastoma multiforme, astrocytoma, non-small-cell lung carcinoma."( Successful treatment with temozolomide in an elderly woman with advanced pulmonary large-cell neuroendocrine carcinoma: A case report.
Dong, XF; Hu, ZL; Lu, YF; Tang, S; Wei, J, 2018
)
1.5
"Temozolomide (TMZ) has been proposed in patients with aggressive pituitary neuroendocrine tumors (PitNETs) who do not respond to conventional treatments."( Temozolomide cytoreductive treatment in a giant cabergoline-resistant prolactin-secreting pituitary neuroendocrine tumor.
Albiger, N; Ceccato, F; Lombardi, G; Mazzai, L; Pambuku, A; Rolma, G; Scaroni, C; Zagonel, V, 2019
)
2.68
"Temozolomide (TMZ) has been the first-line chemotherapeutic agent used, although to achieve a satisfactory clinical effect."( Synergistic Suppression of Glioblastoma Cell Growth by Combined Application of Temozolomide and Dopamine D2 Receptor Antagonists.
Gao, L; Huang, X; Jiang, X; Li, J; Liu, X; Liu, Z; Zeng, T, 2019
)
1.46
"Temozolomide has an established role in treating pituitary tumours which have demonstrated metastases or which are refractory and progressing, despite all conventional treatment (so-called salvage treatment)."( How and when to use temozolomide to treat aggressive pituitary tumours.
Whitelaw, BC, 2019
)
1.56
"Temozolomide (TMZ) has established antineoplastic activity in the central nervous system in other disease states, with a favorable toxicity profile."( Temozolomide as a Single Agent Maintenance Therapy in Elderly Patients With Primary CNS Lymphoma.
Brenner, A; Butler, MJ; Faivre, G; Le, I, 2019
)
2.68
"Temozolomide (TMZ) has been proposed as a therapeutic option in aggressive pituitary tumors. "( Aggressive silent GH pituitary tumor resistant to multiple treatments, including temozolomide.
Batisse, M; Chazal, J; Durando, X; Kemeny, JL; Maqdasy, S; Montoriol, PF; Raverot, G; Sturm, N; Tauveron, I; Trouillas, J, 2013
)
2.06
"Temozolomide (TMZ) has received much attention, notably in the treatment of malignant glioma and malignant melanoma. "( Efficacy and safety between temozolomide alone and temozolomide-based double therapy for malignant melanoma: a meta-analysis.
Jia, HY; Jiang, G; Lei, TC; Li, RH; Liu, YQ; Sun, C, 2014
)
2.14
"Temozolomide (TMZ) alone has been proposed as a promising alternative to radiotherapy (RT) in elderly glioblastoma (GBM) patients. "( A meta-analysis of temozolomide versus radiotherapy in elderly glioblastoma patients.
Cai, S; Cheng, JX; Dong, Y; Liu, BL; Yin, AA; Zhang, LH; Zhang, X, 2014
)
2.17
"Temozolomide (TMZ) has remained the chemotherapy of choice in patients with glioblastoma multiforme (GBM) primarily due to the lack of more effective drugs. "( Oxidative cytotoxic agent withaferin A resensitizes temozolomide-resistant glioblastomas via MGMT depletion and induces apoptosis through Akt/mTOR pathway inhibitory modulation.
Cohen, MS; Grogan, PT; Sarkaria, JN; Timmermann, BN, 2014
)
2.1
"Temozolomide has recently emerged as an alternative option for PCNSL treatment."( MGMT promoter methylation and correlation with protein expression in primary central nervous system lymphoma.
Canal, F; Cavallin, S; Dei Tos, AP; Gherlinzoni, F; Scarpa, M; Scquizzato, E; Stefani, PM; Toffolatti, L, 2014
)
1.12
"Temozolomide has demonstrated a 2.5-month increase in overall survival and a 1.9-month increase in progression-free survival, relative to radiotherapy alone."( A review of the economic burden of glioblastoma and the cost effectiveness of pharmacologic treatments.
Hay, JW; Messali, A; Villacorta, R, 2014
)
1.12
"Temozolomide (TMZ) has been used for the treatment of glioblastoma."( Inhibition of JNK potentiates temozolomide-induced cytotoxicity in U87MG glioblastoma cells via suppression of Akt phosphorylation.
Chun, W; Kim, SS; Lee, HJ; Lee, JW; Lim, SY; Vo, VA, 2014
)
1.41
"Temozolomide (TMZ) has been showed to be an effective chemotherapeutic agent for glioblastoma treatment; however, the response rate is not satisfactory."( Synergistic Anti-Cancer Effects of Icariin and Temozolomide in Glioblastoma.
Guo, H; Guo, M; Wang, Y; Yang, L, 2015
)
1.4
"Temozolomide (TMZ) has been widely used in the treatment of glioblastoma (GBM), although inherent or acquired resistance restricts the application. "( Sulforaphane enhances temozolomide-induced apoptosis because of down-regulation of miR-21 via Wnt/β-catenin signaling in glioblastoma.
Lan, F; Pan, Q; Yu, H; Yue, X, 2015
)
2.17
"As temozolomide has activity in SCLC, we investigated combination therapy with talazoparib and found marked synergy in vitro and efficacy in vivo, which did not solely depend on SLFN11 or MGMT status."( PARP Inhibitor Activity Correlates with SLFN11 Expression and Demonstrates Synergy with Temozolomide in Small Cell Lung Cancer.
de Stanchina, E; Desmeules, P; Gardner, EE; Lok, BH; Ni, A; Poirier, JT; Powell, SN; Rekhtman, N; Riaz, N; Rudin, CM; Schneeberger, VE; Teicher, BA, 2017
)
1.19
"Temozolomide (TMZ) has been shown as an effective treatment option in aggressive pituitary adenomas and carcinomas. "( How effective is temozolomide for treating pituitary tumours and when should it be used?
Halevy, C; Whitelaw, BC, 2017
)
2.24
"Temozolomide (TMZ) has demonstrated clinical antitumor activity. "( The safety of temozolomide in the treatment of malignancies.
Hwu, WJ; Patel, SP; Trinh, VA, 2009
)
2.16
"Temozolomide has been used with thalidomide to treat gliomas."( Mechanism of thalidomide to enhance cytotoxicity of temozolomide in U251-MG glioma cells in vitro.
Gao, S; Ji, YW; Pan, Q; Yang, XJ; Zhang, WG, 2009
)
1.32
"Temozolomide has been used as a standard therapy for the treatment of newly diagnosed glioblastoma multiforme since 2005. "( Effectiveness of temozolomide for primary glioblastoma multiforme in routine clinical practice.
Baumert, BG; Leffers, P; Tjon-A-Fat, H; Twijnstra, A; van Genugten, JA, 2010
)
2.14
"Temozolomide has recently been used for atypical adenomas or pituitary carcinomas."( [Recent trends in the pathophysiology and treatment of pituitary adenomas].
Matsuno, A, 2009
)
1.07
"Temozolomide (TMZ) has been accepted as a standard antitumor drug for glioma worldwide. "( Flow cytometric analysis for the mechanism of the new antineoplastic agent temozolomide in glioma cells.
Kawamoto, K; Li, Y; Numa, Y; Oishi, T; Oshige, H; Yamahara, T; Zhen, Y, 2010
)
2.03
"Temozolomide chemotherapy has become part of the therapy used to treat glioblastoma multiforme and refractory anaplastic astrocytoma. "( Emergence of cytomegalovirus disease in patients receiving temozolomide: report of two cases and literature review.
Aguado, JM; García-Reyne, A; Juan, RS; Lalueza, A; Lizasoain, M; López-Medrano, F; Martínez, P; Meije, Y; Rodríguez, V, 2010
)
2.05
"Temozolomide (TM) has anti-tumor activity in patients with malignant glioma. "( Temozolomide/PLGA microparticles plus vatalanib inhibits tumor growth and angiogenesis in an orthotopic glioma model.
Liu, JM; Tang, GS; Wang, Y; Yue, ZJ; Zhang, H; Zhang, YH, 2010
)
3.25
"Temozolomide treatment has a wide range of efficacy in patients with pituitary carcinoma or locally aggressive pituitary adenoma. "( Salvage therapy with temozolomide in patients with aggressive or metastatic pituitary adenomas: experience in six cases.
Cangi, MG; Gill, AJ; Losa, M; Mazza, E; McCormack, A; Mortini, P; Motta, M; Reni, M; Talarico, A; Terreni, MR, 2010
)
2.12
"Temozolomide has significantly improved the outcome of patients with glioblastoma. "( [Benefit of a prolonged adjuvant treatment with temozolomide for the management of patients with glioblastoma].
Auberdiac, P; Cartier, L; Chargari, C; Forest, F; Fotso, MJ; Magné, N; Malkoun, N; Nuti, C; Pacaut, C; Peoc'h, M; Schmitt, T; Thorin, J, 2011
)
2.07
"Temozolomide has shown clinical effect in well-differentiated endocrine carcinomas."( Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy.
Busch, C; Knappskog, S; Oberg, K; Sebjornsen, S; Sorbye, H; Welin, S, 2011
)
1.42
"Oral temozolomide has shown similar efficacy to dacarbazine in phase III trials with median progression-free survival (PFS) of 2.1 months. "( First-line temozolomide combined with bevacizumab in metastatic melanoma: a multicentre phase II trial (SAKK 50/07).
Cathomas, R; Dummer, R; Gillessen, S; Goldinger, SM; Mamot, C; Michielin, O; Mjhic-Probst, D; Moch, H; Ochsenbein, A; Schläppi, M; Schönewolf, N; Schraml, PH; Seifert, B; Simcock, M; von Moos, R, 2012
)
1.28
"Temozolomide has an acceptable tolerance in elderly patients with GBM and KPS less than 70. "( Temozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trial.
Barrie, M; Beauchesne, P; Campello, C; Cartalat-Carel, S; Catry-Thomas, I; Chinot, O; Delattre, JY; Ducray, F; Gállego Pérez-Larraya, J; Guillamo, JS; Honnorat, J; Huchet, A; Matta, M; Mokhtari, K; Monjour, A; Taillandier, L; Tanguy, ML, 2011
)
3.25
"Temozolomide (TMZ) has recently been recommended as a novel approach in the management of aggressive pituitary tumors. "( Aggressive silent corticotroph adenoma progressing to pituitary carcinoma: the role of temozolomide therapy.
Cusimano, M; Fadul, CE; Gonzalez, R; Horvath, E; Kovacs, K; Moshkin, O; Ortiz, LD; Rotondo, F; Scheithauer, BW; Syro, LV; Uribe, H,
)
1.8
"Temozolomide has activity in relapsed SCLC, particularly for brain metastases. "( Phase II trial of temozolomide in patients with relapsed sensitive or refractory small cell lung cancer, with assessment of methylguanine-DNA methyltransferase as a potential biomarker.
Azzoli, CG; Chan, TA; Fiore, JJ; Ginsberg, MS; Heguy, A; Holodny, AI; Huberman, K; Kadota, K; Kris, MG; Krug, LM; Pietanza, MC; Riely, GJ; Rizvi, NA; Sima, CS; Sumner, DK; Travis, WD, 2012
)
2.16
"Temozolomide has quickly become part of the standard of care for the modern treatment of stage IV glioblastoma multiforme since its approval in 2005."( Temozolomide and other potential agents for the treatment of glioblastoma multiforme.
Chow, F; Cremer, N; Kim, W; Nagasawa, DT; Yang, I; Yew, A, 2012
)
2.54
"Temozolomide (TMZ) has anti-tumor activity in patients with malignant glioma. "( Combination hyperbaric oxygen and temozolomide therapy in C6 rat glioma model.
Bilir, A; Bozkurt, ER; Dagıstan, Y; Karaca, I; Ozar, E; Toklu, A; Yagmurlu, K, 2012
)
2.1
"Temozolomide (TMZ) has become a key therapeutic agent in patients with malignant gliomas; however, its survival benefit remains unsatisfactory. "( Valproic acid downregulates the expression of MGMT and sensitizes temozolomide-resistant glioma cells.
Hou, Y; Jeong, CH; Jeun, SS; Kim, SM; Lim, JY; Park, KY; Ryu, CH; Woo, JS; Yoon, WS, 2012
)
2.06
"Temozolomide monotherapy has limited effect in treatment of recurrent neuroendocrine carcinomas. "( Temozolomide as second or third line treatment of patients with neuroendocrine carcinomas.
Federspiel, B; Hansen, CP; Kjaer, A; Knigge, U; Langer, SW; Olsen, IH; Sørensen, JB, 2012
)
3.26
"Temozolomide (Temodar) has demonstrated clinical activity against melanoma equivalent to that of intravenous dacarbazine (DTIC). "( Phase II evaluation of temozolomide in metastatic choroidal melanoma.
Bedikian, AY; Eton, O; Papadopoulos, N; Plager, C; Ring, S, 2003
)
2.07
"Temozolomide has activity and a favorable safety profile in all dosing schedules tested."( Temozolomide: realizing the promise and potential.
Dolan, ME; Nagasubramanian, R, 2003
)
2.48
"Temozolomide has single-agent activity in patients with WHO grade II cerebral glioma, with modest improvement in quality of life and improvement in epilepsy control. "( Phase II study of primary temozolomide chemotherapy in patients with WHO grade II gliomas.
Abson, C; Ashley, S; Brada, M; Britton, J; Gonsalves, A; Hines, F; Sardell, S; Traish, D; Viviers, L; Westbury, C; Wilkins, P, 2003
)
2.06
"Temozolomide has shown activity and limited toxicity in patients with primary brain tumors at doses of 150-200 mg/m(2)/day on days 1-5 every 4 weeks. "( Dose-dense regimen of temozolomide given every other week in patients with primary central nervous system tumors.
Abdulkarim, B; Armand, JP; Cioloca, C; Djafari, L; Djazouli, K; Faivre, S; Guillamo, JS; Osorio, M; Parker, F; Raymond, E; Vera, K, 2004
)
2.08
"Temozolomide has shown antineoplastic activity against malignant gliomas and more recently was beneficial in one patient with gliomatosis cerebri."( Gliomatosis cerebri: clinical, neurochemical and neuroradiological response to temozolomide administration.
Ambrosetto, P; Barbiroli, B; Cortelli, P; Crinò, L; Franceschi, E; Lodi, R; Setola, E; Tonon, C, 2003
)
1.27
"Temozolomide has established activity in the treatment of recurrent glioblastoma multiforme (GBM). "( Phase 2 study of temozolomide and Caelyx in patients with recurrent glioblastoma multiforme.
Ashley, DM; Cher, LM; Chua, SL; Dowling, A; Rosenthal, MA; Wong, SS; Woods, AM, 2004
)
2.11
"Temozolomide (TMZ) has demonstrated activity and acceptable toxicity for the treatment of recurrent high-grade gliomas in prospective phase II studies. "( Temozolomide for the treatment of recurrent supratentorial glioma: results of a compassionate use program in Belgium.
Branle, F; Everaert, E; Joosens, E; Menten, J; Neyns, B; Strauven, T, 2004
)
3.21
"Temozolomide has been studied in soft-tissue sarcomas with varying dosing schedules. "( Temozolomide in uterine leiomyosarcomas.
Aghajanian, C; Anderson, S, 2005
)
3.21
"Temozolomide has promising therapeutic benefit and is well tolerated in patients with metastatic unresectable leiomyosarcoma. "( Temozolomide in uterine leiomyosarcomas.
Aghajanian, C; Anderson, S, 2005
)
3.21
"Temozolomide has an acceptable safety profile and may be regarded as the preferred treatment for recurrent anaplastic gliomas after radiotherapy."( [Favourable result for temozolomide in recurrent high-grade glioma].
Enting, RH; Kros, JM; Sillevis Smitt, PA; Taal, W; van den Bent, MJ; van der Rijt, CD; van Heuvel, I, 2005
)
1.36
"Temozolomide (TMZ) has shown efficacy in metastatic melanoma equal to that of dacarbazine (DTIC), the standard chemotherapeutic agent for melanoma. "( Temozolomide in combination with interferon-alfa versus temozolomide alone in patients with advanced metastatic melanoma: a randomized, phase III, multicenter study from the Dermatologic Cooperative Oncology Group.
Becker, JC; Dummer, R; Garbe, C; Kaufmann, R; Krengel, S; Kretschmer, L; Leiter, U; Linse, R; Mauch, C; Schadendorf, D; Sebastian, G; Spieth, K; Tilgen, W; Vogt, T; von den Driesch, P, 2005
)
3.21
"Temozolomide (TMZ) has shown modest efficacy in the treatment of recurrent brain metastasis (BM). "( Vinorelbine combined with a protracted course of temozolomide for recurrent brain metastases: a phase I trial.
Abrey, LE; Demopoulos, A; Malkin, MG; Omuro, AM; Raizer, JJ, 2006
)
2.03
"Temozolomide (TMZ) has shown efficacy in the treatment of metastatic melanoma."( Whole brain irradiation and temozolomide based chemotherapy in melanoma brain metastases.
Castel, T; Conill, C; Domingo-Doménech, J; Gallego, R; Jorcano, S; Malvehy, J; Puig, S; Sánchez, M; Vilella, R, 2006
)
1.35
"Temozolomide (TMZ) has demonstrated activity against melanoma and has been investigated as single agent or in combination."( Temozolomide in combination with celecoxib in patients with advanced melanoma. A phase II study of the Hellenic Cooperative Oncology Group.
Fountzilas, G; Frangia, K; Gogas, H; Mantzourani, M; Markopoulos, C; Middleton, M; Panagiotou, P; Papadopoulos, O; Pectasides, D; Polyzos, A; Stavrinidis, I; Tsoutsos, D; Vaiopoulos, G, 2006
)
2.5
"Temozolomide has shown some efficacy in metastatic melanoma and recently received extended approval to treat brain tumours. "( Dose-intensified bi-weekly temozolomide in patients with asymptomatic brain metastases from malignant melanoma: a phase II DeCOG/ADO study.
Egberts, F; Garbe, C; Hauschild, A; Kreissig, M; Linse, R; Mohr, P; Schadendorf, D; Thoelke, A; Tilgen, W; Trefzer, U; Ugurel, S; Vogt, T, 2006
)
2.07
"Temozolomide (TMZ) has demonstrated activity and acceptable toxicity for the treatment of recurrent malignant gliomas in carious prospective phase II studies. "( [Temozolomide in the treatment of recurrent malignant glioma].
Ishii, N; Iwasaki, Y; Kobayashi, H; Murata, J; Sawamura, Y, 2006
)
2.69
"Temozolomide has emerged as a new alternative treatment for PCNSL and constitutes an attractive option for the elderly because of its favorable toxicity profile."( Temozolomide and methotrexate for primary central nervous system lymphoma in the elderly.
Barrie, M; Carnin, C; Chinot, O; Hoang-Xuan, K; Omuro, AM; Taillandier, L, 2007
)
2.5
"Temozolomide has proven benefit in grade II/III gliomas progressive following standard therapy and when added to radiation for glioblastoma."( Temozolomide and radiation in low-grade and anaplastic gliomas: temoradiation.
Schiff, D, 2007
)
2.5
"Temozolomide (TMZ) has emerged as an active agent against malignant gliomas."( Management of glioblastoma.
Aoki, T; Hashimoto, N; Matsutani, M, 2007
)
1.06
"Temozolomide has become the standard of care in newly diagnosed glioblastoma."( [Chemotherapy for brain tumors in adult patients].
Weller, M, 2008
)
1.07
"Temozolomide has recently been introduced by Schering-Plough Ltd (Welwyn Garden City, UK) as a new treatment which merits further investigation in this situation."( Temozolomide (Temodal) for treatment of primary brain tumours.
MacConnachie, AM, 2000
)
2.47
"Temozolomide, a new drug, has shown promise in treating malignant gliomas and other difficult-to-treat tumors."( Temozolomide and treatment of malignant glioma.
Calvert, H; Friedman, HS; Kerby, T, 2000
)
2.47
"Temozolomide has an evolving role in the treatment of high grade gliomas. "( An Australian experience with temozolomide for the treatment of recurrent high grade gliomas.
Ashley, DL; Cher, L; Harris, MT; Rosenthal, MA, 2001
)
2.04
"Temozolomide has the potential to be a useful agent in the treatment of a variety of cancers."( Future directions for temozolomide therapy.
Yung, WK, 2001
)
1.35
"Temozolomide has shown efficacy in the treatment of metastatic melanoma similar to that of dacarbazine (DTIC), the standard chemotherapy, but with the added benefit of penetration into the central nervous system (CNS). "( Effect of temozolomide on central nervous system relapse in patients with advanced melanoma.
Brampton, MH; Calvert, AH; Middleton, MR; Paul, MJ; Rustin, G; Summers, Y; Thatcher, N, 2002
)
2.16

Actions

Temozolomide was chosen because of its previous success when combined with PARP inhibitors, dissimilar to other commonly used chemotherapies. The drug can cause early radiation induced injury which can mimic progressive tumor.

ExcerptReferenceRelevance
"Temozolomide was chosen because of its previous success when combined with PARP inhibitors, dissimilar to other commonly used chemotherapies."( Does PARP Inhibition Sensitize Chondrosarcoma Cell Lines to Chemotherapy or Radiotherapy? Results From a Three-dimensional Spheroid Cell Model.
Boveé, JVMG; Briaire-de Bruijn, IH; Palubeckaitė, I; van den Akker, BEWM; Venneker, S, 2023
)
1.63
"Temozolomide plays a role in treating melanoma refractory to immunomodulatory and mitogen-activated protein kinase-targeted approaches, but its efficacy is limited. "( Preclinical Activity of 4-Demethyl-4-cholesteryloxycarbonylpenclomedine in Melanoma.
Benes, EN; Friedlander, P; Jursic, B; Morgan, LR; Rodgers, AH, 2023
)
2.35
"Temozolomide may cause thrombocytopenia or neutropenia in 3-4% of glioblastoma patients, respectively. "( MGMT gene variants, temozolomide myelotoxicity and glioma risk. A concise literature survey including an illustrative case.
Altinoz, MA; Bolukbasi, FH; Ekmekci, CG; Elmaci, I; Sari, R; Sav, A; Yenmis, G, 2017
)
2.22
"Temozolomide (TMZ) may enhance antitumor immunity in patients with glioblastoma multiforme (GBM). "( Phase I/IIa trial of fractionated radiotherapy, temozolomide, and autologous formalin-fixed tumor vaccine for newly diagnosed glioblastoma.
Abe, T; Hashimoto, K; Ikuta, S; Ishihara, T; Ishikawa, E; Karasawa, K; Maruyama, T; Matsuda, M; Matsumura, A; Matsutani, M; Muragaki, Y; Nakazato, Y; Ohno, T; Tsuboi, K; Uemae, Y; Yamamoto, T, 2014
)
2.1
"Temozolomide plays a critical role in curing glioma at present. "( Liposome encapsulated of temozolomide for the treatment of glioma tumor: preparation, characterization and evaluation.
Gao, J; Huang, G; Liu, H; Wang, L; Wang, Z, 2015
)
2.16
"Temozolomide can cause early radiation induced injury which can mimic progressive tumor. "( Radiation induced early necrosis in patients with malignant gliomas receiving temozolomide.
Akmansu, M; Benekli, M; Buyukberber, S; Coskun, U; Kaya, AO; Oner, Y; Ozturk, B; Uncu, D; Yaman, E; Yildiz, R, 2010
)
2.03
"Temozolomide can cause Pneumocystis pneumonia."( Fatal pneumonia associated with temozolomide therapy in patients with malignant glioma.
Abe, S; Azuma, A; Fujita, K; Gemma, A; Hayashi, H; Kobayashi, K; Kokuho, N; Morimoto, T; Saito, Y; Tanaka, T, 2012
)
1.38
"Temozolomide (TMZ) displays efficacy for the treatment of metastatic melanoma."( [Temozolomide in patients with melanoma brain metastases treated with whole brain irradiation].
Castel, T; Conill, C; Fernández-Ibiza, J; Malvehy, J; Puig, S; Sánchez, M, 2004
)
1.96
"Temozolomide appears to produce few serious adverse effects and may also have a positive impact on health-related quality of life."( A rapid and systematic review of the effectiveness of temozolomide for the treatment of recurrent malignant glioma.
Cave, C; Dinnes, J; Huang, S; Milne, R, 2002
)
1.28

Treatment

Temozolomide (TMZ) treatment efficacy in glioblastoma (GBM) has been limited by resistance. Treatment induced cell cycle arrest, diminished cell viability, migration, invasion, and angiogenesis.

ExcerptReferenceRelevance
"Temozolomide-based treatment was administered to 64 of 68 (94.1%) patients, whereas RT + procarbazine, lomustine (CCNU), and vincristine (PCV) were applied in 2 of 69 (3.5%) patients."( Early Postoperative Treatment versus Initial Observation in CNS WHO Grade 2 and 3 Oligodendroglioma: Clinical Outcomes and DNA Methylation Patterns.
Aichholzer, M; Berghoff, AS; Dieckmann, K; Furtner, J; Goldberger, S; Hatziioannou, T; Heller, G; Leibetseder, A; Mair, MJ; Pichler, J; Preusser, M; Puhr, R; Tomasich, E; von Oertzen, T; Weis, S; Widhalm, G; Wöhrer, A, 2022
)
1.44
"Temozolomide treatment in 156/171 patients resulted in complete response in 9.6%, partial response in 30.1%, stable disease in 28.1%, and progressive disease in 32.2% of the patients."( Aggressive pituitary tumours and carcinomas, characteristics and management of 171 patients.
Burman, P; Dekkers, OM; Losa, M; McCormack, A; Petersenn, S; Popovic, V; Raverot, G; Theodoropoulou, M; Trouillas, J, 2022
)
1.44
"Temozolomide treatment induced cell cycle arrest, diminished cell viability, migration, invasion, and angiogenesis, and increased apoptosis and autophagy in glioblastoma, which was counteracted by overexpressing LINC00470 or SOX4 but was further promoted by LINC00470 knockdown."( Temozolomide protects against the progression of glioblastoma via SOX4 downregulation by inhibiting the LINC00470-mediated transcription factor EGR2.
Cui, Y; He, J; Li, W; Liu, P; Ma, W; Wang, M; Zhang, M, 2023
)
3.07
"Temozolomide (TMZ) treatment is the mainstay drug for GBM despite the rapid development of resistance in mesenchymal GBM."( NADPH Oxidase Subunit CYBB Confers Chemotherapy and Ferroptosis Resistance in Mesenchymal Glioblastoma via Nrf2/SOD2 Modulation.
Fong, IH; Lin, CM; Liu, HW; Setiawan, SA; Su, IC; Su, YK; Yadav, VK; Yeh, CT, 2023
)
1.63
"Temozolomide (TMZ) treatment efficacy in glioblastoma (GBM) has been limited by resistance. "( EPIC-0307-mediated selective disruption of PRADX-EZH2 interaction and enhancement of temozolomide sensitivity to glioblastoma via inhibiting DNA repair and MGMT.
Cui, X; Fang, C; Hong, B; Kang, C; Tan, Y; Tian, S; Wang, C; Wang, Q; Xiao, M; Xin, L; Xu, C; Xu, J; Yuan, X; Zhao, J; Zhu, Y, 2023
)
2.58
"Pre-temozolomide treatment, these data were 5.2 cm3 (IQR 1.9-12.3), 89.3% and 14.3% (2 intracranial and 2 spinal metastases)."( Efficacy and safety of temozolomide in the treatment of aggressive pituitary neuroendocrine tumours in Spain.
Araujo-Castro, M; Biagetti, B; Cámara, R; Fajardo, C; García-Centeno, R; Guerrero-Pérez, F; Hanzu, F; Iglesias, P; Lamas, C; Mora, M; Remon-Ruiz, P; Soto, A, 2023
)
1.7
"Temozolomide (TMZ) treatment efficacy in glioblastoma (GBM) patients has been limited by resistance in the clinic. "( Albumin-bound paclitaxel augment temozolomide treatment sensitivity of glioblastoma cells by disrupting DNA damage repair and promoting ferroptosis.
Huang, G; Li, Z; Qi, S; Qu, S; Wang, K; Ye, R; Yi, GZ; Zhang, H; Zhang, W; Zhu, T, 2023
)
2.63
"Temozolomide is used for treatment in high-risk groups with low treatment response of neuroblastomas."( Selenium enhances TRPA1 channel-mediated activity of temozolomide in SH-SY5Y neuroblastoma cells.
Övey, İS; Özkal, B, 2020
)
1.53
"Temozolomide (TMZ) treatment failed to induce apoptotic death in shBeclin1-transfected cells, contrary to control."( Autophagy inhibition reinforces stemness together with exit from dormancy of polydisperse glioblastoma stem cells.
Antonietti, P; Barthout, E; Battu, S; Begaud, G; Bessette, B; Brunel, A; Deluche, E; Durand, S; Hombourger, S; Jauberteau, MO; Kögel, D; Lalloué, F; Saada, S; Verdier, M, 2021
)
1.34
"SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. "( Modulation of temozolomide dose differentially affects T-cell response to immune checkpoint inhibition.
Azari, H; Dastmalchi, F; Flores, C; Huang, J; Karachi, A; Long, Y; Mitchell, DA; Rahman, M; Sayour, EJ; Yang, C, 2019
)
1.5
"Temozolomide treatment inhibited p50 recruitment to its cognate element as a function of Ser329 phosphorylation while concomitantly increasing p53 recruitment."( Temozolomide Treatment Induces lncRNA MALAT1 in an NF-κB and p53 Codependent Manner in Glioblastoma.
Bernal, GM; Cahill, KE; Crawley, CD; Khodarev, NN; Larsen, G; Mansour, NM; Nunez, L; Raleigh, DR; Spretz, R; Szymura, SJ; Uppal, A; Voce, DJ; Weichselbaum, RR; Wu, L; Yamini, B; Zhang, W, 2019
)
2.68
"Temozolomide treatment is continued for more than six cycles by 80.5 % of neuro-oncologists: 44.4 % only if there is residual disease; 27.8 % for 12 cycles even in the absence of residual disease; and 8.3 % until progression. "( Should we continue temozolomide beyond six cycles in the adjuvant treatment of glioblastoma without an evidence of clinical benefit? A cost analysis based on prescribing patterns in Spain.
Arranz, JL; Balañá, C; Benavides, M; Bugés, C; Cano, JM; de la Peñas, R; García-Bueno, JM; Gil, M; Lopez, D; Martin, JM; Molina-Garrido, MJ; Perez-Segura, P; Rodriguez, A; Sanz, SM; Sepúlveda, JM; Vaz, MA, 2014
)
2.17
"Temozolomide-treated cells had increased methylation of the cytosine-phosphate-guanine islands in the Wnt3a gene promoter, decreased expression of Wnt3a, disrupted glycogen synthase-3 kinase/β-catenin axis, reduced transcriptional activation of ABCB1, and a lower amount and activity of Pgp."( Temozolomide downregulates P-glycoprotein expression in glioblastoma stem cells by interfering with the Wnt3a/glycogen synthase-3 kinase/β-catenin pathway.
Annovazzi, L; Bosia, A; Caldera, V; Campia, I; Ghigo, D; Kopecka, J; Mellai, M; Riganti, C; Salaroglio, IC; Schiffer, D, 2013
)
2.55
"Temozolomide treatment of 293_pcDNA3.1 cells intensified the stochastic punctuated genome changes and CNAs, and significantly reduced viability and CFE."( Step-wise and punctuated genome evolution drive phenotype changes of tumor cells.
Andreieva, S; Huleyuk, N; Kavsan, V; Korets, K; Mykytenko, D; Stepanenko, A; Vassetzky, Y, 2015
)
1.14
"Temozolomide treatment significantly induced mRNA expression of nearly all investigated EMT markers in T98G glioma cells."( Epithelial-to-mesenchymal transition in paired human primary and recurrent glioblastomas.
Hattermann, K; Held-Feindt, J; Kubelt, C; Mehdorn, HM; Sebens, S, 2015
)
1.14
"Temozolomide-treated patients with the rs16906252 T genotype have better survival, irrespective of tumor methylation status."( The MGMT promoter SNP rs16906252 is a risk factor for MGMT methylation in glioblastoma and is predictive of response to temozolomide.
Cloughesy, TF; Ha, W; Hitchins, MP; Lai, A; McDonald, KL; Nguyen, HN; Nowak, AK; Rapkins, RW; Wang, F, 2015
)
1.35
"Temozolomide treatment did not affect TPM plasma concentrations in chronically treated patients. "( Temozolomide treatment does not affect topiramate and oxcarbazepine plasma concentrations in chronically treated patients with brain tumor-related epilepsy.
Albani, F; Baruzzi, A; Contin, M; Dinapoli, L; Fabi, A; Jandolo, B; Maschio, M; Pace, A; Zarabla, A, 2008
)
3.23
"Temozolomide treatment increased the number of SP cells, and this corresponded to more progenitor-like cells, concurrent with elevated expression of several ABC transporters."( Characterization of a side population of astrocytoma cells in response to temozolomide.
Ang, BT; Chong, KH; Chua, C; See, SJ; Tang, C; Wong, MC; Zaiden, N, 2008
)
1.3
"Temozolomide, the standard treatment of gliomas, although not an ABCG2 substrate, increases the SP in glioma cells, especially in cells missing PTEN."( PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells.
Bleau, AM; Brennan, CW; Fomchenko, EI; Hambardzumyan, D; Holland, EC; Huse, JT; Ozawa, T, 2009
)
1.07
"Temozolomide treatment may be an effective option for some aggressive pituitary tumors or carcinomas. "( Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience.
Assaker, R; Bernier, M; Borson-Chazot, F; Brue, T; Caron, P; Chabre, O; Chanson, P; Cornélius, A; Cortet-Rudelli, C; de Fraipont, F; Dufour, H; Figarella-Branger, D; François, P; Gaillard, S; Jouanneau, E; Muller, M; Passagia, JG; Raverot, G; Salenave, S; Sturm, N; Trouillas, J, 2010
)
3.25
"Temozolomide treatment has a wide range of efficacy in patients with pituitary carcinoma or locally aggressive pituitary adenoma. "( Salvage therapy with temozolomide in patients with aggressive or metastatic pituitary adenomas: experience in six cases.
Cangi, MG; Gill, AJ; Losa, M; Mazza, E; McCormack, A; Mortini, P; Motta, M; Reni, M; Talarico, A; Terreni, MR, 2010
)
2.12
"Temozolomide treatment was significantly decreased in tumor tissue intratumoral vessel number / total tumor area level."( Combination hyperbaric oxygen and temozolomide therapy in C6 rat glioma model.
Bilir, A; Bozkurt, ER; Dagıstan, Y; Karaca, I; Ozar, E; Toklu, A; Yagmurlu, K, 2012
)
1.38
"Temozolomide-treated control cells activated the DNA damage signal transducers Chk1, Chk2, and p38, leading to Cdc25C and Cdc2 inactivation, prolonged G2 arrest, and loss of clonagenicity by a combination of senescence and mitotic catastrophe."( Akt activation suppresses Chk2-mediated, methylating agent-induced G2 arrest and protects from temozolomide-induced mitotic catastrophe and cellular senescence.
Berger, MS; Hirose, Y; Katayama, M; Mirzoeva, OK; Pieper, RO, 2005
)
1.27
"Temozolomide (TMZ) treatment and hyperthermia served as controls."( Irradiation and hypoxia promote homing of haematopoietic progenitor cells towards gliomas by TGF-beta-dependent HIF-1alpha-mediated induction of CXCL12.
Frank, B; Möhle, R; Tabatabai, G; Weller, M; Wick, W, 2006
)
1.06
"Temozolomide treatment of high-grade tv-a gliomas provided a 14-day growth delay compared with vehicle controls."( Magnetic resonance imaging determination of tumor grade and early response to temozolomide in a genetically engineered mouse model of glioma.
Hambardzumyan, D; Holland, EC; Kreger, AR; Leopold, WR; McConville, P; Moody, JB; Rehemtulla, A; Ross, BD; Woolliscroft, MJ, 2007
)
1.29
"Temozolomide treatment caused a decrease in cellular NAD levels, and this was abolished by the PADPRP inhibitors."( Potentiation of temozolomide-induced cytotoxicity: a comparative study of the biological effects of poly(ADP-ribose) polymerase inhibitors.
Boulton, S; Curtin, NJ; Durkacz, BW; Golding, BT; Griffin, RJ; Pemberton, LC; Porteous, JK, 1995
)
1.36
"Temozolomide treatment of TK6 cells brought about efficient cell growth inhibition, G2/M arrest, and apoptosis, as indicated by the results of cytofluorimetric analysis of 5-bromo-2'-deoxyuridine incorporation and DNA content and evaluation of DNA fragmentation."( Involvement of the mismatch repair system in temozolomide-induced apoptosis.
Benincasa, E; Bonmassar, E; D'Atri, S; Graziani, G; Jiricny, J; Lacal, PM; Pagani, E; Tentori, L; Zambruno, G, 1998
)
1.28
"Mice treated with temozolomide showed recognition memory impairment, along with hippocampal TNF-α and BDNF mRNA expression level raise, and detection of histological insults in hematoxylin and eosin hippocampal slides."( Effect of l -Dopa in acute temozolomide-induced cognitive impairment in male mice: a possible antineuroinflammatory role.
Ahmadi-Zeidabadi, M; Asadi-Shekaari, M; Esmaeilpour, K; Jafarinejad-Farsangi, S; Pardakhty, A; Salarinejad, A; Shabani, M, 2023
)
1.53
"Treatment with Temozolomide and Lomeguatrib, a potent MGMT inhibitor, produced a huge, although transient, blastolysis and complete disappearance of all skin lesions."( Triazene compounds in the treatment of acute myeloid leukemia: a short review and a case report.
Aquino, A; Bernardini, S; Bianchi, A; Bonmassar, E; Bonmassar, L; D'Atri, S; Franzese, O; Lattuada, D; Marchesi, F; Margison, GP; Pascale, E, 2013
)
0.73
"Treatment with temozolomide was given."( Nonanaplastic pleomorphic xanthoastrocytoma with meningeal dissemination presenting with bilateral visual loss.
Berciano, J; Delgado-Alvarado, M; García-Castaño, A; Gómez-Román, J; Polo, JM; Rodríguez-Rodríguez, E; Sánchez-Salmón, E,
)
0.47
"Retreatment with temozolomide (TMZ) is one treatment option."( Dendritic cell vaccination combined with temozolomide retreatment: results of a phase I trial in patients with recurrent glioblastoma multiforme.
Ancelet, LR; Bauer, E; Dzhelali, M; Findlay, MP; Gasser, O; Hamilton, DA; Hermans, IF; Hunn, MK; Mester, B; Sharples, KJ; Wood, CE, 2015
)
1.01
"When treated with temozolomide (TMZ), an oral alkylating chemotherapy drug, most low-grade gliomas (LGG) show an initial volume decrease but this effect is rarely long lasting."( Analysis of temozolomide resistance in low-grade gliomas using a mechanistic mathematical model.
Alentorn, A; Delattre, JY; Ducray, F; Grenier, E; Honnorat, J; Idbaih, A; Kaloshi, G; Mazzocco, P; Ollier, E; Psimaras, D; Ricard, D; Samson, A, 2017
)
1.16
"Treatment with temozolomide resulted in a significant improvement in symptoms, a reduction of plasma ACTH to 389 pmol/l and regression of tumour on magnetic resonance imaging scan after four cycles of treatment."( Treatment of Nelson's syndrome with temozolomide.
Alusi, G; Berney, DM; Drake, WM; Evanson, J; Kovacs, K; Monson, JP; Moyes, VJ; Plowman, PN; Sabin, HI, 2009
)
0.97
"Treatment with temozolomide alone or in combination with capecitabine and bevacizumab resulted in objective response or stabilization in 71% of PDEC patients who failed on first-line chemotherapy. "( Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy.
Busch, C; Knappskog, S; Oberg, K; Sebjornsen, S; Sorbye, H; Welin, S, 2011
)
1.05
"Treatment with temozolomide was stopped in 12 patients due to side-effects in general, whereas in only three patients (7.1%) treatment had to be discontinued due to haematological side-effects."( Prolonged administration of temozolomide in adult patients with anaplastic glioma.
Freyschlag, CF; Janzen, E; Lohr, F; Schmieder, K; Seiz, M; Smolczyk, DR; Thomé, C; Tuettenberg, J; Weiss, C; Wenz, F, 2011
)
1
"Treatment with temozolomide alone or in combination with capecitabine and bevacizumab has recently shown promising results."( Temozolomide as second or third line treatment of patients with neuroendocrine carcinomas.
Federspiel, B; Hansen, CP; Kjaer, A; Knigge, U; Langer, SW; Olsen, IH; Sørensen, JB, 2012
)
2.16
"Treatment with temozolomide and thalidomide was associated with an objective biochemical (chromogranin A) response rate of 40%, and a radiologic response rate of 25% (45% among pancreatic endocrine tumors, 33% among pheochromocytomas, and 7% among carcinoid tumors). "( Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors.
Clark, JW; Enzinger, PC; Fuchs, CS; Kulke, MH; Michelini, A; Muzikansky, A; Ryan, DP; Stuart, K; Vincitore, M, 2006
)
1.02
"Treatment with temozolomide resulted in relief of clinical symptoms and stabilization of tumour growth for 8 months."( Metastatic medulloblastoma in an adult; treatment with temozolomide.
Bernsen, HJ; Poelen, J; Prick, MJ, 2007
)
0.93
"Treatment with temozolomide combined with AMG 102 resulted in increased inhibition of cell growth in vitro compared with treatment with either single agent alone. "( AMG 102, a fully human anti-hepatocyte growth factor/scatter factor neutralizing antibody, enhances the efficacy of temozolomide or docetaxel in U-87 MG cells and xenografts.
Burgess, TL; Coxon, A; Jun, HT; Kendall, R; Radinsky, R; Rex, K; Sun, J, 2007
)
0.9
"Treatment with temozolomide was generally well tolerated, with occasionally moderate toxicity. "( A phase II study of temozolomide in hormone-refractory prostate cancer.
Busstra, MB; Catsburg, T; Lang, MS; Mickisch, GH; Schröder, FH; van Brussel, JP, 2000
)
0.98
"Treatment with temozolomide was well tolerated."( Phase II study of temozolomide in heavily pretreated cancer patients with brain metastases.
Bacoyiannis, C; Bafaloukos, D; Bamias, A; Christodoulou, C; Karabelis, A; Kosmidis, P; Papakostas, P; Samantas, E; Skarlos, DV, 2001
)
0.98
"Treatment with temozolomide is now being explored in patients with newly diagnosed malignant glioma in several ongoing clinical trials, and alternative treatment schedules are being evaluated."( New approaches for temozolomide therapy: use in newly diagnosed glioma.
Newlands, E; Stupp, R, 2001
)
0.98

Toxicity

Temozolomide (TMZ) is a widely used oral alkylating agent that has been associated with the development of severe hematologic adverse events (HAEs) In this project, we evaluated the effects of silibinin, a natural plant component of milk thistle seeds, to potentiate toxic effects of chemotherapy drugs.

ExcerptReferenceRelevance
"The effects of treatment of mice with O6-benzylguanine (O6-BeG) on the levels of O6-alkylguanine-DNA alkyltransferase (ATase) in the hematopoietic compartment and on the in vivo sensitivity of hematopoietic progenitor cells to the toxic and clastogenic effects of the antitumor agents 1,3-bis(2-chloroethyl)-nitrosourea (BCNU) and temozolomide were studied."( O6-benzylguanine potentiates the in vivo toxicity and clastogenicity of temozolomide and BCNU in mouse bone marrow.
Ashby, J; Chinnasamy, N; Dexter, TM; Fairbairn, LJ; Hickson, I; Margison, GP; Rafferty, JA; Tinwell, H, 1997
)
0.7
" Similar degrees of protection were seen for the methylating agent streptozotocin, but no protection was detected for the chloroethylating agents carmustine or mitozolomide in the samples for which there was protection against the toxic effects of Tz."( Macrophage inflammatory protein 1alpha attenuates the toxic effects of temozolomide in human bone marrow granulocyte-macrophage colony-forming cells.
Chang, J; Clemons, M; Dexter, TM; Heyworth, C; Howell, A; Lord, B; Margison, G; Testa, N; Watson, A, 2000
)
0.54
" Potentiation of cytotoxicity was obtained at concentrations of NU1025 and NU1085 that were not toxic per se; however, NU1085 alone was 3-fold more cytotoxic (LC50 values ranged from 83 to 94 microM) than NU1025 alone (LC50 > 900 microM)."( Potentiation of temozolomide and topotecan growth inhibition and cytotoxicity by novel poly(adenosine diphosphoribose) polymerase inhibitors in a panel of human tumor cell lines.
Calvert, AH; Curtin, NJ; Delaney, CA; Durkacz, BW; Hostomsky, Z; Kyle, S; Newell, DR; Wang, LZ; White, AW, 2000
)
0.65
"The DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT) has been shown to protect cells from the toxic and mutagenic effect of alkylating agents by removing lesions from the O6 position of guanine."( Effect of O6-benzylguanine on alkylating agent-induced toxicity and mutagenicity. In Chinese hamster ovary cells expressing wild-type and mutant O6-alkylguanine-DNA alkyltransferases.
Cai, Y; Dolan, ME; Ludeman, SM; Pegg, AE; Wu, MH; Xu-Welliver, M, 2000
)
0.31
" Temozolomide was safe and well tolerated."( Safety and efficacy of temozolomide in patients with recurrent anaplastic oligodendrogliomas after standard radiotherapy and chemotherapy.
Barrie, M; Braguer, D; Chinot, OL; Dufour, H; Figarella-Branger, D; Grisoli, F; Honore, S; Martin, PM; Muracciole, X, 2001
)
1.53
"Temozolomide is safe and effective in the treatment of recurrent AO and AOA."( Safety and efficacy of temozolomide in patients with recurrent anaplastic oligodendrogliomas after standard radiotherapy and chemotherapy.
Barrie, M; Braguer, D; Chinot, OL; Dufour, H; Figarella-Branger, D; Grisoli, F; Honore, S; Martin, PM; Muracciole, X, 2001
)
2.06
" This study also analyzed the ratio of the toxic effect of TMZ on MNC and on tumor cells (i."( DNA repair enzymes and cytotoxic effects of temozolomide: comparative studies between tumor cells and normal cells of the immune system.
Alvino, E; Bonmassar, L; D'Atri, S; Falcinelli, S; Fuggetta, MP; Guadagni, F; Lacal, PM; Pagani, E; Passarelli, F; Pepponi, R; Prete, SP; Turriziani, M, 2003
)
0.58
"Postoperative radiochemotherapy with 30-33 daily doses of temozolomide (75 mg/m(2)) is safe in patients with malignant glioma."( Efficacy and toxicity of postoperative temozolomide radiochemotherapy in malignant glioma.
Eich, HT; Kocher, M; Kunze, S; Müller, RP; Semrau, R, 2005
)
0.84
" CB related adverse events occurring in more than one patient were fatigue, gait disturbance, nystagmus, and confusion."( Convection-enhanced delivery of cintredekin besudotox (interleukin-13-PE38QQR) followed by radiation therapy with and without temozolomide in newly diagnosed malignant gliomas: phase 1 study of final safety results.
Asher, AL; Chang, SM; Croteau, D; Grahn, AY; Husain, SR; Kunwar, S; Lang, FF; Parker, K; Puri, RK; Sampson, JH; Shaffrey, M; Sherman, JW; Vogelbaum, MA, 2007
)
0.55
" Thus, the implantation of BCNU wafers prior to TMZ and radiotherapy appears safe in newly diagnosed GBM patients."( A retrospective study of the safety of BCNU wafers with concurrent temozolomide and radiotherapy and adjuvant temozolomide for newly diagnosed glioblastoma patients.
Mitchell, SB; Pan, E; Tsai, JS, 2008
)
0.58
" The adverse effects related with TMZ administration are divided in three categories: myelosuppression, non haematologic toxicity, and infections."( The safety of the temozolomide in patients with malignant glioma.
Dario, A; Tomei, G, 2006
)
0.67
" Although TMZ is generally safe and acute toxicity is well documented, there are limited data on long-term toxicities."( Long-term use of temozolomide: could you use temozolomide safely for life in gliomas?
Bell, D; Khasraw, M; Wheeler, H, 2009
)
0.69
" There were no toxic deaths."( Temozolomide: a safe and effective treatment for malignant digestive endocrine tumors.
Couvelard, A; Faivre, S; Hammel, P; Hentic, O; Larroque, B; Lévy, P; Maire, F; Raymond, E; Ruszniewski, P; Yapur, L; Zappa, M, 2009
)
1.8
"The primary objective of this augmental, prospective, uncontrolled phase II multicentre trial was to assess adverse events (AE) associated with malignant glioma resection using 5-aminolevulinic (5-ALA)."( Favorable outcome in the elderly cohort treated by concomitant temozolomide radiochemotherapy in a multicentric phase II safety study of 5-ALA.
Kern, BC; Krex, D; Mehdorn, HM; Nestler, U; Pichlmeier, U; Stockhammer, F; Stummer, W; Vince, GH, 2011
)
0.61
" When this regimen was administered to mice containing humanized bone marrow, flow cytometric analyses indicated that the human bone marrow cells were significantly more sensitive to treatment than the murine bone marrow cells and that the regimen was highly toxic to human-derived hematopoietic cells of all lineages (progenitor, lymphoid, and myeloid)."( Humanized bone marrow mouse model as a preclinical tool to assess therapy-mediated hematotoxicity.
Bailey, B; Baluyut, AR; Cai, S; Chan, RJ; Ernstberger, A; Goebel, WS; Jones, DR; Juliar, BE; Mayo, LD; Pollok, KE; Sinn, AL; Wang, H, 2011
)
0.37
"Temozolomide (TMZ) is a widely used oral alkylating agent that has been associated with the development of severe hematologic adverse events (HAEs)."( Hematologic adverse events associated with temozolomide.
Abdur, S; Bressler, LR; Letarte, N; Villano, JL; Yu, JM, 2012
)
2.08
" Although there is possibly an indication that metronomic regimens of TMZ result in better PFS and response rate when compared to the conventional standard 5 day regimen, insufficient available data and study heterogeneity preclude any safe conclusions."( Using different schedules of Temozolomide to treat low grade gliomas: systematic review of their efficacy and toxicity.
Athanasiou, T; Lashkari, HP; Moreno, L; Saso, S; Zacharoulis, S, 2011
)
0.66
"The results suggest that Cyberknife re-treatments are relatively safe using selected dose/fraction schemes."( Efficacy and toxicity of CyberKnife re-irradiation and "dose dense" temozolomide for recurrent gliomas.
Arpa, D; Cardali, S; Conti, A; De Renzis, C; Granata, F; Pontoriero, A; Romanelli, P; Siragusa, C; Tomasello, C; Tomasello, F, 2012
)
0.61
" The survival rate, progression-free survival, overall survival time and adverse reactions were observed."( [Efficacy and safety of 3-dimensional conformal radiotherapy combined with temozolomide for glioma].
Tu, Q; Wang, L; Zhou, R; Zhou, W, 2011
)
0.6
"The adverse reactions were mild and tolerable."( [Efficacy and safety of 3-dimensional conformal radiotherapy combined with temozolomide for glioma].
Tu, Q; Wang, L; Zhou, R; Zhou, W, 2011
)
0.6
" Magnetic resonance imaging and laboratory tests were performed to evaluate the efficacy and adverse reactions."( [Safety and efficacy of three-dimensional conformal radiotherapy combined with temozolomide in treatment of diffuse brainstem gliomas].
Cai, CL; Fang, HH; Kang, JB; Li, FM; Nie, Q, 2011
)
0.6
" Hematotoxicity is listed as a frequent adverse drug reaction in the US prescribing information and hepatotoxicity has been reported infrequently in the postmarketing period."( Severe sustained cholestatic hepatitis following temozolomide in a patient with glioblastoma multiforme: case study and review of data from the FDA adverse event reporting system.
Bronder, E; Garbe, E; Herbst, H; Kauffmann, W; Klimpel, A; Orzechowski, HD; Sarganas, G; Thomae, M, 2012
)
0.63
"Conformal Radiation yields low grades of MRI assessed neurotoxicity and cognitive disturbance in patients of HGG with no adverse impact on local control and survival."( Survival outcome and neurotoxicity in patients of high-grade gliomas treated with conformal radiation and temozolamide.
Aggarwal, HN; Anand, AK; Babu, AG; Chaudhory, AR; Chaudhury, PS; Jena, A; Negi, PS; Rao, A; Sachdeva, PK; Sinha, SN,
)
0.13
"Combining Gliadel wafers and radiochemotherapy with TMZ may carry the risk of increased adverse events (AE)."( Safety and efficacy of Gliadel wafers for newly diagnosed and recurrent glioblastoma.
Anile, C; Balducci, M; Chiesa, S; De Bonis, P; Fiorentino, A; Maira, G; Mangiola, A; Pompucci, A, 2012
)
0.38
" The incidence of hematological adverse effects (AE) was recorded for all patients."( Toxicity and survival in primary glioblastoma patients treated with concomitant plus adjuvant temozolomide versus adjuvant temozolomide: results of a single-institution, retrospective, matched-pair analysis.
Bock, HC; Brück, W; Giese, A; Gutenberg, A; Reifenberger, G, 2013
)
0.61
" However severe hematologic adverse events (HAEs), including myelodysplastic syndrome and aplastic anemia, have also been reported."( Temozolomide-related hematologic toxicity.
De Sanctis, V; Enrici, RM; Minniti, G; Scaringi, C, 2013
)
1.83
"Central diabetes insipidus is a rare but reversible side effect of treatment with TMZ."( Central diabetes insipidus: a previously unreported side effect of temozolomide.
Faje, AT; Klibanski, A; Makimura, H; Miller, KK; Nachtigall, L; Wexler, D, 2013
)
0.63
" Although TMZ is considered to be a safe drug, it has been demonstrated to cause severe myelotoxicity; in particular, some case reports and small series studies have reported severe myelotoxicity developing during TMZ and concomitant RT."( Clinical and Genetic Factors Associated With Severe Hematological Toxicity in Glioblastoma Patients During Radiation Plus Temozolomide Treatment: A Prospective Study.
Amadori, A; Berti, F; Bertorelle, R; Della Puppa, A; Farina, P; Lombardi, G; Marcato, R; Rumiato, E; Sacchetto, V; Saggioro, D; Zagonel, V; Zustovich, F, 2015
)
0.62
" The evaluating indicators were overall response rate (ORR), 1-year survival rate, and several of the most frequent adverse events."( Efficacy and safety between temozolomide alone and temozolomide-based double therapy for malignant melanoma: a meta-analysis.
Jia, HY; Jiang, G; Lei, TC; Li, RH; Liu, YQ; Sun, C, 2014
)
0.7
" The aim of this study was to investigate the benefits and adverse effects of this combined therapy in elderly patients with glioblastoma."( Toxicity and outcome of radiotherapy with concomitant and adjuvant temozolomide in elderly patients with glioblastoma: a retrospective study.
Mukasa, A; Narita, Y; Saito, K; Saito, N; Shibui, S; Shinoura, N; Tabei, Y, 2014
)
0.64
"8% of the 618 toxicities were Common Terminology Criteria for Adverse Events (CTCAE) grade 1 or 2, while 15."( Toxicity profile of temozolomide in the treatment of 300 malignant glioma patients in Korea.
Bae, SH; Cho, SY; Kim, CY; Kim, TM; Kim, YH; Kim, YJ; Lee, MM; Lee, SH; Park, CK; Park, MJ, 2014
)
0.73
" In this project, we evaluated the effects of silibinin, a natural plant component of milk thistle seeds, to potentiate toxic effects of chemotherapy drugs such as temozolomide, etoposide and irinotecan on LN229, U87 and A172 (P53 and phosphatase and tensin homolog (PTEN) -tumor suppressor-mutated) glioma cell lines."( The effect of silibinin in enhancing toxicity of temozolomide and etoposide in p53 and PTEN-mutated resistant glioma cell lines.
Elhag, R; Mazzio, EA; Soliman, KF, 2015
)
0.87
" BBBD temozolomide was toxic in the efficacy study, but there was no evidence of symptomatic neurotoxicity in rats given IA temozolomide."( Intra-arterial administration improves temozolomide delivery and efficacy in a model of intracerebral metastasis, but has unexpected brain toxicity.
Muldoon, LL; Netto, JP; Neuwelt, EA; Pagel, MA, 2016
)
1.18
" Data on adverse events (AEs) were collected throughout."( Bevacizumab, temozolomide, and radiotherapy for newly diagnosed glioblastoma: comprehensive safety results during and after first-line therapy.
Chinot, OL; Cloughesy, T; Dhar, S; Garcia, J; Henriksson, R; Mason, W; Nishikawa, R; Pozzi, E; Saran, F; Wick, W, 2016
)
0.8
" Patients reported adverse effects in Common Toxicity Criteria for Adverse Events diaries; recorded vomiting, nausea, and rescue medication use in diaries (which were used to assess cRINV-CR); and reported QoL 4 days/week using the Modified Functional Living Index-Emesis (M-FLIE) and Osoba nausea and vomiting/retching modules."( Phase II study to evaluate the safety and efficacy of intravenous palonosetron (PAL) in primary malignant glioma (MG) patients receiving standard radiotherapy (RT) and concomitant temozolomide (TMZ).
Affronti, ML; Allen, K; Desjardins, A; Friedman, HS; Healy, PN; Herndon, JE; Kirkpatrick, J; McSherry, F; Peters, KB; Vredenburgh, JJ; Woodring, S, 2016
)
0.63
"Single-dose weekly PAL is a safe and tolerable antiemetic for cRINV prevention in MG patients receiving standard RT and concomitant TMZ."( Phase II study to evaluate the safety and efficacy of intravenous palonosetron (PAL) in primary malignant glioma (MG) patients receiving standard radiotherapy (RT) and concomitant temozolomide (TMZ).
Affronti, ML; Allen, K; Desjardins, A; Friedman, HS; Healy, PN; Herndon, JE; Kirkpatrick, J; McSherry, F; Peters, KB; Vredenburgh, JJ; Woodring, S, 2016
)
0.63
" Commenced 15 years ago, PRRT is now becoming established as first- and second-line therapy for gastroentero pancreatic neuroendocrine tumors (GEPNETs), and early treatment minimizes myelotoxicity, which is the most significant potential adverse event following PRRT."( Myelotoxicity of Peptide Receptor Radionuclide Therapy of Neuroendocrine Tumors: A Decade of Experience.
Kesavan, M; Turner, JH, 2016
)
0.43
" In conclusion, this work reports at least four compounds (5b, 5e, 5g and 6e) with potential anti-tumor effect against glioblastoma multiform cell presenting activity at low concentrations and safe profile of cytotoxicity."( Thiazolidin-4-ones from 4-(methylthio)benzaldehyde and 4-(methylsulfonyl)benzaldehyde: Synthesis, antiglioma activity and cytotoxicity.
Azambuja, JH; Braganhol, E; Cunico, W; da Silva, CEH; da Silva, DS; de Carvalho, TR; Frizzo, CP; Soares, MSP; Spanevello, RM; Zimmer, GC, 2016
)
0.43
" The most common treatment emergent adverse events were ocular: blurred vision, dry eye, keratitis, photophobia, and eye pain."( Efficacy and safety results of ABT-414 in combination with radiation and temozolomide in newly diagnosed glioblastoma.
Ansell, P; Fichtel, L; Fischer, J; Gan, HK; Gomez, E; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Mandich, H; Merrell, R; Munasinghe, W; Reardon, DA; Roberts-Rapp, L; Scott, AM; Sulman, EP; van den Bent, M; Xiong, H, 2017
)
0.69
" ATOR showed similar cytotoxic effect as TMZ to glioma cells, and it may be a safer drug, regarding side effect induction, than chemotherapic agents."( Atorvastatin Promotes Cytotoxicity and Reduces Migration and Proliferation of Human A172 Glioma Cells.
Dal-Cim, T; Lopes, FG; Ludka, FK; Nedel, CB; Oliveira, KA; Tasca, CI, 2018
)
0.48
" We aimed to determine whether or not pulse high-dose lapatinib was a safe and tolerable regimen in addition to standard therapy."( Report of safety of pulse dosing of lapatinib with temozolomide and radiation therapy for newly-diagnosed glioblastoma in a pilot phase II study.
Cloughesy, TF; Faiq, N; Green, R; Green, S; Hu, J; Lai, A; Mellinghoff, I; Nghiemphu, PL; Yu, A, 2017
)
0.71
"Extended cycles of temozolomide are safe and feasible for Chinese patients with disease responsive to temozolomide."( Feasibility and safety of extended adjuvant temozolomide beyond six cycles for patients with glioblastoma.
Chan, DT; Hsieh, SY; Kam, MK; Loong, HH; Ng, SC; Poon, DM; Poon, WS; Tsang, WK, 2017
)
1.04
"00%) had grade 3 adverse events."( Safety and Efficacy of the S-1/Temozolomide Regimen in Patients with Metastatic Neuroendocrine Tumors.
Chi, Y; Zhao, H; Zhao, J, 2018
)
0.77
" This review summarizes the mechanism of action, efficacy, and adverse events based on pre-clinical studies and clinical trials for TTF in GBM."( Tumor treating fields: a novel and effective therapy for glioblastoma: mechanism, efficacy, safety and future perspectives.
Zhu, JJ; Zhu, P, 2017
)
0.46
" For recurrent GBM, the efficacy of TTF monotherapy was shown to be equivalent in PFS and OS without systemic adverse events when compared to the control group that received best physicians-chosen chemotherapies (EF-11 trial)."( Tumor treating fields: a novel and effective therapy for glioblastoma: mechanism, efficacy, safety and future perspectives.
Zhu, JJ; Zhu, P, 2017
)
0.46
"Radiochemotherapy involving cisplatinum-based polychemotherapy is more toxic than radiotherapy in combination with temozolomide."( Concurrent radiotherapy with temozolomide vs. concurrent radiotherapy with a cisplatinum-based polychemotherapy regimen : Acute toxicity in pediatric high-grade glioma patients.
Bison, B; Bojko, S; Gielen, GH; Hoffmann, M; Kortmann, RD; Kramm, CM; Pietsch, T; Seidel, C; von Bueren, AO; Warmuth-Metz, M, 2018
)
0.98
" Keratitis was the most common grade 3 adverse event observed in 6/38 (16%) patients; thrombocytopenia was the most common grade 4 event seen in 5/38 (13%) patients."( Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma.
Ansell, PJ; Butowski, N; Fichtel, L; Fischer, J; Gan, HK; Gomez, EJ; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Mandich, H; Merrell, R; Munasinghe, WP; Reardon, DA; Roberts-Rapp, LA; Scott, AM; van den Bent, M; Wheeler, H; Xiong, H, 2018
)
0.69
" Compelling clinical data also support the safety of carmustine wafer implantation (grade A recommendation) in these patients and suggest that observed adverse events can be avoided in experienced neurosurgeon hands."( Carmustine wafer implantation for high-grade gliomas: Evidence-based safety efficacy and practical recommendations from the Neuro-oncology Club of the French Society of Neurosurgery.
Caire, F; Guyotat, J; Menei, P; Metellus, P; Pallud, J; Roux, A, 2017
)
0.46
" The observation index of both groups included the short- and long-term clinical efficacies, improvement of symptoms and signs, quality of life (QOL), and adverse responses."( Efficacy and safety of temozolomide plus whole-brain radiotherapy in the treatment of intracranial metastases.
Liu, HP; Wang, JW; Zheng, KB, 2017
)
0.77
" Although temozolomide is generally a well tolerated drug, with rare severe toxic effects, sometimes certain toxicities can overcome the life risk of the underlying malignancy."( Severe hematologic temozolomide-related toxicity and lifethreatening infections.
Nikitovic, M; Stepanovic, A,
)
0.86
" Common adverse events (AEs) were blurred vision (63%), fatigue (38%), and photophobia (35%)."( Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: results from an international phase I multicenter trial.
Ansell, PJ; Bain, E; Butowski, N; Gan, HK; Gomez, E; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Maag, D; Merrell, R; Mikkelsen, T; Nabors, LB; Papadopoulos, KP; Penas-Prado, M; Reardon, DA; Roberts-Rapp, L; Scott, AM; Simes, J; van den Bent, MJ; Walbert, T; Wheeler, H; Xiong, H, 2019
)
0.77
" Adverse events were as expected."( Safety and Feasibility of Integrating Yttrium-90 Radioembolization With Capecitabine-Temozolomide for Grade 2 Liver-Dominant Metastatic Neuroendocrine Tumors.
Cengel, KA; Damjanov, N; Metz, DC; Soulen, MC; Teitelbaum, UR; van Houten, D, 2018
)
0.7
"CapTemY90 is feasible and safe for grade 2 NETs."( Safety and Feasibility of Integrating Yttrium-90 Radioembolization With Capecitabine-Temozolomide for Grade 2 Liver-Dominant Metastatic Neuroendocrine Tumors.
Cengel, KA; Damjanov, N; Metz, DC; Soulen, MC; Teitelbaum, UR; van Houten, D, 2018
)
0.7
"Treatments for advanced melanoma are associated with different adverse events (AEs), which may be costly to manage."( Direct costs associated with adverse events of systemic therapies for advanced melanoma: Systematic literature review.
Chmielowski, B; Copley-Merriman, C; Liu, FX; Mauskopf, J; Stevinson, K; Wang, J; Zimovetz, EA, 2018
)
0.48
"CAPTEM is effective and relatively safe for treating patients with advanced NENs."( Safety and efficacy of combining capecitabine and temozolomide (CAPTEM) to treat advanced neuroendocrine neoplasms: A meta-analysis.
Fu, W; Li, W; Lu, L; Lu, Y; Lv, W; Wang, J; Zhao, Z, 2018
)
0.73
" Together, our results suggest that trehalose, a safe nutrient supplement able to cross the blood-brain barrier, is a promising candidate, worthy to be further explored in vivo, to augment the therapeutic efficacy of TMZ and RT in melanoma brain metastases."( Trehalose inhibits cell proliferation and amplifies long-term temozolomide- and radiation-induced cytotoxicity in melanoma cells: A role for autophagy and premature senescence.
Allavena, G; Del Bello, B; Maellaro, E; Miracco, C; Pirtoli, L; Tini, P; Valacchi, G; Volpi, N, 2019
)
0.75
"our data underline re-RT as a safe and feasible treatment with limited rate of toxicity, and a combined ones as a better option for selected patients."( Re-irradiation for recurrent glioma: outcome evaluation, toxicity and prognostic factors assessment. A multicenter study of the Radiation Oncology Italian Association (AIRO).
Alongi, F; Amelio, D; Borzillo, V; Ciammella, P; Clerici, E; di Monale, MB; Draghini, L; Fariselli, L; Ferrarese, F; Fiorentino, A; Galaverni, M; Krengli, M; Livi, L; Magrini, S; Maranzano, E; Masini, L; Minniti, G; Muto, P; Navarria, P; Pasqualetti, F; Pinzi, V; Scartoni, D; Scoccianti, S; Scorsetti, M; Tomatis, S, 2019
)
0.51
" The procedure was well-tolerated, with no adverse clinical or radiologic events related to the procedure."( Blood-Brain Barrier Opening in Primary Brain Tumors with Non-invasive MR-Guided Focused Ultrasound: A Clinical Safety and Feasibility Study.
Alkins, R; Bethune, A; Heyn, C; Huang, Y; Hynynen, K; Ironside, S; Lipsman, N; Mainprize, T; Meng, Y; Perry, J; Sahgal, A; Trudeau, M, 2019
)
0.51
"CAPTEM treatment can be an effective and safe treatment even after prolonged administration for patients with NENs of various sites and Ki67 labeling index, associated with significant favorable responses and PFS."( Activity and Safety of Standard and Prolonged Capecitabine/Temozolomide Administration in Patients with Advanced Neuroendocrine Neoplasms.
Alexandraki, KI; Angelousi, A; Chatzellis, E; Daskalakis, K; Gross, D; Grozinsky-Glasberg, S; Kaltsas, G; Kos-Kudła, B; Koumarianou, A; Maimon, O; Meirovitz, A; Tsoli, M; Wachuła, E, 2019
)
0.76
" 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
"00001), with no increase in adverse events (RR = 0."( The efficacy and safety of radiotherapy with adjuvant temozolomide for glioblastoma: A meta-analysis of randomized controlled studies.
Feng, Y; Wang, Y, 2020
)
0.81
" The aim of this study was to introduce the Z-scan technique as a fast, accurate, inexpensive, and safe in vitro method to distinguish the cytotoxic effects of various treatments."( Z-scan method to measure the nonlinear optical behavior of cells for evaluating the cytotoxic effects of chemotherapy and hyperthermia treatments.
Ara, MHM; Ardakani, AA; Asgari, H; Ghader, A; Khoei, S; Minaei, SE, 2021
)
0.62
" Common nonocular treatment-emergent adverse events (TEAEs) with both second-line and first-line Depatux-M included lymphopenia (42%, 33%, respectively), thrombocytopenia (39%, 47%), alanine aminotransferase increase (29%, 47%), and aspartate aminotransferase increase (24%, 60%); incidence of grade ≥3 TEAEs was 66% and 53%, respectively."( Safety and efficacy of depatuxizumab mafodotin in Japanese patients with malignant glioma: A nonrandomized, phase 1/2 trial.
Asai, K; Beppu, T; Date, I; Kagawa, N; Kanamori, M; Kasai, S; Kobayashi, H; Kumabe, T; Kuroda, J; Matsuda, M; Mishima, K; Muragaki, Y; Nagane, M; Narita, Y; Nishimura, Y; Ocampo, C; Ueki, K; Xiong, H; Yamada, M, 2021
)
0.62
"Hematological adverse events (HAEs) are common during treatment for glioblastoma (GBM), usually associated with temozolomide (TMZ)."( Hematological adverse events in the management of glioblastoma.
Butts, AR; Garcia, CR; Jayswal, R; Morgan, RM; Myint, ZW; Villano, JL; Wang, C; Weiss, HL, 2022
)
0.93
"9 months) compared to those with other or no adverse events (OS 14."( Hematological adverse events in the management of glioblastoma.
Butts, AR; Garcia, CR; Jayswal, R; Morgan, RM; Myint, ZW; Villano, JL; Wang, C; Weiss, HL, 2022
)
0.72
" Only one possibly treatment-related treatment emergent adverse event (TEAE), Grade 1 gingival swelling, was observed."( Safety and tolerability of asunercept plus standard radiotherapy/temozolomide in Asian patients with newly-diagnosed glioblastoma: a phase I study.
Chen, KT; Hsu, PW; Huang, HL; Jung, SM; Ke, YX; Lin, YJ; Toh, CH; Tsai, HC; Tseng, CK; Wei, KC, 2021
)
0.86
" In addition, OS, PFS and adverse event (AE) data on ndGBM and recurrent GBM (rGBM) were assessed."( Comparative efficacy and safety of therapeutics for elderly glioblastoma patients: A Bayesian network analysis.
Li, H; Ma, W; Qu, T; Wang, Y; Wu, J; Xia, Y; Xing, H; Zhao, B, 2022
)
0.72
"5%) had treatment-emergent adverse events (TEAEs); 72."( Efficacy and Safety of Lanreotide Autogel and Temozolomide Combination Therapy in Progressive Thoracic Neuroendocrine Tumors (Carcinoid): Results from the Phase 2 ATLANT Study.
Amoroso, V; Berruti, A; Brizzi, MP; Colao, A; Faggiano, A; Fazio, N; Ferolla, P; Ghizzoni, S; Giuffrida, D; Houchard, A; Ibrahim, T; La Salvia, A; Marconcini, R; Mazzanti, P; Spada, F; Vaccaro, V; Volante, M, 2023
)
1.17
" No unexpected adverse events clinically, or permanent end-organ damage during postmortem examination was identified in glioma subjects who had received standard or prolonged duration of BEV, TMZ or TIB regimen-based chemotherapies except rare events of bone marrow suppression."( Postmortem study of organ-specific toxicity in glioblastoma patients treated with a combination of temozolomide, irinotecan and bevacizumab.
Ballester, LY; Bhattacharjee, MB; Brown, RE; Buja, LM; Chen, L; Glass, WF; Hergenroeder, GW; Hunter, RL; Linendoll, N; Lu, G; Pilichowska, M; Pillai, AK; Rao, M; Tian, X; Wu, JK; Zhang, R; Zhu, JJ; Zhu, P, 2022
)
0.94
" The patient's progression free survival time (PFS), total survival time (OS) and adverse reactions were observed by telephone, outpatient reexamination and other follow-up methods."( The efficacy and safety of low-dose temozolomide maintenance therapy in elderly patients with glioblastoma: a retrospective cohort study.
Ge, X; Gong, S; Guo, J; Tao, Q; Zhu, T, 2022
)
1
" The main adverse reactions were digestive tract reactions and hematological toxicity."( The efficacy and safety of low-dose temozolomide maintenance therapy in elderly patients with glioblastoma: a retrospective cohort study.
Ge, X; Gong, S; Guo, J; Tao, Q; Zhu, T, 2022
)
1
"In general, for elderly patients with good Karnofsky Performance Scale (KPS) scores, further reducing TMZ to maintain chemotherapy after the standard STUPP regimen may improve the PFS and OS to a certain extent, with tolerable adverse reactions and reduced cost."( The efficacy and safety of low-dose temozolomide maintenance therapy in elderly patients with glioblastoma: a retrospective cohort study.
Ge, X; Gong, S; Guo, J; Tao, Q; Zhu, T, 2022
)
1
" A multicenter phase II trial was designed to determine: (1) the success rate of manufacturing the Aivita GBM vaccine (AV-GBM-1), (2) Adverse Events (AE) associated with AV-GBM-1 administration, and (3) survival."( Phase 2 study of AV-GBM-1 (a tumor-initiating cell targeted dendritic cell vaccine) in newly diagnosed Glioblastoma patients: safety and efficacy assessment.
Abedi, M; Aiken, RD; Bota, DA; Bota, PG; Carrillo, JA; Dillman, RO; Duma, CM; Hsieh, C; Hsu, FPK; Keirstead, HS; Kesari, S; Kong, XT; LaRocca, RV; Nistor, GI; Piccioni, DE; Taylor, TH, 2022
)
0.72
" The primary endpoints were skin, neurological and psychiatric adverse events."( Safety and efficacy of tumour-treating fields (TTFields) therapy for newly diagnosed glioblastoma in Japanese patients using the Novo-TTF System: a prospective post-approval study.
Arakawa, Y; Kanamori, M; Mukasa, A; Muragaki, Y; Narita, Y; Nishikawa, R; Tanaka, S; Yamaguchi, S; Yamasaki, F, 2023
)
0.91
" The most common tumour-treating-fields-therapy-related adverse event was beneath-array local skin reaction (60% of patients)."( Safety and efficacy of tumour-treating fields (TTFields) therapy for newly diagnosed glioblastoma in Japanese patients using the Novo-TTF System: a prospective post-approval study.
Arakawa, Y; Kanamori, M; Mukasa, A; Muragaki, Y; Narita, Y; Nishikawa, R; Tanaka, S; Yamaguchi, S; Yamasaki, F, 2023
)
0.91
" Safety was determined by the number of treatment-related adverse events."( Antisecretory factor is safe to use as add-on treatment in newly diagnosed glioblastoma.
Belting, M; Bengzon, J; Cederberg, D; Darabi, A; Edvardsson, C; Ehinger, E; Kopecky, J; Siesjö, P; Tomasevic, G; Visse, E, 2023
)
0.91
"No serious treatment-related adverse events were observed."( Antisecretory factor is safe to use as add-on treatment in newly diagnosed glioblastoma.
Belting, M; Bengzon, J; Cederberg, D; Darabi, A; Edvardsson, C; Ehinger, E; Kopecky, J; Siesjö, P; Tomasevic, G; Visse, E, 2023
)
0.91
"We conclude that Salovum is safe to use as an add-on treatment for GBM."( Antisecretory factor is safe to use as add-on treatment in newly diagnosed glioblastoma.
Belting, M; Bengzon, J; Cederberg, D; Darabi, A; Edvardsson, C; Ehinger, E; Kopecky, J; Siesjö, P; Tomasevic, G; Visse, E, 2023
)
0.91
" The most common adverse events were leukocytopenia (66."( Safety and Efficacy of Anlotinib Hydrochloride Plus Temozolomide in Patients with Recurrent Glioblastoma.
Bu, L; Cai, J; Chen, Q; Huang, K; Meng, X; Weng, Y; Xu, Q; Zhan, R; Zhang, L; Zheng, X, 2023
)
1.16
" Adverse effects occurred in 18 patients (14 mild and 4 moderate or severe)."( Efficacy and safety of temozolomide in the treatment of aggressive pituitary neuroendocrine tumours in Spain.
Araujo-Castro, M; Biagetti, B; Cámara, R; Fajardo, C; García-Centeno, R; Guerrero-Pérez, F; Hanzu, F; Iglesias, P; Lamas, C; Mora, M; Remon-Ruiz, P; Soto, A, 2023
)
1.22

Pharmacokinetics

This method was used successfully to perform brain and plasma pharmacokinetic studies of temozolomide in mice after intraperitoneal administration. The enhanced hematologic toxicity resulting from combining O(6)BG with temozlomide does not appear to be the result of a pharmacokinetics interaction between the agents.

ExcerptReferenceRelevance
" The half-life of the drug in the tumors was approximately 60 min."( Pharmacokinetics of the 13C labeled anticancer agent temozolomide detected in vivo by selective cross-polarization transfer.
Artemov, D; Bhujwalla, ZM; Glickson, JD; Griffiths, JR; Judson, IR; Leach, MO; Maxwell, RJ, 1995
)
0.54
"We conclude that spectral analysis provides important pharmacokinetic information about radiolabeled anti-cancer drugs with relatively few model assumptions."( Pharmacokinetic assessment of novel anti-cancer drugs using spectral analysis and positron emission tomography: a feasibility study.
Brock, CS; Cunningham, VJ; Harte, RJ; Jones, T; Matthews, JC; Meikle, SR; Price, P; Wells, P, 1998
)
0.3
" Plasma samples were obtained on days 1 and 2 to evaluate the pharmacokinetic parameters of TMZ alone and in combination with CDDP."( A Phase I and pharmacokinetic study of temozolomide and cisplatin in patients with advanced solid malignancies.
Agarwala, SS; Baker, SD; Barrington, R; Britten, CD; Diab, SG; Eckardt, JR; Eckhardt, SG; Fraass, U; Hammond, LA; Johnson, T; Rowinsky, EK; Statkevich, P; Villalona-Calero, M; Von Hoff, DD, 1999
)
0.57
"Fifteen patients with advanced cancer were enrolled of which 12 were evaluable, all of whom had pharmacokinetic blood sampling."( Effect of gastric pH on the relative oral bioavailability and pharmacokinetics of temozolomide.
Beale, P; Brada, M; Cutler, DL; Judson, I; Marco, A; Moore, S; Reidenberg, P; Statkevich, P, 1999
)
0.53
" There was no difference in the pharmacokinetic parameters of temozolomide or MTIC with or without the concomitant administration of ranitidine."( Effect of gastric pH on the relative oral bioavailability and pharmacokinetics of temozolomide.
Beale, P; Brada, M; Cutler, DL; Judson, I; Marco, A; Moore, S; Reidenberg, P; Statkevich, P, 1999
)
0.77
" Statistical analyses of pharmacokinetic and pharmacodynamic end points in the control and TNP-470 treatment groups were completed by nonparametric tests."( Pharmacodynamic-mediated reduction of temozolomide tumor concentrations by the angiogenesis inhibitor TNP-470.
Chu, J; Gallo, JM; Li, S; Ma, J; Pulfer, S; Reed, K, 2001
)
0.58
" It is proposed that the net balance of antiangiogenic drug-mediated pharmacodynamic actions will determine how drug disposition in tumors may be affected."( Pharmacodynamic-mediated effects of the angiogenesis inhibitor SU5416 on the tumor disposition of temozolomide in subcutaneous and intracerebral glioma xenograft models.
Gallo, JM; Guo, P; Li, S; Ma, J; Reed, K, 2003
)
0.54
"To construct a population pharmacokinetic model for temozolomide (TMZ), a novel imidazo-tetrazine methylating agent and its metabolites MTIC and AIC in infants and children with primary central nervous system tumors."( Population pharmacokinetics of temozolomide and metabolites in infants and children with primary central nervous system tumors.
Fouladi, M; Gajjar, A; Heideman, RL; Kirstein, MN; Nair, G; Panetta, JC; Stewart, CF; Wilkinson, M, 2003
)
0.86
"We evaluated the pharmacokinetics of TMZ and MTIC in 39 children (20 boys and 19 girls) with 132 pharmacokinetic studies (109 in the training set and 23 in the validation set)."( Population pharmacokinetics of temozolomide and metabolites in infants and children with primary central nervous system tumors.
Fouladi, M; Gajjar, A; Heideman, RL; Kirstein, MN; Nair, G; Panetta, JC; Stewart, CF; Wilkinson, M, 2003
)
0.6
"To characterize and compare pharmacokinetic parameters in children and adults treated with temozolomide (TMZ) administered for 5 days in three doses daily, and to evaluate the possible relationship between AUC values and hematologic toxicity."( Pharmacokinetics of temozolomide given three times a day in pediatric and adult patients.
Barone, C; Caldarelli, M; Cefalo, G; Garrè, ML; Lazzareschi, I; Madon, E; Maira, G; Massimino, M; Mastrangelo, S; Mazzarella, G; Riccardi, A; Riccardi, R; Ridola, V; Ruggiero, A; Sandri, A, 2003
)
0.86
"TMZ pharmacokinetic parameters were characterized in pediatric and adult patients with primary central nervous system tumors treated with doses ranging from 120 to 200 mg/m2 per day, divided into three doses daily for 5 days."( Pharmacokinetics of temozolomide given three times a day in pediatric and adult patients.
Barone, C; Caldarelli, M; Cefalo, G; Garrè, ML; Lazzareschi, I; Madon, E; Maira, G; Massimino, M; Mastrangelo, S; Mazzarella, G; Riccardi, A; Riccardi, R; Ridola, V; Ruggiero, A; Sandri, A, 2003
)
0.64
"No difference appears to exist between pharmacokinetic parameters in adults and children when TMZ is administered in three doses daily."( Pharmacokinetics of temozolomide given three times a day in pediatric and adult patients.
Barone, C; Caldarelli, M; Cefalo, G; Garrè, ML; Lazzareschi, I; Madon, E; Maira, G; Massimino, M; Mastrangelo, S; Mazzarella, G; Riccardi, A; Riccardi, R; Ridola, V; Ruggiero, A; Sandri, A, 2003
)
0.64
" The population pharmacokinetic analysis was performed with nonlinear mixed-effect modeling software."( Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients.
Buclin, T; Csajka, C; Decosterd, LA; Lejeune, F; Leyvraz, S; Ostermann, S; Stupp, R, 2004
)
0.56
"This is the first human pharmacokinetic study on TMZ to quantify CSF penetration."( Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients.
Buclin, T; Csajka, C; Decosterd, LA; Lejeune, F; Leyvraz, S; Ostermann, S; Stupp, R, 2004
)
0.56
" To that end, a novel method, based on physiologically based hybrid pharmacokinetic models, is presented to predict human tumor drug concentrations."( Pharmacokinetic model-predicted anticancer drug concentrations in human tumors.
Bookman, MA; Gallo, JM; Guo, P; Li, S; Ma, J; Orlansky, A; Pulfer, S; Vicini, P; Zhou, F, 2004
)
0.32
"To develop a pharmacokinetic limited sampling model (LSM) for temozolomide and its metabolite MTIC in infants and children."( Development of a pharmacokinetic limited sampling model for temozolomide and its active metabolite MTIC.
Freeman, BB; Gajjar, A; Iacono, LC; Kirstein, MN; Nair, G; Panetta, JC; Stewart, CF, 2005
)
0.81
" This accounted for prior distribution of temozolomide and MTIC pharmacokinetic parameters based on full pharmacokinetic sampling from 38 patients with 120 pharmacokinetic studies (dosage range 145-200 mg/m(2) per day orally)."( Development of a pharmacokinetic limited sampling model for temozolomide and its active metabolite MTIC.
Freeman, BB; Gajjar, A; Iacono, LC; Kirstein, MN; Nair, G; Panetta, JC; Stewart, CF, 2005
)
0.84
" Furthermore, PARP activity was not inhibited by temozolomide treatment and this newly validated pharmacodynamic assay is therefore suitable for use in a proof-of-principle phase I trial a PARP-1 inhibitor in combination with temozolomide."( Temozolomide pharmacodynamics in patients with metastatic melanoma: dna damage and activity of repair enzymes O6-alkylguanine alkyltransferase and poly(ADP-ribose) polymerase-1.
Boddy, AV; Calvert, AH; Curtin, NJ; Harris, AL; Hickson, I; Jones, C; Margison, GP; McGown, G; McHugh, P; Middleton, MR; Newell, DR; Olsen, A; Plummer, ER; Thorncroft, M; Watson, AJ, 2005
)
2.03
" Although this approach is promising, there has not been any attempt to define optimal metronomic dosing regimens by integrating pharmacokinetic (PK) with pharmacodynamic (PD) measurements."( Preclinical pharmacokinetic and pharmacodynamic evaluation of metronomic and conventional temozolomide dosing regimens.
Gallo, JM; Guo, P; Nuthalapati, S; Wang, X; Zhou, Q, 2007
)
0.56
" Because significant obstacles prevent making these measurements in humans, development of a predictive pharmacokinetic model would be of great value to the translation of preclinical data to the clinic."( Predicting human tumor drug concentrations from a preclinical pharmacokinetic model of temozolomide brain disposition.
Gallo, JM; Guo, P; Kruh, GD; Vicini, P; Wang, X; Zhou, Q, 2007
)
0.56
" The resultant data provided the framework to develop a hybrid physiologically based pharmacokinetic model for temozolomide in brain."( Predicting human tumor drug concentrations from a preclinical pharmacokinetic model of temozolomide brain disposition.
Gallo, JM; Guo, P; Kruh, GD; Vicini, P; Wang, X; Zhou, Q, 2007
)
0.77
"A physiologically based pharmacokinetic modeling approach offers a means to translate preclinical to clinical characteristics of drug disposition in target tissues and, thus, a means to select appropriate drug dosing regimens for achieving optimal target tissue drug concentrations."( Predicting human tumor drug concentrations from a preclinical pharmacokinetic model of temozolomide brain disposition.
Gallo, JM; Guo, P; Kruh, GD; Vicini, P; Wang, X; Zhou, Q, 2007
)
0.56
"The pharmacokinetic and safety profile of TMZ in Japanese patients was comparable to that in Caucasians."( Pharmacokinetic study of temozolomide on a daily-for-5-days schedule in Japanese patients with relapsed malignant gliomas: first study in Asians.
Adachi, J; Aoki, T; Matsutani, M; Mishima, K; Mizutani, T; Nishikawa, R; Nojima, K, 2007
)
0.64
" The mean clearance of temozolomide was 107 mL/min/m2, with a volume of distribution of 20 L/m2 and half-life of 109 minutes."( Phase I pharmacokinetic and pharmacodynamic study of temozolomide in pediatric patients with refractory or recurrent leukemia: a Children's Oncology Group Study.
Adamson, PC; Berg, SL; Blaney, SM; Delaney, SM; Dolan, ME; Hedge, M; Horton, TM; Ingle, AM; Thompson, PA; Weiss, HL; Wu, MF, 2007
)
0.9
" Pharmacokinetic analyses revealed lowered exposures and enhanced clearance among patients on EIAEDs."( Safety and pharmacokinetics of dose-intensive imatinib mesylate plus temozolomide: phase 1 trial in adults with malignant glioma.
Desjardins, A; Egorin, MJ; Friedman, AH; Friedman, HS; Gururangan, S; Herndon, JE; Lagattuta, TF; McLendon, R; Quinn, JA; Reardon, DA; Rich, JN; Salvado, AJ; Sathornsumetee, S; Vredenburgh, JJ, 2008
)
0.58
" pharmacokinetic data on 670 drugs representing, to our knowledge, the largest publicly available set of human clinical pharmacokinetic data."( Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds.
Lombardo, F; Obach, RS; Waters, NJ, 2008
)
0.35
"We conducted a phase I and pharmacokinetic study of the epidermal growth factor receptor (EGFR) inhibitor erlotinib as a single agent and in combination with temozolomide in children with refractory solid tumors."( Pediatric phase I and pharmacokinetic study of erlotinib followed by the combination of erlotinib and temozolomide: a Children's Oncology Group Phase I Consortium Study.
Adamson, PC; Blaney, SM; Dancey, JE; Gilbertson, RJ; Hamilton, M; Ingle, AM; Jakacki, RI; Krailo, MD; Tersak, J; Voss, SD, 2008
)
0.76
" Pharmacokinetic studies and ERBB-receptor expression and signaling studies were performed."( Pediatric phase I and pharmacokinetic study of erlotinib followed by the combination of erlotinib and temozolomide: a Children's Oncology Group Phase I Consortium Study.
Adamson, PC; Blaney, SM; Dancey, JE; Gilbertson, RJ; Hamilton, M; Ingle, AM; Jakacki, RI; Krailo, MD; Tersak, J; Voss, SD, 2008
)
0.56
"The enhanced hematologic toxicity resulting from combining O(6)BG with temozolomide does not appear to be the result of a pharmacokinetic interaction between the agents."( Pharmacokinetics of temozolomide administered in combination with O6-benzylguanine in children and adolescents with refractory solid tumors.
Aikin, AA; Balis, FM; Cole, DE; Fox, E; Meany, HJ; Warren, KE, 2009
)
0.91
" This method was used successfully to perform brain and plasma pharmacokinetic studies of temozolomide in mice after intraperitoneal administration."( Development of a new UPLC-MSMS method for the determination of temozolomide in mice: application to plasma pharmacokinetics and brain distribution study.
Farinotti, R; Fernandez, C; Goldwirt, L; Zahr, N, 2013
)
0.85
"Cabozantinib at a dose of 40 mg daily with RT plus TMZ and post-RT TMZ for patients with newly diagnosed high-grade glioma was generally well tolerated, and demonstrated no pharmacokinetic interactions with concurrent TMZ."( Phase 1 dose escalation trial of the safety and pharmacokinetics of cabozantinib concurrent with temozolomide and radiotherapy or temozolomide after radiotherapy in newly diagnosed patients with high-grade gliomas.
Chamberlain, MC; Cloughesy, T; Desjardins, A; Glantz, M; Mikkelsen, T; Reardon, DA; Schiff, D; Wen, PY, 2016
)
0.65
" Pharmacokinetic parameters were estimated using non-compartmental analysis."( Plasma and cerebrospinal fluid pharmacokinetics of select chemotherapeutic agents following intranasal delivery in a non-human primate model.
Cruz, R; Figg, WD; League-Pascual, JC; Lester-McCully, CM; Peer, CJ; Rodgers, L; Ronner, L; Shandilya, S; Warren, KE, 2017
)
0.46
" This phase I study evaluated the pharmacokinetics and mass balance of veliparib administered alone and in combination with temozolomide, and assessed any potential pharmacokinetic drug-drug interaction between veliparib and temozolomide."( Clinical Pharmacokinetics and Mass Balance of Veliparib in Combination with Temozolomide in Subjects with Nonhematologic Malignancies.
Giranda, V; Munasinghe, W; Nuthalapati, S; Xiong, H, 2018
)
0.92
" No pharmacokinetic interaction was observed when veliparib and temozolomide were administered together."( Clinical Pharmacokinetics and Mass Balance of Veliparib in Combination with Temozolomide in Subjects with Nonhematologic Malignancies.
Giranda, V; Munasinghe, W; Nuthalapati, S; Xiong, H, 2018
)
0.95
"We recently reported an acceptable safety and pharmacokinetic profile of depatuxizumab mafodotin (depatux-m), formerly called ABT-414, plus radiation and temozolomide in newly diagnosed glioblastoma (arm A)."( Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma.
Ansell, PJ; Butowski, N; Fichtel, L; Fischer, J; Gan, HK; Gomez, EJ; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Mandich, H; Merrell, R; Munasinghe, WP; Reardon, DA; Roberts-Rapp, LA; Scott, AM; van den Bent, M; Wheeler, H; Xiong, H, 2018
)
0.89
" Depatux-m demonstrated a linear pharmacokinetic profile."( Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma.
Ansell, PJ; Butowski, N; Fichtel, L; Fischer, J; Gan, HK; Gomez, EJ; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Mandich, H; Merrell, R; Munasinghe, WP; Reardon, DA; Roberts-Rapp, LA; Scott, AM; van den Bent, M; Wheeler, H; Xiong, H, 2018
)
0.69
"Depatux-m alone or in combination with temozolomide demonstrated an acceptable safety and pharmacokinetic profile in glioblastoma."( Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma.
Ansell, PJ; Butowski, N; Fichtel, L; Fischer, J; Gan, HK; Gomez, EJ; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Mandich, H; Merrell, R; Munasinghe, WP; Reardon, DA; Roberts-Rapp, LA; Scott, AM; van den Bent, M; Wheeler, H; Xiong, H, 2018
)
0.96
" We aimed to describe the development and comparison of 2 population pharmacokinetic modeling approaches."( An Integrated Population Pharmacokinetic Model Versus Individual Models of Depatuxizumab Mafodotin, an Anti-EGFR Antibody Drug Conjugate, in Patients With Solid Tumors Likely to Overexpress EGFR.
Bain, E; Friedel, A; Menon, RM; Mensing, S; Mittapalli, RK; Stodtmann, S; Xiong, H, 2019
)
0.51
"Preclinical pharmacokinetic studies evaluated olaparib tissue distribution in rats and tumor-bearing mice."( Pharmacokinetics, safety, and tolerability of olaparib and temozolomide for recurrent glioblastoma: results of the phase I OPARATIC trial.
Carruthers, R; Chalmers, AJ; Cruickshank, G; Dunn, L; Erridge, S; Godfrey, L; Halford, S; Hanna, C; Jackson, A; Jefferies, S; Kurian, KM; McBain, C; McCormick, A; Pittman, M; Sleigh, R; Strathdee, K; Watts, C; Williams, K, 2020
)
0.8
" TMZ concentration in blood, brain and tumor was detected using liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) and then pharmacokinetic parameters were calculated."( Essential oil of Ligusticum chuanxiong Hort. Regulated P-gp protein and tight junction protein to change pharmacokinetic parameters of temozolomide in blood, brain and tumor.
Hu, PY; Liu, SS; Liu, XJ; Shuai, SY; Yang, M; Yue, PF; Zhang, GS; Zheng, Q, 2022
)
0.92

Compound-Compound Interactions

Zotiraciclib combined with temozolomide is safe in patients with recurrent high-grade astrocytomas. This phase I study evaluated the pharmacokinetics and mass balance of veliparib.

ExcerptReferenceRelevance
" The analysis of cell cycle indicated that the drug combination of TZM and PADPRP inhibitors provoked G1 arrest only in p53+ cells."( Role of wild-type p53 on the antineoplastic activity of temozolomide alone or combined with inhibitors of poly(ADP-ribose) polymerase.
Benincasa, E; Bonmassar, E; Faraoni, I; Franco, D; Graziani, G; Lacal, PM; Tentori, L, 1998
)
0.55
" AGT depleting agents in combination with methylating and chloroethylating agents are now in clinical testing, and may result in greater clinical efficacy in metastatic malignant melanoma."( New cytotoxic agents for the treatment of metastatic malignant melanoma: temozolomide and related alkylating agents in combination with guanine analogues to abrogate drug resistance.
Gerson, S; Liu, L; Spiro, T,
)
0.36
"The activity of temozolomide combined with irinotecan (CPT-11) was evaluated against eight independent xenografts (four neuroblastomas, three rhabdomyosarcomas, and one glioblastoma)."( Antitumor activity of temozolomide combined with irinotecan is partly independent of O6-methylguanine-DNA methyltransferase and mismatch repair phenotypes in xenograft models.
Brent, TP; Cheshire, PJ; Friedman, HS; Houghton, PJ; Kirstein, MN; Poquette, CA; Richmond, LB; Stewart, CF; Tan, M, 2000
)
0.97
" Preclinical and phase I trials have shown the additive or synergistic activity of temozolomide combined with carmustine against solid tumors, including malignant glioma, and the sequence-dependent effects of the combination."( Temozolomide in combination with other cytotoxic agents.
Prados, M, 2001
)
1.98
" These results indicate that a careful selection of the antitumor agent has to be made when antitelomerase therapy is combined with chemotherapy."( Inhibition of telomerase increases resistance of melanoma cells to temozolomide, but not to temozolomide combined with poly (adp-ribose) polymerase inhibitor.
Balduzzi, A; Barbarino, M; Biroccio, A; Gold, B; Graziani, G; Levati, L; Lombardi, ML; Portarena, I; Tentori, L; Vergati, M, 2003
)
0.56
" Therefore, the authors initiated a Phase I study to determine the pharmacokinetics and safety profile of temozolomide (TMZ), a novel oral alkylating agent known to cross the blood-brain barrier, in combination with interferon alpha-2b (IFN-alpha2b)."( Temozolomide in combination with interferon alpha-2b in patients with metastatic melanoma: a phase I dose-escalation study.
Agarwala, SS; Kirkwood, JM, 2003
)
1.98
" This phase I study was designed to evaluate the efficacy and safety of TMZ alone (course 1) and TMZ in combination with PCB in subsequent courses in chemotherapy-naïve patients with malignant glioma."( Phase I study of temozolamide (TMZ) combined with procarbazine (PCB) in patients with gliomas.
Foster, T; Newlands, ES; Zaknoen, S, 2003
)
0.32
"To evaluate the efficacy of temozolomide (TMZ) combined with cisplatin (CDDP) in terms of response rate, time to progression (TTP) and overall survival (OS), as well as the tolerability of the regimen in patients with brain metastases from solid tumors."( Temozolomide (TMZ) combined with cisplatin (CDDP) in patients with brain metastases from solid tumors: a Hellenic Cooperative Oncology Group (HeCOG) Phase II study.
Aravantinos, G; Bafaloukos, D; Bamias, A; Carina, M; Christodoulou, C; Klouvas, G; Linardou, H; Skarlos, D, 2005
)
2.07
"Patients (n=32) with brain metastases were treated with TMZ 150 mg/m2/day (chemotherapy-pretreated) or 200 mg/m2/day (chemotherapy-naive) for 5 days, combined with CDDP 75 mg/m2 on day 1, every 28 days."( Temozolomide (TMZ) combined with cisplatin (CDDP) in patients with brain metastases from solid tumors: a Hellenic Cooperative Oncology Group (HeCOG) Phase II study.
Aravantinos, G; Bafaloukos, D; Bamias, A; Carina, M; Christodoulou, C; Klouvas, G; Linardou, H; Skarlos, D, 2005
)
1.77
"TMZ combined with CDDP is an active and well-tolerated combination in patients with brain metastases from solid tumors."( Temozolomide (TMZ) combined with cisplatin (CDDP) in patients with brain metastases from solid tumors: a Hellenic Cooperative Oncology Group (HeCOG) Phase II study.
Aravantinos, G; Bafaloukos, D; Bamias, A; Carina, M; Christodoulou, C; Klouvas, G; Linardou, H; Skarlos, D, 2005
)
1.77
"The aim of this study was to evaluate the efficacy and safety of carmustine (BCNU) in combination with temozolomide as first-line chemotherapy before and after radiotherapy (RT) in patients with inoperable, newly diagnosed glioblastoma multiforme (GBM)."( Temozolomide in combination with BCNU before and after radiotherapy in patients with inoperable newly diagnosed glioblastoma multiforme.
Barrié, M; Braguer, D; Chinot, O; Couprie, C; Dufour, H; Figarella-Branger, D; Grisoli, F; Hoang-Xuan, K; Martin, PM; Muracciole, X; Peragut, JC, 2005
)
1.99
" Furthermore, we show that a human haemopoietic cell line (K562) transduced with a retroviral vector encoding MGMT(P140K) is highly resistant to the cytotoxic effects of PaTrin-2 in combination with the methylating agent temozolomide, and that cells expressing MGMT(P140K) can be effectively enriched in vitro following challenge with this drug combination."( The P140K mutant of human O(6)-methylguanine-DNA-methyltransferase (MGMT) confers resistance in vitro and in vivo to temozolomide in combination with the novel MGMT inactivator O(6)-(4-bromothenyl)guanine.
Fairbairn, LJ; Margison, GP; Milsom, MD; Southgate, TD; Woolford, LB, 2006
)
0.73
" As the combination with interferon-alfa (IFN-alpha) appears superior to single-agent DTIC regarding response rates, the purpose of this study was to compare TMZ alone and TMZ plus IFN-alpha in terms of objective response (OR), overall survival, and safety in a prospective, randomized, multicenter trial."( Temozolomide in combination with interferon-alfa versus temozolomide alone in patients with advanced metastatic melanoma: a randomized, phase III, multicenter study from the Dermatologic Cooperative Oncology Group.
Becker, JC; Dummer, R; Garbe, C; Kaufmann, R; Krengel, S; Kretschmer, L; Leiter, U; Linse, R; Mauch, C; Schadendorf, D; Sebastian, G; Spieth, K; Tilgen, W; Vogt, T; von den Driesch, P, 2005
)
1.77
"Two hundred ninety-four patients with untreated stage IV metastatic melanoma (American Joint Committee on Cancer staging system) were randomly assigned to receive either oral TMZ alone (200 mg/m2/day; days 1 through 5 every 28 days) or in combination with subcutaneous IFN-alpha (5 MU/m2; days 1, 3, and 5 every week)."( Temozolomide in combination with interferon-alfa versus temozolomide alone in patients with advanced metastatic melanoma: a randomized, phase III, multicenter study from the Dermatologic Cooperative Oncology Group.
Becker, JC; Dummer, R; Garbe, C; Kaufmann, R; Krengel, S; Kretschmer, L; Leiter, U; Linse, R; Mauch, C; Schadendorf, D; Sebastian, G; Spieth, K; Tilgen, W; Vogt, T; von den Driesch, P, 2005
)
1.77
" In conclusion, temozolomide combined with cisplatin is an active and safe first-line chemotherapy regimen with acceptable and easily manageable toxicities in patients with metastatic melanoma."( Temozolomide in combination with cisplatin in patients with metastatic melanoma: a phase II trial.
Argon, A; Camlica, H; Tas, F; Topuz, E, 2005
)
2.12
" Patients with stable or progressive malignant primary glioma received erlotinib alone or combined with temozolomide in this dose-escalation study."( Phase 1 study of erlotinib HCl alone and combined with temozolomide in patients with stable or recurrent malignant glioma.
Burton, E; Butowski, N; Chang, S; Fedoroff, A; Kapadia, A; Kelley, SK; Lamborn, KR; Malec, M; Page, MS; Prados, MD; Rabbitt, J; Xie, D, 2006
)
0.8
" The main aims of this phase I trial were to determine an ATase-depleting dose (ADD) of lomeguatrib, a potent pseudosubstrate inhibitor, and to define a suitable dose of temozolomide to be used in combination with lomeguatrib in patients with advanced cancer."( Lomeguatrib, a potent inhibitor of O6-alkylguanine-DNA-alkyltransferase: phase I safety, pharmacodynamic, and pharmacokinetic trial and evaluation in combination with temozolomide in patients with advanced solid tumors.
Bridgewater, J; Dawson, M; Donnelly, D; Gumbrell, L; Halbert, G; Jowle, D; Lee, SM; Margison, GP; McElhinney, RS; McGrath, H; McMurry, TB; Middleton, MR; Ranson, M; Waller, S, 2006
)
0.72
"Lomeguatrib was administered at dose levels of 10 to 40 mg/m2 days 1 to 5, as a single agent, and also in combination with temozolomide."( Lomeguatrib, a potent inhibitor of O6-alkylguanine-DNA-alkyltransferase: phase I safety, pharmacodynamic, and pharmacokinetic trial and evaluation in combination with temozolomide in patients with advanced solid tumors.
Bridgewater, J; Dawson, M; Donnelly, D; Gumbrell, L; Halbert, G; Jowle, D; Lee, SM; Margison, GP; McElhinney, RS; McGrath, H; McMurry, TB; Middleton, MR; Ranson, M; Waller, S, 2006
)
0.74
" Groups treated with regional temozolomide alone (350 mg/kg), systemic temozolomide with O6BG, or vehicle combined with O6BG showed no significant tumor responses compared with controls."( Modulation of chemotherapy resistance in regional therapy: a novel therapeutic approach to advanced extremity melanoma using intra-arterial temozolomide in combination with systemic O6-benzylguanine.
Abdel-Wahab, OI; Abdel-Wahab, Z; Augustine, C; Cheng, TY; Friedman, HS; Grubbs, E; Ko, SH; Pruitt, SK; Tyler, DS; Ueno, T; Yoshimoto, Y, 2006
)
0.82
" We designed a new regimen utilizing dose-intensified, protracted course of TMZ in combination with vinorelbine, a lipophilic large-spectrum agent, in an attempt to improve the efficacy of TMZ."( Vinorelbine combined with a protracted course of temozolomide for recurrent brain metastases: a phase I trial.
Abrey, LE; Demopoulos, A; Malkin, MG; Omuro, AM; Raizer, JJ, 2006
)
0.59
" A multicentre phase II study was performed to assess the activity and toxicity of temozolomide in combination with interferon alpha-2b."( Phase II multicentre study of temozolomide in combination with interferon alpha-2b in metastatic malignant melanoma.
Crespo, C; del Muro, XG; Filipovich, E; García, M; Germà-Lluch, JR; López, JJ; Pérez, X; Rifà, J; Tres, A; Valladares, M, 2006
)
0.85
" Previously untreated patients received temozolomide administered orally at a dose of 150 mg/m/day for 5 days every 4 weeks, in combination with interferon given continuously subcutaneously twice a week at a dose of 10 MU/m."( Phase II multicentre study of temozolomide in combination with interferon alpha-2b in metastatic malignant melanoma.
Crespo, C; del Muro, XG; Filipovich, E; García, M; Germà-Lluch, JR; López, JJ; Pérez, X; Rifà, J; Tres, A; Valladares, M, 2006
)
0.89
"Temozolomide in combination with interferon is a well-tolerated palliative regimen that has moderate activity against metastatic melanoma."( Phase II multicentre study of temozolomide in combination with interferon alpha-2b in metastatic malignant melanoma.
Crespo, C; del Muro, XG; Filipovich, E; García, M; Germà-Lluch, JR; López, JJ; Pérez, X; Rifà, J; Tres, A; Valladares, M, 2006
)
2.07
" In contrast, the cytotoxic drug temozolomide, when used in combination with HIF-1alpha knockdown, exhibited a superadditive and likely synergistic therapeutic effect compared with the monotherapy of either treatment alone in the D54MG glioma model."( Hypoxia-inducible factor-1 inhibition in combination with temozolomide treatment exhibits robust antitumor efficacy in vivo.
Albert, DH; Fesik, SW; Li, L; Lin, X; Shen, Y; Shoemaker, AR, 2006
)
0.86
"Our results show that the DNA alkylating agent temozolomide exhibits robust antitumor efficacy when used in combination with HIF-1 inhibition in D54MG-derived tumors, suggesting that the combination of temozolomide with HIF-1 inhibitors might be an effective regimen for cancer therapy."( Hypoxia-inducible factor-1 inhibition in combination with temozolomide treatment exhibits robust antitumor efficacy in vivo.
Albert, DH; Fesik, SW; Li, L; Lin, X; Shen, Y; Shoemaker, AR, 2006
)
0.83
" The observed antitumor activity warrants further evaluation of this combination as an alternative to or in combination with whole-brain radiation therapy for the treatment of multiple brain metastases."( Phase I study of capecitabine in combination with temozolomide in the treatment of patients with brain metastases from breast carcinoma.
Arun, B; Broglio, K; Buchholz, T; Francis, D; Groves, M; Hortobagyi, GN; Meyers, C; Rivera, E; Valero, V; Yin, G, 2006
)
0.59
" Three MMR-proficient melanoma cell clones with low or no MGMT activity were treated daily for 5 days with 50 micromol/l TMZ, alone or in combination with 5 micromol/l BG."( A single cycle of treatment with temozolomide, alone or combined with O(6)-benzylguanine, induces strong chemoresistance in melanoma cell clones in vitro: role of O(6)-methylguanine-DNA methyltransferase and the mismatch repair system.
Alvino, E; Bonmassar, E; Caporali, S; Caporaso, P; Castiglia, D; D'Atri, S; Fischer, F; Jiricny, J; Lacal, PM; Marra, G; Pepponi, R; Zambruno, G, 2006
)
0.62
" Randomized studies are needed to explore the role of celecoxib in combination with chemotherapy or as maintenance treatment in these patients."( Temozolomide in combination with celecoxib in patients with advanced melanoma. A phase II study of the Hellenic Cooperative Oncology Group.
Fountzilas, G; Frangia, K; Gogas, H; Mantzourani, M; Markopoulos, C; Middleton, M; Panagiotou, P; Papadopoulos, O; Pectasides, D; Polyzos, A; Stavrinidis, I; Tsoutsos, D; Vaiopoulos, G, 2006
)
1.78
"The aim of this study was to investigate the effect of temozolomide (TZM) in combination with X-rays on proliferation and migration in human glioma spheroids."( The inhibition of proliferation and migration of glioma spheroids exposed to temozolomide is less than additive if combined with irradiation.
Fehlauer, F; Muench, M; Rades, D; Richter, E, 2007
)
0.82
"To compare results of treatment with temozolomide or dacarbazine, in combination with an anthracycline, in dogs with relapsed or refractory lymphoma."( Efficacy of temozolomide or dacarbazine in combination with an anthracycline for rescue chemotherapy in dogs with lymphoma.
Cadile, CD; Dervisis, NG; Dominguez, PA; Kitchell, BE; Newman, RG; Sarbu, L; Swanson, CN, 2007
)
0.99
"The regimen, temozolomide combined with fotemustine, is an active and moderately safe first-line chemotherapy regimen with acceptable and easily manageable toxicities in patients with metastatic melanoma."( Temozolomide in combination with fotemustine in patients with metastatic melanoma.
Camlica, H; Tas, F; Topuz, E, 2008
)
2.16
" The effect of ZD6474, a potent inhibitor of VEGF-receptor-2, was evaluated in combination with either radiotherapy or temozolomide."( Effects of the VEGFR inhibitor ZD6474 in combination with radiotherapy and temozolomide in an orthotopic glioma model.
Bergenheim, AT; Bergström, P; Henriksson, R; Johansson, M; Sandström, M, 2008
)
0.78
" ZD6474 30 mg/kg was given alone or in combination with radiotherapy 12 Gy x 1 or with temozolomide 100 mg/kg for 3 days."( Effects of the VEGFR inhibitor ZD6474 in combination with radiotherapy and temozolomide in an orthotopic glioma model.
Bergenheim, AT; Bergström, P; Henriksson, R; Johansson, M; Sandström, M, 2008
)
0.8
"ZD6474 in combination with radiotherapy significantly decreased tumour area by 66% compared with controls whereas the combination with temozolomide decreased tumour area by 74%."( Effects of the VEGFR inhibitor ZD6474 in combination with radiotherapy and temozolomide in an orthotopic glioma model.
Bergenheim, AT; Bergström, P; Henriksson, R; Johansson, M; Sandström, M, 2008
)
0.78
"ZD6474 in combination with two standard modalities in the treatment of malignant glioma, radiotherapy and chemotherapy, markedly decreased the growth of an intracerebral experimental glioma."( Effects of the VEGFR inhibitor ZD6474 in combination with radiotherapy and temozolomide in an orthotopic glioma model.
Bergenheim, AT; Bergström, P; Henriksson, R; Johansson, M; Sandström, M, 2008
)
0.58
"A phase II trial was initiated to analyze the activity of continuously administered pioglitazone and rofecoxib combined with low-dose chemotherapy (capecitabine or temozolomide) in patients with high-grade gliomas (glioblastoma or anaplastic glioma)."( Low-dose chemotherapy in combination with COX-2 inhibitors and PPAR-gamma agonists in recurrent high-grade gliomas - a phase II study.
Baumgart, U; Bogdahn, U; Hau, P; Hirschmann, B; Kunz-Schughart, L; Muhleisen, H; Reichle, A; Ruemmele, P; Steinbrecher, A; Weimann, E, 2007
)
0.54
"To assess interim safety and tolerability of a 10-patient, Phase II pilot study using bevacizumab (BV) in combination with temozolomide (TMZ) and regional radiation therapy (RT) in the up-front treatment of patients with newly diagnosed glioblastoma."( Phase II pilot study of bevacizumab in combination with temozolomide and regional radiation therapy for up-front treatment of patients with newly diagnosed glioblastoma multiforme: interim analysis of safety and tolerability.
Bergsneider, M; Cloughesy, T; Filka, E; Graham, C; Lai, A; Liau, LM; McGibbon, B; Mischel, P; Nghiemphu, PL; Pope, W; Selch, M; Yong, WH, 2008
)
0.8
" At this time data are not sufficient to encourage routine off-label use of BV combined with TMZ/RT in the setting of newly diagnosed glioblastoma without longer follow-up, enrollment of additional patients, and thorough efficacy assessment."( Phase II pilot study of bevacizumab in combination with temozolomide and regional radiation therapy for up-front treatment of patients with newly diagnosed glioblastoma multiforme: interim analysis of safety and tolerability.
Bergsneider, M; Cloughesy, T; Filka, E; Graham, C; Lai, A; Liau, LM; McGibbon, B; Mischel, P; Nghiemphu, PL; Pope, W; Selch, M; Yong, WH, 2008
)
0.59
"6% and 1/6 rat survived more than 1 year in case of MRT combined with gadolinium injection."( Enhancement of survival of 9L gliosarcoma bearing rats following intracerebral delivery of drugs in combination with microbeam radiation therapy.
Bräuer-Krisch, E; Bravin, A; Keyriläinen, J; Le Duc, G; Régnard, P; Troprès, I, 2008
)
0.35
" The article reports safety, efficacy, pharmacokinetic, and pharmacodynamic results of the first-in-class trial of a PARP inhibitor, AG014699, combined with temozolomide in adults with advanced malignancy."( Phase I study of the poly(ADP-ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors.
Boddy, A; Calvert, H; Curtin, N; Dewji, R; Evans, J; Harris, A; Johnson, P; Jones, C; McHugh, P; Middleton, M; Newell, D; Olsen, A; Plummer, R; Robson, L; Steinfeldt, H; Wang, D; Wilson, R, 2008
)
0.76
" Dasatinib in combination with temozolomide more effectively increased the therapeutic efficacy of temozolomide than when dasatinib was combined with carboplatin or irinotecan."( Dasatinib-induced autophagy is enhanced in combination with temozolomide in glioma.
de Groot, J; LaFortune, T; Milano, V; Piao, Y, 2009
)
0.88
"To evaluate the toxicity and efficacy of preoperative intensity-modulated radiotherapy (IMRT) combined with temozolomide to improve local tumor control in soft-tissue sarcoma (STS)."( Preoperative intensity-modulated radiotherapy combined with temozolomide for locally advanced soft-tissue sarcoma.
Dinter, DJ; Hohenberger, P; Jakob, J; Ströbel, P; Wenz, F, 2009
)
0.81
" Drug combination of TMZ plus BG and PHA-848125 produced additive or synergistic effects on cell growth, depending on the cell line."( The cyclin-dependent kinase inhibitor PHA-848125 suppresses the in vitro growth of human melanomas sensitive or resistant to temozolomide, and shows synergistic effects in combination with this triazene compound.
Alvino, E; Bonmassar, E; Brasca, MG; Caporali, S; Castiglia, D; Ciomei, M; Covaciu, C; D'Atri, S; Garbin, A; Levati, L; Starace, G, 2010
)
0.57
" After irradiation of the symptomatic sites, intrathecal liposomal Ara-C every 2-4 weeks was combined with temozolomide 100 mg/m(2) day 1-5/7."( Neoplastic meningitis from breast cancer: feasibility and activity of long-term intrathecal liposomal Ara-C combined with dose-dense temozolomide.
Buhk, JH; Hoffmann, AL; Strik, H, 2009
)
0.77
"To evaluate maximum tolerated dose and recommended dose (RD) for phase II studies of topotecan (TPT) combined with temozolomide (TMZ) (TOTEM) in children and adolescents with relapsed or refractory solid malignancies."( Phase I study of topotecan in combination with temozolomide (TOTEM) in relapsed or refractory paediatric solid tumours.
Aerts, I; Chastagner, P; Chatelut, E; Corradini, N; Dias, N; Djafari, L; Frappaz, D; Gentet, JC; Geoerger, B; Landman-Parker, J; Le Deley, MC; Leblond, P; Ndiaye, A; Paci, A; Pasquet, M; Rubie, H; Schmitt, A; Vassal, G, 2010
)
0.83
" Our results demonstrate the potential of the anti-IGF-1R antibody alone and in combination with alkylating agents and support the therapeutic development of the AVE1642 for aggressive neuroblastoma."( Anti-insulin-like growth factor 1 receptor antibody EM164 (murine AVE1642) exhibits anti-tumour activity alone and in combination with temozolomide against neuroblastoma.
Brasme, JF; Daudigeos-Dubus, E; Debussche, L; Geoerger, B; Opolon, P; Vassal, G; Venot, C; Vrignaud, P, 2010
)
0.56
" Metronomic dosing of temozolomide (TMZ) combined with standard radiotherapy may improve survival by increasing the therapeutic index and anti-angiogenic effect of TMZ."( A multi-centre Canadian pilot study of metronomic temozolomide combined with radiotherapy for newly diagnosed paediatric brainstem glioma.
Bartels, U; Baruchel, S; Bouffet, E; Eisenstat, D; Gammon, J; Huang, A; Hukin, J; Johnston, DL; Samson, Y; Sharp, JR; Stempak, D; Stephens, D; Tabori, U, 2010
)
0.93
" This factorial design phase I/II protocol tested dose-dense temozolomide alone and combined with cytostatic agents."( A phase I factorial design study of dose-dense temozolomide alone and in combination with thalidomide, isotretinoin, and/or celecoxib as postchemoradiation adjuvant therapy for newly diagnosed glioblastoma.
Chang, E; Colman, H; Conrad, C; de Groot, J; Giglio, P; Gilbert, MR; Gonzalez, J; Groves, MD; Hess, K; Hunter, K; Levin, V; Mahajan, A; Puduvalli, V; Woo, S; Yung, WK, 2010
)
0.86
" On the basis of promising preclinical data, the safety and tolerability of therapy with the mTOR inhibitor RAD001 in combination with radiation (RT) and temozolomide (TMZ) was evaluated in this Phase I study."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.79
"All patients received weekly oral RAD001 in combination with standard chemoradiotherapy, followed by RAD001 in combination with standard adjuvant temozolomide."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.79
" On the basis of these results, the recommended Phase II dosage currently being tested is RAD001 70 mg/week in combination with standard chemoradiotherapy."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.59
"RAD001 in combination with RT/TMZ and adjuvant TMZ was reasonably well tolerated."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.59
"The phase II trial has been prepared to assess the effectiveness of BPA (250 mg/kg)-based NCT combined with X-ray irradiation and temozolomide (75 mg/m(2)) for the treatment of newly diagnosed GBM."( The status of Tsukuba BNCT trial: BPA-based boron neutron capture therapy combined with X-ray irradiation.
Aiyama, H; Endo, K; Ishikawa, E; Kumada, H; Matsumura, A; Mizumoto, M; Nakai, K; Nariai, T; Okumura, T; Shibata, Y; Takada, T; Tsuboi, K; Yamamoto, T; Yoshida, F; Zaboronok, A, 2011
)
0.57
" The major differences of our protocol from the other past studies were simultaneous use of both sodium borocapate and boronophenylalanine, and combination with fractionated X-ray irradiation."( Phase II clinical study of boron neutron capture therapy combined with X-ray radiotherapy/temozolomide in patients with newly diagnosed glioblastoma multiforme--study design and current status report.
Hiramatsu, R; Hirota, Y; Kawabata, S; Kirihata, M; Kuroiwa, T; Maruhashi, A; Miyata, S; Miyatake, S; Ono, K; Sakurai, Y; Takekita, Y, 2011
)
0.59
" Here, we describe 3 cases of GBM patients treated with autologous formalin-fixed tumor vaccine (AFTV) combined with TMZ."( Pathological changes after autologous formalin-fixed tumor vaccine therapy combined with temozolomide for glioblastoma - three case reports - .
Enomoto, T; Ishikawa, E; Matsumura, A; Morishita, Y; Nakai, K; Ohno, T; Sakamoto, N; Sato, M; Satomi, K; Takano, S; Tsuboi, K; Yamamoto, T, 2011
)
0.59
"The primary end point was surpassed showing promising activity of this bevacizumab/temozolomide combination with a favourable toxicity profile."( First-line temozolomide combined with bevacizumab in metastatic melanoma: a multicentre phase II trial (SAKK 50/07).
Cathomas, R; Dummer, R; Gillessen, S; Goldinger, SM; Mamot, C; Michielin, O; Mjhic-Probst, D; Moch, H; Ochsenbein, A; Schläppi, M; Schönewolf, N; Schraml, PH; Seifert, B; Simcock, M; von Moos, R, 2012
)
0.99
" The aim of the present study was to evaluate the efficacy and side effects of nimotuzumab in combination with chemotherapy for patients with malignant gliomas."( [Nimotuzumab in combination with chemotherapy for patients with malignant gliomas].
Chen, ZP; Jiang, XB; Mu, YG; Sai, K; Shen, D; Yang, QY; Zhang, XH, 2011
)
0.37
" Individualized chemotherapy was administered based on O(6)-methylguanine-DNA methyltransferase (MGMT) expression and previous chemotherapy responses in combined with nimotuzumab."( [Nimotuzumab in combination with chemotherapy for patients with malignant gliomas].
Chen, ZP; Jiang, XB; Mu, YG; Sai, K; Shen, D; Yang, QY; Zhang, XH, 2011
)
0.37
"Nimotuzumab in combination with chemotherapy has moderate activity in patients with malignant gliomas and the toxicities are well tolerable, therefore, worth further investigation."( [Nimotuzumab in combination with chemotherapy for patients with malignant gliomas].
Chen, ZP; Jiang, XB; Mu, YG; Sai, K; Shen, D; Yang, QY; Zhang, XH, 2011
)
0.37
" In vitro Southern Blot analysis and cytopathic effect assays demonstrate high anti-glioma potency, which was significantly increased in combination with temozolomide (TMZ), daunorubicin and cisplatin."( YB-1 dependent virotherapy in combination with temozolomide as a multimodal therapy approach to eradicate malignant glioma.
Anton, M; Gänsbacher, B; Haczek, C; Holm, PS; Holzmüller, R; Kasajima, A; Lage, H; Mantwill, K; Rognoni, E; Schlegel, J; Schuster, T; Treue, D; Weichert, W, 2011
)
0.82
"To study the efficacy and safety of 3-dimensional conformal radiotherapy combined with temozolomide (TMZ) for gliomas."( [Efficacy and safety of 3-dimensional conformal radiotherapy combined with temozolomide for glioma].
Tu, Q; Wang, L; Zhou, R; Zhou, W, 2011
)
0.82
" The patients received temozolomide alone,3-dimensional conformal radiotherapy alone,3-dimensional conformal radiotherapy combined with temozolomide in the chemotherapy group,the radiotherapy group and the comprehensive therapy group respectively."( [Efficacy and safety of 3-dimensional conformal radiotherapy combined with temozolomide for glioma].
Tu, Q; Wang, L; Zhou, R; Zhou, W, 2011
)
0.91
"Three-dimensional conformal radiotherapy combined with temozolomide is more effective for gliomas than the simple 3-dimensional conformal radiotherapy and the temozolomide chemotherapy alone."( [Efficacy and safety of 3-dimensional conformal radiotherapy combined with temozolomide for glioma].
Tu, Q; Wang, L; Zhou, R; Zhou, W, 2011
)
0.85
"To study the safety and efficacy of three-dimensional conformal radiotherapy in combination with temozolomide in treatment of patients with diffuse brainstem glioma."( [Safety and efficacy of three-dimensional conformal radiotherapy combined with temozolomide in treatment of diffuse brainstem gliomas].
Cai, CL; Fang, HH; Kang, JB; Li, FM; Nie, Q, 2011
)
0.81
" It is likely that clinical testing of these agents will be in combination with standard therapies to harness the apoptotic potential of both the agents."( The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models.
Alferez, D; Davies, BR; Heaton, SP; Heier, A; Holt, SV; Logié, A; Odedra, R; Smith, PD; Wilkinson, RW, 2012
)
0.38
"The aim of this study was to identify agents, which would be likely to offer clinical benefit when combined with selumetinib."( The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models.
Alferez, D; Davies, BR; Heaton, SP; Heier, A; Holt, SV; Logié, A; Odedra, R; Smith, PD; Wilkinson, RW, 2012
)
0.38
" In several models we observed that continuous exposure to selumetinib in combination with docetaxel results in tumour regression."( The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models.
Alferez, D; Davies, BR; Heaton, SP; Heier, A; Holt, SV; Logié, A; Odedra, R; Smith, PD; Wilkinson, RW, 2012
)
0.38
"The data presented suggests that MEK inhibition in combination with several standard chemotherapeutics or an Aurora B kinase inhibitor is a promising clinical strategy."( The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models.
Alferez, D; Davies, BR; Heaton, SP; Heier, A; Holt, SV; Logié, A; Odedra, R; Smith, PD; Wilkinson, RW, 2012
)
0.38
" As HSP27 and AKT interact to regulate the activity of each other, we determined whether inhibition of HSP27 was better than targeting SPARC as a therapeutic approach to inhibit both SPARC-induced glioma cell invasion and survival."( Inhibition of HSP27 alone or in combination with pAKT inhibition as therapeutic approaches to target SPARC-induced glioma cell survival.
Brodie, C; Brown, SL; Golembieski, WA; King, DA; Rempel, SA; Schultz, CR, 2012
)
0.38
"We conclude that inhibition of HSP27 alone, or in combination with pAKT inhibitor IV, may be an effective therapeutic approach to inhibit SPARC-induced glioma cell invasion and survival in SPARC-positive/PTEN-wildtype and SPARC-positive/PTEN-null tumors, respectively."( Inhibition of HSP27 alone or in combination with pAKT inhibition as therapeutic approaches to target SPARC-induced glioma cell survival.
Brodie, C; Brown, SL; Golembieski, WA; King, DA; Rempel, SA; Schultz, CR, 2012
)
0.38
"A retrospective study assessing treatment-related toxicities in tumor-bearing cats treated with temozolomide (TMZ) alone or in combination with doxorubicin was conducted."( Treatment-related toxicities in tumor-bearing cats treated with temozolomide alone or in combination with doxorubicin: a pilot assessment.
Dervisis, NG; Gagnon, J; Kitchell, BE, 2012
)
0.84
" The primary objectives of this randomized phase 2 trial were to determine the safety and efficacy of cilengitide when combined with radiation and temozolomide for patients with newly diagnosed glioblastoma multiforme and to select a dose for comparative clinical testing."( A safety run-in and randomized phase 2 study of cilengitide combined with chemoradiation for newly diagnosed glioblastoma (NABTT 0306).
Batchelor, T; Brem, S; Fisher, JD; Grossman, SA; Hegi, ME; Lesser, G; Mikkelsen, T; Nabors, LB; Olsen, J; Peereboom, D; Rosenfeld, MR; Ye, X, 2012
)
0.58
"Cilengitide was well tolerated when combined with standard chemoradiation and may improve survival for patients newly diagnosed with glioblastoma multiforme regardless of MGMT methylation status."( A safety run-in and randomized phase 2 study of cilengitide combined with chemoradiation for newly diagnosed glioblastoma (NABTT 0306).
Batchelor, T; Brem, S; Fisher, JD; Grossman, SA; Hegi, ME; Lesser, G; Mikkelsen, T; Nabors, LB; Olsen, J; Peereboom, D; Rosenfeld, MR; Ye, X, 2012
)
0.38
" At the time of last relapse, temozolomide was administered alone and combined with external beam radiation therapy."( Temozolomide combined with radiotherapy in the treatment of recurrent cranial meningioma previously treated with multiple surgical resections and two sessions of radiosurgery: a case report and literature review.
Alongi, F; Clerici, E; Navarria, P; Rognone, E; Santoro, A; Scorsetti, M; Simonelli, M,
)
1.86
"To evaluate the toxicity and maximum tolerated dose (MTD) of arsenic trioxide (ATO) in combination with temozolomide (TMZ) and radiation therapy (RT) in malignant gliomas."( Phase I study of arsenic trioxide and temozolomide in combination with radiation therapy in patients with malignant gliomas.
Chandler, JP; Grimm, SA; Jovanovic, B; Levy, RM; Marymont, M; McCarthy, K; Muro, K; Newman, SB; Raizer, JJ, 2012
)
0.86
"A phase I, dose-finding study of vorinostat in combination with temozolomide (TMZ) was conducted to determine the maximum tolerated dose (MTD), safety, and pharmacokinetics in patients with high-grade glioma (HGG)."( Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03.
Ames, MM; Chang, SM; Cloughesy, TF; Desideri, S; Drappatz, J; Espinoza-Delgado, I; Gilbert, MR; Kuhn, JG; Lamborn, KR; Lassman, AB; Lee, EQ; Lieberman, FS; McGovern, RM; Prados, MD; Puduvalli, VK; Reid, JM; Robins, HI; Wen, PY; Xu, J; Ye, X; Yung, WK, 2012
)
0.87
" Part 1 was a dose-escalation study of vorinostat in combination with TMZ 150 mg/m(2)/day for 5 days every 28 days."( Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03.
Ames, MM; Chang, SM; Cloughesy, TF; Desideri, S; Drappatz, J; Espinoza-Delgado, I; Gilbert, MR; Kuhn, JG; Lamborn, KR; Lassman, AB; Lee, EQ; Lieberman, FS; McGovern, RM; Prados, MD; Puduvalli, VK; Reid, JM; Robins, HI; Wen, PY; Xu, J; Ye, X; Yung, WK, 2012
)
0.63
"In part 1, the MTD of vorinostat administered on days 1 to 7 and 15 to 21 of every 28-day cycle, in combination with TMZ, was 500 mg daily."( Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03.
Ames, MM; Chang, SM; Cloughesy, TF; Desideri, S; Drappatz, J; Espinoza-Delgado, I; Gilbert, MR; Kuhn, JG; Lamborn, KR; Lassman, AB; Lee, EQ; Lieberman, FS; McGovern, RM; Prados, MD; Puduvalli, VK; Reid, JM; Robins, HI; Wen, PY; Xu, J; Ye, X; Yung, WK, 2012
)
0.63
"Vorinostat in combination with temozolomide is well tolerated in patients with HGG."( Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03.
Ames, MM; Chang, SM; Cloughesy, TF; Desideri, S; Drappatz, J; Espinoza-Delgado, I; Gilbert, MR; Kuhn, JG; Lamborn, KR; Lassman, AB; Lee, EQ; Lieberman, FS; McGovern, RM; Prados, MD; Puduvalli, VK; Reid, JM; Robins, HI; Wen, PY; Xu, J; Ye, X; Yung, WK, 2012
)
0.92
" One of the most interesting combinations that have started to become part of the therapeutic arsenal in the daily clinic is dose-dense temozolomide in combination with bevacizumab."( A review of dose-dense temozolomide alone and in combination with bevacizumab in patients with first relapse of glioblastoma.
Bergqvist, M; Bergström, S; Blomquist, E; Ekman, S; Henriksson, R; Johansson, F, 2012
)
0.89
" (177)Lu-octreotate, in combination with capecitabine and temozolomide, is well tolerated in patients with advanced low-grade NETs, and shows substantial tumor control rates."( Phase I-II study of radiopeptide 177Lu-octreotate in combination with capecitabine and temozolomide in advanced low-grade neuroendocrine tumors.
Claringbold, PG; Price, RA; Turner, JH, 2012
)
0.85
" This drug combination significantly impaired the sphere-forming ability of GSCs in vitro and tumor formation in vivo, leading to increase in the overall survival of mice bearing orthotopic inoculation of GSCs."( Effective elimination of cancer stem cells by a novel drug combination strategy.
Chen, G; Colman, H; Feng, L; Huang, P; Keating, MJ; Li, X; Wang, F; Wang, J; Wang, L; Xu, RH; Yuan, S; Zhang, H, 2013
)
0.39
" We hypothesized that epigenetic modulators will reverse chemotherapy resistance, and in this article, we report studies that sought to determine the recommended phase 2 dose (RP2D), safety, and efficacy of decitabine (DAC) combined with TMZ."( Safety and efficacy of decitabine in combination with temozolomide in metastatic melanoma: a phase I/II study and pharmacokinetic analysis.
Beumer, JH; Buch, SC; Christner, S; Egorin, MJ; Kirkwood, JM; Lin, Y; Moschos, S; Tarhini, AA; Tawbi, HA, 2013
)
0.64
"This study suggests that ABT-888 has the clinical potential to enhance the current standard treatment for GBM, in combination with conventional chemo-radiotherapy."( Evaluation of poly (ADP-ribose) polymerase inhibitor ABT-888 combined with radiotherapy and temozolomide in glioblastoma.
Barazzuol, L; Burnet, NG; Jena, R; Jeynes, JC; Kirkby, KJ; Kirkby, NF; Meira, LB, 2013
)
0.61
"Daily oral everolimus (10 mg) combined with both concurrent radiation and temozolomide followed by adjuvant temozolomide is well tolerated, with an acceptable toxicity profile."( RTOG 0913: a phase 1 study of daily everolimus (RAD001) in combination with radiation therapy and temozolomide in patients with newly diagnosed glioblastoma.
Chinnaiyan, P; Corn, BW; Dipetrillo, TA; Mehta, MP; Rojiani, AM; Wen, PY; Wendland, M; Won, M, 2013
)
0.84
"To evaluate the efficacy of pulsed low-dose radiation therapy (PLRT) combined with temozolomide (TMZ) as a novel treatment approach for radioresistant glioblastoma multiforme (GBM) in a murine model."( Pulsed versus conventional radiation therapy in combination with temozolomide in a murine orthotopic model of glioblastoma multiforme.
Chunta, JL; Grills, IS; Huang, J; Krueger, SA; Lee, DY; Marples, B; Martinez, AA; Park, SS; Wilson, GD, 2013
)
0.85
"Patients treated within a Phase I/II Trial with a carbon ion boost were compared retrospectively with randomly chosen patients treated with photons or photons in combination with TMZ in a retrospective analysis."( Comparison of carbon ion radiotherapy to photon radiation alone or in combination with temozolomide in patients with high-grade gliomas: explorative hypothesis-generating retrospective analysis.
Bruckner, T; Combs, SE; Debus, J; Kamada, T; Kieser, M; Mizoe, JE; Tsujii, H, 2013
)
0.61
"To assess objective response rate (ORR) after two cycles of temozolomide in combination with topotecan (TOTEM) in children with refractory or relapsed neuroblastoma."( Phase II study of temozolomide in combination with topotecan (TOTEM) in relapsed or refractory neuroblastoma: a European Innovative Therapies for Children with Cancer-SIOP-European Neuroblastoma study.
Aerts, I; Amoroso, L; Boubaker, A; Casanova, M; Chastagner, P; Courbon, F; Devos, A; Di Giannatale, A; Dias-Gastellier, N; Ducassoul, S; Geoerger, B; Landman-Parker, J; Le Deley, MC; Malekzadeh, K; Mc Hugh, K; Munzer, C; Riccardi, R; Rubie, H; Verschuur, A; Zwaan, CM, 2014
)
0.98
" In the present analysis, we retrospectively investigated the feasibility and effectiveness of bevacizumab combined with ICE in patients with glioblastoma at second relapse during ICE treatment."( Retrospective analysis of bevacizumab in combination with ifosfamide, carboplatin, and etoposide in patients with second recurrence of glioblastoma.
Arakawa, Y; Fujimoto, K; Kikuchi, T; Kunieda, T; Miyamoto, S; Mizowaki, T; Murata, D; Takagi, Y; Takahashi, JC, 2013
)
0.39
"To determine the maximum tolerated dose of irinotecan administered every 2 weeks, in combination with a fixed and continuous administration of temozolomide, in patients with glioblastoma at first relapse."( A phase I study of irinotecan in combination with metronomic temozolomide in patients with recurrent glioblastoma.
Balañá, C; Gallego, O; García, JL; Iglesias, L; Pérez, P; Reynés, G, 2014
)
0.84
" More generally, these results suggest that traditional therapy in combination with local, as opposed to systemic, delivery of angiogenesis inhibitors may be able to increase median survival for patients with glioblastoma."( Local delivery of angiogenesis-inhibitor minocycline combined with radiotherapy and oral temozolomide chemotherapy in 9L glioma.
Bow, H; Brem, H; Hwang, LS; Murray, L; Salditch, Q; Schildhaus, N; Tyler, B; Weingart, J; Xing, J; Ye, X; Zhang, Y, 2014
)
0.62
"To evaluate the safety and efficacy of nimotuzumab, a humanized monoclonal antibody specific for the epidermal growth factor receptor (EGFR), in combination with temozolomide (TMZ) and radiation therapy (RT) in the treatment of newly diagnosed glioblastoma (GBM) in Chinese patients."( Nimotuzumab, a humanized monoclonal antibody specific for the EGFR, in combination with temozolomide and radiation therapy for newly diagnosed glioblastoma multiforme: First results in Chinese patients.
Chen, S; Dai, JZ; Pan, L; Sheng, XF; Wang, Y, 2016
)
0.85
" During RT, concurrent TMZ was given daily at 75 mg/m(2) for 40-42 days, combined with six weekly infusions of nimotuzumab at a 200 mg dose."( Nimotuzumab, a humanized monoclonal antibody specific for the EGFR, in combination with temozolomide and radiation therapy for newly diagnosed glioblastoma multiforme: First results in Chinese patients.
Chen, S; Dai, JZ; Pan, L; Sheng, XF; Wang, Y, 2016
)
0.66
" This phase I open-label, noncontrolled dose escalation study was performed to determine the safety and maximum tolerated dose (MTD) of Sb in combination with radiation therapy (RT) and temozolomide (TMZ) in 17 patients with newly diagnosed high-grade glioma."( Phase I study of sorafenib combined with radiation therapy and temozolomide as first-line treatment of high-grade glioma.
Ben Aissa, A; Bodmer, A; Dietrich, PY; Dunkel, N; Espeli, V; Hottinger, AF; Hundsberger, T; Mach, N; Schaller, K; Squiban, D; Vargas, MI; Weber, DC, 2014
)
0.83
"Patients were treated with RT (60 Gy in 2 Gy fractions) combined with TMZ 75 mg m(-2) daily, and Sb administered at three dose levels (200 mg daily, 200 mg BID, and 400 mg BID) starting on day 8 of RT."( Phase I study of sorafenib combined with radiation therapy and temozolomide as first-line treatment of high-grade glioma.
Ben Aissa, A; Bodmer, A; Dietrich, PY; Dunkel, N; Espeli, V; Hottinger, AF; Hundsberger, T; Mach, N; Schaller, K; Squiban, D; Vargas, MI; Weber, DC, 2014
)
0.64
"Although Sb can be combined with RT and TMZ, significant side effects and moderate outcome results do not support further clinical development in malignant gliomas."( Phase I study of sorafenib combined with radiation therapy and temozolomide as first-line treatment of high-grade glioma.
Ben Aissa, A; Bodmer, A; Dietrich, PY; Dunkel, N; Espeli, V; Hottinger, AF; Hundsberger, T; Mach, N; Schaller, K; Squiban, D; Vargas, MI; Weber, DC, 2014
)
0.64
"Twenty-six patients received CEP-9722 150-1,000 mg/day combined with temozolomide."( Phase 1 dose-escalation study of the PARP inhibitor CEP-9722 as monotherapy or in combination with temozolomide in patients with solid tumors.
Aissat-Daudigny, L; Brown, PD; Cambois, A; Campone, M; Moachon, G; Plummer, R; Stephens, P, 2014
)
0.85
"WBRT followed by IMBRT combined with concomitant TMZ is well tolerated, yielding an encouraging objective response rate; however, overall survival improves slightly comparing with RTOG 9508 randomized trial."( Whole brain radiation therapy followed by intensity-modulated boosting treatment combined with concomitant temozolomide for brain metastases from non-small-cell lung cancer.
Jiang, Z; Leng, C; Liang, S; Liu, H; Lu, F; Lu, S; Qi, X; Shi, J; Wang, Q; Wang, S, 2014
)
0.62
"2 mg/kg three times weekly for 2 weeks (starting on day 1), in combination with oral panobinostat 10, 20, or 30 mg every 96 h (starting on day 8), and oral temozolomide 150 mg/m(2)/day on days 9 through 13."( Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide).
Deutsch, J; Frees, M; Laux, D; Leon-Ferre, R; Milhem, M; Smith, BJ; Xia, C, 2014
)
0.8
"This triple agent of dual epigenetic therapy in combination with traditional chemotherapy was generally well tolerated by the cohort and appeared safe to be continued in a Phase II trial."( Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide).
Deutsch, J; Frees, M; Laux, D; Leon-Ferre, R; Milhem, M; Smith, BJ; Xia, C, 2014
)
0.61
" Data from phase 2 trials suggest that it has antitumour activity as a single agent in recurrent glioblastoma and in combination with standard temozolomide chemoradiotherapy in newly diagnosed glioblastoma (particularly in tumours with methylated MGMT promoter)."( Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): a multicentre, randomised, open-label, phase 3 trial.
Adamska, K; Aldape, KD; Brandes, AA; Erridge, SC; Gorlia, T; Grujicic, D; Gupta, T; Hau, P; Hegi, ME; Herrlinger, U; Hicking, C; Hong, YK; Kim, CY; Kortmann, RD; Lhermitte, B; Markivskyy, A; McBain, C; Nabors, LB; Nam, DH; Perry, J; Picard, M; Pietsch, T; Rao, N; Reardon, DA; Schnell, O; Shen, CC; Steinbach, JP; Stupp, R; Taphoorn, MJ; Tarnawski, R; Thurzo, L; Tonn, JC; van den Bent, MJ; Weller, M; Weyerbrock, A; Wick, W; Wiegel, T, 2014
)
0.6
" In the present work, TMZ was combined with a specific SKI, and the cytotoxic effect of each drug alone or in combination was tested on GBM cell lines."( A sphingosine kinase inhibitor combined with temozolomide induces glioblastoma cell death through accumulation of dihydrosphingosine and dihydroceramide, endoplasmic reticulum stress and autophagy.
Choi, J; Kopp-Schneider, A; Noack, J; Régnier-Vigouroux, A; Richter, K, 2014
)
0.66
" We reasoned this could be more effective if combined with a vaccine that preferentially targeted TMZ-resistant cells."( Dendritic cell vaccination combined with temozolomide retreatment: results of a phase I trial in patients with recurrent glioblastoma multiforme.
Ancelet, LR; Bauer, E; Dzhelali, M; Findlay, MP; Gasser, O; Hamilton, DA; Hermans, IF; Hunn, MK; Mester, B; Sharples, KJ; Wood, CE, 2015
)
0.68
"After a biopsy (6 patients) or a resection (13 patients) of a newly diagnosed GBM, 19 patients received radiotherapy (30 fractions of 2 Gy) in combination with daily TMZ 75 mg/m(2) and BV 10 mg/kg on days 1, 14, and 28, followed by 6 monthly cycles of TMZ 150-200 mg/m(2) on days 1-5."( Bevacizumab in combination with radiotherapy and temozolomide for patients with newly diagnosed glioblastoma multiforme.
Reijneveld, JC; Richel, DJ; Stalpers, LJ; van Furth, WR; van Linde, ME; Verheul, HM; Verhoeff, JJ, 2015
)
0.67
"Wee1 regulates key DNA damage checkpoints, and in this study, the efficacy of the Wee1 inhibitor MK-1775 was evaluated in glioblastoma multiforme (GBM) xenograft models alone and in combination with radiation and/or temozolomide."( The Efficacy of the Wee1 Inhibitor MK-1775 Combined with Temozolomide Is Limited by Heterogeneous Distribution across the Blood-Brain Barrier in Glioblastoma.
Agar, NY; Bakken, KK; Calligaris, D; Carlson, BL; Decker, PA; Eckel-Passow, JE; Elmquist, WF; Evans, DL; Gupta, SK; Iyekegbe, DO; Lou, Z; Ma, B; Mueller, D; Pokorny, JL; Pucci, V; Sarkaria, JN; Schroeder, MA; Shumway, SD, 2015
)
0.85
"In vitro MK-1775 efficacy alone and in combination with temozolomide, and the impact on DNA damage, was analyzed by Western blotting and γH2AX foci formation."( The Efficacy of the Wee1 Inhibitor MK-1775 Combined with Temozolomide Is Limited by Heterogeneous Distribution across the Blood-Brain Barrier in Glioblastoma.
Agar, NY; Bakken, KK; Calligaris, D; Carlson, BL; Decker, PA; Eckel-Passow, JE; Elmquist, WF; Evans, DL; Gupta, SK; Iyekegbe, DO; Lou, Z; Ma, B; Mueller, D; Pokorny, JL; Pucci, V; Sarkaria, JN; Schroeder, MA; Shumway, SD, 2015
)
0.91
" However, there was no sensitizing effect of MK-1775 when combined with temozolomide in vitro."( The Efficacy of the Wee1 Inhibitor MK-1775 Combined with Temozolomide Is Limited by Heterogeneous Distribution across the Blood-Brain Barrier in Glioblastoma.
Agar, NY; Bakken, KK; Calligaris, D; Carlson, BL; Decker, PA; Eckel-Passow, JE; Elmquist, WF; Evans, DL; Gupta, SK; Iyekegbe, DO; Lou, Z; Ma, B; Mueller, D; Pokorny, JL; Pucci, V; Sarkaria, JN; Schroeder, MA; Shumway, SD, 2015
)
0.89
" Primary GBM cells were treated with VPA as a monotherapy and in combination with temozolomide and irradiation."( The effect of valproic acid in combination with irradiation and temozolomide on primary human glioblastoma cells.
Cosgrove, L; Day, B; Fay, M; Head, R; Hosein, AN; Lim, YC; Martin, JH; Rose, S; Sminia, P; Stringer, B, 2015
)
0.88
" This phase II, randomized, open-label, multicenter trial investigated the efficacy and safety of 2 dose regimens of the selective integrin inhibitor cilengitide combined with standard chemoradiotherapy in patients with newly diagnosed glioblastoma and an unmethylated MGMT promoter."( Two cilengitide regimens in combination with standard treatment for patients with newly diagnosed glioblastoma and unmethylated MGMT gene promoter: results of the open-label, controlled, randomized phase II CORE study.
Ashby, L; Depenni, R; Fink, KL; Grujicic, D; Hegi, ME; Hicking, C; Lhermitte, B; Mazurkiewicz, M; Mikkelsen, T; Nabors, LB; Nam, DH; Perry, JR; Picard, M; Reardon, DA; Salacz, M; Tarnawski, R; Zagonel, V, 2015
)
0.42
" Cilengitide was administered intravenously in combination with daily temozolomide (TMZ) and concomitant radiotherapy (RT; wk 1-6), followed by TMZ maintenance therapy (TMZ/RT→TMZ)."( Two cilengitide regimens in combination with standard treatment for patients with newly diagnosed glioblastoma and unmethylated MGMT gene promoter: results of the open-label, controlled, randomized phase II CORE study.
Ashby, L; Depenni, R; Fink, KL; Grujicic, D; Hegi, ME; Hicking, C; Lhermitte, B; Mazurkiewicz, M; Mikkelsen, T; Nabors, LB; Nam, DH; Perry, JR; Picard, M; Reardon, DA; Salacz, M; Tarnawski, R; Zagonel, V, 2015
)
0.65
"Standard and intensive cilengitide dose regimens were well tolerated in combination with TMZ/RT→TMZ."( Two cilengitide regimens in combination with standard treatment for patients with newly diagnosed glioblastoma and unmethylated MGMT gene promoter: results of the open-label, controlled, randomized phase II CORE study.
Ashby, L; Depenni, R; Fink, KL; Grujicic, D; Hegi, ME; Hicking, C; Lhermitte, B; Mazurkiewicz, M; Mikkelsen, T; Nabors, LB; Nam, DH; Perry, JR; Picard, M; Reardon, DA; Salacz, M; Tarnawski, R; Zagonel, V, 2015
)
0.42
" Abemaciclib antitumor activity was assessed in subcutaneous and orthotopic glioma models alone and in combination with standard of care temozolomide (TMZ)."( Brain Exposure of Two Selective Dual CDK4 and CDK6 Inhibitors and the Antitumor Activity of CDK4 and CDK6 Inhibition in Combination with Temozolomide in an Intracranial Glioblastoma Xenograft.
Ajamie, RT; De Dios, A; Gelbert, LM; Kulanthaivel, P; Raub, TJ; Sanchez-Martinez, C; Sawada, GA; Shannon, HE; Staton, BA; Wishart, GN, 2015
)
0.82
" In this study, we found that PI3K inhibitor combined with miR-125b inhibitor caused a marked increase of TMZ-induced GSC proliferation and invasiveness inhibition."( PI3K inhibitor combined with miR-125b inhibitor sensitize TMZ-induced anti-glioma stem cancer effects through inactivation of Wnt/β-catenin signaling pathway.
Fei, X; Shi, L; Wang, Z; You, Y, 2015
)
0.42
"The primary aim of this Phase I study was to determine the maximum tolerated dose (MTD) of TPI 287 and the safety and tolerability of TPI 287 alone and in combination with temozolomide (TMZ) in pediatric patients with refractory or recurrent neuroblastoma or medulloblastoma."( A Phase 1 Trial of TPI 287 as a Single Agent and in Combination With Temozolomide in Patients with Refractory or Recurrent Neuroblastoma or Medulloblastoma.
Ashikaga, T; Bergendahl, G; DeSarno, M; Eslin, D; Ferguson, W; Hanna, GK; Higgins, T; Kaplan, J; Kraveka, J; Mitchell, D; Roberts, W; Sholler, GL; Werff, AV, 2016
)
0.86
"Eighteen patients were enrolled to a phase I dose escalation trial of weekly intravenous infusion of TPI 287 for two 28-day cycles with toxicity monitoring to determine the MTD, followed by two cycles of TPI 287 in combination with TMZ."( A Phase 1 Trial of TPI 287 as a Single Agent and in Combination With Temozolomide in Patients with Refractory or Recurrent Neuroblastoma or Medulloblastoma.
Ashikaga, T; Bergendahl, G; DeSarno, M; Eslin, D; Ferguson, W; Hanna, GK; Higgins, T; Kaplan, J; Kraveka, J; Mitchell, D; Roberts, W; Sholler, GL; Werff, AV, 2016
)
0.67
"We determined the MTD of TPI 287 alone and in combination with temozolomide to be 125 mg/m(2)."( A Phase 1 Trial of TPI 287 as a Single Agent and in Combination With Temozolomide in Patients with Refractory or Recurrent Neuroblastoma or Medulloblastoma.
Ashikaga, T; Bergendahl, G; DeSarno, M; Eslin, D; Ferguson, W; Hanna, GK; Higgins, T; Kaplan, J; Kraveka, J; Mitchell, D; Roberts, W; Sholler, GL; Werff, AV, 2016
)
0.91
" Pharmacokinetic testing did not show evidence of drug-drug interaction between irinotecan and alisertib."( Phase I Study of the Aurora A Kinase Inhibitor Alisertib in Combination With Irinotecan and Temozolomide for Patients With Relapsed or Refractory Neuroblastoma: A NANT (New Approaches to Neuroblastoma Therapy) Trial.
Bagatell, R; Courtier, J; Czarnecki, S; DuBois, SG; Fox, E; Goodarzian, F; Groshen, S; Hawkins, R; Kudgus, RA; Lai, H; Malvar, J; Marachelian, A; Maris, JM; Matthay, KK; Mosse, YP; Reid, JM; Shimada, H; Tsao-Wei, D; Wagner, L, 2016
)
0.65
" This phase I study aims to evaluate its safety, maximum tolerated dose (MTD), pharmacodynamic effect, and preliminary efficacy when combined with adjuvant temozolomide in GBM patients after standard chemoradiotherapy."( A phase I study to repurpose disulfiram in combination with temozolomide to treat newly diagnosed glioblastoma after chemoradiotherapy.
Campian, JL; DeWees, TA; Gujar, AD; Huang, J; Kim, AH; Lockhart, AC; Tran, DD; Tsien, CI, 2016
)
0.87
" We show that GSI in combination with RT and TMZ attenuates proliferation, decreases 3D spheroid growth and results into a marked reduction in clonogenic survival in primary and established glioma cell lines."( NOTCH blockade combined with radiation therapy and temozolomide prolongs survival of orthotopic glioblastoma.
Barbeau, LM; Chalmers, AJ; Eekers, DB; Granton, PV; Groot, AJ; Habets, R; Iglesias, VS; King, H; Prickaerts, J; Short, SC; Theys, J; van Hoof, SJ; Verhaegen, F; Vooijs, M; Yahyanejad, S, 2016
)
0.69
" We investigated the pharmacologic characteristics of OTX015 as a single agent and combined with targeted therapy or conventional chemotherapies in glioblastoma cell lines."( OTX015 (MK-8628), a novel BET inhibitor, displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma models.
Astorgues-Xerri, L; Bekradda, M; Berenguer-Daizé, C; Cayol, M; Cvitkovic, E; Lokiec, F; MacKenzie, S; Noel, K; Odore, E; Ouafik, L; Rezai, K; Riveiro, ME, 2016
)
0.43
" Subsequently, each agent was used in combination with CNDAC at fixed concentration ratios."( Mechanism-Based Drug Combinations with the DNA Strand-Breaking Nucleoside Analog CNDAC.
Hargis, S; Jiang, Y; Liu, X; Nowak, B; Plunkett, W, 2016
)
0.43
"We observed no survival benefit in young adults (age < 50) with anaplastic glioma when treated with TMZ combined with RT."( Radiation combined with temozolomide contraindicated for young adults diagnosed with anaplastic glioma.
Cai, J; Jiang, T; Li, S; Li, W; Peng, X; Qiu, X; Wang, Y; Wu, C; Yang, P; Yao, K; You, G; Zhang, C; Zhang, W, 2016
)
0.74
"To observe the effect of RITA, a small molecule that targets p53, combined with temozolomide (TMZ) on proliferation, colony formation and apoptosis of human glioblastoma U87 cells and explore the underlying mechanism."( [RITA combined with temozolomide inhibits the proliferation of human glioblastoma U87 cells].
Cao, ZX; Feng, XL; He, XY; Song, XP; Wu, QH; Xiao, WW; Zeng, HC; Zhang, B, 2016
)
0.98
" RITA combined with TMZ caused a more significant inhibition of U87 cells (29."( [RITA combined with temozolomide inhibits the proliferation of human glioblastoma U87 cells].
Cao, ZX; Feng, XL; He, XY; Song, XP; Wu, QH; Xiao, WW; Zeng, HC; Zhang, B, 2016
)
0.76
" The present study used a patient-derived orthotopic xenograft (PDOX) nude-mouse model of melanoma with a BRAF-V600E mutation to determine the efficacy of temozolomide (TEM) combined with tumor-targeting Salmonella typhimurium A1-R."( Tumor-targeting Salmonella typhimurium A1-R combined with temozolomide regresses malignant melanoma with a BRAF-V600E mutation in a patient-derived orthotopic xenograft (PDOX) model.
Chmielowski, B; Dry, SM; Eilber, FC; Hoffman, RM; Igarashi, K; Kawaguchi, K; Kiyuna, T; Li, Y; Murakami, T; Nelson, SD; Russell, TA; Singh, A; Unno, M; Zhang, Y; Zhao, M, 2016
)
0.88
" This phase I study evaluated the pharmacokinetics and mass balance of veliparib administered alone and in combination with temozolomide, and assessed any potential pharmacokinetic drug-drug interaction between veliparib and temozolomide."( Clinical Pharmacokinetics and Mass Balance of Veliparib in Combination with Temozolomide in Subjects with Nonhematologic Malignancies.
Giranda, V; Munasinghe, W; Nuthalapati, S; Xiong, H, 2018
)
0.92
"This was an open-label, dose-escalation study of veliparib in combination with temozolomide in 42 subjects with nonhematologic malignancies."( Clinical Pharmacokinetics and Mass Balance of Veliparib in Combination with Temozolomide in Subjects with Nonhematologic Malignancies.
Giranda, V; Munasinghe, W; Nuthalapati, S; Xiong, H, 2018
)
0.94
"New therapeutic agents in combination with the standard Stupp protocol (a protocol about the temozolomide combined with radiotherapy treatment with glioblastoma was research by Stupp R in 2005) were assessed to evaluate whether they were superior to the Stupp protocol alone, to determine the optimum treatment regimen for patients with newly diagnosed glioblastoma."( The interventional effect of new drugs combined with the Stupp protocol on glioblastoma: A network meta-analysis.
Chen, T; Fu, A; Li, J; Li, M; Song, X; Zhu, J, 2017
)
0.67
"The use of novel therapeutic agents in combination with the Stupp protocol were all shown to be superior than the Stupp protocol alone for the treatment of newly diagnosed glioblastoma, ranked as follows: cilengitide 2000mg/5/week, bevacizumab in combination with irinotecan, nimotuzumab, bevacizumab, cilengitide 2000mg/2/week, cytokine-induced killer cell immunotherapy, and the Stupp protocol."( The interventional effect of new drugs combined with the Stupp protocol on glioblastoma: A network meta-analysis.
Chen, T; Fu, A; Li, J; Li, M; Song, X; Zhu, J, 2017
)
0.46
"All intervention drugs evaluated in our study were superior to the Stupp protocol alone when used in combination with it."( The interventional effect of new drugs combined with the Stupp protocol on glioblastoma: A network meta-analysis.
Chen, T; Fu, A; Li, J; Li, M; Song, X; Zhu, J, 2017
)
0.46
"We evaluated stereotactic volume modulated arc radiotherapy (VMAT) for canine gliomas, alone (radiotherapy [RT]) and in combination with temozolomide (RT + TMZ), compared with palliation."( Frameless stereotactic radiotherapy alone and combined with temozolomide for presumed canine gliomas.
Bianchi, C; Carrara, N; Dolera, M; Finesso, S; Malfassi, L; Marcarini, S; Mazza, G; Pavesi, S; Sala, M; Urso, G, 2018
)
0.93
" The aim of this retrospective study was to evaluate the efficacy and toxicity of TMZ in dogs with relapsed multicentric lymphoma that failed multi-agent chemotherapy protocols, and compare the outcome to a group of dogs receiving the same drug in combination with DOX."( Temozolomide alone or in combination with doxorubicin as a rescue agent in 37 cases of canine multicentric lymphoma.
Baines, SJ; Blackwood, L; Elliott, JW; Treggiari, E, 2018
)
1.92
" Proliferation, cell cycle, and apoptotic assays were performed with ML00253764, whereas the synergism of the simultaneous combination with temozolomide was evaluated by the combination index method."( Melanocortin Receptor-4 and Glioblastoma Cells: Effects of the Selective Antagonist ML00253764 Alone and in Combination with Temozolomide In Vitro and In Vivo.
Bocci, G; Di Desidero, T; Giuliani, D; Guarini, S; Orlandi, P; Ottani, A; Pacini, S; Pardini, C; Pasqualetti, F; Vaglini, F, 2018
)
0.89
" This open-label, single-arm trial evaluated the maximum tolerated dose (MTD; phase I) and efficacy (phase II) of vorinostat combined with standard chemoradiation in newly diagnosed glioblastoma."( Phase I/II trial of vorinostat combined with temozolomide and radiation therapy for newly diagnosed glioblastoma: results of Alliance N0874/ABTC 02.
Ahluwalia, MS; Anderson, SK; Ballman, KV; Buckner, JC; Cerhan, J; Galanis, E; Gerstner, ER; Giannini, C; Grossman, SA; Jaeckle, K; Lee, EQ; Lesser, GJ; Ligon, KL; Loboda, A; Miller, CR; Moore, DF; Nebozhyn, M; Prados, M; Sarkaria, JN; Schiff, D; Wen, PY, 2018
)
0.74
"Vorinostat combined with standard chemoradiation had acceptable tolerability in newly diagnosed glioblastoma."( Phase I/II trial of vorinostat combined with temozolomide and radiation therapy for newly diagnosed glioblastoma: results of Alliance N0874/ABTC 02.
Ahluwalia, MS; Anderson, SK; Ballman, KV; Buckner, JC; Cerhan, J; Galanis, E; Gerstner, ER; Giannini, C; Grossman, SA; Jaeckle, K; Lee, EQ; Lesser, GJ; Ligon, KL; Loboda, A; Miller, CR; Moore, DF; Nebozhyn, M; Prados, M; Sarkaria, JN; Schiff, D; Wen, PY, 2018
)
0.74
" The aim of our research was the synthesis of a nanocarrier of quercetin combined with temozolomide, to enhance the specificity and efficacy of this anticancer drug commonly used in glioblastoma treatment."( Novel nanohydrogel of hyaluronic acid loaded with quercetin alone and in combination with temozolomide as new therapeutic tool, CD44 targeted based, of glioblastoma multiforme.
Armenia, E; Barbarisi, A; Barbarisi, M; De Sena, G; Iaffaioli, RV; Quagliariello, V; Schiavo, L; Tafuto, S, 2018
)
0.92
" The purpose of this study was to evaluate the safety and pharmacokinetics of depatux-m, either in combination with temozolomide in newly diagnosed or recurrent glioblastoma (arm B) or as monotherapy in recurrent glioblastoma (arm C)."( Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma.
Ansell, PJ; Butowski, N; Fichtel, L; Fischer, J; Gan, HK; Gomez, EJ; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Mandich, H; Merrell, R; Munasinghe, WP; Reardon, DA; Roberts-Rapp, LA; Scott, AM; van den Bent, M; Wheeler, H; Xiong, H, 2018
)
0.9
"Depatux-m alone or in combination with temozolomide demonstrated an acceptable safety and pharmacokinetic profile in glioblastoma."( Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma.
Ansell, PJ; Butowski, N; Fichtel, L; Fischer, J; Gan, HK; Gomez, EJ; Holen, KD; Kumthekar, P; Lassman, AB; Lee, HJ; Lwin, Z; Mandich, H; Merrell, R; Munasinghe, WP; Reardon, DA; Roberts-Rapp, LA; Scott, AM; van den Bent, M; Wheeler, H; Xiong, H, 2018
)
0.96
" In vivo, systemic treatment with pacritinib demonstrated blood-brain barrier penetration and led to improved overall median survival in combination with TMZ, in mice orthotopically xenografted with an aggressive recurrent GBM BTIC culture."( The JAK2/STAT3 inhibitor pacritinib effectively inhibits patient-derived GBM brain tumor initiating cells in vitro and when used in combination with temozolomide increases survival in an orthotopic xenograft model.
Aman, A; Cseh, O; Jensen, KV; Luchman, HA; Weiss, S, 2017
)
0.65
"This preclinical study demonstrates the efficacy of pacritinib and supports the feasibility of testing pacritinib for the treatment of GBM, in combination with the standard of care TMZ."( The JAK2/STAT3 inhibitor pacritinib effectively inhibits patient-derived GBM brain tumor initiating cells in vitro and when used in combination with temozolomide increases survival in an orthotopic xenograft model.
Aman, A; Cseh, O; Jensen, KV; Luchman, HA; Weiss, S, 2017
)
0.65
"We performed C57BL/6 mouse orthotopic glioma model by stereotactic intracranial implantation of glioma cell line GL261, mice were randomly divided into four groups: (1) control group; (2) TMZ group; (3) anti-PD-1 antibody group; (4) TMZ combined with anti-PD-1 antibody group."( Temozolomide combined with PD-1 Antibody therapy for mouse orthotopic glioma model.
Dai, B; Li, J; Qi, N; Zhang, G, 2018
)
1.92
"Anti-PD1 antibody combined with TMZ therapy for orthotopic mouse glioma model could significantly improve the survival time of tumor-bear mice."( Temozolomide combined with PD-1 Antibody therapy for mouse orthotopic glioma model.
Dai, B; Li, J; Qi, N; Zhang, G, 2018
)
1.92
"To investigate the clinical efficacy of stereotactic radiation therapy combined with temozolomide on recurrent glioma."( [Clinical efficacy of stereotactic radiation therapy combined with temozolomide on recurrent brain glioma].
Jiang, C; Li, X; Liu, S; Tang, S; Zhao, H, 2018
)
0.94
"We retrospectively analyzed the safety and efficacy of hypofractionated radiotherapy (45 Gy/15 fr) combined with temozolomide (TMZ) followed by bevacizumab (BEV) salvage treatment in 18 glioblastoma patients aged > 75 years."( Treatment outcomes of hypofractionated radiotherapy combined with temozolomide followed by bevacizumab salvage therapy in glioblastoma patients aged > 75 years.
Kayama, T; Matsuda, KI; Nemoto, K; Sakurada, K; Sonoda, Y, 2018
)
0.93
"Hypofractionated radiotherapy combined with TMZ and BEV salvage treatment was found to be safe and effective in glioblastoma patients aged > 75 years."( Treatment outcomes of hypofractionated radiotherapy combined with temozolomide followed by bevacizumab salvage therapy in glioblastoma patients aged > 75 years.
Kayama, T; Matsuda, KI; Nemoto, K; Sakurada, K; Sonoda, Y, 2018
)
0.72
" In addition, TMZ could increase the levels of miR-505 and combination with pri-miR-505 and TMZ promoted the suppressive role mediated by miR-505 in GBM cells."( Combination with TMZ and miR-505 inhibits the development of glioblastoma by regulating the WNT7B/Wnt/β-catenin signaling pathway.
Fu, C; Liu, X; Yang, X; Zhang, C, 2018
)
0.48
"We performed a retrospective analysis to compare the efficacy of whole brain radiotherapy (WBRT) combined with temozolomide (TMZ) versus WBRT alone as first-line treatment for brain metastases (BM)."( Effectiveness of temozolomide combined with whole brain radiotherapy for non-small cell lung cancer brain metastases.
Fu, L; Guo, D; Jing, W; Kong, L; Yu, J; Zhu, Y, 2018
)
1.03
" To evaluate antitumor activity in vivo, 5-aza was administered alone and in combination with temozolomide (TMZ) in a PDX glioma model harboring IDH1 R132H mutation."( Demethylation and epigenetic modification with 5-azacytidine reduces IDH1 mutant glioma growth in combination with temozolomide.
Borodovsky, A; Chan, T; da Costa Rosa, M; Festuccia, WT; Riggins, GJ; Yamashita, AS, 2019
)
0.94
"5-Aza provided a survival benefit as a single agent but worked best in combination with TMZ in 2 different IDH1 R132H mutant glioma models."( Demethylation and epigenetic modification with 5-azacytidine reduces IDH1 mutant glioma growth in combination with temozolomide.
Borodovsky, A; Chan, T; da Costa Rosa, M; Festuccia, WT; Riggins, GJ; Yamashita, AS, 2019
)
0.72
" Temozolomide (TMZ) is one of them, widely used even in combination with ionizing radiation."( Drug resistance in glioblastoma and cytotoxicity of seaweed compounds, alone and in combination with anticancer drugs: A mini review.
Almeida, T; Azqueta, A; Ferreira, J; Ramos, AA; Rocha, E, 2018
)
1.39
" However, their effects and mechanisms of action, alone or in combination with anticancer drugs, namely TMZ, in glioblastoma cell, still few explored and require more attention due to the unquestionable high potential of these marine compounds."( Drug resistance in glioblastoma and cytotoxicity of seaweed compounds, alone and in combination with anticancer drugs: A mini review.
Almeida, T; Azqueta, A; Ferreira, J; Ramos, AA; Rocha, E, 2018
)
0.48
" Six patients received temozolomide and 8 received dacarbazine + 5-FU, combined with Endostar."( Effect of Endostar combined with chemotherapy in advanced well-differentiated pancreatic neuroendocrine tumors.
Bai, CM; Cheng, YJ; Gao, X; Meng, CT; Yan, XY; Ying, HY; Zhou, JF; Zhou, N, 2018
)
0.79
" Further clinical investigations aimed at elucidating the veliparib exposure-efficacy/safety relationship and optimizing dosing recommendations for maximizing benefit-risk in patients with advanced myeloid malignancies should study veliparib doses ranging up to 120 mg in combination with temozolomide."( Population pharmacokinetics and exposure-response assessment of veliparib co-administered with temozolomide in patients with myeloid leukemias.
Beumer, JH; Chen, A; Gobburu, J; Gojo, I; Gopalakrishnan, M; Greer, JM; Karp, JE; Kiesel, BF; Mehrotra, S; Piekarz, R; Rudek, MA; Singh, R, 2019
)
0.91
" This is the first study to evaluate the in vivo efficacy of the intravenous idasanutlin prodrug, RO6839921 (RG7775), both alone and in combination with temozolomide in TP53 wt orthotopic neuroblastoma models."( Preclinical evaluation of the first intravenous small molecule MDM2 antagonist alone and in combination with temozolomide in neuroblastoma.
Berry, P; Bonner, J; Chen, L; Daga, A; Kirk, C; Lunec, J; Newell, DR; Pastorino, F; Ponzoni, M; Thomas, HD; Tweddle, DA; Veal, GJ; Wood, KM; Zhao, Y, 2019
)
0.92
" This study aimed to investigate the antitumor effects of natural killer cells in combination with temozolomide as the standard chemotherapeutic agent for glioblastoma."( Ex vivo-expanded highly purified natural killer cells in combination with temozolomide induce antitumor effects in human glioblastoma cells in vitro.
Matsuda, R; Morita, H; Motoyama, Y; Murakami, T; Nakagawa, I; Nakamura, M; Nakase, H; Nakazawa, T; Nishimura, F; Omoto, K; Shida, Y; Tanaka, Y; Tsujimura, T, 2019
)
0.96
" The purpose of our study was to explore if PDT combined with TMZ can effectively inhibit glioma cells by influencing NHE1 in vitro."( Photodynamic therapy combined with temozolomide inhibits C6 glioma migration and invasion and promotes mitochondrial-associated apoptosis by inhibiting sodium-hydrogen exchanger isoform 1.
Chen, L; Chi, D; Cong, D; Gao, S; Hu, S; Ji, H; Jia, Y; Jin, J; Liang, B; Zhou, P, 2019
)
0.79
"These results demonstrate that PDT combined with TMZ can inhibit C6 cell migration and invasion and promote mitochondrial-associated apoptosis by inhibiting NHE1."( Photodynamic therapy combined with temozolomide inhibits C6 glioma migration and invasion and promotes mitochondrial-associated apoptosis by inhibiting sodium-hydrogen exchanger isoform 1.
Chen, L; Chi, D; Cong, D; Gao, S; Hu, S; Ji, H; Jia, Y; Jin, J; Liang, B; Zhou, P, 2019
)
0.79
" The aim of this study was to evaluate the efficacy of moderately hypofractionated simultaneous integrated boost intensity-modulated radiotherapy (SIB-IMRT) combined with temozolomide (TMZ) for the postoperative treatment of GBM."( Efficacy of moderately hypofractionated simultaneous integrated boost intensity-modulated radiotherapy combined with temozolomide for the postoperative treatment of glioblastoma multiforme: a single-institution experience.
Chen, G; Chen, L; Li, G; Li, Q; Luo, W; Lv, S; Zhong, L; Zhou, P, 2019
)
0.92
"Moderately hypofractionated SIB-IMRT combined with TMZ is a feasible and safe treatment option with mild toxicity and good PFS and OS."( Efficacy of moderately hypofractionated simultaneous integrated boost intensity-modulated radiotherapy combined with temozolomide for the postoperative treatment of glioblastoma multiforme: a single-institution experience.
Chen, G; Chen, L; Li, G; Li, Q; Luo, W; Lv, S; Zhong, L; Zhou, P, 2019
)
0.72
"A novel core‑shell type thermo‑nanoparticle (CSTNP) co‑loaded with temozolomide (TMZ) and the fluorescein new indocyanine green dye IR820 (termed IT‑CSTNPs) was designed and combined with a near‑infrared (NIR) laser to realize its photothermal conversion."( Core‑shell type thermo‑nanoparticles loaded with temozolomide combined with photothermal therapy in melanoma cells.
Hou, X; Jiang, G; Li, X; Liu, W; Liu, Y; Pang, Y; Yang, C, 2019
)
1
"The purpose of this study was to evaluate the outcomes of elderly patients (aged ≥75 years) with newly diagnosed glioblastoma (GBM), who were treated with hypofractionated radiotherapy comprising 45 Gy in 15 fractions combined with temozolomide (TMZ) or TMZ and bevacizumab (TMZ/Bev)."( Survival benefits of hypofractionated radiotherapy combined with temozolomide or temozolomide plus bevacizumab in elderly patients with glioblastoma aged ≥ 75 years.
Ichimura, K; Igaki, H; Matsushita, Y; Miyakita, Y; Narita, Y; Ohno, M; Takahashi, M, 2019
)
0.94
"Our hypofractionated radiotherapy regimen combined with TMZ or TMZ/Bev showed benefits in terms of OS, PFS, and KPS maintenance with acceptable toxicities in elderly patients with GBM aged ≥75 years."( Survival benefits of hypofractionated radiotherapy combined with temozolomide or temozolomide plus bevacizumab in elderly patients with glioblastoma aged ≥ 75 years.
Ichimura, K; Igaki, H; Matsushita, Y; Miyakita, Y; Narita, Y; Ohno, M; Takahashi, M, 2019
)
0.75
" In clinical practice, Chinese doctors often use radiotherapy combined with temozolomide (TMZ) to treat these patients, although large-scale prospective studies are lacking."( Radiotherapy versus radiotherapy combined with temozolomide in high-risk low-grade gliomas after surgery: study protocol for a randomized controlled clinical trial.
Guan, H; He, L; He, Y; Mu, X; Peng, X; Wang, J; Wang, Y, 2019
)
1
" The efficacy was significantly improved after lobaplatin combined with pemetrexed, temozolomide and bevacizumab."( Chemotherapy combined with bevacizumab for the treatment of advanced lung adenocarcinoma cancer harboring EGFR-ANXA2, EGFR-RAD51, ATR and BRCA2 mutations: A case report.
Cheng, Y; Huang, Z; Li, H; Liu, J; Liu, Y; Zhang, S; Zhong, R, 2020
)
0.78
" Because the free drug cannot pass the blood-brain barrier (BBB), we investigated the use of nanocarriers for transport of the drug through the BBB and its efficacy when combined with radiotherapy and temozolomide (TMZ) in glioma spheroids."( Preclinical evaluation of binimetinib (MEK162) delivered via polymeric nanocarriers in combination with radiation and temozolomide in glioma.
Becerril Aragon, G; Bikhezar, F; de Kruijff, RM; de Vries, HE; Denkova, AG; Gasol Garcia, A; Narayan, RS; Slotman, BJ; Sminia, P; Torrelo Villa, G; van der Meer, AJGM; van der Pol, SMA, 2020
)
0.96
" A synergistic effect was found in combination with fractionated irradiation and an additive effect with TMZ on spheroid volume reduction."( Preclinical evaluation of binimetinib (MEK162) delivered via polymeric nanocarriers in combination with radiation and temozolomide in glioma.
Becerril Aragon, G; Bikhezar, F; de Kruijff, RM; de Vries, HE; Denkova, AG; Gasol Garcia, A; Narayan, RS; Slotman, BJ; Sminia, P; Torrelo Villa, G; van der Meer, AJGM; van der Pol, SMA, 2020
)
0.77
" Methods This is an open-label, 2-arm Phase 1b/2a study (N = 56) of galunisertib (intermittent dosing: 14 days on/14 days off per cycle of 28 days) in combination with TMZ/RTX (n = 40), versus a control arm (TMZ/RTX, n = 16)."( Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma.
Burkholder, T; Capper, D; Cleverly, AL; Desjardins, A; Estrem, ST; Forsyth, P; Guba, SC; Gueorguieva, I; Lahn, MM; Rodon, J; Suarez, C; Wang, S; Wick, A, 2020
)
0.76
" We observe the effects of Si wei xiao xiu yin combined with chemotherapy on the growth of subcutaneous xenografts in nude mice and the expression of miRNA-21 and miRNA-221 in tumor tissues."( New advances on the inhibition of Siwei Xiaoliuyin combined with Temozolomide in glioma based on the regulatory mechanism of miRNA21/221.
Chen, H; Chen, Y; Li, C; Sharma, A; Sharma, HS; Tan, Q; Xie, C; Yang, Y; Zhan, W; Zhang, Z, 2020
)
0.8
" They were randomly divided into saline group, traditional Chinese medicine group, temozolomide group and traditional Chinese medicine combined with temozolomide group to observe the changes in body weight, and the tumor weight, length, short diameter, volume of mice."( New advances on the inhibition of Siwei Xiaoliuyin combined with Temozolomide in glioma based on the regulatory mechanism of miRNA21/221.
Chen, H; Chen, Y; Li, C; Sharma, A; Sharma, HS; Tan, Q; Xie, C; Yang, Y; Zhan, W; Zhang, Z, 2020
)
1.02
"To some extent, Si wei xiao xiu yin combined with temozolomide can inhibit the growth of subcutaneous xenografts in glioma nude mice."( New advances on the inhibition of Siwei Xiaoliuyin combined with Temozolomide in glioma based on the regulatory mechanism of miRNA21/221.
Chen, H; Chen, Y; Li, C; Sharma, A; Sharma, HS; Tan, Q; Xie, C; Yang, Y; Zhan, W; Zhang, Z, 2020
)
1.05
"9% NaCl at a volume of 100 mL administered over a 30-min period every 3 weeks, combined with temozolomide or albumin-bound paclitaxel (nab-paclitaxel) in four patients with ARMM."( Transcatheter arterial infusion of anti-programmed cell death 1 antibody pembrolizumab combined with temozolomide or nab-paclitaxel in patient with primary anorectal malignant melanoma: Four case reports.
Cao, F; Chen, S; Fan, W; Li, D; Ma, W; Qi, H; Shen, L; Song, Z; Wen, X; Wu, Y; Xie, L; Zhang, X, 2020
)
0.99
"This was a phase I, two-stage, multicentre, open-label, dose-escalation study of buparlisib in combination with temozolomide and radiotherapy in patients with newly diagnosed glioblastoma."( Phase I, open-label, multicentre study of buparlisib in combination with temozolomide or with concomitant radiation therapy and temozolomide in patients with newly diagnosed glioblastoma.
Beck, JT; DeGroot, J; Donnet, V; El-Hashimy, M; Mason, W; Mills, D; Rodon, JA; Rosenthal, M; Wen, PY, 2020
)
1
"The MTD of buparlisib in combination with temozolomide at stage I (adjuvant phase only) was 80 mg/day, which was used as the starting dose in stage II."( Phase I, open-label, multicentre study of buparlisib in combination with temozolomide or with concomitant radiation therapy and temozolomide in patients with newly diagnosed glioblastoma.
Beck, JT; DeGroot, J; Donnet, V; El-Hashimy, M; Mason, W; Mills, D; Rodon, JA; Rosenthal, M; Wen, PY, 2020
)
1.05
"Considering that the primary objective of estimating the MTD was not achieved in addition to the observed challenging safety profile of buparlisib in combination with radiotherapy and temozolomide, Novartis decided not to pursue the development of buparlisib in newly diagnosed glioblastoma."( Phase I, open-label, multicentre study of buparlisib in combination with temozolomide or with concomitant radiation therapy and temozolomide in patients with newly diagnosed glioblastoma.
Beck, JT; DeGroot, J; Donnet, V; El-Hashimy, M; Mason, W; Mills, D; Rodon, JA; Rosenthal, M; Wen, PY, 2020
)
0.98
"Temozolomide (TEM) has been reported to be active alone or in combination with capecitabine (CAP) in patients with neuroendocrine neoplasms (NENs)."( Temozolomide alone or in combination with capecitabine in patients with advanced neuroendocrine neoplasms: an Italian multicenter real-world analysis.
Antonuzzo, L; Barberis, M; Campana, D; Faviana, P; Fazio, N; Fumagalli, C; Gelsomino, F; Maisonneuve, P; Marconcini, R; Messerini, L; Puliafito, I; Rossi, G; Spada, F, 2021
)
3.51
"Patients received oral TEM alone or in combination with CAP."( Temozolomide alone or in combination with capecitabine in patients with advanced neuroendocrine neoplasms: an Italian multicenter real-world analysis.
Antonuzzo, L; Barberis, M; Campana, D; Faviana, P; Fazio, N; Fumagalli, C; Gelsomino, F; Maisonneuve, P; Marconcini, R; Messerini, L; Puliafito, I; Rossi, G; Spada, F, 2021
)
2.06
" This study evaluated the efficacy of atorvastatin in combination with standard therapy in patients with glioblastoma."( Atorvastatin in combination with radiotherapy and temozolomide for glioblastoma: a prospective phase II study.
Aldanan, S; Alghareeb, WA; Alhussain, H; AlNajjar, FH; Alsaeed, E; Alsharm, AA; Altwairgi, AK; Balbaid, AAO; Orz, Y, 2021
)
0.87
" Therefore, we analyzed the cytotoxic effect of methadone alone and in combination with temozolomide, a DNA-alkylating drug that is first-line used in GBM treatment, utilizing GBM-derived cell lines and a human fibroblast cell line."( Cytotoxic and Senolytic Effects of Methadone in Combination with Temozolomide in Glioblastoma Cells.
Beltzig, L; Haas, B; Kaina, B; Piee-Staffa, A, 2020
)
1.02
" Current treatments for glioblastoma patients consist of surgery followed by radiation in combination with temozolomide."( The effect of temozolomide in combination with doxorubicin in glioblastoma cells
Alexandru, O; Artene, SA; Danoiu, S; Dricu, A; Elena Cioc, C; Horescu, C; Sevastre, AS; Stefana Oana, P; Tache, DE; Tuta, C, 2020
)
1.13
"To investigate the efficacy and safety of synchronous stereotactic radiotherapy (SRT) with temozolomide (TMZ) combined with whole brain radiotherapy (WBRT) in treating brain metastases originating from non-small cell lung cancer (NSCLC)."( Efficacy of synchronous stereotactic radiotherapy with temozolomide combined with whole brain radiotherapy in treating brain metastases originating from non-small cell lung cancer.
Liu, J; Liu, P; Ren, R; You, D,
)
0.6
"Synchronous SRT with TMZ combined with WBRT is effective in treating patients with brain metastases originating from NSCLC, which can effectively improve the survival of patients and has tolerable adverse reactions."( Efficacy of synchronous stereotactic radiotherapy with temozolomide combined with whole brain radiotherapy in treating brain metastases originating from non-small cell lung cancer.
Liu, J; Liu, P; Ren, R; You, D,
)
0.38
"In this work, we constructed LPHNs-cRGD for targeting delivery of the CRISPR/Cas9 system, in combination with FUS-MBs to open the BBB."( Gene Therapy for Drug-Resistant Glioblastoma via Lipid-Polymer Hybrid Nanoparticles Combined with Focused Ultrasound.
Chen, J; Cheng, Y; Huang, N; Wang, Z; Yang, Q; Zhou, Y, 2021
)
0.62
"Purpose: To investigate the clinical therapeutic effect and safety of thalidomide combined with temozolomide (TMZ) and three-dimensional conformal radiotherapy for patients with high-grade gliomas after operation."( Therapeutic effect of thalidomide combined with temozolomide and three-dimensional conformal radiotherapy for patients with high-grade gliomas after operation.
Li, Y; Shen, X; Sui, L; Xu, Z,
)
0.61
"Methods: The clinical data of 108 patients with high-grade gliomas undergoing operation in our hospital from September 2014 to December 2016 were retrospectively analyzed, of which 54 received thalidomide combined with TMZ and three-dimensional conformal radiotherapy (thalidomide group) and 54 received TMZ combined with three-dimensional conformal radiotherapy (control group)."( Therapeutic effect of thalidomide combined with temozolomide and three-dimensional conformal radiotherapy for patients with high-grade gliomas after operation.
Li, Y; Shen, X; Sui, L; Xu, Z,
)
0.39
"Conclusions: The application of thalidomide combined with TMZ and three-dimensional conformal radiotherapy for high-grade glioma patients after operation can prominently enhance the clinical therapeutic effect, improve patient quality of life, prolong survival, and produce tolerable adverse reactions."( Therapeutic effect of thalidomide combined with temozolomide and three-dimensional conformal radiotherapy for patients with high-grade gliomas after operation.
Li, Y; Shen, X; Sui, L; Xu, Z,
)
0.39
"To explore the efficacy and safety of 125 I radioactive seed implantation combined with intermittent hormonal therapy (IHT) in the clinical treatment of moderate- and high-risk non-metastatic prostate cancer."( Therapeutic effect of thalidomide combined with temozolomide and three-dimensional conformal radiotherapy for patients with high-grade gliomas after operation.
Li, Y; Shen, X; Sui, L; Xu, Z,
)
0.39
"125 I seed implantation combined with IHT is safe and effective in the clinical treatment of patients with moderate- and high-risk non-metastatic prostate cancer."( Therapeutic effect of thalidomide combined with temozolomide and three-dimensional conformal radiotherapy for patients with high-grade gliomas after operation.
Li, Y; Shen, X; Sui, L; Xu, Z,
)
0.39
"To explore the clinical efficacy and safety of bevacizumab combined with temozolomide dose-dense regimen in the treatment of recurrent glioma."( Efficacy of bevacizumab combined with temozolomide dose-dense regimen on recurrent glioma.
Huang, W; Ji, S; Li, J; Liu, Z; Piao, M; Xue, J; Zhu, X,
)
0.63
" There were 51 patients undergoing bevacizumab combined with temozolomide treatment (Bevacizumab group), and the remaining 51 patients received temozolomide treatment alone (Control group)."( Efficacy of bevacizumab combined with temozolomide dose-dense regimen on recurrent glioma.
Huang, W; Ji, S; Li, J; Liu, Z; Piao, M; Xue, J; Zhu, X,
)
0.64
"Bevacizumab combined with temozolomide can significantly improve the clinical efficacy, increase the quality of life of patients, and delay the progression of recurrent glioma, with tolerable adverse reactions."( Efficacy of bevacizumab combined with temozolomide dose-dense regimen on recurrent glioma.
Huang, W; Ji, S; Li, J; Liu, Z; Piao, M; Xue, J; Zhu, X,
)
0.7
"This two-stage phase I trial determined the MTD of zotiraciclib combined with either dose-dense (Arm1) or metronomic (Arm2) temozolomide using a Bayesian Optimal Interval design; then a randomized cohort expansion compared the progression-free survival rate at 4 months (PFS4) of the two arms for an efficient determination of a temozolomide schedule to combine with zotiraciclib at MTD."( Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas.
Aboud, O; Ahmad, S; Antony, R; Armstrong, TS; Boris, L; Bryla, C; Burton, EM; Butler, MK; Calvo, KR; Cordova, C; Figg, WD; Fink, D; Gallin, JI; Garren, N; Gilbert, MR; Gonzales, J; Grajkowska, E; Kuhns, DB; Leeper, H; Lindsley, M; Lollo, N; Long Priel, DA; Mendoza, TR; Mentges, K; Pang, Y; Peer, CJ; Penas-Prado, M; Siegel, C; Sissung, TM; Su, YT; Theeler, BJ; Vera, E; Wu, J; Yu, G; Yuan, Y, 2021
)
1.08
"Zotiraciclib combined with temozolomide is safe in patients with recurrent high-grade astrocytomas."( Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas.
Aboud, O; Ahmad, S; Antony, R; Armstrong, TS; Boris, L; Bryla, C; Burton, EM; Butler, MK; Calvo, KR; Cordova, C; Figg, WD; Fink, D; Gallin, JI; Garren, N; Gilbert, MR; Gonzales, J; Grajkowska, E; Kuhns, DB; Leeper, H; Lindsley, M; Lollo, N; Long Priel, DA; Mendoza, TR; Mentges, K; Pang, Y; Peer, CJ; Penas-Prado, M; Siegel, C; Sissung, TM; Su, YT; Theeler, BJ; Vera, E; Wu, J; Yu, G; Yuan, Y, 2021
)
1.17
"The purpose of this study was to compare the clinical efficacy and safety of temozolomide (TMZ) combined with three-dimensional conformal radiotherapy (3D-CRT) and radiotherapy alone after surgery in patients with high-risk low-grade gliomas (LGGs)."( Temozolomide chemotherapy combined with radiotherapy versus radiotherapy alone after surgery in patients with high-risk low-grade gliomas.
Hou, Y; Huan, Y; Li, Y; Liu, X; Zhang, Y,
)
1.8
" Patients receiving TMZ chemotherapy combined with radiotherapy were considered as combination group (n=55), while those treated with radiotherapy alone were regarded as control group (n=55)."( Temozolomide chemotherapy combined with radiotherapy versus radiotherapy alone after surgery in patients with high-risk low-grade gliomas.
Hou, Y; Huan, Y; Li, Y; Liu, X; Zhang, Y,
)
1.57
" Besides, age <40 years old, complete tumor resection and TMZ chemotherapy combined with radiotherapy after surgery were independent factors affecting the three-year PFS of patients with high-risk LGGs."( Temozolomide chemotherapy combined with radiotherapy versus radiotherapy alone after surgery in patients with high-risk low-grade gliomas.
Hou, Y; Huan, Y; Li, Y; Liu, X; Zhang, Y,
)
1.57
"TMZ chemotherapy combined with radiotherapy after surgery in patients with high-risk LGGs can prominently improve clinical efficacy, prolong PFS, and facilitate tolerance to adverse reactions, but not prolong the OS of patients."( Temozolomide chemotherapy combined with radiotherapy versus radiotherapy alone after surgery in patients with high-risk low-grade gliomas.
Hou, Y; Huan, Y; Li, Y; Liu, X; Zhang, Y,
)
1.57
" Recent reports suggested that TMZ combined with capecitabine (CAPTEM) can be effective for the treatment of aggressive pituitary tumors."( Efficacy of temozolomide combined with capecitabine (CAPTEM) on refractory prolactinomas as assessed using an ex vivo 3D spheroid assay.
Fukuoka, H; Inoshita, N; Ishida, A; Ogawa, W; Shichi, H; Yamada, S, 2022
)
1.1
" The current study evaluated the role and molecular mechanisms of anlotinib in glioblastoma, and the effects of anlotinib in combination with temozolomide (TMZ)."( Anlotinib combined with temozolomide suppresses glioblastoma growth via mediation of JAK2/STAT3 signaling pathway.
Chen, J; Deng, C; Pan, H; Wang, H; Xu, P, 2022
)
1.23
" In our study, we aim to explore the efficacy and safety of temozolomide combined with radiotherapy in the treatment of malignant glioma (MG) and its influence on postoperative complications and survival rate of patients."( Efficacy and Safety of Temozolomide Combined with Radiotherapy in the Treatment of Malignant Glioma.
Li, J; Wei, S, 2022
)
1.27
" Non-ionising electromagnetic fields represent an emerging option given the potential advantages of safety, low toxicity and the possibility to be combined with other therapies."( Selective cell cycle arrest in glioblastoma cell lines by quantum molecular resonance alone or in combination with temozolomide.
Astori, G; Belli, R; Bernardi, M; Bozza, A; Catanzaro, D; Celli, P; Chieregato, K; Menarin, M; Merlo, A; Milani, G; Peroni, D; Pozzato, A; Pozzato, G; Raneri, FA; Ruggeri, M; Volpin, L, 2022
)
0.93
"It was found that radiotherapy combined with temozolomide administration often increased the size of the original lesion or produced a new glioblastoma lesion."( Apatinib combined with temozolomide treatment for pseudoprogression in glioblastoma: A case report.
Cheng, P; Han, Q; Ma, H; Yang, H; Zhao, M; Zhao, Y, 2022
)
1.29
" Based on the genetic testing results, almonertinib combined with anlotinib and temozolomide was administered and obtained 12 months of progression-free survival after the diagnosis of recurrence as the fourth-line treatment."( Almonertinib Combined with Anlotinib and Temozolomide in a Patient with Recurrent Glioblastoma with EGFR L858R Mutation.
Dong, S; Hou, Z; Li, S; Luo, N; Tao, R; Wu, H; Zhang, H; Zhang, X; Zhu, D, 2023
)
1.4
" In this paper, we present the effects of juglone alone and in combination with temozolomide on glioblastoma cells."( Juglone in Combination with Temozolomide Shows a Promising Epigenetic Therapeutic Effect on the Glioblastoma Cell Line.
Barciszewska, AM; Belter, A; Gawrońska, I; Giel-Pietraszuk, M; Naskręt-Barciszewska, MZ, 2023
)
1.43
" CCK-8 was used to determine the inhibitory rate of Coix group, TMZ group and drug combination group on GBM cells, and the combination index (CI) was calculated to determine whether they had synergistic effect."( Coixendide efficacy in combination with temozolomide in glioblastoma and transcriptome analysis of the mechanism.
Ban, X; Jin, P; Li, Y; Liu, S; Yue, Y; Zhang, L; Zhang, X; Zhao, C; Zhao, Z, 2023
)
1.18
"To investigate the effect of Temozolomide combined with intensity modulated radiation therapy on serum factor, immune function and clinical efficacy in postoperative glioma patients."( Effect of Temozolomide Combined with Intensity Modulated Radiation Therapy on Serum Factor, Immune Function and Clinical Efficacy in Postoperative Glioma Patients.
Fan, R; Liu, J; Liu, Z; Yuan, J, 2023
)
1.6

Bioavailability

Temozolomide (TMZ) has become the more commonly used analog of DTIC-related oral agents because of its greater bioavailability and ability to cross the blood brain barrier. 10b is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model.

ExcerptReferenceRelevance
" This study evaluated the effect of an increase in gastric pH, through the use of ranitidine, on the oral bioavailability and plasma pharmacokinetics of temozolomide and MTIC."( Effect of gastric pH on the relative oral bioavailability and pharmacokinetics of temozolomide.
Beale, P; Brada, M; Cutler, DL; Judson, I; Marco, A; Moore, S; Reidenberg, P; Statkevich, P, 1999
)
0.73
"Temozolomide, an oral cytotoxic agent with approximately 100% bioavailability after one administration, has demonstrated schedule-dependent clinical activity against highly resistant cancers."( Phase I dose-escalation and pharmacokinetic study of temozolomide (SCH 52365) for refractory or relapsing malignancies.
Batra, V; Beale, P; Brada, M; Cutler, D; Dugan, M; Judson, I; Moore, S; Reidenberg, P; Statkevich, P, 1999
)
2
"Temozolomide (TMZ) is a new, orally administered, second-generation imidazotetrazine prodrug with essentially 100% oral bioavailability that has demonstrated meaningful efficacy and an acceptable safety profile in the treatment of patients with recurrent glioblastoma multiforme."( Future directions in the treatment of malignant gliomas with temozolomide.
Prados, MD, 2000
)
1.99
" Predictable bioavailability and minimal toxicity make temozolomide a candidate for a wide range of clinical testing to evaluate the potential of combination treatments in different tumor types."( Temozolomide and treatment of malignant glioma.
Calvert, H; Friedman, HS; Kerby, T, 2000
)
2
" TMZ is an alkylating agent chemically similar to dacarbazine (DTIC) with good oral bioavailability and CNS penetration."( Temozolomide and whole brain irradiation in melanoma metastatic to the brain: a phase II trial of the Cytokine Working Group.
Atkins, B; Clark, I; Dutcher, P; Ernstoff, S; Flaherty, L; Gollob, J; II Smith, W; Johnson, D; Longmate, J; Margolin, K; Sosman, J; Thompson, A; Weber, J; Weiss, G, 2002
)
1.76
" We treated 25 stage IV patients with temozolomide - a cytostatic drug with 100% oral bioavailability and considerable penetration of CNS tissue."( [Temozolomide as therapeutic option for patients with metastatic melanoma and poor prognosis].
Christophers, E; Frick, S; Haacke, TC; Hauschild, A; Lischner, S; Rosien, F; Schäfer, F, 2002
)
1.5
" Temozolomide, an orally bioavailable alkylating agent that crosses the blood-brain barrier, has activity against brain metastases from both small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) when used as a single agent, but response rates are low."( Use of temozolomide with other cytotoxic chemotherapy in the treatment of patients with recurrent brain metastases from lung cancer.
Ebert, BL; Niemierko, E; Salgia, R; Shaffer, K, 2003
)
1.68
" In this work we investigated the effect of association of temozolomide (TMZ), an orally bioavailable alkylating agent, with three chemotherapeutic drugs, liposomal doxorubicin (DOXO), cis-platinum (CDDP)."( Effect of association of temozolomide with other chemotherapic agents on cell growth inhibition in glioma cell lines.
Balzarotti, M; Boiardi, A; Calatozzolo, C; Ciusani, E; Croci, D; Salmaggi, A, 2004
)
0.87
" Pharmacokinetics studies revealed that GPI 15427 possesses a substantial oral bioavailability (plasma Cmax after a single dose of 40 mg/kg: 1041+/-516 ng/ml)."( Brain distribution and efficacy as chemosensitizer of an oral formulation of PARP-1 inhibitor GPI 15427 in experimental models of CNS tumors.
Alemu, C; Calvin, D; Graziani, G; Hoover, R; Lapidus, R; Leonetti, C; Morgan, L; Scarsella, M; Tang, Z; Tentori, L; Vergati, M; Woznizk, K; Xu, W; Zhang, J, 2005
)
0.33
" TMZ is able to cross the blood brain barrier and is stable at gastric acid pH so it has almost 100% oral bioavailability and is rapidly absorbed after it is taken orally."( Role of temozolomide in pediatric brain tumors.
Barone, G; Maurizi, P; Riccardi, R; Tamburrini, G, 2006
)
0.77
" 10b is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide (TMZ) and in an MX-1 breast xenograph model in combination with cisplatin."( Discovery and SAR of 2-(1-propylpiperidin-4-yl)-1H-benzimidazole-4-carboxamide: A potent inhibitor of poly(ADP-ribose) polymerase (PARP) for the treatment of cancer.
Bontcheva-Diaz, V; Bouska, JJ; Donawho, CK; Frost, DJ; Fry, EH; Gandhi, VB; Giranda, VL; Gong, J; Grandel, R; Johnson, EF; Klinghofer, V; Liu, X; Lubisch, W; Luo, Y; Marsh, KC; Olson, AM; Park, CH; Penning, TD; Rosenberg, SH; Shi, Y; Thomas, S; Wernet, W; Zhu, GD, 2008
)
0.54
"ABT-888 is a potent, orally bioavailable PARP-1/2 inhibitor shown to potentiate DNA damaging agents."( The PARP inhibitor, ABT-888 potentiates temozolomide: correlation with drug levels and reduction in PARP activity in vivo.
Bontcheva-Diaz, VD; Bouska, JJ; Bukofzer, G; Colon-Lopez, M; Donawho, CK; Frost, DJ; Giranda, VL; Guan, R; Jarvis, K; Johnson, EF; Klinghofer, V; Liu, X; Luo, Y; Olson, A; Palma, JP; Penning, TD; Rodriguez, LE; Rosenberg, SH; Saltarelli, MJ; Shi, Y; Stavropoulos, JA; Zhu, GD,
)
0.4
" Although the bioavailability of temozolomide is approximately 100%, pathology or anatomical changes of the gastrointestinal tract may adversely affect absorption, and consequently therapeutic response."( Disposition of temozolomide in a patient with glioblastoma multiforme after gastric bypass surgery.
Beumer, JH; Egorin, MJ; Park, DM; Shah, DD, 2009
)
0.99
" In addition, 3a is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide (TMZ) and in an MX-1 breast cancer xenograft model in combination with either carboplatin or cyclophosphamide."( Discovery of the Poly(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide (ABT-888) for the treatment of cancer.
Bontcheva-Diaz, V; Bouska, JJ; Donawho, CK; Frost, DJ; Gandhi, VB; Giranda, VL; Gong, J; Johnson, EF; Klinghofer, V; Liu, X; Luo, Y; Marsh, KC; Olson, AM; Osterling, DJ; Penning, TD; Shi, Y; Zhu, GD, 2009
)
0.54
" Thus, our results show that polymeric nanocapsules are able to increase the intratumoral bioavailability of indomethacin and reduce the growth of implanted gliomas."( Indomethacin-loaded nanocapsules treatment reduces in vivo glioblastoma growth in a rat glioma model.
Battastini, AM; Bernardi, A; Braganhol, E; Edelweiss, MI; Figueiró, F; Guterres, SS; Jäger, E; Pohlmann, AR, 2009
)
0.35
" Temozolomide's characteristics which make it a candidate for a wide range of clinical testing to evaluate the potential of combination treatments in different tumor types are its predictable bioavailability and minimal toxicity."( Temozolomide with radiation therapy in high grade brain gliomas: pharmaceuticals considerations and efficacy; a review article.
Beli, I; Chaldeopoulos, D; Fotineas, A; Koukourakis, GV; Kouloulias, V; Kouvaris, J; Maravelis, G; Pantelakos, P; Papadimitriou, C; Zacharias, G, 2009
)
2.71
" In addition, 22b is orally bioavailable across multiple species, crosses the blood-brain barrier, and appears to distribute into tumor tissue."( Optimization of phenyl-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase inhibitors: identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (A-966492), a highly potent and efficacious inhibitor.
Bontcheva-Diaz, V; Bouska, JJ; Buchanan, FG; Bukofzer, GT; Donawho, CK; Frost, DJ; Fry, EH; Gandhi, VB; Giranda, VL; Gong, J; Johnson, EF; Klinghofer, V; Liu, X; Luo, Y; Marsh, KC; Olson, AM; Osterling, DJ; Park, CH; Penning, TD; Rodriguez, LE; Shi, Y; Thomas, S; Zhu, GD, 2010
)
0.36
" Recently, the clinical use of temozolomide (TMZ) has become the more commonly used analog of DTIC-related oral agents because of its greater bioavailability and ability to cross the blood brain barrier."( Quercetin abrogates chemoresistance in melanoma cells by modulating deltaNp73.
Burd, R; Limesand, KH; Mendoza, EE; Mitchell, GC; Radhakrishnan, VM; Sittadjody, S; Thangasamy, T, 2010
)
0.65
" Further in vivo experiments on laboratory animals and analysis of absorption rate and side effects are required."( Arsenic trioxide sensitizes cancer stem cells to chemoradiotherapy. A new approach in the treatment of inoperable glioblastoma multiforme.
Cernea, D; Cocis, A; Fischer-Fodor, E; Florian, IS; Ioani, H; Irimie, A; Kacso, G; Petrescu, M; Soritau, O; Timis, T; Tomuleasa, C; Virag, P,
)
0.13
" We evaluated the preclinical potential of a novel, orally bioavailable PI3K/mTOR dual inhibitor (XL765) in in vitro and in vivo studies."( Inhibition of PI3K/mTOR pathways in glioblastoma and implications for combination therapy with temozolomide.
Aftab, DT; Berger, MS; Haas-Kogan, DA; James, CD; Mueller, S; Ozawa, T; Polley, MY; Prados, MD; Prasad, G; Sottero, T; Weiss, WA; Yang, X, 2011
)
0.59
" Future studies are needed to determine whether VPA increases TMZ bioavailability or acts as an inhibitor of histone deacetylases and thereby sensitizes for radiochemotherapy in vivo."( Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma.
Belanger, K; Bogdahn, U; Brandes, AA; Cairncross, JG; Forsyth, P; Gorlia, T; Lacombe, D; Macdonald, DR; Mason, W; Mirimanoff, RO; Rossetti, AO; Stupp, R; van den Bent, MJ; Vecht, CJ; Weller, M, 2011
)
0.61
" This preclinical study sought to test the efficacy of the food additive Triacetin (glyceryl triacetate, GTA) as a novel therapy to increase acetate bioavailability in glioma cells."( Triacetin-based acetate supplementation as a chemotherapeutic adjuvant therapy in glioma.
Davies, MT; Driscoll, HE; Jaworski, DM; Lawler, SE; Long, PM; Penar, PL; Pendlebury, WW; Spees, JL; Teasdale, BA; Tsen, AR; Viapiano, MS, 2014
)
0.4
"CNDAC (2'-C-cyano-2'-deoxy-1-β-d-arabino-pentofuranosyl-cytosine, DFP10917) and its orally bioavailable prodrug, sapacitabine, are undergoing clinical trials for hematologic malignancies and solid tumors."( Mechanism-Based Drug Combinations with the DNA Strand-Breaking Nucleoside Analog CNDAC.
Hargis, S; Jiang, Y; Liu, X; Nowak, B; Plunkett, W, 2016
)
0.43
" Its high bioavailability (40%~100%) and high tissue distribution in both monkeys and rats were its most important pharmacokinetic features."( Novel PARP1/2 inhibitor mefuparib hydrochloride elicits potent in vitro and in vivo anticancer activity, characteristic of high tissue distribution.
Chen, CH; Chen, XY; Chen, Y; Ding, J; Gao, ZW; He, JX; He, Q; Huan, XJ; Li, XH; Liao, XM; Lu, XL; Miao, ZH; Shen, YY; Song, SS; Su, Y; Sun, YM; Tan, C; Tong, LJ; Wang, M; Wang, YQ; Wang, YT; Xiong, B; Yang, CH; Yang, XY, 2017
)
0.46
"Veliparib is a Biopharmaceutical Classification System (BCS) Class 1 compound, with no less than 90% of the dose absorbed and an oral bioavailability of at least 73%."( Clinical Pharmacokinetics and Mass Balance of Veliparib in Combination with Temozolomide in Subjects with Nonhematologic Malignancies.
Giranda, V; Munasinghe, W; Nuthalapati, S; Xiong, H, 2018
)
0.71
" Doxorubicin (DOX) is not used in gliomas due to its low bioavailability in the brain; however, new delivery strategies and low doses may be effective in the long term, especially as part of a drug cocktail."( Low Dose of Doxorubicin Potentiates the Effect of Temozolomide in Glioblastoma Cells.
Kipper, FC; Lenz, G; Lopez, PLDC; Silva, AO; Villodre, ES, 2018
)
0.73
" Delivery outcomes are evaluated by the bioavailability of free temozolomide across time."( Delivery of liposome encapsulated temozolomide to brain tumour: Understanding the drug transport for optimisation.
Zhan, W, 2019
)
1.03
" However, its pharmacological activity is reduced due MTIC low bioavailability in the brain."( Biophysical interaction of temozolomide and its active metabolite with biomembrane models: The relevance of drug-membrane interaction for Glioblastoma Multiforme therapy.
Andrade, S; Coelho, MÁN; Loureiro, JA; Pereira, MC; Ramalho, MJ, 2019
)
0.81
"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
" We reported that copper, for the first time, is found to recognize and bind MTIC in the process of TMZ degradation, which means copper can play an important role in enhancing the bioavailability of MTIC derived from TMZ."( Enhanced Copper-Temozolomide Interactions by Protein for Chemotherapy against Glioblastoma Multiforme.
Du, K; Feng, F; Li, X; Shao, F; Sun, J; Sun, Y, 2019
)
0.86
"Temozolomide is a drug approved for treating glioblastomas, which has 100% oral bioavailability but gets degraded at physiological pH thus having very short half-life and only 20-30% brain bioavailability."( Design of temozolomide-loaded proliposomes and lipid crystal nanoparticles with industrial feasible approaches: comparative assessment of drug loading, entrapment efficiency, and stability at plasma pH.
Dubey, SK; Gorantla, S; Khosa, A; Rapalli, VK; Saha, RN; Singhvi, G; Waghule, T, 2021
)
2.47
" However, low bioavailability and extractive yield limit the clinical applications of XN."( Xanthohumol regulates miR-4749-5p-inhibited RFC2 signaling in enhancing temozolomide cytotoxicity to glioblastoma.
Chen, KC; Chen, PH; Cheng, CH; Ho, KH; Kuo, TC; Lee, CC; Lee, YT; Liu, AJ; Shih, CM, 2020
)
0.79
" The main advantages are its high oral bioavailability (almost 100% although the concentration found in the cerebrospinal fluid was approximately 20% of the plasma concentration of TMZ), its lipophilic properties, and small size that confer the ability to cross the blood-brain barrier."( Temozolomide: An Updated Overview of Resistance Mechanisms, Nanotechnology Advances and Clinical Applications.
Cabeza, L; Jiménez-Luna, C; Luque, R; Melguizo, C; Ortiz, R; Perazzoli, G; Prados, J, 2021
)
2.06
"Liposomal formulations may improve the solubility and bioavailability of drugs potentially increasing their ability to cross the blood-brain barrier."( Nanoliposomal Irinotecan and Metronomic Temozolomide for Patients With Recurrent Glioblastoma: BrUOG329, A Phase I Brown University Oncology Research Group Trial.
Baekey, J; Carcieri, A; Cielo, D; Disano, D; Donnelly, J; Elinzano, H; MacKinnon, K; Mohler, A; Robison, J; Safran, H; Sturtevant, A; Toms, S; Vatketich, J; Wood, R, 2021
)
0.89
" The efficacy and optimal deployment schedule of the orally bioavailable small-molecule tumor checkpoint controller lisavanbulin alone, and in combination with, standards of care were assessed using a panel of IDH-wildtype GBM patient-derived xenografts."( Preclinical modeling in glioblastoma patient-derived xenograft (GBM PDX) xenografts to guide clinical development of lisavanbulin-a novel tumor checkpoint controller targeting microtubules.
Bachmann, F; Bakken, KK; Burgenske, DM; Carlson, BL; Decker, PA; Eckel-Passow, JE; Elmquist, WF; Gampa, G; Giannini, C; He, L; Hu, Z; Kitange, GJ; Kosel, ML; Lane, HA; McSheehy, P; Mladek, AC; Pokorny, JL; Sarkaria, JN; Schmitt-Hoffmann, A; Schroeder, MA; Talele, S; Vaubel, RA, 2022
)
0.72
" Therefore, localised approaches that treat GB straight into the tumour site provide an alternative to enhance chemotherapy bioavailability and efficacy, reducing systemic toxicity."( Nek1-inhibitor and temozolomide-loaded microfibers as a co-therapy strategy for glioblastoma treatment.
Arantes, PR; Borges, GR; Braganhol, E; Dalanhol, CS; de Barros Dias, MCH; de Oliveira Merib, J; de Souza, PO; Ferro, MB; Henn, JG; Morás, AM; Moura, DJ; Nugent, M; Reinhardt, LS, 2022
)
1.05
" Although these beneficial effects are promising, the efficacy of natural compounds in glioblastoma is limited due to their bioavailability and blood-brain barrier permeability."( Natural Compounds as Promising Adjuvant Agents in The Treatment of Gliomas.
Gigli, G; Leporatti, S; Persano, F, 2022
)
0.72
" TMZ is an orally bioavailable prodrug, which is well absorbed from the gastrointestinal tract and is converted to its active alkylating metabolite 5-(3-methyl triazen-1-yl)imidazole-4-carbozamide (MTIC) spontaneously in physiological condition that does not require hepatic involvement."( Temozolomide Efficacy and Metabolism: The Implicit Relevance of Nanoscale Delivery Systems.
Alyautdin, R; Chubarev, V; Grigorevskikh, E; Ismail, N; Merkulov, V; Petrenko, D; Smolyarchuk, E; Sologova, S; Syzrantsev, N, 2022
)
2.16
" Although TMZ has shown 100% oral bioavailability and crosses the blood-brain barrier effectively, however it suffers from limitations such as a short half-life (∼1."( Polymeric and small molecule-conjugates of temozolomide as improved therapeutic agents for glioblastoma multiforme.
Chitkara, D; Jatyan, R; Karthik, YG; Mittal, A; Sahel, DK; Singh, P, 2022
)
0.98
" However, poor site-specific delivery and bioavailability significantly restrict the efficient permeability of regorafenib to brain lesions and compromise its treatment efficacy."( Brain-Targeted HFn-Cu-REGO Nanoplatform for Site-Specific Delivery and Manipulation of Autophagy and Cuproptosis in Glioblastoma.
Ding, N; Huang, C; Jia, W; Jiang, J; Li, L; Luo, M; Nice, EC; Tian, H; Zhang, H; Zhou, L, 2023
)
0.91
"Brain bioavailability of the KDM1A inhibitor (NCD38) was established using LS-MS/MS."( Lysine-specific histone demethylase 1A (KDM1A/LSD1) inhibition attenuates DNA double-strand break repair and augments the efficacy of temozolomide in glioblastoma.
Alejo, S; Brenner, AJ; Chen, Y; Clarke, K; Gilbert, AR; He, Y; Jayamohan, S; Johnson, JD; Lai, Z; Li, W; Lv, Y; Palacios, BE; Pratap, UP; Sareddy, GR; Suzuki, T; Tekmal, RR; Vadlamudi, RK; Venkata, PP; Viswanadhapalli, S; Weldon, K; Ye, Z; Zhao, W; Zheng, S; Zou, Y, 2023
)
1.11
" However, due to the short plasma half-life, only 20-30 % brain bioavailability can be achieved using traditional formulations."( Exploring temozolomide encapsulated PEGylated liposomes and lyotropic liquid crystals for effective treatment of glioblastoma: in-vitro, cell line, and pharmacokinetic studies.
Laxmi Swetha, K; Narayan Saha, R; Roy, A; Singhvi, G; Waghule, T, 2023
)
1.31

Dosage Studied

Temozolomide has been studied in soft-tissue sarcomas with varying dosing schedules. Pretreatment of mice with O6-benzylguanine increased temozolmide-induced mortality, requiring reduction of the dosage from 1200 to 750 mg/m2 on the single-day regimen.

ExcerptRelevanceReference
" Pretreatment of mice with O6-benzylguanine increased temozolomide-induced mortality, requiring reduction of the dosage from 1200 to 750 mg/m2 on the single-day regimen."( Activity of temozolomide in the treatment of central nervous system tumor xenografts.
Bigner, DD; Catino, JJ; Dolan, ME; Friedman, HS; Keir, S; Marcelli, S; Pegg, AE; Schold, SC, 1995
)
0.92
" Comparisons with BCNU were made on both single and multiple dosing schedules, since temozolomide cytotoxicity is highly schedule dependent."( Potentiation of temozolomide and BCNU cytotoxicity by O(6)-benzylguanine: a comparative study in vitro.
May, BL; Newlands, ES; Porteus, JK; Wedge, SR, 1996
)
0.86
") was examined using single and daily x5 dosing regimens in athymic mice bearing subcutaneous A375P xenografts."( Effect of single and multiple administration of an O6-benzylguanine/temozolomide combination: an evaluation in a human melanoma xenograft model.
Newlands, ES; Porteous, JK; Wedge, SR, 1997
)
0.53
"Thirty-three patients with newly diagnosed glioblastoma multiforme (GBM) and five patients with newly diagnosed anaplastic astrocytoma (AA) were treated with Temodal at a starting dose of 200 mg/m2 daily for 5 consecutive days with repeat dosing every 28 days after the first daily dose."( DNA mismatch repair and O6-alkylguanine-DNA alkyltransferase analysis and response to Temodal in newly diagnosed malignant glioma.
Ashley, DM; Bigner, DD; Bigner, SH; Cokgor, I; Colvin, OM; Dugan, M; Friedman, AH; Friedman, HS; Haglund, MM; Henry, AJ; Kerby, T; Krischer, J; Lovell, S; Marchev, F; McLendon, RE; Modrich, PL; Provenzale, JM; Rasheed, K; Rich, J; Seman, AJ; Stewart, E, 1998
)
0.3
" This review discusses the mechanism of action of TMZ and strategies for overcoming pathways of resistance to this promising agent, including the use of TMZ in combination with other chemotherapeutic agents or radiation therapy, and exploration of alternate dosing schedules."( Future directions in the treatment of malignant gliomas with temozolomide.
Prados, MD, 2000
)
0.55
" One day after the last dose of vehicle or TNP-470, a steady-state dosing regimen of TMZ was administered with subsequent collection and high-performance liquid chromatography analysis of plasma and either tumor homogenate or tumor microdialysis steady-state TMZ concentrations, and in some cases [5-(3-methyltriazen-1-yl)imidazole-4-carboximide] MTIC, its active metabolite."( Pharmacodynamic-mediated reduction of temozolomide tumor concentrations by the angiogenesis inhibitor TNP-470.
Chu, J; Gallo, JM; Li, S; Ma, J; Pulfer, S; Reed, K, 2001
)
0.58
" In this study, we evaluated the relation between TMZ dosing and AGT depletion in patients with deep visceral tumors and in peripheral blood mononuclear cells (PBMCs) to determine whether the dose of TMZ was sufficient to inactivate AGT and lead to therapeutic efficacy."( Temozolomide: the effect of once- and twice-a-day dosing on tumor tissue levels of the DNA repair protein O(6)-alkylguanine-DNA-alkyltransferase.
Gerson, SL; Haaga, J; Liu, L; Majka, S; Spiro, TP; Willson, JK, 2001
)
1.75
" For patients with recurrent malignant glioma, temozolomide provides a therapeutic option with a predictable safety profile, clinical efficacy, and convenient dosing that can provide important quality-of-life benefits."( Temozolomide for recurrent high-grade glioma.
Macdonald, DR, 2001
)
2.01
" Also under investigation are modifications to the temozolomide dosing schedule, other routes of administration, and treatment regimens that include temozolomide in combination with other chemotherapeutic and biologic agents."( Future directions for temozolomide therapy.
Yung, WK, 2001
)
0.88
" The recommended dosage for TEMO for a phase II study of this combination is 200 mg/m2 per day for 5 days."( Phase I study of Gliadel wafers plus temozolomide in adults with recurrent supratentorial high-grade gliomas.
Affronti, ML; Cokgor, L; Early, M; Edwards, S; Friedman, AH; Friedman, HS; Gururangan, S; Herndon, JE; McLendon, RE; Provenzale, JM; Quinn, JA; Rich, JN; Sampson, JH; Stafford-Fox, V; Tourt-Uhlig, S; Zaknoen, S, 2001
)
0.58
" The standard dosage is 150-200 mg/m2 per day for 5 days in a 28-day cycle."( A phase II study of extended low-dose temozolomide in recurrent malignant gliomas.
Abrey, LE; Bazylewicz, KA; Khan, RB; Malkin, MG; Raizer, JJ, 2002
)
0.59
"To evaluate the antitumor effects and toxicities of whole brain irradiation (WBI) with temozolomide (TMZ) administered by prolonged oral dosing in patients with melanoma metastatic to the brain."( Temozolomide and whole brain irradiation in melanoma metastatic to the brain: a phase II trial of the Cytokine Working Group.
Atkins, B; Clark, I; Dutcher, P; Ernstoff, S; Flaherty, L; Gollob, J; II Smith, W; Johnson, D; Longmate, J; Margolin, K; Sosman, J; Thompson, A; Weber, J; Weiss, G, 2002
)
1.98
" This review will focus on the different pharmacological strategies aimed at overcoming tumor resistance to TMZ such as new formulations of the drug or dosing schedules, and combined treatments with other chemotherapeutic agents, modulators of DNA repair systems, or gene therapy."( Pharmacological strategies to increase the antitumor activity of methylating agents.
Graziani, G; Tentori, L, 2002
)
0.31
" The drug is well tolerated with dose limiting myelosuppression and thrombocytopenia occurring in less than 10% of patients at current dosage schedules."( The use of temozolomide in recurrent malignant gliomas.
Gaya, A; Greenstein, A; Rees, J; Stebbing, J, 2002
)
0.7
" Body surface area-based dosing was statistically significantly associated with a reduction in interpatient variability in drug clearance for only five of the 33 agents: docosahexaenoic acid (DHA)-paclitaxel, 5-fluorouracil/eniluracil, paclitaxel, temozolomide, and troxacitabine."( Role of body surface area in dosing of investigational anticancer agents in adults, 1991-2001.
Baker, SD; Donehower, RC; Grochow, LB; Rowinsky, EK; Schellens, JH; Sparreboom, A; Verweij, J, 2002
)
0.5
" For example, Abdelbasit and Plackett proposed an optimal design assuming that the dose-response relationship follows some specified linear models."( Experimental design and sample size determination for testing synergism in drug combination studies based on uniform measures.
Fang, HB; Houghton, PJ; Tan, M; Tian, GL, 2003
)
0.32
" The model is fit to serial ANC measurements obtained after TMZ dosing and it is able to explain, among other things, the lag in ANC reduction following a dose of TMZ, the ANC nadir, and the 'rebound effect' observed where the ANC recovers to levels greater than that observed pre-TMZ dose."( A mechanistic mathematical model of temozolomide myelosuppression in children with high-grade gliomas.
Fouladi, M; Gajjar, AJ; Kirstein, MN; Nair, G; Panetta, JC; Stewart, CF, 2003
)
0.59
" In the last 2 years, studies have focused on exploring strategies to optimize the efficacy of temozolomide, including evaluating different temozolomide dosing schedules and combining temozolomide with other antineoplastic agents, radiation therapy, or drug resistance-modifying agents."( Temozolomide: realizing the promise and potential.
Dolan, ME; Nagasubramanian, R, 2003
)
1.98
" Temozolomide has activity and a favorable safety profile in all dosing schedules tested."( Temozolomide: realizing the promise and potential.
Dolan, ME; Nagasubramanian, R, 2003
)
2.67
" Extended TMZ dosing regimens may be superior by delivering the drug continuously at a higher dose over time."( Selective CD4+ lymphopenia in melanoma patients treated with temozolomide: a toxicity with therapeutic implications.
Chapman, PB; Foster, T; Krown, SE; Livingston, PO; Quinn, C; Sepkowitz, KA; Sohn, S; Su, YB; Williams, L; Wolchok, JD, 2004
)
0.56
" Alternate temozolomide dosing schedules such as continuous daily administration may enhance antitumor activity through sustained depletion of the DNA repair protein O6-alkylguanine DNA alkyltransferase."( Temozolomide in patients with advanced cancer: phase I and pharmacokinetic study.
Baker, SD; Batra, VK; Cutler, DL; Donehower, RC; Rudek, MA; Statkevich, P, 2004
)
2.16
" In phase II of the study, four weeks after completion of RT, a monochemotherapy using TMZ was administered at the dosage of 200 mg/m2/day per 5 days every 28 days for 6 cycles."( Temozolomide in radio-chemotherapy combined treatment for newly-diagnosed glioblastoma multiforme: phase II clinical trial.
Campanella, C; Costa, A; Fedele, F; Frati, A; Frati, L; Gagliardi, FM; Innocenzi, G; Lanzetta, G; Minniti, G; Nappa, M; Rozzi, A; Salvati, M; Vecchione, A,
)
1.57
" Median thalidomide dosage was 200 mg/day."( Combined thalidomide and temozolomide treatment in patients with glioblastoma multiforme.
Baumann, F; Baumert, BG; Bernays, RL; Bjeljac, M; Brandner, S; Kollias, SS; Rousson, V; Yonekawa, Y,
)
0.43
"The response rate, efficacy, side-effects, reasons for discontinuation of therapy and survival rate of 47 patients treated with temozolomide in combination with two different dosing regimens of IFN-alpha 2b were documented."( Temozolomide and interferon alpha 2b in metastatic melanoma stage IV.
Fialla, R; Forstinger, Ch; Fritsch, P; Hofmann-Wellenhof, R; Kehrer, H; Kerl, H; Kindermann-Glebowski, E; Klein, G; Koller, J; Konrad, K; Kos, C; Lang, A; Mischer, P; Pachinger, W; Pehamberger, H; Raml, J; Ratzinger, G; Richtig, E; Seeber, A; Smolle, J; Steiner, A; Ulmer, H; Wolff, K; Zelger, B, 2004
)
1.97
"We conducted a study to determine the dose-limiting toxicity of an extended dosing schedule of temozolomide (TMZ) when used with a fixed dose of BCNU, or 1,3-bis(2-chloroethyl)-1-nitrosourea (carmustine), taking advantage of TMZ's ability to deplete O6-alkylguanine-DNA-alkyltransferase and the synergistic activity of these two agents."( Phase 1 study of 28-day, low-dose temozolomide and BCNU in the treatment of malignant gliomas after radiation therapy.
Abrey, LE; Kleber, M; Malkin, MG; Raizer, JJ, 2004
)
0.82
" We develop a maximum likelihood method based on the expectation/conditional maximization (ECM) algorithm to estimate the dose-response relationship while accounting for the informative censoring and the constraints of model parameters."( Repeated-measures models with constrained parameters for incomplete data in tumour xenograft experiments.
Fang, HB; Houghton, PJ; Tan, M; Tian, GL, 2005
)
0.33
" Attempts to maximise efficacy have led to manipulation of both dosage and drug scheduling and the evidence for the various strategies is reviewed."( Temozolomide in the treatment of solid tumours: current results and rationale for dosing/scheduling.
Middleton, MR; Payne, MJ; Pratap, SE, 2005
)
1.77
"Temozolomide has been studied in soft-tissue sarcomas with varying dosing schedules."( Temozolomide in uterine leiomyosarcomas.
Aghajanian, C; Anderson, S, 2005
)
3.21
" Cisplatin-temozolomide combinations are well tolerated without additional toxicity to single-agent treatments; the recommended phase II dosage is 80 mg m(-2) cisplatin and 150 mg m(-2) x 5 temozolomide in heavily treated, and 200 mg m(-2) x 5 temozolomide in less-heavily pretreated children."( Dose finding and O6-alkylguanine-DNA alkyltransferase study of cisplatin combined with temozolomide in paediatric solid malignancies.
Chastagner, P; Couanet, D; Djafari, L; Doz, F; Frappaz, D; Gentet, JC; Geoerger, B; Geoffray, A; Margison, GP; O'Quigley, J; Pein, F; Raquin, MA; Rubie, H; Vassal, G; Wartelle, M; Watson, AJ, 2005
)
0.94
"The objective of the study was to evaluate the efficacy and toxicity of Temozolomide (TMZ) administered for 5 consecutive days in three daily dosing in children with recurrent or refractory high-grade glioma."( Phase II trial of temozolomide in children with recurrent high-grade glioma.
Abate, ME; Attinà, G; Caldarelli, M; Cefalo, G; Clerico, A; Colosimo, C; Di Rocco, C; Garré, ML; Lazzareschi, I; Madon, E; Massimino, M; Maurizi, P; Mazzarella, G; Riccardi, R; Ridola, V; Ruggiero, A; Sandri, A, 2006
)
0.9
"We identified a transcriptomic signature that predicts a common in vitro and in vivo resistance phenotype to these agents, a proportion of which is imprinted recurrently by gene dosage changes in the resistant glioblastoma genome."( Tumor necrosis factor-alpha-induced protein 3 as a putative regulator of nuclear factor-kappaB-mediated resistance to O6-alkylating agents in human glioblastomas.
Bredel, C; Bredel, M; Duran, GE; Harsh, GR; Juric, D; Recht, LD; Scheck, AC; Sikic, BI; Vogel, H; Yu, RX, 2006
)
0.33
" Extended dosing has met with early favourable results."( Exploiting the role of O6-methylguanine-DNA-methyltransferase (MGMT) in cancer therapy.
Middleton, MR; Sabharwal, A, 2006
)
0.33
" Temozolomide and capecitabine were administered concomitantly to 4 sequential cohorts at different dosing levels on Days 1-5 and Days 8-12, with cycles repeated every 21 days until disease progression."( Phase I study of capecitabine in combination with temozolomide in the treatment of patients with brain metastases from breast carcinoma.
Arun, B; Broglio, K; Buchholz, T; Francis, D; Groves, M; Hortobagyi, GN; Meyers, C; Rivera, E; Valero, V; Yin, G, 2006
)
1.5
" Three patients (12%) were changed to standard temozolomide dosing due to side effects, including intractable nausea (n = 2) and multiple cytopenias (n = 1)."( Toxicity and efficacy of protracted low dose temozolomide for the treatment of low grade gliomas.
Gasco, J; Pouratian, N; Schiff, D; Shaffrey, ME; Sherman, JH, 2007
)
0.86
"Dose-limiting adverse effects of thrombocytopenia and leukopenia prevent augmentation of current temozolomide (TMZ) dosing protocols; therefore, we hypothesized that the direct intracranial delivery of TMZ would lead to improved efficacy in an animal model of malignant glioma in an animal model."( Local delivery of temozolomide by biodegradable polymers is superior to oral administration in a rodent glioma model.
Brem, H; Brem, S; Caplan, J; Legnani, F; Li, K; Pradilla, G; Tyler, B, 2007
)
0.89
"Metronomic dosed (MD) chemotherapy as opposed to conventional dosed (CD) chemotherapy is considered an alternate strategy to target angiogenesis and limit host toxicity."( Preclinical pharmacokinetic and pharmacodynamic evaluation of metronomic and conventional temozolomide dosing regimens.
Gallo, JM; Guo, P; Nuthalapati, S; Wang, X; Zhou, Q, 2007
)
0.56
" Subsequently the tumor recurred and the patient had a dramatic and durable response to standard 5 day dosing of adjuvant temozolomide."( Durable response of a radiation-induced, high-grade cerebellar glioma to temozolomide.
Doherty, LM; Drappatz, J; Kesari, S; Monje, ML; Ramakrishna, NR; Wen, PY; Young, G, 2007
)
0.78
" Depending on the intended indication and dosing regimen, PPL can delay or stop development of a compound in the drug discovery process."( Evaluation of a published in silico model and construction of a novel Bayesian model for predicting phospholipidosis inducing potential.
Gehlhaar, D; Greene, N; Johnson, TO; Pelletier, DJ; Tilloy-Ellul, A,
)
0.13
"The efficacy of LM and TMZ in the current dosing schedule is similar to that of TMZ alone."( Randomized trial of the combination of lomeguatrib and temozolomide compared with temozolomide alone in chemotherapy naive patients with metastatic cutaneous melanoma.
Baka, S; Beith, J; Davis, ID; Harris, PA; Haydon, A; Hersey, P; Kefford, RF; Margison, GP; McArthur, GA; Middleton, MR; Mortimer, P; Ranson, M; Sabharwal, A; Seebaran, A; Thompson, D; Watson, AJ, 2007
)
0.59
" The preclinical pharmacokinetic model was scaled to predict temozolomide concentrations in human CSF, normal brain, and brain tumor, and through a series of Monte Carlo simulations, the accumulation of temozolomide in brain tumors under conditions of altered blood-brain barrier permeability, fractional blood volume, and clinical dosing schedules was evaluated."( Predicting human tumor drug concentrations from a preclinical pharmacokinetic model of temozolomide brain disposition.
Gallo, JM; Guo, P; Kruh, GD; Vicini, P; Wang, X; Zhou, Q, 2007
)
0.8
" Through a series of model simulations, it was shown that the brain tumor accumulation of temozolomide varied substantially based on changes in blood-brain barrier permeability and fractional tumor blood volume but minimally based on clinical dosing regimens."( Predicting human tumor drug concentrations from a preclinical pharmacokinetic model of temozolomide brain disposition.
Gallo, JM; Guo, P; Kruh, GD; Vicini, P; Wang, X; Zhou, Q, 2007
)
0.78
"A physiologically based pharmacokinetic modeling approach offers a means to translate preclinical to clinical characteristics of drug disposition in target tissues and, thus, a means to select appropriate drug dosing regimens for achieving optimal target tissue drug concentrations."( Predicting human tumor drug concentrations from a preclinical pharmacokinetic model of temozolomide brain disposition.
Gallo, JM; Guo, P; Kruh, GD; Vicini, P; Wang, X; Zhou, Q, 2007
)
0.56
"The effects of 'metronomic' or extended chemotherapy dosing schedules (ECS) are mediated through poorly understood anti-angiogenic mechanisms."( Metronomic chemotherapy dosing-schedules with estramustine and temozolomide act synergistically with anti-VEGFR-2 antibody to cause inhibition of human umbilical venous endothelial cell growth.
Greenman, J; Hetherington, JW; Lam, T; Little, S; Maraveyas, A, 2007
)
0.58
" Single-dose temozolomide at five dosage levels (267, 355, 472, 628, and 835 mg/m(2)) was given at least 6 h after completion of O(6)-benzylguanine bolus."( Phase I trial of single-dose temozolomide and continuous administration of o6-benzylguanine in children with brain tumors: a pediatric brain tumor consortium report.
Boyett, JM; Broniscer, A; Danks, MK; Friedman, HS; Gajjar, A; Goldman, S; Gururangan, S; Kun, LE; MacDonald, TJ; Packer, RJ; Poussaint, TY; Stewart, CF; Wallace, D, 2007
)
1
" Because preclinical studies suggested that a twice-daily dosing schedule might be more effective, the safety and efficacy of twice-daily dosing of temozolomide were studied in patients with recurrent gliomas at their first, second, or third recurrence."( Multi-institutional phase II study of temozolomide administered twice daily in the treatment of recurrent high-grade gliomas.
Alavi, J; Balmaceda, C; Chen, J; Cheung, YK; Fine, RL; Fisher, PG; Pannullo, S; Peereboom, D; Sisti, M, 2008
)
0.82
"Twice-daily dosing may enhance the efficacy of temozolomide in the treatment of recurrent gliomas without increasing toxicity."( Multi-institutional phase II study of temozolomide administered twice daily in the treatment of recurrent high-grade gliomas.
Alavi, J; Balmaceda, C; Chen, J; Cheung, YK; Fine, RL; Fisher, PG; Pannullo, S; Peereboom, D; Sisti, M, 2008
)
0.87
" It may also allow a reduction in dosage and a decrease in systemic toxicity."( Solid lipid nanoparticles of temozolomide: potential reduction of cardial and nephric toxicity.
Bi, X; Dou, M; Huang, G; Zhang, N, 2008
)
0.64
" Lower dosing of TMZ also is associated with a more beneficial toxicity profile."( Radiochemotherapy in patients with primary glioblastoma comparing two temozolomide dose regimens.
Bischof, M; Combs, SE; Debus, J; Edler, L; Rausch, R; Schulz-Ertner, D; Wagner, F; Wagner, J; Welzel, T; Zabel-du Bois, A, 2008
)
0.58
" Accordingly, a clinical trial using oral temozolomide (TMZ) and subcutaneous PEG-interferon alpha-2b (PEG) in patients with metastatic melanoma was designed to determine the maximal tolerated dosage of both drugs and the antitumoral response."( Temozolomide associated with PEG-interferon in patients with metastatic melanoma: a multicenter prospective phase I/II study.
Bedane, C; Cupissol, D; Delaunay, M; Dereure, O; Dreno, B; Guillot, B; Khamari, A; Picot, MC, 2008
)
2.05
" Further studies using ATO and AA with TMZ with this dosing schedule in advanced melanoma are not warranted."( Phase II trial of arsenic trioxide and ascorbic acid with temozolomide in patients with metastatic melanoma with or without central nervous system metastases.
Bael, TE; Gollob, JA; Peterson, BL, 2008
)
0.59
" Imatinib doses up to 1,000 mg/day for 8 consecutive days are well tolerated when combined with standard TMZ dosing for MG patients."( Safety and pharmacokinetics of dose-intensive imatinib mesylate plus temozolomide: phase 1 trial in adults with malignant glioma.
Desjardins, A; Egorin, MJ; Friedman, AH; Friedman, HS; Gururangan, S; Herndon, JE; Lagattuta, TF; McLendon, R; Quinn, JA; Reardon, DA; Rich, JN; Salvado, AJ; Sathornsumetee, S; Vredenburgh, JJ, 2008
)
0.58
" Noninvasive imaging of apoptosis facilitates optimization of therapeutic protocols regarding dosing and schedule and enables identification of efficacious combination therapies."( Noninvasive imaging of apoptosis and its application in cancer therapeutics.
Coppola, JM; Rehemtulla, A; Ross, BD, 2008
)
0.35
" If further studies are to be performed, emerging data suggest that higher daily doses of lomeguatrib and a dosing period beyond that of TMZ should be evaluated."( A phase II trial of lomeguatrib and temozolomide in metastatic colorectal cancer.
Khan, OA; Levitt, NC; Margison, GP; Michael, M; Middleton, MR; Midgley, R; Mortimer, P; Olver, I; Ranson, M; Watson, AJ, 2008
)
0.62
" Several preliminary studies have been initiated to address the issue of resistance and suppression of MGMT activity, and have used alternative temozolomide dosing schedules and O(6)-guanine mimetic agents as substrates for MGMT."( Mechanisms of disease: temozolomide and glioblastoma--look to the future.
Chamberlain, MC; Mrugala, MM, 2008
)
0.86
" A variety of dosing schedules that increase the duration of exposure and the cumulative dose of temozolomide are currently being investigated for the treatment of glioma, with the goal of improving antitumor activity and overcoming resistance."( New (alternative) temozolomide regimens for the treatment of glioma.
Platten, M; Weller, M; Wick, W, 2009
)
0.9
" The toxicity of the dose-intense pre-RT regimen used in this study may warrant evaluation of other, less intense dosing strategies."( Phase II trial of preirradiation and concurrent temozolomide in patients with newly diagnosed anaplastic oligodendrogliomas and mixed anaplastic oligoastrocytomas: RTOG BR0131.
Berkey, B; Biggs, C; Blumenthal, DT; Brown, P; Giannini, C; Herman, J; Jenkins, R; Macdonald, D; Mehta, M; Peereboom, D; Schultz, C; Suh, JH; Vogelbaum, MA, 2009
)
0.61
" Consistent with a functional significance of MGMT induction, treatment of GBM43 with a protracted low-dose TMZ regimen was significantly less effective than a shorter high-dose regimen, while survival for GBM14 was improved with the protracted dosing regimen."( Induction of MGMT expression is associated with temozolomide resistance in glioblastoma xenografts.
Carlson, BL; Decker, PA; Grogan, PT; James, CD; Kitange, GJ; Lamont, JD; Sarkaria, JN; Schroeder, MA; Wu, W, 2009
)
0.61
" Extended ABT-888 dosing schedules showed no advantage compared to simultaneous TMZ administration."( The PARP inhibitor, ABT-888 potentiates temozolomide: correlation with drug levels and reduction in PARP activity in vivo.
Bontcheva-Diaz, VD; Bouska, JJ; Bukofzer, G; Colon-Lopez, M; Donawho, CK; Frost, DJ; Giranda, VL; Guan, R; Jarvis, K; Johnson, EF; Klinghofer, V; Liu, X; Luo, Y; Olson, A; Palma, JP; Penning, TD; Rodriguez, LE; Rosenberg, SH; Saltarelli, MJ; Shi, Y; Stavropoulos, JA; Zhu, GD,
)
0.4
" The system analysis technique, embodied in the convolution integral, generated an impulse response function that, when convolved with temozolomide plasma concentration input functions, yielded predicted normal brain and brain tumor temozolomide concentration profiles for different temozolomide dosing regimens (75-200 mg/m(2)/d)."( A new model for prediction of drug distribution in tumor and normal tissues: pharmacokinetics of temozolomide in glioma patients.
Aboagye, EO; Brock, CS; Gallo, JM; Price, PM; Rosso, L; Saleem, A; Turkheimer, FE, 2009
)
0.77
"O(6)-BG when added to a 1-day dosing regimen of temozolomide was able to restore temozolomide sensitivity in patients with temozolomide-resistant anaplastic glioma, but there seemed to be no significant restoration of temozolomide sensitivity in patients with temozolomide-resistant GBM."( Phase II trial of temozolomide plus o6-benzylguanine in adults with recurrent, temozolomide-resistant malignant glioma.
Bigner, DD; Desjardins, A; Friedman, AH; Friedman, HS; Gururangan, S; Herndon, JE; Jiang, SX; McLendon, RE; Quinn, JA; Reardon, DA; Rich, JN; Sampson, JH; Vredenburgh, JJ; Walker, A, 2009
)
0.94
" It has become common practice to re-expose patients to TMZ who had been previously treated with TMZ, or to switch patients to alternative dosing regimens of TMZ when there are signs of relapse or progress on standard TMZ therapeutic regimens."( Rechallenge with temozolomide in patients with recurrent gliomas.
Bogdahn, U; Hau, P; Jauch, T; Pascher, C; Weller, M; Wick, A; Wick, W, 2009
)
0.69
"This phase I clinical trial conducted with patients who had recurrent or progressive malignant glioma (MG) was designed to determine the maximum tolerated dose (MTD) and toxicity of three different 5-day dosing regimens of temozolomide (TMZ) in combination with O(6)-benzylguanine (O(6)-BG)."( Phase I trial of temozolomide plus O6-benzylguanine 5-day regimen with recurrent malignant glioma.
Bigner, DD; Desjardins, A; Friedman, AH; Friedman, HS; Gururangan, S; Herndon, JE; Jiang, SX; McLendon, RE; Quinn, JA; Reardon, DA; Rich, JN; Sampson, JH; Vredenburgh, JJ; Walker, A, 2009
)
0.88
"Lomeguatrib, an O(6)-methylguanine-DNA methyltransferase inactivator, was evaluated in an extended dosing regimen with temozolomide, designed according to pharmacodynamic data from previous studies."( A phase I study of extended dosing with lomeguatrib with temozolomide in patients with advanced melanoma.
Abdi, E; Beith, J; Corrie, PG; Kefford, RF; Kotasek, D; Margison, GP; Middleton, MR; Mortimer, P; Palmer, C; Ranson, M; Thomas, NP; Watson, AJ, 2009
)
0.81
" Early recovery of MGMT activity in tumours suggested that more protracted dosing with LM is required."( O(6)-methylguanine-DNA methyltransferase depletion and DNA damage in patients with melanoma treated with temozolomide alone or with lomeguatrib.
Beith, J; Davis, ID; Haydon, A; Hayward, O; Hersey, P; Kefford, R; Lorigan, P; Margison, GP; McArthur, G; McGown, G; Middleton, MR; Mortimer, P; Ranson, M; Sabharwal, A; Thomson, D; Thorncroft, M; Watson, AJ, 2009
)
0.57
" Regarding the dosing schedule and administration scheme, as well as the co-administration with other anticancer drugs, a C score was attributed for the off label situations."( [Prescription guidebook for temozolomide usage in brain tumors].
Borget, I; Brignone, M; Cartalat-Carel, S; Chinot, O; Hassani, Y; Taillandier, L; Taillibert, S; Tilleul, P, 2009
)
0.65
"Alternative dosing schedules of temozolomide may improve survival in patients with newly diagnosed glioblastoma (GBM) by increasing the therapeutic index, overcoming common mechanisms of temozolomide resistance, or both."( Randomized phase II trial of chemoradiotherapy followed by either dose-dense or metronomic temozolomide for newly diagnosed glioblastoma.
Abrey, LE; Clarke, JL; DeAngelis, LM; Gavrilovic, I; Hormigo, A; Iwamoto, FM; Karimi, S; Lassman, AB; Nolan, CP; Panageas, K; Sul, J, 2009
)
0.86
" Alternative dosing regimens, such as 1-week on/1-week off, or 3-week on/1-week off, that deliver more prolonged exposure have been observed to result in higher cumulative doses than the standard 5-day regimen and may deplete tumor-derived O6-methylguanine-DNA methyltransferase (MGMT) in tumor cells, thus sensitizing tumor cells to the effects of TMZ."( [Treatment of glioma with temozolomide].
Nishikawa, R, 2009
)
0.65
" Neither the extent of the initial resection nor dexamethasone dosing was associated with pseudoprogression."( Population-based study of pseudoprogression after chemoradiotherapy in GBM.
Cairncross, JG; de Robles, PA; Dharmawardene, M; Easaw, JC; Forsyth, PA; Hamilton, MG; Magliocco, AM; McIntyre, JB; Parney, IF; Roldán, GB; Scott, JN; Yan, ES, 2009
)
0.35
" Afterward, adjuvant TMZ chemotherapy was discontinued in one patient and the dosage of TMZ was reduced in the other."( Patient-tailored, imaging-guided, long-term temozolomide chemotherapy in patients with glioblastoma.
Backes, H; Brunn, A; Burghaus, L; Galldiks, N; Heiss, WD; Jacobs, AH; Kracht, LW; Ullrich, RT, 2010
)
0.62
" 44 of the patients underwent chemo-radiotherapy with Temodal dosed 75 mg/m2, and the rest 135 ones received RT alone."( Postoperative chemo-radiotherapy with temodal in patients with glioblastoma multiforme--survival rates and prognostic factors.
Radev, LR; Semerdjieva, ML; Vlaikova, MI; Yaneva, MP,
)
0.13
" In addition, various protracted temozolomide dosing schedules have been evaluated as a strategy to further enhance its anti-tumor activity."( Effect of CYP3A-inducing anti-epileptics on sorafenib exposure: results of a phase II study of sorafenib plus daily temozolomide in adults with recurrent glioblastoma.
Bigner, DD; Desjardins, A; Friedman, AH; Friedman, HS; Gururangan, S; Herndon, JE; Janney, D; Marcello, J; McLendon, RE; Peters, K; Reardon, DA; Sampson, JH; Vredenburgh, JJ, 2011
)
0.86
" The originally approved temozolomide dosing regimen is 150 to 200 mg/m(2) per day (Days 1 to 5 every 28-day cycle [5 of 28 days])."( Dose-dense temozolomide regimens: antitumor activity, toxicity, and immunomodulatory effects.
Hwu, WJ; Neyns, B; Reardon, DA; Tosoni, A, 2010
)
1.05
" Metronomic dosing of temozolomide (TMZ) combined with standard radiotherapy may improve survival by increasing the therapeutic index and anti-angiogenic effect of TMZ."( A multi-centre Canadian pilot study of metronomic temozolomide combined with radiotherapy for newly diagnosed paediatric brainstem glioma.
Bartels, U; Baruchel, S; Bouffet, E; Eisenstat, D; Gammon, J; Huang, A; Hukin, J; Johnston, DL; Samson, Y; Sharp, JR; Stempak, D; Stephens, D; Tabori, U, 2010
)
0.93
"Chemoradiotherapy with metronomic dosing of TMZ showed similar toxicity to previous TMZ regimens, and does not appear to improve survival in paediatric DIBSG."( A multi-centre Canadian pilot study of metronomic temozolomide combined with radiotherapy for newly diagnosed paediatric brainstem glioma.
Bartels, U; Baruchel, S; Bouffet, E; Eisenstat, D; Gammon, J; Huang, A; Hukin, J; Johnston, DL; Samson, Y; Sharp, JR; Stempak, D; Stephens, D; Tabori, U, 2010
)
0.61
" These combinations appear to be the most promising for in vivo pre-clinical studies, with a view to testing in melanoma patients as a continuous dosing strategy, due to the in vitro additive inhibitory effect on growth seen in both endothelial and cancer cells."( Sorafenib enhances the in vitro anti-endothelial effects of low dose (metronomic) chemotherapy.
Cawkwell, L; Little, SJ; Maraveyas, A; Murray, A; Stanley, P, 2010
)
0.36
" On the basis of these results, the recommended Phase II dosage currently being tested is RAD001 70 mg/week in combination with standard chemoradiotherapy."( North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme.
Brown, PD; Buckner, JC; Galanis, E; Giannini, C; Jaeckle, KA; McGraw, S; Peller, PJ; Sarkaria, JN; Uhm, JH; Wu, W, 2011
)
0.59
"After diagnostic surgery or biopsy, treatment with tipifarnib started 5 to 9 days before initiating radiotherapy, twice daily, in 4-week cycles using discontinuous dosing (21 out of 28 days), until toxicity or progression."( A phase I trial of tipifarnib with radiation therapy, with and without temozolomide, for patients with newly diagnosed glioblastoma.
Abrey, L; Chang, SM; Cloughesy, TF; DeAngelis, LM; Demopoulos, A; Drappatz, J; Fine, HA; Fink, K; Kesari, S; Lamborn, KR; Lassman, AB; Lieberman, FS; Malkin, MG; Mehta, MP; Nghiemphu, PL; Prados, MD; Robins, HI; Torres-Trejo, A; Wen, PY, 2011
)
0.6
" All patients received TMZ at a dosage of 90 mg/m(2)/day for 42 days to a dose of 59."( Temozolomide in the treatment of children with newly diagnosed diffuse intrinsic pontine gliomas: a report from the Children's Oncology Group.
Bouffet, E; Cohen, KJ; Heideman, RL; Holmes, EJ; Lavey, RS; Pollack, IF; Zhou, T, 2011
)
1.81
" We identified melanoma cell lines with different sensitivities to single versus prolonged clinical dosing regimens of temozolomide treatment and assessed a variety of potential resistance mechanisms using this model."( Temozolomide chemoresistance heterogeneity in melanoma with different treatment regimens: DNA damage accumulation contribution.
Boeckmann, L; Emmert, S; Kuschal, C; Nickel, AC; Schaefer, A; Schön, MP; Thomale, J; Thoms, KM, 2011
)
2.02
" Cediranib was dosed at 3 mg/kg daily five times a week orally for 2 weeks."( Cediranib enhances control of wild type EGFR and EGFRvIII-expressing gliomas through potentiating temozolomide, but not through radiosensitization: implications for the clinic.
Andersen, B; Dicker, AP; Lawrence, RY; Liu, Y; Wachsberger, PR; Xia, X, 2011
)
0.59
" In our case, TMZ administration, despite changing the TMZ dosing regimen to prompt a drug response, was incapable of depleting MGMT stores."( Aggressive silent corticotroph adenoma progressing to pituitary carcinoma: the role of temozolomide therapy.
Cusimano, M; Fadul, CE; Gonzalez, R; Horvath, E; Kovacs, K; Moshkin, O; Ortiz, LD; Rotondo, F; Scheithauer, BW; Syro, LV; Uribe, H,
)
0.35
"We conducted a phase I clinical trial of the combination of SCH 66336 with temozolomide administered on the standard 5-day dosing schedule."( A phase I trial of the farnesyl transferase inhibitor, SCH 66336, with temozolomide for patients with malignant glioma.
Coan, AD; Desjardins, A; Friedman, AH; Friedman, HS; Herndon, JE; Peters, KB; Reardon, DA; Threatt, S; Vredenburgh, JJ, 2011
)
0.83
" Additional trials are needed to better define the optimal dosing in such patients."( A phase I study of bortezomib and temozolomide in patients with advanced solid tumors.
Chow, W; Chung, V; Cristea, M; Frankel, P; Koehler, S; Leong, L; Lim, D; Martel, C; Morgan, R; Portnow, J; Reckamp, K; Shibata, S; Synold, TW; Twardowski, P, 2012
)
0.66
" The growing body of evidence demonstrating the clinical importance of O6-methylguanine methyltransferase (MGMT) has generated a considerable interest in the exploration of strategies to overcome MGMT-mediated resistance to alkylating agents; for example protracted administration of Temozolomide (TMZ) may result in more extensive and sustained depletion of MGMT; for this reason a variety of dosing schedules that increase the duration of exposure and the cumulative dose of TMZ are being investigated for the treatment of patient with recurrent malignant glioma after standard treatment."( Rechallenge with temozolomide in recurrent glioma.
Botturi, A; Fariselli, L; Ferrari, D; Gaviani, P; Lamperti, E; Salmaggi, A; Silvani, A; Simonetti, G, 2011
)
0.89
" These may include TMZ concentrations in the brain parenchyma, TMZ dosing schemes, hypoxic microenvironments, niche factors, and the re-acquisition of stem cell properties by non-stem cells."( Chemoresistance of glioblastoma cancer stem cells--much more complex than expected.
Beier, CP; Beier, D; Schulz, JB, 2011
)
0.37
" Temozolomide, dosed according to MGMT methylation status, demonstrated modest clinical activity in elderly patients with AML, especially in those presenting with fewer comorbidities and low disease burden."( Tailored temozolomide therapy according to MGMT methylation status for elderly patients with acute myeloid leukemia.
Arber, DA; Coutre, SE; Gotlib, J; Kohrt, HE; Medeiros, BC; Zehnder, JL; Zhang, B, 2012
)
1.71
"Seventy-one eligible patients 70 years of age or older with newly diagnosed GBM and a Karnofsky performance status ≥60 were treated with a short course of RT (40 Gy in 15 fractions over 3 weeks) plus TMZ at the dosage of 75 mg/m(2) per day followed by 12 cycles of adjuvant TMZ (150-200 mg/m(2) for 5 days during each 28-day cycle)."( Phase II study of short-course radiotherapy plus concomitant and adjuvant temozolomide in elderly patients with glioblastoma.
Arcella, A; Caporello, P; De Sanctis, V; Enrici, RM; Giangaspero, F; Lanzetta, G; Minniti, G; Salvati, M; Scaringi, C, 2012
)
0.61
"A dosage of 10 mg everolimus daily with TMZ 150 mg/m(2)/day for five consecutive days every 28 days in patients is the recommended dose for this regimen."( A phase I study of temozolomide and everolimus (RAD001) in patients with newly diagnosed and progressive glioblastoma either receiving or not receiving enzyme-inducing anticonvulsants: an NCIC CTG study.
Easaw, J; Eisenhauer, E; Kavan, P; Lwin, Z; Macdonald, D; Macneil, M; Mason, WP; McIntosh, L; Thiessen, B; Urva, S, 2012
)
0.71
" Scheduling of docetaxel before selumetinib was more beneficial than when selumetinib was dosed before docetaxel and demonstrated a pro-apoptotic phenotype."( The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models.
Alferez, D; Davies, BR; Heaton, SP; Heier, A; Holt, SV; Logié, A; Odedra, R; Smith, PD; Wilkinson, RW, 2012
)
0.38
" It could therefore be used as an important platform for better prediction of drug dosing and schedule towards personalized medicine."( Towards personalized medicine with a three-dimensional micro-scale perfusion-based two-chamber tissue model system.
Barker, J; Foltz, G; Honkakoski, P; Küblbeck, J; Li, W; Lin, B; Ma, L; Zhang, J; Zhou, C, 2012
)
0.38
" Moreover, this class of compounds possesses a generally favorable in vitro ADME profile, along with good exposure levels in plasma and brain following intraperitoneal dosing (30 mg/kg body weight) in mice."( Synthesis, biological evaluation, and structure-activity relationships of a novel class of apurinic/apyrimidinic endonuclease 1 inhibitors.
Dorjsuren, D; Jadhav, A; Maloney, DJ; Rai, G; Simeonov, A; Vyjayanti, VN; Wilson, DM, 2012
)
0.38
" These results suggest that GBMs with EGFR amplification are a heterogenous group of tumors and that behavior might differ according to the degree of amplification, although not in a straightforward dose-response manner."( Paradoxical relationship between the degree of EGFR amplification and outcome in glioblastomas.
Bortoluzzi, S; Cieply, K; Fardo, DW; Hamilton, RL; Hobbs, J; Horbinski, C; Nikiforova, MN, 2012
)
0.38
"The effectiveness of temozolomide (TMZ) dosing schemes and the "rechallenge" of recurrent glioblastoma (GBM) with TMZ are controversial."( Efficacy of clinically relevant temozolomide dosing schemes in glioblastoma cancer stem cell lines.
Beier, CP; Beier, D; Brawanski, K; Hau, P; Schriefer, B; Schulz, JB; Weis, J, 2012
)
0.98
" These data reassuringly suggest that BEV does not significantly change the ECF tumor concentrations of TMZ in either tumor-bearing or normal brain when dosed 36 h prior to TMZ."( The impact of bevacizumab on temozolomide concentrations in intracranial U87 gliomas.
Blakeley, JO; Brastianos, H; Brem, H; Grossman, R; Rudek, MA; Tyler, B; Zadnik, P, 2012
)
0.67
" Dose-response and cellular growth assays indicate that erlotinib reduces cell proliferation in all tested cell lines without inducing cytotoxic effects."( EGFR inhibition in glioma cells modulates Rho signaling to inhibit cell motility and invasion and cooperates with temozolomide to reduce cell growth.
Fernández de Mattos, S; Ramis, G; Rodríguez, J; Thomàs-Moyà, E; Villalonga, P, 2012
)
0.59
" No DLTs were encountered, but vorinostat dosing could not be escalated further due to thrombocytopenia."( Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03.
Ames, MM; Chang, SM; Cloughesy, TF; Desideri, S; Drappatz, J; Espinoza-Delgado, I; Gilbert, MR; Kuhn, JG; Lamborn, KR; Lassman, AB; Lee, EQ; Lieberman, FS; McGovern, RM; Prados, MD; Puduvalli, VK; Reid, JM; Robins, HI; Wen, PY; Xu, J; Ye, X; Yung, WK, 2012
)
0.63
" Treatment was well tolerated in all dosage groups."( Phase I-II study of radiopeptide 177Lu-octreotate in combination with capecitabine and temozolomide in advanced low-grade neuroendocrine tumors.
Claringbold, PG; Price, RA; Turner, JH, 2012
)
0.6
" It was designed to maximize cytoreduction via high dosing of synergistically interacting agents, while minimizing morbidity in patients with resistant neuroblastoma (NB) and ineligible for clinical trials due to myelosuppression from previous therapy."( 5-day/5-drug myeloablative outpatient regimen for resistant neuroblastoma.
Basu, EM; Cheung, NK; Kramer, K; Kushner, BH; Modak, S; Roberts, SS, 2013
)
0.39
" This regimen was based on our studies with carcinoid cell lines that showed synergistic cytotoxicity with sequence-specific dosing of 5-fluorouracil preceding temozolomide (TMZ)."( Capecitabine and temozolomide (CAPTEM) for metastatic, well-differentiated neuroendocrine cancers: The Pancreas Center at Columbia University experience.
Allendorf, J; Chabot, JA; Dinnen, RD; Fine, RL; Gulati, AP; Krantz, BA; Lee, JA; Mao, Y; Moss, RA; Mowatt, KB; Schreibman, S; Schrope, B; Sherman, WH; Stevens, PD; Tsushima, DA, 2013
)
0.93
" In a human GBM xenograft model, a single daily dosage of MB does not activate AMP-activated protein kinase signaling, and no tumor regression was observed."( Reversing the Warburg effect as a treatment for glioblastoma.
Bigner, DD; Choudhury, GR; Ghorpade, A; Keir, ST; Li, W; Liu, R; Poteet, E; Ryou, MG; Simpkins, JW; Tang, L; Wen, Y; Winters, A; Yan, H; Yang, SH; Yuan, F, 2013
)
0.39
" Sufficient information on steroid dosing was available in 72 patients included in the final analysis."( Steroid management in newly diagnosed glioblastoma.
Deangelis, LM; Deutsch, MB; Lassman, AB; Panageas, KS, 2013
)
0.39
"Temozolomide (TMZ) is important chemotherapy for glioblastoma multiforme (GBM), but the optimal dosing schedule is unclear."( Efficacy of protracted temozolomide dosing is limited in MGMT unmethylated GBM xenograft models.
Anderson, SK; Ballman, KV; Carlson, BL; Cen, L; Decker, PA; Giannini, C; Grogan, PT; Kitange, GJ; Mladek, AC; Pokorny, JL; Sarkaria, JN; Schroeder, MA; Wu, W, 2013
)
2.14
"The efficacies of different clinically relevant dosing regimens were compared in a panel of 7 primary GBM xenografts in an intracranial therapy evaluation model."( Efficacy of protracted temozolomide dosing is limited in MGMT unmethylated GBM xenograft models.
Anderson, SK; Ballman, KV; Carlson, BL; Cen, L; Decker, PA; Giannini, C; Grogan, PT; Kitange, GJ; Mladek, AC; Pokorny, JL; Sarkaria, JN; Schroeder, MA; Wu, W, 2013
)
0.7
" There was no clear relationship between vorinostat dosage and drug exposure over the dose range studied."( A pediatric phase 1 trial of vorinostat and temozolomide in relapsed or refractory primary brain or spinal cord tumors: a Children's Oncology Group phase 1 consortium study.
Ahern, C; Ames, MM; Blaney, SM; Fouladi, M; Gilbertson, RJ; Horton, T; Hummel, TR; Ingle, AM; McGovern, RM; Reid, JM; Wagner, L; Weigel, B, 2013
)
0.65
" Comparisons included no chemotherapy, non-temozolomide chemotherapy or different dosing schedules of temozolomide."( Temozolomide for high grade glioma.
Garside, R; Grant, R; Hart, MG; Rogers, G; Stein, K, 2013
)
2.1
" We report five patients who received long-term treatment with TMZ chemotherapy at normal dosing levels."( Long-term treatment with temozolomide in malignant glioma.
Defrates, SR; Lightner, DD; Mannas, JP; Pittman, T; Villano, JL, 2014
)
0.71
"Mutagenic and clastogenic effects of some DNA damaging agents such as methyl methanesulfonate (MMS) and ethyl methanesulfonate (EMS) have been demonstrated to exhibit a nonlinear or even "thresholded" dose-response in vitro and in vivo."( Quantitative assessment of the dose-response of alkylating agents in DNA repair proficient and deficient ames tester strains.
Guérard, M; Tang, L; Zeller, A, 2014
)
0.4
" Overall, we have verified that TMZ in addition to being an alkylating and cytotoxic chemotherapy, also possess immune modulatory effect in MM patients treated with standard dosage of TMZ."( Immune modulations during chemoimmunotherapy & novel vaccine strategies--in metastatic melanoma and non small-cell lung cancer.
Iversen, TZ, 2013
)
0.39
" Patients aged over 70 years with favorable KPS, or patients aged 60-70 years with borderline KPS, should be considered for monotherapy utilizing standard TMZ dosing for patients with MGMT-methylated tumors, and hypofractionated RT (34 Gy in ten fractions or 40 Gy in 15 fractions) for patients with MGMT-unmethylated tumors."( Treatment options and outcomes for glioblastoma in the elderly patient.
Arvold, ND; Reardon, DA, 2014
)
0.4
"Temozolomide, when dosed at 50, 25, 10, or 5 mg/kg, 5 days per week, beginning 3 days after inoculation, completely prevented the formation of experimental brain metastases from MGMT-negative 231-BR-EGFP cells."( Profound prevention of experimental brain metastases of breast cancer by temozolomide in an MGMT-dependent manner.
Biernat, W; Duchnowska, R; Gril, B; Hewitt, SM; Hua, E; Jassem, J; Liewehr, DJ; Palmieri, D; Qian, Y; Sosińska-Mielcarek, K; Stark, AM; Steeg, PS; Steinberg, SM; Woditschka, S, 2014
)
2.08
" The ECM algorithm for incomplete data is applied to estimating the dose-response relationship in the proposed model."( Modeling sustained treatment effects in tumor xenograft experiments.
Deng, D; Fang, HB; Tan, M; Zhang, T, 2014
)
0.4
" In vivo drug efficacy, pharmacokinetics, and pharmacodynamics were analyzed using clinically relevant dosing regimens."( Discordant in vitro and in vivo chemopotentiating effects of the PARP inhibitor veliparib in temozolomide-sensitive versus -resistant glioblastoma multiforme xenografts.
Bakken, KK; Boakye-Agyeman, F; Carlson, BL; Gupta, SK; Kizilbash, SH; Mladek, AC; Reid, J; Sarkaria, JN; Schroeder, MA, 2014
)
0.62
"In vitro cytotoxicity assays do not adequately model the therapeutic index of PARP inhibitors, as concentrations of veliparib and TMZ required to sensitize TMZ-resistant cancer cells in vivo cannot be achieved using a tolerable dosing regimen."( Discordant in vitro and in vivo chemopotentiating effects of the PARP inhibitor veliparib in temozolomide-sensitive versus -resistant glioblastoma multiforme xenografts.
Bakken, KK; Boakye-Agyeman, F; Carlson, BL; Gupta, SK; Kizilbash, SH; Mladek, AC; Reid, J; Sarkaria, JN; Schroeder, MA, 2014
)
0.62
"The EORTC-NCIC regimen for glioblastoma requires different dosing of temozolomide (TMZ) during radiation and maintenance therapy."( Analyzing temozolomide medication errors: potentially fatal.
Bressler, LR; Gabay, MP; Letarte, N; Long, KE; Stachnik, JM; Villano, JL, 2014
)
1.04
" In phase II, participants were randomized (stratified by age and KPS) to receive A, T or AT; A was dosed at 40 mg/day and T at 75 mg/m(2) for 21 of 28 days."( Phase I/randomized phase II study of afatinib, an irreversible ErbB family blocker, with or without protracted temozolomide in adults with recurrent glioblastoma.
Cong, J; Cseh, A; Eisenstat, DD; Fu, Y; Kavan, P; Mason, WP; Mathieu, D; Nabors, LB; Perry, JR; Phuphanich, S; Reardon, DA; Shapiro, W; Wind, S, 2015
)
0.63
" We therefore evaluated a 5-day dosing schedule of temozolomide and examined MGMT as a predictive biomarker for temozolomide treatment in SCLC."( Trial of a 5-day dosing regimen of temozolomide in patients with relapsed small cell lung cancers with assessment of methylguanine-DNA methyltransferase.
Bergagnini, I; Drilon, A; Ginsberg, MS; Heguy, A; Holodny, AI; Huberman, K; Kadota, K; Kris, MG; Krug, LM; Pietanza, MC; Riely, GJ; Sima, CS; Sumner, DK; Travis, WD; Zauderer, MG, 2014
)
0.93
" Because tasisulam is highly albumin-bound, patients in the tumor-specific confirmation arms were dosed targeting specific albumin-corrected exposure ranges (AUCalb) identified during dose-escalation (3,500 h*μg/mL [75th percentile] for docetaxel, temozolomide, and cisplatin; 4,000 h*μg/mL for gemcitabine and erlotinib)."( An innovative, multi-arm, complete phase 1b study of the novel anti-cancer agent tasisulam in patients with advanced solid tumors.
Becerra, CR; Braiteh, F; Chen, J; Chow, KH; Conkling, PR; Garbo, L; Ilaria, R; Jotte, RM; Richards, DA; Robert-Vizcarrondo, F; Smith, DA; Stephenson, J; Tai, DF; Turner, PK; Von Hoff, DD, 2015
)
0.6
" Dose-response analysis of cultured GBM cells revealed that DP68 is more potent than DP86 and TMZ and that DP68 was effective even in cell lines resistant to TMZ."( Evaluation of novel imidazotetrazine analogues designed to overcome temozolomide resistance and glioblastoma regrowth.
Gynther, M; Mladek, AC; Phillips, RM; Ramirez, YP; Rautio, J; Ross, AH; Sakaria, JN; Wheelhouse, RT, 2015
)
0.65
" All patients received conformal WBRT (3 Gy × 10-30 Gy), with or without concomitant TMZ administered at a dosage of 75 mg/m(2)/day during the irradiation period."( Phase II randomized study of whole-brain radiation therapy with or without concurrent temozolomide for brain metastases from breast cancer.
Bourgier, C; Cao, KI; Gerber, S; Gobillion, A; Kirova, YM; Le Scodan, R; Lebas, N; Levy, C; Pierga, JY; Savignoni, A, 2015
)
0.64
" All patients received the first cycle of TMZ at a dosage of 150 mg/m(2) starting on the second or third postsurgical day."( Long-term therapy with temozolomide is a feasible option for newly diagnosed glioblastoma: a single-institution experience with as many as 101 temozolomide cycles.
Albanese, V; Barbagallo, GM; Caltabiano, R; Certo, F; Lanzafame, S; Longo, A; Motta, F; Palmucci, S; Paratore, S; Parra, HS; Privitera, G; Scaglione, G, 2014
)
0.71
"01), and higher posttreatment dosing of corticosteroid (P=."( Standard (60 Gy) or short-course (40 Gy) irradiation plus concomitant and adjuvant temozolomide for elderly patients with glioblastoma: a propensity-matched analysis.
Arcella, A; Bozzao, A; Enrici, RM; Esposito, V; Giangaspero, F; Lanzetta, G; Minniti, G; Pace, A; Scaringi, C; Terrenato, I, 2015
)
0.64
" Since TMZ is the most active cytotoxic agent against GBM, and the standard dosing of TMZ has shown favorable safety profile in clinical trials, re-challenge with TMZ in increased dose density schedules for recurrent tumors that have evaded from prior standard TMZ therapy appears to be a rational approach and has been intensively exploited."( Dose-dense temozolomide: is it still promising?
Nagane, M, 2015
)
0.81
" Even though dosing and timing might have been the reasons for this failure, it might also be that TREO does not reach the brain in sufficient amount."( Transport of treosulfan and temozolomide across an in-vitro blood-brain barrier model.
Hupert, M; Linz, U; Santiago-Schübel, B; Stab, J; Wagner, S; Wien, S, 2015
)
0.71
"8- and 21-fold, respectively, when similarly dosed in P-gp-deficient mice."( Brain Exposure of Two Selective Dual CDK4 and CDK6 Inhibitors and the Antitumor Activity of CDK4 and CDK6 Inhibition in Combination with Temozolomide in an Intracranial Glioblastoma Xenograft.
Ajamie, RT; De Dios, A; Gelbert, LM; Kulanthaivel, P; Raub, TJ; Sanchez-Martinez, C; Sawada, GA; Shannon, HE; Staton, BA; Wishart, GN, 2015
)
0.62
" We investigated the maximum tolerated dose (MTD) of iniparib with monthly (m) and continuous (c) temozolomide (TMZ) dosing schedules in patients with malignant gliomas (MG)."( Phase I study of iniparib concurrent with monthly or continuous temozolomide dosing schedules in patients with newly diagnosed malignant gliomas.
Blakeley, JO; Chi, AS; Desideri, S; Emmons, G; Garcia Ribas, I; Grossman, SA; Mikkelsen, T; Nabors, LB; Peereboom, D; Rosenfeld, MR; Supko, JG; Ye, X, 2015
)
0.87
" We used these screening methods to evaluate the dependencies of seven patient-derived cell models: three grown on laminin and four grown as neurospheres, against 56 agents in 17-point dose-response curves in 384-well format in triplicate."( High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
deCarvalho, AC; Mikkelsen, T; Quartararo, CE; Reznik, E; Stockwell, BR, 2015
)
0.42
"Midazolam causes a hormetic dose-response relationship in human neuroblastoma cells."( Pretreatment but not subsequent coincubation with midazolam reduces the cytotoxicity of temozolomide in neuroblastoma cells.
Bauer, I; Braun, S; Pannen, B; Werdehausen, R, 2015
)
0.64
" We confirmed in these studies that after completion of the Q7D×3 dosing of IrC™, but not IRN, the tumor-associated vascular was normalized as compared to untreated tumors."( Determination of an optimal dosing schedule for combining Irinophore C™ and temozolomide in an orthotopic model of glioblastoma.
Anantha, M; Backstrom, I; Bally, MB; Chu, F; Kalra, J; Masin, D; Strutt, D; Verreault, M; Walker, D; Waterhouse, D; Wehbe, M; Yapp, DT, 2015
)
0.65
" In comparing DNA damage signaling after dosing with veliparib/TMZ or TMZ alone, increased phosphorylation of damage-responsive proteins (KAP1, Chk1, Chk2, and H2AX) was observed only in MGMT promoter-hypermethylated lines."( Delineation of MGMT Hypermethylation as a Biomarker for Veliparib-Mediated Temozolomide-Sensitizing Therapy of Glioblastoma.
Bakken, KK; Ballman, KV; Boakye-Agyeman, F; Carlson, BL; Cen, L; Decker, PA; Eckel-Passow, JE; Gupta, SK; Jenkins, RB; Kitange, GJ; Kizilbash, SH; Mladek, AC; Pokorny, JL; Reid, JM; Sarkar, G; Sarkaria, JN; Schroeder, MA; Sulman, EP; Verhaak, RG, 2016
)
0.66
" This study provides strong scientific rationale for the development of an optimized dosing regimen for a PARP inhibitor with TMZ/IR for upfront treatment of GBM."( Evaluation of Concurrent Radiation, Temozolomide and ABT-888 Treatment Followed by Maintenance Therapy with Temozolomide and ABT-888 in a Genetically Engineered Glioblastoma Mouse Model.
Chenevert, TL; Galbán, CJ; Galbán, S; Heist, KA; Holland, EC; Lemasson, B; Li, Y; Rehemtulla, A; Ross, BD; Tsein, C; Wang, H; Zhu, Y, 2016
)
0.71
" Treatment of TMZ along with a sublethal dosage range of SU1498, a chemical inhibitor of the VEGF receptor signaling, induced significant cell death in both TMZ-sensitive and TMZ-resistant GBM cells without changing the status of the MGMT promoter methylation."( Combined inhibition of vascular endothelial growth factor receptor signaling with temozolomide enhances cytotoxicity against human glioblastoma cells via downregulation of Neuropilin-1.
Choi, C; Choi, K; Kim, E; Lee, J; Ryu, SW, 2016
)
0.66
" So far, inhibition of angiogenesis by compounds such as bevacizumab, cediranib, enzastaurin or cilengitide as well as alternative dosing schedules of temozolomide did not prolong survival, neither at primary diagnosis nor at recurrent disease."( Pharmacotherapies for the treatment of glioblastoma - current evidence and perspectives.
Gramatzki, D; Roth, P; Seystahl, K; Weller, M, 2016
)
0.63
" According with the poor Performance Status (PS = 2) and to reduce the toxicity of the treatment was chosen an intermittent dosing regimen of metronomic temozolomide (75 mg/m(2)/day-one-week-on/on-week-off)."( Metronomic temozolomide as second line treatment for metastatic poorly differentiated pancreatic neuroendocrine carcinoma.
Arcella, A; Ascierto, PA; Cicala, D; De Divitiis, C; Grimaldi, AM; Iaffaioli, RV; Romano, GM; Simeone, E; Tafuto, S; Tatangelo, F; von Arx, C, 2016
)
1.02
" In vivo, we implanted the cells orthotopically in nude mice and administered CBL0137 in various dosing regimens to assess brain and tumor accumulation of CBL0137, its effect on tumor cell proliferation and apoptosis, and on survival of mice with and without temozolomide (TMZ)."( Anticancer drug candidate CBL0137, which inhibits histone chaperone FACT, is efficacious in preclinical orthotopic models of temozolomide-responsive and -resistant glioblastoma.
Barone, TA; Burkhart, CA; Gudkov, AV; Gurova, KV; Haderski, G; Plunkett, RJ; Purmal, AA; Safina, A, 2017
)
0.84
" As a comparison group 33 patients with bevacizumab for at least 6 months continuously dosed at 10 mg/kg every 2 weeks were selected."( Impact of tapering and discontinuation of bevacizumab in patients with progressive glioblastoma.
Hertenstein, A; Hielscher, T; Menn, O; Platten, M; Wick, A; Wick, W; Wiestler, B; Winkler, F, 2016
)
0.43
" Dose-response curves were constructed using cell proliferation and sphere-forming assays."( The effects of tumor treating fields and temozolomide in MGMT expressing and non-expressing patient-derived glioblastoma cells.
Clark, PA; Deming, DA; Gaal, JT; Kuo, JS; Pasch, CA; Robins, HI; Strebe, JK, 2017
)
0.72
" Metronomic dosing of cytotoxic chemotherapy has emerged as a promising option to achieve this objective."( Phase I study of low-dose metronomic temozolomide for recurrent malignant gliomas.
Alsop, DC; Callahan, A; Giarusso, B; O'Loughlin, L; Timmons, J; Wong, ET, 2016
)
0.71
" As a part of the study, two DNA alkylating agents, methylnitrosourea (MNU) and temozolomide (TMZ), were dosed by single oral gavage at 25, 50, and 100mg/kg body weight."( Evaluation of the mutagenicity of alkylating agents, methylnitrosourea and temozolomide, using the rat Pig-a assay with total red blood cells or reticulocytes.
Ando, M; Inoue, Y; Iwase, Y; Kato, T; Muto, S; Uno, Y; Yamada, K, 2016
)
0.89
"6 μg/day) with negligible leakage into the peripheral blood (<100 ng) rendering ~1000 fold differential drug dosage in tumor versus peripheral blood."( Theranostic 3-Dimensional nano brain-implant for prolonged and localized treatment of recurrent glioma.
Ashokan, A; Gowd, GS; Junnuthula, VR; Koyakutty, M; Nair, SV; Panikar, D; Peethambaran, R; Ramachandran, R; Thomas, A; Thomas, J; Unni, AK, 2017
)
0.46
"Temozolomide (TMZ) for malignant gliomas is traditionally dosed in 5 out of a 28-day cycle, however alternative regimens exist, including dose-dense."( Long-term daily temozolomide with dose-dependent efficacy in MGMT promotor methylation negative recurrent high-grade astrocytoma.
Howard, TA; Villano, JL; Zhou, Z, 2017
)
2.24
" At recurrence, alternate dosing of temozolomide has shown to further deplete methyl-guanine-methyltransferase (MGMT) conferring added activity for patients who have progressed on the standard dosing regimen."( Phase II study of bi-weekly temozolomide plus bevacizumab for adult patients with recurrent glioblastoma.
Ahmadi, MM; Badruddoja, MA; Kuzma, K; Mahadevan, D; Norton, T; Pazzi, M; Sanan, A; Schroeder, K; Scully, T, 2017
)
1.02
" Modern techniques, such as intensity-modulated radiation therapy (IMRT) and proton therapy allow for modifications in radiation dosing and delivery while improving conformality and limiting irradiation of normal tissue."( Fractionated Radiotherapy of Intracranial Gliomas.
Ghia, AJ, 2018
)
0.48
" Temozolomide has several effects on the immune system that are dependent on mode of delivery and the dosing strategy, which may have unpredicted effects on immunotherapy."( Temozolomide for immunomodulation in the treatment of glioblastoma.
Dastmalchi, F; Karachi, A; Mitchell, DA; Rahman, M, 2018
)
2.83
" Dosing optimization (frequency and sequence) is needed prior to additional efficacy studies."( Phase II Study of Iniparib with Concurrent Chemoradiation in Patients with Newly Diagnosed Glioblastoma.
Ahluwalia, MS; Blakeley, JO; Chi, AS; Desideri, S; Eichler, A; Grossman, SA; Mikkelsen, T; Nabors, LB; Ribas, IG; Rosenfeld, MR; Ye, X, 2019
)
0.51
" Our results demonstrate benefit of ddTMZ after previous treatment with standard TMZ dosing with no apparent increase in treatment-related toxicities."( Dose-dense temozolomide for recurrent high-grade gliomas: a single-center retrospective study.
Garcia, CR; Gruber, L; Kumar, SS; Lightner, DD; Morgan, RM; Slone, SA; Villano, JL, 2018
)
0.87
" To facilitate optimal dosing for such studies, we evaluated the plasma and brain pharmacokinetics (PK) of letrozole in NOD-scid gamma (NSG) mice, which are frequently employed for assessing efficacy against patient-derived tumor cells."( Plasma and brain pharmacokinetics of letrozole and drug interaction studies with temozolomide in NOD-scid gamma mice and sprague dawley rats.
Adams, CH; Arora, P; DasGupta, B; Desai, PB; Gudelsky, G, 2019
)
0.74
"These results will guide the optimization of dosing regimen for further development of letrozole for HGG treatment."( Plasma and brain pharmacokinetics of letrozole and drug interaction studies with temozolomide in NOD-scid gamma mice and sprague dawley rats.
Adams, CH; Arora, P; DasGupta, B; Desai, PB; Gudelsky, G, 2019
)
0.74
" Further clinical investigations aimed at elucidating the veliparib exposure-efficacy/safety relationship and optimizing dosing recommendations for maximizing benefit-risk in patients with advanced myeloid malignancies should study veliparib doses ranging up to 120 mg in combination with temozolomide."( Population pharmacokinetics and exposure-response assessment of veliparib co-administered with temozolomide in patients with myeloid leukemias.
Beumer, JH; Chen, A; Gobburu, J; Gojo, I; Gopalakrishnan, M; Greer, JM; Karp, JE; Kiesel, BF; Mehrotra, S; Piekarz, R; Rudek, MA; Singh, R, 2019
)
0.91
" Overall, RO6839921 had a favourable pharmacokinetic profile consistent with intermittent dosing and was well tolerated alone and in combination."( Preclinical evaluation of the first intravenous small molecule MDM2 antagonist alone and in combination with temozolomide in neuroblastoma.
Berry, P; Bonner, J; Chen, L; Daga, A; Kirk, C; Lunec, J; Newell, DR; Pastorino, F; Ponzoni, M; Thomas, HD; Tweddle, DA; Veal, GJ; Wood, KM; Zhao, Y, 2019
)
0.73
" Combinatorial therapy of diosgenin and TMZ significantly reduced the dosage regimen of TMZ and also showed affectivity in hitherto TMZ resistant GBM cells."( Targeting NFE2L2, a transcription factor upstream of MMP-2: A potential therapeutic strategy for temozolomide resistant glioblastoma.
Banerjee, I; Banik, P; Bharti, R; Biswas, A; Das, S; Ghosh, SK; Kumar, U; Mandal, M; Nayak, S; Rajesh, Y, 2019
)
0.73
"Using patient-derived glioblastoma stem cell (GSC) cultures from 15 GBM patients, we described stem cell properties of individual cultures, determined the dose-response relationships of the drugs in the CUSP9, and assessed the efficacy the CUSP9 combination with TMZ in concentrations clinically achievable."( The efficacy of a coordinated pharmacological blockade in glioblastoma stem cells with nine repurposed drugs using the CUSP9 strategy.
Grieg, Z; Langmoen, IA; Sandberg, CJ; Skaga, E; Skaga, IØ; Vik-Mo, EO, 2019
)
0.51
" These results form part of the basis for the translation of the therapy to patients with GBM but the dosing and timing of delivery will have to be explored in depth both experimentally and clinically."( Convection-enhanced delivery of temozolomide and whole cell tumor immunizations in GL261 and KR158 experimental mouse gliomas.
Darabi, A; Enríquez Pérez, J; Kopecky, J; Siesjö, P; Visse, E, 2020
)
0.84
" Side effects associated with chemotherapy delays or modifications included thrombocytopenia (28%) and nausea/vomiting (19%), with temozolomide dosing most frequently modified."( Children with DIPG and high-grade glioma treated with temozolomide, irinotecan, and bevacizumab: the Seattle Children's Hospital experience.
Browd, SR; Cole, BL; Crotty, EE; Ellenbogen, RG; Ermoian, RP; Geyer, JR; Hauptman, JS; Leary, SES; Lee, A; Lockwood, CM; Millard, NE; Ojemann, JG; Olson, JM; Paulson, VA; Sato, AA; Vitanza, NA, 2020
)
1.01
"The interaction of temozolomide (TMZ) (the main chemotherapeutic agent for brain tumors) with blood components has not been studied at the molecular level to date, even though such information is essential in the design of dosage forms for optimal therapy."( The Interaction of Temozolomide with Blood Components Suggests the Potential Use of Human Serum Albumin as a Biomimetic Carrier for the Drug.
Encinar, JA; Esquembre, R; Martínez-Tomé, MJ; Mateo, CR; Rubio-Camacho, M, 2020
)
1.22
" We have successfully employed temozolomide desensitization followed by metronomic dosing of temozolomide."( Temozolomide desensitization followed by metronomic dosing in patients with hypersensitivity.
Divekar, RD; Johnson, DR; Maddox, DE; Neth, BJ; Ruff, MW; Uhm, JH, 2020
)
2.29
"We performed an observational study of 15 patients at Mayo Clinic (Rochester) with a diagnosis of glioma who underwent temozolomide desensitization with subsequent metronomic dosing from May 2012 to January 2017."( Temozolomide desensitization followed by metronomic dosing in patients with hypersensitivity.
Divekar, RD; Johnson, DR; Maddox, DE; Neth, BJ; Ruff, MW; Uhm, JH, 2020
)
2.21
"8% of actual body weight calculated body surface area dosing was determined for concurrent phase temozolomide."( Actual body weight dosing of temozolomide and overall survival in patients with glioblastoma.
Chambers, C; Coppens, R; de Robles, P; Dersch-Mills, D; Folkman, F; Ghosh, S; Hsu, PYH; Leckie, C, 2021
)
1.13
"Temozolomide doses at full actual body weight calculated body surface area dosing during the concurrent phase is required to achieve a similar median OS as seen in the pivotal trial by Stupp et al."( Actual body weight dosing of temozolomide and overall survival in patients with glioblastoma.
Chambers, C; Coppens, R; de Robles, P; Dersch-Mills, D; Folkman, F; Ghosh, S; Hsu, PYH; Leckie, C, 2021
)
2.36
" However, clinical trials show that treatment schedule and drug dosage significantly affect patient survival."( A neuro evolutionary algorithm for patient calibrated prediction of survival in Glioblastoma patients.
Ebrahimi Zade, A; Shahabi Haghighi, S; Soltani, M, 2021
)
0.62
"To investigate the toxicity profile and establish an optimal dosing schedule of zotiraciclib with temozolomide in patients with recurrent high-grade astrocytoma."( Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas.
Aboud, O; Ahmad, S; Antony, R; Armstrong, TS; Boris, L; Bryla, C; Burton, EM; Butler, MK; Calvo, KR; Cordova, C; Figg, WD; Fink, D; Gallin, JI; Garren, N; Gilbert, MR; Gonzales, J; Grajkowska, E; Kuhns, DB; Leeper, H; Lindsley, M; Lollo, N; Long Priel, DA; Mendoza, TR; Mentges, K; Pang, Y; Peer, CJ; Penas-Prado, M; Siegel, C; Sissung, TM; Su, YT; Theeler, BJ; Vera, E; Wu, J; Yu, G; Yuan, Y, 2021
)
1.09
" Once validated, polymorphisms predicting drug metabolism may allow personalized dosing of zotiraciclib."( Phase I Study of Zotiraciclib in Combination with Temozolomide for Patients with Recurrent High-grade Astrocytomas.
Aboud, O; Ahmad, S; Antony, R; Armstrong, TS; Boris, L; Bryla, C; Burton, EM; Butler, MK; Calvo, KR; Cordova, C; Figg, WD; Fink, D; Gallin, JI; Garren, N; Gilbert, MR; Gonzales, J; Grajkowska, E; Kuhns, DB; Leeper, H; Lindsley, M; Lollo, N; Long Priel, DA; Mendoza, TR; Mentges, K; Pang, Y; Peer, CJ; Penas-Prado, M; Siegel, C; Sissung, TM; Su, YT; Theeler, BJ; Vera, E; Wu, J; Yu, G; Yuan, Y, 2021
)
0.87
" Prolonged lisavanbulin dosing from RT start until moribund was required for maximal benefit (GBM6: median survival lisavanbulin/RT 90 vs."( Preclinical modeling in glioblastoma patient-derived xenograft (GBM PDX) xenografts to guide clinical development of lisavanbulin-a novel tumor checkpoint controller targeting microtubules.
Bachmann, F; Bakken, KK; Burgenske, DM; Carlson, BL; Decker, PA; Eckel-Passow, JE; Elmquist, WF; Gampa, G; Giannini, C; He, L; Hu, Z; Kitange, GJ; Kosel, ML; Lane, HA; McSheehy, P; Mladek, AC; Pokorny, JL; Sarkaria, JN; Schmitt-Hoffmann, A; Schroeder, MA; Talele, S; Vaubel, RA, 2022
)
0.72
" Severe myelotoxicity is rare; the risk of grade 4 cytopenias is significantly increased in women, and therefore sex-based dosing should be considered."( Efficacy and Toxicity Analysis of Capecitabine and Temozolomide in Neuroendocrine Neoplasms.
Al-Toubah, T; Haider, M; Pelle, E; Strosberg, JR; Valone, T, 2021
)
0.87
"GBM cells proliferation speed is inversely proportional to the irradiation dose and recedes when the dosage is increased, as expected."( Effectiveness of bortezomib and temozolomide for eradication of recurrent human glioblastoma cells, resistant to radiation.
Bryukhovetskiy, I; Pak, O; Sharma, A; Sharma, HS; Shevchenko, V; Zaitsev, S, 2021
)
0.9
" Isoeffective dosing to 52."( Dose-escalated accelerated hypofractionation for elderly or frail patients with a newly diagnosed glioblastoma.
Ammirati, M; Arnett, A; Beyer, S; Blakaj, DM; Brown, PD; Chakravarti, A; Elder, JB; Giglio, P; Gondi, V; Goranovich, J; Grecula, J; Hardesty, D; Klamer, B; Lonser, R; Matsui, J; Ong, S; Palmer, JD; Perlow, HK; Pillainayagam, C; Raval, RR; Yaney, A; Yang, M, 2022
)
0.72
"Isoeffective dosing to 52."( Dose-escalated accelerated hypofractionation for elderly or frail patients with a newly diagnosed glioblastoma.
Ammirati, M; Arnett, A; Beyer, S; Blakaj, DM; Brown, PD; Chakravarti, A; Elder, JB; Giglio, P; Gondi, V; Goranovich, J; Grecula, J; Hardesty, D; Klamer, B; Lonser, R; Matsui, J; Ong, S; Palmer, JD; Perlow, HK; Pillainayagam, C; Raval, RR; Yaney, A; Yang, M, 2022
)
0.72
" There is insufficient data to make a recommendation about which alternative TMZ dosing provides the best benefits."( Congress of Neurological Surgeons systematic review and evidence-based guidelines update on the role of cytotoxic chemotherapy and other cytotoxic therapies in the management of progressive glioblastoma in adults.
Germano, IM; Olson, JJ; Ormond, DR; Wen, P; Ziu, M, 2022
)
0.72
" Furthermore, the non-cytolytic and non-cytotoxic metronomic dosage of hydroxyurea and temozolomide had increased the DBM therapy outcome by strengthening anti-tumor capability."( A retrospective observational study on cases of anaplastic brain tumors treated with the Di Bella Method: A rationale and effectiveness.
Borghetto, V; Costanzo, E; Di Bella, G, 2021
)
0.84
"8%), requiring a repeated dosing regimen that limits its efficacy and increases adverse events."( Temozolomide: An Overview of Biological Properties, Drug Delivery Nanosystems, and Analytical Methods.
Carvalho, SG; Chorilli, M; Di Filippo, LD; Dutra, JAP; Luiz, MT; Tavares Junior, AG, 2022
)
2.16
" Additionally, by lowering the effective dosage of TMZ, the combination liposomes reduced systemic TMZ-induced toxicity, highlighting the preclinical potential of this novel integrative strategy to deliver combination therapies to brain tumors."( Targeted liposomes for combined delivery of artesunate and temozolomide to resistant glioblastoma.
Chai, T; Du, Q; Hanif, S; Ismail, M; Li, Y; Muhammad, P; Shi, B; Yang, W; Zhang, D; Zheng, M, 2022
)
0.96
" In a drug screening assay, diffusion of temozolomide and carmustine to hydrogel-encapsulated U87 cells from the perfusion solution is measured, and dose-response curves are generated, demonstrating utility as an in vitro mimic of the glioblastoma microenvironment."( Hydrogel-based microfluidic device with multiplexed 3D in vitro cell culture.
Bruns, J; Chen, D; Clancy, A; Nadella, J; Stealey, S; Timperman, A; Zhang, Y; Zustiak, SP, 2022
)
0.99
" We present a novel case of the challenges of dosing temozolomide in a patient with end stage renal disease on peritoneal hemodialysis with unpredictable clearance and toxicities."( Safe administration of temozolomide in end-stage renal disease patients.
Hundal, J; Pereira, MK; Singh, A; Vredenburg, J, 2023
)
1.47
" A stepwise increase in the dosage of temozolomide did not increase the risk of toxicity with HD."( Safe administration of temozolomide in end-stage renal disease patients.
Hundal, J; Pereira, MK; Singh, A; Vredenburg, J, 2023
)
1.49
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (3)

RoleDescription
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
prodrugA compound that, on administration, must undergo chemical conversion by metabolic processes before becoming the pharmacologically active drug for which it is a prodrug.
alkylating agentHighly reactive chemical that introduces alkyl radicals into biologically active molecules and thereby prevents their proper functioning. It could be used as an antineoplastic agent, but it might be very toxic, with carcinogenic, mutagenic, teratogenic, and immunosuppressant actions. It could also be used as a component of poison gases.
[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 (3)

ClassDescription
imidazotetrazineAny organic heterobicyclic compound containing ortho-fused imidazole and tetrazine rings.
monocarboxylic acid amideA carboxamide derived from a monocarboxylic acid.
triazene derivativeA nitrogen molecular entity resulting from the formal substitution of one or more of the hydrogens of triazene.
[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 (13)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
DNA polymerase kappa isoform 1Homo sapiens (human)Potency21.19230.031622.3146100.0000AID588579
muscleblind-like protein 1 isoform 1Homo sapiens (human)Potency28.18380.00419.962528.1838AID2675
[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)
polyadenylate-binding protein 1Homo sapiens (human)IC50 (µMol)54.72504.910023.702976.1900AID602259; AID602260
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
Carbonic anhydrase 12Homo sapiens (human)Ki50.00000.00021.10439.9000AID1425990
Bile salt export pumpHomo sapiens (human)IC50 (µMol)133.00000.11007.190310.0000AID1473738
Tyrosine-protein kinase ABL1Homo sapiens (human)IC50 (µMol)100.00000.00010.712810.0000AID228828
Carbonic anhydrase 1Homo sapiens (human)Ki50.00000.00001.372610.0000AID1425987
Carbonic anhydrase 2Homo sapiens (human)Ki50.00000.00000.72369.9200AID1425988
Tyrosine-protein kinase ABL2Homo sapiens (human)IC50 (µMol)100.00000.00050.14921.0000AID228828
Carbonic anhydrase 9Homo sapiens (human)Ki50.00000.00010.78749.9000AID1425989
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (178)

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)
estrous cycleCarbonic anhydrase 12Homo sapiens (human)
chloride ion homeostasisCarbonic anhydrase 12Homo sapiens (human)
one-carbon metabolic processCarbonic anhydrase 12Homo 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)
one-carbon metabolic processCarbonic anhydrase 1Homo sapiens (human)
morphogenesis of an epitheliumCarbonic anhydrase 2Homo sapiens (human)
positive regulation of synaptic transmission, GABAergicCarbonic anhydrase 2Homo sapiens (human)
positive regulation of cellular pH reductionCarbonic anhydrase 2Homo sapiens (human)
angiotensin-activated signaling pathwayCarbonic anhydrase 2Homo sapiens (human)
regulation of monoatomic anion transportCarbonic anhydrase 2Homo sapiens (human)
secretionCarbonic anhydrase 2Homo sapiens (human)
regulation of intracellular pHCarbonic anhydrase 2Homo sapiens (human)
neuron cellular homeostasisCarbonic anhydrase 2Homo sapiens (human)
positive regulation of dipeptide transmembrane transportCarbonic anhydrase 2Homo sapiens (human)
regulation of chloride transportCarbonic anhydrase 2Homo sapiens (human)
carbon dioxide transportCarbonic anhydrase 2Homo sapiens (human)
one-carbon metabolic processCarbonic anhydrase 2Homo sapiens (human)
positive regulation of cytosolic calcium ion concentrationTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of phospholipase C activityTyrosine-protein kinase ABL2Homo sapiens (human)
negative regulation of Rho protein signal transductionTyrosine-protein kinase ABL2Homo sapiens (human)
exploration behaviorTyrosine-protein kinase ABL2Homo sapiens (human)
cell adhesionTyrosine-protein kinase ABL2Homo sapiens (human)
signal transductionTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of autophagyTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of neuron projection developmentTyrosine-protein kinase ABL2Homo sapiens (human)
peptidyl-tyrosine phosphorylationTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of endocytosisTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of cell adhesionTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of actin cytoskeleton organizationTyrosine-protein kinase ABL2Homo sapiens (human)
protein modification processTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of oxidoreductase activityTyrosine-protein kinase ABL2Homo sapiens (human)
cellular response to retinoic acidTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of establishment of T cell polarityTyrosine-protein kinase ABL2Homo sapiens (human)
regulation of cell motilityTyrosine-protein kinase ABL2Homo sapiens (human)
positive regulation of T cell migrationTyrosine-protein kinase ABL2Homo sapiens (human)
epidermal growth factor receptor signaling pathwayTyrosine-protein kinase ABL2Homo sapiens (human)
protein phosphorylationTyrosine-protein kinase ABL2Homo sapiens (human)
response to hypoxiaCarbonic anhydrase 9Homo sapiens (human)
morphogenesis of an epitheliumCarbonic anhydrase 9Homo sapiens (human)
response to xenobiotic stimulusCarbonic anhydrase 9Homo sapiens (human)
response to testosteroneCarbonic anhydrase 9Homo sapiens (human)
secretionCarbonic anhydrase 9Homo sapiens (human)
one-carbon metabolic processCarbonic anhydrase 9Homo 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)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (55)

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)
zinc ion bindingCarbonic anhydrase 12Homo sapiens (human)
carbonate dehydratase activityCarbonic anhydrase 12Homo 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)
arylesterase activityCarbonic anhydrase 1Homo sapiens (human)
carbonate dehydratase activityCarbonic anhydrase 1Homo sapiens (human)
protein bindingCarbonic anhydrase 1Homo sapiens (human)
zinc ion bindingCarbonic anhydrase 1Homo sapiens (human)
hydro-lyase activityCarbonic anhydrase 1Homo sapiens (human)
cyanamide hydratase activityCarbonic anhydrase 1Homo sapiens (human)
arylesterase activityCarbonic anhydrase 2Homo sapiens (human)
carbonate dehydratase activityCarbonic anhydrase 2Homo sapiens (human)
protein bindingCarbonic anhydrase 2Homo sapiens (human)
zinc ion bindingCarbonic anhydrase 2Homo sapiens (human)
cyanamide hydratase activityCarbonic anhydrase 2Homo sapiens (human)
magnesium ion bindingTyrosine-protein kinase ABL2Homo sapiens (human)
phosphotyrosine residue bindingTyrosine-protein kinase ABL2Homo sapiens (human)
actin monomer bindingTyrosine-protein kinase ABL2Homo sapiens (human)
protein kinase activityTyrosine-protein kinase ABL2Homo sapiens (human)
protein tyrosine kinase activityTyrosine-protein kinase ABL2Homo sapiens (human)
non-membrane spanning protein tyrosine kinase activityTyrosine-protein kinase ABL2Homo sapiens (human)
protein bindingTyrosine-protein kinase ABL2Homo sapiens (human)
ATP bindingTyrosine-protein kinase ABL2Homo sapiens (human)
manganese ion bindingTyrosine-protein kinase ABL2Homo sapiens (human)
actin filament bindingTyrosine-protein kinase ABL2Homo sapiens (human)
carbonate dehydratase activityCarbonic anhydrase 9Homo sapiens (human)
protein bindingCarbonic anhydrase 9Homo sapiens (human)
zinc ion bindingCarbonic anhydrase 9Homo sapiens (human)
molecular function activator activityCarbonic anhydrase 9Homo 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)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (35)

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)
plasma membraneCarbonic anhydrase 12Homo sapiens (human)
membraneCarbonic anhydrase 12Homo sapiens (human)
basolateral plasma membraneCarbonic anhydrase 12Homo sapiens (human)
apical plasma membraneCarbonic anhydrase 12Homo sapiens (human)
plasma membraneCarbonic anhydrase 12Homo 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)
cytosolCarbonic anhydrase 1Homo sapiens (human)
extracellular exosomeCarbonic anhydrase 1Homo sapiens (human)
cytoplasmCarbonic anhydrase 2Homo sapiens (human)
cytosolCarbonic anhydrase 2Homo sapiens (human)
plasma membraneCarbonic anhydrase 2Homo sapiens (human)
myelin sheathCarbonic anhydrase 2Homo sapiens (human)
apical part of cellCarbonic anhydrase 2Homo sapiens (human)
extracellular exosomeCarbonic anhydrase 2Homo sapiens (human)
cytoplasmCarbonic anhydrase 2Homo sapiens (human)
plasma membraneCarbonic anhydrase 2Homo sapiens (human)
apical part of cellCarbonic anhydrase 2Homo sapiens (human)
cytosolTyrosine-protein kinase ABL2Homo sapiens (human)
actin cytoskeletonTyrosine-protein kinase ABL2Homo sapiens (human)
plasma membraneTyrosine-protein kinase ABL2Homo sapiens (human)
nucleolusCarbonic anhydrase 9Homo sapiens (human)
plasma membraneCarbonic anhydrase 9Homo sapiens (human)
membraneCarbonic anhydrase 9Homo sapiens (human)
basolateral plasma membraneCarbonic anhydrase 9Homo sapiens (human)
microvillus membraneCarbonic anhydrase 9Homo sapiens (human)
plasma membraneCarbonic anhydrase 9Homo 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)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (549)

Assay IDTitleYearJournalArticle
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.
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.
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.
AID1347117qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1347127qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347122qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
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.
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.
AID1347114qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347111qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
AID1347113qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347129qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1347118qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347128qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347115qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347123qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347109qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
AID1347125qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
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.
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.
AID1347126qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1347121qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347110qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for A673 cells)2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
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.
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.
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.
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.
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.
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.
AID1347119qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347112qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
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.
AID1347116qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID1347124qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory 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.
AID504812Inverse Agonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID504810Antagonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID282551Antitumor activity against human NCI non-small cell lung cancer cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1079935Cytolytic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is > 5 (see ACUTE). Value is number of references indexed. [column 'CYTOL' in source]
AID1683791Antitumor activity against human NCI-H209 cells xenografted in BALB/c nude mouse assessed as PR rate at 50 mg/kg, po QD for 5 consecutive days at start of study and again 14 days later2020Journal of medicinal chemistry, 12-24, Volume: 63, Issue:24
Discovery of Pamiparib (BGB-290), a Potent and Selective Poly (ADP-ribose) Polymerase (PARP) Inhibitor in Clinical Development.
AID1697834Antitumour activity against human DBTRG-05MG cells xenografted in BALB/c nude mouse assessed as reduction in tumour growth at 5 mg/kg, ip for 5 days and measured every 2 days2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1474445Toxicity in BALB/c mouse assessed as change in body weight at 50 mg/kg, po administered once daily via gavage for 5 days measured twice every week up to 11 days (Rvb = 5.32 g)2017European journal of medicinal chemistry, May-26, Volume: 132Discovery of 2-substituted 1H-benzo[d]immidazole-4-carboxamide derivatives as novel poly(ADP-ribose)polymerase-1 inhibitors with in vivo anti-tumor activity.
AID1497082Cytotoxicity against human MDA-MB-231 cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1697710Effect on ERK expression in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697755Induction of apoptosis in human DBTRG-05MG cells assessed as cleaved PARP expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID661465Cytotoxicity against mouse astrocytes assessed as decrease in cell viability at 1 to 100 uM after 48 hrs by Hoechst 33342 staining2012Journal of medicinal chemistry, Mar-08, Volume: 55, Issue:5
Oxaphosphinanes: new therapeutic perspectives for glioblastoma.
AID1537890Cytotoxicity against P-gp knockout patient-isolated primary glioblastoma cells derived neurospheres assessed as reduction in cell viability at 50 uM measured after 72 hrs by chemiluminescence-based assay2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1736286Selectivity index, ratio of CC50 for Wistar rat astrocytes to EC50 for human U87MG cells2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID1697774Induction of apoptosis in rat RG2 cells assessed as phosphorylated caspace-9 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697687Inhibition of ERK phosphorylation at Tyr204 residues in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1079936Choleostatic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is < 2 (see ACUTE). Value is number of references indexed. [column 'CHOLE' in source]
AID1697639Induction of cell cycle arrest in human DBTRG-05MG cells assessed as p53 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1876511Induction of apoptosis in human U-87 MG cells assessed as early apoptotic cells at 1.20 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb = 2.53 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1825322Cytotoxicity against human U-87 MG cells assessed as cell viability using raw interpretation method2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1697684Effect on JNK expression in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
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).
AID1767414Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as decrease in PGE2 in tumor tissue at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation by West
AID1172642Inhibition of cell proliferation of rat C6 cells assessed as cell viability after 72 hrs by sulforhodamine B assay2014Bioorganic & medicinal chemistry letters, Nov-15, Volume: 24, Issue:22
Bioactive triterpenoid saponins and phenolic compounds against glioma cells.
AID1697800Induction of DNA damage in human DBTRG-05MG cells assessed as ATM phosphorylation at ser1981 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1079942Steatosis, proven histopathologically. Value is number of references indexed. [column 'STEAT' in source]
AID1888669Antiproliferative activity against human LN-229 cells assessed as reduction in cell viability at 80 uM up to 7 days by incucyte live cell assay2022Bioorganic & medicinal chemistry, 01-01, Volume: 53Novel structural-related analogs of PFI-3 (SRAPs) that target the BRG1 catalytic subunit of the SWI/SNF complex increase the activity of temozolomide in glioblastoma cells.
AID1697617Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G0/G1 phase at 206 uM after 24 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 57.81 +/- 1.9%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1876525Induction of apoptosis in human U-251 cells assessed as late apoptotic cells at 6 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb =1.31 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1497081Cytotoxicity against human MCF7 cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1736284Antiproliferative activity against human U251MG cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID667958Growth inhibition of human Hs683 cells after 72 hrs by MTT assay2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID1697733Induction of apoptosis in rat RG2 cells assessed as increase in apoptotic body formation at 206 uM after 48 hrs by TUNEL assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID661466Cytotoxicity against mouse astrocytes assessed as decrease in cell viability at 1 to 100 uM after 48 hrs by propidium iodide incorporation assay2012Journal of medicinal chemistry, Mar-08, Volume: 55, Issue:5
Oxaphosphinanes: new therapeutic perspectives for glioblastoma.
AID1851872Cytotoxicity against human GSC#83 cells assessed as reduction in cell viability at 5 to 50 uM incubated for 24 hrs by MTS assay
AID1876524Induction of apoptosis in human U-251 cells assessed as early apoptotic cells at 6 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb =1.96 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
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).
AID348241Antitumor activity against mouse B16F10 cells xenografted in C57BL/6 mouse assessed as growth inhibition at 50 mg/kg/day, po after 14 days relative to control2009Journal of medicinal chemistry, Jan-22, Volume: 52, Issue:2
Discovery of the Poly(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide (ABT-888) for the treatment of cancer.
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).
AID1736311Cytotoxicity against Wistar rat astrocytes assessed as cell growth inhibition at 100 uM measured after 72 hrs by SRB assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID102166Cytotoxic potentiation of Topotecan (TP) by the compound in (human colorectal cancer LoVo cell line2002Journal of medicinal chemistry, Nov-07, Volume: 45, Issue:23
Novel tricyclic poly(ADP-ribose) polymerase-1 inhibitors with potent anticancer chemopotentiating activity: design, synthesis, and X-ray cocrystal structure.
AID282550Antitumor activity against human NCI leukemia cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1054218Induction of apoptosis in human SK-MEL-30 cells assessed as apoptotic/necrotic cells at 100 uM after 48 hrs by APC-labeled annexin V and 7-AAD staining-based flow cytometric analysis relative to control2013European journal of medicinal chemistry, , Volume: 70Synthesis and evaluation of N-acylamino acids derivatives of triazenes. Activation by tyrosinase in human melanoma cell lines.
AID1777597Stability of the compound in potassium phosphate buffer at pH 7.4 assessed as half life2021Bioorganic & medicinal chemistry letters, 10-15, Volume: 50Model studies towards prodrugs of the glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) containing a diazo precursor.
AID1697344Acute toxicity in Balb/c mouse xenografted with SW-620 cells assessed as mean weight loss at 50 mg/kg, po qd administered for 5 days measured on day 6 relative to control2020Journal of medicinal chemistry, 10-08, Volume: 63, Issue:19
Discovery of SK-575 as a Highly Potent and Efficacious Proteolysis-Targeting Chimera Degrader of PARP1 for Treating Cancers.
AID282485Half life of hydrolysis in phosphate buffer at pH 7.42004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1697465Cytotoxicity against rat RG2 cells assessed as reduction in cell viability after 72 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1683790Antitumor activity against human NCI-H209 cells xenografted in BALB/c nude mouse assessed as tumor relapse at 50 mg/kg, po QD for 5 consecutive days at start of study and again 14 days later and measured after 48 days2020Journal of medicinal chemistry, 12-24, Volume: 63, Issue:24
Discovery of Pamiparib (BGB-290), a Potent and Selective Poly (ADP-ribose) Polymerase (PARP) Inhibitor in Clinical Development.
AID1316634Drug level in Sprague-Dawley rat brain at 10 mg/kg, iv after 30 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID73182In vitro cytotoxicity against human lymphoblast cell line (GM892 A).1995Journal of medicinal chemistry, Apr-28, Volume: 38, Issue:9
Antitumor imidazotetrazines. 32. Synthesis of novel imidazotetrazinones and related bicyclic heterocycles to probe the mode of action of the antitumor drug temozolomide.
AID1876510Induction of apoptosis in human U-87 MG cells assessed as viable cells at 1.20 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb = 96.84 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1172641Inhibition of cell proliferation of human U251 cells assessed as cell viability after 72 hrs by sulforhodamine B assay2014Bioorganic & medicinal chemistry letters, Nov-15, Volume: 24, Issue:22
Bioactive triterpenoid saponins and phenolic compounds against glioma cells.
AID94799Cytotoxic effect in K562 cells2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
Synthesis of pyrimidinopyridine-triazene conjugates targeted to abl tyrosine kinase.
AID1736297Antitumor activity against C57BL/6 mouse GL261 cells xenografted in C57BL/6 mouse assessed as reduction in tumor volume at 20 mg/kg, ip administered once daily for 10 days starting from 7 days post tumor cell inoculation and measured on day 172020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID1595390Induction of morphological changes in mouse GL261 cells assessed as increase in cell circularity at 10 uM incubated for 48 hrs by Hoechst 33258 staining based fluorescence microscopic analysis2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1316653Ratio of AUC in human brain to plasma by reversed-phase HPLC analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1763423Cytotoxicity against human HL-60 cells by MTT assay2021Bioorganic & medicinal chemistry letters, 07-01, Volume: 43Chemical modifications of ergostane-type triterpenoids from Antrodia camphorata and their cytotoxic activities.
AID1888353Induction of DNA damage in human T98G cells assessed as increase in gammaH2AX foci number and size at 94 uM measured after 96 hrs by DAPI staining based immunofluorescence assay
AID1079939Cirrhosis, proven histopathologically. Value is number of references indexed. [column 'CIRRH' in source]
AID1308011Growth inhibition of human U87MG cells at 50 uM after 72 hrs by MTS assay relative to control2016Journal of medicinal chemistry, 05-26, Volume: 59, Issue:10
Lead Optimization of 2-Phenylindolylglyoxylyldipeptide Murine Double Minute (MDM)2/Translocator Protein (TSPO) Dual Inhibitors for the Treatment of Gliomas.
AID1763424Cytotoxicity against human U-251 cells by MTT assay2021Bioorganic & medicinal chemistry letters, 07-01, Volume: 43Chemical modifications of ergostane-type triterpenoids from Antrodia camphorata and their cytotoxic activities.
AID7471Potentiation factor at 50% growth inhibition (IC50 of compound to that of compound along with PARP inhibitor) in experiment 22000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
AID282552Antitumor activity against human NCI colon cancer cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1167096Inhibition of cell proliferation of human WI38 cells by CCK8 assay2014Journal of medicinal chemistry, Oct-23, Volume: 57, Issue:20
Inhibition of cancer-associated mutant isocitrate dehydrogenases: synthesis, structure-activity relationship, and selective antitumor activity.
AID231797Ratio of cytotoxicity against Raji cells to that of GM892 A cells.1995Journal of medicinal chemistry, Apr-28, Volume: 38, Issue:9
Antitumor imidazotetrazines. 32. Synthesis of novel imidazotetrazinones and related bicyclic heterocycles to probe the mode of action of the antitumor drug temozolomide.
AID1633573Toxicity in human Capan1 cells xenografted Balb athymic Nu/Nu mouse assessed as maximum weight loss at 62.5 mg/kg, iv qd for 5 days2019ACS medicinal chemistry letters, Apr-11, Volume: 10, Issue:4
Discovery of Stereospecific PARP-1 Inhibitor Isoindolinone NMS-P515.
AID1474441Anti-tumor activity against human MX1 cells xenografted in BALB/c mouse assessed as tumor growth inhibition at 50 mg/kg, po administered once daily via gavage for 5 days measured after 11 days relative to control2017European journal of medicinal chemistry, May-26, Volume: 132Discovery of 2-substituted 1H-benzo[d]immidazole-4-carboxamide derivatives as novel poly(ADP-ribose)polymerase-1 inhibitors with in vivo anti-tumor activity.
AID1537888Cytotoxicity against P-gp knockout patient-isolated primary glioblastoma cells derived neurospheres assessed as increase in LDH release at 50 uM measured after 48 hrs by spectrophotometric method2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1697688Effect on p38 expression in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697628Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G2/M phase at 206 uM after 48 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.04 +/- 0.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1769430Cytotoxicity against human HaCaT cells assessed as reduction in cell viability incubated for 72 hrs by MTS assay2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Sulfur Analogues of Tyrosine in the Development of Triazene Hybrid Compounds: A New Strategy against Melanoma.
AID1064453Growth inhibition of human MDA-MB-436 cells after 72 hrs by SRB assay2014Bioorganic & medicinal chemistry letters, Jan-15, Volume: 24, Issue:2
Novel PARP-1 inhibitors based on a 2-propanoyl-3H-quinazolin-4-one scaffold.
AID1697467Cytotoxicity against rat CTX TNA2 cells assessed as reduction in cell viability after 48 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID766233Chemosensitization of human A2780 cells after 5 days by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1767413Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as increase in E-cadherin in tumor tissue at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation b
AID1425998Cytotoxicity against MDCK cells expressing carbonic anhydrase 9 assessed as reduction in cell viability after 72 hrs under normoxic condition by Alamar blue assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID282556Antitumor activity against human NCI renal cancer cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1595381Cytotoxicity against mouse GL261 cells assessed as reduction in cell viability incubated for 48 hrs by MTS assay2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1546668Cytotoxicity against human SW480 cells incubated for 24 hrs by MTT assay2020Journal of natural products, 01-24, Volume: 83, Issue:1
Antcamphorols A-K, Cytotoxic and ROS Scavenging Triterpenoids from
AID1542800Cytotoxicity against human U251MG cells assessed as reduction in cell survival measured after 72 hrs by MTT assay2019Bioorganic & medicinal chemistry, 05-15, Volume: 27, Issue:10
Chemical modifications of imidazole-containing alkoxyamines increase C-ON bond homolysis rate: Effects on their cytotoxic properties in glioblastoma cells.
AID52527Level of AGT activity in COLO 205 cell line2000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 8. Inhibition of O(6)-alkylguanine-DNA alkyltransferase by O(6)-alkenyl-, O(6)-cycloalkenyl-, and O(6)-(2-oxoalkyl)guanines and potentiation of temozolomide cytotoxicity in vitro by O(6)-(1-cyclopentenylmethyl)guanine.
AID1697466Cytotoxicity against rat CTX TNA2 cells assessed as reduction in cell viability after 24 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316633Plasma concentration in Sprague-Dawley rat plasma at 10 mg/kg, iv after 180 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
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).
AID1697640Induction of cell cycle arrest in human DBTRG-05MG cells assessed as p21 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID282487Half life of hydrolysis in deuterated phosphate buffer at pD 7.82004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1697778Induction of apoptosis in rat RG2 cells assessed as phosphorylated PARP expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID474430Antitumor activity against mouse B16F10 cells transplanted in C57BL/6 mouse assessed as tumor volume at 50 mg/kg/day, po administered from day 6 to 10 post transplantation measured on day 18 relative to control2010Journal of medicinal chemistry, Apr-22, Volume: 53, Issue:8
Optimization of phenyl-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase inhibitors: identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (A-966492), a highly potent and efficacious inhibitor.
AID1683795Antitumor activity against human NCI-H209 cells xenografted in BALB/c nude mouse assessed as inhibition of tumor growth at 50 mg/kg, po QD for 5 consecutive days at start of study and again 14 days later2020Journal of medicinal chemistry, 12-24, Volume: 63, Issue:24
Discovery of Pamiparib (BGB-290), a Potent and Selective Poly (ADP-ribose) Polymerase (PARP) Inhibitor in Clinical Development.
AID1079948Times to onset, minimal and maximal, observed in the indexed observations. [column 'DELAI' in source]
AID1426003Cytotoxicity against carbonic anhydrase 9 knockdown MDCK cells assessed as reduction in cell viability incubated for 24 hrs measured after 7 days under hypoxic condition by methylene blue staining based clonogenic survival assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1054217Induction of apoptosis in human M8 cells assessed as apoptotic/necrotic cells at 50 to 100 uM after 48 hrs by APC-labeled annexin V and 7-AAD staining-based flow cytometric analysis2013European journal of medicinal chemistry, , Volume: 70Synthesis and evaluation of N-acylamino acids derivatives of triazenes. Activation by tyrosinase in human melanoma cell lines.
AID1623800Toxicity in nude BALB/c mouse xenografted with human U87MG cells assessed as reduction in body weight at 50 mg/kg, ip administered for 5 days/week for two cycles2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
AID1416899Growth inhibition of MGMT-transfected human SNB19 cells expressing MGMT after 7 days by MTT assay
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).
AID1167095Inhibition of neurosphere formation of mouse BXD3752 cells by CCK8 assay2014Journal of medicinal chemistry, Oct-23, Volume: 57, Issue:20
Inhibition of cancer-associated mutant isocitrate dehydrogenases: synthesis, structure-activity relationship, and selective antitumor activity.
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).
AID1763426Cytotoxicity against human MCF7 cells by MTT assay2021Bioorganic & medicinal chemistry letters, 07-01, Volume: 43Chemical modifications of ergostane-type triterpenoids from Antrodia camphorata and their cytotoxic activities.
AID282555Antitumor activity against human NCI ovarian carcinoma cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1497079Cytotoxicity against human U87 cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1697709Inhibition of JNK phosphorylation at Thr183/Tyr185 residues in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1167093Inhibition of neurosphere formation of human BT142 cells by CCK8 assay2014Journal of medicinal chemistry, Oct-23, Volume: 57, Issue:20
Inhibition of cancer-associated mutant isocitrate dehydrogenases: synthesis, structure-activity relationship, and selective antitumor activity.
AID1317836Antiproliferative activity against drug-resistant human T98G cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1317832Antiproliferative activity against drug-sensitive human Hs683 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1697707Inhibition of AKT phosphorylation at S473 residues in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID667956Growth inhibition of human U373 cells after 72 hrs by MTT assay2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID1876492Antiproliferative activity against rat C6 cells assessed as inhibition of cell proliferation measured after 72 hrs by CCK8 assay2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1697750Induction of apoptosis in human DBTRG-05MG cells assessed as phosphorylated caspace-9 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697610Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G2/M phase at 206 uM after 12 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.04 +/- 0.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697753Induction of apoptosis in human DBTRG-05MG cells assessed as cleaved caspace-3 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697581Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G0/G1 phase at 206 uM after 24 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 47.16 +/- 1.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1497080Cytotoxicity against human SH-SY5Y cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1545832Antiproliferative activity against human GBM2 cells assessed as reduction in cell viability after 72 hrs by MTT assay2019European journal of medicinal chemistry, Dec-01, Volume: 1831,2,3-Triazole-containing hybrids as potential anticancer agents: Current developments, action mechanisms and structure-activity relationships.
AID1425999Cytotoxicity against carbonic anhydrase 9 knockdown MDCK cells assessed as reduction in cell viability after 72 hrs under hypoxic condition by Alamar blue assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1895190Inhibition of sulfatase in human T98G cells assessed as reduction in fluorescence intensity at 25 uM using 2-(2-Morpholinoethyl)-6-(((2S,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)-1H-benzo[de]-isoquinoline-1,3(2H)-dione a2021Journal of medicinal chemistry, 06-24, Volume: 64, Issue:12
New Protocol-Guided Exploitation of a Lysosomal Sulfatase Inhibitor to Suppress Cell Growth in Glioblastoma Multiforme.
AID1697461Cytotoxicity against human DBTRG-05MG cells assessed as reduction in cell viability after 48 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1317834Antiproliferative activity against drug-resistant human U251 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1697573Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at S phase at 206 uM after 12 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 22.76 +/- 1.56%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1623802Anti-angiogenic activity in tumor of human U87MG cells xenografted in nude BALB/c mouse assessed as reduction in number of blood vessels at 50 mg/kg, ip administered for 14 days by hematoxylin and eosin-staining based light microscopy2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
AID1888667Induction of cell death in human MT330 cells assessed as reduction in cell viability at 50 uM after 3 days by ELISA2022Bioorganic & medicinal chemistry, 01-01, Volume: 53Novel structural-related analogs of PFI-3 (SRAPs) that target the BRG1 catalytic subunit of the SWI/SNF complex increase the activity of temozolomide in glioblastoma cells.
AID1697618Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at S phase at 206 uM after 24 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.15 +/- 2.44%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697469Toxicity in BALB/c mouse xenografted with DBTRG-05MG cells assessed as reduction in body weight at 5 mg/kg, ip after 5 days measured every 2 days2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID282558Antitumor activity against human NCI breast cancer cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1876563Antitumor activity against human U-87 MG cells xenografted in BALB/c mouse assessed as decrease in tumor volume at 10 mg/kg,ip bid for 1 month2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1262128Antiproliferative activity against human U118MG cells assessed as inhibition of cell growth at 100 uM after 72 hrs by WST1 assay2015European journal of medicinal chemistry, Nov-13, Volume: 105Design and synthesis of 2-oxindole based multi-targeted inhibitors of PDK1/Akt signaling pathway for the treatment of glioblastoma multiforme.
AID1851874Cytotoxicity against human U-251 cells assessed as reduction in cell viability at 10 uM incubated for 24 hrs by MTS assay
AID282554Antitumor activity against NCI human melanoma cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1633569Antitumor activity against human Capan1 cells xenografted in Balb athymic Nu/Nu mouse assessed as maximal tumor growth inhibition at 62.5 mg/kg, iv qd for 5 days2019ACS medicinal chemistry letters, Apr-11, Volume: 10, Issue:4
Discovery of Stereospecific PARP-1 Inhibitor Isoindolinone NMS-P515.
AID668219Antiproliferative activity against human A172 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Jul-01, Volume: 22, Issue:13
New substituted 4H-chromenes as anticancer agents.
AID1767417Toxicity in rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as body weight change at 50 mg/kg, ip twice a day administered for 13 days of tumor implantation
AID766225Chemosensitization of human HCT116 cells after 5 days by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1316632Plasma concentration in Sprague-Dawley rat plasma at 10 mg/kg, iv after 90 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1851859Cytotoxicity against human T98G cells assessed as reduction in cell viability at 5 to 50 uM incubated for 24 hrs by MTS assay
AID1697627Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at S phase at 206 uM after 48 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.15 +/- 2.44%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697772Induction of apoptosis in rat RG2 cells assessed as Bcl2 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1254863Tumoricidal effect in patient derived GBM 047T cells assessed as reduction in neurosphere formation at 20 uM after 1 to 2 weeks by 3D tumor clonogenic assay2015European journal of medicinal chemistry, Oct-20, Volume: 103Discovery of potent and selective cytotoxic activity of new quinazoline-ureas against TMZ-resistant glioblastoma multiforme (GBM).
AID1697582Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at S phase at 206 uM after 24 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 22.76 +/- 1.56%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID386713Antitumor activity in human SW620 cells xenografted mouse assessed as days to double the relative tumor volume at 50 mg/kg, po once daily for 5 consecutive days2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
4-[3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin-1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1.
AID441949Antitumor activity against mouse B16F10 cells implanted in C57BL/6 mouse at 50 mg/kg, po QD for 5 days measured on day 122009Journal of medicinal chemistry, Nov-12, Volume: 52, Issue:21
Synthesis and evaluation of a new generation of orally efficacious benzimidazole-based poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors as anticancer agents.
AID348240Antitumor activity against mouse B16F10 cells xenografted in C57BL/6 mouse assessed as tumor volume at 50 mg/kg/day, po after 14 days2009Journal of medicinal chemistry, Jan-22, Volume: 52, Issue:2
Discovery of the Poly(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide (ABT-888) for the treatment of cancer.
AID540210Clearance in human after iv administration2008Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 36, Issue:7
Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds.
AID667962Selectivity index, ratio of of IC50 for mouse astrocytes to IC50 for human Hs683 cells2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID712153Prodrug conversion in citrate-phosphate buffer at pH 8 assessed as pseudo-first-order rate constant2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1729709Inhibition of colony formation in human GSC-18 cells assessed as tumor spheres damage at 15 uM after 2 weeks by crystal violet staining based assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Structures/cytotoxicity/selectivity relationship of natural steroidal saponins against GSCs and primary mechanism of tribulosaponin A.
AID1809515Cytotoxicity against human U-87 MG cells assessed as inhibition of cell growth measured after 48 hrs by MTT assay2021Bioorganic & medicinal chemistry letters, 11-01, Volume: 51Non-alkylator anti-glioblastoma agents induced cell cycle G2/M arrest and apoptosis: Design, in silico physicochemical and SAR studies of 2-aminoquinoline-3-carboxamides.
AID1425987Binding affinity to recombinant human carbonic anhydrase 1 after 15 mins by stopped-flow CO2 hydration assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1079931Moderate liver toxicity, defined via clinical-chemistry results: ALT or AST serum activity 6 times the normal upper limit (N) or alkaline phosphatase serum activity of 1.7 N. Value is number of references indexed. [column 'BIOL' in source]
AID7467Potentiation of growth inhibition of A2780 cells by compound alone in experiment 22000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1876512Induction of apoptosis in human U-87 MG cells assessed as late apoptotic cells at 1.20 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb = 0.42 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1697600Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at S phase at 206 uM after 6 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.15 +/- 2.44%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1079940Granulomatous liver disease, proven histopathologically. Value is number of references indexed. [column 'GRAN' in source]
AID1317833Antiproliferative activity against drug-resistant human U373 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1697591Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at S phase at 206 uM after 48 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 22.76 +/- 1.56%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1317835Antiproliferative activity against drug-resistant human U87 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1697706Effect on AKT expression in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
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.
AID1697601Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G2/M phase at 206 uM after 6 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.04 +/- 0.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697729Induction of apoptosis in human DBTRG-05MG cells assessed as increase in DNA fragmentation at 206 uM after 48 hrs by TUNEL assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1426000Cytotoxicity against MDCK cells expressing carbonic anhydrase 9 assessed as reduction in cell viability after 72 hrs under hypoxic condition by Alamar blue assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1054220Induction of apoptosis in human MNT1 cells assessed as apoptotic/necrotic cells at 100 uM after 48 hrs by APC-labeled annexin V and 7-AAD staining-based flow cytometric analysis relative to control2013European journal of medicinal chemistry, , Volume: 70Synthesis and evaluation of N-acylamino acids derivatives of triazenes. Activation by tyrosinase in human melanoma cell lines.
AID475198Antitumor activity against mouse B16F10 cells transplanted in C57BL/6 mouse assessed as tumor volume at 50 mg/kg/day, po administered from day 6 to 10 post transplantation measured on day 16 relative to control2010Journal of medicinal chemistry, Apr-22, Volume: 53, Issue:8
Optimization of phenyl-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase inhibitors: identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (A-966492), a highly potent and efficacious inhibitor.
AID454060Cytotoxicity against human HCT116 cells after 4 days2009Bioorganic & medicinal chemistry, Nov-01, Volume: 17, Issue:21
Synthesis of isoquinolinone-based tetracycles as poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors.
AID1507125Antiproliferative activity in human A431 Cells after 72 hrs by SRB assay2017European journal of medicinal chemistry, Aug-18, Volume: 136Designing multi-targeted agents: An emerging anticancer drug discovery paradigm.
AID1830962Antitumor activity against human U-87MG cells xenografted in BALB/c mouse assessed as reduction in tumor volume at 30 mg/kg, po administered once daily for 5 days and measured on day 8 by MRI scanner (Rvb = 202 +/- 120.4 mm3)
AID1697608Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G0/G1 phase at 206 uM after 12 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 57.81 +/- 1.9%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1537861Cytotoxicity against patient-isolated primary glioblastoma cells derived adherent cells assessed as increase in LDH release at 50 uM measured after 48 hrs by spectrophotometric method2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1693365Antitumour activity against human U251 cells xenografted in NCr athymic nude nu/nu mouse assessed as decrease in tumour growth at 50 mg/kg once in a week2021Bioorganic & medicinal chemistry, 01-01, Volume: 29The 3-D conformational shape of N-naphthyl-cyclopenta[d]pyrimidines affects their potency as microtubule targeting agents and their antitumor activity.
AID1825321Cytotoxicity against human SH-SY5Y cells assessed as cell viability using raw interpretation method2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1697809Induction of DNA damage in rat RG2 cells assessed as H2A.X phosphorylation at ser139 at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID474425Antitumor activity against mouse B16F10 cells transplanted in C57BL/6 mouse assessed as tumor growth inhibition at 50 mg/kg/day, po administered from day 6 to 10 post transplantation measured on day 12 relative to control2010Journal of medicinal chemistry, Apr-22, Volume: 53, Issue:8
Optimization of phenyl-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase inhibitors: identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (A-966492), a highly potent and efficacious inhibitor.
AID1697662Induction of cell cycle arrest in rat RG2 cells assessed as CDK4 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1729711Inhibition of colony formation in human GSC-18 cells assessed as decrease in number of tumor spheres at 15 uM after 2 weeks by crystal violet staining based assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Structures/cytotoxicity/selectivity relationship of natural steroidal saponins against GSCs and primary mechanism of tribulosaponin A.
AID7469Potentiation of growth inhibition of A2780 cells along with 10 uM NU1085 in experiment 22000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
AID1583449Permeability in dog MDCK2 cells assessed as reduction in barrier integrity measured within 7 hrs by transendothelial electric resistance measurement assay2020Journal of medicinal chemistry, 02-27, Volume: 63, Issue:4
Identification of Parthenolide Dimers as Activators of Pyruvate Kinase M2 in Xenografts of Glioblastoma Multiforme in Vivo.
AID1825342Induction of morphological changes in human U-87 MG cells assessed as membrane blebbing at 1408 uM measured after 72 hrs by phase contrast microscopy2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1876523Induction of apoptosis in human U-251 cells assessed as viable cells at 6 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb = 96.22 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1697641Induction of cell cycle arrest in human DBTRG-05MG cells assessed as CDK4 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316635Drug level in Sprague-Dawley rat brain at 10 mg/kg, iv after 90 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID712156Cytotoxicity against human U251 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1546666Cytotoxicity against human U251 cells incubated for 24 hrs by MTT assay2020Journal of natural products, 01-24, Volume: 83, Issue:1
Antcamphorols A-K, Cytotoxic and ROS Scavenging Triterpenoids from
AID1697754Induction of apoptosis in human DBTRG-05MG cells assessed as phosphorylated PARP expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID766232Chemosensitization of MMR-deficient human A2780/CP70 cells after 5 days by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1317842Antiproliferative activity against human MES-SA cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1317839Antiproliferative activity against patient-derived TMZ-resistant GBM 031810 cells over-expressing MGMT after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1697642Induction of cell cycle arrest in human DBTRG-05MG cells assessed as CyclinD1 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697728Induction of apoptosis in human DBTRG-05MG cells assessed as increase in chromatin condensation at 206 uM after 48 hrs by TUNEL assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697660Induction of cell cycle arrest in rat RG2 cells assessed as p53 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1054219Induction of apoptosis in human SK-MEL-30 cells assessed as apoptotic/necrotic cells at 50 uM after 48 hrs by APC-labeled annexin V and 7-AAD staining-based flow cytometric analysis relative to control2013European journal of medicinal chemistry, , Volume: 70Synthesis and evaluation of N-acylamino acids derivatives of triazenes. Activation by tyrosinase in human melanoma cell lines.
AID712157Prodrug conversion in citrate-phosphate buffer at pH 8 assessed as half life of compound2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1079932Highest frequency of moderate liver toxicity observed during clinical trials, expressed as a percentage. [column '% BIOL' in source]
AID1697665Induction of cell cycle arrest in rat RG2 cells assessed as CyclinA expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID667957Growth inhibition of human T98G cells after 72 hrs by MTT assay2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID1537887Cytotoxicity against patient-isolated primary glioblastoma cells derived adherent cells assessed as reduction in cell viability at 50 uM measured after 72 hrs by chemiluminescence-based assay2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1623804In-vivo inhibition of cell proliferation in human U87MG cells xenografted in BALB/c nude mouse at 50 mg/kg, ip after 14 days by Ki-67b-staining based assay2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
AID1497085Cytotoxicity against human L02 cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1697464Cytotoxicity against rat RG2 cells assessed as reduction in cell viability after 48 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316647Antitumor activity against human U87MG cells orthotopically xenografted in Harlan nude mouse brain assessed as increase in mouse survival at 50 umol/kg, iv administered once daily for 5 days2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1240666Antitumor activity against human Capan1 cells xenografted in athymic nu/nu mouse assessed as tumor growth inhibition at 50 mg/kg administered for 5 days2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
Discovery of 2-[1-(4,4-Difluorocyclohexyl)piperidin-4-yl]-6-fluoro-3-oxo-2,3-dihydro-1H-isoindole-4-carboxamide (NMS-P118): A Potent, Orally Available, and Highly Selective PARP-1 Inhibitor for Cancer Therapy.
AID1736280Selectivity index, ratio of CC50 for Wistar rat astrocytes to EC50 for human U-251 MG cells2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID1250159Cytotoxicity against human Glioma cells (HF2885) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
AID1079944Benign tumor, proven histopathologically. Value is number of references indexed. [column 'T.BEN' in source]
AID101599Inhibitory concentration of compound against Mer+ breast tumor cell line MCF-72000Bioorganic & medicinal chemistry letters, Oct-16, Volume: 10, Issue:20
Design and synthesis of a novel water soluble benzotetrazepinone.
AID1830966Antitumor activity against human U-87MG cells xenografted in BALB/c mouse assessed as reduction in tumor volume at 30 mg/kg, po administered once daily for 5 days and measured on day 32 by MRI scanner
AID1416901Growth inhibition of MGMT-transfected human U373 cells expressing MGMT after 7 days by MTT assay
AID209544In vitro cytotoxicity against mouse TLX5 lymphoma cells1995Journal of medicinal chemistry, Apr-28, Volume: 38, Issue:9
Antitumor imidazotetrazines. 32. Synthesis of novel imidazotetrazinones and related bicyclic heterocycles to probe the mode of action of the antitumor drug temozolomide.
AID668217Antiproliferative activity against human U87 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Jul-01, Volume: 22, Issue:13
New substituted 4H-chromenes as anticancer agents.
AID1697779Induction of apoptosis in rat RG2 cells assessed as cleaved PARP expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1305718Antitumor activity against MCT1 expressing mouse GL261 cells coexpressing luc2 syngrafted in C57BL/6 mouse assessed as tumor growth inhibition at 20 mg/kg, ip qd2016Bioorganic & medicinal chemistry letters, 07-15, Volume: 26, Issue:14
Coumarin carboxylic acids as monocarboxylate transporter 1 inhibitors: In vitro and in vivo studies as potential anticancer agents.
AID1623803Anti-angiogenic activity in tumor of human U87MG cells xenografted in nude BALB/c mouse assessed as reduction in CD31 staining at 50 mg/kg, ip administered for 14 days by immunohistochemistry2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
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).
AID1250157Cytotoxicity against human Glioma cells (HF2790) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
AID1545833Antiproliferative activity against human GBM95 cells assessed as reduction in cell viability after 72 hrs by MTT assay2019European journal of medicinal chemistry, Dec-01, Volume: 1831,2,3-Triazole-containing hybrids as potential anticancer agents: Current developments, action mechanisms and structure-activity relationships.
AID1697619Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G2/M phase at 206 uM after 24 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.04 +/- 0.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID712164Cytotoxicity against human DLD1 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1200320Antiproliferative activity against human MES-SA/Dx5 cells assessed as reduction in cell viability after 48 hrs by MTT assay2015Journal of medicinal chemistry, Mar-12, Volume: 58, Issue:5
Activity of 2-aryl-2-(3-indolyl)acetohydroxamates against drug-resistant cancer cells.
AID1595387Induction of apoptosis in human SH-SY5Y cells assessed as increase in early apoptotic cells at 10 uM incubated for 48 hrs by annexinV-PE/7-AAD staining based flow cytometric analysis2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1079947Comments (NB not yet translated). [column 'COMMENTAIRES' in source]
AID1416898Growth inhibition of vehicle-transfected human SNB19 cells after 7 days by MTT assay
AID1054221Induction of apoptosis in human MNT1 cells assessed as apoptotic/necrotic cells at 50 uM after 48 hrs by APC-labeled annexin V and 7-AAD staining-based flow cytometric analysis relative to control2013European journal of medicinal chemistry, , Volume: 70Synthesis and evaluation of N-acylamino acids derivatives of triazenes. Activation by tyrosinase in human melanoma cell lines.
AID1876564Antitumor activity against human U-87 MG cells xenografted in BALB/c mouse assessed as decrease in tumor weight at 10 mg/kg,ip bid for 1 month2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1497083Cytotoxicity against human A549 cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1767410Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as reduction tumor weight at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation
AID1079946Presence of at least one case with successful reintroduction. [column 'REINT' in source]
AID673621Antitumor activity against B16F10 tumor xenografted in C57BL/6 mouse assessed as tumor growth inhibition at 50 mg/kg/day, po administered QD from day 5 to day 10 post-tumor implantation2012Bioorganic & medicinal chemistry, Aug-01, Volume: 20, Issue:15
Discovery and SAR of orally efficacious tetrahydropyridopyridazinone PARP inhibitors for the treatment of cancer.
AID1317841Antiproliferative activity against patient-derived paclitaxel-resistant NCI-H2073 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1771786Cytotoxicity against human A 172 cells assessed as cell viability measured after 96 hrs by XTT assay2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1079934Highest frequency of acute liver toxicity observed during clinical trials, expressed as a percentage. [column '% AIGUE' in source]
AID766231Chemosensitization of human A2780 cells after 5 days by MTT assay in presence of MGMT inactivator PaTrin22013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID683689Cytotoxicity against human U138MG cells incubated for 48 hrs by MTT assay2012European journal of medicinal chemistry, Nov, Volume: 57Selective cytotoxicity and apoptosis induction in glioma cell lines by 5-oxygenated-6,7-methylenedioxycoumarins from Pterocaulon species.
AID1697713Inhibition of p38 phosphorylation at Tyr182 residues in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1769428Cytotoxicity against mouse B16-F10 cells assessed as reduction in cell viability incubated for 72 hrs by MTS assay2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Sulfur Analogues of Tyrosine in the Development of Triazene Hybrid Compounds: A New Strategy against Melanoma.
AID712167Cytotoxicity against human A2780 cells after 5 days by MTT assay in presence of 10 uM MGMT inactivator Patrin22012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1697666Induction of cell cycle arrest in rat RG2 cells assessed as CyclinB1 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1335506Cytotoxicity against Wistar rat C6 cells assessed as decrease in cell viability at 100 uM after 72 hrs by MTT assay relative to control2016European journal of medicinal chemistry, Nov-29, Volume: 124Thiazolidin-4-ones from 4-(methylthio)benzaldehyde and 4-(methylsulfonyl)benzaldehyde: Synthesis, antiglioma activity and cytotoxicity.
AID1697463Cytotoxicity against rat RG2 cells assessed as reduction in cell viability after 24 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1809101Cytotoxicity effect against human LN-229 cells assessed as cell growth inhibition incubated for 48 hrs by trypan blue exclusion assay2021Journal of medicinal chemistry, 08-12, Volume: 64, Issue:15
Synthesis and Preclinical Validation of Novel Indole Derivatives as a GPR17 Agonist for Glioblastoma Treatment.
AID1317838Antiproliferative activity against patient-derived TMZ-sensitive GBM 090909 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1697643Induction of cell cycle arrest in human DBTRG-05MG cells assessed as CDK2 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID386714Toxicity in human SW620 cells xenografted mouse assessed as maximum mean body weight loss after 3 days2008Journal of medicinal chemistry, Oct-23, Volume: 51, Issue:20
4-[3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin-1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1.
AID712162Cytotoxicity against human PANC1 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID52525Cytotoxicity of compound in COLO 205 cell line2000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 8. Inhibition of O(6)-alkylguanine-DNA alkyltransferase by O(6)-alkenyl-, O(6)-cycloalkenyl-, and O(6)-(2-oxoalkyl)guanines and potentiation of temozolomide cytotoxicity in vitro by O(6)-(1-cyclopentenylmethyl)guanine.
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).
AID1595388Induction of apoptosis in human SH-SY5Y cells assessed as reduction in cell viability at 10 uM incubated for 48 hrs by annexinV-PE/7-AAD staining based flow cytometric analysis2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1697563Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G0/G1 phase at 206 uM after 6 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 47.16 +/- 1.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID672002Toxicity in C57BL/6 mouse bearing B16F10 tumor assessed as body weight loss at 50 mg/kg/day, po administered QD from day 5 to day 10 post-tumor implantation relative to vehicle-treated control2012Bioorganic & medicinal chemistry, Aug-01, Volume: 20, Issue:15
Discovery and SAR of orally efficacious tetrahydropyridopyridazinone PARP inhibitors for the treatment of cancer.
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).
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.
AID712168Cytotoxicity against human A2780 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1079933Acute liver toxicity defined via clinical observations and clear clinical-chemistry results: serum ALT or AST activity > 6 N or serum alkaline phosphatases activity > 1.7 N. This category includes cytolytic, choleostatic and mixed liver toxicity. Value is
AID661464Cytotoxicity against mouse astrocytes assessed as decrease in cell viability at 1 to 100 uM after 48 hrs by neutral red incorporation assay2012Journal of medicinal chemistry, Mar-08, Volume: 55, Issue:5
Oxaphosphinanes: new therapeutic perspectives for glioblastoma.
AID1876490Antiproliferative activity against human U-87 MG cells assessed as inhibition of cell proliferation measured after 72 hrs by CCK8 assay2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1697645Induction of cell cycle arrest in human DBTRG-05MG cells assessed as CyclinB1 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1767416Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as decrease in p-STAT3 in tumor tissue at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation by W
AID1254859Tumoricidal effect in patient derived GBM 464T cells assessed as reduction in neurosphere formation at 20 uM after 1 to 2 weeks by 3D tumor clonogenic assay2015European journal of medicinal chemistry, Oct-20, Volume: 103Discovery of potent and selective cytotoxic activity of new quinazoline-ureas against TMZ-resistant glioblastoma multiforme (GBM).
AID282553Antitumor activity against human NCI CNS cancer cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1771789Cytotoxicity against human MDA-MB-238 cells assessed as cell viability measured after 96 hrs by XTT assay2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1697749Induction of apoptosis in human DBTRG-05MG cells assessed as Bax expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID667960Selectivity index, ratio of IC50 for mouse astrocytes to IC50 for human U373 cells2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID1697752Induction of apoptosis in human DBTRG-05MG cells assessed as phosphorylated caspace-3 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1661315Antiproliferative activity against rat C6 cells assessed as inhibition of cell proliferation at 500 uM after 24 hrs by MTT assay relative to control2020ACS medicinal chemistry letters, Jul-09, Volume: 11, Issue:7
Antiglioma Activity of Aryl and Amido-Aryl Acetamidine Derivatives Targeting iNOS: Synthesis and Biological Evaluation.
AID1317837Antiproliferative activity against drug-resistant human A549 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1250160Cytotoxicity against human Glioma cells (HF3013) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
AID1736296Antitumor activity against C57BL/6 mouse GL261 cells xenografted in C57BL/6 mouse assessed as reduction in tumor volume at 20 mg/kg, po administered via gavage once daily for 10 days starting from 7 days post tumor cell inoculation and measured on day 172020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID1783998Cytotoxicity against cold atmospheric plasma (CAP)-treated human U-251MG cells assessed as decrease in cell viability treated with CAP for 30 secs followed by incubated with compound for 6 days by alamar blue assay2021European journal of medicinal chemistry, Nov-15, Volume: 224Enhanced pyrazolopyrimidinones cytotoxicity against glioblastoma cells activated by ROS-Generating cold atmospheric plasma.
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).
AID712159Cytotoxicity against human A549 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1767411Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as reduction tumor volume at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation
AID766228Ratio of IC50 for human A2780 cells to IC50 for human A2780 cells in presence of MGMT inactivator PaTrin22013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1316637Ratio of AUC in brain to plasma of Sprague-Dawley rat at 10 mg/kg, iv after 30 to 180 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1697807Induction of DNA damage in rat RG2 cells assessed as Chk2 phosphorylation at Thr68 at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID766226Chemosensitization of MGMT-deficient human SNB19 cells after 5 days by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1497084Cytotoxicity against human A375 cells assessed as reduction in cell viability after 72 hrs by MTT assay2018Bioorganic & medicinal chemistry letters, 07-15, Volume: 28, Issue:13
Design, synthesis and biological evaluation of artemisinin derivatives containing fluorine atoms as anticancer agents.
AID1537885Substrate activity at P-gp in patient-isolated primary glioblastoma cells derived neurospheres assessed as reduction in P-gp ATPase activity at 50 uM after 48 hrs by spectrophotometric method2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1825316Cytotoxicity against human SH-SY5Y cells assessed as cell viability at 2000 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1697609Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at S phase at 206 uM after 12 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 20.15 +/- 2.44%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697803Induction of DNA damage in rat RG2 cells assessed as ATR phosphorylation at ser428 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID766227Chemosensitization of human A2058 cells after 5 days by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1697730Induction of apoptosis in human DBTRG-05MG cells assessed as increase in apoptotic body formation at 206 uM after 48 hrs by TUNEL assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
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).
AID712158Prodrug conversion in citrate-phosphate buffer at pH 7.4 assessed as half life of compound2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID766230Chemoseinsitizatioin of MMR-deficieint humain A2780/CP70 cells after 5 days by MTT assay in preseince of MGMT iinactivator PaTriin22013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1316642Cytotoxicity against human U87MG cells assessed as inhibition of cell growth at 100 uM after 72 hrs by MTS assay2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID672001Toxicity in C57BL/6 mouse bearing B16F10 tumor assessed as body weight loss at 50 mg/kg/day, po administered QD from day 5 to day 9 post-tumor implantation relative to vehicle-treated control2012Bioorganic & medicinal chemistry, Aug-01, Volume: 20, Issue:15
Discovery and SAR of orally efficacious tetrahydropyridopyridazinone PARP inhibitors for the treatment of cancer.
AID1316655AUC in Sprague-Dawley rat plasma at 10 mg/kg, iv after 30 to 180 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1181988Cytotoxicity against human HeLa cells assessed as reduction in cell viability after 24 to 48 hrs by MTT assay2014Bioorganic & medicinal chemistry letters, Aug-01, Volume: 24, Issue:15
New benzothieno[3,2-d]-1,2,3-triazines with antiproliferative activity: synthesis, spectroscopic studies, and biological activity.
AID1553327Antiproliferative activity against patient-derived GBM cells assessed as cell viability after 48 hrs by 5-Ethynyl-2'-deoxyuridine incorporation assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
The synthesis of a novel Crizotinib heptamethine cyanine dye conjugate that potentiates the cytostatic and cytotoxic effects of Crizotinib in patient-derived glioblastoma cell lines.
AID1426002Cytotoxicity against MDCK cells expressing carbonic anhydrase 9 assessed as reduction in cell viability incubated for 24 hrs measured after 7 days under normoxic condition by methylene blue staining based clonogenic survival assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1316652Drug level in mouse brain assessed as compound level in soluble tissue fraction at 10 uM after 1 hr by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1623798Antitumor activity against human U87MG cells xenografted in BALB/c nude mouse assessed as tumor growth inhibition 50 mg/kg/day, ip administered for 14 days2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
AID1546667Cytotoxicity against human MCF7 cells incubated for 24 hrs by MTT assay2020Journal of natural products, 01-24, Volume: 83, Issue:1
Antcamphorols A-K, Cytotoxic and ROS Scavenging Triterpenoids from
AID15820Alkylating activity (0.1 mM) was determined at pH 7.4; expressed as OD (540n m)/mM of drug)2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
Synthesis of pyrimidinopyridine-triazene conjugates targeted to abl tyrosine kinase.
AID1426001Cytotoxicity against carbonic anhydrase 9 knockdown MDCK cells assessed as reduction in cell viability incubated for 24 hrs measured after 7 days under normoxic condition by methylene blue staining based clonogenic survival assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1426004Cytotoxicity against MDCK cells expressing carbonic anhydrase 9 assessed as reduction in cell viability incubated for 24 hrs measured after 7 days under hypoxic condition by methylene blue staining based clonogenic survival assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID668218Antiproliferative activity against human LN18 cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Jul-01, Volume: 22, Issue:13
New substituted 4H-chromenes as anticancer agents.
AID1486366Cytotoxicity against human MNT1 cells after 72 hrs by MTS assay2017Bioorganic & medicinal chemistry, 08-01, Volume: 25, Issue:15
The selective cytotoxicity of new triazene compounds to human melanoma cells.
AID1316631Plasma concentration in Sprague-Dawley rat plasma at 10 mg/kg, iv after 30 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID712161Cytotoxicity against human HCT116 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID588210Human drug-induced liver injury (DILI) modelling dataset from Ekins et al2010Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 38, Issue:12
A predictive ligand-based Bayesian model for human drug-induced liver injury.
AID1663800Antiproliferative activity against human patient derived GBM cells assessed as reduction in cell proliferation up to 100 uM measured after 48 hrs by Edu incorporation assay2020Bioorganic & medicinal chemistry letters, 07-15, Volume: 30, Issue:14
PARP inhibitor cyanine dye conjugate with enhanced cytotoxic and antiproliferative activity in patient derived glioblastoma cell lines.
AID1771779Induction of DNA methylation in human HCT-116 cells assessed as reduction of MGMT expression at 200 uM measured after 48 hrs by Western blot assay relative to control2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1697340Antitumor activity against SW-620 cells xenografted in Balb/c mouse assessed as tumor growth suppression at 50 mg/kg, po administered once daily for 5 days2020Journal of medicinal chemistry, 10-08, Volume: 63, Issue:19
Discovery of SK-575 as a Highly Potent and Efficacious Proteolysis-Targeting Chimera Degrader of PARP1 for Treating Cancers.
AID712163Cytotoxicity against human HT-29 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1888341Antiproliferative activity against human T98G cells assessed as cell growth inhibition incubated for 96 hrs by CellTiter-Glo luminescent cell viability assay
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).
AID1851871Cytotoxicity against human U-251 cells assessed as reduction in cell viability at 5 to 50 uM incubated for 24 hrs by MTS assay
AID1736285Antiproliferative activity against rat C6 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID7468Potentiation of growth inhibition of A2780 cells along with 10 uM NU1085 in experiment 12000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
AID1771792Induction of DNA methylation in human HCT-116 cells assessed as reduction of MGMT expression at 400 uM measured after 48 hrs by Western blot assay relative to control2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1697775Induction of apoptosis in rat RG2 cells assessed as cleaved caspace-9 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1595389Induction of apoptosis in human SH-SY5Y cells assessed as increase in late necrotic cells at 10 uM incubated for 48 hrs by annexinV-PE/7-AAD staining based flow cytometric analysis2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1623805Induction of apoptosis in human U87MG cells xenografted in BALB/c nude mouse assessed as caspase-3 levels at 50 mg/kg, ip after 14 days by immunohistochemistry2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
AID1697773Induction of apoptosis in rat RG2 cells assessed as Bax expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1851873Cytotoxicity against human T98G cells assessed as reduction in cell viability at 10 uM incubated for 24 hrs by MTS assay
AID1809102Cytotoxicity effect against human SNB-19 cells assessed as cell growth inhibition incubated for 48 hrs by trypan blue exclusion assay2021Journal of medicinal chemistry, 08-12, Volume: 64, Issue:15
Synthesis and Preclinical Validation of Novel Indole Derivatives as a GPR17 Agonist for Glioblastoma Treatment.
AID1079938Chronic liver disease either proven histopathologically, or through a chonic elevation of serum amino-transferase activity after 6 months. Value is number of references indexed. [column 'CHRON' in source]
AID766229Ratio of IC50 for MMR-deficient human A2780/CP70 cells to IC50 for human A2780 cells in presence of MGMT inactivator PaTrin22013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1825341Induction of morphological changes in human U-87 MG cells assessed as appearance of shrunken cell bodies at 1408 uM measured after 72 hrs by phase contrast microscopy2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1316636Drug level in Sprague-Dawley rat brain at 10 mg/kg, iv after 180 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1825320Cytotoxicity against human U-87 MG cells assessed as cell viability at 10000 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1474439Anti-tumor activity against human MX1 cells xenografted in BALB/c mouse assessed as tumor weight at 50 mg/kg, po administered once daily via gavage for 5 days measured after 11 days (Rvb = 4.09 +/- 0.64 g)2017European journal of medicinal chemistry, May-26, Volume: 132Discovery of 2-substituted 1H-benzo[d]immidazole-4-carboxamide derivatives as novel poly(ADP-ribose)polymerase-1 inhibitors with in vivo anti-tumor activity.
AID540209Volume of distribution at steady state in human after iv administration2008Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 36, Issue:7
Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds.
AID474420Antitumor activity against mouse B16F10 cells transplanted in C57BL/6 mouse assessed as tumor volume at 50 mg/kg/day, po administered from day 6 to 10 post transplantation measured on day 12 relative to control2010Journal of medicinal chemistry, Apr-22, Volume: 53, Issue:8
Optimization of phenyl-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase inhibitors: identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (A-966492), a highly potent and efficacious inhibitor.
AID1317843Antiproliferative activity against human MES-SA/Dx5 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID1888345Antiproliferative activity against human T98G cells assessed as cell viability at 94 uM incubated for 96 hrs by CellTiter-Glo luminescent cell viability assay relative to control
AID1079941Liver damage due to vascular disease: peliosis hepatitis, hepatic veno-occlusive disease, Budd-Chiari syndrome. Value is number of references indexed. [column 'VASC' in source]
AID712149Ratio of IC50 for human A2780 cells to IC50 for MMR-deficient human A2780/CP70 cells in presence of 10 uM MGMT inactivator Patrin22012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1079945Animal toxicity known. [column 'TOXIC' in source]
AID1767412Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as tumor growth inhibition at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation
AID209983In vitro cytotoxicity against mouse TLX5 lymphoma cells2002Journal of medicinal chemistry, Dec-05, Volume: 45, Issue:25
Antitumor imidazotetrazines. 41. Conjugation of the antitumor agents mitozolomide and temozolomide to peptides and lexitropsins bearing DNA major and minor groove-binding structural motifs.
AID1697776Induction of apoptosis in rat RG2 cells assessed as phosphorylated caspace-3 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316639Induction of DNA alkylation in human U87MG cells assessed as increase in O6-MedG formation at 10 to 100 uM after 3 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1763425Cytotoxicity against human SW480 cells by MTT assay2021Bioorganic & medicinal chemistry letters, 07-01, Volume: 43Chemical modifications of ergostane-type triterpenoids from Antrodia camphorata and their cytotoxic activities.
AID7470Potentiation factor at 50% growth inhibition (IC50 of compound to that of compound along with PARP inhibitor) in experiment 12000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
AID1651753Cytotoxicity against human U87MG cells assessed as reduction in cell viability after 48 hrs under hypoxia condition by MTT assay2020Journal of natural products, 04-24, Volume: 83, Issue:4
Boholamide A, an APD-Class, Hypoxia-Selective Cyclodepsipeptide.
AID1316640Induction of DNA alkylation in human U87MG cells assessed as increase in N7-MedG formation after 3 hrs by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1697732Induction of apoptosis in rat RG2 cells assessed as increase in DNA fragmentation at 206 uM after 48 hrs by TUNEL assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697682Effect on AKT expression in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1830975Toxicity in BALB/c mouse xenografted with human U-87MG cells assessed as effect on body weight at 30 mg/kg, po administered once daily for 5 days and measured on day 8 (Rvb = 19 +/- 1 g)
AID26696Half-life period in 95% ethanol in diffuse laboratory light at 28 degrees celsius1984Journal of medicinal chemistry, Feb, Volume: 27, Issue:2
Antitumor imidazotetrazines. 1. Synthesis and chemistry of 8-carbamoyl-3-(2-chloroethyl)imidazo[5,1-d]-1,2,3,5-tetrazin-4(3 H)-one , a novel broad-spectrum antitumor agent.
AID1697806Induction of DNA damage in human DBTRG-05MG cells assessed as Chk2 phosphorylation at Thr68 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID668220Cytotoxicity against mouse primary astrocytes after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Jul-01, Volume: 22, Issue:13
New substituted 4H-chromenes as anticancer agents.
AID1888347Antiproliferative activity against human T98G cells assessed as cell viability at 375 uM incubated for 96 hrs by CellTiter-Glo luminescent cell viability assay relative to control
AID1825318Cytotoxicity against human U-87 MG cells assessed as cell viability at 100 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1200321Antiproliferative activity against human GBM 031810 cells assessed as reduction in cell viability after 48 hrs by MTT assay2015Journal of medicinal chemistry, Mar-12, Volume: 58, Issue:5
Activity of 2-aryl-2-(3-indolyl)acetohydroxamates against drug-resistant cancer cells.
AID1697644Induction of cell cycle arrest in human DBTRG-05MG cells assessed as CyclinA expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1595392Antiinvasive activity in mouse GL261 cells assessed as reduction in cell migration at 10 uM preincubated for 48 hrs and measured after 6 hrs by giemsa staining based Boyden chamber assay2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1274559Antiproliferative activity against human H4 cells after 48 hrs by MTT assay2015Journal of natural products, Dec-24, Volume: 78, Issue:12
Phomanolides A and B from the Fungus Phoma sp.: Meroterpenoids Derived from a Putative Tropolonic Sesquiterpene via Hetero-Diels-Alder Reactions.
AID1262127Antiproliferative activity against human ANGM-CSS cells assessed as inhibition of cell growth at 100 uM after 72 hrs by WST1 assay2015European journal of medicinal chemistry, Nov-13, Volume: 105Design and synthesis of 2-oxindole based multi-targeted inhibitors of PDK1/Akt signaling pathway for the treatment of glioblastoma multiforme.
AID1783996Cytotoxicity against human U-251MG cells assessed as decrease in cell viability incubated for 6 days by alamar blue assay2021European journal of medicinal chemistry, Nov-15, Volume: 224Enhanced pyrazolopyrimidinones cytotoxicity against glioblastoma cells activated by ROS-Generating cold atmospheric plasma.
AID1250158Cytotoxicity against human Glioma cells (HF2876) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
AID1697664Induction of cell cycle arrest in rat RG2 cells assessed as CDK2 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697574Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G2/M phase at 206 uM after 12 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 30.08 +/- 0.1%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID661450Antiproliferative activity against human SF295 cells at 10 uM after 24 hrs by SRB assay2012Journal of medicinal chemistry, Mar-08, Volume: 55, Issue:5
Oxaphosphinanes: new therapeutic perspectives for glioblastoma.
AID1697663Induction of cell cycle arrest in rat RG2 cells assessed as CyclinD1 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1595380Stability of the compound in PBS buffer at pH 7.4 assessed as half life by HPLC analysis2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID26695Half-life period in 95% ethanol in dark at 28 degrees celsius.1984Journal of medicinal chemistry, Feb, Volume: 27, Issue:2
Antitumor imidazotetrazines. 1. Synthesis and chemistry of 8-carbamoyl-3-(2-chloroethyl)imidazo[5,1-d]-1,2,3,5-tetrazin-4(3 H)-one , a novel broad-spectrum antitumor agent.
AID1274558Antiproliferative activity against human U251 cells after 48 hrs by MTT assay2015Journal of natural products, Dec-24, Volume: 78, Issue:12
Phomanolides A and B from the Fungus Phoma sp.: Meroterpenoids Derived from a Putative Tropolonic Sesquiterpene via Hetero-Diels-Alder Reactions.
AID1767415Antitumor activity against rat C6 cells inoculated orthotopic Sprague-Dawley rat model assessed as decrease in VEGF-A in tumor tissue at 50 mg/kg, ip twice a day treated 3 days after tumor cell injection measured for 13 days after tumor implantation by We
AID1697805Induction of DNA damage in rat RG2 cells assessed as Chk1 phosphorylation at ser345 at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697590Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G0/G1 phase at 206 uM after 48 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 47.16 +/- 1.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697777Induction of apoptosis in rat RG2 cells assessed as cleaved caspace-3 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697808Induction of DNA damage in human DBTRG-05MG cells assessed as H2A.X phosphorylation at ser139 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697683Inhibition of AKT phosphorylation at S473 residues in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1876526Induction of apoptosis in human U-251 cells assessed as necrotic cells at 6 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb =0.51 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID661451Antiproliferative activity against human SNB75 cells at 10 uM after 24 hrs by SRB assay2012Journal of medicinal chemistry, Mar-08, Volume: 55, Issue:5
Oxaphosphinanes: new therapeutic perspectives for glioblastoma.
AID1729708Inhibition of colony formation in human GSC-3 cells assessed as tumor spheres damage at 15 uM after 2 weeks by crystal violet staining based assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Structures/cytotoxicity/selectivity relationship of natural steroidal saponins against GSCs and primary mechanism of tribulosaponin A.
AID1697685Inhibition of JNK phosphorylation at Thr183/Tyr185 residues in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316656AUC in Sprague-Dawley rat brain at 10 mg/kg, iv after 30 to 180 mins by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID712165Cytotoxicity against MMR-deficient human A2780/CP70 cells after 5 days by MTT assay2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID7466Potentiation of growth inhibition of A2780 by compound alone in experiment 12000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
AID1079937Severe hepatitis, defined as possibly life-threatening liver failure or through clinical observations. Value is number of references indexed. [column 'MASS' in source]
AID1200319Antiproliferative activity against human MES-SA cells assessed as reduction in cell viability after 48 hrs by MTT assay2015Journal of medicinal chemistry, Mar-12, Volume: 58, Issue:5
Activity of 2-aryl-2-(3-indolyl)acetohydroxamates against drug-resistant cancer cells.
AID1697802Induction of DNA damage in human DBTRG-05MG cells assessed as ATR phosphorylation at ser428 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1416900Growth inhibition of vehicle-transfected human U373 cells after 7 days by MTT assay
AID1825323Cytotoxicity against human U-87 MG cells using best fit nonlinear regression analysis2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1697686Effect on ERK expression in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1254861Tumoricidal effect in patient derived GBM 025T cells assessed as reduction in neurosphere formation at 20 uM after 1 to 2 weeks by 3D tumor clonogenic assay2015European journal of medicinal chemistry, Oct-20, Volume: 103Discovery of potent and selective cytotoxic activity of new quinazoline-ureas against TMZ-resistant glioblastoma multiforme (GBM).
AID1697751Induction of apoptosis in human DBTRG-05MG cells assessed as cleaved caspace-9 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697592Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G2/M phase at 206 uM after 48 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 30.08 +/- 0.1%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1876491Antiproliferative activity against human U-251 cells assessed as inhibition of cell proliferation measured after 72 hrs by CCK8 assay2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID1316646Antitumor activity against human U87MG cells orthotopically xenografted in Harlan nude mouse brain assessed as induction of slow tumor growth at 50 umol/kg, iv administered once daily for 5 days2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1274557Antiproliferative activity against human SH-SY5Y cells after 48 hrs by MTT assay2015Journal of natural products, Dec-24, Volume: 78, Issue:12
Phomanolides A and B from the Fungus Phoma sp.: Meroterpenoids Derived from a Putative Tropolonic Sesquiterpene via Hetero-Diels-Alder Reactions.
AID1651754Selectivity index, ratio of IC50 for cytotoxicity against human U87MG cells under normoxia condition to IC50 for cytotoxicity against human U87MG cells under hypoxia condition incubated for 48 hrs by MTT assay2020Journal of natural products, 04-24, Volume: 83, Issue:4
Boholamide A, an APD-Class, Hypoxia-Selective Cyclodepsipeptide.
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.
AID282486Half life in plasma2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID588208Literature-mined public compounds from Lowe et al phospholipidosis modelling dataset2010Molecular pharmaceutics, Oct-04, Volume: 7, Issue:5
Predicting phospholipidosis using machine learning.
AID1079943Malignant tumor, proven histopathologically. Value is number of references indexed. [column 'T.MAL' in source]
AID1888356Induction of apoptosis in human T98G cells assessed as increase in early apoptotic cells at 94 uM measured after 48 to 96 hrs by Annexin V-FITC/PI staining based flow cytometry analysis
AID1736283Antiproliferative activity against human U87MG cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID1335505Cytotoxicity against Wistar rat C6 cells assessed as decrease in cell viability at 100 uM after 48 hrs by MTT assay relative to control2016European journal of medicinal chemistry, Nov-29, Volume: 124Thiazolidin-4-ones from 4-(methylthio)benzaldehyde and 4-(methylsulfonyl)benzaldehyde: Synthesis, antiglioma activity and cytotoxicity.
AID1202313Cytotoxicity against human WiDr cells overexpressing MGMT after 48 hrs by trypan blue staining-based hemocytometric analysis2015European journal of medicinal chemistry, , Volume: 96Synthesis and antiproliferative activity of 3-(2-chloroethyl)-5-methyl-6-phenyl-8-(trifluoromethyl)-5,6-dihydropyrazolo[3,4-f][1,2,3,5]tetrazepin-4-(3H)-one.
AID1697599Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G0/G1 phase at 206 uM after 6 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 57.81 +/- 1.9%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316644Oral bioavailability in human for 1 to 2 hrs2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1167094Inhibition of neurosphere formation of mouse BXD4687 cells by CCK8 assay2014Journal of medicinal chemistry, Oct-23, Volume: 57, Issue:20
Inhibition of cancer-associated mutant isocitrate dehydrogenases: synthesis, structure-activity relationship, and selective antitumor activity.
AID441950Antitumor activity against mouse B16F10 cells implanted in C57BL/6 mouse at 50 mg/kg, po QD for 5 days measured on day 162009Journal of medicinal chemistry, Nov-12, Volume: 52, Issue:21
Synthesis and evaluation of a new generation of orally efficacious benzimidazole-based poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors as anticancer agents.
AID1697708Effect on JNK expression in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1316650Drug level in rat brain at 103 umol/kg, iv measured 3 hrs post dose by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1736281Selectivity index, ratio of CC50 for Wistar rat astrocytes to EC50 for rat C6 cells2020European journal of medicinal chemistry, Mar-01, Volume: 189Efficient identification of novel anti-glioma lead compounds by machine learning models.
AID1425988Binding affinity to recombinant human carbonic anhydrase 2 after 15 mins by stopped-flow CO2 hydration assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1769427Cytotoxicity against human MNT-1 cells assessed as reduction in cell viability incubated for 72 hrs by MTS assay2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Sulfur Analogues of Tyrosine in the Development of Triazene Hybrid Compounds: A New Strategy against Melanoma.
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.
AID477295Octanol-water partition coefficient, log P of the compound2010European journal of medicinal chemistry, Apr, Volume: 45, Issue:4
QSPR modeling of octanol/water partition coefficient of antineoplastic agents by balance of correlations.
AID540213Half life in human after iv administration2008Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 36, Issue:7
Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds.
AID712154Prodrug conversion in citrate-phosphate buffer at pH 7.4 assessed as pseudo-first-order rate constant2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1811267In vivo inhibition of COX2 in ip dosed orthotopic Sprague-Dawley rat model inoculated with C6 cells assessed as reduction in PGE2 production by ELISA2021European journal of medicinal chemistry, Dec-15, Volume: 226N-2-(phenylamino) benzamide derivatives as novel anti-glioblastoma agents: Synthesis and biological evaluation.
AID1697837Antitumour activity against human DBTRG-05MG cells xenografted in BALB/c nude mouse assessed as increase in survival time at 5 mg/kg, ip for 5 days and measured every 2 days2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1623801Anti-angiogenic activity in tumor of human U87MG cells xenografted in nude BALB/c mouse assessed as reduction in VEGF staining at 50 mg/kg, ip administered for 14 days by immunohistochemistry2019Journal of medicinal chemistry, 02-14, Volume: 62, Issue:3
Design of Novel Inhibitors of Human Thymidine Phosphorylase: Synthesis, Enzyme Inhibition, in Vitro Toxicity, and Impact on Human Glioblastoma Cancer.
AID1697731Induction of apoptosis in rat RG2 cells assessed as increase in chromatin condensation at 206 uM after 48 hrs by TUNEL assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1172643Inhibition of cell proliferation of human U87MG cells assessed as cell viability after 72 hrs by sulforhodamine B assay2014Bioorganic & medicinal chemistry letters, Nov-15, Volume: 24, Issue:22
Bioactive triterpenoid saponins and phenolic compounds against glioma cells.
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.
AID84439Cytotoxicity of compound in HT-29 cell line2000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 8. Inhibition of O(6)-alkylguanine-DNA alkyltransferase by O(6)-alkenyl-, O(6)-cycloalkenyl-, and O(6)-(2-oxoalkyl)guanines and potentiation of temozolomide cytotoxicity in vitro by O(6)-(1-cyclopentenylmethyl)guanine.
AID1697583Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G2/M phase at 206 uM after 24 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 30.08 +/- 0.1%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1425997Cytotoxicity against carbonic anhydrase 9 knockdown MDCK cells assessed as reduction in cell viability after 72 hrs under normoxic condition by Alamar blue assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID667959Growth inhibition of mouse B16F10 cells after 72 hrs by MTT assay2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID1697460Cytotoxicity against human DBTRG-05MG cells assessed as reduction in cell viability after 24 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697572Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G0/G1 phase at 206 uM after 12 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 47.16 +/- 1.55%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID661463Antiproliferative activity against rat C6 cells at 100 uM after 48 hrs by neutral red incorporation assay2012Journal of medicinal chemistry, Mar-08, Volume: 55, Issue:5
Oxaphosphinanes: new therapeutic perspectives for glioblastoma.
AID1697564Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at S phase at 206 uM after 6 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 22.76 +/- 1.56%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1254862Tumoricidal effect in patient derived GBM 448T cells assessed as reduction in neurosphere formation at 20 uM after 1 to 2 weeks by 3D tumor clonogenic assay2015European journal of medicinal chemistry, Oct-20, Volume: 103Discovery of potent and selective cytotoxic activity of new quinazoline-ureas against TMZ-resistant glioblastoma multiforme (GBM).
AID1537886Cytotoxicity against patient-isolated primary glioblastoma cells derived adherent cells assessed as increase in caspase3 cleavage at 50 uM measured after 48 hrs by immunoblotting method2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1771785Inhibition of DNA Topoisomerase-2 in human MDA-MB-238 cells assessed as generation of DNA double strand breaks measuring gamma-H2AX at incubated for 24 hrs followed by drug washout and measured after 24 to 96 hrs by immunofluorescence assay2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1651752Cytotoxicity against human U87MG cells assessed as reduction in cell viability after 48 hrs under normoxia condition by MTT assay2020Journal of natural products, 04-24, Volume: 83, Issue:4
Boholamide A, an APD-Class, Hypoxia-Selective Cyclodepsipeptide.
AID1316651Fraction unbound in mouse brain assessed as compound level in insoluble tissue fraction at 10 uM after 1 hr by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1769429Cytotoxicity against human A-375 cells assessed as reduction in cell viability incubated for 72 hrs by MTS assay2021ACS medicinal chemistry letters, Nov-11, Volume: 12, Issue:11
Sulfur Analogues of Tyrosine in the Development of Triazene Hybrid Compounds: A New Strategy against Melanoma.
AID1825343Induction of morphological changes in human U-87 MG cells assessed as appearance of undergoing apoptosis at 1408 uM measured after 72 hrs by phase contrast microscopy2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID712155Prodrug conversion in citrate-phosphate buffer at pH 4 assessed as pseudo-first-order rate constant2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID766234Half life of the compound at pH 7.42013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID84467Level of AGT activity in HT-29 cell line2000Journal of medicinal chemistry, Nov-02, Volume: 43, Issue:22
Resistance-modifying agents. 8. Inhibition of O(6)-alkylguanine-DNA alkyltransferase by O(6)-alkenyl-, O(6)-cycloalkenyl-, and O(6)-(2-oxoalkyl)guanines and potentiation of temozolomide cytotoxicity in vitro by O(6)-(1-cyclopentenylmethyl)guanine.
AID683688Cytotoxicity against rat C6 cells incubated for 48 hrs by MTT assay2012European journal of medicinal chemistry, Nov, Volume: 57Selective cytotoxicity and apoptosis induction in glioma cell lines by 5-oxygenated-6,7-methylenedioxycoumarins from Pterocaulon species.
AID165776In vitro cytotoxicity against Raji cell line1995Journal of medicinal chemistry, Apr-28, Volume: 38, Issue:9
Antitumor imidazotetrazines. 32. Synthesis of novel imidazotetrazinones and related bicyclic heterocycles to probe the mode of action of the antitumor drug temozolomide.
AID1697626Induction of cell cycle arrest in rat RG2 cells assessed as accumulation at G0/G1 phase at 206 uM after 48 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 57.81 +/- 1.9%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1825311Cytotoxicity against human SH-SY5Y cells using best fit nonlinear regression analysis2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1697711Inhibition of ERK phosphorylation at Tyr204 residues in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1811285Toxicity in Sprague-Dawley rat inoculated with C6 cells assessed as reduction in body weight at 50 mg/kg, ip bid administered for 14 days2021European journal of medicinal chemistry, Dec-15, Volume: 226N-2-(phenylamino) benzamide derivatives as novel anti-glioblastoma agents: Synthesis and biological evaluation.
AID1697712Effect on p38 expression in rat RG2 cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1771788Cytotoxicity against human NCI-H1299 cells assessed as cell viability measured after 96 hrs by XTT assay2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1425990Binding affinity to recombinant human carbonic anhydrase 12 after 15 mins by stopped-flow CO2 hydration assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID712166Cytotoxicity against MMR-deficient human A2780/CP70 cells after 5 days by MTT assay in presence of 10 uM MGMT inactivator Patrin22012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID1697462Cytotoxicity against human DBTRG-05MG cells assessed as reduction in cell viability after 72 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697804Induction of DNA damage in human DBTRG-05MG cells assessed as Chk1 phosphorylation at ser345 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1771787Cytotoxicity against human WM 266-4 cells assessed as cell viability measured after 96 hrs by XTT assay2021European journal of medicinal chemistry, Dec-05, Volume: 225Targeted methylation facilitates DNA double strand breaks and enhances cancer suppression: A DNA intercalating/methylating dual-action chimera Amonafidazene.
AID1254860Tumoricidal effect in patient derived GBM 096T cells assessed as reduction in neurosphere formation at 20 uM after 1 to 2 weeks by 3D tumor clonogenic assay2015European journal of medicinal chemistry, Oct-20, Volume: 103Discovery of potent and selective cytotoxic activity of new quinazoline-ureas against TMZ-resistant glioblastoma multiforme (GBM).
AID1825317Cytotoxicity against human SH-SY5Y cells assessed as cell viability at 500 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1729710Inhibition of colony formation in human GSC-3 cells assessed as decrease in number of tumor spheres at 15 uM after 2 weeks by crystal violet staining based assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Structures/cytotoxicity/selectivity relationship of natural steroidal saponins against GSCs and primary mechanism of tribulosaponin A.
AID1697748Induction of apoptosis in human DBTRG-05MG cells assessed as Bcl2 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1876513Induction of apoptosis in human U-87 MG cells assessed as necrotic cells at 1.20 uM incubated for 72 hrs by Annexin V-FITC/PI staining based flow cytometry method (Rvb = 0.21 %)2022European journal of medicinal chemistry, Jan-05, Volume: 227Novel piperazine based benzamide derivatives as potential anti-glioblastoma agents inhibiting cell proliferation and cell cycle progression.
AID712160Prodrug conversion in citrate-phosphate buffer at pH 4 assessed as half life of compound2012ACS medicinal chemistry letters, Dec-13, Volume: 3, Issue:12
Strategy for Imidazotetrazine Prodrugs with Anticancer Activity Independent of MGMT and MMR.
AID668216Antiproliferative activity against human T98G cells after 5 days by MTT assay2012Bioorganic & medicinal chemistry letters, Jul-01, Volume: 22, Issue:13
New substituted 4H-chromenes as anticancer agents.
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.
AID1595391Induction of morphological changes in mouse GL261 cells assessed as decrease in cell aspect ratio at 10 uM incubated for 48 hrs by Hoechst 33258 staining based fluorescence microscopic analysis2019European journal of medicinal chemistry, Jun-15, Volume: 172Targeting gliomas with triazene-based hybrids: Structure-activity relationship, mechanistic study and stability.
AID1316648Increase in O6-MedG formation in rat brain at 51.5 to 206 umol/kg, iv measured 3 hrs post dose by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1603447Cytotoxicity in TRPV3 expressing human PC3 cells assessed as reduction in cell viability at 200 uM incubated for 24 to 72 hrs by Cell-titer blue assay2019Bioorganic & medicinal chemistry, 06-15, Volume: 27, Issue:12
Natural product-drug conjugates for modulation of TRPV1-expressing tumors.
AID1486367Cytotoxicity against mouse B16F10 cells after 72 hrs by MTS assay2017Bioorganic & medicinal chemistry, 08-01, Volume: 25, Issue:15
The selective cytotoxicity of new triazene compounds to human melanoma cells.
AID1697661Induction of cell cycle arrest in rat RG2 cells assessed as p21 expression at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1537879Down regulation of P-gp expression in patient-isolated primary glioblastoma cells derived neurospheres at 50 uM after 48 hrs by immunoblotting analysis2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID1825319Cytotoxicity against human U-87 MG cells assessed as cell viability at 2000 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1250154Cytotoxicity against human Glioma cells (HF2303) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
AID348242Antitumor activity against mouse B16F10 cells xenografted in C57BL/6 mouse assessed as tumor volume at 50 mg/kg/day, po after 17 days2009Journal of medicinal chemistry, Jan-22, Volume: 52, Issue:2
Discovery of the Poly(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide (ABT-888) for the treatment of cancer.
AID667961Selectivity index, ratio of IC50 for mouse astrocytes to IC50 for human T98G cells2012European journal of medicinal chemistry, Aug, Volume: 54N-Aryl-N'-(chroman-4-yl)ureas and thioureas display in vitro anticancer activity and selectivity on apoptosis-resistant glioblastoma cells: screening, synthesis of simplified derivatives, and structure-activity relationship analysis.
AID1783997Cytotoxicity against cold atmospheric plasma (CAP)-treated human U-251MG cells assessed as decrease in cell viability treated with CAP for 15 secs followed by incubated with compound for 6 days by alamar blue assay2021European journal of medicinal chemistry, Nov-15, Volume: 224Enhanced pyrazolopyrimidinones cytotoxicity against glioblastoma cells activated by ROS-Generating cold atmospheric plasma.
AID540235Phospholipidosis-negative literature compound
AID228828Inhibition of Abl tyrosine kinase2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
Synthesis of pyrimidinopyridine-triazene conjugates targeted to abl tyrosine kinase.
AID1825335Cytotoxicity against human U-87 MG cells assessed as cell death at 352 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1697801Induction of DNA damage in rat RG2 cells assessed as ATM phosphorylation at ser1981 residues at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697565Induction of cell cycle arrest in human DBTRG-05MG cells assessed as accumulation at G2/M phase at 206 uM after 6 hrs by propidium iodide staining based flow cytometric analysis (Rvb = 30.08 +/- 0.1%)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1697689Inhibition of p38 phosphorylation at Tyr182 residues in human DBTRG-05MG cells at 206 uM after 48 hrs by Western blot analysis (Rvb = 1 No_unit)2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
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).
AID1079949Proposed mechanism(s) of liver damage. [column 'MEC' in source]
AID1425989Binding affinity to recombinant human carbonic anhydrase 9 after 15 mins by stopped-flow CO2 hydration assay2017European journal of medicinal chemistry, Feb-15, Volume: 127New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs.
AID1317840Antiproliferative activity against patient-derived paclitaxel-sensitive NCI-H1993 cells after 48 to 96 hrs by MTT assay2016European journal of medicinal chemistry, Sep-14, Volume: 1205,10b-Ethanophenanthridine amaryllidaceae alkaloids inspire the discovery of novel bicyclic ring systems with activity against drug resistant cancer cells.
AID540212Mean residence time in human after iv administration2008Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 36, Issue:7
Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 670 drug compounds.
AID1250155Cytotoxicity against human Glioma cells (HF2381) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
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).
AID1537889Cytotoxicity against P-gp knockout patient-isolated primary glioblastoma cells derived neurospheres assessed as increase in caspase3 cleavage at 50 uM measured after 48 hrs by immunoblotting method2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Carbonic Anhydrase XII Inhibitors Overcome Temozolomide Resistance in Glioblastoma.
AID282557Antitumor activity against human NCI prostate cancer cell line panel2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis and antitumor activity of methyltriazene prodrugs simultaneously releasing DNA-methylating agents and the antiresistance drug O(6)-benzylguanine.
AID1316654Ratio of AUC in brain to plasma of Sprague-Dawley rat administered as single dose2016Bioorganic & medicinal chemistry letters, 10-15, Volume: 26, Issue:20
Temozolomide analogs with improved brain/plasma ratios - Exploring the possibility of enhancing the therapeutic index of temozolomide.
AID1250156Cytotoxicity against human Glioma cells (HF2476) at 50 uM after 72 hrs by CelltiterGlo assay2015ACS medicinal chemistry letters, Aug-13, Volume: 6, Issue:8
High-Throughput Screening of Patient-Derived Cultures Reveals Potential for Precision Medicine in Glioblastoma.
AID1825315Cytotoxicity against human SH-SY5Y cells assessed as cell viability at 100 uM incubated upto 72 hrs by MTT assay2022Journal of natural products, 01-28, Volume: 85, Issue:1
Cyclotides Chemosensitize Glioblastoma Cells to Temozolomide.
AID1888346Antiproliferative activity against human T98G cells assessed as cell viability at 188 uM incubated for 96 hrs by CellTiter-Glo luminescent cell viability assay relative to control
AID766224Chemosensitization of human SNB19 cells expressing MGMT after 5 days by MTT assay2013Journal of medicinal chemistry, Sep-12, Volume: 56, Issue:17
Synthesis and quantitative structure-activity relationship of imidazotetrazine prodrugs with activity independent of O6-methylguanine-DNA-methyltransferase, DNA mismatch repair, and p53.
AID1697468Cytotoxicity against rat CTX TNA2 cells assessed as reduction in cell viability after 72 hrs by MTT assay2020Journal of natural products, 10-23, Volume: 83, Issue:10
Cedrol, a Sesquiterpene Alcohol, Enhances the Anticancer Efficacy of Temozolomide in Attenuating Drug Resistance via Regulation of the DNA Damage Response and MGMT Expression.
AID1851875Cytotoxicity against human GSC#83 cells assessed as reduction in cell viability at 10 uM incubated for 24 hrs by MTS assay
AID1347411qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Mechanism Interrogation Plate v5.0 (MIPE) Libary2020ACS 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.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
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.
AID1159607Screen for inhibitors of RMI FANCM (MM2) intereaction2016Journal of biomolecular screening, Jul, Volume: 21, Issue:6
A High-Throughput Screening Strategy to Identify Protein-Protein Interaction Inhibitors That Block the Fanconi Anemia DNA Repair Pathway.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (5,827)

TimeframeStudies, This Drug (%)All Drugs %
pre-199013 (0.22)18.7374
1990's99 (1.70)18.2507
2000's1139 (19.55)29.6817
2010's3154 (54.13)24.3611
2020's1422 (24.40)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 79.23

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 Index79.23 (24.57)
Research Supply Index8.83 (2.92)
Research Growth Index6.03 (4.65)
Search Engine Demand Index142.86 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (79.23)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials826 (13.73%)5.53%
Reviews623 (10.36%)6.00%
Case Studies541 (9.00%)4.05%
Observational22 (0.37%)0.25%
Other4,002 (66.54%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (854)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Phase 3, Open Label, Randomized, Comparative Study Of CP-675,206 And Either Dacarbazine Or Temozolomide In Patients With Advanced Melanoma [NCT00257205]Phase 3655 participants (Actual)Interventional2006-03-31Completed
A Multi-Center Phase I Dose Escalation Trial to Evaluate Safety and Tolerability of Intra-Arterial Temozolomide for Patients With Advanced Extremity Melanoma Using Normothermic Isolated Limb Infusion [NCT01127594]Phase 129 participants (Actual)Interventional2010-07-31Completed
Phase 11 Study of Cilengitide in Combination With Concurrent Chemotherapy and Radiotherapy Followed by Protracted Daily Low Dose Temozolomide and Low Dose Procarbazine D1 - 20 in Newly Diagnosed Glioblastoma Without Methylation of the MGMT Promoter Gene [NCT01124240]Phase 248 participants (Anticipated)Interventional2009-11-30Recruiting
A Phase 1 Study of Temsirolimus in Combination With Irinotecan and Temozolomide in Children, Adolescents, and Young Adults With Relapsed or Refractory Solid Tumors [NCT01141244]Phase 172 participants (Actual)Interventional2010-06-30Completed
Phase III Trial Comparing Conventional Adjuvant Temozolomide With Dose-Intensive Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT00304031]Phase 31,173 participants (Actual)Interventional2006-01-31Completed
A Phase II Clinical Trial of the Histone Deacetylase Inhibitor Valproic Acid in Combination With Temodar and Radiation Therapy in Patients With High Grade Gliomas: Multi-Institutional Trial [NCT00302159]Phase 243 participants (Actual)Interventional2006-03-31Completed
A Phase I/II Study of High-Dose Calcitriol in Combination With Temozolomide for Patients With Metastatic Melanoma [NCT00301067]Phase 1/Phase 220 participants (Actual)Interventional2005-01-30Completed
A Phase 1/2 Trial Evaluating the Combination of Temozolomide and the Ataxia Telangiectasia and Rad3-Related Inhibitor M1774 [NCT05691491]Phase 1/Phase 258 participants (Anticipated)Interventional2023-09-28Recruiting
Quantitative Magnetic Resonance Spectroscopic Imaging (MRSI) to Predict Early Response to Standard Radiation Therapy (RT)/Temozolomide (TMZ) ± Belinostat Therapy in Newly-Diagnosed Glioblastomas (GBM) [NCT02137759]Phase 229 participants (Actual)Interventional2014-05-07Active, not recruiting
Phase II Trial of Pulse Dosing of Lapatinib in Combination With Temozolomide and Regional Radiation Therapy for Upfront Treatment of Patients With Newly-Diagnosed Glioblastoma Multiforme [NCT01591577]Phase 250 participants (Actual)Interventional2012-12-07Active, not recruiting
An International, Multi-Center, Randomized, Double Blind Placebo Controlled Phase II Study to Evaluate the Safety and Efficacy of Lucanthone Administered as an Adjunct to Radiation and Temozolomide for Primary Therapy of Glioblastoma Multiforme [NCT01587144]Phase 218 participants (Actual)Interventional2012-06-19Terminated(stopped due to Sponsor's decision)
A Phase II Study of Temozolomide for Injection Combined With Epirubicin in the First-line Treatment of Leiomyosarcoma [NCT05204524]28 participants (Anticipated)Interventional2021-08-27Recruiting
A Phase II/III Study of Non Small Cell Lung Cancer Patient With/Without Synchronous Oral Temozolomide Capsules Combine With Intensity Modulated Radiation Therapy With Simultaneous Integrated Boost With Hippocampus Protection [NCT03732482]Phase 2/Phase 3400 participants (Anticipated)Interventional2019-03-01Not yet recruiting
A Phase II Study of Temozolomide, Cisplatin and Nivolumab in MMR-Proficient Colorectal Cancer [NCT04457284]Phase 218 participants (Actual)Interventional2020-11-18Active, not recruiting
A Phase I Study of BRCX014 to Investigate Dose-Ranging Safety and Pharmacokinetics in Adults With Glioblastoma (GBM) and Non-Methylated MGMT Gene Status [NCT03687034]Phase 121 participants (Anticipated)Interventional2019-06-01Not yet recruiting
First Line Standard Therapy of Glioblastoma Multiforme With or Without add-on Treatment With Trivax, an Anti-tumour Immune Therapy Based on Tumour-lysate Charged Dendritic Cells [NCT01213407]Phase 287 participants (Actual)Interventional2010-04-30Completed
A Cancer Research UK Phase I Trial of IMA950 (A Novel Multi-Peptide Vaccine) Plus GM-CSF in Patients With Newly Diagnosed Glioblastoma [NCT01222221]Phase 145 participants (Actual)Interventional2010-07-31Completed
Randomized Trial Assessing the Significance of Bevacizumab in Recurrent Grade II and Grade III Gliomas - The TAVAREC Trial [NCT01164189]Phase 2155 participants (Actual)Interventional2011-02-28Completed
High-Dose Vitamin D in Combination With Chemoradiotherapy in the Treatment of Glioblastoma Multiforme [NCT01181193]Phase 1/Phase 220 participants (Anticipated)Interventional2011-03-31Recruiting
Efficacy and Safety of Tunlametinib Capsules Versus Combination Chemotherapy of Investigator's Choice in Advanced NRAS-mutant Melanoma Patients Who Had Previously Received Immunotherapy [NCT06008106]Phase 3165 participants (Anticipated)Interventional2023-09-22Not yet recruiting
Phase I Trial of Zotiraciclib (TG02) Plus Dose-Dense or Metronomic Temozolomide Followed by Randomized Phase II Trial of Zotiraciclib (TG02) Plus Temozolomide Versus Temozolomide Alone in Adults With Recurrent Anaplastic Astrocytoma and Glioblastoma [NCT02942264]Phase 1/Phase 253 participants (Actual)Interventional2016-12-14Completed
PI3K/mTOR Pathway Activation Selected Phase II Study of Everolimus (RAD001) With and Without Temozolomide in the Treatment of Adult Patients With Supratentorial Low-Grade Glioma [NCT02023905]Phase 227 participants (Actual)Interventional2014-03-19Terminated(stopped due to Sponsor decision)
Phase II Trial of Low-Dose Whole Brain Radiotherapy With Concurrent Temozolomide and Adjuvant Temozolomide in Patients With Newly-Diagnosed Glioblastoma Multiforme [NCT01822275]Phase 224 participants (Actual)Interventional2013-05-31Terminated(stopped due to Poor accrual)
A Phase II, Open-Label Study to Evaluate the Safety, Tolerability, and Efficacy of Disulfiram and Copper Gluconate When Added to Standard Temozolomide Treatment in Patients With Newly Diagnosed Resected Unmethylated Glioblastoma Multiforme [NCT03363659]Phase 215 participants (Actual)Interventional2018-03-28Terminated(stopped due to Investigator decision)
Phase II Study Incorporating Pegylated Interferon In the Treatment For Children With High-Risk Melanoma [NCT00539591]Phase 229 participants (Actual)Interventional2008-05-09Active, not recruiting
International Randomized Phase II Trial of the Combination of Vincristine and Irinotecan With or Without Temozolomide (VI or VIT) in Children and Adults With Refractory or Relapsed Rhabdomyosarcoma [NCT01355445]Phase 2120 participants (Actual)Interventional2012-01-31Completed
A Single-blind, Randomized, Clinical Trial Comparing the Efficacy of 6 Cycles Versus 12 Cycles Temozolomide Regimens in Adjuvant Treatment of Patients With Brain High Grade Glioma [NCT03633552]Phase 362 participants (Anticipated)Interventional2018-03-03Recruiting
Temozolomide and Concomitant Whole Brain Radiotherapy in NSCLC Patients With Brain Metastases: A Randomized Trial [NCT02385136]Phase 1/Phase 2135 participants (Anticipated)Interventional2015-04-30Not yet recruiting
A Phase I/II Study of Ex-Vivo Expanded Allogeneic Universal Donor (UD) TGFbi NK Cell Infusions in Combination With Temozolomide as a Lymphodepleting Agent in Patients With Melanoma Metastatic to the Brain [NCT05588453]Phase 1/Phase 230 participants (Anticipated)Interventional2023-01-01Recruiting
IDH Mutated 1p/19q Intact Lower Grade Glioma Following Resection: Wait Or Treat? IWOT - a Phase III Study [NCT03763422]Phase 319 participants (Actual)Interventional2020-03-16Terminated(stopped due to Poorly recruiting)
A Pilot Study of Chimeric Antigen Receptor (CAR) T Cells Targeting B7-H3 Antigen in Treating Patients With Recurrent and Refractory Glioblastoma [NCT04385173]Phase 112 participants (Anticipated)Interventional2022-12-01Recruiting
Phase 1b Trial of 5-fluorouracil, Leucovorin, Irinotecan in Combination With Temozolomide (FLIRT) and Bevacizumab for the First-line Treatment of Patients With MGMT Silenced, Microsatellite Stable Metastatic Colorectal Cancer. [NCT04689347]Phase 118 participants (Anticipated)Interventional2021-01-01Recruiting
Image-Based Quantitative Assessment of Acute Radiation-Induced Changes in Glioma [NCT03168919]5 participants (Actual)Interventional2016-10-06Terminated(stopped due to PI is leaving institution)
RAPID Feasibility Study: A Pilot Study for the Rapid Infusion of Dinutuximab [NCT05421897]Phase 411 participants (Anticipated)Interventional2022-10-24Recruiting
A Phase I Study of Methoxyamine and Temozolomide in Patients With Advanced Solid Tumors [NCT00892385]Phase 166 participants (Actual)Interventional2007-08-16Completed
A Phase II, Multicenter, Randomized, Double-blind, Placebo-controlled Clinical Study to Evaluate the Efficacy and Safety of Toripalimab Combined With Temozolomide and Cisplatin in the Postoperative Adjuvant Treatment of Completely Resected Mucosal Melanom [NCT04462965]Phase 2294 participants (Anticipated)Interventional2020-05-30Recruiting
Phase I/II Randomized Prospective Trial for Newly Diagnosed GBM, With Upfront Gross Total Resection, Gliadel®, Followed by Temodar® With Concurrent IMRT Versus GK [NCT02085304]Phase 1/Phase 280 participants (Anticipated)Interventional2012-10-31Recruiting
B7-H3-Targeted Chimeric Antigen Receptor (CAR) T Cells in Treating Patients With Recurrent or Refractory Glioblastoma [NCT04077866]Phase 1/Phase 240 participants (Anticipated)Interventional2023-06-01Recruiting
A Pilot Study of the Combination of Nab-paclitaxel, Temozolomide and Bevacizumab in Patients With Metastatic Melanoma With Brain Metastases [NCT02065466]Phase 1/Phase 20 participants (Actual)Interventional2014-07-31Withdrawn(stopped due to lack of accrual)
A Phase I Study of Mebendazole for the Treatment of Pediatric Gliomas [NCT01837862]Phase 1/Phase 236 participants (Anticipated)Interventional2013-10-22Recruiting
PV-10 Intralesional Injection vs Systemic Chemotherapy or Oncolytic Viral Therapy for Treatment of Locally Advanced Cutaneous Melanoma [NCT02288897]Phase 320 participants (Actual)Interventional2015-04-30Terminated(stopped due to Inadequate rate of enrollment)
Expedited Laser Interstitial Thermal Therapy and Chemoradiation for Patients With Newly Diagnosed High Grade Gliomas [NCT02970448]Early Phase 145 participants (Anticipated)Interventional2017-08-24Recruiting
Phase II Trial of Extended-Dosing Temozolomide in Patients With Melanoma [NCT00591370]Phase 251 participants (Actual)Interventional2005-01-31Completed
Linking the Gut Microbiota to the Prognosis of Glioblastoma Multiforme [NCT03631823]200 participants (Anticipated)Observational2018-08-10Not yet recruiting
Safety and Tolerability of Tumor Treating Fields (TTFields) Combined With Chemoradiation in Newly Diagnosed Glioblastoma (Unity) [NCT03705351]Phase 17 participants (Actual)Interventional2019-12-02Active, not recruiting
A Phase 2 Study of Verubulin With Radiation Therapy and Temozolomide in Subjects Newly Diagnosed With Glioblastoma Multiforme [NCT01285414]Phase 25 participants (Actual)Interventional2010-12-31Completed
Phase I Trial of Super-Selective Intraarterial Cerebral Infusion of Temozolomide (Temodar) for Treatment of Newly Diagnosed Glioblastoma Multiforme and Anaplastic Astrocytoma [NCT01180816]Phase 121 participants (Actual)Interventional2010-08-31Completed
Phase 2 Single-Arm, Open Label Study Of Irinotecan In Combination With Temozolomide In Children With Recurrent Or Refractory Medulloblastoma And In Children With Newly Diagnosed High-Grade Glioma. [NCT00404495]Phase 283 participants (Actual)Interventional2007-04-30Completed
A Randomized, Factorial-Design, Phase II Trial of Temozolomide Alone and in Combination With Possible Permutations of Thalidomide, Isotretinoin and/or Celecoxib as Post-Radiation Adjuvant Therapy of Glioblastoma Multiforme [NCT00112502]Phase 2178 participants (Actual)Interventional2005-09-30Completed
Phase I/II Study Of Pre-Irradiation Chemotherapy With Methotrexate, Rituximab, And Temozolomide And Post -Irradiation Temozolomide For Primary Central Nervous System Lymphoma [NCT00068250]Phase 1/Phase 260 participants (Actual)Interventional2003-07-31Completed
The Temozolomide RESCUE Study: A Phase II Trial of Continuous (28/28) Dose-intense Temozolomide (CDIT) Chemotherapy After Progression on Conventional 5/28 Day Temozolomide in Patients With Recurrent Malignant Glioma [NCT00392171]Phase 2120 participants (Actual)Interventional2006-06-09Completed
Extended Schedule, Escalated Dose Temozolomide Versus Dacarbazine in Stage IV Metastatic Melanoma: A Randomized Phase III Study of the EORTC Melanoma Group [NCT00091572]Phase 3859 participants (Actual)Interventional2004-10-20Completed
Intensive Chemotherapy And Immunotherapy In Patients With Newly Diagnosed Primary CNS Lymphoma [NCT00098774]Phase 247 participants (Actual)Interventional2004-10-31Completed
A Phase II Single Arm Interventional Trial Evaluating the Activity and Safety of Combination Between Cabozantinib and Temozolomide in Lung and GEP-NENS Progressive After Everolimus, Sunitinib or PRRT (CABOTEM) [NCT04893785]Phase 235 participants (Anticipated)Interventional2021-06-15Recruiting
A Phase II Pilot Study Of Thalidomide With Temozolomide In Patients With Relapsed Or Progressive Brain Tumors Or Neuroblastoma [NCT00098865]Phase 215 participants (Actual)Interventional2002-09-30Completed
Phase I/IIa Study of Concomitant Radiotherapy With Olaparib and Temozolomide in Unresectable High Grade Gliomas Patients [NCT03212742]Phase 1/Phase 291 participants (Anticipated)Interventional2017-09-04Recruiting
Radiation Versus Temozolomide in Preventing Brain Metastases in Limited Stage Small Cell Lung Cancer [NCT02605811]Phase 2426 participants (Anticipated)Interventional2015-09-30Recruiting
A Phase Ib/II Randomized, Open Label Drug Repurposing Trial of Glutamate Signaling Inhibitors in Combination With Chemoradiotherapy in Patients With Newly Diagnosed Glioblastoma [NCT05664464]Phase 1/Phase 2120 participants (Anticipated)Interventional2023-01-01Recruiting
A Phase 1 Study of Tumor Treating Fields With 5 Day Hypofractionated Stereotactic Radiosurgery and Concurrent and Maintenance Temozolomide in Newly Diagnosed Glioblastoma [NCT04474353]Phase 112 participants (Anticipated)Interventional2021-05-21Recruiting
PHASE 1/2 STUDY TO EVALUATE PALBOCICLIB (IBRANCE®) IN COMBINATION WITH IRINOTECAN AND TEMOZOLOMIDE OR IN COMBINATION WITH TOPOTECAN AND CYCLOPHOSPHAMIDE IN PEDIATRIC PATIENTS WITH RECURRENT OR REFRACTORY SOLID TUMORS [NCT03709680]Phase 2184 participants (Anticipated)Interventional2019-05-24Recruiting
A Pivotal Randomized, Controlled Trial of VAL-083 in Patients With Recurrent Glioblastoma Who Have Failed Standard Temozolomide/Radiation Therapy and Bevacizumab (STAR-3) [NCT03149575]Phase 32 participants (Actual)Interventional2017-10-27Terminated(stopped due to Change in clinical development plan)
Phase 1b/2a Study Combining LY2157299 With Standard Temozolomide-based Radiochemotherapy in Patients With Newly Diagnosed Malignant Glioma [NCT01220271]Phase 1/Phase 275 participants (Actual)Interventional2011-04-30Completed
An International, Randomized, Double-Blind, Controlled Study of Rindopepimut/GM-CSF With Adjuvant Temozolomide in Patients With Newly Diagnosed, Surgically Resected, EGFRvIII-positive Glioblastoma [NCT01480479]Phase 3745 participants (Actual)Interventional2011-11-30Completed
A Phase 2 Randomized Clinical Trial of ABT-888 in Combination With Temozolomide Versus Pegylated Liposomal Doxorubicin Alone in Subjects With Recurrent High Grade Serous Ovarian Cancer [NCT01113957]Phase 2168 participants (Actual)Interventional2010-03-31Completed
Naxitamab and Granulocyte-Macrophage Colony Stimulating Factor in Combination With Irinotecan and Temozolomide in Patients With High-Risk Neuroblastoma With Primary Refractory Disease or in First Relapse. An International, Single-Arm, Multicenter Phase 2 [NCT04560166]Phase 22 participants (Actual)Interventional2021-11-08Terminated(stopped due to Due to business priorities)
Phase I/II Study of Temozolomide and Gleevec (Imatinib Mesylate, Formerly Known as STI571) in Advanced Melanoma [NCT00667953]Phase 1/Phase 232 participants (Actual)Interventional2003-01-31Terminated(stopped due to Difficulty enrolling subjects)
A Phase II Study of CDX-110 With Radiation and Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00458601]Phase 282 participants (Actual)Interventional2007-08-31Completed
A Phase II Randomized,Controlled,Open Label,Multicentre Study of Tegafur Combined With Temozolomide Versus Tegafur Combined With Temozolomide and Thalidomide in Subjects With Advanced Extrapancreatic Neuroendocrine Tumor [NCT03204032]Phase 260 participants (Anticipated)Interventional2016-10-31Recruiting
Phase Ib/II Multicentric Study Combining Glasdegib (PF-04449913) With Temozolomide in Patients With Newly Diagnosed Glioblastoma, Safety and Preliminary Efficacy for the Combination [NCT03466450]Phase 1/Phase 275 participants (Actual)Interventional2018-03-15Active, not recruiting
Pre-operative Radiation Therapy (RT) and Temozolomide (TMZ) in Patients With Newly Diagnosed Glioblastoma. A Phase I Study. (PARADIGMA) [NCT03480867]Phase 10 participants (Actual)Interventional2017-03-31Withdrawn(stopped due to competing study was opened by the surgeon after this trial was opened)
A Phase 1/1b Adaptive Dose Escalation Study of Mycophenolate Mofetil (MMF) in Combination With Standard of Care for Patients With Glioblastoma [NCT05236036]Phase 160 participants (Anticipated)Interventional2022-08-08Recruiting
An Extension Study to Evaluate the Safety of Veliparib in Combination With Temozolomide in Subjects With Solid Tumors. [NCT01193140]Phase 224 participants (Actual)Interventional2010-07-31Completed
Phase I/II Study of LY2228820 With Radiotherapy Plus Concomitant TMZ in the Treatment of Newly Diagnosed Glioblastoma [NCT02364206]Phase 1/Phase 218 participants (Actual)Interventional2015-06-08Completed
Efficacy and Safety of Apatinib Combined With Dose-dense Temozolomide in Recurrent Glioblastoma [NCT03660761]Phase 225 participants (Actual)Interventional2016-03-03Completed
A Large-scale Research for Immunotherapy of Glioblastoma With Autologous Heat Shock Protein gp96 [NCT03650257]Phase 2150 participants (Anticipated)Interventional2019-08-21Recruiting
Bortezomib Sensitization of Recurrent Glioblastoma With Unmethylated MGMT Promoter to Temozolomide Phase 1B/II Study [NCT03643549]Phase 1/Phase 263 participants (Anticipated)Interventional2018-08-30Recruiting
Individualized (Timed) Temozolomide Administration as a Means of Immune Reconstitution in Patients With Metastatic Melanoma [NCT01328535]Phase 225 participants (Actual)Interventional2011-01-31Completed
Randomized, Phase II, Double-Blind, Placebo-Controlled Trial of Conventional Chemoradiation and Adjuvant Temozolomide Plus Cediranib Versus Conventional Chemoradiation and Adjuvant Temozolomide Plus Placebo in Patients With Newly Diagnosed Glioblastoma [NCT01062425]Phase 2261 participants (Actual)Interventional2010-02-26Completed
A Phase 2 Study of Apatinib Combined With Temozolomide in the Treatment of Advanced Melanoma Patients After Conventional Treatment Failure [NCT03422445]Phase 230 participants (Anticipated)Interventional2018-01-08Recruiting
Randomized Double-Blind, Placebo-Controlled Parallel Multi-Center Study to Assess the Efficacy of Cannabidiol (BRCX014) Combined With Standard-Of-Care Treatment in Subjects With Multiple Myeloma, Glioblastoma Multiforme, and GI Malignancies [NCT03607643]Phase 1/Phase 2160 participants (Anticipated)Interventional2019-01-15Not yet recruiting
Prospective Study of Stereotactic Radiosurgery Using Diffusion-Weighted Abnormality Versus Chemotherapy for Recurrent/Progressive Glioblastoma After Second-line Chemotherapy [NCT05718466]Phase 335 participants (Actual)Interventional2010-11-30Completed
Phase II, Multicentre, Open Label Study to Evaluate the Efficacy of the Combination of Lanreotide Autogel 120mg and Temozolomide in Patients With Progressive Gastro-entero-pancreatic Neuroendocrine Tumours (GEP-NET) G1/G2 - A Pilot-Study [NCT02231762]Phase 257 participants (Actual)Interventional2014-10-31Completed
Phase I Pilot Study of Preoperative Chemoradiation for Glioblastoma [NCT02092038]Phase 11 participants (Actual)Interventional2014-11-30Completed
Phase II Trial of Temozolomide for the Treatment of Mycosis Fungoides and the Sezary Syndrome [NCT00004106]Phase 229 participants (Actual)Interventional1998-05-31Terminated(stopped due to Per Data Monitoring Committee given the poor/inadequate accrual.)
Phase I/II Trial of Concurrent RAD001 (Everolimus) With Temozolomide/Radiation Followed by Adjuvant RAD001/Temozolomide in Newly Diagnosed Glioblastoma [NCT01062399]Phase 1/Phase 2279 participants (Actual)Interventional2010-12-31Completed
Phase II Study of Anti-GD2 3F8 Antibody and GM-CSF for High-Risk Neuroblastoma [NCT00072358]Phase 2291 participants (Actual)Interventional2003-07-31Completed
A Study on the Safety and Effectiveness of Temozolomide for Neoadjuvant Treatment of Pheochromocytoma or Paraganglioma Patients [NCT05885386]Phase 220 participants (Anticipated)Interventional2023-04-01Recruiting
Phase I/Ib Study of Sarasar and Temodar in Patients With Recurrent or Temodar-Refractory Glioblastoma Multiforme [NCT00102648]Phase 135 participants (Anticipated)Interventional2004-12-21Active, not recruiting
A Study of BPM31510 With Vitamin K1 in Subjects With Newly Diagnosed Glioblastoma (GB) [NCT04752813]Phase 250 participants (Anticipated)Interventional2023-09-28Recruiting
Phase I/II Study of BGB-290 With Temozolomide in Recurrent Gliomas With IDH1/2 Mutations [NCT03914742]Phase 1/Phase 260 participants (Actual)Interventional2020-01-03Suspended(stopped due to NCI decided to terminate ABTC Consortium due to NCI moving in different direction for Brain Cancer])
A Phase II, Multicenter, Open-Label, Single-Arm Study to Evaluate the Safety, Tolerability, and Efficacy of DIsulfiram and Copper Gluconate in Recurrent Glioblastoma [NCT03034135]Phase 223 participants (Actual)Interventional2017-03-09Completed
A Retrospective Clinical Study of Postoperative Concurrent Chemoradiotherapy Combined With Anti-angiogenic Drugs in the Treatment of Glioblastoma. [NCT03567135]60 participants (Anticipated)Observational2017-10-01Recruiting
Phase 2 Study of Intraventricular Omburtamab-based Radioimmunotherapy for Pediatric Patients With Recurrent Medulloblastoma and Ependymoma [NCT04743661]Phase 262 participants (Anticipated)Interventional2022-04-04Active, not recruiting
A Prospective Cohort to Study the Effect of Postoperative Upfront Temozolomide Chemotherapy on IDH Mutational Low Grade Gliomas in Eloquent Areas [NCT02209428]Phase 254 participants (Actual)Interventional2014-06-30Active, not recruiting
A Randomized Phase II Study of Temozolomide Monotherapy or in Combination With Olaparib in Patients With METHYLATED 06-Methylguanine-DNA Methyltransferase (MGMT) Pre-Treated Triple Negative Breast Cancer (TNBC) [NCT05128734]Phase 240 participants (Anticipated)Interventional2023-12-01Not yet recruiting
A Phase I Trial of Chlorpromazine Together With Standard of Care in New Diagnosis of Glioblastoma [NCT05190315]Phase 110 participants (Actual)Interventional2022-01-28Active, not recruiting
Phase 2 Trial of a Novel Peptide Vaccine (PEP-CMV) Targeting CMV Antigen for Newly Diagnosed Pediatric High-grade Glioma and Diffuse Intrinsic Pontine Glioma and Recurrent Medulloblastoma [NCT05096481]Phase 2120 participants (Anticipated)Interventional2023-12-15Not yet recruiting
Efficacy and Safety of Anlotinib, Irinotecan and Temozolomide in the Treatment of Refractory or Recurrent Neuroblastoma in Children: an Open, Single Arm, Single Center, Phase II Clinical Study [NCT04842526]Phase 227 participants (Anticipated)Interventional2021-04-12Not yet recruiting
A Phase 1b/2, Dose-finding and Expansion Study to Assess the Safety, Tolerability, Pharmacokinetics and Preliminary Efficacy of HTMC0435 With Temozolomide in Patients With Small Cell Lung Cancer [NCT05728619]Phase 1/Phase 264 participants (Anticipated)Interventional2023-02-03Recruiting
A Randomized, Phase I/II Trial of Irinotecan and Temozolomide Compared to Irinotecan and Temozolomide in Combination With TPI 287 in Patients With Primary Refractory or Early Relapsed Neuroblastoma [NCT01505608]Phase 1/Phase 214 participants (Actual)Interventional2011-12-31Terminated(stopped due to Lack of Enrollment)
Randomized Phase III Study of Conventional Versus Hypofractionated Radiotherapy Combined With Temozolomide in Elderly Glioblastoma Patients [NCT05439278]Phase 3268 participants (Anticipated)Interventional2022-08-01Not yet recruiting
Improvement of Functional Outcome for Patients With Newly Diagnosed Grade II or III Glioma With Co-deletion of 1p/19q - IMPROVE CODEL: the NOA-18 Trial [NCT05331521]Phase 3406 participants (Anticipated)Interventional2021-04-07Recruiting
Preoperative Radiosurgery in High Grade Glioma: A Phase I/IIA Clinical Trial: The NeoGlioma Study [NCT05030298]Phase 1/Phase 240 participants (Anticipated)Interventional2023-05-23Recruiting
An Open Label Single Arm Phase II Trial in Patients With Advanced Unresectable Previously Treated Oesophagogastric Adenocarcinoma Which is MGMT Deficient [NCT04984733]Phase 218 participants (Anticipated)Interventional2021-09-28Recruiting
A Phase I Clinical Trial of Selinexor (KPT-330) in Combination With Temozolomide and Radiation Therapy in Patients With Newly Diagnosed Glioblastoma [NCT04216329]Phase 124 participants (Anticipated)Interventional2020-07-07Recruiting
A Multicenter, Randomized, Controlled, Phase III Trial Comparing High-Dose IFN-a2b With Temozolomide Plus Cisplatin as Systemic Adjuvant Therapy for Resected Mucosal Melanoma [NCT03435302]Phase 3204 participants (Anticipated)Interventional2014-02-28Recruiting
A Phase 2, Randomized, Open-Label Study of Anhydrous Enol-Oxaloacetate in Subjects With Newly Diagnosed Glioblastoma Multiforme [NCT04450160]Phase 280 participants (Anticipated)Interventional2020-12-31Not yet recruiting
Phase II Single-Center Study of Bevacizumab in Combination With Temozolomide in Patients With First-Line Metastatic Uveal Melanoma [NCT01217398]Phase 235 participants (Anticipated)Interventional2009-10-31Recruiting
Cilengitide and Metronomic Temozolomide for Relapsed or Refractory High Grade Gliomas or Diffuse Intrinsic Pontine Gliomas in Children and Adolescents - A Phase II Study HIT-HGG-CilMetro - A Clinical Phase II Trial of the HIT-HGG Study Group - [NCT01517776]Phase 228 participants (Actual)Interventional2012-01-31Terminated(stopped due to due to an altered benefit/risk assessment.)
A Pilot Study Using Carboplatin, Vincristine And Temozolomide For Children ≤ 10 Years With Progressive/Symptomatic Low-Grade Gliomas [NCT00077207]66 participants (Actual)Interventional2004-07-31Completed
A Phase Ib/II Study of AZD2171 in Combination With Daily Temozolomide and Radiation in Patients With Newly Diagnosed Glioblastoma Not Taking Enzyme-Inducing Anti-epileptic Drugs [NCT00662506]Phase 1/Phase 246 participants (Actual)Interventional2008-04-30Completed
A Phase I Trial of High-Dose Ascorbate in Glioblastoma Multiforme [NCT01752491]Phase 113 participants (Actual)Interventional2013-04-01Active, not recruiting
A Phase II Trial of Talampanel in Conjunction With Radiation Therapy With Concurrent and Adjuvant Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00267592]Phase 272 participants (Actual)Interventional2005-12-31Completed
A Phase Ⅰb/Ⅱ Study of Fluzoparib (SHR-3162) and Temozolomide With or Without SHR-1316 in Treating Patients With Replapsed Small Cell Lung Cancer [NCT04400188]Phase 1/Phase 225 participants (Actual)Interventional2020-06-11Active, not recruiting
Bioequivalence Study Between Temozolomide Oral Suspension (Ped-TMZ) and Temodal® Capsules [NCT04467346]Phase 136 participants (Actual)Interventional2020-09-25Completed
Comparative Assessment of Methods to Analyze MGMT as a Predictive Factor of Response to Temozolomide in Glioblastomas. [NCT01345370]300 participants (Actual)Observational2009-03-31Completed
Phase II Trial O6-benzylguanine(BG) and Temozolomide(TMZ) Therapy of Glioblastoma Multiforme (GBM) With Infusion of Autologous P140K MGMT+Hematopoietic Progenitors to Protect Hematopoiesis [NCT05052957]Phase 216 participants (Anticipated)Interventional2023-01-20Recruiting
Phase 1/2 Study of TPX-0005 (Repotrectinib) in Combination With Chemotherapy in Pediatric and Young Adult Subjects With Advanced or Metastatic Solid Tumors and Primary Central Nervous System Tumors [NCT05004116]Phase 1/Phase 250 participants (Anticipated)Interventional2021-08-09Recruiting
A Phase 1-2 Dose-escalation and Expansion Study of ST101 in Patients With Advanced Unresectable and Metastatic Solid Tumors [NCT04478279]Phase 1/Phase 2162 participants (Anticipated)Interventional2020-07-01Recruiting
A Pilot Study of Dinutuximab, Sargramostim (GM-CSF), and Isotretinoin in Combination With Irinotecan and Temozolomide in the Post-Consolidation Setting for High-Risk Neuroblastoma [NCT04385277]Phase 234 participants (Actual)Interventional2020-12-31Active, not recruiting
A Phase 2 Randomized Study of Irinotecan/Temozolomide/Dinutuximab With or Without Eflornithine (DFMO) in Children With Relapsed, Refractory or Progressive Neuroblastoma [NCT03794349]Phase 295 participants (Anticipated)Interventional2019-07-08Recruiting
A Phase II/III Randomized Trial of Veliparib or Placebo in Combination With Adjuvant Temozolomide in Newly Diagnosed Glioblastoma With MGMT Promoter Hypermethylation [NCT02152982]Phase 2/Phase 3447 participants (Actual)Interventional2014-12-15Active, not recruiting
A Randomized Phase II/III Study of Vorinostat and Local Irradiation OR Temozolomide and Local Irradiation OR Bevacizumab and Local Irradiation Followed by Maintenance Bevacizumab and Temozolomide in Children With Newly Diagnosed High-Grade Gliomas [NCT01236560]Phase 2/Phase 3101 participants (Actual)Interventional2010-11-15Active, not recruiting
Quality of Life and Neurocognitive Functioning in Diffuse Low-grade Glioma (TEMOIN) [NCT03257618]26 participants (Actual)Interventional2017-07-27Completed
Phase III Trial of CCNU/Temozolomide (TMZ) Combination Therapy vs. Standard TMZ Therapy for Newly Diagnosed MGMT-methylated Glioblastoma Patients [NCT01149109]Phase 3141 participants (Actual)Interventional2010-10-31Completed
A Randomized Phase 3 Study of Antineoplastons A10 and AS2-1 vs. Temozolomide in Subjects With Recurrent and / or Progressive Optic Pathway Glioma After Carboplatin or Cisplatin Therapy [NCT01260103]Phase 30 participants (Actual)Interventional2011-12-31Withdrawn(stopped due to Withdrawn: study halted prematurely, prior to enrollment of first participant)
Phase I/II Study of AGuIX Nanoparticles With Radiotherapy Plus Concomitant Temozolomide in the Treatment of Newly Diagnosed Glioblastoma [NCT04881032]Phase 1/Phase 266 participants (Anticipated)Interventional2022-03-07Recruiting
A Phase I Study of MK-4827 in Combination With Temozolomide in Patients With Advanced Cancer [NCT01294735]Phase 119 participants (Actual)Interventional2011-02-28Completed
A Phase II Open Label Randomised Multicentric Study in Patients With Unresectable Glioblastoma Using Neo-adjuvant Treatment With Two Cycles of Temozolomide Previous Temozolomide Plus Radiation Therapy and Adjuvant Temozolomide vs. Neo-adjuvant Treatment W [NCT01102595]Phase 2102 participants (Actual)Interventional2009-12-31Completed
Phase I Trial of Oral Topotecan Plus Temodar in the Treatment of Patients With Malignant Gliomas [NCT00610571]Phase 162 participants (Actual)Interventional2004-04-30Completed
Phase 1 Trial of RO4929097 in Combination With Standard Radiotherapy and Temozolomide for Newly Diagnosed Malignant Glioma: A Pharmacokinetic and Pharmacodynamic Study [NCT01119599]Phase 122 participants (Actual)Interventional2010-05-31Completed
First-line Chemotherapy With Temozolomide Alone for Non-enhancing Adult Brainstem Gliomas, With a Diffuse Subtype and Showing Clinical and/or Radiological Infiltrative Pattern of Progression [NCT03932981]Phase 260 participants (Anticipated)Interventional2019-07-26Recruiting
Irinotecan and Temozolomide for Advanced Ewing Sarcoma After Failure of Standard Multimodal Therapy [NCT03359005]Phase 260 participants (Anticipated)Interventional2018-02-07Recruiting
NIVOLUMAB Plus IPILIMUMAB and TEMOZOLOMIDE in Combination in Microsatellite Stable (MSS), MGMT Silenced Metastatic Colorectal Cancer (mCRC): the MAYA Study [NCT03832621]Phase 2135 participants (Actual)Interventional2019-03-25Completed
Early Post-surgical Temozolomide Therapy in Patients With High-Grade Gliomas Admitted to Acute Rehabilitation: A Feasibility Study [NCT03796507]Early Phase 10 participants (Actual)Interventional2021-09-01Withdrawn(stopped due to Hiring freeze of research personnel due to COVID pandemic)
Randomized Phase II Study Comparing Two Schedules Of Temozolomide In Combination With Bay 43-9006 In Patients With Advanced Melanoma [NCT00602576]Phase 2169 participants (Actual)Interventional2005-01-01Completed
A Phase II Study of a Temozolomide-Based Chemoradiotherapy Regimen for High-Risk Low-Grade Gliomas [NCT00114140]Phase 2136 participants (Actual)Interventional2005-01-31Completed
A Multicenter Randomized Phase III Study for Recurrent Glioblastoma Comparing Bevacizumab Alone With Dose-dense Temozolomide Followed by Bevacizumab (JCOG1308C, RE-GEND-pIII) [NCT02761070]Phase 3146 participants (Anticipated)Interventional2016-07-11Active, not recruiting
Study of TG02 in Elderly Newly Diagnosed or Adult Relapsed Patients With Anaplastic Astrocytoma or Glioblastoma: A Phase Ib Study [NCT03224104]Phase 171 participants (Actual)Interventional2018-06-12Completed
Dose Finding Phase 1 Study of the Treatment of Metastatic Melanoma With MPC-6827 in Combination With Temozolomide [NCT00609011]Phase 1/Phase 222 participants (Actual)Interventional2008-03-31Completed
Efficacy and Safety of Nimotuzumab in Addition to Radiotherapy and Temozolomide for Cerebral Glioblastoma [NCT03388372]Phase 239 participants (Actual)Interventional2010-08-18Completed
A Randomized Phase II Trial of Capecitabine and Temozolomide (CAPTEM) or FOLFIRI as SEcond-line Therapy in NEuroendocrine CArcinomas and Exploratory Analysis of Predictive Role of Positron Emission Tomography (PET) Imaging and Biological Markers [NCT03387592]Phase 2112 participants (Anticipated)Interventional2017-03-06Recruiting
A Pilot Study to Assess the Safety, Feasibility, and Preliminary Efficacy of a Neoepitope-based Personalized Vaccine Approach in Patients With Newly Diagnosed Glioblastoma [NCT02510950]Phase 11 participants (Actual)Interventional2015-12-03Terminated(stopped due to Low accrual)
A Study of CIK in Combination With Temozolomide With and Without Radiation in Adults With Stage I-II Malignant Gliomas [NCT02494804]Phase 1/Phase 280 participants (Anticipated)Interventional2015-07-31Not yet recruiting
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
An Open-Label, Multi-Center Trial of INO-5401 and INO-9012 Delivered by Electroporation (EP) in Combination With REGN2810 in Subjects With Newly-Diagnosed Glioblastoma (GBM) [NCT03491683]Phase 1/Phase 252 participants (Actual)Interventional2018-05-31Active, not recruiting
Randomized Phase 2 Trial Of Two Chemotherapy Regimens Plus Or Minus Bevacizumab In Patients With Well Differentiated Pancreatic Neuroendocrine Tumors [NCT03351296]Phase 2140 participants (Anticipated)Interventional2018-06-26Recruiting
A Randomized, Double Blind Phase II Trial of Surgery, Radiation Therapy Plus Temozolomide and Pembrolizumab With and Without HSPPC-96 in Newly Diagnosed Glioblastoma (GBM) [NCT03018288]Phase 290 participants (Actual)Interventional2017-09-21Completed
A Study of CIK in Combination With Temozolomide With and Without Radiation in Adults With Advanced Malignant Gliomas [NCT02496988]Phase 4120 participants (Anticipated)Interventional2015-07-31Not yet recruiting
A Prospective Trial of NovoTTF-200A Together With Temozolomide and Radiotherapy in Patients With Newly Diagnosed GBM [NCT03780569]10 participants (Anticipated)Interventional2017-04-27Active, not recruiting
Hypofractionated Radiotherapy With Concurrent Temozolomide for Large Brain Metastases: a Multi-center Randomized Phase III Trial [NCT03778541]Phase 3224 participants (Anticipated)Interventional2018-12-03Recruiting
Phase II Clinical Trial of Concomitant Association of Ultrafractionated Brain Irradiation - Temozolomide in Inoperable Primary Glioblastoma Multiforme [NCT03310372]Phase 246 participants (Actual)Interventional2008-02-13Completed
Short Course Chemo-Radiation Therapy for Patients With Newly Diagnosed Glioblastoma: Comparison of Two Established Schedules [NCT04019262]Phase 1/Phase 240 participants (Anticipated)Interventional2021-12-14Recruiting
An Open Label, Multicenter, Phase II Study Evaluating the Safety and Efficacy of Temozolomide Treatment in Patients With Invasive Pituitary Tumors [NCT00601289]Phase 20 participants (Actual)Interventional2009-12-31Withdrawn(stopped due to funding term ended)
A Randomized Phase I/II Study of ABT-888 in Combination With Temozolomide in Recurrent (Temozolomide Resistant) Glioblastoma [NCT01026493]Phase 1/Phase 2257 participants (Actual)Interventional2010-07-31Completed
A Phase 1/2 Open-label Study of Debio 0123 in Combination With Temozolomide in Adult Participants With Recurrent or Progressive Glioblastoma and of Debio 0123 in Combination With Temozolomide and Radiotherapy in Adult Participants With Newly Diagnosed Gli [NCT05765812]Phase 1/Phase 289 participants (Anticipated)Interventional2023-03-01Recruiting
A Phase 2a Study of the Addition of Temozolomide to a Standard Conditioning Regimen for Autologous Stem Cell Transplantation in Relapsed and Refractory Central Nervous System (CNS) Lymphoma [NCT01235793]Phase 211 participants (Actual)Interventional2010-10-14Terminated(stopped due to The clinical trial was terminated due to poor enrollment)
Phase I, Open Label Trial to Explore Safety of Combining BIBW 2992 and Radiotherapy With or Without Temozolomide in Newly Diagnosed GBM [NCT00977431]Phase 136 participants (Actual)Interventional2009-09-17Completed
A Phase I Study of Safety and Tolerability of Acetazolamide With Temozolomide in Adults With Newly Diagnosed MGMT Promoter-Methylated IDH Wildtype Glioblastoma [NCT03011671]Phase 160 participants (Anticipated)Interventional2018-10-03Recruiting
CPT-SIOP-2009: Intercontinental Multidisciplinary Registry and Treatment Optimization Study for Patients With Choroid Plexus Tumors [NCT01014767]Phase 327 participants (Actual)Interventional2009-11-30Terminated(stopped due to PI departure from coordinating institution)
Phase II Trial of Bevacizumab in Combination With Temozolomide and Regional Radiation Therapy for Upfront Treatment of Patients With Newly-diagnosed Glioblastoma Multiforme [NCT01013285]Phase 270 participants (Actual)Interventional2006-06-30Completed
A Phase I Study of Hydroxychloroquine in Combination With Temozolomide in Patients With Advanced Solid Tumors [NCT00714181]Phase 138 participants (Actual)Interventional2008-06-30Completed
Therapeutic Trial for Patients With Ewing Sarcoma Family of Tumor and Desmoplastic Small Round Cell Tumors [NCT01946529]Phase 224 participants (Actual)Interventional2013-12-27Active, not recruiting
Phase II Portion of Multi Phase Study of Sorafenib With Radiation and Temozolomide in Newly Diagnosed Glioblastoma or Gliosarcoma [NCT02599090]Phase 20 participants (Actual)Interventional2008-12-31Withdrawn(stopped due to Multi phase study did not progress to Phase II. Phase I registered as NCT00734526.)
Prospective Pilot Trial to Assess a Multimodal Molecular Targeted Therapy in Children, Adolescent and Young Adults With Relapsed or Refractory High-grade Pineoblastoma [NCT02596828]Phase 24 participants (Actual)Interventional2016-04-30Completed
Cilengitide for Subjects With Newly Diagnosed Glioblastoma and Methylated MGMT Gene Promoter - A Multicenter, Open-label, Controlled Phase III Study, Testing Cilengitide in Combination With Standard Treatment (Temozolomide With Concomitant Radiation Thera [NCT00689221]Phase 3545 participants (Actual)Interventional2008-09-30Completed
A Clinical Study of Standard TEMODAL® Regimen Versus Standard Regimen Plus Early Post-Surgery TEMODAL® Chemotherapy in Treatment on Patients With Newly Diagnosed Glioblastoma Multiforme (GBM) [NCT00686725]Phase 499 participants (Actual)Interventional2008-06-24Completed
A Randomized, Double-Blind, Placebo-Controlled, Multicenter Phase III Trial of Bevacizumab, Temozolomide and Radiotherapy, Followed by Bevacizumab and Temozolomide Versus Placebo, Temozolomide and Radiotherapy Followed by Placebo and Temozolomide in Patie [NCT00943826]Phase 3921 participants (Actual)Interventional2009-06-29Completed
Open-label, Single Center, Uncontrolled Phase I/II Study Evaluating the Safety and Maximum Tolerated Dose of Daily Sorafenib Administered in Combination With Prolonged Temozolomide in Patients With Metastatic Melanoma [NCT00811759]Phase 1/Phase 258 participants (Anticipated)Interventional2007-06-30Active, not recruiting
Phase Ib Study To Evaluate The Safety Of Combining IGF-1R Antagonist R1507 With Multiple Standard Chemotherapy Drug Treatments In Patients With Advanced Malignancies [NCT00811993]Phase 1104 participants (Actual)Interventional2009-02-28Terminated
Randomized Study of Normal-fractionated Radiotherapy Versus Hypofractionated Radiotherapy Versus Chemotherapy in Patients Over 60 Years With Malignant Glioma [NCT00820963]Phase 370 participants (Anticipated)Interventional2006-07-31Completed
A First-in-Human Clinical Trial of Pharmacologic Ascorbate and Ferumoxytol Combined With Concomitant Temozolomide and External Beam Radiation Therapy for Newly Diagnosed Glioblastoma [NCT04900792]Phase 112 participants (Anticipated)Interventional2023-02-28Active, not recruiting
A Randomized Phase I/II Study of Talazoparib or Temozolomide in Combination With Onivyde in Children With Recurrent Solid Malignancies and Ewing Sarcoma [NCT04901702]Phase 1/Phase 2160 participants (Anticipated)Interventional2021-06-09Recruiting
Phase II Study of Short Course Hypofractionated Proton Beam Therapy Incorporating 18F-DOPA-PET/MRI for Elderly Patients With Newly Diagnosed Glioblastoma [NCT03778294]Phase 243 participants (Actual)Interventional2019-03-28Completed
A Phase I Study of Temozolomide and RAD001C in Patients With Malignant Glioblastoma Multiforme [NCT00387400]Phase 132 participants (Actual)Interventional2007-03-20Completed
Efficacy and Tolerability of Cisplatin Plus Alternating Weekly Temozolomide in Recurrent High-grade Gliomas: A Single-arm Prospective Phase II Clinical Study [NCT02263105]Phase 267 participants (Actual)Interventional2014-10-31Completed
Pilot Study of Concomitant NovoTTF-200A and Temozolomide Chemoradiation for Newly Diagnosed Glioblastoma [NCT03232424]Phase 112 participants (Actual)Interventional2017-07-26Completed
Phase I/II Study to Evaluate the Safety and Clinical Efficacy of Atezolizumab (aPDL1) in Combination With Temozolomide and Radiation in Patients With Newly Diagnosed Glioblastoma (GBM) [NCT03174197]Phase 1/Phase 280 participants (Actual)Interventional2017-06-30Active, not recruiting
A Phase 1b, Open-label, Dose-Finding Study of CC-90010 in Combination With Temozolomide With or Without Radiation Therapy in Subjects With Newly Diagnosed Glioblastoma [NCT04324840]Phase 1194 participants (Anticipated)Interventional2020-07-10Active, not recruiting
A Phase II Trial of Irinotecan and Temozolomide in Combination With Existing High Dose Alkylator Based Chemotherapy for Treatment of Patients With Newly Diagnosed Ewing Sarcoma [NCT01864109]Phase 283 participants (Actual)Interventional2013-05-31Active, not recruiting
A Phase 2 Study of ABT-888 and Temozolomide for Metastatic Breast Cancer and an Expansion Cohort in BRCA1/2 Mutation Carriers [NCT01009788]Phase 264 participants (Actual)Interventional2009-11-30Active, not recruiting
Phase 3b Study for Management of Ocular Side Effects in Subjects With EGFR-amplified Glioblastoma Receiving Depatuxizumab Mafodotin (ABT-414) [NCT03419403]Phase 340 participants (Actual)Interventional2018-07-30Terminated(stopped due to The study was terminated because clinical development of depatuxizumab mafodotin in glioblastoma was stopped due to lack of survival benefit.)
A Phase II Study of Concurrent Radiation and Temozolomide Followed By Temozolomide and CCNU in the Treatment of Children With High-Grade Glioma [NCT00100802]Phase 2118 participants (Actual)Interventional2005-03-21Completed
Addition of Anlotinib Hydrochloride to the Stupp Regimen Versus the Stupp Regimen Alone for Newly Diagnosed Glioblastoma: A Randomized Double-blind Multicenter Prospective Phase II Study [NCT04959500]Phase 2150 participants (Anticipated)Interventional2021-06-10Recruiting
Application of Proteome Profiler Antibody Arrays to Find Angiogenetic Predictors in Glioma Paitents. [NCT03225963]60 participants (Anticipated)Observational2015-08-01Recruiting
INTELLANCE-2: ABT-414 Alone or ABT-414 Plus Temozolomide Versus Lomustine or Temozolomide for Recurrent Glioblastoma: A Randomized Phase 2 Study of the EORTC Brain Tumor Group [NCT02343406]Phase 2266 participants (Actual)Interventional2015-02-17Completed
Iodine-125 Brachytherapy Together With Chemotherapy Compared With Surgical Resection Followed by Concomitant Radiochemotherapy in Patients With Newly Diagnosed Glioblastoma,a Randomized, Open-label, Multi-center Clinical Trial [NCT04856852]84 participants (Anticipated)Interventional2021-06-28Not yet recruiting
A Phase 1 Study to Assess BDTX-1535, an Oral EGFR Inhibitor, in Patients With Glioblastoma or Non-Small Cell Lung Cancer [NCT05256290]Phase 1120 participants (Anticipated)Interventional2022-03-31Recruiting
A Phase 3 Study of Dinutuximab Added to Intensive Multimodal Therapy for Children With Newly Diagnosed High-Risk Neuroblastoma [NCT06172296]Phase 3478 participants (Anticipated)Interventional2024-02-14Not yet recruiting
A Phase II, Open-label, Single-arm, Multi-center Pilot Study to Evaluate the Efficacy and Safety of Whole-brain Radiotherapy With Concomitant Temozolomide in Lung Cancer and Breast Cancer Patients With Brain Metastases [NCT02133677]Phase 224 participants (Anticipated)Interventional2014-05-31Not yet recruiting
A Randomized, Controlled, Open, Multicenter, Phase II/III Clinical Study to Evaluate the Safety and Efficacy of Vebreltinib Enteric Capsules in the Treatment of sGBM/IDH Mutant Glioblastoma Patients With the ZM Fusion Gene (FUGEN). [NCT06105619]Phase 2/Phase 384 participants (Actual)Interventional2018-10-08Active, not recruiting
A Phase Ib Trial of Eribulin in Combination With Irinotecan and Temozolamide in Children With Relapsed or Refractory Solid Tumors [NCT06006273]Phase 1/Phase 248 participants (Anticipated)Interventional2023-08-16Recruiting
CAMPFIRE: Children's and Young Adult Master Protocol for Innovative Pediatric Research [NCT05999994]Phase 2105 participants (Anticipated)Interventional2020-01-22Recruiting
A Phase II Study of Temozolomide and Everolimus (RAD001) Therapy for Metastatic Melanoma [NCT00521001]Phase 249 participants (Actual)Interventional2008-01-31Completed
Preoperative Chemoradiotherapy With Capecitabine With or Without Temozolomide in Patients With Locally Advanced Rectal Cancer; A Prospective Randomised Phase 2 Study Stratified by MGMT (O6-methylguanine DNA Methyltransferase) Status [NCT03156036]Phase 264 participants (Actual)Interventional2017-11-30Completed
A Phase III Trial Comparing Whole Brain Radiation and Stereotactic Radiosurgery Alone Versus With Temozolomide or Erlotinib in Patients With Non-Small Cell Lung Cancer and 1-3 Brain Metastases [NCT00096265]Phase 3126 participants (Actual)Interventional2004-10-06Terminated
A Phase II Study of Pembrolizumab, Olaparib, and Temozolomide in Patients With Glioma [NCT05188508]Phase 257 participants (Anticipated)Interventional2022-01-14Recruiting
A Randomized Phase II Study of Temozolomide or Temozolomide and Capecitabine in Patients With Advanced Pancreatic Neuroendocrine Tumors [NCT01824875]Phase 2144 participants (Actual)Interventional2013-08-08Active, not recruiting
Phase I/II Study of ZD6474 (Vandetanib) With Radiation Therapy and Concomitant and Adjuvant Temozolomide in Patients With Newly-Diagnosed Glioblastoma [NCT00441142]Phase 1/Phase 2119 participants (Actual)Interventional2007-05-25Completed
Therapy-Optimization Trial and Phase II Study for the Treatment of Relapsed or Refractory of Primitive Neuroectodermal Brain Tumors and Ependymomas in Children and Adolescents [NCT00749723]Phase 2/Phase 3174 participants (Actual)Interventional2006-02-01Completed
Phase I/II Trial for Vaccine Therapy With Dendritic Cells - Transfected With hTERT-, Survivin- and Tumor Cell Derived mRNA + ex Vivo T Cell Expansion and Reinfusion in Patients With Metastatic Malignant Melanoma [NCT00961844]Phase 1/Phase 215 participants (Actual)Interventional2009-08-31Terminated(stopped due to Logistical problems)
A Phase 1 Dose Finding Study of the Safety and Pharmacokinetics of XL184 Administered Orally in Combination With Temozolomide and Radiation Therapy in the First Line Treatment of Subjects With Glioblastoma [NCT00960492]Phase 126 participants (Actual)Interventional2009-09-30Completed
Phase II Study of Temozolomide in Previously Untreated Acute Myeloid Leukemia (AML)/Myelodysplastic Syndrome (MDS) Subjects Unsuitable for Standard Induction Therapy Exhibiting Low MGMT Expression [NCT00687323]Phase 247 participants (Actual)Interventional2007-07-30Completed
Avastin in Combination With Radiation and Temozolomide Followed by Avastin, Temozolomide, and Topotecan for Glioblastoma Multiformes and Gliosarcomas [NCT01004874]Phase 280 participants (Actual)Interventional2009-12-30Completed
A Multi-centre, Two-arm, Randomized, Open, Phase II Study Investigating SentoClone® Compared to Reference Treatment in Advanced Malignant Melanoma [NCT00991250]Phase 2140 participants (Anticipated)Interventional2009-10-31Recruiting
A Phase I/II Trial of Temozolomide and ABT-888 in Subjects With Newly Diagnosed Glioblastoma Multiforme [NCT00770471]Phase 124 participants (Actual)Interventional2009-07-13Completed
Phase 1, Open Label, Multi-Center Study To Evaluate The Safety And Tolerability of CT-322 Administered In Combination With Focal Brain Radiotherapy And Temozolomide To Subjects With Newly Diagnosed Glioblastoma Multiforme [NCT00768911]Phase 130 participants (Anticipated)Interventional2008-10-31Active, not recruiting
"Phase II Pilot Trial of Chemo-Switch Regimen of Biochemotherapy Followed by Daily Low-Dose Temozolomide Plus Sorafenib in Advanced Melanoma" [NCT00673361]Phase 29 participants (Actual)Interventional2007-03-31Terminated
Dose Escalation Radiotherapy With Modulation of Intensity and Integrated Boost in Association With a Temozolomide in the Treatment of Glioblastoma [NCT01043536]Phase 110 participants (Actual)Interventional2009-09-30Completed
A Phase I/II Trial of Hydroxychloroquine in Conjunction With Radiation Therapy and Concurrent and Adjuvant Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00486603]Phase 1/Phase 292 participants (Actual)Interventional2007-10-29Completed
Releasing the Cancer Patient's Immune System From Down-regulation With Timed Delivery of Standard Chemotherapy [NCT00568451]Phase 212 participants (Actual)Interventional2006-06-30Terminated(stopped due to Slow accrual.)
A Randomized Phase II Trial of Temozolomide (TMZ) and Bevacizumab or ABI-007 (ABX)/Carboplatin (CBDCA) and Bevacizumab in Patients With Unresectable Stage IV Malignant Melanoma [NCT00626405]Phase 295 participants (Actual)Interventional2008-08-31Completed
A Phase I/II Trial Of Temozolomide And Vinorelbine For Patients With Recurrent Brain Metastases [NCT00026494]Phase 1/Phase 249 participants (Actual)Interventional2001-07-31Completed
A Phase II Study of the Safety and Efficacy of SVN53-67/M57-KLH (SurVaxM) in Survivin-Positive Newly Diagnosed Glioblastoma [NCT02455557]Phase 266 participants (Actual)Interventional2015-05-04Active, not recruiting
A Phase 2, Multiple-Cohort, Open-Label, International Study of Talazoparib Monotherapy and Talazoparib Plus Temozolomide in Women With Relapsed Ovarian, Fallopian Tube, and Peritoneal Cancer [NCT02836028]Phase 20 participants (Actual)Interventional2016-10-31Withdrawn
A Phase 1/2, Open-label, Dose-exploration and Expansion Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of NTX-301 Monotherapy in Advanced Solid Tumours, and in Combination With Platinum-based Chemotherapy in Advanced Ova [NCT04851834]Phase 1/Phase 212 participants (Actual)Interventional2021-08-25Terminated(stopped due to Study was terminated by IP Holder (collaborator), PinotBio Inc.)
A Prospective Study of Concurrent Chemoradiotherapy With Temozolomide Versus Radiation Therapy Alone in Patients With IDH Wild-type/TERT Promoter Mutation Grade II/III Gliomas [NCT02766270]Early Phase 130 participants (Anticipated)Interventional2016-09-26Recruiting
Radiotherapy Plus Concomitant Temozolomide for Refractory Pituitary Adenomas,A Randomized,Double-blind, Placebo-controlled Phase II Trial [NCT04244708]Phase 2/Phase 3150 participants (Anticipated)Interventional2020-02-10Not yet recruiting
A Randomized Phase 3 Open Label Study of Nivolumab vs Temozolomide Each in Combination With Radiation Therapy in Newly Diagnosed Adult Subjects With Unmethylated MGMT (Tumor O-6-methylguanine DNA Methyltransferase) Glioblastoma (CheckMate 498: CHECKpoint [NCT02617589]Phase 3560 participants (Actual)Interventional2016-03-01Completed
Randomized Phase II Trial of Hypofractionated Dose-Escalated Photon IMRT or Proton Beam Therapy Versus Conventional Photon Irradiation With Concomitant and Adjuvant Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT02179086]Phase 2624 participants (Actual)Interventional2014-10-27Active, not recruiting
A Randomised Phase II Study of NivolUmab and TeMozolomide vs Temozolomide Alone in Newly Diagnosed Elderly Patients With Glioblastoma (NUTMEG) [NCT04195139]Phase 2103 participants (Actual)Interventional2018-02-22Active, not recruiting
Phase II Study of Pembrolizumab (MK-3475) in Combination With Standard Therapy for Newly Diagnosed Glioblastoma [NCT03197506]Phase 252 participants (Actual)Interventional2017-09-15Suspended(stopped due to Temporary closure per protocol design)
A Phase I Study of a Personalized NeoAntigen Cancer Vaccine With Radiotherapy Plus Pembrolizumab/MK-3475 Among Newly Diagnosed Glioblastoma Patients [NCT02287428]Phase 156 participants (Anticipated)Interventional2014-11-30Recruiting
A Randomized Phase III Study of Temozolomide and Short-Course Radiation Versus Short-Course Radiation Alone In The Treatment of Newly Diagnosed Glioblastoma Multiforme in Elderly Patients [NCT00482677]Phase 3562 participants (Actual)Interventional2007-11-14Completed
Phase 2 Study of Nimotuzumab in Combination With Radio-chemotherapy for the Treatment of Brainstem Tumor in Children [NCT02672241]Phase 230 participants (Anticipated)Interventional2016-01-31Recruiting
Clinical Study of Apatinib Combined With Temozolomide in the Treatment of Uncontrolled or Repeated High-grade Gliomas [NCT04253873]Phase 240 participants (Anticipated)Interventional2019-12-16Recruiting
A Phase III Randomized Study of Neuradiab in Combination With External Beam Radiation and Temozolomide Versus External Beam Radiation and Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00615186]Phase 39 participants (Actual)Interventional2008-06-30Terminated(stopped due to Due to delay in site initiation and funding considerations)
Phase I Trail of Temodar Plus O6-Benzylguanine (O6-BG) (NSC 637037) Plus Irinotecan (CPT-11) (NSC 616348) in the Treatment of Patients With Recurrent / Progressive Cerebral Anaplastic Gliomas [NCT00612638]Phase 196 participants (Anticipated)Interventional2005-01-31Completed
A Phase II Study Of Gliadel, Concomitant Temozolomide And Radiation, Followed By Dose Dense Therapy With Temozolomide Plus Bevacizumab For Newly Diagnosed Malignant High Grade Glioma [NCT00660621]Phase 240 participants (Anticipated)Interventional2008-04-30Recruiting
Phase 1 Study of the Combination of Lapatinib and Temozolomide for the Treatment of Progressive Brain Disease in HER-2 Positive Breast Cancer [NCT00614978]Phase 118 participants (Actual)Interventional2008-01-31Completed
A Phase II Study of Multiparametric MR-Guided High Dose Adaptive Radiotherapy With Concurrent Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT04574856]Phase 230 participants (Anticipated)Interventional2020-11-04Recruiting
A 2-part Trial Comparing Overall Survival of Patients With Metastatic Ewing's Sarcoma Treated With Vigil Versus Gemcitabine and Docetaxel and to Determine Safety Profile of Vigil in Combination With Irinotecan and Temozolomide. [NCT02511132]Phase 222 participants (Actual)Interventional2016-02-10Completed
Effectiveness and Safety of Whole-brain Radiotherapy With/Without TMZ Concurrent Chemotherapy or Avoidance of Hippocampus for Patients of Brain Metastases: a Multi-institutional, Randomized Controlled Clinical Trial [NCT02832635]Phase 2256 participants (Anticipated)Interventional2016-07-31Not yet recruiting
Regorafenib in Combination With Temozolomide With or Without Radiotherapy in Patients With Newly Diagnosed MGMT-Methylated, IDH Wild-type Glioblastoma. A Phase I Dose-finding Study [NCT06095375]Phase 136 participants (Anticipated)Interventional2022-07-04Recruiting
A Randomized, Open-Label, Phase 2 Study Evaluating Abemaciclib in Combination With Irinotecan and Temozolomide in Participants With Relapsed or Refractory Ewing's Sarcoma [NCT05440786]Phase 245 participants (Anticipated)Interventional2022-09-20Recruiting
Phase I/II Multicenter Study to Assess Efficacy and Safety of Ribociclib (LEE011) in Combination With Topotecan and Temozolomide (TOTEM) in Pediatric Patients With Relapsed or Refractory Neuroblastoma and Other Solid Tumors [NCT05429502]Phase 1/Phase 2231 participants (Anticipated)Interventional2022-12-27Recruiting
Phase II Clinical Trial of Bavituximab With Radiation and Temozolomide for Patients With Newly Diagnosed Glioblastoma [NCT03139916]Phase 236 participants (Actual)Interventional2017-09-13Completed
Adjuvant Dendritic-Cell Immunotherapy Plus Temozolomide Following Surgery and Chemoradiation in Patients With Newly Diagnosed Glioblastoma [NCT02649582]Phase 1/Phase 220 participants (Anticipated)Interventional2015-12-31Recruiting
Fractionated Stereotactic Radiotherapy Combined With Temozolomide for Refractory Brain Metastases: A Single-arm, Single-center Phase II Trial [NCT02654106]Phase 265 participants (Actual)Interventional2011-10-31Completed
Genetic Variates of Response to Cisplatin, Vinblastine, and Temozolomide (CVT) in Patients With Metastatic Melanoma [NCT00885534]Phase 27 participants (Actual)Interventional2009-04-30Completed
Addition of Anlotinib Hydrochloride to the Stupp Regimen Versus the Stupp Regimen Alone for Newly Diagnosed Glioblastoma: A Randomized Multicenter Prospective Phase II Study [NCT04157478]Phase 2464 participants (Anticipated)Interventional2020-01-31Not yet recruiting
[NCT02772107]Phase 2/Phase 3132 participants (Anticipated)Interventional2015-12-31Recruiting
Multisite Open-label Randomized Phase II Clinical Trial in Newly Diagnosed Glioblastoma Treated by Concurrent Temoradiation and Adjuvant Temozolomide +/- Ultrasound-induced Blood Brain Barrier Opening. [NCT04614493]Phase 266 participants (Anticipated)Interventional2021-09-11Recruiting
Multicenter Phase 2 Evaluation of Temozolomide for Treatment of Brain Metastases of Either Malignant Melanoma, Breast and Non-small Cell Lung Cancer. [NCT00831545]Phase 2162 participants (Actual)Interventional2000-12-01Completed
Vaccination With Autologous Dendritic Cells Loaded With Autologous Tumour Homogenate in Glioblastoma: a Phase II Study [NCT04523688]Phase 228 participants (Anticipated)Interventional2021-03-25Recruiting
A Study to Evaluate the Safety and Efficacy of the Tumor-targeting Human Antibody-cytokine Fusion Protein L19TNF Plus Standard Temozolomide Chemoradiotherapy in Patients With Newly Diagnosed Glioblastoma [NCT04443010]Phase 1/Phase 2226 participants (Anticipated)Interventional2021-01-20Recruiting
A Phase III Randomized Trial of Repeated Superselective Intraarterial Cerebral Infusion (SIACI) of Bevacizumab (Avastin) With Temozolomide and Radiation Compared to Temozolomide and Radiation Alone in Newly Diagnosed Glioblastoma (GBM) [NCT05271240]Phase 3432 participants (Anticipated)Interventional2022-04-27Recruiting
A Phase Ib/II Study of Intermittent Talazoparib Plus Temozolomide in Patients With Metastatic Castration Resistant Prostate Cancer and No Mutations in DNA Damage Repair [NCT04019327]Phase 1/Phase 244 participants (Anticipated)Interventional2019-07-11Recruiting
European Proof-of-Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory Tumors [NCT02813135]Phase 1/Phase 2460 participants (Anticipated)Interventional2016-08-03Recruiting
A Randomized Phase II Study of Subventricular Zone (SVZ) Irradiation Plus Temozolomide in Newly Diagnosed Glioblastoma Multiforme [NCT02177578]Phase 260 participants (Anticipated)Interventional2014-07-08Recruiting
A Phase II Randomized Trial of Proton Vs. Photon Therapy (IMRT) for Cognitive Preservation in Patients With IDH Mutant, Low to Intermediate Grade Gliomas [NCT03180502]Phase 2120 participants (Anticipated)Interventional2017-08-02Recruiting
Pilot Study of Anlotinib in Combination With STUPP Regimen for Treatment of Patients With Newly Diagnosed Glioblastoma [NCT04119674]Phase 1/Phase 233 participants (Actual)Interventional2019-03-19Completed
A Phase I Study of Oral Carboxyamidotriazole Orotate (CTO) Titrated as a Single Agent in Patients With Advanced or Metastatic Solid Tumors and Titrated in Combination Therapy With Temodar® for Patients With Glioblastoma and Other Recurrent Malignant Gliom [NCT01107522]Phase 1100 participants (Anticipated)Interventional2010-05-31Active, not recruiting
Treatment Strategy for Low-grade Gliomas [NCT00897377]Phase 3500 participants (Anticipated)Interventional2007-12-31Terminated(stopped due to Difficulty in recruiting patients)
(Inhibition of NF-kB Signaling in Melanoma Therapy) A Phase I/II Clinical Trial of PS-341, a Proteasome Inhibitor, in Combination With an Extended Continuous Oral Schedule of Temozolomide in Patients With Advanced Refractory Solid Tumors With the Phase II [NCT00512798]Phase 1/Phase 247 participants (Actual)Interventional2003-06-30Terminated(stopped due to This study was terminated due to lack of efficacy)
A Phase II Study of Irinotecan + Temozolomide in Children With Recurrent Neuroblastoma [NCT00311584]Phase 259 participants (Actual)Interventional2006-04-30Completed
A Phase II, Multicenter, Study for Newly Diagnosed Glioblastomas Using Boron Neutron Capture Therapy, Additional X-ray Treatment and Chemotherapy [NCT00974987]Phase 232 participants (Actual)Interventional2009-09-01Completed
A Phase II Trial of Concurrent Radiation Therapy and Temozolomide Followed by Temozolomide Plus Sorafenib in the First-Line Treatment of Patients With Glioblastoma Multiforme [NCT00544817]Phase 247 participants (Actual)Interventional2007-04-30Completed
Randomized Phase II Study: Temozolomide (TMZ) Concomitant to Radiotherapy Followed by Sequential TMZ in Advanced Non-Small Cell Lung Cancer (NSCLC) Patients With Central Nervous System (CNS) Metastasis Versus Radiotherapy Alone [NCT00266812]Phase 235 participants (Actual)Interventional2005-03-08Terminated(stopped due to Patient target could not be reached within the planned timeframe.)
Phase I Dose Escalation Study of Temozolomide With Autologous Stem Cell Rescue for Patients With Relapsed/Refractory CNS Malignancy Including Isolated Metastatic Disease [NCT00629187]Phase 12 participants (Actual)Interventional2004-04-30Terminated(stopped due to The study was stopped due to lack of adequate enrollment.)
A Single-center, Open-label, Phase 1 Study of Macitentan, Radiotherapy and Temozolomide Concurrent Therapy Followed by Maintenance Therapy With Macitentan and Temozolomide in Subjects With Newly Diagnosed Glioblastoma [NCT02254954]Phase 130 participants (Actual)Interventional2015-01-08Terminated(stopped due to Sponsor decision due to low recruitment)
UPCC 04219 Phase 2 Study of Capecitabine-Temozolomide(CapTem) With Yttrium-90 Radioembolization in the Treatment of Patients With Unresectable Metastatic Grade 2 Neuroendocrine Tumors [NCT04339036]Phase 250 participants (Anticipated)Interventional2021-10-07Recruiting
A Phase II/III Randomized, Open-Label Study of Toca 511, A Retroviral Replicating Vector, Combined With Toca FC With Temozolomide and Radiation Followed by Adjuvant Temozolomide and Toca FC Compared to Temozolomide and Radiation Followed by Adjuvant Temoz [NCT04105374]Phase 2/Phase 30 participants (Actual)Interventional2020-01-31Withdrawn(stopped due to NCI approval withdrawn)
Phase 2 Study of Radiation Therapy and Combination Chemotherapy Following Surgery in Treating Children With Newly Diagnosed Medulloblastoma [NCT02681705]Phase 260 participants (Anticipated)Interventional2010-01-31Recruiting
DIRECT (DIsulfiram REsponse as add-on to ChemoTherapy in Recurrent) Glioblastoma: A Randomized Controlled Trial [NCT02678975]Phase 2/Phase 388 participants (Actual)Interventional2017-01-31Completed
A Phase II Study of Dose-Dense Temozolomide and Lapatinib for Recurrent Low-Grade and Anaplastic Supratentorial, Infratentorial and Spinal Cord Ependymoma [NCT00826241]Phase 258 participants (Actual)Interventional2009-01-31Completed
Efficacy and Safety of AP 12009 in Adult Patients With Recurrent or Refractory Anaplastic Astrocytoma or Secondary Glioblastoma as Compared to Standard Chemotherapy Treatment: A Randomized, Actively Controlled, Open Label Clinical Phase III Study. [NCT00761280]Phase 327 participants (Actual)Interventional2008-12-31Terminated(stopped due to Unable to recruit the projected patient number. All analyses are descriptive, only.)
A Randomized, Double-Blind, Placebo-Controlled, Phase 2 Study Evaluating the Efficacy of ABT-888 in Combination With Temozolomide Versus Temozolomide Alone in Subjects With Metastatic Melanoma [NCT00804908]Phase 2346 participants (Actual)Interventional2009-02-28Completed
Phase III Double-blind Placebo-Controlled Trial of Conventional Concurrent Chemoradiation and Adjuvant Temozolomide Plus Bevacizumab Versus Conventional Concurrent Chemoradiation and Adjuvant Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT00884741]Phase 3637 participants (Actual)Interventional2009-04-15Completed
A Phase 2 Study of Pamiparib (BGB-290) Plus Temozolomide for Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) [NCT04603365]Phase 20 participants (Actual)Interventional2021-10-18Withdrawn(stopped due to enrollment)
Temodal Capsule All-Case-Registered Surveillance (Designated Drug Use Investigation) - Evaluation of The Safety and Efficacy of Temodal in Patients With Newly Diagnosed Malignant Glioma (Concomitant With Radiotherapy and Then as Monotherapy) and Relapsed [NCT00705198]1,804 participants (Actual)Observational2006-09-30Completed
A Randomized, Multicenter, Double-Blind, Placebo-Controlled, Phase 2b Study to Assess the Safety and Efficacy of IGV-001, an Autologous Cell Immunotherapy With Antisense Oligonucleotide (IMV-001) Targeting IGF-1R, in Newly Diagnosed Patients With Glioblas [NCT04485949]Phase 293 participants (Anticipated)Interventional2023-03-20Recruiting
Effect of rhIL-7-hyFc on Increasing Lymphocyte Counts in Patients With Newly Diagnosed Non-severe Lymphopenic Gliomas Following Radiation and Temzolomide [NCT03687957]Phase 1/Phase 270 participants (Anticipated)Interventional2019-01-04Recruiting
[NCT02330991]Phase 2204 participants (Actual)Interventional2015-01-31Completed
Avastin and Temozolomide Following Radiation and Chemotherapy for Newly Diagnosed Glioblastoma Multiforme: A Phase II Study [NCT00590681]Phase 262 participants (Actual)Interventional2007-02-28Completed
A Non-Randomized, Open-Label, Multi-Center Phase 1/2 Study Evaluating the Safety, Pharmacokinetics and Efficacy of ABT-414 in Japanese Subjects With Malignant Glioma [NCT02590263]Phase 1/Phase 253 participants (Actual)Interventional2015-08-24Completed
Temodal (TMZ) in Concomitant Radiochemotherapy Followed by Sequential TMZ Chemotherapy in Newly Diagnosed Glioblastoma Multiforme Patients - an Observational Program [NCT00725010]64 participants (Actual)Observational2007-04-30Completed
Phase II Trial of Neoadjuvant Temozolomide in Melanoma Patients With Palpable Stage III or IV Disease Undergoing Complete Surgical Resection [NCT00588341]Phase 224 participants (Actual)Interventional2005-09-30Completed
A Phase II Study of Temozolomide (SCH 52365) in the Treatment of Patients With Relapsed Small Cell Lung Cancer [NCT00022711]Phase 27 participants (Actual)Interventional2002-01-31Completed
A Phase 2 Study of Continuous Talazoparib Plus Intermittent Low-Dose Temozolomide in Patients With Relapsed or Refractory Extensive-Stage Small Cell Lung Cancer (TRIO-US L-07) [NCT03672773]Phase 228 participants (Anticipated)Interventional2018-10-31Recruiting
Phase II Study of Temozolomide for Relapsed Sensitive or Refractory Small Cell Lung Cancer [NCT00740636]Phase 292 participants (Actual)Interventional2008-08-31Completed
Phase 2 Single- Arm Studies of Temozolomide in Combination With Topotecan in Refractory and Relapsed Neuroblastoma and Other Paediatric Solid Tumours [NCT00918320]Phase 2129 participants (Actual)Interventional2009-06-30Completed
Aerosol Liposomal 9-Nitro-20(S)-Camptothecin (L9-NC) and Temozolomide in Ewing's Sarcoma and Other Solid Tumors With Lung Involvement [NCT00492141]Phase 1/Phase 210 participants (Actual)Interventional2006-06-30Completed
SCH 52365 Phase II Clinical Study in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00684567]Phase 230 participants (Actual)Interventional2005-09-27Completed
Phase II Trial of Carboplatin, Paclitaxel, and Temozolomide for Patients With Metastatic Melanoma [NCT01009515]Phase 219 participants (Actual)Interventional2009-08-31Terminated(stopped due to Low accrual; target accrual not met)
Benzylguanine-Mediated Tumor Sensitization With Chemoprotected Autologous Stem Cells for Patients With Malignant Gliomas [NCT00669669]Phase 1/Phase 212 participants (Actual)Interventional2009-02-25Terminated(stopped due to Terminated due to loss in funding.)
A Randomized Phase II Trial of Bevacizumab With Irinotecan or Bevacizumab With Temozolomide in Recurrent Glioblastoma [NCT00433381]Phase 2123 participants (Actual)Interventional2007-03-01Completed
Safety and Efficacy of LITT Combined With Early Use of Temozolomide for Recurrent Glioblastomas [NCT05663125]Phase 1/Phase 210 participants (Anticipated)Interventional2022-12-01Active, not recruiting
A Pilot Study of the Addition of Bevacizumab to Vincristine, Oral Irinotecan, and Temozolomide (VOIT Regimen) for Relapsed/Refractory Pediatric Solid Tumors [NCT00786669]Phase 113 participants (Actual)Interventional2008-10-31Completed
SCH 52365 Phase II Clinical Study: A Study on the Efficacy and Safety of Monotherapy With SCH 52365 in Patients With First Relapsed Anaplastic Astrocytoma [NCT00783393]Phase 232 participants (Actual)Interventional2003-05-27Completed
"Randomized Multicentric Phase II Study of Prolonged Adjuvant Temozolomide or Stop and Go in Glioblastoma Patients: The PATSGO Study" [NCT00643825]Phase 264 participants (Anticipated)Interventional2008-01-31Recruiting
Phase Ib/II: Epigenetic Modification of Chemosensitivity and Apoptosis in Metastatic Melanoma: Treatment of a Resistant Disease Using Decitabine, Temozolomide and Panobinostat [NCT00925132]Phase 1/Phase 239 participants (Actual)Interventional2009-12-31Terminated(stopped due to Change in the number of approved drugs for metastatic melanoma)
A Prospective, Multi-center Trial of NovoTTF-100A Together With Temozolomide Compared to Temozolomide Alone in Patients With Newly Diagnosed GBM. [NCT00916409]Phase 3700 participants (Anticipated)Interventional2009-06-30Completed
Phase I Trial of Aflibercept (VEGF Trap) With Radiation Therapy and Concomitant and Adjuvant Temozolomide in Patients With Malignant Glioma [NCT00650923]Phase 161 participants (Actual)Interventional2008-07-31Completed
A Phase 1/1B Dose-Escalation Study to Determine the Safety and Tolerability of SCH 717454 Administered in Combination With Chemotherapy in Pediatric Subjects With Advanced Solid Tumors (Protocol No. 05883) [NCT00960063]Phase 14 participants (Actual)Interventional2009-11-11Terminated(stopped due to Study was terminated for business reasons.)
An Open-label, Phase 1b Study to Evaluate the Safety and Tolerability of Eflornithine Plus Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT05879367]Phase 166 participants (Anticipated)Interventional2023-07-24Recruiting
A Phase I Study of Temozolomide, Oral Irinotecan, and Vincristine for Children With Refractory Solid Tumors [NCT00138216]Phase 142 participants (Actual)Interventional2005-10-31Completed
NOA-04 Randomized Phase III Study of Sequential Radiochemotherapy of Anaplastic Glioma With PCV or Temozolomide [NCT00717210]Phase 3318 participants (Actual)Interventional1999-06-30Completed
A Phase I Study of the Combination of Radiation Therapy (RT), Arsenic Trioxide (ATO) and Temozolomide (TMZ) in Patients With Newly-Diagnosed Glioblastoma Multiforme (GBM) [NCT00720564]Phase 118 participants (Anticipated)Interventional2008-04-30Completed
Phase 2 Study of Dose-Intense Temozolomide in Recurrent Glioblastoma [NCT00657267]Phase 258 participants (Actual)Interventional2008-05-31Completed
A Phase I Trial of Dose Escalation of Metformin in Combination With Vincristine, Irinotecan, and Temozolomide in Children With Relapsed or Refractory Solid Tumors [NCT01528046]Phase 126 participants (Actual)Interventional2012-09-24Completed
A Phase I Study of Sorafenib With Radiation and Temozolomide in Newly Diagnosed Glioblastoma or Gliosarcoma [NCT00734526]Phase 118 participants (Actual)Interventional2008-12-18Terminated
Clinical Study of Evaluating the Safety and Initial Efficacy of XS005 Cell Injection Combined With Stupp Regimen for Adjuvant Chemotherapy in Subjects With Primary Glioblastoma [NCT06147505]Phase 1/Phase 230 participants (Anticipated)Interventional2023-10-29Recruiting
A Phase I Study of Allogeneic Ex Vivo Expanded Gamma Delta (γδ) T Cells (IND # 28460) in Combination With Dinutuximab, Temozolomide, Irinotecan, and Zoledronate in Children With Refractory, Relapsed, or Progressive Neuroblastoma [NCT05400603]Phase 124 participants (Anticipated)Interventional2023-11-06Recruiting
A Randomized, Open-label Pilot Trial to Evaluate the Safety and Efficacy of Carmustine Wafer in Combination With Retifanlimab and Standard Radiation With or Without Temozolomide in Newly-Diagnosed Adult Subjects With Glioblastoma [NCT05083754]Phase 150 participants (Anticipated)Interventional2022-08-31Recruiting
Phase I/II Trial of Dasatinib (Sprycel) With Radiation Therapy and Concomitant and Adjuvant Temozolomide in Patients With Newly-Diagnosed Glioblastoma [NCT00895960]Phase 116 participants (Actual)Interventional2009-05-07Terminated(stopped due to Sponsor withdrew support; Study did not progress to Phase II.)
Radiotherapy Versus Radiotherapy Combined With Temozolomide in High-risk Low-grade Gliomas After Surgery [NCT04316039]Phase 2/Phase 3250 participants (Anticipated)Interventional2018-04-10Recruiting
A Phase 1, Multicenter, Open-Label, Dose Escalation Study of Tandutinib (Formerly MLN518) in Combination With Temozolomide and Bevacizumab Following Concurrent Radiation Therapy and Temozolomide in the Treatment of Patients With Newly Diagnosed Glioblasto [NCT00904852]Phase 130 participants (Anticipated)Interventional2009-06-30Withdrawn
Phase 2 Clinical Trial Treating Relapsed/Recurrent/Refractory Pediatric Solid Tumors With the Genomically-Targeted Agent Erlotinib in Combination With Temozolomide [NCT02689336]Phase 20 participants (Actual)Interventional2016-08-06Withdrawn(stopped due to Could not recruit any participants)
Use of PET/CT Imaging With 18F-fluoroethylcholine (FEC) in the Evaluation of Patients Treated With Radiotherapy and Temozolomide Following a Diagnosis of Glioblastoma Multiforme [NCT00943462]0 participants (Actual)Observational2009-06-30Withdrawn(stopped due to No eligible patients could be recruited.)
A Phase I Study of Cyclic Oral Administration of SCH 52365 for 21 of 28 Days in Patients With Advanced Solid Malignancies [NCT00003708]Phase 132 participants (Actual)Interventional1998-07-31Completed
Phase 1b, Multicenter, Open-Label Study of Marizomib With Temozolomide and Radiotherapy in Patients With Newly Diagnosed WHO Grade IV Malignant Glioma [NCT02903069]Phase 166 participants (Actual)Interventional2016-08-17Completed
A Cancer Research UK Phase I Trial of Olaparib (AZD2281), an Oral PARP Inhibitor, in Combination With Extended Low-Dose Oral Temozolomide in Patients With Relapsed Glioblastoma [NCT01390571]Phase 134 participants (Anticipated)Interventional2011-07-31Completed
A Pilot Study Investigating Neoadjuvant Temozolomide-based Proton Chemoradiotherapy for High-Risk Soft Tissue Sarcomas [NCT00881595]Phase 20 participants (Actual)Interventional2009-02-28Withdrawn(stopped due to No patients accrued since study opened)
A Protocol for the Treatment of Newly Diagnosed Rhabdomyosarcoma Using Molecular Risk Stratification and Liposomal Irinotecan Based Therapy in Children With Intermediate and High Risk Disease [NCT06023641]Phase 1/Phase 2100 participants (Anticipated)Interventional2023-11-30Not yet recruiting
Phase II Study of Two Distinct Tailored Temozolomide Regimens for Patients With Acute Myeloid Leukemia Age > 60 Years and Poor Risk/Refractory Disease [NCT00611247]Phase 242 participants (Actual)Interventional2007-12-31Completed
The Prospective Study of FTD Program and HD-MTX-Ara-C Program Contrast in the Treatment of PCNSL Lymphoma. [NCT05274139]Phase 220 participants (Actual)Interventional2017-03-02Completed
A Phase 2 Clinical Trial of Neoadjuvant Camrelizumab Plus Apatinib and Temozolomide in High Risk Clinical Stage Ⅱ-Ⅲ Acral Melanoma [NCT05512481]Phase 240 participants (Anticipated)Interventional2022-09-13Recruiting
Evaluating the Impact of 18F-DOPA-PET on Radiotherapy Planning for Newly Diagnosed Gliomas [NCT01991977]Phase 291 participants (Actual)Interventional2013-12-31Active, not recruiting
VITAS: Atezolizumab in Combination With Chemotherapy for Pediatric Relapsed/Refractory Solid Tumors: An Open-label, Phase II, Single-arm, Multi-center Trial [NCT04796012]Phase 1/Phase 223 participants (Anticipated)Interventional2023-04-18Recruiting
IPI-Biochemotherapy for Chemonaive Patients With Metastatic Melanoma [NCT01409174]Phase 119 participants (Actual)Interventional2013-02-28Terminated(stopped due to Slow accrual, closed in Phase I.)
A Phase II Study of Gliadel, Concomitant Temozolomide and Radiation, Followed by Metronomic Therapy With Temozolomide for Newly Diagnosed Malignant High Grade Glioma [NCT00548938]Phase 240 participants (Anticipated)Interventional2007-10-31Terminated(stopped due to Principal Investigator has left the institution)
Phase I Biochemotherapy With Cisplatin, Temozolomide, With Increasing Doses of Abraxane, Combined With Interleukin-2 and Interferon in Patients With Metastatic Melanoma [NCT00970996]Phase 110 participants (Actual)Interventional2009-09-30Completed
Radiation Therapy and Concurrent Plus Adjuvant Temsirolimus (CCI-779) Versus Chemo-Irradiation With Temozolomide in Newly Diagnosed Glioblastoma Without Methylation of the MGMT Gene Promoter - A Randomized Multicenter, Open-Label, Phase II Study. [NCT01019434]Phase 2111 participants (Actual)Interventional2009-10-31Completed
Phase II Study of Bi-Weekly Temozolomide Plus Bevacizumab for Adult Patients With Recurrent Glioblastoma Multiforme [NCT00883298]Phase 230 participants (Actual)Interventional2009-04-30Completed
Dose-intensified Rechallenge With Temozolomide, One Week On One Week Off Versus Three Weeks On One Week Off in Patients With Progressive or Recurrent Glioblastoma [NCT00941460]Phase 2105 participants (Actual)Interventional2009-09-30Completed
A Pilot Study of Bevacizumab-Based Therapy in Patients With Newly Diagnosed High-Grade Gliomas and Diffuse Intrinsic Pontine Gliomas [NCT00890786]Early Phase 127 participants (Actual)Interventional2009-05-31Completed
Feasibility Study on Laser Interstitial Thermal Therapy Ablation for the Treatment of Newly Diagnosed Glioblastoma [NCT02880410]Phase 13 participants (Actual)Interventional2017-08-15Terminated(stopped due to Terminated (failure to enroll))
Open-label, Non-randomized, Two-stage Study to Evaluate the Efficacy, Safety, Pharmacokinetics and Pharmacodynamic Effects of PQR309 in Patients With Progressive Glioblastoma [NCT02850744]Phase 210 participants (Actual)Interventional2015-07-31Terminated
Phase I-II Study of Nivolumab in Combination With Temozolomide and Radiotherapy in Children and Adolescents With Newly Diagnosed High-grade Glioma [NCT04267146]Phase 1/Phase 240 participants (Anticipated)Interventional2019-07-15Recruiting
Phase II Study of Capecitabine and Temozolomide for Progressive, Differentiated, Metastatic Neuroendocrine Cancers [NCT00869050]Phase 241 participants (Actual)Interventional2005-08-31Completed
A Phase 1 Dose-Escalation Study of XL765 (SAR245409) in Combination With Temozolomide With and Without Radiation in Subjects With Malignant Gliomas [NCT00704080]Phase 154 participants (Actual)Interventional2008-08-31Completed
A Phase I Dose Per Fraction Escalation Study of Hypofractionated Intensity-Modulated Radiation Therapy (Hypo-IMRT) Combining With Temozolomide (TMZ) Chemotherapy for Patients With Newly Diagnosed Glioblastoma Multiforme (GBM) [NCT00792012]Phase 137 participants (Actual)Interventional2005-11-30Completed
Repurposing the Antipsychotic Drug Chlorpromazine as a Therapeutic Agent in the Combined Treatment of Newly Diagnosed Glioblastoma Multiforme [NCT04224441]Phase 241 participants (Anticipated)Interventional2019-12-15Recruiting
A Phase I Study of ABT-888, an Oral Inhibitor of Poly (ADP-Ribose) Polymerase and Temozolomide in Children With Recurrent/Refractory CNS Tumors [NCT00946335]Phase 131 participants (Actual)Interventional2009-07-31Completed
A Phase I Trial of the Addition of the Farnesyl Transferase Inhibitor, SCH 66336, to Temodar for Patients With Grade 3 and 4 Malignant Gliomas [NCT00612651]Phase 137 participants (Actual)Interventional2005-10-31Completed
A Randomized Phase II Trial With Bevacizumab, Irinotecan and Cerebral Radiotherapy Versus Bevacizumab, Temozolomide and Cerebral Radiotherapy as First Line Treatment for Patients With Glioblastoma Multiforme [NCT00817284]Phase 260 participants (Actual)Interventional2008-11-30Completed
A Phase I Trial of the Protease Inhibitor Nelfinavir and Concurrent Radiation and Temozolomide in Patients With WHO Grade IV Glioma [NCT00915694]Phase 115 participants (Actual)Interventional2009-04-30Terminated(stopped due to Insufficient accrual)
Phase II Treatment of Adults With Primary Malignant Glioma With Irinotecan Plus Temozolomide [NCT00616005]Phase 241 participants (Anticipated)Interventional2005-11-30Completed
Phase I Study of Temozolomide and Intrathecal DepoCyt in Patients With Neoplastic Meningitis [NCT00515788]Phase 111 participants (Actual)Interventional2006-02-28Terminated(stopped due to Study terminated due to slow accrual with no expansion to additional phase.)
A Randomized, Placebo Controlled Phase 3 Study of ABT-414 With Concurrent Chemoradiation and Adjuvant Temozolomide in Subjects With Newly Diagnosed Glioblastoma (GBM) With Epidermal Growth Factor Receptor (EGFR) Amplification (Intellance1) [NCT02573324]Phase 3691 participants (Actual)Interventional2015-01-04Completed
Phase II Trial of Pemetrexed and Temozolomide in Treating Patients With Relapsed PCNSL [NCT01985451]Phase 215 participants (Anticipated)Interventional2013-03-31Active, not recruiting
ESP1/SARC025 Global Collaboration: A Phase I Study of a Combination of the PARP Inhibitor, Niraparib and Temozolomide and/or Irinotecan in Patients With Previously Treated,Incurable Ewing Sarcoma [NCT02044120]Phase 134 participants (Actual)Interventional2014-05-31Completed
A Phase I/II Trial of Temodar in Pediatric Patients and Young Adults With High-Risk or Recurrent Solid Tumors [NCT00005952]Phase 1/Phase 230 participants (Anticipated)Interventional2000-08-31Completed
Phase II Treatment of Children With Newly Diagnosed Malignant Central Nervous System Tumors With Temozolomide Prior to Radiation Therapy [NCT00005955]Phase 20 participants Interventional2000-08-31Completed
A Phase I Study of Temozolomide and CCNU in Pediatric Patients With Newly Diagnosed Incompletely Resected Non-Brainstem High-Grade Gliomas [NCT00006024]Phase 132 participants (Actual)Interventional2000-11-30Completed
Lobaplatin Combined With Etoposide for First-line Treatment in Extensive Stage Sclc Then Benefit Patients Follow up Temozolomide Maintain Therapeutic [NCT02972320]Phase 260 participants (Anticipated)Interventional2016-06-30Recruiting
Phase I Trial of DNA-PK Inhibitor (M3814) in Combination With Radiation and Adjuvant Temozolomide in Newly Diagnosed MGMT Unmethylated Glioblastoma [NCT04555577]Phase 129 participants (Anticipated)Interventional2020-09-20Recruiting
A Phase I, Open Label Study of AT-101 Plus Radiotherapy and Temozolomide and of AT-101 Plus Adjuvant Temozolomide for Patients With Newly-Diagnosed Glioblastoma Multiforme [NCT00390403]Phase 150 participants (Anticipated)Interventional2007-02-28Completed
Phase I Treatment of Adults With Primary Malignant Glioma With Irinotecan (CPT-11) (NSC- #6616348) Plus Temodar (NSC #362856) [NCT00005951]Phase 10 participants Interventional2000-08-31Completed
Phase II Treatment of Adults With Brain Metastases With Temodar [NCT00005954]Phase 20 participants Interventional2000-08-31Completed
Treatment of Patients With Metastatic Malignant Melanoma With Chemobiotherapy With Temozolomide, GM-CSF, IL2, and Interferon Alfa-2b Phase II Trial [NCT00014092]Phase 20 participants Interventional1999-12-31Completed
Intensive Dose Temozolomide Treatment or Temozolomide With Thalidomide Treatment in Recurrent Glioblastoma After Standard Therapy:a Randomized Phase II Trial [NCT00521482]Phase 240 participants (Anticipated)Interventional2007-09-30Not yet recruiting
Phase I Study of Enzastaurin and Temozolomide in Patients With Gliomas [NCT00516607]Phase 128 participants (Actual)Interventional2007-07-31Active, not recruiting
Phase I Study of TTC (Taxotere/Temodar/Cisplatin) in Metastatic Melanoma Patients [NCT00527761]Phase 123 participants (Actual)Interventional2004-08-31Completed
Phase II Study of Gamma Knife Radiosurgery and Temozolomide (Temodar) for Newly Diagnosed Brain Metastases [NCT00582075]Phase 225 participants (Actual)Interventional2002-07-31Completed
Monitoring Anti-angiogenic Therapy in Brain Tumors by Advanced MRI [NCT02843230]30 participants (Actual)Observational2016-08-01Completed
A Phase II Study of Prolonged Daily Temozolomide for Low-Grade Glioma [NCT00165360]Phase 246 participants (Actual)Interventional2001-09-30Completed
"A Pilot Study of Abraxane® (Albumin-bound Paclitaxel) and Temodar® (Temozolomide) Plus Genasense® (Oblimersen Sodium) in Subjects With Advanced Melanoma (The ATG Study)." [NCT00409383]Phase 128 participants (Anticipated)Interventional2006-11-30Active, not recruiting
A Phase II Study of Temozolomide (TMZ) Following Stereotactic Radiosurgery (SRS) for Patients With Newly Diagnosed Brain Metastases [NCT00717275]Phase 23 participants (Actual)Interventional2008-09-30Terminated(stopped due to Slow accrual.)
A Phase II Feasibility Study of Adjuvant Intra-Nodal Autologous Dendritic Cell Vaccination for Newly Diagnosed Glioblastoma Multiforme [NCT00323115]Phase 211 participants (Actual)Interventional2006-05-31Completed
A Phase 2 Study of Temozolomide (SCH 52365) in Subjects With Advanced Aerodigestive Tract Cancers Selected for Methylation of O6-Methyl-Guanine-DNA Methyltransferase (MGMT) Promoter [NCT00423150]Phase 286 participants (Actual)Interventional2007-01-26Terminated
Evaluation of the Irinotecan/Bevacizumab Association as Neo-adjuvant and Adjuvant Treatment of Chemoradiation With Temozolomide for Naive Unresectable Glioblastoma. Phase II Randomized Study With Comparison to Chemoradiation With Temozolomide [NCT01022918]Phase 2134 participants (Actual)Interventional2009-01-31Completed
A Phase 2 Study of O-Benzylguanine (O-BG) and Temozolomide in Patients With Glioblastoma Progressing at Least 3 Months After Completion of Primary Treatment With Radiation Therapy and Temozolomide [NCT00436436]Phase 212 participants (Actual)Interventional2006-11-13Terminated(stopped due to Study closed to accrual after the company chose to stop development of the drug.)
A Phase II Trial of O6-Benzylguanine and Temozolomide in Pediatric Patients With Recurrent or Progressive High-Grade Gliomas and Recurrent or Progressive Brainstem Tumors [NCT00275002]Phase 241 participants (Actual)Interventional2006-02-28Completed
Phase 1/2 Study of Enzastaurin Plus Temozolomide During and Following Radiation Therapy in Patients With Newly Diagnosed Glioblastoma Multiforme or Gliosarcoma [NCT00402116]Phase 1/Phase 272 participants (Actual)Interventional2006-09-30Completed
Phase II Study of Tarceva Plus Temodar During and Following Radiation Therapy in Patients With Newly Diagnosed Glioblastoma Multiforme and Gliosarcoma [NCT00187486]Phase 266 participants (Actual)Interventional2004-08-31Completed
A Phase II Trial of Temozolomide Plus Bevacizumab in Patients With Metastatic Melanoma Involving the Central Nervous System [NCT01048554]Phase 234 participants (Actual)Interventional2009-11-30Completed
TEMOkids Study: A Population Pharmacokinetic, Acceptability and Safety Study for KIMOZO, a Paediatric Oral Suspension of Temozolomide [NCT04610736]Phase 149 participants (Actual)Interventional2021-03-16Active, not recruiting
A Phase I Study of SAHA and Temozolomide in Children With Relapsed or Refractory Primary Brain or Spinal Cord Tumors [NCT01076530]Phase 127 participants (Actual)Interventional2010-02-28Completed
A Modular Phase I/IIa, Open-label, Multi-centre Study to Assess the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Preliminary Efficacy of Ascending Doses of AZD9574 as Monotherapy and in Combination With Anti-cancer Agents in Patients With [NCT05417594]Phase 1/Phase 2490 participants (Anticipated)Interventional2022-06-24Recruiting
A Phase 0/1 Pharmacokinetic and Pharmacodynamics and Safety and Tolerability Study of Letrozole in Combination With Standard Therapy in Recurrent High Grade Gliomas [NCT03122197]Early Phase 139 participants (Anticipated)Interventional2017-05-16Recruiting
A PHASE I/II TRIAL OF TEMOZOLOMIDE, MOTEXAFIN GADOLINIUM, AND 60 GY FRACTIONATED RADIATION FOR NEWLY DIAGNOSED SUPRATENTORIAL GLIOBLASTOMA MULTIFORME [NCT00305864]Phase 1/Phase 2118 participants (Actual)Interventional2006-02-09Completed
A Pilot Study Combining ABT-888, an Oral PARP Inhibitor, With Temozolomide in Patients With Metastatic Castration Resistant Prostate Cancer Who Have Failed Up to Two Non-hormonal Systemic Therapies [NCT01085422]Phase 135 participants (Actual)Interventional2010-04-30Completed
A Randomized Controlled Clinical Trial of Temozolomide Plus Apatinib in Newly Diagnosed High-grade Glioma [NCT03741244]Phase 2211 participants (Anticipated)Interventional2019-05-06Recruiting
A Phase I/II Open-Label Study TPI 287 in Combination With Temozolomide in Patients With Metastatic Melanoma [NCT01067066]Phase 121 participants (Actual)Interventional2010-02-03Terminated(stopped due to Phase I/II study halted as Phase I, Low Response Rate)
A Multicenter Randomized Phase III Trial on INTraoperative RAdiotherapy in Newly Diagnosed GliOblastoma Multiforme (INTRAGO II) [NCT02685605]Phase 3314 participants (Anticipated)Interventional2016-12-09Recruiting
Phase 2 Study of Temozolomide as Maintenance Therapy After Initial Induction Chemotherapy in Small Cell Lung Cancer [NCT01900951]Phase 260 participants (Anticipated)Interventional2013-01-31Active, not recruiting
A Pharmacodynamic Study of Proteasome Inhibition by Disulfiram in Patients With Glioblastoma [NCT01907165]Early Phase 121 participants (Actual)Interventional2013-10-10Completed
A Phase 1 Study of ABI-009 (NAB-RAPAMYCIN) in Pediatric Patients With Recurrent or Refractory Solid Tumors, Including CNS Tumors as a Single Agent and in Combination With Temozolomide and Irinotecan [NCT02975882]Phase 133 participants (Actual)Interventional2017-08-15Active, not recruiting
A Phase I Study of Vincristine, Escalating Doses of Irinotecan, Temozolomide and Bevacizumab (Vit-b) in Pediatric and Adolescent Patients With Recurrent or Refractory Solid Tumors of Non-hematopoietic Origin [NCT00993044]Phase 113 participants (Actual)Interventional2009-09-30Completed
An Open-label, Single Arm Study to Explore Whether Potential Image Biomarkers Correlate With Efficacy of Bevacizumab Combined With Conventional Therapy in Newly Diagnosed Glioblastoma [NCT01939574]20 participants (Actual)Interventional2013-08-31Active, not recruiting
Whole Brain Radiotherapy in Combination With Gefitinib (Iressa) or Temozolomide (Temodal) for Brain Metastases From Non-Small Lung Cancer (NSCLC) A Randomized Phase II Trial [NCT00238251]Phase 259 participants (Actual)Interventional2005-05-31Completed
Peptide Targets for Glioblastoma Against Novel Cytomegalovirus Antigens [NCT02864368]Phase 127 participants (Actual)Interventional2016-12-07Terminated(stopped due to Funding)
A Phase 1b Study to Assess the Safety, Tolerability and Clinical Activity of BGB-290 in Combination With Temozolomide (TMZ) in Subjects With Locally Advanced or Metastatic Solid Tumors [NCT03150810]Phase 1/Phase 2139 participants (Actual)Interventional2017-06-28Completed
Phase I Trial of Combination of DNX-2401 (Formerly Named Delta-24-RGD) Oncolytic Adenovirus With a Short Course of Temozolomide for Treatment of Glioblastoma at First Recurrent [NCT01956734]Phase 131 participants (Anticipated)Interventional2013-09-30Completed
A Phase I Study of Metronomic Temozolomide With Abexinostat (PCI-24781) for Patients With Recurrent High Grade Glioma [NCT05698524]Phase 118 participants (Anticipated)Interventional2023-06-26Recruiting
A Phase I/ II Study of Hypofractionated Radiotherapy With Concurrent Temozolomide Followed by Adjuvant Temozolomide in Patients Over 70 Years Old With Newly Diagnosed Glioblastoma [NCT01985087]Phase 1/Phase 240 participants (Anticipated)Interventional2014-09-30Recruiting
Temozolomide Combined With Bevacizumab in Metastatic Melanoma. A Multicenter Phase II Trial [NCT00568048]Phase 262 participants (Actual)Interventional2007-12-31Completed
A Phase I Study Of Irinotecan and Bevacizumab With Temozolomide in Children With Recurrent/Refractory Central Nervous System Tumors [NCT00876993]Phase 126 participants (Actual)Interventional2008-09-30Completed
The Feasibility of Selecting Patient-Specific Biologically Targeted Therapy With Sorafenib, Everolimus, Erlotinib or Dasatinib for Pediatric Patients With Refractory Or Recurrent Brain Tumors [NCT02015728]20 participants (Anticipated)Interventional2013-12-31Active, not recruiting
Phase II Study of Atorvastatin in Combination With Radiotherapy and Temozolomide in Glioblastoma [NCT02029573]Phase 236 participants (Actual)Interventional2014-01-01Completed
Phase I/II Prospective Trial for Newly Diagnosed GBM, With Upfront Gross or Subtotal Resection, Followed by Ketogenic Diet With Radiotherapy and Concurrent Temodar(R) Chemotherapy Followed by Adjuvant Temodar(R) Chemotherapy. [NCT02046187]Phase 1/Phase 214 participants (Actual)Interventional2013-10-31Terminated(stopped due to excessive protocol deviations due to strict nature of diet requirements)
Phase I-II Trial of CPT-11 and Temozolomide (Temodar) in Patients With Recurrent Malignant Glioma [NCT00006025]Phase 10 participants Interventional2001-01-05Completed
Phase I Trial of Temodar Plus O6-Benzylguanine (O6-BG) (NSC 637037) in the Treatment of Patients With Newly Diagnosed (Part 1) or Recurrent/Progressive (Parts 1 and 2) Cerebral Anaplastic Gliomas [NCT00006474]Phase 10 participants Interventional2001-03-31Completed
Phase II Trial of Temozolomide in Patients With Cisplatin-Refractory Germ Cell Tumors [NCT00006043]Phase 20 participants Interventional2000-02-29Completed
Phase II Evaluation of Temozolomide (SCH52365) and Thalidomide for the Treatment of Recurrent and Progressive Glioblastoma Multiforme [NCT00006358]Phase 244 participants (Actual)Interventional2000-06-13Completed
A Phase II Study Of Temozolomide In The Treatment Of Patients With Metastatic Non-Small Cell Lung Cancer [NCT00006877]Phase 230 participants (Actual)Interventional2000-07-31Completed
Phase 1/2 Study of Vincristine and Temozolomide in Combination With PEN-866 for Adolescents and Young Adults With Relapsed or Refractory Solid Tumors [NCT04890093]Phase 1/Phase 264 participants (Anticipated)Interventional2024-01-03Not yet recruiting
Phase II Study Temozolomide for Retinoblastoma Metastatic to the Central Nervous System for Patients From Guatemala [NCT01857752]Phase 23 participants (Actual)Interventional2012-03-31Terminated(stopped due to accrual)
Phase II Treatment of Adults With Newly Diagnosed Supratentorial Glioblastoma Multiforme Treated With Gliadel Wafers, Surgery and Limited Field Radiation Plus Concomitant Temozolomide Followed by Temozolomide [NCT00574964]Phase 250 participants (Anticipated)Interventional2005-10-31Terminated(stopped due to PI left the institution.)
A Pilot Study of Elemene Injectable Emulsion in Treating Patients With Refractory Glioblastoma [NCT04674527]Phase 230 participants (Anticipated)Interventional2021-06-01Not yet recruiting
A Trail of Fluzoparil in Combination With Temozolomide in Patients With Recurrent Glioblastoma [NCT04552977]Phase 250 participants (Anticipated)Interventional2020-09-30Not yet recruiting
Phase II Trial of High Dose Interleukin-2 Followed by Intermittent Low Dose Temozolomide in Patients With Metastatic Malignant Melanoma [NCT01124734]Phase 217 participants (Actual)Interventional2010-05-31Completed
Randomized Phase II Trial of Temozolomide Versus Hyd-Sulfate AZD6244 [NSC 748727] in Patients With Metastatic Uveal Melanoma [NCT01143402]Phase 2120 participants (Actual)Interventional2010-06-30Completed
A Phase II Study of Anlotinib Combined With Dose-dense Temozolomide for the First Recurrent or Progressive Glioblastoma After STUPP Regimen [NCT04547855]Phase 254 participants (Anticipated)Interventional2020-09-11Recruiting
The Combination of Hypofractionated Stereotactic Radiotherapy and Chemoradiotherapy Using Intensity-Modulated Radiotherapy for Newly Diagnosed Glioblastoma Multiforme: A Prospective, Single-Center, Single-Arm Phase II Clinical Trial [NCT04547621]Phase 1/Phase 250 participants (Anticipated)Interventional2020-09-01Active, not recruiting
Phase Ib/II Study of the Gamma-Secretase Inhibitor (GSI) RO4929097 in Combination With Cisplatin, Vinblastine, and Temozolomide (CVT) in Patients With Metastatic Melanoma [NCT01196416]Phase 1/Phase 214 participants (Actual)Interventional2010-08-31Completed
A Phase III Trial of Gleostine® (Lomustine)-Temozolomide Combination Therapy Versus Standard Temozolomide in Patients With Methylated MGMT Promoter Glioblastoma [NCT05095376]Phase 3306 participants (Anticipated)Interventional2021-11-29Recruiting
Apatinib Mesylate Combined With IT Regimen for the Treatment of Recurrent or Refractory Pediatric Neuroblastoma: Multi-center, Single-arm, Phase II Clinical Study. [NCT05027386]Phase 262 participants (Anticipated)Interventional2021-08-26Recruiting
A Phase 0/2 Clinical Trial of Pamiparib in Newly-Diagnosed and Recurrent Glioblastoma Patients [NCT04614909]Early Phase 130 participants (Anticipated)Interventional2021-01-11Recruiting
A Phase I/II Safety Lead in Study of Ex-Vivo Expanded Allogeneic Universal Donor TGFβi NK Cell Infusions in Combination With Irinotecan, Temozolomide, and Dinutuximab in Patients With Relapsed or Refractory Neuroblastoma: The Allo - STING Trial [NCT04211675]Phase 1/Phase 231 participants (Anticipated)Interventional2022-09-01Recruiting
Pilot Study of Safety and Feasibility of Acquiring Hyperpolarized Imaging in Patients With Gliomas [NCT03739411]Phase 1140 participants (Anticipated)Interventional2015-12-09Recruiting
Phase III Trial on Concurrent and Adjuvant Temozolomide Chemotherapy in Non-1p/19q Deleted Anaplastic Glioma. The CATNON Intergroup Trial. [NCT00626990]Phase 3751 participants (Actual)Interventional2007-12-31Active, not recruiting
An International, Randomized, Open-label Phase I/II Study of Vismodegib in Combination With Temozolomide Versus Temozolomide Alone in Adult Patients With Recurrent or Refractory Medulloblastomas Presenting an Activation of the Sonic Hedgehog (SHH) Pathway [NCT01601184]Phase 1/Phase 224 participants (Actual)Interventional2012-06-30Terminated(stopped due to The number of successes is not reached at the end of first stage of the phase II. The study is stopped.)
Phase I Study of ZD6474 and Temozolomide in Patients With Advanced Cancer [NCT00601614]Phase 10 participants (Actual)Interventional2008-01-31Withdrawn(stopped due to no enrollment)
A Single Arm Phase II Study of Bevacizumab and Extended Treatment of Temozolomide in Patients With Recurrent Glioblastoma Multiforme [NCT01115491]Phase 232 participants (Actual)Interventional2010-06-30Completed
Phase II Randomized Multicenter Study Comparing Brain Radiation in Combination With Temozolomide or Radiation Alone in Patients With Brain Metastases From Breast Cancer [NCT00875355]Phase 2100 participants (Anticipated)Interventional2007-11-30Recruiting
A Phase I/II Study of Daily Oral Dosing With Temozolomide and Sunitinib Malate for 6 Weeks of an 8-Week Cycle in Patients With Metastatic and Unresectable Locally-Advanced Malignant Melanoma [NCT01005472]Phase 116 participants (Actual)Interventional2008-12-01Completed
A Phase Ib, Open-Label, Multicenter Trial of Intravenous INO-1001 Plus Oral Temozolomide to Evaluate the Tolerability, Safety, and Pharmacokinetics in Subjects With Newly-Diagnosed or Recurrent Unresectable Stage III or Stage IV Melanoma [NCT00272415]Phase 139 participants (Anticipated)Interventional2005-10-31Terminated
A Phase II, Multi-centre, Open-Label, Parallel Group, Randomised Study To Compare the Efficacy of AZD6244 vs Temozolomide in Patients With Unresectable AJCC Stage 3 or 4 Malignant Melanoma [NCT00338130]Phase 2239 participants (Actual)Interventional2006-07-31Completed
Phase I Trial of a 5-day Regimen of Temodar Plus O6-Benzylguanine (O6-BG) in the Treatment of Patients With Recurrent / Progressive Glioblastoma Multiforme [NCT00612989]Phase 142 participants (Anticipated)Interventional2005-02-28Completed
A Multicenter Randomized Double-blinded Controlled Phase 2 Study Evaluating the Efficacy of Valganciclovir as add-on Therapy in Glioblastoma Patients [NCT04116411]Phase 2220 participants (Anticipated)Interventional2019-09-04Recruiting
Randomized Phase II Neoadjuvant Study of Temozolomide Alone or With Pegylated Interferon-alpha 2b in Patients With Resectable American Joint Committee on Cancer (AJCC) Stage IIIB/IIIC or Stage IV (M1a) Metastatic Melanoma [NCT00525031]Phase 255 participants (Actual)Interventional2006-08-31Completed
Phase III Trial Evaluating Maintenance Treatment Versus Observation in Elderly Patients Suffering From Primary Central Nervous System Lymphoma in Complete Remission After High Dose Methotrexate Based Chemotherapy in First Line [NCT02313389]Phase 3428 participants (Actual)Interventional2015-09-30Active, not recruiting
A Phase II Trial Evaluating the Effects of Bortezomib in Patients With Recurrent Malignant Gliomas Treated Prior to Surgery and Then Bortezomib and Temozolomide Post-operatively [NCT00990652]Phase 210 participants (Actual)Interventional2009-05-31Completed
Phase II Trial of Temodar Plus O6-Benzylguanine (O6-BG) (NSC 637037) in the Treatment of Patients With Temodar-Resistant Malignant Glioma [NCT00613093]Phase 267 participants (Actual)Interventional2002-10-31Completed
A Phase I Dose-Escalation Study Of Azacitidine In Combination With Temozolomide In Patients With Unresectable Or Metastatic Soft Tissue Sarcoma or Malignant Mesothelioma [NCT00629343]Phase 128 participants (Actual)Interventional2007-10-31Active, not recruiting
Phase II Trial of Vaccination With Lysate-loaded, Mature Dendritic Cells Integrated Into Standard Radiochemotherapy in Newly Diagnosed Glioblastoma [NCT03395587]Phase 2136 participants (Anticipated)Interventional2018-03-06Recruiting
A Phase II Study of Gliadel, Concomitant Temozolomide and Radiation, Followed By Dose Dense Therapy With Temozolomide For Newly Diagnosed Malignant High Grade Glioma [NCT00660283]Phase 220 participants (Anticipated)Interventional2009-01-31Active, not recruiting
An Open Label, Randomised, Phase II Study to Investigate the Efficacy and Safety of ALECSAT Treatment as an add-on Therapy to Radiotherapy and Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT02799238]Phase 262 participants (Actual)Interventional2016-03-31Completed
Phase I/Randomized Phase II Trial of Either Dasatinib or Placebo Combined With Standard Chemo-Radiotherapy for Newly Diagnosed Glioblastoma Multiforme (GBM) [NCT00869401]Phase 1/Phase 2217 participants (Actual)Interventional2009-06-30Completed
A Proof-of-concept Clinical Trial Assessing the Safety of the Coordinated Undermining of Survival Paths by 9 Repurposed Drugs Combined With Metronomic Temozolomide (CUSP9v3 Treatment Protocol) for Recurrent Glioblastoma [NCT02770378]Phase 1/Phase 210 participants (Actual)Interventional2016-11-30Completed
Zenapax®-Activated Peptide ImmunoTherapy [ZAP IT] [NCT00626015]Phase 116 participants (Actual)Interventional2007-03-31Completed
Phase II Trial of Molecularly Determined Treatment of Children and Young Adults With Newly Diagnosed Diffuse Intrinsic Pontine Gliomas [NCT01182350]Phase 253 participants (Actual)Interventional2011-09-30Terminated(stopped due to Per design: Stop if fewer than 3 cohorts by molecular classification were with 10 participants or only 1 cohort enrolled at least 15 participants of 50.)
Phase I Study of the Combination of Sorafenib and Radiation Therapy -/+ Temozolomide for the Treatment of Patients With Brain Metastases and Primary Brain Tumors [NCT00639262]Phase 135 participants (Actual)Interventional2008-03-31Completed
[NCT02663440]Phase 241 participants (Anticipated)Interventional2016-01-31Recruiting
Cilengitide in Subjects With Newly Diagnosed Glioblastoma and Unmethylated MGMT Gene Promoter - a Multicenter, Open-label Phase II Study, Investigating Two Cilengitide Regimens in Combination With Standard Treatment (Temozolomide With Concomitant Radiatio [NCT00813943]Phase 2265 participants (Actual)Interventional2009-03-31Completed
Combination of Checkpoint Inhibition and IDO1 Inhibition Together With Standard Radiotherapy or Chemoradiotherapy in Newly Diagnosed Glioblastoma. A Phase 1 Clinical and Translational Trial [NCT04047706]Phase 118 participants (Actual)Interventional2019-08-13Active, not recruiting
[NCT00967330]Phase 2182 participants (Actual)Interventional2010-06-30Completed
A Phase 1 Study of Veliparib (ABT-888) in Combination With Capecitabine and Temozolomide in Advanced Well-Differentiated Neuroendocrine Tumors [NCT02831179]Phase 10 participants (Actual)Interventional2017-12-31Withdrawn(stopped due to Loss of funding support)
A Multicenter, Open-Label, Randomized, Active-Controlled Parallel Groups Study Comparing the Efficacy and Safety of Temodal vs Semustine in the Treatment of Subjects With Recurrent Glioblastoma or Anaplastic Astrocytoma [NCT00335075]Phase 3151 participants (Actual)Interventional2005-03-02Completed
Phase II Study of Temozolomide (Temodal) in Children Over 1 Year of Age With Relapsed or Refractory High Risk Neuroblastoma [NCT00276679]Phase 20 participants Interventional2003-04-30Completed
Continuous Temozolomide (SCH 52365) in Patients With Advanced or Metastatic Soft Tissue Sarcoma or Metastatic Breast Cancer, Phase II [NCT00194766]Phase 235 participants (Actual)Interventional2000-07-31Completed
Phase II Trial of Temozolomide, Carboplatin and Neupogen in High-Grade Gliomas, Both Newly-Diagnosed and Recurrent [NCT00006263]Phase 20 participants (Actual)Interventional1997-11-30Withdrawn
Phase II Study of 7 Days On/7 Days Off Temozolomide in Patients With High-Grade Glioma [NCT00619112]Phase 260 participants (Actual)Interventional2007-10-31Completed
Phase I/II Trial of the Combination of Decitabine and Temozolomide in the Treatment of Patients With Metastatic Melanoma [NCT00715793]Phase 1/Phase 239 participants (Actual)Interventional2008-06-30Completed
A Phase I Study of ABT-888, an Oral Inhibitor of Poly(ADP-Ribose) Polymerase and Temozolomide in Children With Recurrent/Refractory CNS Tumors [NCT00994071]Phase 19 participants (Actual)Interventional2009-09-22Completed
A Prospective, Single-arm, Single-center, Multi-cohort Phase II Clinical Study of HER2-positive and Triple-negative Breast Cancer Brain Metastases [NCT04303988]Phase 259 participants (Anticipated)Interventional2020-03-30Not yet recruiting
A Randomised Controlled Phase II Trial of Temozolomide With or Without Cannabinoids in Patients With Recurrent Glioblastoma [NCT05629702]Phase 2234 participants (Anticipated)Interventional2023-02-03Recruiting
Phase I/II Study of Vorinostat (Suberoylanilide Hydroxamic Acid [SAHA]), Temozolomide, and Radiation Therapy in Patients With Newly Diagnosed Glioblastoma [NCT00731731]Phase 1/Phase 2125 participants (Actual)Interventional2009-07-10Completed
Randomized Phase II/III Trial of Radiotherapy Plus Concomitant and Adjuvant Temozolomide With or Without Hydroxychloroquine, Rapamycin for Newly Diagnosed Glioblastoma [NCT03008148]Phase 2/Phase 3288 participants (Anticipated)Interventional2018-10-11Recruiting
Primary Chemotherapy With Temozolomide Versus Radiotherapy in Patients With Low Grade Gliomas After Stratification for Genetic 1p Loss: A Phase III Study [NCT00182819]Phase 3709 participants (Actual)Interventional2005-07-31Completed
Phase 1 Study of Temozolomide Associated With Topotecan in Refractory or Relapsed Malignant Tumors in Children and Adolescents [NCT00412503]Phase 10 participants InterventionalCompleted
A Phase 1 Study of Temozolomide in Combination With Targeted Therapy for NSCLC Patients With CNS Progression on Either Osimertinib or Lorlatinib [NCT04541407]Phase 11 participants (Actual)Interventional2020-10-29Completed
Pilot Study of Spectroscopic MRI-Guided, Dose-Escalated Radiation Therapy for Newly-Diagnosed Glioblastoma [NCT03137888]Phase 234 participants (Actual)Interventional2017-09-20Active, not recruiting
The Efficacy and Safety of Temozolomide in Patients With Relapsed or Advanced Anaplastic Oligodendroglioma and Oligoastrocytoma: a Multicenter, Single-arm, Phase II Trial [NCT01847235]Phase 223 participants (Actual)Interventional2013-05-31Completed
IIT2016-17-HU-KETORADTMZ: A Phase 1 Study of a 4-month Ketogenic Diet in Combination With Standard-of-care Radiation and Temozolomide for Patients With Newly/Recently Diagnosed Glioblastoma [NCT03451799]Phase 121 participants (Actual)Interventional2018-04-13Active, not recruiting
Phase I Study of Olaparib and Temozolomide in Adult Patients With Recurrent/Metastatic Ewing's Sarcoma or Rhabdomyosarcoma Following Failure of Prior Chemotherapy [NCT01858168]Phase 193 participants (Anticipated)Interventional2013-07-31Recruiting
A Double-Blind, Placebo-Controlled, Randomised, Phase II Study Evaluating the Efficacy and Safety of Addition of Continuous Multiple Line Bevacizumab Treatment to Lomustine in Second (2nd)-Line Followed by Standard of Care (SOC) in Third (3rd)-Line and Be [NCT01860638]Phase 2296 participants (Actual)Interventional2013-08-19Completed
Positron Emission Tomography With Fluoro-misonidazole (PET-FMISO) in High Grade Gliomas: Assessment of Tumor Hypoxia and Effect of Spinal Cord Stimulation [NCT01868906]Phase 26 participants (Actual)Interventional2013-06-30Terminated(stopped due to Isotope (FMISO) production is no longer available in our country.)
Zanubrutinib Combined With Rituximab and Chemotherapy for Newly-Diagnosed Primary Central Nervous System Large B-Cell Lymphoma Patients Intolerant to Haematopoietic Stem Cell Transplantation [NCT05896007]Phase 229 participants (Anticipated)Interventional2023-07-13Recruiting
A Phase 1/2 Study of Selinexor in Combination With Standard of Care (SoC) Therapy for Newly Diagnosed or Recurrent Glioblastoma [NCT04421378]Phase 1/Phase 274 participants (Actual)Interventional2020-06-08Terminated(stopped due to Upon further consideration of the existing data and the competitive landscape, Karyopharm Therapeutics Inc has decided not to pursue the ongoing development of Selinexor in GBM at this time.)
Avastin in Combination With Temozolomide and Irinotecan for Unresectable or Multifocal Glioblastoma Multiformes and Gliosarcomas [NCT00979017]Phase 241 participants (Actual)Interventional2009-11-30Completed
Phase II Quality of Life Study of Stereotactic RadioSurgery, Temozolomide and Erlotinib Chemotherapy for the Treatment of 1-3 Brain Metastases in Non-small Cell Lung Cancer [NCT00385398]Phase 20 participants (Actual)Interventional2008-07-31Withdrawn(stopped due to lack of funding and drug supply)
High-Dose Methotrexate Plus Steroid Followed by Concurrent Whole Brain Chemoradiation With Temozolomide for Immunocompetent Patients With Primary Central Nervous System Lymphoma - a Phase II Study [NCT00455286]Phase 225 participants Interventional2006-11-30Recruiting
A Phase I Study of Imatinib Mesylate in Combination With Temozolomide in Patients With Malignant Glioma [NCT00354068]Phase 165 participants (Actual)Interventional2004-07-31Completed
A Single-Center, Open-Label, Phase II, Safety and Efficacy Study of Panzem Nanocrystal Colloidal Dispersion Administered Orally in Combination With Protracted Oral Fixed-Dose Temozolomide to Patients With Recurrent Glioblastoma Multiforme [NCT00481455]Phase 215 participants (Actual)Interventional2007-04-30Completed
Phase I Trial Of Temozolomide Combined With The Histone Deacetylase Inhibitor Valproic Acid (VPA) And Whole Brain Radiation Therapy (WBR) For Brain Metastases From Solid Tumors In Adults [NCT00437957]Phase 110 participants (Actual)Interventional2006-12-31Terminated(stopped due to Lack of enrollment)
A Phase II Pilot Trial of Radiation Therapy With Concurrent and Adjuvant Temozolomide, Tamoxifen and Carboplatin (T2C) in the Treatment of Patients With Primary Central Nervous System Malignant Gliomas [NCT00492687]Phase 280 participants (Anticipated)Interventional2006-12-31Recruiting
A Pharmacokinetic and Pharmacogenomic Study of Patients With High-Grade Gliomas Receiving Daily Radiation Therapy and Temozolomide [NCT00471653]10 participants (Actual)Observational2006-11-11Terminated(stopped due to low accrual)
Phase II Trial of Temozolomide in Elderly Patients With Glioblastoma and Poor Performance Status (KPS<70). [NCT01242566]Phase 270 participants (Actual)Interventional2007-07-31Completed
A Randomized Phase II/III Open-Label Study of Ipilimumab and Nivolumab Versus Temozolomide in Patients With Newly Diagnosed MGMT (Tumor O-6-Methylguanine DNA Methyltransferase) Unmethylated Glioblastoma [NCT04396860]Phase 2/Phase 3485 participants (Anticipated)Interventional2020-09-01Active, not recruiting
An Open Label, Multicentre, Long-Term Extension Study of Tislelizumab- Containing Treatment and/or Pamiparib-Containing Treatment in Patients With Advanced Malignancies [NCT04164199]Phase 3300 participants (Anticipated)Interventional2019-12-19Enrolling by invitation
A Phase IIa Study of AdV-tk + Valacyclovir Gene Therapy in Combination With Standard Radiation Therapy for Malignant Gliomas [NCT00589875]Phase 252 participants (Actual)Interventional2007-03-31Completed
Multicenter Randomized Phase II Study of Methotrexate (MTX) and Temozolomide Versus MTX, Procarbazine, Vincristine and Cytarabine for Primary CNS Lymphoma (PCNSL) in the Elderly [NCT00503594]Phase 292 participants (Anticipated)Interventional2007-07-31Recruiting
A Phase I Study of ABT-888 in Combination With Temozolomide (TMZ) in Subjects With Non-Hematologic Malignancies (NHM) and Metastatic Melanoma (MM) [NCT00526617]Phase 141 participants (Actual)Interventional2007-08-31Completed
A Phase II Multi-Centre Study of Concomitant and Prolonged Adjuvant Temozolomide With Radiotherapy in Diffuse Pontine Gliomas [NCT00514397]Phase 243 participants (Anticipated)Interventional2008-01-31Recruiting
A Phase I Study of Temozolomide, Thalidomide, and Lomustine (TTL) in Patients With Metastatic Melanoma in the Brain [NCT00527657]Phase 117 participants (Actual)Interventional2006-02-09Completed
Clinical Analysis of Naxitamab (hu3F8) in the Treatment of Pediatric High Risk or Refractory/ Relapsed Neuroblastoma [NCT06013618]Phase 230 participants (Anticipated)Interventional2023-06-19Recruiting
A Phase I/II Combination Study of NMS-03305293 and Temozolomide in Adult Patients With Recurrent Glioblastoma [NCT04910022]Phase 1/Phase 275 participants (Anticipated)Interventional2021-12-01Recruiting
Temozolomide and Olaparib for O6-Methylguanine DNA Methyltransferase Promoter Hypermethylated Colorectal Cancer [NCT04166435]Phase 211 participants (Actual)Interventional2020-06-17Completed
A Pilot Trial of Irinotecan, Temozolomide and Bevacizumab in Combination With Existing High Dose Alkylator Based Chemotherapy for Treatment of Newly Diagnosed Patients With Desmoplastic Small Round Cell Tumor [NCT01189643]Early Phase 115 participants (Actual)Interventional2010-08-31Active, not recruiting
Temozolomide in Treatment of Patients With Primary Central Nervous System Lymphoma [NCT02934204]Phase 251 participants (Anticipated)Interventional2016-02-29Recruiting
A Phase II Pilot Study of Cyclophosphamide, Doxorubicin, Vincristine Alternating With Irinotecan and Temozolomide in Patients With Newly Diagnosed Metastatic Ewing's Sarcoma [NCT01313884]Phase 23 participants (Actual)Interventional2011-05-31Terminated(stopped due to Study did not reach primary objective; study did not accrue enough patients.)
A Pilot Feasibility Study of Intracerebral Microdialysis to Determine the Neuropharmacokinetics of Temozolomide [NCT00547131]Phase 112 participants (Anticipated)Interventional2006-01-31Completed
A Phase I Study of Bortezomib and Temozolomide in Patients With Refractory Solid Tumors [NCT00544284]Phase 125 participants (Actual)Interventional2005-01-31Completed
Temozolomide Plus Bevacizumab Chemotherapy in Supratentorial Glioblastoma in 70 Years and Older Patients With an Impaired Functional Status (KPS<70) [NCT02898012]Phase 270 participants (Actual)Interventional2010-10-31Completed
A Phase II Trial of Concurrent Sunitinib, Temozolomide and Radiation Therapy Followed by Adjuvant Temozolomide for Newly Diagnosed Glioblastoma Patients With an Unmethylated MGMT Gene Promoter [NCT02928575]Phase 245 participants (Anticipated)Interventional2012-08-31Recruiting
A Phase I/II Study of the Combination of Indoximod and Temozolomide for Adult Patients With Temozolomide-Refractory Primary Malignant Brain Tumors [NCT02052648]Phase 1/Phase 2160 participants (Actual)Interventional2014-03-31Completed
Phase I / II Adaptive Randomized Trial of Vorinostat, Isotretinoin and Temozolomide in Adults With Recurrent Glioblastoma Multiforme [NCT00555399]Phase 1/Phase 2135 participants (Anticipated)Interventional2007-11-28Active, not recruiting
Temozolomide With Irinotecan Versus Temozolomide, Irinotecan Plus Bevacizumab (NSC# 704865) for Recurrent/Refractory Medulloblastoma/CNS PNET of Childhood, a COG Randomized Phase II Screening Trial [NCT01217437]Phase 2108 participants (Actual)Interventional2010-11-22Completed
A Phase 1b/2 Study of Abemaciclib in Combination With Irinotecan and Temozolomide (Part A) and Abemaciclib in Combination With Temozolomide (Part B) in Pediatric and Young Adult Patients With Relapsed/Refractory Solid Tumors and Abemaciclib in Combination [NCT04238819]Phase 1/Phase 2117 participants (Anticipated)Interventional2020-11-09Recruiting
Phase I/II Study of T-DM1 Alone Versus T-DM1 and Metronomic Temozolomide in Secondary Prevention of HER2-Positive Breast Cancer Brain Metastases Following Stereotactic Radiosurgery [NCT03190967]Phase 1/Phase 212 participants (Actual)Interventional2018-04-18Terminated(stopped due to The phase II portion was never started as we could no longer get the drug from the manufacturer.)
A Pilot Study to Assess Feasibility of 5 Fraction Hypofractionated Stereotactic Radiosurgery Along With Standard Temozolomide as a Lymphocyte Sparing Therapy for Glioblastoma Multiforme [NCT03291990]Early Phase 13 participants (Actual)Interventional2017-10-18Completed
Phase I Trial of Vaccination With Autologous Dendritic Cells Pulsed With Lysate Derived From an Allogeneic Glioblastoma Stem-like Cell Line for Patients With Newly Diagnosed or Recurrent Glioblastoma [NCT02010606]Phase 139 participants (Actual)Interventional2014-01-08Completed
Treatment of Newly Diagnosed High-Grade Gliomas in Patients Ages Greater Than or Equal to 3 and Less Than or Equal to 21 Years With a Phase II Irinotecan Window Followed by Radiation Therapy and Temozolomide [NCT00004068]Phase 253 participants (Actual)Interventional1999-03-31Completed
A Randomized Double-Blind, Placebo-Controlled Phase II Study of the Matrix Metalloprotease Inhibitor Prinomastat in Combination With Temozolomide Following Radiation Therapy in Patients Having Newly Diagnosed Glioblastoma Multiforme [NCT00004200]Phase 20 participants Interventional1999-10-31Completed
A Phase 1B, Open-Label, Dose Escalation Study Evaluating the Safety of BSI-201 in Combination With Chemotherapeutic Regimens in Subjects With Advanced Solid Tumors [NCT00422682]Phase 1136 participants (Actual)Interventional2007-01-31Completed
A Phase I, Two-stage, Multi-center, Open Label, Dose-escalation Study of BKM120 in Combination With Adjuvant Temozolomide and With Concomitant Radiation Therapy and Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT01473901]Phase 138 participants (Actual)Interventional2011-12-30Completed
A Phase I Open Label Safety Study to Evaluate the Pharmacokinetic Profile and Tolerance of Mibefradil Dose Finding in Subjects With Recurrent High-Grade Glioma Undergoing Standard, Repeated Temozolomide Treatment [NCT01480050]Phase 128 participants (Actual)Interventional2012-05-31Completed
A Phase I Study Of Cloretazine™ (VNP40101M) And Temozolomide In Patients With Hematologic Malignancies [NCT00098436]Phase 125 participants (Anticipated)Interventional2004-09-30Completed
Phase I Dose Finding Clinical Trial of Combination Paclitaxel, Carboplatin and Temozolomide for Subjects With Solid Tumor Malignancies. [NCT00249964]Phase 115 participants (Actual)Interventional2003-11-30Completed
A Systemic Temozolomide Treatment Of Melanoma Present In The Central Nervous System [NCT00068666]Phase 241 participants (Actual)Interventional2004-01-31Terminated
A Phase II Trial of Combination Thalidomide Plus Temozolomide in Patients With Metastatic Malignant Melanoma [NCT00104988]Phase 264 participants (Actual)Interventional2005-06-30Completed
A Phase III Trial Comparing Whole Brain Radiation (WBRT) and Stereotactic Radiosurgery (SRS) Alone Versus With Temozolomide or Erlotinib in Patients With Non-Small Cell Lung Cancer and 1-3 Brain Metastases [NCT00268684]Phase 3381 participants Interventional2005-05-31Recruiting
A Randomized Controlled Trial of Chemo-Radiotherapy Versus Biomarker-Guided Therapy for Elderly and Frail Patients With Newly Diagnosed Glioblastoma [NCT04765514]Phase 2121 participants (Anticipated)Interventional2022-07-27Recruiting
A Randomized, Open-Label Phase 2 Study of Temozolomide Added to Whole Brain Radiation Therapy Versus Whole Brain Radiation Therapy Alone for the Treatment of Brain Metastasis From Non-Small Cell Lung Cancer [NCT00076856]Phase 295 participants (Actual)Interventional2004-03-31Completed
A Phase II Study of Radiation With Concomitant and Then Sequential Temozolomide in Patients With Newly Diagnosed Supratentorial Malignant Glioma Who Have Undergone Surgery With Gliadel Wafer Insertion [NCT00283543]Phase 240 participants (Anticipated)Interventional2002-09-30Active, not recruiting
A Phase I Trial and Pharmacokinetic Study of Talabostat (PT-100, Val-Boro-Pro) in Combination With Temozolomide or Carboplatin in Pediatric Patients With Relapsed or Refractory Solid Tumors Including Brain Tumors [NCT00303940]Phase 126 participants (Anticipated)Interventional2005-12-31Completed
Phase I Study Of SCH66336 (Lonafarnib), A Farnesyl Protein Transferase Inhibitor In Combination With Temozolomide In Gliomas [NCT00083096]Phase 130 participants (Anticipated)Interventional2004-03-31Active, not recruiting
Treatment of Primary Glioblastoma Multiforme With Cetuximab, Radiotherapy and Temozolomide (GERT) - Phase I/II Trial [NCT00311857]Phase 1/Phase 246 participants Interventional2006-02-28Recruiting
Phase I Study of Convection Enhanced Delivery (CED) of IL13-PE38QQR Infusion After Resection Followed by Radiation Therapy With or Without Temozolomide in Patients With Newly Diagnosed Supratentorial Malignant Glioma [NCT00089427]Phase 124 participants Interventional2004-07-31Completed
A Randomized, Double-Blind, Placebo-Controlled Phase 3 Study of Enzastaurin Added to Temozolomide During and Following Radiation Therapy in Newly Diagnosed Glioblastoma Patients Who Possess the Novel Genomic Biomarker DGM1 [NCT03776071]Phase 3300 participants (Anticipated)Interventional2020-12-16Active, not recruiting
Phase I Study of Ibrutinib With Radiation and Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT03535350]Phase 136 participants (Anticipated)Interventional2018-08-24Active, not recruiting
Phase I Study of Ruxolitinib With Radiation and Temozolomide in Patients With Newly Diagnosed Grade III Gliomas and Glioblastoma [NCT03514069]Phase 160 participants (Actual)Interventional2018-06-05Active, not recruiting
A Phase I Study of CCI-779, and Temozolomide in Combination With Radiation Therapy in Glioblastoma Multiforme [NCT00316849]Phase 156 participants (Actual)Interventional2006-05-31Completed
Doxorubicin Hydrochloride Liposome Combined With Irinotecan (AI Regimen) Versus VIT Regimen in the Treatment of First Relapsed and Refractory Pediatric Rhabdomyosarcoma: a Prospective, Open-label, Randomized Controlled, Phase II Clinical Study [NCT05457829]Phase 288 participants (Anticipated)Interventional2023-12-30Not yet recruiting
Randomized Phase II of TARCEVA™ (Erlotinib) Versus Temozolomide Or BCNU in Patients With Recurrent Glioblastoma Multiforme [NCT00086879]Phase 2110 participants (Actual)Interventional2004-05-31Completed
A Multi-Center Phase III, Randomized, Open-Label Trial of Vigil (Bi-shRNAfurin and GMCSF Augmented Autologous Tumor Cell Immunotherapy) in Combination With Irinotecan and Temozolomide as a Second-Line Regimen for Ewing's Sarcoma [NCT03495921]Phase 332 participants (Actual)Interventional2018-08-21Terminated(stopped due to Slow accrual and as a result, a strategic business decision was made to terminate enrollment.)
DC Migration Study to Evaluate TReg Depletion In GBM Patients With and Without Varlilumab [NCT03688178]Phase 243 participants (Actual)Interventional2020-08-26Active, not recruiting
A Phase I Trial of Pembrolizumab and Vorinostat Combined With Temozolomide and Radiation Therapy for Newly Diagnosed Glioblastoma [NCT03426891]Phase 121 participants (Actual)Interventional2018-03-16Completed
A Phase I Study Of Oral Irinotecan, Temozolomide, Cefixime In Children With Recurrent/Resistant High-Risk Neuroblastoma [NCT00093353]Phase 130 participants (Anticipated)Interventional2004-05-31Completed
A Pilot Study to Evaluate Novel Agents (Temozolomide and Cixutumumab [IMC-A12, Anti-IGF-IR Monoclonal Antibody NSC # 742460]) in Combination With Intensive Multi-agent Interval Compressed Therapy for Patients With High-Risk Rhabdomyosarcoma [NCT01055314]Phase 2175 participants (Actual)Interventional2010-01-31Completed
A Phase II Study Of Radiation Therapy Plus Low Dose Temozolomide Followed By Temozolomide Plus Irinotecan For Glioblastoma Multiforme [NCT00099125]Phase 2170 participants (Actual)Interventional2004-11-30Completed
Phase I Trial of Weekly Docetaxel and Daily Temozolomide in Patients With Metastatic Disease [NCT00401180]Phase 125 participants (Actual)Interventional2002-06-30Completed
A Randomized, Controlled Phase II Study to Compare Capecitabine Combined With Dacarbazine(CAPDTIC) Versus Capecitabine Combined Temozolomide(CAPTEM) in Advanced and Metastatic Gastrointestinal Pancreatic and Esophageal Neuroendocrine Tumor [NCT03279601]Phase 2148 participants (Anticipated)Interventional2017-09-01Recruiting
A Randomized, Multi-Cohort Phase II Trial of Temozolomide and Atezolizumab as Second or Third Line Treatment for Small Cell Lung Cancer [NCT04919382]Phase 256 participants (Anticipated)Interventional2022-01-26Recruiting
A Phase II Study of Bevacizumab in Combination With Temozolomide in Patients With Advanced Neuroendocrine Tumors [NCT00137774]Phase 234 participants (Actual)Interventional2004-11-30Completed
A Phase I Study of Dietary Methionine Restriction and Temodar® (Temozolomide) for the Treatment of Recurrent and Progressive Glioblastoma Multiforme [NCT00508456]Phase 118 participants (Actual)Interventional2004-08-31Terminated(stopped due to Low Accrual)
A Randomized Phase 2/3 Study of Olaparib Plus Temozolomide Versus Investigator's Choice for the Treatment of Patients With Advanced Uterine Leiomyosarcoma After Progression on Prior Chemotherapy [NCT05432791]Phase 2/Phase 3190 participants (Anticipated)Interventional2023-03-30Recruiting
A Phase II Trial to Evaluate the Effect of Low Dose Temozolomide (TMZ) for 2 Weeks on Brain Tumor O-6-methylguanine-DNA Methyltransferase (MGMT) Activity in Patients With Gliomas. [NCT00424554]Phase 240 participants (Actual)Interventional2006-09-26Completed
Combination of 6-Thioguanine, Capecitabine, Celecoxib and Temozolomide or CCNU for Recurrent Anaplastic Glioma and Glioblastoma Multiforme [NCT00504660]Phase 275 participants (Actual)Interventional2003-09-30Completed
Avastin in Combination With Radiation and Temozolomide, Followed by Avastin, Temozolomide and Irinotecan for Glioblastoma Multiformes and Gliosarcomas [NCT00597402]Phase 2125 participants (Actual)Interventional2007-07-31Completed
Phase II Trial of Velcade (Bortezomib) in Combination With Temozolomide and Regional Radiation Therapy for Upfront Treatment of Patients With Newly-diagnosed Glioblastoma Multiforme [NCT00998010]Phase 224 participants (Actual)Interventional2011-10-03Completed
A Phase II Study of ABT-888, an Inhibitor of Poly(ADP-ribose) Polymerase (PARP) in Combination With Temozolomide in Patients With Heavily Pretreated, Metastatic Colorectal Cancer [NCT01051596]Phase 275 participants (Actual)Interventional2009-09-30Completed
Phase II Trial of Treatment Intensification for IDH Wildtype, Non-Histological Glioblastoma, Gliomas (IDH Wildtype Lower Grade Glioma Treatment Intensification) [NCT04623931]Phase 240 participants (Anticipated)Interventional2020-01-30Recruiting
Phase I / II Adaptive Randomized Trial of Vorinostat, Erlotinib and Temozolomide in Adults With Recurrent Glioblastoma Multiforme [NCT01110876]Phase 1/Phase 221 participants (Actual)Interventional2011-06-30Terminated(stopped due to Unanticipated Toxicities)
Phase I/II Intraventricular DepoCyt (OD # 06-2348) in Glioblastoma (76,730, 11/06) [NCT01044966]Phase 1/Phase 212 participants (Actual)Interventional2009-09-30Terminated(stopped due to The study was terminated due to lack of adequate patient enrollment into trial.)
A Phase II Study of Thalidomide in Combination With Temodar in Patients With Metastatic Neuroendocrine Tumors [NCT00165230]Phase 232 participants Interventional2002-05-31Completed
Neoadjuvant PD-1 in Newly Diagnosed Glioblastoma: A Phase 2 Clinical Trial [NCT04583020]Phase 242 participants (Anticipated)Interventional2020-11-12Recruiting
Multi-national, Open-label, Active-controlled, Randomized Dose-finding Study to Evaluate Efficacy of 2 Doses of AP 12009 in Recurrent Glioma, Administered Intratumorally as Continuous High-flow Microperfusion Over 7 Days Every Other Week [NCT00431561]Phase 2141 participants (Actual)Interventional2003-04-30Completed
A Phase II Study of the PARP Inhibitor Olaparib in Combination With the DNA Damaging Agent Temozolomide for the Treatment of Advanced Uterine Leiomyosarcoma [NCT03880019]Phase 223 participants (Actual)Interventional2019-08-19Active, not recruiting
A Phase I/II Trial of Cloretazine® (VNP40101M) and Temodar® (Temozolomide) for Patients With Malignant Glioma in First Relapse or Progression [NCT00516282]Phase 114 participants (Actual)Interventional2007-08-31Terminated(stopped due to The pharmaceutical collaborator filed for bankruptcy and as a result, the study was unable to move into the phase II portion.)
Temozolomide and Irinotecan Consolidation in Patients With MGMT Silenced, Microsatellite Stable Colorectal Cancer With Persistence of Minimal Residual Disease in Liquid Biopsy After Standard Adjuvant Chemotherapy: the ERASE-TMZ Study [NCT05031975]Phase 235 participants (Anticipated)Interventional2022-05-02Recruiting
An Exploratory Study of PD-1 Antibody(Toripalimab) or Combining With Temozolomide for Injection in the Treatment of Advanced/Metastatic Malignant Melanoma [NCT04884997]Phase 290 participants (Anticipated)Interventional2021-03-07Recruiting
A Phase II Study of Optune (NovoTTF) in Combination With Bevacizumab (BEV) and Temozolomide (TMZ) in Patients With Newly Diagnosed Unresectable Glioblastoma (GBM) [NCT02343549]Phase 213 participants (Actual)Interventional2015-01-31Terminated(stopped due to closed to accrual due to low accrual)
Stereotactic Accelerated Radiotherapy in GlioblastomA (SAGA) [NCT05781321]Phase 2170 participants (Anticipated)Interventional2023-03-23Recruiting
A Randomized Phase 2 Study to Evaluate the Efficacy Between Only Radiotherapy Versus CCRT With Temozolomide in Newly Diagnosed Grade III Gliomas Without 1p/19q Codeletion [NCT01534845]Phase 290 participants (Actual)Interventional2012-03-31Active, not recruiting
Combination of IDO/Survivin Peptide Vaccine, GM-CSF, Imiquimod and Temozolomide Chemotherapy for Patients With Metastatic Malignant Melanoma [NCT01543464]Phase 241 participants (Actual)Interventional2012-05-31Terminated(stopped due to Diminished rectruitment)
Concomitant and Adjuvant Temozolomide and Radiotherapy for Newly Diagnosed Glioblastoma Multiforme - A Randomized Phase III Study [NCT00006353]Phase 3575 participants (Actual)Interventional2000-07-31Completed
Phase II Trial of Continuous Dose Temozolomide in Patients With Newly Diagnosed Pure and Mixed Anaplastic Oligodendroglioma [NCT00400816]Phase 229 participants (Actual)Interventional2005-08-31Completed
Phase II Trial of Salvage Chemotherapy With Temozolomide in Combination With Topotecan for Primary CNS Lymphoma [NCT00109798]0 participants (Actual)Interventional2005-03-31Withdrawn(stopped due to Sub-Investigator reloacted to another institution)
Phase 2 Study of Temozolomide Plus Capecitabine in Patients With Grade 3 and Low Ki-67 Gastroenteropancreatic Neuroendocrine Tumors [NCT03079440]Phase 231 participants (Actual)Interventional2017-05-15Completed
Phase 2 Study to Evaluate the Efficacy and Safety of Sintilimab in Combination With Bevacizumab and Temozolomide in Recurrent Glioblastoma (GBM) Patients [NCT05638451]Phase 230 participants (Anticipated)Interventional2023-01-01Not yet recruiting
Temozolomide Versus Temozolomide + Whole Brain Radiation In Stage IV Melanoma Patients With Asymptomatic Brain Metastases [NCT00020839]Phase 323 participants (Actual)Interventional2001-04-30Terminated(stopped due to low accrual)
Phase I/II Trial Of Temozolomide And Carboplatin In Recurrent Glioblastoma Multiforme [NCT00021307]Phase 1/Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to Study not activated.)
Dose Escalation of Temozolomide in Combination With Thiotepa and Carboplatin With Autologous Stem Cell Rescue in Patients With Malignant Brain Tumors With Minimal Residual Disease [NCT00025558]Phase 10 participants Interventional2000-10-31Completed
A Phase I Study Of ZD 1839 And Temozolomide For The Treatment Of Gliomas [NCT00027625]Phase 10 participants Interventional2002-01-28Completed
A Phase II Study of Temozolomide (Temodar) and Peglated Interferon Alfa-2B (PEGIntron) in the Treatment of Advanced Melanoma [NCT00027742]Phase 20 participants Interventional2001-05-31Completed
A Phase II Study of Temozolomide in the Treatment of Children With High Grade Glioma [NCT00028795]Phase 2170 participants (Actual)Interventional2002-12-31Completed
A Phase II Trial Of Pre-Irradiation And Concurrent Temozolomide In Patients With Newly Diagnosed Anaplastic Oligodendrogliomas And Mixed Anaplastic Oligoastrocytomas [NCT00033280]Phase 242 participants (Actual)Interventional2002-07-31Completed
A Phase II Study of Combination Therapy of a Protracted Oral Schedule of Temozolomide and Thalidomide as First-Line or Subsequent Therapy for Patients With Metastatic, Locally Advanced or Unresectable Leiomyosarcoma [NCT00033709]Phase 20 participants Interventional2002-03-31Active, not recruiting
A Phase II Study of Temozolomide and O6-Benzylguanine (O6-BG) in Patients With Temozolomide-Resistant Anaplastic Glioma [NCT00389090]Phase 232 participants (Actual)Interventional2006-10-31Terminated(stopped due to AOI Pharma terminated the license agreement. IND Transferred to NCI)
Phase I Trial of R115777 With Radiation Therapy and Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00049387]Phase 119 participants (Actual)Interventional2002-09-30Completed
Phase I Trial of Temozolomide and O6-Benzylguanine in Pediatric Patients With Recurrent Brain Tumors [NCT00052780]Phase 172 participants (Actual)Interventional2002-10-31Completed
A Phase II Open-label Study Investigating the Efficacy, Safety and Pharmacokinetic Properties of OKN-007 Combined With Temozolomide in Patients With Recurrent Glioblastoma [NCT04388475]Phase 257 participants (Actual)Interventional2020-06-12Active, not recruiting
A Phase II Study Of Temozolomide And Thalidomide In Patients With Metastatic Melanoma In The Brain [NCT00072163]Phase 250 participants (Actual)Interventional2003-10-31Completed
Dose Intensive Chemotherapy for Children Less Than Ten Years of Age Newly-Diagnosed With Malignant Brain Tumors: A Pilot Study of Two Alternative Intensive Induction Chemotherapy Regimens, Followed by Consolidation With Myeloablative Chemotherapy (Thiotep [NCT00392886]Phase 3120 participants (Anticipated)Interventional2004-03-31Active, not recruiting
Investigation of Cannabis For Tolerability and Feasibility in Patients Receiving Concurrent Chemoradiation for Glioblastoma. [NCT03246113]Phase 11 participants (Actual)Interventional2018-03-19Terminated(stopped due to Lack of subject interest due to study design)
Temozolomide For Patients With Cerebral Metastases Who Have Failed Radiation Therapy [NCT00012116]Phase 24 participants (Actual)Interventional2000-10-31Completed
Phase I/II Trial of Temozolomide and Carboplatin in Recurrent Glioblastoma Multiforme [NCT00014105]Phase 1/Phase 20 participants Interventional2000-12-31Active, not recruiting
A Phase-I Study Of Cyclical Oral Administration Of Temozolomide In Combination With PEG12000-Interferon Alfa-2B In Patients With Refractory And/Or Advanced Solid Tumors [NCT00014261]Phase 10 participants Interventional2000-10-31Completed
A Phase I/II Trial of Arsenic Trioxide and Temozolomide in Combination With Radiation Therapy for Patients With Malignant Gliomas [NCT00275067]Phase 1/Phase 250 participants (Anticipated)Interventional2005-05-31Active, not recruiting
Interleukin 12-Primed Activated T Cells As Therapy For Patients With Metastatic Melanoma (Phase I) [NCT00016055]Phase 118 participants (Anticipated)Interventional2000-11-30Active, not recruiting
Phase I/II Study on Concomitant and Adjuvant Temozolomide and Radiotherapy With or Without PTK787/ZK222584 in Newly Diagnosed GBM [NCT00128700]Phase 1/Phase 220 participants (Actual)Interventional2005-06-30Completed
A Phase 3 Clinical Trial to Evaluate the Safety and Efficacy of Treatment With 2 mg Intralesional Allovectin-7® Compared to Dacarbazine (DTIC) or Temozolomide (TMZ) in Subjects With Recurrent Metastatic Melanoma [NCT00395070]Phase 3390 participants (Actual)Interventional2006-10-31Completed
Phase II Evaluation Temozolomide and Farnesyl Transferase Inhibitor (SCH66336) for the Treatment of Recurrent and Progressive Glioblastoma Multiforme [NCT00038493]Phase 223 participants (Actual)Interventional2001-09-21Completed
An Open-label, Phase I Clinical Trial to Assess the Maximum Tolerated Dose (MTD), Safety and Efficacy of BEY1107 in Combination With Temozolomide in Patient With Recurrent or Progressive Glioblastoma Multiforme (GBM) [NCT05769660]Phase 112 participants (Anticipated)Interventional2022-11-29Recruiting
Pilot Trial of an Implantable Microdevice for In Vivo Drug Sensitivity Testing in Patients With Sarcomas [NCT04199026]Early Phase 120 participants (Anticipated)Interventional2024-01-31Not yet recruiting
Phase I Trial and Pharmacokinetic Study of Temozolomide and O6-Benzylguanine in Childhood Solid Tumors [NCT00020150]Phase 10 participants Interventional2000-06-30Completed
A Phase 1b/2a, Multicenter, Open-Label Study of AQ4N in Combination With Radiation Therapy and Temozolomide, to Evaluate the Safety, Tolerability, and Efficacy in Subjects With Newly Diagnosed Glioblastoma Multiforme [NCT00394628]Phase 1/Phase 260 participants (Anticipated)Interventional2006-10-31Recruiting
A Prospective Randomised Trial Comparing Temozolomide With PCV In The Treatment Of Recurrent WHO Astrocytic Tumours Grades III And IV [NCT00052455]Phase 3500 participants (Anticipated)Interventional2002-10-31Completed
A Phase II Study of Temozolomide (SCH 52365) in Subjects With Brain Metastasis From Non-Small-Cell Lung Cancer [NCT00034697]Phase 224 participants (Actual)Interventional2001-06-28Terminated(stopped due to Slow Enrollment)
A Phase II Study of Temozolomide, Thalidomide, and Lomustine in the Treatment of Advanced Melanoma [NCT00072345]Phase 20 participants Interventional2003-07-31Completed
A Pilot and Phase II Study of OSI-774 and Temozolomide in Combination With Radiation Therapy in Glioblastoma Multiforme [NCT00039494]Phase 2171 participants (Anticipated)Interventional2002-12-31Completed
A Phase III, Randomized, Open-Label Study Of IV Edotecarin Vs Temozolomide Or Carmustine (BCNU) Or Lomustine (CCNU) In Patients With Glioblastoma Multiforme At First Relapse After Alkylator-Based (NEO) Adjuvant Chemotherapy [NCT00068952]Phase 3118 participants Interventional2003-08-31Completed
A Phase I Study of Single Agent OSI-774 (Tarceva) (NSC# 718781, IND# 63383) Followed by OSI-774 With Temozolomide for Patients With Selected Recurrent/Refractory Solid Tumors, Including Brain Tumors [NCT00077454]Phase 195 participants (Actual)Interventional2004-02-29Completed
Phase II Study Of Temozolomide, Thalidomide And Celecoxib In Patients With Newly Diagnosed Glioblastoma Multiforme In The Post-Radiation Setting [NCT00047294]Phase 20 participants Interventional2001-04-30Completed
Phase II Study of the Combination of Cisplatin + Temozolomide in Malignant Glial Tumours in Children and Adolescents at Diagnosis or in Relapse [NCT00147160]Phase 287 participants (Anticipated)Interventional2003-10-31Completed
A Phase II Study Of Whole-Brain Radiation Therapy With Thalidomide And Temozolomide In Patients With Newly Diagnosed Brain Metastases [NCT00049361]Phase 20 participants Interventional2004-01-01Completed
Anlotinib Combined With STUPP Protocol as First-line Regimen for MGMT Nonmethylated Glioblastoma: a Multicenter, Open-label, Single-arm, Phase II Clinical Trial [NCT04725214]Phase 233 participants (Anticipated)Interventional2021-01-15Recruiting
A Phase I Trial Of Temozolomide In Pediatric Patients With Refractory/Recurrent Leukemias [NCT00083070]Phase 116 participants (Actual)Interventional2004-03-31Completed
A Phase II Study of Temozolomide (TEMODAR) in the Treatment of Adult Patients With Supratentorial Low Grade Glioma [NCT00313729]Phase 2120 participants (Actual)Interventional1999-05-31Completed
A Phase 1b Trial of Telaglenastat (CB-839) HCI in Combination With Radiation Therapy and Temozolomide in Patients With IDH-Mutated Diffuse Astrocytoma and Anaplastic Astrocytoma [NCT03528642]Phase 140 participants (Anticipated)Interventional2019-05-01Active, not recruiting
A Multicenter, Open-label, Phase II Pan-Tumor Study in Patients Who Have Participated in Trials to Investigate Efficacy and Safety of ONO-4538 as Monotherapy or in Combination With Other Therapies and Are Continuing ONO-4538 Treatment (ONO-4538-98) [NCT04566380]Phase 259 participants (Anticipated)Interventional2020-09-10Recruiting
A Randomized Phase 3 Single Blind Study of Temozolomide Plus Radiation Therapy Combined With Nivolumab or Placebo in Newly Diagnosed Adult Subjects With MGMT-Methylated (Tumor O6-methylguanine DNA Methyltransferase) Glioblastoma [NCT02667587]Phase 3716 participants (Actual)Interventional2016-05-09Active, not recruiting
"A Surgical Window-of-Opportunity and Phase II Trial of Pembrolizumab, Olaparib and Temozolomide in Recurrent Glioblastoma" [NCT05463848]Phase 278 participants (Anticipated)Interventional2022-10-21Recruiting
An Open-Label Phase 1b Dose-Finding Trial Evaluating the Safety of Niraparib and Temozolomide and Atezolizumab in Participants With Advanced Solid Tumors and Expansion to a Phase 2 Trial Comparing the Effects of Niraparib and Temozolomide Plus Atezolizuma [NCT03830918]Phase 1/Phase 274 participants (Anticipated)Interventional2019-03-06Recruiting
Efficacy of a Protracted Temozolomide Schedule in Patients With Progression After Standard Dose Temozolomide for High-grade Gliomas [NCT00575887]Phase 225 participants (Actual)Interventional2006-08-31Completed
A Phase II Study of Irinotecan and Temozolomide in Breast Cancer Patients With Brian Metastases That Have Progressed After Stereotactic Radiosurgery or Whole Brain Radiation [NCT00617539]Phase 230 participants (Actual)Interventional2005-02-28Completed
Phase 0/I Dose Escalation Study of Mycophenolate Mofetil Combined With Radiation Therapy in Glioblastoma [NCT04477200]Phase 168 participants (Anticipated)Interventional2020-08-05Recruiting
A Phase I Study of Metronomic Temozolomide and Intravenous Ascorbic Acid for Patients With Recurrent High Grade Glioma [NCT02168270]Phase 14 participants (Actual)Interventional2014-06-16Terminated
Phase 2 Study of Sorafenib Plus Protracted Temozolomide in Recurrent Glioblastoma Multiforme [NCT00597493]Phase 232 participants (Actual)Interventional2007-09-30Completed
A Complementary Trial of an Immunotherapy Vaccine Against Tumor-Specific EGFRvIII [NCT00643097]Phase 240 participants (Actual)Interventional2007-09-30Completed
Temodal (Temozolomide) Post Marketing Surveillance Protocol [NCT00723827]682 participants (Actual)Observational2008-03-31Completed
Avastin in Combination With Temozolomide for Unresectable or Multifocal Glioblastoma Multiformes and Gliosarcomas [NCT00612339]Phase 241 participants (Actual)Interventional2007-08-31Completed
A Randomized, Open Label, Two-way Crossover, Single Dose Bioequivalence Study Comparing Dralitem® Capsules to the Reference Drug Temodal® Capsules in Patients With Primary Tumors of the Central Nervous System Under Fasting Conditions [NCT02343081]Phase 424 participants (Actual)Interventional2012-01-31Completed
A Phase I Trial of Hypofraction Radiotherapy + Temozolomide in the Treatment of Patients With Glioblastoma Multiforme and Anaplastic Astrocytoma of the Brain [NCT00841555]Phase 19 participants (Actual)Interventional2009-02-13Completed
Phase I Study of Ipilimumab, Nivolumab, and the Combination in Patients With Newly Diagnosed Glioblastoma [NCT02311920]Phase 132 participants (Actual)Interventional2015-04-16Completed
Phase II Trial of Bevacizumab, Radiation Therapy and Temodar Followed by Bevacizumab and Temodar With Continuation of Bevacizumab Beyond Progression (BBP-Bevacizumab Beyond Progression) [NCT01740258]Phase 268 participants (Actual)Interventional2013-01-31Completed
An Open-Label, Phase 2 Efficacy Study of Temozolomide (TMZ) In Advanced Succinate Dehydrogenase (SDH)-Mutant/Deficient Gastrointestinal Stromal Tumor (GIST) [NCT03556384]Phase 223 participants (Actual)Interventional2018-09-12Active, not recruiting
Phase II Study of Combined Temozolomide and Targeted P53 Gene Therapy (SGT-53) for Treatment of Patients With Recurrent Glioblastoma [NCT02340156]Phase 21 participants (Actual)Interventional2014-12-31Terminated
Phase II Study of TRC102 in Combination With Temozolomide for Recurrent Glioblastoma [NCT02395692]Phase 220 participants (Actual)Interventional2015-12-18Terminated(stopped due to Pre-specified response criteria not met to proceed to next stage of study.)
Phase 1-2 Trial Evaluating Metronomic Chemotherapy in Patients With a Relapsed or Refractory Wilms Tumor [NCT05384821]Phase 1/Phase 228 participants (Anticipated)Interventional2022-09-14Recruiting
Phase II Study of Temozolomide and Radiation in Newly Diagnosed GBM Patients After Resection and Insertion of Gliadel® Wafers [NCT00238277]Phase 215 participants (Actual)Interventional2005-02-15Terminated(stopped due to Terminated - due to low accrual)
Clinical Study of an Dendritic and Glioma Cells Fusion Vaccine With IL-12 for Treatment-naïve GBM Patients. [NCT04388033]Phase 1/Phase 210 participants (Anticipated)Interventional2020-12-31Recruiting
Efficacy of Post-radiation Adjuvant Temozolomide Chemotherapy in Residue Low-grade Glioma [NCT01649830]Phase 3290 participants (Anticipated)Interventional2012-07-31Recruiting
A Phase II Trial of Erlotinib With Temozolomide and Concurrent Radiation Therapy Post-Operatively in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00274833]Phase 227 participants (Actual)Interventional2005-10-31Completed
A Pilot Study of Temozolomide and O-Benzylguanine for Treatment of High-Grade Glioma, Using Autologous Peripheral Blood Stem Cells Genetically Modified for Chemoprotection [NCT00253487]0 participants Interventional2005-08-31Completed
Phase 1 Trial in Newly Diagnosed High Grade Glioma With Temozolomide, Radiation, and Minocycline Followed by Adjuvant Minocycline/Temozolomide. (D-TERMINED) [NCT02272270]Phase 125 participants (Actual)Interventional2014-10-27Completed
Intensive Chemotherapy and Immunotherapy in Patients With Newly Diagnosed Primary CNS Lymphoma: A Pilot Study [NCT00416819]10 participants (Actual)Interventional2003-09-30Completed
Phase II Study of Neoadjuvant Chemoradiation for Resectable Glioblastoma (NeoGlio) [NCT04209790]Phase 230 participants (Anticipated)Interventional2020-04-01Recruiting
A Phase Ib Dose Finding Study Assessing Safety and Activity of [177Lu]Lu-DOTA-TATE in Newly Diagnosed Glioblastoma in Combination With Radiotherapy With or Without Temozolomide and in Recurrent Glioblastoma as Single Agent [NCT05109728]Phase 160 participants (Anticipated)Interventional2022-05-10Recruiting
Phase I Trial of Radiation Therapy Plus Temozolomide With MK-3475 in Patients With Newly Diagnosed Glioblastoma (GBM) [NCT02530502]Phase 14 participants (Actual)Interventional2015-09-30Terminated(stopped due to The protocol was amendment to be stopped after phase I (phase 2 removed from protocol))
Pilot Clinical Trial of Allogeneic Tumor Lysate-Pulsed Autologous Dendritic Cell Vaccination in Newly Diagnosed Glioblastoma [NCT01957956]Early Phase 121 participants (Actual)Interventional2013-11-11Completed
Temodar and Sutent as Therapy for Patients With Malignant Melanoma, a Phase I/II Study [NCT00304200]Phase 1/Phase 27 participants (Actual)Interventional2006-03-31Terminated(stopped due to Funding was inadequate to continue; Companies requested closure.)
Phase Ib Dose Finding Study Assessing Safety and Activity of [177Lu]Lu-NeoB in Combination With Radiotherapy and Temozolomide in Subjects With Newly Diagnosed Glioblastoma and as a Single Agent in Recurrent Glioblastoma [NCT05739942]Phase 148 participants (Anticipated)Interventional2024-04-01Not yet recruiting
A Phase 1/1b, Open-label Study in Patients With Recurrent Glioblastoma to Assess the Safety and Tolerability of Macitentan in Combination With Dose-dense Temozolomide [NCT01499251]Phase 175 participants (Anticipated)Interventional2012-01-31Terminated(stopped due to Results did not clearly support continuing development in recurrent GBM)
Temozolomid (One Week on/One Week Off) Versus Strahlentherapie in Der Primärtherapie Anaplastischer Astrozytome Und Glioblastome Bei älteren Patienten: Eine Randomisierte Phase III-Studie (Methvsalem) [NCT01502241]Phase 3412 participants (Actual)Interventional2005-01-31Completed
A Phase 2 Study to Evaluate the Efficacy and Safety of Temozolomide in Advanced Gastrointestinal Stromal Tumor Patients With SDH Deficiency [NCT05661643]Phase 229 participants (Anticipated)Interventional2023-06-30Not yet recruiting
Phase I Trial of Dimethyl Fumarate, Temozolomide, and Radiation Therapy in Glioblastoma Multiforme [NCT02337426]Phase 112 participants (Actual)Interventional2015-02-13Completed
A Target Validation/Phase1 Study of BGB-290 in Combination With Temozolomide in Adolescent and Young Adult IDH1/2 Newly Diagnosed and Recurrent Mutant Gliomas [NCT03749187]Phase 178 participants (Anticipated)Interventional2019-04-03Recruiting
[NCT01702610]50 participants (Actual)Interventional2008-12-31Completed
Phase Ⅱ Trial of Temozolomide Plus Concurrent Whole-Brain Radiation Followed by TNV Regimen as Adjuvant Therapy for Patients With Newly Diagnosed Primary Central Nervous System (CNS) Lymphoma (PCNSL) [NCT01735747]Phase 216 participants (Anticipated)Interventional2008-06-30Active, not recruiting
Phase II Trial of Chemotherapy With Temozolomide in Combination With Topotecan for Central Nervous System (CNS) Metastasis of Solid Tumors [NCT01736800]Phase 20 participants (Actual)Interventional2007-03-31Withdrawn(stopped due to No enrollment and PI requested study termination per IRB system.)
A Phase 1 and Randomized Phase 2 Trial of Selinexor and Temozolomide in Recurrent Glioblastoma [NCT05432804]Phase 1/Phase 297 participants (Anticipated)Interventional2023-03-20Recruiting
Phase 2 Trial of Indoximod With Chemotherapy and Radiation for Children With Progressive Brain Tumors or Newly Diagnosed DIPG [NCT04049669]Phase 2140 participants (Anticipated)Interventional2019-10-02Recruiting
GBM AGILE: Global Adaptive Trial Master Protocol: An International, Seamless Phase II/III Response Adaptive Randomization Platform Trial Designed To Evaluate Multiple Regimens In Newly Diagnosed and Recurrent GBM [NCT03970447]Phase 2/Phase 31,030 participants (Anticipated)Interventional2019-07-30Recruiting
A Phase III Trial on Adjuvant Standard Temozolomide Chemotherapy With or Without Interferon-alpha in Newly Diagnosed High-grade Gliomas [NCT01765088]Phase 3300 participants (Anticipated)Interventional2012-09-30Recruiting
A PHASE II CLINICAL TRIAL FOR THE EVALUATION OF THE EFFICACY OF DISULFIRAM/COPPER COMBINATION AS AN ADJUVANT AND CONCURRENT CHEMOTHERAPY IN THE TREATMENT OF NEWLY DIAGNOSED GLIOBLASTOMA MULTIFORM [NCT01777919]Phase 232 participants (Anticipated)Interventional2017-01-31Not yet recruiting
Oxidative Phosphorylation Targeting In Malignant Glioma Using Metformin Plus Radiotherapy Temozolomide [NCT04945148]Phase 2640 participants (Anticipated)Interventional2023-05-31Not yet recruiting
A Phase 1 Study Evaluating the Safety and Pharmacokinetics of ABT-414 for Subjects With Glioblastoma Multiforme [NCT01800695]Phase 1202 participants (Actual)Interventional2013-04-02Completed
A Phase II Trial of Low Dose Fractionated Radiation Therapy as a Chemo-Potentiator of Salvage Temozolomide for Recurrent Anaplastic Astrocytoma and Glioblastoma Multiforme [NCT01466686]Phase 231 participants (Actual)Interventional2012-09-30Completed
Clinical Trial Phase IIB Randomized, Multicenter, of Continuation or Non Continuation With 6 Cycles of Temozolomide After the First 6 Cycles of Standard First-line Treatment in Patients With Glioblastoma. [NCT02209948]Phase 2166 participants (Actual)Interventional2014-08-22Completed
Temozolomide or Dacarbazine-based Chemotherapy Plus Endostatin in Advanced Pancreatic Neuroendocrine Tumor [NCT01845675]Phase 214 participants (Actual)Interventional2013-04-30Completed
A Phase 2/3 Randomized, Open-Label Study of Toca 511, a Retroviral Replicating Vector, Combined With Toca FC Versus Standard of Care in Subjects Undergoing Planned Resection for Recurrent Glioblastoma or Anaplastic Astrocytoma [NCT02414165]Phase 2/Phase 3403 participants (Actual)Interventional2015-11-30Terminated(stopped due to Sponsor Decision)
Phase II Trial for Patients With Newly Diagnosed Glioblastoma Multiforme (GBM) Treated With Gliadel Followed by Concurrent Radiation Therapy, Temodar and Avastin, Then Followed by Avastin and Temodar Post-Radiation [NCT01186406]Phase 241 participants (Actual)Interventional2011-04-30Terminated(stopped due to Study ended early due to toxicity)
Feasibility of Autologous Stem Cell Collection and Reinfusion in Newly Diagnosed High Grade Gliomas [NCT02976441]Early Phase 10 participants (Actual)Interventional2017-01-31Withdrawn(stopped due to Physician decided not to go through with study)
Phase II Study of ABT-888 and Temozolomide in Patients With Advanced Hepatocellular Carcinoma (HCC) Progressing Following Sorafenib Treatment or Intolerant to Sorafenib [NCT01205828]Phase 216 participants (Actual)Interventional2010-08-31Terminated(stopped due to Lack of efficacy)
The Efficacy and Safety of Temozolomide in Patients With Metastatic Pheochromocytoma or Paraganglioma [NCT05858177]Phase 262 participants (Actual)Interventional2019-10-01Completed
Phase II Study of Combination Treatment of Methotrexate, Ibrutinib, and Temozolomide (MIT Regimen) in Patients With Newly Diagnosed Primary CNS Lymphoma [NCT04514393]Phase 233 participants (Anticipated)Interventional2020-10-10Recruiting
A Phase I Study of Talazoparib (BMN 673) Plus Irinotecan With or Without Temozolomide in Children With Refractory or Recurrent Solid Malignancies [NCT02392793]Phase 143 participants (Actual)Interventional2015-03-25Completed
I-ATTAC: Improved Anti-Tumor Immunotherapy Targeted Against Cytomegalovirus in Patients With Newly-Diagnosed WHO Grade IV Unmethylated Glioma [NCT03927222]Phase 26 participants (Actual)Interventional2019-09-30Terminated(stopped due to Resource shortage)
Phase I Trial of Temozolomide, Bevacizumab Plus Bortezomib for Patients With Recurrent Glioblastoma Multiforme [NCT01435395]Phase 112 participants (Actual)Interventional2011-12-31Completed
Phase I Clinical Study of Safety & Efficacy of DC Vaccine and TMZ for the Treatment of Newly-diagnosed Glioblastoma After Surgery [NCT04968366]Phase 110 participants (Anticipated)Interventional2021-07-30Recruiting
Temozolomide in the Treatment of Advanced Non-Small Cell Lung Carcinoma: Phase II Evaluation in Previously Treated and Chemo-Naive Patients [NCT00005037]Phase 251 participants (Actual)Interventional2000-01-31Completed
A Phase I/II Dose-escalation and Dose-expansion Study of Disulfiram/Copper With Concurrent Radiation Therapy and Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT02715609]Phase 1/Phase 235 participants (Actual)Interventional2016-06-15Active, not recruiting
Prospective, Open Label, Randomized Phase II Trial to Assess a Multimodal Molecular Targeted Therapy in Children, Adolescent and Young Adults With Relapsed or Refractory High-risk Neuroblastoma [NCT01467986]Phase 2130 participants (Actual)Interventional2013-08-31Completed
Phase II Study of the Combination of Cisplatin + Temozolomide in Malignant Glial Tumours in Children and Adolescents at Diagnosis or in Relapse [NCT00360945]Phase 287 participants (Anticipated)Interventional2004-04-30Active, not recruiting
Combination of Bevacizumab, Irinotecan and Temozolomide for Relapsed or Refractory Neuroblastoma: A Phase II Study [NCT01114555]Phase 234 participants (Actual)Interventional2010-04-29Completed
A Phase 1-2 Trial of Temozolomide and Hypofractionated Radiotherapy in Treatment of Supratentorial Glioblastoma Multiforme [NCT01120639]Phase 1/Phase 230 participants (Actual)Interventional2010-04-30Completed
A Phase 2, Historically Controlled Study Testing the Efficacy of TTFields (Optune®) With Adjuvant Temozolomide in High Risk WHO Grade II and III Astrocytomas (FORWARD) [NCT03906448]Phase 21 participants (Actual)Interventional2019-05-20Terminated(stopped due to The study was terminated because the Study Chair/IDE Sponsor and Novocure determined that conducting this trial was not feasible without CMS approval.)
Phase III Study of Radiation Therapy With or Without Temozolomide for Symptomatic or Progressive Low-Grade Gliomas [NCT00978458]Phase 3540 participants (Actual)Interventional2009-11-17Active, not recruiting
Phase II Treatment of Adults and Children With Progressive Low Grade Gliomas With Temodal [NCT00003466]Phase 2100 participants (Anticipated)Interventional1998-03-31Completed
06-benzylguanine (BG) and Temozolomide (TMZ) Therapy of Glioblastoma Multiforme (GBM) in Patients With MGMT Positive Tumors With Infusion of Autologous P140KMGMT+ Hematopoietic Progenitors to Protect Hematopoiesis [NCT01269424]Phase 110 participants (Actual)Interventional2011-11-22Completed
Phase II Evaluation of Temodal (Temozolomide, Schering) in Previously Treated Advanced Sarcomas [NCT00003718]Phase 225 participants (Anticipated)Interventional1998-09-30Active, not recruiting
Second Line Chemotherapy With Temozolomide in Recurrent Oligodendroglial Tumors After PCV-Chemotherapy [NCT00003304]Phase 229 participants (Anticipated)Interventional1998-04-30Completed
A Phase II Study of Temozolomide in the Treatment of Recurrent Malignant Gliomas [NCT00004204]Phase 20 participants Interventional2000-02-29Active, not recruiting
A Pilot Phase II Trial of Temozolomide in Leptomeningeal Metastases [NCT00005812]Phase 212 participants (Actual)Interventional2000-01-31Terminated(stopped due to No objective response documented, protocol terminated after 12 patients.)
A Phase II Trial of Radiation Plus Temozolomide Followed by Adjuvant Temozolomide and Poly-ICLC in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00262730]Phase 297 participants (Actual)Interventional2006-01-31Completed
FaR-RMS: An Overarching Study for Children and Adults With Frontline and Relapsed RhabdoMyoSarcoma [NCT04625907]Phase 1/Phase 21,672 participants (Anticipated)Interventional2020-09-17Recruiting
A Randomised Phase IIb Trial of Bevacizumab Added to Temozolomide ± Irinotecan for Children With Refractory/Relapsed Neuroblastoma - BEACON-Neuroblastoma Trial [NCT02308527]Phase 2225 participants (Actual)Interventional2013-07-31Active, not recruiting
A Randomized Phase 2 Study of Maintenance Temozolomide Versus Observation in Stable or Responding Stage III/IV Non-Small Cell Lung Cancer Patients [NCT00632203]Phase 253 participants (Actual)Interventional2008-03-04Terminated
Temozolomide in Metastatic Breast Cancer Patients at High Risk of Brain Recurrence: Impact on the Incidence of Brain Metastases (STOP) [NCT00638963]Phase 26 participants (Actual)Interventional2008-10-02Terminated(stopped due to Terminated due to poor accrual)
Safety of TAS-102 in Combination With Temozolomide for Metastatic Pancreatic Neuroendocrine Tumors [NCT02943733]Phase 118 participants (Actual)Interventional2017-08-22Terminated(stopped due to closed per sponsor request, for slow enrollment)
A Phase 2 Trial for Patients With Newly Diagnosed High Grade Glioma Treated With Concurrent Radiation Therapy, Temozolomide, and BMX-001 [NCT02655601]Phase 2160 participants (Actual)Interventional2018-09-25Active, not recruiting
Treatment of Adults With Newly Diagnosed Glioblastoma With Partial Brain Radiation Therapy Plus Temozolomide and Chloroquine Followed by Tumor Treating Fields Plus Temozolomide and Chloroquine -- A Pilot Study [NCT04397679]Phase 110 participants (Anticipated)Interventional2021-08-12Recruiting
Phase 1 Study of 9-ING-41, a Glycogen Synthase Kinase 3 Beta (GSK 3β) Inhibitor, as a Single Agent or With Irinotecan, Irinotecan Plus Temozolomide, or With Cyclophosphamide Plus Topotecan in Pediatric Patients With Refractory Malignancies. [NCT04239092]Phase 168 participants (Anticipated)Interventional2020-06-05Active, not recruiting
A Phase I/II Clinical Trial of Autologous CMV-Specific Cytotoxic T Cells for GBM Patients [NCT02661282]Phase 1/Phase 265 participants (Actual)Interventional2016-06-01Completed
A Phase III Trial of Marizomib in Combination With Standard Temozolomide-based Radiochemotherapy Versus Standard Temozolomide-based Radiochemotherapy Alone in Patients With Newly Diagnosed Glioblastoma [NCT03345095]Phase 3749 participants (Actual)Interventional2018-07-26Active, not recruiting
A Phase I/II Study of High-dose L-methylfolate in With Combination Temozolomide and Bevacizumab in Recurrent High Grade Glioma. [NCT01891747]Phase 114 participants (Actual)Interventional2013-07-31Completed
Phase II Study of Ipilimumab Plus Temozolomide in Patients With Metastatic Melanoma [NCT01119508]Phase 264 participants (Actual)Interventional2010-05-31Completed
A Phase II Single Arm Trial Evaluating the Efficacy and Safety of Temozolomide for Second-Line Treatment of Neuroendocrine Carcinomas Progressing After First-Line Platinum- Based Therapy [NCT04122911]Phase 225 participants (Actual)Interventional2017-01-29Completed
A Phase I/II Study of Local Field Irradiation and Temozolomide Followed by Continuous Infusion Plerixafor as an Upfront Therapy for Newly Diagnosed Glioblastoma GBM [NCT01977677]Phase 1/Phase 230 participants (Actual)Interventional2014-11-30Completed
Phase I Trial of BMN 673 and Selected Cytotoxics in Patients With Advanced Solid Tumors [NCT02049593]Phase 144 participants (Actual)Interventional2014-06-12Completed
Phase I/II Study of the Poly (ADP-ribose) Polymerase-1 (PARP-1) Inhibitor BSI-201 in Patients With Newly Diagnosed Malignant Glioma [NCT00687765]Phase 1/Phase 2126 participants (Actual)Interventional2008-07-31Completed
A Randomized Phase II Trial of Concurrent Temozolomide and Radiotherapy Followed by Dose Dense Versus Metronomic Temozolomide and Maintenance Cis-Retinoic Acid for Patients With Newly Diagnosed Glioblastoma and Other Malignant Gliomas [NCT00200161]Phase 2127 participants (Actual)Interventional2005-08-09Completed
Phase I/II Study of Olaparib and Temozolomide in Patients With Recurrent Small Cell Lung Cancer Following Failure of Prior Chemotherapy [NCT02446704]Phase 1/Phase 266 participants (Actual)Interventional2015-10-13Active, not recruiting
Randomized, Phase II Study of Pembrolizumab (MK-3475) Versus Chemotherapy in Patients With Advanced Melanoma (KEYNOTE 002) [NCT01704287]Phase 2540 participants (Actual)Interventional2012-11-20Completed
Preoperative Chemoradiation With Capecitabine Plus Temozolomide in Patients With Locally Advanced Resectable Rectal Cancer: Phase I Study [NCT01781403]Phase 122 participants (Actual)Interventional2013-05-10Completed
A Phase I Trial of Neoadjuvant Chemoimmunotherapy in Recurrent Glioblastoma [NCT05700955]Phase 130 participants (Anticipated)Interventional2022-11-01Recruiting
An Italian Multicenter Phase II Trial of Metronomic Temozolomide in Unfit Patients With Advanced Neuroendocrine Neoplasms (NENs): MeTe Study [NCT05554003]Phase 246 participants (Anticipated)Interventional2022-01-14Recruiting
A Randomized Phase III Study of Re-Irradiation in Recurrent Glioblastoma [NCT01830101]Phase 30 participants (Actual)Interventional2014-02-28Withdrawn(stopped due to This study will not be proceeding due to lack of funding from anticipated sources)
Efficacy and Safety of Lanreotide ATG 120 mg in Combination With Temozolomide in Subjects With Progressive Well Differentiated Thoracic Neuroendocrine Tumors. A Phase II, Multicentre, Single Arm, Open-label Trial. [NCT02698410]Phase 240 participants (Actual)Interventional2016-07-31Completed
Phase I/II Study of Bevacizumab Plus Daily Temozolomide and Vorinostat for Recurrent Malignant Glioma Patients [NCT00939991]Phase 1/Phase 248 participants (Actual)Interventional2009-10-31Completed
Adjuvant Dendritic Cell Immunotherapy Complementing Conventional Therapy for Pediatric Patients With High-grade Glioma and Diffuse Intrinsic Pontine Glioma [NCT04911621]Phase 1/Phase 210 participants (Anticipated)Interventional2021-09-10Active, not recruiting
A Safety Run-in/Randomized Phase II Trial of EMD 121974 in Conjunction With Concomitant and Adjuvant Temozolomide With Radiation Therapy in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00085254]Phase 1/Phase 2112 participants (Actual)Interventional2005-04-30Completed
Temozolomide, Bevacizumab, Lithium and Radiation Treatment for Newly Diagnosed High Grade Glioma: A Phase II Study [NCT01105702]Phase 228 participants (Actual)Interventional2010-05-31Terminated(stopped due to Due to slow accrual)
A Pilot Phase II Trial of Hypofractionated Intensity-Modulated Radiation Therapy (Hypo-IMRT) Combining With Temozolomide (TMZ) and Bevacizumab for Patients With Newly Diagnosed Glioblastoma Multiforme (GBM) [NCT01209442]Phase 230 participants (Actual)Interventional2010-09-16Completed
Pamiparib and Low Dose Temozolomide In Patients With Platinum Sensitive Biliary Tract Cancer [NCT04796454]Phase 20 participants (Actual)Interventional2022-05-31Withdrawn(stopped due to Company decision)
Patients With IDH1 Positive Recurrent Grade II Glioma Enrolled in a Safety and Immunogenicity Study of Tumor-Specific Peptide Vaccine [NCT02193347]Phase 124 participants (Actual)Interventional2016-01-28Completed
A Phase III Study of Postoperative Early Temozolomide Treatment Plus STUPP Regimen for Newly Diagnosed GBM Multiforme [NCT05600491]Phase 378 participants (Anticipated)Interventional2015-11-08Recruiting
Multicenter Phase II Study of Preoperative Chemoradiotherapy With CApecitabine Plus Temozolomide in Patients With MGMT Silenced and Microsatellite Stable Locally Advanced RecTal Cancer: the CATARTIC Trial [NCT05136326]Phase 221 participants (Anticipated)Interventional2021-12-01Recruiting
A Phase II Study of TEMOZOLOMIDE in Advanced Non-Small Cell Lung Cancer With and Without Brain Metastases [NCT00003062]Phase 270 participants (Anticipated)Interventional1997-07-31Completed
Phase I/II Study of MLN8237 in Combination With Irinotecan and Temozolomide for Patients With Relapsed or Refractory Neuroblastoma [NCT01601535]Phase 1/Phase 254 participants (Actual)Interventional2012-05-31Completed
A Phase II Trial of Temozolomide and BCNU for Anaplastic Gliomas [NCT00003176]Phase 282 participants Interventional1998-03-25Completed
Phase II Treatment of Adults With Newly Diagnosed Primary Malignant Glioblastoma Multiforme With Temodal [NCT00003464]Phase 250 participants (Anticipated)Interventional1997-09-30Completed
Phase II Trial of Temodal 4 Hourly in Progressive Breast Cancer [NCT00005054]Phase 20 participants Interventional1998-05-31Active, not recruiting
A Randomized Phase II Trial of ATN-224 in Combination With Temozolomide or Temozolomide Followed by ATN-224 in Patients With Advanced Melanoma [NCT00383851]Phase 260 participants (Anticipated)Interventional2006-09-30Active, not recruiting
A Phase I/II Study of Temozolamide and Thalidomide in the Treatment of Advanced Melanoma [NCT00005815]Phase 1/Phase 20 participants Interventional1999-12-31Completed
STRONG Trial - Stem Cell Radiotherapy (ScRT) and Temozolomide for Newly Diagnosed High-grade Glioma (HGG): A Prospective, Phase I/II Trial [NCT02039778]4 participants (Actual)Interventional2013-12-31Terminated(stopped due to Poor Accural)
NYU 05-40 PBMTC ONC-032P:High Dose Temozolomide,Thiotepa and Carboplatin With Autologous Stem Cell Rescue (ASCR) Followed by Continuation Therapy With 13-cis-retinoic Acid in Patients With Recurrent/Refractory Malignant Brain Tumors [NCT00528437]Phase 246 participants (Actual)Interventional2005-10-31Completed
Phase II Study of Dose Escalated, Targeted Radiation Therapy Using 3D Magnetic Resonance Spectroscopy Imaging (MRSI) in Newly Diagnosed Glioblastoma [NCT02394665]Phase 21 participants (Actual)Interventional2015-03-31Terminated(stopped due to Lack of Accrual/Enrollment)
Phase II Study of High Dose Radiotherapy and Concurrent Temozolomide Using Biologically-Based Target Volume Definition in Patients With Newly Diagnosed Glioblastoma [NCT02805179]Phase 226 participants (Actual)Interventional2016-09-22Completed
International Cooperative Phase III Trial of the HIT-HGG Study Group for the Treatment of High Grade Glioma, Diffuse Intrinsic Pontine Glioma, and Gliomatosis Cerebri in Children and Adolescents < 18 Years.(HIT-HGG-2013) [NCT03243461]Phase 3167 participants (Anticipated)Interventional2018-07-17Recruiting
Phase Ib Study of Fluzoparib in Combination With Camrelizumab and Temozolomide in Advanced Melanoma With Homologous Recombination (HR) Mutation ,a Single-center Open-label Exploratory Clinical Trial [NCT05983237]Phase 1/Phase 250 participants (Anticipated)Interventional2023-08-31Not yet recruiting
A Randomized Surgical Window of Opportunity Study With Dose Escalation to Evaluate Whether Oral Fluoxetine Can Induce Cytotoxic Lysosomal Stress and Enhance Temozolomide Efficacy in Clinical Glioma [NCT05634707]Early Phase 130 participants (Anticipated)Interventional2023-08-05Recruiting
A Phase II Study of Cabozantinib and Temozolomide in Patients With Unresectable or Metastatic Leiomyosarcoma and Other Soft Tissue Sarcomas [NCT04200443]Phase 272 participants (Actual)Interventional2020-01-14Active, not recruiting
A Phase I/II Trial of Maximal Resection, Local Radiation Boost With Concomitant Temozolomide, Followed by External Radiation Therapy With Concomitant Temozolomide for the Treatment of Newly Diagnosed Glioblastoma Multiforme [NCT00376103]Phase 1/Phase 23 participants (Actual)Interventional2006-08-31Terminated(stopped due to Loss of funding from sponsor)
Phase I Study of Neoadjuvant GMCI Plus Immune Checkpoint Inhibitor Combined With Standard of Care for Newly Diagnosed High-Grade Gliomas [NCT03576612]Phase 136 participants (Anticipated)Interventional2018-02-27Active, not recruiting
A Phase II Study of Temozolomide and Radiation Therapy in Patients With Brain Metastasis From Non-small Cell Lung Cancer (NSCLC) [NCT00080938]Phase 226 participants (Actual)Interventional2005-12-20Completed
Phase II Trial of Continuous Dose Temozolomide in Patients With Newly Diagnosed Anaplastic Oligodendroglioma and Mixed Oligoastrocytoma [NCT00362570]Phase 21 participants (Actual)Interventional2005-05-31Completed
A Multi-Center, Randomized, Double-Blind Phase II Study Comparing ABT-888, a PARP Inhibitor, Versus Placebo With Temozolomide in Patients With Relapsed Sensitive or Refractory Small Cell Lung Cancer [NCT01638546]Phase 297 participants (Actual)Interventional2012-06-30Completed
BrUOG 329: Onivyde (Nanoliposomal Irinotecan) and Metronomic Temozolomide for Patients With Recurrent Glioblastoma: A Phase IB/IIA Brown University Oncology Research Group Study [NCT03119064]Phase 1/Phase 212 participants (Actual)Interventional2017-11-30Terminated(stopped due to lack of response to study therapy)
Dose Intensive Chemotherapy for Children Less Than Ten Years of Age Newly-Diagnosed With Malignant Brain Tumors: A Pilot Study of Two Alternative Intensive Induction Chemotherapy Regimens, Followed by Consolidation With Myeloablative Chemotherapy (Thiotep [NCT00003273]Phase 20 participants (Actual)Interventional1997-11-30Withdrawn
Phase II Treatment of Adults With Newly Diagnosed, Progressive or Recurrent Primary Malignant Anaplastic Oligodendroglioma With Temodal [NCT00003465]Phase 260 participants (Anticipated)Interventional1998-03-31Completed
Mutant MGMT Gene Transfer Into Human Hematopoietic Progenitors to Protect Hematopoiesis During O6-Benzylguanine (BG, NSC 637037) and Carmustine Followed by Temozolomide Therapy of Advanced Solid Tumors [NCT00003567]Phase 18 participants (Actual)Interventional1999-05-31Terminated(stopped due to slow accrual)
A Phase II Study of Temozolomide (SCH 52365, Temodal(R)) for the Treatment of Recurrent Malignant Glioma [NCT00004113]Phase 20 participants Interventional1999-06-30Completed
Phase I (Tumour Site Specific) Study of Carboplatin and Temozolomide in Patients With Advanced Melanoma [NCT00003747]Phase 130 participants (Anticipated)Interventional1998-10-31Active, not recruiting
A Phase I Study of Extended Low Dose Temozolomide (SCH 52365, Temodar (R)) and Carmustine (BCNU) in the Treatment of Malignant Gliomas After Radiation Therapy [NCT00005637]Phase 10 participants Interventional1999-12-31Completed
A Phase II Trial of Intensive Chemotherapy and Autotransplantation for Patients With Newly Diagnosed Anaplastic Oligodendroglioma [NCT00588523]Phase 260 participants (Actual)Interventional2002-09-30Completed
Phase II Study of Dose Intensive Temozolomide in Elderly Adults With Newly Diagnosed Glioblastoma [NCT00365222]Phase 21 participants (Actual)Interventional2006-07-31Terminated(stopped due to IRB Study Closure)
Capecitabine ON Temozolomide Radionuclide Therapy Octreotate Lutetium-177 NeuroEndocrine Tumours Study [NCT02358356]Phase 275 participants (Actual)Interventional2015-11-30Completed
The Efficacy and Safety of Neoadjuvant Toripalimab Combined With Temozolomide in Resectable Stage III Melanoma: A Prospective, Single-center, Phase 2 Clinical Trial [NCT05827770]Phase 220 participants (Anticipated)Interventional2023-05-01Not yet recruiting
Pilot Study to Evaluate the Safety, Tolerability and Effectiveness of Intranasal Administration of Temozolomide in Patients With Glioblastoma (Phase I) [NCT04091503]Phase 130 participants (Actual)Interventional2019-12-20Completed
A Phase I/II Study of ABT-888, An Oral Poly(ADP-ribose) Polymerase Inhibitor, and Concurrent Radiation Therapy, Followed by ABT-888 and Temozolomide, in Children With Newly Diagnosed Diffuse Pontine Gliomas (DIPG) [NCT01514201]Phase 1/Phase 266 participants (Actual)Interventional2012-02-01Completed
A Phase I Trial for the Addition of Chloroquine, an Autophagy Inhibitor, to Concurrent Chemoradiation for Newly Diagnosed Glioblastoma [NCT02378532]Phase 113 participants (Actual)Interventional2016-08-31Completed
A Phase II, Multi-Cohort Trial of Combination Nivolumab and Temozolomide in Recurrent/Refractory Small-Cell Lung Cancer and Advanced Neuroendocrine Tumors [NCT03728361]Phase 255 participants (Actual)Interventional2018-12-31Active, not recruiting
Phase I/II Multicenter Trial of Intra-Arterial Carboplatin and Oral Temozolomide for the Treatment of Recurrent and Symptomatic Residual Brain Metastases. [NCT00362817]Phase 1/Phase 217 participants (Actual)Interventional2004-10-31Completed
SPARE-Scalp Preservation and Radiation Plus Alternating Electric Tumor Treatment Field (NovoTTF, Optune) for Patients With Glioblastoma: A Pilot Study [NCT03477110]Early Phase 130 participants (Actual)Interventional2018-05-04Active, not recruiting
A Phase II Study of the Efficacy of Hypofractionated Radiation Therapy With Bevacizumab and Temozolomide Followed by Maintenance Temozolomide and Bevacizumab for Recurrent High-Grade Gliomas [NCT01478321]Phase 254 participants (Actual)Interventional2011-12-14Terminated(stopped due to Slow accrual to some cohorts)
Temozolomide Plus Vorinostat in Patients With Relapse/Refractory Acute Myeloid Leukemia (AML) [NCT01550224]Phase 223 participants (Actual)Interventional2013-05-01Completed
A Phase II Randomized,Controlled,Open Label,Multicentre Study of Tegafur Combined With Temozolomide Versus Tegafur Combined With Temozolomide and Thalidomide in Subjects With Advanced Pancreatic Neuroendocrine Tumor [NCT03204019]Phase 260 participants (Anticipated)Interventional2016-10-31Recruiting
Phase II Trial of Temozolomide in Patients Affected by Relapsed Sensitive or Refractory Small Cell Lung Cancer With MGMT Methylation (TeRes). GOPAV03 [NCT02477813]Phase 26 participants (Actual)Interventional2015-01-31Terminated(stopped due to Lack of eligible patient recruitment)
Efficacy and Safety of Low Dose Temozolomide Plus Metformin as Combination Chemotherapy Compared With Low Dose Temozolomide Plus Placebo in Patient With Recurrent or Refractory Glioblastoma [NCT03243851]Phase 281 participants (Actual)Interventional2016-11-21Completed
A Prospective Multicenter Randomized Controlled Clinical Trial of Neoadjuvant Temozolomide Combined With Simultaneous Increased Intensity-modulated Radiotherapy in the Treatment of Glioblastoma [NCT04829097]Phase 380 participants (Anticipated)Interventional2020-11-01Recruiting
Open-label, Randomized, Multicenter, Phase II Trial to Compare Efficacy of CAPTEM Versus FOLFIRI as Second Line in Patients Progressed on or After First-line Oxaliplatin Chemo for Advanced, MGMT Methylated, RAS Mutated Colorectal Cancer [NCT02414009]Phase 282 participants (Actual)Interventional2014-09-30Completed
MT-R Followed by Autologous Stem Cells Transplantation in Newly-diagnosed Primary Central Nervous System Lymphoma [NCT02399189]Phase 239 participants (Anticipated)Interventional2014-05-31Recruiting
A Phase I Lead-In to a 2x2x2 Factorial Trial of Temozolomide, Memantine, Mefloquine, and Metformin as Post-Radiation Adjuvant Therapy of Glioblastoma Multiforme [NCT01430351]Phase 1144 participants (Anticipated)Interventional2011-09-14Active, not recruiting
Phase 1 Study of Pazopanib in Combination With Irinotecan and Temozolomide (PAZIT) for Children and Young Adults With Relapsed or Refractory Sarcoma [NCT03139331]Phase 116 participants (Actual)Interventional2017-06-06Completed
Ultra-low Dose Bevacizumab Plus Temozolomide for Recurrent High-grade Gliomas [NCT02416999]30 participants (Anticipated)Interventional2015-05-31Not yet recruiting
A Prospective, Single-center Clinical Study to Explore the Efficacy and Safety of SHR-1701 Combined With Temozolomide in the Treatment of Advanced Melanoma [NCT05106023]Phase 231 participants (Anticipated)Interventional2022-01-21Recruiting
A Phase 2, Open-label Study of ABI-009 (Nab-Rapamycin) in Patients With Recurrent High-grade Glioma and Patients With Newly Diagnosed Glioblastoma [NCT03463265]Phase 262 participants (Actual)Interventional2018-08-01Completed
Evaluation of Overcoming Limited Migration and Enhancing Cytomegalovirus-specific Dendritic Cell Vaccines With Adjuvant TEtanus Pre-conditioning in Patients With Newly-diagnosed Glioblastoma [NCT02366728]Phase 264 participants (Actual)Interventional2015-10-12Completed
Phase 1B Trial of ADI-PEG 20 Plus Radiotherapy and Temozolomide in Subjects With Newly Diagnosed Glioblastoma Multiforme [NCT04587830]Phase 132 participants (Anticipated)Interventional2020-09-14Recruiting
Randomized Phase II Trial of Postoperative Adjuvant Capecitabine and Temozolomide Versus Observation in High-Risk Pancreatic Neuroendocrine Tumors [NCT05040360]Phase 2141 participants (Anticipated)Interventional2022-05-05Recruiting
Pilot Pharmacokinetic Study of VAL-413 (Orotecan®) in Patients With Recurrent Pediatric Solid Tumors [NCT04337177]Phase 120 participants (Anticipated)Interventional2021-10-25Recruiting
Randomized Phase II Study of Cisplatin and Etoposide Versus Temozolomide and Capecitabine in Patients With Advanced G3 Non-small Cell Gastroenteropancreatic Neuroendocrine Carcinomas [NCT02595424]Phase 2126 participants (Anticipated)Interventional2016-04-06Recruiting
Image-derived Prediction of Response to Chemo-radiation in Patients With Glioblastoma [NCT02329795]16 participants (Actual)Observational2014-10-31Terminated(stopped due to Slow accrual)
Phase I Study of Escalated Pharmacologic Dose, of Oral Folinic Acid in Combination With Temozolomide, According to Stupp R. Regimen, in Patients With Operated Grade-IV Astocytoma and a Non-methylated Gene Status of MGMT. [NCT01700569]Phase 124 participants (Actual)Interventional2013-01-31Terminated(stopped due to changing the standard of care)
A Phase II Trial of Continuous Low-Dose Temozolomide for Patients With Recurrent Malignant Glioma [NCT00498927]Phase 247 participants (Actual)Interventional2007-06-30Completed
A Phase II Study of Bevacizumab in Combination With Metronomic Temozolomide for Recurrent Malignant Glioma [NCT00501891]Phase 232 participants (Actual)Interventional2007-07-31Completed
Biochemotherapy With Temozolomide, Velban, Cisplatin, Interleukin-2, Interferon-alpha and Thalidomide for Metastatic Melanoma With Optional Intrathecal Interleukin-2 Treatment [NCT00505635]Phase 25 participants (Actual)Interventional2007-03-31Terminated(stopped due to Slow accrual)
A Phase II Study of Bevacizumab Plus Temodar and Tarceva After Radiation Therapy and Temodar in Patients With Newly Diagnosed Glioblastoma or Gliosarcoma Who Are Stable Following Radiation [NCT00525525]Phase 274 participants (Actual)Interventional2007-09-30Completed
A Longitudinal Assessment of Tumor Evolution in Patients With Brain Cancer [NCT03425292]Phase 149 participants (Actual)Interventional2018-03-01Completed
PPX and Concurrent Radiation for Newly Diagnosed Glioblastoma Without MGMT Methylation: A Randomized Phase II Study [NCT01402063]Phase 263 participants (Actual)Interventional2011-09-30Completed
A Phase 2 Study of Capecitabine, Temozolomide, and Bevacizumab for Metastatic or Unresectable Pancreatic Neuroendocrine Tumors [NCT01525082]Phase 220 participants (Actual)Interventional2012-12-31Completed
A Phase II Study of Rituximab and Temozolomide in Recurrent Primary CNS Lymphoma [NCT00248534]Phase 216 participants (Actual)Interventional2005-09-30Terminated(stopped due to slow accrual/lack of resources/low priority due to combining 2 consortia)
PHASE 2, Multi-center, Single Arm Investigation of HSPPC-96 Vaccine With Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT00905060]Phase 270 participants (Actual)Interventional2009-06-29Completed
A Phase 1 Study of Maprotiline in Combination With Tamoxifen and Temozolomide for Recurrent Glioblastoma [NCT04200066]Phase 10 participants (Actual)Interventional2022-06-01Withdrawn(stopped due to One of the drugs for the study is not commercially available.)
Efficacy and Safety of Salvage Treatment With Dose-dense TMZ Plus CDDP in the Patients With Recurrent Malignant Gliomas: a Multicentre,Prospective Clinical Study [NCT01670890]Phase 2120 participants (Anticipated)Interventional2012-08-31Recruiting
A Phase II Study of Intrathecal and Systemic Chemotherapy With Radiation Therapy for Children With Central Nervous System Atypical Teratoid/Rhabdoid Tumor (AT/RT) Tumor [NCT00084838]Phase 225 participants (Actual)Interventional2003-02-28Completed
A Phase II Randomized Trial of Irinotecan/Temozolomide With Temsirolimus (NSC# 683864) or Chimeric 14.18 Antibody (Ch14.18) (NSC# 764038) in Children With Refractory, Relapsed or Progressive Neuroblastoma [NCT01767194]Phase 273 participants (Actual)Interventional2013-02-12Completed
Camrelizumab Plus Apatinib and Temozolomide as First Line Therapy in Advanced Acral Melanoma:a Single-center, Exploratory Clinical Study [NCT04397770]Phase 240 participants (Anticipated)Interventional2020-05-31Not yet recruiting
Treatment of High-grade Gliomas Using Escalating Doses of Hypofractionated Simultaneous Integrated Boost-intensity Modulated Radiation Therapy in Combination With Temozolomide - a Modified Phase I Clinical Trial [NCT03082846]16 participants (Actual)Interventional2014-01-01Completed
A Randomized, Phase 2 Study of the Efficacy and Tolerability of Veliparib in Combination With Temozolomide or Veliparib in Combination With Carboplatin and Paclitaxel Versus Placebo Plus Carboplatin and Paclitaxel in Subjects With BRCA1 or BRCA2 Mutation [NCT01506609]Phase 2294 participants (Actual)Interventional2012-01-23Completed
Phase I/II Evaluation Temozolomide and Farnesyl Transferase Inhibitor ZARNESTRA (R115777) for the Treatment of Recurrent and Progressive Glioblastoma Multiforme [NCT00050986]Phase 1/Phase 255 participants (Actual)Interventional2002-12-31Completed
A Single Arm Clinical Phase Ⅱ Study of Apatinib Combined With Temozolomide and Etoposide Capsules in the Treatment of Recurrent Medulloblastoma in Children [NCT04501718]Phase 244 participants (Anticipated)Interventional2020-10-28Recruiting
A Multi-Center Pilot/Phase I and Phase II Clinical Trial of NG101m Adjuvant Therapy in Newly Diagnosed Glioblastoma Patients [NCT04373785]Phase 1/Phase 252 participants (Anticipated)Interventional2024-01-01Not yet recruiting
Open-label Prospective Study of Recombinant Human Endostatin Combined With Cytotoxic Chemotherapy Regimen in the Treatment of Recurrent Gliomas [NCT04267978]Phase 2109 participants (Anticipated)Interventional2020-02-13Recruiting
Randomized Phase 2b Study of Safety And Efficacy Of TVI-Brain-1 Combined With Conformal Radiotherapy And Temozolomide Vs Standard Therapy In Newly Diagnosed MGMT Negative Glioblastoma Multiforme (GBM) [NCT05685004]Phase 2/Phase 396 participants (Anticipated)Interventional2023-09-15Recruiting
A Phase Ib/II Clinical Study of BBI608 in Combination With Temozolomide for Adult Patients With Recurrent or Progressed Glioblastoma [NCT02315534]Phase 1/Phase 234 participants (Actual)Interventional2015-03-31Completed
A Phase I/II Study of the Combination of Temozolomide and Pazopanib in Advanced Pancreatic Neuroendocrine Tumors (PNET) [NCT01465659]Phase 1/Phase 229 participants (Actual)Interventional2011-12-12Completed
Capecitabine and Temozolomide for Treatment of Recurrent Pituitary Adenomas [NCT03930771]Phase 21 participants (Actual)Interventional2019-05-21Terminated(stopped due to the study had a low accrual rate)
A Phase 1 Trial of RRx-001 in Combination With Irinotecan and Temozolomide for Pediatric Patients With Recurrent or Progressive Malignant Solid and Central Nervous System Tumors [NCT04525014]Phase 124 participants (Anticipated)Interventional2023-01-26Active, not recruiting
A Phase Ib/II, Open-Label, Multi-Center, Dose Escalation and Expansion Study to Evaluate Safety, Tolerability, Pharmacokinetics, and Anti-tumor Activity of IMP4297 in Combination With Temozolomide in Patients With Advanced Solid Tumors and Small Cell Lung [NCT04434482]Phase 1/Phase 2113 participants (Anticipated)Interventional2020-08-07Recruiting
Safety Run-In and Phase II Trial of M7824 and Topotecan or Temozolomide in Relapsed Small Cell Cancers [NCT03554473]Phase 1/Phase 267 participants (Anticipated)Interventional2018-09-11Recruiting
Phase Ib, Open-label, Multicenter, Intrapatient Dose-escalation Clinical Trial to Assess the Safety Profile of the TN-TC11G (THC+CBD) Combination With Temozolomide and Radiotherapy in Patients With Newly-diagnosed Glioblastoma [NCT03529448]Phase 1/Phase 230 participants (Anticipated)Interventional2023-08-18Recruiting
Pembrolizumab in MMR-Proficient Metastatic Colorectal Cancer Pharmacologically Primed to Trigger Dynamic Hypermutation Status [NCT03519412]Phase 2102 participants (Anticipated)Interventional2019-01-23Active, not recruiting
Molecular Profiling-Based Assignment of Cancer Therapy for Patients With Advanced Solid Tumors [NCT01827384]Phase 2208 participants (Actual)Interventional2014-01-07Completed
An Open Label Phase 1b/2 Study of Orally Administered PLX3397 in Combination With Radiation Therapy and Temozolomide in Patients With Newly Diagnosed Glioblastoma [NCT01790503]Phase 1/Phase 265 participants (Actual)Interventional2013-07-18Completed
Quantitative Assessment of the Early and Late Effects of Radiation and Chemotherapy on Glioblastoma Using Multiple MRI Techniques [NCT00756106]15 participants (Actual)Interventional2008-07-31Terminated(stopped due to Funding ended)
Phase II Study of Bevacizumab and Temozolomide in Elderly Patients With Newly-Diagnosed Glioblastoma [NCT01149850]Phase 250 participants (Actual)Interventional2010-04-28Active, not recruiting
Open Label Randomized Phase II/III Trial of Dendritic Cell Immunotherapy Against Cancer Stem Cells in Glioblastoma Patients Receiving Standard Therapy (DEN-STEM) [NCT03548571]Phase 2/Phase 360 participants (Anticipated)Interventional2018-04-26Active, not recruiting
Phase II Randomized Study: Whole Brain Radiotherapy and Concomitant Temozolomide, Compared With Whole Brain Radiotherapy for Brain Metastases Treatment [NCT01015534]Phase 255 participants (Actual)Interventional2006-01-31Completed
Feasibility Pilot Study of OKN-007 in Combination With Adjuvant Temozolomide Chemoradiotherapy in Patients With Newly Diagnosed Glioblastoma [NCT03587038]Phase 127 participants (Actual)Interventional2018-09-03Active, not recruiting
A Pilot Study of SGT-53 in Conjunction With Irradiation and Chemotherapy in Children With Recurrent or Progressive CNS Malignancies [NCT03554707]Early Phase 16 participants (Anticipated)Interventional2022-06-30Not yet recruiting
A Prospective, Randomised, Controlled, Open-label, Multicentre Study to Evaluate Efficacy, Safety and Patient-Reported Outcomes of Peptide Receptor Radionuclide Therapy (PRRT) With 177Lu-Edotreotide Compared to Best Standard of Care in Patients With Well- [NCT04919226]Phase 3202 participants (Anticipated)Interventional2021-12-21Recruiting
An Open-Label, Multicenter, First-in-Human, Phase 1 Dose-Escalation and Multicohort Expansion Study of INBRX-109 in Subjects With Locally Advanced or Metastatic Solid Tumors Including Sarcomas [NCT03715933]Phase 1240 participants (Anticipated)Interventional2018-10-10Recruiting
Phase II Study of Temozolomide in Unresectable or Metastatic Gastrointestinal Stromal Tumors (GIST) [NCT00005597]Phase 225 participants (Actual)Interventional2000-04-30Completed
A Phase I Study of PRK787/ZK 222584 in Combination With Daily Temozolomide and Radiation in Patients With Newly Diagnosed Glioblastoma [NCT00385853]Phase 119 participants (Actual)Interventional2006-09-30Completed
First Line Chemotherapy With Temozolomide in Recurrent Oligodendroglial Tumors, a Phase II Trial [NCT00003731]Phase 239 participants (Actual)Interventional1998-12-31Completed
A Prospective, Multi-Institutional Phase II Trial Evaluating Temozolomide vs. Temozolomide and Olaparib for Advanced Pheochromocytoma and Paraganglioma [NCT04394858]Phase 276 participants (Anticipated)Interventional2021-03-17Recruiting
A Randomized, Open-label, Multicentric, Two-arm Pivotal Trial of SonoCloud-9 Combined With Carboplatin (CBDCA) vs Standard of Care Lomustine (CCNU) or Temozolomide (TMZ) in Patients Undergoing Planned Resection for First Recurrence Glioblastoma. [NCT05902169]Phase 3560 participants (Anticipated)Interventional2023-11-05Not yet recruiting
Phase I/II Study of RAD001 in Combination With Temozolomide in Patients With Advanced Pancreatic Neuroendocrine Tumors [NCT00576680]Phase 1/Phase 243 participants (Actual)Interventional2008-05-31Completed
Phase II Study of Bevacizumab Plus Either Temozolomide or Etoposide for (GBM) Patients Who Have Failed Bevacizumab Plus Irinotecan [NCT00613028]Phase 223 participants (Actual)Interventional2008-04-30Completed
A Phase 1 Multicenter, Dose-escalation Study of LY573636-sodium in Combination With 1) Gemcitabine HCl or 2) Docetaxel or 3) Temozolomide or 4) Cisplatin, or 5) Erlotinib in Patients With Advanced Solid Tumors [NCT01284335]Phase 1234 participants (Actual)Interventional2008-07-31Terminated(stopped due to Study was terminated due to the termination of tasisulam development.)
A Phase II Study of Bevacizumab, Temozolomide and Hypofractionated Radiotherapy for Patients With Newly Diagnosed Malignant Glioma [NCT00782756]Phase 240 participants (Actual)Interventional2008-10-28Completed
Using Proton Magnetic Resonance Spectroscopy (MRS) to Predict Response of Vorinostat Treatment in Glioblastoma [NCT01342757]12 participants (Actual)Interventional2010-12-31Completed
A Phase II Open-Label, Randomized, Multi-Centre Comparative Study Of Bevacizumab-Based Therapy In Paediatric Patients With Newly Diagnosed Supratentorial, Infratentorial Cerebellar, or Peduncular High-Grade Glioma [NCT01390948]Phase 2124 participants (Actual)Interventional2011-10-18Completed
Everolimus and Temozolomide as 1-line Treatment in Advanced Gastroenteropancreatic Neuroendocrine Carcinoma (G3) With a Ki67 of 20-55% [NCT02248012]Phase 238 participants (Actual)Interventional2014-12-31Completed
A Multicenter, Open-Label, Randomized Study of NovoTTF-200A Alone and Combined With Temozolomide in Patients With Low-Grade Gliomas [NCT02507232]Early Phase 12 participants (Actual)Interventional2017-04-17Terminated(stopped due to low enrollment)
A Phase I Trial of Indoximod and Temozolomide-Based Therapy for Children With Progressive Primary Brain Tumors [NCT02502708]Phase 181 participants (Actual)Interventional2015-10-31Completed
An Open, Single-center Clinical Study of Surufatinib Combined With Temozolomide and S-1 in the First-line Treatment of Advanced Neuroendocrine Tumors [NCT06038461]Phase 1/Phase 240 participants (Anticipated)Interventional2023-09-15Not yet recruiting
A Phase II Study Analyzing Pulsed Reduced Dose Radiotherapy in Upfront Glioblastoma (PRORADGLIO Study) [NCT04747145]Phase 238 participants (Anticipated)Interventional2021-06-03Recruiting
Phase 1b/2a Single-center, Open-label, Dose Escalation Study to Evaluate the Safety, Tolerability, and Efficacy of MN-166 (Ibudilast) and Temozolomide Combination Treatment in Patients With Newly Diagnosed or Recurrent Glioblastoma [NCT03782415]Phase 1/Phase 250 participants (Anticipated)Interventional2018-12-29Active, not recruiting
Phase 2, Single Arm, Historically Controlled Study Testing The Safety and Efficacy of Adjuvant Temozolomide Plus TTFields (Optune®) Plus Pembrolizumab in Patients With Newly Diagnosed Glioblastoma (2-THE-TOP) [NCT03405792]Phase 231 participants (Actual)Interventional2018-02-23Active, not recruiting
Phase II Study of Hu3F8, Irinotecan/Temozolomide and Sargramostim (HITS) Chemoimmunotherapy for High-Risk Neuroblastoma [NCT03189706]Early Phase 148 participants (Actual)Interventional2017-06-12Active, not recruiting
Post Marketing Surveillance of Radiotherapy With Concomitant and Adjuvant Chemotherapy With Temozolomide for Patients With Newly Diagnosed and Operated Glioblastoma Multiforme [NCT00704808]180 participants (Actual)Observational2006-05-31Completed
Phase I/II Evaluation of Everolimus (RAD001), Radiation and Temozolomide (TMZ) Followed by Adjuvant Temozolomide and Everolimus in Newly Diagnosed Glioblastoma [NCT00553150]Phase 1/Phase 2122 participants (Actual)Interventional2009-03-31Completed
Study With High Doses of Chemotherapy, Radiotherapy and Consolidation Therapy With Ciclofosfamide and Anticyclooxygenase 2, for the Metastatic Ewing Sarcoma [NCT02727387]Phase 2155 participants (Actual)Interventional2009-06-01Completed
Phase II Randomized, Prospective Trial of Lutetium Lu 177 Dotatate PRRT Versus Capecitabine and Temozolomide in Well-Differentiated Pancreatic Neuroendocrine Tumors [NCT05247905]Phase 2198 participants (Anticipated)Interventional2022-03-16Recruiting
Phase III Intergroup Study of Radiotherapy With Concomitant and Adjuvant Temozolomide Versus Radiotherapy With Adjuvant PCV Chemotherapy in Patients With 1p/19q Co-deleted Anaplastic Glioma or Low Grade Glioma [NCT00887146]Phase 3360 participants (Anticipated)Interventional2009-09-30Recruiting
Phase I Trial of R115777 With Radiation Therapy and Temozolomide in Patients With Newly Diagnosed Glioblastoma Multiforme [NCT02227901]Phase 119 participants (Actual)Interventional2002-09-30Completed
A Phase 1B Study of the Safety of LAM561 Administered Orally in Combination With Temozolomide (TMZ) and Radiation Therapy or With TMZ Alone in the First Line Treatment of Subjects With Glioblastoma [NCT03867123]Phase 118 participants (Actual)Interventional2018-12-04Completed
A Multi-Center, Open-Label, Phase I/II Study of GEM1777 in Combination With Temozolomide Administered Every 4 Weeks to Patients With Metastatic Melanoma [NCT00724841]Phase 1/Phase 21 participants (Actual)Interventional2008-06-30Terminated(stopped due to Study terminated prematurely due to financial contraints.)
A Randomized, Open-Label, Multicenter, Phase III Study of HX008 (a Humanized Monoclonal Antibody Against PD-1) Plus Transcatheter Arterial Chemoembolization (TACE) in the First-Line Treatment of Subjects With Stage IV (M1c) Melanoma That is Metastatic to [NCT05647954]Phase 3350 participants (Anticipated)Interventional2022-12-31Not yet recruiting
INdividualized Screening Trial of Innovative Glioblastoma Therapy (INSIGhT) [NCT02977780]Phase 2250 participants (Anticipated)Interventional2017-02-09Recruiting
High-Risk Neuroblastoma Study 2 of SIOP-Europa-Neuroblastoma (SIOPEN) [NCT04221035]Phase 3800 participants (Anticipated)Interventional2019-11-05Recruiting
"Medical Treatment of High-Risk Neurofibromas in Patients With Type 1 Neurofibromatosis: A Clinical Trial of Sequential Medical Therapies" [NCT00846430]Phase 29 participants (Actual)Interventional2008-10-31Completed
A Phase II Study of Concurrent Radiation Therapy, Temozolomide, and Bevacizumab Followed by Bevacizumab/Everolimus in the First-line of Treatment of Patients With Glioblastoma Multiforme [NCT00805961]Phase 268 participants (Actual)Interventional2009-01-31Completed
International Multicenter Phase I Trial of Hydroxyurea in Combination With Dose-Intense Temozolomide in Recurrent Glioblastoma [NCT03463733]Phase 154 participants (Anticipated)Interventional2018-03-02Recruiting
A Clinical Study of Supra-early Post-Surgery Chemotherapy Plus Standard TEMODAL® Regimen Versus Standard TEMODAL® Regimen in the Treatment on Patients With Newly Diagnosed Glioblastoma Multiforme [NCT02520635]180 participants (Anticipated)Observational2015-01-31Recruiting
G-FORCE-1: An Open-Label Phase 1 Two Part Dose Escalation Trial of RRx-001 Concurrent With Radiation and Temozolomide and RRx-001 + Temozolomide Post-RT In Newly Diagnosed Glioblastoma and Anaplastic Gliomas With Intact 1p/19q Chromosomes [NCT02871843]Phase 119 participants (Actual)Interventional2017-02-14Completed
Rapid Analysis and Response Evaluation of Combination Anti-Neoplastic Agents in Rare Tumors (RARE CANCER) Trial: RARE 2 Talazoparib and Temozolomide [NCT05142241]Phase 234 participants (Anticipated)Interventional2022-07-11Recruiting
Anticancer Therapeutic Vaccination Using Telomerase-derived Universal Cancer Peptides in Glioblastoma [NCT04280848]Phase 256 participants (Anticipated)Interventional2020-05-26Active, not recruiting
A Phase 2 Study of Veliparib (ABT-888) and Local Irradiation, Followed by Maintenance Veliparib and Temozolomide, in Patients With Newly Diagnosed High-Grade Glioma (HGG) Without H3 K27M or BRAFV600 Mutations [NCT03581292]Phase 238 participants (Actual)Interventional2018-11-06Active, not recruiting
A Phase 1 Study of Pevonedistat (MLN4924), a NEDD8 Activating Enzyme (NAE) Inhibitor, in Combination With Temozolomide and Irinotecan in Pediatric Patients With Recurrent or Refractory Solid Tumors [NCT03323034]Phase 130 participants (Actual)Interventional2018-01-11Active, not recruiting
Phase I/II Study of Oral Capecitabine and Temozolomide (CAPTEM) for Newly Diagnosed Glioblastoma (GBM) [NCT03213002]Phase 1/Phase 267 participants (Anticipated)Interventional2017-06-13Recruiting
Pharmacological Ascorbate Combined With Radiation and Temozolomide in Glioblastoma Multiforme: A Phase 2 Trial [NCT02344355]Phase 290 participants (Anticipated)Interventional2017-03-13Active, not recruiting
Phase I Study of AZD1775 (Adavosertib) With Radiation and Temozolomide in Patients With Newly Diagnosed Glioblastoma and Evaluation of Intratumoral Drug Distribution in Patients With Recurrent Glioblastoma [NCT01849146]Phase 174 participants (Actual)Interventional2013-08-19Active, not recruiting
A Phase I Study of the PARP Inhibitor ABT-888 in Combination With Temozolomide in Acute Leukemias [NCT01139970]Phase 166 participants (Actual)Interventional2010-05-21Active, not recruiting
A Phase I Study of Vorinostat (Suberoylanilide Hydroxamic Acid [SAHA]) in Combination With Temozolomide in Patients With Malignant Gliomas [NCT00268385]Phase 183 participants (Actual)Interventional2005-12-16Active, not recruiting
A Phase Ib/II, Open-label Study of NBM-BMX as Monotherapy or in Combination With Radiotherapy and Temozolomide in Subjects With Solid Tumors or Newly Diagnosed Glioblastoma [NCT06012695]Phase 1/Phase 279 participants (Anticipated)Interventional2023-08-11Recruiting
Feasibility of Evaluating Abemaciclib Neuropharmacokinetics of Diffuse Midline Glioma Using Intratumoral Microdialysis [NCT05413304]Phase 17 participants (Anticipated)Interventional2023-04-07Recruiting
Randomized Phase II Study Comparing the MET Inhibitor Cabozantinib to Temozolomide/Dacarbazine in Ocular Melanoma [NCT01835145]Phase 247 participants (Actual)Interventional2013-07-31Completed
A Phase 1/2 Study of BMN 673, an Oral Poly(ADP-Ribose) Polymerase Inhibitor, Plus Temozolomide in Children With Refractory or Recurrent Malignancies [NCT02116777]Phase 1/Phase 240 participants (Actual)Interventional2014-05-16Completed
Phase II Study of High-Dose Rituximab Combined With Temozolomide as Treatment for Patients With Primary CNS Lymphoma [NCT02113007]Phase 22 participants (Actual)Interventional2014-07-31Terminated(stopped due to Closed early due to slow accrual.)
Brain Interstitium Temozolomide Concentration Pre and Post Regadenoson Administration [NCT02389738]Early Phase 16 participants (Actual)Interventional2015-02-28Completed
A Randomized Feasibility Study Evaluating Temozolomide Chronotherapy for High Grade Glioma [NCT02781792]Phase 242 participants (Actual)Interventional2016-08-11Active, not recruiting
A Follow-Up Study to Add Whole Brain Radiotherapy (WBRT) to Standard Temozolomide Chemo-Radiotherapy in Newly Diagnosed Glioblastoma (GBM) Treated With 4 Weeks of Continuous Infusion Plerixafor [NCT03746080]Phase 220 participants (Anticipated)Interventional2018-12-04Recruiting
Phase I / II Study of the Combination of Doxycycline With Temozolomide and Ipilimumab in Patients With Metastatic Melanoma [NCT01590082]Phase 1/Phase 212 participants (Actual)Interventional2012-11-30Terminated(stopped due to Accrual slow due to alternate trials and treatment options.)
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00026494 (1) [back to overview]Radiographic Response Assessed by Macdonald Criteria Every 2 Months
NCT00050986 (1) [back to overview]Maximal Tolerating Dose (MTD for Phase I)
NCT00068250 (4) [back to overview]Phase II: Pre-irradiation Chemotherapy Tumor Response Rate (Including Phase I Patients at Same Dose)
NCT00068250 (4) [back to overview]Number of Phase I Participants Experiencing Toxicity
NCT00068250 (4) [back to overview]Phase II: Overall Survival Rate at 2 Years (Including Phase I Patients at Same Dose)
NCT00068250 (4) [back to overview]Phase II: Progression-free Survival (Including Phase I Patients at Same Dose)
NCT00077207 (6) [back to overview]Number of Participants Who Experienced a Grade 3 or 4 Thrombocytopenia and/or Neutropenia.
NCT00077207 (6) [back to overview]Number of Participants Who Experienced Toxic Death
NCT00077207 (6) [back to overview]Percent Probability of Progression-free Survival (PFS)
NCT00077207 (6) [back to overview]Percentage Probability of Event-free Survival (EFS)
NCT00077207 (6) [back to overview]Short Term Feasibility Success
NCT00077207 (6) [back to overview]Long Term Feasibility Success
NCT00080938 (4) [back to overview]1-year Neurologic (Central Nervous System, CNS) Progression Free Rate
NCT00080938 (4) [back to overview]Number of Patients With Intracranial Response
NCT00080938 (4) [back to overview]Overall Survival Time
NCT00080938 (4) [back to overview]Time to Non-CNS (Systemic) Progression
NCT00084838 (19) [back to overview]Grade 3-4 Pulmonary Events
NCT00084838 (19) [back to overview]2-yr Overall Survival
NCT00084838 (19) [back to overview]Grade 3-4 Allergy/Immunology
NCT00084838 (19) [back to overview]Grade 3-4 Auditory/Hearing Events
NCT00084838 (19) [back to overview]Grade 3-4 Blood/Bone Marrow Events
NCT00084838 (19) [back to overview]Grade 3-4 Hepatic Events
NCT00084838 (19) [back to overview]Grade 3-4 Pain Events
NCT00084838 (19) [back to overview]Grade 3-4 Cardiovascular Events
NCT00084838 (19) [back to overview]Grade 3-4 Constitutional Events
NCT00084838 (19) [back to overview]Pre-Radiation Therapy Chemotherapeutic Response
NCT00084838 (19) [back to overview]Grade 3-4 Renal/Genitourinary Events
NCT00084838 (19) [back to overview]Grade 3-4 Dermatology Events
NCT00084838 (19) [back to overview]Grade 3-4 Neurology Events
NCT00084838 (19) [back to overview]Grade 3-4 Muscloskeletal Events
NCT00084838 (19) [back to overview]Grade 3-4 Gastrointestinal Events
NCT00084838 (19) [back to overview]Grade 3-4 Hemorrhage Events
NCT00084838 (19) [back to overview]Grade 3/4 Events
NCT00084838 (19) [back to overview]Grade 3-4 Metabolic/Laboratory Events
NCT00084838 (19) [back to overview]Grade 3-4 Infection/Febrile Neutropenia Events
NCT00085254 (5) [back to overview]Dose Limiting Toxicities of EMD + RT and TMZ
NCT00085254 (5) [back to overview]Overall Survival (Phase II)
NCT00085254 (5) [back to overview]Overall Survival Based on Dose Level - Phase 2
NCT00085254 (5) [back to overview]Maximum Tolerated or Tolerable Dose (MTD) - 3 Pre-defined Doses
NCT00085254 (5) [back to overview]Frequency of Hematologic and Nonhematologic Adverse Events
NCT00091572 (4) [back to overview]Progression Free Survival
NCT00091572 (4) [back to overview]Overall Survival
NCT00091572 (4) [back to overview]Objective Response Rate in Subjects With Measurable Lesions
NCT00091572 (4) [back to overview]Duration of Objective Response
NCT00096265 (6) [back to overview]Change in Steroid Dependence at Six Months
NCT00096265 (6) [back to overview]Change in Performance Status at Six Months
NCT00096265 (6) [back to overview]Change in Functional Assessment of Cancer Therapy-Brain (FACT-Br) Score at 3 Months
NCT00096265 (6) [back to overview]Quality-adjusted Survival as Measured by EuroQol 5-dimension Instrument
NCT00096265 (6) [back to overview]Rate of CNS Progression (One Year)
NCT00096265 (6) [back to overview]Overall Survival
NCT00098774 (4) [back to overview]4 Year Overall Survival Rate
NCT00098774 (4) [back to overview]Change From Baseline in Mini-Mental Status Evaluation at 4 Months
NCT00098774 (4) [back to overview]Complete Response Rate After Remission Induction
NCT00098774 (4) [back to overview]4 Year Progression Free Rate
NCT00098865 (3) [back to overview]Overall Response
NCT00098865 (3) [back to overview]Therapy Completion Rate
NCT00098865 (3) [back to overview]Overall Survival
NCT00100802 (2) [back to overview]One Year Overall Survival
NCT00100802 (2) [back to overview]Occurrence of Death Attributable to Complications of Protocol Therapy
NCT00112502 (10) [back to overview]Median Overall Survival (OS) Comparison of Doublet Versus Triplet Therapy
NCT00112502 (10) [back to overview]Median Overall Survival (OS) Comparison of Celecoxib Arms Versus no Celecoxib Arms
NCT00112502 (10) [back to overview]Median Progression-Free Survival (PFS) of Individual Arms
NCT00112502 (10) [back to overview]Median Overall Survival (OS) Comparison of Thalidomide Arms Versus no Thalidomide Arms
NCT00112502 (10) [back to overview]Median Progression-Free Survival (PFS) Comparison of Celecoxib Arms Versus no Celecoxib Arms
NCT00112502 (10) [back to overview]Median Progression-Free Survival (PFS) Comparison of Doublet Versus Triplet Therapy
NCT00112502 (10) [back to overview]Median Overall Survival (OS) Comparison of Isotretinoin Arms Versus no Isotretinoin Arms
NCT00112502 (10) [back to overview]Overall Survival of Individual Arms
NCT00112502 (10) [back to overview]Median Progression-Free Survival (PFS) Comparison of Isotretinoin Arms Versus no Isotretinoin Arms
NCT00112502 (10) [back to overview]Median Progression-Free Survival (PFS) Comparison of Thalidomide Arms Versus no Thalidomide Arms
NCT00114140 (5) [back to overview]Neurocognitive Function
NCT00114140 (5) [back to overview]Progression-free Survival
NCT00114140 (5) [back to overview]Overall Survival Rate at 3 Years
NCT00114140 (5) [back to overview]Survival and Progression-free Survival by O(6)-Methylguanine-DNA Methyltransferase (MGMT) Methylation Status
NCT00114140 (5) [back to overview]Quality of Life as Measured by the Functional Assessment of Cancer Therapy Scale With Brain Module (FACT-BR)
NCT00187486 (2) [back to overview]Overall Survival
NCT00187486 (2) [back to overview]Progression Free Survival
NCT00200161 (2) [back to overview]Progression Free Survival at 6 Months
NCT00200161 (2) [back to overview]12 Month Overall Survival of Patients With Newly Diagnosed Glioblastoma Multiforme Treated With Concurrent Temozolomide and Radiotherapy Followed by Dose Dense or Metronomic Dosing of Temozolomide and Maintenance Cis-retinoic Acid.
NCT00248534 (4) [back to overview]Number of Participants Alive at 3 Years
NCT00248534 (4) [back to overview]Percentage of Participants With Objective Response
NCT00248534 (4) [back to overview]6-month Progression-free Survival
NCT00248534 (4) [back to overview]1 Year Overall Survival Rate
NCT00262730 (1) [back to overview]Survival
NCT00266812 (1) [back to overview]Number of Participants With Progression-free Survival (6 Month)
NCT00275002 (2) [back to overview]Percentage of Participants With an Objective Response (Complete Response or Partial Response)
NCT00275002 (2) [back to overview]Number of Patients With Grade 3 or 4 Adverse Events at Least Possibly Related to the Combination of O6-benzylguanine and Temozolomide
NCT00301067 (6) [back to overview]Overall Response Rate
NCT00301067 (6) [back to overview]Number and Frequency of Dose Limiting Toxicities (DLTs) With High-dose Calcitriol in Combination With Temozolomide
NCT00301067 (6) [back to overview]Overall Survival (OS) Stratified by Vitamin D-Receptor (VDR) Gene Polymorphisms
NCT00301067 (6) [back to overview]Number of Patients With Toxicity
NCT00301067 (6) [back to overview]Time to Progression
NCT00301067 (6) [back to overview]Overall Survival
NCT00302159 (6) [back to overview]Percentage of Participants With Overall Survival at 6, 12, and 24 Months
NCT00302159 (6) [back to overview]Percentage of Participants With Progression Free Survival at 6, 12, and 24 Months
NCT00302159 (6) [back to overview]Median Progression Free Survival.
NCT00302159 (6) [back to overview]Median Overall Survival
NCT00302159 (6) [back to overview]Number of Participants With Adverse Events
NCT00302159 (6) [back to overview]Number of Participants With Best Response
NCT00304031 (18) [back to overview]Mean Neurocognitive Function (NCF) Composite Score Over Time
NCT00304031 (18) [back to overview]Median Progression-free Survival Time by MGMT Status
NCT00304031 (18) [back to overview]Determination of Impactful Baseline Instruments on Overall Survival
NCT00304031 (18) [back to overview]Median Overall Survival Time by MGMT Status
NCT00304031 (18) [back to overview]Median Progression-free Survival (PFS) Time
NCT00304031 (18) [back to overview]Mean MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score Over Time
NCT00304031 (18) [back to overview]Change From Baseline in Mean EORTC QLQ-C30 Global Health Status
NCT00304031 (18) [back to overview]Overall Survival Status by Progression Status at 6 Months
NCT00304031 (18) [back to overview]Number of Participants With Deterioration From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Cycle 4
NCT00304031 (18) [back to overview]Number of Participants With Deterioration From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Interference Score at Cycle 4
NCT00304031 (18) [back to overview]Median Overall Survival Time
NCT00304031 (18) [back to overview]Mean Neurocognitive Function (NCF) Composite Score at Cycle 10 for Participants Without Progression After 6 Months of Adjuvant Therapy
NCT00304031 (18) [back to overview]Mean MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Cycle 10 for Participants Without Progression After 6 Months of Adjuvant Therapy
NCT00304031 (18) [back to overview]Mean EORTC QLQ-C30 Global Health Status Score at Cycle 10 for Participants Without Progression After 6 Months of Adjuvant Therapy
NCT00304031 (18) [back to overview]Mean Change From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Mid-cyle for Cycle 4
NCT00304031 (18) [back to overview]Mean Change From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Mid-cyle for Cycle 1
NCT00304031 (18) [back to overview]Mean Change From Baseline in EORTC QLQ-C30 Global Health Status Score at Mid-cyle for Cycle 4
NCT00304031 (18) [back to overview]Mean Change From Baseline in EORTC QLQ-C30 Global Health Status Score at Mid-cyle for Cycle 1
NCT00305864 (3) [back to overview]Progression-free Survival (Phase II)
NCT00305864 (3) [back to overview]Median Overall Survival (Phase II)
NCT00305864 (3) [back to overview]Maximum Tolerated Dose of MGd (Phase I)
NCT00311584 (1) [back to overview]Overall Response - Complete Response (CR), Very Good Partial Response (VGPR) and Partial Response (PR)
NCT00313729 (3) [back to overview]Response Rate (Complete and Partial Response)
NCT00313729 (3) [back to overview]Safety Profile
NCT00313729 (3) [back to overview]Time to Tumor Progression
NCT00323115 (13) [back to overview]Number of Enzyme-linked Immunosorbent Spots (ELISPOT) - Correlation Between Immunological Parameters and Efficacy - Median
NCT00323115 (13) [back to overview]Number of Participants With Evaluable Data: Feasibility of Vaccination
NCT00323115 (13) [back to overview]Evaluation of T Cell Characteristics
NCT00323115 (13) [back to overview]Number of Participants With Significant Difference in Tumor Volume Size Pre- and Postvaccination: Neuroimaging and Tumor Assessment
NCT00323115 (13) [back to overview]Overall Survival Duration: Efficacy Parameters
NCT00323115 (13) [back to overview]Progression Free Survival (PFS)
NCT00323115 (13) [back to overview]Frequency of CD4+ and CD8+ T Cells - the Proportion of Cells in the Parent Population Responding to Glioblastoma Multiforme (GBM) - Mean
NCT00323115 (13) [back to overview]Frequency of CD4+ and CD8+ T Cells - the Proportion of Cells in the Parent Population Responding to Glioblastoma Multiforme (GBM) - Median
NCT00323115 (13) [back to overview]Percentage of Tumor-specific T-cells - Correlation Between Immunological Parameters and Efficacy- Mean
NCT00323115 (13) [back to overview]Percentage of Tumor-specific T-cells - Correlation Between Immunological Parameters and Efficacy- Median
NCT00323115 (13) [back to overview]Tumor-specific Cytotoxic T-cell Response
NCT00323115 (13) [back to overview]Number of Adverse Events: Toxicity Profile of Intra-nodal DC/Tumor Lysate Vaccination
NCT00323115 (13) [back to overview]Number of Enzyme-linked Immunosorbent Spots (ELISPOT) - Correlation Between Immunological Parameters and Efficacy - Mean
NCT00362817 (5) [back to overview]Analyze Patients Time to Progression
NCT00362817 (5) [back to overview]Determine the Overall Survival of Patients
NCT00362817 (5) [back to overview]The Incidence and Severity of Centeral Nervous System (CNS) Toxicities
NCT00362817 (5) [back to overview]Determine the Cause of Death of Patients After Treatment
NCT00362817 (5) [back to overview]Affects of Response Rate of Chemotherapy With Intra-arterial Carboplatin and Oral Temozolomide
NCT00392171 (1) [back to overview]Percentage of Participants Surviving at Six Months of Treatment Without Evidence of Disease Progression.
NCT00402116 (11) [back to overview]Number of Participants Undergoing Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy (MRI/MRS) for Clinical Evaluation at Baseline
NCT00402116 (11) [back to overview]Phase 1 and 2- Pharmacokinetics: Area Under the Concentration-Time Curve During 1 Dosing Interval at Steady State (AUCτ,ss) for Enzastaurin, LY326020 and Total Analyte (Enzastaurin + LY326020) When Enzastaurin Administered With or Without Temozolomide
NCT00402116 (11) [back to overview]Phase 1 - Number of Participants With Adverse Events (AEs)
NCT00402116 (11) [back to overview]M.D. Anderson Symptom Inventory - Brain Tumor (MDASI-BT)
NCT00402116 (11) [back to overview]Phase 1 and 2 - Progression-Free Survival (PFS)
NCT00402116 (11) [back to overview]Phase 1- Pharmacokinetics (PK): Maximum Observed Drug Concentration During 1 Dosing Interval at Steady State (Cmax,ss) for Enzastaurin, LY326020, and Total Analyte (Enzastaurin + LY326020) When Enzastaurin Administered With or Without Temozolomide
NCT00402116 (11) [back to overview]Functional Assessment of Cancer Therapy - Brain (FACT-Br)
NCT00402116 (11) [back to overview]Phase 1 and Phase 2 - Overall Survival (OS)
NCT00402116 (11) [back to overview]Phase 1- Determination of the Maximum Tolerated Dose (MTD) of Enzastaurin
NCT00402116 (11) [back to overview]Phase 2 - Number of Participants With Adverse Events (AEs)
NCT00402116 (11) [back to overview]Phase 1 and 2: Association Between Biomarkers and Clinical Outcome
NCT00404495 (6) [back to overview]Percentage of Participants With Objective Response of Complete Response or Partial Response
NCT00404495 (6) [back to overview]Percentage of Participants With Objective Response of Complete Response or Partial Response, Investigator's Assessment
NCT00404495 (6) [back to overview]Time to Treatment Failure (TTF)
NCT00404495 (6) [back to overview]Time to Tumor Progression (TTP)
NCT00404495 (6) [back to overview]Duration of Response
NCT00404495 (6) [back to overview]Overall Survival (OS)
NCT00423150 (1) [back to overview]Tumor Responses (Complete and Partial Response)
NCT00424554 (3) [back to overview]Tolerability: Number of Participants Discontinuing Treatment Due to Adverse Events (AE)
NCT00424554 (3) [back to overview]MethylGuanine-DNA MethylTransferase [MGMT] Activity Measured From the Tumor Tissue During Surgery
NCT00424554 (3) [back to overview]Safety: Number of Participants Who Experienced Grade 3 or 4 Toxicities
NCT00433381 (8) [back to overview]Change in Perfusion MRI Markers at Week 8 as Predictors of 12mo Overall Survival (OS)
NCT00433381 (8) [back to overview]Agreement Between Local Interpretation and Central Interpretation of Standard MRI
NCT00433381 (8) [back to overview]Change in Perfusion MRI Markers at Week 16 as Predictors of 12mo Overall Survival (OS)
NCT00433381 (8) [back to overview]Accuracy of Local PFS 6-mo Interpretation Using Central Review PFS-6 as the Reference Standard
NCT00433381 (8) [back to overview]Count/Percentage of Patients Discontinuing Treatment Due to Treatment-related Medical Complications(Bevacizumab and Temozolomide Arm)
NCT00433381 (8) [back to overview]Count/Percentage of Patients Progression-free at 6 Months for Bevacizumab and Irinotecan Hydrochloride Arm
NCT00433381 (8) [back to overview]Count/Percentage of Patients Progression-free at 6 Months for Bevacizumab and Temozolomide Arm
NCT00433381 (8) [back to overview]Change in Perfusion MRI Markers at Week 2 as Predictors of 12mo Overall Survival (OS)
NCT00436436 (1) [back to overview]Toxicity as Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v 3)
NCT00441142 (5) [back to overview]PHASE I: Percentage of Grade 3-5 Treatment-Related Adverse Events
NCT00441142 (5) [back to overview]Number of Participants That Experienced a Dose-limiting Toxicity (DLT)
NCT00441142 (5) [back to overview]Median Progression-free Survival (PFS), as Calculated by the # of Months Patients Remain Progression-free
NCT00441142 (5) [back to overview]Median Overall Survival (OS) of Phase II Patients
NCT00441142 (5) [back to overview]PHASE II: Percentage of Grade 3-5 Treatment-Related Adverse Events
NCT00482677 (3) [back to overview]Methylation Status of the O6-methylguanine-DNA Methyltransferase Promoter
NCT00482677 (3) [back to overview]Overall Survival
NCT00482677 (3) [back to overview]Progression-free Survival
NCT00486603 (10) [back to overview](Phase II) Overall Survival
NCT00486603 (10) [back to overview](Phase II) Number of Participants With Grade 3 and 4 Toxicity
NCT00486603 (10) [back to overview](Phase I) Number of Participants Who Experienced Dose Limiting Toxicity (DLT)
NCT00486603 (10) [back to overview]Pharmocodynamics as Determined by Number of Participants With Autophagy Inhibition in Relation to Maximal Concentration (Cmax) of HCQ
NCT00486603 (10) [back to overview]PK of Hydroxychloroquine as Measured by Oral Clearance (Liters/Hour) From Central Compartment (CL/F)
NCT00486603 (10) [back to overview]PK of Hydroxychloroquine as Measured by Volume of Distribution of Central Compartment (V/F)
NCT00486603 (10) [back to overview](Phase I) Maximum Tolerated Dose (MTD) of Hydroxychloroquine (HCQ)
NCT00486603 (10) [back to overview]PK of Hydroxychloroquine as Measured by First-order Absorption Rate Constant (Ka)
NCT00486603 (10) [back to overview]PK of Hydroxychloroquine as Measured by Distribution Volume of Peripheral Compartment (V2/F)
NCT00486603 (10) [back to overview]Pharmacokinetics (PK) of Hydroxychloroquine as Measured by Lag Time (Tlag)
NCT00498927 (2) [back to overview]Progression-free Survival (PFS) Rate at 6 Months
NCT00498927 (2) [back to overview]Overall Survival
NCT00501891 (4) [back to overview]Incidence and Severity of CNS Hemorrhage and Systemic Hemorrhage
NCT00501891 (4) [back to overview]Incidence of Grade ≥ 4 Hematologic or Grade ≥ 3 Non-hematologic Toxicity
NCT00501891 (4) [back to overview]6-Month Progression-free Survival
NCT00501891 (4) [back to overview]Response Rate
NCT00504660 (2) [back to overview]6 Month Progression-free Survival for Participants With Glioblastoma
NCT00504660 (2) [back to overview]12 Month-progression-free Survival for Participants With Anaplastic Tumors
NCT00505635 (1) [back to overview]Time to Progression (TTP)
NCT00512798 (5) [back to overview]Optimal Doses of Temozolomide and Bortezomib (Phase I)
NCT00512798 (5) [back to overview]Patients With Clinical Anti-tumor Activity (Phase I)
NCT00512798 (5) [back to overview]Patients With Inhibition of NF-kB (Phase II)
NCT00512798 (5) [back to overview]Patients With Inhibition in NF-kB Activation (Phase I)
NCT00512798 (5) [back to overview]Number of Patients With Clinical Anti-tumor Activity Phase II)
NCT00521001 (4) [back to overview]9-week Progression-free Survival Rate
NCT00521001 (4) [back to overview]Confirmed Response Rate (Complete Response and Partial Response)
NCT00521001 (4) [back to overview]Survival Time
NCT00521001 (4) [back to overview]Time to Disease Progression
NCT00525525 (3) [back to overview]Unexpected Toxicities During First 2 Cycles of Study Drug
NCT00525525 (3) [back to overview]Progression-free Survival
NCT00525525 (3) [back to overview]Overall Survival (OS)
NCT00539591 (23) [back to overview]Mean Total PedsQL 3.0 Scores for Child Cancer Quality of Life (QoL) Assessments (Stratum B)
NCT00539591 (23) [back to overview]Mean Total PedsQL 3.0 Scores for Parent Cancer Quality of Life (QoL) Assessments (Stratum A)
NCT00539591 (23) [back to overview]Mean Total PedsQL 3.0 Scores for Parent Cancer Quality of Life (QoL) Assessments (Stratum B)
NCT00539591 (23) [back to overview]Apparent Clearance (CL) of Pegylated Interferon ɑ-2B
NCT00539591 (23) [back to overview]Mean Total PedsQL 4.0 Scores for Child Quality of Life (QoL) Assessments (Stratum A)
NCT00539591 (23) [back to overview]Mean Total PedsQL 4.0 Scores for Child Quality of Life (QoL) Assessments (Stratum B)
NCT00539591 (23) [back to overview]Volume of Central Compartment (Vc) of Interferon ɑ-2b
NCT00539591 (23) [back to overview]Tumor Response Rate
NCT00539591 (23) [back to overview]Mean Total PedsQL 4.0 Scores for Parent Quality of Life Assessments (Stratum A)
NCT00539591 (23) [back to overview]Mean Total PedsQL 4.0 Scores for Parent Quality of Life Assessments (Stratum B)
NCT00539591 (23) [back to overview]Median Steady State Trough Concentration of Pegylated Interferon ɑ-2B
NCT00539591 (23) [back to overview]Number of Patients Who Experience Toxicity at or Above the Target Toxicity for Stratum A Patients
NCT00539591 (23) [back to overview]Half-Life of Interferon ɑ-2b
NCT00539591 (23) [back to overview]Volume of Central Compartment (Vc) of Pegylated Interferon ɑ-2B
NCT00539591 (23) [back to overview]Systemic Clearance (CL) of Interferon ɑ-2B
NCT00539591 (23) [back to overview]Probability of Event-free Survival (EFS) of Stratum A Participants
NCT00539591 (23) [back to overview]Number of Patients Who Experience Toxicity at or Above the Target Toxicity for Strata B1 and B2
NCT00539591 (23) [back to overview]Mean Total PedsQL 3.0 Scores for Child Cancer Quality of Life (QoL) Assessments (Stratum A)
NCT00539591 (23) [back to overview]BRIEF Psychological Assessment (Stratum A)
NCT00539591 (23) [back to overview]BASC-2 Psychological Assessment (Stratum A)
NCT00539591 (23) [back to overview]ɑ Half Life of Pegylated Interferon ɑ-2B
NCT00539591 (23) [back to overview]Area Under the Curve (AUC) of Pegylated Interferon ɑ-2B
NCT00539591 (23) [back to overview]Area Under the Curve (AUC) of Interferon ɑ-2b
NCT00544817 (3) [back to overview]Progression-free Survival
NCT00544817 (3) [back to overview]Objective Response
NCT00544817 (3) [back to overview]Overall Survival
NCT00553150 (6) [back to overview]Overall Survival Time
NCT00553150 (6) [back to overview]Overall Survival at 12 Months (Phase II)
NCT00553150 (6) [back to overview]Maximum Tolerated Dose (MTD) of Everolimus (RAD001) in Combination With Temozolomide (TMZ) and 3D-conformal Radiotherapy (RT) or Intensity-modulated Radiotherapy (IMRT) Followed by Adjuvant TMZ With or Without RAD001 (Phase I)
NCT00553150 (6) [back to overview]Time to Progression (Phase II)
NCT00553150 (6) [back to overview]Response Rate, as Measured in Patients Receiving FLT-PET Imaging (Phase II)
NCT00553150 (6) [back to overview]Progression-free-survival at 6 Months (Phase II)
NCT00568451 (3) [back to overview]Time to Disease Progression
NCT00568451 (3) [back to overview]Survival Time
NCT00568451 (3) [back to overview]Number of Participants With an Objective Tumor Status of Either a Complete Response(CR) or Partial Response (PR), According to RECIST (Response Evaluation Criteria in Solid Tumors) Criteria
NCT00575887 (1) [back to overview]Progression-free Survival at 6-months
NCT00576680 (3) [back to overview]To Determine the Safety and Tolerability of This Drug Combination.
NCT00576680 (3) [back to overview]Progression-free Survival
NCT00576680 (3) [back to overview]Response Rate
NCT00582075 (2) [back to overview]Percentage of Participants With Distant Brain Failure (DBF) at One Year
NCT00582075 (2) [back to overview]Overall Survival
NCT00588341 (1) [back to overview]Overall Objective Response (Complete Response or Partial Response)
NCT00588523 (2) [back to overview]Progession Free Survival
NCT00588523 (2) [back to overview]Number of Participants Evaluated for Toxicities
NCT00590681 (5) [back to overview]Objective Response
NCT00590681 (5) [back to overview]Overall Survival
NCT00590681 (5) [back to overview]Safety of Avastin in Combination With Temozolomide in This Study Population
NCT00590681 (5) [back to overview]Duration of Response
NCT00590681 (5) [back to overview]Progression-free Survival (PFS)
NCT00591370 (3) [back to overview]Duration of Objective Clinical Responses
NCT00591370 (3) [back to overview]Overall Survival
NCT00591370 (3) [back to overview]Determine the Overall Objective Response Rate (CR and PR).
NCT00597402 (4) [back to overview]Number of Patients Experiencing a Central Nervous System (CNS) Hemorrhage or a Systemic Hemorrhage
NCT00597402 (4) [back to overview]12-month Progression-free Survival (PFS)
NCT00597402 (4) [back to overview]16-month Overall Survival (OS)
NCT00597402 (4) [back to overview]Number of Patients Experiencing a Grade ≥ 4 Hematologic or Grade ≥ 3 Non-hematologic Toxicity
NCT00597493 (5) [back to overview]Safety and Toxicity of Combination
NCT00597493 (5) [back to overview]Pharmacokinetics: T-max
NCT00597493 (5) [back to overview]Pharmacokinetics: C-max
NCT00597493 (5) [back to overview]Pharmacokinetics: AUC-24
NCT00597493 (5) [back to overview]6 Month Progression Free Survival (PFS)
NCT00602576 (3) [back to overview]Overall Survival Rate
NCT00602576 (3) [back to overview]Rate of 6 Month Progression-Free Survival
NCT00602576 (3) [back to overview]Response Rate
NCT00611247 (3) [back to overview]Toxicity Profile: Total Number of Drug-related Serious Adverse Events
NCT00611247 (3) [back to overview]Toxicity Profile: Individual Subjects With Drug-related SAEs
NCT00611247 (3) [back to overview]Response Rate (CR + CRi + LFS)
NCT00612339 (1) [back to overview]Response Rate
NCT00613028 (5) [back to overview]Radiographic Response
NCT00613028 (5) [back to overview]Median Progression-free Survival (PFS)
NCT00613028 (5) [back to overview]Median Overall Survival (OS)
NCT00613028 (5) [back to overview]Grade 3 or Greater, Treatment Related, Non-hematologic Toxicities.
NCT00613028 (5) [back to overview]The Primary Outcome Measure is 6 Month Progression-free Survival.
NCT00617539 (5) [back to overview]Overall Time of Survival
NCT00617539 (5) [back to overview]Number of Patients Whose Circulating Tumor Cells (CTCs) Decreased From >5 to <5 CTCs Per 7.5 mL
NCT00617539 (5) [back to overview]Number of Patients With Objective Treatment Response (Complete or Partial) in the CNS
NCT00617539 (5) [back to overview]Time to First Progression in CNS
NCT00617539 (5) [back to overview]Number of Patients Experiencing a Clinical Benefit
NCT00619112 (7) [back to overview]Progression-free Survival (PFS) Based on Tumor MGMT (O(6)-Methylguanine-DNA Methyltransferase) Promoter Methylation Status.
NCT00619112 (7) [back to overview]Patients With Tumors With Functional Alterations of the Mismatch Repair (MMR) System
NCT00619112 (7) [back to overview]6 Month Progression-free Survival
NCT00619112 (7) [back to overview]Patients Progressing 6 Months After Temozolomide is Voluntarily Discontinued
NCT00619112 (7) [back to overview]Overall Survival
NCT00619112 (7) [back to overview]Patients Progressing After Two First-line Adjuvant Courses of Temozolomide
NCT00619112 (7) [back to overview]Patients Progressing Within 6 Months After 6th Adjuvant Course of Temozolomide
NCT00626405 (3) [back to overview]Tumor Response Rate, Calculated as a Percentage Along With it's 95% Confidence Interval
NCT00626405 (3) [back to overview]Progression-free Survival at 6 Months
NCT00626405 (3) [back to overview]Overall Survival
NCT00626990 (2) [back to overview]Progression-free Survival
NCT00626990 (2) [back to overview]Overall Survival as Measured From the Day of Randomization
NCT00632203 (5) [back to overview]Tolerability of Maintenance Temozolomide
NCT00632203 (5) [back to overview]Time to Radiological Central Nervous System (CNS) Progression
NCT00632203 (5) [back to overview]Overall Survival
NCT00632203 (5) [back to overview]Number of Participants Who Had Brain Metastases
NCT00632203 (5) [back to overview]Time to Progression
NCT00638963 (5) [back to overview]Number of Days on Temozolomide Treatment
NCT00638963 (5) [back to overview]Number of Participants Who Had at Least One Treatment Omission During Treatment
NCT00638963 (5) [back to overview]Total Dose of Temozolomide Taken
NCT00638963 (5) [back to overview]Number of Participants Who Completed the Third Cycle of Treatment
NCT00638963 (5) [back to overview]Number of Participants Who Had at Least One Dose Reduction During Treatment
NCT00643097 (4) [back to overview]Humoral and Cellular Immune Response
NCT00643097 (4) [back to overview]Clinical Efficacy of Vaccination, in Terms of Progression-free Survival (PFS)
NCT00643097 (4) [back to overview]Response to Vaccination
NCT00643097 (4) [back to overview]Toxicity to PEP-3 Vaccine Immunization
NCT00657267 (4) [back to overview]Radiographic Response
NCT00657267 (4) [back to overview]6 Month Progression Free Survival
NCT00657267 (4) [back to overview]Time to Progression.
NCT00657267 (4) [back to overview]Overall Survival
NCT00667953 (2) [back to overview]Response
NCT00667953 (2) [back to overview]Safety - Grade 3 or 4 Adverse Events
NCT00669669 (10) [back to overview]Response Rate
NCT00669669 (10) [back to overview]Time to Progression
NCT00669669 (10) [back to overview]Duration of Response
NCT00669669 (10) [back to overview]Gene Transfer Efficiency After Chemotherapy
NCT00669669 (10) [back to overview]Gene Transfer Efficiency
NCT00669669 (10) [back to overview]Number of Participants Dose-limiting Toxicity (DLT)
NCT00669669 (10) [back to overview]Number of Participants That Survived
NCT00669669 (10) [back to overview]Number of Participants With Chemoprotection
NCT00669669 (10) [back to overview]Number of Participants With Chemoselection
NCT00669669 (10) [back to overview]Number of Participants With Retrovirus or Leukemia
NCT00684567 (5) [back to overview]Adverse Events With an Incidence of Greater Than or Equal to 20%
NCT00684567 (5) [back to overview]Number of Participants With Progression Free Survival (PFS) for 1 Year
NCT00684567 (5) [back to overview]Number of Participants With a Response (Complete Response [CR] + Partial Response [PR]) in Terms of Overall Tumor Response
NCT00684567 (5) [back to overview]Abnormal Changes in Laboratory Test Values With an Incidence of Greater Than or Equal to 20%
NCT00684567 (5) [back to overview]Adverse Drug Reactions With an Incidence of Greater Than or Equal to 20%
NCT00686725 (9) [back to overview]Relationship Between MGMT Status and Therapy Response: Overall Survival Rate for the MGMT Positive Group
NCT00686725 (9) [back to overview]Relationship Between MGMT Status and Therapy Response: Overall Survival Rate for the MGMT Negative Group
NCT00686725 (9) [back to overview]Relationship Between O6-methylguanine-DNA Methyltransferase (MGMT) Status and Therapy Response: Overall Survival for the MGMT Positive Group
NCT00686725 (9) [back to overview]Relationship Between MGMT Status and Therapy Response: PFS for the MGMT Positive Group
NCT00686725 (9) [back to overview]Relationship Between MGMT Status and Therapy Response: PFS for the MGMT Negative Group
NCT00686725 (9) [back to overview]Objective Tumor Assessment After Surgery: Overall Response
NCT00686725 (9) [back to overview]Relationship Between MGMT Status and Therapy Response: Overall Survival for the MGMT Negative Group
NCT00686725 (9) [back to overview]Progression-Free Survival (PFS)
NCT00686725 (9) [back to overview]Overall Survival (OS)
NCT00687323 (7) [back to overview]Number of Participants With CR, PR, or MLFS Who Received Modified Low Dose Maintenance Therapy (100 mg/m^2/Day x21 Days of Each 28 Day Cycle) and Experienced Toxicity
NCT00687323 (7) [back to overview]Number of Previously Untreated Participants With Low O6-Methylguanine Methyltransferase (MGMT) Expression
NCT00687323 (7) [back to overview]Clinical Response at the End of Temozolomide Induction
NCT00687323 (7) [back to overview]Progression-free Survival for Participants Achieving PR, MLSF, or MR
NCT00687323 (7) [back to overview]Overall Survival (OS) in Participants Achieving CR or CRp and Proceeding to Reduced Dose-intensity Maintenance Therapy With Temozolomide
NCT00687323 (7) [back to overview]Duration of Response in Participants Achieving Complete Response (CR) and Proceeding to Reduced Dose-intensity Maintenance Therapy With Temozolomide
NCT00687323 (7) [back to overview]Relapse-free Survival in Participants Achieving CR or CRp and Proceeding to Reduced Dose-intensity Maintenance Therapy With Temozolomide
NCT00689221 (11) [back to overview]European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Brain Module (EORTC QLQ-BN20) Sub-scale Scores
NCT00689221 (11) [back to overview]European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) Sub-scale Scores
NCT00689221 (11) [back to overview]Number of Participants With Adverse Events (AEs), Serious AEs, Treatment-Related AEs, Treatment-Related Serious AEs, AEs Leading to Death, Treatment Related AEs Leading to Death, AEs of Grade 3 or 4 and Treatment Related AEs of Grade 3 or 4
NCT00689221 (11) [back to overview]Number of Participants With AEs Belonging to Standardized Medical Dictionary for Regulatory Activities (MedDRA) Queries (SMQs) Thromboembolic Events and Hemorrhage With NCI-CTC Toxicity Grade 3 or 4
NCT00689221 (11) [back to overview]Number of Participants With Change From Baseline in Work Status at End of Study
NCT00689221 (11) [back to overview]Progression Free Survival (PFS) Time - Investigator and Independent Read
NCT00689221 (11) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT00689221 (11) [back to overview]EuroQol 5-Dimensions (EQ-5D) Questionnaire Index
NCT00689221 (11) [back to overview]Overall Survival (OS) Time
NCT00689221 (11) [back to overview]Area Under the Plasma Concentration Curve From Time 0 to 6 Hours (AUC [0-6]) After Dose
NCT00689221 (11) [back to overview]Time to Maximum Plasma Concentration (Tmax)
NCT00704808 (1) [back to overview]Median Progression Free Survival After Primary Surgical Treatment, Concomitant and Adjuvant Chemotherapy With Temozolomide, for Patients With Newly Diagnosed Glioblastoma Multiforme
NCT00715793 (8) [back to overview]Disease Control Rate (DCR)
NCT00715793 (8) [back to overview]Percentage of Participants That Experienced a Dose Limiting Toxicity (DLT)
NCT00715793 (8) [back to overview]Recommended Phase 2 Dose (RP2D) of DAC + TMZ
NCT00715793 (8) [back to overview]Progression-free Survival (PFS)
NCT00715793 (8) [back to overview]Overall Response Rate (ORR)
NCT00715793 (8) [back to overview]Overall Survival (OS)
NCT00715793 (8) [back to overview]1-year Overall Survival (OS) Rate
NCT00715793 (8) [back to overview]6-month Progression-free Survival (PFS) Rate
NCT00723827 (5) [back to overview]Number of Participants Experiencing Unexpected Adverse Drug Reactions (ADRs)
NCT00723827 (5) [back to overview]Number of Temozolomide Drug Interactions
NCT00723827 (5) [back to overview]Number of Temozolomide Misuse or Abuse Events
NCT00723827 (5) [back to overview]Efficacy: Number of Participants Experiencing Complete Response (CR), Partial Response (PR), or Stable Disease(SD)
NCT00723827 (5) [back to overview]Number of Participants Experiencing Adverse Events (AEs)
NCT00725010 (2) [back to overview]Number of Participants Who Discontinued Due to Toxicity
NCT00725010 (2) [back to overview]Safety: Number of Adverse Events in the Indicated Categories
NCT00731731 (5) [back to overview]Incidence of Adverse Events, Based on CTC (Common Toxicity Criteria) Severity Grade
NCT00731731 (5) [back to overview]Overall Survival at 15 Months (Phase II)
NCT00731731 (5) [back to overview]Time to Tumor Progression (Phase II)
NCT00731731 (5) [back to overview]Maximum Tolerated Dose of Vorinostat, Defined as the Dose at Which Fewer Than One-third of Patients Experience DLTs, Graded According to NCI CTCAE (Common Toxicity Criteria for Adverse Effects) Version 3.0 (Phase I)
NCT00731731 (5) [back to overview]Incidence of Adverse Events, as Per NCI CTCAE Version 3.0 (Phase II)
NCT00740636 (1) [back to overview]The Objective Overall Response
NCT00756106 (4) [back to overview]Permeability-surface Area Product Before, During, and After Chemoradiotherapy
NCT00756106 (4) [back to overview]Relative Cerebral Blood Volume as Measured by Perfusion-weighted MRI Before, During, and After Chemoradiotherapy
NCT00756106 (4) [back to overview]Relative Cerebral Blood Flow as Measured by Perfusion-weighted MRI Before, During, and After Chemoradiotherapy
NCT00756106 (4) [back to overview]Apparent Diffusion Coefficient Before, During, and After Chemoradiotherapy
NCT00761280 (12) [back to overview]Disease Progression at 10, 12, 14, 16, 18, 21, and 24 Months by Independent Review in the Intent-to-treat Population - Number of Participants
NCT00761280 (12) [back to overview]Survival Rate at 12, 18, and 21 Months in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Survival Rate at 24 Months in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Survival at 24 Months in the Intent-to-treat Population - Number of Participants
NCT00761280 (12) [back to overview]Tumor Control Rate (CR+PR+SD) by Independent Review in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Survival at 12, 18, and 21 Months in the Intent-to-treat Population - Number of Participants
NCT00761280 (12) [back to overview]Median Duration of Response (Days) by Independent Review (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Median Overall Survival (Days) From Randomization in the Intent-to-treat Population (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Median Time to Progression (Days) by Independent Review for the Intent-to-treat Population (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Response Category by Independent Review in the Intent-to-treat Population - Number of Participants
NCT00761280 (12) [back to overview]Overall Response Rate (CR+PR) by Independent Review in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)
NCT00761280 (12) [back to overview]Disease Progression Rate at 10, 12, 14, 16, 18, 21, and 24 Months by Independent Review in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)
NCT00782756 (2) [back to overview]Progression Free Survival
NCT00782756 (2) [back to overview]Number of Participants With Adverse Events
NCT00804908 (8) [back to overview]12-Month Overall Survival (OS) Rate
NCT00804908 (8) [back to overview]Disease Control Rate
NCT00804908 (8) [back to overview]Objective Response Rate
NCT00804908 (8) [back to overview]Overall Survival (OS): Time to Event
NCT00804908 (8) [back to overview]Progression-Free Survival (PFS): Time to Event
NCT00804908 (8) [back to overview]Time to Disease Progression
NCT00804908 (8) [back to overview]Time to Neurological/Brain Metastases Progression
NCT00804908 (8) [back to overview]6-month Progression-Free Survival Rate
NCT00805961 (4) [back to overview]Number of Participants Experiencing Toxicity After This Novel Multimodality Regimen
NCT00805961 (4) [back to overview]Objective Response Rate of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen
NCT00805961 (4) [back to overview]Overall Survival of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen
NCT00805961 (4) [back to overview]Progression-free Survival (PFS)
NCT00813943 (12) [back to overview]Overall Survival (OS) Time
NCT00813943 (12) [back to overview]Number of Participants With AEs Belonging to Standardized Medical Dictionary for Regulatory Activities (MedDRA) Queries (SMQs) Thromboembolic Events and Hemorrhage With NCI-CTC Toxicity Grade 3 or 4
NCT00813943 (12) [back to overview]Apparent Terminal Rate Constant
NCT00813943 (12) [back to overview]Apparent Volume of Distribution During the Terminal Phase (Vz) and Apparent Volume of Distribution at Steady State (Vss)
NCT00813943 (12) [back to overview]Area Under the Plasma Concentration-Time Curve From Time 0 to Infinity (AUC [0-infinity]) and Area Under the Plasma Concentration-Time Curve From Time 0 to 24 Hours (AUC [0-24])
NCT00813943 (12) [back to overview]Plasma Clearance (CL)
NCT00813943 (12) [back to overview]Maximum Observed Plasma Concentration (Cmax)
NCT00813943 (12) [back to overview]Progression Free Survival (PFS) Time - Investigator and Independent Read
NCT00813943 (12) [back to overview]Time to Maximum Plasma Concentration (Tmax) and Terminal Elimination Half-Life (t1/2)
NCT00813943 (12) [back to overview]Number of Participants With Adverse Events (AEs), Serious AEs, Treatment-Related AEs, Treatment-Related Serious AEs, AEs Leading to Death, Treatment-Related AEs Leading to Death, AEs of Grade 3 or 4 and Treatment-Related AEs of Grade 3 or 4
NCT00813943 (12) [back to overview]Mean Residence Time From Time 0 to Infinity (MRT [0-infinity])
NCT00813943 (12) [back to overview]Plasma Concentration at Pre-dose (Cpre) and Plasma Concentration at End of Infusion (CT)
NCT00826241 (3) [back to overview]Number of Participants With an Overall Response (Complete Response or Partial Response) Assessed by the MacDonald Criteria
NCT00826241 (3) [back to overview]Time to Progression
NCT00826241 (3) [back to overview]Number of Participants With Serious and Non-Serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0)
NCT00841555 (3) [back to overview]Time Spent in a Karnofsky Performance Status of 60-100%
NCT00841555 (3) [back to overview]Survival Time
NCT00841555 (3) [back to overview]Maximum Tolerated Dose(MTD)of Temozolomide(TMZ)
NCT00846430 (4) [back to overview]Response by MRI Measurements
NCT00846430 (4) [back to overview]No Reported Psychological Toxicity Based Upon Psychological Evaluations
NCT00846430 (4) [back to overview]Improvement of Symptoms and Pain
NCT00846430 (4) [back to overview]At Least 50% Shrinkage in Tumor Measurements by Physical Examination
NCT00869050 (2) [back to overview]Number of Participants With Complete Response (CR)
NCT00869050 (2) [back to overview]Number of Participants With Partial Response (PR)
NCT00869401 (4) [back to overview]Overall Survival
NCT00869401 (4) [back to overview]Objective Response
NCT00869401 (4) [back to overview]Progression-free Survival
NCT00869401 (4) [back to overview]The Number of Dose Limiting Toxicities(DLT) in Order to Determine Maximum Tolerable Dose(MTD) of Dasatinib Combined With Radiation and Temozolomide in This Patient Population.
NCT00876993 (3) [back to overview]Measurement of Number of Adverse Events
NCT00876993 (3) [back to overview]To Provide Safety and Efficacy Data for to Recommend Further Larger Studies.
NCT00876993 (3) [back to overview]2 Year Event Free Survival With Children Treated With This Regimen.
NCT00884741 (3) [back to overview]Progression-free Survival (PFS)
NCT00884741 (3) [back to overview]Overall Survival (OS)
NCT00884741 (3) [back to overview]Incidence of Grade 3 and Higher Treatment-related Toxicity as Assessed by the National Cancer Institute Common Terminology Criteria for Adverse Events (AEs) Version 3.0
NCT00885534 (1) [back to overview]Overall Response to CVT Chemotherapy.
NCT00905060 (4) [back to overview]Median Overall Survival
NCT00905060 (4) [back to overview]Median PD-L1 Positivity in Circulating Myeloid Cells
NCT00905060 (4) [back to overview]Median Progression Free Survival (PFS)
NCT00905060 (4) [back to overview]Number of Participants With Treatment-Related Adverse Events of Any Grade
NCT00925132 (2) [back to overview]Phase 2 -Number of Patients With a Decrease in Tumor Size Using RECIST and CHOI's Criteria
NCT00925132 (2) [back to overview]Phase I - Number of Participants With Dose Limiting Toxicities (DLTs) at a Given Dose Level
NCT00939991 (6) [back to overview]Phase II: Median Overall Survival (OS)
NCT00939991 (6) [back to overview]Phase II: Median Progression-free Survival (PFS)
NCT00939991 (6) [back to overview]Phase II: Radiographic Response.
NCT00939991 (6) [back to overview]Phase II: Number of Patients With Grade 2 or Greater, Treatment-related Toxicities
NCT00939991 (6) [back to overview]Phase I: Determination of the Maximum Tolerated Dose (MTD)
NCT00939991 (6) [back to overview]Phase II: 6-month Progression-free Survival (PFS)
NCT00943826 (7) [back to overview]Co-Primary: Overall Survival (OS)
NCT00943826 (7) [back to overview]Co-Primary: Progression-free Survival (PFS) as Assessed by Investigator
NCT00943826 (7) [back to overview]Kaplan-Meier (KM) Estimate of One Year Overall Survival
NCT00943826 (7) [back to overview]Kaplan-Meier (KM) Estimate of Two Year Overall Survival
NCT00943826 (7) [back to overview]PFS as Assessed by an Independent Review Facility
NCT00943826 (7) [back to overview]PFS in Participants With Stable/Improved Health Related Quality of Life (HRQoL) Based on European Organization for Research & Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) Core 30 (C30)(EORTC QLQ-C30) & EORTC QLQ Brain Neoplasm 20 (BN20)
NCT00943826 (7) [back to overview]Number of Participants With Non-Serious Adverse Events, Serious Adverse Events and Death
NCT00960063 (3) [back to overview]Best Overall Response Based on Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00960063 (3) [back to overview]Number of Participants Who Developed Anti-robatumumab Antibodies
NCT00960063 (3) [back to overview]Number of Participants With Dose Limiting Toxicities
NCT00967330 (12) [back to overview]Percentage of Participants Who Discontinued
NCT00967330 (12) [back to overview]Overall Survival (OS)
NCT00967330 (12) [back to overview]Change From Baseline for Mini-Mental Status Examination (MMSE) at Baseline, Post-Baseline (up to Month 30)
NCT00967330 (12) [back to overview]Percentage of Participants Achieving Progression-Free Survival (PFS) Without Disease Progression or Death at 6 Months
NCT00967330 (12) [back to overview]Percentage of Participants Who Received Corticosteroid for Glioblastoma
NCT00967330 (12) [back to overview]Number of Participants With A Best Overall Response (BOR) of Complete Response (CR) and With A BOR of CR or Partial Response (PR)
NCT00967330 (12) [back to overview]Change From Baseline in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ - C30) at Baseline, Post-Baseline (up to Month 30)
NCT00967330 (12) [back to overview]Progression-Free Survival (PFS)
NCT00967330 (12) [back to overview]Percentage of Participants With Response on FLAIR Imaging
NCT00967330 (12) [back to overview]Time to Treatment Failure
NCT00967330 (12) [back to overview]Change From Baseline for EORTC QLQ Brain Neoplasm 20 (BN20) at Baseline, Post-Baseline (up to Month 30)
NCT00967330 (12) [back to overview]Change From Baseline for Karnofsky Performance Status (KPS) Score at Baseline, Post-Baseline (up to Month 30)
NCT00977431 (5) [back to overview]Concentration of Afatinib in Plasma at Steady State Pre-dose on Days 8, 15 and 29
NCT00977431 (5) [back to overview]Number of Patients With Investigator Defined Dose Limiting Toxicities (DLT) During the RT Phase
NCT00977431 (5) [back to overview]The Objective Tumour Response According to the Macdonald Criteria
NCT00977431 (5) [back to overview]Maximum Tolerated Dose (MTD) of Afatinib
NCT00977431 (5) [back to overview]Incidence and Intensity of Adverse Events (AE) According to Common Terminology Criteria of Adverse Events (CTCAE v.3.0)
NCT00979017 (5) [back to overview]Response Rate
NCT00979017 (5) [back to overview]Incidence and Severity of Central Nervous System (CNS) Hemorrhage and Systemic Hemorrhage
NCT00979017 (5) [back to overview]Median Overall Survival (OS)
NCT00979017 (5) [back to overview]Median Progression-free Survival (PFS)
NCT00979017 (5) [back to overview]Incidence of Grade ≥ 4 Hematologic and ≥ Grade 3 Non-hematologic Toxicities
NCT00990652 (5) [back to overview]Number of Patients Surviving Without Disease Progression After 6 Months
NCT00990652 (5) [back to overview]Number of Participants Achieving a Response to Treatment (Either Complete or Partial Response) as Defined by MacDonald Criteria
NCT00990652 (5) [back to overview]Number of Grade 1, 2, 3, 4, and 5 Adverse Events Observed During Study Treatment (Defined by CTCAE v 3.0)
NCT00990652 (5) [back to overview]Overall Survival Rate at 6 Months
NCT00990652 (5) [back to overview]Overall Survival (in Days)
NCT00993044 (1) [back to overview]Dose Limiting Toxicity
NCT00998010 (5) [back to overview]Overall Survival
NCT00998010 (5) [back to overview]Survival at 1 Year
NCT00998010 (5) [back to overview]Toxicity Assessed According to the NCI Common Terminology Criteria for Adverse Events (CTCAE) Version 3.
NCT00998010 (5) [back to overview]Tumor Progression as Assessed by Magnetic Resonance Imaging (MRI) and Neurologic Exam
NCT00998010 (5) [back to overview]Time to Progression
NCT01004874 (6) [back to overview]One and Two Year Overall Survival
NCT01004874 (6) [back to overview]Median Overall Survival
NCT01004874 (6) [back to overview]6-month Progression-free Survival
NCT01004874 (6) [back to overview]Median Progression-free Survival
NCT01004874 (6) [back to overview]Number of Patients Experiencing a Greater Than or Equal to Grade 4 Hematologic or a Greater Than or Equal to Grade 3 Non-hematologic Toxicity
NCT01004874 (6) [back to overview]Number of Patients Experiencing a Central Nervous System (CNS) Hemorrhage or a Systemic Hemorrhage
NCT01009515 (4) [back to overview]Objective Response Rate (ORR)
NCT01009515 (4) [back to overview]Overall Survival
NCT01009515 (4) [back to overview]Time to Progression
NCT01009515 (4) [back to overview]Safety Profile
NCT01013285 (5) [back to overview]Median Overall Survival (OS) Based on the MGMT Promoter Methylation Status
NCT01013285 (5) [back to overview]Overall Survival
NCT01013285 (5) [back to overview]Radiographic Response (When Evaluable)
NCT01013285 (5) [back to overview]Time to Disease Progression
NCT01013285 (5) [back to overview]Progression-free Survival at 6 Months
NCT01015534 (4) [back to overview]Objective Response Rates. Assessed With Cranial MRI
NCT01015534 (4) [back to overview]Overall Survival
NCT01015534 (4) [back to overview]Survival Free of Brain Metastases Progression (PFS of BM)
NCT01015534 (4) [back to overview]Number of Grade 3-4 Adverse Events (AE) That Are Definitely or Probably Related to Both Groups of Treatment.
NCT01026493 (4) [back to overview]Phase 1: Maximum Tolerated Dose (MTD)
NCT01026493 (4) [back to overview]Phase II: 6-month Progression-free Survival (PFS) Rate for Patients With Measurable Disease After Surgery
NCT01026493 (4) [back to overview]Phase II: Objective Response (Partial and Complete Response) Rate for Patients With Measurable Disease After Surgery
NCT01026493 (4) [back to overview]Phase II: Overall Survival (OS)
NCT01044966 (5) [back to overview]Progression Free Survival
NCT01044966 (5) [back to overview]Proportion of Patients With Recurrent GBM Treated With ITV DepoCyt in Combination With Oral Temozolomide Who Are Progression-free at 16 Weeks.
NCT01044966 (5) [back to overview]Response Rate of Drug Treatment
NCT01044966 (5) [back to overview]Quality of Life Outcomes Measurement
NCT01044966 (5) [back to overview]Number of Participants With Treatment-related Adverse Events as Assessed by CTCAE v4.0
NCT01051596 (4) [back to overview]Percent of Patients With Disease Control
NCT01051596 (4) [back to overview]Percent of Patients With an Objective Response
NCT01051596 (4) [back to overview]Median Progression-free Survival Time
NCT01051596 (4) [back to overview]Overall Survival
NCT01055314 (5) [back to overview]Feasibility of the Addition of Cixutumumab to Chemotherapy Determined by Patient Enrollment
NCT01055314 (5) [back to overview]Event-Free Survival
NCT01055314 (5) [back to overview]Response Rate (CR + PR)
NCT01055314 (5) [back to overview]Incidence of Adverse Events Assessed by Common Terminology Criteria for Adverse Events Version 4.0
NCT01055314 (5) [back to overview]Feasibility of the Addition of Temozolomide to Chemotherapy Determined by Patient Enrollment
NCT01062399 (5) [back to overview]Phase II: Overall Survival (OS)
NCT01062399 (5) [back to overview]Phase I: Distribution of Worst Adverse Event Grade
NCT01062399 (5) [back to overview]Phase II: Distribution of Worst Adverse Event Grade
NCT01062399 (5) [back to overview]Phase I: Number of Patients With Dose-limiting Toxicity (DLT)
NCT01062399 (5) [back to overview]Phase II: Progression-free Survival (PFS)
NCT01062425 (4) [back to overview]6-month Progression-free Survival Rate
NCT01062425 (4) [back to overview]Overall Survival (OS)
NCT01062425 (4) [back to overview]Progression-free Survival (PFS)
NCT01062425 (4) [back to overview]Incidence of Grade 3+ Toxicities
NCT01105702 (3) [back to overview]Median Progression-Free Survival (PFS)
NCT01105702 (3) [back to overview]Median Overall Survival (OS)
NCT01105702 (3) [back to overview]Number of Patients With Grade 3 or 4 Adverse Events
NCT01110876 (1) [back to overview]Maximum Tolerated Dose (MTD) of Vorinostat in Combination With Escalating Doses of Erlotinib and Temozolomide
NCT01114555 (1) [back to overview]Number of Participants With Treatment Related Toxicity
NCT01115491 (6) [back to overview]Overall Survival - Percentage of Participants With an Event
NCT01115491 (6) [back to overview]Progression-Free Survival (PFS) - Percentage of Participants With an Event
NCT01115491 (6) [back to overview]PFS: Probability of Remaining Progression Free at 24 Weeks After Beginning the Study
NCT01115491 (6) [back to overview]PFS - Time to Event
NCT01115491 (6) [back to overview]Percentage of Participants Achieving an Overall Response of Complete Response (CR) or Partial Response (PR)
NCT01115491 (6) [back to overview]Overall Survival - Time to Event
NCT01119508 (1) [back to overview]6-Month Progression-Free Survival (PFS) Rate
NCT01120639 (10) [back to overview]Quality of Life by Brain-20 Survey
NCT01120639 (10) [back to overview]Quality of Life by European Organisation for Research and Treatment of Cancer (EORTC-QLQ C30) Survey
NCT01120639 (10) [back to overview]Quality of Life by MD Anderson Symptom Inventory - Brain Tumor (MDASI-BT) Survey
NCT01120639 (10) [back to overview]Overall Survival (OS)
NCT01120639 (10) [back to overview]Number of Acute Toxicity Within 30 Days
NCT01120639 (10) [back to overview]Long-term Toxicity After More Than 30 Days
NCT01120639 (10) [back to overview]Treatment Failure Analysis
NCT01120639 (10) [back to overview]Progression-free Survival
NCT01120639 (10) [back to overview]Number of Dose-limiting Toxicities (DLTs)
NCT01120639 (10) [back to overview]Percent of Participants With Radiographic Response
NCT01124734 (3) [back to overview]Duration of Response to High-Dose Interleukin-2 (H-D IL-2) Followed by Low Dose Temozolomide
NCT01124734 (3) [back to overview]Safety and Toxicity of H-D IL-2 Followed by Low Dose Temozolomide
NCT01124734 (3) [back to overview]Effect of High Dose IL2 Followed by Low Dose Temozolomide on Lymphocyte Subsets (Autoimmune Biomarkers)
NCT01143402 (2) [back to overview]Progression-free Survival (PFS) (Evaluable Randomized Patients)
NCT01143402 (2) [back to overview]Median Overall Survival (Evaluable Randomized Patients)
NCT01182350 (8) [back to overview]Grade 3-4 Post-Procedural Surgery-Related Toxicity Rate
NCT01182350 (8) [back to overview]9-month Overall Survival (OS) Rate
NCT01182350 (8) [back to overview]Feasibility Rate of Molecular Approach to Therapy
NCT01182350 (8) [back to overview]Rate of Lethal Complications From Surgery
NCT01182350 (8) [back to overview]Number of Participants With Grade 3-4 Treatment-Related Toxicity Over Chemoradiation Therapy
NCT01182350 (8) [back to overview]Median Progression Free Survival (PFS)
NCT01182350 (8) [back to overview]Delay in Radiation Therapy Start
NCT01182350 (8) [back to overview]9-month Overall Survival (OS) Rate by Molecular Cohort
NCT01186406 (4) [back to overview]Unacceptable Toxicity Related to the Treatment Regimen
NCT01186406 (4) [back to overview]Median Progression-free Survival
NCT01186406 (4) [back to overview]Median Overall Survival
NCT01186406 (4) [back to overview]21-month Overall Survival
NCT01196416 (9) [back to overview]Cycle 1 Mean Day 2 Trough/Pharmacokinetics of Gamma-secretase Inhibitor RO4929097 in Combination With Temozolomide (Phase IB)
NCT01196416 (9) [back to overview]Participants Evaluated for Toxicity
NCT01196416 (9) [back to overview]Progression-free Survival (Phase II)
NCT01196416 (9) [back to overview]Maximum Tolerated Dose for RO4929097
NCT01196416 (9) [back to overview]Number of Participants With Presence or Absence of Markers of Notch Signalling Pathway Inhibition in Patient Tumors (Phase Ib)
NCT01196416 (9) [back to overview]Overall Survival (Phase II)
NCT01196416 (9) [back to overview]Cycle 1 AUC/Pharmacokinetics of Gamma-secretase Inhibitor RO4929097 in Combination With Temozolomide (Phase IB)
NCT01196416 (9) [back to overview]Cycle 1 C Max/Pharmacokinetics of Gamma-secretase Inhibitor RO4929097 in Combination With Temozolomide (Phase IB)
NCT01196416 (9) [back to overview]Maximum-tolerated Dose for Cisplatin, Vinblastine and TMZ
NCT01205828 (4) [back to overview]Overall Survival
NCT01205828 (4) [back to overview]Clinical Benefit Rate
NCT01205828 (4) [back to overview]Progression Free Survival
NCT01205828 (4) [back to overview]Number of Participants Who Had Grade 3 or 4 Adverse Events
NCT01209442 (2) [back to overview]6-month Progression-free Survival
NCT01209442 (2) [back to overview]Overall Survival, Measured From the Day of Initial Diagnosis (Biopsy or Surgery) to the Time of Death From Any Cause.
NCT01217437 (3) [back to overview]Response
NCT01217437 (3) [back to overview]Event-free Survival
NCT01217437 (3) [back to overview]Overall Survival
NCT01235793 (2) [back to overview]One-year Progression-free Survival and Overall Survival
NCT01235793 (2) [back to overview]Safest Dose of Temozolomide for the DRBEAT Regimen
NCT01236560 (4) [back to overview]Event-free Survival
NCT01236560 (4) [back to overview]Cumulative Incidence of Disease Progression in Each Treatment Arm
NCT01236560 (4) [back to overview]Maximum Tolerated Dose (MTD) of Vorinostat
NCT01236560 (4) [back to overview]Overall Survival
NCT01284335 (8) [back to overview]Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)
NCT01284335 (8) [back to overview]Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)
NCT01284335 (8) [back to overview]Pharmacokinetic (PK): Concentration Maximum (Cmax)
NCT01284335 (8) [back to overview]Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)
NCT01284335 (8) [back to overview]PK: Area Under the Curve Albumin (AUCalb)
NCT01284335 (8) [back to overview]Number of Participants With Dose-Limiting Toxicities Cycle 1
NCT01284335 (8) [back to overview]Number of Participants With a Clinically Significant Effects
NCT01284335 (8) [back to overview]Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)
NCT01328535 (4) [back to overview]Toxicity, Assessed Using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 (v4)
NCT01328535 (4) [back to overview]Progression-Free Survival at 4 Months
NCT01328535 (4) [back to overview]Progression-Free Survival
NCT01328535 (4) [back to overview]Overall Survival
NCT01342757 (3) [back to overview]Measurable Change on Magnetic Resonance Spectroscopy Imaging After Vorinostat Administration
NCT01342757 (3) [back to overview]Proportion of Patients With Magnetic Resonance Spectroscopy (MRS) Response to Initial Vorinostat by MRI and MRS Scans as Determined by Spectroscopic Index
NCT01342757 (3) [back to overview]Mean Change in Metabolite Levels
NCT01390948 (16) [back to overview]Percentage of Participants With EFS as Determined by the CRRC at 1 Year
NCT01390948 (16) [back to overview]Health Status as Measured by the Health Utility Index (HUI)
NCT01390948 (16) [back to overview]Health Status as Measured by the Health Utility Index (HUI)
NCT01390948 (16) [back to overview]Number of Dose Administrations of TMZ and Bevacizumab in the Concurrent Phase
NCT01390948 (16) [back to overview]Percentage of Participants Who Completed >/= 90% of Planned Radiotherapy and TMZ Administrations
NCT01390948 (16) [back to overview]Percentage of Participants With a Treatment Delay or Discontinuation
NCT01390948 (16) [back to overview]Percentage of Participants With EFS as Determined by the CRRC at 6 Months
NCT01390948 (16) [back to overview]Concordance Between Structural Versus Multimodal Imaging for CRRC-Assessed Event-Free Survival
NCT01390948 (16) [back to overview]EFS as Assessed by the Investigator
NCT01390948 (16) [back to overview]Event-Free Survival (EFS) as Assessed by the Central Radiology Review Committee (CRRC)
NCT01390948 (16) [back to overview]Neurological Psychological Function as Measured by the Wechsler Scale
NCT01390948 (16) [back to overview]Number of Radiotherapy Dose Administrations in the Concurrent Phase
NCT01390948 (16) [back to overview]Objective Response Rate (ORR)
NCT01390948 (16) [back to overview]Overall Survival
NCT01390948 (16) [back to overview]Percentage of Participants With 1-Year Survival
NCT01390948 (16) [back to overview]Percentage of Participants With an Adverse Event (AE)
NCT01402063 (1) [back to overview]Progression Free Survival PPX/RT Versus TMZ/RT for Patients With GBM Without Methylation
NCT01465659 (7) [back to overview]Number of Patients Who Experience Toxicity Events Undergoing This Treatment.
NCT01465659 (7) [back to overview]Progression Free Survival (PFS)
NCT01465659 (7) [back to overview]Determine the Maximum Tolerated Dose (MTD) of Temozolomide in Combination With 400 mg Pazopanib in Patients With Advanced Pancreatic Neuroendocrine Tumor (PNET) in Phase I
NCT01465659 (7) [back to overview]Plasma Temozolomide Concentration in the Blood at Various Timepoints After Administration
NCT01465659 (7) [back to overview]Overall Survival (OS)
NCT01465659 (7) [back to overview]Overall Response Rate (ORR) in Patients With Advanced Neuroendocrine Tumors (PNET) Treated With Temozolomide and Pazopanib Combination Treatment at the RP2D in Phase II
NCT01465659 (7) [back to overview]Number of Patients Experiencing Response to Treatment or Stable Disease (Disease Control Rate)
NCT01466686 (4) [back to overview]Overall Survival Rate
NCT01466686 (4) [back to overview]Number of Patients With Neurologic Toxicity
NCT01466686 (4) [back to overview]Number of Patients With Hematologic Toxicities
NCT01466686 (4) [back to overview]Progression Free Survival Rate
NCT01478321 (6) [back to overview]Safety Profile for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab
NCT01478321 (6) [back to overview]Overall Survival (OS) for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab.
NCT01478321 (6) [back to overview]Median Progression Free Survival (PFS) for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab
NCT01478321 (6) [back to overview]Percentage of Patients With Progression Free Survival (PFS) at 6 Months and 12 Months
NCT01478321 (6) [back to overview]Patient Reported Quality of Life (QOL)
NCT01478321 (6) [back to overview]Overall Survival (OS) at 6 Months and 12 Months for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab.
NCT01505608 (3) [back to overview]Overall Response Rate (ORR) of Participants Using RECIST Criteria
NCT01505608 (3) [back to overview]Number of Participants With Adverse Events as a Measure of Safety and Tolerability
NCT01505608 (3) [back to overview]Progression Free Survival (PFS) of Participants Using Days Until Progression
NCT01506609 (5) [back to overview]Progression-Free Survival (PFS)
NCT01506609 (5) [back to overview]Clinical Benefit Rate (CBR) at Week 18
NCT01506609 (5) [back to overview]Overall Survival (OS)
NCT01506609 (5) [back to overview]Objective Response Rate (ORR)
NCT01506609 (5) [back to overview]Change From Baseline at Week 18 in European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Chemotherapy-Induced Peripheral Neuropathy Module (EORTC QLQ-CIPN20) Sensory Subscale Score
NCT01514201 (14) [back to overview]Overall Survival
NCT01514201 (14) [back to overview]Trough for Veliparib [Pharmacokinetic Parameter]
NCT01514201 (14) [back to overview]Levels of Urinary Biomarkers
NCT01514201 (14) [back to overview]Maximum Concentration of Veliparib (Cmax) on Days 1 and 4 (Measured in ng/mL) [Pharmacokinetic Parameter]
NCT01514201 (14) [back to overview]Percentage of Participants With Significant Changes in Poly(ADP-ribose) Polymerase (PARP) Levels Post-Veliparib, as Measured in Peripheral Blood Monocytes (PBMCs)
NCT01514201 (14) [back to overview]Percentage of Participants Observed to Have Unacceptable Toxicity During the Intra-patient Dose Escalation of Temozolomide During Maintenance Therapy (Feasibility Analysis Population)
NCT01514201 (14) [back to overview]Terminal Half-life (t1/2) for Veliparib [Pharmacokinetic Parameter]
NCT01514201 (14) [back to overview]Mean Apparent Clearance (CL/F) for Veliparib [Pharmacokinetic Parameter]
NCT01514201 (14) [back to overview]Progression-free Survival (PFS)
NCT01514201 (14) [back to overview]Percentage of Patients With Pseudo Progression
NCT01514201 (14) [back to overview]Apparent Volume of Distribution (Vd/F) for Veliparib [Pharmacokinetic Parameter]
NCT01514201 (14) [back to overview]Maximum Concentration of Veliparib (Cmax) on Day 1 (Measured in μM) [Pharmacokinetic Parameter]
NCT01514201 (14) [back to overview]Maximum-tolerated Dose of Veliparib Defined as Highest Dose Level With Fewer Than 2 Dose Limiting Toxicities in 6 Patients as Assessed by the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (Phase I)
NCT01514201 (14) [back to overview]Number of Phase I Patients Who Experienced Dose Limiting Toxicities (DLTs)
NCT01525082 (6) [back to overview]Treatment-related Toxicity
NCT01525082 (6) [back to overview]O6-methylguanine DNA Methyltransferase (MGMT) Status by Immunohistochemistry (IHC)
NCT01525082 (6) [back to overview]O6-methylguanine DNA Methyltransferase (MGMT) by Promoter Methylation
NCT01525082 (6) [back to overview]Progression-free Survival (PFS)
NCT01525082 (6) [back to overview]Overall Survival (OS)
NCT01525082 (6) [back to overview]Radiographic Response (RR)
NCT01550224 (9) [back to overview]Treatment Failure (TF)
NCT01550224 (9) [back to overview]Relapse-Free Survival (RFS) at 2 Years
NCT01550224 (9) [back to overview]Partial Remission (PR)
NCT01550224 (9) [back to overview]Overall Survival (OS) at 2 Years
NCT01550224 (9) [back to overview]Disease-free Survival (DFS) at 2 Years
NCT01550224 (9) [back to overview]Cytogenetic Response (CyR)
NCT01550224 (9) [back to overview]Morphologic Leukemia-free State (MLFS)
NCT01550224 (9) [back to overview]Complete Remission (CR)
NCT01550224 (9) [back to overview]Complete Remission With Incomplete Blood Count Recovery (CRp)
NCT01590082 (1) [back to overview]Overall Participant Response
NCT01601535 (13) [back to overview]Maximum Tolerated Dose (MTD) When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma
NCT01601535 (13) [back to overview]One Year Progression Free Survival Rate
NCT01601535 (13) [back to overview]Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Alisertib AUC
NCT01601535 (13) [back to overview]Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Irinotecan Clearance
NCT01601535 (13) [back to overview]AURKA Genotype
NCT01601535 (13) [back to overview]Aurora A Expression
NCT01601535 (13) [back to overview]Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Alisertib Day 4 Trough, Day 5 Trough and Cmax
NCT01601535 (13) [back to overview]Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Alisertib Tmax and Half-life
NCT01601535 (13) [back to overview]Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Irinotecan AUC, APC AUC, SN-38 AUC, and SN-38G AUC
NCT01601535 (13) [back to overview]Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Irinotecan Cmax, APC Cmax, SN-38 Cmax, and SN-38G Cmax
NCT01601535 (13) [back to overview]Response Rate for Patients With Relapsed or Refractory Neuroblastoma Treated With MLN8237, Irinotecan, and Temozolomide at the Identified MTD
NCT01601535 (13) [back to overview]UGT1A1 Genotype
NCT01601535 (13) [back to overview]Dose Limiting Toxicity (DLT) Data Associated With the Determination of the Recommended Phase 2 Dose
NCT01638546 (4) [back to overview]Number of Participants With Adverse Events
NCT01638546 (4) [back to overview]Overall Response (ORR) by RECIST 1.1 Criteria
NCT01638546 (4) [back to overview]Overall Survival
NCT01638546 (4) [back to overview]Progression-free Survival, Calculated as the Proportion of Patients Alive and Without Evidence of Disease
NCT01704287 (10) [back to overview]Best Overall Response (BOR) - Initial Treatment Period
NCT01704287 (10) [back to overview]Progression-free Survival (PFS) - Initial Treatment Period
NCT01704287 (10) [back to overview]Overall Response Rate (ORR) - Initial Treatment Period
NCT01704287 (10) [back to overview]Number of Participants Who Experienced an Adverse Event (AE) - Overall Study
NCT01704287 (10) [back to overview]Number of Participants Who Discontinued Study Drug Due to an Adverse Event (AE) - Overall Study
NCT01704287 (10) [back to overview]Interim Overall Survival (OS) - Initial Treatment Period
NCT01704287 (10) [back to overview]Final Overall Survival (OS) - Initial Treatment Period
NCT01704287 (10) [back to overview]Duration of Response (DOR) - Initial Treatment Period
NCT01704287 (10) [back to overview]Best Overall Response (BOR) - Switch-to-Pembrolizumab Treatment Period
NCT01704287 (10) [back to overview]Final Overall Survival (OS) By Programmed Cell Death-Ligand 1 (PD-L1) Tumor Expression Status - Initial Treatment Period
NCT01740258 (3) [back to overview]Progression-free Survival (PFS)
NCT01740258 (3) [back to overview]Overall Survival
NCT01740258 (3) [back to overview]Toxicity: Percentage of Subjects With Unacceptable Toxicities
NCT01767194 (2) [back to overview]Percentage of Randomized Patients Who Are Responders
NCT01767194 (2) [back to overview]Percentage of Patients in the Dinutuximab Arm Who Are Responders
NCT01781403 (4) [back to overview]Pathological Complete Response
NCT01781403 (4) [back to overview]Toxicity(Adeverse Event)
NCT01781403 (4) [back to overview]Recommended Dose (RD)
NCT01781403 (4) [back to overview]Maximum Tolerated Dose (MTD)
NCT01790503 (11) [back to overview]Summary of the Median Progression-free Survival (mPFS) in the Study Group Compared With Historical Control From Medical Literature
NCT01790503 (11) [back to overview]Number of Patients With Clinically Significant Abnormal Chemistry and Hematology Values Reported as Adverse Events by Preferred Term in Phase 1b of the Modified Intent-To-Treat Population
NCT01790503 (11) [back to overview]Summary of the Median Progression-free Survival (mPFS) by Subgroups in the Modified Intent-To-Treat Population on the Recommended Phase 2 Dose
NCT01790503 (11) [back to overview]Overview of Most Frequent System Organ Classes Reported in Phase 2 of the Modified Intent-To-Treat Population
NCT01790503 (11) [back to overview]Overview of Most Frequent System Organ Classes Reported in Phase 1b of the Modified Intent-To-Treat Population
NCT01790503 (11) [back to overview]Summary of the Median Progression-free Survival (mPFS) in the Combined 800 mg, 5 Days/Week Dose Group
NCT01790503 (11) [back to overview]Number of Patients With Clinically Significant Abnormal Chemistry and Hematology Values Reported as Adverse Events by Preferred Term in Phase 2, the Combined RP2D Groups in Phase 1b, and in the Study Overall Modified Intent-To-Treat Population
NCT01790503 (11) [back to overview]Summary of the Overall Survival in the Study Group Compared With Historical Control From Medical Literature
NCT01790503 (11) [back to overview]Summary of Best Overall Response by Subgroups in the Modified Intent-To-Treat Population on the Recommended Phase 2 Dose
NCT01790503 (11) [back to overview]Summary of the Overall Survival in The Study Population
NCT01790503 (11) [back to overview]Summary of the Overall Survival by Subgroups in the Modified Intent-To-Treat Population on the Recommended Phase 2 Dose
NCT01822275 (2) [back to overview]Ability to Calculate the Efficacy of Low-dose Whole Brain RT (WBRT) in Patients With GBM
NCT01822275 (2) [back to overview]Radiographic (CT-MRI) Response Assessment and Treatment Failure Patterns of Patients With GBM
NCT01824875 (3) [back to overview]Proportion of Patients With Response
NCT01824875 (3) [back to overview]Progression-free Survival
NCT01824875 (3) [back to overview]Overall Survival
NCT01827384 (3) [back to overview]Number of Participants With an Objective Response
NCT01827384 (3) [back to overview]Proportion of Participants With 4 Month Progression-free Survival (PFS)
NCT01827384 (3) [back to overview]Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v5.0)
NCT01835145 (5) [back to overview]Proportion of Patients Without a Progression Free Survival Event at 4 Months (PFS4)
NCT01835145 (5) [back to overview]Percentage of Patients Who Experienced Grade 3+ Adverse Events Regardless of Attribution
NCT01835145 (5) [back to overview]Overall Survival (OS)
NCT01835145 (5) [back to overview]Confirmed Response Rate as Determined by the RECIST Criteria (Version 1.1)
NCT01835145 (5) [back to overview]PFS
NCT01946529 (1) [back to overview]Response to Window Therapy (2 Courses) for Group B (High-risk) - ESFT Participants
NCT01977677 (2) [back to overview]Participants Alive and Without Disease Progression At 6 Months After the Start of the Irradiation
NCT01977677 (2) [back to overview]Dose-limiting Toxicity
NCT01991977 (1) [back to overview]Proportion of Grade IV MGMT Un-methylated Patients That Experience Confirmed-progression-free Survival at 6 Months (CPFS6)
NCT02023905 (6) [back to overview]Progression-Free Survival Rate (PFS) (Arm 3)
NCT02023905 (6) [back to overview]Progression-Free Survival Rate (PFS) (Arms 1 and 2)
NCT02023905 (6) [back to overview]Rate of Reduction in Seizures
NCT02023905 (6) [back to overview]Median Progression Free Survival (PFS)
NCT02023905 (6) [back to overview]Objective Response Rate (ORR)
NCT02023905 (6) [back to overview]Overall Survival Rate (OS)
NCT02039778 (2) [back to overview]Number of Participants With Adverse Events as a Measure of Safety and Tolerability
NCT02039778 (2) [back to overview]Overall Survival
NCT02113007 (1) [back to overview]Number of Participants With Serious and Non-serious Adverse Events as a Measure of Safety.
NCT02116777 (18) [back to overview]C Max of Talazoparib
NCT02116777 (18) [back to overview]C Max of Talazoparib in Combination With Temozolomide
NCT02116777 (18) [back to overview]C Max of Temozolomide in Combination With Talazoparib
NCT02116777 (18) [back to overview]Clearance of Temozolomide in Combination With Talazoparib
NCT02116777 (18) [back to overview]Half-life of Temozolomide in Combination With Talazoparib
NCT02116777 (18) [back to overview]Number of Ewing/Peripheral PNET Participants in Phase 2 With Complete Response (CR) or Partial Response (PR)
NCT02116777 (18) [back to overview]Number of Solid Tumor Patients With Complete Response (CR) or Partial Response (PR)
NCT02116777 (18) [back to overview]T Max of Talazoparib
NCT02116777 (18) [back to overview]T Max of Talazoparib in Combination With Temozolomide
NCT02116777 (18) [back to overview]T Max of Temozolomide in Combination With Talazoparib
NCT02116777 (18) [back to overview]The Number of Participants With Dose Limiting Toxicities to Determine the Maximum Tolerated Dose of Temozolomide and Talazoparib Combination Therapy
NCT02116777 (18) [back to overview]Clearance of Talazoparib in Combination With Temozolomide
NCT02116777 (18) [back to overview]AUC of Talazoparib in Combination With Temozolomide
NCT02116777 (18) [back to overview]Accumulation Half-life of Talazoparib in Combination With Temozolomide.
NCT02116777 (18) [back to overview]Accumulation Ratio of Talazoparib in Combination With Temozolomide
NCT02116777 (18) [back to overview]All Cycle 1 Toxicities >=Grade 3
NCT02116777 (18) [back to overview]AUC of Talazoparib
NCT02116777 (18) [back to overview]AUC of Temozolomide in Combination With Talazoparib
NCT02152982 (5) [back to overview]Overall Survival (OS)
NCT02152982 (5) [back to overview]Objective Tumor Response
NCT02152982 (5) [back to overview]Interaction With Optune Device
NCT02152982 (5) [back to overview]Progression-free Survival (PFS)
NCT02152982 (5) [back to overview]Overall Adverse Event Rates for Grade 3 or Higher Adverse Events
NCT02193347 (2) [back to overview]Percentage of Participants With an Unacceptable Toxicity
NCT02193347 (2) [back to overview]Percentage of Patients With a Positive Vaccine Response After 3 Post-Surgery Vaccines as Measured by IFNγ ELIspot
NCT02209948 (6) [back to overview]Median Overall Survival (OS) by Arm and MGMT Methylation Status
NCT02209948 (6) [back to overview]Progresion Free Survival Median Values
NCT02209948 (6) [back to overview]Overall Survival
NCT02209948 (6) [back to overview]Progression Free Survival at 6 Month
NCT02209948 (6) [back to overview]Number of Participants With Adverse Effects
NCT02209948 (6) [back to overview]Median Progression-free Survival (PFS) by Arm and MGMT Methylation Status
NCT02231762 (18) [back to overview]DCR After 12 Months
NCT02231762 (18) [back to overview]Disease Control Rate (DCR) After 6 Months
NCT02231762 (18) [back to overview]Duration of Response (DoR) Within 12 Months
NCT02231762 (18) [back to overview]Progression-Free Survival (PFS) Within 12 Months
NCT02231762 (18) [back to overview]Time To Response (TtR) Within 12 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Symptomatic Response After 12 Months - Maintenance Phase
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using CgA Levels After 12 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using 5-Hydroxy-Indol-Amino-Acid (HIAA) Levels After 6 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Symptomatic Response After 6 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using 5-HIAA Levels After 12 Months
NCT02231762 (18) [back to overview]DCR by O6-methylguanine-DNA Methyl-transferase (MGMT) Expression and Methylation and Somatostatin Receptor (SSTR) Expression After 6 Months
NCT02231762 (18) [back to overview]Quality of Life Gastrointestinal Neuroendocrine Tumour 21 Questionnaire (QLQ-GI.NET21): Mean Change From Baseline at 6 Months
NCT02231762 (18) [back to overview]QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months
NCT02231762 (18) [back to overview]Pharmacokinetic (PK) Results: Lanreotide ATG 120 mg Serum Concentrations Within 12 Months
NCT02231762 (18) [back to overview]European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QoL) Core 30 Questionnaire (QLQ-C30): Mean Change From Baseline at 6 Months
NCT02231762 (18) [back to overview]EORTC QoL Questionnaire QLQ-C30: Mean Change From Baseline at 12 Months
NCT02231762 (18) [back to overview]QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months
NCT02231762 (18) [back to overview]The Number of Subjects With a Biochemical Response Using Chromogranin-A (CgA) Levels After 6 Months
NCT02288897 (6) [back to overview]Number of Participants With Adverse Events
NCT02288897 (6) [back to overview]Overall Survival (OS)
NCT02288897 (6) [back to overview]Complete Response Rate (CRR)
NCT02288897 (6) [back to overview]Progression-free Survival (PFS)
NCT02288897 (6) [back to overview]Change in Domain Scores From Baseline Using the Patient Reported Skindex-16 Instrument
NCT02288897 (6) [back to overview]Duration of Complete Response (DCR)
NCT02315534 (7) [back to overview]Pharmacokinetic Profile of BBI608 and Temozolomide When Administered in Combination With Temozolomide as Assessed by the Area Under the Curve
NCT02315534 (7) [back to overview]Dose-limiting Toxicities (DLTs)
NCT02315534 (7) [back to overview]Progression Free Survival (PFS)-6
NCT02315534 (7) [back to overview]Overall Survival (OS)
NCT02315534 (7) [back to overview]Disease Control Rate (DCR)
NCT02315534 (7) [back to overview]Progression Free Survival (PFS)-12
NCT02315534 (7) [back to overview]Overall Response Rate (ORR)
NCT02340156 (1) [back to overview]Incidence of Treatment-Emergent Adverse Events
NCT02343081 (5) [back to overview]AUC0-∞
NCT02343081 (5) [back to overview]Kel
NCT02343081 (5) [back to overview]AUC0-t
NCT02343081 (5) [back to overview]T1/2
NCT02343081 (5) [back to overview]Cmax
NCT02343406 (11) [back to overview]Pediatric Study: Half-life (t1/2) Observed for ABT-414
NCT02343406 (11) [back to overview]Pediatric Study: Area Under the Concentration-time-curve (AUC) Observed for Unconjugated Cys-mcMMAF
NCT02343406 (11) [back to overview]Pediatric Study: Area Under the Concentration-time Curve (AUC) Observed for ABT-414
NCT02343406 (11) [back to overview]Adult Study: Objective Response Rate (ORR)
NCT02343406 (11) [back to overview]Adult Study: Progression-Free Survival (PFS)
NCT02343406 (11) [back to overview]Adult Study: Overall Survival in the Subgroup With Epidermal Growth Factor Receptor (EGFRvIII) Mutation
NCT02343406 (11) [back to overview]Pediatric Study: Percentage of Participants With Adverse Events From the First Visit Until 49 Days After the Last Dose of Study Drug
NCT02343406 (11) [back to overview]Adult Study: Overall Survival (OS)
NCT02343406 (11) [back to overview]Pediatric Study: Maximum Observed Serum Concentration (Cmax) of ABT-414
NCT02343406 (11) [back to overview]Pediatric Study: Maximum Observed Plasma Concentration (Cmax) of Cys-mcMMAF
NCT02343406 (11) [back to overview]Pediatric Study: Half-life (t1/2) Observed for Cys-mcMMAF
NCT02343549 (7) [back to overview]Overall Survival (OS)
NCT02343549 (7) [back to overview]Progression Free Survival (PFS)
NCT02343549 (7) [back to overview]Duration of Response
NCT02343549 (7) [back to overview]Number of Participants With 12-Month Survival
NCT02343549 (7) [back to overview]Duration of Disease Control
NCT02343549 (7) [back to overview]Number of Participants With Disease Control
NCT02343549 (7) [back to overview]Number of Participants With Objective Response
NCT02366728 (7) [back to overview]Median Overall Survival in CMV Negative Participants
NCT02366728 (7) [back to overview]Median Overall Survival
NCT02366728 (7) [back to overview]Percentage of 111In-labeled Dendritic Cells Migrating to the Inguinal Lymph Nodes
NCT02366728 (7) [back to overview]Median Progression-free Survival in CMV Positive Participants
NCT02366728 (7) [back to overview]Median Progression-free Survival in CMV Negative Participants
NCT02366728 (7) [back to overview]Median Progression-free Survival
NCT02366728 (7) [back to overview]Median Overall Survival in CMV Positive Participants
NCT02389738 (1) [back to overview]Change in AUC0-18 of the Temozolomide Concentration (AUC-T) in Brain Interstitium Before and After Regadenoson Infusion
NCT02395692 (5) [back to overview]Progression-free Survival at 6 Months
NCT02395692 (5) [back to overview]Toxicity as Assessed by Number of Participants Who Experienced Adverse Events
NCT02395692 (5) [back to overview]Progression-free Survival
NCT02395692 (5) [back to overview]Overall Survival
NCT02395692 (5) [back to overview]Objective Response as Assessed by Response Assessment in Neuro-Oncology (RANO) Criteria (Arm 1 and Arm 2)
NCT02455557 (2) [back to overview]Overall Survival
NCT02455557 (2) [back to overview]Progression-free Survival (PFS)
NCT02511132 (3) [back to overview]Progression Free Survival
NCT02511132 (3) [back to overview]Overall Survival
NCT02511132 (3) [back to overview]Number of Participants With Adverse Events Determined by Laboratory Assessments and Physical Examinations
NCT02573324 (9) [back to overview]Deterioration Free Survival in M.D. Anderson Symptom Inventory Brain Tumor Module (MDASI-BT) Symptom Severity Score
NCT02573324 (9) [back to overview]Progression-Free Survival (PFS)
NCT02573324 (9) [back to overview]PFS for EGFRvIII-Mutated Tumor Subgroup
NCT02573324 (9) [back to overview]Overall Survival (OS)
NCT02573324 (9) [back to overview]OS for the O6-methylguaninemethlytransferese (MGMT) Unmethylated Group
NCT02573324 (9) [back to overview]OS for the MGMT Methylated Group
NCT02573324 (9) [back to overview]OS for the Epidermal Growth Factor Receptor (EGFR)vIII-Mutated Tumor Subgroup
NCT02573324 (9) [back to overview]Deterioration Free Survival in Neurocognitive Functioning on the Hopkins Verbal Learning Test Revised (HVLT-R) Total Recall Score
NCT02573324 (9) [back to overview]Deterioration Free Survival in MDASI-BT Symptom Interference Score
NCT02617589 (4) [back to overview]Kaplan-Meier Plot of Overall Survival (OS) - Extended Collection
NCT02617589 (4) [back to overview]Kaplan-Meier Plot of Progression Free Survival
NCT02617589 (4) [back to overview]Overall Survival Rate at 24 Months
NCT02617589 (4) [back to overview]Overall Survival (OS)
NCT02661282 (8) [back to overview]Progression Free Survival (PFS) (Newly Diagnosed Glioblastoma Cohort)- Phase II
NCT02661282 (8) [back to overview]Progression Free Survival (PFS) (Recurrent Glioblastoma Cohort) at 6 Months- Phase II
NCT02661282 (8) [back to overview]Time to Progression (Recurrent Glioblastoma Cohort)- Phase II
NCT02661282 (8) [back to overview]Overall Survival (OS) (Newly Diagnosed Glioblastoma Cohort)- Phase II
NCT02661282 (8) [back to overview]Maximum Tolerated Dose (MTD) (Recurrent Glioblastoma Participant Cohort)- Phase I
NCT02661282 (8) [back to overview]Median Duration of Response (Newly Diagnosed Glioblastoma Cohort)- Phase II
NCT02661282 (8) [back to overview]Number of Participants With Immunological Effects in Tumor Tissue (Recurrent Glioblastoma Cohort)- Phase II
NCT02661282 (8) [back to overview]Overall Objective Response Rate (ORR) (Newly Diagnosed Glioblastoma Cohort)- Phase II
NCT02667587 (5) [back to overview]Progression-free Survival (PFS) Determined by BICR
NCT02667587 (5) [back to overview]Progression Free Survival (PFS) Based on Investigator Assessment
NCT02667587 (5) [back to overview]Overall Survival (OS) Rates at 12 Months
NCT02667587 (5) [back to overview]Overall Survival (OS)
NCT02667587 (5) [back to overview]Overall Survival (OS) Rates at 24 Months
NCT02698410 (15) [back to overview]Median Time to Progression (TTP) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]Median Progression Free Survival (PFS) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]DCR Assessed Centrally at Month 9
NCT02698410 (15) [back to overview]Median Time to Response (TTR) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]Neuron-Specific Enolase (NSE) and CgA Biomarker Levels
NCT02698410 (15) [back to overview]Percentage of Biochemical Responders According to Chromogranin A (CgA) Plasma Levels
NCT02698410 (15) [back to overview]Best Overall Response (BOR) Assessed Locally and Centrally
NCT02698410 (15) [back to overview]DCR Assessed Locally and Centrally at Month 12
NCT02698410 (15) [back to overview]Disease Control Rate (DCR) Assessed Locally at Month 9
NCT02698410 (15) [back to overview]DCR Assessed Locally and Centrally at Month 9 by Carcinoid Type
NCT02698410 (15) [back to overview]Coefficient of Agreement Between Central and Local Assessment of Tumor Radiological Response at Month 9
NCT02698410 (15) [back to overview]Objective Response Rate (ORR) Assessed Locally and Centrally at Months 9 and 12
NCT02698410 (15) [back to overview]Influence of Biomarkers Expression on Locally and Centrally Assessed DCR at Months 9 and 12
NCT02698410 (15) [back to overview]Influence of Biomarkers Expression on Locally and Centrally Assessed PFS
NCT02698410 (15) [back to overview]Median Duration of Response (DOR) Assessed Locally and Centrally
NCT02805179 (6) [back to overview]Median Change in Tumor Volume From Baseline to Mid-radiation Treatment (Week 4)
NCT02805179 (6) [back to overview]Median Progression-free Survival
NCT02805179 (6) [back to overview]Percentage of Patients With Failure; Central or In-field vs. Marginal or Distant
NCT02805179 (6) [back to overview]Percentage of Patients That Experienced Deterioration in Quality of Life (QOL)
NCT02805179 (6) [back to overview]Overall Survival at 12 Months
NCT02805179 (6) [back to overview]Median Overall Survival
NCT02864368 (2) [back to overview]Immunologic Response as Measured by Peak Number of T Cells That Secrete IFNγ by ELISPOT in Response to Component A of PEP-CMV
NCT02864368 (2) [back to overview]Percentage of Participants With Treatment-related Adverse Events
NCT02942264 (6) [back to overview]% of Participants Free of Disease Progression at 4mos Treated w/Zotiraciclib at the Maximum Tolerated Dose in Combination Temozolomide w/Dose Dense Temozolomide Schedules in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma
NCT02942264 (6) [back to overview]Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0)
NCT02942264 (6) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Zotiraciclib (TG02) in Combination of Metronomic (mn) Temozolomide (TMZ) in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma
NCT02942264 (6) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Zotiraciclib in Combination With Dose Dense (dd) Temozolomide (TMZ) in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma
NCT02942264 (6) [back to overview]Phase I: Number of Participants With a Dose-limiting Toxicity (DLT)
NCT02942264 (6) [back to overview]% of Participants Free of Disease Progression at 4mos Treated w/Zotiraciclib at the Maximum Tolerated Dose (MTD) in Combination With the Metronomic (mn) Temozolomide (TMZ) in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma
NCT02975882 (4) [back to overview]Area Under the Serum of Nanoparticle Albumin-bound Rapamycin Concentration Curve
NCT02975882 (4) [back to overview]Number of Patients With Cycle 1 and 2 Dose Limiting Toxicities Attributable to Nanoparticle Albumin-bound Rapamycin
NCT02975882 (4) [back to overview]Number of Patients With Adverse Events
NCT02975882 (4) [back to overview]Number of Patients With Antitumor Activity of Nanoparticle Albumin-bound Rapamycin
NCT03034135 (6) [back to overview]Objective Response Rate
NCT03034135 (6) [back to overview]Overall Survival
NCT03034135 (6) [back to overview]Median Duration of Overall Survival
NCT03034135 (6) [back to overview]Progression Free Survival
NCT03034135 (6) [back to overview]Median Progression Free Survival
NCT03034135 (6) [back to overview]Number of Participants With Serious Adverse Events
NCT03119064 (3) [back to overview]Toxicities
NCT03119064 (3) [back to overview]Response
NCT03119064 (3) [back to overview]Determination of Maximum Tolerated Dose (MTD)
NCT03168919 (4) [back to overview]Time to Progression
NCT03168919 (4) [back to overview]Changes of Tumor Angiogenesis
NCT03168919 (4) [back to overview]Changes of Tumor Cellularity
NCT03168919 (4) [back to overview]Changes of Tumor Volume
NCT03190967 (6) [back to overview]Phase I: Number of Participants With Serious and/or Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0)
NCT03190967 (6) [back to overview]Phase I: Number of Participants With Dose Limiting Toxicity (DLTs) at Each Dose Level
NCT03190967 (6) [back to overview]Phase I: Standard Time to Progression (TTP)
NCT03190967 (6) [back to overview]Phase I: Median Survival
NCT03190967 (6) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Temozolomide (TMZ) When Used With T-DM1 (Ado-trastuzumab)
NCT03190967 (6) [back to overview]Phase I: Number of Participants With Grade 3 and/or Grade 4 Adverse Events
NCT03323034 (8) [back to overview]Number of Participants With Grade 3 or Greater Toxicities Associated With Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]Anti-tumor Activity of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]AUC of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]C Max of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]Clearance of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]Half-life of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]MTD/RP2D of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]T Max of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03363659 (2) [back to overview]6 Months Progression Free Survival (PFS)
NCT03363659 (2) [back to overview]Overall Survival
NCT03419403 (10) [back to overview]Number of Participants With Depatuxizumab Mafodotin Dose Modifications to Ocular Side Effects After Bandage Contact Lens (BCL) Intervention
NCT03419403 (10) [back to overview]Percentage of Participants Who Required a Change in Ocular Side Effect (OSE) Management
NCT03419403 (10) [back to overview]Maximum Change From Baseline on the Logarithm of the Minimum Angle of Resolution (LogMAR) Scale
NCT03419403 (10) [back to overview]Change From Baseline In Logarithm of the Minimum Angle of Resolution (LogMAR) Scale After Bandage Contact Lens (BCL) Intervention
NCT03419403 (10) [back to overview]Time to Bandage Contact Lens (BCL) Intervention
NCT03419403 (10) [back to overview]Cumulative Dose of Depatuxizumab Mafodotin Received During Chemoradiation and During Adjuvant Treatment
NCT03419403 (10) [back to overview]Treatment-Emergent Corneal Epithelial Adverse Event (CEAE) Grade at Each Visit
NCT03419403 (10) [back to overview]Treatment Emergent Corneal Epithelial Adverse Event (CEAE) Grade at Each Visit After Bandage Contact Lens (BCL) Intervention
NCT03419403 (10) [back to overview]Number of Participants With Depatuxizumab Mafodotin Dose Modifications Due to Ocular Side Effects (OSE)
NCT03419403 (10) [back to overview]Change From Baseline In Logarithm of the Minimum Angle of Resolution (LogMAR) Scale After Bandage Contact Lens (BCL) Intervention
NCT03463265 (5) [back to overview]PFS Rate at 6 Months and 12 Months
NCT03463265 (5) [back to overview]OS at 12 Months
NCT03463265 (5) [back to overview]OS
NCT03463265 (5) [back to overview]ORR
NCT03463265 (5) [back to overview]Median PFS
NCT03495921 (4) [back to overview]Overall Response Rate (ORR)
NCT03495921 (4) [back to overview]Overall Survival (OS)
NCT03495921 (4) [back to overview]Vigil Manufacture Success Rate: Number of Participants Eligible for Treatment on the Main Study.
NCT03495921 (4) [back to overview]Progression Free Survival (PFS)
NCT03778294 (2) [back to overview]Overall Survival (OS)
NCT03778294 (2) [back to overview]Progression Free Survival
NCT03880019 (2) [back to overview]Number of Patients Experiencing Adverse Events
NCT03880019 (2) [back to overview]Confirmed Objective Response Rate (ORR) (Complete Response + Partial Response)
NCT03930771 (4) [back to overview]Number of Subjects With Radiographic Response, as Defined by the RECIST Criteria.
NCT03930771 (4) [back to overview]Safety, as Measured by the Number of Subjects With at Least One AE
NCT03930771 (4) [back to overview]Tolerability of the TMZ and Capecitabine Combination, as Measured by Number of Participants With a Dose-limiting Toxicity
NCT03930771 (4) [back to overview]Effect of the CAP and TMZ Combination on Pituitary Function, Measured by Changes in Pituitary Hormone Secretion in Patients
NCT04166435 (3) [back to overview]Progression Free Survival
NCT04166435 (3) [back to overview]Count of Participants With Adverse Events Greater or Equal to 3
NCT04166435 (3) [back to overview]Overall Survival
NCT05718466 (3) [back to overview]Overall Survival
NCT05718466 (3) [back to overview]Local Tumor Control
NCT05718466 (3) [back to overview]Progression Free Survival

Radiographic Response Assessed by Macdonald Criteria Every 2 Months

All patients will have their tumor measurements recorded at baseline and at the time of each MRI scan. Lesions must be measured in two dimensions. (NCT00026494)
Timeframe: 2 years

,,,
Interventionparticipants (Number)
Stable Disease (SD)Progression of Disease (POD)Partial Response (PR)Minor Response (MR)Complete Response (CR)
15mg/m2 - Vinorelbine32000
20mg/m2 - Vinorelbine21100
25mg/m2 - Vinorelbine411010
30mg/m2 - Vinorelbine312001

[back to top]

Maximal Tolerating Dose (MTD for Phase I)

"Phase I Dose limiting toxicity evaluation at end of first cycle based on blood tests every two weeks and participants' subjective and objective symptoms.~Start Dose Level 100 mg/m² Temozolomide once daily + 400 mg ZARNESTRA twice daily; Dose Level 1 100 mg/m² Temozolomide once daily + 500 mg ZARNESTRA twice daily; Dose Level 2 150 mg/m² Temozolomide once daily + 500 mg ZARNESTRA twice daily; Dose Level 3 150 mg/m² Temozolomide once daily + 600 mg ZARNESTRA twice daily; Dose Level 4 150 mg/m² Temozolomide once daily + 800 mg ZARNESTRA twice daily" (NCT00050986)
Timeframe: End of first cycle (4 weeks) evaluation

Interventionparticipants (Number)
Dose Level 1Dose Level 2Dose Level 3Dose Level 4
Temozolomide and R1157773363

[back to top]

Phase II: Pre-irradiation Chemotherapy Tumor Response Rate (Including Phase I Patients at Same Dose)

Tumor response was centrally reviewed. Complete response: Disappearance of all enhancing tumor, the patient must be off steroid therapy and neurologically stable or improved; partial response: ≥ 50% decrease in enhancing tumor; progressive disease: ≥ 25% increase in a lesion, progressive or newly emergent meningeal or ocular disease. (Please note that this outcome measure is considered a secondary endpoint for the Phase II component of the study, but that the patients from Phase I that were treated at the same dose level are included, as indicated in the treatment arm descriptions. ) (NCT00068250)
Timeframe: From start of treatment to 10 weeks if RT received, to 15 weeks if not.

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseProgressive DiseaseNot evaluable
Combined Temozolomide 100mg Arms181223

[back to top]

Number of Phase I Participants Experiencing Toxicity

A dose limiting toxicity (DLT) is defined as any grade 3 or 4 non-hematological toxicity (other than grade 3 nausea/vomiting) or any hematological toxicity resulting in the discontinuation of temozolomide. Toxicity evaluation for this dose escalation includes all toxicities occurring prior to the start of radiation therapy. If the patient did not receive radiation therapy, then toxicity evaluation included all toxicities occurring through week 15. Any grade 5 toxicity would result in immediate suspension of accrual. (NCT00068250)
Timeframe: From start of treatment to 10 weeks if radiation therapy received, to 15 weeks if not.

InterventionParticipants (Count of Participants)
Phase I: Temozolomide 100mg1
Phase I: Temozolomide150 mg3

[back to top]

Phase II: Overall Survival Rate at 2 Years (Including Phase I Patients at Same Dose)

Survival time is defined as time from registration to date of death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. (Please note that this outcome measure is considered the primary endpoint for the Phase II component of the study, but that the patients from Phase I that were treated at the same dose level are included, as indicated in the treatment arm descriptions. ) (NCT00068250)
Timeframe: Analysis occured after all patients have been on study for 2 years. Maximum follow-up at time of analysis was 8.5 years.

Interventionpercentage of participants (Number)
Combined Temozolomide 100mg Arms80.8

[back to top]

Phase II: Progression-free Survival (Including Phase I Patients at Same Dose)

Progression is defined as greater than 25% increase in enhancing tumor or the appearance of new lesions in the brain, eye, or the appearance of a new positive cerebrospinal fluid (CSF) cytology. The patient may be neurologically stable or worse and on stable or increasing doses of corticosteroid. Progression-free survival time is defined as time from registration to the date of progression, death, or last known follow-up (censored). Progression-free survival rates are estimated using the Kaplan-Meier method. (Please note that this outcome measure is considered a secondary endpoint for the Phase II component of the study, but that the patients from Phase I that were treated at the same dose level are included, as indicated in the treatment arm descriptions. ) (NCT00068250)
Timeframe: Analysis occured after all patients have been on study for 2 years. Maximum follow-up at time of analysis was 8.5 years.

Interventionyears (Median)
Combined Temozolomide 100mg Arms5.4

[back to top]

Number of Participants Who Experienced a Grade 3 or 4 Thrombocytopenia and/or Neutropenia.

Occurence of grade 3 or 4 thrombocytopenia or neutropenia while receiving protocol therapy. (NCT00077207)
Timeframe: Up to 18 months of protocol therapy

InterventionParticipants (Count of Participants)
Carboplatin, Vincristine Sulfate, Temozolomide)43

[back to top]

Number of Participants Who Experienced Toxic Death

Primary safety endpoints are (1) the occurrence of toxic death, which is death during treatment that is not primarily attributable to disease progression, and (2) the occurrence of grade 4 allergy to carboplatin. (NCT00077207)
Timeframe: Up to 6 years after the start of protocol therapy

InterventionParticipants (Count of Participants)
Carboplatin, Vincristine Sulfate, Temozolomide)0

[back to top]

Percent Probability of Progression-free Survival (PFS)

Percentage probability of being alive and without the occurrence of disease progression 3 years following enrollment. (NCT00077207)
Timeframe: 3 years

InterventionPercent probability PFS (Number)
Carboplatin, Vincristine Sulfate, Temozolomide)60.59

[back to top]

Percentage Probability of Event-free Survival (EFS)

Percentage probability of being alive and without the occurrence of disease progression or second malignant neoplasm 6 years following enrollment. (NCT00077207)
Timeframe: Six years

Interventionpercent probability EFS (Number)
Carboplatin, Vincristine Sulfate, Temozolomide)40.89

[back to top]

Short Term Feasibility Success

"Success is defined as the completion of induction plus one cycle of maintenance within 24 weeks of enrollment without more than a 25% reduction in either carboplatin or temozolomide dosage.~Failure to complete the induction and one cycle of maintenance within 24 weeks counts as a short-term-feasibility failure." (NCT00077207)
Timeframe: 24 weeks

Interventionparticipants (Number)
Treatment (Carboplatin, Vincristine Sulfate, Temozolomide)25

[back to top]

Long Term Feasibility Success

"Success is defined as the completion of induction plus four cycles of maintenance within 60 weeks of enrollment without more than a 25% reduction in either carboplatin or temozolomide dosage.~If the participant completes all therapy within 60 weeks the patient is a long-term feasibility success. As such, a patient who experiences short term feasibility failure can be classified as a long-term feasibility success." (NCT00077207)
Timeframe: 60 weeks

Interventionparticipants (Number)
Treatment (Carboplatin, Vincristine Sulfate, Temozolomide)41

[back to top]

1-year Neurologic (Central Nervous System, CNS) Progression Free Rate

1-year CNS progression free rate is the percentage of patients who had no CNS progression after being followed for 1 year . Progressive disease (CNS) was defined as a 25% or greater increase in the sum of the product(s) of the maximal cross-sections on MRI scan, reappearance of any lesion that has disappeared, development of any new lesion(s), stable disease with a deterioration of neurologic exam, or clear worsening of any evaluable disease. (NCT00080938)
Timeframe: assessed every 3 months for 2 years

Interventionpercentage of participants (Number)
Temozolamide and Radiation90.5

[back to top]

Number of Patients With Intracranial Response

Response was assessed per Response Evaluation Criteria in Solid Tumor (RECIST) by brain MRI in the 21 eligible and treated patients.Complete response (CR): complete disappearance of the clinically detectable malignant brain metastasis(es) being followed on MRI scan off corticosteroids and a stable or improving neurologic exam. Partial response (PR): greater than or equal to a 50% reduction in the sum of the product(s) of the maximal cross-sections on MRI scan with a stable or decreasing dose of corticosteroids and a stable or improving neurologic exam. Response = CR + PR (NCT00080938)
Timeframe: assessed every cycle while on treatment, then every 3 months for 2 years

Interventionparticipants (Number)
Temozolamide and Radiation3

[back to top]

Overall Survival Time

Overall survival (months) was calculated from time of protocol entry to time of death from any cause. Patients alive at last follow-up were censored. The 21 eligible and treated patients were included in the analysis. (NCT00080938)
Timeframe: assessed every 3 months for 2 years

Interventionmonths (Median)
Temozolamide and Radiation7

[back to top]

Time to Non-CNS (Systemic) Progression

Time to non-CNS progression was calculated from time of protocol entry to time of first systemic progressive disease or death. Patients alive and non-CNS progression-free at last follow-up were censored. Disease progression was defined as at least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the baseline sum longest diameter (per RECIST criteria). Development of new lesions in non-CNS sites also constituted non-CNS progression. The 21 eligible and treated patients were included in the analysis. (NCT00080938)
Timeframe: assessed every 3 months for 2 years

Interventionmonths (Median)
Temozolamide and Radiation3.2

[back to top]

Grade 3-4 Pulmonary Events

All Grade 3-4 Pulmonary events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)4

[back to top]

2-yr Overall Survival

Overall survival is defined as the time from date of diagnosis to death or date of last follow-up. 2-year overall survival is the probability of patients remaining alive at 2-years from study entry estimated using Kaplan-Meier (KM) methods which censors patients at date of last follow-up. Precision of this conditional probability estimate was measured in terms of standard error. Median OS, the original primary endpoint, was not estimable based on the Kaplan-Meier method because of insufficient follow-up. (NCT00084838)
Timeframe: Patients are followed for survival up to 5 yrs post-therapy completion or death; As of this analysis, median follow-up among survivors was 31 months with the longest follow-up being 40 months.

Interventionprobability (Number)
Multi-agent Intrathecal and Systemic CT With RT (Modified IRS0.70

[back to top]

Grade 3-4 Allergy/Immunology

All Grade 3-4 Allergy/Immunology events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)1

[back to top]

Grade 3-4 Auditory/Hearing Events

All Grade 3-4 Auditory/Hearing events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)8

[back to top]

Grade 3-4 Blood/Bone Marrow Events

"All Grade 3-4 Blood/Bone Marrow events based on CTCAEv2 as reported on case report forms.~Arm Name" (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)564

[back to top]

Grade 3-4 Hepatic Events

All Grade 3-4 Hepatic events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)8

[back to top]

Grade 3-4 Pain Events

All Grade 3-4 Pain events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)31

[back to top]

Grade 3-4 Cardiovascular Events

All Grade 3-4 Cardiovascular events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)6

[back to top]

Grade 3-4 Constitutional Events

All Grade 3-4 Constitutional events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)22

[back to top]

Pre-Radiation Therapy Chemotherapeutic Response

"Response pre-RT/post-CT was defined as follows with overall response defined as achieving PR or CR.~Complete Response (CR): Complete resolution of all initially demonstrable tumor on MRI or CT evaluation w/o appearance of any new areas of disease; negative CSF cytology. Partial Response (PR): >/= 50% decrease in the sum of the products of the maximum perpendicular diameters of the tumor (sum LD) relative to baseline w/o appearance of any new areas of disease; CSF cytology unchanged from that at diagnosis or clearing after being initially positive Stable Disease (SD): <50% decrease in the sum LD w/o appearance of any new areas of disease; CSF cytology unchanged from that at diagnosis or clearing after being initially positive Progressive Disease (PD): >/= 25% increase in the sum LD relative to baseline, or the appearance of any new areas of disease or appearance of positive cytology after two consecutive negative samples." (NCT00084838)
Timeframe: Assessed at study entry and pre-RT/post-CT at week 7.

Interventionproportion of evaluable patients (Number)
Multi-agent Intrathecal and Systemic CT With RT (Modified IRS0.58

[back to top]

Grade 3-4 Renal/Genitourinary Events

All Grade 3-4 Renal/Genitourinary events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)4

[back to top]

Grade 3-4 Dermatology Events

All Grade 3-4 Dermatology events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)3

[back to top]

Grade 3-4 Neurology Events

All Grade 3-4 Neurology events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)45

[back to top]

Grade 3-4 Muscloskeletal Events

All Grade 3-4 Muscloskeletal events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)8

[back to top]

Grade 3-4 Gastrointestinal Events

All Grade 3-4 Gastrointestinal events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)139

[back to top]

Grade 3-4 Hemorrhage Events

All Grade 3-4 Hemorrhage events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)1

[back to top]

Grade 3/4 Events

All Grade 3-4 events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)1021

[back to top]

Grade 3-4 Metabolic/Laboratory Events

All Grade 3-4 Metabolic/Laboratory events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)128

[back to top]

Grade 3-4 Infection/Febrile Neutropenia Events

All Grade 3-4 Infection/Febrile Neutropenia events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)49

[back to top]

Dose Limiting Toxicities of EMD + RT and TMZ

"pts will be evaluated from first dose through end of initiation cycle. (6 weeks of RT+TMZ +EMD and 4 weeks of EMD alone) to review dose limiting toxicity (DLT) using Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (Phase I)~DLT defined as: Known TMZ hematological toxicities will not be considered dose limiting.~Nonhematological toxicities Grades 3-4 severity (except nausea and vomiting without sufficient antiemetic prophylaxis)" (NCT00085254)
Timeframe: 10 weeks

Interventionparticipants (Number)
Arm 1 500mg (Safety Run In)0
ARM 2 1000mg (Safety run-in)0
Arm 3 2000mg (Safety Run-In)0

[back to top]

Overall Survival (Phase II)

The overall survival is calculated from time of histological diagnosis to death occurance - median based on all 112 patients, all dose levels (NCT00085254)
Timeframe: up to 36 months

Interventionmonths (Median)
Arm 4 (Overall Study)19.7

[back to top]

Overall Survival Based on Dose Level - Phase 2

survival calculated from date of initial histologic diagnosis and occurence of death. Pts at 500mg dose compared against Pts treated at 2000mg dose. Calculated using median (NCT00085254)
Timeframe: Up to 3 years

Interventionmonths (Median)
Arm 1 - Phase 2 (Treatment 1)17.4
Arm 2 - Phase 2 (Treatment 2)20.8

[back to top]

Maximum Tolerated or Tolerable Dose (MTD) - 3 Pre-defined Doses

"pts will be evaluated from first dose through end of initiation cycle. (6 weeks of RT+TMZ +EMD and 4 weeks of EMD alone) to review any dose limiting toxicity (DLT) using Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (safety run-in)~DLT defined as: Known TMZ hematological toxicities will not be considered dose limiting.~cohorts at these 3 defined doses: 500mg, 1000mg and 2000mg MTD defined as: dose producing DLT in 2 out of 6 patients or dose level below the dose which produced DLT in >/= 2 out of 3 patients, or in >/= 3 out of 6 patients If no MTD (maximum tolerable dose) was defined through 3 steps of dose escalation, phase 2 will proceed with a randomized treatment allocation of the two pre-specified dosage arms: low dose; 500mg and high dose; 2000mg" (NCT00085254)
Timeframe: 10 weeks

Interventionmg (Number)
Arm 1 - Safety Run InNA

[back to top]

Frequency of Hematologic and Nonhematologic Adverse Events

The proportion of patients with grade 3 and grade 4 hematologic and non hematologic adverse events per CTCAE 4.0 (NCT00085254)
Timeframe: Up to 1 year

InterventionNumber of grade 3 or 4 events (Number)
Arm 1- Phase 2 (500mg)48
Arm 2 - Phase 2 (2000mg)35

[back to top]

Progression Free Survival

Progression free survival was defined as the time from the date of randomization to the date of disease progression or the date of death regardless of the cause. (NCT00091572)
Timeframe: Treatment continued until disease progression or unacceptable toxicity. Patients will be followed for survival.

InterventionMonths (Median)
Temozolomide2.30
Dacarbazine2.17

[back to top]

Overall Survival

Overall Survival was defined as the time from the date of randomization to the date of death from any cause. (NCT00091572)
Timeframe: The final analysis was to be performed when at least 616 deaths had occurred.

InterventionMonths (Median)
Temozolomide9.13
Dacarbazine9.36

[back to top]

Objective Response Rate in Subjects With Measurable Lesions

Based on investigator's assessment of response in subjects with measurable lesions. Objective response = complete response + partial response. Complete response = disappearance of all target lesions. Partial response = at least a 30% decrease in the sum of longest diameter of target lesions taking as reference the baseline sum longest diameter. (NCT00091572)
Timeframe: Treatment continued until disease progression or unacceptable toxicity.

InterventionRatio (Median)
Temozolomide0.14
Dacarbazine0.10

[back to top]

Duration of Objective Response

Duration of objective response was measured from the time the criteria were met for complete response or partial response to the first date that recurrent or progressive disease was objectively documented. (NCT00091572)
Timeframe: Treatment continued until disease progression or unacceptable toxicity.

InterventionMonths (Median)
Temozolomide4.34
Dacarbazine8.31

[back to top]

Change in Steroid Dependence at Six Months

Daily steroid dose will be collected at baseline and follow-up, as one of the following: 0-4 mg, >4 to ≤ 8 mg, >8 to ≤12 mg, and >12 mg. Change from baseline at six months will be evaluated to have decreased, remained stable, or increased, based on these categories. (NCT00096265)
Timeframe: From randomization to six months.

,,
InterventionParticipants (Count of Participants)
DecreaseStableIncrease
Erlotinib + WBRT + SRS1061
Temozolomide + WBRT + SRS1044
WBRT + SRS12104

[back to top]

Change in Performance Status at Six Months

Compared between two treatment arms using a two-group chi-squared test. Zubrod score will be collected at baseline and follow-up. The Zubrod performance score runs from 0 to 5, with 0 denoting perfect health and 5 death. Change from baseline is calculated as 6-month value - baseline value. Patients with a baseline score who have died by six months will be included in the analysis with a score of 5 at six months. (NCT00096265)
Timeframe: From randomization to six months.

,,
InterventionParticipants (Count of Participants)
Improvement (decrease)Stable (no change)Deterioration (increase)
Erlotinib + WBRT + SRS3230
Temozolomide + WBRT + SRS1430
WBRT + SRS01921

[back to top]

Change in Functional Assessment of Cancer Therapy-Brain (FACT-Br) Score at 3 Months

The Functional Assessment of Cancer Therapy-Brain (FACT-Br) is a 19-item self-report instrument designed to measure multidimensional quality of life in patients with brain cancer. It is to be administered with the FACT-General. There are 5 responses options, with 0=Not a lot and 4=Very much. All items are added together to obtain a total score, which ranges from 0 to 76. Certain items must be reversed before it is added by subtracting the response from 4. It requires at least 50% of the items to be completed while the overall response rate of the FACT-Br including the FACT-G must be greater than 80%. If items are missing, the subscale scores can be prorated. A higher score indicates better QOL. A change of 5 points will be considered a minimal clinically meaningful change. Change from baseline at three months (3 month score - baseline score) will be categorized as improvement if increased, stable if no change, or deterioration if decreased. (NCT00096265)
Timeframe: From randomization to three months.

,,
InterventionParticipants (Count of Participants)
Deterioration/DecreaseStableImprovement/Increase
Erlotinib + WBRT + SRS1161
Temozolomide + WBRT + SRS1024
WBRT + SRS1286

[back to top]

Quality-adjusted Survival as Measured by EuroQol 5-dimension Instrument

Quality-adjusted life years (QALY) incorporate the societal-based utilities of health states into expected life years for a health condition. The QALY model is QALY(h,y) where h is a health state and y is the years of life. Higher quality-adjusted life year values represent a better outcome. A patient's health state will be determined from the index score of the EQ-5D-5L patient questionnaire.The EQ-5D-5L is a 2-part self-assessment questionnaire, a 5-item index score and a visual analogue scale, but only the index score is used for quality-adjusted survival. The index score has 5 items (mobility, self care, usual activities, pain/discomfort, anxiety/depression) each with 5 problem levels (1-none to 5-extreme). The 5-item index score is transformed into a utility score between 0 (worst health state) and 1 (best health state). (NCT00096265)
Timeframe: From randomization to last follow-up, up to 48.1 months. Analysis occurs after all patients have been potentially followed for 9 months.

InterventionQuality-adjusted life years (Mean)
WBRT + SRS16.9
Temozolomide + WBRT + SRS15.9
Erlotinib + WBRT + SRS14.4

[back to top]

Rate of CNS Progression (One Year)

CNS progression is defined as any increase in perpendicular bi-dimensional tumor area for any of the 1-3 tracked brain metastases, by any amount, or the appearance of any new brain metastasis on a follow-up MRI (SRS planning scan will not be used to evaluate CNS progression). For lesions smaller than 1 cm in maximum diameter, a maximum increase of 50% in perpendicular bi-dimensional treatment area is necessary to score as progression. This caveat is included to account for potential variability in measurement, which is most susceptible to proportionate errors at smaller sizes. For greater than 1 cm lesions, the definition uses a 25% rule for change. Rates of CNS progression estimated by the cumulative incidence method, with death treated as a competing risk. (NCT00096265)
Timeframe: From randomization to last follow-up, up to 48.1 months. Analysis occurs after all patients have been potentially followed for 9 months.

Interventionpercentage of participants (Number)
WBRT + SRS34.1
Temozolomide + WBRT + SRS47.4
Erlotinib + WBRT + SRS27.4

[back to top]

Overall Survival

Survival time is defined as time from randomization to date of death from any cause and estimated by the Kaplan-Meier method. Patients last known to be alive are censored at date of last contact. (NCT00096265)
Timeframe: From randomization to date of death or last follow-up, up to 48.1 months. Analysis occurs after all patients have been potentially followed for 9 months.

Interventionmonths (Median)
WBRT + SRS13.4
Temozolomide + WBRT + SRS6.3
Erlotinib + WBRT + SRS6.1

[back to top]

4 Year Overall Survival Rate

Percentage of patients who were alive at 4 years. The 4-year survival rate was estimated using the Kaplan Meier method. (NCT00098774)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Intensive Combination Chemo & Immunotherapy65

[back to top]

Change From Baseline in Mini-Mental Status Evaluation at 4 Months

Neurologic functioning will be assessed using the Mini-Mental Status Evaluation (MMSE), a standardized, bedside tool for evaluation of higher mental function. This assessment is based on a 30-point scale (0-30) with higher scores associated with better performance. (NCT00098774)
Timeframe: Baseline & month 4

Interventionunits on a scale (Median)
Baseline Score4 month ScoreChange from Baseline
Intensive Combination Chemo & Immunotherapy27281

[back to top]

Complete Response Rate After Remission Induction

Response is assessed by investigator according to Revised Response Criteria for Malignant Lymphoma. Complete response requires disappearance of all evidence of disease. (NCT00098774)
Timeframe: 4 months

Interventionpercentage of participants (Number)
Intensive Combination Chemo & Immunotherapy66

[back to top]

4 Year Progression Free Rate

"Percentage of patients who were progression free at 4 years. The 4-year progression free rate was estimated using the Kaplan Meier method.~Relapse was assessed by investigator according to Revised Response Criteria for Malignant Lymphoma. Progression required a 25% increase of previous area of gadolinium enhancement, appearance of new areas of T1 gadolinium enhancement or new appearance of malignant cells in the spinal fluid or new tumor appearance in other sites of the body" (NCT00098774)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Intensive Combination Chemo & Immunotherapy48

[back to top]

Overall Response

"Overall response is the best response during 6 months of therapy measured by radiographic response.~Complete Response (CR): Disappearance of all detectable tumors by imaging, if initially positive, as well as 2 consecutively negative CSF cytologic examinations (if the initial cytology was positive).~Partial Response (PR): > 50% reduction in the sum of the products of the maximum perpendicular diameter of all measurable lesions; or 2 consecutively negative CSF cytologies and a < 50% reduction in tumor size.~Stable Disease (SD): < 50% reduction in the sum of the products of the maximum perpendicular diameters of all measurable lesions, and persistently negative or positive CSF cytology Progressive Disease (PD): > 25% increase in the size of any measurable lesion, the appearance of a new radiographically demonstrable lesion, or the conversion of negative CSF cytology to positive, as confirmed by at least one repeat CSF cytology" (NCT00098865)
Timeframe: Assessed every 8 weeks while on treatment and every 3 months for one year off-study

Interventionparticipants (Number)
Partial ResponseStable DiseaseProgressive DiseaseUnevaluable
Thalidomide and Temozolomide1941

[back to top]

Therapy Completion Rate

Feasibility in this study was defined as completion of 6 months of thalidomide with temozolomide therapy. The corresponding therapy completion rate is defined as the proportion of patients who completed 6 months of therapy. (NCT00098865)
Timeframe: 6 months

Interventionproportion of participants (Number)
Thalidomide and Temozolomide.40

[back to top]

Overall Survival

Time from registration to death. Patients alive at last follow-up were censored. (NCT00098865)
Timeframe: Assessed after treatment discontinued every 3 months up to 2 years.

Interventionmonths (Median)
Thalidomide and Temozolomide12.8

[back to top]

One Year Overall Survival

Estimated one year survival using the Kaplan-Meier methodology. (NCT00100802)
Timeframe: One year

InterventionEstimated probability (Number)
Surgery, Chemoradiotherapy, Rest, Maintenance, FUP0.7208

[back to top]

Occurrence of Death Attributable to Complications of Protocol Therapy

Number of deaths due to complications of protocol therapy. (NCT00100802)
Timeframe: While receiving protocol therapy (up to 301 days excluding delays) or within 30 days of Termination of Protocol Therapy

Interventionpatients (Number)
Surgery, Chemoradiotherapy, Rest, Maintenance, FUP1

[back to top]

Median Overall Survival (OS) Comparison of Doublet Versus Triplet Therapy

Doublet (2 agents) versus Triplet (3 agents) therapy analysis: We compared the median OS outcome of participants in arms II, III, IV, versus participants in arms V, VI and VII. Median OS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 3 months from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Doublet (2 Agents): Arm II, Arm III and Arm IV17.0
Triplet (3 Agents): Arm V, Arm VI and Arm VII20.1

[back to top]

Median Overall Survival (OS) Comparison of Celecoxib Arms Versus no Celecoxib Arms

Celecoxib versus not Celecoxib analysis: We compared the median OS outcome of participants in arms III, V, VI and VIII, versus participants in arms I, II, IV and VII. Median OS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 3 months from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Celecoxib: Arm III, Arm V, Arm VI and Arm VIII20.2
No Celecoxib: Arm I, Arm II, Arm IV and Arm VII17.1

[back to top]

Median Progression-Free Survival (PFS) of Individual Arms

Median PFS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 2 cycles (1 cycle = 28 days) from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Arm I: TMZ10.5
Arm II: TMZ + Thalidomide7.7
Arm III: TMZ + Celecoxib13.4
Arm IV: TMZ + Isotretinoin6.5
Arm V: TMZ + Isotretinoin + Celecoxib11.6
Arm VI: TMZ + Thalidomide + Celecoxib7.9
Arm VII: TMZ + Thalidomide + Isotretinoin6.2
Arm VIII: TMZ + Thalidomide + Isotretinoin + Celecoxib5.8

[back to top]

Median Overall Survival (OS) Comparison of Thalidomide Arms Versus no Thalidomide Arms

Thalidomide versus not Thalidomide analysis: We compared the median OS outcome of participants in arms II, VI, VII and VIII, versus participants in arms I, III, IV and V. Median OS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 3 months from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Thalidomide: Arm II, Arm VI, Arm VII and Arm VIII18.3
No Thalidomide: Arm I, Arm III, Arm IV and Arm V17.4

[back to top]

Median Progression-Free Survival (PFS) Comparison of Celecoxib Arms Versus no Celecoxib Arms

Celecoxib versus not Celecoxib analysis: We compared the median PFS outcome of participants in arms III, V, VI and VIII, versus participants in arms I, II, IV and VII. Median PFS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 2 cycles (1 cycle = 28 days) from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Celecoxib: Arm III, Arm V, Arm VI and Arm VIII8.3
No Celecoxib: Arm I, Arm II, Arm IV and Arm VII7.4

[back to top]

Median Progression-Free Survival (PFS) Comparison of Doublet Versus Triplet Therapy

Doublet (2 agents) versus Triplet (3 agents) therapy analysis: We compared the median PFS outcome of participants in arms II, III, IV, versus participants in arms V, VI and VII. Median PFS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 2 cycles (1 cycle = 28 days) from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Doublet (2 Agents): Arm II, Arm III and Arm IV8.3
Triplet (3 Agents): Arm V, Arm VI and Arm VII8.2

[back to top]

Median Overall Survival (OS) Comparison of Isotretinoin Arms Versus no Isotretinoin Arms

Isotretinoin versus not Isotretinoin analysis: We compared the median OS outcome of participants in arms IV, V, VII and VIII, versus participants in arms I, II, III and VI. Median OS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 3 months from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Isotretinoin: Arm IV, Arm V, Arm VII and ARM VIII17.1
No Isotretinoin: Arm I, Arm II, Arm III and ARM VI19.9

[back to top]

Overall Survival of Individual Arms

Overall Survival (OS) was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 3 months from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Arm I: TMZ21.2
Arm II: TMZ + Thalidomide17.4
Arm III: TMZ + Celecoxib18.1
Arm IV: TMZ + Isotretinoin11.7
Arm V: TMZ + Isotretinoin + Celecoxib23.1
Arm VI: TMZ + Thalidomide + Celecoxib20.2
Arm VII: TMZ + Thalidomide + Isotretinoin17.9
Arm VIII: TMZ + Thalidomide + Isotretinoin + Celecoxib18.5

[back to top]

Median Progression-Free Survival (PFS) Comparison of Isotretinoin Arms Versus no Isotretinoin Arms

Isotretinoin versus not Isotretinoin analysis: We compared the median PFS outcome of participants in arms IV, V, VII and VIII, versus participants in arms I, II, III and VI. Median PFS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 2 cycles (1 cycle = 28 days) from randomization until progression of disease, death or last follow-up.

Interventionmonths (Median)
Isotretinoin: Arm IV, Arm V, Arm VII and Arm VIII6.6
No Isotretinoin: Arm I, Arm II, Arm III and Arm VI9.1

[back to top]

Median Progression-Free Survival (PFS) Comparison of Thalidomide Arms Versus no Thalidomide Arms

Thalidomide versus not Thalidomide analysis: Comparison of median PFS outcome of participants in arms II, VI, VII and VIII, versus participants in arms I, III, IV and V. Median PFS was estimated using the Kaplan-Meier method from time of randomization to time of progression, death, or last follow-up. Progression defined as 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00112502)
Timeframe: Every 2 cycles (1 cycle = 28 days) from randomization until progression of disease, death or last follow-up, up to one year (12 study cycles).

Interventionmonths (Median)
Thalidomide: Arm II, Arm VI, Arm VII and Arm VIII7.6
No Thalidomide: Arm I, Arm III, Arm IV and Arm V8.7

[back to top]

Neurocognitive Function

"Hopkins Verbal Learning Test (HVLT) is a test measuring learning memory retrieval, and memory consolidation processes.; Controlled Oral Word Association Test (COWAT) is a test of phonemic verbal fluency. The patient produces as many words as possible in 1 min. (each) for a specific letter (C, F, L or P, R, W).; Trail Making Test (TMT) is a measure of visuospatial scanning, attention, sequencing, and speed in Part A (TMT A) and executive function in Part B (TMT B). Patients must connect the dots either in a numbered sequence or alternating letters and numbers. Difference between pre-treatment baseline and follow-up assessment scores determined by the reliable change (RC) index, using a 90% confidence interval to designate statistically significant change." (NCT00114140)
Timeframe: Baseline, 6 months, and 12 months.

,
Interventionparticipants (Number)
HVLT Recall Status: DeteriorationHVLT Recall Status: No changeHVLT Recall Status: ImprovementCOWA Status: DeteriorationCOWA Status: No changeCOWA Status: ImprovementTMT A Status: DeteriorationTMT A Status: No changeTMT A Status: ImprovementTMT B Status: DeteriorationTMT B Status: No changeTMT B Status: Improvement
Temozolomide + Radiation Therapy (RT) at 12 Months82215328127241472215
Temozolomide + Radiation Therapy (RT) at 6 Months1126131426152411112711

[back to top]

Progression-free Survival

"Progressive Disease (PD) is defined as 25% or > increase in the cross-sectional area of enhancing or non-enhancing tumor on consecutive MRI scans, or any new area(s) of tumor. Under exceptional circumstances, disease progression may be declared in the absence of an increase in tumor size based on clinical deterioration including the need for increasing doses of steroid and/or a worsening Karnofsky Performance Status(KPS) / Neurologic Function Score(NFS). Progression-free survival time is defined as time from registration to date of progressive disease or death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. Median survival time is reported." (NCT00114140)
Timeframe: From registration to last follow-up, up to 7.1 years. Analysis occurs after all patients have been on study for at least 3 years.

Interventionyears (Median)
Temozolomide + Radiation Therapy (RT)4.5

[back to top]

Overall Survival Rate at 3 Years

Survival time is defined as time from registration to date of death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. This analysis was planned to occur when all patients had been potentially followed for at least 3 years. (NCT00114140)
Timeframe: Registration to 3 years

Interventionpercentage of participants (Number)
Temozolomide + Radiation Therapy (RT)73.1

[back to top]

Survival and Progression-free Survival by O(6)-Methylguanine-DNA Methyltransferase (MGMT) Methylation Status

"Survival time is defined as time from registration to date of death from any cause. Progressive Disease (PD) is defined as 25% or > increase in the cross-sectional area of enhancing or non-enhancing tumor on consecutive MRI scans, or any new area(s) of tumor. Under exceptional circumstances, disease progression may be declared in the absence of an increase in tumor size based on clinical deterioration including the need for increasing doses of steroid and/or a worsening Karnofsky Performance Status(KPS) / Neurologic Function Score(NFS). Survival and progression-free survival are estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact." (NCT00114140)
Timeframe: Registration to 3 years

,
Interventionyears (Median)
Overall SurvivalProgression-free Survival
Temozolomide + Radiation Therapy (RT) - MethylatedNANA
Temozolomide + Radiation Therapy (RT) - Unmethylated3.02.0

[back to top]

Quality of Life as Measured by the Functional Assessment of Cancer Therapy Scale With Brain Module (FACT-BR)

Functional Assessment of Cancer Therapy Scale with brain module (FACT-BR): a 50-question self-report questionnaire contains the following domains (scales): Physical well-being (7 questions totalling 0-28), social/family well-being (7 questions totalling 0-28), emotional well-being (6 questions totalling 0-24), functional well-being (7 questions totalling 0-28) and brain cancer subscale which contains concerns relevant to patients with brain tumors (19 questions totalling 0-76). Each question has a value 0-4. For some questions a higher indicates better outcome and others are the opposite. The former are summed as is, the latter are reversed in value before adding, such that each domain ranges from 0 to 4 multiplied by the number of questions in the domain, with 0 indicating worst and the highest possible value indicating best outcome. The FACT-Br total (0-184) is obtained by adding all domains together if the overall question response rate is greater than 80%. (NCT00114140)
Timeframe: Baseline, 6 months, and 12 months.

,,
Interventionunits on a scale (Median)
Physical Well-BeingSocial/Family Well-BeingEmotional Well-BeingFunctional Well-BeingBrain Cancer SubscaleFACT-Br Total
Temozolomide + Radiation Therapy (RT) at 12 Months2324.323.019.559.0140.0
Temozolomide + Radiation Therapy (RT) at 6 Months2224.020.018.052.0129.9
Temozolomide + Radiation Therapy (RT) at Baseline2324.518.517.054.0135.3

[back to top]

Overall Survival

Patients were monitored until death (NCT00187486)
Timeframe: assessment of survival was every 2 months, up to 181 weeks

Interventionmonths (Median)
Temodar Plus Tarceva Plus Radiotherapy19

[back to top]

Progression Free Survival

Progression based on MR imaging using the Modified McDonnald Criteria defined as 25% increase in sum of products of all measured lesions or any new lesion (NCT00187486)
Timeframe: every 2 months measure by MR imaging, up to 39 months

Interventionmonths (Median)
Temodar Plus Tarceva Plus Radiotherapy8.2

[back to top]

Progression Free Survival at 6 Months

(NCT00200161)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Metronomic Therapy Cohort46
Dose-Dense Therapy Cohort56

[back to top]

12 Month Overall Survival of Patients With Newly Diagnosed Glioblastoma Multiforme Treated With Concurrent Temozolomide and Radiotherapy Followed by Dose Dense or Metronomic Dosing of Temozolomide and Maintenance Cis-retinoic Acid.

(NCT00200161)
Timeframe: until death or date of last follow up, an average of 12 months

Interventionpercentage of participants (Number)
Metronomic Therapy Cohort69
Dose-Dense Therapy Cohort80

[back to top]

Number of Participants Alive at 3 Years

The intent was to measure Median Overall Survival at 3 years, however only one participant was analyzable at this time point. Therefore, the number of participants who survived is reported instead. (NCT00248534)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
IV Rituximab1

[back to top]

Percentage of Participants With Objective Response

Objective response rate of the combination of Rituximab and TMZ (NCT00248534)
Timeframe: 2 months

Interventionpercent of participants (Number)
IV Rituximab14

[back to top]

6-month Progression-free Survival

"Scan at 6 months~Complete response: Complete disappearance of all tumor on MRI scan, off all glucocorticoids with stable or improving neurological exam minimum of 4 wks~Partial response: Greater than or equal 50% reduction in tumor size on MRI, on sable or decreasing glucocorticoids with stable or improving neurological exam for a minimum of 4 wks.~Progressive disease: Progressive neurological abnormalities not explained by other causes or greater than 25% increase in size of tumor or if new lesion.~Stable disease: Clinical status and MRI does not qualify for complete response, partial response or progression" (NCT00248534)
Timeframe: 6 months

Interventionpercentage of participants (Number)
IV Rituximab13

[back to top]

1 Year Overall Survival Rate

(NCT00248534)
Timeframe: 1 year

Interventionpercentage of participants (Number)
IV Rituximab71

[back to top]

Survival

survival time is defined from time of histological diagnosis to death occurrence. (NCT00262730)
Timeframe: 30 months

Interventionmonths (Mean)
Treatment Arm - All Subjects17.2

[back to top]

Number of Participants With Progression-free Survival (6 Month)

The occurrence of progression will be compared between the study groups by Kaplan-Meier curves. Responsiveness to temozolomide will be evaluated by brain Magnetic Resonance Imanging (MRI)/Computed Tomography (CT), thorax CT, and Quality of Life (QoL) assessments. Progression-free is defined as <25% increase in tumor size on CT or MRI. (NCT00266812)
Timeframe: 6 months

InterventionParticipants (Number)
Chemotherapy With Temozolomide and Radiotherapy8
Radiotherapy Alone8

[back to top]

Percentage of Participants With an Objective Response (Complete Response or Partial Response)

The primary endpoint is to assess the percentage of participants with a sustained objective response (complete response (CR) or partial response (PR)). Response is assessed by magnetic resonance imaging (MRI) per the following criteria: CR - disappearance of tumor and PR - ≥50% reduction in tumor based on the maximal cross-sectional measurements. The response must be sustained for at least 8 weeks, and the date of the confirmed sustained response is the date at which the response was first noted by MRI. (NCT00275002)
Timeframe: Week 8, 16, 24, 32, and 40 after starting therapy

InterventionPercent of Participants (Number)
Recurrent High-Grade Gliomas4
Recurrent Brain Stem Tumors0

[back to top] [back to top]

Overall Response Rate

"Overall Response Rate (ORR) is defined as percentage of patients who's best response to treatment is complete response plus those with partial response.~Complete Response (CR): Disappearance of all target lesions.~Partial Response (PR): At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD." (NCT00301067)
Timeframe: From the start of treatment, every 2 cycles where 1 cycle equals 28 days, for a maximum of 12 cycles

InterventionParticipants (Count of Participants)
Temozolomide and Calcitriol (Cohort 1-3+Expansion)2

[back to top]

Number and Frequency of Dose Limiting Toxicities (DLTs) With High-dose Calcitriol in Combination With Temozolomide

"Determine number and frequency of dose limiting toxicities (DLT) of high-dose calcitriol when administered with temozolomide in patients with metastatic melanoma for up to 12 cycles of therapy, where 1 cycle equals 28 days.~3 patients per dose cohort will be entered into the trial at doses of 0.2, 0.3, and 0.5 mcg/kg of calcitriol administered orally. If 1 patient experiences dose limiting toxicity (DLT) at any dose, that dose cohort will be expanded to a maximum of 6 patients. If 1 additional patient experiences DLT at that dose stratum, further dose escalation will cease and the dose cohort immediately preceding the dose cohort where the 2 experiences of DLT occurred will be considered the MTD. If no additional patients experience DLT, dose escalation to the next higher dose stratum will take place.~DLT is defined as National Cancer Institute Common Toxicity Criteria, version 3.0 grade 3 toxicity determined to be related to calcitriol." (NCT00301067)
Timeframe: From start of treatment, up to 12 cycles where 1 cycle equals 28 days

InterventionDLT (Number)
Cohort 1 - Temozolomide and Calcitriol0
Cohort 2 - Temozolomide and Calcitriol0
Cohort 3 - Temozolomide and Calcitriol0

[back to top]

Overall Survival (OS) Stratified by Vitamin D-Receptor (VDR) Gene Polymorphisms

Investigate the relationship between vitamin D-receptor (VDR) gene polymorphisms in Taq1 and Fok1 (analyzed from baseline blood sample) and Overall Survival (OS). VDR gene analysis was completed using PCR-RFLP based assays. (NCT00301067)
Timeframe: at baseline and until death from any cause up to 6 and half years

InterventionMonths (Median)
VDR genotype (tt+/-ff)non tt+/-ff VDR genotype
Temozolomide and Calcitriol (Cohort 1-3+Expansion)3.87.4

[back to top]

Number of Patients With Toxicity

"Toxicity will be assessed for each patient on a seven-day on/seven-day off temozolomide in combination with high-dose calcitriol for every 2 weeks for up to 12 cycles where 1 cycle equals 28 days. Toxicity will be assessed during treatment according to the National Cancer Institute's Common Toxicity Criteria for adverse events version 3.0 (CTCAE v3.0) and defined by any toxicity determined to be at least possibly related to either study drug (temozolomide or calcitriol).~In general adverse events (AEs) will be graded according to the following:~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~Grade 3 and grade 4 toxicities where relatedness to either study drug could not be ruled out were collected and recorded only." (NCT00301067)
Timeframe: From the start of treatment and every 2 weeks for a maximum of 12 cycles, and 30 days post last treatment, where 1 cycle equals 28 days

,,,
Interventionparticipants (Number)
ThrombocytopeniaVascularNauseaVomitingLeukopeniaFatigueAnemiaLymphopeniaHemorrhageRashAnorexia
Cohort 1 - Temozolomide and Calcitriol11000000000
Cohort 2 - Temozolomide and Calcitriol00000000000
Cohort 3 - Temozolomide and Calcitriol00111000000
Expansion - Temozolomide and Calcitriol11001122111

[back to top]

Time to Progression

Time to progression (TTP) is measured from the start of treatment until the time of first documentation of disease progression. (NCT00301067)
Timeframe: From the start of treatment, until progressive disease, up to 12 months

InterventionMonths (Median)
Temozolomide and Calcitriol (Cohort 1-3+Expansion)1.81

[back to top]

Overall Survival

Overall Survival (OS) will be measured from first day of treatment until death of any cause. Patients still alive at the last data cut off point will be censored. (NCT00301067)
Timeframe: From the first day of treatment until death from any cause, up to a maximum of 6 and half years

InterventionMonths (Median)
Temozolomide and Calcitriol (Cohort 1-3+Expansion)5.5

[back to top]

Percentage of Participants With Overall Survival at 6, 12, and 24 Months

Percentage of participants who were alive at 6, 12, and 24 months. (NCT00302159)
Timeframe: 6, 12, and 24 months

Interventionpercentage of participants (Number)
6 months12 months24 months
Valproic Acid978656

[back to top]

Percentage of Participants With Progression Free Survival at 6, 12, and 24 Months

Percentage of participants who were progression free by 6, 12, or 24 months. Progressive disease is a >25% increase in contrast enhancing tumor volume documented at the initiation of treatment on protocol. (NCT00302159)
Timeframe: 6, 12, and 24 months

Interventionpercentage of participants (Number)
6 months12 months24 months
Valproic Acid704338

[back to top]

Median Progression Free Survival.

Progression free survival is the interval from initiation of treatment on protocol to symptomatic or radiographic progression. Progressive disease is a >25% increase in contrast enhancing tumor volume documented at the initiation of treatment on protocol. (NCT00302159)
Timeframe: up to 51 months

Interventionmonths (Median)
Valproic Acid10.5

[back to top]

Median Overall Survival

Survival is the interval from the initiation of treatment on protocol to date of death. (NCT00302159)
Timeframe: up to 63.8 months

Interventionmonths (Median)
Valproic Acid29.6

[back to top]

Number of Participants With Adverse Events

Here is the number of participants with adverse events. For a detailed list of adverse events, see the adverse event module. (NCT00302159)
Timeframe: 6 years, 7 months and 27 days

Interventionparticipants (Number)
Valproic Acid43

[back to top]

Number of Participants With Best Response

Best response recorded from the start of treatment until disease progression/recurrence. Complete response is complete resolution of all contrast enhancing tumor documented at initiation of treatment on protocol, with no appearance of new lesions. Partial response is a >50% reduction in the contrast enhancing tumor volume documented at the initiation of treatment on protocol. Minor response is a >25%, but <50% reduction in the contrast enhancing tumor volume documented at the initiation of treatment on protocol. Stable disease is a change in tumor size less than MR but not demonstrating progressive disease. Progressive disease is a >25% increase in contrast enhancing tumor volume documented at the initiation of treatment on protocol. Not evaluable means the participant cannot be evaluated (e.g., quality of scan). (NCT00302159)
Timeframe: up to 63.8 months

Interventionparticipants (Number)
Complete ResponsePartial ResponseMinor ResponseStable DiseaseProgressive DiseaseNot Evaluable
Valproic Acid0002779

[back to top]

Mean Neurocognitive Function (NCF) Composite Score Over Time

The NCF Composite score is the arithmetic mean of the HVLT-R (Free Recall, Delayed Recall, Delayed Recognition), TMTA, TMTB, and COWA scores, all of which are standardized, adjusting for age, education, and gender as necessary, such that mean is 0 and standard deviation is 1. A participant must have at least 5 of the 6 scores. A higher composite score indicates better neurocognitive function. (NCT00304031)
Timeframe: Baseline, 10, 22, and 46 weeks

,
Interventionscore on a scale (Mean)
BaselineWeek 10 (Cycle 1)Week 22 (Cycle 4)Week 46 (Cycle 10)
Conventional Adjuvant TMZ-1.2-1.3-1.1-1.0
Dose-dense Adjuvant TMZ-1.5-1.45-1.3-1.2

[back to top]

Median Progression-free Survival Time by MGMT Status

Progression is defined as greater than 25% increase in tumor area (two diameters) provided that the patient has not had his/her dose of steroids decreased since the last evaluation period. Progression-free survival time is defined as time from registration to the date of first progression, death, or last known follow-up (censored). Progression-free survival rates are estimated using the Kaplan-Meier method. A concomitant decrease in steroid dose will rule out a progression designation during the first 2 months after completion of radiation therapy. Tumor tissue submitted at baseline was analyzed to determine MGMT (O[6]-methylguanine-DNA methyltransferase) promoter methylation status (methylated / unmethylated). Analysis occurred after 647 deaths were reported. (NCT00304031)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 4.4 years.

,
Interventionmonths (Median)
Unmethylated MGMTMethylated MGMT
Conventional Adjuvant TMZ5.16.5
Dose-dense Adjuvant TMZ6.010.1

[back to top]

Determination of Impactful Baseline Instruments on Overall Survival

Overall survival time is defined as time from registration/randomization to the date of death from any cause or last known follow-up (censored). Overall survival rates are estimated by the Kaplan-Meier method. The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire [EORTC QLQ]-C30 subscales are calculated as the mean of component items, then standardized such that subscale scores range from 0 to 100. A high score for a functional scale represents a healthy level of functioning. Controlled Oral Word Association (COWA) score is the sum of correct responses with a range of 0 to infinity. A higher score indicates better functioning. Hopkins Verbal Learning Test - Revised (HVLT-R) score ranges from 0 to 36 for total recall is 0 to 36, 0 to 12 for delayed recall, and -12 to 12 for recognition. A higher score indicates better functioning. (NCT00304031)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 4.4 years.

Interventionmonths (Median)
Both Arms Combined17.5

[back to top]

Median Overall Survival Time by MGMT Status

Overall survival time is defined as time from randomization to the date of death from any cause. Overall survival rates are estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. Tumor tissue submitted at baseline was analyzed to determine MGMT (O[6]-methylguanine-DNA methyltransferase) promoter methylation status (methylated / unmethylated). Analysis occurred after 647 deaths were reported. (NCT00304031)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 4.4 years.

,
Interventionmonths (Median)
Unmethylated MGMTMethylated MGMT
Conventional Adjuvant TMZ14.621.4
Dose-dense Adjuvant TMZ13.320.2

[back to top]

Median Progression-free Survival (PFS) Time

Progression is defined as greater than 25% increase in tumor area (two diameters) provided that the patient has not had his/her dose of steroids decreased since the last evaluation period. Progression-free survival time is defined as time from registration to the date of first progression, death, or last known follow-up (censored). Progression-free survival rates are estimated using the Kaplan-Meier method. A concomitant decrease in steroid dose will rule out a progression designation during the first 2 months after completion of radiation therapy. Analysis occurred after 647 deaths were reported. (NCT00304031)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 4.4 years.

Interventionmonths (Median)
Conventional Adjuvant TMZ5.5
Dose-dense Adjuvant TMZ6.7

[back to top]

Mean MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score Over Time

The MDASI-BT is a 28-item patient-reported outcome measure assessing symptom severity and resulting interference with daily living in brain cancer patients. All items range from 0 (not present) to 10 (as bad as you can imagine). The symptom severity score is the average of the symptom severity items, given a specified minimum numbers were completed. (NCT00304031)
Timeframe: Baseline, 10, 12, 22, 24, and 46 weeks

,
Interventionscore on a scale (Mean)
BaselineWeek 10 (Cycle 1)Week 12 (Cycle 1.5)Week 22 (Cycle 4)Week 24 (Cycle 4.5)Week 46 (Cycle 10)
Conventional Adjuvant TMZ1.31.41.61.01.01.2
Dose-dense Adjuvant TMZ1.11.41.51.21.11.1

[back to top]

Change From Baseline in Mean EORTC QLQ-C30 Global Health Status

"Global Health Status is calculated from two questions on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire [EORTC QLQ]-C30. The question responses range from 1 very poor to 7 excellent such that a higher response indicates better quality of life (QOL). The mean of these responses is linearly transformed to a range of 0 (worst) to 100 (best). Change from baseline was calculated as time point value - baseline value with a positive change value indicating improved QOL from baseline." (NCT00304031)
Timeframe: Baseline, 10,12, 22, 24, and 46 weeks

,
Interventionscore on a scale (Mean)
Week 10 (Cycle 1)Week 12 (Cycle 1.5)Week 22 (Cycle 4)Week 24 (Cycle 4.5)Week 46 (Cycle 10)
Conventional Adjuvant TMZ0.0-4.63.9-2.85.4
Dose-dense Adjuvant TMZ-2.9-2.7-4.4-0.7-1.9

[back to top]

Overall Survival Status by Progression Status at 6 Months

Overall survival time is defined as time from registration to the date of death from any cause or last known follow-up (censored). Overall survival rates are estimated by the Kaplan-Meier method. Progression is defined as greater than 25% increase in tumor area (two diameters) provided that the patient has not had his/her dose of steroids decreased since the last evaluation period. (NCT00304031)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 4.4 years.

Interventionmonths (Median)
No Progression at 6 Months20.7
Progression at 6 Months10.1

[back to top]

Number of Participants With Deterioration From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Cycle 4

The MDASI-BT is a 28-item patient-reported outcome measure assessing symptom severity and resulting interference with daily living in brain cancer patients. All items range from 0 (not present) to 10 (as bad as you can imagine). The symptom severity score is the average of the symptom severity items, given a specified minimum numbers were completed. A score worse than baseline by at least one is considered deterioration. (NCT00304031)
Timeframe: baseline and cycle 4 (approximately 22 weeks)

InterventionParticipants (Count of Participants)
Conventional Adjuvant TMZ5
Dose-dense Adjuvant TMZ11

[back to top]

Number of Participants With Deterioration From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Interference Score at Cycle 4

The MDASI-BT is a 28-item patient-reported outcome measure assessing symptom severity and resulting interference with daily living in brain cancer patients. All items range from 0 (did not interfere) to 10 (interfered completely). The symptom interference score is the average of the symptom interference items, given a specified minimum numbers were completed. A score worse than baseline by at least one is considered deterioration. (NCT00304031)
Timeframe: baseline and cycle 4 (approximately 22 weeks)

InterventionParticipants (Count of Participants)
Conventional Adjuvant TMZ7
Dose-dense Adjuvant TMZ13

[back to top]

Median Overall Survival Time

Overall survival time is defined as time from registration/randomization to the date of death from any cause. Overall survival rates are estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. Analysis occurred after 647 deaths were reported. (NCT00304031)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 4.4 years.

Interventionmonths (Median)
Conventional Adjuvant TMZ16.6
Dose-dense Adjuvant TMZ14.9

[back to top]

Mean Neurocognitive Function (NCF) Composite Score at Cycle 10 for Participants Without Progression After 6 Months of Adjuvant Therapy

The NCF Composite score is the arithmetic mean of the Hopkins Verbal Learning Test - Revised (HVLT-R) (Free Recall, Delayed Recall, Delayed Recognition), Trail Making Test Part A (TMTA), Trail Making Test Part B (TMTB), and Controlled Oral Word Association (COWA) test scores, all of which are standardized, adjusting for age, education, and gender as necessary, such that mean is 0 and standard deviation is 1. A participant must have at least 5 of the 6 scores. A higher composite score indicates better neurocognitive function. (NCT00304031)
Timeframe: Baseline and cycle 10 (approximately 46 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ-0.95
Dose-dense Adjuvant TMZ-1.19

[back to top]

Mean MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Cycle 10 for Participants Without Progression After 6 Months of Adjuvant Therapy

The MDASI-BT is a 28-item patient-reported outcome measure assessing symptom severity and resulting interference with daily living in brain cancer patients. All items range from 0 (not present) to 10 (as bad as you can imagine). The symptom severity score is the average of the symptom severity items, given a specified minimum numbers were completed. (NCT00304031)
Timeframe: Baseline and cycle 10 (approximately 46 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ1.17
Dose-dense Adjuvant TMZ1.18

[back to top]

Mean EORTC QLQ-C30 Global Health Status Score at Cycle 10 for Participants Without Progression After 6 Months of Adjuvant Therapy

"Global Health Status is calculated from two questions on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire [EORTC QLQ]-C30. The question responses range from 1 very poor to 7 excellent such that a higher response indicates better quality of life (QOL). The mean of these responses is linearly transformed to a range of 0 (worst) to 100 (best)." (NCT00304031)
Timeframe: Baseline and cycle 10 (approximately 46 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ73.3
Dose-dense Adjuvant TMZ69.7

[back to top]

Mean Change From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Mid-cyle for Cycle 4

The MDASI-BT is a 28-item patient-reported outcome measure assessing symptom severity and resulting interference with daily living in brain cancer patients. All items range from 0 (not present) to 10 (as bad as you can imagine). The symptom severity score is the average of the symptom severity items, given a specified minimum numbers were completed. A score worse than baseline by at least one is considered deterioration. Change is calculated as time point - baseline such that a positive change value indicates worse symptoms compared to baseline. (NCT00304031)
Timeframe: Baseline and mid-cycle 4 (approximately 24 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ-0.23
Dose-dense Adjuvant TMZ0.19

[back to top]

Mean Change From Baseline in MD Anderson Symptom Inventory Brain Tumor (MDASI-BT) Symptom Severity Score at Mid-cyle for Cycle 1

The MDASI-BT is a 28-item patient-reported outcome measure assessing symptom severity and resulting interference with daily living in brain cancer patients. All items range from 0 (not present) to 10 (as bad as you can imagine). The symptom severity score is the average of the symptom severity items, given a specified minimum numbers were completed. A score worse than baseline by at least one is considered deterioration. Change is calculated as time point - baseline such that a positive change value indicates worse symptoms compared to baseline. (NCT00304031)
Timeframe: Baseline and mid-cycle 1 (approximately 12 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ0.48
Dose-dense Adjuvant TMZ0.39

[back to top]

Mean Change From Baseline in EORTC QLQ-C30 Global Health Status Score at Mid-cyle for Cycle 4

"Global Health Status is calculated from two questions on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire [EORTC QLQ]-C30. The question responses range from 1 very poor to 7 excellent such that a higher response indicates better quality of life (QOL). The mean of these responses is linearly transformed to a range of 0 (worst) to 100 (best). Change is calculated as time point - baseline such that a positive change value indicates worse symptoms compared to baseline." (NCT00304031)
Timeframe: Baseline and mid-cycle 4 (approximately 24 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ-2.78
Dose-dense Adjuvant TMZ-0.72

[back to top]

Mean Change From Baseline in EORTC QLQ-C30 Global Health Status Score at Mid-cyle for Cycle 1

"Global Health Status is calculated from two questions on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire [EORTC QLQ]-C30. The question responses range from 1 very poor to 7 excellent such that a higher response indicates better quality of life (QOL). The mean of these responses is linearly transformed to a range of 0 (worst) to 100 (best). Change is calculated as time point - baseline such that a positive change value indicates worse symptoms compared to baseline." (NCT00304031)
Timeframe: Baseline and mid-cycle 1 (approximately 12 weeks)

Interventionscore on a scale (Mean)
Conventional Adjuvant TMZ-4.58
Dose-dense Adjuvant TMZ-2.63

[back to top]

Progression-free Survival (Phase II)

Progression will be defined as a > 25% increase in tumor area. Progression-free survival time was defined as the time from baseline to date of death from any cause. Patients last known to be alive are censored at date of last contact. (NCT00305864)
Timeframe: From randomization to date of progression, death, or last follow-up. Analysis occurs after all patients have been potentially followed for at least 18 months. Patients were followed up to 54.3 months.

Interventionmonths (Median)
All MGd 5mg/kg Patients (Phase I and II Arms Combined)7.6

[back to top]

Median Overall Survival (Phase II)

Survival time was defined as the time from baseline to date of death from any cause. Patients last known to be alive are censored at date of last contact. (NCT00305864)
Timeframe: From randomization to date of death or last follow-up. Analysis occurs after all patients have been potentially followed for at least 18 months. Patients were followed up to 54.3 months

InterventionMonths (Median)
All MGd 5mg/kg Patients (Phase I and II Arms Combined)15.6

[back to top]

Maximum Tolerated Dose of MGd (Phase I)

"Patients were to be followed for a minimum of 90 days from the start of radiation therapy (RT) and carefully evaluated with respect to treatment morbidity. A dose limiting toxicity (DLT) was defined as a grade 4 neurologic adverse event (AE) considered to be related to treatment occurring within 21 days of the conclusion of RT. For each dose level, up to seven patients were to be accrued to assure that there would be six eligible for treatment adverse event evaluation. A dose level of MGd was considered acceptable if no more than 1 patient of the 6 experience a DLT. If the current level was considered acceptable, then dose escalation occurred. Otherwise, the preceding dose level would be declared the maximum tolerated dose (MTD). The MTD would be used for the Phase II arm.~Rating scale: 0 = not the MTD, 1 = MTD" (NCT00305864)
Timeframe: From start of radiation therapy to 90 days,

Interventionunits on a scale (Number)
Phase I: MGd 3 mg/kg0
Phase I: MGd 4 mg/kg0
Phase I: 5 mg/kg1

[back to top]

Overall Response - Complete Response (CR), Very Good Partial Response (VGPR) and Partial Response (PR)

"The patient's best overall response obtained during Reporting Periods 1 and 2 will be scored as best response. Patients enrolled on Stratum 1 with bone marrow disease, a responder has no tumor cells detectable by routine morphology on 2 subsequent bilateral bone marrow aspirates and biopsies done at least 3 weeks apart. For patients enrolled on stratum 1 with MIBG only disease, response will be assessed using the Curie scale. Patients who have complete resolution of all MIBG positive lesions (CR) or resolution of at least one MIBG positive lesion with persistence of other lesions (PR) will be considered responders. For Stratum 2 a responder is defined to be a patient who achieves a best overall response of CR, VGPR or PR from CT/MRI scans from central review using (RECIST) Response Evaluation Criteria in Solid Tumor. A responder is defined to be a patient who achieves a best overall response of CR (Complete Response), VGPR (Very Good Partial Response) or PR (Partial Response)." (NCT00311584)
Timeframe: up to 6 courses of therapy, or about 6 months

Interventionparticipants (Number)
Disease Eval by Bone Marrow or MIBG (Irinotecan/Temozolomide)5
Disease Measurable by CT or MRI Scan (Irinotecan/Temozolomide)3

[back to top]

Response Rate (Complete and Partial Response)

"Assessment of treatment response was determined by MRI in conjunction with neurological examination and steroid requirement assessment derived from Macdonald's criteria. Complete response was defined as complete disappearance of lesion on consecutive MRI scans with stable or improved neuro exam and steroids. Partial response was defined as a 50% reduction in lesion size or that tumor burden was definitely better than prior scan with stable or improved neuro exam and steroids." (NCT00313729)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Temozolomide7

[back to top]

Safety Profile

Number of participants with treatment related grade 2-4 adverse events as defined by CTCAE 3.0 (NCT00313729)
Timeframe: Time from registration up to 13 months

InterventionParticipants (Count of Participants)
Temozolomide12

[back to top]

Time to Tumor Progression

Progressive disease was defined as definite enlargement of any existing lesion or any new lesion based on modified Macdonald's criteria. (NCT00313729)
Timeframe: time from registration until date of the first documented progression, an average of 1 year

Interventionyears (Median)
Temozolomide3.8

[back to top]

Number of Enzyme-linked Immunosorbent Spots (ELISPOT) - Correlation Between Immunological Parameters and Efficacy - Median

Pre- and post-vaccine immune assay results (Tumor-specific T-cell Responses) are summarized on a continuous scale as mean. (NCT00323115)
Timeframe: Day 7 (pre-vaccination) and Day 42 (post-vaccination)

Interventionspots (Median)
Pre-Vaccine0
Post-Vaccine0

[back to top]

Number of Participants With Evaluable Data: Feasibility of Vaccination

To determine the feasibility of this approach, the investigators hypothesize that at least 2/3 of the patients included in the study will be evaluable, meaning that the participants would have received the 3 vaccinations with immunologic outcome parameters measured before and after vaccination. Therefore a maximum of 15 patients would be enrolled in the study to obtain 10 evaluable patients. If after enrolling 15 patients the investigators are unable to obtain 10 evaluable patients, the investigators would consider this approach not feasible. (NCT00323115)
Timeframe: Through enrollment, approximately 2 years

InterventionParticipants (Count of Participants)
Vaccine10

[back to top]

Evaluation of T Cell Characteristics

Peripheral blood obtained before starting radiation/ temozolomide (TMZ), and at first and second leukapheresis will be used to do lymphocyte phenotyping. We will determine percentages of CD3+/CD8+/CD45RO+ (memory T-cells), CD3+/CD8+/CD28- (CD8 suppressor T cell phenotype), and CD4+/CD25+ cells at those 3 time points. An anti-human Foxp3 antibody will be used to determine if the CD4+/CD25+ cells are T regulatory cells (TREG) and how the compartmental shift correlates with immunoresponse by other immune parameters as well as to efficacy. (NCT00323115)
Timeframe: Before starting radiation/Temozolomide and at Day 7 and Day 42.

,,
Interventionpercentage of cells (Mean)
% of CD3+/CD8+/CD45RO+ (memory T-cells)% CD3+/CD8+/CD28- (CD8 suppressor T cell phenotype%CD4+/CD25+ cells% of are T regulatory cells (TREG)
Vaccine - Before Starting Radiation/TMZ30.834.614.94.0
Vaccine - First Leukapheresis25.830.514.13.5
Vaccine - Second Leukapheresis19.323.711.81.2

[back to top]

Number of Participants With Significant Difference in Tumor Volume Size Pre- and Postvaccination: Neuroimaging and Tumor Assessment

Patients with evidence of evaluable enhancing disease on contrast-enhanced MRI performed within four weeks of study entry will be evaluated for response rate. Patients will be evaluated for objective tumor assessments by gadolinium-enhanced magnetic resonance imaging (Gd-MRI). Comparisons of objective assessments, excluding progressive disease, are based upon major changes in tumor size on the Gd-MRI compared to the baseline scan. Determination of progressive disease is based upon comparison to the previous scan with volumetric analysis. (NCT00323115)
Timeframe: baseline and 4 weeks

InterventionParticipants (Count of Participants)
Vaccine0

[back to top]

Overall Survival Duration: Efficacy Parameters

Overall survival will also be followed. Survival will be assessed from the date of surgery to the date of patient death, due to any cause, or to the last date the patient was known to be alive. (NCT00323115)
Timeframe: Approximately 42 months

InterventionMonths (Median)
Vaccine28

[back to top]

Progression Free Survival (PFS)

Progression-free survival will be assessed for each patient as the time from surgery until the patient reaches objective disease progression by MRI criteria. Death will be regarded as a progression event in those patients that die before disease progression. Patients without documented objective progression at the time of the analysis will be censored at the date of their last objective tumor assessment. Since disease free survival and overall survival are secondary endpoints all patients will be followed until death or for a period of 5 years following enrollment. (NCT00323115)
Timeframe: Approximately 42 months

InterventionMonths (Median)
Vaccine9.5

[back to top]

Frequency of CD4+ and CD8+ T Cells - the Proportion of Cells in the Parent Population Responding to Glioblastoma Multiforme (GBM) - Mean

Pre- and post-vaccine immune assay results (Tumor-specific T-cells ) are summarized on a continuous scale as mean. (NCT00323115)
Timeframe: Day 7 (pre-vaccination) and Day 42 (post-vaccination)

,
Interventionproportion of cells (Mean)
Precursor frequency of CD4+ T cellsPrecursor frequency of CD8+ T cells
Post-Vaccine0.010.003
Pre-Vaccine0.0050.001

[back to top]

Frequency of CD4+ and CD8+ T Cells - the Proportion of Cells in the Parent Population Responding to Glioblastoma Multiforme (GBM) - Median

Pre- and post-vaccine immune assay results (Tumor-specific T-cells ) are summarized on a continuous scale as median. (NCT00323115)
Timeframe: Day 7 (pre-vaccination) and Day 42 (post-vaccination).

,
Interventionproportion of cells (Median)
Precursor frequency of CD4+ T cellsPrecursor frequency of CD8+ T cells
Post-Vaccine0.010.001
Pre-Vaccine0.0030.001

[back to top]

Percentage of Tumor-specific T-cells - Correlation Between Immunological Parameters and Efficacy- Mean

Pre- and post-vaccine immune assay results (Tumor-specific T-cell Responses) are summarized on a continuous scale as median. IFN = interferon. (NCT00323115)
Timeframe: Day 7 (pre-vaccination) and Day 42 (post-vaccination)

,
Interventionpercentage of cells (Mean)
Percentage of CD4+ proliferating and IFNPercentage of CD8+ proliferating and IFN
Post-Vaccine0.880.92
Pre-Vaccine0.380.45

[back to top]

Percentage of Tumor-specific T-cells - Correlation Between Immunological Parameters and Efficacy- Median

Pre- and post-vaccine immune assay results (Tumor-specific T-cell Responses) are summarized on a continuous scale as median. (NCT00323115)
Timeframe: Day 7 (pre-vaccination) and Day 42 (post-vaccination)

,
Interventionpercentage of cells (Median)
Percentage of CD4+ proliferating and IFNPercentage of CD8+ proliferating and IFN
Post-Vaccine0.250.25
Pre-Vaccine0.150.27

[back to top]

Tumor-specific Cytotoxic T-cell Response

MRI & pheresis post vaccine (NCT00323115)
Timeframe: Day 42

Intervention10^9 cells/L (Median)
No. CD4+No. CD8+
Vaccine0.4960.4836

[back to top]

Number of Adverse Events: Toxicity Profile of Intra-nodal DC/Tumor Lysate Vaccination

Adverse events attributed to vaccination. Collected and attributed adverse events at each study visit; monitored participants for adverse events for two hours following vaccination procedure. (NCT00323115)
Timeframe: Until death or approximately 24 months after diagnosis

Interventionattributable adverse events (Number)
Vaccine1

[back to top]

Number of Enzyme-linked Immunosorbent Spots (ELISPOT) - Correlation Between Immunological Parameters and Efficacy - Mean

Pre- and post-vaccine immune assay results (Tumor-specific T-cell Responses) are summarized on a continuous scale as mean. (NCT00323115)
Timeframe: Day 7 (pre-vaccination) and Day 42 (post-vaccination)

Interventionspots (Mean)
Pre-Vaccine1.40
Post-Vaccine44.2

[back to top]

Analyze Patients Time to Progression

"Responses to treatment was determined by comparing new enhanced MRI scans with those obtained at the previous evaluation (i.e., 2 treatment cycles ago) or with the pre-IA chemotherapy baseline scan, if it is the first follow-up MRI scan during treatment.~MRI is the neuro-imaging modality of choice, since it is more accurate than CT for small tumors, multiple tumors, and tumors in the posterior fossa.58 The methodology used (techniques and equipment) must be identical for all scans. Lesions should be measured as the largest diameter seen on scan and the largest diameter perpendicular to that dimension." (NCT00362817)
Timeframe: up to 60 weeks

Interventionweeks (Mean)
Temozolomide & Intra-Arterial (IA) Carboplatin22.6

[back to top]

Determine the Overall Survival of Patients

From the time of protocol initiation (NCT00362817)
Timeframe: up to 64 weeks

Interventionweeks (Mean)
Temozolomide & Intra-Arterial (IA) Carboplatin25.2

[back to top]

The Incidence and Severity of Centeral Nervous System (CNS) Toxicities

To determine the incidence and severity of CNS toxicity in patients treated with intra-arterial carboplatin and oral temozolomide. (NCT00362817)
Timeframe: up to 24 weeks

Interventionpatients (Number)
Temozolomide & Intra-Arterial (IA) Carboplatin0

[back to top]

Determine the Cause of Death of Patients After Treatment

To determine the cause of death (i.e., CNS tumor versus systemic disease progression) in patients after treatment. (NCT00362817)
Timeframe: up to 1 year

Interventionpatients (Number)
CNS tumorSystemic disease progression
Temozolomide & Intra-Arterial (IA) Carboplatin07

[back to top]

Affects of Response Rate of Chemotherapy With Intra-arterial Carboplatin and Oral Temozolomide

Response was evaluated by MRI Criteria (MacDonald Criteria). The MacDonald criteria for determining tumor progression is determined through assessing the increase in size of an enhancing tumor on consecutive MRI scans and clinical assessment. Complete response occurs when there is a disappearance of all enhancing tumor on consecutive MRI scans at least one month apart. Partial response occurs at a >50% reduction in size of enhancing tumor on consecutive MRI scans at least one month apart. Progressive disease occurs when there is a >25% increase in size of enhancing tumor on consecutive MRI scans. Stable disease occurs in all remaining situations. (NCT00362817)
Timeframe: up to 1 year

Interventionpercentage of patients with response (Number)
Temozolomide & Intra-Arterial (IA) Carboplatin42.8

[back to top]

Percentage of Participants Surviving at Six Months of Treatment Without Evidence of Disease Progression.

Progression-free survival as determined by Kaplan-Meier method. (NCT00392171)
Timeframe: 6 months

InterventionPercentage of Participants (Number)
Anaplastic Glioma (n=28)Early Glioblastoma Multiforme (GBM) (n=33)Extended Glioblastoma Multiforme (GBM) (n=27)Rechallenge Glioblastoma Multiforme (GBM) (n=28)
Temozolomide35.727.37.435.7

[back to top]

Number of Participants Undergoing Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy (MRI/MRS) for Clinical Evaluation at Baseline

Number of patients having MRI/MRS for clinical evaluation with baseline assessment. (NCT00402116)
Timeframe: Each radiologic assessment (up to 12 cycles, 28 days per cycle)

InterventionParticipants (Count of Participants)
Phase 2 - 250 mg35

[back to top]

Phase 1 and 2- Pharmacokinetics: Area Under the Concentration-Time Curve During 1 Dosing Interval at Steady State (AUCτ,ss) for Enzastaurin, LY326020 and Total Analyte (Enzastaurin + LY326020) When Enzastaurin Administered With or Without Temozolomide

AUCτ,ss was calculated using concentration versus time data of enzastaurin, LY326020, and total analyte (enzastaurin + LY326020). Data are reported as Geometric Mean and Geometric Coefficient of Variation (%). (NCT00402116)
Timeframe: Phase 1, Cycle 1 Day 22, Cycle 2 Day 5 of a 28 day Cycle; Phase 2, Cycle 1 Day 22 of a 28 day Cycle

,,,,
Interventionnmol•h/L (Geometric Mean)
EnzastaurinLY326020Total Analyte
Enzastaurin 250 mg + Temozolomide5270738012800
Enzastaurin 250 mg Phase 15390726012800
Enzastaurin 250 mg Phase 277201280021400
Enzastaurin 500 mg16600424021300
Enzastaurin 500 mg + Temozolomide10100865018900

[back to top]

Phase 1 - Number of Participants With Adverse Events (AEs)

Summaries of serious AEs (SAEs) and all other non-serious AEs are located in the Reported Adverse Event Module. (NCT00402116)
Timeframe: Every cycle (up to 12 cycles, 28 days per cycle)

InterventionParticipants (Count of Participants)
Phase 1- Cohort 1 (250 mg Enzastaurin)0
Phase 1 Cohort 2 (500 mg Enzastaurin )1

[back to top]

M.D. Anderson Symptom Inventory - Brain Tumor (MDASI-BT)

General and brain tumor-specific symptoms each assessed on a scale of 0 to 10, with a higher score representing higher symptom burden. The 4 symptom scales reported as changing over the course of the study are listed here. (NCT00402116)
Timeframe: Every cycle (up to 12 cycles, 28 days per cycle)

,,,
Interventionunits on a scale (Mean)
BaselineRadiation therapy visitCycle 1Cycle 2Cycle 3Cycle 4Cycle 5Cycle 6Cycle 7Cycle 8Cycle 9Cycle 10Cycle 11Cycle 12
Enzastaurin 250 mg Phase 1 and 2 Combined-Appetite0.31.01.61.81.41.00.60.70.70.51.00.40.20.4
Enzastaurin 250 mg Phase 1 and 2 Combined-Concentration1.21.41.81.91.51.51.61.21.31.11.11.20.91.2
Enzastaurin 250 mg Phase 1 and 2 Combined-Memory1.82.22.42.52.01.92.11.61.41.41.81.51.61.2
Enzastaurin 250 mg Phases 1 and 2 Combined-Fatigue2.43.33.63.73.22.63.32.92.82.62.81.93.01.7

[back to top]

Phase 1 and 2 - Progression-Free Survival (PFS)

PFS was defined as the time from date of first dose to the first observation of disease progression, or death due to any cause. (NCT00402116)
Timeframe: Baseline to measured progressive disease (up to 12 cycles, 28 days per cycle)

Interventionmonths (Median)
250 mg Enzastaurin Phases 1 and 2 Combined10.6

[back to top]

Phase 1- Pharmacokinetics (PK): Maximum Observed Drug Concentration During 1 Dosing Interval at Steady State (Cmax,ss) for Enzastaurin, LY326020, and Total Analyte (Enzastaurin + LY326020) When Enzastaurin Administered With or Without Temozolomide

Cmax,ss was calculated using concentration versus time data of enzastaurin, LY326020, and Total Analyte (enzastaurin + LY326020) when 250 mg or 500 mg enzastaurin was administered alone or with 75 mg/m^2 temozolomide. Data are reported as Geometric Mean and Geometric Coefficient of Variation (%). (NCT00402116)
Timeframe: Cycle 1 Day 22, Cycle 2 Day 5, 28 days per Cycle

,,,
Interventionnanomole per liter (nmol/L) (Geometric Mean)
EnzastaurinLY326020Total Analyte
Enzastaurin 250 mg504370853
Enzastaurin 250 mg + Temozolomide458392821
Enzastaurin 500 mg13501981570
Enzastaurin 500 mg + Temozolomide7194651010

[back to top]

Functional Assessment of Cancer Therapy - Brain (FACT-Br)

Total FACT-Br score includes physical well-being, social/family well-being, emotional well-being, functional well-being and additional concerns related to brain tumors. The score ranges 0 to 200, with a higher score representing better quality of life. (NCT00402116)
Timeframe: Every cycle (up to 12 cycles, 28 days per cycle)

Interventionunits on a scale (Mean)
Baseline visitRadiation therapy visitCycle 1Cycle 2Cycle 3Cycle 4Cycle 5Cycle 6Cycle 7Cycle 8Cycle 9Cycle 10Cycle 11Cycle 12
250 mg Enzastaurin Phases 1 and 2 Combined153.5156.1151.9152.0158.9158.8158.3159.0164.1163.6162.0159.3158.2163.5

[back to top]

Phase 1 and Phase 2 - Overall Survival (OS)

OS is the time from surgical diagnosis to the date of death from any cause. For participants who were alive, OS was censored at the last contact. (NCT00402116)
Timeframe: Baseline to death from any cause (Up to 48 weeks)

Interventionmonths (Median)
250 mg Enzastaurin Phases 1 and 2 Combined18.3

[back to top]

Phase 1- Determination of the Maximum Tolerated Dose (MTD) of Enzastaurin

Phase 1- dose escalation of enzastaurin in 2 cohorts up to 6 participants each in order to assess MTD. After radiation/enzastaurin 250 mg per day/temozolomide 75 mg/m^2 therapy, if no more than 1 of 6 patients experienced a dose-limiting toxicity (DLT) or tumor progression, participants completed one 28-day cycle. If no significant toxicity after the first cycle, participants received subsequent cycles. If no more than 1 of the 6 patients treated at 250 mg of enzastaurin experienced a DLT, up to 6 more patients could be entered at escalated dose cohort of enzastaurin (500 mg). (NCT00402116)
Timeframe: Until MTD can be determined (up to 12 cycles, 28 days per cycle)

Interventionmilligrams (mg) (Number)
Phase 1 Participants250

[back to top]

Phase 2 - Number of Participants With Adverse Events (AEs)

Summaries of serious AEs (SAEs) and all other non-serious AEs are located in the Reported Adverse Event Module. (NCT00402116)
Timeframe: Every cycle (28 days per cycle)

InterventionParticipants (Count of Participants)
Phase 2 - 250 mg Enzastaurin22

[back to top]

Phase 1 and 2: Association Between Biomarkers and Clinical Outcome

Phosphorylated-S6 (pS6) ribosomal protein is a biomarker that's being investigated as a potential marker for clinical outcome using 2211 or 2215 antibody to pS6. Reported here are the hazard ratios and 95% confidence intervals (CIs) for participants for whom an pS6 immunohistochemistry (IHC) score was available. The IHC assays were scored using a 0 to +3 scoring system (no positive staining was scored 0; at least 25% immunoreactivity of cells was scored +1; 26% to 75% was scored +2; and 76% or greater was scored +3). Hazard ratio (HR) > 1 indicates worse outcome for that IHC score. (NCT00402116)
Timeframe: Baseline, Cycle 2, end of study (up to 12 cycles, 28 days per cycle)

InterventionHazard ratio (Number)
S6 2211 scoreS6 2215 score
250 mg Enzastaurin Phases 1 and 2 Combined1.701.69

[back to top]

Percentage of Participants With Objective Response of Complete Response or Partial Response

Percentage of participants with objective response based assessment of confirmed complete response (CR) or confirmed partial response (PR). CR persisted on repeat imaging study at least (≥) 4 weeks after initial documentation of response. PR, for bidimensionally measurable disease, was a decrease by ≥50% of the sum of the products of the largest perpendicular diameters of all measurable lesions as determined by 2 observations not less than 4 weeks apart. Best overall response recorded any time while the participant was receiving treatment. External Response Review Committee (ERRC) assessment. (NCT00404495)
Timeframe: Baseline to 1 Year (medulloblastoma), Baseline to 6 Weeks (high-grade glioma)

Interventionpercentage of participants (Number)
Temozolomide + Irinotecan for Medulloblastoma32.6
Temozolomide + Irinotecan for High-Grade Glioma0

[back to top]

Percentage of Participants With Objective Response of Complete Response or Partial Response, Investigator's Assessment

Percentage of participants with objective response based assessment of confirmed CR or confirmed PR. CR persisted on repeat imaging study ≥4 weeks after initial documentation of response. PR, in case of bidimensionally measurable disease, was a decrease by ≥50% of the sum of the products of the largest perpendicular diameters of all measurable lesions as determined by 2 observations not less than 4 weeks apart. Best overall response could be recorded any time while the participant was receiving treatment. Investigator's assessment. (NCT00404495)
Timeframe: Baseline to 1 Year (medulloblastoma), Baseline to 6 Weeks (high-grade glioma)

Interventionpercentage of participants (Number)
Temozolomide + Irinotecan for Medulloblastoma34.9
Temozolomide + Irinotecan for High-Grade Glioma11.8

[back to top]

Time to Treatment Failure (TTF)

TTF was defined as the time from the date of first dose of study treatment to the date of the first documentation of progressive disease (PD), the date of treatment discontinuation except completion of treatment, or date of death due to cancer. Investigator's assessment. (NCT00404495)
Timeframe: Baseline to Date of Treatment Failure (Up to 1 Year)

Interventionmonths (Median)
Temozolomide + Irinotecan for Medulloblastoma3.8
Temozolomide + Irinotecan for High-Grade Glioma1.6

[back to top]

Time to Tumor Progression (TTP)

TTP was defined as the time in months from start of study treatment to first documentation of objective tumor progression or death due to cancer, whichever came first. TTP was calculated as (first event date minus the date of first dose of study medication plus 1) divided by 7 multiplied by 4.33. Tumor progression was determined from oncologic assessment data (where data met the criteria for PD). Investigator's assessment. (NCT00404495)
Timeframe: Baseline to Date of Progression (Up to 1 Year)

Interventionmonths (Median)
Temozolomide + Irinotecan for Medulloblastoma5.6
Temozolomide + Irinotecan for High-Grade Glioma1.6

[back to top]

Duration of Response

Median duration (50%) of tumor response for participants with objective disease response: who have not progressed or died due to any cause; with a response and subsequent progression or death due to any cause for duration of response (DR). DR was defined as time from start of first documented objective tumor response (CR or PR) to first documented objective tumor progression or death due to any cause, whichever occurred first. DR (calculated in Weeks) = (the end date for DR minus first subsequent confirmed CR or PR plus 1) divided by 7. Investigator's assessment. (NCT00404495)
Timeframe: Baseline to Date of Tumor Response (Up to 1 Year)

Interventionweeks (Median)
Temozolomide + Irinotecan for Medulloblastoma22.4
Temozolomide + Irinotecan for High-Grade Glioma36.3

[back to top]

Overall Survival (OS)

Time in months from the start of study treatment to date of death due to any cause. OS was calculated as (the death date minus the date of first dose of study medication plus 1) divided by 7 multiplied by 4.33. Death was determined from adverse event data (where outcome was death) or from follow-up contact data (where the participant current status was death). Investigator's assessment. (NCT00404495)
Timeframe: Baseline to Date of Death (Up to 1 Year After Treatment)

Interventionmonths (Median)
Temozolomide + Irinotecan for Medulloblastoma16.7
Temozolomide + Irinotecan for High-Grade Glioma9.4

[back to top]

Tumor Responses (Complete and Partial Response)

"Tumor response rate was based on Response Evaluation Criteria in Solid Tumors (RECIST).~Complete Response (CR): Disappearance of all target lesions.~Partial Response (PR): At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD." (NCT00423150)
Timeframe: From start of treatment until participant's disease progression, intolerable toxicity or death, which ever comes first

InterventionParticipants (Number)
Number of participants with Complete ResponseNumber of participants with Partial Response
Temozolomide05

[back to top]

Tolerability: Number of Participants Discontinuing Treatment Due to Adverse Events (AE)

An AE was defined as any event which was adverse, including what were commonly described as adverse or undesirable experiences, adverse events, adverse reactions, side effects, or death due to any cause associated with, or observed in conjunction with the use of a drug, biological product, or device in humans, whether or not considered related to the use of that product. Additionally, any event which was associated with, or observed in conjunction with product overdose whether accidental or intentional, or product abuse and/or withdrawal was also considered an AE. (NCT00424554)
Timeframe: 12 months

Interventionparticipants (Number)
Temozolomide (TMZ)0
No Intervention0

[back to top]

MethylGuanine-DNA MethylTransferase [MGMT] Activity Measured From the Tumor Tissue During Surgery

An experimental assay was developed to measure MGMT levels. (NCT00424554)
Timeframe: 14 days

Interventionfmol/mg of proteins (Mean)
Temozolomide (TMZ)333.7
No Intervention105.1

[back to top]

Safety: Number of Participants Who Experienced Grade 3 or 4 Toxicities

"Grade 3 was defined as severe per Common Terminology Criteria for Adverse Events (CTCAE).~Grade 4 was defined as life-threatening per CTCAE." (NCT00424554)
Timeframe: 12 months

,
Interventionparticipants (Number)
Grade 3Grade 4
No Intervention00
Temozolomide (TMZ)40

[back to top]

Change in Perfusion MRI Markers at Week 8 as Predictors of 12mo Overall Survival (OS)

To assess the potential role of perfusion MRI as a prognostic indicator for 12-month OS based on the change in MRI markers evaluated before treatment and 8 weeks following initiation of protocol treatment. Percent change in mean tumor relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) MRI normalized to white matter (nRCBV) and standardized rCBV (sRCBV) between baseline and week 8 are the prognostic indicators. (NCT00433381)
Timeframe: Baseline and 8 weeks

InterventionArea Under the Curve (AUC) (Number)
nRCBV (%change @ week8)sRCBV (%change @ week8)
DSC 8-week Participants0.4700.561

[back to top]

Agreement Between Local Interpretation and Central Interpretation of Standard MRI

Local and central interpretations of the standard MRI were assessed for progression and survival at all available imaging (baseline visit, week 2, and after every 2 cycles of treatment, and at termination of treatment). Patients who suffer clinical progression without radiographic confirmation of progression were considered to have progressive disease in determination of PFS-6. Subjects participated in the MR substudies regardless of therapeutic intervention (NCT00433381)
Timeframe: baseline visit, week 2, after every 2 cycles of treatment, and at termination of treatment

InterventionParticipants (Count of Participants)
Local Read72402959Central Read72402959
PFS <= 6moPFS >= 6mo
Standard MRI Local and Central Evaluation62
Standard MRI Local and Central Evaluation41
Standard MRI Local and Central Evaluation55
Standard MRI Local and Central Evaluation48

[back to top]

Change in Perfusion MRI Markers at Week 16 as Predictors of 12mo Overall Survival (OS)

To assess the potential role of perfusion MRI as a prognostic indicator for 12-month OS based on the change in MRI markers evaluated before treatment and 16 weeks following initiation of protocol treatment. Percent change in mean tumor relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) MRI normalized to white matter (nRCBV) and standardized rCBV (sRCBV) between baseline and week 16 are the prognostic indicators. (NCT00433381)
Timeframe: Baseline and 16 Weeks

InterventionArea Under the Curve (AUC) (Number)
nRCBV (%change @ week16)sRCBV (%change @ week16)
DSC 16-week Participants0.7620.905

[back to top]

Accuracy of Local PFS 6-mo Interpretation Using Central Review PFS-6 as the Reference Standard

Local reads were treated as the test and central reads were treated as the reference standard. Thus, a participant meeting the definition of progression on any standard MRI central interpretation (baseline visit, week 2, after every 2 cycles of treatment, and at termination of treatment) was considered positive for PFS-6. Therefore, a true positive is defined as a positive local interpretation for a subject with a positive central read. (NCT00433381)
Timeframe: baseline visit, week 2, after every 2 cycles of treatment, and at termination of treatment

InterventionParticipants (Count of Participants)
Local Read (Test)72402959Central Read (Reference)72402959
PFS-6 NegativePFS-6 Positive
Standard MRI Local and Central Evaluation62
Standard MRI Local and Central Evaluation41
Standard MRI Local and Central Evaluation48
Standard MRI Local and Central Evaluation55

[back to top] [back to top]

Count/Percentage of Patients Progression-free at 6 Months for Bevacizumab and Irinotecan Hydrochloride Arm

Progression defined as ≥ 25% increase in the size of enhancing tumor or any new tumor; or neurologically worse, and steroids stable or increased. Percentage is calculated by taking the number of patients who have survived 6 months without progression of study disease after study registration in the numerator. The denominator consists of all patients except those who were found retrospectively to be ineligible or who were lost to follow-up after less than 6 months. (NCT00433381)
Timeframe: From randomization to six months.

InterventionParticipants (Count of Participants)
Arm II (Bevacizumab and Irinotecan Hydrochloride)22

[back to top]

Count/Percentage of Patients Progression-free at 6 Months for Bevacizumab and Temozolomide Arm

Progression defined as ≥ 25% increase in the size of enhancing tumor or any new tumor; or neurologically worse, and steroids stable or increased. Percentage is calculated by taking the number of patients who have survived 6 months without progression of study disease after study registration in the numerator. The denominator consists of all patients except those who were found retrospectively to be ineligible or who were lost to follow-up after less than 6 months. (NCT00433381)
Timeframe: From randomization to six months.

InterventionParticipants (Count of Participants)
Arm I (Bevacizumab and Temozolomide)23

[back to top]

Change in Perfusion MRI Markers at Week 2 as Predictors of 12mo Overall Survival (OS)

To assess the potential role of perfusion MRI as a prognostic indicator for 12-month OS based on the change in MRI markers evaluated before treatment and 2 weeks following initiation of protocol treatment. Percent change in mean tumor relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) MRI normalized to white matter (nRCBV) and standardized rCBV (sRCBV) between baseline and week 2 are the prognostic indicators. (NCT00433381)
Timeframe: Baseline and 2 Weeks

InterventionArea Under the Curve (AUC) (Number)
nRCBV (%change @ week2)sRCBV (%change @ week2)
DSC 2-week Participants0.850.825

[back to top]

Toxicity as Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v 3)

Here is the number of participants with adverse events. For a detailed list of adverse events, see the adverse event module. Adverse events were assessed by the Common Terminology Criteria in Adverse Events (CTCAE)v3. (NCT00436436)
Timeframe: 18 months and 4 days

InterventionParticipants (Count of Participants)
O6-benzylguanine & Temozolomide in Glioblastoma12

[back to top] [back to top]

Number of Participants That Experienced a Dose-limiting Toxicity (DLT)

The primary outcome of Phase I of this trial was to determine the maximum tolerated dose (MTD) of ZD6474 (Vandetanib) in patients with newly-diagnosed glioblastomas multiforme (GBM) and gliosarcomas who are also receiving radiation therapy with concomitant and adjuvant temozolomide. The MTD is the dose level at which 0/6 or 1/6 patients experience a dose-limiting toxicity (DLT) with the next higher dose having at least 2/3 or 2/6 patients encountering DLT. (NCT00441142)
Timeframe: 2 years

InterventionParticipants (Number)
Phase I: Dose Level -1: RT + TMZ + Vandetanib @ 200 mg/Day3
Phase I: Dose Level -2: RT + TMZ + Vandetanib @ 100 mg/Day0

[back to top]

Median Progression-free Survival (PFS), as Calculated by the # of Months Patients Remain Progression-free

A secondary outcome of Phase II of this trial is the median progression-free survival (PFS), as calculated by the # of months patients remain progression-free (NCT00441142)
Timeframe: 3 years

Interventionmonths (Median)
Phase II: Arm A (Control Group: RT + TMZ)6.2
Phase II: Arm B (Experimental Group: RT + TMZ + Vandetanib)7.7

[back to top]

Median Overall Survival (OS) of Phase II Patients

The primary outcome of Phase II of this trial was to determine the efficacy of ZD6474 (Vandetanib) in combination with radiation therapy and concomitant and adjuvant temozolomide in patients with newly-diagnosed GBM and gliosarcomas as measured by overall survival and median survival. (NCT00441142)
Timeframe: 3 years

Interventionmonths (Median)
Phase II: Arm A (Control Group: RT + TMZ)15.9
Phase II: Arm B (Experimental Group: RT + TMZ + Vandetanib)16.6

[back to top] [back to top]

Methylation Status of the O6-methylguanine-DNA Methyltransferase Promoter

Overall survival for patients by Methylation status of the O6-methylguanine-DNA methyltransferase promoter (NCT00482677)
Timeframe: 7 years

InterventionMonths (Median)
Temozolomide13.47
Radiation7.69

[back to top]

Overall Survival

Time from date of randomization to the date of death of any causes, or censored at last known alive date. (NCT00482677)
Timeframe: 7 years

InterventionMonths (Median)
Temozolomide9.33
Radiation7.62

[back to top]

Progression-free Survival

Time from date of randomization to the date of disease progression or death whichever came first, or censored at last disease assessment date. (NCT00482677)
Timeframe: 7 years

InterventionMonths (Median)
Temozolomide5.29
Radiation3.94

[back to top]

(Phase II) Overall Survival

Number of months alive after end of study participation (NCT00486603)
Timeframe: 2 years

Interventionmonths (Median)
Phase 2: RT + TMZ + HCQ15.6

[back to top]

(Phase II) Number of Participants With Grade 3 and 4 Toxicity

Number of participants experiencing Grade 3 and 4 toxicity, as defined by CTCAE v3.0, with a possible, probable or definite relationship to HCQ, TMZ or both (NCT00486603)
Timeframe: up to 2 years

InterventionParticipants (Count of Participants)
Phase 2: RT + TMZ + HCQ22

[back to top]

(Phase I) Number of Participants Who Experienced Dose Limiting Toxicity (DLT)

Dose limiting toxicity defined as: Any DLT must be a toxicity considered at least possibly related to HCQ. DLTs will include any possibly, probably, or definitely HCQ-related Grade 3 or 4 toxicity. Known or reasonably suspected TMZ hematological toxicities will not be considered dose limiting unless the treating physician considers the toxicity to be exacerbated by HCQ. Nonhematological toxicities: Any Grades 3-4 severity (except nausea and vomiting without sufficient antiemetic prophylaxis) (NCT00486603)
Timeframe: 10 weeks

InterventionParticipants (Count of Participants)
Phase 1: RT+TMZ+HCQ - 200mg0
Phase 1: RT+TMZ+HCQ - 400mg0
Phase 1: RT+TMZ+HCQ - 600mg0
Phase 1: RT+TMZ+HCQ - 800mg3

[back to top]

Pharmocodynamics as Determined by Number of Participants With Autophagy Inhibition in Relation to Maximal Concentration (Cmax) of HCQ

Autophagy inhibition is represented by an increase in autophagic vacuoles (AV) in participants with at least 2 peripheral blood mononuclear cell samples that were amenable to EM. (NCT00486603)
Timeframe: up to 9 weeks

,
InterventionParticipants (Count of Participants)
AV IncreaseNo AV Increase
HCQ Cmax <= 1785 ng/mL1012
HCQ Cmax>1785 ng/mL126

[back to top]

PK of Hydroxychloroquine as Measured by Oral Clearance (Liters/Hour) From Central Compartment (CL/F)

The population model PK parameters do not specifically represent steady-state values, as they were determined from multiple repeated single doses taken from multiple repeated doses taken by the individual patient during their period on the study. To obtain steady state PK parameters, individual estimates were simulated from the population model. (NCT00486603)
Timeframe: up to 276 days

InterventionL/hr (Mean)
Phase 2: RT + TMZ + HCQ11.85

[back to top]

PK of Hydroxychloroquine as Measured by Volume of Distribution of Central Compartment (V/F)

The population model PK parameters do not specifically represent steady-state values, as they were determined from multiple repeated single doses taken from multiple repeated doses taken by the individual patient during their period on the study. To obtain steady state PK parameters, individual estimates were simulated from the population model. (NCT00486603)
Timeframe: up to 276 days

InterventionLiters (Mean)
Phase 2: RT + TMZ + HCQ483.96

[back to top]

(Phase I) Maximum Tolerated Dose (MTD) of Hydroxychloroquine (HCQ)

Number of participants who tolerated doses of HCQ without dose limiting toxicity. The highest dose at which participants did not experience dose limiting toxicity was determined as the MTD. (NCT00486603)
Timeframe: 10 weeks

InterventionParticipants (Count of Participants)
200mg400mg600mg800mg
Phase 1 - Dose Finding3730

[back to top]

PK of Hydroxychloroquine as Measured by First-order Absorption Rate Constant (Ka)

The population model PK parameters do not specifically represent steady-state values, as they were determined from multiple repeated single doses taken from multiple repeated doses taken by the individual patient during their period on the study. To obtain steady state PK parameters, individual estimates were simulated from the population model. (NCT00486603)
Timeframe: up to 276 days

Interventionhours (Mean)
Phase 2: RT + TMZ + HCQ0.51

[back to top]

PK of Hydroxychloroquine as Measured by Distribution Volume of Peripheral Compartment (V2/F)

The population model PK parameters do not specifically represent steady-state values, as they were determined from multiple repeated single doses taken from multiple repeated doses taken by the individual patient during their period on the study. To obtain steady state PK parameters, individual estimates were simulated from the population model. (NCT00486603)
Timeframe: up to 276 days

InterventionLiters (Mean)
Phase 2: RT + TMZ + HCQ963

[back to top]

Pharmacokinetics (PK) of Hydroxychloroquine as Measured by Lag Time (Tlag)

The population model PK parameters do not specifically represent steady-state values, as they were determined from multiple repeated single doses taken from multiple repeated doses taken by the individual patient during their period on the study. To obtain steady state PK parameters, individual estimates were simulated from the population model. (NCT00486603)
Timeframe: up to 276 days

Interventionhour (Mean)
Phase 2: RT + TMZ + HCQ1.06

[back to top]

Progression-free Survival (PFS) Rate at 6 Months

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. (NCT00498927)
Timeframe: at 6 months

Interventionpercentage of participants (Number)
Temozolomide19

[back to top]

Overall Survival

All patients will have their tumor measurements recorded at baseline and at the time of each MRI scan. Lesions must be measured in two dimensions. The dose of gadolinium must be held constant from scan to scan. Macdonald criteria will be used for assessment of tumor response. (NCT00498927)
Timeframe: 2 years

Interventionmonths (Median)
Temozolomide7

[back to top]

Incidence and Severity of CNS Hemorrhage and Systemic Hemorrhage

Number of participants experiencing a Central Nervous System (CNS) hemorrhage or systemic hemorrhage (NCT00501891)
Timeframe: 27 months

Interventionparticipants (Number)
CNS HemorrhageSystemic Hemorrhage
Bevacizumab and Metronomic Temozolomide00

[back to top]

Incidence of Grade ≥ 4 Hematologic or Grade ≥ 3 Non-hematologic Toxicity

Number of participants experiencing a grade ≥4 hematologic or grade ≥3 non-hematologic toxicity (NCT00501891)
Timeframe: 27 months

Interventionparticipants (Number)
Grade ≥ 4 hematologic toxicitiesGrade ≥ 3 non-hematologic toxicities
Bevacizumab and Metromonic Temozolomide014

[back to top]

6-Month Progression-free Survival

Percentage of participants surviving six months from the start of study treatment without progression of disease. PFS was defined as the time from the date of study treatment initiation to the date of the first documented progression according to the Macdonald criteria, or to death due to any cause. [Optional: Macdonald criteria are standard criteria in neuro-oncology. Tumor assessment was made according to the adapted MacDonald criteria based on the combined evaluation of: 1) assessment of the MRI scan for measurable, evaluable, and new lesions (made by the independent external expert too), 2) overall assessment of neurological performance (made by the investigator), 3) concomitant steroid use (as reported by the investigator).] (NCT00501891)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Bevacizumab and Metronomic Temozolomide18.8

[back to top]

Response Rate

The number of participants with complete or partial response as determined by a modification of the Macdonald criteria. Complete response was defined as complete disappearance on MR/CT of all enhancing tumor and mass effect, off all corticosteroids (or receiving only adrenal replacement doses), accompanied by a stable or improving neurologic examination, and maintained for at least 4 weeks. Partial Response was defined as greater than or equal to 50% reduction in tumor size on MR/CT by bi-dimensional measurement, on a stable or decreasing dose of corticosteroids, accompanied by a stable or improving neurologic examination, and maintained for at least 4 weeks. (NCT00501891)
Timeframe: 27 months

InterventionNumber of participants (Number)
Bevacizumab and Metronomic Temozolomide9

[back to top]

6 Month Progression-free Survival for Participants With Glioblastoma

Progression-free Survival (PFS) at 6 months measured as percentage of participants that are alive and progression-free at 6 months (glioblastoma multiforme). A combination of neurological examination and MRI brain scan used to define overall response or progression. (NCT00504660)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Participants With Glioblastoma Multiforme14

[back to top]

12 Month-progression-free Survival for Participants With Anaplastic Tumors

Progression-free Survival (PFS) at 12 months measured as percentage of participants that are alive and progression-free at 12 months (anaplastic tumors). A combination of neurological examination and MRI brain scan used to define overall response or progression. (NCT00504660)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Participants With Recurrent Anaplastic Glioma44

[back to top]

Time to Progression (TTP)

TTP defined as the time from date of first dose of study medication to first documentation of objective tumor progression in days. Response evaluation by Response Evaluation Criteria in Solid Tumors (RECIST) done following 2 cycles and 3 cycles. Progression is defined, using RECIST, as a measurable increase in the smallest dimension of any target or non-target lesion, or the appearance of new lesions, since baseline. (NCT00505635)
Timeframe: Following two 21 day cycles until disease progression

Interventiondays (Geometric Mean)
Biochemotherapy With Temozolomide93.2

[back to top]

Optimal Doses of Temozolomide and Bortezomib (Phase I)

The optimal biologic dose (OBD) defined as the dose that achieves the greatest degree of inhibition of NF-κB activation in peripheral blood mononuclear cells when co-administered with Temozolomide (NCT00512798)
Timeframe: up to 42 days

Interventionmg/m2 (Number)
bortezomibtemozolomide
Phase I1.375

[back to top]

Patients With Clinical Anti-tumor Activity (Phase I)

Per RECIST criteria v. 1.0: measurable lesions: complete response (CR) disappearance of target lesions, partial response (PR) > 30% decrease in the sum of the longest diameter (LD) of target lesions, progressive disease (PD) > 20% increase in the sum of the LD of target lesions or appearance of new lesions, stable disease (SD) neither sufficient decrease nor increase of the sum of smallest sum of the LD of target lesions (NCT00512798)
Timeframe: every 9 weeks up to a maximum of 54 weeks

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Phase I04114

[back to top]

Patients With Inhibition of NF-kB (Phase II)

Patient with a minimum of 50% reduction from baseline on day 8 or day 29 in NF-kB, measured by picograms/milliliter in peripheral mononuclear blood cells (NCT00512798)
Timeframe: at baseline, on day 8 and on day 29

Interventionparticipants (Number)
Phase II0

[back to top]

Patients With Inhibition in NF-kB Activation (Phase I)

Patient with a minimum of 50% reduction from baseline on day 8 or day 29 in NF-kB, measured by picograms/milliliter in peripheral mononuclear blood cells (NCT00512798)
Timeframe: at baseline, on day 8 and on day 29

Interventionparticipants (Number)
Phase I0

[back to top]

Number of Patients With Clinical Anti-tumor Activity Phase II)

Per RECIST criteria v. 1.0: measurable lesions: complete response (CR) disappearance of target lesions, partial response (PR) > 30% decrease in the sum of the longest diameter (LD) of target lesions, progressive disease (PD) > 20% increase in the sum of the LD of target lesions or appearance of new lesions, stable disease (SD) neither sufficient decrease nor increase of the sum of smallest sum of the LD of target lesions. Patients with CR + PR + SD (NCT00512798)
Timeframe: every 9 weeks to a maximum of 54 weeks

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Phase II01522

[back to top]

9-week Progression-free Survival Rate

The primary endpoint of this trial is the 9 week PFS rate. A patient is a success if they are progression free at their cycle 2 evaluation (approximately 9 weeks post registration). All patients, who meet the eligibility criteria, sign a consent form, and start treatment will be included in the evaluation of the 9-week PFS rate (evaluable patients). The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated using the properties of the binomial distribution. If some patients are lost to follow up prior to their cycle 2 evaluation, the Kaplan-Meier method will be used to estimate the 9 week PFS rate. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00521001)
Timeframe: at 9 weeks

Interventionproportion of patients (Number)
Everolimus + Temozolomide0.44

[back to top]

Confirmed Response Rate (Complete Response and Partial Response)

Confirmed response rates will be evaluated by dividing the number of confirmed responders (i.e. patients that achieve a CR or PR on consecutive evaluations) by the total number of evaluable patients. Confidence intervals for the true response rate will be calculated using the properties of the binomial distribution. (NCT00521001)
Timeframe: Up to 5 years

Interventionpercentage of confirmed responses (Number)
Everolimus + Temozolomide8.3

[back to top]

Survival Time

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT00521001)
Timeframe: Time from registration to death due to any cause; Up to 5 years

Interventionmonths (Median)
Everolimus + Temozolomide8.6

[back to top]

Time to Disease Progression

Time to disease progression is defined as the time from registration to the earliest date documentation of disease progression. If a patient dies without a documentation of disease progression the patient will be considered to have had tumor progression at the time of their death unless there is sufficient documented evidence to conclude no progression occurred prior to death. The distribution of time to progression will be estimated using the method of Kaplan-Meier. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00521001)
Timeframe: Time from registration to the earliest date documentation of disease progression; Up to 5 years

Interventionmonths (Median)
Everolimus + Temozolomide2.4

[back to top]

Unexpected Toxicities During First 2 Cycles of Study Drug

Unexpected severe study-related adverse events (NCT00525525)
Timeframe: Within 8 weeks of initiating study therapy

InterventionEvents (Number)
Safety Lead-in Group0

[back to top]

Progression-free Survival

Progression-free survival was defined from the date of diagnosis to the date that progressive disease was first observed on imaging, or the date at which nonreversible neurologic progression or permanently increased corticosteroid requirement, death from any cause, or early discontinuation of treatment. Imaging guidelines were used to evaluate progression: (i) 25% increase in the sum of products of all measurable lesions over the smallest sum observed (over baseline if no decrease) using the same techniques as baseline; (ii) clear worsening of any assessable disease; (iii) appearance of any new lesion/site; and (iv) clear clinical worsening or failure to return for evaluation as a result of death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00525525)
Timeframe: Approximately 6 months to 1 year

Interventionmonths (Median)
Efficacy Group13.5

[back to top]

Overall Survival (OS)

Overall survival was defined from the date of diagnosis to date of death from any cause (NCT00525525)
Timeframe: Approximately 6-24 months

Interventionmonths (Median)
Efficacy Group19.8

[back to top]

Mean Total PedsQL 3.0 Scores for Child Cancer Quality of Life (QoL) Assessments (Stratum B)

QoL assessments were completed using Pediatrics Cancer Quality of Life Inventory (PedsQL v3.0). Scale range is 0-100 with higher scores reflecting better quality of life. (NCT00539591)
Timeframe: Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Number)
Week 292.8
Week 490.1
Week 893.2
Week 1279.6
End of Therapy67.4

[back to top]

Mean Total PedsQL 3.0 Scores for Parent Cancer Quality of Life (QoL) Assessments (Stratum A)

QoL assessments were completed using Pediatrics Cancer Quality of Life Inventory (PedsQL v3.0). Scale range is 0-100 with higher scores reflecting better quality of life. (NCT00539591)
Timeframe: Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Mean)
Week 273.2
Week 475.1
Week 881.4
Week 1278.7
Week 2481.6
End of Therapy85.6
6 Months After End of Therapy85.0
12 Months After End of Therapy89.1

[back to top]

Mean Total PedsQL 3.0 Scores for Parent Cancer Quality of Life (QoL) Assessments (Stratum B)

QoL assessments were completed using Pediatrics Cancer Quality of Life Inventory (PedsQL v3.0). Scale range is 0-100 with higher scores reflecting better quality of life. (NCT00539591)
Timeframe: Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Number)
Week 267.7
Week 471.4

[back to top]

Apparent Clearance (CL) of Pegylated Interferon ɑ-2B

"Pharmacokinetic (PK) analysis of pegylated ɑ-2b included only Stratum A patients who had PK studies performed.~Samples were analyzed for pegylated interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM." (NCT00539591)
Timeframe: Before first dose, and 24, 96 and 168 hours after dose during weeks 5 and 28

Interventionml/hr/kg (Median)
Peginterferon ɑ-2b/Non-Pegylated Interferon ɑ-2b19.8

[back to top]

Mean Total PedsQL 4.0 Scores for Child Quality of Life (QoL) Assessments (Stratum A)

QoL assessments were completed using Pediatrics Quality of Life Inventory (PedsQL v4.0). Scale range is 0-100 with higher scores reflecting better quality of life. PedsQL 4.0 healthy sample normative mean ± SD for child report = 83.0 ± 14.8. (NCT00539591)
Timeframe: Pretherapy; Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Mean)
Pretherapy75.5
Week 271.6
Week 477.2
Week 879.3
Week 1277.8
Week 2480.6
End of Therapy80.4
6 Months After End of Therapy87.5
12 Months After End of Therapy91.0

[back to top]

Mean Total PedsQL 4.0 Scores for Child Quality of Life (QoL) Assessments (Stratum B)

QoL assessments were completed using Pediatrics Quality of Life Inventory (PedsQL v4.0). Scale range is 0-100 with higher scores reflecting better quality of life. PedsQL 4.0 healthy sample normative mean ± SD for child report = 83.0 ± 14.8. (NCT00539591)
Timeframe: Pretherapy; Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Number)
Pretherapy90.3
Week 293.1
Week 472.8
Week 879.2
Week 1268.1
End of Therapy65.6

[back to top]

Volume of Central Compartment (Vc) of Interferon ɑ-2b

Samples were analyzed for interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM. (NCT00539591)
Timeframe: Before first dose, and 1, 2, 4, 6, 8, 12, and 24 hours postinfusion

Interventionl/m^2 (Median)
Interferon ɑ-2b25.1

[back to top]

Tumor Response Rate

Tumor response rate of stratum B1 participants was evaluated after 1 treatment course of temozolomide plus peginterferon ɑ-2b. Complete response (CR) and partial response (PR) confirmed with repeated scan at least 4 weeks apart following completion of course 1 therapy. CR defined as disappearance of all target and non-target lesions with no new lesions detected. If available, no disease must be detected by immunocytology or serum tumor markers. PR defined as at least 30% decrease in disease measurement compared to disease measurement at study entry with no new lesions detected. Progressive disease (PD) defined as at least 20% increase in the disease measurement compared to the smallest disease measurement recorded since start of treatment, or appearance of one or more new lesions. Stable disease defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD compared to smallest disease measurement since start of treatment. (NCT00539591)
Timeframe: 8 weeks

Interventionparticipants (Number)
Progressive DiseaseClinical Remission
Stratum B120

[back to top]

Mean Total PedsQL 4.0 Scores for Parent Quality of Life Assessments (Stratum A)

QoL assessments were completed using Pediatrics Quality of Live Inventory (PedsQL v4.0). Scale range is 0-100 with higher scores reflecting better quality of life. PedsQL 4.0 healthy sample normative mean ± SD for parent report = 87.6 ± 12.3. (NCT00539591)
Timeframe: Pretherapy; Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Mean)
Pretherapy70.3
Week 271.8
Week 474.4
Week 879.1
Week 1279.0
Week 2482.2
End of Therapy87.5
6 Months After End of Therapy86.0
12 Months After End of Therapy87.3

[back to top]

Mean Total PedsQL 4.0 Scores for Parent Quality of Life Assessments (Stratum B)

QoL assessments were completed using Pediatrics Quality of Live Inventory (PedsQL v4.0). Scale range is 0-100 with higher scores reflecting better quality of life. PedsQL 4.0 healthy sample normative mean ± SD for parent report = 87.6 ± 12.3. (NCT00539591)
Timeframe: Pretherapy; Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Number)
Pretherapy77.6
Week 272.2
Week 489.1

[back to top]

Median Steady State Trough Concentration of Pegylated Interferon ɑ-2B

"The pharmacokinetic (PK) analysis of pegylated ɑ-2b included only patients within Stratum A who had PK studies performed.~Samples were analyzed for pegylated interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM." (NCT00539591)
Timeframe: Before first dose, and 24, 96 and 168 hours after dose during weeks 5 and 28

Interventionpcg/ml (Median)
Peginterferon ɑ-2b/Non-Pegylated Interferon ɑ-2b52.8

[back to top]

Number of Patients Who Experience Toxicity at or Above the Target Toxicity for Stratum A Patients

"The objective was to study the feasibility and safety of administering peginterferon a-2b weekly for 48 weeks following the initial induction phase to Stratum A participants.~Accrual was suspended during the 48-week course if 2 or more of 6, 4 or more of 12, 6 or more of 18, 8 or more of 24, 10 or more of 30 participants experienced target toxicity defined as:~Grade 4 non-hematologic (non-hem) toxicity that does not resolve to ≤grade 1 within 2 weeks from the time next dose is due and is determined to be probably or definitely related to protocol therapy~Grade 4 non-hem toxicity that is NOT constitutional symptoms (fever, chills, fatigue and/or pain)~Grade 3 elevations in creatinine or BUN that are determined to be probably or definitely related to protocol therapy~Grade 4 cardiopulmonary toxicity that is determined to be probably or definitely related to protocol therapy~Grade 4 mood alteration (suicidal ideation; danger to self or others)" (NCT00539591)
Timeframe: 52 weeks

Interventionparticipants (Number)
Grade 4 non-hem toxicityGrade 4 non-hem/NOT constitutionalGrade 3 elevations in creatinine or BUNGrade 4 cardiopulmonary toxicityGrade 4 mood alteration
Peginterferon ɑ-2b/Non-pegylated Interferon ɑ-2b20001

[back to top]

Half-Life of Interferon ɑ-2b

Samples were analyzed for interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM. (NCT00539591)
Timeframe: Before first dose, and 1, 2, 4, 6, 8, 12, and 24 hours postinfusion

Interventionhours (Median)
ɑ half-lifeß half-life
Peginterferon ɑ-2b/Non-Pegylated Interferon ɑ-2b0.714.7

[back to top]

Volume of Central Compartment (Vc) of Pegylated Interferon ɑ-2B

"Pharmacokinetic (PK) analysis of pegylated ɑ-2b included only Stratum A patients who had PK studies performed.~Samples were analyzed for pegylated interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM." (NCT00539591)
Timeframe: Before first dose, and 24, 96 and 168 hours after dose during weeks 5 and 28

Interventionml/kg (Median)
Peginterferon ɑ-2b/Non-Pegylated Interferon ɑ-2b772

[back to top]

Systemic Clearance (CL) of Interferon ɑ-2B

Samples were analyzed for interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM. (NCT00539591)
Timeframe: Before first dose, and 1, 2, 4, 6, 8, 12, and 24 hours postinfusion

Interventionl/hr/m^2 (Median)
Interferon ɑ-2b15.3

[back to top]

Probability of Event-free Survival (EFS) of Stratum A Participants

The probability of EFS was estimated as time to first event (relapse, death or second malignancy). As of April 2016, 21 out of 23 participants had no events. The EFS rate was estimated by Kaplan-Meier method. (NCT00539591)
Timeframe: 3 years from diagnosis

Interventionprobability (Number)
Peginterferon ɑ-2b/Non-pegylated Interferon ɑ-2b0.913

[back to top]

Number of Patients Who Experience Toxicity at or Above the Target Toxicity for Strata B1 and B2

"The objective was to assess the safety of temozolomide administered in combination with peginterferon a-2b in Stratum B participants.~Accrual was suspended any time during therapy if 2 or more of 6, 4 or more of 12, 6 or more of 18, 8 or more of 24, 10 or more of 30 participants experienced target toxicity defined as:~Grade 4 non-hematologic (non-hem) toxicity that does not resolve to ≤grade 1 within 2 weeks from the time next dose is due and is determined to be probably or definitely related to protocol therapy~Grade 4 non-hem toxicity that is NOT constitutional symptoms (fever, chills, fatigue and/or pain)~Grade 3 elevations in creatinine or BUN that are determined to be probably or definitely related to protocol therapy~Grade 4 cardiopulmonary toxicity that is determined to be probably or definitely related to protocol therapy~Grade 4 mood alteration (suicidal ideation; danger to self or others)" (NCT00539591)
Timeframe: 52 weeks

Interventionparticipants (Number)
Temozolomide/Peginterferon ɑ-2b With Measureable Disease0
Temozolomide/Peginterferon ɑ-2b Without Measureable Disease0

[back to top]

Mean Total PedsQL 3.0 Scores for Child Cancer Quality of Life (QoL) Assessments (Stratum A)

QoL assessments were completed using Pediatrics Cancer Quality of Life Inventory (PedsQL v3.0). Scale range is 0-100 with higher scores reflecting better quality of life. (NCT00539591)
Timeframe: Weeks 2, 4, 8, 12, and 24; and End of therapy at 6 months and 12 months post

Interventionunits on a scale (Mean)
Week 271.1
Week 476.1
Week 879.2
Week 1278.5
Week 2477.1
End of Therapy77.0
6 Months After End of Therapy83.7
12 Months After End of Therapy85.4

[back to top]

BRIEF Psychological Assessment (Stratum A)

The Behavioral Rating Inventory of Executive Function (BRIEF) was administered to parents, assessing for any effects on behavior or mood in children undergoing study therapy. The global executive composite (GEC) T-score (range 0-100) is reported for the BRIEF assessment. Higher scores reflect poorer executive function. (NCT00539591)
Timeframe: Pretherapy, Week 4, Week 24, End of Therapy, and 6 Months Post End of Therapy

InterventionT score (Mean)
Pretherapy47.9
Week 450.8
Week 2448.6
End of Therapy47.6
6 Months After End of Therapy42.6

[back to top]

BASC-2 Psychological Assessment (Stratum A)

The Behavioral Assessment System for Children, 2nd Edition (BASC-2) was administered to parents, assessing for any effects on behavior or mood in children undergoing study therapy. The behavior system index (BSI) T-score (range 0-100) is reported for the BASC-2 assessment. Higher scores reflect greater behavioral problems. (NCT00539591)
Timeframe: Pretherapy, Week 4, Week 24, End of Therapy, and 6 Months Post End of Therapy

InterventionT score (Mean)
Pretherapy44.9
Week 445.9
Week 2444.2
End of Therapy47.2
6 Months After End of Therapy42.3

[back to top]

ɑ Half Life of Pegylated Interferon ɑ-2B

"Pharmacokinetic (PK) analysis of pegylated ɑ-2b included only Stratum A patients who had PK studies performed.~Samples were analyzed for pegylated interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM." (NCT00539591)
Timeframe: Before first dose, and 24, 96 and 168 hours after dose during weeks 5 and 28

Interventionhours (Median)
Peginterferon ɑ-2b/Non-Pegylated Interferon ɑ-2b24.8

[back to top]

Area Under the Curve (AUC) of Pegylated Interferon ɑ-2B

"Pharmacokinetic (PK) analysis of pegylated ɑ-2b included only Stratum A patients who had PK studies performed.~Samples were analyzed for pegylated interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM. AUC is given as Time 0 through infinity." (NCT00539591)
Timeframe: Before first dose, and 24, 96 and 168 hours after dose during weeks 5 and 28

Interventionpcg * hr/ml (Median)
Week 5 - First Dose50556
Week 28 - Steady State48480

[back to top]

Area Under the Curve (AUC) of Interferon ɑ-2b

Samples were analyzed for interferon ɑ-2b concentrations by using the VeriKine Human Interferon Alpha ELISA Kit following the manufacturer's instructions, and concentration-time data were analyzed by nonlinear-mixed effects modeling as implemented in NONMEM. AUC is given as Time 0 to infinity. (NCT00539591)
Timeframe: Before first dose, and 1, 2, 4, 6, 8, 12, and 24 hours postinfusion

Interventionpcg * hr/ml (Median)
Peginterferon ɑ-2b/Non-Pegylated Interferon ɑ-2b5026

[back to top]

Progression-free Survival

Defined as the duration of time from start of treatment to time of progression or death, whichever comes first. (NCT00544817)
Timeframe: 18 months

InterventionMonths (Median)
Combination Therapy6

[back to top]

Objective Response

"The number of patients with complete or partial responses measured from the time of initial response to documented tumor progression. Radiologic response was defined using the Macdonald criteria.~The Macdonald criteria divides response into 4 types of response based on imaging (MRI) and clinical features, as follows: 1) complete response (CR); 2) partial response (PR); 3) stable disease (SD); and 4) progression (PD).~Criteria:~CR: disappearance of all enhancing disease (measurable and non-measurable) sustained for at least 4 weeks, no new lesions. No corticosteroids, clinically stable or improved.~PR: >=50% decrease of all measurable enhancing lesions, sustained for at least 4 weeks, no new lesions. Stable or reduced corticosteroids, clinically stable or improved.~SD: does not qualify for complete response, partial response or progression. Clinically stable.~PD: >= 25% increase in enhancing lesions, any new lesions. Clinical deterioration." (NCT00544817)
Timeframe: every 8 weeks until disease progression, estimated 18 months

Interventionparticipants (Number)
Combination Therapy13

[back to top]

Overall Survival

Defined as Day 1 of protocol treatment to date of death from any cause. (NCT00544817)
Timeframe: 18 months

InterventionMonths (Median)
Combination Therapy12

[back to top]

Overall Survival Time

Overall survival: The overall survival or survival time is defined as the time from registration to death due to any cause. The distribution of overall survival will be estimated using the method of Kaplan-Meier method. (NCT00553150)
Timeframe: Up to 15 years

Interventionmonths (Median)
Phase II15.8

[back to top]

Overall Survival at 12 Months (Phase II)

"The primary endpoint is overall survival at 12 months (OS12) after entry into this study. The proportion of successes will be estimated using the binomial point estimator (number of successes divided by the total number of evaluable patients) and the binomial 95% confidence interval estimated. A patient who is evaluable and survive more than 12 months (i.e. 365 days or more) after start of therapy will be classified as a success. Patients who die within 12 months after start of therapy will be considered to have failed." (NCT00553150)
Timeframe: at 12 months

Interventionproportion of participants (Number)
Phase II0.64

[back to top]

Maximum Tolerated Dose (MTD) of Everolimus (RAD001) in Combination With Temozolomide (TMZ) and 3D-conformal Radiotherapy (RT) or Intensity-modulated Radiotherapy (IMRT) Followed by Adjuvant TMZ With or Without RAD001 (Phase I)

Patients were assessed during RT for dose-limiting toxicities (DLT), which were defined as failure to deliver greater than 75% of the planned doses of TMZ or RAD001 during RT, interruption of RT for more than 5 days because of toxicity, or the following: >= Grade 3 diarrhea or skin rash; >= Grade 4 neutropenia, leukopenia, or thrombocytopenia; >= Grade 4 hypertriglyceridemia, hypercholesterolemia, or hyperglycemia despite optimal medial management, other >= 3 non-hematologic events; or >= Grade 4 radiation dermatitis. Maximum tolerated dose (MTD) was defined a priori as the highest dose level at which 0 or 1 of 6 patients developed DLTs. The number of patients who developed DLTs are reported here by dose level, with the MTD reported in the statistical analysis section. (NCT00553150)
Timeframe: Up to 49 days

Interventionparticipants who developed DLTs (Number)
Phase I: Dose Level 01
Phase I: Dose Level 11
Phase I: Dose Level 21

[back to top]

Time to Progression (Phase II)

Time-to-disease progression is defined as the time from start of study therapy to documentation of disease progression. Patients who die without documentation of progression will be considered to have had tumor progression at the time of death unless there is documented evidence that no progression occurred before death. Patients who fail to return for evaluation after beginning therapy will be censored for progression on the last day of therapy. Patients who experience major treatment violations will be censored for progression on the date of treatment violation occurred. The time-to-progression distribution will be estimated using the Kaplan-Meier method. Progression is defined as at least a 25% increase in product of perpendicular diameters of contrast enhancement or mass or unequivocal increase in size of contrast enhancement or increase in mass effect as agreed upon independently by primary physician and quality control physicians or appearance of new lesions. (NCT00553150)
Timeframe: Up to 5 years

Interventionmonths (Median)
Phase II6.4

[back to top]

Response Rate, as Measured in Patients Receiving FLT-PET Imaging (Phase II)

The response rate is defined as the percentage of patients receiving F-fluorothymidine positron emission tomography (FLT-PET) imaging whose cancer shrinks or disappears after treatment. A reduction in standardized uptake value (SUV) of 30% or greater in the T1-post-gadolinium scan volume of interest (T1-gad VOI) or the total tumor VOI will be considered a responsive tumor. (NCT00553150)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Phase II44.4

[back to top]

Progression-free-survival at 6 Months (Phase II)

Progression-free-survival at 6 months: is the proportion of patients alive and progression-free at 6 months after start of regimen. This proportion will be estimated using the binomial point estimator and the binomial 95% confidence interval estimated. Progression is defined as at least a 25% increase in product of perpendicular diameters of contrast enhancement or mass or unequivocal increase in size of contrast enhancement or increase in mass effect as agreed upon independently by primary physician and quality control physicians or appearance of new lesions. (NCT00553150)
Timeframe: at 6 months

Interventionproportion of participants (Number)
Phase II0.52

[back to top]

Time to Disease Progression

Time to disease progression was defined as the time from registration to documentation of disease progression. Disease progression was measured according to the RECIST criteria. Progression: At least a 20 percent increase in the sum of of target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. (NCT00568451)
Timeframe: up to 2 years

InterventionDays (Median)
Overall74

[back to top]

Survival Time

Survival time was defined as the time from registration to death due to any cause. (NCT00568451)
Timeframe: up to 2 years

InterventionMonths (Median)
Overall12.5

[back to top]

Number of Participants With an Objective Tumor Status of Either a Complete Response(CR) or Partial Response (PR), According to RECIST (Response Evaluation Criteria in Solid Tumors) Criteria

"Response that was noted on 2 consecutive evaluations for at least 4 weeks apart.~CR: Disappearance of all target lesions; PR: At least a 30 percent of decrease in the sum of the longest diameter (LD) of target lesions taking as reference the baseline sum LD. Target lesions: All measurable lesions up to a maximum of 10 lesions representative of all involved organs." (NCT00568451)
Timeframe: Every other cycle of therapy (cycle=4 weeks) for the first 6 cycles of treatment

Interventionparticipants (Number)
TMZ (Previously Treated)0
TMZ (Chemo Naive)2

[back to top]

Progression-free Survival at 6-months

(NCT00575887)
Timeframe: Until progression

Interventionpercentage of participants (Number)
Temozolomide17.3

[back to top]

To Determine the Safety and Tolerability of This Drug Combination.

To determine the safety and tolerability of RAD001 when given in combination with temozolomide in patients with advanced pancreatic neuroendocrine tumors. (NCT00576680)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Grade 3 or 4 LymphopeniaGrade 3 or 4 thrombocytopeniaGrade 3 or 4 mucositisGrade 3 or 4 hyperglycemiaGrade 3 or 4 AST IncreaseGrade 3 or 4 Leukocyte decrease
Temozolomide and Everolimus1971847

[back to top]

Progression-free Survival

To determine progression-free survival when RAD001 is given in combination with temozolomide in patients with advanced pancreatic neuroendocrine tumors. Progression-free survival is defined as time from start of therapy until disease progression, as defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST), as 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since the treatment started or the appearance of one or more new lesions, or death. (NCT00576680)
Timeframe: 2 years

Interventionmonths (Median)
Temozolomide and Everolimus15.4

[back to top]

Response Rate

To determine the objective response rate by RECIST criteria of RAD001 in combination with temozolomide in patients with advanced pancreatic neuroendocrine tumors. Partial response (PR) by these criteria is defined as at least a 30% decrease in the sum of the longest diameter of target lesions, taking as reference the baseline sum longest diameter. Progressive disease (PD) is defined as at least a 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since the treatment started or the appearance of one or more new lesions. Stable Disease (SD) is defined as neither sufficient decrease to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum longest diameter since the treatment started. (NCT00576680)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Temozolomide and Everolimus16

[back to top]

Percentage of Participants With Distant Brain Failure (DBF) at One Year

Patients developing distant brain failure (DBF) at one year. An approximation method was used to arrive at the reported percentage. (NCT00582075)
Timeframe: 1 years

Interventionpercentage of participants (Number)
Radiosurgery 15-24 Gy + Adjuvant Temozolomide37

[back to top]

Overall Survival

(NCT00582075)
Timeframe: 2 years

Interventionweeks (Median)
Radiosurgery 15-24 Gy + Adjuvant Temozolomide31

[back to top]

Overall Objective Response (Complete Response or Partial Response)

"The Response Evaluation Criteria in Solid Tumors (RECIST) will be used to determine treatment response.~Clinical Complete Response (CRc) Disappearance of all target lesions and non-measurable disease.~Pathological Complete Response (CRp) A CRc in which a lymph node dissection done after completing temozolomide treatment shows no pathological evidence of melanoma. Partial Response (PR) A greater or equal then 30% in the sum of the longest diameter of all target lesions relative to baseline measurement" (NCT00588341)
Timeframe: 2 years

Interventionparticipants (Number)
Complete Response (CR)Partial Response (PR)Non Responder
Treatment2215

[back to top]

Progession Free Survival

To determine the duration of disease control of newly diagnosed pure and mixed anaplastic oligodendrogliomas treated with dose-intensive chemotherapy requiring hematopoietic stem cell support. (NCT00588523)
Timeframe: 2 years

Intervention% of participants without progression (Number)
Participants With Newly Diagnosed Anaplastic Oligodendroglioma85.7

[back to top]

Number of Participants Evaluated for Toxicities

(NCT00588523)
Timeframe: up to 2 years

InterventionParticipants (Count of Participants)
Participants With Newly Diagnosed Anaplastic Oligodendroglioma60

[back to top]

Objective Response

The best clinical response rates determined and 95% confidence intervals obtained using the exact binomial distribution. (NCT00590681)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Complete ResponsePartial ResponseStable DiseaseProgression
Treatment22.519.411.36.5

[back to top]

Overall Survival

(NCT00590681)
Timeframe: Up to 3 years

Interventionyears (Median)
Treatment1.68

[back to top]

Safety of Avastin in Combination With Temozolomide in This Study Population

Toxicity rates (limited toxicity to grade 4+) (NCT00590681)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
ApneaBone infectionCreatine phosphokinase increasedDICDeathDepressionFatigueFebrile neutropeniaHypocalcemiaIschemia cerebrovascularLeukocytesLeukopeniaMetabolicMusculoskeletalNeutrophilsPlateletsPleural effusionProteinuriaSeizureSoft tissue infectionThrombosisLymphopenia
Treatment1111111111121122121111

[back to top]

Duration of Response

Duration of response time from the first assessment of CR or PR until disease progression or death from any cause, whichever occurs first, event-free survival probability at two years assessed by Kaplan-Meier survivor function (NCT00590681)
Timeframe: Up to 2 years

Interventionprobability (Number)
Treatment38.8

[back to top]

Progression-free Survival (PFS)

(NCT00590681)
Timeframe: Up to 3 years

Interventionyears (Median)
Treatment0.96

[back to top]

Duration of Objective Clinical Responses

Duration of Response (Objective Clinical Responses) (NCT00591370)
Timeframe: 24 weeks after ending treatment

Interventionmonths (Median)
Temozolomide (TMZ)7.7

[back to top]

Overall Survival

Overall survival at 18 months post treatment (NCT00591370)
Timeframe: 18 months after ending treatment

Interventionpercentage of participants (Number)
Temozolomide (TMZ)27

[back to top]

Determine the Overall Objective Response Rate (CR and PR).

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. (NCT00591370)
Timeframe: From start of treatment through 24 weeks after ending treatment

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgression of Disease
Temozolomide (TMZ)061627

[back to top]

Number of Patients Experiencing a Central Nervous System (CNS) Hemorrhage or a Systemic Hemorrhage

Number of times a CNS hemorrhage or systemic hemorrhage was experienced (NCT00597402)
Timeframe: 55 months

Interventionparticipants (Number)
Gr.2 Central Nervous System (CNS) HemorrhageGr.3 Central Nervous System (CNS) HemorrhageGr.4 Central Nervous System (CNS) HemorrhageGr.5 Central Nervous System (CNS) HemorrhageGr.2 Systemic HemorrhageGr.3 Systemic HemorrhageGr.4 Systemic HemorrhageGr.5 Systemic Hemorrhage
Avastin, Radiation, Temozolomide, and Irinotecan10101000

[back to top]

12-month Progression-free Survival (PFS)

Percentage of participants surviving twelve months from the start of study treatment without progression of disease. PFS was defined as the time from the date of study treatment initiation to the date of the first documented progression according to the Macdonald criteria, or to death due to any cause. (NCT00597402)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Avastin, Radiation, Temozolomide, and Irinotecan63.2

[back to top]

16-month Overall Survival (OS)

Percentage of participants surviving sixteen months from the start of study treatment. OS was defined as the time from the date of study treatment initiation to the date of death due to any cause. (NCT00597402)
Timeframe: 16 months

Interventionpercentage of participants (Number)
Avastin, Radiation, Temozolomide, and Irinotecan64.8

[back to top]

Number of Patients Experiencing a Grade ≥ 4 Hematologic or Grade ≥ 3 Non-hematologic Toxicity

Number of times a grade ≥ 4 hematologic or grade ≥ 3 non-hematologic toxicity was experienced (NCT00597402)
Timeframe: 55 months

Interventionparticipants (Number)
Grade ≥ 4 Hematologic ToxicitiesGrade ≥3 Non-hematologic Toxicities
Avastin, Radiation, Temozolomide, and Irinotecan2049

[back to top]

Safety and Toxicity of Combination

Number of participants experiencing a toxicity of at least grade 3 that was deemed possibly, probably, or definitely related to the treatment. (NCT00597493)
Timeframe: 16 months

Interventionparticipants (Number)
Sorafenib + Temozolomide19

[back to top]

Pharmacokinetics: T-max

Blood sampling for sorafenib pharmacokinetics was performed on days 1 and 28 of cycle 1 and was obtained before and at 0.5, 1, 2, 4, 6, 8, and 24 h after the morning dose. T-max refers to time to maximum concentration. The pharmacokinetics of those patients taking enzyme-inducing antiepileptic drugs (EIAED) and those who were not were analyzed separately. (NCT00597493)
Timeframe: 13 months

Interventionhours (Median)
EIAEDs-Day 18.2
EIAEDs-Day 282.1
Non-EIAEDs-Day 124.0
Non-EIAEDs-Day 284.2

[back to top]

Pharmacokinetics: C-max

Blood sampling for sorafenib pharmacokinetics was performed on days 1 and 28 of cycle 1 and was obtained before and at 0.5, 1, 2, 4, 6, 8, and 24 h after the morning dose. C-max refers to maximum plasma concentration. The pharmacokinetics of those patients taking enzyme-inducing antiepileptic drugs (EIAED) and those who were not were analyzed separately. (NCT00597493)
Timeframe: 13 months

Interventionug/L (Geometric Mean)
EIAEDs-Day 13397.3
EIAEDs-Day 283813.9
Non-EIAEDs-Day 13155.1
Non-EIAEDs-Day 288118.8

[back to top]

Pharmacokinetics: AUC-24

Blood sampling for sorafenib pharmacokinetics was performed on days 1 and 28 of cycle 1 and was obtained before and at 0.5, 1, 2, 4, 6, 8, and 24 h after the morning dose. AUC-24 refers to area under the plasma concentration-time curve from 0 to 24 hours. The pharmacokinetics of those patients taking enzyme-inducing antiepileptic drugs (EIAEDs) and those who were not were analyzed separately. (NCT00597493)
Timeframe: 13 months

Interventionug*H/L (Geometric Mean)
EIAEDs-Day 145309.7
EIAEDs-Day 2847148.2
Non-EIAEDs-Day 145238.7
Non-EIAEDs-Day 28128820.8

[back to top]

6 Month Progression Free Survival (PFS)

Percentage of participants surviving six months from the start of study treatment without progression of disease. PFS was defined as the time from the date of study treatment initiation to the date of the first documented progression according to the Macdonald criteria, or to death due to any cause. (NCT00597493)
Timeframe: 6 months

Interventionpercentage of patients (Number)
Sorafenib + Temozolomide9.4

[back to top]

Overall Survival Rate

(NCT00602576)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Arm A12
Arm B18
Arm C5
Arm D17

[back to top]

Rate of 6 Month Progression-Free Survival

Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT00602576)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Arm A18
Arm B15
Arm C4
Arm D12

[back to top]

Response Rate

Response Rate as defined by 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. (NCT00602576)
Timeframe: Approximately 3 years

InterventionParticipants (Count of Participants)
Arm A8
Arm B6
Arm C0
Arm D8

[back to top] [back to top] [back to top]

Response Rate (CR + CRi + LFS)

"Response determined per European LeukemiaNet response criteria:~CR = bone marrow blasts <5%; absence of blasts with Auer rods; absence of extramedullary disease; absolute neutrophil count > 1.0 x 10e9/L; platelet count > 100 x 10e9/L; and independence of red cell transfusions.~CRi = all CR criteria except for residual neutropenia (< 1.0 x 10e9/L) or thrombocytopenia (< 100 x 10e9/L)].~Morphologic leukemia-free state (LFS) = bone marrow blasts <5%; absence of blasts with Auer rods; absence of extramedullary disease; with no hematologic recovery required.~Relapse = bone marrow blasts >5%; reappearance of blasts in the blood; or development of extramedullary disease." (NCT00611247)
Timeframe: up to 2 months

Interventionparticipants (Number)
Methylated AGAT Promoter (Group 1)3
Un-Methylated AGAT Promoter (Group 2)10

[back to top]

Response Rate

The proportion of subjects with complete or partial response as determined by a modification of the RANO (Response Assessment in Neuro-Oncology) criteria. A confirmation of response was not required. Complete Response was defined as complete disappearance on MR/CT of all enhancing tumor and mass effect, off all corticosteroids (or receiving only adrenal replacement doses), accompanied by a stable or improving neurologic examination, and maintained for at least 4 weeks. Partial Response was defined as greater than or equal to 50% reduction in tumor size on MR/CT by bi-dimensional measurement, on a stable or decreasing dose of corticosteroids, accompanied by a stable or improving neurologic examination, and maintained for at least 4 weeks. (NCT00612339)
Timeframe: 4 months

Interventionpercentage of patients (Number)
Avastin and Temozolomide24.4

[back to top]

Radiographic Response

Percentage of participants with an objective response (complete response or partial response) based on modified Macdonald criteria. (NCT00613028)
Timeframe: 41 months

Interventionpercentage of participants (Number)
Temozolomide Arm0
Etoposide Arm0

[back to top]

Median Progression-free Survival (PFS)

Time in months from the start of study treatment to the date of first progression according to Macdonald criteria, or to death due to any cause. Patients alive who had not progressed as of the last follow-up had PFS censored at the last follow-up date. Median PFS was estimated using a Kaplan-Meier curve. (NCT00613028)
Timeframe: 41 months

Interventionweeks (Median)
Temozolomide Arm4.1
Etoposide Arm8.1

[back to top]

Median Overall Survival (OS)

Time in months from the start of study treatment to date of death due to any cause. Patients alive as of the last follow-up had OS censored at the last follow-up date. Median OS was estimated using a Kaplan-Meier curve. (NCT00613028)
Timeframe: 41 months

Interventionweeks (Median)
Temozolomide Arm12.6
Etoposide Arm19

[back to top]

Grade 3 or Greater, Treatment Related, Non-hematologic Toxicities.

Incidence of ≥Grade 3 treatment related, non-hematologic toxicity (NCT00613028)
Timeframe: 41 months

Interventionparticipants (Number)
Temozolomide Arm1
Etoposide Arm2

[back to top]

The Primary Outcome Measure is 6 Month Progression-free Survival.

Percentage of participants surviving six months from the start of study treatment without progression of disease. PFS was defined as the time from the date of study treatment initiation to the date of the first documented progression according to the Macdonald criteria, or to death due to any cause. (NCT00613028)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Temozolomide Arm0
Etoposide Arm7.7

[back to top]

Overall Time of Survival

Time from initiation of study participation until death (NCT00617539)
Timeframe: Time from initiation of study participation until death or up to 3 years

InterventionDays (Median)
Irinotecan and Temozolomide146

[back to top]

Number of Patients Whose Circulating Tumor Cells (CTCs) Decreased From >5 to <5 CTCs Per 7.5 mL

CTCs were measured in blood using the Cellsearch(R) assay in 14 of the 20 patients measured at baseline (NCT00617539)
Timeframe: CTCs drawn on cycle 1 day 1, collection at 8 week intervals on patients who did not progress on their 8 week scans up to 2 years

Interventionparticipants (Number)
Irinotecan and Temozolomide1

[back to top]

Number of Patients With Objective Treatment Response (Complete or Partial) in the CNS

Imaging was performed at 8-week intervals to assess response to treatment. Patients with known or suspected leptomeningeal disease were deemed to have a complete response if CSF cytology converted to negative (if positive at baseline) and all meningeal enhancement or nodularity of brain and/or spine MRI resolved. A modified RECIST 1.0 criteria was used to assess CNS response for patients with new or progressing brain metastases. In this modified RECIST criteria, CNS lesions <1cm were not considered measurable, but were considered evaluable for response and progression. Progressive disease for patients with lesions <1 cm was defined as follows: growth of a lesion from less than or equal to 5 mm to greater than or equal to 10mm; or, growth of a 6-9 mm lesion by at least 5 mm in the case of non-target parenchymal brain metastases. (NCT00617539)
Timeframe: Baseline scan prior to study entry was performed within 14 days of cycle 1 day 1, then every 8 weeks from then until disease progression or up to 2 years

InterventionParticipants (Count of Participants)
Irinotecan and Temozolomide2

[back to top]

Time to First Progression in CNS

"Imaging at 8-week intervals to assess response to treatment. A modified RECIST 1.0 criteria was used to assess response and time to progression in the CNS for patients with progressing brain metastases. In this modified RECIST criteria, CNS lesions <1cm were not considered measurable, but were considered evaluable for response and progression. Progressive disease for patients with lesions <1 cm was defined as follows: growth of a lesion from less than or equal to 5 mm to greater than or equal to 10mm; or, growth of a 6-9 mm lesion by at least 5 mm in the case of non-target parenchymal brain metastases.~If patient did not come back for a follow up scan after clinical deterioration, patient was only considered stable up to the time of the last scan per protocol and time to progression would be from cycle 1 day 1 to the last scan they completed that was stable." (NCT00617539)
Timeframe: Baseline scan prior to study entry was performed within 14 days of cycle 1 day 1, then every 8 weeks from then until disease progression or up to 2 years

InterventionDays (Median)
Irinotecan and Temozolomide70

[back to top]

Number of Patients Experiencing a Clinical Benefit

The number of patients experiencing a clinical benefit is the sum of patients with an objective response plus patients with stable disease at ≥ 16 weeks from cycle 1 day 1 (first day of treatment). If a patient did not come back for a follow up scan after clinical deterioration, then they were only considered stable up to the time of the last scan they had per protocol. (NCT00617539)
Timeframe: From 1 day 1 (first day of treatment) every 8 weeks until scan shows disease progression or up to 2 years

InterventionParticipants (Count of Participants)
Irinotecan and Temozolomide7

[back to top]

Progression-free Survival (PFS) Based on Tumor MGMT (O(6)-Methylguanine-DNA Methyltransferase) Promoter Methylation Status.

Progression-free survival data (obtained for Primary Outcome Measure) was correlated with tumor MGMT (O(6)-methylguanine-DNA methyltransferase) promoter methylation status, obtained from patients as part of the study. (NCT00619112)
Timeframe: First day of treatment until progression or until 6 months mark

Interventionweeks (Median)
Glioblastoma With Methylated MGMT8.14
Glioblastoma With Unmethylated MGMT7.57
Grade III Glioma With Methylated MGMT38.1
Grade III Glioma With Unmethylated MGMT48.6

[back to top]

Patients With Tumors With Functional Alterations of the Mismatch Repair (MMR) System

PCR analysis of tumor tissue for microsatellite instability (MSI). Tissue was obtained during surgeries prior this study. (NCT00619112)
Timeframe: prior to start of study

InterventionParticipants (Count of Participants)
Glioblastoma0
Grade III Glioma0

[back to top]

6 Month Progression-free Survival

Efficacy of dose-intense temozolomide treatment schedule, as measured by 6 months progression-free survival (NCT00619112)
Timeframe: First day of treatment until progression or until 6 months mark

Interventionpercentage of patients (Number)
Glioblastoma10
Grade III Glioma50

[back to top]

Patients Progressing 6 Months After Temozolomide is Voluntarily Discontinued

(NCT00619112)
Timeframe: From beginning of voluntarily temozolomide discontinued up to 6 months

InterventionParticipants (Count of Participants)
Glioblastoma4
Grade III Glioma4

[back to top]

Overall Survival

(NCT00619112)
Timeframe: up to 2 years after treatment

Interventionweeks (Median)
Glioblastoma21.6
Grade III Glioma100.6

[back to top]

Patients Progressing After Two First-line Adjuvant Courses of Temozolomide

(NCT00619112)
Timeframe: After two first-line adjuvant courses of temozolomide

InterventionParticipants (Count of Participants)
Glioblastoma3
Grade III Glioma0

[back to top]

Patients Progressing Within 6 Months After 6th Adjuvant Course of Temozolomide

(NCT00619112)
Timeframe: Within 6 months after 6th adjuvant course of temozolomide

InterventionParticipants (Count of Participants)
Glioblastoma4
Grade III Glioma1

[back to top]

Tumor Response Rate, Calculated as a Percentage Along With it's 95% Confidence Interval

"A confirmed tumor response is defined to be a Complete Response or Partial Response noted~> as the objective status on 2 consecutive evaluations at least 8~> weeks apart. The proportion of tumor responses will be~> estimated by the number of confirmed tumor responses divided~> by the total number of evaluable patients.~> Complete Response (CR): Disappearance of all target lesions~> Partial Response (PR): At least a 30% decrease in the sum of the LD of target lesions taking as reference the baseline sum LD.~> Progression (PD): At least a 20% increase in the sum of target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions.~> Stable Disease (SD): Neither sufficient shrinkage to Qualify for PR nor sufficient increase to Qualify for PD taking as reference the smallest sum LD. responses will be calculated assuming that the number of~> confirmed tumor responses follows a binomial distribution." (NCT00626405)
Timeframe: Up to 5 years

Interventionpercentage of patients with response (Number)
Arm I23.8
Arm II33.3

[back to top]

Progression-free Survival at 6 Months

The primary endpoint is the 6 month post registration Progression-free survival (PFS) rate. Progression-free survival time is defined as the time from registration to documentation of disease progression using the RECIST criteria. Patients who died without documentation of disease progression will be considered to have progressed at death unless there is sufficient documented evidence to conclude no progression occurred prior to death. All patients, who meet the eligibility criteria, sign a consent form, and start treatment will be included in the evaluation of the 6 month PFS rate. (NCT00626405)
Timeframe: at 6 months

Intervention% of patients alive and progression free (Number)
Arm I32.8
Arm II56.1

[back to top]

Overall Survival

Overall 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. (NCT00626405)
Timeframe: Up to 5 years

InterventionMonths (Median)
Arm I12.3
Arm II13.9

[back to top]

Progression-free Survival

Disease progression is defined as radiological or neurological/clinical progression (whichever occurs first); progression free survival (PFS) is the time interval between the date of randomization and the date of disease progression or death whichever occurs first. If neither event has been observed, the patient is censored at the date of the last follow up examination. Radiological progression was defined as increase of contrast enhancing area on MRI or CT scans of more than 25% as measured by two perpendicular diameters compared to the smallest measurements ever recorded for the same lesion by the same technique. The appearance of new lesions with or without contrast enhancement Neurological/clinical progression was defined as:decrease in WHO performance status,deterioration of neurological functions,appearance of signs/symptoms of increased intracranial pressure,and/or start of corticosteroid or increase of corticosteroid dosage by 50% for control of neurological symptoms. (NCT00626990)
Timeframe: from randomization till the date of disease progression or death (time till death is up to 10.9 years after patient enrollment in the study)

InterventionMonths (Median)
Absence of Concomitant TMZ20.9
Presence of Concomitant TMZ33.02
Absence of Adjuvant TMZ19.09
Presence of Adjuvant TMZ42.81

[back to top]

Overall Survival as Measured From the Day of Randomization

The duration of survival is the time interval between randomization and the date of death due to any cause. Patients not reported dead or lost to follow up will be censored at the date of the last follow up examination. (NCT00626990)
Timeframe: from date from enrollment till the date of death (time till death is up to 10.9 years after patient enrollment in the study)

InterventionMonths (Median)
Absence of Concomitant Temozolomide (TMZ)60.42
Presence of Concomitant Temozolomide (TMZ)66.92
Absence of Adjuvant Temozolomide (TMZ)46.92
Presence of Adjuvant Temozolomide (TMZ)82.33

[back to top]

Tolerability of Maintenance Temozolomide

Tolerability was defined as number of participants with any adverse event leading to study discontinuation and/or study drug discontinuation. (NCT00632203)
Timeframe: from Cycle 1 Day 1 of Standard First Line Systemic Therapy to the last time of follow-up (up to 6 cycles (168 days) of study treatment)

Interventionparticipants (Number)
Temozolomide Treatment5
ObservationNA

[back to top]

Time to Radiological Central Nervous System (CNS) Progression

"Defined as CNS progression as measured by MRI.~Time to CNS progression was analyzed using the Kaplan-Meier method." (NCT00632203)
Timeframe: from Cycle 1 Day 1 of Standard First Line Systemic Therapy to radiological progression or the last known CNS progression-free date

Interventionmonths (Median)
Temozolomide TreatmentNA
ObservationNA

[back to top]

Overall Survival

The overall survival was analyzed using the Kaplan-Meier method. (NCT00632203)
Timeframe: from Cycle 1 Day 1 of Standard First Line Systemic Therapy to the last time of follow-up

Interventionmonths (Median)
Temozolomide Treatment27.14
Observation22.54

[back to top]

Number of Participants Who Had Brain Metastases

Brain Metastases were defined as radiological evidence of brain metastases on magnetic resonance imaging (MRI). (NCT00632203)
Timeframe: Up to 12 months (as measured from day 1 of cycle 1 of standard first-line systemic chemotherapy)

Interventionparticipants (Number)
Temozolomide Treatment4
Observation3

[back to top]

Time to Progression

"The time to progression (per response evaluation criteria in solid tumors [RECIST]) was analyzed using the Kaplan-Meier method.~Definitions of response per RECIST:~Complete Response (CR): Disappearance of all target lesions.~Partial Response (PR): A decrease of at least 30% in the sum of the longest~diameter of target lesions.~Progressive Disease (PD): An increase of at least 20% in the sum of the longest~diameter of target lesions.~Stable Disease (SD): Neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease." (NCT00632203)
Timeframe: from Cycle 1 Day 1 of Standard First Line Systemic Therapy to progression or up to 6 cycles (168 days) of study treatment

Interventionmonths (Median)
Temozolomide Treatment11.70
Observation10.68

[back to top]

Number of Days on Temozolomide Treatment

This outcome measure was only applicable to the temozolomide arm; the observation arm was therefore not analyzed. (NCT00638963)
Timeframe: Baseline to 24 Weeks

InterventionDays (Mean)
Temozolomide75

[back to top]

Number of Participants Who Had at Least One Treatment Omission During Treatment

This outcome measure was only applicable to the temozolomide arm; the observation arm was therefore not analyzed. (NCT00638963)
Timeframe: Baseline to 24 Weeks

InterventionParticipants (Number)
Temozolomide2

[back to top]

Total Dose of Temozolomide Taken

This outcome measure was only applicable to the temozolomide arm; the observation arm was therefore not analyzed. (NCT00638963)
Timeframe: Baseline to 24 Weeks

InterventionMilligrams (Mean)
Temozolomide9791.9

[back to top]

Number of Participants Who Completed the Third Cycle of Treatment

This outcome measure was only applicable to the temozolomide arm; the observation arm was therefore not analyzed. (NCT00638963)
Timeframe: Baseline to 24 Weeks

InterventionParticipants (Number)
Temozolomide2

[back to top]

Number of Participants Who Had at Least One Dose Reduction During Treatment

This outcome measure was only applicable to the temozolomide arm; the observation arm was therefore not analyzed. (NCT00638963)
Timeframe: Baseline to 24 Weeks

InterventionParticipants (Number)
Temozolomide3

[back to top]

Humoral and Cellular Immune Response

Number of patients that developed a delayed-type hypersensitivity (DTH) response at following vaccination. Any skin reaction in response to the intradermal injection of the antigen was measured and recorded. A positive skin test was defined as > 5 mm induration (swelling). (NCT00643097)
Timeframe: 26 months

Interventionparticipants (Number)
Arm I (ACTIVATE)3
Arm II (ACT II STD)0
Arm III (ACT II DI)7

[back to top]

Clinical Efficacy of Vaccination, in Terms of Progression-free Survival (PFS)

"Time in months from the start of study treatment to the date of first progression according to Macdonald criteria, or to death due to any cause. Patients alive who had not progressed as of the last follow-up had PFS censored at the last follow-up date. Median PFS was estimated using a Kaplan-Meier curve.~Macdonald criteria are standard criteria in neuro-oncology. Tumor assessment was made according to the adapted MacDonald criteria based on the combined evaluation of: 1) assessment of the MRI scan for measurable, evaluable, and new lesions (made by the independent external expert too), 2) overall assessment of neurological performance (made by the investigator), 3) concomitant steroid use (as reported by the investigator)." (NCT00643097)
Timeframe: 58 months

Interventionmonths (Median)
Arm I (ACTIVATE)14.2
Arm II (ACT II STD)12.1
Arm III (ACT II DI)11.6

[back to top]

Response to Vaccination

The objective is to assess the duration of immunosuppressive cytokine secretion and to identify a receptive interval for active immunotherapy. Immunosuppression will determined by monitoring a panel of immunosuppressive serum/plasma cytokines longitudinally and by determining the response of each patient to Recombivax Hepatitis B (HB) vaccination. Response is defined as seropositive or seronegative to the Hepatitis B surface antigen. (NCT00643097)
Timeframe: 26 months

InterventionMonths (Mean)
Arm I (ACTIVATE)NA
Arm II (ACT II STD)NA
Arm III (ACT II DI)NA

[back to top]

Toxicity to PEP-3 Vaccine Immunization

To assess for any potential toxicity to the PEP-3 vaccine immunization in patients with newly diagnosed glioblastoma, Common Terminology Criteria for Adverse Events (CTCAE) version 3.0 was used to tabulate any toxicities attributable to PEP-3. The number of patients with toxicity attributable to vaccine while on study are tabulated. (NCT00643097)
Timeframe: 26 months

Interventionparticipants (Number)
Arm I (ACTIVATE)4
Arm II (ACT II STD)1
Arm III (ACT II DI)7

[back to top]

Radiographic Response

Responders on study are those with a best response of either CR or PR. Per Modified Macdonald Criteria for lesions assessed by MRI/CT: Complete Response (CR) = Complete disappearance of all measurable and evaluable disease, no new lesions, no evidence of non-evaluable disease, with no steroids. Partial Response (PR) >/= 50% decrease under baseline in the sum of products of perpendicular diameters of all measurable lesions, no progression of evaluable disease, no new lesions, with steroid dose @ time of response NCT00657267)
Timeframe: From patient registration until end of study, assessed up to 54 months

Interventionpercentage of participants evaluated (Number)
Partial Response13
Complete Response0

[back to top]

6 Month Progression Free Survival

Progression is defined using Modified Macdonald Criteria , using a >/= 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR clear clinical worsening or failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00657267)
Timeframe: 6 months

Interventionpercentage of evaluable participants (Number)
12 Cycles of Dose-Dense Temozolomide (Single Arm Study)11

[back to top]

Time to Progression.

Progression is defined using Modified Macdonald Criteria , using a >/= 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR clear clinical worsening or failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00657267)
Timeframe: From patient registration until end of study, assessed up to 54 months

Interventiondays (Median)
12 Cycles of Dose-Dense Temozolomide (Single Arm Study)56

[back to top]

Overall Survival

(NCT00657267)
Timeframe: From patient registration until end of study, assessed up to 54 months

Interventionmonths (Median)
12 Cycles of Dose-Dense Temozolomide (Single Arm Study)11.7

[back to top]

Response

Response rate by RECIST (NCT00667953)
Timeframe: through study completion, an average of 1 year

InterventionParticipants (Count of Participants)
Arm A:1

[back to top]

Safety - Grade 3 or 4 Adverse Events

Number of reported grade 3 or 4 adverse events (NCT00667953)
Timeframe: through study completion, an average of 1 year

InterventionAdverse Events (Number)
Arm A:20

[back to top]

Response Rate

Number of patients with reduction in tumor burden of a predefined amount (NCT00669669)
Timeframe: Up to 66 months

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)1

[back to top]

Time to Progression

From the first day of treatment (transplant) until unequivocal progression is documented, assessed up to 66 months. (NCT00669669)
Timeframe: Up to 66 months.

Interventionmonths (Median)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)5.5

[back to top]

Duration of Response

From the onset of temozolomide to the date at which unequivocal disease progression, assessed up to 65 months. (NCT00669669)
Timeframe: Up to 65 months

Interventionmonths (Median)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)4.5

[back to top]

Gene Transfer Efficiency After Chemotherapy

Assessed by gene marking in peripheral blood after chemoselection. Gene marking is assessed in whole blood by quantitative PCR and reported as a vector copy number (VCN) or the average copies of integrated transgene per cell. The units here will be reported as copies/cell. (NCT00669669)
Timeframe: Up to 59 months

Interventioncopies/cell (Mean)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)0.50

[back to top]

Gene Transfer Efficiency

Assessed by gene marking in peripheral blood prior to chemoselection. Gene marking is assessed in whole blood by quantitative PCR and reported as a vector copy number (VCN) or the average copies of integrated transgene per cell. The units here will be reported as copies/cell. (NCT00669669)
Timeframe: Up to 59 months

Interventioncopies/cell (Mean)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)0.78

[back to top]

Number of Participants Dose-limiting Toxicity (DLT)

Defined as any grade 4 nonhematopoietic toxicity that is likely related to the investigational procedures (Part I) (NCT00669669)
Timeframe: Up to 6 weeks after infusion

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)1

[back to top]

Number of Participants That Survived

From the first day of treatment until death, assessed up to 74 months. (NCT00669669)
Timeframe: Up to 74 months

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)0

[back to top]

Number of Participants With Chemoprotection

assessed by the ability to increase the Temozolomide dose beyond 472 mg/m^2 (NCT00669669)
Timeframe: Up to 66 months

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)2

[back to top]

Number of Participants With Chemoselection

assessed by the increase in peripheral blood Vector Copy Number (VCN), the average copies of integrated transgene per cell, after chemotherapy (NCT00669669)
Timeframe: Up to 59 months

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)4

[back to top]

Number of Participants With Retrovirus or Leukemia

Replication competent retrovirus or diagnosis of leukemia (NCT00669669)
Timeframe: Up to 2 years after infusion

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Autologous Stem Cell Transplant)0

[back to top]

Adverse Events With an Incidence of Greater Than or Equal to 20%

Safety was assessed from the start of administration during the concomitant radiotherapy phase until 30 days after the completion of administration of monotherapy. Adverse events were classified under the system organ class using MedDRA-J Version 11.0. (NCT00684567)
Timeframe: until 30 days after the completion of administration of monotherapy

InterventionParticipants (Number)
alopecianauseaanorexiaconstipationmalaiserashweight decreasedheadachevomitingdry skinnasopharyngitisdiarrheaconvulsionpruritusneutrophil count decreasedwound complication
Radiotherapy/Temozolomide251717151411111098877666

[back to top]

Number of Participants With Progression Free Survival (PFS) for 1 Year

Administration of SCH 52365 was continued until progression was observed (progression was judged by the investigator based on MRI and clinical symptoms). (NCT00684567)
Timeframe: 1 year after the start of admininstration in the concomitant radiotherapy phase

InterventionParticipants (Number)
Radiotherapy/Temozolomide11

[back to top]

Number of Participants With a Response (Complete Response [CR] + Partial Response [PR]) in Terms of Overall Tumor Response

"CR = measurable lesion disappeared.~PR = total sum of lesions measurable in bidimension decreased by 50% or more on whole and no secondary progression attributable to tumor was noted. No onset of new lesion." (NCT00684567)
Timeframe: 1 year after the start of administration in the concomitant radiotherapy phase

InterventionParticipants (Number)
Radiotherapy/Temozolomide6

[back to top]

Abnormal Changes in Laboratory Test Values With an Incidence of Greater Than or Equal to 20%

Safety was assessed from the start of administration during the concomitant radiotherapy phase until 30 days after the completion of administration of monotherapy. (NCT00684567)
Timeframe: until 30 days after the completion of administration of monotherapy

InterventionParticipants (Number)
lymphocyte count decreasedwhite blood cell count decreasedeosinophil percentage increased
Radiotherapy/Temozolomide241410

[back to top]

Adverse Drug Reactions With an Incidence of Greater Than or Equal to 20%

Safety was assessed from the start of administration during the concomitant radiotherapy phase until 30 days after the completion of administration of monotherapy. (NCT00684567)
Timeframe: until 30 days after the completion of administration of monotherapy

InterventionParticipants (Number)
constipationnauseavomitingneutrophil count decreasedweight decreasedmalaiseanorexia
Radiotherapy/Temozolomide151166679

[back to top]

Relationship Between MGMT Status and Therapy Response: Overall Survival Rate for the MGMT Positive Group

"MGMT was measured by IHC.~OS rate was defined as the percentage of participants who were still alive 6, 12, & 18 months after starting study treatment.~OS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: 6, 12, & 18 months

,
Interventionpercentage of participants (Number)
6 months12 months18 months
Temozolomide + Radiation80.869.336.9
Temozolomide Alone, Then Temozolomide + Radiation89.282.866.2

[back to top]

Relationship Between MGMT Status and Therapy Response: Overall Survival Rate for the MGMT Negative Group

"MGMT was measured by IHC.~OS rate was defined as the percentage of participants who were still alive 6, 12, & 18 months after starting study treatment.~OS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: 6, 12, & 18 months

,
Interventionpercentage of participants (Number)
6 months12 months18 months
Temozolomide + Radiation87.557.744.9
Temozolomide Alone, Then Temozolomide + Radiation95.881.426.7

[back to top]

Relationship Between O6-methylguanine-DNA Methyltransferase (MGMT) Status and Therapy Response: Overall Survival for the MGMT Positive Group

"MGMT was measured by immunohistochemistry (IHC).~OS was defined as the length of time from the start of treatment that 1/2 of the participants were still alive.~OS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: Up to 2 years

Interventionmonths (Median)
Temozolomide + Radiation14.4
Temozolomide Alone, Then Temozolomide + Radiation17.49

[back to top]

Relationship Between MGMT Status and Therapy Response: PFS for the MGMT Positive Group

"MGMT was measured by IHC.~PFS: The length of time during and after treatment that a participant lived with the cancer but it does not get worse.~PFS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: Up to 2 years

Interventionmonths (Median)
Temozolomide + Radiation10.04
Temozolomide Alone, Then Temozolomide + Radiation11.19

[back to top]

Relationship Between MGMT Status and Therapy Response: PFS for the MGMT Negative Group

"MGMT was measured by IHC.~PFS: The length of time during and after treatment that a participant lived with the cancer but it does not get worse.~PFS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: Up to 2 years

Interventionmonths (Median)
Temozolomide + Radiation11.11
Temozolomide Alone, Then Temozolomide + Radiation10.46

[back to top]

Objective Tumor Assessment After Surgery: Overall Response

"Overall response was based on neuroimaging (magnetic resonance imaging [MRI]), clinical neurological examination, and steroid administration.~It was assessed as follows:~Complete Response (CR): Disappearance of all enhancing tumor (measurable~or non-measurable), no corticosteroid use, and neurologically stable or~improved.~Partial Response (PR): ≥50% reduction in size of enhancing tumor~(measurable or non-measurable) for any measurable lesions or definite~improvement for any non-measurable lesions, corticosteroid dosage stable or~reduced, and neurologically stable or improved.~Progressive Disease (PD): ≥25% increase in contrast enhancement for any~measurable lesions or definite worsening for any non-measurable lesions, or~any new tumor on MRI scans, at an increased dose of corticosteroid, with or without neurologic progression. Clinical or radiological worsening resulting from other than tumor factors were excluded.~Stable Disease (SD): All other situations." (NCT00686725)
Timeframe: Up to 2 years

,
Interventionparticipants (Number)
CRPRSDPDNo overall response data available
Temozolomide + Radiation103835
Temozolomide Alone, Then Temozolomide + Radiation004741

[back to top]

Relationship Between MGMT Status and Therapy Response: Overall Survival for the MGMT Negative Group

"MGMT was measured by IHC.~OS was defined as the length of time from the start of treatment that 1/2 of the participants were still alive.~OS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: Up to 2 years

Interventionmonths (Median)
Temozolomide + Radiation13.81
Temozolomide Alone, Then Temozolomide + Radiation15.12

[back to top]

Progression-Free Survival (PFS)

"PFS was defined as the length of time from randomization to disease progression (the length of time during which the cancer did not get worse) or death.~PFS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: Up to 2 years

Interventionmonths (Median)
Temozolomide + Radiation10.38
Temozolomide Alone, Then Temozolomide + Radiation8.74

[back to top]

Overall Survival (OS)

"OS was defined as the time from randomization to death.~OS was calculated by the Kaplan-Meier method." (NCT00686725)
Timeframe: Up to 2 years

Interventionmonths (Median)
Temozolomide + Radiation13.17
Temozolomide Alone, Then Temozolomide + Radiation17.58

[back to top]

Number of Participants With CR, PR, or MLFS Who Received Modified Low Dose Maintenance Therapy (100 mg/m^2/Day x21 Days of Each 28 Day Cycle) and Experienced Toxicity

Toxicity was defined as any adverse event experienced by a participant regardless of causal relationship with study treatment. An adverse event (AE) was defined as any untoward medical occurrence in a participant administered a pharmaceutical product, biologic (at any dose), or medical device, which did not necessarily have a causal relationship with the treatment. Adverse events may have included the onset of new illness and/or the exacerbation of preexisting conditions. (NCT00687323)
Timeframe: From first dose to 30 days after last dose of study drug (up to 67 weeks)

Interventionparticipants (Number)
Temozolomide1

[back to top]

Number of Previously Untreated Participants With Low O6-Methylguanine Methyltransferase (MGMT) Expression

Low MGMT expression was defined as MGMT/β-actin ratio < 0.2. MGMT & β-actin are cancer biomarkers. (NCT00687323)
Timeframe: Baseline

Interventionparticipants (Number)
Temozolomide81

[back to top]

Clinical Response at the End of Temozolomide Induction

"Complete Response (CR): < 5% blasts in normocellular bone marrow (BM); Absolute Neutrophil Count (ANC) > 1.0 x 10^9/L, platelets > 100 x 10^9/L, and no extramedullary disease.~CR with incomplete platelet recovery (CRp): All the criteria of CR but with platelets < 100 x 10^9/L but ≥ 50 x 10^9/L and platelet transfusion independent.~Morphologic leukemia-free state (MLFS): complete clearance of blasts from marrow and blood, but criteria for CR or CRp not met.~Partial response (PR): decrease ≥ 50% BM blasts. Minimal Response (MR): decrease ≥ 25% but <50% BM blasts." (NCT00687323)
Timeframe: at the end of each cycle (approximately 4 weeks post start of cycle), up to a maximum 63 weeks

Interventionparticipants (Number)
CRCRpMLFSPRMR
Temozolomide102487

[back to top]

Progression-free Survival for Participants Achieving PR, MLSF, or MR

Progression-free survival was defined as time to disease progression. Morphologic leukemia-free state (MLFS): complete clearance of blasts from marrow and blood, but criteria for CR or CRp not met. Partial response (PR): decrease ≥ 50% BM blasts. Minimal Response (MR): decrease ≥ 25% but <50% BM blasts. (NCT00687323)
Timeframe: Start of treatment until disease progression [up to 1 year after treatment ends (up to 115 weeks)]

Interventionmonths (Median)
Temozolomide1.6

[back to top]

Overall Survival (OS) in Participants Achieving CR or CRp and Proceeding to Reduced Dose-intensity Maintenance Therapy With Temozolomide

"OS was defined as the time from start of treatment until death or end of study.~Complete Response (CR): < 5% blasts in normocellular bone marrow (BM); Absolute Neutrophil Count (ANC) > 1.0 x 10^9/L, platelets > 100 x 10^9/L, and no extramedullary disease. CR with incomplete platelet recovery (CRp): All the criteria of CR but with platelets < 100 x 10^9/L but ≥ 50 x 10^9/L and platelet transfusion independent." (NCT00687323)
Timeframe: Start of treatment until death or end of study [up to 1 year after treatment ends (up to 115 weeks)]

Interventionmonths (Median)
Temozolomide21.4

[back to top]

Duration of Response in Participants Achieving Complete Response (CR) and Proceeding to Reduced Dose-intensity Maintenance Therapy With Temozolomide

Complete Response (CR): < 5% blasts in normocellular bone marrow (BM); Absolute Neutrophil Count (ANC) > 1.0 x 10^9/L, platelets > 100 x 10^9/L, and no extramedullary disease. (NCT00687323)
Timeframe: Up to 1 year after treatment ends (up to 115 weeks)

InterventionDays (Median)
Temozolomide408

[back to top]

Relapse-free Survival in Participants Achieving CR or CRp and Proceeding to Reduced Dose-intensity Maintenance Therapy With Temozolomide

Relapse-free survival was defined as time to disease progression. Complete Response (CR): < 5% blasts in normocellular bone marrow (BM); Absolute Neutrophil Count (ANC) > 1.0 x 10^9/L, platelets > 100 x 10^9/L, and no extramedullary disease. CR with incomplete platelet recovery (CRp): All the criteria of CR but with platelets < 100 x 10^9/L but ≥ 50 x 10^9/L and platelet transfusion independent. (NCT00687323)
Timeframe: Start of treatment until disease progression [up to 1 year after treatment ends (up to 115 weeks)]

Interventionmonths (Median)
Temozolomide10.5

[back to top]

European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Brain Module (EORTC QLQ-BN20) Sub-scale Scores

The QLQ-BN20 is a questionnaire specifically designed as the QLQ-C30 supplement for the evaluation of quality of life in brain tumor participants. It includes 4 multi-item sub-scales: future uncertainty, visual disorder, motor dysfunction, communication deficits, and 7 single-item scales: headaches, seizures, drowsiness, itchy skin, hair loss, weakness of legs, and bladder control. All items are rated on a 4-point Likert-type scale ('1=not at all', '2=a little', '3=quite a bit' and '4=very much'), and are linearly transformed to a 0-100 scale, with higher scores indicating more severe symptoms. (NCT00689221)
Timeframe: Up to 50 months

,
Interventionunits on a scale (Mean)
Future Uncertainty (n=68, 86)Visual Disorder (n=68, 85)Motor Dysfunction (n=68, 86)Communication Deficit (n=68, 86)Headaches (n=68, 86)Seizures (n=68, 87)Drowsiness (n=66, 87)Itchy Skin (n=68, 86)Hair Loss (n=66, 86)Weakness of Legs (n=67, 85)Bladder Control (n=67, 85)
Cilengitide + Temozolomide + Radiotherapy44.4912.9927.4526.1425.989.3138.389.8013.1324.3819.40
Temozolomide + Radiotherapy39.3117.7823.3919.9621.718.0535.2513.5715.1220.3910.20

[back to top]

European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) Sub-scale Scores

The EORTC QLQ-C30 is a questionnaire including following sub-scales: global health status, functional scales (physical functioning, role functioning, emotional functioning, cognitive functioning, and social activity), symptom scales (fatigue, nausea and vomiting, and pain) and single items (dyspnoea, insomnia, appetite loss, constipation, diarrhoea and financial difficulties). Scores are averaged for each scale and transformed to 0-100 scale; higher score indicates better quality of life on global health status and functional scales and worse quality of life on symptom scales and financial difficulty scale. (NCT00689221)
Timeframe: Up to 50 months

,
Interventionunits on a scale (Mean)
Global Health Status (n=71, 92)Physical Functioning (n=71, 92)Role Functioning (n=71, 92)Emotional Functioning (n=71, 93)Cognitive Functioning (n=70, 93)Social Activity (n=71, 93)Fatigue (n=71, 92)Nausea and Vomiting (n=71, 93)Pain (n=71, 93)Dyspnoea (n=71, 92)Insomnia (n=71, 91)Appetite Loss (n=71, 92)Constipation (n=71, 93)Diarrhoea (n=70, 92)Financial Difficulties (n=71, 93)
Cilengitide + Temozolomide + Radiotherapy54.3465.7056.3467.4964.0556.3444.3710.3322.3015.9620.6621.1318.786.6727.23
Temozolomide + Radiotherapy55.4367.4656.3467.0065.4162.7239.737.7124.3713.0420.5115.9413.984.3522.94

[back to top] [back to top]

Number of Participants With AEs Belonging to Standardized Medical Dictionary for Regulatory Activities (MedDRA) Queries (SMQs) Thromboembolic Events and Hemorrhage With NCI-CTC Toxicity Grade 3 or 4

Thromboembolic events (standardized MedDRA query [SMQ]) Grade 3 or 4 AEs encompassed hemiparesis and cerebrovascular accident, pulmonary embolism, and deep vein thrombosis. Thromboembolic events (SMQ) of any grade and of Grade 3 or 4 were generally more frequent in the Cilengitide + Temozolomide/Radiotherapy group than in the Temozolomide/Radiotherapy group but were still in the expected range of this patient population The severity of AEs was assessed according to the National Cancer Institute-Common Toxicity Criteria (NCI-CTCAE) (version 3.0): Grade 1=mild, Grade 2=moderate, Grade 3=severe, Grade 4=life threatening or disabling. Note: Death (Grade 5) was regarded as an outcome. (NCT00689221)
Timeframe: Time from first dose up to 28 days after last dose of study treatment, reported between day of first participant randomized, that is, Sep 2008 until cut-off date (19 Nov 2012)

,
InterventionParticipants (Number)
SMQ:Thromboembolic eventsSMQ: Hemorrhage
Cilengitide + Temozolomide + Radiotherapy354
Temozolomide + Radiotherapy234

[back to top]

Number of Participants With Change From Baseline in Work Status at End of Study

Number of participants with change from baseline in work status (working full time [FT], part-time [PT], unemployed/retired [U/R]) at end of study (EOS) (up to cut-off date, [19 Nov 2012]) was reported. For the category 'part-time', the following sub-categories were defined: part-time due to basic disease (PT1); part-time not due to basic disease (PT2); part-time reason not known (PT3). (NCT00689221)
Timeframe: Baseline, End of study (up to cut-off date, [19 Nov 2012])

,
Interventionparticipants (Number)
Baseline: FT, EOS: FTBaseline: FT, EOS: PT1Baseline: FT, EOS: PT2Baseline: FT, EOS: PT3Baseline: FT, EOS: U/RBaseline: PT1, EOS: FTBaseline: PT1, EOS: PT1Baseline: PT1, EOS: PT2Baseline: PT1, EOS: PT3Baseline: PT1, EOS: U/RBaseline: PT2, EOS: FTBaseline: PT2, EOS: PT1Baseline: PT2, EOS: PT2Baseline: PT2, EOS: PT3Baseline: PT2, EOS: U/RBaseline: PT3, EOS: FTBaseline: PT3, EOS: PT1Baseline: PT3, EOS: PT2Baseline: PT3, EOS: PT3Baseline: PT3, EOS: U/RBaseline: U/R, EOS: FTBaseline: U/R, EOS: PT1Baseline: U/R, EOS: PT2Baseline: U/R, EOS: PT3Baseline: U/R, EOS: U/RBaseline: Missing, EOS: FTBaseline: Missing, EOS: PT1Baseline: Missing, EOS: PT2Baseline: Missing, EOS: PT3Baseline: Missing, EOS: U/RBaseline: Missing, EOS: Missing
Cilengitide + Temozolomide + Radiotherapy3210243300900015000005510199000011
Temozolomide + Radiotherapy61002221001210004000008710191000011

[back to top]

Progression Free Survival (PFS) Time - Investigator and Independent Read

"The PFS time is defined as the duration from randomization to either first observation of progressive disease (PD) or occurrence of death due to any cause. Investigator read is the assessment of all imaging by the treating physician at the local trial site and Independent Read is the assessment of all imaging centrally by an Independent Review Committee (IRC). Investigator's assessed progression according to MacDonald criteria and IRC by Response Assessment in Neuro-Oncology Working Group (RANO) criteria using Gadolinium-enhanced magnetic resonance imaging.~Investigator and IRC read: Progression is defined as greater than 25 percent increase in the sum of the product of the largest perpendicular diameters of enhancing tumor compared to the smallest prior sum, or Worsening of an evaluable lesion(s),or Marked increase in T2/FLAIR non-enhancing lesions (IRC only) or Any new lesion" (NCT00689221)
Timeframe: Time from randomization to disease progression, death or last tumor assessment, reported between day of first participant randomized, that is, Sep 2008 until cut-off date, (19 Nov 2012)

,
InterventionMonths (Median)
PFS Time: Investigator readPFS Time: Independent read
Cilengitide + Temozolomide + Radiotherapy13.510.6
Temozolomide + Radiotherapy10.77.9

[back to top]

Maximum Observed Plasma Concentration (Cmax)

The Cmax for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2.1. (NCT00689221)
Timeframe: Day 1 of Week -1

Interventionnanogram per milliliter (ng/mL) (Mean)
Cilengitide + Temozolomide + Radiotherapy167363.2

[back to top]

EuroQol 5-Dimensions (EQ-5D) Questionnaire Index

The EuroQuol-5D (EQ-5D) questionnaire is a measure of health status that provides a simple descriptive profile and a single index value. The optional part of the questionnaire was not applied. The EQ-5D defines health in terms of mobility, self-care, usual activities, pain/discomfort and anxiety/depression. The 5 items are combined to generate health profiles. These profiles were converted to a continuous single index score using a one to one matching. The lowest possible score is -0.594 (death) and the highest is 1.00 (full health). (NCT00689221)
Timeframe: Up to 50 months

Interventionunits on a scale (Mean)
Cilengitide + Temozolomide + Radiotherapy0.598
Temozolomide + Radiotherapy0.623

[back to top]

Overall Survival (OS) Time

The OS time is defined as the time (in months) from randomization to death or last day known to be alive. Participants without event are censored at the last date known to be alive or at the clinical cut-off date, whatever is earlier. (NCT00689221)
Timeframe: Time from randomization to death or last day known to be alive, reported between day of first participant randomized, that is, Sep 2008 until cut-off date, (19 Nov 2012)

InterventionMonths (Median)
Cilengitide + Temozolomide + Radiotherapy26.3
Temozolomide + Radiotherapy26.3

[back to top]

Area Under the Plasma Concentration Curve From Time 0 to 6 Hours (AUC [0-6]) After Dose

The AUC (0-6) for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2.1. (NCT00689221)
Timeframe: Day 1 of Week -1

Interventionhour*ng/mL (Mean)
Cilengitide + Temozolomide + Radiotherapy295171.2

[back to top]

Time to Maximum Plasma Concentration (Tmax)

The Tmax for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2.1. (NCT00689221)
Timeframe: Day 1 of Week -1

Interventionhours (Mean)
Cilengitide + Temozolomide + Radiotherapy1.029

[back to top]

Median Progression Free Survival After Primary Surgical Treatment, Concomitant and Adjuvant Chemotherapy With Temozolomide, for Patients With Newly Diagnosed Glioblastoma Multiforme

(NCT00704808)
Timeframe: After primary surgical treatment and concomitant and adjuvant chemotherapy with temozolomide

InterventionMonths (Median)
Temozolomide9.57

[back to top]

Disease Control Rate (DCR)

Using RECIST v1.0 criteria, disease control rate (DCR) was determined by the number of participants with complete response (CR) + the number of participants with partial response (PR) + the number of participants with stable disease (SD) / the number of participants with complete response (CR) + the number of participants with partial response (PR) + the number of participants with stable disease (SD) + the number of participants with progressive disease (PD). Per RECIST v1.0 criteria (assessed by MRI or CT): Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD), neither sufficient shrinkage to qualify for a Partial Response (PR) nor sufficient increase to to qualify for Progressive Disease (PD); PD, 20% increase in the sum if target lesion or the appearance of new lesions; Complete Response (CR), Disappearance of all target lesions. (NCT00715793)
Timeframe: Up to 30 months

Interventionpercentage of participants (Number)
DAC (Decitabine) + TMZ (Temozolomide)61

[back to top]

Percentage of Participants That Experienced a Dose Limiting Toxicity (DLT)

Dose-limiting toxicities (DLTs) were defined as grade 4 neutropenia or thrombocytopenia which lasts >7 days; grade 3 or 4 febrile neutropenia; grade 3 or greater non-hematological toxic effects. (NCT00715793)
Timeframe: Up to 26 months

Interventionpercentage of participants (Number)
Dose Level 1:DAC (Decitabine) 0.075 mg/kg + TMZ (Temozolomide)0
Dose Level 2:DAC (Decitabine) 0.15 mg/kg + TMZ (Temozolomide)17

[back to top] [back to top]

Progression-free Survival (PFS)

PFS was defined as the time from study entry until the documented radiological or symptomatic progression. Per RECIST v1.0 criteria (assessed by MRI or CT): Progressive Disease (PD); PD, 20% increase in the sum if target lesion or the appearance of new lesions. (NCT00715793)
Timeframe: Up to 42 months

Interventionmonths (Median)
DAC (Decitabine) + TMZ (Temozolomide)3.4

[back to top]

Overall Response Rate (ORR)

Using RECIST v1.0 criteria, overall response rate (ORR) was determined by the number of participants with complete response (CR) + the number of participants with partial response (PR) / the number of participants with complete response (CR) + the number of participants with partial response (PR) + the number of participants with stable disease (SD) + the number of participants with progressive disease (PD), multiplied by 100. Per RECIST v1.0 criteria (assessed by MRI or CT): Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD), neither sufficient shrinkage to qualify for a Partial Response (PR) nor sufficient increase to to qualify for Progressive Disease (PD); PD, 20% increase in the sum if target lesion or the appearance of new lesions; Complete Response (CR), Disappearance of all target lesions. (NCT00715793)
Timeframe: Up to 30 months

Interventionpercentage of participants (Number)
DAC (Decitabine) + TMZ (Temozolomide)18

[back to top]

Overall Survival (OS)

OS was defined as the time from study entry until the death or date of last contract. (NCT00715793)
Timeframe: Up to 42 months

Interventionmonths (Median)
DAC (Decitabine) + TMZ (Temozolomide)12.4

[back to top]

1-year Overall Survival (OS) Rate

Percentage of patients alive at one year (number of patients alive / total number of evaluable (analyzed) patients). (NCT00715793)
Timeframe: 12 months

Interventionpercentage of participants (Number)
DAC (Decitabine) + TMZ (Temozolomide)56

[back to top]

6-month Progression-free Survival (PFS) Rate

(NCT00715793)
Timeframe: 6 months

Interventionpercentage of participants (Number)
DAC (Decitabine) + TMZ (Temozolomide)32.4

[back to top]

Number of Participants Experiencing Unexpected Adverse Drug Reactions (ADRs)

An unexpected ADR was defined as an adverse reaction, whose nature, severity, specificity, or outcome is not consistent with the term or description used in the applicable product information. (NCT00723827)
Timeframe: Complete study duration & 30 days after completion (up to approximately 7.5 months)

Interventionparticipants (Number)
All Participants49

[back to top]

Number of Temozolomide Drug Interactions

Drug interaction was defined as a chemical or physiological reaction that can occur when two different drugs are taken together. (NCT00723827)
Timeframe: Complete study duration & 30 days after completion (up to approximately 7.5 months)

Interventionevents (Number)
All Participants5

[back to top]

Number of Temozolomide Misuse or Abuse Events

"Drug abuse was defined as the use of the study drug for a non-therapeutic effect.~Misuse was defined as use of the study medication in a way that was not prescribed." (NCT00723827)
Timeframe: Complete study duration & 30 days after completion (up to approximately 7.5 months)

InterventionEvents (Number)
All Participants1

[back to top]

Efficacy: Number of Participants Experiencing Complete Response (CR), Partial Response (PR), or Stable Disease(SD)

The response ratings were based on the judgment of the investigator. (NCT00723827)
Timeframe: Complete study duration (up to approximately 6.5 months)

Interventionparticipants (Number)
CRPRSD
All Participants82143322

[back to top]

Number of Participants Experiencing Adverse Events (AEs)

An AE was defined as any unfavorable and unintended sign, symptom, or disease temporally associated with the use of vaccine, whether or not considered related to the medicinal product. (NCT00723827)
Timeframe: Complete study duration & 30 days after completion (up to approximately 7.5 months)

Interventionparticipants (Number)
All Participants324

[back to top]

Number of Participants Who Discontinued Due to Toxicity

(NCT00725010)
Timeframe: Weekly during the concomitant treatment phase, and then monthly during the monotherapy phase

InterventionParticipants (Number)
Temozolomide+Radiotherapy8
Temozolomide Alone1

[back to top]

Safety: Number of Adverse Events in the Indicated Categories

(NCT00725010)
Timeframe: Weekly during the concomitant treatment phase, and then monthly during the monotherapy phase

,
InterventionAdverse Events (Number)
Unlikely Related to TemozolomidePossibly Related to TemozolomideProbably Related to Temozolomide
Temozolomide Alone101
Temozolomide+Radiotherapy351422

[back to top]

Incidence of Adverse Events, Based on CTC (Common Toxicity Criteria) Severity Grade

Safety variables will be summarized by descriptive statistics. Adverse Events (AEs) that occur will be reported for each phase and dose level and described in terms of incidence and severity. Parameters will be described based on the CTC severity grading. Distribution by CTC severity grade and clinical relevance will be given. (NCT00731731)
Timeframe: Up to 5 years

Interventionparticipants evaluable for toxicity (Number)
Phase I, Dose Level 012
Phase I, Dose Level 13
Phase II107

[back to top]

Overall Survival at 15 Months (Phase II)

The primary endpoint will be survival status at 15 months (OS15). In addition, survival will be estimated using a Kaplan-Meier curve. For this analysis, patients who are still alive at the time of analysis have survival time censored at the last contact date. (NCT00731731)
Timeframe: Time from study registration to the date of death from any cause, assessed up to 5 years

Interventionpercentage of phase II patients (Number)
Phase II54.6

[back to top]

Time to Tumor Progression (Phase II)

Progression free survival time will be defined from date of registration to date of progression or death. (NCT00731731)
Timeframe: Up to 5 years

Interventionmonths (Median)
Phase II8.05

[back to top] [back to top]

Incidence of Adverse Events, as Per NCI CTCAE Version 3.0 (Phase II)

"The maximum grade for each type of treatment-related adverse event will be recorded for each patient, and frequency tables for each arm will be reviewed to determine patterns. In addition, we will review all adverse event data that is graded as 3, 4, or 5 and classified as either unrelated or unlikely to be related to study treatment in the event of an actual relationship developing." (NCT00731731)
Timeframe: Up to 5 years

Interventionparticipants evaluable for toxicity (Number)
Phase I, Dose Level 012
Phase I, Dose Level 13
Phase II107

[back to top]

The Objective Overall Response

"The objective response is defined as all complete responses and partial responses based on the modified RECIST.Complete Response (CR): Disappearance of all target lesions Partial Response (PR): At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD. Progressive Disease (PD): At least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions.~Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum LD since the treatment started." (NCT00740636)
Timeframe: 2 years

,
Interventionparticipants (Number)
Partial Response (PR)Stable Disease (SD)Progression of Disease (POD)Complete Response (CR)
200 mg/m2/Day for 5 Days (23 Days Off tx). 28 Day Cycles.7790
75 mg/m2/Day for 21 Days (7 Days Off tx). 28 Day Cycles.1219311

[back to top]

Permeability-surface Area Product Before, During, and After Chemoradiotherapy

"Permeability-surface Area Product (Ktrans). Ktrans reflects the efflux rate of contrast from blood plasma into the tissue extravascular extracellular space (EES). Ktrans was assessed using post-contrast T1-weighted images. Multiple images were used to assess each participant at every time-point and the median value for each participant was calculated by time-point. The data presented represent the average of those median values at each time-point.~CRT: Chemoradiotherapy Cx: The cycle number TMZ: temozolomide" (NCT00756106)
Timeframe: Baseline, weekly during treatment, monthly following treatment for up to six months

Interventionmin ^-1 (Mean)
Baseline 1Baseline 2Week 1 CRTWeek 2 CRTWeek 3 CRTWeek 4 CRTWeek 5 CRTWeek 6 CRTPre-C1 TMZPre-C2 TMZPre-C3 TMZPre-C4 TMZPre-C5 TMZPre-C6 TMZPost-TMZ
Temozolomide and Radiation Therapy0.0530.0390.0550.0480.0550.0530.0580.0590.0630.0420.0500.0580.0560.0600.032

[back to top]

Relative Cerebral Blood Volume as Measured by Perfusion-weighted MRI Before, During, and After Chemoradiotherapy

"Relative cerebral blood volume (rCBV) is the blood volume in the region of interest (ROI) divided by the blood volume in the symmetrical region on the other side of the normal brain (control region). CBV was assessed using spin-echo post-contrast T1-weighted images. Multiple images were used to assess each participant at every time-point and the median value for each participant was calculated by time-point. The data presented represent the average of those median values at each time-point. The baseline value was measured twice (representing baseline 1 and 2) to make sure that the value was reproducible and to account for any variation attributable to measurement variation.~CRT: Chemoradiotherapy Cx: The cycle number TMZ: temozolomide" (NCT00756106)
Timeframe: Baseline, weekly during treatment, monthly following treatment for up to six months

Interventionratio (Mean)
Baseline 1Baseline 2Week 1 CRTWeek 2 CRTWeek 3 CRTWeek 4 CRTWeek 5 CRTWeek 6 CRTPre-C1 TMZPre-C2 TMZPre-C3 TMZPre-C4 TMZPre-C5 TMZPre-C6 TMZPost-TMZ
Temozolomide and Radiation Therapy0.880.900.951.00.880.830.860.760.680.610.590.650.610.640.32

[back to top]

Relative Cerebral Blood Flow as Measured by Perfusion-weighted MRI Before, During, and After Chemoradiotherapy

"Relative cerebral blood flow (rCBF) is the blood flow rate (the volume of blood passing through the specified are over a specified period of time) in the region of interest (ROI) divided by the blood flow rate in the symmetrical region on the other side of the normal brain (control region). CBF was assessed using spin-echo post-contrast T1-weighted images. CBF was assessed using spin-echo post-contrast T1-weighted images. Multiple images were used to assess each participant at every time-point and the median value for each participant was calculated by time-point. The data presented represent the average of those median values at each time-point. The baseline value was measured twice (representing baseline 1 and 2) to make sure that the value was reproducible and to account for any variation attributable to measurement variation.~CRT: Chemoradiotherapy Cx: The cycle number TMZ: temozolomide" (NCT00756106)
Timeframe: Baseline, weekly during treatment, monthly following treatment for up to six months

Interventionratio (Mean)
Baseline 1Baseline 2Week 1 CRTWeek 2 CRTWeek 3 CRTWeek 4 CRTWeek 5 CRTWeek 6 CRTPre-C1 TMZPre-C2 TMZPre-C3 TMZPre-C4 TMZPre-C5 TMZPre-C6 TMZPost-TMZ
Temozolomide and Radiation Therapy0.830.860.920.940.800.790.830.730.630.590.600.600.520.470.37

[back to top]

Apparent Diffusion Coefficient Before, During, and After Chemoradiotherapy

"Apparent diffusion coefficient (ADC) is a measure of the magnitude of diffusion (of water molecules) within tissue. ADC was assessed using post-contrast T1-weighted images. Multiple images were used to assess each participant at every time-point and the median value for each participant was calculated by time-point. The data presented represent the average of those median values at each time-point.~CRT: Chemoradiotherapy Cx: The cycle number TMZ: temozolomide" (NCT00756106)
Timeframe: Baseline, weekly during treatment, monthly following treatment for up to six months

Interventionmm2/s (Mean)
Baseline 1Baseline 2Week 1 CRTWeek 2 CRTWeek 3 CRTWeek 4 CRTWeek 5 CRTWeek 6 CRTPre-C1 TMZPre-C2 TMZPre-C3 TMZPre-C4 TMZPre-C5 TMZPre-C6 TMZPost-TMZ
Temozolomide and Radiation Therapy0.001100.001130.001060.001100.001180.001220.001250.001250.001320.001270.001080.001330.001480.001790.00140

[back to top]

Disease Progression at 10, 12, 14, 16, 18, 21, and 24 Months by Independent Review in the Intent-to-treat Population - Number of Participants

"Tumor response was classified based on the (neuro-)radiologist's evaluation:~Complete Response (CR): Disappearance of all enhancing tumor on consecutive MRI scans at least 1 month apart, off steroids, neurologically stable or improved.~Partial Response (PR): ≥50% reduction in size of enhancing tumor on consecutive MRI scans at least 1 month apart, steroids stable or reduced, neurologically stable or improved.~Progressive Disease (PD): ≥25% increase in size of enhancing tumor or any new tumor on MRI scans, steroids stable or increased, neurologically worse.~Stable Disease (SD): all other situations.~Based on clinical and imaging data, an independent neuro-oncologist made the final assessment of Progressed versus Not Progressed. Participants who had MRI assessment results missing or unknown were UNK or missing, and were treated as Progressed for the purposes of the calculation." (NCT00761280)
Timeframe: 10, 12, 14, 16, 18, 21, and 24 months

,
Interventionparticipants (Number)
Progressed at 10 monthsNot progressed at 10 monthsStatus unknown / missing at 10 monthsLost to follow-up at 10 monthsProgressed at 12 monthsNot progressed at 12 monthsStatus unknown / missing at 12 monthsLost to follow-up at 12 monthsProgressed at 14 monthsNot progressed at 14 monthsStatus unknown / missing at 14 monthsLost to follow-up at 14 monthsProgressed at 16 monthsNot progressed at 16 monthsStatus unknown / missing at 16 monthsLost to follow-up at 16 monthsProgressed at 18 monthsNot progressed at 18 monthsStatus unknown / missing at 18 monthsLost to follow-up at 18 monthsProgressed at 21 monthsNot progressed at 21 monthsStatus unknown / missing at 21 monthsLost to follow-up at 21 monthsProgressed at 24 monthsNot progressed at 24 monthsStatus unknown / missing at 24 monthsLost to follow-up at 24 months
Chemotherapy5071507150715071507150715071
Trabedersen 10 µM7340725072507250725071607160

[back to top]

Survival Rate at 12, 18, and 21 Months in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)

Survival rate was defined as the proportion of participants known to be alive at each time-point from randomization. Participants with unknown or missing status were considered treatment failures, i.e., assumed to be dead. (NCT00761280)
Timeframe: 12, 18, and 21 months

,
Interventionpercentage of participants (Number)
Survival rates at 12 monthsSurvival rates at 18 monthsSurvival rates at 21 months
Chemotherapy53.838.523.1
Trabedersen 10 µM57.142.935.7

[back to top]

Survival Rate at 24 Months in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)

Survival rate was defined as the proportion of participants known to be alive at 24 months from randomization. If a participant's status was unknown and there was no follow-up information available, they were categorized as 'Died' for the purposes of the analysis. (NCT00761280)
Timeframe: 24 months

Interventionpercentage of participants (Number)
Trabedersen 10 µM28.6
Chemotherapy15.4

[back to top]

Survival at 24 Months in the Intent-to-treat Population - Number of Participants

"Survival status was assessed at 24 months from randomization. Participants with unknown or missing status were considered treatment failures, i.e., assumed to be dead. The category Lost to / insufficient follow-up includes participants who were alive at last data collection point but did not yet have enough follow-up time to reach the 24 month time point." (NCT00761280)
Timeframe: 24 months

,
Interventionparticipants (Number)
AliveDiedLost to/insufficient follow-up
Chemotherapy256
Trabedersen 10 µM473

[back to top]

Tumor Control Rate (CR+PR+SD) by Independent Review in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)

Tumor control rate was defined as the proportion of participants assessed as having Complete Response (CR), Partial Response (PR), or Stable Disease (SD). Participants with unknown or missing response were treated as non-responders. (NCT00761280)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
Trabedersen 10 µM28.6
Chemotherapy53.8

[back to top]

Survival at 12, 18, and 21 Months in the Intent-to-treat Population - Number of Participants

"Survival status was assessed at each time-point from randomization. Participants with unknown or missing status were considered treatment failures, i.e., assumed to be dead. The category Lost to follow-up for each time-point includes participants who were alive at the last data collection point but did not yet have enough follow-up time to reach the time point." (NCT00761280)
Timeframe: 12, 18, and 21 months

,
Interventionparticipants (Number)
Alive at 12 monthsDied at 12 monthsLost to follow-up at 12 monthsAlive at 18 monthsDied at 18 monthsLost to follow-up at 18 monthsAlive at 21 monthsDied at 21 monthsLost to follow-up at 21 months
Chemotherapy715535355
Trabedersen 10 µM851671572

[back to top]

Median Duration of Response (Days) by Independent Review (Descriptive Analysis, Only)

"Duration of response was defined as the time from the first documentation of confirmed response (Complete Response, CR, or Partial Response, PR) to the first signs of Progressive Disease (PD), as assessed by the study neuro-oncologist. Median Duration of Response was calculated by Kaplan-Meier estimate.~Censoring rules were:~at the date of randomization -- participants without baseline assessments, or for those with no post-baseline timor assessments who were discontinued for other than progressive disease or death.~at the date of last tumor assessment -- discontinuation other than PD or death, or if a new treatment was started prior to disease progression~at the date of death or last tumor assessment -- death or PD after one missed tumor assessment~at the date of last tumor assessment -- death or PD after more than one missed tumor assessment~at the date of last tumor assessment -- participants on ongoing treatment at data cut-off" (NCT00761280)
Timeframe: Up to 24 months

Interventiondays (Median)
Trabedersen 10 µMNA
ChemotherapyNA

[back to top]

Median Overall Survival (Days) From Randomization in the Intent-to-treat Population (Descriptive Analysis, Only)

Median overall survival was defined as the date of randomization to the date of death. If a participant's status was unknown and there was no follow-up information available, they were categorized as 'Died' for the purposes of the analysis. Analysis was by Kaplan-Meier estimation. (NCT00761280)
Timeframe: Up to 24 months

Interventiondays (Median)
Trabedersen 10 µM458.0
Chemotherapy584.5

[back to top]

Median Time to Progression (Days) by Independent Review for the Intent-to-treat Population (Descriptive Analysis, Only)

Time to progression was calculated from the date of randomization to the date of the first documented tumor progression. Participants who did not progress or died were censored at the last tumor assessment date or the date of start of a new anti-tumor treatment or death. (NCT00761280)
Timeframe: Up to 24 months

Interventiondays (Median)
Trabedersen 10 µM57.0
Chemotherapy153.5

[back to top]

Response Category by Independent Review in the Intent-to-treat Population - Number of Participants

"Tumor response was classified based on the (neuro-)radiologist's evaluation according to the Macdonald Response Criteria for bidimensionally measurable disease as outlined below:~Complete Response (CR): Disappearance of all enhancing tumor on consecutive MRI scans at least 1 month apart, off steroids, neurologically stable or improved.~Partial Response (PR): ≥50% reduction in size of enhancing tumor on consecutive MRI scans at least 1 month apart, steroids stable or reduced, neurologically stable or improved.~Progressive Disease (PD): ≥25% increase in size of enhancing tumor or any new tumor on MRI scans, steroids stable or increased, neurologically worse.~Stable Disease (SD): all other situations.~Two qualified neuro-radiologists reviewed scans at each MRI time point, with adjudication of discrepancies by a third reviewer. Their findings and clinical information were independently reviewed by a neuro-oncologist, who made the assessment of overall response." (NCT00761280)
Timeframe: Up to 24 months

,
Interventionparticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)UnknownMissing
Chemotherapy016132
Trabedersen 10 µM022712

[back to top]

Overall Response Rate (CR+PR) by Independent Review in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)

Overall response rate was the proportion of participants with a best response of Complete Response (CR) or Partial Response (PR) observed from the start of treatment until disease progression. (NCT00761280)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
Trabedersen 10 µM14.3
Chemotherapy7.7

[back to top]

Disease Progression Rate at 10, 12, 14, 16, 18, 21, and 24 Months by Independent Review in the Intent-to-treat Population - Percentage of Participants (Descriptive Analysis, Only)

"Tumor response was classified based on the (neuro-)radiologist's evaluation:~Complete Response (CR): Disappearance of all enhancing tumor on consecutive MRI scans at least 1 month apart, off steroids, neurologically stable or improved.~Partial Response (PR): ≥50% reduction in size of enhancing tumor on consecutive MRI scans at least 1 month apart, steroids stable or reduced, neurologically stable or improved.~Progressive Disease (PD): ≥25% increase in size of enhancing tumor or any new tumor on MRI scans, steroids stable or increased, neurologically worse.~Stable Disease (SD): all other situations.~Based on clinical and imaging data, an independent neuro-oncologist made the final assessment of Progressed versus Not Progressed. Participants who had MRI assessment results missing or unknown were UNK or missing, and were treated as Progressed for the purposes of the calculation." (NCT00761280)
Timeframe: 10, 12, 14, 16, 18, 21 and 24 months

,
Interventionpercentage of participants (Number)
Progression rate at 10 monthsProgression rate at 12 monthsProgression rate at 14 monthsProgression rate at 16 monthsProgression rate at 18 monthsProgression rate at 21 monthsProgression rate at 24 months
Chemotherapy100.0100.0100.0100.0100.0100.0100.0
Trabedersen 10 µM78.685.785.785.785.792.992.9

[back to top]

Progression Free Survival

(NCT00782756)
Timeframe: through study completion, an average of 1 year

Interventionmonths (Median)
RT, With Temozolomide and Bevacizumab10

[back to top]

Number of Participants With Adverse Events

Safety assessments and toxicity grading will follow CTCAE Version 4 Grade (NCT00782756)
Timeframe: through study completion, an average of 1 year

InterventionParticipants (Count of Participants)
RT, With Temozolomide and Bevacizumab40

[back to top]

12-Month Overall Survival (OS) Rate

The 12-month overall survival rate was defined as the percentage of participants surviving at 12 months. The distribution of 12-month OS rate was estimated using Kaplan-Meier methodology. Point estimates and 95% CIs for the quartiles for the PFS distribution are provided. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ treatment groups were statistically significantly better than the Placebo + TMZ treatment group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints, regardless of the observed P values. (NCT00804908)
Timeframe: Per protocol, survival was to be assessed every 4 weeks or as needed after participant is registered as off-study for up to 18 months. The maximum observed follow-up at the overall survival analysis time was 21.0 months.

Interventionpercentage of participants (Number)
Placebo for ABT-888 BID + TMZ QD52.6
ABT-888 20 mg BID + TMZ QD43.5
ABT-888 40 mg BID + TMZ QD54.1

[back to top]

Disease Control Rate

The disease control rate was defined as the percentage of participants who had at least stable disease (complete response, partial response, or stable disease) through the end of Week 8. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ treatment groups were statistically significantly better than the Placebo + TMZ treatment group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints, regardless of the observed P values. (NCT00804908)
Timeframe: Week 8

Interventionpercentage of participants (Number)
Placebo for ABT-888 BID + TMZ QD48.7
ABT-888 20 mg BID + TMZ QD62.9
ABT-888 40 mg BID + TMZ QD59.1

[back to top]

Objective Response Rate

The objective response rate was defined as the percentage of participants with a confirmed CR or PR per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by computed tomography (CT) scan: complete response (CR), disappearance of all target lesions; partial response (PR), ≥30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ treatment groups were statistically significantly better than the Placebo + TMZ treatment group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints, regardless of the observed P values. (NCT00804908)
Timeframe: Every 2 cycles (8 weeks) until disease progression was observed or another reason for discontinuation of assessments was identified by the investigator. The maximum observed followup duration at the progression-free survival analysis time was 9.7 months.

Interventionpercentage of participants (Number)
Placebo for ABT-888 BID + TMZ QD7.0
ABT-888 20 mg BID + TMZ QD10.3
ABT-888 40 mg BID + TMZ QD9.6

[back to top]

Overall Survival (OS): Time to Event

OS was defined as the number of days from the date the participant was randomized to the date of death. All deaths were included, whether the participant was still taking or had discontinued study drug. If a participant had not died and was lost to follow-up, then data were censored at the last study visit or contact date, or date the participant was last known to be alive, whichever was later; if the participant was not lost to follow-up, then data were censored at the last study visit or contact date, whichever was later. The distribution of OS was estimated for each treatment group using Kaplan-Meier methodology. Point estimates and 95% CIs for the quartiles for the OS distribution are provided. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ groups were statistically significantly better than the Placebo + TMZ group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints. (NCT00804908)
Timeframe: Per protocol, survival follow-up information was to be obtained every 3 months for up to 18 months after the final visit for the subject. The maximum observed follow-up at the overall survival analysis time was 21.0 months.

,,
Interventiondays (Number)
25th Percentile50th percentile75th percentile
ABT-888 20 mg BID + TMZ QD204327NA
ABT-888 40 mg BID + TMZ QD181412NA
Placebo for ABT-888 BID + TMZ QD207390559

[back to top]

Progression-Free Survival (PFS): Time to Event

PFS: the number of days from the date that the participant was randomized to the date the participant experienced a confirmed event of disease progression (radiological, as determined by the central imaging center; or clinical, as determined by the investigator), or to the date of death (all causes of mortality) if disease progression was not reached. All events were included whether the participant was still taking or had discontinued study drug. Events of death were included for participants who had not experienced a confirmed event of disease progression, provided the death occurred within 8 weeks of the last available disease progression assessment. The distribution of PFS, as determined by the central imaging center (radiological)/ investigator (clinical), was estimated for each treatment group using Kaplan-Meier methodology. Point estimates and 95% confidence intervals (95% CIs) for the quartiles for the PFS distribution are provided. (NCT00804908)
Timeframe: Every Cycle (28 Days) until disease progression was observed or another reason for discontinuation of assessments was identified by the investigator. The maximum observed followup duration at the progression-free survival analysis time was 9.7 months.

,,
Interventiondays (Number)
25th Percentile50th Percentile75th Percentile
ABT-888 20 mg BID + TMZ QD56113225
ABT-888 40 mg BID + TMZ QD53110226
Placebo for ABT-888 BID + TMZ QD5460163

[back to top]

Time to Disease Progression

The distribution of time to disease progression, as determined by the central imaging center (radiological)/ investigator (clinical), was estimated for each treatment group using Kaplan-Meier methodology. Point estimates and 95% CIs for the quartiles for the PFS distribution are provided. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ treatment groups were statistically significantly better than the Placebo + TMZ treatment group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints, regardless of the observed P values. (NCT00804908)
Timeframe: Every Cycle (28 Days), until disease progression was observed or another reason for discontinuation of assessments was identified by the investigator. The maximum observed followup duration at the progression-free survival analysis time was 9.7 months.

,,
Interventiondays (Number)
25th Percentile50th percentile75th percentile
ABT-888 20 mg BID + TMZ QD56113225
ABT-888 40 mg BID + TMZ QD53110226
Placebo for ABT-888 BID + TMZ QD5460163

[back to top]

Time to Neurological/Brain Metastases Progression

Time to neurological/brain metastases progression, defined as the number of days from the date of randomization to the date the participant experienced an event of neurological/brain metastases progression, was estimated using Kaplan-Meier methodology. Point estimates and 95% CIs for the quartiles for the distribution are provided. All events of progression were included, regardless of whether the event occurred while the participant was still taking study drug. If a participant did not experience an event, data were censored at the date of the last available brain CT scan. For participants with no postbaseline brain CT scans, data were censored at randomization. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ groups were statistically significantly better than the Placebo + TMZ group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints, regardless of the observed P values. (NCT00804908)
Timeframe: Every 2 cycles (8 weeks) until disease progression was observed or another reason for discontinuation of assessments was identified by the investigator. The maximum observed followup duration at the progression-free survival analysis time was 9.7 months.

,,
Interventiondays (Number)
25th Percentile50th percentile75th percentile
ABT-888 20 mg BID + TMZ QD119NANA
ABT-888 40 mg BID + TMZ QD184184NA
Placebo for ABT-888 BID + TMZ QD60NANA

[back to top]

6-month Progression-Free Survival Rate

The 6-month progression-free survival rate was defined as the percentage of participants without disease progression at 6 months.The distribution of 6-month progression-free survival rate, as determined by the central imaging center (radiological)/ investigator (clinical), was estimated using Kaplan-Meier methodology. Point estimates and 95% CIs for the quartiles for the PFS distribution are provided. Per protocol, because neither the ABT-888 20 mg BID + TMZ nor ABT-888 40 mg BID + TMZ treatment groups were statistically significantly better than the Placebo + TMZ treatment group for the primary endpoint of PFS, confirmatory statistical testing was not continued for any secondary endpoints, regardless of the observed P values. (NCT00804908)
Timeframe: Every Cycle (28 Days) until disease progression was observed or another reason for discontinuation of assessments was identified by the investigator. The maximum observed followup duration at the progression-free survival analysis time was 9.7 months.

Interventionpercentage of participants (Number)
Placebo for ABT-888 BID + TMZ QD19.1
ABT-888 20 mg BID + TMZ QD32.8
ABT-888 40 mg BID + TMZ QD30.7

[back to top]

Number of Participants Experiencing Toxicity After This Novel Multimodality Regimen

The toxicity assessments were made according to the common terminology criteria for adverse events (CTCAE version 3.0) of the National Cancer Institute. Number of participants with Grade 1 to 5 adverse events are reported here. (NCT00805961)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Overall Study53

[back to top]

Objective Response Rate of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen

Response to treatment was assessed by MRI using the MacDonald criteria based on the assessment of the MRI scan for measurable, evaluable, and new lesions. The objective response rate is defined as the proportion of patients with improvement and or decreased extent of lesions compared to baseline. (NCT00805961)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Overall Study31

[back to top]

Overall Survival of Patients With Glioblastoma Multiforme Following Treatment With This Novel Multimodality Regimen

Overall survival was defined as the interval from the first day of study treatment until the date of death. (NCT00805961)
Timeframe: 18 months

InterventionMonths (Number)
Overall Study13.9

[back to top]

Progression-free Survival (PFS)

Progression-free survival is defined as the duration of time from start of treatment to time of progression or death, whichever comes first. (NCT00805961)
Timeframe: 18 months

InterventionMonths (Median)
Overall Study11.3

[back to top]

Overall Survival (OS) Time

The OS time is defined as the time (in months) from randomization to death or last day known to be alive. Participants without event are censored at the last date known to be alive or at the clinical cut-off date, whatever is earlier. (NCT00813943)
Timeframe: Time from randomization to death or last day known to be alive, reported between day of first participant randomized, that is, Jun 2009 until cut-off date, (07 Feb 2013)

InterventionMonths (Median)
Cilengitide (2-times Weekly) + Temozolomide + Radiotherapy16.3
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy14.5
Temozolomide + Radiotherapy13.4

[back to top]

Number of Participants With AEs Belonging to Standardized Medical Dictionary for Regulatory Activities (MedDRA) Queries (SMQs) Thromboembolic Events and Hemorrhage With NCI-CTC Toxicity Grade 3 or 4

Thromboembolic events (standardized MedDRA query [SMQ]) Grade 3 or 4 AEs encompassed hemiparesis and cerebrovascular accident, pulmonary embolism, and deep vein thrombosis. Thromboembolic events (SMQ) of any grade and of Grade 3 or 4 were generally more frequent in the Cilengitide + Temozolomide/Radiotherapy group than in the Temozolomide/Radiotherapy group but were still in the expected range of this patient population The severity of AEs was assessed according to the National Cancer Institute-Common Toxicity Criteria (NCI-CTCAE) (version 3.0): Grade 1=mild, Grade 2=moderate, Grade 3=severe, Grade 4=life threatening or disabling. Note: Death (Grade 5) was regarded as an outcome. (NCT00813943)
Timeframe: Time from first dose up to 28 days after last dose of study treatment, reported between day of first participant randomized, that is, Jun 2009 until cut-off date (07 Feb 2013)

,,
InterventionParticipants (Number)
SMQ: Thromboembolic eventsSMQ:Hemorrhage
Cilengitide (2-times Weekly) + Temozolomide + Radiotherapy173
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy100
Temozolomide + Radiotherapy121

[back to top]

Apparent Terminal Rate Constant

The apparent terminal rate constant for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionper hour (Mean)
Day 1 (Single dose)Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy0.320.32

[back to top]

Apparent Volume of Distribution During the Terminal Phase (Vz) and Apparent Volume of Distribution at Steady State (Vss)

The Vz (after single dose) and Vss (after repeated doses) for cilengitide were calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionliter (Mean)
VzVss
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy24.719.2

[back to top]

Area Under the Plasma Concentration-Time Curve From Time 0 to Infinity (AUC [0-infinity]) and Area Under the Plasma Concentration-Time Curve From Time 0 to 24 Hours (AUC [0-24])

The AUC (0-infinity) and AUC (0-24) for cilengitide were calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionhour*ng/mL (Mean)
AUC (0-infinity): Day 1 (Single dose)AUC (0-infinity): Day 5 (Repeated Doses)AUC (0-24): Day 1 (Single dose)AUC (0-24): Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy280944335263269941316137

[back to top]

Plasma Clearance (CL)

The CL for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionmilliliter per minute (Mean)
Day 1 (Single dose)Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy125.7109.3

[back to top]

Maximum Observed Plasma Concentration (Cmax)

The Cmax for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionnanogram per milliliter (ng/mL) (Mean)
Day 1 (Single dose)Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy108527150873

[back to top]

Progression Free Survival (PFS) Time - Investigator and Independent Read

The PFS time is defined as the duration from randomization to either first observation of progressive disease (PD) or occurrence of death due to any cause. Investigator read is the assessment of all imaging by the treating physician at the local trial site. Independent Read is the assessment of all imaging centrally by an Independent Review Committee (IRC). (NCT00813943)
Timeframe: Time from randomization to disease progression, death or last tumor assessment, reported between day of first participant randomized, that is, Jun 2009 until cut-off date, (07 Feb 2013)

,,
InterventionMonths (Median)
PFS Time: Independent readPFS Time: Investigator read
Cilengitide (2-times Weekly) + Temozolomide + Radiotherapy5.66.4
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy5.97.5
Temozolomide + Radiotherapy4.16.0

[back to top]

Time to Maximum Plasma Concentration (Tmax) and Terminal Elimination Half-Life (t1/2)

The Tmax and t1/2 for cilengitide were calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionhours (Mean)
Tmax: Day 1 (Single dose)Tmax: Day 5 (Repeated doses)t1/2: Day 1 (Single dose)t1/2: Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy0.971.172.382.44

[back to top] [back to top]

Mean Residence Time From Time 0 to Infinity (MRT [0-infinity])

The MRT (0-infinity) for cilengitide was calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionhour (Mean)
Day 1 (Single dose)Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy2.82.9

[back to top]

Plasma Concentration at Pre-dose (Cpre) and Plasma Concentration at End of Infusion (CT)

The Cpre and CT for cilengitide were calculated by non-compartmental analysis using the computer program WinNonlin, Version 6.2. Cilengitide plasma concentrations were determined after dosing on Day 1 (single dose) and Day 5 (repeated doses) of Week 1. (NCT00813943)
Timeframe: Days 1 and 5 of Week 1

Interventionng/mL (Mean)
Cpre: Day 1 (Single dose)Cpre: Day 5 (Repeated doses)CT: Day 1 (Single dose)CT: Day 5 (Repeated doses)
Cilengitide (5-times Weekly) + Temozolomide + Radiotherapy6372.7286.0108045.5157470.0

[back to top]

Number of Participants With an Overall Response (Complete Response or Partial Response) Assessed by the MacDonald Criteria

Anti-tumor activity as determined by the overall response (Complete response (CR) or partial response (PR)) was assessed by the MacDonald criteria. Complete response is complete resolution of all lesions. The patient cannot be on any corticosteroids with the exception of adrenal replacement doses. Partial response is ≥50% reduction in the sum of products of all measurable lesions over baseline sum observed using the same techniques as baseline. The patient must be on a stable or decreased dose of corticosteroids to be evaluable for response. (NCT00826241)
Timeframe: 4 weeks

InterventionParticipants (Count of Participants)
Complete ResponsePartial Response
Temozolomide + Lapatinib28

[back to top]

Time to Progression

Time to progression defined as progressive disease, toxicity at a level of severity that precludes the patient continuing on the protocol, or death. Progression is a 25% increase in the sum of products of all measurable lesions over smallest sum observed (over baseline if no decrease) using the same techniques as baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer). (NCT00826241)
Timeframe: Assessed every two months till disease progression, up to 4 years

InterventionMonths (Median)
Temozolomide + Lapatinib7.8

[back to top]

Number of Participants With Serious and Non-Serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0)

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. (NCT00826241)
Timeframe: Date treatment consent signed to date off study, approximately 111 months and 26 days

InterventionParticipants (Count of Participants)
Temozolomide + Lapatinib50

[back to top]

Time Spent in a Karnofsky Performance Status of 60-100%

Time spent in a KPS ≥70 was calculated from the date of diagnosis of Karonofsky Performance Status decline (KPS<70) or censored at the last date the patient was known with KPS ≥70. The KPS higher scores indicates normal activity status. (NCT00841555)
Timeframe: up to 12-16 months

Interventionmonths (Median)
Hypofractionation Radiotherapy+Temozolomide8.1

[back to top]

Survival Time

All patients will be followed to death. Active follow-up with disease evaluation with scans will be terminated if the patient's physician deems it in the patient's interest not to continue or upon patient request. (NCT00841555)
Timeframe: up to 2 years

Interventionmonths (Median)
Hypofractionation Radiotherapy+Temozolomide12.7

[back to top]

Maximum Tolerated Dose(MTD)of Temozolomide(TMZ)

This study is designed as a phase I dose escalation trial using the Standard Method of dose escalation of three patients per dose level to determine the MTD of TMZ (up to 75 mg/m 2 /day) when TMZ is used with HIMRT for patients with glioblastoma multiforme(GBM) or Anaplastic Astrocytoma(AA)of the brain. The 3 dose levels will be evaluated using the standard method to determine if either represents an MTD based on DLT. If DLT is not observed at all doses level, the greater of the three levels will be recommended for phase II evaluations of treatment effect. (NCT00841555)
Timeframe: up to 12-16 months

Interventionmg/m^2 (Number)
Hypofractionation Radiotherapy+Temozolomide75

[back to top]

Response by MRI Measurements

partial response by RICST criteria is defined as >50% tumor shrinkage (NCT00846430)
Timeframe: evaluated 6, 12 and 24 months compared to baseline

Interventionparticipants (Number)
Partial Response at 6 monthsComplete Response at 6 monthsLess than Partial Response at 6 monthsPartial Response at 12 monthsComplete Response at 12 monthsLess than Partial Response at 12 monthsPartial Response at 24 monthsComplete Response at 24 monthsLess than Partial Response at 24 monthsno longer participating at 24 months
Open-Label Intervention0182164113

[back to top]

No Reported Psychological Toxicity Based Upon Psychological Evaluations

Psychological toxicity defined as suicidal ideation (NCT00846430)
Timeframe: Psychological evaluation at 24 months

InterventionParticipants (Count of Participants)
Open-Label Intervention8

[back to top]

Improvement of Symptoms and Pain

Subjects will be evaluated for pain and Quality of Life scores (NCT00846430)
Timeframe: Monthly physical exam first three months and then every three months after, for up to 36 months

InterventionParticipants (Count of Participants)
Open-Label Intervention9

[back to top]

At Least 50% Shrinkage in Tumor Measurements by Physical Examination

(NCT00846430)
Timeframe: Monthly physical exam first three months and then every three months after, for up to 36 months

InterventionParticipants (Count of Participants)
Open-Label Intervention7

[back to top]

Number of Participants With Complete Response (CR)

CR according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, which is defined as disappearance of all target lesions (primary and metastases), signs, symptoms, and biochemical changes related to the tumor for >4 weeks, during which no new lesions may appear and no existing lesion may enlarge. (NCT00869050)
Timeframe: 12 months

Interventionparticipants (Number)
Capecitabine and Temozolomide3

[back to top]

Number of Participants With Partial Response (PR)

PR according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, which is defined as a reduction of ≥ 30% in the sum of the longest diameter for all target lesions lasting > 4 weeks, during which no new lesions may appear, when compared with with pretreatment measurements. (NCT00869050)
Timeframe: 12 months

Interventionparticipants (Number)
Capecitabine and Temozolomide9

[back to top]

Overall Survival

Overall survival (OS) is the primary endpoint and is defined as the time from study registration to time of death due to any cause. All patients who meet the eligibility criteria, have signed a consent form, and have received at least one dose of the regimens will be considered evaluable. Patients who are lost to follow-up will be censored at the date of their last follow-up. Patients still alive at the time of analysis will be censored. Only Phase II was evaluated for survival (NCT00869401)
Timeframe: Up to 5 years post treatment

InterventionMonths (Median)
Group 1 (Phase II) Dasatinib + Radiation + Temozolomide15.6
Group 2 (Phase II) Placebo + Radiation + Temozolomide19.3

[back to top]

Objective Response

Objective response to treatment will be determined by a combination of the results of neurological exam and the MRI and/or CT measurement of the tumor at each evaluation as is used for all NCCTG neuro-oncology trials. The proportion of patients in each response category will be summarized. Only phase II patients were evaluated for response. (NCT00869401)
Timeframe: Up to 5 years post treatment

,
InterventionProportion of participants (Number)
ProgressionStableREGRPartial ResponseComplete Response
Group 1 (Phase II) Dasatinib + Radiation + Temozolomide.31.59.08.020
Group 2 (Phase II) Placebo + Radiation + Temozolomide.19.73.02.060

[back to top]

Progression-free Survival

Progression-free survival (PFS) is defined as the time from study registration to the date of first observation of disease progression or death due to any cause (whichever comes first). If a patient has not progressed or died, progression-free survival is censored at the time of last follow-up. Only Phase II patients were evaluated for Progression-free survival (NCT00869401)
Timeframe: Up to 5 years post treatment

InterventionMonths (Median)
Group 1 (Phase II) Dasatinib + Radiation + Temozolomide6.2
Group 2 (Phase II) Placebo + Radiation + Temozolomide7.8

[back to top]

The Number of Dose Limiting Toxicities(DLT) in Order to Determine Maximum Tolerable Dose(MTD) of Dasatinib Combined With Radiation and Temozolomide in This Patient Population.

Doselimiting toxicity will be defined as: Adverse event at least possibly related to the study medication. All by CTCAE v3.0 criteria: Greater than or equal to grade 3: diarrhea or skin rash or desquamation or (other) clinically relevant non-hematological adverse event or non-hematologic adverse event at least possibly due to drug therapy. Or greater than or equal to grade 4: neutropenia or leukopenia or thrombocytopenia or radiation dermatitis or hematologic adverse event OR failure to administer greater than 75% of dasatinib TMZ or interruption of RT for more than 5 days due to adverse events.OR severe acute central nervous system deterioration attributable to TMZ, RT and or dasatinib which cannot be controlled with corticosteroid administration. The MTD for this study will be defined as the highest safely tolerated dose level where at most 1 out of 6 patients experience DLT with the next higher dose having at least 2 patients out of a maximum of 6 patients experience DLT. (NCT00869401)
Timeframe: Every cycle from first dose to end of rest period prior cycle 3

Interventionparticipants with Dose Limiting Toxicits (Number)
Dose Level 0 Phase I1
Dose Level 0-A Phase I0
Dose Level 1 Phase I1

[back to top]

Measurement of Number of Adverse Events

Collect and grade the all of the adverse events to evaluate for safety. This data was collected for the first 2 cycles for each participant. (NCT00876993)
Timeframe: Two 28-day cycles

,,,,
InterventionAdverse events (Number)
Grade 1 adverse eventsGrade 2 adverse eventsGrade 3 adverse eventsGrade 4 adverse events
Cohort 1 - Dose Level 14431
Cohort 2 - Dose Level 06310
Cohort 3 - Dose Level 13310
Cohort 4 - Dose Level 25540
Cohort 5 - Dose Level 13210

[back to top]

To Provide Safety and Efficacy Data for to Recommend Further Larger Studies.

Number participants with grade 3 and 4 hematologic and non-hematologic toxicities. All toxicities are for end of cycle 2. (NCT00876993)
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 Other
Cohort 1 - Dose Level 120001101
Cohort 2 - Dose Level 000000010
Cohort 3 - Dose Level 101000000
Cohort 4 - Dose Level 211113200
Cohort 5 - Dose Level 110000000

[back to top]

2 Year Event Free Survival With Children Treated With This Regimen.

2 year actual event free survival.with children treated with this protocol (NCT00876993)
Timeframe: 2 year

InterventionCount of participants (Number)
Cohort 1 - Dose Level 14
Cohort 2 - Dose Level 06
Cohort 3 - Dose Level 13
Cohort 4 - Dose Level 25
Cohort 5 - Dose Level 13

[back to top]

Progression-free Survival (PFS)

Progression-free survival was defined as time from randomization to date of progression, death, or last follow-up, and was estimated by the Kaplan-Meier method. Patients last known to be alive were censored at the date of last contact. This analysis was planned to occur when 390 deaths had been reported. (NCT00884741)
Timeframe: From randomization to date of progression, death, or last follow-up for progression-free survival. Analysis occurs after all 390 deaths have been reported.

Interventionmonths (Median)
Randomized Arm 1: TMZ+RT + Placebo7.3
Randomized Arm 2: TMZ+RT + Bevacizumab10.7

[back to top]

Overall Survival (OS)

Survival time was defined as time from randomization to date of death from any cause and was estimated by the Kaplan-Meier method. Patients last known to be alive were censored at the date of last contact. This analysis was planned to occur when 390 deaths had been reported. (NCT00884741)
Timeframe: From randomization to date of death or last follow-up. Analysis occurs after all 390 deaths have been reported.

Interventionmonths (Median)
Randomized Arm 1: TMZ+RT + Placebo16.1
Randomized Arm 2: TMZ+RT + Bevacizumab15.7

[back to top] [back to top]

Overall Response to CVT Chemotherapy.

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 (NCT00885534)
Timeframe: 2 years

Interventionparticipants (Number)
Partial ResponseStable Disease
Cisplatin, Vinblastine, Temozolomide15

[back to top]

Median Overall Survival

Overall survival is defined as the time from surgical resection to death of any cause. (NCT00905060)
Timeframe: Up to 3 years

Interventionmonths (Median)
Protein Peptide-Complex (HSPPC-96)23.8

[back to top]

Median PD-L1 Positivity in Circulating Myeloid Cells

Circulating myeloid cells (CD45+/CD11b+) obtained from patients at the time of surgery were analyzed for PD-L1 expression to determine the percent of myeloid cells positive for PD-L1 (cut off for positivity determined relative to Fluorescence Minus One (FMO) and isotype control). (NCT00905060)
Timeframe: Up to 53 Weeks

Interventionpercentage of PD-L1 positivity (Median)
Protein Peptide-Complex (HSPPC-96)54.5

[back to top]

Median Progression Free Survival (PFS)

PFS is defined as the duration of progression-free survival from the time from resection until either documented disease progression or death (NCT00905060)
Timeframe: Up to 3 years

Interventionmonths (Median)
Protein Peptide-Complex (HSPPC-96)18

[back to top] [back to top]

Phase 2 -Number of Patients With a Decrease in Tumor Size Using RECIST and CHOI's Criteria

"Tumor response rate was assessed using RECIST criteria in which a complete response was the disappearance of all target lesions; Partial response was a 30% decrease in the sum of the longest dimension (LD) of target lesions, relative to baseline measurement; Progressive disease was an increase of 20% or more in the sum of the LD of target lesions; and Stable disease was a decrease in tumor size of less than 30% or increase of less than 20%.~Tumor response rate was also assessed using CHOI's criteria in which a response was a 10% decrease in tumor size or a 15% decrease in tumor density on contrast-enhanced computed tomography scan.~Tumor response rates were assessed in order to determine effectiveness of the treatment regimen which was defined by the response rate of at least 30% as measured by either the RECIST or Choi's criteria, and ineffective if the rate is less than 15% on both." (NCT00925132)
Timeframe: 12 weeks (2 cycles)

InterventionParticipants (Count of Participants)
Progressive Disease (PD)Stable Disease (SD)
Phase II:Decitabine 0.2 mg/kg +Panobinostat 30mg +Temozolomide125

[back to top]

Phase I - Number of Participants With Dose Limiting Toxicities (DLTs) at a Given Dose Level

"A DLT was any Grade 4 toxicity for neutrophils or platelets for ≥ 7 days, Grade 3 toxicity for neutrophils for ≥ 21 days, any Grade 3 solid organ toxicity not explainable by another cause (e.g. neurotoxicity, GI toxicity), metabolic/laboratory toxicity (≥10 x ULN AST) for ≥ 14 days, any Grade 4 infection or QTcF> 500msec EKG. DLTs were assessed according to the Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0.~All adverse events were collected and graded to determine DLTs as assessed according to the Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0. DLTs were assessed to determine the recommended Decitabine and Panobinostat dose for the Phase II portion of the trial." (NCT00925132)
Timeframe: 6 weeks (one full cycle)

InterventionParticipants (Count of Participants)
Cohort 1: Experienced DLTCohort 2: Experienced DLTCohort 3: Experienced DLTCohort 4: Experienced DLT
Phase I Dose Escalation0000

[back to top]

Phase II: Median Overall Survival (OS)

Phase II: Time in months from the start of study treatment to date of death due to any cause. Patients alive as of the last follow-up had OS censored at the last follow-up date. Median OS was estimated using a Kaplan-Meier curve. (NCT00939991)
Timeframe: 3 years

Interventionmonths (Median)
Phase II12.5

[back to top]

Phase II: Median Progression-free Survival (PFS)

Phase II: Time in months from the start of study treatment to the date of first progression according to RANO criteria, or to death due to any cause. Per RANO, progression is a ≥ 25% increase in the sum of the products of perpendicular diameters of enhancing lesions, any new lesion or clinical deterioration. Patients alive who had not progressed as of the last follow-up had PFS censored at the last follow-up date. Median PFS was estimated using a Kaplan-Meier curve. (NCT00939991)
Timeframe: 3 years

Interventionmonths (Median)
Phase II6.7

[back to top]

Phase II: Radiographic Response.

The percentage of participants with a complete or partial response as determined by modified Response Assessment in Neuro-Oncology (RANO) criteria. A confirmation of response was not required. Complete Response (CR) was defined as complete disappearance on MR/CT of all enhancing tumor and mass effect, off all corticosteroids (or receiving only adrenal replacement doses) and accompanied by a stable or improving neurologic examination. Partial Response (PR) was defined as greater than or equal to 50% reduction in tumor size on MR/CT by bi-dimensional measurement, on a stable or decreasing dose of corticosteroids and accompanied by a stable or improving neurologic examination. Tumor assessments were done at baseline, the end of the first cycle (4 weeks), then the end of every second cycle (every 8 weeks) thereafter. (NCT00939991)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Phase II56

[back to top] [back to top]

Phase I: Determination of the Maximum Tolerated Dose (MTD)

The MTD is based upon dose-limiting toxicities (DLTs) experienced during Cycle 1 of treatment. The MTD is the dose level at which 0/6 or 1/6 patients experience DLT with at least two patients experiencing DLT at the next higher dose level. Using Common Terminology Criteria for Adverse Events (CTCAE) version 3.0, DLTs are defined as: Grade 4 neutropenia lasting greater than 5 days; Grade 3 thrombocytopenia; the occurrence of non-hematologic Grade 3 or greater drug-related adverse events excluding Grade ≥ 3 elevation in alkaline phosphatase, Grade ≥ 3 nausea or vomiting unless occurring despite the use of standard anti-emetics or Grade 3 diarrhea unless occurring despite standard anti-diarrheal therapy; > 14 day delay to re-treat due to failure to resolve drug-related toxicity to re-treatment criteria or pre-treatment baseline. (NCT00939991)
Timeframe: Cycle 1 (28 days)

Interventionmg (Number)
Phase I Dose Escalation400

[back to top]

Phase II: 6-month Progression-free Survival (PFS)

Phase II: Percentage of participants surviving six months from the start of study treatment without progression of disease. PFS was defined as the time from the date of study treatment initiation to the date of the first documented progression according to RANO criteria, or to death due to any cause. Per RANO, progression is a ≥ 25% increase in the sum of the products of perpendicular diameters of enhancing lesions, any new lesion or clinical deterioration. (NCT00939991)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Phase II53.8

[back to top]

Co-Primary: Overall Survival (OS)

Overall Survival was defined as the time from randomization to death due to any cause. (NCT00943826)
Timeframe: Randomization until OS Event (Until data cutoff= 28 February 2013 [up to 42.2 months])

InterventionMonths (Median)
Bevacizumab + RT + Temozolomide16.8
Placebo + RT + Temozolomide16.7

[back to top]

Co-Primary: Progression-free Survival (PFS) as Assessed by Investigator

PFS is defined as time from randomization to disease progression (PD) or death. PD was assessed using adapted Macdonald response criteria (modified World Health Organization [WHO] criteria) based on 3 components: radiological tumor assessments using Magnetic Resonance Imaging [MRI] scans,neurological assessment and changes in corticosteroid use. PD is assessed as greater than or equal to(>=) 25% increase in sum of products of the longest diameters of all index lesions (enhancing,measurable) compared with the smallest recorded sum (nadir); or unequivocal PD of existing non-index lesions (non-enhancing and enhancing,non-measurable); or unequivocal appearance of new lesions); or neurological worsening (if corticosteroid dose is stable or increased) compared to neurological evaluation at previous disease assessment with no need for a confirmatory scan. Participants without a PFS event were censored at last disease assessment. (NCT00943826)
Timeframe: Randomization until PFS Event [Until data cutoff= 31 March 2012 (up to 31.4 months)

InterventionMonths (Median)
Bevacizumab + RT + Temozolomide10.6
Placebo + RT + Temozolomide6.2

[back to top]

Kaplan-Meier (KM) Estimate of One Year Overall Survival

KM estimate of one year overall survival (probability to survive for at least 1 year) was reported. Corresponding 95% confidence interval (CI) was calculated using Greenwood's formula. (NCT00943826)
Timeframe: Randomization until Overall Survival Event (Until data cutoff= 28 February 2013 [up to 42.2 months])

Interventionprobability of being alive (Number)
Bevacizumab + RT + Temozolomide0.72
Placebo + RT + Temozolomide0.66

[back to top]

Kaplan-Meier (KM) Estimate of Two Year Overall Survival

KM estimate of two year overall survival was reported (probability to survive for at least 2 years). Corresponding 95% CI was calculated using Greenwood's formula. (NCT00943826)
Timeframe: Randomization until Overall Survival Event (Until data cutoff= 28 February 2013 [up to 42.2 months])

Interventionprobability of being alive (Number)
Bevacizumab + RT + Temozolomide0.34
Placebo + RT + Temozolomide0.30

[back to top]

PFS as Assessed by an Independent Review Facility

An Independent Review Facility reviewed the MRI scans used by investigator to evaluate radiological tumor response. PFS is defined as time from randomization to PD or death. PD was assessed using adapted Macdonald response (modified WHO) criteria based on 3 components: radiological tumor assessments using MRI scans, neurological assessment and changes in corticosteroid use. PD is assessed as >=25% increase in sum of products of the longest diameters of all index lesions (enhancing, measurable) compared with the smallest recorded sum (nadir); or unequivocal PD of existing non-index lesions (non-enhancing and enhancing, non-measurable); or unequivocal appearance of new lesions); or neurological worsening (if corticosteroid dose is stable or increased) compared to neurological evaluation at previous disease assessment with no need for a confirmatory scan. Participants without a PFS event were censored at last disease assessment. (NCT00943826)
Timeframe: Randomization until PFS Event (Until data cutoff= 31 March 2012 [up to 29.5 months])

InterventionMonths (Median)
Bevacizumab + RT +Temozolomide8.4
Placebo + RT + Temozolomide4.3

[back to top] [back to top]

Number of Participants With Non-Serious Adverse Events, Serious Adverse Events and Death

An adverse event (AE) was considered any unfavorable and unintended sign, symptom, or disease associated with the use of the study drug, whether or not considered related to the study drug. Preexisting conditions that worsened during the study were reported as AE.A serious adverse event (SAE) is any experience that suggests a significant hazard,contraindication, side effect or precaution that: results in death, is life-threatening, required in-patient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or is medically significant. Non-serious adverse events (Non-SAEs) included all AEs except SAEs (non-SAEs = all AEs - SAEs). Nine participants randomized to the Placebo+RT+Temozolomide arm incorrectly received at least 1 dose of bevacizumab and were added to the Bevacizumab+RT+Temozolomide arm for Safety. (NCT00943826)
Timeframe: Randomization until study completion (Until data cutoff= 09 Sep 2015 [up to 64 months])

,
InterventionParticipants (Number)
Non-SAEsSAEsDeath
Bevacizumab + RT + Temozolomide437179335
Placebo + RT + Temozolomide412115337

[back to top]

Best Overall Response Based on Response Evaluation Criteria in Solid Tumors (RECIST)

All measurable lesions up to a maximum of 5 lesions per organ and 10 lesions in total, representative of all involved organs were to be identified as target lesions. Data were to be collected at Screening, every 6 weeks and at 30 days after the final dose of robatumumab or the standard treatment assigned (whichever was last). The best overall response was to be the best response recorded from the start of the treatment until disease progression/recurrence. Complete Response (CR): Disappearance of all target lesions; Partial Response (PR): >30% decrease in the sum of the longest diameter (LD) of target lesions; Progressive Disease (PD): >20% increase in the sum of the LD of target lesions or the appearance of one or more new lesions; or Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT00960063)
Timeframe: Screening, every 6 weeks and at ~30 days after last dose of study drug (Up to ~10.3 months)

,
InterventionParticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)
Ifosfamide+Etoposide+Robatumumab0201
Temozolomide+Irinotecan+Robatumumab0100

[back to top]

Number of Participants Who Developed Anti-robatumumab Antibodies

The incidence of anti-robatumumab antibodies was to be assessed. Only participants who had a negative pre-treatment sample and a post-treatment sample were considered to be evaluable. If a participant had a single sample considered positive in the anti-robatumumab antibody assay (with the exception of pre-treatment positive participants), then they would be counted as positive in the immunogenicity assessment. (NCT00960063)
Timeframe: Prior to 1st and 8th doses of robatumumab, ~30 days after last dose of study drug, and 4 months after last dose of study drug (Up to ~13.3 months)

InterventionParticipants (Number)
Temozolomide+Irinotecan+Robatumumab0
Ifosfamide+Etoposide+Robatumumab0

[back to top]

Number of Participants With Dose Limiting Toxicities

Dose-limiting toxicity was defined by the following adverse events (AEs) that were considered possibly or probably related to either robatumumab or to its interaction with the chemotherapy regimen assigned: neutropenia (Grade 4 for >1 week that did not resolve prior to Day 1 of the next cycle; Grade 3-4 neutropenia with Grade ≥2 fever lasting 3 days; neutropenic infection; failure to recover to study entry/eligibility criteria laboratory requirement levels that resulted in a delay of 14 days between treatment cycles), thrombocytopenia (Grade 4 for >1 week that did not resolve prior to Day 1 of the next cycle; Grade 3-4 requiring a platelet transfusion on 2 separate days within a cycle) or all other AEs (Grade ≥3 [any duration] not ameliorable by supportive or symptomatic measures). The National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE version 4.0) was to be used to grade AEs. (NCT00960063)
Timeframe: Up to ~30 days after last dose of study drug (Up to ~10.3 months)

InterventionParticipants (Number)
Temozolomide+Irinotecan+Robatumumab1
Ifosfamide+Etoposide+Robatumumab3

[back to top]

Percentage of Participants Who Discontinued

Discontinuation was defined as the percentage of participants who permanently discontinued treatment in either treatment arm. Percentage of participant with individual discontinuation reason are reported. CNS: central nervous system; CTCAE: Common Terminology Criteria for Adverse Events . Other reason refers to any other reason than the specified ones. (NCT00967330)
Timeframe: From baseline until death (up to 4.5 years)

,
Interventionpercentage of participants (Number)
Persisting non-hematological toxicity CTCAE Grade3CNS hemorrhagic event (CTCAE Grade >1)Gastro-intestinal perforation (CTCAE Grade 1-4)OtherParticipant's wishProgressive diseaseProteinuria (nephrotic syndrome) (CTCAE Grade 4)RegularRepeated CTCAE Grade 4 hematological toxicityVenous thrombosis/embolismWound dehiscence requiring medical interventionWound dehiscence requiring surgical intervention
Bevacizumab + Irinotecan00.90.99.5674.10.91.700.90.94.3
Temozolomide1.9005.65.657.4027.80.9000

[back to top]

Overall Survival (OS)

Overall survival was defined as the time from randomization to death from any cause. OS was estimated using Kaplan-Meier method. (NCT00967330)
Timeframe: From baseline until death (up to 4.5 years)

InterventionMonths (Median)
Bevacizumab + Irinotecan16.64
Temozolomide17.30

[back to top]

Change From Baseline for Mini-Mental Status Examination (MMSE) at Baseline, Post-Baseline (up to Month 30)

The MMSE briefly measures orientation to time and place, immediate recall, short-term verbal memory, calculation, language and construct ability. Each area tested had a designated point value, the total score can range from 0 to 30, with a higher score indicating better function. (NCT00967330)
Timeframe: Baseline, Post-Baseline (up to Month 30)

,
Interventionunits on a scale (Least Squares Mean)
Orientation to time and placeImmediate recallRepetitions requiredCalculationsShort-term verbal memoryLanguage and construct abilityTotal Score
Bevacizumab + Irinotecan-0.01771-0.00264-0.05763-0.21530.2012-0.1254-0.2871
Temozolomide-0.2110-0.032190.08530-0.21200.1634-0.2057-0.5999

[back to top]

Percentage of Participants Achieving Progression-Free Survival (PFS) Without Disease Progression or Death at 6 Months

Progression-free survival was defined as the time from randomization to objective tumor progression or death from any cause, whichever came first. Progression was defined as 25 percent (%) increase in size of enhancing tumor or any new tumor on gadolinium contrast agent magnetic resonance imaging (Gd-MRI) scans, or neurologically worse, and steroids stable or increased. Percentage of participants achieving PFS without disease progression or death was reported. (NCT00967330)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Bevacizumab + Irinotecan79.31
Temozolomide42.59

[back to top]

Percentage of Participants Who Received Corticosteroid for Glioblastoma

Participants used corticosteroids for the glioblastoma condition. Corticosteroids included dexamethasone, methylprednisone, fortecortin, hydrocortisone, urbason, and prednisolone. (NCT00967330)
Timeframe: From baseline to Month 6

Interventionpercentage of participants (Number)
Bevacizumab + Irinotecan80.0
Temozolomide78.7

[back to top]

Number of Participants With A Best Overall Response (BOR) of Complete Response (CR) and With A BOR of CR or Partial Response (PR)

BOR was defined as the best response observed for a participant during assessment. Number of participants who had BOR as CR and number of participants who had BOR as CR or PR were reported. Complete response was defined as disappearance of all enhancing tumor on consecutive Gd-MRI scans at least 1 month apart, off steroids, and neurologically stable or improved. Partial response was defined as 50% reduction in size of enhancing tumor on consecutive Gd-MRI scans at least 1 month apart, steroids stable or reduced, and neurologically stable or improved. (NCT00967330)
Timeframe: 4 week after radiotherapy (RT) (up to Week 4), >4 Week after RT (up to Week 8) and Month 6

,
Interventionparticipants (Number)
CR at 4 weeks after RT (n=110,46)CR at >4 weeks after RT (n=95,35)CR at Month 6 (n=91,28)CR or PR at 4 Week after RT (n=110,46)CR or PR at >4 Week after RT (n=95,35)CR or PR at Month 6 (n=91,28)
Bevacizumab + Irinotecan1111342185
Temozolomide211633

[back to top]

Change From Baseline in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ - C30) at Baseline, Post-Baseline (up to Month 30)

The EORTC QLQ-C30 incorporates: 5 functional scales (physical, role, cognitive, emotional, and social); 9 symptom scales (fatigue, pain, nausea and vomiting, dyspnea, insomnia, appetite loss, constipation, diarrhea and financial difficulties); and a global health and quality-of-life scale. 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 were averaged and transformed to 0-100 scale; higher score for Global Qol/functional scales=better level of functioning or a higher score for symptom scale=greater degree of symptoms. The change in global health status was determined to be the difference in values at baseline and each specific visit. The term ''baseline'' refers to the time of randomization to the maintenance phase. (NCT00967330)
Timeframe: Baseline, Post-Baseline (up to Month 30)

,
Interventionunits on a scale (Least Squares Mean)
Physical FunctioningRole FunctioningEmotional FunctioningCognitive FunctioningSocial FunctioningGlobal health Status /QoL (ql)FatiqueNausea/VomittingPainDyspnoeaInsomniaAppetite lossConstipationDiarrhoeaFinancial Problems
Bevacizumab + Irinotecan-8.3513-0.76352.2774-2.0188-6.2324-3.11345.52288.955710.68763.7134-2.626613.74238.02306.02304.8435
Temozolomide-6.2511-2.23392.2547-3.8401-4.61980.38552.17794.75971.59260.5046-7.502610.96014.0855-0.14552.1140

[back to top]

Progression-Free Survival (PFS)

Progression-free survival was defined as the time from randomization to objective tumor progression or death from any cause, whichever came first. Progression was defined as 25% increase in size of enhancing tumor or any new tumor on Gd-MRI scans, or neurologically worse, and steroids stable or increased. PFS was estimated using Kaplan-Meier method. (NCT00967330)
Timeframe: From baseline to the end of the study (up to 4.5 years)

InterventionMonths (Median)
Bevacizumab + Irinotecan9.74
Temozolomide5.99

[back to top]

Percentage of Participants With Response on FLAIR Imaging

"FLAIR lesions were determined as stable, progressive or decreased. FLAIR lesions was determined as progressive only if they were not be attributed to causes apart from tumor infiltration (sequelae of radiation therapy, demyelination, ischemia, infection, seizures, or other treatment effects). Percentage of participants are based on ITT population.~Dis.=Discontinuation." (NCT00967330)
Timeframe: At screening, Baseline, Month 6 and Therapy Discontinuation (Up to 4.5 years)

,
Interventionpercentage of participants (Number)
Screening: Initial Flair Lesion (n=116,54)Screening:Stable Flair Lesion (n=116,54)Baseline:Decreased FLAIR Lesions (n=105,46)Baseline:Initial FLAIR Lesions (n=105,46)Baseline:Progressive FLAIR Lesions (n=105,46)Baseline: Stable FLAIR Lesions (n=105,46)Month 6:Progressive FLAIR Lesions (n=91,28)Month 6: Stable FLAIR Lesions (n=91,28)Therapy Dis.:Decreased FLAIR Lesions (n=55,31)Therapy Dis.:Progressiv FLAIR Lesions (n=55,31)Therapy Dis.:Stable FLAIR Lesions (n=55,31)
Bevacizumab + Irinotecan72.417.216.418.114.741.416.462.10.929.317.2
Temozolomide72.216.720.418.511.135.222.229.60.027.829.6

[back to top]

Time to Treatment Failure

(NCT00967330)
Timeframe: From baseline until end of study (up to 4.5 years)

Interventionyears (Median)
Bevacizumab + IrinotecanNA
TemozolomideNA

[back to top]

Change From Baseline for EORTC QLQ Brain Neoplasm 20 (BN20) at Baseline, Post-Baseline (up to Month 30)

EORTC QLQ-BN20 consisted of 20 items assessing visual disorders, motor dysfunction, communication deficit, various disease symptoms (e.g. headaches and seizures), treatment toxicities (e.g. hair loss) and future uncertainty. All of the 20 items are rated on a 4 point Likert scale from 1=not at all, 2=a little, 3=quite a bit and 4=very much, and were linearly transformed to a 0-100 scale, with higher scores indicating more severe symptoms. (NCT00967330)
Timeframe: Baseline, Post-Baseline (up to Month 30)

,
Interventionunits on a scale (Least Squares Mean)
Future uncertaintyVisual disorderMotor dysfunctionCommunication deficitHeadachesDrowsinesHair lossItchy skinWeakness of legsBladder control
Bevacizumab + Irinotecan-5.2779-2.08695.44164.74404.390511.720411.92355.48828.95861.5020
Temozolomide-8.5478-3.2026.54294.6431-3.93898.28057.33286.46907.92451.9710

[back to top]

Change From Baseline for Karnofsky Performance Status (KPS) Score at Baseline, Post-Baseline (up to Month 30)

KPS is an 11-level score (0, 10, 20, 30, 40, 50, 60, 70, 80, 90, and 100) which ranges between 0 (death) to 100 (complete healthy status); a higher score represents a higher ability to perform daily tasks. Deterioration in KPS was defined as decrease of 20 or more points in KPS score. (NCT00967330)
Timeframe: Baseline, Post-Baseline (up to Month 30)

Interventionunits on a scale (Least Squares Mean)
Bevacizumab + Irinotecan-3.3399
Temozolomide-5.4909

[back to top]

Concentration of Afatinib in Plasma at Steady State Pre-dose on Days 8, 15 and 29

Concentration of afatinib in plasma at steady state pre-dose (Cpre,ss) on days 8, 15 and 29. (NCT00977431)
Timeframe: Pharmacokinetic blood sample were taken at 5 minutes before drug on days 8, 15 and 29 and 1, 3 and 6 hours after drug administration on day 15

,,,,
Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Cpre, ss, 8Cpre, ss, 15Cpre, ss, 29
Afatinib 20 mg, Radiotherapy - Regimen U4.94.45.0
Afatinib 20 Milligram, Radiotherapy + Temozolomide - Regimen M4.45.65.3
Afatinib 30 mg, Radiotherapy + Temozolomide - Regimen M10.79.617.8
Afatinib 40 mg, Radiotherapy - Regimen U16.718.916.1
Afatinib 40 mg, Radiotherapy + Temozolomide - Regimen M15.716.817.4

[back to top]

Number of Patients With Investigator Defined Dose Limiting Toxicities (DLT) During the RT Phase

"Adverse event (AE) related to afatinib with any one criteria; Hematological: Common terminology criteria for adverse events (CTCAE) Grade 4 neutropenia (Absolute neutrophil count, including bands <500/cubic millimeter (mm³)) for >7 days, CTCAE Grade 3 or 4 neutropenia of any duration associated with fever >38.3 Celsius, CTCAE Grade 3 thrombocytopenia (platelet count <50000 - 25000/mm³), All other toxicities of CTCAE Grade ≥3 leading interruption of treatment > 14 days.~Non-hematological: CTCAE Grade ≥3 nausea or vomiting despite appropriate use of standard anti-emetics for ≥3 days, CTCAE Grade ≥3 diarrhea despite appropriate use of standard anti-diarrheal therapy for ≥3 days, CTCAE Grade ≥3 rash despite standard medical management and lasting >7 days, CTCAE Grade ≥2 cardiac left ventricular function, CTCAE Grade ≥2 worsening of renal function as measured by serum creatinine, newly developed proteinuria or decrease in glomerular filtration rate, All other toxicities of CTCAE Grade ≥3. (NCT00977431)
Timeframe: 6 weeks

InterventionParticipants (Number)
Afatinib 20 Milligram, Radiotherapy + Temozolomide - Regimen M1
Afatinib 30 mg, Radiotherapy + Temozolomide - Regimen M0
Afatinib 40 mg, Radiotherapy + Temozolomide - Regimen M2
Afatinib 20 mg, Radiotherapy - Regimen U0
Afatinib 40 mg, Radiotherapy - Regimen U1

[back to top]

The Objective Tumour Response According to the Macdonald Criteria

Objective response was defined as a best overall response of complete response (CR) or partial response (PR). The best overall response was the best overall response to trial medication according to the Macdonald criteria recorded since the first administration of trial medication and until the earliest of disease progression, death, or start of further anti-cancer treatment. Tumour response was assessed based on local radiological image evaluation by the investigators according to the Macdonald criteria: Complete Response (CR): Disappearance of all enhancing tumour on consecutive Magnetic resonance imaging (MRI) scans at least 28 days apart, off steroids, and neurologically stable or improved. Partial Response (PR): At least 50% reduction in size of enhancing tumour on consecutive MRI scans at least 28 days apart, steroids stable or reduced, and neurologically stable or improved. (NCT00977431)
Timeframe: From the first administration of trial medication until 4 weeks after the last administration of trial medication, up to approximately 338 weeks

,,,,
InterventionParticipants (Number)
NoYesMissing
Afatinib 20 mg, Radiotherapy - Regimen U300
Afatinib 20 Milligram, Radiotherapy + Temozolomide - Regimen M520
Afatinib 30 mg, Radiotherapy + Temozolomide - Regimen M330
Afatinib 40 mg, Radiotherapy - Regimen U1012
Afatinib 40 mg, Radiotherapy + Temozolomide - Regimen M502

[back to top]

Maximum Tolerated Dose (MTD) of Afatinib

The MTD was defined as the highest afatinib dose level, at which no more than 1 out of 6 patients experienced drug-related DLT, i.e. the highest afatinib dose with a DLT incidence ≤17%. A separate MTD was determined for afatinib and RT (Regimen U), and for afatinib, TMZ, and RT (Regimen M). (NCT00977431)
Timeframe: 6 weeks

InterventionMilligram (mg) (Number)
Total - Regimen M30
Total - Regimen U40

[back to top]

Incidence and Intensity of Adverse Events (AE) According to Common Terminology Criteria of Adverse Events (CTCAE v.3.0)

Incidence and intensity of adverse events (AE) according to Common Terminology Criteria of Adverse Events (CTCAE v.3.0). The CTCAE grades are: 1 (mild AE), 2 (moderate AE), 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related to AE). (NCT00977431)
Timeframe: From the first administration of trial medication until 4 weeks after the last administration of trial medication, up to approximately 338 weeks

,,,,
InterventionParticipants (Number)
Grade 1Grade 2Grade 3Grade 4Grade 5
Afatinib 20 mg, Radiotherapy - Regimen U01200
Afatinib 20 Milligram, Radiotherapy + Temozolomide - Regimen M00430
Afatinib 30 mg, Radiotherapy + Temozolomide - Regimen M02400
Afatinib 40 mg, Radiotherapy - Regimen U12523
Afatinib 40 mg, Radiotherapy + Temozolomide - Regimen M11410

[back to top]

Response Rate

The percentage of participants with a complete or partial response as determined by a modification of the Response Assessment in Neuro-Oncology (RANO) criteria. Complete Response (CR) was defined as complete disappearance on MR/CT of all enhancing tumor and mass effect, off all corticosteroids (or receiving only adrenal replacement doses) and accompanied by a stable or improving neurologic examination. Partial Response (PR) was defined as greater than or equal to 50% reduction in tumor size on MR/CT by bi-dimensional measurement, on a stable or decreasing dose of corticosteroids and accompanied by a stable or improving neurologic examination. Per the criteria, confirmation of response was required. Response rate = CR+PR. (NCT00979017)
Timeframe: 4 months

Interventionpercentage of participants (Number)
Avastin in Combination With Temozolomide and Irinotecan22

[back to top]

Incidence and Severity of Central Nervous System (CNS) Hemorrhage and Systemic Hemorrhage

Incidence and severity of CNS hemorrhage and systemic hemorrhage- The adverse events for this study were collected using Common Terminology Criteria for Adverse Events (CTCAE) version 3.0, and have been converted to CTCAE version 4.0 for entry into ClinicalTrials.gov. (NCT00979017)
Timeframe: 4 months

Interventionparticipants (Number)
CNS hemorrhage (grade 3)Systemic hemorrhage (all grade 3)
Avastin in Combination With Temozolomide and Irinotecan13

[back to top]

Median Overall Survival (OS)

Time in months from the start of study treatment to the date of death due to any cause. Patients alive as of the last follow-up had OS censored at the last follow-up date. Median OS was estimated using a Kaplan-Meier curve. (NCT00979017)
Timeframe: 36 months

Interventionmonths (Median)
Avastin in Combination With Temozolomide and Irinotecan12

[back to top]

Median Progression-free Survival (PFS)

Time in months from the start of study treatment to the date of first progression according to RANO criteria, or to death due to any cause. Per RANO, progression is a ≥ 25% increase in the sum of the products of perpendicular diameters of enhancing lesions, worsening T2/FLAIR, any new lesion, or clinical deterioration. Patients alive who had not progressed as of the last follow-up had PFS censored at the last follow-up date. Median PFS was estimated using a Kaplan-Meier curve. (NCT00979017)
Timeframe: 36 months

Interventionmonths (Median)
Avastin in Combination With Temozolomide and Irinotecan8.6

[back to top]

Incidence of Grade ≥ 4 Hematologic and ≥ Grade 3 Non-hematologic Toxicities

Incidence of treatment-related, grade ≥ 4 hematologic and ≥ grade 3 non-hematologic toxicities- The adverse events for this study were collected using Common Terminology Criteria for Adverse Events (CTCAE) version 3.0, and have been converted to CTCAE version 4.0 for entry into ClinicalTrials.gov. (NCT00979017)
Timeframe: 4 months

Interventionparticipants (Number)
Grade > or = to 4 hematologic toxicityGrade > or = to 3 non-hematologic toxicity
Avastin in Combination With Temozolomide and Irinotecan717

[back to top]

Number of Patients Surviving Without Disease Progression After 6 Months

"Patients will be monitored from date of first treatment to the date of first observation of progressive disease, non-reversible neurologic progression or increasing steroid requirements, death due to any cause, or early discontinuation of treatment. Progression-free survival will be defined as the absence of any of the above after 6 months.~Progression (defined by MacDonald Criteria) is a 25% increase in the sum of products of all measurable lesions over smallest sum observed compared to baseline, OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to the cancer)." (NCT00990652)
Timeframe: From date of first treatment until disease progression, death, or early discontinuation of treatment (up to 24 months)

Interventionparticipants (Number)
Bortezomib + Temozolomide0

[back to top]

Number of Participants Achieving a Response to Treatment (Either Complete or Partial Response) as Defined by MacDonald Criteria

"This measure was assessed only in patients who had residual tumor post-operatively. Per MacDonald Criteria:~Complete Response requires complete disappearance of all measurable & evaluable disease, no new lesions, no evidence of non-evaluable disease, and only minimal or no use of steroids.~Partial Response is defined as >= 50% decrease compared to baseline in the sum of products of perpendicular diameters of all measurable lesions, no progression of evaluable disease, and no new lesions. Responders must be on the same or decreasing doses of steroid.~Response was assessed by imaging (MRI or CT with contrast)." (NCT00990652)
Timeframe: Day of treatment post-surgery and then approximately every 8 weeks thereafter until off treatment

Interventionparticipants (Number)
Bortezomib + Temozolomide0

[back to top]

Number of Grade 1, 2, 3, 4, and 5 Adverse Events Observed During Study Treatment (Defined by CTCAE v 3.0)

"Adverse events (AEs) were graded according to the National Cancer Institute's Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0. In general, AEs are graded according to the following:~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" (NCT00990652)
Timeframe: Days 1, 4, 8 pre-surgery, and then at the start of every cycle (approximately every 4 weeks) post-surgery while on treatment

Interventionadverse events (Number)
Grade 1Grade 2Grade 3Grade 4Grade 5
Bortezomib + Temozolomide80301110

[back to top]

Overall Survival Rate at 6 Months

The rate of overall survival at 6 months (regardless of disease progression) was calculated. (NCT00990652)
Timeframe: After 6 months on study

Interventionpercentage of participants (Number)
Bortezomib + Temozolomide69

[back to top]

Overall Survival (in Days)

(NCT00990652)
Timeframe: Days 1, 4, 8 pre-surgery, once per cycle (every 4 weeks) while on treatment post-surgery, and then every 3 months up to 2 years during follow-up

Interventiondays (Median)
Bortezomib + Temozolomide248

[back to top]

Dose Limiting Toxicity

Number of participants with dose limiting toxicity events (NCT00993044)
Timeframe: 2 years

Interventionparticipants (Number)
Single Arm2

[back to top]

Overall Survival

Estimate the overall survival in subjects with newly-diagnosed glioblastoma (GBM) treated with bortezomib/temozolomide/radiation followed by bortezomib/temozolomide for 24 cycles until progression is detected or for up to 24 cycles (~2 years). (NCT00998010)
Timeframe: 2 years

Interventionmonths (Mean)
Experimental19.1

[back to top]

Survival at 1 Year

Overall Survival at 12 months from completion of radiation treatment (NCT00998010)
Timeframe: 1 year

Interventionpercentage of participants (Median)
Experimental87.5

[back to top]

Toxicity Assessed According to the NCI Common Terminology Criteria for Adverse Events (CTCAE) Version 3.

(NCT00998010)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Radiotherapy phase72241098Post Radiotherapy phase72241098Follow-up phase72241098
Grade 4 Toxicity< Grade 3 ToxicityGrade 3 toxicity
Experimental6
Experimental1
Experimental17
Experimental7
Experimental0
Experimental24

[back to top]

Tumor Progression as Assessed by Magnetic Resonance Imaging (MRI) and Neurologic Exam

MRI will be done 2 weeks after completion of radiation and then every 8 weeks. Neurologic exam to be performed every 2 weeks during radiation therapy, then every every 4 weeks after radiation is completed. (NCT00998010)
Timeframe: at 6, 12, 18 and 24 months from completion of radiation treatment.

Interventionpercentage of participants (Median)
Progression Free Survival rate at 6 monthsProgression Free Survival rate at 12 monthsProgression Free Survival rate at 18 monthsProgression Free Survival rate at 24 moths
Experimental54.229.225.025

[back to top]

Time to Progression

"Median time to tumor progression. Because many newly-diagnosed patients are likely not to have evaluable disease due to gross total resections. A 7-point scale was used to guide Magnetic resonance imaging (MRI) assessment to determine progression in this study. A -2 or -3 assessment will be taken as progression.~The 7-point scale is listed below. complete resolution of tumor: 3 tumor definitely smaller: 2 tumor probably smaller: 1 tumor unchanged: 0 tumor probably worse: -1 tumor definitely worse: -2 new lesion: -3" (NCT00998010)
Timeframe: From the completion of radiation treatment to tumor progression

Interventionmonths (Median)
Experimental6.2

[back to top]

One and Two Year Overall Survival

Time in months from the start of study treatment to date of death due to any cause. Patients alive as of the last follow-up had OS censored at the last follow-up date. (NCT01004874)
Timeframe: One year and two years

Interventionpercentage of participants (Number)
One year Overall SurvivalTwo year Overall Survival
Bevacizumab, XRT, Temozolomide, Topotecan73.835.6

[back to top]

Median Overall Survival

OS was defined as time in months from the start of study treatment to date of death due to any cause. Patients alive as of the last follow-up had OS censored at the last follow-up date. Median OS was estimated using a Kaplan-Meier curve. (NCT01004874)
Timeframe: 27 months

Interventionmonths (Median)
Bevacizumab, XRT, Temozolomide, Topotecan17.2

[back to top]

6-month Progression-free Survival

Percentage of participants surviving six months from the start of study treatment without progression of disease. PFS was defined as the time from the date of study treatment initiation to the date of the first documented progression according to the Macdonald criteria, or to death due to any cause. (NCT01004874)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Bevacizumab, XRT, Temozolomide, Topotecan88.8

[back to top]

Median Progression-free Survival

PFS was defined as time in months from the start of study treatment to the date of first progression according to Macdonald criteria, or to death due to any cause. Patients alive who had not progressed as of the last follow-up had PFS censored at the last follow-up date. Median PFS was estimated using a Kaplan-Meier curve. (NCT01004874)
Timeframe: 27 months

Interventionmonths (Median)
Bevacizumab, XRT, Temozolomide, Topotecan11.1

[back to top]

Number of Patients Experiencing a Greater Than or Equal to Grade 4 Hematologic or a Greater Than or Equal to Grade 3 Non-hematologic Toxicity

Number of times a grade ≥4 hematologic or grade ≥3 non-hematologic toxicity was experienced (NCT01004874)
Timeframe: 27 months

Interventionparticipants (Number)
Greater than equal Gr.4 hematologic toxicitesGreater than equal Gr.3 non-hematologic toxicites
Bevacizumab, XRT, Temozolomide, Topotecan2317

[back to top]

Number of Patients Experiencing a Central Nervous System (CNS) Hemorrhage or a Systemic Hemorrhage

Number of times a CNS hemorrhage or systemic hemorrhage was experienced (NCT01004874)
Timeframe: 27 months

Interventionparticipants (Number)
Grade 2 central nervous system (CNS) hemorrhageGrade 3 CNS hemorrhageGrade 4 CNS hemorrhage
Bevacizumab, XRT, Temozolomide, Topotecan102

[back to top]

Objective Response Rate (ORR)

Tumor response is evaluated according to Response Evaluation Criteria in Solid Tumors (RECIST) (version 1.0). Target lesions are assessed by physical exam and/or computerized tomography (CT): Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD), neither sufficient decrease in the sum of the longest diameter of target lesions to qualify for PR nor sufficient increase in the sum of the longest diameter of target lesions to qualify for Progressive Disease; Progressive Disease (PD), 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. The Objective Response Rate (ORR) is the sum of the percentages of patients achieving CR or PR. (NCT01009515)
Timeframe: 6 months

Interventionparticipants (Number)
Complete response (CR)Partial response (PR)Stable diseaseProgressive disease (PD)ORR (CR + PR)
Chemotherapy Combination130124

[back to top]

Overall Survival

The time from treatment initiation to death by any cause. (NCT01009515)
Timeframe: 2 years

Interventionweeks (Median)
Chemotherapy Combination47

[back to top]

Time to Progression

The time from treatment initiation to disease progression or death by any cause. Progression is evaluated according to Response Evaluation Criteria in Solid Tumors (RECIST) (version 1.0). Target lesions are assessed by physical exam or computerized tomography (CT): Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD), neither sufficient decrease in the sum of the longest diameter of target lesions to qualify for PR nor sufficient increase in the sum of the longest diameter of target lesions to qualify for Progressive Disease; Progressive Disease (PD), 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. (NCT01009515)
Timeframe: 2 years

Interventionweeks (Median)
Chemotherapy Combination31

[back to top]

Safety Profile

All toxicities encountered during the study by patients who receive at least one on-study treatment will be graded according to the NCI CTCAE (Version 3.0). The number of patients experiencing adverse events will be reported according to grade. (NCT01009515)
Timeframe: Up to 30 days after last on-study treatment, for up to 2 years

Interventionparticipants (Number)
Fatigue (Grade 1-2)Fatigue (Grade 3-4)Fever (Grade 1-2)Fever (Grade 3-4)Weight loss (Grade 1-2)Weight loss (Grade 3-4)Anorexia (Grade 1-2)Anorexia (Grade 3-4)Diarrhea (Grade 1-2)Diarrhea (Grade 3-4)Aspartate aminotransferase increased (Grade 1-2)Aspartate aminotransferase increased (Grade 3-4)Vomiting (Grade 1-2)Vomiting (Grade 3-4)Gastrointestinal Bleed (Grade 1-2)Gastrointestinal Bleed (Grade 3-4)Nausea (Grade 1-2)Nausea (Grade 3-4)Alanine aminotransferase increased (Grade 1-2)Alanine aminotransferase increased (Grade 3-4)Hyperbilirubinemia (Grade 1-2)Hyperbilirubinemia (Grade 3-4)Neutropenia (Grade 1-2)Neutropenia (Grade 3-4)Anemia (Grade 1-2)Anemia (Grade 3-4)Lymphopenia (Grade 1-2)Lymphopenia (Grade 3-4)Leukopenia (Grade 1-2)Leukopenia (Grade 3-4)Thrombocytopenia (Grade 1-2)Thrombocytopenia (Grade 3-4)Hyperglycemia (Grade 1-2)Hyperglycemia (Grade 3-4)Hypophosphatemia (Grade 1-2)Hypophosphatemia (Grade 3-4)Hypokalemia (Grade 1-2)Hypokalemia (Grade 3-4)Hyponatremia (Grade 1-2)Hyponatremia (Grade 3-4)Chest wall pain (Grade 1-2)Chest wall pain (Grade 3-4)Bone pain (Grade 1-2)Bone pain (Grade 3-4)Muscle weakness, lower limb (Grade 1-2)Muscle weakness, lower limb (Grade 3-4)Joint pain (Grade 1-2)Joint pain (Grade 3-4)Drowsiness (Grade 1-2)Drowsiness (Grade 3-4)Peripheral Sensory Neuropathy (Grade 1-2)Peripheral Sensory Neutropenia (Grade 3-4)Pericardial effusion (Grade 1-2)Pericardial effusion (Grade 3-4)Edema limbs (Grade 1-2)Edema limbs (Grade 3-4)Alopecia (Grade 1-2)Alopecia (Grade 3-4)Rash (Grade 1-2)Rash (Grade 3-4)Dry mouth (Grade 1-2)Dry mouth (Grade 3-4)
Chemotherapy Combination910101105121210112020103240417030310130301120201001300110804030

[back to top]

Median Overall Survival (OS) Based on the MGMT Promoter Methylation Status

IDH1 and MGMT methylation are important independent prognostic biomarkers that have to be included in a Cox regression model. In addition, subgroup analysis could reveal differential sensitivities to the treatment arm. The MGMT promoter methylation status, IDH1 mutation status were not available for all of the control samples. Therefore, only the samples with both info available were included in the analysis. Baseline analysis results are included in the Baseline Characteristics module. (NCT01013285)
Timeframe: 2 years

,
Interventionmonths (Median)
Median Overall survival of MGMT MethylatedMedian Overall survival of MGMT Unmethylated
Historical Control UCLA/KPLA26.718.2
Treatment Arm24.715.9

[back to top]

Overall Survival

(NCT01013285)
Timeframe: 2 years

InterventionMonths (Median)
Treatment Arm19.6
Historical Control UCLA/KPLA21.1

[back to top]

Radiographic Response (When Evaluable)

"Radiation therapy (RT) median from diagnosis to RT. Patients will have brain MRI evaluation of response and progression every 8 weeks starting from the day 56 scan obtained 2 weeks after completion of radiation and daily temozolomide and assessment will be conducted on a 7-point scale. This scale is expected to be more useful in this study because many newly-diagnosed patients are likely not to have evaluable disease due to gross total resections. Determination of whether progression occurs based on the day 56 scan will take into account the untreated window between baseline MRI and day of 1 of study.~7 Point Likert Scale: 3 to -3, 3 means complete resolution of tumor, and -3 means new lesion. A -2 or -3 assessment will be taken as tumor progression.~complete resolution of tumor: 3~tumor resolved 3 tumor definitely smaller: 2 tumor probably smaller: 1 tumor unchanged: 0 tumor probably worse: -1 tumor definitely worse: -2 New Lesion: -3" (NCT01013285)
Timeframe: 2 years

InterventionWeeks (Median)
Treatment Arm4.14
Historical Control UCLA/KPLA5

[back to top]

Time to Disease Progression

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. (NCT01013285)
Timeframe: 2 years

InterventionMonths (Median)
Treatment Arm13.6
Historical Control UCLA/KPLA7.6

[back to top]

Progression-free Survival at 6 Months

participants who were alive and disease progression free at 6 months (NCT01013285)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Treatment Arm88.4
Historical Control UCLA/KPLA58.3

[back to top]

Objective Response Rates. Assessed With Cranial MRI

"Objective Response (OR) encompassed the number of participants with Complete Response (CR) and the number of participants with Partial Response (PR). CR is the disappearance of all brain metastases, assessed between two or more cranial MRI. PR is at least a 30% decrease in the sum of the longest diameter of the brain metastases, taking as reference the baseline sum longest diameter, assessed between two or more cranial MRI.~Objective Response Rate (ORR) is the ratio between the number of participants with objective response and the total number of participants." (NCT01015534)
Timeframe: 90 days

InterventionPercentage of participants with OR (Number)
Whole Brain Irradiation and Temozolomide78.6
Whole Brain Irradiation48.1

[back to top]

Overall Survival

Overall survival:Time in months measured from treatment initiation until the date of death or the date of last follow-up. (NCT01015534)
Timeframe: 1 year

InterventionMonths of Overall Survival (Median)
Whole Brain Irradiation and Temozolomide8
Whole Brain Irradiation8.1

[back to top]

Survival Free of Brain Metastases Progression (PFS of BM)

Progression free survival of brain metastases is the survival of participants without progressive brain metastases or without neurological symptoms. The progressive brain metastases (PBM) were evaluated with cranial MRI. The PBM were defined as an increase of at least 20% in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since the treatment started or the appearance of one or more new metastases. (NCT01015534)
Timeframe: at 90 days

InterventionPercentage of Participants (Number)
Whole Brain Irradiation and Temozolomide88.7
Whole Brain Irradiation83.7

[back to top] [back to top]

Phase 1: Maximum Tolerated Dose (MTD)

Dose limiting toxicity (DLT) = any of the following events within 1st 8 weeks of treatment attributable to study drugs: Any grade (gr) 3/4 thrombocytopenia, gr 4 anemia, gr 3 neutropenia with fever (>100.4). gr 4 neutropenia lasting > 7 days; Any non-hematologic (NH) gr 3+ toxicity (TOX), excluding alopecia, despite maximal medical therapy (MLT); NH TOX such as rash, nausea, vomiting, diarrhea, mucositis, hypophosphatemia, and hypertension will only be considered DLTs if they remain gr 3+ despite MLT; 2nd occurrence of thromboembolism; Failure to recover from TOX (<= gr 1) to be eligible for re-treatment with study drugs <= 14 days of last dose of either drug; Any episode of non-infectious radiologically observed pneumonitis gr 2-4 any duration. Dose level will be considered acceptable if <= 1 of the 1st 6 eligible patients experiences a DLT. If current level is considered acceptable, dose escalation occurs. Otherwise preceding acceptable dose level will be declared the MTD. (NCT01026493)
Timeframe: Start of treatment to 8 weeks.

Interventionparticipants (Number)
Phase I: Dose Level 11
Phase I: Dose Level 2a0
Phase I: Dose Level 2b1
Phase I: Dose Level 31

[back to top]

Phase II: 6-month Progression-free Survival (PFS) Rate for Patients With Measurable Disease After Surgery

For patients with measureable disease after surgery: Progression defined as ≥ 25% increase in size of enhancing tumor or any new tumor; or neurologically worse, and steroids stable/increased. Bevacizumab (BEV)-naïve group: p0= 15% as estimate of 6-mo. PFS [null hypothesis (NH)], p1= 30%, with a 15% absolute increase [alternative hypothesis (AH)]. Error rates of 10% alpha and 10% beta. If <= 11 patients experience 6-month PFS of the first 53 analyzable patients, then do not reject the null hypothesis that the 6-month PFS rate of experimental arm is less than 15%; BEV-failure group: p0 = 2% as a conservative estimate of 6-month PFS [NH], p1 = 15%, with a 13% absolute increase [AH]. Using first 26 analyzable subjects for each experimental arm, there is >= 90% power to detect >= 15% increase at a significance level of 0.10, using a 1-sided binomial test. If >= 2 patients (8%) are progression free at 6 mo., then claim this regimen to be promising in the patient group. (NCT01026493)
Timeframe: Randomization to 6 months.

Interventionparticipants (Number)
Phase II: Arm 1/BEV-NAIVE9
Phase II: Arm 2/BEV-NAIVE9
Phase II: Arm 1/BEV-FAILURE1
Phase II: Arm 2/BEV-FAILURE1

[back to top]

Phase II: Objective Response (Partial and Complete Response) Rate for Patients With Measurable Disease After Surgery

Response and progression will be evaluated using standard criteria for patients with malignant gliomas (Macdonald 1990). Partial response and complete response are centrally reviewed. (NCT01026493)
Timeframe: Analysis occurs after all patients have been on study for at 6 months. (Patients are followed from randomization to death or study termination whichever occurs first.)

Interventionpercentage of participants (Number)
Phase II: Arm 1/BEV-NAIVE0
Phase II: Arm 2/BEV-NAIVE3.8
Phase II: Arm 1/BEV-FAILURE5.3
Phase II: Arm 2/BEV-FAILURE0

[back to top]

Phase II: Overall Survival (OS)

Survival time is defined as time from randomization to date of death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. This analysis was planned to occur when all patients had been potentially followed for at least 6 months. (NCT01026493)
Timeframe: Analysis occurs after all patients have been on study for at 6 months. (Patients are followed from randomization to death or study termination whichever occurs first.)

Interventionmonths (Median)
Phase II: Arm 1/BEV-NAIVE10.3
Phase II: Arm 2/BEV-NAIVE10.7
Phase II: Arm 1/BEV-FAILURE4.7
Phase II: Arm 2/BEV-FAILURE4.7

[back to top]

Progression Free Survival

The progression free survival of patients receiving study drug will be recorded. 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. (NCT01044966)
Timeframe: At 6 months

InterventionParticipants (Count of Participants)
ITV DepoCyt + Temozolomide2

[back to top]

Proportion of Patients With Recurrent GBM Treated With ITV DepoCyt in Combination With Oral Temozolomide Who Are Progression-free at 16 Weeks.

Eligibility of patients with GBM that are able to receive study drug to estimate the proportion of patients with recurrent GBM treated with ITV DepoCyt in combination with oral temozolomide who are progression-free at 16 weeks. 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. (NCT01044966)
Timeframe: 16 weeks

InterventionParticipants (Count of Participants)
ITV DepoCyt + Temozolomide12

[back to top]

Response Rate of Drug Treatment

Those responding to study drug will be recorded. Response will be defined as stable neurological examination in conjunction with the absence of progression as defined above. (NCT01044966)
Timeframe: 52 weeks

InterventionParticipants (Count of Participants)
ITV DepoCyt + Temozolomide2

[back to top]

Quality of Life Outcomes Measurement

"Participants recorded are those who had an improvement in QOL score, QOL outcomes will be assessed and recorded using the EORTC QLQ C30 version 3.~This 30 question questionnaire will be used to asses our patient overall feeling of well-being during the trial. Questions to assess quality of life are measured from 1-4 with the following graded values:~Not at all~A little~Quite a bit~Very much Lower total scores are consistent with better quality of life and changes of greater or equal to 10 points are considered a significant change in quality of life." (NCT01044966)
Timeframe: 52 weeks

InterventionParticipants (Count of Participants)
ITV DepoCyt + Temozolomide2

[back to top] [back to top]

Percent of Patients With Disease Control

Disease control rate defined as stable disease, partial response, or complete response according to the Response Evaluation Criteria in Solid Tumors (RECIST). (NCT01051596)
Timeframe: 2 months

InterventionParticipants (Count of Participants)
ABT-888 and Temozolomide18

[back to top]

Percent of Patients With an Objective Response

Objective response rate defined as partial response or complete response according to RECIST criteria (NCT01051596)
Timeframe: 2 months

InterventionParticipants (Count of Participants)
ABT-888 and Temozolomide2

[back to top]

Median Progression-free Survival Time

Progression-free survival defines as the time in days from study study entry until progression or death (NCT01051596)
Timeframe: 1 year

InterventionMonths (Median)
ABT-888 and Temozolomide1.8

[back to top]

Overall Survival

Overall survival defined as the time in days from study entry until death (NCT01051596)
Timeframe: 1 year

InterventionMonths (Median)
ABT-888 and Temozolomide6.6

[back to top]

Feasibility of the Addition of Cixutumumab to Chemotherapy Determined by Patient Enrollment

Proportion of no Grade 3+ cardiac toxicity. (NCT01055314)
Timeframe: From start to week 26 of therapy

InterventionProportion of Participants (Number)
IMC-A120.9390

[back to top]

Event-Free Survival

Probability of no relapse, secondary malignancy, or death after 3 years in the study. (NCT01055314)
Timeframe: 3 years

InterventionProbability (Number)
IMC-A120.1627
Temozolomide0.1919

[back to top]

Response Rate (CR + PR)

Proportion of patients with complete or partial response. Complete Response (CR): Complete disappearance of the tumor confirmed at > 4 weeks. Partial Response (PR): At least 64% decrease in volume compared to the measurement obtained at study enrollment; Overall Response (OR) = CR + PR. (NCT01055314)
Timeframe: From the start of treatment until a maximum of 2 cycles (21 days per cycle) of treatment in the absence of disease progression or unacceptable toxicities

InterventionProportion of Participants (Number)
IMC-A120.7667
Temozolomide0.7846

[back to top]

Incidence of Adverse Events Assessed by Common Terminology Criteria for Adverse Events Version 4.0

Number of patients with grade 3+ adverse events (AE) during therapy. (Grade 3+) = (Grade 3 + Grade 4 + Grade 5) . Grade 3: Severe and undesirable AE; Grade 4: Life threatening or disabling AE; Grade 5: Death related to AE. (NCT01055314)
Timeframe: Up to 54 weeks

InterventionParticipants (Number)
IMC-A1289
Temozolomide61

[back to top]

Feasibility of the Addition of Temozolomide to Chemotherapy Determined by Patient Enrollment

Proportion of no Grade 4+ non-hematologic toxicity. (NCT01055314)
Timeframe: From start to week 26 of therapy

InterventionProportion of Participants (Number)
Temozolomide0.7097

[back to top]

Phase II: Overall Survival (OS)

Overall survival time is defined as time from/randomization to the date of death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive are censored at the date of last contact. (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

Interventionmonths (Median)
Ph II: RT + TMZ21.2
Ph II: RT + TMZ + RAD00116.5

[back to top]

Phase I: Distribution of Worst Adverse Event Grade

"AE reporting in Phase I was split up by treatment timing: concurrent treatment (RT, TMZ, RAD001); post-RT treatment (TMZ, RAD001) along with all AE's reported in follow-up.~The worst/highest grade of any adverse event reported in each time period was determined for each patient. The percentage of patients in each grade level is reported. Adverse events are graded using CTCAE v4.0. Grade refers to the severity of the AE. The CTCAE v4.0 assigns Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grade 1 Mild AE, Grade 2 Moderate AE, Grade 3 Severe AE, Grade 4 Life-threatening or disabling AE, Grade 5 Death related to AE." (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

,,
Interventionpercentage of participants (Number)
Concurrent treatment: Grade 1Concurrent treatment: Grade 2Concurrent treatment: Grade 3Concurrent treatment: Grade 4Concurrent treatment: Grade 5Post-RT treatment: Grade 1Post-RT treatment: Grade 2Post-RT treatment: Grade 3Post-RT treatment: Grade 4Post-RT treatment: Grade 5
Ph I: RT + TMZ + RAD001 10 mg/Day12.50.087.50.00.00.0100.00.00.00.0
Ph I: RT + TMZ + RAD001 2.5 mg/Day0.00.050.050.00.00.00.071.414.314.3
Ph I: RT + TMZ + RAD001 5 mg/Day11.133.344.411.10.00.042.928.60.00.0

[back to top]

Phase II: Distribution of Worst Adverse Event Grade

The worst/highest grade of any adverse event reported was determined for each patient. The percentage of patients in each grade level is reported. Adverse events are graded using CTCAE v4.0. Grade refers to the severity of the AE. The CTCAE v4.0 assigns Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grade 1 Mild AE, Grade 2 Moderate AE, Grade 3 Severe AE, Grade 4 Life-threatening or disabling AE, Grade 5 Death related to AE. (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

,
Interventionpercentage of patients (Number)
Grade 1Grade 2Grade 3Grade 4Grade 5
Ph II: RT + TMZ00332
Ph II: RT + TMZ + RAD00103501

[back to top]

Phase I: Number of Patients With Dose-limiting Toxicity (DLT)

DLT is defined as any of the following events occurring during the first 8 weeks of treatment with RAD001 and temozolomide and attributable to the study drugs: any grade 3 or 4 thrombocytopenia, grade 4 anemia, or grade 4 neutropenia lasting more than 7 days; any non-hematologic grade 3 or greater adverse event (AE), excluding alopecia, despite maximal medical therapy; any grade 4 radiation-induced skin changes; failure to recover from adverse events to be eligible for re-treatment with RAD001 and temozolomide within 14 days of the last dose of either drug; or any episode of non-infectious pneumonitis grade 2, 3, or 4 of any duration. Adverse events are graded using CTCAE v4.0. Grade refers to the severity of the AE. The CTCAE v4.0 assigns Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline: Grade 1 Mild AE, Grade 2 Moderate AE, Grade 3 Severe AE, Grade 4 Life-threatening or disabling AE, Grade 5 Death related to AE (NCT01062399)
Timeframe: From start of treatment to eight weeks.

InterventionParticipants (Count of Participants)
Ph I: RT + TMZ + RAD001 2.5 mg/Day2
Ph I: RT + TMZ + RAD001 5 mg/Day2
Ph I: RT + TMZ + RAD001 10 mg/Day2

[back to top]

Phase II: Progression-free Survival (PFS)

Using the Response Assessment in Neuro- Oncology (RANO) criteria, the progression is defined by any of the following: > 25% increase in sum of the products of perpendicular diameters of enhancing lesions compared to the smallest tumor measurement obtained either at baseline (if no decrease) or best response, on stable or increasing doses of corticosteroids; Significant increase in T2/FLAIR non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation of therapy, not due to co-morbid events; Any new lesion; Clear clinical deterioration not attributable to other causes apart from the tumor or changes in corticosteroid dose; Failure to return for evaluation due to death or deteriorating condition; Clear progression of non-measurable disease. PFS time is defined as time from registration to date of progression, death, or last known follow-up (censored). PFS rates are estimated using the Kaplan-Meier method. (NCT01062399)
Timeframe: Analysis occured after 134 events (progression or death) were reported. Patients were followed from randomization to death or study termination whichever occurs first, up to 36.7 months.

Interventionmonths (Median)
Ph II: RT + TMZ10.2
Ph II: RT + TMZ + RAD0018.2

[back to top]

6-month Progression-free Survival Rate

Six-month progression-free survival is the rate of patients who have NOT progressed at six months, where progressive disease is defined as any of the following: ≥ 25% increase in sum of the products of perpendicular diameters of enhancing lesions; any new lesion; or clinical deterioration. Progression will be determined by central review of MRI exams, assessed using MacDonald criteria for progression versus response on 2D T1 and T2 weighted images. (NCT01062425)
Timeframe: From randomization to 6 months.

Interventionpercentage of participants (Number)
Placebo, TMZ, and RT24.5
Cediranib, TMZ, and RT46.6

[back to top]

Overall Survival (OS)

OS will be estimated using the Kaplan-Meier method and differences between treatment arms will be tested using the log rank test. Multivariate analyses with the Cox proportional hazard model for OS will be performed with the stratification variables as fixed variables to assess the treatment effect adjusting patient-specific risk factors. (NCT01062425)
Timeframe: From randomization to time of death due to any cause. Patients are followed until death. Analysis occurs after all patients have been potentially followed for six months.

InterventionMonths (Median)
Placebo, TMZ, and RT13.8
Cediranib, TMZ, and RT14.5

[back to top]

Progression-free Survival (PFS)

Progression-free survival time is defined as time from randomization to date of first progression or death from any cause and is estimated by the Kaplan-Meier method. Patients last known to be alive without progression are censored at the date of last contact. Progression is defined as any of the following: ≥ 25% increase in sum of the products of perpendicular diameters of enhancing lesions; any new lesion; or clinical deterioration. (NCT01062425)
Timeframe: From randomization to time of first progression or death due to any cause. Patients are followed until death. Analysis occurs after all patients have been potentially followed for six months.

InterventionMonths (Median)
Placebo, TMZ, and RT2.7
Cediranib, TMZ, and RT6.2

[back to top]

Incidence of Grade 3+ Toxicities

The number of patients with reported grade 3 and higher treatment-related toxicities as assessed by Common Terminology Criteria for Adverse Events version 4.0 (NCT01062425)
Timeframe: From randomization to six months.

Interventionparticipants (Number)
Placebo, TMZ, and RT35
Cediranib, TMZ, and RT77

[back to top]

Median Progression-Free Survival (PFS)

PFS defined as time from date of diagnosis to most recent follow up, disease progression, or death. Disease progress defined as either clinical deterioration or radiographic progressive disease on magnetic resonance imaging (MRI) per updated response assessment in neuro-oncology criteria (Wen, et al). (NCT01105702)
Timeframe: Up to 50 months

Interventionmonths (Median)
TBL/RT9.4

[back to top]

Median Overall Survival (OS)

OS defined as time from diagnosis to most recent follow up or death. (NCT01105702)
Timeframe: Up to 50 months

Interventionmonths (Median)
TBL/RT18.5

[back to top]

Number of Patients With Grade 3 or 4 Adverse Events

Adverse events evaluated per CTCAE 3 (NCT01105702)
Timeframe: The whole time while on treatment and 30 days after the treatment

,
Interventionparticipants (Number)
DermatitisDVTNeurologicPneumoniaRashCoughHyponatremiaGastrointestinalFebril NeutropeniaNeutropeniaThrombocytopeniaSomnolence
Grade 3113111120111
Grade 4000000001010

[back to top]

Maximum Tolerated Dose (MTD) of Vorinostat in Combination With Escalating Doses of Erlotinib and Temozolomide

"Phase I assessment MTD Vorinostat in combination with escalating doses of Erlotinib and Temozolomide using conventional phase I design where 3 enrolled into first dose level, monitored for 3 weeks and if no dose-limiting toxicity (DLT) seen, 3 more enrolled at next dosage level. If 2/6 participants experience DLT, the previous (lower) dosage level declared MTD of vorinostat in combination with Erlotinib and Temozolomide.~A maximum of 4 dosage levels utilized with deescalation by 2 dose levels if DLT is seen at the starting dose level. If no DLT noted after dose escalation to Level 4, these doses utilized as MTD for phase II." (NCT01110876)
Timeframe: Evaluated with each 28 day (+2 days) cycle, up to 24 weeks

Interventionmg (Number)
Vorinostat (mg) twice/dayErlotinib (mg)
Phase I Group 1: Vorinostat + Erlotinib + Temozolomide200200

[back to top] [back to top]

Overall Survival - Percentage of Participants With an Event

Overall survival was defined as the time transpired (in weeks) between the date of the participant's inclusion in the trial until the date of his/her death by any cause. Participants that were alive at the time the analysis was performed were censored on the date of last contact. (NCT01115491)
Timeframe: BL, every 28 days, until death or end-of-study, an average of 32 weeks

Interventionpercentage of participants (Number)
Bevacizumab + Temozolomide75.00

[back to top]

Progression-Free Survival (PFS) - Percentage of Participants With an Event

PFS was defined as the time, in weeks, from the date of inclusion in the study to the date of the first documentation of disease progression or death of the participant due to any cause. Participants that did not have an event at the time the analysis was performed were censored at the date of last contact. Participants that began a treatment other than those planned in this study (bevacizumab or temozolomide) were censored on the start date of the new treatment. (NCT01115491)
Timeframe: Baseline (BL), every 28 days, until progression, death or end-of-study, an average of 32 weeks

Interventionpercentage of participants (Number)
Bevacizumab + Temozolomide96.88

[back to top]

PFS: Probability of Remaining Progression Free at 24 Weeks After Beginning the Study

(NCT01115491)
Timeframe: BL, 24 weeks (after 6th cycle)

Interventionsurvival probability (Number)
Bevacizumab + Temozolomide0.30000

[back to top]

PFS - Time to Event

PFS was defined as the time, in weeks, from the date of inclusion in the study to the date of the first documentation of disease progression or death of the participant due to any cause. Participants that did not have an event at the time the analysis was performed were censored at the date of last contact. Participants that began a treatment other than those planned in this study (bevacizumab or temozolomide) were censored on the start date of the new treatment. PFS was estimated using the Kaplan-Meier method. (NCT01115491)
Timeframe: BL, every 28 days, until progression, death or end-of-study, an average of 32 weeks

Interventionweeks (Median)
Bevacizumab + Temozolomide18.29

[back to top]

Percentage of Participants Achieving an Overall Response of Complete Response (CR) or Partial Response (PR)

Overall response was defined as the percentage of participants who obtained CR or PR using adapted MacDonald criteria. CR: disappearance of all index and non-index lesions, confirmed no less than 4 weeks after assessment, no evidence of disease progression; corticosteroid dosage at or below 20 mg hydrocortisone daily; no neurological changes or an improvement as compared to last disease assessment. PR was defined as: Fifty percent or greater decrease in the sum of products of the larger diameter and the larger perpendicular diameter of all index lesions confirmed no less than 4 weeks after assessment, no evidence of disease progression and the absence of progressive, or non-evaluable disease status for non-index legions; unchanged, or decreased corticosteroid dose as compared to the last disease assessment; no neurological changes or an improvement as compared to the neurological examination at last disease assessment. (NCT01115491)
Timeframe: BL, every 28 days, until progression, death or end-of-study, an average of 32 weeks

Interventionpercentage of participants (Number)
Bevacizumab + Temozolomide40.6

[back to top]

Overall Survival - Time to Event

Overall survival was defined as the time transpired (in weeks) between the date of the participant's inclusion in the trial until the date of his/her death by any cause. Participants that were alive at the time the analysis was performed were censored on the date of last contact. Median overall survival was estimated using the Kaplan-Meier method. (NCT01115491)
Timeframe: BL, every 28 days, until death or end-of-study, an average of 32 weeks

Interventionweeks (Median)
Bevacizumab + Temozolomide31.43

[back to top]

6-Month Progression-Free Survival (PFS) Rate

6-month progression free survival rate is defined as the number of participants without progression per RECIST 1.0 6 months after starting study treatment. (NCT01119508)
Timeframe: 6 Months

Interventionparticipants (Number)
Ipilimumab + Temozolomide29

[back to top]

Quality of Life by Brain-20 Survey

"Brain-20 (BN-20) quality of life surveys were administered at study entry and 12 months after treatment initiation to assess health-related quality of life (HR-QOL). The Brain-20 (BN-20) quality of life survey has 20 questions and responses are on scale of 1 to 4 with 1 indicating not at all (most favorable) and 4 indicating very much (least favorable). The total score can range from 20 to 80, and the result is expressed as the difference from baseline (study entry) to 12 months after the start of treatment. The outcome is stratified by Planning Target Volume (PTV) < 60 cm³ or 60 to 150 cm³, and expressed as the mean with 95% confidence interval." (NCT01120639)
Timeframe: 12 months

,,,
Interventionscore on a scale (Mean)
Planning Target Volume (PTV) <60 CM3Planning Target Volume (PTV) 60-150CM3
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide66.640.7
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide40.755.5
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide18.559.2
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide44.425.9

[back to top]

Quality of Life by European Organisation for Research and Treatment of Cancer (EORTC-QLQ C30) Survey

"European Organization for Research and Treatment of Cancer (EORTC-QLQ C30) quality of life surveys were administered at study entry and 12 months after treatment initiation to assess health-related quality of life (HR-QOL). The EORTC-QLQ C30 survey has 30 questions and responses are on scale of 1 to 4 with 1 indicating not at all and 4 indicating very much. The total score can range from 30 to 120. The outcome is stratified by Planning Target Volume (PTV) < 60 cm³ or 60 to 150 cm³, and is expressed as the mean of the difference from baseline to 12 months, with 95% confidence interval." (NCT01120639)
Timeframe: 12 Months

,,,
Interventionscore on a scale (Mean)
Planning Target Volume (PTV) <60 CM3Planning Target Volume (PTV) 60-150CM3
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide41.641.6
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide58.352.7
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide80.544.4
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide53.351.3

[back to top]

Quality of Life by MD Anderson Symptom Inventory - Brain Tumor (MDASI-BT) Survey

"MD Anderson Symptom Inventory - Brain Tumor (MDASI-BT) quality of life surveys were administered at study entry and 12 months after treatment initiation to assess health-related quality of life (HR-QOL). MDASI-BT quality of life survey has 23 questions and responses are on scale of 0 to 10 with 0 indicating did not interfere (most favorable) and 10 indicating interfered completely (least favorable). A participant's overall score is computed as the mean of that participant's individual scores, and can range 0 to 10. The outcome is stratified by Planning Target Volume (PTV) < 60 cm³ or 60 to 150 cm³, and expressed as the mean difference from baseline with 95% confidence interval. A positive value for the mean indicates worsening quality of life." (NCT01120639)
Timeframe: 12 months

,,,
Interventionscore on a scale (Mean)
Planning Target Volume (PTV) <60 CM3Planning Target Volume (PTV) 60-150CM3
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide5.23.7
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide2.03.2
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide2.82.3
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide3.23.6

[back to top]

Overall Survival (OS)

Overall survival (OS) was assessed as those participants remaining alive with any tumor status following radiotherapy after 20 months. The outcome is stratified by Planning Target Volume (PTV) < 60 cm³ or 60 to 150 cm³, and expressed as the median value with 95% confidence interval. (NCT01120639)
Timeframe: 20 Months.

,,,
InterventionMonths (Median)
Planning Target Volume (PTV) <60 CM3Planning Target Volume (PTV) 60-150CM3
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide17.610.1
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide19.911.3
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide48.317.6
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide10.211.6

[back to top]

Number of Acute Toxicity Within 30 Days

"Acute toxicity is defined as treatment-related adverse events that occur within 30 days of receiving radiotherapy. National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) v4.0 is used to grade adverse events. The non-stratified outcome is reported as number of treatment-related adverse events observed for each radiotherapy dose level.~Acute toxicity is based on radiotherapy dose level not tumor volume, and is reported by radiotherapy dose level only." (NCT01120639)
Timeframe: 30 days

,,,
Interventionadverse events (Number)
CTCAE Grade 1CTCAE Grade 2CTCAE Grade 3CTCAE Grade 4CTCAE Grade 5All grades
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide500005
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide200002
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide211004
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide400116

[back to top]

Long-term Toxicity After More Than 30 Days

"Long-term toxicity is defined as treatment-related adverse events (any grade or any Body System) that occur ≥ 30 days after receiving radiotherapy. National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) v4.0 is used to grade adverse events. The non-stratified outcome is reported as number of treatment-related adverse events observed for each dose level.~Long-term toxicity is based on radiotherapy dose level not tumor volume, and is reported by radiotherapy dose level only." (NCT01120639)
Timeframe: 12 months

,,,
InterventionNumber of adverse events (Number)
CTCAE Grade 1CTCAE Grade 2CTCAE Grade 3CTCAE Grade 4All grades
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide00303
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide01405
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide11103
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide11316

[back to top]

Treatment Failure Analysis

"Treatment failure in individual participants, ie, tumor recurrence or metastasis, can be described by the location relative to the first treatment failure (ie, infield, marginal, or distal), as further defined below.~Failure pattern is defined as tumor recurrence or metastasis relative to the primary lesion that is~Infield: at tumor or within 5 mm~Marginal: > 5 mm or ≤ 20 mm from tumor~Distal: > 20 mm from tumor~The outcome will be reported as the number of participants who failed treatment for each type of failure, ie, infield, marginal, or distal failure." (NCT01120639)
Timeframe: 18 months

,,,
InterventionParticipants (Count of Participants)
In-FieldMarginalDistal
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide213
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide501
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide211
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide812

[back to top]

Progression-free Survival

Progression-free survival (PFS) following radiotherapy, measured in months. Progressive disease (PD) is defined as: New tumor lesion, or > 25% increase in the product of the 2 greatest diameters of target lesion, as determined by computed tomography (CT) or magnetic resonance imaging (MRI), provided that within 2 months of completion of radiotherapy, the participant has not had a decrease in steroid dose since the last evaluation. The outcome is expressed as the median with 95% confidence interval for each cohort. (NCT01120639)
Timeframe: 18 Months.

,,,
InterventionMonths (Median)
Planning Target Volume (PTV) <60 CM3Planning Target Volume (PTV) 60-150CM3
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide13.47.2
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide10.56.5
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide33.58.8
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide3.94.0

[back to top]

Number of Dose-limiting Toxicities (DLTs)

"The maximum-tolerated dose (MTD) of study treatment (temozolomid plus hypofractionated radiotherapy administered as 5 fractions) is defined as either:~The highest radiation dose per protocol, or~The radiation dose at which dose-limiting toxicities (DLTs) occurred in ≥ 2 of 3 participants at a dose level, and/or ≥ 2 of 6 participants, at a dose level.~Dose-limiting toxicity (DLT) was defined as a treatment-related (with possible, probable or definite attribution) Grade 3 to 5 CNS toxicity [Common Terminology Criteria for Adverse Events (CTCAE) v4] occurring within 30 days of stereotactic radiosurgery (SRS).~The non-stratified outcome is reported as the number of DLTs observed in by radiation dose and by strata (Planning Target Volume (PTV) < 60 cm³ and from 60 to 150 cm³)." (NCT01120639)
Timeframe: 30 days

,,,
InterventionNumber of DLT observed (Number)
Planning Target Volume (PTV) < 60 cm³Planning Target Volume (PTV) 60 to 150 cm³
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide00
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide00
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide00
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide11

[back to top]

Percent of Participants With Radiographic Response

"Radiographic response rate was assessed following radiotherapy until disease progression. Response is considered to be the sum and proportion participants that achieved a complete response (CR); partial response (PR); or minor response (MR). The outcome is expressed as a number without dispersion for each cohort.~CR: Tumor is no longer detected by computed tomography (CT) or magnetic resonance imaging (MRI).~PR: Decrease in the product of the two greatest diameters > 50%, as determined by CT or MRI, with no new lesions, and the same or lower dose of dexamethasone.~MR: Decrease in the product of the two greatest diameters < 50%, as determined by CT or MRI, and neither PR nor PD.~PD: New tumor lesion, or > 25% increase in the product of the two greatest diameters of target lesion, as determined by CT or MRI, provided that within 2 months of completion of radiotherapy, the participant has not had a decrease in steroid dose since the last evaluation." (NCT01120639)
Timeframe: 6 months

,,,
InterventionPercentage of participants (Number)
Planning Target Volume (PTV) <60 CM3Planning Target Volume (PTV) 60-150CM3
Stereotactic Radiosurgery (25 Gray x 5 Fractions)+Temozolomide10066.7
Stereotactic Radiosurgery (30 Gray x 5 Fractions)+Temozolomide10066.7
Stereotactic Radiosurgery (35 Gray x 5 Fractions)+Temozolomide10066.7
Stereotactic Radiosurgery (40 Gray x 5 Fractions)+Temozolomide33.316.7

[back to top]

Duration of Response to High-Dose Interleukin-2 (H-D IL-2) Followed by Low Dose Temozolomide

Duration of response is defined as the length (measured in days) from the date of best response to the date of progression (if any), or to the date of last follow-up (if no progression is observed). The duration of response is applicable for those CR/MR/PR/SD subjects only. (NCT01124734)
Timeframe: 8 years

InterventionDays (Mean)
Course 1 Cycle 1 and Cycle 2432.88

[back to top]

Safety and Toxicity of H-D IL-2 Followed by Low Dose Temozolomide

Safety and toxicity in this study population was evaluated using the NCI Common Toxicity Criteria. The unit of measure is the number of study participants with one or more unexpected and related (even remotely) SAE. (NCT01124734)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Course 1 Cycle 10
Course 1 Cycle 20

[back to top]

Effect of High Dose IL2 Followed by Low Dose Temozolomide on Lymphocyte Subsets (Autoimmune Biomarkers)

The effect outcome is measured by the change in percentage of circulating lymphocyte cells (autoimmune biomarkers) that express the noted phenotype. This percentage change is determined by comparing the values obtained within 7 days of participant going off treatment against the baseline values. (NCT01124734)
Timeframe: 2 years

,
InterventionPercentage of Cells (Mean)
%CCR7- CD45RO+ of CD4+ Effector Memory%CCR7- CD45RO+ of CD8+ Effector Memory%CCR7- CD45RO- of CD4+ Effector Memory RA+%CCR7- CD45RO- of CD8+ Effector Memory RA+%CD25+FoxP3+ of CD4+ (Tregs)
% of Cells Expressing Phenotype Pre-treatment (BASELINE).36.31.09.46.10
% of Cells Expressing Phenotype Within 7 Days of Off-treatment.36.34.08.40.09

[back to top]

Progression-free Survival (PFS) (Evaluable Randomized Patients)

The primary analysis will be performed among the Gnaq/Gna11 mutant patients. A stratified logrank test will be performed stratified by mutation status, M stage, and number of prior systemic therapies for metastatic disease. Due to the potential for a large number of strata and small strata sizes, the standard asymptotic stratified logrank test will be verified for robustness utilizing a permutation reference distribution. (NCT01143402)
Timeframe: The time from randomization to the earlier date of objective disease progression per Response Evaluation Criteria In Solid Tumors (RECIST) criteria or death due to any cause in the absence of progression, assessed up to 5 years

Interventionweeks (Median)
Arm I (Temozolomide)7
Arm II (Selumetinib)15.9

[back to top]

Median Overall Survival (Evaluable Randomized Patients)

The primary analysis will be performed among the Gnaq/Gna11 mutant patients. A stratified logrank test will be performed stratified by mutation status, M stage, and number of prior systemic therapies for metastatic disease. Due to the potential for a large number of strata and small strata sizes, the standard asymptotic stratified logrank test will be verified for robustness utilizing a permutation reference distribution. (NCT01143402)
Timeframe: The time from randomization to death due to any cause, assessed up to 5 years

InterventionMonths (Median)
Arm I (Temozolomide)9.1
Arm II (Selumetinib)11.8

[back to top]

Grade 3-4 Post-Procedural Surgery-Related Toxicity Rate

Grade 3-4 post-procedural surgery-related toxicity rate is the percentage of participants experiencing at least one grade 3-4 adverse event (AE) during the post-procedural time frame of 14 days attributable to the surgical procedure based on NCI Common Toxicity Criteria for Adverse Events version 4 (CTCAEv4). (NCT01182350)
Timeframe: 2 weeks

Interventionpercentage of participants (Number)
All Biopsied Participants10.0

[back to top]

9-month Overall Survival (OS) Rate

9-month overall survival is the percentage of participants remaining alive 9 months from registration. (NCT01182350)
Timeframe: 9 months

Interventionpercentage of participants (Number)
All Patients Starting Assigned Chemoradiation64.4

[back to top]

Feasibility Rate of Molecular Approach to Therapy

Feasibility rate of the molecular strategy is based on the percentage of participants either with inadequate tissue from surgical biopsy to confirm DIPG diagnosis and/or with uninterpretable results for identification of EGFR overexpression and MGMT methylation status. (NCT01182350)
Timeframe: 3 weeks

Interventionpercentage of participants (Number)
All Biopsied Participants92.0

[back to top]

Rate of Lethal Complications From Surgery

The rate of lethal complications from surgery is the percentage of participants dying as a result of surgery. (NCT01182350)
Timeframe: 2 weeks

Interventionpercentage of participants (Number)
All Biopsied Participants0.0

[back to top] [back to top]

Median Progression Free Survival (PFS)

PFS based on the Kaplan-Meier method is defined as the duration of time [months (m)] from study entry to documented disease progression (PD) or death. Participants alive without PD are censored at the date of last disease assessment. For intrinsic pontine brainstem gliomas, only one lesion/mass is present at diagnosis. Comparisons of maximal 2-dimensional measurements, TxW (product of the longest diameter [width (W)] and its longest perpendicular diameter [transverse (T)]) are used to assess response for this target lesion. PD is 25% or more increase, taking as reference the smallest product observed since the start of treatment, or the appearance of one or more new lesions. (NCT01182350)
Timeframe: Disease assessments using a standard CNS imaging protocol occurred chemoradiation cycles 1 and 2, every other maintenance cycle, every 3 months post-treatment year 1 then annually until PD or therapy change; In this study cohort, follow-up was up to 34m.

Interventionmonths (Median)
All Patients Starting Assigned Chemoradiation8

[back to top]

Delay in Radiation Therapy Start

The number of participants delaying the start of radiation therapy by more than 3 weeks due to complications as a result of surgical biopsy to obtain diagnostic tumor sample. (NCT01182350)
Timeframe: 3 weeks

InterventionParticipants (Count of Participants)
All Biopsied Participants1

[back to top]

9-month Overall Survival (OS) Rate by Molecular Cohort

9-month OS rate is the percentage of participants alive at 9 months from registration. (NCT01182350)
Timeframe: 9 months

Interventionpercentage of participants (Number)
Cohort 1: MGMT-/EGFR-66.7
Cohort 2: MGMT-/EGFR+57.1
Cohort 3. MGMT+/EGFR-50.0
Cohort 4. MGMT+/EGFR+100

[back to top] [back to top]

Median Progression-free Survival

Progression-free survival was defined as the time in months from the date study treatment started until the date of progression or the date of death if death occurred before progression, or until the date of last follow-up if alive without progression. Kaplan-Meier methods were used to estimate progression-free survival. (NCT01186406)
Timeframe: 21 months

Interventionmonths (Median)
Gliadel, Radiation Therapy, Avastin, Temodar11.3

[back to top]

Median Overall Survival

Overall survival was defined as the time in months from the start of SRS to the date of death or last contact if alive. Kaplan-Meier methods were used to estimate overall survival. (NCT01186406)
Timeframe: 21 months

Interventionmonths (Median)
Gliadel, Radiation Therapy, Avastin, Temodar19.4

[back to top]

21-month Overall Survival

The percentage of participants alive at 21 months after the start of study treatment. Overall survival was calculated from the date study treatment started until the date of death or the date of last follow-up if alive. Kaplan-Meier methods were used to estimate overall survival. (NCT01186406)
Timeframe: 21 months

Interventionpercentage of participants (Number)
Gliadel, Radiation Therapy, Avastin, Temodar40.9

[back to top]

Cycle 1 Mean Day 2 Trough/Pharmacokinetics of Gamma-secretase Inhibitor RO4929097 in Combination With Temozolomide (Phase IB)

(NCT01196416)
Timeframe: At Day 2 of Cycle 1

Interventionng/mL (Mean)
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)129

[back to top]

Participants Evaluated for Toxicity

Number of patients with AE's as assessed by NCI CTCAE v. 4.0 Please see Adverse Events section for specifics. (NCT01196416)
Timeframe: Up to 30 days post-treatment

InterventionParticipants (Count of Participants)
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)14

[back to top]

Progression-free Survival (Phase II)

Progression-free survival curves will be generated using Kaplan-Meier methodology. (NCT01196416)
Timeframe: Time from start of treatment to time of progression or death, whichever occurs first, assessed up to 2 years

Interventionmonths (Median)
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)2.7

[back to top]

Maximum Tolerated Dose for RO4929097

based on the incidence of dose-limiting toxicity as assessed the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (Phase IB) (NCT01196416)
Timeframe: 21 days

Interventionmg/day (Number)
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)15

[back to top]

Number of Participants With Presence or Absence of Markers of Notch Signalling Pathway Inhibition in Patient Tumors (Phase Ib)

The association of response or clinical benefit with the presence or absence of markers of pathway inhibition in patient tumors will be tested using Fisher's exact test. (NCT01196416)
Timeframe: 2 weeks

InterventionParticipants (Count of Participants)
Insufficient tissue to asses any changeSig decrease in post-treatment cleaved Notch
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)14

[back to top]

Overall Survival (Phase II)

Overall response rate (complete [CR] or partial response [PR]) according to RECIST version 1.1 (NCT01196416)
Timeframe: Up to 2 years

Interventionparticipants (Number)
Partial ResponseStable DiseaseProgression of Disease
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)356

[back to top]

Cycle 1 AUC/Pharmacokinetics of Gamma-secretase Inhibitor RO4929097 in Combination With Temozolomide (Phase IB)

(NCT01196416)
Timeframe: Days 4 and 5

Interventionhr·ng/mL (Mean)
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)5410

[back to top]

Cycle 1 C Max/Pharmacokinetics of Gamma-secretase Inhibitor RO4929097 in Combination With Temozolomide (Phase IB)

(NCT01196416)
Timeframe: At Cycle 1

Interventionng/mL (Mean)
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)301

[back to top]

Maximum-tolerated Dose for Cisplatin, Vinblastine and TMZ

"based on the incidence of dose-limiting toxicity as assessed the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (Phase IB)~Data is not yet available, as it's currently being analyzed." (NCT01196416)
Timeframe: 21 days

Interventionmg/m2 (Number)
CisplatinVinblastineTemozolomide
Treatment (RO4929097, Cisplatin, Vinblastine, Temozolomide)201.2150

[back to top]

Overall Survival

the number of months between a patient's enrollment and his/her date of death (NCT01205828)
Timeframe: 2 years

Interventionmonths (Median)
ABT-888 and Temozolomide13.1

[back to top]

Clinical Benefit Rate

complete response at any time + partial response at any time + stable disease after 8 weeks of treatment based on RECIST Criteria (NCT01205828)
Timeframe: 8 weeks

Interventionparticipants (Number)
ABT-888 and Temozolomide3

[back to top]

Progression Free Survival

The number of months between a patient's enrollment and his/her disease progression (NCT01205828)
Timeframe: 2 years

Interventionmonths (Median)
ABT-888 and Temozolomide1.9

[back to top]

Number of Participants Who Had Grade 3 or 4 Adverse Events

Record of all toxicities graded according to the NCI CTCAE version 3.0 (NCT01205828)
Timeframe: 6 months

Interventionparticipants (Number)
ABT-888 and Temozolomide5

[back to top]

6-month Progression-free Survival

To use 6-month progression-free survival to assess the efficacy of the combination of hypofractionated IMRT delivering 60 Gy over 2 weeks with concurrent bevacizumab and temozolomide followed by 6 cycles of adjuvant bevacizumab and temozolomide. (NCT01209442)
Timeframe: 6 months

Interventionmonths (Median)
RT With Temozolomide and Bevacizumab90

[back to top]

Overall Survival, Measured From the Day of Initial Diagnosis (Biopsy or Surgery) to the Time of Death From Any Cause.

(NCT01209442)
Timeframe: follow up for life

Interventionmonths (Median)
RT With Temozolomide and Bevacizumab16.3

[back to top]

Response

Patient's best response during protocol therapy coded as complete response, partial response or no response. (NCT01217437)
Timeframe: Up to 12 cycles of therapy (11 months)

,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseNo Response
Arm I (Temozolomide, Irinotecan Hydrochloride)01632
Arm II (Temozolomide, Irinotecan Hydrochloride, Bevacizumab)81424

[back to top]

Event-free Survival

Percentage Probability of remaining event-free 5 years after enrollment estimated by the method of Kaplan and Meier (NCT01217437)
Timeframe: Up to 5 years after enrollment

Interventionpercent probability (Number)
Arm I (Temozolomide, Irinotecan Hydrochloride)1.85
Arm II (Temozolomide, Irinotecan Hydrochloride, Bevacizumab)15.4

[back to top]

Overall Survival

Percentage Probability of remaining alive 5 years after enrollment estimated by the method of Kaplan and Meier (NCT01217437)
Timeframe: Up to 5 years after enrollment

Interventionpercent probability (Number)
Arm I (Temozolomide, Irinotecan Hydrochloride)2.43
Arm II (Temozolomide, Irinotecan Hydrochloride, Bevacizumab)13.6

[back to top]

One-year Progression-free Survival and Overall Survival

"Efficacy of the DRBEAT Regimen will be assessed by analysis of~one-year progression-free survival (PFS), defined as the time interval from maximal response from therapy to tumor regrowth, progression*, or death, (*Progression is defined as meeting the response criteria listed in Table 4: Response Criteria for Primary Central Nervous System Lymphoma according to Abrey LE, Batchelor TT, Ferreri AJM et al.)~and~Overall survival, defined as the time interval between the date of transplant and the date of death from any cause." (NCT01235793)
Timeframe: (1) One Year (2) Until date of death from any cause, assessed up to 2 years

InterventionDays (Median)
Progression Free SurvivalOverall Survival
DRBEAT Regimen132564

[back to top]

Safest Dose of Temozolomide for the DRBEAT Regimen

Safety will be assessed using a dose escalation design for temozolomide's use to determine the target dose and also to evaluate any and all acute treatment related toxicities. During the course of patient follow up and therapy, toxicities will be evaluated, particularly as the investigators will be determining the target dose of temozolomide. One of the major criteria for dose limiting toxicity for the study will be any Grade 3 or 4 nonhematologic toxicity from a list of commonly expected toxicities associated with autologous transplantation and temozolomide. (NCT01235793)
Timeframe: One Year

Interventiondose in mg/m^2 (Number)
DRBEAT Regimen773.25

[back to top]

Event-free Survival

Time from enrollment to disease progression, diagnosis of a second malignant neoplasm, death or last follow-up, whichever occurs first. Disease progression is evaluated according to the COG criteria for measurement of brain tumors and is defined to be a ≥ 25% increase in the product of perpendicular diameters of ANY target lesion, taking as reference the smallest product observed since the start of treatment; OR the appearance of one or more new lesions, OR worsening neurologic status not explained by causes unrelated to tumor progression (e.g., anticonvulsant or corticosteroid toxicity, electrolyte disturbances, sepsis, hyperglycemia, presumed post-therapy swelling etc) PLUS any increase in tumor cross-sectional area (or tumor volume). (NCT01236560)
Timeframe: 1 year after enrollment

Interventionpercent probability (Number)
Feasibility (Vorinostat)33.3
Arm I (Vorinostat, Phase II Arm A)41.3
Arm II (Temozolomide, Phase II Arm B)59.3
Arm III (Bevacizumab, Phase II Arm C)43.8

[back to top]

Cumulative Incidence of Disease Progression in Each Treatment Arm

Cumulative incidence of progression where death from any cause prior to progression and diagnosis of a second malignant neoplasm prior to progression are considered competing events. Disease progression is evaluated according to the COG criteria for measurement of brain tumors and is defined to be a ≥ 25% increase in the product of perpendicular diameters of ANY target lesion, taking as reference the smallest product observed since the start of treatment; OR the appearance of one or more new lesions, OR worsening neurologic status not explained by causes unrelated to tumor progression (e.g., anticonvulsant or corticosteroid toxicity, electrolyte disturbances, sepsis, hyperglycemia, presumed post-therapy swelling etc) PLUS any increase in tumor cross-sectional area (or tumor volume). (NCT01236560)
Timeframe: 1 year after enrollment

Interventionpercent probability (Number)
Feasibility (Vorinostat)67
Arm I (Vorinostat, Phase II Arm A)59
Arm II (Temozolomide, Phase II Arm B)53
Arm III (Bevacizumab, Phase II Arm C)37

[back to top]

Maximum Tolerated Dose (MTD) of Vorinostat

The dose of vorinostat in mg/sq m/day to be administered with combination chemotherapy and radiation therapy. (NCT01236560)
Timeframe: 10 weeks

Interventionmg/sq m (Number)
Feasibility (Vorinostat)230

[back to top]

Overall Survival

Time from enrollment to death or last follow-up, whichever occurs first. (NCT01236560)
Timeframe: 1 year after enrollment

Interventionpercent probability (Number)
Feasibility (Vorinostat)33.3
Arm I (Vorinostat, Phase II Arm A)82.2
Arm II (Temozolomide, Phase II Arm B)85.2
Arm III (Bevacizumab, Phase II Arm C)67.3

[back to top]

Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)

Best overall tumor response was evaluated using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Complete Response (CR) was defined as the disappearance of all target lesions; Partial Response (PR) was defined as at least a 30% decrease in sum of longest diameter of target lesions. (NCT01284335)
Timeframe: Baseline to Study Completion (Up to 2 years)

Interventionpercentage of participants (Number)
Non-Small Cell Lung Cancer (NSCLC)Other
Arm B* Tasisulam + Docetaxel Dose Confirmation20.06.3

[back to top]

Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)

Best overall tumor response was evaluated using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Complete Response (CR) was defined as the disappearance of all target lesions; Partial Response (PR) was defined as at least a 30% decrease in sum of longest diameter of target lesions. (NCT01284335)
Timeframe: Baseline to Study Completion (Up to 2 years)

Interventionpercentage of participants (Number)
Non-Small Cell Lung Cancer (NSCLC)OtherPancreasSmall Cell Lung Cancer (SCLC)
Arm D Tasisulam + Cisplatin Dose Confirmation5.010.007.1

[back to top]

Pharmacokinetic (PK): Concentration Maximum (Cmax)

Cycle 2: predose,1h start of infusion, end of infusion, 30min,2h,4h,6h,22h,166h,334h,698h end of infusion. (NCT01284335)
Timeframe: Cycle 1: predose,0,30min,1h start of infusion, end of infusion, 30min,2h,4h,6h,22h,166h,334h,698h end of infusion.

Interventionmicrograms per milliliter (µg/mL) (Geometric Mean)
Cycle 1Cycle 2
Tasisulam306250

[back to top]

Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)

Best overall tumor response was evaluated using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Complete Response (CR) was defined as the disappearance of all target lesions; Partial Response (PR) was defined as at least a 30% decrease in sum of longest diameter of target lesions. (NCT01284335)
Timeframe: Baseline to Study Completion (Up to 2 years)

,
Interventionpercentage of participants (Number)
Non-Small Cell Lung Cancer (NSCLC)OtherPancreas
Arm A Tasisulam + Gemcitabine Dose Confirmation0.014.313.3
Arm E Tasisulam + Erlotinib Dose Confirmation0.014.30.0

[back to top]

PK: Area Under the Curve Albumin (AUCalb)

Cycle 2: predose,1h start of infusion, end of infusion, 30min,2h,4h,6h,22h,166h,334h,698h end of infusion. (NCT01284335)
Timeframe: Cycle 1: predose,0,30min,1h start of infusion, end of infusion, 30min,2h,4h,6h,22h,166h,334h,698h end of infusion.

Interventionmicrograms*hour/milliliter (µg*h/mL) (Geometric Mean)
Cycle 1Cycle 2
Tasisulam946648

[back to top]

Number of Participants With Dose-Limiting Toxicities Cycle 1

A Dose-Limiting Toxicity (DLT) is defined as an Adverse Event (AE) that is likely related to the study medication or combination, and fulfills any one of the following criteria: Common Terminology Criteria for Adverse Events (CTCAE, Version 3.0) Grade 4 neutropenia lasting more than 5 days. Grade 4 neutropenia with fever or Grade 4 thrombocytopenia, regardless of duration; Grade ≥3 thrombocytopenia with bleeding, regardless of duration; Grade ≥3 nonhematologic toxicity (excluding nausea/vomiting or diarrhea that can be controlled with medication, and alopecia). Grade 3 electrolyte toxicity (for example, hypokalemia, hypophosphatemia) will not be considered a DLT unless it is considered related to the study drug or combination and does not resolve with standard replacement treatments within 42 days after Cycle 1 Day 1. A summary of other nonserious AEs and all Serious Adverse Events (SAE), regardless of causality is located in the Reported Adverse Event section. (NCT01284335)
Timeframe: Baseline to Cycle 1 (Up to Day 28)

InterventionParticipants (Count of Participants)
Arm A Tasisulam + Gemcitabine Dose Escalation4
Arm A Tasisulam + Gemcitabine Dose Confirmation0
Arm B* Tasisulam + Docetaxel Dose Escalation4
Arm B1 Tasisulam + Docetaxel Dose Escalation4
Arm B2 Tasisulam + Docetaxel Dose Escalation3
Arm B2 Tasisulam + Docetaxel Dose Confirmation0
Arm C Tasisulam + Temozolomide Dose Escalation3
Arm C Tasisulam + Temozolomide Dose Confirmation0
Arm D* Tasisulam + Cisplatin Dose Escalation0
Arm D Tasisulam + Cisplatin Dose Escalation2
Arm D Tasisulam + Cisplatin Dose Confirmation0
Arm E Tasisulam + Erlotinib Dose Escalation1
Arm E Tasisulam + Erlotinib Dose Confirmation0

[back to top]

Number of Participants With a Clinically Significant Effects

Clinically significant effects are reported if a Grade 3 or higher treatment emergent adverse event (TEAE) and observed in ≥10% of participants or a toxicity possibly related to study drug based on Common Terminology Criteria for Adverse Events (CTCAE). A summary of other nonserious AEs and all SAEs, regardless of causality is located in the Reported Adverse Event section. (NCT01284335)
Timeframe: Baseline to Study Completion (Up to 2 years)

,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
TEAE >/= Grade 3Toxicity >/= Grade 3
Arm A Tasisulam + Gemcitabine Dose Confirmation1818
Arm A Tasisulam + Gemcitabine Dose Escalation1513
Arm B* Tasisulam + Docetaxel Dose Escalation04
Arm B1 Tasisulam + Docetaxel Dose Escalation05
Arm B2 Tasisulam + Docetaxel Dose Confirmation1926
Arm B2 Tasisulam + Docetaxel Dose Escalation1618
Arm C Tasisulam + Temozolomide Dose Confirmation66
Arm C Tasisulam + Temozolomide Dose Escalation1113
Arm D Tasisulam + Cisplatin Dose Confirmation3030
Arm D Tasisulam + Cisplatin Dose Escalation87
Arm D* Tasisulam + Cisplatin Dose Escalation01
Arm E Tasisulam + Erlotinib Dose Confirmation77
Arm E Tasisulam + Erlotinib Dose Escalation55

[back to top]

Percentage of Participants With a Complete (CR) or Partial Response (PR) (Best Overall Tumor Response)

Best overall tumor response was evaluated using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Complete Response (CR) was defined as the disappearance of all target lesions; Partial Response (PR) was defined as at least a 30% decrease in sum of longest diameter of target lesions. (NCT01284335)
Timeframe: Baseline to Study Completion (Up to 2 years)

Interventionpercentage of participants (Number)
Other
Arm C Tasisulam + Temozolomide Dose Confirmation0.0

[back to top]

Toxicity, Assessed Using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 (v4)

"The maximum grade for each type of toxicity will be recorded for each patient, and frequency tables will be reviewed to determine toxicity patterns within patient groups. In addition, we will review all adverse event data that is graded as 3, 4, or 5 and classified as either unrelated or unlikely to be related to study treatment in the event of an actual relationship developing. The overall toxicity rates (percentages) for grade 3 or higher adverse events considered at least possibly related to treatment are reported below." (NCT01328535)
Timeframe: Up to 2 years

Interventionpercentage of patients (Number)
FatigueFebrile NeutropeniaNeutrophil Count DecreasedPlatelet Count DecreasedSepsisUrinary Tract InfectionWhite Blood Cell DecreasedAbdominal Infection
Treatment (Individualized Chemotherapy)4.28.38.3254.24.28.34.2

[back to top]

Progression-Free Survival at 4 Months

The distribution of time to progression will be estimated using the method of Kaplan-Meier and the 4 month progression-free rate (percentage) will be provided. Progression is defined as: At least one of the following must be true: At least one new malignant lesion, which also includes any lymph node that was normal at baseline (< 1.0 cm short axis) and increased to ≥ 1.0 cm short axis during follow-up. At least a 20% increase in PBSD (sum of the longest diameter for all target lesions plus the sum of the short axis of all the target lymph nodes at current evaluation) taking as reference the MSD. In addition, the PBSD must also demonstrate an absolute increase of at least 0.5 cm from the MSD. (NCT01328535)
Timeframe: Time from registration to the earliest date of documentation of disease progression, assessed at 4 months

Interventionpercentage of patients (Number)
Treatment (Timed Individualized Chemotherapy)22.2

[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 is defined as:At least one of the following must be true: At least one new malignant lesion, which also includes any lymph node that was normal at baseline (< 1.0 cm short axis) and increased to ≥ 1.0 cm short axis during follow-up. At least a 20% increase in PBSD (sum of the longest diameter for all target lesions plus the sum of the short axis of all the target lymph nodes at current evaluation) taking as reference the MSD (Section 11.41). In addition, the PBSD must also demonstrate an absolute increase of at least 0.5 cm from the MSD. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01328535)
Timeframe: Up to 2 years

Interventionmonths (Median)
Treatment (Timed Individualized Chemotherapy)3.4
Treatment (Untimed Individualized Chemotherapy)7.2

[back to top]

Overall Survival

Overall survival time is defined as the time from randomization to death due to any cause. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01328535)
Timeframe: Up to 2 years

Interventionmonths (Median)
Treatment (Timed Individualized Chemotherapy)23.1
Treatment (Untimed Individualized Chemotherapy)17.5

[back to top]

Measurable Change on Magnetic Resonance Spectroscopy Imaging After Vorinostat Administration

Changes in magnetic resonance spectroscopic imaging signal and semiquantitative analysis of inositol, choline, lactate, and N-acetyl aspartate (NAA) signal are measured. The values for each of these metabolites are normalized to baseline at one and nine weeks. The values of all the metabolites at one week are added and this is the magnetic resonance spectroscopic index for one week. This is done again at nine weeks. The number is unitless and there is no range limit. Positive values represent normalization of tumor metabolism and negative values suggest no improvement or worsening of metabolic character. (NCT01342757)
Timeframe: 9 weeks

InterventionSpectroscopic index (Mean)
Arm I0

[back to top]

Proportion of Patients With Magnetic Resonance Spectroscopy (MRS) Response to Initial Vorinostat by MRI and MRS Scans as Determined by Spectroscopic Index

Changes in magnetic resonance spectroscopic imaging signal and semiquantitative analysis of inositol, choline, lactate, and N-acetylaspartate signal are measured. The values for each of these metabolites are normalized to baseline at one and nine weeks. The values of all the metabolites at one week are added and this is the magnetic resonance spectroscopic index for one week. This is done again at nine weeks. The number is unitless and there is no range limit. Positive values represent normalization of tumor metabolism and negative values suggest no improvement or worsening of metabolic character. (NCT01342757)
Timeframe: 9 weeks

Interventionparticipants (Number)
Arm I0

[back to top]

Mean Change in Metabolite Levels

Baseline MRS was performed 1-3 days before initiation of treatment. Follow-up MRS studies were performed at day 7. A standard quadrature head coil was used to collect MR data. The change of metabolite level in choline (Cho) and N-acetyl aspartate (NAA) were calculated in ratio by (metabolite after treatment / metabolite before treatment - 1). The reported ratios represent the Cho-to-NAA ratio at day 7 compared with day 0. (NCT01342757)
Timeframe: Baseline to 1 week

InterventionCho/NAA Ratios (Mean)
Metabolic RespondersMetabolic Non-Responders
Arm I-0.150.29

[back to top]

Percentage of Participants With EFS as Determined by the CRRC at 1 Year

EFS was defined as the time from randomisation to the earliest occurrence of any of the following: tumor progression, tumor recurrence, second primary non- HGG malignancy or death attributable to any cause. Tumor assessments were conducted using MRI and reviewed by the site-independent CRRC using RANO criteria. Tumor progression was defined as clear clinical progression or >/= 25% increase in the sum of the products of perpendicular diameters of the contrast enhancing lesions compared with the smallest tumor measurement obtained either at baseline (if no decrease was observed) or best response and with the subject on stable or increasing doses of corticosteroids. Tumor recurrence was defined as recurrence after tumor was completely resected (no disease present at baseline). EFS was estimated using the Kaplan-Meier method. (NCT01390948)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Chemoradiation + TMZ48.37
Chemoradiation + Bevacizumab + TMZ38.28

[back to top]

Health Status as Measured by the Health Utility Index (HUI)

HUI is a preference-based, multi-attitude, health-related instrument specifically developed for use with children. HUI consists of eight attributes of health status: vision, hearing, speech, ambulation, dexterity, emotion, cognition and pain. Each attribute had 5 or 6 levels varying from highly impaired to normal. Each of the eight health dimensions was tested separately and a composite score ranging between 1 (perfect health) and 0 (death) was obtained for participants aged 5 years or older. (NCT01390948)
Timeframe: Baseline, Cycle 6 of the adjuvant phase, end of treatment (approximately 58 weeks post-baseline), and yearly during the follow-up period (maximum 5 years in follow-up)

Interventionunits on a scale (Mean)
BaselineCycle 6, Day 1End of TreatmentYearly Follow-Up 1Yearly Follow-up 2Additional Safety Follow-Up (Visit 2)Additional Safety Follow-Up (Visit 4)Additional Safety Follow-Up (Visit 6)Additional Safety Follow-Up (Visit 8)End of Study
Chemoradiation + Bevacizumab + TMZ0.7300.7790.8200.9260.7930.9010.8300.4900.9300.790

[back to top]

Health Status as Measured by the Health Utility Index (HUI)

HUI is a preference-based, multi-attitude, health-related instrument specifically developed for use with children. HUI consists of eight attributes of health status: vision, hearing, speech, ambulation, dexterity, emotion, cognition and pain. Each attribute had 5 or 6 levels varying from highly impaired to normal. Each of the eight health dimensions was tested separately and a composite score ranging between 1 (perfect health) and 0 (death) was obtained for participants aged 5 years or older. (NCT01390948)
Timeframe: Baseline, Cycle 6 of the adjuvant phase, end of treatment (approximately 58 weeks post-baseline), and yearly during the follow-up period (maximum 5 years in follow-up)

Interventionunits on a scale (Mean)
BaselineCycle 6, Day 1End of TreatmentYearly Follow-Up 1Yearly Follow-up 2Additional Safety Follow-Up (Visit 2)Additional Safety Follow-Up (Visit 4)Additional Safety Follow-Up (Visit 6)End of Study
Chemoradiation + TMZ0.7130.7850.8320.9060.7370.7840.8141.0000.647

[back to top]

Number of Dose Administrations of TMZ and Bevacizumab in the Concurrent Phase

Number of doses were assessed for the concurrent phase, which is the treatment period after the initial treatment phase and including the subsequent treatment break of approximately 4 weeks. (NCT01390948)
Timeframe: Beginning of the concurrent phase to end of treatment break (10 weeks)

,
Interventionnumber of dose administrations (Median)
TMZBevacizumab
Chemoradiation + Bevacizumab + TMZ42.06.0
Chemoradiation + TMZ42.0NA

[back to top]

Percentage of Participants Who Completed >/= 90% of Planned Radiotherapy and TMZ Administrations

(NCT01390948)
Timeframe: From the time of randomization of the first participant to the date of clinical cutoff (approximately 60 months)

,
Interventionpercentage of participants (Number)
RadiotherapyTMZ
Chemoradiation + Bevacizumab + TMZ98.388.3
Chemoradiation + TMZ94.685.7

[back to top]

Percentage of Participants With a Treatment Delay or Discontinuation

(NCT01390948)
Timeframe: From the time of randomization of the first participant to the date of clinical cutoff (approximately 60 months)

,
Interventionpercentage of participants (Number)
AE leading to dose modification/interruptionAE leading to withdrawal from treatment
Chemoradiation + Bevacizumab + TMZ71.721.7
Chemoradiation + TMZ60.75.4

[back to top]

Percentage of Participants With EFS as Determined by the CRRC at 6 Months

EFS was defined as the time from randomisation to the earliest occurrence of any of the following: tumor progression, tumor recurrence, second primary non- HGG malignancy or death attributable to any cause. Tumor assessments were conducted using MRI and reviewed by the site-independent CRRC using RANO criteria. Tumor progression was defined as clear clinical progression or >/= 25% increase in the sum of the products of perpendicular diameters of the contrast enhancing lesions compared with the smallest tumor measurement obtained either at baseline (if no decrease was observed) or best response and with the subject on stable or increasing doses of corticosteroids. Tumor recurrence was defined as recurrence after tumor was completely resected (no disease present at baseline). EFS was estimated using the Kaplan-Meier method. (NCT01390948)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Chemoradiation + TMZ66.46
Chemoradiation + Bevacizumab + TMZ68.43

[back to top]

Concordance Between Structural Versus Multimodal Imaging for CRRC-Assessed Event-Free Survival

Concordance is presented as the percentage of participants with concordance between assessments. EFS concordance was defined as event Structural assessment and Diffusion Perfusion assessment occurs within 28 days or no event Structural and no Diffusion Perfusion. (NCT01390948)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Chemoradiation + TMZ96.6
Chemoradiation + Bevacizumab + TMZ87.1

[back to top]

EFS as Assessed by the Investigator

EFS was defined as the time from randomisation to the earliest occurrence of any of the following: tumor progression, tumor recurrence, second primary non-HGG malignancy or death attributable to any cause. Tumor assessments were conducted using MRI and reviewed by the investigator using RANO criteria. Tumor progression was defined as clear clinical progression or >/= 25% increase in the sum of the products of perpendicular diameters of the contrast enhancing lesions compared with the smallest tumor measurement obtained either at baseline (if no decrease was observed) or best response and with the participant on stable or increasing doses of corticosteroids. Tumor recurrence was defined as recurrence after tumor was completely resected (no disease present at baseline). EFS was estimated using the Kaplan-Meier method. (NCT01390948)
Timeframe: From the time of randomization to the date of any defined event (up to 12 months)

Interventionmonths (Median)
Chemoradiation + TMZ11.79
Chemoradiation + Bevacizumab + TMZ11.27

[back to top]

Event-Free Survival (EFS) as Assessed by the Central Radiology Review Committee (CRRC)

EFS was defined as the time from randomisation to the earliest occurrence of any of the following: tumor progression, tumor recurrence, second primary non- HGG malignancy or death attributable to any cause. Tumor assessments were conducted using magnetic resonance imaging (MRI) and reviewed by the site-independent CRRC using Response Assessment in Neuro-Oncology (RANO) criteria. Tumor progression was defined as clear clinical progression or >/= 25% increase in the sum of the products of perpendicular diameters of the contrast enhancing lesions compared with the smallest tumor measurement obtained either at baseline (if no decrease was observed) or best response and with the subject on stable or increasing doses of corticosteroids. Tumor recurrence was defined as recurrence after tumor was completely resected (no disease present at baseline). EFS was estimated using the Kaplan-Meier method. (NCT01390948)
Timeframe: From the time of randomization to the date of any defined event (up to 12 months)

Interventionmonths (Median)
Chemoradiation + TMZ11.79
Chemoradiation + Bevacizumab + TMZ8.21

[back to top]

Neurological Psychological Function as Measured by the Wechsler Scale

The Wechsler Intelligence Scale for Children version IV (WISC-IV) was used to generate a full scale intelligence quotient (IQ) which represents a child's general intellectual ability. The average IQ score is 100, with lower scores representing lower intellectual ability. (NCT01390948)
Timeframe: End of treatment (approximately 58 weeks post-baseline)

Interventionunits on a scale (Mean)
Chemoradiation + TMZ92.0
Chemoradiation + Bevacizumab + TMZ97.0

[back to top]

Number of Radiotherapy Dose Administrations in the Concurrent Phase

Number of doses were assessed for the concurrent phase, which is the treatment period after the initial treatment phase and including the subsequent treatment break of approximately 4 weeks. (NCT01390948)
Timeframe: Beginning of the concurrent phase to end of treatment break (10 weeks)

InterventionGrays (Median)
Chemoradiation + TMZ54.0
Chemoradiation + Bevacizumab + TMZ54.0

[back to top]

Objective Response Rate (ORR)

ORR was defined as the percentage of participants with a complete response (CR) or partial response (PR) determined on two consecutive occasions >/= 4 weeks apart. Tumor assessments were conducted using MRI and reviewed by the site-independent CRRC using RANO criteria. The following were needed to qualify as CR: complete disappearance of all measurable enhancing lesions sustained for at least 4 weeks by MRI, no steroids above physiological levels, clinical status stable or improved compared to baseline. The following were needed to qualify as PR: ≥ 50% decrease from baseline in the sum of products of perpendicular diameters of all measurable enhancing lesions sustained for at least 4 weeks by MRI, steroid dose not increased compared to baseline, clinical status stable or improved compared to baseline. (NCT01390948)
Timeframe: From the time of randomization to the date of any defined event (up to 12 months)

Interventionpercentage of participants (Number)
Chemoradiation + TMZ40
Chemoradiation + Bevacizumab + TMZ41.7

[back to top]

Overall Survival

Overall Survival was defined as the time of diagnosis to the date of death due to any cause. Overall Survival was estimated using the Kaplan-Meier method. (NCT01390948)
Timeframe: From the time of randomization to the date of death (up to approximately 60 months)

Interventionmonths (Median)
Chemoradiation + TMZ20.27
Chemoradiation + Bevacizumab + TMZ18.30

[back to top]

Percentage of Participants With 1-Year Survival

1-year survival was estimated using the Kaplan-Meier method. (NCT01390948)
Timeframe: 1 year after end of treatment

Interventionpercentage of participants (Number)
Chemoradiation + TMZ67.69
Chemoradiation + Bevacizumab + TMZ74.83

[back to top]

Percentage of Participants With an Adverse Event (AE)

An AE was defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT01390948)
Timeframe: From the time of randomization of the first participant to the date of clinical cutoff (approximately 60 months)

Interventionpercentage of participants (Number)
Chemoradiation + TMZ100
Chemoradiation + Bevacizumab + TMZ98.3

[back to top]

Progression Free Survival PPX/RT Versus TMZ/RT for Patients With GBM Without Methylation

"MRI response evaluated by RANO criteria~Complete Response (CR): Circumstance when the enhancing tumor is no longer seen by neuroimaging, with the patient off all steroids or on adrenal maintenance only; CR will be coded only if confirmed by a second CT/MR scan performed a minimum of 4 weeks after the initial scan coding a response.~Partial Response (PR): Decrease of > 50% in the product of two diameters. Patients should be receiving stable or decreasing doses of steroids. PR will be coded only if confirmed by a second CT/MR scan performed a minimum of 4 weeks after the initial scan.~Progression (P): A > 25% increase in tumor area (two diameters) provided that the patient has not had his/her dose of steroids decreased since the last evaluation period. This will not need a confirmatory scan. A concomitant decrease in steroid dose will rule out a progression designation during the first 2 months after completion of XRT." (NCT01402063)
Timeframe: Q 3 months on study then Q3 months in f/u for yr 1, q 4 months yr 2, q 6 months for approximately 4 ys.

Interventionparticipants (Number)
Radiation Plus PPX(CT210331
Radiation + Temozolomide15

[back to top]

Number of Patients Who Experience Toxicity Events Undergoing This Treatment.

"Safety and toxicity will be reported in the number of patients who experience adverse events during treatment, graded using CTCAE 4.03.~In general the severity of an AE is graded as follows:~Mild (grade 1): the event causes discomfort without disruption of normal daily activities.~Moderate (grade 2): the event causes discomfort that affects normal daily activities.~Severe (grade 3): the event makes the patient unable to perform normal daily activities or significantly affects his/her clinical status.~Life-threatening (grade 4): the patient was at risk of death at the time of the event. Fatal (grade 5): the event caused death." (NCT01465659)
Timeframe: During treatment and up to one month post last dose of study drug. Range of cycles completed by patients was 1-41 where one cycle =28 days.

,,,
Interventionpatients (Number)
Any AEAE related to treatmentSAESAE related to treatmentGrade 5 SAECame off treatment due to an AE
Cohort -1 -Temozolomide 100 mg/m2 and Pazopanib 400 mg553113
Cohort -2 - Temozolomide 75 mg/m2 and Pazopanib 400 mg661103
Cohort 1 - Temozolomide 150 mg/m2 and Pazopanib 400 mg773303
Phase II -Temozolomide 75 mg/m2 and Pazopanib 400 mg11111102

[back to top]

Progression Free Survival (PFS)

PFS will be defined as will be defined as the time from the first study treatment to the first occurrence of progression or death. Progressive disease will be assessed using RECIST v1.1 criteria where in general the following definition is true: At least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum recorded on study AND an absolute increase in the sum of at least 5 mm OR the appearance of one or more new lesions (NCT01465659)
Timeframe: Baseline and after every 2 cycles of treatment (8 weeks) for up to 40 months

Interventionmonths (Median)
Cohort 1/Cohort -1/Cohort -2 - Temozolomide and Pazopanib9.38

[back to top]

Determine the Maximum Tolerated Dose (MTD) of Temozolomide in Combination With 400 mg Pazopanib in Patients With Advanced Pancreatic Neuroendocrine Tumor (PNET) in Phase I

"MTD and recommended phase II dose (RP2D) determination for the combination of temozolomide in combination with 400mg pazopanib in patients with advanced PNET will be achieved using a standard 3+3 dose escalation/de-escalation design. After each 3 patients are enrolled into the study, further enrollment will be temporarily suspended until safety has been reviewed for the first 28 days of treatment to determine if dose limiting toxicities have been experienced by patients and if a further 3 patients should be enrolled at the current dose or dose escalation/de-escalation for the next 3 patients should occur." (NCT01465659)
Timeframe: After 28 days (1 cycle of treatment)

Interventionmg/m2 (Number)
Cohort 1/Cohort -1/Cohort -2 - Temozolomide and Pazopanib75

[back to top]

Plasma Temozolomide Concentration in the Blood at Various Timepoints After Administration

"For the Six Patients in Phase I portion who are enrolled in the PK cohort: Blood will be drawn on Day 1 before beginning treatment and again at 10 minutes, 30 minutes, 1, 2, 3, 4, 6 and 8 hours after beginning treatment. On Day 2- 24 hours after the first dose from Day 1, and again 10 minutes, 30 minutes, 1, 2, 3, 4, 6 and 8 hours after taking the second dose. Day 3 - 24 hours after the second dose from Day 2.~Cycle 1 Day 1 only temozolomide will be taken by the patient and on Cycle 2 Day 2, temozolomide and pazopanib will be taken by the patient. Data that was collected but not analyzed. The data that was collected is reported below in raw form." (NCT01465659)
Timeframe: Multiple timepoints during Days 1-3 of cycle 1 and cycle 2 (1 cycle =28 days)

Interventionng/mL (Number)
Patient #1 : Cycle 1 Day 1 at time 0 hoursPatient #1 : Cycle 1 Day 1 at time 0.17 hoursPatient #1 : Cycle 1 Day 1 at time 0.5 hoursPatient #1 : Cycle 1 Day 1 at time 1 hoursPatient #1 : Cycle 1 Day 1 at time 2 hoursPatient #1 : Cycle 1 Day 1 at time 3 hoursPatient #1 : Cycle 1 Day 1 at time 4 hoursPatient #1 : Cycle 1 Day 1 at time 6 hoursPatient #1 : Cycle 1 Day 1 at time 8 hoursPatient #1 : Cycle 1 Day 1 at time 24 hoursPatient #1 : Cycle 2 Day 1 at time 0 hoursPatient #1 : Cycle 2 Day 1 at time 0.17 hoursPatient #1 : Cycle 2 Day 1 at time 0.5 hoursPatient #1 : Cycle 2 Day 1 at time 1 hoursPatient #1 : Cycle 2 Day 1 at time 2 hoursPatient #1 : Cycle 2 Day 1 at time 3 hoursPatient #1 : Cycle 2 Day 1 at time 4 hoursPatient #1 : Cycle 2 Day 1 at time 6 hoursPatient #1 : Cycle 2 Day 1 at time 8 hoursPatient #1 : Cycle 2 Day 1 at time 24 hoursPatient #2 : Cycle 1 Day 1 at time 0 hoursPatient #2 : Cycle 1 Day 1 at time 0.17 hoursPatient #2 : Cycle 1 Day 1 at time 0.5 hoursPatient #2 : Cycle 1 Day 1 at time 1 hoursPatient #2 : Cycle 1 Day 1 at time 2 hoursPatient #2 : Cycle 1 Day 1 at time 3 hoursPatient #2 : Cycle 1 Day 1 at time 4 hoursPatient #2 : Cycle 1 Day 1 at time 6 hoursPatient #2 : Cycle 1 Day 1 at time 8 hoursPatient #2 : Cycle 1 Day 1 at time 24 hoursPatient #2 : Cycle 2 Day 1 at time 0 hoursPatient #2 : Cycle 2 Day 1 at time 0.17 hoursPatient #2 : Cycle 2 Day 1 at time 0.5 hoursPatient #2 : Cycle 2 Day 1 at time 1 hoursPatient #2 : Cycle 2 Day 1 at time 2 hoursPatient #2 : Cycle 2 Day 1 at time 3 hoursPatient #2 : Cycle 2 Day 1 at time 4 hoursPatient #2 : Cycle 2 Day 1 at time 6 hoursPatient #2 : Cycle 2 Day 1 at time 8 hoursPatient #2 : Cycle 2 Day 1 at time 24 hoursPatient #3 : Cycle 1 Day 1 at time 0 hoursPatient #3 : Cycle 1 Day 1 at time 0.17 hoursPatient #3 : Cycle 1 Day 1 at time 0.5 hoursPatient #3 : Cycle 1 Day 1 at time 1 hoursPatient #3 : Cycle 1 Day 1 at time 2 hoursPatient #3 : Cycle 1 Day 1 at time 3 hoursPatient #3 : Cycle 1 Day 1 at time 4 hoursPatient #3 : Cycle 1 Day 1 at time 6 hoursPatient #3 : Cycle 1 Day 1 at time 8 hoursPatient #3 : Cycle 1 Day 1 at time 24 hoursPatient #3 : Cycle 2 Day 1 at time 0 hoursPatient #3 : Cycle 2 Day 1 at time 0.17 hoursPatient #3 : Cycle 2 Day 1 at time 0.5 hoursPatient #3 : Cycle 2 Day 1 at time 1 hoursPatient #3 : Cycle 2 Day 1 at time 2 hoursPatient #3 : Cycle 2 Day 1 at time 3 hoursPatient #3 : Cycle 2 Day 1 at time 4 hoursPatient #3 : Cycle 2 Day 1 at time 6 hoursPatient #3 : Cycle 2 Day 1 at time 8 hoursPatient #3 : Cycle 2 Day 1 at time 24 hoursPatient #4 : Cycle 1 Day 1 at time 0 hoursPatient #4 : Cycle 1 Day 1 at time 0.17 hoursPatient #4 : Cycle 1 Day 1 at time 0.5 hoursPatient #4 : Cycle 1 Day 1 at time 1 hoursPatient #4 : Cycle 1 Day 1 at time 2 hoursPatient #4 : Cycle 1 Day 1 at time 3 hoursPatient #4 : Cycle 1 Day 1 at time 4 hoursPatient #4 : Cycle 1 Day 1 at time 6 hoursPatient #4 : Cycle 1 Day 1 at time 8 hoursPatient #4 : Cycle 1 Day 1 at time 24 hoursPatient #4 : Cycle 2 Day 1 at time 0 hoursPatient #4 : Cycle 2 Day 1 at time 0.17 hoursPatient #4 : Cycle 2 Day 1 at time 0.5 hoursPatient #4 : Cycle 2 Day 1 at time 1 hoursPatient #4 : Cycle 2 Day 1 at time 2 hoursPatient #4 : Cycle 2 Day 1 at time 3 hoursPatient #4 : Cycle 2 Day 1 at time 4 hoursPatient #4 : Cycle 2 Day 1 at time 6 hoursPatient #4 : Cycle 2 Day 1 at time 8 hoursPatient #4 : Cycle 2 Day 1 at time 24 hoursPatient #5 : Cycle 1 Day 1 at time 0 hoursPatient #5 : Cycle 1 Day 1 at time 0.17 hoursPatient #5 : Cycle 1 Day 1 at time 0.5 hoursPatient #5 : Cycle 1 Day 1 at time 1 hoursPatient #5 : Cycle 1 Day 1 at time 2 hoursPatient #5 : Cycle 1 Day 1 at time 3 hoursPatient #5 : Cycle 1 Day 1 at time 4 hoursPatient #5 : Cycle 1 Day 1 at time 6 hoursPatient #5 : Cycle 1 Day 1 at time 8 hoursPatient #5 : Cycle 1 Day 1 at time 24 hoursPatient #5 : Cycle 2 Day 1 at time 0 hoursPatient #5 : Cycle 2 Day 1 at time 0.17 hoursPatient #5 : Cycle 2 Day 1 at time 0.5 hoursPatient #5 : Cycle 2 Day 1 at time 1 hoursPatient #5 : Cycle 2 Day 1 at time 2 hoursPatient #5 : Cycle 2 Day 1 at time 3 hoursPatient #5 : Cycle 2 Day 1 at time 4 hoursPatient #5 : Cycle 2 Day 1 at time 6 hoursPatient #5 : Cycle 2 Day 1 at time 8 hoursPatient #5 : Cycle 2 Day 1 at time 24 hoursPatient #6a : Cycle 1 Day 1 at time 0 hoursPatient #6a : Cycle 1 Day 1 at time 0.17 hoursPatient #6a : Cycle 1 Day 1 at time 0.5 hoursPatient #6a : Cycle 1 Day 1 at time 1 hoursPatient #6a : Cycle 1 Day 1 at time 2 hoursPatient #6a : Cycle 1 Day 1 at time 3 hoursPatient #6a : Cycle 1 Day 1 at time 4 hoursPatient #6a : Cycle 1 Day 1 at time 6 hoursPatient #6a : Cycle 1 Day 1 at time 8 hoursPatient #6a : Cycle 1 Day 1 at time 24 hoursPatient #6a : Cycle 2 Day 1 at time 0 hoursPatient #6a : Cycle 2 Day 1 at time 0.17 hoursPatient #6a : Cycle 2 Day 1 at time 0.5 hoursPatient #6a : Cycle 2 Day 1 at time 1 hoursPatient #6a : Cycle 2 Day 1 at time 2 hoursPatient #6a : Cycle 2 Day 1 at time 3 hoursPatient #6a : Cycle 2 Day 1 at time 4 hoursPatient #6a : Cycle 2 Day 1 at time 6 hoursPatient #6a : Cycle 2 Day 1 at time 8 hoursPatient #6a : Cycle 2 Day 1 at time 24 hours
Temozolomide 75 mg/m2 and Pazopanib 400 mg0091.8425.5462583210.51054400025.290.8516.525551915499.900054.9494660.5557.599105.5109.500041.61151490961.5382.5510.5109.500052716702440202414667183174.34.124190921302490193315867613284.7503.64923025280517651430686.53831.22.42.33.01.61.0123360261012253285019.1177227275829.59251145819.544.20108166.5188.5422.5716.510701180744.536.70459.530552720173514701195499.52511.5067.740952135209514451025.5324.5192.51.3

[back to top]

Overall Survival (OS)

OS is defined as the time from first study treatment until death from any cause. (NCT01465659)
Timeframe: Baseline and after every 2 cycles of treatment (8 weeks) and up to 60 months

Interventionmonths (Median)
Cohort 1/Cohort -1/Cohort -2 - Temozolomide and Pazopanib24.2

[back to top]

Overall Response Rate (ORR) in Patients With Advanced Neuroendocrine Tumors (PNET) Treated With Temozolomide and Pazopanib Combination Treatment at the RP2D in Phase II

"Overall response rate will be determined by the number of patients who's best response as assessed by RECIST 1.1 is complete response (CR) and partial response (PR) in patients with PNET that are enrolled at the recommended phase II dose (RP2D) (PK cohort included).~CR= Disappearance of all target lesions PR = At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters." (NCT01465659)
Timeframe: After two cycles of treatment (8 weeks)

InterventionParticipants (Count of Participants)
Phase II -Temozolomide 75 mg/m2 and Pazopanib 400 mg0

[back to top]

Number of Patients Experiencing Response to Treatment or Stable Disease (Disease Control Rate)

"Disease Control Rate (DCR) is defined as the number of patients demonstrating the complete response, partial response or stable disease.~In general the following is true:~Complete Response (CR) Disappearance of all target lesions. Disappearance of all non-target lesions and normalization of tumor marker level.~Partial Response (PR) At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters Stable Disease (SD) Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum of diameters while on study" (NCT01465659)
Timeframe: After every 2 courses of treatment (8 weeks) for up to 41 cycles where 1 cycle =28 days.

InterventionParticipants (Count of Participants)
Cohort 1/Cohort -1/Cohort -2 - Temozolomide and Pazopanib20

[back to top]

Overall Survival Rate

Overall survival rate is calculated as the median number of months that patients were alive for the cohort (NCT01466686)
Timeframe: up to 12 months after completion of temozolomide (48 weeks of treatment)

Interventionmonths (Median)
Temozolomide With Low Dose Fractionated Radiation Therapy9.6

[back to top]

Number of Patients With Neurologic Toxicity

The number of patients with reported grade 3+ neurologic toxicities (NCT01466686)
Timeframe: Approximately every month from study start until 48 weeks, and then up to 12 months after completion of temozolomide at 3, 6, and 12 months follow up

InterventionParticipants (Count of Participants)
Temozolomide With Low Dose Fractionated Radiation Therapy6

[back to top]

Number of Patients With Hematologic Toxicities

The number of patients with grade 3+ hematologic toxicities. (NCT01466686)
Timeframe: Approximately every month from study start until 48 weeks, and then up to 12 months after completion of temozolomide at 3, 6, and 12 months follow up

InterventionParticipants (Count of Participants)
Temozolomide With Low Dose Fractionated Radiation Therapy8

[back to top]

Progression Free Survival Rate

Progression free survival rate is calculated as the median number of months for the cohort until patient's disease worsened/progressed (NCT01466686)
Timeframe: up to 12 months after completion of temozolomide (48 weeks of treatment)

Interventionmonths (Median)
Temozolomide With Low Dose Fractionated Radiation Therapy7.3

[back to top]

Safety Profile for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab

"Toxicity will be assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 4.0 criteria on Day 1 of every treatment cycle and by patient report while on study treatment and up to 30 days after the last treatment. Grade 1 - 4 adverse events (AE) where the relationship between the AE and at least one of the study drugs were considered to be definite, probable or possible, were collected and graded as:~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" (NCT01478321)
Timeframe: Completed weekly during initial phase of 5 weeks and 2 weeks recovery, then every cycle during adjuvant therapy where 1 cycle =8 weeks (for up to 7 cycles)

Interventionparticipants (Number)
Lymphopenia : Grade 1Lymphopenia : Grade 2Lymphopenia : Grade 3Lymphopenia : Grade 4Fatigue : Grade 1Fatigue : Grade 2Fatigue : Grade 3Fatigue : Grade 4Thrombocytopenia : Grade 1Thrombocytopenia : Grade 2Thrombocytopenia : Grade 3Thrombocytopenia : Grade 4Anemia : Grade 1Anemia : Grade 2Anemia : Grade 3Anemia : Grade 4Constipation : Grade 1Constipation : Grade 2Constipation : Grade 3Constipation : Grade 4Hypertension : Grade 1Hypertension : Grade 2Hypertension : Grade 3Hypertension : Grade 4Neutropenia : Grade 1Neutropenia : Grade 2Neutropenia : Grade 3Neutropenia : Grade 4Epistaxis : Grade 1Epistaxis : Grade 2Epistaxis : Grade 3Epistaxis : Grade 4Thromboembolism : Grade 1Thromboembolism : Grade 2Thromboembolism : Grade 3Thromboembolism : Grade 4Proteinuria : Grade 1Proteinuria : Grade 2Proteinuria : Grade 3Proteinuria : Grade 4Wound Complication : Grade 1Wound Complication : Grade 2Wound Complication : Grade 3Wound Complication : Grade 4
Treatment (Radiation, Chemotherapy, Monoclonal Antibody)412419510812171108100061023003000011000200020

[back to top]

Overall Survival (OS) for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab.

Data will be analyzed using Kaplan-Meier curves. OS is defined as the time from first re-irradiation treatment until death from any cause. (NCT01478321)
Timeframe: From treatment initiation and every 8 weeks for up to 53.5 months

InterventionMonths (Median)
Treatment (Radiation, Chemotherapy, Monoclonal Antibody)8.5

[back to top]

Median Progression Free Survival (PFS) for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab

Progression Free Survival (PFS) is defined as the time from the first study treatment to the first occurrence of disease progression or death. Data will be analyzed using Kaplan-Meier curves. Tumor measurements and assessments will be based on Updated Response Assessment Criteria of High Grade Gliomas- Neuro-Oncology Working Group (RANO criteria). Tumor assessments may include either a CT or MRI scan of the brain, however the same method should be used throughout the treatment period for each patient. In general, progressive disease is defined as any of the following: ≥ 25% increase in T1 gadolinium enhancing disease, increase in T2/Flair, new lesions present or decrease in clinical status. (NCT01478321)
Timeframe: Range from treatment initiation 0.4-26.9 months

InterventionMonths (Median)
Treatment (Radiation, Chemotherapy, Monoclonal Antibody)5.5

[back to top]

Percentage of Patients With Progression Free Survival (PFS) at 6 Months and 12 Months

Progression Free Survival is defined as the time from the first study treatment to the first occurrence of disease progression or death. Data will be analyzed using Kaplan-Meier curves. Tumor measurements and assessments will be based on Updated Response Assessment Criteria of High Grade Gliomas- Neuro-Oncology Working Group (RANO criteria). Tumor assessments may include either a CT or MRI scan of the brain, however the same method should be used throughout the treatment period for each patient. In general, progressive disease is defined as any of the following: ≥ 25% increase in T1 gadolinium enhancing disease, increase in T2/Flair, new lesions present or decrease in clinical status (NCT01478321)
Timeframe: At 6 and 12 months after the start of treatment

Interventionpercentage of patients with PFS (Number)
6 Months12 Months
Treatment (Radiation, Chemotherapy, Monoclonal Antibody)4812

[back to top]

Patient Reported Quality of Life (QOL)

"Questionnaires were completed before treatment (baseline) at the end of treatment (EOT) and after Cycle 1 and Cycle 2 of treatment (Initial phase of treatment =5 weeks + approximately 2 weeks recovery then adjuvant therapy where 1 cycle = 8 weeks). The following questionnaires were completed by patients to evaluate quality of life (QOL) at these timepoints:~FACT-Fatigue - scores from 1 to 4 with 1=not at all and 4=very much, the higher the score the more fatigue reported by the patient.~FACT-Brain (FACT Br) which included - Physcial Well-being (PWB), Social/Family Well-being (SWB), Emotional Well-being (EWB), and Functional Well-being (FWB).~Patients gave scores from 0 to 4 with 0=not at all and 4=very much, the higher the score the better the QOL reported by the patient." (NCT01478321)
Timeframe: Completed before treatment (baseline) after Cycle 1 (approximately week 15) and Cycle 2 (approximately week 23)of adjuvant treatment and at the end of treatment (up to 7 cycles of adjuvant treatment, where 1 cycle =8 weeks)

Interventionscore on a scale (Mean)
FACT-BR Total : BaselineFACT-BR Total : EOTFACT-BR Total : Post Cycle 1FACT-BR Total : Post Cycle 2FACT-BR PWB : BaselineFACT-BR PWB : EOTFACT-BR PWB : Post Cycle 1FACT-BR PWB : Post Cycle 2FACT-BR SWB : BaselineFACT-BR SWB : EOTFACT-BR SWB : Post Cycle 1FACT-BR SWB : Post Cycle 2FACT-BR EWB : BaselineFACT-BR EWB : EOTFACT-BR EWB : Post Cycle 1FACT-BR EWB : Post Cycle 2FACT-FWB : BaselineFACT-FWB : EOTFACT-FWB : Post Cycle 1FACT-FWB: Post Cycle 2FACIT-Fatigue: BaselineFACIT-Fatigue: EOTFACIT-Fatigue: Post Cycle 1FACIT-Fatigue: Post Cycle 2
Treatment (Radiation, Chemotherapy, Monoclonal Antibody)130.4106.9133.0131.422.021.621.721.122.222.923.023.715.916.517.217.715.917.817.018.335.630.334.436.2

[back to top]

Overall Survival (OS) at 6 Months and 12 Months for Patients With Recurrent High Grade Malignant Gliomas Treated With Concurrent Radiation, Temozolomide, and Bevacizumab Followed by Adjuvant Temozolomide and Bevacizumab.

Data will be analyzed using Kaplan-Meier curves. OS is defined as the time from first re-irradiation treatment until death from any cause. Percentages of patients alive at that 6 months and 12 months will be calculated from the Kaplan-Meier curve. (NCT01478321)
Timeframe: At 6 and 12 months from start of treatment

Interventionpercentage of patients alive (Number)
6 Months12 Months
Treatment (Radiation, Chemotherapy, Monoclonal Antibody)6728

[back to top]

Overall Response Rate (ORR) of Participants Using RECIST Criteria

"Phase I portion of trial- All patients enrolled to recieve TPI+I+TMZ. These patients will be added to the Phase II patients that were randomized to Arm B- Arm with TPI 287 (recieved same tx as Phase I participatns).~Phase II portion of trial- TPatients enrolled to this portion (different patients than enrolled to Phase 1) were randomized to Arm A: I+TMZ OR Arm B: TPI+I+TMZ 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." (NCT01505608)
Timeframe: 3 years

Interventionparticipants (Number)
Arm A- Temozolomide and Irinotecan0
Phase I Patients + Phase II Assigned Arm B- Tmz +I+ TPI 2872

[back to top]

Number of Participants With Adverse Events as a Measure of Safety and Tolerability

"To determine the safety and tolerability of TPI 287 in combination with Irinotecan and Temozolomide (TPI+I+TMZ) in pediatric and young adult patients with primary refractory or recurrent Neuroblastoma.~Phase I patients were all enrolled to receive TPI 287. Phase 2 is where randomization began. Patients were different patients than the Phase 1 patients. Below all patients that received TPI 287 are included in the TPI 287 group. This includes Phase I patients and the Phase 2 patients randomized to TPI 287." (NCT01505608)
Timeframe: 6 months

Interventionparticipants (Number)
TPI 2877
Arm A- TMZ + Irinotecan Only0

[back to top]

Progression Free Survival (PFS) of Participants Using Days Until Progression

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 (NCT01505608)
Timeframe: 3 years

InterventionDays (Number)
Arm A- Temozolomide and Irinotecan22
Arm B- Temozolomide/Irinotecan + TPI 287125

[back to top]

Progression-Free Survival (PFS)

PFS is defined as the number of months from the date the participant was randomized to the date of radiographic progression as determined by the central imaging center, or to the date of all cause deaths within 63 days of last tumor assessment if disease progression was not reached. (NCT01506609)
Timeframe: Radiographic evaluation every 9 weeks, clinical evaluation every cycle (data cutoff date: 04 March 2016); maximum duration of follow up for PFS was 34 months.

Interventionmonths (Median)
Group 2 Placebo + Carboplatin/Paclitaxel12.3
Group 2 Veliparib + Carboplatin/Paclitaxel14.1
Group 2 Veliparib + TMZ7.4

[back to top]

Clinical Benefit Rate (CBR) at Week 18

"CBR: percentage of participants who were progression-free at 18 weeks, defined as complete response (CR), partial response (PR), stable disease (SD) or non-CR/non-disease progression (PD) per Response Evaluation Criteria in Solid Tumors [RECIST] 1.1.~CR: The disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 0 mm. PR: >= 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters (SOD). PD: >= 20% increase in the SOD of target lesions, taking as reference the smallest SOD recorded since the treatment started (baseline or after) or the appearance of >=1 new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest SOD since the treatment started (baseline or after)." (NCT01506609)
Timeframe: Week 18

Interventionpercentage of participants (Number)
Group 2 Placebo + Carboplatin/Paclitaxel87.0
Group 2 Veliparib + Carboplatin/Paclitaxel90.7
Group 2 Veliparib + TMZ73.0

[back to top]

Overall Survival (OS)

Time to death for a given participant was defined as the number of months from the day the participant is randomized to the date of the participant's death. All events of death were included, regardless of whether the event occurs while the participant was still taking study drug, or after the participant discontinued study drug. If a participant had not died, then the data will be censored at the date when the participant was last known to be alive. (NCT01506609)
Timeframe: From Cycle 1 Day 1 until participant's death or 3 years post discontinuation (data cutoff date: 04 March 2016); maximum duration of follow up for OS was 72 months.

Interventionmonths (Median)
Group 2 Placebo + Carboplatin/Paclitaxel25.4
Group 2 Veliparib + Carboplatin/Paclitaxel28.3
Group 2 Veliparib + TMZ19.1

[back to top]

Objective Response Rate (ORR)

The objective response rate, defined as percentage of participants with a confirmed CR or PR based on RECIST 1.1 criteria. CR: The disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 0 mm. PR: >= 30% decrease in the sum of diameters of target lesions, taking as reference the baseline SODs. (NCT01506609)
Timeframe: Radiographic evaluation every 9 weeks, clinical evaluation every cycle (data cutoff date: 04 March 2016); maximum duration of follow up for ORR was 34 months.

Interventionpercentage of participants (Number)
Group 2 Placebo + Carboplatin/Paclitaxel61.3
Group 2 Veliparib + Carboplatin/Paclitaxel77.8
Group 2 Veliparib + TMZ28.6

[back to top]

Change From Baseline at Week 18 in European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Chemotherapy-Induced Peripheral Neuropathy Module (EORTC QLQ-CIPN20) Sensory Subscale Score

EORTC QLQ-CIPN20 sensory subscale score was calculated following the standard scoring algorithm, transformed to a 0 (low quality of life) to 100 (best quality of life) scale. A positive change from baseline indicates improvement. (NCT01506609)
Timeframe: Baseline, Week 18

Interventionscore on a scale (Mean)
Group 2 Placebo + Carboplatin/Paclitaxel13.94
Group 2 Veliparib + Carboplatin/Paclitaxel11.24

[back to top]

Overall Survival

Overall survival was defined as the interval from date on treatment to date of death from any cause or to date of last follow-up. Patients who had not failed (died) at the time of analyses were censored at their last date of contact. The method of Kaplan and Meier was used to estimate overall survival. The 3-year estimate with a 95% confidence interval is reported. (NCT01514201)
Timeframe: Time from initiation of therapy to the date of death from any cause or to the date patient was known to be alive for surviving patients, assessed to up to 3 years

InterventionPercent probability (Number)
Phase II Patients + Phase I MTD Patients5.3

[back to top]

Trough for Veliparib [Pharmacokinetic Parameter]

During course 1, blood samples were collected pre-veliparib on day 1, at 0.5, 1, 2, and 6-8 hours after the first dose, pre-veliparib on day 4 (steady state), and 2 hours after the morning dose. Veliparib concentrations were measured using a liquid chromatography tandem mass spectrometry assay and pharmacokinetic parameters were evaluated using a non-compartmental analysis. (NCT01514201)
Timeframe: Up to day 4

Interventionng/mL (Mean)
Phase I, Dose Level 1 (50 mg)58
Phase I, Dose Level 2 (65 mg)140
Phase I, Dose Level 3 (85 mg)163
Phase II (MTD)84

[back to top]

Levels of Urinary Biomarkers

Urine samples were analyzed for a panel of biomarkers. Netrin-1 levels were determined by ELISA. Levels of matrix metalloproteinase 3 (MMP3) and basic fibroblast growth factor (bFGF) were analyzed using custom Luminex® screening assays. Tissue inhibitor of metalloproteinase 1 (TIMP1) levels were analyzed using a Luminex® performance assay. Protein concentrations are given in picograms per microgram (pg/μg), and were determined by dividing the concentration of the target protein in the sample (pg/mL) by the concentration of total protein in the sample (μg/mL) as a normalization measure. (NCT01514201)
Timeframe: Baseline to up to 3 years

,,,
Interventionpg/μg (Median)
MMP3 at pre-studyMMP3 at week 10-11MMP3 at week 18MMP3 at week 26Netrin-1 at pre-studyNetrin-1 at week 10-11Netrin-1 at week 18Netrin-1 at week 26TIMP1 at pre-studyTIMP1 at week 10-11TIMP1 at week 18TIMP1 at week 26bFGF at pre-studybFGF at week 10-11bFGF at week 18bFGF at week 26
Phase I, Dose Level 1 (50 mg)2.01.44.32.90.10.10.30.43.46.210.77.33.13.610.37.7
Phase I, Dose Level 2 (65 mg)1.01.01.70.40.10.10.10.29.911.97.75.21.92.13.50.9
Phase I, Dose Level 3 (85 mg)0.00.80.71.00.10.10.00.010.812.314.832.81.21.81.31.1
Phase II (MTD)1.12.62.33.00.10.00.10.07.37.57.110.74.53.54.44.5

[back to top]

Maximum Concentration of Veliparib (Cmax) on Days 1 and 4 (Measured in ng/mL) [Pharmacokinetic Parameter]

During course 1, blood samples were collected pre-veliparib on day 1, at 0.5, 1, 2, and 6-8 hours after the first dose, pre-veliparib on day 4 (steady state), and 2 hours after the morning dose. Veliparib concentrations were measured using a liquid chromatography tandem mass spectrometry assay and pharmacokinetic parameters were evaluated using a non-compartmental analysis. Cmax measures the highest concentration of drug. (NCT01514201)
Timeframe: Up to day 4

,,,
Interventionng/mL (Mean)
Day 1, Cmax (ng/mL)Day 4, Cmax (ng/mL)
Phase I, Dose Level 1 (50 mg)519409
Phase I, Dose Level 2 (65 mg)843788
Phase I, Dose Level 3 (85 mg)1074954
Phase II (MTD)844717

[back to top]

Percentage of Participants With Significant Changes in Poly(ADP-ribose) Polymerase (PARP) Levels Post-Veliparib, as Measured in Peripheral Blood Monocytes (PBMCs)

Blood samples were collected from patients and assessed pre- and post-Veliparib to assess treatment-induced changes. A significant change in PBMC PARP level was arbitrarily defined as a >50% increase or decrease from the pre-treatment level, documented at week 6 and/or week 11 after starting protocol therapy. (NCT01514201)
Timeframe: Baseline and up to 11 weeks

Interventionpercentage of participants (Number)
Phase I, Dose Level 1 (50 mg)100
Phase I, Dose Level 2 (65 mg)100
Phase I, Dose Level 3 (85 mg)75
Phase II (MTD)36

[back to top]

Percentage of Participants Observed to Have Unacceptable Toxicity During the Intra-patient Dose Escalation of Temozolomide During Maintenance Therapy (Feasibility Analysis Population)

Unacceptable toxicities during maintenance included events at least possibly attributable to Veliparib and temozolomide (TMZ) such as any grade 4 non-hematologic toxicity, any grade 3 non-hematologic toxicity with some exceptions (e.g., grade 3 nausea/vomiting <5 days, grade 3 fever or infection <5 days), grade 3+ thrombocytopenia, grade 4 neutropenia, delay >14 days in starting subsequent cycle due to neutrophil <1,000/mm3 or platelet <100,000/mm3. Maintenance therapy was initiated with 25 mg/m2 Veliparib and 135 mg/m2 of TMZ, with the possibility to escalate TMZ to 175 mg/m2 and 200 mg/m2 in courses 2 and 3, respectively, if no unacceptable toxicities occurred following one course of treatment at each of the dose levels to be tested. Intra-patient dose escalation to a given dose (135, 175, or 200 mg/m2) was halted based on rules employed in 3+3 designs. This dose escalation was intended for all patients but was halted early, during the phase I portion, as it was not well tolerated. (NCT01514201)
Timeframe: 28 days per treatment cycle

Intervention% of participants (Number)
Dose Level 1 (135 mg/m2)9
Dose Level 2 (175 mg/m2)40
Dose Level 3 (200 mg/m2)67

[back to top]

Terminal Half-life (t1/2) for Veliparib [Pharmacokinetic Parameter]

During course 1, blood samples were collected pre-veliparib on day 1, at 0.5, 1, 2, and 6-8 hours after the first dose, pre-veliparib on day 4 (steady state), and 2 hours after the morning dose. Veliparib concentrations were measured using a liquid chromatography tandem mass spectrometry assay and pharmacokinetic parameters were evaluated using a non-compartmental analysis. (NCT01514201)
Timeframe: Up to day 4

InterventionHour (Mean)
Phase I, Dose Level 1 (50 mg)5.18
Phase I, Dose Level 2 (65 mg)2.62
Phase I, Dose Level 3 (85 mg)4.45
Phase II (MTD)2.18

[back to top]

Mean Apparent Clearance (CL/F) for Veliparib [Pharmacokinetic Parameter]

During course 1, blood samples were collected pre-veliparib on day 1, at 0.5, 1, 2, and 6-8 hours after the first dose, pre-veliparib on day 4 (steady state), and 2 hours after the morning dose. Veliparib concentrations were measured using a liquid chromatography tandem mass spectrometry assay and pharmacokinetic parameters were evaluated using a non-compartmental analysis. (NCT01514201)
Timeframe: Up to day 4

InterventionL/m^2/h (Mean)
Phase I, Dose Level 1 (50 mg)16.1
Phase I, Dose Level 2 (65 mg)13.2
Phase I, Dose Level 3 (85 mg)15.8
Phase II (MTD)11.7

[back to top]

Progression-free Survival (PFS)

PFS was defined as the interval from date of treatment initiation to date of first event (disease progression or relapse, second malignancy or death from any cause). Patients who had not failed at the time of analyses were censored at their last date of contact. The method of Kaplan and Meier was used to estimate PFS. A 3-year estimate with a 95% confidence interval is reported. (NCT01514201)
Timeframe: Time from initiation of treatment to the earliest date of failure (disease progression, death from any cause, or second malignancy), assessed up to 3 years

InterventionPercent probability (Number)
Phase II Patients + Phase I MTD Patients2.9

[back to top]

Percentage of Patients With Pseudo Progression

For participants that showed possible tumor progression (pseudo progression) on magnetic resonance imaging (MRI) during the first 6 months of therapy, treating physicians had the option of allowing patients to remain on therapy and repeating the disease assessment in 4-6 weeks. If the repeat MRI at 4-6 weeks showed disease progression, the patient was noted to have true disease progression (and the progression date corresponded to that of the first MRI). If the repeat MRI at 4-6 weeks did not show disease progression, then the patient was noted to have pseudo progression. The percentage of patients observed to have experienced pseudo progression was provided with a 95% confidence interval. (NCT01514201)
Timeframe: Up to 6 months

InterventionPercentage of participants (Number)
Phase I, Dose Level 1 (50 mg)33.3
Phase I, Dose Level 2 (65 mg)16.7
Phase I, Dose Level 3 (85 mg)0
Phase II (MTD)12.8

[back to top]

Apparent Volume of Distribution (Vd/F) for Veliparib [Pharmacokinetic Parameter]

During course 1, blood samples were collected pre-veliparib on day 1, at 0.5, 1, 2, and 6-8 hours after the first dose, pre-veliparib on day 4 (steady state), and 2 hours after the morning dose. Veliparib concentrations were measured using a liquid chromatography tandem mass spectrometry assay and pharmacokinetic parameters were evaluated using a non-compartmental analysis. (NCT01514201)
Timeframe: Up to day 4

InterventionL/m^2 (Mean)
Phase I, Dose Level 1 (50 mg)75.4
Phase I, Dose Level 2 (65 mg)56.1
Phase I, Dose Level 3 (85 mg)63.9
Phase II (MTD)73.1

[back to top]

Maximum Concentration of Veliparib (Cmax) on Day 1 (Measured in μM) [Pharmacokinetic Parameter]

During course 1, blood samples were collected pre-veliparib on day 1, at 0.5, 1, 2, and 6-8 hours after the first dose, pre-veliparib on day 4 (steady state), and 2 hours after the morning dose. Veliparib concentrations were measured using a liquid chromatography tandem mass spectrometry assay and pharmacokinetic parameters were evaluated using a non-compartmental analysis. Cmax measures the highest concentration of drug. (NCT01514201)
Timeframe: Day 1

InterventionμM (Mean)
Phase I, Dose Level 1 (50 mg)2.12
Phase I, Dose Level 2 (65 mg)3.45
Phase I, Dose Level 3 (85 mg)4.40
Phase II (MTD)3.45

[back to top]

Maximum-tolerated Dose of Veliparib Defined as Highest Dose Level With Fewer Than 2 Dose Limiting Toxicities in 6 Patients as Assessed by the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (Phase I)

The traditional 3+3 dose finding algorithm was used to estimate the maximum-tolerated dose of veliparib given concurrently with radiation therapy. The dose-limiting toxicity observation period was the first 10 weeks of therapy. Dose-limiting toxicities included any grade 4 non-hematologic toxicity, any grade 3 non-hematologic toxicity with a few exceptions (see section 5.2.1.2 of the protocol document), any grade 2 non-hematologic toxicity that persisted for >7 days and considered medically significant that required treatment interruption; grade 3 or higher thrombocytopenia or grade 4 neutropenia; and any Veliparib related adverse event that led to a dose reduction or the permanent cessation of therapy. (NCT01514201)
Timeframe: 10 weeks

Interventionmg/m2/dose BID (Number)
Phase I Patients65

[back to top]

Number of Phase I Patients Who Experienced Dose Limiting Toxicities (DLTs)

DLTs were defined as any of the following adverse events that were at least possibly attributable to Veliparib observed during the dose finding phase (the first 10 weeks of therapy). Hematologic dose limiting toxicities included grade 3 and higher thrombocytopenia or grade 4 neutropenia. Non-hematologic dose limiting toxicities included any grade 4 non-hematologic toxicity, any grade 3 non-hematologic toxicity with some exceptions (e.g., nausea and vomiting of <5 days; fever or infection of <5 days; hypophosphatemia, hypokalemia, hypocalcemia or hypomagnesemia responsive to oral supplementation; elevation of transaminases that return to levels meeting eligibility criteria within 7 days), or any grade non-hematologic toxicity that persisted for >7 days and considered medically significant or sufficiently intolerable by patients that required treatment interruption. (NCT01514201)
Timeframe: 10 weeks

InterventionParticipants (Count of Participants)
Phase I, Dose Level 1 (50 mg)1
Phase I, Dose Level 2 (65 mg)0
Phase I, Dose Level 3 (85 mg)3

[back to top] [back to top]

O6-methylguanine DNA Methyltransferase (MGMT) Status by Immunohistochemistry (IHC)

"O6-alkylguanine DNA alkyltransferase is a protein that in humans is encoded by the gene O6-methylguanine DNA methyltransferase (MGMT). Deficiency of the gene product, ie, the protein, is refereed to as MGMT deficiency, and is thought to be predictive of response to temozolomide and is more often associated with pancreatic NETs.~MGMT status in pre-treatment biopsy specimens was to be assessed by immuno-histochemistry (IHC), and associated to best clinical response. The outcome is reported by the the number of patients that were IHC-positive (MGMT detected) or IHC-negative (MGMT not detected), and by best clinical response [ie, complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD)]. The outcome is reported as a number without dispersion." (NCT01525082)
Timeframe: 18 months

Interventionparticipants (Number)
IHC-positive and CRIHC-positive and PRIHC-positive and SDIHC-positive and PDIHC-negative and CRIHC-negative and PRIHC-negative and SDIHC-negative and PD
Bevacizumab + Capecitabine + Temozolomide02210420

[back to top]

O6-methylguanine DNA Methyltransferase (MGMT) by Promoter Methylation

"O6-alkylguanine DNA alkyltransferase is a protein that in humans is encoded by the gene O6-methylguanine DNA methyltransferase (MGMT). Deficiency of the gene product, ie, the protein, is refered to as MGMT deficiency, and is thought to be predictive of response to temozolomide and is more often associated with pancreatic NETs.~MGMT status in pre-treatment biopsy specimens was to be assessed by extent of genetic promoter methylation (an analysis of DNA), and correlated to subject survival.~MGMT status in pre-treatment biopsy specimens was to be assessed by promoter methylation assessment (PM), and associated to best clinical response. The outcome is reported by the the number of patients that were PM-positive (MGMT detected) or PM-negative (MGMT not detected), and by best clinical response [ie, complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD)]. The outcome is reported as a number without dispersion." (NCT01525082)
Timeframe: 18 months

Interventionparticipants (Number)
PM-positive and CRPM-positive and PRPM-positive and SDPM-positive and PDPM-negative and CRPM-negative and PRPM-negative and SDPM-negative and PD
Bevacizumab + Capecitabine + Temozolomide01000451

[back to top]

Progression-free Survival (PFS)

Progression-free survival (PFS) means the length of time that participants survive without disease (tumor) progression, and was assessed using Kaplan-Meier analysis. The outcome is given as the mean in months with standard error of the mean. (NCT01525082)
Timeframe: 82 months

Interventionmonths (Mean)
Bevacizumab + Capecitabine + Temozolomide25.2

[back to top]

Overall Survival (OS)

"Time from the date of enrollment to the date of death due to any cause or the last date the patient was known to be alive (censored observation) at the date of data cutoff for the final analysis. The results will be reported as median in months~The length of overall survival (OS) of participants was assessed by Kaplan-Meier analysis. The outcome is given as the median in months with full range." (NCT01525082)
Timeframe: 82 mo

Interventionmonths (Median)
Bevacizumab + Capecitabine + Temozolomide49.8

[back to top]

Radiographic Response (RR)

"Tumor response to treatment with the combination of capecitabine & temozolomide plus bevacizumab in patients with metastatic or unresectable pancreatic neuroendocrine tumors was assessed per Response Evaluation Criteria in Solid Tumors Criteria (RECIST v1.1) for target lesions. Response is defined as:~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~Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions, and/or the appearance of one or more new lesion(s)~Stable disease (SD) = Small changes that do not meet any of the above criteria~The outcome is reported as the number of participants who between 3 and 18 months after treatment initiation achieve CR, PR, or clinical response (PR + CR), each a number without dispersion." (NCT01525082)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Overall response (OR)Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive disease (PD)
Bevacizumab + Capecitabine + Temozolomide90991

[back to top]

Treatment Failure (TF)

"Treatment failure (TF) is defined as failing to achieve either a complete remission (CR) or partial remission (PR) after induction chemotherapy. The outcome is reported as the percentage without dispersion of participants in Groups 1 and 2 that experienced TF. CR and PR are defined as the following.~CR = < 5% blasts in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods; no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL~PR = 5% to 25% blasts (must be ≥ 50% reduction of blasts) in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL" (NCT01550224)
Timeframe: up to 10 weeks

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)3
Participant Group 2 (Non-methylated MGMT Promoter)20

[back to top]

Relapse-Free Survival (RFS) at 2 Years

"Relapse-free survival (RFS) is defined as survival after complete response (CR) or (PR) without further disease progression. RFS is reported as the percentage without dispersion of participants in Groups 1 and 2 that experienced CR or PR, and were alive without progression 2 years after induction chemotherapy. CR and PR are defined as the following.~CR = < 5% blasts in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods; no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL~PR = 5% to 25% blasts (must be ≥ 50% reduction of blasts) in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL" (NCT01550224)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)0

[back to top]

Partial Remission (PR)

"Partial remission (PR) is reported as the percentage without dispersion of participants in Groups 1 and 2 that demonstrated PR. PR is defined as all of the following.~PR = 5% to 25% blasts (must be ≥ 50% reduction of blasts) in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL" (NCT01550224)
Timeframe: up to 10 weeks

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)0

[back to top]

Overall Survival (OS) at 2 Years

Overall Survival (OS) is defined as survival regardless of clinical status. OS is reported as the percentage without dispersion of participants in Groups 1 and 2 that remained alive 2 years after induction chemotherapy. (NCT01550224)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)0

[back to top]

Disease-free Survival (DFS) at 2 Years

"Disease-Free Survival (DFS) is defined as survival after complete response (CR) without disease progression. DFS is reported as the percentage without dispersion of participants in Groups 1 and 2 that experienced CR and were alive without progression (ie, without disease) 2 years after induction chemotherapy. CR is defined as all of the following.~CR = < 5% blasts in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods; no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL" (NCT01550224)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)0

[back to top]

Cytogenetic Response (CyR)

"Cytogenetic response (CyR) is defined as complete remission (CR), PLUS a documented decrease or absence of cytogenetic abnormalities, when analyzed microscopically for 20 cellular metaphases (actively dividing cells). The outcome is reported as the percentage without dispersion of participants in Groups 1 and 2 that demonstrated CyR. CR is defined as all of the following.~CR = < 5% blasts in bone marrow aspirate containing marrow spicules; > 200 nucleated cells no blasts with Auer rods; no persistence of extramedullary disease absolute neutrophil count (ANC) ≥ 1,000/µL platelets (PLT) ≥ 100,000/µL" (NCT01550224)
Timeframe: up to 10 weeks

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)0

[back to top]

Morphologic Leukemia-free State (MLFS)

"The rate of morphologic leukemia-free state (MLFS) is reported as the percentage without dispersion of participants in Groups 1 and 2 that achieve MLFS. The outcome is reported as the percentage without dispersion of participants in Groups 1 and 2 that demonstrated MLFS. This assessment is independent of absolute neutrophil count (ANC) or platelets (PLT) recovery status. MLFS is defined below.~MLFS = < 5% blasts in bone marrow aspirate containing marrow spicules > 200 nucleated cells no blasts with Auer rods no persistence of extramedullary disease" (NCT01550224)
Timeframe: up to 10 weeks

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)1

[back to top]

Complete Remission (CR)

"This study evaluates the clinical efficacy of temozolomide + vorinostat as administered to Groups 1 and 2, assessed as the rate of complete remission [CR, aka morphologic complete remission (mCR)], defined as the morphologic leukemia-free state (MLFS), WITH absolute neutrophil count (ANC) ≥ 1,000/µL AND platelets (PLT) ≥ 100,000/µL. The outcome is reported as the percentage without dispersion of participants in Groups 1 and 2 that demonstrated CR. CR is defined as all of the following.~MLFS = < 5% blasts in bone marrow aspirate containing marrow spicules > 200 nucleated cells no blasts with Auer rods no persistence of extramedullary disease ANC = ≥ 1,000/µL PLT = ≥ 100,000/µL" (NCT01550224)
Timeframe: up to 10 weeks

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)0

[back to top]

Complete Remission With Incomplete Blood Count Recovery (CRp)

"The rate of complete remission with incomplete blood count recovery (CRp) for Groups 1 and 2 was assessed as the rate of morphologic leukemia-free state (MLFS) but with EITHER residual neutropenia (ANC < 1,000/µL) OR residual thrombocytopenia (PLT < 100,000/µL). MLFS is defined as follows.~MLFS = < 5% blasts in bone marrow aspirate containing marrow spicules > 200 nucleated cells no blasts with Auer rods no persistence of extramedullary disease" (NCT01550224)
Timeframe: up to 10 weeks

InterventionParticipants (Count of Participants)
Participant Group 1 (Methylated MGMT Promoter)0
Participant Group 2 (Non-methylated MGMT Promoter)1

[back to top]

Overall Participant Response

Number of participants with response according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 criteria. Complete Response (CR): Disappearance all target lesions; Any pathological lymph nodes must be <10mm in short axis. Partial Response (PR): >30% decrease in sum of diameters of target lesions, reference baseline sum of diameters (e.g. percent change from baseline). Stable Disease (SD): Neither sufficient shrinkage for PR nor sufficient increase for progressive disease. Progressive Disease (PD): >20% increase in sum of diameters of target lesions, reference smallest sum of diameters recorded since treatment started (e.g. percent change from nadir, where nadir defined as smallest sum of diameters recorded since treatment start). In addition, sum must have an absolute increase from nadir of 5mm. Not Applicable (NA): No target lesions at baseline. Not Evaluable (NE): Cannot be classified by 1 of 5 preceding definitions. (NCT01590082)
Timeframe: 2 cycles, up to 7 weeks

InterventionParticipants (Count of Participants)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Not Evaluable (NE)
Phase I: Doxycycline, Ipilimumab, and Temozolomide00361

[back to top]

Maximum Tolerated Dose (MTD) When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma

The MTD was the highest dose level tested at which fewer than two of six patients had first course DLT. Hematologic DLT was defined as grade 4 neutropenia for more than 7 days, need for platelet transfusion for a platelet count of less than 20,000/mL twice within a 7-day period, or greater than 14-day delay in the start of a subsequent course because of neutropenia or thrombocytopenia. Nonhematologic DLT was defined as any nonhematologic toxicity that delayed the start of a subsequent cycle by more than 14 days or any grade ≥3 toxicity with the exception of the following grade 3 toxicities: nausea, vomiting, anorexia, or dehydration resolving to grade ≤ 2 within 72 hours; increase in hepatic transaminase or electrolyte abnormality resolving to grade ≤ 1 within 7 days; diarrhea persisting for less than 72 hours; fever; infection; or febrile neutropenia. DLT definitions included only toxicities deemed at least possibly related to therapy. (NCT01601535)
Timeframe: 21 days, from study day 1

Interventionmg/m^2 (Number)
Phase I60

[back to top]

One Year Progression Free Survival Rate

To determine the progression free survival rates for patients with relapsed or refractory neuroblastoma treated with MLN8237, irinotecan, and temozolomide at the identified MTD (NCT01601535)
Timeframe: 1 Years after completion of study

InterventionParticipants (Count of Participants)
Ph 24

[back to top]

Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Alisertib AUC

Outcomes included Alisertib, irinotecan, APC, SN-38, and SN-38G. APC, SN-38, and SN-38G are metabolites of irinotecan. (NCT01601535)
Timeframe: 1st week of cycle 1

InterventionµM•hour (Median)
DL 128.15
DL 1B21
DL 2B30.71
DL 3B47.73
Oral Solution58.15

[back to top]

Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Irinotecan Clearance

Outcomes included Alisertib, irinotecan, APC, SN-38, and SN-38G. APC, SN-38, and SN-38G are metabolites of irinotecan. (NCT01601535)
Timeframe: 1st week of cycle 1

InterventionL/h (Median)
DL 114.0
DL 1B15.7
DL 2B10.4
DL 3B16.0
Ph 212.6
Oral Solution10.3

[back to top]

AURKA Genotype

To explore whether AURKA genotype is associated with antitumor activity in children with refractory neuroblastoma treated with the combination of MLN8237, irinotecan, and temozolomide. (NCT01601535)
Timeframe: Day 7 of cycle 1

,
Interventionparticipants (Number)
AurkA Codon 31 Summary HAurkA Codon 31 Summary VAurkA Codon 31 Summary WAurkA Codon 31 Summary MissingAurkA Codon 57 Summary HAurkA Codon 57 Summary WAurkA Codon 57 Summary Missing
Objective Non-Responders912210102210
Objective Responders3054354

[back to top]

Aurora A Expression

To explore whether MYCN status and markers of expression of Aurora A in archival tumor tissue are associated with the antitumor activity of the combination of MLN8237, irinotecan, and temozolomide (NCT01601535)
Timeframe: From date of study enrollment to the date of progression or withdrawal from the study, up to 34 cycles (about 2 years).

,
Interventionparticipants (Number)
MYCN AmplifiedMYCN not AmplifiedMYCN MissingMYCN Amplified or Myc PositiveMYCN Non-amplified and Myc NegativeMYCN or Myc MissingAurora A protein PositiveAurora A protein NegativeAurora A protein Missing
Objective Non-Responders14244161115101418
Objective Responders192255246

[back to top]

Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Alisertib Day 4 Trough, Day 5 Trough and Cmax

Outcomes included Alisertib, irinotecan, APC, SN-38, and SN-38G. APC, SN-38, and SN-38G are metabolites of irinotecan. (NCT01601535)
Timeframe: 1st week of cycle 1

,,,,
InterventionµM (Median)
Alisertib Day 4 troughAlisertib Day 5 troughAlisertib Cmax
DL 10.480.372.56
DL 1B0.350.362.39
DL 2B0.30.23.77
DL 3B0.730.694.94
Oral Solution0.470.588.66

[back to top]

Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Alisertib Tmax and Half-life

Outcomes included Alisertib, irinotecan, APC, SN-38, and SN-38G. APC, SN-38, and SN-38G are metabolites of irinotecan. (NCT01601535)
Timeframe: 1st week of cycle 1

,,,,
Interventionhour (Median)
Alisertib TmaxAlisertib Half-life
DL 12.047.20
DL 1B1.748.61
DL 2B2.56.19
DL 3B2.528.54
Oral Solution28.34

[back to top]

Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Irinotecan AUC, APC AUC, SN-38 AUC, and SN-38G AUC

Outcomes included Alisertib, irinotecan, APC, SN-38, and SN-38G. APC, SN-38, and SN-38G are metabolites of irinotecan. (NCT01601535)
Timeframe: 1st week of cycle 1

,,,,,
Interventionh·ng/mL (Median)
Irinotecan AUCAPC AUCSN-38 AUCSN-38G AUC
DL 13,70257163.2206.5
DL 1B2,68047752.997.6
DL 2B3,95751180.8141.8
DL 3B2,61541872.0134.4
Oral Solution3,12161656.7136
Ph 23,53351190.0132

[back to top]

Pharmacokinetics When Given Together With Fixed Doses of Irinotecan and Temozolomide in Children and Young Adults With Relapsed or Refractory Neuroblastoma: Irinotecan Cmax, APC Cmax, SN-38 Cmax, and SN-38G Cmax

Outcomes included Alisertib, irinotecan, APC, SN-38, and SN-38G. APC, SN-38, and SN-38G are metabolites of irinotecan. (NCT01601535)
Timeframe: 1st week of cycle 1

,,,,,
Interventionng/mL (Median)
Irinotecan CmaxAPC CmaxSN-38 CmaxSN-38G Cmax
DL 172261.29.518.2
DL 1B70359.812.616.9
DL 2B1,23855.812.013.8
DL 3B78443.411.713.0
Oral Solution73251.48.2615.2
Ph 288157.110.412.8

[back to top]

Response Rate for Patients With Relapsed or Refractory Neuroblastoma Treated With MLN8237, Irinotecan, and Temozolomide at the Identified MTD

Response was graded according to version 1.2 of the NANT response criteria that classifies patients as having one of the following overall response categories based upon underlying response at soft tissue sites, MIBG positive sites, and bone marrow disease: complete response (CR); CR with minimal residual disease (CR-MRD); partial response (PR); minor response (MR); stable disease (SD); and progressive disease (PD). These criteria utilize RECIST criteria for measurable tumors, Curie score for MIBG scan response, and bone marrow (BM) morphology. BM response was graded as CR (required two time points to confirm), CR unconfirmed (one time point only), CR-MRD (bone marrow involvement < 5% at study entry with negative follow-up biopsies), SD, or PD. Patients with at least SD or better underwent central review of MIBG scans, CT scans, and bone marrow pathology slides. Overall responses of CR, CR-MRD, or PR were considered objective responses. (NCT01601535)
Timeframe: Cycles repeated every 21 days for up to 34 cycles.

InterventionParticipants (Count of Participants)
Complete Response (CR)CR-Minimal Residual Disease (MRD)Partial Response (PR)Minor ResponseStable DiseaseProgressive DiseaseResponse Rate (CR + CR-MRD + PR)
Ph 20042854

[back to top]

UGT1A1 Genotype

To explore whether UGT1A1 genotype is associated with toxicity in children with refractory neuroblastoma treated with the combination of MLN8237, irinotecan, and temozolomide (NCT01601535)
Timeframe: Day 7 of cycle 1

,,,
Interventionparticipants (Number)
UGT1A1 66UGT1A1 67UGT1A1 77UGT1A1 Missing
DLT in Any Cycle No91116
DLT in Any Cycle Yes8649
First Cycle DLT No1514212
First Cycle DLT Yes2333

[back to top] [back to top]

Number of Participants With Adverse Events

Tabulated According to the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0. Summary level. (NCT01638546)
Timeframe: From the start of treatment until 30 days from coming off treatment

InterventionParticipants (Count of Participants)
Arm I (Veliparib and Temozolomide)52
Arm II (Placebo and Temozolomide)45

[back to top]

Overall Response (ORR) by RECIST 1.1 Criteria

Corresponding exact two-sided 95% confidence intervals will be calculated and reported in both arms of the study. Comparisons between treatment arms will be performed using Fisher-exact test. (NCT01638546)
Timeframe: From date of randomization until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 4 months

Interventionparticipants (Number)
Arm I (Veliparib and Temozolomide)20
Arm II (Placebo and Temozolomide)6

[back to top]

Overall Survival

Estimated in each treatment group using Kaplan-Meier method. Group comparisons will be performed using log-rank test. (NCT01638546)
Timeframe: From randomization to time of death

Interventionmonths (Median)
Arm I (Veliparib and Temozolomide)8.2
Arm II (Placebo and Temozolomide)7

[back to top]

Progression-free Survival, Calculated as the Proportion of Patients Alive and Without Evidence of Disease

Compared across the two arms using a Fisher exact test. (NCT01638546)
Timeframe: From randomization to time of progression or death, whichever occurs first, assessed at 4 months

Interventionparticipants (Number)
Arm I (Veliparib and Temozolomide)18
Arm II (Placebo and Temozolomide)11

[back to top]

Best Overall Response (BOR) - Initial Treatment Period

BOR was assessed by independent radiology review using RECIST 1.1 and was recorded from randomization until the last imaging assessment in this period. Response categories included: Complete Response (CR): disappearance of all target lesions; Partial Response (PR): at least a 30% decrease in the sum of diameters of target lesions; Progressive Disease (PD): at least a 20% increase in the sum of diameters of target lesions; and Stable Disease (SD): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. BOR for the Initial Treatment Period was based on IRO. BOR for participants during the Initial Treatment Period is presented. (NCT01704287)
Timeframe: Up to approximately 36 months (Through Final Analysis database cutoff date of 16-Nov-2015)

,,
InterventionPercentage of Participants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot EvaluableNo DiseaseNo Assessment
Investigator-Choice Chemotherapy (ICC)0.04.519.061.515.10.00.0
Pembrolizumab 10 mg/kg7.220.414.947.59.90.00.0
Pembrolizumab 2 mg/kg3.318.916.746.713.30.60.6

[back to top]

Progression-free Survival (PFS) - Initial Treatment Period

PFS was defined as the time from randomization to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per Response Criteria in Solid Tumors version 1.1 (RECIST 1.1), PD was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to the relative increase of 20%, the sum must have also demonstrated an absolute increase of at least 5 mm. Note: The appearance of one or more new lesions was also considered PD. Analysis of PFS was based on an integrated radiology and oncology (IRO) assessment and was not planned or conducted for the switch-to-pembrolizumab treatment groups. Median PFS based on the product limit (Kaplan-Meier) method for censored data is presented. This was the final analysis for PFS. (NCT01704287)
Timeframe: Up to approximately 36 months (Through Final Analysis database cutoff date of 16-Nov-2015)

InterventionMonths (Median)
Pembrolizumab 2 mg/kg2.9
Pembrolizumab 10 mg/kg3.0
Investigator-Choice Chemotherapy (ICC)2.8

[back to top]

Overall Response Rate (ORR) - Initial Treatment Period

ORR was defined as the percentage of participants who had a Complete Response (CR: Disappearance of all target lesions) or a Partial Response (PR: At least a 30% decrease in the sum of diameters of target lesions) as assessed using RECIST 1.1. Analysis of ORR was not planned or conducted for the switch-to-pembrolizumab treatment groups. The percentage of participants who experienced a CR or PR is presented. This was the final analysis for ORR. (NCT01704287)
Timeframe: Up to approximately 36 months (Through Final Analysis database cutoff date of 16-Nov-2015)

InterventionPercentage of Participants (Number)
Pembrolizumab 2 mg/kg22.2
Pembrolizumab 10 mg/kg27.6
Investigator-Choice Chemotherapy (ICC)4.5

[back to top]

Number of Participants Who Experienced an Adverse Event (AE) - Overall Study

An AE was defined as any unfavorable and unintended change in the structure, function, or chemistry of the body temporally associated with the use of study drug, whether or not considered related to the use of the product. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a pre-existing condition which was temporally associated with the use of study drug, was also an AE. Participants were included in the treatment group in which an AE was experienced. The number of participants who experienced at least one AE is presented. (NCT01704287)
Timeframe: Up to approximately 75 months (Through End of Trial Analysis database cutoff date of 31-Jan-2019)

InterventionParticipants (Count of Participants)
Pembrolizumab 2 mg/kg172
Pembrolizumab 10 mg/kg179
ICC Only71
ICC→Pembrolizumab 2 mg/kg52
ICC→Pembrolizumab 10 mg/kg45
ICC→Pembrolizumab 2 mg/kg (After Switch to Pembrolizumab)53
ICC→Pembrolizumab 10 mg/kg (After Switch to Pembrolizumab)41

[back to top]

Number of Participants Who Discontinued Study Drug Due to an Adverse Event (AE) - Overall Study

An AE was defined as any unfavorable and unintended change in the structure, function, or chemistry of the body temporally associated with the use of the study drug, whether or not considered related to the use of the product. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a pre-existing condition which is temporally associated with the use of study drug, was also an AE. The number of participants who discontinued study drug due to an AE is presented. (NCT01704287)
Timeframe: Up to approximately 75 months (Through End of Trial Analysis database cutoff date of 31-Jan-2019)

InterventionParticipants (Count of Participants)
Pembrolizumab 2 mg/kg29
Pembrolizumab 10 mg/kg33
ICC Only13
ICC→Pembrolizumab 2 mg/kg1
ICC→Pembrolizumab 10 mg/kg1
ICC→Pembrolizumab 2 mg/kg (After Switch to Pembrolizumab)4
ICC→Pembrolizumab 10 mg/kg (After Switch to Pembrolizumab)4

[back to top]

Interim Overall Survival (OS) - Initial Treatment Period

OS was defined as the time from randomization to death due to any cause. Analysis of OS was not planned or conducted for the switch-to-pembrolizumab treatment groups. Median OS based on the product-limit (Kaplan-Meier) method for censored data is presented. This was the interim analysis for OS. (NCT01704287)
Timeframe: Up to approximately 36 months (Through Final Analysis database cutoff date of 16-Nov-2015)

InterventionMonths (Median)
Pembrolizumab 2 mg/kg13.4
Pembrolizumab 10 mg/kg14.7
Investigator-Choice Chemotherapy (ICC)11.0

[back to top]

Final Overall Survival (OS) - Initial Treatment Period

OS was defined as the time from randomization to death due to any cause. Analysis of OS was not planned or conducted for the switch-to-pembrolizumab treatment groups. Median OS duration based on the product-limit (Kaplan-Meier) method for censored data is presented. This was the final analysis for OS. (NCT01704287)
Timeframe: Up to approximately 75 months (Through End of Trial Analysis database cutoff date of 31-Jan-2019)

InterventionMonths (Median)
Pembrolizumab 2 mg/kg13.4
Pembrolizumab 10 mg/kg14.7
Investigator-Choice Chemotherapy (ICC)11.0

[back to top]

Duration of Response (DOR) - Initial Treatment Period

For participants who demonstrated a confirmed response (Complete Response [CR]: disappearance of all target lesions or Partial Response [PR]: ≥30% decrease in the sum of diameters of target lesions) per RECIST 1.1, DOR was defined as the time from first documented evidence of CR or PR until disease progression or death. DOR for participants who had not progressed or died at the time of analysis was to be censored at the date of their last tumor assessment. DOR analysis was based on IRO assessment. Analysis of DOR was not planned or analyzed for the switch-to-pembrolizumab treatment groups. Median DOR for participants who demonstrated a confirmed response is presented. (NCT01704287)
Timeframe: Up to approximately 36 months (Through Final Analysis database cutoff date of 16-Nov-2015)

InterventionMonths (Median)
Pembrolizumab 2 mg/kg22.8
Pembrolizumab 10 mg/kgNA
Investigator-Choice Chemotherapy (ICC)6.8

[back to top]

Best Overall Response (BOR) - Switch-to-Pembrolizumab Treatment Period

The BOR was assessed using RECIST 1.1 and was recorded from the start of the second line of study drug (pembrolizumab) until the last imaging assessment in this period. Response categories included: Complete Response (CR): disappearance of all target lesions; Partial Response (PR): at least a 30% decrease in the sum of diameters of target lesions; Progressive Disease (PD): at least a 20% increase in the sum of diameters of target lesions; and Stable Disease (SD): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. For the switch-to-pembrolizumab treatment groups, BOR was based on independent review committee (IRC) assessment. The BOR for switched-to pembrolizumab treatment groups is presented. (NCT01704287)
Timeframe: Up to approximately 36 months (Through Final Analysis database cutoff date of 16-Nov-2015)

,
InterventionPercentage of Participants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot EvaluableNo Assessment
ICC→Pembrolizumab 10 mg/kg4.413.311.155.613.32.2
ICC→Pembrolizumab 2 mg/kg1.917.015.154.711.30.0

[back to top]

Final Overall Survival (OS) By Programmed Cell Death-Ligand 1 (PD-L1) Tumor Expression Status - Initial Treatment Period

OS was defined as the time from randomization to death due to any cause. Participants with an Allred Proportion Score (APS) ≥2 (membranous staining in ≥1% of cells for PD-L1) were considered to be PD-L1 Positive and participants with an APS of 0 or 1 were considered to be PD-L1 Negative. Analysis of OS was not planned or conducted for the switch-to-pembrolizumab treatment groups. Median OS duration based on the product-limit (Kaplan-Meier) method for censored data by PD-L1 tumor expression status is presented. (NCT01704287)
Timeframe: Up to approximately 75 months (Through End of Trial Analysis database cutoff date of 31-Jan-2019)

,,
InterventionMonths (Median)
PD-L1 PositivePD-L1 Negative
Investigator-Choice Chemotherapy (ICC)12.19.3
Pembrolizumab 10 mg/kg17.513.4
Pembrolizumab 2 mg/kg15.010.5

[back to top]

Progression-free Survival (PFS)

To assess the effect on progression-free survival of continuing bevacizumab treatment after disease progression in patients treated with bevacizumab from the time of initiation of treatment to the first occurrence of progression, or death (NCT01740258)
Timeframe: 5 Years

Interventionmonths (Median)
Bevaczimab, Radiation Therapy, Temozolomide9.9

[back to top]

Overall Survival

To assess the effect on overall survival of continuing bevacizumab treatment after disease progression in patients treated with bevacizumab from the time of first diagnosis of grade IV malignant glioma. (NCT01740258)
Timeframe: 5 Years

Interventionmonths (Median)
Bevaczimab, Radiation Therapy, Temozolomide17.8

[back to top]

Toxicity: Percentage of Subjects With Unacceptable Toxicities

"The occurrence of ≥ grade 2 CNS (central nervous system) hemorrhage or grade 4 or 5 non-hematologic toxicity is defined as being unacceptable. Unacceptable toxicity rates of 5% or less are considered desirable, while rates of 20% or greater are considered as undesirable." (NCT01740258)
Timeframe: 5 Years

Interventionpercentage of participants (Number)
Bevaczimab, Radiation Therapy, Temozolomide11.8

[back to top]

Percentage of Randomized Patients Who Are Responders

The percentage of patients who are responders will be tabulated, including a 95% confidence interval on the response rate. Responders are defined as patients who achieve a best overall response of complete response (CR), very good partial response (VGPR), or partial response (PR) per the International Neuroblastoma Response Criteria (INRC). Per INRC: CR= Disappearance of all target lesions. No evidence of tumor at any site; VGPR= >90% decrease of the disease measurement for CT/MRI target lesions. All pre-existing bone lesions with CR by MIBG; MIBG scan can be stable disease (SD) or CR in soft tissue lesions corresponding to lesions on CT/MRI. CR in bone marrow. No new sites of tumor; PR= >=30% decrease in the disease measurement for CT/MRI target lesions. Bone marrow with CR. MIBG with either PR/CR in bone lesions; MIBG may be SD or CR in soft tissue lesions corresponding to lesions on CT/MRI. Homovanillic acid (HVA)/Vanillylmandelic acid (VMA) may still be elevated. (NCT01767194)
Timeframe: Up to the first 6 cycles of treatment

InterventionPercentage of patients (Number)
Arm I (Temozolomide, Irinotecan Hydrochloride, Temsirolimus)5.6
Arm II (Temozolomide, Irinotecan Hydrochloride, Dinutuximab)52.9

[back to top]

Percentage of Patients in the Dinutuximab Arm Who Are Responders

Percentage of patients who are responders to therapy with dinutuximab will be tabulated, including a 95% confidence interval on the response rate. Responders are defined as patients who achieve a best overall response of complete response (CR), very good partial response (VGPR), or partial response (PR) per the International Neuroblastoma Response Criteria (INRC). Per INRC: CR= Disappearance of all target lesions. No evidence of tumor at any site; VGPR= >90% decrease of disease measurement for CT/MRI target lesions. All pre-existing bone lesions with CR by MIBG; MIBG scan can be stable disease (SD) or CR in soft tissue lesions corresponding to lesions on CT/MRI. CR in bone marrow. No new sites of tumor; PR= ≥30% decrease in disease measurement for CT/MRI target lesions. Bone marrow with CR. MIBG with either PR/CR in bone lesions; MIBG may be SD or CR in soft tissue lesions corresponding to lesions on CT/MRI. Homovanillic acid (HVA)/ Vanillylmandelic acid (VMA) may still be elevated. (NCT01767194)
Timeframe: Up to the first 6 cycles of treatment

InterventionPercentage of patients (Number)
Arm II (Temozolomide, Irinotecan Hydrochloride, Dinutuximab)41.2

[back to top]

Pathological Complete Response

"Pathologic responses and stages were classified according to Dworak's classification and the 7th edition of the American Joint Committee on Cancer staging system, respectively.~The pathologic complete response (pCR) was defined as the total regression of the primary tumor regardless of regional lymph nodal status (ypT0), with residual fibrotic mass or acellular mucin pools only, thus without detectable tumor cells." (NCT01781403)
Timeframe: at the time of surgery (6-8 weeks after study treatment)

Interventionparticipants (Number)
Unmethylated MGMT1
Hypermethylated MGMT6

[back to top]

Toxicity(Adeverse Event)

"Toxicity will be monitored and recorded every week during study treatment (5 or 6 weeks) as following according to the NCI-CTCAE version 4.0~An interval history and physical examination with particular attention to drug-induced side effects along with documentation of the patient's weight and performance status will be performed on each visit.~CBC with differential count, blood chemistry including calcium, phosphorus, glucose, BUN, creatinine, total protein, albumin, AST, ALT, alkaline phosphatase, total bilirubin, and electrolyte will be performed before next planned treatment.~All relevant information regarding drug dosage, laboratory examinations, and treatment-related toxicities must be recorded before each treatment is given.~Summaries of the frequency and severity of adverse effects are based on the worst episodes recorded." (NCT01781403)
Timeframe: 5-6 weeks during study treatment

,,
Interventionevents (Number)
Any grade 1 adverse eventAny grade 2 adverse eventAny grade 3 adverse eventAny grade 4 adverse event
Dose Level 16210
Dose Level 28200
Dose Level 3/Recommended Dose601730

[back to top] [back to top]

Maximum Tolerated Dose (MTD)

The MTD is defined as the maximum dose level in the doses of temozolomide tested with capecitabine and radiation in which the incidence proportion of DLT exceeds 30%. (NCT01781403)
Timeframe: 5-6 weeks during study treatment

Interventionmg/m^2 (Number)
Dose Level 10
Dose Level 20
Dose Level 3/Recommended Dose0

[back to top]

Summary of the Median Progression-free Survival (mPFS) in the Study Group Compared With Historical Control From Medical Literature

mPFS was based on model parameters estimated by maximum likelihood with a Newton-Raphson algorithm. The Radiation Therapy Oncology Group (RTOG) 0525 study was the main historical control used for statistical analysis. Additionally, RTOG-0825 was also used to support this outcome measure. (NCT01790503)
Timeframe: Assessed from date of first dose administered to date of first documented progression or date of death from any cause, whichever came first, assessed up to 4 years 4 months.

Interventionmonths (Median)
RP2D7.7
RP2D-0525 (Cycle 1, Day 1)7.6
RP2D-08257.0
RP2D-0525 (Rest Period, Day 15)6.1

[back to top]

Number of Patients With Clinically Significant Abnormal Chemistry and Hematology Values Reported as Adverse Events by Preferred Term in Phase 1b of the Modified Intent-To-Treat Population

(NCT01790503)
Timeframe: Baseline up to 30 days after last dose, up to 4 years 4 months

,,,,,,,,
InterventionParticipants (Count of Participants)
ThrombocytopeniaNeutropeniaAnaemiaLymphopeniaAST increasedPlatelet count decreasedNeutrophil count decreasedLeukopeniaFebrile neutropeniaALT increasedBone marrow failureHaemolysisWhite blood cell decreasedLymphocyte count decreased
Phase 1b Dose Escalation - 600 mg00000000000100
Phase 1b Dose Escalation - 600 mg/1000 mg00001001010000
Phase 1b Dose Escalation - 600 mg/600 mg10110010000000
Phase 1b Dose Escalation - 600 mg/800 mg11010001000000
Phase 1b Dose Escalation - 800 mg (5 Days)11000100000001
Phase 1b Dose Escalation - 800 mg/1000 mg11112100110000
Phase 1b Dose Escalation - 800 mg/600 mg00000000000000
Phase 1b Dose Escalation - 800 mg/800 mg10000000000000
Phase 1b Dose Escation - 800 mg (No Adjuvant Therapy)11100120101010

[back to top] [back to top]

Overview of Most Frequent System Organ Classes Reported in Phase 2 of the Modified Intent-To-Treat Population

(NCT01790503)
Timeframe: Baseline up to 30 days after last dose, up to 4 years 4 months

,,,
InterventionParticipants (Count of Participants)
General Disorders and Administration SiteSkin and Subcutaneous Tissue DisordersNervous System DisordersGastrointestinal DisordersMetabolism and Nutrition DisordersPsychiatric DisordersInvestigationsMusculoskeletal and Connective Tissue DisordersBlood and Lymphatic System DisordersInjury, Poisoning, and Procedural ComplicationsEye DisordersVascular DisordersRespiratory, Thoracic, and Mediastinal DisordersRenal and Urinary DisordersCardiac DisordersEndocrine DisordersEar and Labyrinth DisordersReproductive System and Breast DisordersNeoplasms Benign, Malignant, and UnspecifiedHepatobiliary DisordersImmune System Disorders
Combined 800 mg, 5 Days/Week47454341302727251818161816138753211
Phase 2 - 800 mg1312121286972664322110010
Phase 2 - 800 mg/800 mg2424222116171213141079775532201
Phase 2 Total3736343324232120161613131097642211

[back to top]

Overview of Most Frequent System Organ Classes Reported in Phase 1b of the Modified Intent-To-Treat Population

(NCT01790503)
Timeframe: Baseline up to 30 days after last dose, up to 4 years 4 months

,,,,,,,,
InterventionParticipants (Count of Participants)
General Disorders and Administrative SiteNervous System DisordersSkin and Subcutaneous Tissue DisordersGastrointestinal DisordersMetabolism and Nutrition DisordersPsychiatric DisordersInfections and InfestationsInvestigationsRespiratory, Thoracic, and Mediastinal DisordersMusculoskeletal and Connective Tissue DisordersBlood and Lymphatic Tissue DisordersVascular DisordersRenal and Urinary DisordersEye DisordersInjury, Poisoning, and Procedural ComplicationsEar and LabyrinthEndocrine DisordersCardiac DisordersHepatobiliary DisordersReproductive System and Breast DisordersImmune System DisordersNeoplasms Benign, Malignant, and Unspecified
Phase 1b Dose Escalation - 600 mg1221121101111011000000
Phase 1b Dose Escalation - 600 mg/1000 mg1111111110100000000000
Phase 1b Dose Escalation - 600 mg/600 mg2333321110101110000000
Phase 1b Dose Escalation - 600 mg/800 mg0111110000100000000000
Phase 1b Dose Escalation - 800 mg5443211233121100010000
Phase 1b Dose Escalation - 800 mg/1000 mg5555435432133221100100
Phase 1b Dose Escalation - 800 mg/600 mg1100010101000000000000
Phase 1b Dose Escalation - 800 mg/800 mg1010100000100000000000
Phase 1b Dose Escation - 800 mg3212213321121101101000

[back to top]

Summary of the Median Progression-free Survival (mPFS) in the Combined 800 mg, 5 Days/Week Dose Group

Progression was determined by Response Assessment in Neuro-Oncology (RANO) criteria and progression-free survival (PFS) was analyzed based on the non-parametric Kaplan-Meier method. (NCT01790503)
Timeframe: Assessed from date of first dose administered to date of first documented progression or date of death from any cause, whichever came first, assessed up to 4 years 4 months.

Interventionmonths (Median)
mITT RP2DPP RP2D
Combined 800 mg, 5 Days/Week6.76.9

[back to top]

Number of Patients With Clinically Significant Abnormal Chemistry and Hematology Values Reported as Adverse Events by Preferred Term in Phase 2, the Combined RP2D Groups in Phase 1b, and in the Study Overall Modified Intent-To-Treat Population

(NCT01790503)
Timeframe: Baseline up to 30 days after last dose, up to 4 years 4 months

,,,
InterventionParticipants (Count of Participants)
NeutropeniaThrombocytopeniaAnaemiaALT increasedAST increasedPlatelet count decreasedLymphopeniaWhite blood cell count decreasedNeutrophil count decreasedLeukopeniaFebrile neutropeniaLymphocyte count decreasedBlood alkaline phosphatase increasedBlood creatinine increasedDRESSLiver function test abnormalBone marrow failureHaemolysisBlood bilirubin increasedBlood iron decreased
Combined 800 mg, 5 Days/Week96788745222333110111
Phase 2 - 800 mg11154111011131110010
Phase 2 - 800 mg/800 mg63522424210102000001
Phase 2 Total74676535221233110011

[back to top]

Summary of the Overall Survival in the Study Group Compared With Historical Control From Medical Literature

Overall Survival was calculated using a parametric model. The Radiation Therapy Oncology Group (RTOG) 0525 study was the main historical control used for statistical analysis. Additionally, RTOG-0825 was also used to support this outcome measure (NCT01790503)
Timeframe: Assessed from date of first dose administered to the date of death from any cause, assessed up to 4 years 4 months.

Interventionmonths (Median)
RP2D18.8
RP2D-0525 (Cycle 1, Day 1)20.2
RP2D-082517.0
RP2D-0525 (Rest Period, Day 15)16.9

[back to top] [back to top]

Summary of the Overall Survival in The Study Population

Overall Survival (OS) was defined as the number of days from the first day of treatment (C1D1) to the date of death and analyzed using a non-parametric Kaplan Meier method. (NCT01790503)
Timeframe: Assessed from date of first dose administered to date of death from any cause, assessed up to 4 years 4 months

Interventionmonths (Median)
mITT RP2DPP RP2D
Combined 800 mg, 5 Days/Week13.112.4

[back to top] [back to top]

Ability to Calculate the Efficacy of Low-dose Whole Brain RT (WBRT) in Patients With GBM

"Median survival (in months from time of diagnosis to date of death) was calculated and reported below.~It is very difficult to draw efficacy outcomes for outcome measure #1 and #2 because of incomplete enrollment." (NCT01822275)
Timeframe: 5 years

Interventionmonths (Median)
Low Dose WBRT10

[back to top]

Radiographic (CT-MRI) Response Assessment and Treatment Failure Patterns of Patients With GBM

"To determine the radiographic response and treatment failure patterns with the combination therapy. This would be measured through imaging that was collected from baseline to follow-up. If unable to meet statistical requirements, response assessment and treatment patterns will be unable to be determined.~It is very difficult to draw efficacy outcomes for outcome measure #1 and #2 because of incomplete enrollment." (NCT01822275)
Timeframe: 5 years

Interventionparticipants (Number)
Low Dose WBRT4

[back to top]

Proportion of Patients With Response

"Response was evaluated using the revised Response Evaluation Criteria in Solid Tumors (RECIST) guideline (version 1.1) and defined as either complete response (CR) or partial response (PR).~CR is defined as disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR is defined as at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters." (NCT01824875)
Timeframe: Assessed every 3 months for 3 years and then every 6 months for years 3-5

Interventionproportion of participants (Number)
Arm A (Temozolomide)0.338
Arm B (Temozolomide and Capecitabine)0.397

[back to top]

Progression-free Survival

Progression-free survival (PFS) is defined as the time from randomization to progression or death without evidence of progression. Progression was evaluated using the revised Response Evaluation Criteria in Solid Tumors (RECIST) guideline (version 1.1) and defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. Kaplan-Meier method was used to estimate PFS. (NCT01824875)
Timeframe: Assessed every 3 months for 3 years and then every 6 months for years 3-5

Interventionmonths (Median)
Arm A (Temozolomide)15.1
Arm B (Temozolomide and Capecitabine)23.2

[back to top]

Overall Survival

Overall survival is defined as time from randomization to death or date last known alive. (NCT01824875)
Timeframe: Assessed every 3 months for 3 years and then every 6 months for years 3-5

Interventionmonths (Median)
Arm A (Temozolomide)53.8
Arm B (Temozolomide and Capecitabine)58.7

[back to top]

Number of Participants With an Objective Response

ORR is the proportion of participants with a complete response (CR) or partial response (PR) per the Response Evaluation Criteria in Solid Tumors (RECIST). Complete Response is disappearance of all tumors. Partial Response is at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. (NCT01827384)
Timeframe: Up to 30 days after completion of study treatment, up to 75 months

,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial Response
TAC1 -> TAC400
TAC200
TAC2 -> TAC100
TAC2 -> TAC300
TAC301
TAC3 -> TAC100
TAC3 -> TAC1 -> TAC400
TAC3 -> TAC400
TAC400
TAC4 -> TAC100
TAC4 -> TAC200
TAC4 -> TAC300
Treatment Assignment Code 1 (TAC1)00

[back to top]

Proportion of Participants With 4 Month Progression-free Survival (PFS)

Time from random assignment to progression or death from any cause (whichever comes first). Progression was measured by the Response Evaluation Criteria in Solid Tumors (RECIST). Progression is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study. And the appearance of one or more new lesions is also considered progressions. (NCT01827384)
Timeframe: 4 months

Interventionproportion of participants (Number)
Treatment Assignment Code 1 (TAC1)0.23
TAC1 -> TAC40.00
TAC20.22
TAC2 -> TAC10.00
TAC2 -> TAC31.00
TAC30.41
TAC3 -> TAC10.50
TAC3 -> TAC1 -> TAC40.00
TAC3 -> TAC40.00
TAC40.13
TAC4 -> TAC10.29
TAC4 -> TAC20.00
TAC4 -> TAC30.00

[back to top]

Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v5.0)

Here is the number of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria for Adverse Events (CTCAE v5.0). A non-serious adverse event is any untoward medical occurrence. A serious adverse event is an adverse event or suspected adverse reaction that results in death, a life-threatening adverse drug experience, hospitalization, disruption of the ability to conduct normal life functions, congenital anomaly/birth defect or important medical events that jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the previous outcomes mentioned. (NCT01827384)
Timeframe: Date treatment consent signed to date off study, approx. 73months (m) & 21day (d); 4m & 11d; 61m & 8d; 4m & 11d; 10m & 11d; 72m & 29d; 13m & 11d; 6m & 13d; 29m & 18d; 48m & 25d; 49m & 5d; 4m &7d; 15m & 5d; and 75m &13d, for each group respectively.

InterventionParticipants (Count of Participants)
Treatment Assignment Code 1 (TAC1)13
TAC1 -> TAC41
TAC29
TAC2 -> TAC11
TAC2 -> TAC31
TAC322
TAC3 -> TAC12
TAC3 -> TAC1 -> TAC41
TAC3 -> TAC42
TAC415
TAC4 -> TAC17
TAC4 -> TAC21
TAC4 -> TAC32
Participants Enrolled But Not Treated2

[back to top]

Proportion of Patients Without a Progression Free Survival Event at 4 Months (PFS4)

A patient will be declared a PFS4 success if they are on study and progression free for at least 4 months. Progression is defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study, with an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered progression. The success for each arm will be calculated independently as the number of successes divided by the total number of evaluable patients. A one-sided chi-squared test for a difference in PFS4 proportions will be used to test for a difference between arms. (NCT01835145)
Timeframe: At 4 months

Interventionproportion of participants (Number)
Arm I (Cabozantinib-s-malate).323
Arm II (Temozolomide or Dacarbazine).267

[back to top]

Percentage of Patients Who Experienced Grade 3+ Adverse Events Regardless of Attribution

percentage of patients who experienced grade 3+ adverse events regardless of attribution, graded according to the National Cancer Institute CTCAE version 4.0 (NCT01835145)
Timeframe: Up to 2 years

Interventionpercentage of patients (Number)
Arm I (Cabozantinib-s-malate)51.6
Arm II (Temozolomide or Dacarbazine)20

[back to top]

Overall Survival (OS)

The distribution of OS time will be estimated using the method of Kaplan Meier. (NCT01835145)
Timeframe: Number of days from registration until death, assessed up to 2 years

Interventionmonths (Median)
Arm I (Cabozantinib-s-malate)6.3
Arm II (Temozolomide or Dacarbazine)7.2

[back to top]

Confirmed Response Rate as Determined by the RECIST Criteria (Version 1.1)

The confirmed response rates will be estimated by dividing the number of confirmed responders by the number of evaluable patients. 95% confidence intervals will be calculated. (NCT01835145)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Arm I (Cabozantinib-s-malate)0
Arm II (Temozolomide or Dacarbazine)0

[back to top]

PFS

The distribution of PFS time will be estimated using the method of Kaplan Meier and is defined as the number of days from registration until disease progression (or death). Progression is defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study, with an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered progression. (NCT01835145)
Timeframe: Number of days from registration until disease progression (or death), assessed up to 2 years

Interventionmonths (Median)
Arm I (Cabozantinib-s-malate)2.0
Arm II (Temozolomide or Dacarbazine)1.9

[back to top]

Response to Window Therapy (2 Courses) for Group B (High-risk) - ESFT Participants

Response rate will be defined as the proportion of patients who achieved complete response or partial response (CR+PR) using the World Health Organization (WHO) criteria evaluated after two initial courses of temsirolimus, temozolomide and irinotecan in previously untreated patients with high risk Ewing Sarcoma Family of Tumor (ESFT). Participants who are treated in Group B with Desmoplastic Small Round Cell Tumor (DSRCT) or those who do not receive window therapy will not be included in this analysis. (NCT01946529)
Timeframe: at 6 weeks after start of therapy (after 2 initial courses)

Interventionparticipants (Number)
Partial Response (PR)Stable disease (no response) (NR)Progressive Disease (PD)
Group B (High Risk) - ESFT381

[back to top]

Participants Alive and Without Disease Progression At 6 Months After the Start of the Irradiation

Progression free survival based on the Response Assessment for Neuro-Oncology (RANO) criteria, using both clinical examinations and MRIs with and without contrast summarized with Kaplan Meier estimates. (NCT01977677)
Timeframe: 6 months from start of irradiation

InterventionParticipants (Count of Participants)
Escalation: Plerixafor 200 mcg/kg/Day3
Escalation: Plerixafor 400 mcg/kg/Day5
Expansion: Plerixafor 400 mcg/kg/Day19

[back to top]

Dose-limiting Toxicity

Dose Limiting Toxicity is defined as defined as any hematologic or on-hematologic adverse events grade 3 or higher using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 with a suspected causal relationship to Plerixafor (including electrocardiogram changes indicative of ischemia, ventricular tachycardia) (NCT01977677)
Timeframe: Up to 30 days post plerixafor

InterventionParticipants (Count of Participants)
Escalation: Plerixafor 200 mcg/kg/Day0
Escalation: Plerixafor 400 mcg/kg/Day0
Expansion: Plerixafor 400 mcg/kg/Day0

[back to top]

Proportion of Grade IV MGMT Un-methylated Patients That Experience Confirmed-progression-free Survival at 6 Months (CPFS6)

"The proportion of Grade IV MGMT un-methylated patients that experience confirmed-progression-free survival at 6 months (CPFS6). Progression is defined by any of the following:~≥25% increase in the sum of products of perpendicular diameters of enhancing lesions compared to the smallest tumor measurement obtained either at baseline or best response, on stable or increasing doses of corticosteroids~Significant increase in T2/FLAIR non-enhancing lesion on stable or increasing doses of corticosteroids compared to baseline scan or best response following initiation of therapy, not due to co-morbid events~Any new lesion~Clear clinical deterioration not attributable to other causes apart from the tumor or changes in corticosteroid dose.~Failure to return for evaluation due to death or deteriorating condition~Clear progression of non-measurable disease" (NCT01991977)
Timeframe: Time from registration to the confirmed disease progression, assessed at 6 months

Interventionproportion of participants (Number)
Diagnostic (PET, pMRI, DTI, IMRT, Temozolomide)0.795

[back to top]

Progression-Free Survival Rate (PFS) (Arm 3)

Participants will be analyzed based on intention to treat using the Response Assessment in Neuro-Oncology (RANO) criteria for progression. Based on RANO criteria, a responder is defined using both radiographic and clinical criteria. Complete response (CR) or Partial Response (PR) will be first assessed by radiographic changes as determined by an improvement of the bi-dimensional evaluation of the tumor size. In addition, changes in neurologic function and steroid use will be considered. Analyses will be performed after all participants enrolled in arm 3 have completed 38 months on study, or whenever the progression status of all participants has been established, whichever comes first. Kaplan-Meier estimates and the associated 95% CIs will be calculated for the 38-month PFS. (NCT02023905)
Timeframe: Up to 38 Months

Interventionpercentage of participants (Number)
Arm 3: Everolimus (1p/19q Co-deletion Present)55.5

[back to top]

Progression-Free Survival Rate (PFS) (Arms 1 and 2)

Participants will be analyzed based on intention to treat using the Response Assessment in Neuro-Oncology (RANO) criteria for progression. Based on RANO criteria, a responder is defined using both radiographic and clinical criteria. Complete response (CR) or Partial Response (PR) will be first assessed by radiographic changes as determined by an improvement of the bi-dimensional evaluation of the tumor size. In addition, changes in neurologic function and steroid use will be considered. Analyses will be performed after all enrolled participants in arm 1 or arm 2 have completed 33 months on study, or whenever the progression status of all participants has been established, whichever comes first. Kaplan-Meier estimates and the associated 95% confidence intervals (CI) will be calculated for the 33-month PFS separately for the two arms. (NCT02023905)
Timeframe: Up to 33 Months

Interventionpercentage of participants (Number)
Arm 1: Everolimus31
Arm 2: Everolimus and TemozolomideNA

[back to top]

Rate of Reduction in Seizures

To assess the reduction in seizure rate the investigator will compare the seizure rate on study to that experienced one month prior to enrolling in the study. The RANO low grade gliomas (LGG) guideline will be used to assess reduction of seizures, which calls a 50% or more reduction number of monthly seizures an 'improvement'; a 50% or more increase a worsening'; and anything less than 50% in either direction a 'stable seizure rate'. (NCT02023905)
Timeframe: Up to 36 Months

Interventionpercentage of participants (Number)
Arm 1: Everolimus25
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)50

[back to top]

Median Progression Free Survival (PFS)

Participants will be analyzed based on an intention to treat model. PFS is defined as the time from first objective response to the time of disease progression or death using the Response Assessment in Neuro-Oncology (RANO) criteria to determine progression. If the participant does not have an event of disease progression or recurrence nor has the patient died, the participant's data will be censored at the date of last contact with the patient. Kaplan-Meier estimates and the associated 95% CIs will be calculated. (NCT02023905)
Timeframe: Up to 84 Months

Interventionmonths (Median)
Arm 1: Everolimus25.8
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)43.6

[back to top]

Objective Response Rate (ORR)

Overall response rate (ORR) is the percentage of patients who achieved a best response of complete response (CR) or partial response (PR) out of all assigned patients. Based on the best objective status as assessed by the Response Assessment in Neuro-Oncology (RANO) criteria. Using RANO criteria, a responder is defined by radiographic and clinical criteria. Complete response or PR will be first assessed by radiographic changes as determined by an improvement of the bi-dimensional evaluation of the tumor size. In addition, changes in neurologic function and steroid use will be considered. The point estimate and the associated 2-sided 95% CI for the response rate separately for the three arms. (NCT02023905)
Timeframe: Up to 36 Months

Interventionpercentage of participants (Number)
Arm 1: Everolimus0
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)11

[back to top]

Overall Survival Rate (OS)

Participants will be analyzed based on an intention to treat model. Overall survival is defined as the first day of treatment until death or study completion, whichever comes first. Kaplan-Meier estimates and the associated 95% CIs will be calculated by treatment arm. (NCT02023905)
Timeframe: Up to 84 Months

Interventionpercentage of participants (Number)
Arm 1: EverolimusNA
Arm 2: Everolimus and TemozolomideNA
Arm 3: Everolimus (1p/19q Co-deletion Present)NA

[back to top]

Number of Participants With Adverse Events as a Measure of Safety and Tolerability

The short-and long-term toxicity of ScRT (and compare to historical controls). (NCT02039778)
Timeframe: 36 months

InterventionParticipants (Count of Participants)
Stem Cell Radiotherapy and Temozolomide4

[back to top]

Overall Survival

The overall survival of patients with newly diagnosed high-grade glioma (HGG) treated with concurrent ScRT and temozolomide, followed by post-radiation temozolomide (and compare to historical controls). (NCT02039778)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Stem Cell Radiotherapy and Temozolomide1

[back to top]

Number of Participants With Serious and Non-serious Adverse Events as a Measure of Safety.

Patients in the safety lead-in part of the study were monitored for up to 2 treatment cycles (4 weeks) to assure there were no unexpected or prohibitive toxicities. A non-serious adverse event is any untoward medical occurrence. A serious adverse event (SAE) is an event that meets one or more of the following: 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; requires intervention to prevent permanent impairment or damage. Specific AE and SAE terms are provided in the Adverse event module. (NCT02113007)
Timeframe: up to 4 weeks

Interventionparticipants (Number)
Adverse EventsSerious Adverse Events
Rituximab Plus Temozolomide21

[back to top]

C Max of Talazoparib

Median with minimum and maximum for the time at which the maximum (peak) serum concentration occurs. (NCT02116777)
Timeframe: Cycle 1 Day 1 pre-dose, and 1, 2, 4, 8 and 24 hours after the first talazoparib dose.

Interventionng/mL (Median)
Treatment (Talazoparib, Temozolomide): Phase 14670

[back to top]

C Max of Talazoparib in Combination With Temozolomide

Median with minimum and maximum for the time at which the maximum (peak) serum concentration occurs. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the talazoparib dose

Interventionpg/ml (Median)
Treatment (Talazoparib, Temozolomide): Phase 116450

[back to top]

C Max of Temozolomide in Combination With Talazoparib

Median with minimum and maximum for the maximum (peak) serum concentration. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the temozolomide dose

Interventionng/mL (Median)
Treatment (Talazoparib, Temozolomide): Phase 11234.1

[back to top]

Clearance of Temozolomide in Combination With Talazoparib

Median with minimum and maximum for the rate of elimination of the drug. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the temozolomide dose

InterventionL/hr/m² (Median)
Treatment (Talazoparib, Temozolomide): Phase 16.49

[back to top]

Half-life of Temozolomide in Combination With Talazoparib

Median with minimum and maximum for the time required for the serum concentration to fall to 50% of its starting dose. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the temozolomide dose

InterventionHours (Median)
Treatment (Talazoparib, Temozolomide): Phase 11.74

[back to top]

Number of Ewing/Peripheral PNET Participants in Phase 2 With Complete Response (CR) or Partial Response (PR)

Frequency of Ewing sarcoma and peripheral PNET participants with Complete Response (CR) or Partial Response (PR) per the Response Evaluation Criteria In Solid Tumors (RECIST) (NCT02116777)
Timeframe: Up to 24 months

InterventionParticipants (Count of Participants)
Treatment (Talazoparib, Temozolomide): Phase 20

[back to top]

Number of Solid Tumor Patients With Complete Response (CR) or Partial Response (PR)

Frequency of solid tumor participants with Complete Response (CR) or Partial Response (PR) per the Response Evaluation Criteria In Solid Tumors (RECIST) (NCT02116777)
Timeframe: Up to 24 months

InterventionParticipants (Count of Participants)
Treatment (Talazoparib, Temozolomide): Phase 11

[back to top]

T Max of Talazoparib

Median with minimum and maximum for the time at which the maximum (peak) serum concentration occurs. (NCT02116777)
Timeframe: Cycle 1 Day 1 pre-dose, and 1, 2, 4, 8 and 24 hours after the first talazoparib dose.

InterventionHours (Median)
Treatment (Talazoparib, Temozolomide): Phase 12

[back to top]

T Max of Talazoparib in Combination With Temozolomide

Median with minimum and maximum for the time at which the maximum (peak) serum concentration occurs. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the talazoparib dose

InterventionHours (Median)
Treatment (Talazoparib, Temozolomide): Phase 11.04

[back to top]

T Max of Temozolomide in Combination With Talazoparib

Median with minimum and maximum for the time at which the maximum (peak) serum concentration occurs. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the temozolomide dose

InterventionHours (Median)
Treatment (Talazoparib, Temozolomide): Phase 11.03

[back to top]

The Number of Participants With Dose Limiting Toxicities to Determine the Maximum Tolerated Dose of Temozolomide and Talazoparib Combination Therapy

The Maximum Tolerated Dose (MTD) reflects the highest dose of Talazoparib (BMN 673) when combined with a dose of temozolomide that did not cause a Grade 3 or higher toxicity in children with refractory or recurrent solid tumors. (NCT02116777)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
400 mcg/m²/Dose BMN 673 QD+20mg/m²/Dose TEM,Max 800 mcg/Day0
400 mcg/m²/Dose BMN 673 BID+20mg/m²/Dose TEM,Max 800 mcg/Day0
600 mcg/m²/Dose BMN 673 BID+20mg/m²/Dose TEM,Max 1000 mcg/Day1
600 mcg/m²/Dose BMN 673 BID+30mg/m²/Dose TEM, Max 1000 mcg/Day2
600 mcg/m²/Dose BMN 673 BID+40mg/m²/Dose TEM, Max 1000 mcg/Day2
600 mcg/m²/doseBMN 673 BID+55mg/m²/Dose TEM, Max 1000 mcg/Day2
600 mcg/m²/Dose BMN 673 BID+30 mg/m²/Dose TEM,Max 1000 mcg/Day0

[back to top]

Clearance of Talazoparib in Combination With Temozolomide

Median with minimum and maximum for the rate of elimination of the drug. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the talazoparib dose

InterventionL/hr/m² (Median)
Treatment (Talazoparib, Temozolomide): Phase 13.08

[back to top]

AUC of Talazoparib in Combination With Temozolomide

Median with minimum and maximum area under the drug concentration over time curve (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the talazoparib dose

Interventionh•µg/L (Median)
Treatment (Talazoparib, Temozolomide): Phase 182.08
Treatment (Talazoparib, Temozolomide): Phase 1173.08

[back to top]

Accumulation Half-life of Talazoparib in Combination With Temozolomide.

Median with minimum and maximum for the time required for the serum concentration to fall to 50% of its starting dose. (NCT02116777)
Timeframe: Cycle 1 Day 1 pre-dose, and 1, 2, 4 and 8 hours after the first talazoparib dose. Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the talazoparib dose

InterventionHours (Median)
Treatment (Talazoparib, Temozolomide): Phase 146.8

[back to top]

Accumulation Ratio of Talazoparib in Combination With Temozolomide

Median with minimum and maximum of the accumulation ratio. (NCT02116777)
Timeframe: Cycle 1 Day 1 pre-dose, and 1, 2, 4 and 8 hours after the first talazoparib dose. Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the talazoparib dose

InterventionRatio (Median)
Treatment (Talazoparib, Temozolomide): Phase 13.34

[back to top]

All Cycle 1 Toxicities >=Grade 3

The number of patients with at least one toxicity (DLT or non-DLT) in cycle 1 that is at least possibly attributable to study agent (NCT02116777)
Timeframe: Up to 28 days

InterventionParticipants (Count of Participants)
400 mcg/m²/Dose BMN 673 QD+20mg/m²/Dose TEM,Max 800 mcg/Day0
400 mcg/m²/Dose BMN 673 BID+20mg/m²/Dose TEM,Max 800 mcg/Day0
600 mcg/m²/Dose BMN 673 BID+20mg/m²/Dose TEM,Max 1000 mcg/Day2
600 mcg/m²/Dose BMN 673 BID+30mg/m²/Dose TEM, Max 1000 mcg/Day7
600 mcg/m²/Dose BMN 673 BID+40mg/m²/Dose TEM, Max 1000 mcg/Day5
600 mcg/m²/doseBMN 673 BID+55mg/m²/Dose TEM, Max 1000 mcg/Day3
600 mcg/m²/Dose BMN 673 BID+30mg/m²/Dose TEM,Max 1000 mcg/Day5

[back to top]

AUC of Talazoparib

Median with minimum and maximum for the area under the drug concentration over time curve. (NCT02116777)
Timeframe: Cycle 1 Day 1 pre-dose, and 1, 2, 4 and 8 hours after the first talazoparib dose

Interventionh•µg/L (Median)
Treatment (Talazoparib, Temozolomide): Phase 117.55

[back to top]

AUC of Temozolomide in Combination With Talazoparib

Median with minimum and maximum for the area under the drug concentration over time curve. (NCT02116777)
Timeframe: Cycle 1 Day 5 or 6 pre-dose, and 1, 2, 4 and 8 hours after the temozolomide dose.

Interventionh•µg/L (Median)
Treatment (Talazoparib, Temozolomide): Phase 14352.16

[back to top]

Overall Survival (OS)

The distribution of OS for each arm will be estimated using the Kaplan-Meier method and compared with a stratified logrank test. (NCT02152982)
Timeframe: 83 months

Interventionmonths (Median)
Arm I (Temozolomide, Veliparib)24.8
Arm II (Temozolomide, Placebo)28.1

[back to top]

Objective Tumor Response

"Defined as complete response (CR) or partial response (PR) as specified in the Revised Assessment in Neuro-Oncology criteria. An objective tumor response will be evaluated for each patient and the tumor response count will be summarized for each arm and compared using the Chi-square test. For CR, all of the following must be true:~disappearance of all enhancing measurable and non-measurable disease; no new enhancing lesions; stable or improved non-enhancing lesions; patients must be off corticosteroids; stable or improved clinically~A PR requires all of the following: > 50% decrease in sum of products of perpendicular diameters of all measurable enhancing lesions compared with baseline; no progression of non-measurable disease; no new lesions; stable or improved non-enhancing lesions on same or lower dose of corticosteroids compared with baseline scan; steroid dose should be same or lower compared with baseline scan; stable or improved clinically" (NCT02152982)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
Arm I (Temozolomide, Veliparib)34
Arm II (Temozolomide, Placebo)37

[back to top]

Interaction With Optune Device

Cox proportional hazards model will be used to evaluate whether there is a potential interaction between the treatment arm and the Optune device. If an interaction is detected, separate analyses of treatment effect (using Cox models) will be done for patients treated with the Optune device and patients who were not treated with the Optune device. (NCT02152982)
Timeframe: 5 years

,
Interventionparticipants (Number)
Reporting actual Optune useNo actual Optune use
Arm I (Temozolomide, Veliparib)33125
Arm II (Temozolomide, Placebo)33135

[back to top]

Progression-free Survival (PFS)

"The distribution of PFS for each arm will be estimated using the Kaplan-Meier method, and be compared using Cox proportional hazard models with all stratification factors adjusted. Progression (PD): Defined by any of the following:~> 25% increase in sum of products of perpendicular diameters of enhancing lesions, compared with the smallest tumor measurement obtained either at baseline or best response~Significant increase in T2/FLAIR non-enhancing lesion on stable or increasing doses of corticosteroids compared with baseline scan or best response after therapy initiation (stable doses of steroids include patient not on steroids) not caused by comorbid events~Any new lesion~Clear clinical deterioration not attributable to other causes apart from tumor or change in corticosteroid dose~Failure to return for evaluation as a result of death or deteriorating condition~Clear progression of non-measurable disease" (NCT02152982)
Timeframe: 83 months

Interventionmonths (Median)
Arm I (Temozolomide, Veliparib)12.1
Arm II (Temozolomide, Placebo)13.2

[back to top]

Overall Adverse Event Rates for Grade 3 or Higher Adverse Events

Assessed using National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 (version 5 beginning April 1, 2018). The overall adverse event rates for grade 3 or higher adverse events will be summarized and be compared using Chi-Square or Fisher's Exact tests between treatment arms. The maximum grade for each type of treatment-related adverse event will be recorded for each patient, and frequency tables for each arm will be reviewed to determine patterns. Treatment-related adverse events will be tabulated for each arm. (NCT02152982)
Timeframe: 5 years

Interventionparticipants with at least 1 grade 3+ AE (Number)
Arm I (Temozolomide, Veliparib)94
Arm II (Temozolomide, Placebo)137

[back to top]

Percentage of Participants With an Unacceptable Toxicity

The percentage of patients who experience an unacceptable toxicity defined as any Grade 3 toxicity at least possibly attributed to the vaccine (or vaccine + TMZ and/or RT) that does not resolve to baseline within 3 weeks, any Grade 3 hypersensitivity reactions requiring steroids, any Grade 4 toxicity, including neurologic events not due to progressive disease, or any life threatening-event not attributable to concomitant medication, co-morbid event, or disease progression. (NCT02193347)
Timeframe: Date of consent through 2 months after the last vaccination

Interventionpercentage of participants (Number)
PEPIDH1M Vaccine0

[back to top]

Percentage of Patients With a Positive Vaccine Response After 3 Post-Surgery Vaccines as Measured by IFNγ ELIspot

Assess the immunogenicity of the PEPIDH1M vaccine with adjuvant TMZ using ELISpot. This will be done by finding the difference between the number of SFC after 3 post-surgery vaccines and the number of SFC at baseline for each evaluable subject. Immunogenicity will then be summarized by the percentage of evaluable subjects who have a positive vaccine response after 3 post-surgery vaccines. A response will be considered positive if the difference in SFC measurement is greater than 20 SFC per 10^6 lymphocytes after determining the level of detection in the IDH ELISpot. (NCT02193347)
Timeframe: From time of pheresis #1, one day prior to first pre-surgery vaccine dose, until the time of the third post-surgery vaccination, an expected average of 24 weeks after study initiation

InterventionPercentage of evaluable participants (Number)
PEPIDH1M Vaccine42.86

[back to top]

Median Overall Survival (OS) by Arm and MGMT Methylation Status

Median OS depending on treatment arm in patients with methylated MGMT (NCT02209948)
Timeframe: Through the whole study. 4 years. The median follow up for each patient was 33.4 months

Interventionmonths (Median)
Temozolomide20.7
Without Treatment27.1

[back to top]

Progresion Free Survival Median Values

It will be measured following Response assessment in neuro-oncology (RANO) guidelines: progression-free survival (NCT02209948)
Timeframe: Through the whole study. 4 years. The median follow up for each patient was 33.4 months

Interventionmonths (Median)
Temozolomide9.5
Without Treatment7.77

[back to top]

Overall Survival

Time between start of treatment and death (NCT02209948)
Timeframe: Through the whole study. 4 years. The median follow up for each patient was 33.4 months

Interventionmonths (Median)
Temozolomide18.2
Without Treatment23.3

[back to top]

Progression Free Survival at 6 Month

"Percentage of patients without progression of disease and time between start of treatment and progression of disease.~The progression disease is defined as the time from the date of randomization to the date of progression defined according to the RANO criteria." (NCT02209948)
Timeframe: 6 month

Interventionpercentage of patients (Number)
Temozolomide61.3
Without Treatment55.7

[back to top]

Number of Participants With Adverse Effects

Total number of patients presenting adverse events, stratified by type of event and grade. Adverse Events of special interest: Only relevant differences in toxicity by arm. (NCT02209948)
Timeframe: Through the whole study. 4 years

InterventionParticipants (Count of Participants)
Lymphopenia72025836Lymphopenia72025835Thrombocytopenia72025835Thrombocytopenia72025836Nausea and vomiting72025835Nausea and vomiting72025836Fatigue72025836Fatigue72025835Leucopenia72025835Leucopenia72025836
Grade 1-2Grade 3-4not affected
Temozolomide52
Without Treatment33
Temozolomide3
Temozolomide25
Without Treatment46
Temozolomide36
Without Treatment17
Temozolomide2
Temozolomide42
Without Treatment62
Temozolomide30
Without Treatment10
Without Treatment0
Temozolomide50
Without Treatment69
Temozolomide35
Without Treatment21
Temozolomide0
Temozolomide45
Without Treatment58
Temozolomide29
Without Treatment20
Temozolomide1
Without Treatment59

[back to top]

Median Progression-free Survival (PFS) by Arm and MGMT Methylation Status

Median Progression Free Survival depending on treatment arm in patients with MGMT methylation (NCT02209948)
Timeframe: Through the whole study. 4 years. The median follow up for each patient was 33.4 months

Interventionmonths (Median)
Temozolomide11.4
Without Treatment8.5

[back to top]

DCR After 12 Months

"All tumour assessments were performed using the RECIST criteria (1.1). CT-scan or MRI could be used for as method of tumour measurement and the same method of tumour measurement was used throughout the study for each subject. CT scans/MRI were performed at screening or baseline visit then at baseline, weeks 12, 24, 36, 48 (end of study) and at study withdrawal or at anytime during the study in the case of any clinical or biological signs of tumour progression.~The DCR was defined as the proportion of subjects with a response of CR, PR or SD after 6 months combination treatment followed by either 6 months of lanreotide ATG 120 mg maintenance treatment or no treatment. The DCR was described in the ITT population along with its 95% CI and was compared to 45% with an exact binomial proportion test. The LOCF method was used to replace missing assessments at the end of the maintenance phase." (NCT02231762)
Timeframe: 12 months

Interventionpercentage of subjects (Number)
Maintenance Phase - Functioning NET, Lanreotide54.5
Maintenance Phase - Non-functioning NET, Lanreotide71.4
Maintenance Phase - Non-functioning NET, No Treatment41.7

[back to top]

Disease Control Rate (DCR) After 6 Months

"All tumour assessments were performed using the Response Evaluation Criteria In Solid Tumours (RECIST) criteria (1.1). Computer Tomography (CT-scan) or Magnetic Resonance Imaging (MRI) could be used for as method of tumour measurement and the same method of tumour measurement was used throughout the study for each subject. CT scans/MRI were performed at screening or baseline visit then at weeks 12, 24 and at early withdrawal or at anytime during the study in the case of any clinical or biological signs of tumour progression.~The DCR was defined as the proportion of subjects with a response of CR, PR or SD after 6 months of combination treatment and was described in the ITT population along with its 95% Confidence Interval (CI) and was compared to 45% with an exact binomial proportion test. The Last Observation Carried Forward (LOCF) method was used to replace missing assessments at the end of the combination phase." (NCT02231762)
Timeframe: 6 months

Interventionpercentage of subjects (Number)
Combination Phase73.5

[back to top]

Duration of Response (DoR) Within 12 Months

"The DoR is an estimation of the time from first documented objective response (CR or PR) to the first date of progressive disease (PD) or death due to disease progression for subjects who experienced an objective response within the first 12 months of treatment (combination and maintenance phases).~The Kaplan-Meier method was used to estimate the median DoR and its 95% CI for subjects in the ITT population who had an objective response." (NCT02231762)
Timeframe: 12 months

Interventionmonths (Median)
Intention-to-treat (ITT) PopulationNA

[back to top]

Progression-Free Survival (PFS) Within 12 Months

"PFS was defined as the time from the date of treatment start to the date of the first documented disease progression or death due to any cause within the first 12 months of treatment. If a subject had not progressed or died after 12 months of treatment or when any further anti-neoplastic therapy was received, PFS was censored at the time of the last tumour assessment before the analysis cut-off date or the anti-neoplastic therapy date.~A Kaplan-Meier estimate of the PFS was calculated to determine the number of subjects at risk. Median PFS time (50% of subjects who would not progress or die) of the ITT population is presented along with 95 % CI." (NCT02231762)
Timeframe: 12 months

Interventionmonths (Median)
Intention-to-treat (ITT) Population11.1

[back to top]

Time To Response (TtR) Within 12 Months

"TtR was defined as the time from the date of treatment start to the date of the first documented objective response (CR or PR) within the first 12 months of treatment (combination and maintenance phases). A Kaplan Meier estimate of the TtR survival function was constructed.~The Kaplan-Meier method was used to estimate the median TtR and its 95% CI for subjects in the ITT population (50% of subjects were expected to have a CR or PR at this time)." (NCT02231762)
Timeframe: 12 months

Interventionmonths (Median)
Intention-to-treat (ITT) PopulationNA

[back to top]

The Number of Subjects With a Symptomatic Response After 12 Months - Maintenance Phase

"Symptomatic response was evaluated as absolute change from baseline in the number of episodes of the lead symptoms (i.e. diarrhoea and flushing) using the mean of the last 3 days before the visit, at each visit, as compared to baseline.~Symptomatic responses were categorised as: Reduction, Increase or Stability of occurrences of diarrhoea / Reduction, Increase or Stability of occurrences of flushing.~The number of subjects in each response category at week 48 (end of study) is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Diarrhoea - ReductionDiarrhoea - IncreaseDiarrhoea - StabilityDiarrhoea - MissingFlushing - ReductionFlushing - IncreaseFlushing - StabilityFlushing - Missing
Maintenance Phase - Functioning NET, Lanreotide41332333

[back to top]

The Number of Subjects With a Biochemical Response Using CgA Levels After 12 Months

"Blood samples for CgA blood tumour marker analysis were taken at baseline, weeks 12, 24, 36, 48 (end of study) and at early withdrawal. The biochemical response after 12 months combination and maintenance treatment was estimated for subjects with abnormal CgA levels at baseline. Abnormal CgA levels were defined as above the upper limit of normal range (≥100 mcg/L).~Biochemical response based on CgA levels was categorised as: PR (decrease of CgA ≥50 % compared to the baseline CgA), SD (decrease < 50% or an increase ≤ 25% compared to the baseline CgA) or PD (defined as an increase ≥ 25%, compared to the baseline CgA).~The number of subjects in each response category at each time point in the maintenance phase is presented. Analysis was only carried out on subjects in the ITT population who had abnormal CgA at baseline." (NCT02231762)
Timeframe: 12 months

,,
InterventionParticipants (Count of Participants)
Week 24 - PDWeek 24 - SDWeek 24 - PRWeek 24 - MissingWeek 36 - PDWeek 36 - SDWeek 36 - PRWeek 36 - MissingWeek 48 - PDWeek 48 - SDWeek 48 - PRWeek 48 - MissingEarly Withdrawal - PDEarly Withdrawal - SDEarly Withdrawal - PREarly Withdrawal - Missing
Maintenance Phase - Functioning NET, Lanreotide2300120010101002
Maintenance Phase - Non-functioning NET, Lanreotide1431141112202010
Maintenance Phase - Non-functioning NET, No Treatment2232324023101010

[back to top]

The Number of Subjects With a Biochemical Response Using 5-Hydroxy-Indol-Amino-Acid (HIAA) Levels After 6 Months

"Urine samples for 5-HIAA urinary tumour marker analysis were taken at at baseline, weeks 12, 24and early withdrawal.~Biochemical response based on 5-HIAA levels was categorised as: Response (5-HIAA reduction compared to baseline) or Progression (5-HIAA increase compared to baseline).~The number of subjects in each response category at each time point in the combination phase is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Week 12 - ProgressionWeek 12 - ResponseWeek 12 - Not evaluableWeek 12 - MissingWeek 24 - ProgressionWeek 24 - ResponseWeek 24 - Not evaluableWeek 24 - MissingEarly Withdrawal - ProgressionEarly Withdrawal - ResponseEarly Withdrawal - Not EvaluableEarly Withdrawal - Missing
Combination Phase - Functioning NET461663130107

[back to top]

The Number of Subjects With a Symptomatic Response After 6 Months

"Symptomatic response was evaluated as absolute change from baseline in the number of episodes of the lead symptoms (i.e. diarrhoea and flushing) using the mean of the last 3 days before the visit, at each visit, as compared to baseline.~Symptomatic responses were categorised as: Reduction, Increase or Stability of occurrences of diarrhoea / Reduction, Increase or Stability of occurrences of flushing.~The number of subjects in each response category at week 24 (end of the combination phase) is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Diarrhoea - ReductionDiarrhoea - IncreaseDiarrhoea - StabilityDiarrhoea - MissingFlushing - ReductionFlushing - IncreaseFlushing - StabilityFlushing - Missing
Combination Phase - Functioning NET42564436

[back to top]

The Number of Subjects With a Biochemical Response Using 5-HIAA Levels After 12 Months

"Urine samples for 5-HIAA urinary tumour marker analysis were taken at baseline, weeks 12, 24, 36, 48 (end of study) and early withdrawal.~Biochemical response based on 5-HIAA levels was categorised as: Response (5-HIAA reduction compared to baseline) or Progression (5-HIAA increase compared to baseline).~The number of subjects in each response category at each time point in the maintenance phase is presented. Analysis was only carried out on subjects in the ITT population with functioning NET." (NCT02231762)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Week 24 - ProgressionWeek 24 - ResponseWeek 24 - Not evaluableWeek 24 - MissingWeek 36 - ProgressionWeek 36 - ResponseWeek 36 - Not evaluableWeek 36 - MissingWeek 48 - ProgressionWeek 48 - ResponseWeek 48 - Not evaluableWeek 48 - MissingEarly Withdrawal - ProgressionEarly Withdrawal - ResponseEarly Withdrawal - Not evaluableEarly Withdrawal - Missing
Maintenance Phase - Functioning NET, Lanreotide6311330342020003

[back to top]

DCR by O6-methylguanine-DNA Methyl-transferase (MGMT) Expression and Methylation and Somatostatin Receptor (SSTR) Expression After 6 Months

"In all subjects whose tumour tissue was available, MGMT expression/methylation and SSTR expression was analysed. After 6 months, the DCR (SD+PR+CR) by MGMT methylation and expression and by SSTR 2a and SSTR 5 expression was evaluated.~DCR in response to MGMT methylation and expression results are presented. SSTR 2a and SSTR 5 expression is categorised as: No Receptors, Cytoplasmatic Expression (CE), Focal Expression (FE), Complete Circumferent Membrane Expression (CCME).~The DCR was defined as the proportion of subjects with a response of CR, PR or SD after 6 months of combination treatment within each methylation/expression category. The DCR was described in the ITT population along with its 95% CI and was compared to 45% with an exact binomial proportion test." (NCT02231762)
Timeframe: 6 months

Interventionpercentage of subjects (Number)
MGMT MethylationMGMT No methylationMGMT ExpressionMGMT No expressionSSTR 2a FESSTR 2a CCMESSTR 5 - No ReceptorsSSTR 5 CESSTR 5 FE
Combination Phase100.084.690.970.086.772.775.0100.081.8

[back to top]

Quality of Life Gastrointestinal Neuroendocrine Tumour 21 Questionnaire (QLQ-GI.NET21): Mean Change From Baseline at 6 Months

Subjects were instructed to complete the QLQ-GI.NET21 questionnaire at baseline, weeks 12, 24 or at early withdrawal. It contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Each individual subscore was transformed to range from 0 to 100. The mean change from baseline at week 24 (end of combination phase) is presented with a higher score representing a higher level response. Thus, a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 6 months

InterventionUnits on a scale (Mean)
Endocrine symptomsGastrointestinal (G.I.) symptomsTreatment related symptomsSocial functionDisease related worriesMuscle/bone pain symptomsBody imageWeight gainInformation/communication functionSexual function
Combination Phase-1.05.73.62.31.61.00.0-11.8-9.4-4.8

[back to top]

QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months

Subjects were instructed to complete the QLQ-GI.NET21 questionnaire at baseline, weeks 12, 24, 36, 48 (end of study) or at early withdrawal. It contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Answers were converted into grading scale, with values between 0 and 100. Each individual subscore was transformed to range from 0 to 100. The mean change from baseline at week 48 (end of study) is presented with a higher score representing a higher level response. Thus, a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 12 months

Interventionunits on a scale (Mean)
Endocrine symptomsG.I. symptomsSocial functionDisease related worriesMuscle/bone pain symptomsBody imageWeight gainInformation/communication functionSexual function
Maintenance Phase - Non-functioning NET, No Treatment1.66.7-6.3-3.24.84.8-9.5-4.80.0

[back to top]

Pharmacokinetic (PK) Results: Lanreotide ATG 120 mg Serum Concentrations Within 12 Months

"Lanreotide ATG levels were measured in a subset of subjects to evaluate if temozolomide co-treatment had an impact on lanreotide serum concentration over a 12 month period.~Blood samples were collected for the determination of lanreotide ATG in serum at baseline, weeks 4, 12, 24 and 48 (end of study).~The concentrations of lanreotide ATG in serum were determined by a validated radioimmunoassay analysis method with a lower limit of quantitation of 0.08 nanograms [ng]/mL).~Serum concentrations of lanreotide ATG at each of the time points in the combination and maintenance phase are presented. Only subjects with data available for analysis are presented." (NCT02231762)
Timeframe: Baseline (week 1) and weeks 4, 12, 24 and 48

Interventionng/mL (Mean)
BaselineWeek 4Week 12Week 24Week 48
PK Subset0.442.455.065.833.68

[back to top]

European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life (QoL) Core 30 Questionnaire (QLQ-C30): Mean Change From Baseline at 6 Months

Subjects were instructed to complete the QLQ-C30 questionnaire at baseline, weeks 12, 24 or at early withdrawal. The first 28 questions used a 4-point scale (1=not at all, 2=a little, 3=quite a bit, 4=very much) for evaluating 5 functional scales (physical, role, emotional, cognitive, social), 3 symptom scales (fatigue, nausea/vomiting, pain) & 6 other single items. The last 2 questions represented subject's assessment of overall health & quality of life, coded on a 7-point scale (1=very poor to 7=excellent). The mean change from baseline at week 24 (end of the combination phase) is presented for global health status (scoring of questions 29 & 30) and 5 functional scales, 3 symptom scales and other single items (scoring of questions 1 to 28). Each individual subscore was transformed to range from 0 to 100. A higher score represents a higher level response. Thus, a better QoL/a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 6 months

Interventionunits on a scale (Mean)
Global health statusPhysical functioningRole functioningEmotional functioningCognitive functioningSocial functioningFatigueNausea and vomitingPainDyspnoeaInsomniaAppetite lossConstipationDiarrhoeaFinancial difficulties
Combination Phase-4.9-9.6-8.3-4.7-5.9-11.86.96.9-1.012.70.02.06.9-3.92.9

[back to top]

EORTC QoL Questionnaire QLQ-C30: Mean Change From Baseline at 12 Months

Subjects were instructed to complete QLQ-C30 questionnaire at baseline, weeks 12, 24, 36, 48 (end of study) or at early withdrawal. The first 28 questions used a 4-point scale (1=not at all, 2=a little, 3=quite a bit, 4=very much) for evaluating 5 functional scales (physical, role, emotional, cognitive, social), 3 symptom scales (fatigue, nausea/vomiting, pain) & 6 other single items. The last 2 questions represented subject's assessment of overall health & quality of life, coded on a 7-point scale (1=very poor to 7=excellent). The mean change from baseline at week 48 (end of study) is presented for global health status (scoring of questions 29 & 30) and 5 functional scales, 3 symptom scales and other single items (scoring of questions 1 to 28). Each individual subscore was transformed to range from 0 to 100. A higher score represents a higher level response. Thus, a better QoL/a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 12 months

,,
Interventionunits on a scale (Mean)
Global health statusPhysical functioningRole functioningEmotional functioningCognitive functioningSocial functioningFatigueNausea and vomitingPainDyspnoeaInsomniaAppetite lossConstipationDiarrhoeaFinancial difficulties
Maintenance Phase - Functioning NET, Lanreotide-11.78.03.315.03.313.3-22.2-10.0-13.3-8.3-13.30.020.0-40.06.7
Maintenance Phase - Non-functioning NET, Lanreotide-3.1-12.5-2.1-6.2-2.1-22.9-9.74.24.24.2-8.30.0-4.28.34.2
Maintenance Phase - Non-functioning NET, No Treatment-7.1-11.4-7.1-6.02.4-4.84.82.49.59.516.714.3-4.80.09.5

[back to top]

QoL Questionnaire QLQ-GI.NET21: Mean Change From Baseline at 12 Months

Subjects were instructed to complete the QLQ-GI.NET21 questionnaire at baseline, weeks 12, 24, 36, 48 (end of study) or at early withdrawal. It contained 21 questions that used a 4-point scale (1 = Not at all, 2 = A little, 3 = Quite a bit, 4 = Very much) to evaluate 3 defined multi-item symptom scales (endocrine, gastrointestinal and treatment related side effects), 2 single item symptoms (bone/muscle pain and concern about weight loss), 2 psychosocial scales (social function and disease-related worries) and 2 other single items (sexuality and communication). Answers were converted into grading scale, with values between 0 and 100. Each individual subscore was transformed to range from 0 to 100. The mean change from baseline at week 48 (end of study) is presented with a higher score representing a higher level response. Thus, a better level of functioning/a worse level of symptoms. (NCT02231762)
Timeframe: 12 months

,
Interventionunits on a scale (Mean)
Endocrine symptomsG.I. symptomsTreatment related symptomsSocial functionDisease related worriesMuscle/bone pain symptomsBody imageWeight gainInformation/communication functionSexual function
Maintenance Phase - Functioning NET, Lanreotide-13.3-4.00.0-6.7-6.7-6.70.0-20.00.00.0
Maintenance Phase - Non-functioning NET, Lanreotide-5.60.0-11.19.71.44.24.29.50.00.0

[back to top]

The Number of Subjects With a Biochemical Response Using Chromogranin-A (CgA) Levels After 6 Months

"Blood samples for CgA blood tumour marker analysis were taken at baseline, weeks 12, 24 and at early withdrawal. The biochemical response after 6 months combination treatment was estimated for subjects with abnormal CgA levels at baseline. Abnormal CgA levels were defined as above the upper limit of normal range (≥100 micrograms/litre [mcg/L]).~Biochemical response based on CgA levels was categorised as: PR (decrease of CgA ≥ 50%, compared to the baseline CgA), SD (decrease < 50 % or an increase ≤25%, compared to the baseline CgA) or PD (defined as an increase ≥25 %, compared to the baseline CgA).~The number of subjects in each response category at each time point in the combination phase is presented. Analysis was only carried out on subjects in the ITT population who had abnormal CgA at baseline." (NCT02231762)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Week 12- PDWeek 12 - SDWeek 12- PRWeek 12 - MissingWeek 24 - PDWeek 24 - SDWeek 24 - PRWeek 24 - MissingEarly Withdrawal - PDEarly Withdrawal - SDEarly Withdrawal - PREarly Withdrawal - Missing
Combination Phase815101597012114

[back to top]

Number of Participants With Adverse Events

Safety and tolerability were assessed by monitoring the frequency, duration, severity and attribution of adverse events and evaluating changes in laboratory values and vital signs. (NCT02288897)
Timeframe: Assessed every 4 weeks until 28 days after last treatment; median duration of treatment was 11.8 and 9.5 weeks in each treatment group, respectively.

InterventionParticipants (Count of Participants)
PV-1014
Chemotherapy or Oncolytic Viral Therapy8

[back to top]

Overall Survival (OS)

OS was documented at 12 week intervals commencing on withdrawal from active study participation. Documentation was made by subject clinic visit or other personal contact, telephonic contact, review of medical records, or other unequivocal evidence of survival status. OS was estimated via Kaplan-Meier analysis, and was defined as the interval from randomization to death; subjects who did not have an event of death were censored at their last assessment date. (NCT02288897)
Timeframe: Assessed every 12 weeks upon withdrawal from active study participation until death, withdrawal of consent, or study termination; median follow-up time for survival was 82.4 weeks, maximum was 167.0 weeks.

InterventionMonths (Median)
PV-10NA
Chemotherapy or Oncolytic Viral TherapyNA

[back to top]

Complete Response Rate (CRR)

CRR was assessed using RECIST v1.1 criteria, and required disappearance of all target and non-target lesions. (NCT02288897)
Timeframe: Assessed every 12 weeks until disease progression, withdrawal of consent, or study termination; median follow-up time for CRR was 26.6 weeks, maximum was 125.8 weeks.

InterventionParticipants (Count of Participants)
PV-103
Chemotherapy or Oncolytic Viral Therapy2

[back to top]

Progression-free Survival (PFS)

"PFS was estimated via Kaplan-Meier analysis using Response Evaluation Criteria In Solid Tumors (RECIST v1.1) criteria. Subjects who did not have an event of progression or death were censored at their last assessment date.~Assessment of progression was performed by the sponsor based on review of lesion measurement and clinical progression data reported by each investigator. Events signaling progression included increase in size and/or number of study lesions, onset of visceral metastatic disease, and death. For target lesions (assigned prior to randomization), complete response (CR) required disappearance of all target lesions; partial response (PR) required >= 30% decrease in sum of the longest diameter (SLD) of all target lesions; progressive disease (PD) required >=20% increase in SLD of target lesions. Non-target lesions were followed for CR, PD, or non-CR/non-PD status using equivalent thresholds. Progression occurred when PD was observed in either target or non-target lesion." (NCT02288897)
Timeframe: Assessed every 12 weeks until disease progression, withdrawal of consent, or study termination; progression detected between formal assessments was documented at the time of detection; median follow-up time for PFS was 26.6 weeks, maximum was 125.8 weeks.

InterventionMonths (Median)
PV-106.1
Chemotherapy or Oncolytic Viral Therapy8.6

[back to top]

Change in Domain Scores From Baseline Using the Patient Reported Skindex-16 Instrument

In this exploratory endpoint, changes in Skindex-16 self-assessment scores were evaluated vs Day 1 baseline domain scores: symptom domain (items 1-4); emotional domain (items 5-11); and functioning domain (items 12-16). The Skindex-16 instrument solicits response to the extent of bother during the preceding week by 16 items (e.g., itching, hurting, worry, impact on daily activities), using a score from 0 (never bothered) to 6 (always bothered) for each item. Item scores are transformed to 0 to 100 scale, and domain scores are calculated as the average of the item scores comprising the domain. A lower domain score at baseline signifies lower impact of that domain; a decrease in domain score from baseline signifies improvement in that domain. Median baseline score and change from baseline over the study interval is presented for each domain. (NCT02288897)
Timeframe: Assessed at baseline (Day 1 at start of study treatment) and at the end of each treatment cycle (every 4 or 6 weeks) until disease progression, withdrawal of consent, or study termination; median follow-up time was 26.6 weeks, maximum was 125.8 weeks.

,
InterventionScores on a Scale (0-100) (Median)
Baseline Symptom Domain ScoreChange in Symptom Domain Score from BaselineBaseline Emotional Domain ScoreChange in Emotional Domain Score from BaselineBaseline Functioning Domain ScoreChange in Functioning Domain Score from Baseline
Chemotherapy or Oncolytic Viral Therapy18.8-6.316.7-4.88.36.7
PV-102.1-2.120.2-10.11.7-1.7

[back to top]

Duration of Complete Response (DCR)

DCR was estimated via Kaplan-Meier analysis for participants achieving a complete response, and was defined as the interval from first complete response to disease progression or death; responders who did not have an event of progression or death were censored at their last assessment date. Complete response was assessed using RECIST v1.1 criteria, and required disappearance of all target and non-target lesions. (NCT02288897)
Timeframe: Assessed every 12 weeks until disease progression, withdrawal of consent, or study termination; median follow-up time for DCR was 26.7 weeks, maximum was 77.7 weeks.

InterventionMonths (Median)
PV-10NA
Chemotherapy or Oncolytic Viral TherapyNA

[back to top]

Pharmacokinetic Profile of BBI608 and Temozolomide When Administered in Combination With Temozolomide as Assessed by the Area Under the Curve

The area under the curve of BBI608, from time 0 to the last quantifiable concentration, calculated by a combination of linear and logarithmic trapezoidal methods (linear up/log down method) (NCT02315534)
Timeframe: On Day 1 and Day 5 after the first dosing, prior to dosing and 1, 2, 3, 5, 5h40m (day 1 only), 6, 7, 8 and 24 hours after first dose of BBI608

Interventionh*ng/ml (Geometric Mean)
Candidates Whom Surgery is Recommended4820
Candidates Whom Surgery is Not Recommended4200

[back to top]

Dose-limiting Toxicities (DLTs)

Number of patients who experienced a dose limiting toxicity following a dosing of BBI608 (NCT02315534)
Timeframe: 28 days after first administration of combination treatment (BBI608+TMZ)

InterventionParticipants (Count of Participants)
Candidates Whom Surgery is Recommended.0
Candidates Whom Surgery is Not Recommended0

[back to top]

Progression Free Survival (PFS)-6

To assess the effect of BBI608 + temozolomide (TMZ) therapy defined as the percentage of patients who have survived without objective disease progression for at least 6 months after treatment per neuro-oncology (RANO) criteria who had evaluable disease at baseline. PFS-6 is defined as the percentage of patients who survived without objective disease progression per RANO criteria for at least 6 months after treatment. (NCT02315534)
Timeframe: From the time of exposure to study drug to first objective documentation of disease progression or death due to any cause, assessed up to 6 months

Interventionpercentage of participants (Number)
Candidates Whom Surgery is Not Recommended.28.07

[back to top]

Overall Survival (OS)

To assess the effect of BBI608 + temozolomide (TMZ) on the overall survival of patients with recurrent or progressive glioblastoma multiforme (GBM) who had not received prior treatment with bevacizumab or other anti-vascular endothelial growth factor agents who either were eligible or not eligible for surgical resection. (NCT02315534)
Timeframe: From the time of exposure to study drug to death from any cause. If patient discontinued study drug, they were assessed the first 3 months after discontinuation, then every 3 months up to 1 year, then every 6 months thereafter until death.

Interventionmonths (Median)
Candidates Whom Surgery is Not Recommended8.05

[back to top]

Disease Control Rate (DCR)

To assess the percentage of patients that had evaluable disease at baseline with a documented complete response, partial response, and stable disease (CR + PR + SD) based on the Response Assessment in Neuro-Oncology (RANO) criteria out of all patients who received at least one dose of any study drug and had evaluable disease at baseline. (NCT02315534)
Timeframe: 4 weeks

Interventionpercentage of patients (Number)
Candidates Whom Surgery is Not Recommended30

[back to top]

Progression Free Survival (PFS)-12

To assess the effect of BBI608 + temozolomide (TMZ) therapy defined as the percentage of patients who have survived without objective disease progression for at least 12 months after treatment per neuro-oncology (RANO) criteria who had evaluable disease at baseline. PFS-12 is defined as the percentage of patients who survived without objective disease progression per RANO criteria for at least 12 months after treatment. (NCT02315534)
Timeframe: From the time of exposure to study drug to first objective documentation of disease progression or death due to any cause, up to 12 months

Interventionpercentage of participants (Number)
Candidates Whom Surgery is Not Recommended16.84

[back to top]

Overall Response Rate (ORR)

The proportion of patients with a documented complete response and partial response (CR + PR) based on RANO criteria. (NCT02315534)
Timeframe: 4 weeks

Interventionpercentage (Number)
Candidates Whom Surgery is Not Recommended3

[back to top]

Incidence of Treatment-Emergent Adverse Events

The safety of the combination of SGT-53 and Temozolomide was assessed by analysis of adverse experiences, clinical laboratory tests and physical examinations. (NCT02340156)
Timeframe: Study drug initiation through 30 days after the last dose of study drug or EOS, whichever is later, approximately 90 days.

InterventionParticipants (Number)
SeizureElectrocardiogram QT corrected interval prolongedSkin ulceration
SGT-53 With Temozolomide111

[back to top]

AUC0-∞

Extent of absorption of Temozolomide from time (0) to infinity (∞) will be measured after oral administration of the test product (Dralitem®, Monte Verde S.A.) or the reference product (Temodal®, Schering-Plough). (NCT02343081)
Timeframe: 0, 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 1.75, 2.0, 2.5, 3.0, 4.0, 6.0, 8.0, 10.0 hours on Days 3 and 4

Interventionmcg*h/mL (Mean)
Temodal31.817
Dralitem32.290

[back to top]

Kel

Rate at which Temozolomide is removed from the body. (NCT02343081)
Timeframe: 0, 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 1.75, 2.0, 2.5, 3.0, 4.0, 6.0, 8.0, 10.0 hours on Days 3 and 4

Intervention1/h (Mean)
Temodal0.3743
Dralitem0.3691

[back to top]

AUC0-t

Extent of absorption of Temozolomide from time (0) to the last quantifiable concentration (t) will be measured after the oral administration of the test product (Dralitem®, Monte Verde S.A.) or the reference product (Temodal®, Schering-Plough) (NCT02343081)
Timeframe: 0, 0.25, 0.5, 0.75, 1.0, 1.25, 1.75, 2.0, 2.5, 3.0, 4.0, 6.0, 8.0, 10.0 hours on Days 3 and 4

Interventionmcg*h/mL (Mean)
Temodal30.796
Dralitem31.104

[back to top]

T1/2

Time required for Temozolomide plasma concentration to decrease by 50% (NCT02343081)
Timeframe: 0, 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 1.75, 2.0, 2.5, 3.0, 4.0, 6.0, 8.0, 10.0 hours on Days 3 and 4

Interventionhours (Mean)
Temodal1.87
Dralitem1.89

[back to top]

Cmax

Rate of absorption of Temozolomide (Cmax) will be measured after the oral administration of the test product (Dralitem®, Monte Verde S.A.) or the reference product (Temodal®, Schering-Plough). (NCT02343081)
Timeframe: 0, 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 1.75, 2.0, 2.5, 3.0, 4.0, 6.0, 8.0, 10.0 hours on Days 3 and 4

Interventionmcg/mL (Mean)
Temodal11.108
Dralitem10.575

[back to top]

Pediatric Study: Half-life (t1/2) Observed for ABT-414

Half-life is the calculated time it takes for half of the drug to leave the body. (NCT02343406)
Timeframe: Samples collected Cycle 1 Days 1, 2,3,5,8,15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 5 Day 1; Day 1 of every two cycles starting with Cycle 5; and 35 days after the last dose

Interventiondays (Mean)
ABT-414_ Pediatric9.0

[back to top]

Pediatric Study: Area Under the Concentration-time-curve (AUC) Observed for Unconjugated Cys-mcMMAF

AUC is a measure of how long and how much drug or drug metabolite is present in the body after dosing. The AUC of Cys-mcMMAF, a toxic metabolite of depatuxizumab mafodotin, in the pediatric population was measured following treatment to confirm that this was comparable to adults, and that the dosing levels are appropriate for a pediatric population. (NCT02343406)
Timeframe: Samples collected Cycle 1 Days 1, 2, 3, 5, 8

Interventionng*h/mL (Mean)
ABT-414_ Pediatric14.1

[back to top]

Pediatric Study: Area Under the Concentration-time Curve (AUC) Observed for ABT-414

AUC is a measure of how long and how much drug is present in the body after dosing. The AUC of depatuxizumab mafodotin (ABT-414) in the pediatric population was measured following treatment to confirm that this was comparable to adults, and that the dosing levels are appropriate for a pediatric population. (NCT02343406)
Timeframe: Samples collected Cycle 1 Days 1, 2,3,5,8,15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 5 Day 1; Day 1 of every two cycles starting with Cycle 5; and 35 days after the last dose

Interventionµg*h/mL (Mean)
ABT-414_ Pediatric3170

[back to top]

Adult Study: Objective Response Rate (ORR)

The objective response rate (ORR) included best overall responses - complete response (CR) and partial response (PR) - assessed by the independent review committee per response assessment in neurooncology criteria (RANO) criteria from the date of randomization until disease progression or death, whichever came first. All objective responses (CR and PR) must be have been confirmed by repeat MRI 4 weeks after the first time when CR or PR is identified. Any subject who did not meet CR or PR including those who did not have post-baseline radiological assessments was considered a nonresponder. (NCT02343406)
Timeframe: Every 8 weeks at each assessment of disease, up to 28 months

Interventionpercentage of participants (Number)
ABT-414/Temozolomide14.3
ABT-414_adult7.7
Control (Temozolomide/Lomustine)4.4

[back to top]

Adult Study: Progression-Free Survival (PFS)

Progression-free survival was assessed per response assessment in neuro-oncology criteria (RANO) criteria and assessed by an independent review committee and was defined as the length of time during and after the treatment of a disease, that the participant lived with the disease but did not get worse. (NCT02343406)
Timeframe: Measured every 8 weeks from date of randomization until the date of first objective progression or subject's death, whichever occurred first, up to 2 years

,,
Interventionmonths (Number)
25th quartile50th quartile75th quartile
ABT-414_adult1.51.93.5
ABT-414/Temozolomide1.82.74.9
Control (Temozolomide/Lomustine)1.61.94.2

[back to top]

Adult Study: Overall Survival in the Subgroup With Epidermal Growth Factor Receptor (EGFRvIII) Mutation

Overall Survival (OS) was defined as time from randomization to death due to any cause, regardless of whether the event occurred on or off study drug (depatuxizumab mafodotin/temozolomide/lomustine) for all randomized participants that had the Epidermal Growth Factor Receptor (EGFRvIII) mutation. (NCT02343406)
Timeframe: From the date of randomization up to the date of participant's death; participants who completed treatment were to be assessed every 12 weeks, up to 28 months

,,
Interventionmonths (Number)
25th quartile50th quartile75th quartile
ABT-414_adult5.08.413.9
ABT-414/Temozolomide6.39.414.4
Control (Temozolomide/Lomustine)4.77.512.4

[back to top]

Pediatric Study: Percentage of Participants With Adverse Events From the First Visit Until 49 Days After the Last Dose of Study Drug

The severity of each adverse event was rated according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE Version 4.0) (NCT02343406)
Timeframe: From participant's first visit until 49 days after the participant's last dose of study drug, up to 63 weeks

Interventionpercentage of participants (Number)
ABT-414_ Pediatric100

[back to top]

Adult Study: Overall Survival (OS)

Overall Survival (OS) was defined as time from randomization to death due to any cause, regardless of whether the event occurred on or off study drug (depatuxizumab mafodotin/temozolomide/lomustine). (NCT02343406)
Timeframe: From the date of randomization up to the date of participant's death; participants who completed treatment were to be assessed every 12 weeks, up to 28 months.

,,
Interventionmonths (Number)
25th quartile50th quartile75th quartile
ABT-414_adult4.67.915.5
ABT-414/Temozolomide5.79.616.9
Control (Temozolomide/Lomustine)4.98.212.6

[back to top]

Pediatric Study: Maximum Observed Serum Concentration (Cmax) of ABT-414

Cmax is the peak concentration that a drug achieves in a specified compartment after the drug has been administrated and before administration of a second dose. (NCT02343406)
Timeframe: Samples collected Cycle 1 Days 1, 2,3,5,8,15; Cycle 2 Day 1; Cycle 3 Day 1; Cycle 5 Day 1; Day 1 of every two cycles starting with Cycle 5; and 35 days after the last dose

Interventionµg/mL (Mean)
ABT-414_ Pediatric31.4

[back to top]

Pediatric Study: Maximum Observed Plasma Concentration (Cmax) of Cys-mcMMAF

Cmax is the peak concentration that a drug or drug metabolite achieves in a specified compartment after the drug has been administrated and before administration of a second dose. Cys-mcMMAF is a toxic metabolite of depatuxizumab mafodotin. (NCT02343406)
Timeframe: Samples collected Cycle 1 Days 1, 2, 3, 5, 8

Interventionng/mL (Mean)
ABT-414_ Pediatric0.272

[back to top]

Pediatric Study: Half-life (t1/2) Observed for Cys-mcMMAF

Half-life is the calculated time it takes for half of the drug or drug metabolite to leave the body. CysmcMMAF is a toxic metabolite of depatuxizumab mafodotin. (NCT02343406)
Timeframe: Samples collected Cycle 1 Days 1, 2, 3, 5, 8

Interventiondays (Mean)
ABT-414_ Pediatric11.2

[back to top]

Overall Survival (OS)

Overall survival was defined as the duration from the initiation of chemoradiation therapy to the date of death from any cause. Subjects who were alive or lost to follow-up at the time of the analysis were be censored at the last known date they were alive. (NCT02343549)
Timeframe: From date of treatment start to date of death, or censored as described above; assessed for approximately 5 years

Interventionmonths (Median)
Planned RT + TMZ + BEV + NovoTTF100A Device9.9

[back to top]

Progression Free Survival (PFS)

Progression-Free Survival (PFS) is defined as duration of time from initiation of chemoradiation therapy to first occurrence of either progressive disease (PD) or death. PD must be objectively determined as per RANO criteria (Updated Response Assessment Criteria for High Grade Gliomas, where PD is defined as a 25% increase in sum of products of diameters from nadir), where date of PD is date of assessment that identified PD. If subject died without documented PD, date of progression will be date of death. For surviving subjects who do not have documented PD, PFS will be censored at date of last radiologic assessment. For subjects who receive subsequent anti-cancer therapy prior to documented PD, PFS will be censored at the date of last radiologic assessment prior to the commencement of subsequent therapy. Subjects who have an initial PFS event immediately following 2 or more consecutive missed assessments will be censored at date of last assessment prior to missed assessments. (NCT02343549)
Timeframe: From date of treatment start to date of progression/death, or censored as described above; assessed for approximately 2 years.

Interventionmonths (Median)
Planned RT + TMZ + BEV + NovoTTF100A Device7.9

[back to top]

Duration of Response

Duration of response (DOR) was measured from the time RANO criteria are met for CR or PR (whichever is first recorded) until the first occurrence of either progressive disease or death. PD must be objectively determined as per RANO criteria (Updated Response Assessment Criteria for High Grade Gliomas, where PD is defined as a 25% increase in sum of products of diameters from nadir), where PD date is assessment date that identified PD. If subject died without PD, PD date was death date. For surviving subjects who didn't have documented PD, DOR was censored at last radiologic assessment. For subjects who receive subsequent anti-cancer therapy prior to documented PD, DOR was censored at last radiologic assessment prior to commencement of subsequent therapy. Subjects who had an initial PD event immediately following 2 or more consecutive missed assessments were censored at last assessment prior to missed assessments. DOR was only calculated for subjects with a best response of PR or CR. (NCT02343549)
Timeframe: From date of first response to date of progression/death, or censored as described above; assessed for approximately 2 years.

Interventionmonths (Median)
Planned RT + TMZ + BEV + NovoTTF100A Device8.1

[back to top]

Number of Participants With 12-Month Survival

Twelve-month survival was determined for each subject as a binary variable indicating whether or not the subject was alive at 12 months from initiation of chemoradiation. Determination of this endpoint occurs after the subject has at least 12 months of follow-up, unless they have died sooner. (NCT02343549)
Timeframe: Evaluated over 12 months

InterventionParticipants (Count of Participants)
Planned RT + TMZ + BEV + NovoTTF100A Device3

[back to top]

Duration of Disease Control

Duration of disease control (DoDC) was measured from the time of initiation of chemoradiation therapy until the first occurrence of either progressive disease or death. PD must be objectively determined as per RANO criteria (Updated Response Assessment Criteria for High Grade Gliomas, where PD is defined as a 25% increase in sum of products of diameters from nadir), where PD date is assessment date that identified PD. If subject died without PD, PD date was death date. For surviving subjects who didn't have documented PD, DoDC was censored at last radiologic assessment. For subjects who receive subsequent anti-cancer therapy prior to documented PD, DoDC was censored at last radiologic assessment prior to commencement of subsequent therapy. Subjects who had an initial PD event immediately following 2 or more consecutive missed assessments were censored at last assessment prior to missed assessments. DoDC was calculated only for subjects with a best overall response of SD or better. (NCT02343549)
Timeframe: From date of treatment start to date of progression/death, or censored as described above; assessed for approximately 2 years.

Interventionmonths (Median)
Planned RT + TMZ + BEV + NovoTTF100A Device7.9

[back to top]

Number of Participants With Disease Control

Disease control was determined for each subject indicating whether or not they achieved an overall response of stable disease (SD) or better by RANO criteria (where a CR is indicated by disappearance of all target and non target lesions, a PR is indicated by >= 50% decrease in sum of products of diameters of target lesions with baseline as reference, and SD is neither sufficient shrinkage to qualify for PR nor sufficient growth, >=25%, to indicate progression). (NCT02343549)
Timeframe: From enrollment to best response while on study treatment; subjects remained on treatment until disease progression or death or unacceptable toxicity (subjects were on treatment for an average of 7 months).

InterventionParticipants (Count of Participants)
Planned RT + TMZ + BEV + NovoTTF100A Device9

[back to top]

Number of Participants With Objective Response

Objective response was determined for each subject as a binary variable indicating whether or not the subject achieved a best overall response of complete response (CR) or partial response (PR) as determined by the Updated Response Assessment Criteria for High Grade Gliomas (RANO; where CR is indicated by disappearance of all target lesions and PR is indicated by >=50% decrease in the sum of products of diameters of target lesions with baseline as reference). Best responses of CR or PR must be confirmed by a subsequent radiologic assessment. (NCT02343549)
Timeframe: From enrollment to best response while on study treatment; subjects remained on treatment until disease progression or death or unacceptable toxicity (subjects were on treatment for an average of 7 months)

InterventionParticipants (Count of Participants)
Planned RT + TMZ + BEV + NovoTTF100A Device3

[back to top]

Median Overall Survival in CMV Negative Participants

Median overall survival will be estimated in the subset of participants that are CMV negative. Overall survival will be defined as the time in months between randomization and death, or last follow-up if alive. Kaplan-Meier methods will be used to estimate overall survival. (NCT02366728)
Timeframe: Up to 72 months (from the time of randomization of the first patient until approximately 31 months after randomization of the last patient)

Interventionmonths (Median)
Group I: Unpulsed DC Pre-conditioning13.4
Group II: Tetanus Pre-conditioning16.7

[back to top]

Median Overall Survival

Overall survival will be defined as the time in months between randomization and death, or last follow-up if alive. Kaplan-Meier methods will be used to estimate overall survival. (NCT02366728)
Timeframe: Up to 72 months (from the time of randomization of the first patient until approximately 31 months after randomization of the last patient)

Interventionmonths (Median)
Group I: Unpulsed DC Pre-conditioning16
Group II: Tetanus Pre-conditioning20
Group III: Basiliximab and Tetanus Pre-conditioning19

[back to top]

Percentage of 111In-labeled Dendritic Cells Migrating to the Inguinal Lymph Nodes

Within Groups I and II only, the percentage of 111In-labeled DCs migrating to the inguinal lymph nodes from the initial injection sites in the left and right groin at 48 hours post-vaccination #4 will be calculated. (NCT02366728)
Timeframe: For each patient, migration studies will occur after vaccination #4 which occurs approximately 7 months after study consent.

Interventionpercentage of cells (Median)
Group I: Unpulsed DC Pre-conditioning6.0
Group II: Tetanus Pre-conditioning9

[back to top]

Median Progression-free Survival in CMV Positive Participants

Median progression-free survival will be estimated in the subset of participants that are CMV positive. Progression-free survival will be defined as the time in months between randomization and disease progression or death. Participants alive without disease progression will be censored at the time of their last follow-up. Kaplan-Meier methods will be used to estimate progression-free survival. (NCT02366728)
Timeframe: Up to 72 months (from the time of randomization of the first patient until approximately 31 months after randomization of the last patient)

Interventionmonths (Median)
Group I: Unpulsed DC Pre-conditioning6.5
Group II: Tetanus Pre-conditioning6.8

[back to top]

Median Progression-free Survival in CMV Negative Participants

Median progression-free survival will be estimated in the subset of participants that are CMV negative. Progression-free survival will be defined as the time in months between randomization and disease progression or death. Participants alive without disease progression will be censored at the time of their last follow-up. Kaplan-Meier methods will be used to estimate progression-free survival. (NCT02366728)
Timeframe: Up to 72 months (from the time of randomization of the first patient until approximately 31 months after randomization of the last patient)

Interventionmonths (Median)
Group I: Unpulsed DC Pre-conditioning5.9
Group II: Tetanus Pre-conditioning5.8

[back to top]

Median Progression-free Survival

Progression-free survival will be defined as the time in months between randomization and disease progression or death. Participants alive without disease progression will be censored at the time of their last follow-up. Kaplan-Meier methods will be used to estimate progression-free survival. (NCT02366728)
Timeframe: Up to 72 months (from the time of randomization of the first patient until approximately 31 months after randomization of the last patient)

Interventionmonths (Median)
Group I: Unpulsed DC Pre-conditioning6.5
Group II: Tetanus Pre-conditioning6.7
Group III: Basiliximab and Tetanus Pre-conditioning7.1

[back to top]

Median Overall Survival in CMV Positive Participants

Median overall survival will be estimated in the subset of participants that are CMV positive. Overall survival will be defined as the time in months between randomization and death, or last follow-up if alive. Kaplan-Meier methods will be used to estimate overall survival. (NCT02366728)
Timeframe: Up to 72 months (from the time of randomization of the first patient until approximately 31 months after randomization of the last patient)

Interventionmonths (Median)
Group I: Unpulsed DC Pre-conditioning16.5
Group II: Tetanus Pre-conditioning23.8

[back to top]

Change in AUC0-18 of the Temozolomide Concentration (AUC-T) in Brain Interstitium Before and After Regadenoson Infusion

The AUC-T for each day will be estimated by the trapezoid rule, based on the number of time points available for the particular evaluable patient. The PK variables will be tabulated and descriptive statistics calculated pre and post Regadenoson. The difference of AUCs will be summarized by mean and standard deviation or median and range if there is large variation from patient to patient. Means and standard deviation or mean and range will be presented for Cmax and AUC(inf) for each group. Graphic method will be used to display the difference for individual patient and all five patients. (NCT02389738)
Timeframe: 18 hours post temozolomide administration

Interventionpercentage of AUC (Mean)
Temozolomide alone AUC brain/plasmaTemozolomide/Regadenoson AUC brain/plasma
BBB Disruption With Regadenoson19.118.0

[back to top]

Progression-free Survival at 6 Months

Will be analyzed using standard descriptive statistical methods. (NCT02395692)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Treatment (Methoxyamine, Temozolomide)2

[back to top]

Toxicity as Assessed by Number of Participants Who Experienced Adverse Events

Number of participants who experience adverse events graded 3 or higher as defined by National Cancer Institute CTCAE v4.0. (NCT02395692)
Timeframe: Up to 30 days following the last dose of study drug

InterventionParticipants (Count of Participants)
Treatment (Methoxyamine, Temozolomide)19

[back to top]

Progression-free Survival

Will be analyzed using standard descriptive statistical methods. 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 (NCT02395692)
Timeframe: Up to at least 2 years

Interventionmonths (Median)
Treatment (Methoxyamine, Temozolomide)2

[back to top]

Overall Survival

Will be analyzed using standard descriptive statistical methods. (NCT02395692)
Timeframe: Up to at least 2 years

Interventionmonths (Median)
Treatment (Methoxyamine, Temozolomide)11

[back to top]

Objective Response as Assessed by Response Assessment in Neuro-Oncology (RANO) Criteria (Arm 1 and Arm 2)

"To test the hypothesis that the combination treatment of temozolomide and methoxyamine will achieve 30% radiographic response rate (partial response + complete response) in patients with first recurrence of glioblastoma.~Per Response Assessment in Neuro-Oncology (RANO) Criteria: 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." (NCT02395692)
Timeframe: Up to at least 2 years

InterventionParticipants (Count of Participants)
Arm1 Methoxyamine&Temozolomide (Bevacizumab-naïve)0

[back to top]

Overall Survival

The median overall survival estimated using the Kaplan-Meier methods. Estimates of quantities such as median survival will be obtained. Corresponding confidence intervals will be computed. (NCT02455557)
Timeframe: Date of diagnosis until (1) date of death or (2) the last date patient known alive (if death is not observed), assessed up to 2 years

Interventionmonths (Median)
Treatment (SurVaxM, Temozolomide)25.8

[back to top]

Progression-free Survival (PFS)

The 6-month progression-free survival (PFS6) estimated using the Kaplan-Meier methods. PFS6 is defined as the percentage of patients without tumor progression or death from any cause 6 months after the date of diagnosis by biopsy. Corresponding confidence intervals will be computed. (NCT02455557)
Timeframe: Date of diagnosis to the date of first observed disease progression or death due to any cause, assessed at 6 months

Interventionpercentage of participants (Number)
Treatment (SurVaxM, Temozolomide)95

[back to top]

Progression Free Survival

Progression Free Survival (PFS) is defined as the time from randomization to the event of disease recurrence/progression defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) or death due to any cause. To determine the progression free survival of subjects dosed with Vigil immunotherapy in combination with irinotecan and temozolomide. (NCT02511132)
Timeframe: Estimated median 1.3 years

InterventionParticipants (Count of Participants)
Part 1: Vigil Alone5
Part 1: Gemicitabine and Docetaxel6
Part 2: Vigil in Combination With Temozolomide and Irinotecan9

[back to top]

Overall Survival

OS is defined as time from randomization to death or to the date of last follow-up. The date of last follow-up confirming survival will be used as the censoring date for subjects who are alive and/or do not have a known date of death. (NCT02511132)
Timeframe: Estimated median 2 years

InterventionParticipants (Count of Participants)
Part 1: Vigil Alone3
Part 1: Gemcitabine and Docetaxel6
Part 2: Vigil in Combination With Temozolomide and Irinotecan5

[back to top]

Number of Participants With Adverse Events Determined by Laboratory Assessments and Physical Examinations

"To determine safety profile of Vigil immunotherapy in combination with irinotecan and temozolomide with 30 days of last dose in patients with metastatic Ewing's sarcoma refractory or intolerant to at least 1 prior line of systemic chemotherapy.~• To determine safety profile of Vigil immunotherapy in combination with irinotecan and temozolimidetemozolomide in patients with metastatic Ewing's sarcoma refractory or intolerant to at least 1 prior line of systemic chemotherapy." (NCT02511132)
Timeframe: 30 days of last treatment dosing

InterventionParticipants (Count of Participants)
Part 1: Vigil Alone5
Part 1: Gemcitabine and Docetaxel6
Part 2: Vigil in Combination With Temozolomide and Irinotecan9

[back to top]

Deterioration Free Survival in M.D. Anderson Symptom Inventory Brain Tumor Module (MDASI-BT) Symptom Severity Score

The MDASI-BT assesses the severity of multiple brain tumor-related symptoms and the impact of these symptoms on daily functioning in the last 24 hours. It consists of 22 symptom items and 6 interference items, each rated from 0 to 10. MDASI-BT symptom severity score is defined as average over 13 core symptom items and 9 brain tumor symptom items, with a total score of 0 to 10, with higher score indicating worse symptoms/interference. Changes in symptom severity score were classified into 3 categories: improved (≤ -1), stable (> -1 and < 1), and deteriorated (≥ 1). Deterioration is defined as satisfying the deterioration criteria (i.e., increase in symptom severity score by ≥ 1 unit) without further improvement (i.e., failing to satisfy deterioration criteria) within 8 weeks or occurrence of death. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ11.0
Depatuxizumab Mafodotin, Radiation and TMZ6.1

[back to top]

Progression-Free Survival (PFS)

PFS will be defined as the number of days from the date of randomization to the date of earliest disease progression based on Response Assessment in Neuro-Oncology (RANO) criteria (see Wen et al. J Clin Oncol. 2010 Apr 10;28(11):1963-72) or to the date of death, if disease progression does not occur. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ6.3
Depatuxizumab Mafodotin, Radiation and TMZ8.0

[back to top]

PFS for EGFRvIII-Mutated Tumor Subgroup

PFS will be defined as the number of days from the date of randomization to the date of earliest disease progression based on Response Assessment in Neuro-Oncology (RANO) criteria or to the date of death, if disease progression does not occur. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ5.9
Depatuxizumab Mafodotin, Radiation and TMZ8.3

[back to top]

Overall Survival (OS)

Time to OS is defined as the number of days from the date of randomization to the date of death due to any cause. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ18.7
Depatuxizumab Mafodotin, Radiation and TMZ18.9

[back to top]

OS for the O6-methylguaninemethlytransferese (MGMT) Unmethylated Group

"Time to OS is defined as the number of days from the date of randomization to the date of death due to any cause.~Unmethylated MGMT promoter is associated with a worse prognosis in GBM" (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ16.2
Depatuxizumab Mafodotin, Radiation and TMZ16.1

[back to top]

OS for the MGMT Methylated Group

Time to OS is defined as the number of days from the date of randomization to the date of death due to any cause. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZNA
Depatuxizumab Mafodotin, Radiation and TMZ25.4

[back to top]

OS for the Epidermal Growth Factor Receptor (EGFR)vIII-Mutated Tumor Subgroup

Time to OS is defined as the number of days from the date of randomization to the date of death due to any cause. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ18.2
Depatuxizumab Mafodotin, Radiation and TMZ19.8

[back to top]

Deterioration Free Survival in Neurocognitive Functioning on the Hopkins Verbal Learning Test Revised (HVLT-R) Total Recall Score

The HVLT-R consists of 3 parts. Free call has a range of 0 to 36, delayed recall has a range from 0 to 12, and delayed recognition has a range of -12 to 12. Higher scores indicating better function in all 3 parts. When scoring the HVLT-R, the 3 learning trials are combined to calculate a total recall score (range -12 to 60). Deterioration is defined as satisfying the deterioration criteria (i.e., decrease in HVLT-R total recall score by 5 units) without further improvement within 8 weeks or occurrence of death. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ13.2
Depatuxizumab Mafodotin, Radiation and TMZ10.7

[back to top]

Deterioration Free Survival in MDASI-BT Symptom Interference Score

The MDASI-BT assesses the severity of multiple brain tumor-related symptoms and the impact of these symptoms on daily functioning in the last 24 hours. It consists of 22 symptom items and 6 interference items, each rated from 0 to 10. MDASI-BT symptom interference score is defined as an average of 6 interference items, with a total score of 0 to 10, where higher scores indicate worse interference. Changes in symptom interference score were classified into 3 categories: improved (≤ -1), stable (> -1 and < 1), and deteriorated (≥ 1). Deterioration is defined as satisfying the deterioration criteria (i.e., increase in symptom interference score by ≥ 1 unit) without further improvement (i.e., failing to satisfy deterioration criteria) within 8 weeks or occurrence of death. (NCT02573324)
Timeframe: Overall median duration of follow-up was 15.5 months (range: 0.1, 35.6).

Interventionmonths (Median)
Placebo, Radiation and TMZ9.7
Depatuxizumab Mafodotin, Radiation and TMZ6.1

[back to top]

Kaplan-Meier Plot of Overall Survival (OS) - Extended Collection

OS was defined as the time between the date of randomization and the date of death due to any cause. A participant who has not died was censored at the last known alive date. 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 March 4, 2022). (NCT02617589)
Timeframe: From randomization to the date of death due to any cause (up to approximately 6 years)

InterventionMonths (Median)
Nivolumab + Radiation Therapy13.34
Temozolomide + Radiation Therapy14.92

[back to top]

Kaplan-Meier Plot of Progression Free Survival

PFS was defined as the time from randomization to the date of the first documented tumor progression or death due to any cause. Participants who did not have disease progression or who did not die were censored at the date of last tumor assessment. Participants who did not have any on study tumor assessment and did not have tumor progression or die were censored at the randomization date. Participants who started any subsequent anti-cancer therapy without a prior reported progression were censored at the last tumor assessment prior to initiation of the subsequent anti-cancer therapy. Participants who had surgical resection post start of study treatment were censored at the last tumor assessment date prior to initiation of surgical resection. PFS was determined by investigator reported response based on the Radiologic Assessment in Neuro-Oncology criteria. (NCT02617589)
Timeframe: From randomization to the date of the first documented tumor progression or death due to any cause (up to approximately 6 years)

InterventionMonths (Median)
Nivolumab + Radiation Therapy6.01
Temozolomide + Radiation Therapy6.21

[back to top]

Overall Survival Rate at 24 Months

The overall survival (OS) rate of (nivolumab + radiation therapy) and (temozolomide + radiation therapy) estimated as Kaplan-Meier probability of survival at 24 months. OS was defined as the time between the date of randomization and the date of death due to any cause. A participant who has not died was censored at the last known alive date. (NCT02617589)
Timeframe: At 24 Months

InterventionPercentage of participants (Number)
Nivolumab + Radiation Therapy10.6
Temozolomide + Radiation Therapy21.2

[back to top]

Overall Survival (OS)

OS is defined as the time between the date of randomization and the date of death due to any cause. A participant who has not died will be censored at the last known alive date. (NCT02617589)
Timeframe: up to 3 years

InterventionMonths (Median)
Nivolumab + Radiation Therapy13.40
Temozolomide + Radiation Therapy14.88

[back to top]

Progression Free Survival (PFS) (Newly Diagnosed Glioblastoma Cohort)- Phase II

Progression free survival is defined as time in weeks from start of study treatment to first documentation of objective tumor progression or up to death due to any cause, whichever occurs first. (NCT02661282)
Timeframe: At 6 months

Interventionweeks (Median)
Phase II: Newly Diagnosed Dose Level 1 x 10^819

[back to top]

Progression Free Survival (PFS) (Recurrent Glioblastoma Cohort) at 6 Months- Phase II

Progression-free survival (PFS) is defined as the time from study enrollment until the time of first disease progression, relapse, or death due to disease. Patients who are alive without progression or relapse will be censored at the time of last contact. The point estimate of 6-month progression-free survival (PFS6) will be analyzed. Kaplan-Meier curves will be generated and median survival time will be derived. (NCT02661282)
Timeframe: 6 months

Interventionmonths (Number)
Phase II: Recurrent Glioblastoma Dose Level 1 x 10^82.5

[back to top]

Time to Progression (Recurrent Glioblastoma Cohort)- Phase II

The length of time from the date of diagnosis or the start of treatment for a disease until the disease starts to get worse or spread to other parts of the body. (NCT02661282)
Timeframe: Baseline to disease progression, assessed up to 4 years

Interventionmonths (Number)
Phase II: Recurrent Glioblastoma Dose Level 1 x 10^82.5

[back to top]

Overall Survival (OS) (Newly Diagnosed Glioblastoma Cohort)- Phase II

Overall Survival is defined as the time from definitive histological diagnosis until the time of death. (NCT02661282)
Timeframe: Time from definitive histological diagnosis until death

Interventionweeks (Median)
Phase II: Newly Diagnosed Dose Level 1 x 10^824

[back to top]

Maximum Tolerated Dose (MTD) (Recurrent Glioblastoma Participant Cohort)- Phase I

The number of participants who were treated at the respective dose level without DLT (NCT02661282)
Timeframe: Up to 42 days

Interventionparticipants (Number)
Phase I: Recurrent Glioblastoma Dose Level 5 x 10^63
Phase I: Recurrent Glioblastoma Dose Level 1 x 10^73
Phase I: Recurrent Glioblastoma Dose Level 5 x 10^73
Phase I: Recurrent Glioblastoma (MTD) Dose Level 1 x 10^87

[back to top]

Median Duration of Response (Newly Diagnosed Glioblastoma Cohort)- Phase II

Cox proportional hazard regression will be employed for multivariate analysis. (NCT02661282)
Timeframe: Baseline to response, assessed up to 4 years

Interventionmonths (Median)
Phase II: Newly Diagnosed Dose Level 1 x 10^85.3

[back to top]

Number of Participants With Immunological Effects in Tumor Tissue (Recurrent Glioblastoma Cohort)- Phase II

Descriptive statistics will be used to summarize immunological effect. To evaluate the tumor-mediated immune suppression at the effector location, the markers (interferon, interleukin-2, and tumor necrosis factor alpha, perforin, granzyme B) will be measured for immune responses in the tumor microenvironment rather than in the peripheral blood. (NCT02661282)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Phase II: Recurrent Glioblastoma Dose Level 1 x 10^81

[back to top]

Overall Objective Response Rate (ORR) (Newly Diagnosed Glioblastoma Cohort)- Phase II

The number of participants with stable disease according to Response Evaluation Criteria in Solid Tumors (RECIST). (NCT02661282)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Phase II: Newly Diagnosed Dose Level 1 x 10^83

[back to top]

Progression-free Survival (PFS) Determined by BICR

The time from randomization to the date of the first documented tumor progression or death by any cause. PFS will be determined by a Blinded Independent Central Review (BICR) assessed based on Radiologic Assessment in Neuro-Oncology (RANO) criteria. Specifically, RANO response criteria indicates that within the first 12 weeks of completion of radiotherapy, progression can only be assessed if the majority of the new enhancement is outside of the radiation field or if there is pathologic confirmation of progressive disease. (NCT02667587)
Timeframe: From randomization to the date of the first documented tumor progression or death by any cause. (up to approximately 4.5 years)

InterventionMonths (Median)
Radiotherapy, Temozolomide Plus Nivolumab10.64
Radiotherapy, Temozolomide Plus Placebo10.32

[back to top]

Progression Free Survival (PFS) Based on Investigator Assessment

The time from randomization to the date of the first documented tumor progression or death by any cause. PFS will be determined by investigator assessment based Radiologic Assessment in Neuro-Oncology (RANO) criteria. Specifically, RANO response criteria indicates that within the first 12 weeks of completion of radiotherapy, progression can only be assessed if the majority of the new enhancement is outside of the radiation field or if there is pathologic confirmation of progressive disease. (NCT02667587)
Timeframe: From randomization to the date of the first documented tumor progression or death by any cause. (up to approximately 4.5 years)

InterventionMonths (Median)
Radiotherapy, Temozolomide Plus Nivolumab14.09
Radiotherapy, Temozolomide Plus Placebo15.18

[back to top]

Overall Survival (OS) Rates at 12 Months

Overall Survival (OS) rate is defined as the percentage of participants surviving at 12 months (NCT02667587)
Timeframe: From randomization to 12 months after first dose

Interventionpercentage of participants (Number)
Radiotherapy, Temozolomide Plus Nivolumab82.7
Radiotherapy, Temozolomide Plus Placebo87.7

[back to top]

Overall Survival (OS)

The time from the date of randomization to the date of death. who have not died by the end of the study will be censored to last known date alive. OS is assessed in the randomized population with no corticosteroids at baseline population and in the overall randomized population. (NCT02667587)
Timeframe: From randomization to date of death (up to approximately 4.5 years)

,
InterventionMonths (Median)
All randomized participantsAll randomized participants without baseline corticosteroids
Radiotherapy, Temozolomide Plus Nivolumab28.9131.34
Radiotherapy, Temozolomide Plus Placebo32.0732.99

[back to top]

Overall Survival (OS) Rates at 24 Months

Overall Survival (OS) rate is defined as the percentage of participants surviving at 24 months (NCT02667587)
Timeframe: From randomization to 24 months after first dose

Interventionpercentage of participants (Number)
Radiotherapy, Temozolomide Plus Nivolumab55.9
Radiotherapy, Temozolomide Plus Placebo63.3

[back to top]

Median Time to Progression (TTP) Assessed Locally and Centrally

The TTP was defined as the time from first treatment administration to the first objective tumor progression (PD) according to RECIST criteria v 1.1. The TTP was assessed locally by the investigator and centrally by an independent radiologist. The distribution of TTP times was estimated using the Kaplan-Meier method. The TTP of participants who were lost to follow-up, and those who had not progressed at end of study were censored at the date of the last disease assessment. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus Temozolomide37.137.1

[back to top]

Median Progression Free Survival (PFS) Assessed Locally and Centrally

The PFS was defined as the time from the first treatment administration to disease progression according to RECIST criteria v 1.1 or death from any cause. The PFS was assessed locally by the investigator and centrally by an independent radiologist. The distribution of PFS times was estimated using the Kaplan-Meier method. The PFS of participants who were lost to follow-up and those who had not progressed at end of study were censored at the date of the last disease assessment. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus Temozolomide37.137.1

[back to top]

DCR Assessed Centrally at Month 9

The DCR was defined as SD, PR or CR according to RECIST criteria v1.1. A second set of the original computed tomography (CT) scan images were used for a centralized RECIST v1.1 assessment by an independent radiologist. (NCT02698410)
Timeframe: Up to Month 9

Interventionpercentage of participants (Number)
Lanreotide ATG Plus Temozolomide28.2

[back to top]

Median Time to Response (TTR) Assessed Locally and Centrally

The TTR was defined as the time from first treatment administration to the first objective tumor response (PR or CR according to RECIST criteria v 1.1). The TTR was assessed locally by the investigator and centrally by an independent radiologist. The distribution of TTR was estimated using the Kaplan-Meier method. The TTR of participants who were lost to follow-up or died prior to any objective tumor response were censored at the date of the last disease assessment. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus TemozolomideNANA

[back to top]

Neuron-Specific Enolase (NSE) and CgA Biomarker Levels

The NSE and CgA levels were classified according to ULN as follows: < 1 ULN, 1-2 ULN and > 2 ULN. Baseline was defined as value at Day 1. Percentages are based on the number of participants in the ITT/Safety population who attended the visit with non-missing observations. (NCT02698410)
Timeframe: Baseline and Weeks 4, 12, 24, 36 and 52

Interventionpercentage of participants (Number)
CgA: Baseline: <1 ULNCgA: Baseline: 1-2 ULNCgA: Baseline: >2 ULNCgA: Week 4: <1 ULNCgA: Week 4: 1-2 ULNCgA: Week 4: >2 ULNCgA: Week 12: <1 ULNCgA: Week 12: 1-2 ULNCgA: Week 12: >2 ULNCgA: Week 24: <1 ULNCgA: Week 24: 1-2 ULNCgA: Week 24: >2 ULNCgA: Week 36: <1 ULNCgA: Week 36: 1-2 ULNCgA: Week 36: >2 ULNCgA: Week 52: <1 ULNCgA: Week 52: 1-2 ULNCgA: Week 52: >2 ULNNSE: Baseline: <1 ULNNSE: Baseline: 1-2 ULNNSE: Baseline: >2 ULNNSE: Week 4: <1 ULNNSE: Week 4: 1-2 ULNNSE: Week 4: >2 ULNNSE: Week 12: <1 ULNNSE: Week 12: 1-2 ULNNSE: Week 12: >2 ULNNSE: Week 24: <1 ULNNSE: Week 24: 1-2 ULNNSE: Week 24: >2 ULNNSE: Week 36: <1 ULNNSE: Week 36: 1-2 ULNNSE: Week 36: >2 ULNNSE: Week 52: <1 ULNNSE: Week 52: 1-2 ULNNSE: Week 52: >2 ULN
Lanreotide ATG Plus Temozolomide40.015.045.034.322.942.946.417.935.730.025.045.031.312.556.327.39.163.665.025.010.062.925.711.478.617.93.680.010.010.087.56.36.363.618.218.2

[back to top]

Percentage of Biochemical Responders According to Chromogranin A (CgA) Plasma Levels

Participants with baseline CgA plasma levels greater than the upper limit of normal (ULN) were assessed for a biochemical response. A biochemical responder was defined as a participant who had a decrease of CgA >= 50% compared to baseline, while biochemical SD was defined as a decrease < 50% or an increase <= 25% compared to baseline. Biochemical non-responders had an increase >25% compared to baseline. Baseline was defined as value at Day 1. (NCT02698410)
Timeframe: Baseline (Day 1) and Week 4, 12, 24, 36 and 52

Interventionpercentage of participants (Number)
Week 4: RespondersWeek 4: SDWeek 4: Non-respondersWeek 12: RespondersWeek 12: SDWeek 12: Non-respondersWeek 24: RespondersWeek 24: SDWeek 24: Non-respondersWeek 36: RespondersWeek 36: SDWeek 36: Non-respondersWeek 52: RespondersWeek 52: SDWeek 52: Non-responders
Lanreotide ATG Plus Temozolomide27.359.113.637.537.525.023.130.846.236.427.336.412.550.037.5

[back to top]

Best Overall Response (BOR) Assessed Locally and Centrally

The BOR was defined as the highest OR achieved by the participant from the time of first treatment until disease progression/recurrence or the end of study according to RECIST criteria v1.1 and was classified as: CR > PR > Non-CR/Non-progressive disease (NCR/NPD) > SD > PD > ND > not evaluable (NE). The BOR was assessed locally by the investigator and centrally by an independent radiologist. Percentages are based on the number of participants in the ITT/Safety population with non-missing observations. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionpercentage of participants (Number)
Local assessment: CRLocal assessment: PRLocal assessment: SDLocal assessment: NCR/NPDLocal assessment: PDLocal assessment: NELocal assessment: Not applicableCentral assessment: CRCentral assessment: PRCentral assessment: SDCentral assessment: NCR/NPDCentral assessment: PDCentral assessment: NECentral assessment: Not applicable
Lanreotide ATG Plus Temozolomide07.771.8020.500013.265.82.615.802.6

[back to top]

DCR Assessed Locally and Centrally at Month 12

The DCR was defined as SD, PR or CR according to RECIST criteria v1.1. The DCR was assessed locally by the investigator and centrally by an independent radiologist. (NCT02698410)
Timeframe: Month 12

Interventionpercentage of participants (Number)
Local assessmentCentral assessment
Lanreotide ATG Plus Temozolomide17.515.4

[back to top]

Disease Control Rate (DCR) Assessed Locally at Month 9

Responders were participants who showed disease control according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria v 1.1 assessed locally by the investigator. The DCR was defined as complete response (CR), partial response (PR) or stable disease (SD) according to RECIST criteria v1.1. A sensitivity analysis-1 of local DCR was performed excluding participants withdrawn before 9 months with reason other than progressive disease (PD) or missing assessment and considering participants with PD prior or at 9 months as failures. In addition, a sensitivity analysis-2 was performed in order to consider assessments done between 7.5 and 10.5 months as 9 months assessments when 9-month assessment was missing using same methodology, i.e. considering PD prior or at 9 months and participants withdrawn with other or missing reasons as failures. (NCT02698410)
Timeframe: Up to Month 9; for sensitivity analysis-2, up to 10.5 months

Interventionpercentage of participants (Number)
DCR at Month 9Sensitivity analysis-1Sensitivity analysis-2
Lanreotide ATG Plus Temozolomide35.045.245.0

[back to top]

DCR Assessed Locally and Centrally at Month 9 by Carcinoid Type

The influence of type of carcinoid [typical, atypical and undetermined carcinoid neuroendocrine tumors (NET)] on the local and central DCR at 9 months was analysed. Typical carcinoids were defined with absent foci of necrosis and mitotic count < 2 mitoses/2 millimeters^2 (mm^2); atypical carcinoids were defined with presence of foci of necrosis and/or 2 mitoses/2 mm^2 <= mitotic count <= 10 mitoses/2 mm^2; and carcinoid NET were defined as confirmed carcinoid without foci of necrosis and/or mitotic count reported. The DCR was assessed locally by the investigator and centrally by an independent radiologist. (NCT02698410)
Timeframe: Up to Month 9

Interventionpercentage of participants (Number)
Local assessment: Typical carcinoidLocal assessment: Atypical carcinoidLocal assessment: Carcinoid NETCentral assessment: Typical carcinoidCentral assessment: Atypical carcinoidCentral assessment: Carcinoid NET
Lanreotide ATG Plus Temozolomide12.547.627.3035.036.4

[back to top]

Coefficient of Agreement Between Central and Local Assessment of Tumor Radiological Response at Month 9

Differences between central radiology review and local investigator review were assessed according to the DCR status and the number of agreements and disagreements between the evaluators (central versus local) along with the p-values from the kappa test. A kappa statistic was used to evaluate the concordance between the central and the local review. (NCT02698410)
Timeframe: Month 9

Interventionkappa coefficient (Number)
Lanreotide ATG Plus Temozolomide0.71

[back to top]

Objective Response Rate (ORR) Assessed Locally and Centrally at Months 9 and 12

The ORR was defined as the percentage of participants with CR or PR according to RECIST criteria v1.1. The ORR was assessed locally by the investigator and centrally by an independent radiologist. The ORR was based on the participants with PD prior to 9 and 12 months with respectively PD at 9 and 12 months, and participants withdrawn before the assessment for reason other than PD or missing as failures. (NCT02698410)
Timeframe: Months 9 and 12

Interventionpercentage of participants (Number)
Local assessment: Month 9Local assessment: Month 12Central assessment: Month 9Central assessment: Month 12
Lanreotide ATG Plus Temozolomide2.52.55.12.6

[back to top]

Influence of Biomarkers Expression on Locally and Centrally Assessed DCR at Months 9 and 12

The following biomarkers were investigated: Immunohistochemistry assay SSTR2 including HER-2 score (0, 1+, 2+, 3+ and +ve versus -ve); H-score (from 0 to 300 as quantitative variable and +ve versus -ve); IRS score (from 0 to 12 and reference for odds ratio was 0-1). Ki67 index (from 0% to 100% and reference for odds ratio was 4%-25%). MGMT expression including percentage of +ve nuclei stained and methylated sites and H-score (from 0 to 300 as quantitative variable). For all these categories, higher score indicates a higher expression of biomarkers. Carcinoid type (typical or NET) was also investigated. Prognostic value of biomarkers expression at screening on DCR were assessed locally and centrally using univariate logistic regression models. (NCT02698410)
Timeframe: Screening period, Months 9 and 12

Interventionodds ratio (Number)
Month 9: Local: SSTR2: HER-2 score; +ve (2+, 3+)Month 9: Local: SSTR2: H-score; +ve (>= 50)Month 9: Local: SSTR2: IRS score; 2-3Month 9: Local: SSTR2: IRS score; 4-8Month 9: Local: SSTR2: IRS score; 9-12Month 9: Local: Ki67: <4Month 9: Local: Ki67: >=25Month 9:Local: MGMT: +ve nuclei stained; +ve(>=5%)Month 9: Local: MGMT: Methylated sites; +ve (>10%)Month 9: Local: MGMT: H-scoreMonth 9: Local: Carcinoid type: TypicalMonth 9: Local: Carcinoid type: Carcinoid NETMonth 9: Central: SSTR2: HER-2 score; +ve (2+, 3+)Month 9: Central: SSTR2: H-score; +ve (>= 50)Month 9: Central: SSTR2: IRS score; 2-3Month 9: Central: SSTR2: IRS score; 4-8Month 9: Central: SSTR2: IRS score; 9-12Month 9: Central: Ki67: <4Month 9: Central: Ki67: >=25Month 9:Central:MGMT:+ve nuclei stained; +ve(>=5%)Month 9:Central: MGMT: Methylated sites; +ve(>10%)Month 9: Central: MGMT: H-scoreMonth 9: Central: Carcinoid type: TypicalMonth 9: Central: Carcinoid type: Carcinoid NETMonth 12: Local: SSTR2: HER-2 score; +ve (2+, 3+)Month 12: Local: SSTR2: H-score; +ve (>= 50)Month 12: Local: SSTR2: IRS score; 2-3Month 12: Local: SSTR2: IRS score; 4-8Month 12: Local: SSTR2: IRS score; 9-12Month 12: Local: Ki67: <4Month 12: Local: Ki67: >=25Month 12:Local:MGMT: +ve nuclei stained; +ve(>=5%)Month 12: Local: MGMT: Methylated sites; +ve(>10%)Month 12: Local: MGMT: H-scoreMonth 12: Local: Carcinoid type: TypicalMonth 12: Local: Carcinoid type: Carcinoid NETMonth 12: Central: SSTR2: HER-2 score; +ve(2+, 3+)Month 12: Central: SSTR2: H-score; +ve (>= 50)Month 12: Central: SSTR2: IRS score; 2-3Month 12: Central: SSTR2: IRS score; 4-8Month 12: Central: SSTR2: IRS score; 9-12Month 12: Central: Ki67: <4Month 12: Central: Ki67: >=25Month 12:Central:MGMT:+ve nuclei stained;+ve(>=5%)Month 12:Central: MGMT: Methylated sites;+ve(>10%)Month 12: Central: MGMT: H-scoreMonth 12: Central: Carcinoid type: TypicalMonth 12: Central: Carcinoid type: Carcinoid NET
Lanreotide ATG Plus Temozolomide0.900.78NA0.6712.00NA1.67NA3.250.990.160.411.402.253.000.6712.00NANA0.505.001.00NA1.063.004.40NA0.7010.50NANA0.202.130.990.460.322.083.00NANA10.50NANA0.143.000.990.570.40

[back to top]

Influence of Biomarkers Expression on Locally and Centrally Assessed PFS

The following biomarkers were investigated: Immunohistochemistry assay human somatostatin receptors 2 (SSTR2) including human epidermal growth factor receptor 2 (HER-2) score [0, 1+, 2+, 3+ and positive (+ve) versus negative (-ve)]; hormone receptor score (H-score) (from 0 to 300 as quantitative variable and +ve versus -ve); immunoreactive score (IRS) score (from 0 to 12 and reference for hazard ratio was 0-1). Ki67 index (from 0% to 100% and reference for hazard ratio was 4%-25%). O^6-methylguanine-DNA methyltransferase (MGMT) expression including percentage of +ve nuclei stained and methylated sites and H-score (from 0 to 300 as quantitative variable). For all these categories, higher score indicates a higher expression of biomarkers. Carcinoid type (typical or NET) was also investigated. Prognostic value of biomarkers expression at screening on PFS were assessed locally and centrally using univariate cox proportional hazard models. (NCT02698410)
Timeframe: From Screening period (-4 weeks) up to Week 52

Interventionhazard ratio (Number)
Local: SSTR2: HER-2 score; +ve (2+, 3+)Local: SSTR2: H-score; +ve (>= 50)Local: SSTR2: IRS score; 2-3Local: SSTR2: IRS score; 4-8Local: SSTR2: IRS score; 9-12Local: Ki67: <4Local: Ki67: >=25Local: MGMT: +ve nuclei stained; +ve (>=5%)Local: MGMT: Methylated sites; +ve (>10%)Local: MGMT: H-scoreLocal: Carcinoid type: TypicalLocal: Carcinoid type: Carcinoid NETCentral: SSTR2: HER-2 score; +ve (2+, 3+)Central: SSTR2: H-score; +ve (>= 50)Central: SSTR2: IRS score; 2-3Central: SSTR2: IRS score; 4-8Central: SSTR2: IRS score; 9-12Central: Ki67: <4Central: Ki67: >=25Central: MGMT: +ve nuclei stained; +ve (>=5%)Central: MGMT: Methylated sites; +ve (>10%)Central: MGMT: H-scoreCentral: Carcinoid type: TypicalCentral: Carcinoid type: Carcinoid NET
Lanreotide ATG Plus Temozolomide0.710.660.310.900.121.081.682.060.411.001.051.590.500.360.780.880.100.002.752.110.731.000.990.61

[back to top]

Median Duration of Response (DOR) Assessed Locally and Centrally

The DOR was defined as the time from onset of the first objective tumor response (PR or CR) to objective tumor progression (PD) according to RECIST criteria v 1.1. The DOR was assessed locally by the investigator and centrally by an independent radiologist. (NCT02698410)
Timeframe: From Day 1 up to end of study, 52 weeks

Interventionweeks (Median)
Local assessmentCentral assessment
Lanreotide ATG Plus TemozolomideNANA

[back to top]

Median Change in Tumor Volume From Baseline to Mid-radiation Treatment (Week 4)

Tumor volume will be measured by diffusion MRI and perfusion MRI before treatment start and at mid-treatment. (NCT02805179)
Timeframe: Baseline to Week 4

Interventioncubic centimeters (Median)
High Dose Chemoradiation-2.9

[back to top]

Median Progression-free Survival

From start of RT until disease progression or death, or until date of last imaging follow-up, estimated using Kaplan-Meier. Progression is defined by any of the following: >= 25% increase in sum of the products of perpendicular diameters of enhancing lesions (compared with baseline if no decrease) on stable or increasing doses of corticosteroids; a significant increase in T2/FLAIR non-enhancing lesions on stable or increasing doses of corticosteroids compared with baseline scan or best response after initiation of therapy, not due to comorbid events; appearance of any new lesions; clear progression of non-measurable lesions; or definite clinical deterioration not attributable to causes other than tumor, or to decrease in corticosteroid dose. When pathologic confirmation was unavailable, progression was defined as worsening enhancement based on imaging with or without adjunctive advanced imaging including perfusion MRI or magnetic resonance spectroscopy, when clinically indicated. (NCT02805179)
Timeframe: Median follow-up time was 26 months

Interventionmonths (Median)
all eligible patients who completed dose-intensified chemo-radiationonly patients who were boosted to both diffusion and perfusion
High Dose Chemoradiation1012

[back to top]

Percentage of Patients With Failure; Central or In-field vs. Marginal or Distant

"Failures will be classified as central or in-field, marginal or distant based on previously published criteria. 1) central, in which 95% or more of the recurrent tumor volume (Vrecur) was within D95, the region treated to high dose (95% of the prescription dose); 2) in-field, in which 80% or more of Vrecur was within the D95 isodose surface; 3) marginal, when between 20 and 80% of Vrecur was inside the D95 surface; 4) outside, in which less than 20% of Vrecur was inside the D95 surface." (NCT02805179)
Timeframe: Median 26 months

Interventionpercentage of participants (Number)
central or in-fieldnon-central/non-in-field (marginal or distant)
High Dose Chemoradiation3169

[back to top]

Percentage of Patients That Experienced Deterioration in Quality of Life (QOL)

Percentage of patients that experienced deterioration in QOL per the European Organization for Research and Treatment of Cancer, Quality of Life Questionnaire (EORTC QLQ-C30). 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 (dyspnea, 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. (NCT02805179)
Timeframe: Baseline to 1 and 7 months

Interventionpercentage of participants (Number)
at 1 monthat 7 months
High Dose Chemoradiation2633

[back to top]

Overall Survival at 12 Months

Percentage of patients alive at 12 months after completion of chemoradiation (NCT02805179)
Timeframe: 12 months after completion of chemoradiation

Interventionpercentage of participants (Number)
all eligible patients who completed dose-intensified chemo-radiationonly patients who were boosted to both diffusion and perfusion
High Dose Chemoradiation7492

[back to top]

Median Overall Survival

Median overall survival in months (NCT02805179)
Timeframe: Median follow-up time was 26 months

Interventionmonths (Median)
all eligible patients who completed dose-intensified chemo-radiationonly patients who were boosted to both diffusion and perfusion
High Dose Chemoradiation2020

[back to top]

Immunologic Response as Measured by Peak Number of T Cells That Secrete IFNγ by ELISPOT in Response to Component A of PEP-CMV

"The primary analysis will focus on patients who have follow-up immunologic monitoring after the 3rd vaccination with component A alone and before initiation of the second TMZ/vaccine cycle. Such a patient is considered evaluable for the immunologic response primary analyses.~The Wilcoxon rank sum test will compare treatment groups with regard to the median peak number of T cells that secrete IFNγ by ELISPOT in response to component A of PEP-CMV. Analyses will include only those patients who have an assessment of immune response after receiving 3 vaccinations." (NCT02864368)
Timeframe: Through study completion, an average of 1.5 years

InterventionT cells (Median)
5-day TMZ: Component A Alone57.5
21-day TMZ: Component A Alone71.25

[back to top] [back to top]

% of Participants Free of Disease Progression at 4mos Treated w/Zotiraciclib at the Maximum Tolerated Dose in Combination Temozolomide w/Dose Dense Temozolomide Schedules in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma

We first determined the maximum tolerated dose (MTDs) in each ARM and we then performed the cohort expansion at the MTD in both ARMs separately, until we treated a total of 18 participants at this dose in each ARM. PFS is defined as the duration of time from start of registration to time of progression or death, whichever comes first. Progression was assessed by the Response Assessment in Neuro-Oncology Criteria (RANO). Progression is ≥25% increase in tumor volume compared to baseline in the sum of the products of perpendicular diameters of enhancing lesions compared with the smallest measurement obtained either at baseline or best response with the participant on stable or increasing doses of steroids. Significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesions with the participant on stable or increasing doses of steroids (not caused by comorbid events). Any new lesions. (NCT02942264)
Timeframe: 4 months

Interventionpercentage of participants (Number)
All Participants40

[back to top]

Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0)

Here is the number of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0). A non-serious adverse event is any untoward medical occurrence. A serious adverse event is an adverse event or suspected adverse reaction that results in death, a life-threatening adverse drug experience, hospitalization, disruption of the ability to conduct normal life functions, congenital anomaly/birth defect or important medical events that jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the previous outcomes mentioned. (NCT02942264)
Timeframe: Date treatment consent signed to date off study, approximately 15mo(m)/8days(d), 26m, 7m/26d, 13m/17d, 11m/30d, 12m/6d, 19m/11d, 9m/28d and 18m/21d for Group 1-9 respectively.

InterventionParticipants (Count of Participants)
ARM 1 Dose Level 0 - (Starting Dose)6
ARM 1 Dose Level 1 - (Dose Escalation)13
ARM 2 Dose Level 0 (Starting Dose)3
ARM 2 Dose 1 - (Dose Escalation)6
ARM 2 Dose 0 - (Dose De-escalation)3
ARM 2 Dose Level II - (Dose Escalation)2
ARM 2 Dose Level I - (Dose De-escalation)7
ARM 1 Dose Level 1 (MTD Level in ARM1)6
ARM 2 Dose Level 1 (MTD Level in ARM2)7

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Zotiraciclib (TG02) in Combination of Metronomic (mn) Temozolomide (TMZ) in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma

Maximum tolerated dose of metronomic (mn) Zotiraciclib (TG02) was assessed using the Bayesian Optimal Interval (BOIN) design. The MTD is defined as the dose for which the isotonic estimate of the toxicity rate is closest to the target toxicity rate of 0.35. If there are ties, we select the higher dose level when the isotonic estimate is lower than the target toxicity rate; and we select the lower dose level when the isotonic estimate is greater than the target toxicity rate of 0.35. (NCT02942264)
Timeframe: 4 weeks after initiation of treatment

Interventionmg/day (Number)
All Participants250

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Zotiraciclib in Combination With Dose Dense (dd) Temozolomide (TMZ) in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma

Maximum tolerated dose of Zotiraciclib (TG02) in combination with dose dense Temozolomide (TMZ) was assessed using the Bayesian Optimal Interval (BOIN) design. The MTD is defined as the dose for which the isotonic estimate of the toxicity rate is closest to the target toxicity rate of 0.35. If there are ties, we select the higher dose level when the isotonic estimate is lower than the target toxicity rate; and we select the lower dose level when the isotonic estimate is greater than the target toxicity rate of 0.35. (NCT02942264)
Timeframe: 4 weeks after initiation of treatment

Interventionmg/day (Number)
All Participants250

[back to top]

Phase I: Number of Participants With a Dose-limiting Toxicity (DLT)

DLT is defined as any adverse events attributed to the study drug. For example, Grade 4 neutropenia lasting 5 days or more. Febrile neutropenia defined as grade 3-4 neutropenia with fever ≥38.5ºC and/or infection requiring antibiotic or antifungal treatment. Nausea or vomiting that responds to symptomatic therapy and lasts ≤7 days. Fatigue that responds to symptomatic therapy and lasts ≤7 days. And weight gain (in patients on steroids). (NCT02942264)
Timeframe: 4 weeks after initiation of treatment

InterventionParticipants (Count of Participants)
ARM 1 Dose Level 0 - (Starting Dose)1
ARM 1 Dose Level 1 - (Dose Escalation)3
ARM 2 Dose Level 0 (Starting Dose)0
ARM 2 Dose 1 - (Dose Escalation)3
ARM 2 Dose 0 - (Dose De-escalation)1
ARM 2 Dose Level II - (Dose Escalation)1
ARM 2 Dose Level I - (Dose De-escalation)4

[back to top]

% of Participants Free of Disease Progression at 4mos Treated w/Zotiraciclib at the Maximum Tolerated Dose (MTD) in Combination With the Metronomic (mn) Temozolomide (TMZ) in Adult Patients With Recurrent Anaplastic Astrocytoma or Glioblastoma/Gliosarcoma

We first determined the MTDs in each ARM and we then performed the cohort expansion at the MTD in both ARMs separately, until we treated a total of 18 participants at this dose in each ARM. PFS is defined as the duration of time from start of registration to time of progression or death, whichever comes first. Progression was assessed by the Response Assessment in Neuro-Oncology Criteria (RANO). Progression is ≥25% increase in tumor volume compared to baseline in the sum of the products of perpendicular diameters of enhancing lesions compared with the smallest measurement obtained either at baseline or best response with the participant on stable or increasing doses of steroids. Significant increase in T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) non-enhancing lesions with the participant on stable or increasing doses of steroids (not caused by comorbid events). Any new lesions. (NCT02942264)
Timeframe: 4 months

Interventionpercentage of participants (Number)
All Participants25

[back to top]

Area Under the Serum of Nanoparticle Albumin-bound Rapamycin Concentration Curve

Median with minimum and maximum values of the area under the drug concentration over time curve stratified by dose level. (NCT02975882)
Timeframe: Days 1, 2, 3, 4, and 8

Interventionhr*ng/mL (Mean)
Stratum Cohort With Dose Level 113654
Stratum Cohort With Dose Level -19532
Stratum Cohort With Dose Level -28666
PK Cohort With Dose Level -27988.5

[back to top]

Number of Patients With Cycle 1 and 2 Dose Limiting Toxicities Attributable to Nanoparticle Albumin-bound Rapamycin

Number of patients with cycle 1 and 2 dose limiting toxicities possibly, probably, or definitely attributable with nanoparticle albumin-bound rapamycin stratified by dose level. (NCT02975882)
Timeframe: Up to 42 days

InterventionParticipants (Count of Participants)
Stratum Cohort With Dose Level 11
Stratum Cohort With Dose Level -12
Stratum Cohort With Dose Level -21
PK Cohort With Dose Level -21

[back to top]

Number of Patients With Adverse Events

The number of patients with adverse events that are at least possibly attributable to nanoparticle albumin-bound rapamycin stratified by dose level. (NCT02975882)
Timeframe: Up to 24 months

InterventionParticipants (Count of Participants)
Stratum Cohort With Dose Level 14
Stratum Cohort With Dose Level -112
Stratum Cohort With Dose Level -28
PK Cohort With Dose Level -25

[back to top]

Number of Patients With Antitumor Activity of Nanoparticle Albumin-bound Rapamycin

The number of response-evaluable patients with best overall response of complete response (CR), disappearance of all target lesions; or partial response (PR), >=50% decrease in the sum of the products of the two perpendicular diameters of all target lesions (up to 5), while on study therapy stratified by dose level; Overall response (OR)=CR+PR. (NCT02975882)
Timeframe: Up to 24 months

InterventionParticipants (Count of Participants)
Stratum Cohort With Dose Level 11
Stratum Cohort With Dose Level -10
Stratum Cohort With Dose Level -20
PK Cohort With Dose Level -20

[back to top]

Objective Response Rate

ORR will be defined as the percentage of patients with complete response (CR) or partial response (PR) according to the RANO criteria. (NCT03034135)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Complete responsePartial Response
DSF-Cu00

[back to top]

Overall Survival

Percentage of patients that are alive (NCT03034135)
Timeframe: 6 months and 12 months

Interventionpercentage of participants (Number)
6 months12 months
DSF-Cu6135

[back to top]

Median Duration of Overall Survival

Duration of overall survival for patients that are alive (NCT03034135)
Timeframe: 14 months

Interventionmonths (Median)
DSF-Cu7.1

[back to top]

Progression Free Survival

Percentage of patients that are free from progressive disease per RANO criteria (NCT03034135)
Timeframe: 6 months

Interventionpercentage of participants (Number)
DSF-Cu14

[back to top]

Median Progression Free Survival

Duration of progression free survival according to RANO criteria (NCT03034135)
Timeframe: 12 months

Interventionmonths (Median)
DSF-Cu1.7

[back to top]

Number of Participants With Serious Adverse Events

Number of Participants with Grade 3 and 4 serious adverse events (NCT03034135)
Timeframe: 14 months

InterventionParticipants (Count of Participants)
DSF-Cu2

[back to top]

Toxicities

Treatment emergent toxicities of nanoliposomal irinotecan with continuous low-dose temozolomide using CTCAE version 4.03, grades 2 through 4 (NCT03119064)
Timeframe: Baseline through 30 days post off study treatment

,
Interventionevents (Number)
NeutropeniaALT/ASTHypokalemiaHypophosphatemiaNauseaFatigueDiarrheaAnorexiaDehydrationUrticaria
Dose 10211120001
Dose 21110222220

[back to top]

Response

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR." (NCT03119064)
Timeframe: Every 2 months on study treatment then very 3 months once treatment has stopped, until progression of disease up to 2 years.

,
Interventionparticipants (Number)
Partial ResponseProgressive Disease
Dose 118
Dose 212

[back to top]

Determination of Maximum Tolerated Dose (MTD)

To evaluate the maximum tolerated dose of nanoliposomal irinotecan with continuous low-dose temozolomide for patients with recurrent glioblastoma. (NCT03119064)
Timeframe: Every two weeks for 4 weeks

Interventionmg/m^2 (Number)
All Participants50

[back to top]

Time to Progression

Percentage change of the Cho/NAA, ADC and nCBV from baseline to the end of radiation therapy, RT will be used for assessment of time to progression. (NCT03168919)
Timeframe: From Baseline through 24 months.

Interventiondays (Mean)
Chemoradiation Arm465

[back to top]

Changes of Tumor Angiogenesis

Perfusion MRI will be used for assessment of tumor angiogenesis (with median normalized cerbral blood volume, nCBV) during the course of fractionated radiation treatment. (NCT03168919)
Timeframe: From baseline to 6 weeks

InterventionmL/100 gm of brain tissue (Mean)
baselineEarly in therapy (At 2 weeks in therapy)Late therapy (At 4 weeks in therapy)At completion (At 6 weeks from baseline)
Chemoradiation Arm3.783.351.581.2

[back to top]

Changes of Tumor Cellularity

Diffusion MRI will be used for assessment of measures tumor cellularity (with minimum apparent diffusion co-efficent, ADC) during the course of fractionated radiation treatment. (NCT03168919)
Timeframe: From baseline to 6 weeks

Interventionmm^2/sec (Mean)
BaselineEarly in therapy (At 2 weeks in therapy)Late therapy (At 4 weeks in therapy)At completion (At 6 weeks from baseline)
Chemoradiation Arm0.0006750.0007980.0008420.001144

[back to top]

Changes of Tumor Volume

MRI will be used for assessment of measures tumor volume. (NCT03168919)
Timeframe: From baseline Up to 6 weeks

Interventioncm3 (Mean)
BaselineEarly in therapy (At 2 weeks in therapy)Late therapy (At 4 weeks in therapy)At completion (At 6 weeks from baseline)
Chemoradiation Arm6.13.792.662.16

[back to top]

Phase I: 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. (NCT03190967)
Timeframe: Date treatment consent signed to date off study, approximately 44 months and 27 days for level 1, 28 months and 10 days for level 2, and 40 months and 8 days for level 3.

InterventionParticipants (Count of Participants)
Level 1 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 30 mg/m^23
Level 2 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 40 mg/m^23
Level 3 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 50 mg/m^26

[back to top]

Phase I: Number of Participants With Dose Limiting Toxicity (DLTs) at Each Dose Level

Number of participants with DLTs at each dose level 30 days after treatment. A DLT is defined as grade 3 or higher non-hematologic adverse events excluding grade 3 hypertension controlled with anti-hypertensive therapy; or grade 3 asymptomatic electrolytes imbalance; grade 3 endocrinopathy; grade 3 asymptomatic increase in aspartate aminotransferase or alanine aminotransferase; and transient (lasting less than <48 hours) nausea, emesis, or diarrhea if corrected with conservative measures within 24-48 hours. A hematologic grade 4 neutropenia of ≥ 7 days duration, grade ≥ 3 thrombocytopenia, and all other grade 4 hematologic toxicities excluding grade 4 lymphopenia, or leukopenia in the absence of grade 3 or higher neutropenia. Grade 3 is severe. Grade 4 is life-threatening. (NCT03190967)
Timeframe: After first cycle of treatment, up to 30 days

InterventionParticipants (Count of Participants)
Level 1 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 30 mg/m^20
Level 2 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 40 mg/m^20
Level 3 Phase I:Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 50 mg/m^20

[back to top]

Phase I: Standard Time to Progression (TTP)

TTP is the time between the first day of treatment to the day of disease progression. Progression was assessed using the Response Evaluation Criteria in Solid Tumors (RECIST). Progression is at least a 25% increase in the sum of products of all measurable lesions over smallest sum observed, clear worsening of any evaluable disease, appearance of any new lesion/site, or failure to return for evaluation due to death or deteriorating condition. (NCT03190967)
Timeframe: From first day of treatment to the day of disease progression, an average of 15 months.

InterventionMonths (Mean)
Level 1 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 30 mg/m^26.95
Level 2 Phase I:Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 40 mg/m^216.26
Level 3 Phase I:Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 50 mg/m^217.46

[back to top]

Phase I: Median Survival

Median amount of time participants survives after therapy. Survival compared between the two arms and Kaplan-Meier curves constructed with a two-tailed log-rank test used to compare the arms. (NCT03190967)
Timeframe: From date of first therapy until death, an average of 40.79 months

InterventionMonths (Median)
Level 1 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 30 mg/m^237.8
Level 2 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 40 mg/m^248.4
Level 3 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 50 mg/m^238.5

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Temozolomide (TMZ) When Used With T-DM1 (Ado-trastuzumab)

Maximum tolerated dose of TMZ when used in combination with T-DM1. MTD is defined as the dose level at which 0 or 1 participant in 6 has a dose limiting toxicity (DLT). A DLT is defined as grade 3 or higher non-hematologic adverse events excluding grade 3 hypertension controlled with anti-hypertensive therapy; or grade 3 asymptomatic electrolytes imbalance; grade 3 endocrinopathy; grade 3 asymptomatic increase in aspartate aminotransferase or alanine aminotransferase; and transient (lasting less than <48 hours) nausea, emesis, or diarrhea if corrected with conservative measures within 24-48 hours. A hematologic grade 4 neutropenia of ≥ 7 days duration, grade ≥ 3 thrombocytopenia, and all other grade 4 hematologic toxicities excluding grade 4 lymphopenia, or leukopenia in the absence of grade 3 or higher neutropenia. Grade 3 is severe. Grade 4 is life-threatening. (NCT03190967)
Timeframe: first 21 days of treatment

Interventionmg/m^2 (Number)
All Participants40

[back to top]

Phase I: Number of Participants With Grade 3 and/or Grade 4 Adverse Events

Adverse events were assessed by the Common Terminology Criteria for Adverse Events (CTCAE v4.0). Grade 3 is severe. Grade 4 is life-threatening. (NCT03190967)
Timeframe: Evaluated at the beginning of every cycle while on study, for an average of 9.6 months (range 2.8-33.9 months).

,,
InterventionParticipants (Count of Participants)
Grade 3 AnemiaGrade 3 Aspartate aminotransferase increasedGrade 3 CD4 lymphocytes decreasedGrade 3 DysphasiaGrade 3 HypokalemiaGrade 3 Lymphocyte count decreasedGrade 3 Neoplasms benign, malignant and unspecified (incl cysts and polyps) - Malignant Neoplasm-CMLGrade 3 Neutrophil count decreasedGrade 3 Platelet count decreasedGrade 3 Surgical and medical procedures - Other, Lap Cholecystectomy/Hernia RepairGrade 3 White blood cell decreasedGrade 4 White blood cell decreasedGrade 4 CD4 lymphocytes decreasedGrade 4 Lymphocyte count decreasedGrade 5
Level 1 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 30 mg/m^2001101000000000
Level 2 Phase I: Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 40 mg/m^2101002000000000
Level 3 Phase I:Ado-trastuzumab (T-DMI) 3.6 mg/kg + Temozolomide (TMZ) 50 mg/m^2015015111110220

[back to top]

Number of Participants With Grade 3 or Greater Toxicities Associated With Pevonedistat in Combination With Irinotecan and Temozolomide

Frequency of patients who experience at least one grade 3 or higher toxicity that is at least possibly attributable to pevonedistat using the Common Terminology Criteria for Adverse Events version 5.0 by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 3 years 8 months

InterventionParticipants (Count of Participants)
Part A Dose Level 1: 15 mg/m^25
Part A Dose Level 2: 20 mg/m^24
Part A Dose Level 3: 25 mg/m^26
Part A Dose Level 4: 35 mg/m^22
Part A PK Dose Level 4: 35 mg/m^24

[back to top]

Anti-tumor Activity of Pevonedistat in Combination With Irinotecan and Temozolomide

Frequency of disease response (best overall response of partial or complete response) assessed per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR by dose level. (NCT03323034)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Part A Dose Level 1: 15 mg/m^21
Part A Dose Level 2: 20 mg/m^21
Part A Dose Level 3: 25 mg/m^20
Part A Dose Level 4: 35 mg/m^20
Part A PK Dose Level 4: 35 mg/m^20

[back to top]

AUC of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (range) of the area under the drug concentration over time (pre-dose and then 0, 1, 2, 4, 6-8, and 24-hours post-dose infusion) curve stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionhr*ng/mL (Median)
Part A Dose Level 1: 15 mg/m^2803
Part A Dose Level 2: 20 mg/m^2973.5
Part A Dose Level 3: 25 mg/m^21340
Part A Dose Level 4: 35 mg/m^22018.5
Part A PK Dose Level 4: 35 mg/m^21957

[back to top]

C Max of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the maximum (peak) serum concentration by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionng/mL (Median)
Part A Dose Level 1: 15 mg/m^2156
Part A Dose Level 2: 20 mg/m^2218.5
Part A Dose Level 3: 25 mg/m^2303.5
Part A Dose Level 4: 35 mg/m^2419
Part A PK Dose Level 4: 35 mg/m^2363.8

[back to top]

Clearance of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the rate of elimination of the drug by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

InterventionL/hr/m^2 (Median)
Part A Dose Level 1: 15 mg/m^218.8
Part A Dose Level 2: 20 mg/m^220.8
Part A Dose Level 3: 25 mg/m^218.9
Part A Dose Level 4: 35 mg/m^217.5
Part A PK Dose Level 4: 35 mg/m^217.9

[back to top]

Half-life of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the time required for the serum concentration to fall to 50% of its starting dose by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionhours (Median)
Part A Dose Level 1: 15 mg/m^25.3
Part A Dose Level 2: 20 mg/m^25
Part A Dose Level 3: 25 mg/m^25.2
Part A Dose Level 4: 35 mg/m^24.7
Part A PK Dose Level 4: 35 mg/m^26.1

[back to top]

MTD/RP2D of Pevonedistat in Combination With Irinotecan and Temozolomide

The maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) will be the maximum dose at which fewer than one-third of patients experience dose limiting toxicities. (NCT03323034)
Timeframe: Up to 28 days

Interventionmg/m^2 (Number)
Treatment (Pevonedistat, Temozolomide, Irinotecan)35

[back to top]

T Max of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the time at which the maximum (peak) serum concentration occurs by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionhours (Median)
Part A Dose Level 1: 15 mg/m^21.
Part A Dose Level 2: 20 mg/m^21
Part A Dose Level 3: 25 mg/m^21
Part A Dose Level 4: 35 mg/m^21.1
Part A PK Dose Level 4: 35 mg/m^21.1

[back to top]

6 Months Progression Free Survival (PFS)

To determine 6 month PFS of patients with unmethylated glioblastoma treated with DSF-Cu in combination with concurrent radiation and temozolomide. This was assessed by the number of participants who did not have disease progression and were alive at 6 months. (NCT03363659)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Open Label1

[back to top]

Overall Survival

Overall Survival will be assessed as a number of participants alive at 1 and 2 years. (NCT03363659)
Timeframe: 1 and 2 years

InterventionParticipants (Count of Participants)
1 Year2 Years
Open Label92

[back to top]

Number of Participants With Depatuxizumab Mafodotin Dose Modifications to Ocular Side Effects After Bandage Contact Lens (BCL) Intervention

Dose modifications included depatuxizumab mafodotin withdrawal, interruption, and reductions in dose initiated due to OSEs after BCL intervention. (NCT03419403)
Timeframe: From the last assessment prior to BCL intervention to the end of BCL intervention, up to 38 weeks

Interventionparticipants (Number)
Standard Steroids2
Standard Steroids + Vasoconstrictor + Cold Compress1
Enhanced Steroids + Vasoconstrictor + Cold Compress0

[back to top]

Percentage of Participants Who Required a Change in Ocular Side Effect (OSE) Management

Inadequate control of ocular side effects (OSE) was defined as either a ≥ 3-line decline from baseline (≥ +0.3 on LogMAR scale) in visual acuity (with baseline correction determined at the screening ophthalmology visit)) or ≥ Grade 3 OSE severity on the Corneal Epithelial Adverse Event (CEAE) scale. (NCT03419403)
Timeframe: Within 8 weeks after the initial dose of depatuxizumab mafodotin

,,
Interventionpercentage of participants (Number)
Bilateral visionVision in worst eye
Enhanced Steroids + Vasoconstrictor + Cold Compress41.750.0
Standard Steroids50.064.3
Standard Steroids + Vasoconstrictor + Cold Compress27.372.7

[back to top]

Maximum Change From Baseline on the Logarithm of the Minimum Angle of Resolution (LogMAR) Scale

The LogMAR scale measures visual acuity on a continuous scale, with a LogMAR value of 0 equivalent to 20/20 visual acuity. Normal vision is considered to be from -0.2 - 0.1; higher values indicate visual impairment. The baseline observation is defined as the last non-missing measurement collected prior to the first dose of depatuxizumab mafodotin. (NCT03419403)
Timeframe: Within 8 weeks after the initial dose of depatuxizumab mafodotin

,,
Interventionunits on a scale (Mean)
Bilateral visionVision in worst eye
Enhanced Steroids + Vasoconstrictor + Cold Compress0.2720.430
Standard Steroids0.3530.482
Standard Steroids + Vasoconstrictor + Cold Compress0.4020.528

[back to top]

Change From Baseline In Logarithm of the Minimum Angle of Resolution (LogMAR) Scale After Bandage Contact Lens (BCL) Intervention

The change on the LogMAR Scale from last assessment prior to BCL intervention to 2 weeks after BCL intervention was calculated. The LogMAR scale measures visual acuity on a continuous scale, with a LogMAR value of 0 equivalent to 20/20 visual acuity. Normal vision is considered to be from -0.2 - 0.1; higher values indicate visual impairment. (NCT03419403)
Timeframe: From the last assessment prior to BCL intervention to 2 weeks after BCL intervention

,
Interventionunits on a scale (Mean)
Within 8 weeks of first dose: Bilateral visionWithin 8 weeks of first dose: Vision in worst eyeWithin 8 weeks of first adjuvant dose: Bilateral visionWithin 8 weeks of first adjuvant dose: Vision in worst eye
Standard Steroids0.3250.435-0.1-0.1
Standard Steroids + Vasoconstrictor + Cold Compress0.130.520.410.33

[back to top]

Time to Bandage Contact Lens (BCL) Intervention

The time to initiation of bandage contact lenses for those participants who required intervention due to inadequate control of ocular side effects (OSE) was calculated. (NCT03419403)
Timeframe: Up to 9 months after the first dose of depatuxizumab mafodotin

Interventionmonths (Median)
Standard SteroidsNA
Standard Steroids + Vasoconstrictor + Cold Compress3.6
Enhanced Steroids + Vasoconstrictor + Cold Compress2.1

[back to top]

Cumulative Dose of Depatuxizumab Mafodotin Received During Chemoradiation and During Adjuvant Treatment

The cumulative dose of depatuxizumab mafodotin administered was tabulated. (NCT03419403)
Timeframe: Up to 9 months

Interventionmg/kg (Mean)
Standard Steroids8.5
Standard Steroids + Vasoconstrictor + Cold Compress10.5
Enhanced Steroids + Vasoconstrictor + Cold Compress7.0

[back to top]

Treatment-Emergent Corneal Epithelial Adverse Event (CEAE) Grade at Each Visit

The corneal epithelial adverse event (CEAE) rating scale is designed to record symptoms associated with corneal epitheliopathy caused by antibody-drug conjugates and to grade the severity of findings. The overall CEAE grade is measured on a scale of 0 to 5, with higher values being more severe, reflecting the impact of corneal abnormalities on visual activities of daily living (ADLs). Additional detailed information is collected for specific domains that are commonly affected, with the following ranges (each in order of increasing severity): ocular discomfort (0 - 4), photophobia (0 - 3), and reading (1 - 3). (NCT03419403)
Timeframe: Up to 47 weeks

,,
InterventionParticipants (Count of Participants)
Week 1: Grade 0Week 1: Grade 1Week 3: Grade 0Week 3: Grade 1Week 3: Grade 2Week 5: Grade 0Week 5: Grade 1Week 5: Grade 2Week 5: Grade 3Week 7: Grade 0Week 7: Grade 1Week 7: Grade 2Week 7: Grade 3Week 9: Grade 0Week 9: Grade 1Week 9: Grade 2Week 9: Grade 3Week 11: Grade 0Week 11: Grade 1Week 11: Grade 2Week 11: Grade 3Week 11: Grade 4Week 13: Grade 1Adj Week 1: Grade 0Adj Week 1: Grade 1Adj Week 1: Grade 2Adj Week 1: Grade 3Adj Week 5: Grade 0Adj Week 5: Grade 1Adj Week 5: Grade 2Adj Week 5: Grade 3Adj Week 9: Grade 0Adj Week 9: Grade 1Adj Week 9: Grade 2Adj Week 9: Grade 3Adj Week 13: Grade 0Adj Week 13: Grade 1Adj Week 13: Grade 2Adj Week 13: Grade 3Adj Week 17: Grade 0Adj Week 17: Grade 1Adj Week 17: Grade 2Adj Week 21: Grade 1Adj Week 21: Grade 2Adj Week 25: Grade 1Adj Week 25: Grade 2Adj Week 29: Grade 2Adj Week 29: Grade 4
Enhanced Steroids + Vasoconstrictor + Cold Compress517312622205210411031010220201001200020100100000
Standard Steroids10141001714034211320221110131014100210021002020110
Standard Steroids + Vasoconstrictor + Cold Compress835600532046002500100001320230112211220013032101

[back to top]

Treatment Emergent Corneal Epithelial Adverse Event (CEAE) Grade at Each Visit After Bandage Contact Lens (BCL) Intervention

The corneal epithelial adverse event (CEAE) rating scale is designed to record symptoms associated with corneal epitheliopathy caused by antibody-drug conjugates and to grade the severity of findings. The overall CEAE grade is measured on a scale of 0 to 5, with higher values being more severe, reflecting the impact of corneal abnormalities on visual activities of daily living (ADLs). Additional detailed information is collected for specific domains that are commonly affected, with the following ranges (each in order of increasing severity): ocular discomfort (0 - 4), photophobia (0 - 3), and reading (1 - 3). (NCT03419403)
Timeframe: From the last assessment prior to BCL intervention to the end of BCL intervention, up to 38 weeks

,,
InterventionParticipants (Count of Participants)
Week 5: Grade 1Week 7: Grade 2Week 9: Grade 2Week 11: Grade 2Week 11: Grade 3Week 13: Grade 1Adj Week 1: Grade 1Adj Week 1: Grade 2Adj Week 5: Grade 0Adj Week 5: Grade 1Adj Week 5: Grade 2Adj Week 5: Grade 3Adj Week 9: Grade 0Adj Week 9: Grade 1Adj Week 9: Grade 2Adj Week 9: Grade 3Adj Week 13: Grade 0Adj Week 13: Grade 1Adj Week 13: Grade 2Adj Week 13: Grade 3Adj Week 17: Grade 1Adj Week 17: Grade 2Adj Week 21: Grade 2Adj Week 25: Grade 1Adj Week 25: Grade 2Adj Week 29: Grade 2
Enhanced Steroids + Vasoconstrictor + Cold Compress00120011101001100010000000
Standard Steroids21101100001100010011012011
Standard Steroids + Vasoconstrictor + Cold Compress01100000110010002100111100

[back to top]

Number of Participants With Depatuxizumab Mafodotin Dose Modifications Due to Ocular Side Effects (OSE)

Dose modifications included depatuxizumab mafodotin withdrawal, interruption, and reductions in dose initiated due to OSEs. (NCT03419403)
Timeframe: From the first dose of study drug until 49 days after last depatuxizumab mafodotin administration, up to 47 weeks

InterventionParticipants (Count of Participants)
Standard Steroids2
Standard Steroids + Vasoconstrictor + Cold Compress3
Enhanced Steroids + Vasoconstrictor + Cold Compress2

[back to top]

Change From Baseline In Logarithm of the Minimum Angle of Resolution (LogMAR) Scale After Bandage Contact Lens (BCL) Intervention

The change on the LogMAR Scale from last assessment prior to BCL intervention to 2 weeks after BCL intervention was calculated. The LogMAR scale measures visual acuity on a continuous scale, with a LogMAR value of 0 equivalent to 20/20 visual acuity. Normal vision is considered to be from -0.2 - 0.1; higher values indicate visual impairment. (NCT03419403)
Timeframe: From the last assessment prior to BCL intervention to 2 weeks after BCL intervention

Interventionunits on a scale (Mean)
Within 8 weeks of first dose: Bilateral visionWithin 8 weeks of first dose: Vision in worst eye
Enhanced Steroids + Vasoconstrictor + Cold Compress0.540.867

[back to top]

PFS Rate at 6 Months and 12 Months

"Progression-free survival rate at 6 months and 12 months was calculated as the proportion of patients who were progression-free and alive at 6 and 12 months, respectively.~Progression is assessed according to Response Assessment in Neuro-Oncology (RANO) 2010 criteria based on MRI imaging, and includes ≥25% increase in the sum of the products of perpendicular diameters of enhancing lesions (compared with baseline if no decrease) on stable or increasing doses of corticosteroids." (NCT03463265)
Timeframe: 6 and 12 months

,,,,,
Interventionpercentage of patients (Number)
PFS at 6 monthsPFS at 12 months
Arm A, Cohort 1: Nab-sirolimus in Patients With Recurrent High Grade Glioma0.00.0
Arm A, Cohort 2: Nab-sirolimus + Temozolomide in Patients With Recurrent High Grade Glioma75.050.0
Arm A, Cohort 3: Nab-sirolimus + Bevacizumab in Patients With Recurrent High Grade Glioma37.512.5
Arm A, Cohort 4: Nab-sirolimus + Lomustine in Patients With Recurrent High Grade Glioma25.00.0
Arm A, Cohort 5: Nab-sirolimus + Marizomib in Patients With Recurrent High Grade Glioma10.00.0
Arm B: Nab-sirolimus + Temozolomide + Radiotherapy in Patients With Newly Diagnosed Glioblastoma76.936.4

[back to top]

OS at 12 Months

Overall Survival rate at 12 months (NCT03463265)
Timeframe: 12 months

Interventionpercentage of patients (Number)
Arm A, Cohort 1: Nab-sirolimus in Patients With Recurrent High Grade Glioma0.0
Arm A, Cohort 2: Nab-sirolimus + Temozolomide in Patients With Recurrent High Grade Glioma66.7
Arm A, Cohort 3: Nab-sirolimus + Bevacizumab in Patients With Recurrent High Grade Glioma25.0
Arm A, Cohort 4: Nab-sirolimus + Lomustine in Patients With Recurrent High Grade Glioma25.0
Arm A, Cohort 5: Nab-sirolimus + Marizomib in Patients With Recurrent High Grade Glioma0.0
Arm B: Nab-sirolimus + Temozolomide + Radiotherapy in Patients With Newly Diagnosed Glioblastoma53.8

[back to top]

OS

Median Overall Survival (NCT03463265)
Timeframe: Through study completion (up to 48 months)

Interventionmonths (Median)
Arm A, Cohort 1: Nab-sirolimus in Patients With Recurrent High Grade Glioma7.2
Arm A, Cohort 2: Nab-sirolimus + Temozolomide in Patients With Recurrent High Grade Glioma13.8
Arm A, Cohort 3: Nab-sirolimus + Bevacizumab in Patients With Recurrent High Grade Glioma6.8
Arm A, Cohort 4: Nab-sirolimus + Lomustine in Patients With Recurrent High Grade Gliom7.5
Arm A, Cohort 5: Nab-sirolimus + Marizomib in Patients With Recurrent High Grade Glioma6.7
Arm B: Nab-sirolimus + Temozolomide + Radiotherapy in Patients With Newly Diagnosed Glioblastoma13.3

[back to top]

ORR

Objective overall response rate (ORR, according to Response Assessment in Neuro-Oncology [RANO]) by investigator-assessed radiologic review and defined as the proportion of patients who achieved a confirmed partial response (PR) or confirmed complete response (CR) per RANO 2010 criteria. PR is defined as greater than or equal to 50% decrease compared with baseline in the sum of products of perpendicular diameters of all measurable enhancing lesions sustained for at least 4 weeks. (NCT03463265)
Timeframe: Through study completion (up to 48 months)

Interventionpercentage of patients (Number)
Arm A, Cohort 1: Nab-sirolimus in Patients With Recurrent High Grade Glioma0
Arm A, Cohort 2: Nab-sirolimus + Temozolomide in Patients With Recurrent High Grade Glioma0
Arm A, Cohort 3: Nab-sirolimus + Bevacizumab in Patients With Recurrent High Grade Glioma0
Arm A, Cohort 4: Nab-sirolimus + Lomustine in Patients With Recurrent High Grade Glioma0
Arm A, Cohort 5: Nab-sirolimus + Marizomib in Patients With Recurrent High Grade Glioma0
Arm B: Nab-sirolimus + Temozolomide + Radiotherapy in Patients With Newly Diagnosed Glioblastoma11.5

[back to top]

Median PFS

"Progression-free Survival defined as number of months from the date of the first dose of study drug to the first observation of a disease progression or death due to any cause.~Progression is assessed according to Response Assessment in Neuro-Oncology (RANO) 2010 criteria based on MRI imaging, and includes ≥25% increase in the sum of the products of perpendicular diameters of enhancing lesions (compared with baseline if no decrease) on stable or increasing doses of corticosteroids." (NCT03463265)
Timeframe: Through study completion (up to 48 months)

Interventionmonths (Median)
Arm A, Cohort 1: Nab-sirolimus in Patients With Recurrent High Grade Glioma1.7
Arm A, Cohort 2: Nab-sirolimus + Temozolomide in Patients With Recurrent High Grade Glioma11.3
Arm A, Cohort 3: Nab-sirolimus + Bevacizumab in Patients With Recurrent High Grade Glioma3.1
Arm A, Cohort 4: Nab-sirolimus + Lomustine in Patients With Recurrent High Grade Glioma3.8
Arm A, Cohort 5: Nab-sirolimus + Marizomib in Patients With Recurrent High Grade Glioma1.7
Arm B: Nab-sirolimus + Temozolomide + Radiotherapy in Patients With Newly Diagnosed Glioblastoma7.5

[back to top]

Overall Response Rate (ORR)

ORR is defined as the proportion of participants who have prolonged stable disease or a partial or complete response or complete response to therapy according to RECIST 1.1. (NCT03495921)
Timeframe: 6 months after treatment with Vigil.

InterventionProportion of participants. (Number)
Group A: Vigil in Combination With Irinotecan and Temozolomide0
Group B: Irinotecan and Temozolomide0

[back to top]

Overall Survival (OS)

OS is defined as time from randomization to death or to the date of last follow-up. The date of last follow-up confirming survival will be used as the censoring date for subjects who are alive and/or do not have a known date of death. (NCT03495921)
Timeframe: From date of randomization until date of death from any cause, whichever came first (assessed up to 3 years).

InterventionMonths (Median)
Group A: Vigil in Combination With Irinotecan and Temozolomide16.1
Group B: Irinotecan and Temozolomide3.3

[back to top]

Vigil Manufacture Success Rate: Number of Participants Eligible for Treatment on the Main Study.

Participants were considered eligible for treatment, if the tissue submitted to Gradalis met all criteria, including manufacturing product release criteria. (NCT03495921)
Timeframe: From manufacturing start date until 4 weeks post manufacturing for each tissue procurement (assessed up to 17 months).

InterventionParticipants (Count of Participants)
Total Number of Tissue Procurements4

[back to top]

Progression Free Survival (PFS)

Progression Free Survival (PFS) is defined as the time from randomization to the event of disease recurrence/progression according to Response Evaluation Criteria In Solid Tumors (RECIST) criteria (version 1.1) for target lesions and assessed CT/MRI by local investigator. (NCT03495921)
Timeframe: From date of randomization until the date of first documented progression (assessed up to 3 years).

InterventionMonths (Median)
Group A: Vigil in Combination With Irinotecan and Temozolomide2.8
Group B: Irinotecan and Temozolomide9.1

[back to top]

Overall Survival (OS)

The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated utilizing exact binomial methodology. The distribution of survival time will be estimated using the method of Kaplan-Meier (1958). (NCT03778294)
Timeframe: Time from registration to death due to any cause, assessed up to 12 months

Interventionproportion of successes (Number)
Treatment (18F-DOPA, PET/MRI, PET/CT, Temozolomide)0.54

[back to top]

Progression Free Survival

The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated utilizing exact binomial methodology. (NCT03778294)
Timeframe: At 12 months after radiation therapy

Interventionproportion of successes (Number)
Treatment (18F-DOPA, PET/MRI, PET/CT, Temozolomide)0.72

[back to top]

Number of Patients Experiencing Adverse Events

Recorded at each clinical visit and will be categorized according to National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. Number of patients experiencing a grade 3 or greater adverse event will be reported. (NCT03880019)
Timeframe: Up to 2 years after study treatment

InterventionParticipants (Count of Participants)
Treatment (Olaparib, Temozolomide)20

[back to top]

Confirmed Objective Response Rate (ORR) (Complete Response + Partial Response)

Will be measured by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 criteria assessed by MRI: Complete Response (CR), disappearance of all target lesions, any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm (<1 cm); Partial Response (PR), at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters. A response rate of 10% is considered inactive and unworthy of further study. A response rate of 35% would be promising for further study among patients with advanced uterine leiomyosarcoma (LMS) treated with at least one prior systemic regimen. A response rate of 35% for the temozolomide (TMZ) + poly(adenosine diphosphate[ADP]-ribose) polymerase inhibitor (PARPi) combination would also be suggestive of superior efficacy over TMZ monotherapy in sarcoma. Will be reported with a 95% confidence interval. (NCT03880019)
Timeframe: Within first 6 months of study treatment

InterventionParticipants (Count of Participants)
Treatment (Olaparib, Temozolomide)5

[back to top]

Number of Subjects With Radiographic Response, as Defined by the RECIST Criteria.

"Evaluation of Target lesions:~Complete Response (CR):~Disappearance of all target lesions.~Partial Response (PR):~At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.~Progressive Disease (PD):~At least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum onstudy (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progression).~Stable Disease (SD):~Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study." (NCT03930771)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
All Patients0

[back to top]

Safety, as Measured by the Number of Subjects With at Least One AE

(NCT03930771)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
All Patients1

[back to top]

Tolerability of the TMZ and Capecitabine Combination, as Measured by Number of Participants With a Dose-limiting Toxicity

(NCT03930771)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
All Patients1

[back to top]

Effect of the CAP and TMZ Combination on Pituitary Function, Measured by Changes in Pituitary Hormone Secretion in Patients

Serum prolactin, IGF-1, ACTH, FSH, LH and TSH levels are used to assess changes in pituitary hormone function. These tests will only be repeated if found to be abnormal at baseline. (NCT03930771)
Timeframe: At baseline and every 8 weeks, up to 6 months

Interventionng/mL (Number)
BaselineCycle 3 Day 1Cycle 5 Day 1Cycle 6 Day 1End of TreatmentFollow-Up Week 6
All Patients134.786.969.469.880.177.0

[back to top]

Progression Free Survival

To estimate the progression free survival (PFS), patients will be followed for up to 24 months. (NCT04166435)
Timeframe: Up to 2 years from last treatment

Interventionmonths (Median)
Temozolomide + Olaparib3

[back to top]

Count of Participants With Adverse Events Greater or Equal to 3

To determine the safety of TMZ in combination with olaparib, the number of participants with ≥ grade 3 treatment related adverse events by CTCAE v5.0 coding will be assessed. (NCT04166435)
Timeframe: Up to 2 years from last treatment

InterventionParticipants (Count of Participants)
Temozolomide + Olaparib7

[back to top]

Overall Survival

To estimate overall survival (OS), patients will be followed for up to 24 months. (NCT04166435)
Timeframe: Up to 2 years from last treatment

Interventionmonths (Median)
Temozolomide + Olaparib9.4

[back to top]

Overall Survival

(NCT05718466)
Timeframe: 12 months

Interventionmonths (Median)
Fractionated Radiosurgery and Bevacizumab7.2
Bev With Chemo4.8

[back to top]

Local Tumor Control

(NCT05718466)
Timeframe: 2 months

InterventionParticipants (Count of Participants)
Fractionated Radiosurgery and Bevacizumab14
Bev With Chemo4

[back to top]

Progression Free Survival

(NCT05718466)
Timeframe: 12 months

Interventionmonths (Median)
Fractionated Radiosurgery and Bevacizumab5.1
Bev With Chemo1.8

[back to top]