Page last updated: 2024-11-13

pemetrexed

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

Description

pemetrexed disodium : An organic sodium salt that is the disodium salt of N-{4-[2-(2-amino-4-oxo-4,7-dihydro-1H-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl}-L-glutamic acid. Inhibits thymidylate synthase (TS), 421 dihydrofolate reductase (DHFR), and glycinamide ribonucleotide formyltransferase (GARFT). [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]

theonellamide A: from Theonella marine sponges; structure in first source [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

Cross-References

ID SourceID
PubMed CID135410875
CHEMBL ID225072
CHEBI ID63616
SCHEMBL ID7968
MeSH IDM0227083
PubMed CID45382315
CHEMBL ID3908274
MeSH IDM0227083

Synonyms (90)

Synonym
nsc-698037
nsc698037
ly231514
alimta
ly-231514
pemetrexed disodium
alimta (tn)
2-{4-[2-(2-amino-4-oxo-4,7-dihydro-3h-pyrrolo[2,3-d]pyrimidin-5-yl)-ethyl]-benzoylamino}-pentanedioic acid
LYA ,
DB00642
1JUJ
l-glutamic acid,3-d]pyrimidin-5-yl]ethyl]benzoyl]-
alimta (tn) (lilly)
pemetrexed ,
pemetrexed [inn:ban]
n-(4-(2-(2-amino-3,4-dihydro-4-oxo-7h-pyrrolo(2,3-d)pyrimdin-5-yl)ethyl)benzoyl)glutamic acid
hsdb 7316
l-glutamic acid, n-(4-(2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo(2,3-d)pyrimidin-5-yl)ethyl)benzoyl)-
NCGC00167517-01
137281-23-3
(2s)-2-[[4-[2-(2-amino-4-oxo-1,7-dihydropyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]amino]pentanedioic acid
CHEMBL225072
pemetrexed (tn)
D07472
pemetrexed (inn)
chebi:63616 ,
us9422297, pemetrexed
bdbm50027656
ly-2315
A807228
n-[4-[2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid disodium salt;pemetrexed
NCGC00242485-01
n-[4-[2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid
n-[4-[2-(2-amino-4,7-dihydro-4-oxo-3h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid
n-[4-[2(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid
04q9aiz7no ,
unii-04q9aiz7no
premetrexed [common mis-spelling]
HY-10820
CS-1297
(s)-2-(4-(2-(2-amino-4-oxo-4,7-dihydro-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl)benzamido)pentanedioic acid
n-{4-[2-(2-amino-4-oxo-4,7-dihydro-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl}-l-glutamic acid
n-(4-(2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo(2,3-d)pyrimidin-5-yl)ethyl)benzoyl)-l-glutamic acid
pemfexy
l-glutamic acid, n-(4-(2-(2-amino-4,7-dihydro-4-oxo-3h-pyrrolo(2,3-d)pyrimidin-5-yl)ethyl)benzoyl)-
pemetrexed [hsdb]
pemetrexed [orange book]
pemetrexed [vandf]
pemetrexed [mi]
pemetrexed [who-dd]
pemetrexed [inn]
n-(p-(2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo(2,3-d)pyrimidin-5-yl)ethyl)benzoyl)-l-glutamate
pemetrexed [ema epar]
AKOS015896253
S5971
n-(4-[2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl)-l-glutamic acid
n-[4-[2(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo [2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid
n-[4-[2-[2-amino-4,7-dihydro-4-oxo-3h-pyrrolo[2,3-d]pyrimidin-5-yl]ethyl]benzoyl]glutamic acid
WBXPDJSOTKVWSJ-ZDUSSCGKSA-N
SCHEMBL7968
AB01273937-01
(s)-2-(4-(2-(2-amino-4-oxo-4,7-dihydro-3h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl)benzamido)pentanedioic acid
J-502393
DTXSID2048329 ,
P2288
(2s)-2-[[4-[2-(2-amino-4-oxo-3,7-dihydropyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]amino]pentanedioic acid
ly-231514 disodium hydrate
AC-1701
(s)-2-(4-(2-(2-amino-4-oxo-4,7-dihydro-1h-pyrrolo-[2,3-d]pyrimidin-5-yl)ethyl)benzamido)pentanedioic acid
(2s)-2-{[4-(2-{2-amino-4-oxo-3h,4h,7h-pyrrolo[2,3-d]pyrimidin-5-yl}ethyl)phenyl]formamido}pentanedioic acid
n-({4-[2-(2-amino-4-oxo-4,7-dihydro-1h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]phenyl}carbonyl)-l-glutamic acid
Q415220
n-[4-[2-(2-amino-3,4-dihydro-4-oxo-7h-pyrrolo[2,3-d]pyrimi-din-5-yl)ethyl]benzoyl]l-glutamic acid
1006872-74-7
AS-30680
NCGC00166414-11
l-glutamic acid, n-[4-[2-(2-amino-4,7-dihydro-4-oxo-3h-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-
AKOS016842348
pemetrexed- bio-x
BP164240
EN300-7396246
pemetrexedum
premetrexed (common mis-spelling)
multitargeted antifolate
dtxcid7028304
l-glutamic acid, n-(4-(2-(2-amino-4,7-dihydro-4-oxo-1h-pyrrolo(2,3-d)pyrimidin-5-yl)ethyl)benzoyl)
pemetrexed for injection
n-(4-(2-(2-amino-4-oxo-4,7-dihydro-1h-pyrrolo(2,3-d)pyrimidin-5-yl)ethyl)benzoyl)-l-glutamic acid
theonellamide a
CHEMBL3908274

Research Excerpts

Toxicity

Pemetrexed maintenance (median 4 cycles, range 1-44) exhibited a higher incidence of drug-related serious adverse events compared with JMDB patients. Our results would aid clinicians to make decisions of coadministration drugs to avoid drug interaction-induced side effects.

ExcerptReferenceRelevance
" The LD50 occurred at 60- and 250-fold lower doses of LY231514 in DBA/2 and CD1 nu/nu mice, respectively, maintained on low folate diet compared to standard diet."( Role of folic acid in modulating the toxicity and efficacy of the multitargeted antifolate, LY231514.
Schultz, RM; Shih, C; Worzalla, JF,
)
0.13
" No unexpected adverse events were noted."( Phase II study of efficacy and safety of bevacizumab in combination with chemotherapy or erlotinib compared with chemotherapy alone for treatment of recurrent or refractory non small-cell lung cancer.
Belani, CP; Bonomi, PD; Fehrenbacher, L; Hart, L; Herbst, RS; Lin, M; Melnyk, O; O'Neill, VJ; Ramies, D; Sandler, A, 2007
)
0.34
" Many drugs have been used for the treatment of this disease, but there is little information about how predictive factors can be used to aid treatment response and anticipate toxic effects related to anticancer treatment in colorectal cancer."( Predictive factors for chemotherapy-related toxic effects in patients with colorectal cancer.
Pantano, F; Santini, D; Schiavon, G; Tonini, G; Vincenzi, B, 2008
)
0.35
" Main adverse drug reactions of severity grade 3 or 4 were neutrophil count decreased (20."( Efficacy and safety of two doses of pemetrexed supplemented with folic acid and vitamin B12 in previously treated patients with non-small cell lung cancer.
Adachi, S; Fujimoto, T; Fukuoka, M; Ichinose, Y; Kubota, K; Nakagawa, K; Nambu, Y; Nishiwaki, Y; Ohe, Y; Saijo, N; Tamura, T; Yamamoto, N, 2008
)
0.35
"Addition of bevacizumab to various chemotherapy agents or erlotinib in patients with NSCLC and treated brain metastases seems to be safe and is associated with a low incidence of CNS hemorrhage."( Safety of bevacizumab in patients with non-small-cell lung cancer and brain metastases.
Akerley, W; Compton, P; Huang, JE; Kolb, MM; Langer, CJ; Socinski, MA; Wang, L, 2009
)
0.35
" Dermatitis acneiform, diarrhea, nausea, and vomiting were the most frequently reported adverse events with AZD6244, compared with fatigue, anemia, nausea, anorexia, and dermatitis acneiform with pemetrexed."( A phase II, open-label, randomized study to assess the efficacy and safety of AZD6244 (ARRY-142886) versus pemetrexed in patients with non-small cell lung cancer who have failed one or two prior chemotherapeutic regimens.
Cebotaru, CL; Ciuleanu, TE; Damyanov, D; Ganchev, H; Hainsworth, JD; Kanarev, V; Morris, C; Pover, G; Stella, P; Tzekova, V, 2010
)
0.36
" At each cycle, adverse events were assessed, and concomitant medications, transfusions, and hospitalizations were recorded."( Safety and resource utilization by non-small cell lung cancer histology: results from the randomized phase III study of pemetrexed plus cisplatin versus gemcitabine plus cisplatin in chemonaïve patients with advanced non-small cell lung cancer.
Douillard, JY; Eckardt, J; Liepa, AM; Novello, S; O'Brien, M; Paz-Ares, L; Pimentel, FL; Simms, L; Visseren-Grul, C; von Pawel, J, 2010
)
0.36
" The most frequent AEs were gastrointestinal adverse events, skin rash, fever and fatigue."( [The efficacy and safety of pemetrexed as monotherapy for Chinese patients with advanced non-small cell lung cancer].
Li, LY; Wang, MZ; Zhang, L; Zhang, XT; Zhao, J; Zhong, W, 2010
)
0.36
"The no observed adverse effect level of intrathecal administration of pemetrexed was 1 mg/kg in rats."( Safety and pharmacokinetics of intrathecal administration of pemetrexed in rats.
Ahn, JS; Ahn, MJ; Chung, JY; Im, B; Lee, SY; Nam, MH; Park, K; Suh, YL; Sun, JM, 2011
)
0.37
" Safety and tolerability were evaluated by recording the incidence of adverse events (AE) according to Common Toxicity Criteria for AE (CTCAE)."( A phase II, randomized, multicenter study to assess the efficacy, safety, and tolerability of zibotentan (ZD4054) in combination with pemetrexed in patients with advanced non-small cell lung cancer.
Chouaid, C; Morris, T; Nathan, F; Pemberton, K, 2011
)
0.37
"Objective of this indirect economic comparison was to estimate and compare management costs of grade 3/4 adverse events (AEs) reported for first-line erlotinib or pemetrexed maintenance therapy in patients with advanced non-small cell lung cancer (NSCLC)."( Comparison of treatment costs of grade 3/4 adverse events associated with erlotinib or pemetrexed maintenance therapy for patients with advanced non-small-cell lung cancer (NSCLC) in Germany, France, Italy, and Spain.
Banz, K; Bischoff, H; Brunner, M; Chouaid, C; de Castro Carpeño, J; de Marinis, F; Grossi, F; Vergnenègre, A; Walzer, S, 2011
)
0.37
" Taken together, the data suggest that premexetred and sorafenib act synergistically to enhance tumor killing via the promotion of a toxic form of autophagy that leads to activation of the intrinsic apoptosis pathway, and predict that combination treatment represents a future therapeutic option in the treatment of solid tumors."( Sorafenib enhances pemetrexed cytotoxicity through an autophagy-dependent mechanism in cancer cells.
Bareford, MD; Burow, ME; Cruickshanks, N; Dent, P; Eulitt, P; Fisher, PB; Grant, S; Hamed, HA; Hubbard, N; Moran, RG; Nephew, KP; Park, MA; Tang, Y; Tye, G; Yacoub, A, 2011
)
0.37
" Taken together, the data suggest that premexetred and sorafenib act synergistically to enhance tumor killing via the promotion of a toxic form of autophagy that leads to activation of the intrinsic apoptosis pathway, and predict that combination treatment represents a future therapeutic option in the treatment of solid tumors."( Sorafenib enhances pemetrexed cytotoxicity through an autophagy-dependent mechanism in cancer cells.
Bareford, MD; Burow, ME; Cruickshanks, N; Dent, P; Fisher, PB; Grant, S; Hamed, HA; Moran, RG; Nephew, KP; Tang, Y, 2011
)
0.37
" Pemetrexed was reasonably well tolerated with few severe adverse events reported."( Efficacy and safety of pemetrexed maintenance therapy versus best supportive care in patients from East Asia with advanced, nonsquamous non-small cell lung cancer: an exploratory subgroup analysis of a global, randomized, phase 3 clinical trial.
Belani, CP; Chen, YM; Kim, JH; Orlando, M; Peterson, P; Wu, YL; Yang, SH; Zhang, L, 2012
)
0.38
" Rash (98/196 [50%] in the erlotinib group vs 10/213 [5%] in the chemotherapy group for all grades; nine [5%] vs none for grade 3 or 4) and diarrhoea (36 [18%] vs four [2%] for all grades; five [3%] vs none for grade 3 or 4) were the most common treatment-related adverse events with erlotinib, whereas alopecia (none vs 23 [11%] for all grades; none vs one [<1%] for grade 3/4) was the most common treatment-related adverse event with chemotherapy."( Efficacy and safety of erlotinib versus chemotherapy in second-line treatment of patients with advanced, non-small-cell lung cancer with poor prognosis (TITAN): a randomised multicentre, open-label, phase 3 study.
Cicenas, S; Ciuleanu, T; Gonzalez, EE; Grigorescu, AC; Hillenbach, C; Johannsdottir, HK; Klughammer, B; Miliauskas, S; Stelmakh, L, 2012
)
0.38
" The most common adverse reactions (incidence ≥ 20%) with single-agent use are fatigue, nausea, and anorexia."( Pemetrexed-induced cellulitis: a rare toxicity in non-small cell lung cancer treatment.
Katsenos, S; Panagou, C; Psara, A, 2013
)
0.39
" However, it has been reported to have adverse interactions with nonsteroid anti-inflammatory drugs(NSAIDs)."( [Adverse events during pemetrexed administration caused by concomitant nonsteroid anti-inflammatory therapy].
Hiraki, K; Inata, J; Iwamoto, Y; Kanehara, M; Kitaguchi, S; Matsumoto, S; Miyamori, S; Nakashima, K; Sakamoto, S; Sakata, Y, 2012
)
0.38
"Due to the various inter-individual differences in the biological characteristics of tumor cells, as well as issues on the efficacy, adverse reactions, and defects of existing drugs, we compared the clinical efficacy and toxicity of pemetrexed and gemcitabine combined with cisplatin for the treatment of previously untreated advanced non-small cell lung cancer (NSCLC)."( [Efficacy and toxicity of pemetrexed or gemcitabine combined with cisplatin in the treatment of patients with advanced non-small cell lung cancer].
Cheng, G; Hu, C; Hu, X; Huang, C; Jiao, S; Li, K; Luo, R; Lv, W; Ouyang, X; Sun, Y; Wang, J; Wang, M; Wang, Y; Wang, Z; Zhang, S; Zheng, R, 2012
)
0.38
" The rate of adverse reactions, including white blood cell reduction, lower platelet count, lower hemoglobin, and hair loss in the PP group was significantly lower than that in the GP group."( [Efficacy and toxicity of pemetrexed or gemcitabine combined with cisplatin in the treatment of patients with advanced non-small cell lung cancer].
Cheng, G; Hu, C; Hu, X; Huang, C; Jiao, S; Li, K; Luo, R; Lv, W; Ouyang, X; Sun, Y; Wang, J; Wang, M; Wang, Y; Wang, Z; Zhang, S; Zheng, R, 2012
)
0.38
"The clinical efficacy of pemetrexed and gemcitabine combined with cisplatin for the treatment of previously untreated advanced NSCLC was roughly the same, but the adverse reactions decreased significantly in the PP group compared with those in the GP group."( [Efficacy and toxicity of pemetrexed or gemcitabine combined with cisplatin in the treatment of patients with advanced non-small cell lung cancer].
Cheng, G; Hu, C; Hu, X; Huang, C; Jiao, S; Li, K; Luo, R; Lv, W; Ouyang, X; Sun, Y; Wang, J; Wang, M; Wang, Y; Wang, Z; Zhang, S; Zheng, R, 2012
)
0.38
"Rash is a common side effect of pemetrexed(PEM)."( [Effectiveness of steroids for the rash side effect of pemetrexed].
Ishikawa, H; Kobayashi, R; Ohashi, Y; Onishi, T; Shino, M; Suzuki, K; Yamamoto, N, 2013
)
0.39
" In this study, we aimed to determine the risk factors for severe adverse events associated with pemetrexed administration."( Analysis of risk factors for severe adverse events of chemotherapy with pemetrexed and comparison of adverse event occurrence according to renal function.
Abe, K; Hiraki, K; Inata, J; Iwamoto, Y; Kanehara, M; Miyamori, S; Sakata, Y, 2013
)
0.39
"To assess whether adjuvant chemotherapy alters pulmonary function and impacts on treatment-related adverse events."( Impact and safety of adjuvant chemotherapy on pulmonary function in early stage non-small cell lung cancer.
Eberhardt, W; Fischer, JR; Griesinger, F; Herth, FJ; Kreuter, M; Reinmuth, N; Thomas, M; Vansteenkiste, J; Zuna, I, 2014
)
0.4
" The most frequent adverse reactions were: hematological, gastrointestinal and neurological."( Systematic review of efficacy and safety of pemetrexed in non-small-cell-lung cancer.
Bautista-Paloma, FJ; Cotrina-Luque, J; Flores-Moreno, S; Galván-Banqueri, M; Pérez-Moreno, MA; Villalba-Moreno, A, 2014
)
0.4
" Multivariate analysis demonstrated that a heavy smoking history (40 or more pack-year smoking history) was an independent adverse prognostic factor for OS."( Safety and efficacy of gemcitabine or pemetrexed in combination with a platinum in patients with non-small-cell lung cancer and prior interstitial lung disease.
Ahn, JS; Ahn, MJ; Chang, W; Choi, MK; Chung, MP; Hong, JY; Jung, HA; Kim, M; Kim, S; Lee, SJ; Park, K; Park, S; Sun, JM, 2014
)
0.4
" The incidence rate of adverse drug reactions(ADR)was 76."( [Safety and effectiveness of pemetrexed in patients with non-small cell lung cancer in Japan - analysis of post-marketing surveillance].
Enatsu, S; Kaneko, N; Kobayashi, N; Nishiuma, S; Okubo, S; Taketsuna, M, 2014
)
0.4
" Patients who received pemetrexed maintenance (median 4 cycles, range 1-44) following 4 cycles of pemetrexed-cisplatin exhibited a higher incidence of drug-related serious adverse events compared with JMDB patients (median 6 cycles of pemetrexed-cisplatin) (10."( Efficacy and safety of maintenance pemetrexed in patients with advanced nonsquamous non-small cell lung cancer following pemetrexed plus cisplatin induction treatment: A cross-trial comparison of two phase III trials.
Chouaki, N; de Marinis, F; Dediu, M; Gridelli, C; John, W; Manegold, C; Paz-Ares, LG; Peterson, PM; Pujol, JL; San Antonio, B; Scagliotti, GV; Thomas, M; Visseren-Grul, C, 2014
)
0.4
" Adverse events (by maximum Common Terminology Criteria for Adverse Events [CTCAE] grade) and QoL (EuroQol 5-dimensional [EQ-5D] scale) were assessed."( Long-term and low-grade safety results of a phase III study (PARAMOUNT): maintenance pemetrexed plus best supportive care versus placebo plus best supportive care immediately after induction treatment with pemetrexed plus cisplatin for advanced nonsquamou
Bidoli, P; Chouaki, N; Corral, J; de Marinis, F; Dediu, M; Gridelli, C; John, W; Paz-Ares, L; Pujol, JL; San Antonio, B; Thomas, M; Visseren-Grul, C; Zimmermann, A, 2014
)
0.4
" Herein, we retrospectively investigated the background characteristics of patients with a creatinine clearance rate (Ccr) of<45 mL/min, who experienced severe adverse events due to pemetrexed."( [Assessment of risk factors for adverse events due to pemetrexed in patients with reduced renal function].
Abe, K; Funaki, M; Hata, Y; Hiraki, K; Inata, J; Iwamoto, Y; Kanehara, M; Kono, M; Miura, S; Miyamori, S; Sakata, Y; Sugawara, T, 2014
)
0.4
" The main side effect of pemetrexed is myelosuppression, which may be prevented by folinic acid supplementation."( [Pemetrexed nephrotoxicity].
Izzedine, H, 2015
)
0.42
" Therefore, the adverse events were mostly mild."( [Efficacy and safety of cisplatin plus pemetrexed as a first-line treatment for Japanese patients with advanced non-squamous non-small-cell lung cancer -- a retrospective analysis].
Hayashi, M; Hoshino, T; Imaizumi, K; Isogai, S; Kato, A; Mieno, Y; Minezawa, T; Morikawa, S; Morishita, M; Nakanishi, T; Niwa, Y; Okamura, T; Okazawa, M; Takeyama, T; Uozu, S; Yamaguchi, T, 2015
)
0.42
" These results suggest that encapsulation of PMX within "fluid" liposomes might represent a novel strategy to enhance the therapeutic efficacy of PMX while minimizing the side effect encountered by the non selective delivery of free PMX to various body tissues."( Liposomal pemetrexed: formulation, characterization and in vitro cytotoxicity studies for effective management of malignant pleural mesothelioma.
Elnahas, HM; Essam Eldin, N; Ishida, T; Mahdy, MA, 2015
)
0.42
" We analyzed adverse effects, such as myelosuppression, rash, and diarrhea, after 1 cycle of pemetrexed therapy."( [Safety of pemetrexed according to the duration of vitamin B12 and folic acid supplementation prior to the first dose of pemetrexed].
Chohnabayashi, N; Gotoh, K; Jinta, T; Kitamura, A; Koyama, K; Nishimura, N; Ohde, S; Okafuji, K; Takayama, S; Tomishima, Y; Tsuda, Y; Yagi, N, 2015
)
0.42
" Five patients experienced at least 1 treatment-related Grade 3 adverse event."( Preliminary Safety, Pharmacokinetics, and Efficacy of Regorafenib, Cisplatin, and Pemetrexed in Patients With Advanced Nonsquamous Non-Small-Cell Lung Cancers.
Diefenbach, K; Gettinger, SN; Hellmann, MD; Lettieri, J; Rizvi, NA; Sturm, I; Trnkova, ZJ, 2015
)
0.42
"Grade 3 or 4 neoadjuvant-related adverse events included fatigue (3 patients), neutropenia (3 patients), hypertension (1 patient)."( [Safety of Neoadjuvant Bevacizumab plus Pemetrexed and Carboplatin 
in Patients with IIIa Lung Adenocarcinoma].
Ou, W; Xu, P; Yuan, C; Zhang, S, 2015
)
0.42
"The treatment modality of neoadjuvant Bev-PC appears to be safe and tolerant in patients with stage IIIa lung adenocarcinoma."( [Safety of Neoadjuvant Bevacizumab plus Pemetrexed and Carboplatin 
in Patients with IIIa Lung Adenocarcinoma].
Ou, W; Xu, P; Yuan, C; Zhang, S, 2015
)
0.42
"Erlotinib appears to be a useful second-line option in PS0/1 patients with EGFR mutation-negative advanced non-squamous NSCLC given its mild adverse effects."( Retrospective efficacy and safety analyses of erlotinib, pemetrexed, and docetaxel in EGFR-mutation-negative patients with previously treated advanced non-squamous non-small-cell lung cancer.
Fujita, S; Hata, A; Iwasaku, M; Katakami, N; Korogi, Y; Mori, M; Morita, S; Namba, Y; Nishino, K; Nishiyama, A; Okuyama, T; Otsuka, K; Takeshita, J; Uchida, J; Yoshioka, H, 2015
)
0.42
" The two groups were compared in terms of median survival and adverse events to chemotherapy."( The effectiveness and safety of platinum-based pemetrexed and platinum-based gemcitabine treatment in patients with malignant pleural mesothelioma.
Ak, G; Akarsu, M; Metintas, M; Metintas, S, 2015
)
0.42
"The study indicates that platinum-based gemcitabine is effective and a safe schema in malignant pleural mesothelioma."( The effectiveness and safety of platinum-based pemetrexed and platinum-based gemcitabine treatment in patients with malignant pleural mesothelioma.
Ak, G; Akarsu, M; Metintas, M; Metintas, S, 2015
)
0.42
" Grade 3-4 adverse events included leukopenia (2/39), and neutropenia (3/39) in first-line therapy versus neutropenia (1/40) and thrombocytopenia (2/40) in second-line treatment."( A retrospective analysis of efficacy and safety of adding bevacizumab to chemotherapy as first- and second-line therapy in advanced non-small-cell lung cancer (NSCLC).
Chen, N; Fang, W; Hu, Z; Huang, J; Quan, R; Zhan, J; Zhang, H; Zhang, L; Zhou, T, 2016
)
0.43
" Our results would aid clinicians to make decisions of coadministration drugs to avoid drug interaction-induced side effects for achievement of safe and appropriate chemotherapy with pemetrexed."( Lansoprazole Exacerbates Pemetrexed-Mediated Hematologic Toxicity by Competitive Inhibition of Renal Basolateral Human Organic Anion Transporter 3.
Enokiya, T; Fujimoto, H; Hamada, Y; Ikemura, K; Iwamoto, T; Kaya, C; Kobayashi, T; Muraki, Y; Nakahara, H; Okuda, M, 2016
)
0.43
" Four important transporter genes are expressed in the kidney, including organic cation transporter 2 (OCT2), multidrug and toxin extrusion 1 (MATE1), ATP-binding cassette subfamily B member 1 (ABCB1), and ATP-binding cassette subfamily C member 2 (ABCC2), and genetic polymorphisms in these genes may alter the efficacy and adverse effects of platinum drugs."( Associations of genetic polymorphisms of the transporters organic cation transporter 2 (OCT2), multidrug and toxin extrusion 1 (MATE1), and ATP-binding cassette subfamily C member 2 (ABCC2) with platinum-based chemotherapy response and toxicity in non-sma
Chen, J; Liu, JY; Liu, ZQ; Qian, CY; Wang, Y; Yin, JY; Zheng, Y; Zhou, HH, 2016
)
0.43
" Areas covered: There is clinical evidence which suggsest that, like other chemotherapeutic agents, not all HGOS patients are equally responsive to antifolates and do not have the same susceptibility to experience adverse drug-related toxicities."( Pharmacogenomics of genes involved in antifolate drug response and toxicity in osteosarcoma.
Fanelli, M; Hattinger, CM; Picci, P; Serra, M; Tavanti, E; Vella, S, 2017
)
0.46
" Select adverse events (those with a potential immunologic cause) of any grade were observed in six, four, six, and five patients in arms A, B, C, and D, respectively."( Safety and efficacy of nivolumab and standard chemotherapy drug combination in patients with advanced non-small-cell lung cancer: a four arms phase Ib study.
Fujiwara, Y; Goto, K; Horinouchi, H; Hozumi, H; Kanda, S; Kitazono, S; Kubo, E; Mizugaki, H; Nokihara, H; Shiraishi, H; Sunami, K; Tamura, T; Tanaka, A; Utsumi, H; Yamamoto, N, 2016
)
0.43
" A higher incidence of some adverse events was observed with onartuzumab versus placebo, including peripheral edema (30% vs."( Efficacy and Safety of Onartuzumab in Combination With First-Line Bevacizumab- or Pemetrexed-Based Chemotherapy Regimens in Advanced Non-Squamous Non-Small-Cell Lung Cancer.
Boyer, M; Braiteh, F; Cosgriff, T; De Braud, F; Hsu, J; Kaen, D; Kingsley, CD; Lawler, W; Lena, H; Lowe, T; Mekhail, T; Novello, S; Phan, S; Schütte, W; Wakelee, H; Zvirbule, Z, 2017
)
0.46
" We collected patients´ baseline characteristics, diagnostic and treatment data, effectiveness variables (response to treatment with pemetrexed and overallsurvival) and main adverse reactions detected."( Effectiveness and safety of pemetrexed for non-small cell lung cancer in the Andalusian Public Health System.
Bautista-Paloma, FJ; Calleja-Hernández, MÁ; Cotrina-Luque, J; Flores-Moreno, S; Galván-Banqueri, M; Pérez-Moreno, MA, 2016
)
0.43
" Main adverse reactions detected were asthenia, haematological reactions, gastrointestinal reactions and dermal o mucous toxicity."( Effectiveness and safety of pemetrexed for non-small cell lung cancer in the Andalusian Public Health System.
Bautista-Paloma, FJ; Calleja-Hernández, MÁ; Cotrina-Luque, J; Flores-Moreno, S; Galván-Banqueri, M; Pérez-Moreno, MA, 2016
)
0.43
" Patient characteristics and incidence of adverse events (AEs) were described for each cohort."( Safety profiles of non-small cell lung cancer patients treated with pemetrexed plus carboplatin: a real-world retrospective, observational, cohort study.
Chen, J; Chen, L; Goodloe, RJ; John, WJ; San Antonio, B; Yan, Y, 2017
)
0.46
"This meta-analysis compared safety profiles (selected drug-related treatment-emergent adverse events [TEAEs]) of first-line pemetrexed plus carboplatin (PCb) area under the concentration-time curve 5 mg/min•mL (PCb5) or 6 mg/min•mL (PCb6), two widely used regimens in clinical practice for advanced non-squamous non-small cell lung cancer."( Meta-analysis of pemetrexed plus carboplatin doublet safety profile in first-line non-squamous non-small cell lung cancer studies.
Chen, J; John, WJ; Liu, J; Okamoto, I; Rodrigues-Pereira, J; San Antonio, B; Schuette, WH; Stinchcombe, TE; Zinner, RG, 2017
)
0.46
"In a phase III study, maintenance pemetrexed showed superior survival over placebo (PARAMOUNT) for patients with advanced non-squamous non-small cell lung cancer (NSCLC) who completed 4 cycles of pemetrexed plus cisplatin (PC) induction therapy, with low incidence of treatment-emergent adverse events (TEAEs) generally associated with pemetrexed."( Safety Analyses of Pemetrexed-cisplatin and Pemetrexed Maintenance Therapies in Patients With Advanced Non-squamous NSCLC: Retrospective Analyses From 2 Phase III Studies.
Chen, J; de Marinis, F; Gridelli, C; Langer, CJ; Liu, J; Oton, AB; Paz-Ares, LG; Pujol, JL; San Antonio, B; Scagliotti, GV; Visseren-Grul, C; Wozniak, AJ, 2017
)
0.46
"All 66 treated patients experienced at least one adverse event (AE)."( A phase IB dose-escalation study of the safety and pharmacokinetics of pictilisib in combination with either paclitaxel and carboplatin (with or without bevacizumab) or pemetrexed and cisplatin (with or without bevacizumab) in patients with advanced non-s
Adjei, AA; Bahleda, R; Besse, B; Dy, GK; Ferte, C; Groen, HJM; Lin, W; Morrissey, K; Planchard, D; Schutzman, JL; Shankar, G; Soria, JC; Ware, J; Zhou, J, 2017
)
0.46
" Pemetrexed is generally well tolerated, but individual-patient differences exist in severity of adverse events."( Severity and predictive factors of adverse events in pemetrexed-containing chemotherapy for non-small cell lung cancer.
Abe, T; Iwanaga, K; Kimura, S; Komiya, K; Matsunaga, H; Miyahara, T; Nakamura, T; Nakashima, C; Sueoka-Aragane, N; Ureshino, N, 2017
)
0.46
" However, polymorphisms of MTHFR C677T were not significantly associated with other adverse events and clinical outcomes."( Correlation Between Methylenetetrahydrofolate Reductase (MTHFR) C677T Polymorphisms and Pemetrexed Chemotherapy Efficacy/Toxicity in Non-Squamous Non-Small Cell Lung Cancer.
Chen, L; Chen, X; Lan, G; Lin, L, 2017
)
0.46
" Bevacizumab was administered at a dose of 100 mg intravenously once a week combined with one or two types of chemotherapeutic drugs until confirmed disease progression or an intolerable adverse event was observed."( Analysis of the activity and safety of weekly low-dose bevacizumab-based regimens in heavily pretreated patients with metastatic breast cancer.
Chen, J; Fan, Y; Hong, R; Ma, F; Ou, K; Sang, D; Wang, J; Xu, B; Yuan, P; Zhai, X; Zhao, C, 2018
)
0.48
" The most common hematological adverse events were neutropenia, anemia, and thrombocytopenia."( Analysis of the activity and safety of weekly low-dose bevacizumab-based regimens in heavily pretreated patients with metastatic breast cancer.
Chen, J; Fan, Y; Hong, R; Ma, F; Ou, K; Sang, D; Wang, J; Xu, B; Yuan, P; Zhai, X; Zhao, C, 2018
)
0.48
"Recombinant human endostatin (rh-endostatin) plus standard chemotherapy in advanced non-small cell lung cancer (NSCLC) patients has shown improved efficacy; however, it is unclear whether it is effective and safe when added to pemetrexed/cisplatin and used as maintenance therapy."( Efficacy and safety of rh-endostatin (Endostar) combined with pemetrexed/cisplatin followed by rh-endostatin plus pemetrexed maintenance in non-small cell lung cancer: A retrospective comparison with standard chemotherapy.
He, X; Jin, J; Pi, J; Shi, Y; Zhou, S; Zuo, L, 2018
)
0.48
"032) or in the incidence of drug-related or grade 3-4 adverse events."( Efficacy and safety of rh-endostatin (Endostar) combined with pemetrexed/cisplatin followed by rh-endostatin plus pemetrexed maintenance in non-small cell lung cancer: A retrospective comparison with standard chemotherapy.
He, X; Jin, J; Pi, J; Shi, Y; Zhou, S; Zuo, L, 2018
)
0.48
" Some studies have suggested that concomitant drugs may be the risk factors for severe adverse events."( Effects of proton pump inhibitors on severe haematotoxicity induced after first course of pemetrexed/carboplatin combination chemotherapy.
Araki, R; Hamamoto, T; Keira, T; Masuda, Y; Tanaka, T; Yamada, H, 2019
)
0.51
" Adverse events were recorded."( Second-line erlotinib after failure of pemetrexed-containing chemotherapy in advanced non-small cell lung cancer (NSCLC): Real-world effectiveness, safety and tolerability.
Germonpré, P; Van den Wyngaert, T, 2019
)
0.51
"9%) of patients due to adverse events, which were treatment related in 7%."( Second-line erlotinib after failure of pemetrexed-containing chemotherapy in advanced non-small cell lung cancer (NSCLC): Real-world effectiveness, safety and tolerability.
Germonpré, P; Van den Wyngaert, T, 2019
)
0.51
" No unexpected severe adverse events or treatment-related deaths occurred."( Five-year safety and efficacy data from a phase Ib study of nivolumab and chemotherapy in advanced non-small-cell lung cancer.
Fujiwara, Y; Goto, Y; Horinouchi, H; Kanda, S; Nokihara, H; Ohe, Y; Tamura, T; Yamamoto, N; Yamamoto, T, 2020
)
0.56
" This review aims to describe the underlying mechanisms of the renal adverse effects caused by chemotherapy and ICI therapy, leading to a suggested diagnostic and treatment algorithm on the basis of clinical, laboratory, radiographic, and pathologic parameters."( Renal Toxicity From Pemetrexed and Pembrolizumab in the Era of Combination Therapy in Patients With Metastatic Nonsquamous Cell NSCLC.
Aerts, JGJV; Cornelissen, R; Dumoulin, DW; van Gelder, T; Vansteenkiste, J; Visser, S; von der Thusen, J, 2020
)
0.56
" As it is primarily eliminated by renal excretion, adequate renal function is essential to prevent toxic exposure."( Cumulative pemetrexed dose increases the risk of nephrotoxicity.
Biesma, B; Boosman, RJ; Burger, DM; de Rouw, N; Derijks, HJ; Hilbrands, LB; Ter Heine, R; van de Bruinhorst, H; van den Heuvel, MM, 2020
)
0.56
"Search the online electronic databases on comparison the effectiveness and adverse effects of pemetrexed versus gefitinib in therapy outcomes of pre-treated NSCLC to September 2019."( A meta-analysis of the safety and effectiveness of pemetrexed compared with gefitinib for pre-treated advanced or metastatic NSCLC.
Chen, W; Li, F; Li, S; Li, Y; Lu, X; Zheng, D, 2020
)
0.56
" The incidence of grade 3 or higher adverse events was 61."( Efficacy and Safety of Sintilimab Plus Pemetrexed and Platinum as First-Line Treatment for Locally Advanced or Metastatic Nonsquamous NSCLC: a Randomized, Double-Blind, Phase 3 Study (Oncology pRogram by InnovENT anti-PD-1-11).
Bi, M; Cang, S; Chen, G; Cheng, Y; Fang, J; Feng, J; Han, B; He, Z; Li, B; Li, J; Li, W; Lu, Y; Ma, R; Mei, X; Ren, X; Song, Y; Wang, L; Wang, S; Wang, Y; Wang, Z; Xie, C; Yang, N; Yang, Y; Yang, Z; Yu, Q; Yu, Z; Zhang, L; Zhang, W; Zhang, Y; Zhao, Y; Zhou, H; Zhou, J; Zhou, R; Zhu, D, 2020
)
0.56
" Bevacizumab plus cisplatin/pemetrexed has been shown to be safe and effective in non-small cell lung cancer, however, there are no efficacy or safety data in Japanese patients with MPM treated with this regimen."( Bevacizumab plus cisplatin/pemetrexed then bevacizumab alone for unresectable malignant pleural mesothelioma: A Japanese safety study.
Hayashi, M; Hirabayashi, M; Hirano, K; Kondo, M; Kuribayashi, K; Morise, M; Nakano, T; Tada, Y; Tanaka, M, 2021
)
0.62
"Eligible patients (n = 7) received bevacizumab plus cisplatin/pemetrexed (up to six cycles), then single-agent bevacizumab until disease progression or onset of unacceptable adverse events (AEs), according to the 3+3 design analogy."( Bevacizumab plus cisplatin/pemetrexed then bevacizumab alone for unresectable malignant pleural mesothelioma: A Japanese safety study.
Hayashi, M; Hirabayashi, M; Hirano, K; Kondo, M; Kuribayashi, K; Morise, M; Nakano, T; Tada, Y; Tanaka, M, 2021
)
0.62
" For patients with evacuated third-space fluid, these adverse effects were nullified."( Haematological toxicity of pemetrexed in patients with metastatic non-squamous non-small cell carcinoma of lung with third-space fluid.
Cheong, TF; Chiang, KY; Ho, JCM; Ip, MSM; Kwok, WC; Lam, DCL; Tam, TCC, 2021
)
0.62
" We present two cases of RION that happened in seemingly safe radiation doses."( Necrosis of the optic nerve and chiasm with safe radiation doses: Report of two rare cases.
Abri Aghdam, K; Aghajani, A; Khosravi Farsani, M; Moghadasi, M; Soltan Sanjari, M, 2022
)
0.72
"The primary endpoint was the 1-year survival rate, with secondary endpoints of response rate (RR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and adverse event rate."( Efficacy and safety of carboplatin and pemetrexed followed by maintenance with pemetrexed for elderly patients with advanced non-squamous non-small cell lung cancer: A single-arm, open-label, multicenter, phase II study.
Hara, Y; Kaneko, T; Kobayashi, N; Manabe, S; Miyazawa, N; Murakami, S; Nishikawa, M; Saito, H; Shinkai, M; Shinoda, M; Tomaru, K, 2021
)
0.62
"This post hoc analysis assessed the safety of pemetrexed and platinum in combination with pembrolizumab, including time-to-onset and time-to-resolution of all-cause any-grade and grade ≥3 adverse events (AEs) and renal AEs."( Safety of pemetrexed plus platinum in combination with pembrolizumab for metastatic nonsquamous non-small cell lung cancer: A post hoc analysis of KEYNOTE-189.
Aerts, J; Gadgeel, SM; Garon, EB; Kim, JS; Muehlenbein, CE; Peterson, P; Rizzo, MT, 2021
)
0.62
" These results support the safe use of the KEYNOTE-189 regimen in clinical practice."( Safety of pemetrexed plus platinum in combination with pembrolizumab for metastatic nonsquamous non-small cell lung cancer: A post hoc analysis of KEYNOTE-189.
Aerts, J; Gadgeel, SM; Garon, EB; Kim, JS; Muehlenbein, CE; Peterson, P; Rizzo, MT, 2021
)
0.62
"A monocentric comparative before/after study was carried out in an oncology day hospital to evaluate the efficacy of Safe Infusion Devices in reducing drug exposure compared to usual infusion practices."( Evaluation of a safe infusion device on reducing occupational exposure of nurses to antineoplastic drugs: a comparative prospective study. Contamoins-1.
Blanc, E; Bouleftour, W; Forges, F; Guitton, J; Hugues, M; Macé, A; Macron, C; Menguy, S; Raymond, B; Simoëns, X; Tinquaut, F, 2021
)
0.62
"3% of contaminated samples while Safe Infusion Devices to a rate of 15%: Safe Infusion Devices reduced the risk of gloves contamination by 85% in multivariate analysis (Odds ratio = 0."( Evaluation of a safe infusion device on reducing occupational exposure of nurses to antineoplastic drugs: a comparative prospective study. Contamoins-1.
Blanc, E; Bouleftour, W; Forges, F; Guitton, J; Hugues, M; Macé, A; Macron, C; Menguy, S; Raymond, B; Simoëns, X; Tinquaut, F, 2021
)
0.62
"Despite the current practice of using neutral solvent-purged infusers, the occupational exposure remains high for nurses and Safe Infusion Devices significantly reduced this risk of exposure."( Evaluation of a safe infusion device on reducing occupational exposure of nurses to antineoplastic drugs: a comparative prospective study. Contamoins-1.
Blanc, E; Bouleftour, W; Forges, F; Guitton, J; Hugues, M; Macé, A; Macron, C; Menguy, S; Raymond, B; Simoëns, X; Tinquaut, F, 2021
)
0.62
" Treatment response, European Organization for Research and Treatment of Cancer QoL Questionnaire-Core 30 (EORTC QLQ-C30), progression-free survival (PFS), overall survival (OS), and adverse events were assessed during the follow-up."( Drug-eluting bead bronchial arterial chemoembolization vs. chemotherapy in treating advanced non-small cell lung cancer: comparison of treatment efficacy, safety and quality of life.
Bao, PT; Lin, H; Liu, XF; Mu, M; Pan, P; Tian, FF; Wang, Q; Zhang, R; Zhao, WG, 2021
)
0.62
" Furthermore, two groups all exhibited mild and tolerable adverse events."( Drug-eluting bead bronchial arterial chemoembolization vs. chemotherapy in treating advanced non-small cell lung cancer: comparison of treatment efficacy, safety and quality of life.
Bao, PT; Lin, H; Liu, XF; Mu, M; Pan, P; Tian, FF; Wang, Q; Zhang, R; Zhao, WG, 2021
)
0.62
" Overall survival and adverse events (AEs) were evaluated as secondary end points."( Efficacy and Safety of Intrathecal Pemetrexed Combined With Dexamethasone for Treating Tyrosine Kinase Inhibitor-Failed Leptomeningeal Metastases From EGFR-Mutant NSCLC-a Prospective, Open-Label, Single-Arm Phase 1/2 Clinical Trial (Unique Identifier: Chi
Du, Y; Fan, C; Gao, F; Gao, N; Huang, D; Jiang, Q; Jiang, Z; Jin, Y; Li, L; Lv, Y; Shen, W; Shi, T; Song, X; Teng, C; Wei, L; Xin, T; Zhao, Q; Zhao, X, 2021
)
0.62
" The endpoints were progression-free survival (PFS), overall survival (OS) and adverse events (AEs)."( The efficacy and toxicity of maintenance therapy with bevacizumab plus pemetrexed versus bevacizumab/pemetrexed alone for stage IIIB/IV nonsquamous non-small cell lung cancer: A meta-analysis of randomized controlled trials.
Fang, Z; Liu, M; Liu, Q; Luo, N; Wei, Y; Yi, F; Zhang, W; Zhang, X, 2022
)
0.72
" Regarding adverse events (AEs), the Bev+Pem group exhibited an increased occurrence of anaemia, fatigue, thrombocytopenia and anorexia."( The efficacy and toxicity of maintenance therapy with bevacizumab plus pemetrexed versus bevacizumab/pemetrexed alone for stage IIIB/IV nonsquamous non-small cell lung cancer: A meta-analysis of randomized controlled trials.
Fang, Z; Liu, M; Liu, Q; Luo, N; Wei, Y; Yi, F; Zhang, W; Zhang, X, 2022
)
0.72
" OS and treatment-emergent adverse events (TEAEs) were analyzed in both groups by treatment duration."( Overall Survival and Safety With Pemetrexed/Platinum ± Anti-VEGF Followed by Pemetrexed ± Anti-VEGF Maintenance in Advanced Nonsquamous Non-Small-Cell Lung Cancer: A Pooled Analysis of 4 Randomized Studies.
Garon, EB; Kim, JS; Peterson, P; Rizzo, MT, 2022
)
0.72
" Safety was assessed by adverse events (AEs), treatment-related adverse events (TRAEs), serious adverse events (SAEs), and adverse events of special interest (AESIs)."( Safety and efficacy of dendritic cell-based immunotherapy (DCVAC/LuCa) combined with carboplatin/pemetrexed for patients with advanced non-squamous non-small-cell lung cancer without oncogenic drivers.
Cao, S; Han, B; Ling, X; Wang, H; Xu, J; Zhang, B; Zhang, X; Zhong, H; Zhong, R, 2022
)
0.72
" In consideration of safety, the most frequent adverse events were peripheral neuropathy (37."( Efficacy, safety and prognostic factors of camrelizumab plus carboplatin and pemetrexed chemotherapy in advanced lung adenocarcinoma patients.
Bao, Q; Chen, Y; Deng, R; Fu, Y; He, Y; Huang, M; Li, C; Liu, M; Liu, Y; Lv, J; Lv, Z; Miao, Y; Wang, F; Wang, L; Wang, Q; Zhang, C; Zhang, T; Zhang, W; Zhang, Z; Zhao, X; Zhou, H, 2022
)
0.72
"Camrelizumab plus CP chemotherapy achieves favourable efficacy and tolerable adverse events in advanced lung adenocarcinoma patients."( Efficacy, safety and prognostic factors of camrelizumab plus carboplatin and pemetrexed chemotherapy in advanced lung adenocarcinoma patients.
Bao, Q; Chen, Y; Deng, R; Fu, Y; He, Y; Huang, M; Li, C; Liu, M; Liu, Y; Lv, J; Lv, Z; Miao, Y; Wang, F; Wang, L; Wang, Q; Zhang, C; Zhang, T; Zhang, W; Zhang, Z; Zhao, X; Zhou, H, 2022
)
0.72
" However, its development is still restricted by low targeting, high dose and toxic side effects."( A multimodal Metal-Organic framework based on unsaturated metal site for enhancing antitumor cytotoxicity through Chemo-Photodynamic therapy.
Ding, Q; Li, B; Li, W; Liu, J; Muddassir, M; Ouyang, Q; Rao, C; Sakiyama, H; Xu, Z; Zhou, L, 2022
)
0.72
" Moreover, the most common adverse events related to camrelizumab plus CP were fatigue (45."( Camrelizumab plus chemotherapy in advanced non-squamous non-small cell lung cancer: Treatment response, survival pattern, and safety.
Cao, H; Lu, Y; Wei, W, 2022
)
0.72
"Camrelizumab plus chemotherapy exhibits good efficacy and manageable adverse events in treating advanced non-squamous NSCLC patients."( Camrelizumab plus chemotherapy in advanced non-squamous non-small cell lung cancer: Treatment response, survival pattern, and safety.
Cao, H; Lu, Y; Wei, W, 2022
)
0.72
" Among the various adverse events, renal toxicity can be a relevant safety issue."( Patterns of renal toxicity from the combination of pemetrexed and pembrolizumab for advanced nonsquamous non-small-cell lung cancer (NSCLC): A single-center experience.
Aprile, M; Ardizzoni, A; Borelli, G; Busutti, M; Campus, A; Croci Chiocchini, AL; De Giglio, A; Di Federico, A; Gelsomino, F; Grandinetti, V; La Manna, G; Melotti, B; Sperandi, F; Vischini, G, 2022
)
0.72
"Renal toxicity represents a challenging adverse event that could negatively impact outcomes of metastatic nonsquamous NSCLC patients receiving CT/ICI demanding a multidisciplinary approach."( Patterns of renal toxicity from the combination of pemetrexed and pembrolizumab for advanced nonsquamous non-small-cell lung cancer (NSCLC): A single-center experience.
Aprile, M; Ardizzoni, A; Borelli, G; Busutti, M; Campus, A; Croci Chiocchini, AL; De Giglio, A; Di Federico, A; Gelsomino, F; Grandinetti, V; La Manna, G; Melotti, B; Sperandi, F; Vischini, G, 2022
)
0.72
"6%) had a grade 3 or 4 treatment-related adverse event (AE), and one (1."( Final results of DIADEM, a phase II study to investigate the efficacy and safety of durvalumab in advanced pretreated malignant pleural mesothelioma.
Belletti, M; Bono, F; Bonomi, M; Canova, S; Carlucci, L; Ceresoli, GL; Cortinovis, DL; D'Aveni, A; De Angelis, A; De Simone, I; Galli, F; Gelsomino, F; Grosso, F; Mencoboni, M; Pasello, G; Perrino, M; Rulli, E; Zucali, PA, 2022
)
0.72
" Patients who experienced toxicity of grade 3 or 4 according to the Common Terminology Criteria for Adverse Events v5."( Predictive factors associated with pemetrexed acute toxicity.
Arrondeau, J; Batista, R; Blanchet, B; Bonnet, M; Boudou-Rouquette, P; Goldwasser, F; Jouinot, A; Seif, V; Thomas-Schoemann, A; Vidal, M; Villeminey, C; Wislez, M, 2023
)
0.91

Pharmacokinetics

The objectives of these population pharmacokinetic analyses were to assess the overall disposition of pemetrexed. The study showed a significant inverse correlation between the AUC and the creatinine clearance.

ExcerptReferenceRelevance
" infusion over 10 minutes for 5 days, repeated every 3 weeks, was conducted to evaluate the maximum tolerated dose, pharmacokinetic profile, and antitumor activity of the drug using this schedule."( A phase I and pharmacokinetic study of LY231514, the multitargeted antifolate.
Abrahams, T; Adams, L; Bailey, NP; Calvert, AH; Cassidy, J; Kaye, SB; Lind, MJ; McCarthy, S; McDonald, AC; Siddiqui, N; Twelves, CJ; Vasey, PA; Walling, J; Woodworth, JR, 1998
)
0.3
" Plasma concentration-time data were analyzed by nonlinear mixed-effect modeling using NONMEM to estimate pemetrexed disodium pharmacokinetic parameters (mean, and between- and within-patient variability) as well as relationships between the pharmacokinetic parameters and various patient-specific factors (demographic and physiologic data)."( Population pharmacokinetics of pemetrexed disodium (ALIMTA) in patients with cancer.
Johnson, RD; Lalonde, RL; Ouellet, D; Periclou, AP; Woodworth, JR, 2000
)
0.31
" In conclusion, population pharmacokinetic modeling revealed relationships between pharmacokinetic parameters and various patient specific factors."( Population pharmacokinetics of pemetrexed disodium (ALIMTA) in patients with cancer.
Johnson, RD; Lalonde, RL; Ouellet, D; Periclou, AP; Woodworth, JR, 2000
)
0.31
" The half-life of MTA in the peritoneal fluid after intraperitoneal infusion was approximately 2 hours."( Multi-targeted antifolate (MTA): pharmacokinetics of intraperitoneal administration in a rat model.
Pestieau, SR; Stuart, OA; Sugarbaker, PH, 2000
)
0.31
"This phase I and pharmacokinetic study of pemetrexed in combination with oxaliplatin was performed to determine the maximum tolerated dose (MTD), and to evaluate safety and pharmacokinetics in patients with metastatic solid tumors."( Phase I and pharmacokinetic study of the multitargeted antifolate pemetrexed in combination with oxaliplatin in patients with advanced solid tumors.
Bozec, L; Campone, M; Delaloge, S; Fumoleau, P; Gamelin, E; Latz, JE; Misset, JL, 2004
)
0.32
" Pharmacokinetic analyses of plasma gemcitabine and pemetrexed concentrations were performed."( A phase IB study of the pharmacokinetics of gemcitabine and pemetrexed, when administered in rapid sequence to patients with advanced solid tumors.
Adjei, AA; Alberts, SR; Atherton, PJ; Burch, PA; Dy, GK; Erlichman, C; Goldberg, RM; Hanson, LJ; Pitot, HC; Reid, JM; Rubin, J; Sloan, JA; Suri, A, 2005
)
0.33
" There was no pharmacokinetic interaction between the two drugs."( A phase IB study of the pharmacokinetics of gemcitabine and pemetrexed, when administered in rapid sequence to patients with advanced solid tumors.
Adjei, AA; Alberts, SR; Atherton, PJ; Burch, PA; Dy, GK; Erlichman, C; Goldberg, RM; Hanson, LJ; Pitot, HC; Reid, JM; Rubin, J; Sloan, JA; Suri, A, 2005
)
0.33
"The objectives of these analyses were to (1) develop a semimechanistic-physiologic population pharmacokinetic/pharmacodynamic (PK/PD) model to describe neutropenic response to pemetrexed and to (2) identify influential covariates with respect to pharmacodynamic response."( A semimechanistic-physiologic population pharmacokinetic/pharmacodynamic model for neutropenia following pemetrexed therapy.
Ghosh, A; Johnson, RD; Karlsson, MO; Latz, JE; Rusthoven, JJ, 2006
)
0.33
"The objectives of these population pharmacokinetic analyses were to (1) assess the overall disposition of pemetrexed, (2) characterize between-patient and within-patient variability and identify influential covariates with respect to pemetrexed pharmacokinetics; and, (3) provide individual empirical Bayesian estimates of pharmacokinetic parameters for use in a subsequent pharmacokinetic/pharmacodynamic evaluation of neutropenia following pemetrexed administration."( Population pharmacokinetic analysis of ten phase II clinical trials of pemetrexed in cancer patients.
Chaudhary, A; Ghosh, A; Johnson, RD; Latz, JE, 2006
)
0.33
"Data from 287 patients who received 441 cycles without folic acid or vitamin B12 supplementation during participation in one of ten phase II cancer trials were evaluated by population pharmacokinetic analysis using NONMEM."( Population pharmacokinetic analysis of ten phase II clinical trials of pemetrexed in cancer patients.
Chaudhary, A; Ghosh, A; Johnson, RD; Latz, JE, 2006
)
0.33
" Since pharmacodynamic analyses have shown that AUC and not C (max) is the primary determinant of neutropenic response to pemetrexed, this suggests that dose adjustments based on renal function, rather than body surface area, might be considered for pemetrexed."( Population pharmacokinetic analysis of ten phase II clinical trials of pemetrexed in cancer patients.
Chaudhary, A; Ghosh, A; Johnson, RD; Latz, JE, 2006
)
0.33
"Pemetrexed disappearance from plasma was best described by a two compartment model with a mean distribution half-life of 13."( Plasma and cerebrospinal fluid pharmacokinetics of pemetrexed after intravenous administration in non-human primates.
Ames, MM; Blaney, SM; Dauser, R; McGovern, RM; McGuffey, L; Nuchtern, J; Reid, JM; Stapleton, SL; Thompson, PA, 2007
)
0.34
" The study also sought to detect major pharmacokinetic drug-drug interactions between these agents and preliminary evidence of antitumor activity in patients with advanced solid malignancies."( A phase I and pharmacokinetic study of pemetrexed plus irinotecan in patients with advanced solid malignancies.
Beeram, M; Chu, Q; De Bono, J; Forouzesh, B; Hammond, LA; Hong, S; John, W; Latz, JE; Nguyen, B; Rowinsky, EK; Schwartz, G, 2007
)
0.34
" No major pharmacokinetic interactions between the agents were evident."( A phase I and pharmacokinetic study of pemetrexed plus irinotecan in patients with advanced solid malignancies.
Beeram, M; Chu, Q; De Bono, J; Forouzesh, B; Hammond, LA; Hong, S; John, W; Latz, JE; Nguyen, B; Rowinsky, EK; Schwartz, G, 2007
)
0.34
"We report results of a phase I trial and pharmacokinetic study of pemetrexed (LY231514) in children and adolescents with refractory solid tumors."( Phase I trial and pharmacokinetic study of pemetrexed in children with refractory solid tumors: the Children's Oncology Group.
Adamson, PC; Ames, MM; Blaney, SM; Ingle, AM; Krailo, M; Malempati, S; McGovern, R; Melemed, AS; Nicholson, HS; Reid, JM; Safgren, S; Stork, LC, 2007
)
0.34
" Pharmacokinetic studies were performed during the first course of treatment."( Phase I trial and pharmacokinetic study of pemetrexed in children with refractory solid tumors: the Children's Oncology Group.
Adamson, PC; Ames, MM; Blaney, SM; Ingle, AM; Krailo, M; Malempati, S; McGovern, R; Melemed, AS; Nicholson, HS; Reid, JM; Safgren, S; Stork, LC, 2007
)
0.34
"The purpose of this study was to investigate the utility of plasma pharmacokinetic and pharmacodynamic measures including plasma deoxynucleosides, homocysteine and methylmalonic acid concentrations in understanding the time course and extent of the inhibition of thymidylate synthase (TS) by pemetrexed in the context of a phase I/II combination study with vinorelbine."( Pemetrexed pharmacokinetics and pharmacodynamics in a phase I/II study of doublet chemotherapy with vinorelbine: implications for further optimisation of pemetrexed schedules.
Clarke, SJ; Li, KM; Rivory, LP, 2007
)
0.34
" The pemetrexed pharmacokinetic results were consistent with those from previous single-agent pemetrexed studies and a previous study of pemetrexed in combination with cisplatin."( Pharmacokinetic evaluation of platinum derived from cisplatin administered alone and with pemetrexed in head and neck cancer patients.
Ciuleanu, T; Darstein, CL; Latz, JE; Musib, LC; Specenier, PM; Vermorken, JB, 2009
)
0.35
" As Cmax is a determinant of pemetrexed toxicity, intrapleural administration might offer a means of widening the effective therapeutic index of the drug by improving tolerability."( Intrapleural administration of pemetrexed: a pharmacokinetic study in an animal model.
Astoul, P; Bouvenot, J; Coltel, N; Devictor-Pierre, B; Fraticelli, A; Greillier, L; Lamarche, G; Monjanel-Mouterde, S, 2009
)
0.35
" Pharmacodynamic separation by intermittent delivery of epidermal growth factor receptor tyrosine kinase inhibitors with chemotherapy may increase efficacy by overcoming hypothesized antagonism."( Intermittent erlotinib in combination with pemetrexed: phase I schedules designed to achieve pharmacodynamic separation.
Beckett, L; Davies, AM; Gandara, DR; Ho, C; Lara, PN; Lau, D; Perkins, N; Scudder, SA, 2009
)
0.35
"This article focuses on all of the currently published pharmacokinetic data of pemetrexed reviewing a number of different scenarios and patient populations."( Pharmacokinetic evaluation of pemetrexed.
Sørensen, JB, 2011
)
0.37
" The pharmacokinetic study showed a significant inverse correlation between the AUC of pemetrexed and the creatinine clearance."( Dose escalation and pharmacokinetic study of carboplatin plus pemetrexed for elderly patients with advanced nonsquamous non-small-cell lung cancer: Kumamoto thoracic oncology study group trial 1002.
Akaike, K; Fujii, S; Hamada, A; Hirosako, S; Iriki, T; Kishi, H; Kohrogi, H; Nakamura, K; Notsute, D; Saeki, S; Sakata, S; Saruwatari, K; Sasaki, J; Sato, R; Tanaka, H, 2015
)
0.42
" The in vitro to in vivo extrapolation approach used in this work was developed to predict possible drug-drug interactions (DDIs) that may occur after coadministration of pemetrexed and nonsteroidal anti-inflammatory drugs (NSAIDs), and it included in vitro assays, risk assessment models, and physiologically based pharmacokinetic (PBPK) models."( Prediction of renal transporter mediated drug-drug interactions for pemetrexed using physiologically based pharmacokinetic modeling.
Bacon, JA; Hall, SD; Higgins, JW; Hillgren, KM; Kim, RB; Pak, YA; Posada, MM; Schneck, KB; Tirona, RG, 2015
)
0.42
" Regorafenib had acceptable tolerability and minor pharmacokinetic interactions in combination with standard doses of cisplatin and pemetrexed in patients with advanced nsNSCLCs."( Preliminary Safety, Pharmacokinetics, and Efficacy of Regorafenib, Cisplatin, and Pemetrexed in Patients With Advanced Nonsquamous Non-Small-Cell Lung Cancers.
Diefenbach, K; Gettinger, SN; Hellmann, MD; Lettieri, J; Rizvi, NA; Sturm, I; Trnkova, ZJ, 2015
)
0.42
"In patients with unselected or EGFR wild-type advanced nonsquamous NSCLC, pharmacodynamic separation of pemetrexed and intercalated erlotinib had promising antitumor activity without new safety concerns."( Randomized Phase 2 Trial of Pharmacodynamic Separation of Pemetrexed and Intercalated Erlotinib Versus Pemetrexed Alone for Advanced Nonsquamous, Non-small-cell Lung Cancer.
Bathini, VG; Beckett, LA; Gajavelli, S; Gandara, DR; Gucalp, R; Haigentz, M; Kim, M; Lara, PN; Li, T; Pasquinelli, PB; Perez-Soler, R; Piperdi, B; Sreedhara, M; Walsh, WV; Wen, H; Xie, X; Zhou, K, 2017
)
0.46
"The pharmacokinetic target was an area under the concentration-time curve (AUC) of 123-205 mg·h/L."( Pharmacokinetically-guided dosing of pemetrexed in a patient with renal impairment and a patient requiring hemodialysis.
Agterhuis, DE; Burger, DM; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AM; Posthuma, R; Schoenmaekers, JJAO; Ter Heine, R, 2019
)
0.51
" Serial samples of cerebrospinal fluid (CSF) and plasma were obtained for pharmacokinetic studies."( Safety, Pharmacokinetic and Clinical Activity of Intrathecal Chemotherapy With Pemetrexed via the Ommaya Reservoir for Leptomeningeal Metastases From Lung Adenocarcinoma: A Prospective Phase I Study.
Jiang, C; Li, H; Lin, Y; Liu, X; Qian, X; Xie, Y; Yin, Z; Yu, T; Zheng, S, 2023
)
0.91
" Pharmacokinetic analysis showed that using Ommaya reservoirs, higher pemetrexed concentrations and prolonged half-lives were achieved in the CSF compared with lumbar puncture (LP)."( Safety, Pharmacokinetic and Clinical Activity of Intrathecal Chemotherapy With Pemetrexed via the Ommaya Reservoir for Leptomeningeal Metastases From Lung Adenocarcinoma: A Prospective Phase I Study.
Jiang, C; Li, H; Lin, Y; Liu, X; Qian, X; Xie, Y; Yin, Z; Yu, T; Zheng, S, 2023
)
0.91

Compound-Compound Interactions

Pemetrexed combined with cisplatin for the first-line chemotherapy of patients with advanced non-small-cell lung cancer (NSCLC) and maintenance treatment.Continuous- or pulsed-dose afatinib combined with pemetrexing exhibited a manageable safety profile.

ExcerptReferenceRelevance
" This study was designed to (1) determine the maximum-tolerated dose (MTD), dose-limiting toxicities (DLT), and pharmacokinetics of MTA combined with cisplatin; (2) determine a recommended dose for phase II studies; and (3) collect anecdotal information on the antitumor activity of MTA combined with cisplatin."( Clinical and pharmacokinetic phase I study of multitargeted antifolate (LY231514) in combination with cisplatin.
Blatter, J; Depenbrock, H; Dumez, H; Hanauske, AR; Johnson, RD; Thödtmann, R; van Oosterom, A, 1999
)
0.3
" Therefore, we evaluated the anti-proliferative potential of MTA combined with drugs known to exert therapeutic activity against colon cancer, including 5-fluorouracil, oxaliplatin, and SN38, the active metabolite of irinotecan."( Pemetrexed disodium combined with oxaliplatin, SN38, or 5-fluorouracil, based on the quantitation of drug interactions in human HT29 colon cancer cells.
Coudray, AM; De Gramont, A; Faivre, S; Gespach, C; Louvet, C; Raymond, E; Tournigand, C, 2002
)
0.31
" Several phase I/II trials of pemetrexed as a single agent or in combination with a platinum drug have demonstrated considerable activity in mesothelioma."( Pemetrexed alone and in combination with platinum compounds in the management of malignant mesothelioma.
Green, MR; Meyer, ML; Suwanrusme, H, 2004
)
0.32
"This phase I and pharmacokinetic study of pemetrexed in combination with oxaliplatin was performed to determine the maximum tolerated dose (MTD), and to evaluate safety and pharmacokinetics in patients with metastatic solid tumors."( Phase I and pharmacokinetic study of the multitargeted antifolate pemetrexed in combination with oxaliplatin in patients with advanced solid tumors.
Bozec, L; Campone, M; Delaloge, S; Fumoleau, P; Gamelin, E; Latz, JE; Misset, JL, 2004
)
0.32
" In this study, we investigated the anticancer activity of BGC9331 either alone or combined with 5-fluorouracil (5-FU), MTA (multi-target antifolate), oxali-platin and SN-38, the active metabolite of the topoisomerase I inhibitor CPT-11."( Increased anticancer activity of the thymidylate synthase inhibitor BGC9331 combined with the topoisomerase I inhibitor SN-38 in human colorectal and breast cancer cells: induction of apoptosis and ROCK cleavage through caspase-3-dependent and -independen
André, T; Coudray, AM; De Gramont, A; Faivre, S; Gespach, C; Kornprobst, M; Larsen, AK; Louvet, C; Raymond, E; Tournigand, C, 2005
)
0.33
"Patients received pemetrexed 500 mg/m2 alone or in combination with cisplatin 75 mg/m2 once every 21 days for > or = 6 cycles."( Open-label study of pemetrexed alone or in combination with cisplatin for the treatment of patients with peritoneal mesothelioma: outcomes of an expanded access program.
Ashland, J; Belani, CP; Jänne, PA; Keohan, ML; Mintzer, DM; Monberg, MJ; Obasaju, CK; Polikoff, JA; Ross, HJ; Taylor, L; Wozniak, AJ; Ye, Z, 2005
)
0.33
" Pemetrexed is the first and only chemotherapy agent that has been granted a marketing approval for use in combination with cisplatin for the treatment of chemo-naïve patients with unresectable MPM."( Pemetrexed disodium in combination with cisplatin versus other cytotoxic agents or supportive care for the treatment of malignant pleural mesothelioma.
Dickson, R; Dodd, S; Dundar, Y; Green, J; Walley, T, 2007
)
0.34
"To examine evidence on the clinical effectiveness of pemetrexed disodium used in combination with cisplatin for the treatment of unresectable malignant pleural mesothelioma in chemotherapy naïve patients compared with other cytotoxic agents used alone or in combination, or supportive care."( Pemetrexed disodium in combination with cisplatin versus other cytotoxic agents or supportive care for the treatment of malignant pleural mesothelioma.
Dickson, R; Dodd, S; Dundar, Y; Green, J; Walley, T, 2007
)
0.34
"Pemetrexed disodium in combination with cisplatin and with folic acid and vitamin B(12 )supplementation may improve survival when used in combination with cisplatin in good performance status patients."( Pemetrexed disodium in combination with cisplatin versus other cytotoxic agents or supportive care for the treatment of malignant pleural mesothelioma.
Dickson, R; Dodd, S; Dundar, Y; Green, J; Walley, T, 2007
)
0.34
" In the present trial we investigated pemetrexed in combination with paclitaxel as front-line treatment in advanced or metastatic NSCLC."( Pemetrexed combined with paclitaxel in patients with advanced or metastatic non-small-cell lung cancer: a phase I-II trial.
Dimitroulis, J; Karaindros, D; Katis, C; Koutandos, J; Stathopoulos, GP; Stathopoulos, J; Toubis, M, 2007
)
0.34
" Patients were treated every 21 days with pemetrexed 500 mg/m alone (n = 91) or in combination with cisplatin 75 mg/m (n = 96) for a maximum of six cycles."( Pemetrexed alone or in combination with cisplatin in previously treated malignant pleural mesothelioma: outcomes from a phase IIIB expanded access program.
Belani, CP; Jänne, PA; Keohan, ML; Mintzer, DM; Monberg, MJ; Obasaju, CK; Polikoff, JA; Ross, HJ; Wozniak, AJ; Ye, Z, 2006
)
0.33
"The data from this EAP study suggest that patients with previously treated MPM can benefit from treatment with pemetrexed alone or in combination with cisplatin."( Pemetrexed alone or in combination with cisplatin in previously treated malignant pleural mesothelioma: outcomes from a phase IIIB expanded access program.
Belani, CP; Jänne, PA; Keohan, ML; Mintzer, DM; Monberg, MJ; Obasaju, CK; Polikoff, JA; Ross, HJ; Wozniak, AJ; Ye, Z, 2006
)
0.33
" The schedule was pemetrexed 500 mg/m(2) in combination with carboplatin area under the curve 5, every 21 days."( Phase II study of pemetrexed in combination with carboplatin in patients with malignant pleural mesothelioma (MPM).
Aitini, E; Alabiso, O; Botta, M; Buosi, R; Carbone, R; Castagneto, B; Degiovanni, D; Galbusera, V; Giaretto, L; Mencoboni, M; Muzio, A; Piccolini, E; Rebella, L; Serra, M; Spigno, F, 2008
)
0.35
"Pemetrexed in combination with cisplatin (Pem/Cis) is used globally for the treatment of malignant pleural mesothelioma (MPM)."( Efficacy and safety of pemetrexed in combination with cisplatin for malignant pleural mesothelioma: a phase I/II study in Japanese patients.
Adachi, S; Fukuoka, M; Gemba, K; Hida, T; Ichinose, Y; Kunitoh, H; Nakagawa, K; Nambu, Y; Saijo, N; Shinkai, T; Yamazaki, K, 2008
)
0.35
" Limited pharmacokinetic profiling of PTK787/ZK222584 revealed no evidence of drug-drug interactions."( A phase I trial of PTK787/ZK222584 in combination with pemetrexed and cisplatin in patients with advanced solid tumors.
Berg, W; Freeman, B; Manno, P; Sharma, S; Symanowski, J; Turner, J; Vogelzang, N, 2009
)
0.35
"This clinical trial assessed the efficacy of pemetrexed combined with oxaliplatin (PEMOX) in patients with advanced gastric cancer (AGC)."( Pemetrexed in combination with oxaliplatin as a first-line therapy for advanced gastric cancer: a multi-institutional phase II study.
Amoroso, V; Bajetta, E; Barone, C; Cascinu, S; Celio, L; Cetto, GL; Di Costanzo, F; La Torre, I; Labianca, R; Pinotti, G; Sternberg, CN, 2009
)
0.35
" The goal of this study was to evaluate the anticancer effect of a histone deacetylase inhibitor, valproate, on mesothelioma cells in combination with pemetrexed and cisplatin, the usual first-line regimen of chemotherapy for this tumor."( Valproate, in combination with pemetrexed and cisplatin, provides additional efficacy to the treatment of malignant mesothelioma.
Burny, A; Delvenne, P; Grigoriu, B; Hubert, P; Mascaux, C; Scherpereel, A; Vandermeers, F; Willems, L, 2009
)
0.35
" As expected, valproate alone or combined with pemetrexed and cisplatin triggers hyperacetylation of histone H3."( Valproate, in combination with pemetrexed and cisplatin, provides additional efficacy to the treatment of malignant mesothelioma.
Burny, A; Delvenne, P; Grigoriu, B; Hubert, P; Mascaux, C; Scherpereel, A; Vandermeers, F; Willems, L, 2009
)
0.35
"These observations support the potential additional efficacy of valproate in combination with pemetrexed and cisplatin for treatment of malignant mesothelioma."( Valproate, in combination with pemetrexed and cisplatin, provides additional efficacy to the treatment of malignant mesothelioma.
Burny, A; Delvenne, P; Grigoriu, B; Hubert, P; Mascaux, C; Scherpereel, A; Vandermeers, F; Willems, L, 2009
)
0.35
") in combination with pemetrexed."( Phase Ib safety and pharmacokinetic evaluation of daily and twice daily oral enzastaurin in combination with pemetrexed in advanced/metastatic cancer.
Giaccone, G; Graefe, T; Hanauske, AR; Kuenen, B; Lahn, M; McNealy, P; Musib, LC; Thornton, D; Weigang-Köhler, K; Yilmaz, E, 2009
)
0.35
" This review outlines the current preclinical and clinical studies using pemetrexed in combination with targeted agents in advanced NSCLC."( The role of pemetrexed combined with targeted agents for non-small cell lung cancer.
Jassem, J; Konopa, K, 2010
)
0.36
" Twenty-three patients experiencing progression following 6 months after concluding platinum-based chemotherapy were managed with second-line treatment with carboplatin combined with gemcitabine or pemetrexed."( High response of second-line chemotherapy with pemetrexed or gemcitabine combined with carboplatin in patients with non-small-cell lung cancer experiencing progression following 6 months after concluding platinum-based chemotherapy.
Arrieta, O; Astorga, A; Flores-Estrada, D; Martinez-Barrera, L; Michel Ortega, RM; Pachuca, D; Villarreal-Garza, C, 2011
)
0.37
"An oral daily dose of 500 mg enzastaurin was administered once daily (QD) or twice daily (BID) in combination with 500 mg/m pemetrexed on day 1 in repeated 21-day cycles."( A phase I study of enzastaurin combined with pemetrexed in advanced non-small cell lung cancer.
Baldwin, JR; Koshiji, M; Kunitoh, H; Murakami, H; Nakamura, Y; Nokihara, H; Ohe, Y; Shukuya, T; Takahashi, T; Tamura, T; Tanai, C; Yamamoto, N, 2010
)
0.36
"Both schedules of enzastaurin in combination with pemetrexed were well tolerated and clinically active in patients with advanced non-small cell lung cancer."( A phase I study of enzastaurin combined with pemetrexed in advanced non-small cell lung cancer.
Baldwin, JR; Koshiji, M; Kunitoh, H; Murakami, H; Nakamura, Y; Nokihara, H; Ohe, Y; Shukuya, T; Takahashi, T; Tamura, T; Tanai, C; Yamamoto, N, 2010
)
0.36
" In this study, using an intrathoracic murine model of malignant mesothelioma (MM), we provide evidence suggesting that Treg blockade could enhance survival when combined with pemetrexed in established tumor."( Synergistic antitumor effects of regulatory T cell blockade combined with pemetrexed in murine malignant mesothelioma.
Anraku, M; de Perrot, M; Johnston, MR; Keshavjee, S; Tagawa, T; Wu, L; Yun, Z; Zhang, L, 2010
)
0.36
"A phase I trial escalating doses of sorafenib in combination with fixed doses of PE (Arm A) or CbP (Arm B) was performed using a 3-patient cohort design to determine the maximum tolerated dose (MTD) and dose-limiting toxicities (DLT); DLT were assessed in the first cycle."( A phase I trial of sorafenib combined with cisplatin/etoposide or carboplatin/pemetrexed in refractory solid tumor patients.
Bernard, S; Chiu, M; Davies, JM; Dees, EC; Dhruva, NS; Hayes, DN; Hilbun, LR; Ivanova, A; Keller, K; Kim, WY; Socinski, MA; Stinchcombe, TE; Walko, CM, 2011
)
0.37
"The MTD of sorafenib was 200 mg BID continuously in combination with carboplatin (AUC of 5) and pemetrexed 500 mg/m² every 3 weeks."( A phase I trial of sorafenib combined with cisplatin/etoposide or carboplatin/pemetrexed in refractory solid tumor patients.
Bernard, S; Chiu, M; Davies, JM; Dees, EC; Dhruva, NS; Hayes, DN; Hilbun, LR; Ivanova, A; Keller, K; Kim, WY; Socinski, MA; Stinchcombe, TE; Walko, CM, 2011
)
0.37
"This phase I study evaluated the safety, tolerability, preliminary antitumor activity, and pharmacokinetic interaction of weekly topotecan (days 1 and 8) in combination with pemetrexed (day 1 only) in patients with advanced solid tumors."( A phase I study of weekly topotecan in combination with pemetrexed in patients with advanced malignancies.
Burris, HA; Greco, FA; Infante, JR; Jewell, RC; Jones, SF; Spigel, DR; Thompson, DS, 2010
)
0.36
" We have performed a feasibility study to investigate the safety of sunitinib in combination with pemetrexed for treatment of advanced refractory solid tumors."( Feasibility study of two schedules of sunitinib in combination with pemetrexed in patients with advanced solid tumors.
Fumita, S; Hashimoto, J; Ichikawa, Y; Kimura, N; Miyazaki, M; Nakagawa, K; Ohki, E; Okamoto, I; Shimizu, T; Takeda, M; Terashima, M; Tsurutani, J, 2012
)
0.38
"5 mg/day) or a 2-weeks-on, 1-week-off treatment schedule (50 mg/day, Schedule 2/1) in combination with pemetrexed at 500 mg/m(2) on day 1 of repeated 21-day cycles."( Feasibility study of two schedules of sunitinib in combination with pemetrexed in patients with advanced solid tumors.
Fumita, S; Hashimoto, J; Ichikawa, Y; Kimura, N; Miyazaki, M; Nakagawa, K; Ohki, E; Okamoto, I; Shimizu, T; Takeda, M; Terashima, M; Tsurutani, J, 2012
)
0.38
" Pharmacokinetic analysis did not reveal any substantial drug-drug interaction."( Feasibility study of two schedules of sunitinib in combination with pemetrexed in patients with advanced solid tumors.
Fumita, S; Hashimoto, J; Ichikawa, Y; Kimura, N; Miyazaki, M; Nakagawa, K; Ohki, E; Okamoto, I; Shimizu, T; Takeda, M; Terashima, M; Tsurutani, J, 2012
)
0.38
"This study evaluated overall survival (OS) of patients with advanced non-squamous NSCLC following treatment with the specific endothelin A receptor antagonist, zibotentan in combination with pemetrexed compared with pemetrexed monotherapy."( A phase II, randomized, multicenter study to assess the efficacy, safety, and tolerability of zibotentan (ZD4054) in combination with pemetrexed in patients with advanced non-small cell lung cancer.
Chouaid, C; Morris, T; Nathan, F; Pemberton, K, 2011
)
0.37
"In this double-blinded, placebo-controlled study, patients with advanced NSCLC with non-squamous histology who had failed first-line platinum-based chemotherapy were randomized to receive either once-daily zibotentan 10 mg in combination with 3-weekly pemetrexed 500 mg/m(2) or placebo plus 3-weekly pemetrexed 500 mg/m(2)."( A phase II, randomized, multicenter study to assess the efficacy, safety, and tolerability of zibotentan (ZD4054) in combination with pemetrexed in patients with advanced non-small cell lung cancer.
Chouaid, C; Morris, T; Nathan, F; Pemberton, K, 2011
)
0.37
"There was no survival signal in patients with NSCLC following treatment with zibotentan in combination with pemetrexed."( A phase II, randomized, multicenter study to assess the efficacy, safety, and tolerability of zibotentan (ZD4054) in combination with pemetrexed in patients with advanced non-small cell lung cancer.
Chouaid, C; Morris, T; Nathan, F; Pemberton, K, 2011
)
0.37
" The aim of this study is to evaluate the efficacy and side effects of pemetrexed combined with cisplatin/carboplatin in the treatment of advanced recurrent or metastasis NSCLC."( [Pemetrexed combined with cisplatin or carboplatin regimen in the treatment of advanced recurrent or metastasis non-small cell lung cancer: analysis of 63 cases].
Li, J; Li, X; Liu, J; Shang, L; Wang, W; Wen, F, 2011
)
0.37
"Pemetrexed combined with cisplatin/carboplatin is effective and feasible for advanced recurrent or metastasis NSCLC."( [Pemetrexed combined with cisplatin or carboplatin regimen in the treatment of advanced recurrent or metastasis non-small cell lung cancer: analysis of 63 cases].
Li, J; Li, X; Liu, J; Shang, L; Wang, W; Wen, F, 2011
)
0.37
" A Bayesian dose-escalation model was used to determine the feasible doses of daily or weekly everolimus combined with pemetrexed (500 mg/m q3w)."( Everolimus in combination with pemetrexed in patients with advanced non-small cell lung cancer previously treated with chemotherapy: a phase I study using a novel, adaptive Bayesian dose-escalation model.
Anrys, B; Boyer, M; Di Scala, L; Dimitrijevic, S; Laack, E; Miller, N; Petrovic, K; Pylvaenaeinen, I; Solomon, B; Vansteenkiste, J; Wolf, J, 2011
)
0.37
" The aim of this study is to compare the efficacies and safeties of pemetrexed alone with pemetrexed combined with oxaliplatin as salvage therapy in stage IV lung adenocarcinoma to provide evidences for combination therapy."( [Pemetrexed alone versus pemetrexed combined with oxaliplatin as salvage therapy in stage IV lung adenocarcinoma].
Gao, Z; Han, B; Jiang, L; Jiang, Y; Liu, Y; Wang, X, 2011
)
0.37
"From January 2009 to February 2011, 83 patients with stage IV lung adenocarcinoma received pemetrexed alone (single agent arm, n=47) or pemetrexed combined with oxaliplatin (combination arm, n=36) as salvage therapy."( [Pemetrexed alone versus pemetrexed combined with oxaliplatin as salvage therapy in stage IV lung adenocarcinoma].
Gao, Z; Han, B; Jiang, L; Jiang, Y; Liu, Y; Wang, X, 2011
)
0.37
"For salvage therapy, pemetrexed combined with oxaliplatin is tolerable in stage IV lung adenocarcinoma patients with good PS scores."( [Pemetrexed alone versus pemetrexed combined with oxaliplatin as salvage therapy in stage IV lung adenocarcinoma].
Gao, Z; Han, B; Jiang, L; Jiang, Y; Liu, Y; Wang, X, 2011
)
0.37
" No significant drug-drug interactions were identified."( A phase I dose-escalation and pharmacokinetic study of sunitinib in combination with pemetrexed in patients with advanced solid malignancies, with an expanded cohort in non-small cell lung cancer.
Blais, N; Camidge, DR; Canil, C; Chao, RC; Chow, LQ; Diab, SG; Jonker, DJ; Laurie, SA; McWilliam, M; Ruiz-Garcia, A; Thall, A; Tye, L; Zhang, K, 2012
)
0.38
" Pemetrexed in combination with carboplatin has been shown to be feasible in a phase I study in PSOC."( A phase II trial of pemetrexed in combination with carboplatin in patients with recurrent ovarian or primary peritoneal cancer.
Alvarez, AM; Bauknecht, T; Ghatage, P; Look, KY; Manouchehrpour, S; Oskay-Öezcelik, G; Sehouli, J; Szczylik, C; Zimmermann, A, 2012
)
0.38
" Defining the platinum-based combination with the best therapeutic index would require a prospective phase III study."( A phase II trial of pemetrexed in combination with carboplatin in patients with recurrent ovarian or primary peritoneal cancer.
Alvarez, AM; Bauknecht, T; Ghatage, P; Look, KY; Manouchehrpour, S; Oskay-Öezcelik, G; Sehouli, J; Szczylik, C; Zimmermann, A, 2012
)
0.38
" This phase II study aimed at evaluating its use in combination with cisplatin for recurrent or metastatic nasopharyngeal carcinoma (NPC)."( A phase II study of pemetrexed combined with cisplatin in patients with recurrent or metastatic nanopharyngeal carcinoma.
Chan, L; Lee, AW; Ng, WT; Shum, T; Yau, TK; Yeung, MW, 2012
)
0.38
" Pemetrexed has shown single-agent activity in SCCHN and in combination with cisplatin for other tumors."( Pemetrexed in combination with cisplatin versus cisplatin monotherapy in patients with recurrent or metastatic head and neck cancer: final results of a randomized, double-blind, placebo-controlled, phase 3 study.
Chang, SC; Frimodt-Moller, B; Hitt, R; Hong, RL; Hossain, AM; Koustenis, A; Licitra, L; Mezei, K; Reuter, C; Russo, F; Sahoo, TP; Shin, DM; Urba, S; van Herpen, CM, 2012
)
0.38
"Pemetrexed has radiosensitizing potential when evaluated in vitro in combination with platinum-containing compounds and radiation."( Dose-escalation study of thoracic radiotherapy in combination with pemetrexed plus Cisplatin followed by pemetrexed consolidation therapy in Japanese patients with locally advanced nonsquamous non-small-cell lung cancer.
Enatsu, S; Funai, J; Kubota, K; Nihei, K; Niho, S; Ohe, Y; Sekiguchi, R; Sekine, I; Sumi, M; Tamura, T, 2013
)
0.39
"Expected toxicities from concurrent chemoradiation were not worsened with concurrent TRT at a total dose of 66 Gy combined with pemetrexed in Japanese patients with locally advanced (LA) nonsquamous NSCLC."( Dose-escalation study of thoracic radiotherapy in combination with pemetrexed plus Cisplatin followed by pemetrexed consolidation therapy in Japanese patients with locally advanced nonsquamous non-small-cell lung cancer.
Enatsu, S; Funai, J; Kubota, K; Nihei, K; Niho, S; Ohe, Y; Sekiguchi, R; Sekine, I; Sumi, M; Tamura, T, 2013
)
0.39
"To evaluate the safety, pharmacokinetics (PKs), and pharmacodynamics of aflibercept, and to identify the recommended phase II dose (RP2D) of aflibercept in combination with pemetrexed and cisplatin."( A phase I dose-escalation study of aflibercept administered in combination with pemetrexed and cisplatin in patients with advanced solid tumours.
Colevas, AD; Diaz-Padilla, I; Gao, B; Lawson, EB; Leighl, NB; Lisano, J; Liu, L; Neal, JW; Razak, AR; San Pedro-Salcedo, M; Shepherd, FA; Siu, LL; Thibault, A; Wakelee, HA, 2012
)
0.38
"Aflibercept was administered at escalating doses of 2, 4, or 6 mg kg(-1) in combination with fixed doses of pemetrexed (500 mg m(-2)) plus cisplatin (75 mg m(-2)) every 3 weeks."( A phase I dose-escalation study of aflibercept administered in combination with pemetrexed and cisplatin in patients with advanced solid tumours.
Colevas, AD; Diaz-Padilla, I; Gao, B; Lawson, EB; Leighl, NB; Lisano, J; Liu, L; Neal, JW; Razak, AR; San Pedro-Salcedo, M; Shepherd, FA; Siu, LL; Thibault, A; Wakelee, HA, 2012
)
0.38
" The RP2D of aflibercept was 6 mg kg(-1), to be administered intravenously every 3 weeks in combination with pemetrexed and cisplatin."( A phase I dose-escalation study of aflibercept administered in combination with pemetrexed and cisplatin in patients with advanced solid tumours.
Colevas, AD; Diaz-Padilla, I; Gao, B; Lawson, EB; Leighl, NB; Lisano, J; Liu, L; Neal, JW; Razak, AR; San Pedro-Salcedo, M; Shepherd, FA; Siu, LL; Thibault, A; Wakelee, HA, 2012
)
0.38
"Due to the various inter-individual differences in the biological characteristics of tumor cells, as well as issues on the efficacy, adverse reactions, and defects of existing drugs, we compared the clinical efficacy and toxicity of pemetrexed and gemcitabine combined with cisplatin for the treatment of previously untreated advanced non-small cell lung cancer (NSCLC)."( [Efficacy and toxicity of pemetrexed or gemcitabine combined with cisplatin in the treatment of patients with advanced non-small cell lung cancer].
Cheng, G; Hu, C; Hu, X; Huang, C; Jiao, S; Li, K; Luo, R; Lv, W; Ouyang, X; Sun, Y; Wang, J; Wang, M; Wang, Y; Wang, Z; Zhang, S; Zheng, R, 2012
)
0.38
"251 patients were randomly divided into pemetrexed combined with cisplatin group (PP group) with 127 cases and gemcitabine combined with cisplatin group (GP group) with 124 cases."( [Efficacy and toxicity of pemetrexed or gemcitabine combined with cisplatin in the treatment of patients with advanced non-small cell lung cancer].
Cheng, G; Hu, C; Hu, X; Huang, C; Jiao, S; Li, K; Luo, R; Lv, W; Ouyang, X; Sun, Y; Wang, J; Wang, M; Wang, Y; Wang, Z; Zhang, S; Zheng, R, 2012
)
0.38
"The clinical efficacy of pemetrexed and gemcitabine combined with cisplatin for the treatment of previously untreated advanced NSCLC was roughly the same, but the adverse reactions decreased significantly in the PP group compared with those in the GP group."( [Efficacy and toxicity of pemetrexed or gemcitabine combined with cisplatin in the treatment of patients with advanced non-small cell lung cancer].
Cheng, G; Hu, C; Hu, X; Huang, C; Jiao, S; Li, K; Luo, R; Lv, W; Ouyang, X; Sun, Y; Wang, J; Wang, M; Wang, Y; Wang, Z; Zhang, S; Zheng, R, 2012
)
0.38
" There were no clinically significant drug-drug interactions."( Sunitinib combined with pemetrexed and cisplatin: results of a phase I dose-escalation and pharmacokinetic study in patients with advanced solid malignancies, with an expanded cohort in non-small cell lung cancer and mesothelioma.
Blais, N; Camidge, DR; Chao, RC; Chow, LQ; Doebele, RC; Jonker, DJ; Laurie, SA; Ruiz-Garcia, A; Soulières, D; Thall, A; Zhang, K, 2013
)
0.39
" Its activity on brain metastases makes pemetrexed attractive in combination with whole-brain radiation therapy (WBRT), but it could also potentially increase toxicity."( First assessment of whole-brain radiation therapy combined with pemetrexed-based chemotherapy in non-small-cell lung carcinoma: data on safety and efficacy.
Bauduceau, O; Ceccaldi, B; Chargari, C; Dulou, R; Fournel, P; Guy, JB; Jacob, J; Le Moulec, S; Magné, N; Moriceau, G; Moussaid, Y; Pacaut, C; Rivoirard, R; Védrine, L, 2013
)
0.39
"5%) alone, or combined with distant metastases (48."( Phase II study of pemetrexed in combination with cisplatin and cetuximab in recurrent or metastatic squamous cell carcinoma of the head and neck.
Chang, SC; Dietz, A; Gauler, TC; Hamid, O; Hossain, AM; Licitra, L; Lopez-Picazo, JM; Stöhlmacher-Williams, J; Vermorken, JB, 2013
)
0.39
"To observe the clinical efficacy and safety of pemetrexed or gemcitabine combined with carboplatin as the first-line therapy in elderly patients with advanced non-small cell lung cancer (NSCLC)."( [Efficacy and safety of pemetrexed or gemcitabine combined with carboplatin as the first-line therapy in elderly patients with advanced non-small cell lung cancer].
Shi, X; Yu, XM; Zhang, YP; Zhao, J, 2013
)
0.39
" Thus, pemetrexed combined with carboplatin is an effective chemotherapeutic regimen for advanced NSCLC in elderly patients."( [Efficacy and safety of pemetrexed or gemcitabine combined with carboplatin as the first-line therapy in elderly patients with advanced non-small cell lung cancer].
Shi, X; Yu, XM; Zhang, YP; Zhao, J, 2013
)
0.39
" Pharmacokinetic data revealed no clinically significant drug-drug interactions."( Sunitinib combined with pemetrexed and carboplatin in patients with advanced solid malignancies--results of a phase I dose-escalation study.
Blais, N; Camidge, DR; Chao, RC; Chow, LQ; Diab, SG; Jonker, DJ; Laurie, SA; Ruiz-Garcia, A; Soulières, D; Thall, A; Zhang, K, 2013
)
0.39
" Application of (177)Lu-EC0800 with PMXther resulted in a two- to four-fold enhanced tumor growth delay and a prolonged survival of KB and IGROV-1 tumor-bearing mice, as compared to the combination with PMXsubther or untreated control mice."( 177Lu-EC0800 combined with the antifolate pemetrexed: preclinical pilot study of folate receptor targeted radionuclide tumor therapy.
Haller, S; Leamon, CP; Müller, C; Reber, J, 2013
)
0.39
"To investigate the efficacy and safety of Javanica oil emulsion injection (Yadanzi®) combined with pemetrexed and platinum (PP) for treating patients with advanced lung cancer."( Phase II study on Javanica oil emulsion injection (Yadanzi®) combined with chemotherapy in treating patients with advanced lung adenocarcinoma.
Cao, J; Huang, XE; Liu, J; Lu, YY; Wu, XY; Xiang, J; Xu, L; Xu, X, 2013
)
0.39
"Javanica oil emulsion injection combined with chemotherapy could be considered as a safe and effective regimen in treating patients with advanced lung adenocarcinoma."( Phase II study on Javanica oil emulsion injection (Yadanzi®) combined with chemotherapy in treating patients with advanced lung adenocarcinoma.
Cao, J; Huang, XE; Liu, J; Lu, YY; Wu, XY; Xiang, J; Xu, L; Xu, X, 2013
)
0.39
"This study evaluated the efficacy and safety of ziv-aflibercept in combination with cisplatin and pemetrexed in non-small cell lung cancer (NSCLC)."( A phase II multicentre study of ziv-aflibercept in combination with cisplatin and pemetrexed in patients with previously untreated advanced/metastatic non-squamous non-small cell lung cancer.
Adjei, AA; Brahmer, JR; Chen, H; Dicioccio, AT; Gao, B; Jotte, RM; Kuo, CJ; Leighl, NB; Liu, L; Modiano, MR; Neal, JW; Riess, JW; Rigas, JR; Wakelee, HA, 2014
)
0.4
"This randomized open-label phase II study evaluated the efficacy, safety, and tolerability of pazopanib in combination with pemetrexed compared with the standard cisplatin/pemetrexed doublet in patients with previously untreated, advanced, nonsquamous non-small-cell lung cancer."( An open-label, multicenter, randomized, phase II study of pazopanib in combination with pemetrexed in first-line treatment of patients with advanced-stage non-small-cell lung cancer.
Besse, B; Bosquee, L; Chouaid, C; Felip, E; Lechevalier, T; Lianes-Barragán, P; Mellemgaard, A; Ottesen, LH; Paul, EM; Reck, M; Ruiz-Soto, R; Scagliotti, GV; Sigal, E; von Pawel, J, 2013
)
0.39
" Although most patients eligible for surgery undergo cytoreductive surgery in combination with hyperthermic intraperitoneal chemotherapy, the role of perioperative systemic chemotherapy still remains undefined."( Complete pathological response to neoadjuvant pemetrexed/cisplatin in combination with regional hyperthermia in a patient with sarcomatoid peritoneal mesothelioma.
Albertsmeier, M; Angele, MK; Boeck, S; Graser, A; Haas, M; Heinemann, V; Kruger, S; Lindner, LH; Reu, S; Sotlar, K; Stemmler, HJ, 2014
)
0.4
" This study was conducted to elucidate the safety and efficacy of palliative chemotherapy with gemcitabine or pemetrexed, both in combination with a platinum agent in NSCLC patients with ILD."( Safety and efficacy of gemcitabine or pemetrexed in combination with a platinum in patients with non-small-cell lung cancer and prior interstitial lung disease.
Ahn, JS; Ahn, MJ; Chang, W; Choi, MK; Chung, MP; Hong, JY; Jung, HA; Kim, M; Kim, S; Lee, SJ; Park, K; Park, S; Sun, JM, 2014
)
0.4
"Patients with advanced or recurrent NSCLC and ILD who received gemcitabine or pemetrexed in combination with a platinum agent as first-line chemotherapy were retrospectively analyzed."( Safety and efficacy of gemcitabine or pemetrexed in combination with a platinum in patients with non-small-cell lung cancer and prior interstitial lung disease.
Ahn, JS; Ahn, MJ; Chang, W; Choi, MK; Chung, MP; Hong, JY; Jung, HA; Kim, M; Kim, S; Lee, SJ; Park, K; Park, S; Sun, JM, 2014
)
0.4
"Our results suggest that gemcitabine or pemetrexed in combination with platinum agents could be a feasible option for advanced NSCLC with ILD with some risk of AE-ILD or early death."( Safety and efficacy of gemcitabine or pemetrexed in combination with a platinum in patients with non-small-cell lung cancer and prior interstitial lung disease.
Ahn, JS; Ahn, MJ; Chang, W; Choi, MK; Chung, MP; Hong, JY; Jung, HA; Kim, M; Kim, S; Lee, SJ; Park, K; Park, S; Sun, JM, 2014
)
0.4
" The aim of this study was to investigate the antitumor activity of recombinant human Apo2L/TRAIL (dulanermin) in combination with chemotherapy in MPM in vitro and in vivo."( Synergistic antitumor activity of recombinant human Apo2L/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in combination with carboplatin and pemetrexed in malignant pleural mesothelioma.
Cavallari, I; Ciminale, V; Favaretto, A; Marulli, G; Nannini, N; Pasello, G; Rea, F; Schiavon, M; Silic-Benussi, M; Urso, L, 2014
)
0.4
"In the present studies, we employed a panel of MPM cell lines to test the antitumor activity of recombinant human Apo2L/TRAIL (T) in combination with carboplatin and pemetrexed (CP) in vitro and SCID mice."( Synergistic antitumor activity of recombinant human Apo2L/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in combination with carboplatin and pemetrexed in malignant pleural mesothelioma.
Cavallari, I; Ciminale, V; Favaretto, A; Marulli, G; Nannini, N; Pasello, G; Rea, F; Schiavon, M; Silic-Benussi, M; Urso, L, 2014
)
0.4
" LY2603618 administered in combination with pemetrexed and cisplatin demonstrated an acceptable safety profile."( Preclinical analyses and phase I evaluation of LY2603618 administered in combination with pemetrexed and cisplatin in patients with advanced cancer.
Barnard, D; Calles, A; Calvo, E; Chen, VJ; Diaz, HB; Dickgreber, N; Huber, L; Hynes, SM; Iversen, P; Kays, L; Kumm, E; Lin, AB; Marshall, M; Merzoug, FF; Ohnmacht, U; Sebastian, M; Voss, B; Wehler, T, 2014
)
0.4
" Erlotinib combined with pemetrexed/cisplatin may be effective in the treatment of LM in EGFR mutation patients after gefitinib failure."( Erlotinib in combination with pemetrexed/cisplatin for leptomeningeal metastases and cerebrospinal fluid drug concentrations in lung adenocarcinoma patients after gefitinib faliure.
Deng, Q; He, J; Liu, X; Xu, X; Yang, H; Yang, X; Zhang, Y; Zhao, M, 2015
)
0.42
"The primary objective of this study was to determine the safety and maximum tolerated dose (MTD) of the antimesothelin immunotoxin SS1(dsFv)PE38 (SS1P) (a recombinant antimesothelin immunotoxin consisting of a murine antimesothelin variable antibody fragment [Fv] linked to PE38, a truncated portion of Pseudomonas exotoxin A) in combination with pemetrexed and cisplatin in chemotherapy-naive patients with advanced malignant pleural mesothelioma (MPM)."( Phase 1 study of the antimesothelin immunotoxin SS1P in combination with pemetrexed and cisplatin for front-line therapy of pleural mesothelioma and correlation of tumor response with serum mesothelin, megakaryocyte potentiating factor, and cancer antigen
Hassan, R; Hollevoet, K; Kreitman, RJ; Ling, A; Miettinen, M; Pastan, I; Sharon, E; Steinberg, SM; Thomas, A; Zhang, J, 2014
)
0.4
"SS1P given with pemetrexed and cisplatin is safe and well tolerated and exhibits significant antitumor activity in patients with unresectable, advanced pleural mesothelioma."( Phase 1 study of the antimesothelin immunotoxin SS1P in combination with pemetrexed and cisplatin for front-line therapy of pleural mesothelioma and correlation of tumor response with serum mesothelin, megakaryocyte potentiating factor, and cancer antigen
Hassan, R; Hollevoet, K; Kreitman, RJ; Ling, A; Miettinen, M; Pastan, I; Sharon, E; Steinberg, SM; Thomas, A; Zhang, J, 2014
)
0.4
"In a 3 + 3 dose-escalation design, patients were given intravenous pemetrexed (500 mg/m(2)) on day 1 of a 21-day cycle (maximum 6 cycles), combined with continuous daily oral afatinib (schedule A [SA]; starting dose 30 mg, escalation to 50 mg) or pulsed-dose daily oral afatinib (schedule B [SB]; starting dose 50 mg, escalation to 70 mg) on days 1-6 of each 21-day cycle."( A phase I, dose-escalation trial of continuous- and pulsed-dose afatinib combined with pemetrexed in patients with advanced solid tumors.
Chand, VK; Chu, QS; Hirte, HW; Hotte, SJ; Sangha, R; Schnell, D; Sergenson, G, 2014
)
0.4
"Continuous- or pulsed-dose afatinib combined with pemetrexed exhibited a manageable safety profile."( A phase I, dose-escalation trial of continuous- and pulsed-dose afatinib combined with pemetrexed in patients with advanced solid tumors.
Chand, VK; Chu, QS; Hirte, HW; Hotte, SJ; Sangha, R; Schnell, D; Sergenson, G, 2014
)
0.4
"Fifty-nine mesothelioma patients (31 men with a median age of 62 years) treated in first-line chemotherapy with platinum in combination with pemetrexed or pemetrexed monotherapy were enrolled."( Correlation between TS, MTHFR, and ERCC1 gene polymorphisms and the efficacy of platinum in combination with pemetrexed first-line chemotherapy in mesothelioma patients.
Dyszkiewicz, W; Kalinka-Warzocha, E; Knetki-Wróblewska, M; Kowalski, DM; Krawczyk, P; Krzakowski, M; Kucharczyk, T; Milanowski, J; Powrózek, T; Ramlau, R; Winiarczyk, K, 2014
)
0.4
" This phase 2 study investigated ramucirumab in combination with first-line pemetrexed and platinum chemotherapy in advanced/metastatic NSCLC."( Phase 2, randomized, open-label study of ramucirumab in combination with first-line pemetrexed and platinum chemotherapy in patients with nonsquamous, advanced/metastatic non-small cell lung cancer.
Alexandris, E; Bonomi, P; Bustin, F; Camidge, DR; Cao, D; Doebele, RC; Eakle, J; Goldschmidt, J; Reck, M; Spigel, D; Tehfe, M; Thomas, S; Verma, S; Yurasov, S, 2015
)
0.42
"The primary endpoint of significant prolongation of PFS was not met; however, ramucirumab showed evidence of clinical activity in combination with pemetrexed and platinum in nonsquamous NSCLC patients."( Phase 2, randomized, open-label study of ramucirumab in combination with first-line pemetrexed and platinum chemotherapy in patients with nonsquamous, advanced/metastatic non-small cell lung cancer.
Alexandris, E; Bonomi, P; Bustin, F; Camidge, DR; Cao, D; Doebele, RC; Eakle, J; Goldschmidt, J; Reck, M; Spigel, D; Tehfe, M; Thomas, S; Verma, S; Yurasov, S, 2015
)
0.42
" Docetaxel 75 mg/m(2) or pemetrexed 500 mg/m(2) once every 21 days per the investigator was administered with apricoxib or placebo 400 mg once per day."( Randomized, double-blind, placebo-controlled, multicenter phase II study of the efficacy and safety of apricoxib in combination with either docetaxel or pemetrexed in patients with biomarker-selected non-small-cell lung cancer.
de Mayolo, JA; Edelman, MJ; Evans, TL; Feliciano, J; Fidler, MJ; Keresztes, R; Medeiros, M; Otterson, G; Rogers, JS; Sanborn, RE; Schneider, BJ; Sequist, LV; Tan, MT; Yang, Y; Zaknoen, SL, 2015
)
0.42
" Fifteen patients were enrolled in a dose escalation of eribulin mesylate in combination with pemetrexed (E+P)."( An open-label, multicenter, randomized phase Ib/II study of eribulin mesylate administered in combination with pemetrexed versus pemetrexed alone as second-line therapy in patients with advanced nonsquamous non-small-cell lung cancer.
Bondarenko, I; Dave, H; Freeman, A; Hodge, JP; Huber, B; Lieberman, R; Shelton, MJ; Shparyk, Y; Vynnychenko, I; Waller, CF, 2015
)
0.42
" The in vitro to in vivo extrapolation approach used in this work was developed to predict possible drug-drug interactions (DDIs) that may occur after coadministration of pemetrexed and nonsteroidal anti-inflammatory drugs (NSAIDs), and it included in vitro assays, risk assessment models, and physiologically based pharmacokinetic (PBPK) models."( Prediction of renal transporter mediated drug-drug interactions for pemetrexed using physiologically based pharmacokinetic modeling.
Bacon, JA; Hall, SD; Higgins, JW; Hillgren, KM; Kim, RB; Pak, YA; Posada, MM; Schneck, KB; Tirona, RG, 2015
)
0.42
" Thus, we examined the feasibility of using thoracic radiotherapy combined with concurrent full-dose pemetrexed as the first-line treatment for advanced nonsquamous NSCLC patients."( Full-dose pemetrexed plus cisplatin combined with concurrent thoracic radiotherapy for previously untreated advanced nonsquamous non-small cell lung cancer.
Cai, XW; Feng, W; Fu, XL; Liu, Q; Xue, MC; Yu, W; Zhang, Q; Zhu, ZF, 2015
)
0.42
" This randomized, phase II trial (n [ 143) investigated volasertib monotherapy or in combination with pemetrexed compared with pemetrexed monotherapy in patients with NSCLC whose disease had progressed after previous platinum-based chemotherapy."( A Randomized, Open-Label Phase II Trial of Volasertib as Monotherapy and in Combination With Standard-Dose Pemetrexed Compared With Pemetrexed Monotherapy in Second-Line Treatment for Non-Small-Cell Lung Cancer.
Blais, N; Chu, Q; Ellis, PM; Gu, Y; Hirsh, V; Leighl, NB; Liu, D; Pilz, K; Reaume, MN; Sadrolhefazi, B; Wierzbicki, R, 2015
)
0.42
" Pharmacokinetics analyses showed no drug-drug interactions between volasertib and pemetrexed."( A Randomized, Open-Label Phase II Trial of Volasertib as Monotherapy and in Combination With Standard-Dose Pemetrexed Compared With Pemetrexed Monotherapy in Second-Line Treatment for Non-Small-Cell Lung Cancer.
Blais, N; Chu, Q; Ellis, PM; Gu, Y; Hirsh, V; Leighl, NB; Liu, D; Pilz, K; Reaume, MN; Sadrolhefazi, B; Wierzbicki, R, 2015
)
0.42
" The objective of this phase 1 dose de-escalation trial was to estimate the minimum tolerated dose of [6R]-MTHF to be used in combination with pemetrexed 500 mg/m(2) in the neoadjuvant treatment of patients with rectal cancer."( Phase 1 dose de-escalation trial of the endogenous folate [6R]-5,10-methylene tetrahydrofolate in combination with fixed-dose pemetrexed as neoadjuvant therapy in patients with resectable rectal cancer.
Björkqvist, H; Carlsson, G; Derwinger, K; Gibson, F; Gustavsson, B; Kurlberg, G; Odin, E; Swartling, T, 2015
)
0.42
"Adult patients (≥18 years) with resectable rectal adenocarcinoma were allocated to [6R]-MTHF doses of 500, 100, 50, and 10 mg/m(2) in combination with pemetrexed 500 mg/m(2)."( Phase 1 dose de-escalation trial of the endogenous folate [6R]-5,10-methylene tetrahydrofolate in combination with fixed-dose pemetrexed as neoadjuvant therapy in patients with resectable rectal cancer.
Björkqvist, H; Carlsson, G; Derwinger, K; Gibson, F; Gustavsson, B; Kurlberg, G; Odin, E; Swartling, T, 2015
)
0.42
"The results of this phase 1 study indicate that the estimated minimum tolerated dose of [6R]-MTHF was 100 mg/m(2) once weekly in combination with pemetrexed 500 mg/m(2)."( Phase 1 dose de-escalation trial of the endogenous folate [6R]-5,10-methylene tetrahydrofolate in combination with fixed-dose pemetrexed as neoadjuvant therapy in patients with resectable rectal cancer.
Björkqvist, H; Carlsson, G; Derwinger, K; Gibson, F; Gustavsson, B; Kurlberg, G; Odin, E; Swartling, T, 2015
)
0.42
"LY2090314 (LY) is a glycogen synthase kinase 3 inhibitor with preclinical efficacy in xenograft models when combined with platinum regimens."( A first-in-human phase I dose-escalation, pharmacokinetic, and pharmacodynamic evaluation of intravenous LY2090314, a glycogen synthase kinase 3 inhibitor, administered in combination with pemetrexed and carboplatin.
Brail, LH; Burris, HA; Chow, KH; Cooksey, JF; Farrington, DL; Gray, JE; Infante, JR; Jackson, KA; Jones, SF; Simon, GR; Yeo, A; Zamek-Gliszczynski, MJ, 2015
)
0.42
"To study the effectiveness of human recombinant endostatin injection (Endostar®) combined with cisplatin doublets in treating advanced non-small cell lung cancer (NSCLC), and to evaluate outcome by CT perfusion imaging."( Human Recombinant Endostatin Combined with Cisplatin Based Doublets in Treating Patients with Advanced NSCLC and Evaluation by CT Perfusion Imaging.
Chen, SQ; Cui, FB; Gao, EY; Jiang, BQ; Li, M; Shu, RB; Sun, P; Tang, W; Wang, H; Zhang, FL; Zhang, Y, 2015
)
0.42
"The response rate with Endostar® administered 4 days before chemotherapy and combined with chemotherapy from day 5 in group A was better than Endostar® combined with chemotherapy from the first day, and CT perfusion imaging could be a reasonable method for evaluation of patient outcomes."( Human Recombinant Endostatin Combined with Cisplatin Based Doublets in Treating Patients with Advanced NSCLC and Evaluation by CT Perfusion Imaging.
Chen, SQ; Cui, FB; Gao, EY; Jiang, BQ; Li, M; Shu, RB; Sun, P; Tang, W; Wang, H; Zhang, FL; Zhang, Y, 2015
)
0.42
"This open-label, phase I, dose-escalation part of a phase I/II study evaluated the safety, pharmacokinetics, and preliminary efficacy of nintedanib, a triple angiokinase inhibitor, combined with pemetrexed in Japanese patients with advanced non-small cell lung cancer (NSCLC) after first-line chemotherapy."( Phase I study of nintedanib in combination with pemetrexed as second-line treatment of Japanese patients with advanced non-small cell lung cancer.
Daga, H; Goto, K; Kaiser, R; Kaneda, H; Konishi, K; Miyazaki, M; Nakagawa, K; Okada, H; Okamoto, I; Sarashina, A; Takeda, K; Tanaka, T; Ueda, S; Yoh, K, 2015
)
0.42
"We investigated the feasibility of cisplatin or carboplatin combined with pemetrexed as adjuvant treatment in patients with completely resected Stage IB/II Non-Small-Cell Lung Cancer (NSCLC)."( A randomized Phase 2 study of pemetrexed in combination with cisplatin or carboplatin as adjuvant chemotherapy in patients with completely resected stage IB or II Non-Small-Cell Lung Cancer.
Chouaid, C; Engel-Riedel, W; Fischer, JR; Mazières, J; Nguyen, T; Reck, M; Ripoche, V; Schmid-Bindert, G; Schuette, W; Soldatenkova, V; Stöhlmacher, J; Vinolas, N; Visseren-Grul, C; Wolf, M, 2015
)
0.42
" Safety and PK of LY2603618 in combination with pemetrexed were favorable."( Phase II evaluation of LY2603618, a first-generation CHK1 inhibitor, in combination with pemetrexed in patients with advanced or metastatic non-small cell lung cancer.
Boyd, TE; Hynes, SM; Kang, JH; Kim, SW; Lin, AB; Lin, J; Myrand, SP; Novello, S; Park, K; Pinder-Schenck, M; Richards, DA; Rosenberg, R; Scagliotti, G; Smith, D; Smyth, EN; Su, WC; Wijayawardana, S, 2016
)
0.43
" CheckMate 012, a phase I, multicohort study, was conducted to explore the safety and efficacy of nivolumab as monotherapy or combined with current standard therapies in first-line advanced NSCLC."( Nivolumab in Combination With Platinum-Based Doublet Chemotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer.
Antonia, S; Borghaei, H; Brahmer, JR; Chen, AC; Chow, LQ; Gerber, DE; Gettinger, S; Goldman, JW; Harbison, CT; Hellmann, MD; Juergens, RA; Laurie, SA; Nathan, FE; Rizvi, NA; Shen, Y; Shepherd, FA, 2016
)
0.43
" This single-center phase Ib study investigated the tolerability, safety, and pharmacokinetics of nivolumab combined with standard chemotherapy in patients with advanced non-small-cell lung cancer (NSCLC)."( Safety and efficacy of nivolumab and standard chemotherapy drug combination in patients with advanced non-small-cell lung cancer: a four arms phase Ib study.
Fujiwara, Y; Goto, K; Horinouchi, H; Hozumi, H; Kanda, S; Kitazono, S; Kubo, E; Mizugaki, H; Nokihara, H; Shiraishi, H; Sunami, K; Tamura, T; Tanaka, A; Utsumi, H; Yamamoto, N, 2016
)
0.43
"Patients with untreated stage IIIB/IV non-squamous NSCLC, stratified by MET diagnostic status, were randomized to receive onartuzumab (15 mg/kg intravenously every 3 weeks) or placebo in combination with either paclitaxel/platinum/bevacizumab (bevacizumab cohort), or in combination with platinum/pemetrexed (pemetrexed cohort) with maintenance bevacizumab or pemetrexed and onartuzumab/placebo as appropriate."( Efficacy and Safety of Onartuzumab in Combination With First-Line Bevacizumab- or Pemetrexed-Based Chemotherapy Regimens in Advanced Non-Squamous Non-Small-Cell Lung Cancer.
Boyer, M; Braiteh, F; Cosgriff, T; De Braud, F; Hsu, J; Kaen, D; Kingsley, CD; Lawler, W; Lena, H; Lowe, T; Mekhail, T; Novello, S; Phan, S; Schütte, W; Wakelee, H; Zvirbule, Z, 2017
)
0.46
"Onartuzumab does not appear to provide any additional clinical benefit when given in combination with current first-line standard-of-care chemotherapy for non-squamous NSCLC."( Efficacy and Safety of Onartuzumab in Combination With First-Line Bevacizumab- or Pemetrexed-Based Chemotherapy Regimens in Advanced Non-Squamous Non-Small-Cell Lung Cancer.
Boyer, M; Braiteh, F; Cosgriff, T; De Braud, F; Hsu, J; Kaen, D; Kingsley, CD; Lawler, W; Lena, H; Lowe, T; Mekhail, T; Novello, S; Phan, S; Schütte, W; Wakelee, H; Zvirbule, Z, 2017
)
0.46
" The primary end point established the maximum tolerated dose in combination with cisplatin-pemetrexed in a dose deescalation scheme."( Phase I Trial of Cediranib in Combination with Cisplatin and Pemetrexed in Chemonaive Patients with Unresectable Malignant Pleural Mesothelioma (SWOG S0905).
Gandara, DR; Kalemkerian, GP; Kelly, K; Moon, J; Redman, MW; Tsao, AS; Vogelzang, NJ; Wistuba, II, 2017
)
0.46
"Cediranib combined with cisplatin-pemetrexed has a reasonable toxicity profile and preliminary promising efficacy."( Phase I Trial of Cediranib in Combination with Cisplatin and Pemetrexed in Chemonaive Patients with Unresectable Malignant Pleural Mesothelioma (SWOG S0905).
Gandara, DR; Kalemkerian, GP; Kelly, K; Moon, J; Redman, MW; Tsao, AS; Vogelzang, NJ; Wistuba, II, 2017
)
0.46
" Chemotherapy-naïve patients with advanced non-squamous non-small-cell lung cancer were enrolled in a dose-escalation study (standard 3 + 3 design) of SASP in combination with cisplatin and pemetrexed."( Phase I study of salazosulfapyridine in combination with cisplatin and pemetrexed for advanced non-small-cell lung cancer.
Fujita, A; Harada, T; Hirai, F; Imamura, CK; Inada, M; Iwama, E; Kishimoto, J; Mushiroda, T; Nagano, O; Nakanishi, Y; Nosaki, K; Ogata, H; Okamoto, I; Otsubo, K; Ozeki, T; Sakata, S; Saya, H; Seto, T; Suina, K; Takenoyama, M; Toyokawa, G; Tsuchihashi, K, 2017
)
0.46
" This study aimed to evaluate the pan-PI3K inhibitor pictilisib in combination with first-line treatment regimens that were the standard of care at the time of study, in patients with NSCLC."( A phase IB dose-escalation study of the safety and pharmacokinetics of pictilisib in combination with either paclitaxel and carboplatin (with or without bevacizumab) or pemetrexed and cisplatin (with or without bevacizumab) in patients with advanced non-s
Adjei, AA; Bahleda, R; Besse, B; Dy, GK; Ferte, C; Groen, HJM; Lin, W; Morrissey, K; Planchard, D; Schutzman, JL; Shankar, G; Soria, JC; Ware, J; Zhou, J, 2017
)
0.46
"To evaluate the clinical effectiveness of pemetrexed combined with cisplatin for the first-line chemotherapy of patients with advanced non-small-cell lung cancer (NSCLC) and maintenance treatment."( Clinical effectiveness of pemetrexed combined with cisplatin chemotherapy for advanced and maintenance treatment for patients with non-small-cell lung cancer.
Chen, WH; Dai, HP; Gan, YL; Xu, HY; Zhu, YM, 2018
)
0.48
"240 advanced NSCLC patients were randomly divided into either a control group (treated with gemcitabine combined with cisplatin) or an observation group (treated with pemetrexed combined with cisplatin)."( Clinical effectiveness of pemetrexed combined with cisplatin chemotherapy for advanced and maintenance treatment for patients with non-small-cell lung cancer.
Chen, WH; Dai, HP; Gan, YL; Xu, HY; Zhu, YM, 2018
)
0.48
"Pemetrexed combined with cisplatin was both safe and efficacious for the first-line chemotherapy of NSCLC patients at a progressive stage and for maintenance treatment."( Clinical effectiveness of pemetrexed combined with cisplatin chemotherapy for advanced and maintenance treatment for patients with non-small-cell lung cancer.
Chen, WH; Dai, HP; Gan, YL; Xu, HY; Zhu, YM, 2018
)
0.48
" Methods Patients were administered escalating twice daily doses of vatalanib in combination with pemetrexed disodium in 21-day cycles."( A phase I study of the vascular endothelial growth factor inhibitor Vatalanib in combination with Pemetrexed disodium in patients with advanced solid tumors.
Adjei, AA; Lim, VS; Molina, J; Satele, D; Wang, F; Yin, J, 2019
)
0.51
"We describe a phase Ib clinical trial evaluating treatment with BI1361849 combined with local radiation in 26 stage IV NSCLC patients with partial response (PR)/stable disease (SD) after standard first-line therapy."( Phase Ib evaluation of a self-adjuvanted protamine formulated mRNA-based active cancer immunotherapy, BI1361849 (CV9202), combined with local radiation treatment in patients with stage IV non-small cell lung cancer.
Alt, J; Bischoff, H; Cathomas, R; Doener, F; Fotin-Mleczek, M; Früh, M; Geißler, M; Gnad-Vogt, U; Griesinger, F; Hilbe, W; Hipp, MM; Hong, HS; Kallen, KJ; Klinkhardt, U; Koch, SD; Muth, A; Pall, G; Papachristofilou, A; Pless, M; Scheel, B; Schröder, A; Sebastian, M; Seibel, T; Stosnach, C; Wehler, T; Weiss, C; Zippelius, A, 2019
)
0.51
" One patient had a partial response in combination with pemetrexed maintenance, and 46."( Phase Ib evaluation of a self-adjuvanted protamine formulated mRNA-based active cancer immunotherapy, BI1361849 (CV9202), combined with local radiation treatment in patients with stage IV non-small cell lung cancer.
Alt, J; Bischoff, H; Cathomas, R; Doener, F; Fotin-Mleczek, M; Früh, M; Geißler, M; Gnad-Vogt, U; Griesinger, F; Hilbe, W; Hipp, MM; Hong, HS; Kallen, KJ; Klinkhardt, U; Koch, SD; Muth, A; Pall, G; Papachristofilou, A; Pless, M; Scheel, B; Schröder, A; Sebastian, M; Seibel, T; Stosnach, C; Wehler, T; Weiss, C; Zippelius, A, 2019
)
0.51
" The aim of this study is to investigate the efficacy of pemetrexed combined with platinum chemotherapy in patients with HER2-mutant and HER2 wild-type lung adenocarcinoma."( [Association between the HER2 Gene Status and the Efficacy of First-line Pemetrexed Combined with Platinum Chemotherapy in Patients with Advanced Lung 
Adenocarcinoma].
Li, B; Li, P; Li, X; Shen, S; Shi, Y; Zhang, F, 2019
)
0.51
"HER2-mutant lung adenocarcinoma patients with first-line pemetrexed combined with platinum chemotherapy have greater clinical benefit than HER2 wild-type patients."( [Association between the HER2 Gene Status and the Efficacy of First-line Pemetrexed Combined with Platinum Chemotherapy in Patients with Advanced Lung 
Adenocarcinoma].
Li, B; Li, P; Li, X; Shen, S; Shi, Y; Zhang, F, 2019
)
0.51
" Conclusion At 15 mg/kg weekly, GSK3052230 was well tolerated in combination with pemetrexed/cisplatin and durable responses were observed."( A phase Ib study of GSK3052230, an FGF ligand trap in combination with pemetrexed and cisplatin in patients with malignant pleural mesothelioma.
Baker-Neblett, K; Bellovin, DI; DeYoung, MP; Dómine, M; Fennell, DA; Gadgeel, S; Kindler, HL; Kostorov, V; Levchenko, E; López, PG; Mitrica, I; Morgensztern, D; Orlov, S; Schellens, JHM; Trigo, J; van Brummelen, EMJ; Vansteenkiste, JF; Vasquez, J; Viteri, S; Wang, X; Yan, L; Zauderer, MG, 2020
)
0.56
" In this study, we evaluated whether MWA combined with chemotherapy could improve progression-free survival (PFS) of patients with stage IV lung adenocarcinoma compared with chemotherapy alone."( Microwave ablation combined with chemotherapy improved progression free survival of IV stage lung adenocarcinoma patients compared with chemotherapy alone.
Li, C; Shao, JB; Sun, ZG; Wang, J; Zhang, N; Zhu, LM, 2019
)
0.51
"A total of 49 patients were enrolled into the study; 21 patients accepted MWA therapy combined with chemotherapy, 28 patients accepted only chemotherapy."( Microwave ablation combined with chemotherapy improved progression free survival of IV stage lung adenocarcinoma patients compared with chemotherapy alone.
Li, C; Shao, JB; Sun, ZG; Wang, J; Zhang, N; Zhu, LM, 2019
)
0.51
" Cox multivariate regression demonstrated that MWA combined with chemotherapy was the independent factor for both the PFS and TTLP."( Microwave ablation combined with chemotherapy improved progression free survival of IV stage lung adenocarcinoma patients compared with chemotherapy alone.
Li, C; Shao, JB; Sun, ZG; Wang, J; Zhang, N; Zhu, LM, 2019
)
0.51
"MWA, as a topical treatment method, when combined with chemotherapy improved the PFS and TTLP of patients with stage IV lung adenocarcinoma."( Microwave ablation combined with chemotherapy improved progression free survival of IV stage lung adenocarcinoma patients compared with chemotherapy alone.
Li, C; Shao, JB; Sun, ZG; Wang, J; Zhang, N; Zhu, LM, 2019
)
0.51
"Antiangiogenic agents combined with chemotherapy have efficacy in the treatment of unresectable malignant pleural mesothelioma (MPM)."( Phase II Trial of Cediranib in Combination With Cisplatin and Pemetrexed in Chemotherapy-Naïve Patients With Unresectable Malignant Pleural Mesothelioma (SWOG S0905).
Box-Noriega, B; Fossella, FV; Gadgeel, S; Gandara, DR; Heymach, JV; Hueftle, JG; Kelly, K; Lu, C; Miao, J; Redman, MW; Tsao, AS; Velasco, MR; Vogelzang, NJ; Wistuba, II, 2019
)
0.51
" 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
)
0.56
"To study the treatment of advanced lung adenocarcinoma in the elderly with pemetrexed combined with platinum drugs."( Study on the treatment of advanced lung adenocarcinoma in the elderly with pemetrexed combined with platinum drugs.
Ba, X; Bian, L; Han, J; Zhao, G, 2019
)
0.51
" This study aimed to investigate VEGFA messenger RNA expression in patients with EGFR mutation, and to further compare the efficacy of bevacizumab combined with platinum-based chemotherapy between EGFR-mutant and wild-type patients."( Potential Benefits of Bevacizumab Combined With Platinum-Based Chemotherapy in Advanced Non-Small-Cell Lung Cancer Patients With EGFR Mutation.
Gen, S; Hase, T; Hasegawa, Y; Hashimoto, N; Kodama, Y; Matsui, A; Miyazawa, A; Morise, M; Sato, M; Tamiya, Y; Tanaka, I, 2020
)
0.56
" The patients in MPC group were treated with microwave ablation (MWA) combined with PC while patients in MGC group were given MWA combined with gemcitabine plus cisplatin (GC)."( Comparative clinical study on microwave ablation combined with gemcitabine and cisplatin or combined with pemetrexed and cisplatin in treatment of advanced NSCLC.
Feng, Y; Wang, L; Wu, Y; Zhang, Y; Zhou, Y, 2020
)
0.56
"We utilized syngeneic mouse models of MPM and lung cancer and assessed the therapeutic effects of anti-PD-1 antibody and its combination with cisplatin (CDDP) and pemetrexed (PEM)."( Anti-PD-1 antibody combined with chemotherapy suppresses the growth of mesothelioma by reducing myeloid-derived suppressor cells.
Goto, H; Hanibuchi, M; Ishizawa, K; Kishuku, M; Koyama, K; Kozai, H; Mitsuhashi, A; Nishioka, Y; Ogawa, H; Ogino, H; Otsuka, K; Saijo, A; Tobiume, M; Yoneda, H, 2020
)
0.56
"For patients with advanced non-small-cell lung cancer and gradual progression who are EGFR-T790M mutation negative after initial EGFR-TKI therapy, EGFR-TKI combined with chemotherapy confers longer PFS and overall survival than sequential EGFR-TKI and chemotherapy does."( EGFR Tyrosine Kinase Inhibitor (TKI) Combined With Concurrent or Sequential Chemotherapy for Patients With Advanced Lung Cancer and Gradual Progression After First-Line EGFR-TKI Therapy: A Randomized Controlled Study.
Chang, Q; Chu, T; Han, B; Lou, Y; Lv, M; Qian, J; Qiang, H; Teng, J; Xu, J; Zhang, Y; Zhao, Y; Zhong, R, 2021
)
0.62
"This phase 2 study explored tislelizumab, an anti-PD-1 antibody, in combination with platinum-based chemotherapy as first-line treatment of advanced lung cancer."( A Phase 2 Study of Tislelizumab in Combination With Platinum-Based Chemotherapy as First-line Treatment for Advanced Lung Cancer in Chinese Patients.
Cheng, Y; Cui, J; Leaw, SJ; Liu, Z; Ma, X; Ma, Y; Ma, Z; Shu, Y; Tan, W; Wang, J; Wang, Z; Wu, Y; Zhang, Y; Zhao, J, 2020
)
0.56
" All patients received tislelizumab 200 mg in combination with 4-6 cycles of platinum-doublet."( A Phase 2 Study of Tislelizumab in Combination With Platinum-Based Chemotherapy as First-line Treatment for Advanced Lung Cancer in Chinese Patients.
Cheng, Y; Cui, J; Leaw, SJ; Liu, Z; Ma, X; Ma, Y; Ma, Z; Shu, Y; Tan, W; Wang, J; Wang, Z; Wu, Y; Zhang, Y; Zhao, J, 2020
)
0.56
"This is the first study to explore the efficacy and safety of osimertinib combined with platinum-based chemotherapy in previously untreated NSCLC patients with EGFR-sensitizing mutations."( A Phase II Study of Osimertinib Combined With Platinum Plus Pemetrexed in Patients With EGFR-Mutated Advanced Non-Small-cell Lung Cancer: The OPAL Study (NEJ032C/LOGIK1801).
Asahina, H; Isobe, T; Kagamu, H; Kikuchi, T; Kobayashi, K; Maemondo, M; Morita, S; Oizumi, S; Okamoto, I; Seike, M; Sugio, K; Takahashi, K; Tanaka, K, 2021
)
0.62
"This post hoc analysis assessed the safety of pemetrexed and platinum in combination with pembrolizumab, including time-to-onset and time-to-resolution of all-cause any-grade and grade ≥3 adverse events (AEs) and renal AEs."( Safety of pemetrexed plus platinum in combination with pembrolizumab for metastatic nonsquamous non-small cell lung cancer: A post hoc analysis of KEYNOTE-189.
Aerts, J; Gadgeel, SM; Garon, EB; Kim, JS; Muehlenbein, CE; Peterson, P; Rizzo, MT, 2021
)
0.62
" To assess the optimal choice of first-line and maintenance treatment regimens, we performed a meta-analysis of prospective randomized controlled clinical trials (RCTs) of patients with NS-NSCLC on bevacizumab combined with chemotherapy."( Clinical option of pemetrexed-based versus paclitaxel-based first-line chemotherapeutic regimens in combination with bevacizumab for advanced non-squamous non-small-cell lung cancer and optimal maintenance therapy: evidence from a meta-analysis of randomi
Cao, R; Guo, YJ; Han, CB; Huang, LT; Jing, W; Ma, J; Song, J; Sun, L; Wang, YR; Zhang, SL; Zhang, XY; Zhao, JZ, 2021
)
0.62
"When the patients of advanced non-squamous non-small cell lung cancer (NSCLC) have achieved remission by induction therapy, it is controversial that combination with bevacizumab is used as maintenance therapy."( Maintenance treatment of combination with bevacizumab vs single agent for advanced non-squamous non-small cell lung cancer: A systematic review and meta-analysis.
Hong, L; Kong, Y; Xu, J; Xu, X, 2021
)
0.62
" The application of combination with bevacizumab, pemetrexed was studied in clinical trials of maintenance treatment for advanced NSCLC."( Maintenance treatment of combination with bevacizumab vs single agent for advanced non-squamous non-small cell lung cancer: A systematic review and meta-analysis.
Hong, L; Kong, Y; Xu, J; Xu, X, 2021
)
0.62
" Compared with bevacizumab and pemetrexed, PFS of combination with bevacizumab was significantly improved (hazard ratio [HR] = 0."( Maintenance treatment of combination with bevacizumab vs single agent for advanced non-squamous non-small cell lung cancer: A systematic review and meta-analysis.
Hong, L; Kong, Y; Xu, J; Xu, X, 2021
)
0.62
"PFS was significantly improved in patients with advanced non-squamous NSCLC who use bevacizumab combination with single-agent as maintenance treatment, but it does not translate into the advantages of OS; compared with bevacizumab, no PFS and OS benefits were found."( Maintenance treatment of combination with bevacizumab vs single agent for advanced non-squamous non-small cell lung cancer: A systematic review and meta-analysis.
Hong, L; Kong, Y; Xu, J; Xu, X, 2021
)
0.62
" This study aimed to explore the real-world use of anti-diabetic agent metformin in combination with pemetrexed-based platinum doublets in a first-line setting."( Benefits of Metformin Combined with Pemetrexed-Based Platinum Doublets as a First-Line Therapy for Advanced Lung Adenocarcinoma Patients with Diabetes.
Chang, GR; Chen, CM; Chen, W; Chong, KY; Cidem, A; Lin, CH; Staniczek, T; Tsai, YT; Wang, JL; Yen, CC, 2021
)
0.62
" However, the clinical significance of immune checkpoint inhibitors combined with chemotherapy in elderly patients with NSCLC has not yet been fully understood."( Clinical impact of pembrolizumab combined with chemotherapy in elderly patients with advanced non-small-cell lung cancer.
Chihara, Y; Goto, Y; Hibino, M; Hiranuma, O; Iwasaku, M; Kaneko, Y; Kijima, T; Morimoto, K; Morimoto, Y; Nakao, A; Takayama, K; Takeda, T; Takeshita, M; Takumi, C; Uchino, J; Yamada, T; Yamaguchi, H; Yokoi, T, 2021
)
0.62
"Pemetrexed (800 mg day 1), cis-platinum (40 mg day 1-3) combined with bevacizumab (400 mg day 1) every 3 weeks were administered to the patient."( Erlotinib combined with bevacizumab and chemotherapy in first line osimertinib-resistant NSCLC patient with leptomeningeal metastasis: A case report.
Li, M; Luo, N; Qi, Y; Wang, M; Zhu, F, 2021
)
0.62
"Our prospective, open-label, single-arm phase II study investigated the safety and efficacy of DCVAC/LuCa (dendritic cell vaccines for lung cancer) combined with standard carboplatin/pemetrexed in advanced non-squamous (nsq) non-small-cell lung cancer (NSCLC)."( Safety and efficacy of dendritic cell-based immunotherapy (DCVAC/LuCa) combined with carboplatin/pemetrexed for patients with advanced non-squamous non-small-cell lung cancer without oncogenic drivers.
Cao, S; Han, B; Ling, X; Wang, H; Xu, J; Zhang, B; Zhang, X; Zhong, H; Zhong, R, 2022
)
0.72
"We aimed to study the activity of chemotherapy in combination with pembrolizumab as first-line treatment in patients with stage IV ERBB2-mutated NSCLC."( Pembrolizumab in Combination with Chemotherapy in Patients with ERBB2-Mutated Non-Small Cell Lung Cancer.
Abu Rous, F; Gadgeel, S; Gutta, R; Halmos, B; Li, P, 2022
)
0.72
" We hypothesize that Src kinase inhibitors, including Bosutinib, may exhibit clinical synergy in combination with the antifolate drug pemetrexed."( A Phase I Study of the Non-Receptor Kinase Inhibitor Bosutinib in Combination with Pemetrexed in Patients with Selected Metastatic Solid Tumors.
Karim, NA; Khaled, A; Morris, JC; Patel, N; Pulliam, S; Shoukier, M; Ullah, A; Wang, H, 2022
)
0.72
" Chemotherapy of pemetrexed and cisplatin combined with antiangiogenic therapy of bevacizumab is recommended as the first-line regimen by guidelines."( Tislelizumab combined with anlotinib in the second-line treatment of malignant pleural mesothelioma.
Guo, W; Huang, X; Liang, J; Lv, Y; Zhang, D, 2022
)
0.72
"He received firstly pemetrexed combined with platinum and bevacizumab, which barely curbed disease progression; When the first line treatment failed, he was switched to tislelizumab combined with anlotinib."( Tislelizumab combined with anlotinib in the second-line treatment of malignant pleural mesothelioma.
Guo, W; Huang, X; Liang, J; Lv, Y; Zhang, D, 2022
)
0.72
"Tislelizumab combined with anlotinib significantly relieved his clinical symptoms, and imaging examination further validated the improvement."( Tislelizumab combined with anlotinib in the second-line treatment of malignant pleural mesothelioma.
Guo, W; Huang, X; Liang, J; Lv, Y; Zhang, D, 2022
)
0.72
"The case firstly demonstrated the efficacy of tislelizumab combined with anlotinib in the second-line management of MPM."( Tislelizumab combined with anlotinib in the second-line treatment of malignant pleural mesothelioma.
Guo, W; Huang, X; Liang, J; Lv, Y; Zhang, D, 2022
)
0.72
" Patients in each group were randomly treated with EGFR-tyrosine kinase inhibitor (TKI) monotherapy or EGFR-TKI combined with pemetrexed in a ratio of 1:1."( EGFR-TKI Combined with Pemetrexed versus EGFR-TKI Monotherapy in Advanced EGFR-mutated NSCLC: A Prospective, Randomized, Exploratory Study.
Feng, W; Gu, W; He, M; Li, M; Li, Z; Liang, J; Lu, Y; Shi, X; Wang, F; Yang, S; Ye, Z; You, D; Zhang, H, 2023
)
0.91
" There were no significant differences in PFS between patients with and without concomitant and between patients received TKI monotherapy and TKI combined with pemetrexed (p=0."( EGFR-TKI Combined with Pemetrexed versus EGFR-TKI Monotherapy in Advanced EGFR-mutated NSCLC: A Prospective, Randomized, Exploratory Study.
Feng, W; Gu, W; He, M; Li, M; Li, Z; Liang, J; Lu, Y; Shi, X; Wang, F; Yang, S; Ye, Z; You, D; Zhang, H, 2023
)
0.91
"Chemotherapy, in combination with immune checkpoint blockade (ICB) targeting to programmed death-1 (PD-1) or its ligand PD-L1, is one of the first-line treatments for patients with advanced non-small-cell lung cancer (NSCLC)."( Pemetrexed combined with dual immune checkpoint blockade enhances cytotoxic T lymphocytes against lung cancer.
Chen, MC; Chiu, SC; Cho, DY; Huang, SW; Hung, MY; Jan, CI; Li, YH; Pan, CM, 2023
)
0.91
"The Atezo-Brain study evaluated atezolizumab combined with chemotherapy in patients with advanced non-small-cell lung cancer (NSCLC) with untreated brain metastases, a population traditionally excluded from trials."( Phase II Trial of Atezolizumab Combined With Carboplatin and Pemetrexed for Patients With Advanced Nonsquamous Non-Small-Cell Lung Cancer With Untreated Brain Metastases (Atezo-Brain, GECP17/05).
Blasco, A; Bruna, J; De Castro, J; Estival, A; Felip, E; Guirado, M; Huidobro, G; Juan, O; López, R; Massutí, B; Mosquera, J; Nadal, E; Navarro, V; Pereira, E; Rodríguez-Abreu, D; Simó, M; Sullivan, I; Vilariño, N, 2023
)
0.91

Bioavailability

ExcerptReferenceRelevance
"The LUX-Lung 3 study investigated the efficacy of chemotherapy compared with afatinib, a selective, orally bioavailable ErbB family blocker that irreversibly blocks signaling from epidermal growth factor receptor (EGFR/ErbB1), human epidermal growth factor receptor 2 (HER2/ErbB2), and ErbB4 and has wide-spectrum preclinical activity against EGFR mutations."( Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations.
Bennouna, J; Boyer, M; Geater, SL; Gorbunova, V; Hirsh, V; Kato, T; Lee, KH; Massey, D; Mok, T; O'Byrne, K; Orlov, S; Schuler, M; Sequist, LV; Shah, R; Shahidi, M; Su, WC; Tsai, CM; Yamamoto, N; Yang, JC; Zazulina, V, 2013
)
0.39
" In mice xenografted with NCI-H2228 cells expressing EML4-ALK, orally administered ASP3026 was well absorbed in tumor tissues, reaching concentrations >10-fold higher than those in plasma, and induced tumor regression with a wide therapeutic margin between efficacious and toxic doses."( The selective anaplastic lymphoma receptor tyrosine kinase inhibitor ASP3026 induces tumor regression and prolongs survival in non-small cell lung cancer model mice.
Doihara, H; Furutani, T; Fushiki, H; Konagai, S; Kondoh, Y; Kudoh, M; Kuromitsu, S; Mori, K; Mori, M; Saito, R; Sakagami, H; Shimada, I; Shindou, N; Soga, T; Ueno, Y, 2014
)
0.4
" However, effective delivery of drugs in combination at the tumor site is marred by low bioavailability and systemic toxicity of individual drugs."( Synergistic activity of combination therapy with PEGylated pemetrexed and gemcitabine for an effective cancer treatment.
Sahoo, SK; Vandana, M, 2015
)
0.42
" The oral administration of drug complex in rats revealed high bioavailability (22."( Oral pemetrexed facilitates low-dose metronomic therapy and enhances antitumor efficacy in lung cancer.
Alam, F; Byun, Y; Choi, JU; Jeon, OC; Lee, DS; Lee, S; Maharjan, R; Mahmud, F; Park, J; Park, JW, 2018
)
0.48
" Therefore, the oral bioavailability of HP-beta-CD/PMX/DCK/P188-NE in rats was evaluated as 26."( Enhanced oral absorption of pemetrexed by ion-pairing complex formation with deoxycholic acid derivative and multiple nanoemulsion formulations: preparation, characterization, and in vivo oral bioavailability and anticancer effect.
Byun, Y; Choi, JU; Pangeni, R; Panthi, VK; Park, JW, 2018
)
0.48
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
" This resulted in a 282% improvement in oral bioavailability in rats."( Metronomic delivery of orally available pemetrexed-incorporated colloidal dispersions for boosting tumor-specific immunity.
Byun, Y; Chang, KY; Choi, JU; Jha, SK; Kang, SH; Maharjan, R; Pangeni, R; Park, JW; Subedi, L, 2021
)
0.62

Dosage Studied

One-month toxicity study confirmed that daily dosing of oral pemetrexed is safe by investigating apoptosis in the gut tissues from mice. Homocysteine blood concentration just before the first cycle dosage was significantly elevated relative to the 2-3 cycle.

ExcerptRelevanceReference
" The 3-weekly dosing schedule was chosen for phase II evaluation."( Clinical studies with MTA.
Calvert, AH; Walling, JM, 1998
)
0.3
" The every three week dosing schedule has proven to be convenient and easy to administer and the clinical toxicities of LY231514 seem to be well tolerated."( Multiple folate enzyme inhibition: mechanism of a novel pyrrolopyrimidine-based antifolate LY231514 (MTA).
Chen, VJ; Habeck, LL; Mendelsohn, LG; Schultz, RM; Shih, C, 1998
)
0.3
" Three dosing schedules have been investigated in the phase I setting: daily x5 every 21 days, weekly x4 every 42 days, and once every 21 days."( Overview of phase I trials of multitargeted antifolate (MTA, LY231514).
Rinaldi, DA, 1999
)
0.3
" The present data suggest the potential clinical efficacy of combining Alimta administration with radiotherapy for Alimta-sensitive cells and indicate that further testing needs to be conducted to optimize the dosing schedule to enhance the interaction between the therapeutic agents."( Treatment of head and neck and esophageal xenografts employing Alimta and concurrent ionizing radiation.
Mauceri, HJ; Salloum, RM; Seetharam, S; Vokes, EE; Weichselbaum, RR, 2001
)
0.31
" The marked improvement in toxicity and tolerance with vitamin supplementation suggests the need to reexamine optimal dosing in pemetrexed combination schedules."( Pemetrexed in advanced colorectal cancer.
de Gramont, A; Louvet, C, 2004
)
0.32
" Ongoing and future studies will establish optimal dosing regimens of pemetrexed and potential benefits of vitamin supplementation in the settings of metastatic breast cancer and gynecologic malignancies."( Phase II studies of pemetrexed in metastatic breast and gynecologic cancers.
Smith, I, 2004
)
0.32
" Additional studies are needed to define appropriate dosing for renally impaired patients receiving higher dose pemetrexed with vitamin supplementation."( Phase I and pharmacokinetic study of pemetrexed administered every 3 weeks to advanced cancer patients with normal and impaired renal function.
Baker, SD; Chaudhary, AK; Chaudhuri, T; Goetz, A; Hammond, LA; Johnson, RD; Latz, JE; Mita, AC; Molpus, K; Patnaik, A; Rowinsky, EK; Sandler, A; Simms, L; Sweeney, CJ; Takimoto, CH; Tolcher, AW; Villalona-Calero, M, 2006
)
0.33
"Pemetrexed (500 mg/m(2)) with vitamin supplementation is well tolerated and requires no dosage adjustment when coadministered with aspirin (in patients with CrCl > or =60 mL/min) or ibuprofen (in patients with CrCl > or =80 mL/min)."( Two drug interaction studies evaluating the pharmacokinetics and toxicity of pemetrexed when coadministered with aspirin or Ibuprofen in patients with advanced cancer.
Baker, SD; Battiato, L; Chaudhary, AK; Chaudhuri, T; Cleverly, A; Fife, K; Krull, JH; Latz, JE; Mita, AC; Murry, DJ; Rowinsky, EK; Sandler, A; Sweeney, CJ; Takimoto, CH, 2006
)
0.33
" Finally, dosing and administration are reviewed, including appropriate premedication."( Pemetrexed, a novel antifolate therapeutic alternative for cancer chemotherapy.
Shah, SR; Stanford, BL; Villela, LR, 2006
)
0.33
" No clear dose-response relationship could be established."( Use of pemetrexed in breast cancer.
Dittrich, C, 2006
)
0.33
" Pemetrexed dosing was 500 mg/m and carboplatin was AUC (area under the curve) 5 once every 3 weeks."( Pemetrexed as second-line treatment in malignant pleural mesothelioma after platinum-based first-line treatment.
Perell, K; Sundstrøm, S; Sørensen, JB; Thielsen, AK, 2007
)
0.34
"Chemonaive patients with stage IIIB/IV NSCLC and an Eastern Cooperative Oncology Group performance status of 0 to 2 received either 500 mg/m2 of pemetrexed (day 1, every 3 weeks) for eight cycles, or the same dosage of pemetrexed for cycles 1 and 2 and then 1200 mg/m2 of gemcitabine (days 1 and 8, every 3 weeks) for cycles 3 and 4 (repeated once for a total of eight cycles)."( Single-agent pemetrexed or sequential pemetrexed/gemcitabine as front-line treatment of advanced non-small cell lung cancer in elderly patients or patients ineligible for platinum-based chemotherapy: a multicenter, randomized, phase II trial.
Caffo, O; Favaretto, A; Gregorc, V; Gridelli, C; Kaukel, E; Manegold, C; Martoni, A; Migliorino, MR; Müller, TR; Muñoz, M; Peterson, P; Reck, M; Rossi, A; Russo, F; Schmittel, A, 2007
)
0.34
" At level 1, 4/6 patients experienced DLTs; dosing decreased to level 0 and 4/5 patients experienced DLTs."( Phase I study of a 3-drug regimen of gemcitabine/cisplatin/pemetrexed in patients with metastatic transitional cell carcinoma of the urothelium.
Atienza, D; Awasthi, S; Berry, W; Delaune, R; Deutsch, M; Dien, PY; Gregory, TF; Hood, K; Hutson, TE; Ilegbodu, D; Kolodziej, MJ; Mull, S; Muscato, JJ; Nicol, S; Raju, RN; Ruxer, RL; Vukelja, S, 2008
)
0.35
"This phase III randomized trial compared pemetrexed 500 mg/m(2) (P500) with pemetrexed 900 mg/m(2) (P900) to determine whether higher dosing benefits non-small-cell lung cancer (NSCLC) patients as second-line therapy."( A randomized phase III trial comparing standard and high-dose pemetrexed as second-line treatment in patients with locally advanced or metastatic non-small-cell lung cancer.
Cullen, MH; Fischer, JR; Iscoe, N; Joy, AA; Novello, S; Peterson, P; Sörenson, S; Visseren-Grul, CM; Zatloukal, P; Zereu, M, 2008
)
0.35
"The Pem/Cis combination provides promising activity and an acceptable safety profile for chemonaive Japanese MPM patients with the same recommend dosage and schedule used in rest of the world."( Efficacy and safety of pemetrexed in combination with cisplatin for malignant pleural mesothelioma: a phase I/II study in Japanese patients.
Adachi, S; Fukuoka, M; Gemba, K; Hida, T; Ichinose, Y; Kunitoh, H; Nakagawa, K; Nambu, Y; Saijo, N; Shinkai, T; Yamazaki, K, 2008
)
0.35
" We defined each agent's clinical potency index (CPI) as the AUC achieved with standard pediatric dosing regimens divided by the in vitro IC50."( Clinical potency of methotrexate, aminopterin, talotrexin and pemetrexed in childhood leukemias.
Adamson, PC; Norris, RE, 2010
)
0.36
" This review summarizes the background, scientific rationale and early clinical data in support of intercalation of intermittent erlotinib dosing with pemetrexed as a means of achieving pharmacodynamic separation."( Intercalation of erlotinib and pemetrexed in the treatment of non-small cell lung cancer.
Gandara, DR; Lara, PN; Li, T; Mack, PC; Perez-Soler, R, 2010
)
0.36
" Hematologic toxicities limiting day 8 gemcitabine dosing were observed in the first 20 patients, prompting a protocol amendment to evaluate pemetrexed 500 mg/m2 followed by gemcitabine 1500 mg/m2 on day 1 of a 14-day cycle."( A phase II trial of pemetrexed and gemcitabine in patients with metastatic breast cancer who have received prior taxane therapy.
Elias, AD; Gralow, J; Muscato, J; Neubauer, M; O'Shaughnessy, JA; Orlando, M; Pippen, J; Shonukan, O; Stokoe, C; Vaughn, LG; Wang, Y, 2010
)
0.36
" The enzastaurin dosing regimen (QD or BID) had no effect on pemetrexed pharmacokinetics."( A phase I study of enzastaurin combined with pemetrexed in advanced non-small cell lung cancer.
Baldwin, JR; Koshiji, M; Kunitoh, H; Murakami, H; Nakamura, Y; Nokihara, H; Ohe, Y; Shukuya, T; Takahashi, T; Tamura, T; Tanai, C; Yamamoto, N, 2010
)
0.36
"Sunitinib was administered once daily on a continuous daily dosing (CDD) schedule (37."( Feasibility study of two schedules of sunitinib in combination with pemetrexed in patients with advanced solid tumors.
Fumita, S; Hashimoto, J; Ichikawa, Y; Kimura, N; Miyazaki, M; Nakagawa, K; Ohki, E; Okamoto, I; Shimizu, T; Takeda, M; Terashima, M; Tsurutani, J, 2012
)
0.38
" Treatment durations and dosing were derived from randomized controlled trials, FDA labeling, and National Comprehensive Cancer Network guidelines."( Budget impact of erlotinib for maintenance therapy in advanced non-small cell lung cancer.
Carlson, JJ; Reyes, C; Veenstra, DL; Wong, WB, 2011
)
0.37
"Using a 3 + 3 dose-escalation design, patients received oral sunitinib qd by continuous daily dosing (CDD schedule; 37."( A phase I dose-escalation and pharmacokinetic study of sunitinib in combination with pemetrexed in patients with advanced solid malignancies, with an expanded cohort in non-small cell lung cancer.
Blais, N; Camidge, DR; Canil, C; Chao, RC; Chow, LQ; Diab, SG; Jonker, DJ; Laurie, SA; McWilliam, M; Ruiz-Garcia, A; Thall, A; Tye, L; Zhang, K, 2012
)
0.38
" The antitumor activity, different dosing sequences, and dosing regimens of TH-302 in combination with commonly used conventional chemotherapeutics were investigated in human tumor xenograft models."( TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules.
Ahluwalia, D; Ammons, WS; Baker, AF; Cranmer, LD; Curd, JG; Duan, JX; Ferraro, D; Hart, CP; Liu, Q; Matteucci, MD; Sun, JD; Wang, J; Wang, Y, 2012
)
0.38
" Simultaneous administration of TH-302 and chemotherapeutics increased toxicity versus schedules with dosing separations."( TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules.
Ahluwalia, D; Ammons, WS; Baker, AF; Cranmer, LD; Curd, JG; Duan, JX; Ferraro, D; Hart, CP; Liu, Q; Matteucci, MD; Sun, JD; Wang, J; Wang, Y, 2012
)
0.38
" Both groups had one patient who reduced dosage because of myelosuppression (grade IV)."( [Effects of EGFR-TKIs on sequential pemetrexed
for advanced pulmonary adenocarcinoma].
Gao, Z; Han, B; Jiang, L; Jiang, Y; Liu, Y; Wang, X, 2012
)
0.38
" One patient dosed at 4 mg kg(-1) experienced grade 3 hypophosphatemia (dose-limiting toxicity; DLT), which prompted a cohort expansion."( A phase I dose-escalation study of aflibercept administered in combination with pemetrexed and cisplatin in patients with advanced solid tumours.
Colevas, AD; Diaz-Padilla, I; Gao, B; Lawson, EB; Leighl, NB; Lisano, J; Liu, L; Neal, JW; Razak, AR; San Pedro-Salcedo, M; Shepherd, FA; Siu, LL; Thibault, A; Wakelee, HA, 2012
)
0.38
" Pemetrexed was given at the dosage of 500 mg/m(2) on day 1, with folic acid and vitamin B12 supplementation, each cycle repeated every 3 weeks."( Open-label, single-arm phase II study of pemetrexed in the treatment of patients with recurrent or metastatic nasopharyngeal carcinoma who have had prior platinum-based chemotherapy.
Huang, P; Huang, Y; Wang, F; Wu, J; Zhang, L; Zhang, Y; Zhao, L, 2012
)
0.38
"5 or 50 mg) qd on a continuous daily dosing (CDD) schedule or Schedule 2/1 (2 weeks on, 1 week off treatment) plus pemetrexed (400 or 500 mg/m(2) IV) and cisplatin (75 mg/m(2) IV) q3w up to 6 cycles."( Sunitinib combined with pemetrexed and cisplatin: results of a phase I dose-escalation and pharmacokinetic study in patients with advanced solid malignancies, with an expanded cohort in non-small cell lung cancer and mesothelioma.
Blais, N; Camidge, DR; Chao, RC; Chow, LQ; Doebele, RC; Jonker, DJ; Laurie, SA; Ruiz-Garcia, A; Soulières, D; Thall, A; Zhang, K, 2013
)
0.39
" Double dosage of gefitinib (500 mg per day) together with pemetrexed were given as the second-line therapy after the patient developed new brain lesions and leptomeningeal metastasis during the maintenance therapy of gefitinib."( Activity of pemetrexed and high-dose gefitinib in an EGFR-mutated lung adenocarcinoma with brain and leptomeningeal metastasis after response to gefitinib.
Fang, X; Hu, Y; Li, M; Ma, S; Shen, H; Tan, C; Yuan, Y, 2012
)
0.38
" However, the dosage and period of medication was not fully verified."( [Effectiveness of steroids for the rash side effect of pemetrexed].
Ishikawa, H; Kobayashi, R; Ohashi, Y; Onishi, T; Shino, M; Suzuki, K; Yamamoto, N, 2013
)
0.39
" Decreasing the price or adjusting the dosage of pemetrexed may be a better option for meeting the treatment demands of Chinese patients."( Cost-effectiveness of continuation maintenance pemetrexed after cisplatin and pemetrexed chemotherapy for advanced nonsquamous non-small-cell lung cancer: estimates from the perspective of the Chinese health care system.
Chen, G; Li, J; Ouyang, L; Peng, L; Tan, C; Wan, X; Wang, S; Zeng, X; Zhao, Z, 2013
)
0.39
"In this phase I dose-escalation study, patients received oral sunitinib on a continuous daily dosing (CDD) schedule (37."( Sunitinib combined with pemetrexed and carboplatin in patients with advanced solid malignancies--results of a phase I dose-escalation study.
Blais, N; Camidge, DR; Chao, RC; Chow, LQ; Diab, SG; Jonker, DJ; Laurie, SA; Ruiz-Garcia, A; Soulières, D; Thall, A; Zhang, K, 2013
)
0.39
" Pulsed dosing conferred no apparent safety or dose advantage."( A phase I, dose-escalation trial of continuous- and pulsed-dose afatinib combined with pemetrexed in patients with advanced solid tumors.
Chand, VK; Chu, QS; Hirte, HW; Hotte, SJ; Sangha, R; Schnell, D; Sergenson, G, 2014
)
0.4
"During phase Ib, E+P was tolerated only at a markedly lower dosing intensity relative to the eribulin monotherapy regimen approved for breast cancer and used in phase II studies of NSCLC."( An open-label, multicenter, randomized phase Ib/II study of eribulin mesylate administered in combination with pemetrexed versus pemetrexed alone as second-line therapy in patients with advanced nonsquamous non-small-cell lung cancer.
Bondarenko, I; Dave, H; Freeman, A; Hodge, JP; Huber, B; Lieberman, R; Shelton, MJ; Shparyk, Y; Vynnychenko, I; Waller, CF, 2015
)
0.42
" Targeted therapies do not require any adjustment of the dosage in case of moderate or severe renal insufficiency but adapting the doses of biphosphonates to renal function is necessary."( [Impact of lung cancer treatments on renal function].
Belaiche, S; Couraud, S; Moro-Sibilot, D; Sakhri, L; Toffart, AC, 2014
)
0.4
" In addition, the homocysteine blood concentration just before the first cycle dosage of pemetrexed was significantly elevated relative to the 2-3 cycle."( Phase II study of oral vitamin B12 supplementation as an alternative to intramuscular injection for patients with non-small cell lung cancer undergoing pemetrexed therapy.
Hosomi, Y; Nagamata, M; Nakahara, Y; Okamura, T; Okuma, Y; Shimokawa, T; Sunami, K; Takagi, Y; Takahashi, S; Watanabe, K; Yomota, M, 2016
)
0.43
"2% completed their planned dosage within a 9-week feasibility time frame."( Reduced folate and serum vitamin metabolites in patients with rectal carcinoma: an open-label feasibility study of pemetrexed with folic acid and vitamin B12 supplementation.
Björkqvist, HG; Carlsson, GU; Gustavsson, BG; Kurlberg, GK; Odin, EA; Stoffregen, CC; Tångefjord, MT, 2016
)
0.43
" Because dosing delays and modifications were associated with the MTD, the recommended phase II dose was declared to be pemetrexed 500 mg/m2 every 14 days with oral sorafenib 400 mg given twice daily on days 1-5."( Phase I study of pemetrexed with sorafenib in advanced solid tumors.
Booth, L; Bose, P; Dent, P; Geyer, CE; Gordon, S; Kmieciak, M; Massey, HD; McGuire, WP; Moran, RG; Poklepovic, A; Quigley, M; Roberts, JD; Shafer, DA; Shrader, E; Strickler, K; Tombes, MB; Wan, W, 2016
)
0.43
" Cohort B involved a modified schedule of sorafenib dosing on days 1-5 of each 14-day pemetrexed cycle."( Phase I study of pemetrexed with sorafenib in advanced solid tumors.
Booth, L; Bose, P; Dent, P; Geyer, CE; Gordon, S; Kmieciak, M; Massey, HD; McGuire, WP; Moran, RG; Poklepovic, A; Quigley, M; Roberts, JD; Shafer, DA; Shrader, E; Strickler, K; Tombes, MB; Wan, W, 2016
)
0.43
" Pemetrexed was added to vinflunine dosed at 280 mg/m2 on day 1 of a 21-day cycle."( Combined treatment with pemetrexed and vinflunine in patients with metastatic urothelial cell carcinoma after prior platinum-containing chemotherapy - results of an exploratory phase I study.
Agerbæk, M; Pappot, H; Ullén, A; von der Maase, H, 2018
)
0.48
"This study has identified a truncated dosing regimen and recommended phase II dose of demcizumab (5 mg/kg q3-weekly ×4) for subsequent clinical evaluation in combination with standard carboplatin and pemetrexed chemotherapy."( Phase IB Trial of the Anti-Cancer Stem Cell DLL4-Binding Agent Demcizumab with Pemetrexed and Carboplatin as First-Line Treatment of Metastatic Non-Squamous NSCLC.
Harris, DL; Hidalgo, M; Hughes, BGM; Jameson, MB; Kapoun, AM; Kotasek, D; Markman, B; McKeage, MJ; Millward, MJ; Stagg, RJ; Xu, L, 2018
)
0.48
" Furthermore, the combination index and dose reduction index values indicated that the cSBL + pemetrexed combination showed the highest synergism, and thus potential for reducing dosage of each drug, compared with the other combinations, including the existing pemetrexed + cisplatin regimen."( Sialic acid-binding lectin from bullfrog eggs inhibits human malignant mesothelioma cell growth in vitro and in vivo.
Hara, A; Hosono, M; Satoh, T; Sugawara, S; Tatsuta, T, 2018
)
0.48
" One-month toxicity study confirmed that daily dosing of oral pemetrexed is safe by investigating apoptosis in the gut tissues from mice."( Oral pemetrexed facilitates low-dose metronomic therapy and enhances antitumor efficacy in lung cancer.
Alam, F; Byun, Y; Choi, JU; Jeon, OC; Lee, DS; Lee, S; Maharjan, R; Mahmud, F; Park, J; Park, JW, 2018
)
0.48
" In patients administered maintenance treatment, rh-endostatin plus pemetrexed was associated with prolonged PFS compared to single-agent pemetrexed when PFS was calculated from first dosing (13."( Efficacy and safety of rh-endostatin (Endostar) combined with pemetrexed/cisplatin followed by rh-endostatin plus pemetrexed maintenance in non-small cell lung cancer: A retrospective comparison with standard chemotherapy.
He, X; Jin, J; Pi, J; Shi, Y; Zhou, S; Zuo, L, 2018
)
0.48
" Dosing is based on body surface are (BSA), while renal function is the only determinant for exposure and thus toxicity."( Pharmacokinetically-guided dosing of pemetrexed in a patient with renal impairment and a patient requiring hemodialysis.
Agterhuis, DE; Burger, DM; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AM; Posthuma, R; Schoenmaekers, JJAO; Ter Heine, R, 2019
)
0.51
"Altogether, we showed that pharmacokinetically-guided dosing of pemetrexed may be a feasible strategy for patients with lung cancer and renal impairment."( Pharmacokinetically-guided dosing of pemetrexed in a patient with renal impairment and a patient requiring hemodialysis.
Agterhuis, DE; Burger, DM; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AM; Posthuma, R; Schoenmaekers, JJAO; Ter Heine, R, 2019
)
0.51
" Results 36 patients were dosed at 10, 15, and 20 mg/kg doses of GSK3052230."( A phase Ib study of GSK3052230, an FGF ligand trap in combination with pemetrexed and cisplatin in patients with malignant pleural mesothelioma.
Baker-Neblett, K; Bellovin, DI; DeYoung, MP; Dómine, M; Fennell, DA; Gadgeel, S; Kindler, HL; Kostorov, V; Levchenko, E; López, PG; Mitrica, I; Morgensztern, D; Orlov, S; Schellens, JHM; Trigo, J; van Brummelen, EMJ; Vansteenkiste, JF; Vasquez, J; Viteri, S; Wang, X; Yan, L; Zauderer, MG, 2020
)
0.56
" Intrathecal chemotherapy is the mainstay of treatment for NSCLC patients with LM, but the optimal drug, administration route and mode, and dosage remain unclear."( [Use of Ommaya Reservoirs to Deliver Pemetrexed in Leptomeningeal Metastasis from Non-small Cell Lung Cancer: A Case Report and Review of the Literature].
Guo, A; Huang, M; Li, H; Lin, Y; Yin, Z, 2019
)
0.51
" Although dosing is based on body surface area (BSA), large interindividual variability in pemetrexed plasma concentrations is observed."( A limited sampling schedule to estimate individual pharmacokinetics of pemetrexed in patients with varying renal functions.
Aerts, JGJV; Burger, DM; de Rouw, N; Derijks, HJ; Koolen, SLW; Ter Heine, R; van den Heuvel, MM; Visser, S, 2020
)
0.56
" Binimetinib 30 mg BID (dose level 1 [DL1]) or 45 mg BID (dose level 2 [DL2]) was given with standard doses of carboplatin and pemetrexed using an intermittent dosing schedule."( A phase I study of binimetinib (MEK 162), a MEK inhibitor, plus carboplatin and pemetrexed chemotherapy in non-squamous non-small cell lung cancer.
Albaba, H; Arif, S; Bradbury, PA; Chan, M; Chen, EX; Effendi, S; Fung, AS; Gill, S; Graham, DM; Law, JH; Le, LW; Leighl, NB; Pisters, KM; Rothenstein, J; Sawczak, M; Stockley, TL; Trinkaus, M; Zawisza, D; Zhang, T; Zurawska, U, 2021
)
0.62
"In this study, we developed oral pemetrexed (PMX) for metronomic dosing to enhance antitumor immunity."( Metronomic delivery of orally available pemetrexed-incorporated colloidal dispersions for boosting tumor-specific immunity.
Byun, Y; Chang, KY; Choi, JU; Jha, SK; Kang, SH; Maharjan, R; Pangeni, R; Park, JW; Subedi, L, 2021
)
0.62
" We postulate that individualized dosing reduces the incidence of neutropenia."( The Pharmacoeconomic Benefits of Pemetrexed Dose Individualization in Patients With Lung Cancer.
Boosman, RJ; Burger, DM; de Boer, M; de Rouw, N; Derijks, HJ; Frederix, GWJ; Lieverse, JE; Ter Heine, R; van den Heuvel, MM, 2022
)
0.72
" The use of a low test dosing of cytotoxic drugs may aid in dose individualization without causing harm."( Prediction of the pharmacokinetics of pemetrexed with a low test dose: A proof-of-concept study.
Boosman, RJ; Burgers, JA; de Rouw, N; Huitema, ADR; Ter Heine, R, 2023
)
0.91
"We show that test dosing of pemetrexed is feasible, but there seems no added value for a low test dosing in the dose individualization of pemetrexed."( Prediction of the pharmacokinetics of pemetrexed with a low test dose: A proof-of-concept study.
Boosman, RJ; Burgers, JA; de Rouw, N; Huitema, ADR; Ter Heine, R, 2023
)
0.91
" Optimized dosing of pemetrexed based on renal function instead of body surface area (BSA) is hypothesized to reduce pharmacokinetic variability in systemic exposure and could therefore improve treatment outcomes."( Renal function-based versus standard dosing of pemetrexed: a randomized controlled trial.
Biesma, B; Boosman, RJ; Burger, DM; Burgers, JA; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AC; Dumoulin, DW; Hendriks, LEL; Huitema, ADR; Mathijssen, RHJ; Piet, B; Pruis, MA; Ter Heine, R; van den Heuvel, MM, 2023
)
0.91
"A multicenter randomized (1:1) controlled trial was performed to assess superiority of optimized dosing versus BSA-based dosing in patients who were eligible for pemetrexed-based chemotherapy."( Renal function-based versus standard dosing of pemetrexed: a randomized controlled trial.
Biesma, B; Boosman, RJ; Burger, DM; Burgers, JA; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AC; Dumoulin, DW; Hendriks, LEL; Huitema, ADR; Mathijssen, RHJ; Piet, B; Pruis, MA; Ter Heine, R; van den Heuvel, MM, 2023
)
0.91
" The AUC of pemetrexed was similar between the optimized dosing arm (n = 37) and the standard of care arm (n = 44) (155 mg × h/L vs 160 mg × h/L (p = 0."( Renal function-based versus standard dosing of pemetrexed: a randomized controlled trial.
Biesma, B; Boosman, RJ; Burger, DM; Burgers, JA; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AC; Dumoulin, DW; Hendriks, LEL; Huitema, ADR; Mathijssen, RHJ; Piet, B; Pruis, MA; Ter Heine, R; van den Heuvel, MM, 2023
)
0.91
"We could not show superiority of optimized dosing of pemetrexed in patients with an adequate renal function does not show added value on the attainment of a pharmacokinetic endpoint, safety, nor QoL compared to standard of care dosing."( Renal function-based versus standard dosing of pemetrexed: a randomized controlled trial.
Biesma, B; Boosman, RJ; Burger, DM; Burgers, JA; Croes, S; de Rouw, N; Derijks, HJ; Dingemans, AC; Dumoulin, DW; Hendriks, LEL; Huitema, ADR; Mathijssen, RHJ; Piet, B; Pruis, MA; Ter Heine, R; van den Heuvel, MM, 2023
)
0.91
" Timely and individualized selection of the hormone dosage is essential for the treatment of immunotherapy-induced multisystem irAEs."( Adverse reactions and efficacy of camrelizumab in patients with lung adenocarcinoma with high PD-L1 expression: A case report.
Liu, X; Wang, Z; Wei, T, 2023
)
0.91
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (5)

RoleDescription
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
antimetaboliteA substance which is structurally similar to a metabolite but which competes with it or replaces it, and so prevents or reduces its normal utilization.
EC 2.1.1.45 (thymidylate synthase) inhibitorAn EC 2.1.1.* (methyltransferases) inhibitor that interferes with the action of thymidylate synthase (EC 2.1.1.45).
EC 1.5.1.3 (dihydrofolate reductase) inhibitorAn EC 1.5.1.* (oxidoreductase acting on donor CH-NH group, NAD(+) or NADP(+) as acceptor) inhibitor that interferes with the action of dihydrofolate reductase (EC 1.5.1.3).
EC 2.1.2.2 (phosphoribosylglycinamide formyltransferase) inhibitorAn EC 2.1.2.* (hydroxymethyl-, formyl- and related transferases) inhibitor that interferes with the action of phosphoribosylglycinamide formyltransferase (EC 2.1.2.2).
[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 (2)

ClassDescription
pyrrolopyrimidine
N-acyl-L-glutamic acidAny optically active N-acylglutamic acid having L-configuration.
[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 (25)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
EWS/FLI fusion proteinHomo sapiens (human)Potency0.39030.001310.157742.8575AID1259252; AID1259253; AID1259255; AID1259256
tyrosine-protein kinase YesHomo sapiens (human)Potency38.70880.00005.018279.2586AID686947
[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)
Chain A, Thymidylate SynthaseEscherichia coliKi0.10900.10900.10900.1090AID977610
Chain A, Thymidylate SynthaseHomo sapiens (human)Ki0.10900.10900.10900.1090AID977610
Chain A, Thymidylate SynthaseEscherichia coliKi0.10900.10900.10900.1090AID977610
Thymidylate synthase Escherichia coliIC50 (µMol)122.23850.01902.589610.0000AID211439; AID211442; AID211446; AID240801; AID241657; AID260014; AID282387; AID282575; AID282789; AID312254; AID341439; AID391733; AID422633
Thymidylate synthase Escherichia coli LF82IC50 (µMol)76.00000.02300.03550.0480AID613094
Dihydrofolate reductaseHomo sapiens (human)IC50 (µMol)7.67620.00060.87267.3000AID1663358; AID1663359; AID240641; AID241717; AID260039; AID282388; AID282571; AID282785; AID312256; AID341462; AID391734; AID422635; AID457195; AID56945; AID56964; AID56978; AID613096
Dihydrofolate reductaseHomo sapiens (human)Ki0.00710.00000.37564.9000AID1894189; AID238926; AID513675
Dihydrofolate reductaseMus musculus (house mouse)Ki0.56000.00000.21713.9000AID57444
Dihydrofolate reductaseLacticaseibacillus caseiIC50 (µMol)173.07500.00130.26964.9000AID241776; AID282573; AID57739; AID57759
Thymidylate synthaseLacticaseibacillus caseiIC50 (µMol)29.00000.00901.93248.8000AID211645; AID211799; AID241751; AID282576
Thymidylate synthaseLacticaseibacillus caseiKi0.55000.01001.65509.3900AID212843
Thymidylate synthaseHomo sapiens (human)IC50 (µMol)15.81180.00662.06379.5000AID1439756; AID1501141; AID1763184; AID212126; AID212157; AID212297; AID212301; AID240604; AID241683; AID260013; AID282386; AID282574; AID282788; AID312253; AID341428; AID391732; AID422632; AID457192; AID476562; AID613093; AID671163; AID740238
Thymidylate synthaseHomo sapiens (human)Ki0.11420.00010.34353.0000AID1894188; AID238909; AID513674
Thymidylate synthaseMus musculus (house mouse)Ki0.34000.00341.73759.3000AID212657
Dihydrofolate reductaseEscherichia coli K-12IC50 (µMol)383.52500.00150.55126.8000AID241682; AID282389; AID282572; AID282786; AID341469; AID391736; AID422636; AID457196; AID57235; AID57240; AID57243; AID613097
Folate receptor betaHomo sapiens (human)IC50 (µMol)0.25850.02200.26580.8630AID1164835; AID1164836; AID1380194; AID1380199; AID1512995; AID1512999
Folate receptor alphaHomo sapiens (human)IC50 (µMol)0.24170.01500.19640.8940AID1164833; AID1164834; AID1197480; AID1380197; AID1380198; AID1512994
Dihydrofolate reductasePneumocystis cariniiIC50 (µMol)230.00000.00060.54766.2000AID55842
Trifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)IC50 (µMol)5.02850.00580.31223.0000AID1197489; AID1501141; AID1513009; AID349815; AID362438; AID459869
Trifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)Ki0.61120.00600.25921.0000AID1252445; AID1631997; AID1894190; AID513677
Bifunctional purine biosynthesis protein ATICHomo sapiens (human)IC50 (µMol)0.07000.01000.26000.7000AID1501141
Bifunctional purine biosynthesis protein ATICHomo sapiens (human)Ki200.00000.40000.64000.8800AID1709484
Reduced folate transporterHomo sapiens (human)IC50 (µMol)0.09280.00070.05010.1380AID1164831; AID1197478; AID1380195; AID1512992; AID1512997
Thymidylate synthase Rattus norvegicus (Norway rat)IC50 (µMol)20.00000.36000.54000.9000AID212677
Bifunctional dihydrofolate reductase-thymidylate synthaseToxoplasma gondiiIC50 (µMol)2.47800.00061.042810.0000AID282787; AID282790; AID341451; AID341470; AID391735; AID391737; AID422634; AID422637; AID457194; AID457197; AID56187; AID56327; AID613095; AID613098; AID671165
Trifunctional purine biosynthetic protein adenosine-3Mus musculus (house mouse)IC50 (µMol)20.00000.78000.78000.7800AID349814; AID362437; AID459868
Proton-coupled folate transporterHomo sapiens (human)IC50 (µMol)0.02290.00830.06120.1210AID1164837; AID1197481; AID1380200; AID1512996; AID1512998
Proton-coupled folate transporterHomo sapiens (human)Ki0.67760.09400.67762.5400AID1380212; AID1513003; AID1631995; AID459865; AID459866; AID695098; AID695099
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Proton-coupled folate transporterHomo sapiens (human)K0.15700.04400.15700.2700AID1252439; AID1252440
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (90)

Processvia Protein(s)Taxonomy
tetrahydrobiopterin biosynthetic processDihydrofolate reductaseHomo sapiens (human)
one-carbon metabolic processDihydrofolate reductaseHomo sapiens (human)
negative regulation of translationDihydrofolate reductaseHomo sapiens (human)
axon regenerationDihydrofolate reductaseHomo sapiens (human)
response to methotrexateDihydrofolate reductaseHomo sapiens (human)
dihydrofolate metabolic processDihydrofolate reductaseHomo sapiens (human)
tetrahydrofolate metabolic processDihydrofolate reductaseHomo sapiens (human)
tetrahydrofolate biosynthetic processDihydrofolate reductaseHomo sapiens (human)
folic acid metabolic processDihydrofolate reductaseHomo sapiens (human)
positive regulation of nitric-oxide synthase activityDihydrofolate reductaseHomo sapiens (human)
regulation of removal of superoxide radicalsDihydrofolate reductaseHomo sapiens (human)
dTMP biosynthetic processThymidylate synthaseHomo sapiens (human)
dTTP biosynthetic processThymidylate synthaseHomo sapiens (human)
circadian rhythmThymidylate synthaseHomo sapiens (human)
response to xenobiotic stimulusThymidylate synthaseHomo sapiens (human)
response to toxic substanceThymidylate synthaseHomo sapiens (human)
negative regulation of translationThymidylate synthaseHomo sapiens (human)
uracil metabolic processThymidylate synthaseHomo sapiens (human)
methylationThymidylate synthaseHomo sapiens (human)
response to progesteroneThymidylate synthaseHomo sapiens (human)
response to vitamin AThymidylate synthaseHomo sapiens (human)
response to cytokineThymidylate synthaseHomo sapiens (human)
tetrahydrofolate interconversionThymidylate synthaseHomo sapiens (human)
response to ethanolThymidylate synthaseHomo sapiens (human)
response to organophosphorusThymidylate synthaseHomo sapiens (human)
developmental growthThymidylate synthaseHomo sapiens (human)
cartilage developmentThymidylate synthaseHomo sapiens (human)
response to glucocorticoidThymidylate synthaseHomo sapiens (human)
response to folic acidThymidylate synthaseHomo sapiens (human)
intestinal epithelial cell maturationThymidylate synthaseHomo sapiens (human)
DNA biosynthetic processThymidylate synthaseHomo sapiens (human)
liver regenerationThymidylate synthaseHomo sapiens (human)
10-formyltetrahydrofolate biosynthetic processDihydrofolate reductaseEscherichia coli K-12
response to xenobiotic stimulusDihydrofolate reductaseEscherichia coli K-12
folic acid biosynthetic processDihydrofolate reductaseEscherichia coli K-12
one-carbon metabolic processDihydrofolate reductaseEscherichia coli K-12
response to methotrexateDihydrofolate reductaseEscherichia coli K-12
tetrahydrofolate biosynthetic processDihydrofolate reductaseEscherichia coli K-12
response to antibioticDihydrofolate reductaseEscherichia coli K-12
dihydrofolate metabolic processDihydrofolate reductaseEscherichia coli K-12
folic acid metabolic processDihydrofolate reductaseEscherichia coli K-12
monocyte chemotaxisFolate receptor betaHomo sapiens (human)
inflammatory responseFolate receptor betaHomo sapiens (human)
positive regulation of cell population proliferationFolate receptor betaHomo sapiens (human)
folic acid transportFolate receptor betaHomo sapiens (human)
cellular response to folic acidFolate receptor betaHomo sapiens (human)
cell adhesionFolate receptor betaHomo sapiens (human)
sperm-egg recognitionFolate receptor betaHomo sapiens (human)
fusion of sperm to egg plasma membrane involved in single fertilizationFolate receptor betaHomo sapiens (human)
heart loopingFolate receptor alphaHomo sapiens (human)
neural crest cell migration involved in heart formationFolate receptor alphaHomo sapiens (human)
cardiac neural crest cell migration involved in outflow tract morphogenesisFolate receptor alphaHomo sapiens (human)
receptor-mediated endocytosisFolate receptor alphaHomo sapiens (human)
folic acid transportFolate receptor alphaHomo sapiens (human)
regulation of transforming growth factor beta receptor signaling pathwayFolate receptor alphaHomo sapiens (human)
axon regenerationFolate receptor alphaHomo sapiens (human)
folic acid metabolic processFolate receptor alphaHomo sapiens (human)
regulation of canonical Wnt signaling pathwayFolate receptor alphaHomo sapiens (human)
pharyngeal arch artery morphogenesisFolate receptor alphaHomo sapiens (human)
anterior neural tube closureFolate receptor alphaHomo sapiens (human)
cellular response to folic acidFolate receptor alphaHomo sapiens (human)
cell adhesionFolate receptor alphaHomo sapiens (human)
fusion of sperm to egg plasma membrane involved in single fertilizationFolate receptor alphaHomo sapiens (human)
sperm-egg recognitionFolate receptor alphaHomo sapiens (human)
brainstem developmentTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
GMP biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
'de novo' IMP biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
glycine metabolic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
purine ribonucleoside monophosphate biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
response to organic substanceTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
response to inorganic substanceTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
cerebellum developmentTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
cerebral cortex developmentTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
'de novo' AMP biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
tetrahydrofolate biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
'de novo' XMP biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
purine nucleotide biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
adenine biosynthetic processTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
brainstem developmentBifunctional purine biosynthesis protein ATICHomo sapiens (human)
nucleobase-containing compound metabolic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
GMP biosynthetic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
'de novo' IMP biosynthetic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
response to inorganic substanceBifunctional purine biosynthesis protein ATICHomo sapiens (human)
cerebellum developmentBifunctional purine biosynthesis protein ATICHomo sapiens (human)
cerebral cortex developmentBifunctional purine biosynthesis protein ATICHomo sapiens (human)
animal organ regenerationBifunctional purine biosynthesis protein ATICHomo sapiens (human)
'de novo' AMP biosynthetic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
dihydrofolate metabolic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
tetrahydrofolate biosynthetic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
'de novo' XMP biosynthetic processBifunctional purine biosynthesis protein ATICHomo sapiens (human)
cellular response to interleukin-7Bifunctional purine biosynthesis protein ATICHomo sapiens (human)
dTMP biosynthetic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
glycine metabolic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
L-serine metabolic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
L-serine catabolic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
purine nucleobase biosynthetic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
negative regulation of translationSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
tetrahydrofolate interconversionSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
tetrahydrofolate metabolic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
folic acid metabolic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
protein homotetramerizationSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
cellular response to tetrahydrofolateSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
cellular response to leukemia inhibitory factorSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
glycine biosynthetic process from serineSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
carnitine biosynthetic processSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
xenobiotic transmembrane transportReduced folate transporterHomo sapiens (human)
female pregnancyReduced folate transporterHomo sapiens (human)
organic anion transportReduced folate transporterHomo sapiens (human)
folic acid transportReduced folate transporterHomo sapiens (human)
folic acid metabolic processReduced folate transporterHomo sapiens (human)
methotrexate transportReduced folate transporterHomo sapiens (human)
monoatomic anion transmembrane transportReduced folate transporterHomo sapiens (human)
folate transmembrane transportReduced folate transporterHomo sapiens (human)
cyclic-GMP-AMP transmembrane import across plasma membraneReduced folate transporterHomo sapiens (human)
positive regulation of cGAS/STING signaling pathwayReduced folate transporterHomo sapiens (human)
transport across blood-brain barrierReduced folate transporterHomo sapiens (human)
folate import across plasma membraneReduced folate transporterHomo sapiens (human)
intracellular iron ion homeostasisProton-coupled folate transporterHomo sapiens (human)
folic acid transportProton-coupled folate transporterHomo sapiens (human)
heme transportProton-coupled folate transporterHomo sapiens (human)
heme metabolic processProton-coupled folate transporterHomo sapiens (human)
folic acid metabolic processProton-coupled folate transporterHomo sapiens (human)
methotrexate transportProton-coupled folate transporterHomo sapiens (human)
intestinal folate absorptionProton-coupled folate transporterHomo sapiens (human)
folate transmembrane transportProton-coupled folate transporterHomo sapiens (human)
proton transmembrane transportProton-coupled folate transporterHomo sapiens (human)
folate import across plasma membraneProton-coupled folate transporterHomo sapiens (human)
transmembrane transportProton-coupled folate transporterHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (44)

Processvia Protein(s)Taxonomy
mRNA regulatory element binding translation repressor activityDihydrofolate reductaseHomo sapiens (human)
mRNA bindingDihydrofolate reductaseHomo sapiens (human)
dihydrofolate reductase activityDihydrofolate reductaseHomo sapiens (human)
folic acid bindingDihydrofolate reductaseHomo sapiens (human)
NADPH bindingDihydrofolate reductaseHomo sapiens (human)
sequence-specific mRNA bindingDihydrofolate reductaseHomo sapiens (human)
NADP bindingDihydrofolate reductaseHomo sapiens (human)
mRNA regulatory element binding translation repressor activityThymidylate synthaseHomo sapiens (human)
thymidylate synthase activityThymidylate synthaseHomo sapiens (human)
folic acid bindingThymidylate synthaseHomo sapiens (human)
protein homodimerization activityThymidylate synthaseHomo sapiens (human)
sequence-specific mRNA bindingThymidylate synthaseHomo sapiens (human)
dihydrofolate reductase activityDihydrofolate reductaseEscherichia coli K-12
protein bindingDihydrofolate reductaseEscherichia coli K-12
folic acid bindingDihydrofolate reductaseEscherichia coli K-12
oxidoreductase activityDihydrofolate reductaseEscherichia coli K-12
NADP bindingDihydrofolate reductaseEscherichia coli K-12
methotrexate bindingDihydrofolate reductaseEscherichia coli K-12
dihydrofolic acid bindingDihydrofolate reductaseEscherichia coli K-12
NADP+ bindingDihydrofolate reductaseEscherichia coli K-12
NADPH bindingDihydrofolate reductaseEscherichia coli K-12
folic acid bindingFolate receptor betaHomo sapiens (human)
folic acid receptor activityFolate receptor betaHomo sapiens (human)
signaling receptor activityFolate receptor betaHomo sapiens (human)
protein bindingFolate receptor alphaHomo sapiens (human)
folic acid bindingFolate receptor alphaHomo sapiens (human)
folic acid receptor activityFolate receptor alphaHomo sapiens (human)
signaling receptor activityFolate receptor alphaHomo sapiens (human)
phosphoribosylamine-glycine ligase activityTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
phosphoribosylformylglycinamidine cyclo-ligase activityTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
phosphoribosylglycinamide formyltransferase activityTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
ATP bindingTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
metal ion bindingTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
IMP cyclohydrolase activityBifunctional purine biosynthesis protein ATICHomo sapiens (human)
protein homodimerization activityBifunctional purine biosynthesis protein ATICHomo sapiens (human)
cadherin bindingBifunctional purine biosynthesis protein ATICHomo sapiens (human)
phosphoribosylaminoimidazolecarboxamide formyltransferase activityBifunctional purine biosynthesis protein ATICHomo sapiens (human)
mRNA regulatory element binding translation repressor activitySerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
glycine hydroxymethyltransferase activitySerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
protein bindingSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
pyridoxal phosphate bindingSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
small molecule bindingSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
identical protein bindingSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
protein homodimerization activitySerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
mRNA 5'-UTR bindingSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
serine bindingSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
aldehyde-lyase activitySerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
organic anion transmembrane transporter activityReduced folate transporterHomo sapiens (human)
folic acid transmembrane transporter activityReduced folate transporterHomo sapiens (human)
folate:monoatomic anion antiporter activityReduced folate transporterHomo sapiens (human)
antiporter activityReduced folate transporterHomo sapiens (human)
methotrexate transmembrane transporter activityReduced folate transporterHomo sapiens (human)
xenobiotic transmembrane transporter activityReduced folate transporterHomo sapiens (human)
2',3'-cyclic GMP-AMP bindingReduced folate transporterHomo sapiens (human)
cyclic-GMP-AMP transmembrane transporter activityReduced folate transporterHomo sapiens (human)
folic acid bindingReduced folate transporterHomo sapiens (human)
heme transmembrane transporter activityProton-coupled folate transporterHomo sapiens (human)
folic acid bindingProton-coupled folate transporterHomo sapiens (human)
folic acid transmembrane transporter activityProton-coupled folate transporterHomo sapiens (human)
proton transmembrane transporter activityProton-coupled folate transporterHomo sapiens (human)
heme transmembrane transporter activityProton-coupled folate transporterHomo sapiens (human)
symporter activityProton-coupled folate transporterHomo sapiens (human)
methotrexate transmembrane transporter activityProton-coupled folate transporterHomo sapiens (human)
folic acid:proton symporter activityProton-coupled folate transporterHomo sapiens (human)
transmembrane transporter activityProton-coupled folate transporterHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (24)

Processvia Protein(s)Taxonomy
mitochondrionDihydrofolate reductaseHomo sapiens (human)
cytosolDihydrofolate reductaseHomo sapiens (human)
mitochondrionDihydrofolate reductaseHomo sapiens (human)
cytosolDihydrofolate reductaseMus musculus (house mouse)
nucleusThymidylate synthaseHomo sapiens (human)
cytoplasmThymidylate synthaseHomo sapiens (human)
mitochondrionThymidylate synthaseHomo sapiens (human)
mitochondrial inner membraneThymidylate synthaseHomo sapiens (human)
mitochondrial matrixThymidylate synthaseHomo sapiens (human)
cytosolThymidylate synthaseHomo sapiens (human)
mitochondrionThymidylate synthaseHomo sapiens (human)
cytosolThymidylate synthaseHomo sapiens (human)
cytosolDihydrofolate reductaseEscherichia coli K-12
cytosolDihydrofolate reductaseEscherichia coli K-12
extracellular regionFolate receptor betaHomo sapiens (human)
plasma membraneFolate receptor betaHomo sapiens (human)
external side of plasma membraneFolate receptor betaHomo sapiens (human)
cell surfaceFolate receptor betaHomo sapiens (human)
external side of plasma membraneFolate receptor betaHomo sapiens (human)
Golgi membraneFolate receptor alphaHomo sapiens (human)
endosomeFolate receptor alphaHomo sapiens (human)
endoplasmic reticulum membraneFolate receptor alphaHomo sapiens (human)
plasma membraneFolate receptor alphaHomo sapiens (human)
external side of plasma membraneFolate receptor alphaHomo sapiens (human)
cell surfaceFolate receptor alphaHomo sapiens (human)
ER to Golgi transport vesicle membraneFolate receptor alphaHomo sapiens (human)
membraneFolate receptor alphaHomo sapiens (human)
basolateral plasma membraneFolate receptor alphaHomo sapiens (human)
apical plasma membraneFolate receptor alphaHomo sapiens (human)
transport vesicleFolate receptor alphaHomo sapiens (human)
clathrin-coated vesicleFolate receptor alphaHomo sapiens (human)
brush border membraneFolate receptor alphaHomo sapiens (human)
endoplasmic reticulum-Golgi intermediate compartment membraneFolate receptor alphaHomo sapiens (human)
extracellular exosomeFolate receptor alphaHomo sapiens (human)
nucleusFolate receptor alphaHomo sapiens (human)
external side of plasma membraneFolate receptor alphaHomo sapiens (human)
cytosolTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
cytosolTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
extracellular exosomeTrifunctional purine biosynthetic protein adenosine-3Homo sapiens (human)
cytosolBifunctional purine biosynthesis protein ATICHomo sapiens (human)
plasma membraneBifunctional purine biosynthesis protein ATICHomo sapiens (human)
membraneBifunctional purine biosynthesis protein ATICHomo sapiens (human)
extracellular exosomeBifunctional purine biosynthesis protein ATICHomo sapiens (human)
cytosolBifunctional purine biosynthesis protein ATICHomo sapiens (human)
nucleoplasmSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
cytosolSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
extracellular exosomeSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
mitochondrionSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
nucleusSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
cytoplasmSerine hydroxymethyltransferase, cytosolicHomo sapiens (human)
plasma membraneReduced folate transporterHomo sapiens (human)
basolateral plasma membraneReduced folate transporterHomo sapiens (human)
apical plasma membraneReduced folate transporterHomo sapiens (human)
brush border membraneReduced folate transporterHomo sapiens (human)
basolateral plasma membraneReduced folate transporterHomo sapiens (human)
plasma membraneReduced folate transporterHomo sapiens (human)
apical plasma membraneReduced folate transporterHomo sapiens (human)
plasma membraneProton-coupled folate transporterHomo sapiens (human)
basolateral plasma membraneProton-coupled folate transporterHomo sapiens (human)
cytoplasmProton-coupled folate transporterHomo sapiens (human)
endosomeProton-coupled folate transporterHomo sapiens (human)
plasma membraneProton-coupled folate transporterHomo sapiens (human)
cell surfaceProton-coupled folate transporterHomo sapiens (human)
endosome membraneProton-coupled folate transporterHomo sapiens (human)
basolateral plasma membraneProton-coupled folate transporterHomo sapiens (human)
apical plasma membraneProton-coupled folate transporterHomo sapiens (human)
brush border membraneProton-coupled folate transporterHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (511)

Assay IDTitleYearJournalArticle
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.
AID1745855NCATS anti-infectives library activity on the primary C. elegans qHTS viability assay2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
AID1745854NCATS anti-infectives library activity on HEK293 viability as a counter-qHTS vs the C. elegans viability qHTS2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
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.
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.
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.
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.
AID1745845Primary qHTS for Inhibitors of ATXN expression
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.
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.
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.
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.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
AID686947qHTS for small molecule inhibitors of Yes1 kinase: Primary Screen2013Bioorganic & medicinal chemistry letters, Aug-01, Volume: 23, Issue:15
Identification of potent Yes1 kinase inhibitors using a library screening approach.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
AID1562587Antiproliferative activity against human KB cells in presence of thymidine2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID1380219Toxicity in folate-deficient diet fed ICR SCID mouse assessed as body weight loss at 8.1 mg/kg, iv administered every 2 days for 4 times starting on day 3 measured twice per week2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1513013Anticlonogenic activity against human IGROV1 cells incubated for 24 hrs followed by compound wash out and measured after 10 days by methylene blue staining-based assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1501142Antiproliferative activity against human SW620 cells in folate free medium after 72 hrs in presence of leucovorin by MTT assay2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID1398150Effect on HSP90 protein expression in human IGROV1 cells by Western blot analysis2018Journal of medicinal chemistry, 08-23, Volume: 61, Issue:16
Conformational Propensity and Biological Studies of Proline Mutated LR Peptides Inhibiting Human Thymidylate Synthase and Ovarian Cancer Cell Growth.
AID282574Inhibition of human recombinant thymidylate synthase2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis of classical, three-carbon-bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID671164Inhibition of Escherichia coli thymidylate synthase2012Bioorganic & medicinal chemistry, Jul-15, Volume: 20, Issue:14
Novel tricyclic indeno[2,1-d]pyrimidines with dual antiangiogenic and cytotoxic activities as potent antitumor agents.
AID1513017Antiproliferative activity against FRalpha knockout human IGROV1 KD4 cells after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1709484Inhibition of human full length N-terminal His-tagged ATIC expressed in Chinese Hamster MTXRII-OuaR2-4 R2 cells assessed as reduction in THF formation using 10-CHOTHF as substrate by spectrophotometric method2021Bioorganic & medicinal chemistry, 05-01, Volume: 37Discovery of 6-substituted thieno[2,3-d]pyrimidine analogs as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis in folate receptor
AID1236755Antiproliferative activity against human HeLa cells at 100 ug/ml after 72 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1198264Cytotoxicity against human A549 cells assessed as growth inhibition after 72 hrs by MTT assay2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
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).
AID1062557Cytotoxicity against chinese hamster R2 cells after 96 hrs by CellTitre-Blue fluorescence assay2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
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.
AID1894190Inhibition of GARFT (unknown origin) assessed as inhibition constant2021European journal of medicinal chemistry, Mar-15, Volume: 214FDA-approved pyrimidine-fused bicyclic heterocycles for cancer therapy: Synthesis and clinical application.
AID1501167Induction of apoptosis in human KB cells assessed as early apoptotic cells at 10 uM after 48 hrs by Annexin V-FITC/propidium iodide staining based flow cytometry (Rvb = 16.9%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID1197482Cytotoxicity against PCFT-deficient Chinese hamster R2(VC) cells assessed as growth inhibition after 96 hrs by CellTiter-Blue assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID1512992Binding affinity to human RFC expressed in Chinese hamster PC43-10 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID605429Growth inhibition of human A549 cells assessed as cell growth at 6.4 uM after 72 hrs by fluorometric microculture cytotoxicity assay in presence of 100 uM hypoxanthine relative to untreated control2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
Antifolate and antiproliferative activity of 6,8,10-triazaspiro[4.5]deca-6,8-dienes and 1,3,5-triazaspiro[5.5]undeca-1,3-dienes.
AID621181Inhibition of GARFTase in chinese hamster R2 cells expressing human PCFT assessed as incorporation of [14C]glycine into [14C]formyl GAR in the presence of 4 uM azaserine2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1360133Selectivity index, ratio of CC50 for human HTB125 cells to IC50 for human A549 cells2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID341472Selectivity for sToxoplasma gondii dihydrofolate reductase over human dihydrofolate reductase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID621123Antiproliferative activity against chinese hamster R2 cells assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID422633Inhibition of Escherichia coli thymidylate synthase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID341470Inhibition of Toxoplasma gondii dihydrofolate reductase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID1062556Cytotoxicity against chinese hamster RT16 cells expressing human FRalpha after 96 hrs by CellTitre-Blue fluorescence assay in presence of folic acid2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID621127Antiproliferative activity against chinese hamster D4 cells expressing human FRbeta assessed as reduction of viable cells after 96 hrs in the presence of 200 nM folic acid2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID391738Selectivity ratio of IC50 for human recombinant DHFR to IC50 for Toxoplasma gondii DHFR2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID72867Compound was evaluated for the inhibitory activity against GAR transformylase purified from mouse L1210 cells1992Journal of medicinal chemistry, Nov-13, Volume: 35, Issue:23
A dideazatetrahydrofolate analogue lacking a chiral center at C-6, N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5- yl)ethyl]benzoyl]-L-glutamic acid, is an inhibitor of thymidylate synthase.
AID1562580Cytotoxicity against HUVEC assessed as reduction in cell viability after 72 hrs by MTT assay2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID1236756Antiproliferative activity against human T47D cells at 100 ug/ml after 24 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1513004Inhibition of human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as reduction in [3H]MTX uptake at pH 5.5 at 1 uM measured over 2 mins relative to control2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1053455Growth inhibition of Chinese hamster PC43-10 cells after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID99486Compound was evaluated for the half-maximal inhibition against mouse L1210 leukemic cell growth1992Journal of medicinal chemistry, Nov-13, Volume: 35, Issue:23
A dideazatetrahydrofolate analogue lacking a chiral center at C-6, N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5- yl)ethyl]benzoyl]-L-glutamic acid, is an inhibitor of thymidylate synthase.
AID362437Inhibition of mouse recombinant GARFTase2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1079944Benign tumor, proven histopathologically. Value is number of references indexed. [column 'T.BEN' in source]
AID1631997Inhibition of recombinant N-terminal 6His-tagged human GARFTase assessed as formation of 5,8-dideazafolate from 10-formyl-5,8-dideazafolic acid measured at every 15 seconds over 20 min by spectrophotometric analysis2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesi
AID1380209Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at 10 uM at pH 5.5 by scintillation counting method relative to control2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1252421Selectivity ratio of IC50 for human RFC expressed in Chinese hamster PC43-10 cells assessed as cell growth inhibition to IC50 for human FRalpha expressed in Chinese hamster RT16 cells assessed as cell growth inhibition2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID221803Compound was evaluated for the half-maximal inhibition against human CCRF-CEM lymphoblastic leukemic cell growth1992Journal of medicinal chemistry, Nov-13, Volume: 35, Issue:23
A dideazatetrahydrofolate analogue lacking a chiral center at C-6, N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5- yl)ethyl]benzoyl]-L-glutamic acid, is an inhibitor of thymidylate synthase.
AID1164833Inhibition of FRalpha (unknown origin) expressed in Chinese hamster RT16 cells assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
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).
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).
AID238926Inhibition of recombinant human dihydrofolate reductase done at 37 degree C in pH 7.4 with compound2005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1198282Cell cycle arrest in human KB cells assessed as accumulation at G2 phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometric analysis (Rvb = 34%)2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID1663358Inhibition of DHFR in human KB cells expressing RFC/FRalpha/PCFT assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID613093Inhibition of human TS assessed as oxidation of tetrahydrofolate to dihydrofolate after 2 to 12 mins by spectrophotometry2011Bioorganic & medicinal chemistry, Jun-01, Volume: 19, Issue:11
Design, synthesis, biological evaluation and X-ray crystal structure of novel classical 6,5,6-tricyclic benzo[4,5]thieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1219361Ratio of efflux clearance to clearance in brain measured in mouse at 0.3 uCi/ml by in vitro brain slice uptake technique2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID57243Inhibitory concentration against isolated Escherichia coli Dihydrofolate reductase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID1062535Induction of apoptosis in human KB cells assessed as late apoptotic cells at 1 uM after 96 hrs by Annexin V-FITC/7-AAD staining-based flow cytometry (Rvb = 2.09%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1079947Comments (NB not yet translated). [column 'COMMENTAIRES' in source]
AID459864Displacement of [3H]folic acid from human FRbeta expressed in Chinese hamster D4 cells relative to unlabeled folic acid2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID1380197Binding affinity to human FR-alpha receptor expressed in Chinese hamster RT16 cells assessed as antiproliferative activity measured as reduction in cell viability after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1380201Growth inhibition of Chinese hamster R2(VC) cells after 96 hrs celltiter-blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1562573Antiproliferative activity against human KB cells assessed as reduction in cell viability after 72 hrs by MTT assay2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID391734Inhibition of human recombinant DHFR2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID1513000Selectivity ratio of IC50 for human RFC2 expressed in human HeLa R1-11 cells to IC50 for human PCFT4 expressed in human HeLa R1-11 cells2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
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]
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]
AID1062530Inhibition of AICARFTase in human KB cells assessed as phosphorylated AMPK at 1 uM after 48 hrs by Western blot analysis2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID56327Inhibitory concentration against isolated Toxoplasma gondii DHFR (dihydrofolate reductase)2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID1053449Growth inhibition of Chinese hamster R2 cells expressing human PCFT4 after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID621169Induction of current in human PCFT expressed in xenopus oocyte at 5 uM at holding potential -90 mV by voltage clamp assay2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID312258Inhibition of Toxoplasma gondii DHFR2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1501147Inhibition of thymidylate synthase in human KB cells assessed as reduction in cell proliferation at 1 to 10 nM after 72 hrs in presence of excess of thymidine by MTT assay2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
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).
AID1501168Induction of apoptosis in human KB cells assessed as late apoptotic cells at 10 uM after 48 hrs by Annexin V-FITC/propidium iodide staining based flow cytometry (Rvb = 5.5%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID1219357Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence of 50 mM unlabelled compou2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1513019Antiproliferative activity against human A2780 cells after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1297310Cell cycle arrest in human SW620 cells assessed as accumulation at G1 phase at 10 uM after 96 hrs by propidium iodide staining based flow cytometry in presence of leucovorin (Rvb = 62.4 %)2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID312253Inhibition of human thymidylate synthase2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1631978Cytotoxicity in RFC-null Chinese hamster R2 cells assessed as reduction in cell viability measured after 96 hrs by Cell-Titer Blue fluorescence analysis2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesi
AID1219372Brain efflux index in Mrp2 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 60 mins2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1164831Inhibition of RFC (unknown origin) expressed in Chinese hamster PC43-10 cells assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID1562574Antiproliferative activity against human SW620 cells assessed as reduction in cell viability after 72 hrs by MTT assay2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID362421Antiproliferative activity against human RFC expressing Chinese hamster PC43-10 cells2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1663361Inhibition of DHFR in human KB cells expressing RFC/FRalpha/PCFT assessed as reduction in cell growth after 96 hrs in presence of AICA by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1631991Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at 1 and 10 uM and pH 5.5 measured after 5 mins2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesi
AID1219362Efflux clearance mouse brain measured per gm of tissue at 0.3 uCi/ml by in vitro brain slice uptake technique2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1062547Cytotoxicity against human KB cells expressing human RFC/FRalpha/PCFT after 96 hrs by CellTitre-Blue fluorescence assay in presence of adenosine/AICA/thymidine2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1236749Antiproliferative activity against African green monkey Vero cells at 100 ug/ml after 72 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1501149Inhibition of TS/AICARFTase/GARFTase in human KB cells assessed as reduction in cell proliferation at 1 to 10 nM after 72 hrs in presence of excess of thymidine and adenosine by MTT assay2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID459868Inhibition of mouse recombinant GARFtase assessed as FGAR formation by spectrophotometry2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID362423Antiproliferative activity against human FRalpha expressing Chinese hamster RT16 cells2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1197478Binding affinity to human RFC expressed in Chinese hamster PC43-10 cells assessed as cell growth inhibition after 96 hrs by CellTiter-Blue assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID1252445Inhibition of human recombinant His-tagged GARFTase assessed as formation of 5,8-dideazafolate from 10-formyl-5,80-dideazafolic acid measured every 15 secs over 20 mins by spectrophotometric analysis2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID1562590Antiproliferative activity against human KB cells in presence of AICA and thymidine2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID1252437Cellular uptake in PCFT-null Chinese hamster R2 cells at 0.5 uM at 37 degC at pH 5.5 measured over 2 mins2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID282573Inhibition of Lactobacillus casei DHFR2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis of classical, three-carbon-bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID212301Inhibitory concentration against isolated Thymidylate synthase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID341451Inhibition of Toxoplasma gondii thymidylate synthase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID22930First-order rate constant of the compound1992Journal of medicinal chemistry, Nov-13, Volume: 35, Issue:23
A dideazatetrahydrofolate analogue lacking a chiral center at C-6, N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5- yl)ethyl]benzoyl]-L-glutamic acid, is an inhibitor of thymidylate synthase.
AID1297295Antiproliferative activity against human SW620 cells after 72 hrs by MTT assay in presence of leucovorin2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID422634Inhibition of Toxoplasma gondii thymidylate synthase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID1079942Steatosis, proven histopathologically. Value is number of references indexed. [column 'STEAT' in source]
AID595567Mixed-type inhibition of human recombinant Serine hydroxymethyltransferase, cytosolic measured by Y-axes intercepts of mixed-type inhibition against compound concentration by spectrophotometry2011European journal of medicinal chemistry, May, Volume: 46, Issue:5
In silico and in vitro validation of serine hydroxymethyltransferase as a chemotherapeutic target of the antifolate drug pemetrexed.
AID362422Antiproliferative activity against human RFC expressing Chinese hamster R2 cells2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID621135Antiproliferative activity against chinese hamster R2 cells expressing human PCFT assessed as inhibition of colony formation after 10 to 14 days2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID282790Inhibition of Toxoplasma gondii thymidylate synthase2005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
Synthesis of N-{4-[(2,4-diamino-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid and N-{4-[(2-amino-4-oxo-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid as dual inhibitors of dihydrofol
AID341469Inhibition of Escherichia coli dihydrofolate reductase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID1360131Selectivity index, ratio of CC50 for human IOSE80 cells to IC50 for human A549 cells2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1236750Antiproliferative activity against human A549 cells at 100 ug/ml after 24 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
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).
AID422638Ratio of IC50 for human recombinant dihydrofolate reductase to IC50 for Toxoplasma gondii dihydrofolate reductase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID1562595Cell cycle arrest in human KB cells assessed as accumulation at G2 phase at 10 uM after 96 hrs by RNase-1 A/propidium iodide staining-based flow cytometric analysis (Rvb = 9.06%)2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID513677Inhibition of GARFT2010Journal of medicinal chemistry, Sep-23, Volume: 53, Issue:18
Novel approaches for targeting thymidylate synthase to overcome the resistance and toxicity of anticancer drugs.
AID1198283Cell cycle arrest in human KB cells assessed as accumulation at G1 phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometric analysis (Rvb = 52.4%)2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID621170Inhibition of human PCFT-mediated [3H]MTX uptake ectopically expressed in chinese hamster R2 cells at 10 uM at pH 5.5 to 7.22011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1853730Induction of hTS expression in human IGROV-1 cells assessed as fold increase at 5 uM measured after 48 hrs by immunoblot quantitative analysis2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Folic Acid-Peptide Conjugates Combine Selective Cancer Cell Internalization with Thymidylate Synthase Dimer Interface Targeting.
AID1512999Binding affinity to human FR2 expressed in human HeLa R1-11 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1079940Granulomatous liver disease, proven histopathologically. Value is number of references indexed. [column 'GRAN' in source]
AID241717Inhibition of human dihydrofolate reductase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1197483Antiproliferative activity against human KB cells expressing RFC/FRalpha/PCFT after 96 hrs by CellTiter-Blue assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID1252438Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as increase in intracellular drug transport at 0.5 uM at 37 degC at pH 5.5 measured over 5 mins in presence of AGF942015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID1219374Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence of 1 mM unlabelled compoun2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1236758Antiproliferative activity against human T47D cells at 100 ug/ml after 72 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1663356Inhibition of DHFR in Chinese Hamster D4 cells expressing human FRbeta assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1252440Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as intracellular drug transport rate at 37 degC at pH 6.8 measured over 2 mins by Lineweaver-Burk plot analysis2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID1513006Inhibition of human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as reduction in [3H]MTX uptake at pH 5.5 at 1 to 10 uM measured over 2 mins2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID621131Antiproliferative activity against human KB cells expressing human RFC, FRalpha and PCFT assessed as reduction of viable cells after 96 hrs in the presence of 200 nM folic acid2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1439757Inhibition of Escherichia coli thymidylate synthase using dUMP/(6R,S)-tetrahydrofolate as substrate/co-factor by spectrophotometric method2017Bioorganic & medicinal chemistry letters, 04-01, Volume: 27, Issue:7
Synthesis and evaluation of 5-(arylthio)-9H-pyrimido[4,5-b]indole-2,4-diamines as receptor tyrosine kinase and thymidylate synthase inhibitors and as antitumor agents.
AID1197481Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as cell growth inhibition after 96 hrs by CellTiter-Blue assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID459867Displacement of [3H]MTX from human RFC expressed in Chinese hamster PC43-10 cells at 10 uM2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID1062549Displacement of [3H]folic acid from human FRalpha expressed in chinese hamster RT16 cells after 15 mins by scintillation counting analysis2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1219353Drug uptake in mouse brain ipsilateral cerebrum assessed as radiolabelled compound after 30 mins at 0.1 uM, intracerebral microinjection in presence 100 mM benzylpenicillin OAT inhibitor2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID282788Inhibition of human thymidylate synthase2005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
Synthesis of N-{4-[(2,4-diamino-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid and N-{4-[(2-amino-4-oxo-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid as dual inhibitors of dihydrofol
AID241682Inhibition of Escherichia coli dihydrofolate reductase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1380203Antiproliferative activity against human KB cells expressing human RFC/FR-alpha/PCFT assessed as reduction in cell viability measured after 96 hrs in presence of folic acid by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1164834Inhibition of FRalpha (unknown origin) expressed in Chinese hamster RT16 cells assessed as cell growth inhibition incubated up to 96 hrs in presence of 200 nM folic acid by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID1197479Cytotoxicity against RFC-deficient Chinese hamster R2 cells assessed as growth inhibition after 96 hrs by CellTiter-Blue assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID312259Selectivity ratio of IC50 for Toxoplasma gondii DHFR over IC50 for human recombinant DHFR2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1918193Dissociation constant, pKa of the compound2022Journal of medicinal chemistry, 11-10, Volume: 65, Issue:21
Identification of Organic Anion Transporter 2 Inhibitors: Screening, Structure-Based Analysis, and Clinical Drug Interaction Risk Assessment.
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]
AID1297306Inhibition of GARFTase/AICARFTase in human SW620 cells assessed as growth inhibition coincubated with adenosine2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID1297301Inhibition of AICARFTase in human SW620 cells assessed as growth inhibition coincubated with AICA2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID1252423Selectivity ratio of IC50 for human RFC expressed in Chinese hamster PC43-10 cells assessed as cell growth inhibition to IC50 for human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as cell growth inhibition2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID1219375Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence of 50 mM unlabelled compou2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID621176Antiproliferative activity against chinese hamster R2 cells expressing human PCFT assessed as growth inhibition in the presence of 10 uM thymidine and 320 uM AICA2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1398149Effect on DHFR protein expression in human IGROV1 cells by Western blot analysis2018Journal of medicinal chemistry, 08-23, Volume: 61, Issue:16
Conformational Propensity and Biological Studies of Proline Mutated LR Peptides Inhibiting Human Thymidylate Synthase and Ovarian Cancer Cell Growth.
AID1198267Inhibition of thymidylate synthase in human KB cells assessed as cell growth inhibition in presence of 10 uM thymidine2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID1164832Cytotoxicity against Chinese hamster R2 cells assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID1198263Cytotoxicity against human KB cells assessed as growth inhibition after 72 hrs by MTT assay2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID241683Inhibition of human thymidylate synthetase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID613096Inhibition of human DHFR by spectrophotometry2011Bioorganic & medicinal chemistry, Jun-01, Volume: 19, Issue:11
Design, synthesis, biological evaluation and X-ray crystal structure of novel classical 6,5,6-tricyclic benzo[4,5]thieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1297300Inhibition of thymidylate synthase/AICARFTase in human SW620 cells assessed as growth inhibition coincubated with AICA and thymine2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID56945Compound was evaluated as inhibitor of human Dihydrofolate reductase2003Journal of medicinal chemistry, Feb-13, Volume: 46, Issue:4
Design, synthesis, and biological activities of classical N-[4-[2-(2-amino-4-ethylpyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid and its 6-methyl derivative as potential dual inhibitors of thymidylate synthase and dihydrofolate reductase and
AID476564Inhibition of Toxoplasma gondii thymidylate synthase2010Journal of medicinal chemistry, Feb-25, Volume: 53, Issue:4
Single agents with designed combination chemotherapy potential: synthesis and evaluation of substituted pyrimido[4,5-b]indoles as receptor tyrosine kinase and thymidylate synthase inhibitors and as antitumor agents.
AID260039Inhibition of recombinant human DHFR2006Journal of medicinal chemistry, Feb-09, Volume: 49, Issue:3
Dual inhibitors of thymidylate synthase and dihydrofolate reductase as antitumor agents: design, synthesis, and biological evaluation of classical and nonclassical pyrrolo[2,3-d]pyrimidine antifolates(1).
AID260013Inhibition of recombinant human TS2006Journal of medicinal chemistry, Feb-09, Volume: 49, Issue:3
Dual inhibitors of thymidylate synthase and dihydrofolate reductase as antitumor agents: design, synthesis, and biological evaluation of classical and nonclassical pyrrolo[2,3-d]pyrimidine antifolates(1).
AID1360123Antiproliferative activity against human SGC7901 cells after 24 hrs by MTT assay2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1894189Inhibition of DHFR (unknown origin) assessed as inhibition constant2021European journal of medicinal chemistry, Mar-15, Volume: 214FDA-approved pyrimidine-fused bicyclic heterocycles for cancer therapy: Synthesis and clinical application.
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).
AID621122Antiproliferative activity against chinese hamster PC43-10 expressing human RFC assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
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).
AID1360132Selectivity index, ratio of CC50 for human GES1 cells to IC50 for human A549 cells2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1062539Cell cycle arrest in human KB cells assessed as accumulation at G1/G0 phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometry (Rvb = 67.4%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1501141Inhibition of TS/AICARFTase/GARFTase in human KB cells assessed as reduction in cell proliferation in folate free medium after 72 hrs in presence of leucovorin by MTT assay2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID595565Binding affinity to human recombinant Serine hydroxymethyltransferase, cytosolic by isothermal titration calorimetry2011European journal of medicinal chemistry, May, Volume: 46, Issue:5
In silico and in vitro validation of serine hydroxymethyltransferase as a chemotherapeutic target of the antifolate drug pemetrexed.
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).
AID1062532Inhibition of GARFTase in human KB cells assessed as incorporation of [14C]-glycine into [14C]-formyl GAR incubated 30 mins prior to [14C]-glycine addition measured after 16 hrs by radiometric assay in presence of azaserin2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1079932Highest frequency of moderate liver toxicity observed during clinical trials, expressed as a percentage. [column '% BIOL' in source]
AID1562594Cell cycle arrest in human KB cells assessed as accumulation at S phase at 10 uM after 96 hrs by RNase-1 A/propidium iodide staining-based flow cytometric analysis (Rvb = 48.26%)2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID422637Inhibition of Toxoplasma gondii dihydrofolate reductase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID1219352Drug uptake in mouse brain ipsilateral cerebrum assessed as radiolabelled compound after 30 mins at 0.1 uM, intracerebral microinjection in presence 100 mM probenecid OAT inhibitor2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
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
AID595566Competitive inhibition of human recombinant Serine hydroxymethyltransferase, cytosolic by spectrophotometry2011European journal of medicinal chemistry, May, Volume: 46, Issue:5
In silico and in vitro validation of serine hydroxymethyltransferase as a chemotherapeutic target of the antifolate drug pemetrexed.
AID621130Antiproliferative activity against human KB cells expressing human RFC, FRalpha and PCFT assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID595568Inhibition of human recombinant Serine hydroxymethyltransferase, cytosolic measured by slope intercepts of mixed-type inhibition against compound concentration by spectrophotometry2011European journal of medicinal chemistry, May, Volume: 46, Issue:5
In silico and in vitro validation of serine hydroxymethyltransferase as a chemotherapeutic target of the antifolate drug pemetrexed.
AID1219355Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins relative to wild type2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID56978Inhibitory concentration against isolated human Dihydrofolate reductase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID1380200Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as antiproliferative activity measured as reduction in cell viability after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID362425Antiproliferative activity against human FRbeta expressing Chinese hamster D4 cells2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1219371Brain efflux index in wild type mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1512994Binding affinity to human FRalpha expressed in Chinese hamster RT16 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1219370Drug uptake in mouse brain ipsilateral cerebrum assessed as radiolabelled compound after 30 mins at 0.1 uM, intracerebral microinjection in presence of 50 mM unlabelled compound2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID282572Inhibition of Escherichia coli DHFR2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis of classical, three-carbon-bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID240883Inhibitory concentration against Escherichia coli dihydrofolate reductase2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Novel 2-amino-4-oxo-5-arylthio-substituted-pyrrolo[2,3-d]pyrimidines as nonclassical antifolate inhibitors of thymidylate synthase.
AID282388Inhibition of human DHFR2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Benzoyl ring halogenated classical 2-amino-6-methyl-3,4-dihydro-4-oxo-5-substituted thiobenzoyl-7H-pyrrolo[2,3-d]pyrimidine antifolates as inhibitors of thymidylate synthase and as antitumor agents.
AID613098Inhibition of Toxoplasma gondii DHFR by spectrophotometry2011Bioorganic & medicinal chemistry, Jun-01, Volume: 19, Issue:11
Design, synthesis, biological evaluation and X-ray crystal structure of novel classical 6,5,6-tricyclic benzo[4,5]thieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1512993Antiproliferative activity in Chinese hamster R2 cells deficient in RFC, PCFT, and FRalpha assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1197490Inhibition of AICARFTase in human KB cells expressing RFC/FRalpha/PCFT assessed as increase in ZAM level at 1 uM after 48 hrs by anion-exchange HPLC analysis relative to control2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID476562Inhibition of human thymidylate synthase2010Journal of medicinal chemistry, Feb-25, Volume: 53, Issue:4
Single agents with designed combination chemotherapy potential: synthesis and evaluation of substituted pyrimido[4,5-b]indoles as receptor tyrosine kinase and thymidylate synthase inhibitors and as antitumor agents.
AID1501148Inhibition of AICARFTase/GARFTase in human KB cells assessed as reduction in cell proliferation at 1 to 10 nM after 72 hrs in presence of excess of adenosine by MTT assay2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID1053451Growth inhibition of Chinese hamster D4 cells after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID1079937Severe hepatitis, defined as possibly life-threatening liver failure or through clinical observations. Value is number of references indexed. [column 'MASS' in source]
AID1164836Inhibition of FRbeta (unknown origin) expressed in Chinese hamster D4 cells assessed as cell growth inhibition incubated up to 96 hrs in presence of 200 nM folic acid by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID1360122Antiproliferative activity against human OVCAR3 cells after 24 hrs by MTT assay2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1236757Antiproliferative activity against human T47D cells at 100 ug/ml after 48 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1164839Cytotoxicity against human KB cells assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID212657Compound was evaluated for competitive inhibition of recombinant mouse thymidylate synthase1992Journal of medicinal chemistry, Nov-13, Volume: 35, Issue:23
A dideazatetrahydrofolate analogue lacking a chiral center at C-6, N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5- yl)ethyl]benzoyl]-L-glutamic acid, is an inhibitor of thymidylate synthase.
AID1062558Cytotoxicity against chinese hamster PC43-10 cells expressing human RFC after 96 hrs by CellTitre-Blue fluorescence assay2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID211446Inhibitory concentration against isolated Escherichia coli Thymidylate synthase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID341471Selectivity for Escherichia coli dihydrofolate reductase over human dihydrofolate reductase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID391733Inhibition of Escherichia coli thymidylate synthase2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID282789Inhibition of Escherichia coli thymidylate synthase2005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
Synthesis of N-{4-[(2,4-diamino-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid and N-{4-[(2-amino-4-oxo-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid as dual inhibitors of dihydrofol
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).
AID241776Inhibition of Lactobacillus casei dihydrofolate reductase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID671163Inhibition of human thymidylate synthase2012Bioorganic & medicinal chemistry, Jul-15, Volume: 20, Issue:14
Novel tricyclic indeno[2,1-d]pyrimidines with dual antiangiogenic and cytotoxic activities as potent antitumor agents.
AID1219363Elimination rate constant mouse brain at 0.3 uCi/ml2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID282386Inhibition of human thymidylate synthase2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Benzoyl ring halogenated classical 2-amino-6-methyl-3,4-dihydro-4-oxo-5-substituted thiobenzoyl-7H-pyrrolo[2,3-d]pyrimidine antifolates as inhibitors of thymidylate synthase and as antitumor agents.
AID1219358Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence 100 mM probenecid OAT inhi2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
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.
AID341428Inhibition of human thymidylate synthase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID1380199Binding affinity to human FR-beta receptor expressed in Chinese hamster D4 cells assessed as antiproliferative activity measured as reduction in cell viability after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1513016Antiproliferative activity against FRalpha knockout human IGROV1 KD10 cells after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
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]
AID1380195Binding affinity to human RFC expressed in Chinese hamster PC43-10 cells assessed as antiproliferative activity measured as reduction in cell viability after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1079934Highest frequency of acute liver toxicity observed during clinical trials, expressed as a percentage. [column '% AIGUE' in source]
AID260046Inhibition of DHFR from Escherichia coli2006Journal of medicinal chemistry, Feb-09, Volume: 49, Issue:3
Dual inhibitors of thymidylate synthase and dihydrofolate reductase as antitumor agents: design, synthesis, and biological evaluation of classical and nonclassical pyrrolo[2,3-d]pyrimidine antifolates(1).
AID282787Inhibition of Toxoplasma gondii DHFR2005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
Synthesis of N-{4-[(2,4-diamino-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid and N-{4-[(2-amino-4-oxo-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid as dual inhibitors of dihydrofol
AID1663359Inhibition of DHFR in human KB cells expressing RFC/FRalpha/PCFT assessed as reduction in cell growth after 96 hrs in presence of folic acid by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1398148Effect on TS protein expression in human IGROV1 cells by Western blot analysis2018Journal of medicinal chemistry, 08-23, Volume: 61, Issue:16
Conformational Propensity and Biological Studies of Proline Mutated LR Peptides Inhibiting Human Thymidylate Synthase and Ovarian Cancer Cell Growth.
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.
AID282786Inhibition of Escherichia coli DHFR2005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
Synthesis of N-{4-[(2,4-diamino-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid and N-{4-[(2-amino-4-oxo-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid as dual inhibitors of dihydrofol
AID1062533Induction of apoptosis in human KB cells assessed as viable cells at 1 uM after 96 hrs by Annexin V-FITC/7-AAD staining-based flow cytometry (RVB = 89.6%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1663353Inhibition of DHFR in Chinese Hamster MTXRII-OuaR2-4/FRalpha-null R2 cells assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID241657Inhibition of Escherichia coli thymidylate synthetase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID238909Inhibition of human thymidylate synthetase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1062555Cytotoxicity against chinese hamster RT16 cells expressing human FRalpha after 96 hrs by CellTitre-Blue fluorescence assay2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID621128Antiproliferative activity against chinese hamster R2 cells expressing human PCFT assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1663362Inhibition of DHFR in human KB cells expressing RFC/FRalpha/PCFT assessed as reduction in cell growth after 96 hrs in presence of thymidine by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1219373Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID459863Displacement of [3H]folic acid from human FRalpha expressed in Chinese hamster RT16 cells relative to unlabeled folic acid2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID459869Inhibition of GARFtase in human KB cells assessed as [14C]glycine incorporation in to [14C]FGAR in folate free RPMI medium with 2 nM LCV by in-situassay2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID341462Inhibition of human dihydrofolate reductase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID621175Antiproliferative activity against chinese hamster R2 cells expressing human PCFT assessed as growth inhibition at <50 nM in the presence of 10 uM thymidine2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID422635Inhibition of human recombinant dihydrofolate reductase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID1197489Inhibition of GARFTase in human KB cells expressing RFC/FRalpha/PCFT assessed as incorporation of [U-14C]-glycine into [14C]-formyl glycinamide ribonucleotide incubated for 1 hr followed by [U-14C]-glycine addition measured after 16 hrs2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID1631995Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at pH 5.5 measured after 5 mins by Dixon plot analysis2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesi
AID1562583Inhibition of colony formation in human KB cells at 100 nM after 10 to 14 days by methylene blue staining based assay2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID1252439Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as intracellular drug transport rate at 37 degC at pH 5.5 measured over 2 mins by Lineweaver-Burk plot analysis2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID1219354Brain efflux index in Mrp2 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 60 mins relative to wild type2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1219368Drug uptake in mouse brain ipsilateral cerebrum assessed as radiolabelled compound after 30 mins at 0.1 uM, intracerebral microinjection2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1164840Cytotoxicity against human KB cells assessed as cell growth inhibition incubated up to 96 hrs in presence of 200 nM folic acid by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID1562588Antiproliferative activity against human KB cells in presence of adenosine2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID362430Antiproliferative activity against human RFC and FRalpha expressing human IGROV1 cells in presence of folic acid2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1663354Inhibition of DHFR in Chinese Hamster PC43-10 cells expressing human RFC assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1219367Elimination half life in mouse brain at 10 nCi/ml, icv2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID391736Inhibition of Escherichia coli DHFR2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID1062537Cell cycle arrest in human KB cells assessed as accumulation at G2/M phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometry (Rvb = 13.2%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1062538Cell cycle arrest in human KB cells assessed as accumulation at S phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometry (Rvb = 15.9%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1663355Inhibition of DHFR in Chinese Hamster RT16 cells expressing human FRalpha assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1053447Growth inhibition of human KB cells expressing human RFC/FRalpha/PCFT after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID1360125Antiproliferative activity against human A549 cells at 0.512 uM after 24 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1053452Growth inhibition of Chinese hamster RT16 cells after 96 hrs by CellTiter-blue assay in presence of folic acid2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID770036Inhibition of Cryptosporidium hominis thymidylate synthase2013Bioorganic & medicinal chemistry letters, Oct-01, Volume: 23, Issue:19
Substituted pyrrolo[2,3-d]pyrimidines as Cryptosporidium hominis thymidylate synthase inhibitors.
AID605430Growth inhibition of human A549 cells assessed as cell growth at 6.4 uM after 72 hrs by fluorometric microculture cytotoxicity assay in presence of 20 uM thymidine relative to untreated control2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
Antifolate and antiproliferative activity of 6,8,10-triazaspiro[4.5]deca-6,8-dienes and 1,3,5-triazaspiro[5.5]undeca-1,3-dienes.
AID513674Inhibition of thymine synthase2010Journal of medicinal chemistry, Sep-23, Volume: 53, Issue:18
Novel approaches for targeting thymidylate synthase to overcome the resistance and toxicity of anticancer drugs.
AID349814Inhibition of mouse recombinant GARFTase2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Synthesis and biological activity of a novel series of 6-substituted thieno[2,3-d]pyrimidine antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors over the reduced folate carrier and proton-coupled folate transpo
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).
AID1062542Cytotoxicity against human KB cells expressing human RFC/FRalpha/PCFT assessed as depletion of ATP pools at 1 uM after 24 hrs by ion-pair HPLC analysis2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1894188Inhibition of thymidylate synthase (unknown origin) assessed as inhibition constant2021European journal of medicinal chemistry, Mar-15, Volume: 214FDA-approved pyrimidine-fused bicyclic heterocycles for cancer therapy: Synthesis and clinical application.
AID1236754Antiproliferative activity against human HeLa cells at 100 ug/ml after 48 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID240801Inhibitory concentration against Escherichia coli thymidylate synthase2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Novel 2-amino-4-oxo-5-arylthio-substituted-pyrrolo[2,3-d]pyrimidines as nonclassical antifolate inhibitors of thymidylate synthase.
AID1236752Antiproliferative activity against human A549 cells at 100 ug/ml after 72 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1380194Binding affinity to human FR-beta receptor expressed in Chinese hamster D4 cells assessed as antiproliferative activity measured as reduction in cell viability after 96 hrs in presence of folic acid by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID362426Antiproliferative activity against human FRbeta expressing Chinese hamster D4 cells in presence of folic acid2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1062553Cytotoxicity against chinese hamster R2(VC) cells after 96 hrs by CellTitre-Blue fluorescence assay2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID312256Inhibition of human recombinant DHFR2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID457194Inhibition of Toxoplasma gondii TS at 30 degC under pH 7.4 by spectrophotometry2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1360126Antiproliferative activity against human A549 cells at 1.28 uM after 24 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1501162Cell cycle arrest in human KB cells assessed as accumulation at G1 phase at 10 uM after 96 hrs by propidium iodide staining based flow cytometry (Rvb = 54.75%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID211799Inhibitory concentration against isolated Lactobacillus casei Thymidylate synthase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID1380196Cytotoxicity in RFC-null Chinese hamster R2 cells assessed as reduction in cell viability measured after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID459866Displacement of [3H]MTX from human PCFT expressed in Chinese hamster R2 cells at pH 5.5 by Dixon plot2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID1360130Selectivity index, ratio of CC50 for HPAEpiC to IC50 for human A549 cells2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1380211Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at 10 uM at pH 6.8 by scintillation counting method relative to control2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID312255Inhibition of Toxoplasma gondii thymidylate synthase2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID613094Inhibition of Escherichia coli TS assessed as oxidation of tetrahydrofolate to dihydrofolate after 2 to 12 mins by spectrophotometry2011Bioorganic & medicinal chemistry, Jun-01, Volume: 19, Issue:11
Design, synthesis, biological evaluation and X-ray crystal structure of novel classical 6,5,6-tricyclic benzo[4,5]thieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1763184Inhibition of human thymidylate synthase expressed in Escherichia coli using 6R,S-tetrahydrofolate and dUMP as substrate by spectrophotometric analysis2021Bioorganic & medicinal chemistry, 05-15, Volume: 38Design, synthesis and molecular docking studies of thymol based 1,2,3-triazole hybrids as thymidylate synthase inhibitors and apoptosis inducers against breast cancer cells.
AID1079946Presence of at least one case with successful reintroduction. [column 'REINT' in source]
AID1512995Binding affinity to human FRbeta expressed in Chinese hamster D4 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1360129Antiproliferative activity against human A549 cells at 20 uM after 24 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID282387Inhibition of Escherichia coli thymidylate synthase2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Benzoyl ring halogenated classical 2-amino-6-methyl-3,4-dihydro-4-oxo-5-substituted thiobenzoyl-7H-pyrrolo[2,3-d]pyrimidine antifolates as inhibitors of thymidylate synthase and as antitumor agents.
AID1198265Cytotoxicity against human HepG2 cells assessed as growth inhibition after 72 hrs by MTT assay2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID1219366Drug elimination in mouse brain at 10 nCi/ml, icv2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1198281Cell cycle arrest in human KB cells assessed as accumulation at S phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometric analysis (Rvb = 13.6%)2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID740237Inhibition of Escherichia coli thymidylate synthase by spectrophotometric analysis2013Bioorganic & medicinal chemistry, Apr-15, Volume: 21, Issue:8
Novel 1,3,4-oxadiazole thioether derivatives targeting thymidylate synthase as dual anticancer/antimicrobial agents.
AID282571Inhibition of human recombinant DHFR2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis of classical, three-carbon-bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1297309Cell cycle arrest in human SW620 cells assessed as accumulation at G2 phase at 10 uM after 96 hrs by propidium iodide staining based flow cytometry in presence of leucovorin (Rvb = 10.3 %)2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID1501161Cell cycle arrest in human KB cells assessed as accumulation at G2 phase at 10 uM after 96 hrs by propidium iodide staining based flow cytometry (Rvb = 9.59%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID621180Decrease in intracellular ATP concentration in chinese hamster R2 cells expressing human PCFT at 1 uM after 24 hrs by HPLC relative to control at pH 6.82011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1053448Growth inhibition of Chinese hamster R2(VC) cells after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID1198269Inhibition of GARFTase/AICARFTase in human KB cells assessed as cell growth inhibition in presence of 10 uM thymidine and 60 uM adenosine2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID1380212Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at pH 5.5 measured after 2 mins by Dixon plot analysis2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID57444Binding affinity against dihydrofolate reductase1994Journal of medicinal chemistry, May-27, Volume: 37, Issue:11
Synthesis and antitumor activities of novel 6-5 fused ring heterocycle antifolates: N-[4-[omega-(2-amino-4-substituted-6,7-dihydrocyclopenta [d]pyrimidin-5-yl)alkyl]benzoyl]-L-glutamic acids.
AID1513009Inhibition of GARFTase in human IGROV1 cells assessed as decrease in incorporation of [14C(U)glycine into [14C]formyl GAR formation after 24 hrs2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1236747Antiproliferative activity against African green monkey Vero cells at 100 ug/ml after 24 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1053453Growth inhibition of Chinese hamster RT16 cells after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID621136Inhibition of human RFC-mediated [3H]MTX uptake in chinese hamster PC43-10 cells at 10 uM after 2 mins relative to control2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1663363Inhibition of DHFR in human KB cells expressing RFC/FRalpha/PCFT assessed as reduction in cell growth after 96 hrs in presence of glycine by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID362429Antiproliferative activity against human RFC and FRalpha expressing human IGROV1 cells2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID312257Inhibition of Escherichia coli DHFR2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID671165Inhibition of Toxoplasma gondii thymidylate synthase2012Bioorganic & medicinal chemistry, Jul-15, Volume: 20, Issue:14
Novel tricyclic indeno[2,1-d]pyrimidines with dual antiangiogenic and cytotoxic activities as potent antitumor agents.
AID212126Inhibitory activity against recombinant Pneumocystis carinii TS2001Journal of medicinal chemistry, Jun-07, Volume: 44, Issue:12
Synthesis, antifolate, and antitumor activities of classical and nonclassical 2-amino-4-oxo-5-substituted-pyrrolo[2,3-d]pyrimidines.
AID57759Inhibitory concentration against isolated Lactobacillus casei Dihydrofolate reductase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID457193Inhibition of Escherichia coli TS at 30 degC under pH 7.4 by spectrophotometry2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1360128Antiproliferative activity against human A549 cells at 8 uM after 24 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID211645Compound was evaluated as inhibitor of Lactobacillus casei thymidylate synthase2003Journal of medicinal chemistry, Feb-13, Volume: 46, Issue:4
Design, synthesis, and biological activities of classical N-[4-[2-(2-amino-4-ethylpyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid and its 6-methyl derivative as potential dual inhibitors of thymidylate synthase and dihydrofolate reductase and
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).
AID605431Growth inhibition of human A549 cells assessed as cell growth at 6.4 uM after 72 hrs by fluorometric microculture cytotoxicity assay in presence of 100 uM hypoxanthine and 20 uM thymidine relative to untreated control2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
Antifolate and antiproliferative activity of 6,8,10-triazaspiro[4.5]deca-6,8-dienes and 1,3,5-triazaspiro[5.5]undeca-1,3-dienes.
AID211439Compound was evaluated as inhibitor of Escherichia coli thymidylate synthase2003Journal of medicinal chemistry, Feb-13, Volume: 46, Issue:4
Design, synthesis, and biological activities of classical N-[4-[2-(2-amino-4-ethylpyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid and its 6-methyl derivative as potential dual inhibitors of thymidylate synthase and dihydrofolate reductase and
AID1079943Malignant tumor, proven histopathologically. Value is number of references indexed. [column 'T.MAL' in source]
AID1062524Cell cycle arrest in human KB cells assessed as accumulation at G1/G0 phase at 1 uM after 48 hrs by propidium iodide staining-based flow cytometry relative to control2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1236748Antiproliferative activity against African green monkey Vero cells at 100 ug/ml after 48 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1512998Binding affinity to human PCFT4 expressed in human HeLa R1-11 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1062527Inhibition of AICARFTase in human KB cells assessed as accumulation of ZMP after 48 hrs by HPLC analysis2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID362440Induction of apoptotic activity in human KB cells at 1 uM after 24 hrs2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID695098Inhibition of [3H]MTX transport at human PCFT expressed in Chinese hamster R2 cells at pH 5.5 by Dixon plot2012Journal of medicinal chemistry, Feb-23, Volume: 55, Issue:4
Synthesis and biological activity of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl regioisomers as inhibitors of de novo purine biosynthesis with selectivity for cellular uptake by high affinity folate receptors and the proton-coupled folate transporter
AID513675Inhibition of DHFR2010Journal of medicinal chemistry, Sep-23, Volume: 53, Issue:18
Novel approaches for targeting thymidylate synthase to overcome the resistance and toxicity of anticancer drugs.
AID1062546Cytotoxicity against human KB cells expressing human RFC/FRalpha/PCFT after 96 hrs by CellTitre-Blue fluorescence assay in presence of thymidine/adenosine2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID613095Inhibition of Toxoplasma gondii TS assessed as oxidation of tetrahydrofolate to dihydrofolate after 2 to 12 mins by spectrophotometry2011Bioorganic & medicinal chemistry, Jun-01, Volume: 19, Issue:11
Design, synthesis, biological evaluation and X-ray crystal structure of novel classical 6,5,6-tricyclic benzo[4,5]thieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
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).
AID1236751Antiproliferative activity against human A549 cells at 100 ug/ml after 48 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID362428Antiproliferative activity against human RFC and FRalpha expressing human KB cells in presence of folic acid2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1079945Animal toxicity known. [column 'TOXIC' in source]
AID476563Inhibition of Escherichia coli thymidylate synthase2010Journal of medicinal chemistry, Feb-25, Volume: 53, Issue:4
Single agents with designed combination chemotherapy potential: synthesis and evaluation of substituted pyrimido[4,5-b]indoles as receptor tyrosine kinase and thymidylate synthase inhibitors and as antitumor agents.
AID1513005Inhibition of human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as reduction in [3H]MTX uptake at pH 5.5 at 10 uM measured over 2 mins relative to control2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1062534Induction of apoptosis in human KB cells assessed as necrotic cells at 1 uM after 96 hrs by Annexin V-FITC/7-AAD staining-based flow cytometry (RVB = 7.80%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID240604Inhibitory concentration against human thymidylate synthase2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Novel 2-amino-4-oxo-5-arylthio-substituted-pyrrolo[2,3-d]pyrimidines as nonclassical antifolate inhibitors of thymidylate synthase.
AID1380217Antitumor activity against human IGROV1 cells xenografted in folate-deficient diet ICR SCID mouse assessed as tumor growth delay time at 8.1 mg/kg, iv administered every 2 days for 4 times starting on day 3 measured twice per week2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID457197Inhibition of Toxoplasma gondii DHFR at 30 degC under pH 7.4 by spectrophotometry2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID422632Inhibition of human thymidylate synthase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID1513018Antiproliferative activity against human IGROV1 cells harboring non targeted control shRNA sequence after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1197480Binding affinity to human FRalpha expressed in Chinese hamster RT16 cells assessed as cell growth inhibition after 96 hrs by CellTiter-Blue assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Novel 5-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and as potential antitumor agents.
AID1164837Inhibition of PCFT (unknown origin) expressed in Chinese hamster R2/PCFT4 cells assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID211442Inhibitory activity against Escherichia coli thymidylate synthase2001Journal of medicinal chemistry, Jun-07, Volume: 44, Issue:12
Synthesis, antifolate, and antitumor activities of classical and nonclassical 2-amino-4-oxo-5-substituted-pyrrolo[2,3-d]pyrimidines.
AID1501169Induction of apoptosis in human KB cells assessed as necrotic cells at 10 uM after 48 hrs by Annexin V-FITC/propidium iodide staining based flow cytometry (Rvb = 3%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID1198268Inhibition of GARFTase/AICARFTase in human KB cells assessed as cell growth inhibition in presence of 320 uM AICA2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID349815Inhibition of GARFTase in human KB cells assessed as inhibition of incorporation of [14C]glycine into [14C]formyl GAR after 30 mins in presence of azaserine2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Synthesis and biological activity of a novel series of 6-substituted thieno[2,3-d]pyrimidine antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors over the reduced folate carrier and proton-coupled folate transpo
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]
AID1380202Antiproliferative activity against human KB cells expressing human RFC/FR-alpha/PCFT assessed as reduction in cell viability measured after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1380210Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at 1 uM at pH 6.8 by scintillation counting method relative to control2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID695100Ratio of Ki for human PCFT at pH 6.8 to Ki for human PCFT at pH 5.5 by [3H]MTX transport assay2012Journal of medicinal chemistry, Feb-23, Volume: 55, Issue:4
Synthesis and biological activity of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl regioisomers as inhibitors of de novo purine biosynthesis with selectivity for cellular uptake by high affinity folate receptors and the proton-coupled folate transporter
AID1219360Volume of distribution in mouse brain per gm of tissue at 0.3 uCi/ml after 1 to 2 hrs by in vitro brain slice uptake technique2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID56187Inhibitory activity against Toxoplasma gondii dihydrofolate reductase2001Journal of medicinal chemistry, Jun-07, Volume: 44, Issue:12
Synthesis, antifolate, and antitumor activities of classical and nonclassical 2-amino-4-oxo-5-substituted-pyrrolo[2,3-d]pyrimidines.
AID1053450Growth inhibition of Chinese hamster D4 cells after 96 hrs by CellTiter-blue assay in presence of folic acid2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
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).
AID422636Inhibition of Escherichia coli dihydrofolate reductase2009Journal of medicinal chemistry, Aug-13, Volume: 52, Issue:15
Design, synthesis, and X-ray crystal structure of classical and nonclassical 2-amino-4-oxo-5-substituted-6-ethylthieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors and as potential antitumor agents.
AID695099Inhibition of [3H]MTX transport at human PCFT expressed in Chinese hamster R2 cells at pH 6.8 by Dixon plot2012Journal of medicinal chemistry, Feb-23, Volume: 55, Issue:4
Synthesis and biological activity of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl regioisomers as inhibitors of de novo purine biosynthesis with selectivity for cellular uptake by high affinity folate receptors and the proton-coupled folate transporter
AID1512996Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1360124Antiproliferative activity against human MDA-MB-231 cells after 24 hrs by MTT assay2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID621125Antiproliferative activity against chinese hamster RT16 cells expressing human FRalpha assessed as reduction of viable cells after 96 hrs in the presence of 200 nM folic acid2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
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).
AID212297Inhibitory activity against human thymidylate synthase2001Journal of medicinal chemistry, Jun-07, Volume: 44, Issue:12
Synthesis, antifolate, and antitumor activities of classical and nonclassical 2-amino-4-oxo-5-substituted-pyrrolo[2,3-d]pyrimidines.
AID621129Antiproliferative activity against chinese hamster R2(VC) cells expressing human PCFT assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID312254Inhibition of Escherichia coli thymidylate synthase2008Journal of medicinal chemistry, Jan-10, Volume: 51, Issue:1
Design, synthesis, and biological evaluation of classical and nonclassical 2-amino-4-oxo-5-substituted-6-methylpyrrolo[3,2-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1513033Inhibition of human RFC2 expressed in human HeLa R1-11 cells assessed as reduction in [3H]MTX uptake at pH 7.2 at 1 uM measured over 2 mins relative to control2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1501143Antiproliferative activity against human MCF7 cells in folate free medium after 72 hrs in presence of leucovorin by MTT assay2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID621177Antiproliferative activity against chinese hamster R2 cells expressing human PCFT assessed as growth inhibition in the presence of 60 uM adenosine2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1663357Inhibition of DHFR in Chinese Hamster MTXRII-OuaR2-4 R2 cells expressing human PCFT assessed as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID1062554Cytotoxicity against chinese hamster R2 cells expressing human PCFT4 after 96 hrs by CellTitre-Blue fluorescence assay2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1663360Inhibition of DHFR in human KB cells expressing RFC/FRalpha/PCFT assessed as reduction in cell growth after 96 hrs in presence of adenosine by Cell-Titer Blue assay2020Bioorganic & medicinal chemistry, 06-15, Volume: 28, Issue:12
Design, synthesis and biological evaluation of novel pyrrolo[2,3-d]pyrimidine as tumor-targeting agents with selectivity for tumor uptake by high affinity folate receptors over the reduced folate carrier.
AID282389Inhibition of Escherichia coli DHFR2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Benzoyl ring halogenated classical 2-amino-6-methyl-3,4-dihydro-4-oxo-5-substituted thiobenzoyl-7H-pyrrolo[2,3-d]pyrimidine antifolates as inhibitors of thymidylate synthase and as antitumor agents.
AID241751Inhibition of Lactobacillus casei thymidylate synthetase at 37 degree C pH 7.42005Journal of medicinal chemistry, Aug-11, Volume: 48, Issue:16
Synthesis of classical, four-carbon bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID621133Antiproliferative activity against human IGROV1 cells expressing human RFC, FRalpha and PCFT assessed as reduction of viable cells after 96 hrs in the presence of 200 nM folic acid2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID362433Inhibition of human RFC-mediated [3H]MTX transport in Chinese hamster PC43-10 cells at 10 uM2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID57235Compound was evaluated as inhibitor of Escherichia coli Dihydrofolate reductase2003Journal of medicinal chemistry, Feb-13, Volume: 46, Issue:4
Design, synthesis, and biological activities of classical N-[4-[2-(2-amino-4-ethylpyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid and its 6-methyl derivative as potential dual inhibitors of thymidylate synthase and dihydrofolate reductase and
AID1297305Inhibition of thymidylate synthase in human SW620 cells assessed as growth inhibition coincubated with thymidine2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID621178Antiproliferative activity against chinese hamster R2 cells expressing human PCFT assessed as growth inhibition in the presence of 60 uM adenosine and 10 uM thymidine2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1562596Cell cycle arrest in human KB cells assessed as accumulation at G1 phase at 10 uM after 96 hrs by RNase-1 A/propidium iodide staining-based flow cytometric analysis (Rvb = 42.69%)2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID605432Growth inhibition of human A549 cells assessed as cell growth at 6.4 uM after 72 hrs by fluorometric microculture cytotoxicity assay relative to untreated control2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
Antifolate and antiproliferative activity of 6,8,10-triazaspiro[4.5]deca-6,8-dienes and 1,3,5-triazaspiro[5.5]undeca-1,3-dienes.
AID740238Inhibition of human thymidylate synthase by spectrophotometric analysis2013Bioorganic & medicinal chemistry, Apr-15, Volume: 21, Issue:8
Novel 1,3,4-oxadiazole thioether derivatives targeting thymidylate synthase as dual anticancer/antimicrobial agents.
AID1297308Cell cycle arrest in human SW620 cells assessed as accumulation at S phase at 10 uM after 96 hrs by propidium iodide staining based flow cytometry in presence of leucovorin (Rvb = 27.3 %)2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID18269Km was determined1992Journal of medicinal chemistry, Nov-13, Volume: 35, Issue:23
A dideazatetrahydrofolate analogue lacking a chiral center at C-6, N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5- yl)ethyl]benzoyl]-L-glutamic acid, is an inhibitor of thymidylate synthase.
AID1198266Inhibition of GARFTase/AICARFTase in human KB cells assessed as cell growth inhibition in presence of 60 uM adenosine2015European journal of medicinal chemistry, Mar-26, Volume: 93Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidines as potential nonclassical antifolates targeting both thymidylate and purine nucleotide biosynthesis.
AID457196Inhibition of Escherichia coli DHFR at 30 degC under pH 7.4 by spectrophotometry2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1053446Growth inhibition of human KB cells expressing human RFC/FRalpha/PCFT after 96 hrs by CellTiter-blue assay in presence of folic acid2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID457195Inhibition of human DHFR at 30 degC under pH 7.4 by spectrophotometry2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1164838Cytotoxicity against Chinese hamster R2 cells expressing empty vector assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID391735Inhibition of Toxoplasma gondii thymidylate synthase2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID1219376Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence 100 mM probenecid OAT inhi2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1918194Lipophilicity, log D of the compound2022Journal of medicinal chemistry, 11-10, Volume: 65, Issue:21
Identification of Organic Anion Transporter 2 Inhibitors: Screening, Structure-Based Analysis, and Clinical Drug Interaction Risk Assessment.
AID1079949Proposed mechanism(s) of liver damage. [column 'MEC' in source]
AID282576Inhibition of Lactobacillus casei thymidylate synthase2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis of classical, three-carbon-bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1360127Antiproliferative activity against human A549 cells at 3.2 uM after 24 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID1513003Inhibition of human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as reduction in [3H]MTX uptake at pH 5.5 measured over 2 mins by Dixon plot analysis2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1501160Cell cycle arrest in human KB cells assessed as accumulation at S phase at 10 uM after 96 hrs by propidium iodide staining based flow cytometry (Rvb = 35.66%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID457192Inhibition of human thymidylate synthase at 30 degC under pH 7.4 by spectrophotometry2010Bioorganic & medicinal chemistry, Jan-15, Volume: 18, Issue:2
2,4-Diamino-5-methyl-6-substituted arylthio-furo[2,3-d]pyrimidines as novel classical and nonclassical antifolates as potential dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1297294Antiproliferative activity against human KB cells after 72 hrs by MTT assay in presence of leucovorin2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID1079939Cirrhosis, proven histopathologically. Value is number of references indexed. [column 'CIRRH' in source]
AID1062536Induction of apoptosis in human KB cells assessed as early apoptotic cells at 1 uM after 96 hrs by Annexin V-FITC/7-AAD staining-based flow cytometry (Rvb = 0.485%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID362427Antiproliferative activity against human RFC and FRalpha expressing human KB cells2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1513020Antiproliferative activity against wild type human IGROV1 cells after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID212157Compound was evaluated as inhibitor of human thymidylate synthase2003Journal of medicinal chemistry, Feb-13, Volume: 46, Issue:4
Design, synthesis, and biological activities of classical N-[4-[2-(2-amino-4-ethylpyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid and its 6-methyl derivative as potential dual inhibitors of thymidylate synthase and dihydrofolate reductase and
AID391732Inhibition of human thymidylate synthase2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID1164835Inhibition of FRbeta (unknown origin) expressed in Chinese hamster D4 cells assessed as cell growth inhibition incubated up to 96 hrs by Celltiter-blue cell viability assay2014Journal of medicinal chemistry, Oct-09, Volume: 57, Issue:19
Structure-activity profiles of novel 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates with modified amino acids for cellular uptake by folate receptors α and β and the proton-coupled folate transporter.
AID1062540Cell cycle arrest in human KB cells assessed as accumulation at sub-G1 phase at 1 uM after 96 hrs by propidium iodide staining-based flow cytometry (Rvb = 3.5%)2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1513015Selectivity ratio of IC50 for human IGROV1 cells harboring non targeted control shRNA sequence to IC50 for FRalpha knockout human IGROV1 KD4 cells2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID362424Antiproliferative activity against human FRalpha expressing Chinese hamster RT16 cells in presence of folic acid2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID1297296Antiproliferative activity against human A549 cells after 72 hrs by MTT assay in presence of leucovorin2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID1219351Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence 100 mM benzylpenicillin OA2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1562575Antiproliferative activity against human A549 cells assessed as reduction in cell viability after 72 hrs by MTT assay2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID1236753Antiproliferative activity against human HeLa cells at 100 ug/ml after 24 hrs by BrdUrd incorporation assay2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Synthesis, structure elucidation and identification of antiproliferative activities of a novel class of thiophene bioisosteres bearing the privileged 7,8-dihydroimidazo[2,1-c][1,2,4]triazin-4(6H)-one scaffold.
AID1512997Binding affinity to human RFC2 expressed in human HeLa R1-11 cells assessed as antiproliferative activity measured as reduction in cell growth after 96 hrs by Cell-Titer Blue assay2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID260014Inhibition of TS from Escherichia coli2006Journal of medicinal chemistry, Feb-09, Volume: 49, Issue:3
Dual inhibitors of thymidylate synthase and dihydrofolate reductase as antitumor agents: design, synthesis, and biological evaluation of classical and nonclassical pyrrolo[2,3-d]pyrimidine antifolates(1).
AID362438Inhibition of GARFTase in human KB cells assessed as inhibition of [14C]glycine incorporation into [14C]formylGAR in presence of azaserine2008Journal of medicinal chemistry, Aug-28, Volume: 51, Issue:16
Synthesis and discovery of high affinity folate receptor-specific glycinamide ribonucleotide formyltransferase inhibitors with antitumor activity.
AID613097Inhibition of Escherichia coli DHFR by spectrophotometry2011Bioorganic & medicinal chemistry, Jun-01, Volume: 19, Issue:11
Design, synthesis, biological evaluation and X-ray crystal structure of novel classical 6,5,6-tricyclic benzo[4,5]thieno[2,3-d]pyrimidines as dual thymidylate synthase and dihydrofolate reductase inhibitors.
AID1501166Induction of apoptosis in human KB cells assessed as live cells at 10 uM after 48 hrs by Annexin V-FITC/propidium iodide staining based flow cytometry (Rvb = 74.6%)2017European journal of medicinal chemistry, Oct-20, Volume: 139Novel 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines as potential antitumor agents with multitargeted inhibition of TS, GARFTase and AICARFTase.
AID459865Displacement of [3H]MTX from human PCFT expressed in Chinese hamster R2 cells at pH 6.8 by Dixon plot2010Journal of medicinal chemistry, Feb-11, Volume: 53, Issue:3
Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduc
AID282575Inhibition of Escherichia coli thymidylate synthase2004Journal of medicinal chemistry, Dec-30, Volume: 47, Issue:27
Synthesis of classical, three-carbon-bridged 5-substituted furo[2,3-d]pyrimidine and 6-substituted pyrrolo[2,3-d]pyrimidine analogues as antifolates.
AID1219359Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence 100 mM benzylpenicillin OA2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1380208Inhibition of [3H]MTX uptake at human PCFT expressed in Chinese hamster R2/PCFT4 cells at 1 uM at pH 5.5 by scintillation counting method relative to control2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID57739Compound was evaluated as inhibitor of Lactobacillus casei Dihydrofolate reductase2003Journal of medicinal chemistry, Feb-13, Volume: 46, Issue:4
Design, synthesis, and biological activities of classical N-[4-[2-(2-amino-4-ethylpyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-l-glutamic acid and its 6-methyl derivative as potential dual inhibitors of thymidylate synthase and dihydrofolate reductase and
AID1513014Selectivity ratio of IC50 for human IGROV1 cells harboring non targeted control shRNA sequence to IC50 for FRalpha knockout human IGROV1 KD10 cells2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID1380198Binding affinity to human FR-alpha receptor expressed in Chinese hamster RT16 cells assessed as antiproliferative activity measured as reduction in cell viability after 96 hrs in presence of folic acid by Cell-Titer Blue assay2018Journal of medicinal chemistry, 03-08, Volume: 61, Issue:5
Tumor Targeting with Novel Pyridyl 6-Substituted Pyrrolo[2,3- d]Pyrimidine Antifolates via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of De Novo Purine Nucleotide Biosynthesis.
AID1219369Drug uptake in mouse brain ipsilateral cerebrum assessed as radiolabelled compound after 30 mins at 0.1 uM, intracerebral microinjection in presence of 1 mM unlabelled compound2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1562589Antiproliferative activity against human KB cells in presence of AICA2019European journal of medicinal chemistry, Sep-15, Volume: 178Targeting dihydrofolate reductase: Design, synthesis and biological evaluation of novel 6-substituted pyrrolo[2,3-d]pyrimidines as nonclassical antifolates and as potential antitumor agents.
AID621124Antiproliferative activity against chinese hamster RT16 cells expressing human FRalpha assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID1079948Times to onset, minimal and maximal, observed in the indexed observations. [column 'DELAI' in source]
AID1439756Inhibition of human thymidylate synthase using dUMP/(6R,S)-tetrahydrofolate as substrate/co-factor by spectrophotometric method2017Bioorganic & medicinal chemistry letters, 04-01, Volume: 27, Issue:7
Synthesis and evaluation of 5-(arylthio)-9H-pyrimido[4,5-b]indole-2,4-diamines as receptor tyrosine kinase and thymidylate synthase inhibitors and as antitumor agents.
AID1053454Growth inhibition of Chinese hamster MTXRII-OuaR2-4 cells after 96 hrs by CellTiter-blue assay2013Journal of medicinal chemistry, Nov-14, Volume: 56, Issue:21
Tumor-targeting with novel non-benzoyl 6-substituted straight chain pyrrolo[2,3-d]pyrimidine antifolates via cellular uptake by folate receptor α and inhibition of de novo purine nucleotide biosynthesis.
AID1513002Inhibition of human RFC2 expressed in human HeLa R1-11 cells assessed as reduction in [3H]MTX uptake at pH 7.2 at 10 uM measured over 2 mins relative to control2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID282785Inhibition of human DHFR2005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
Synthesis of N-{4-[(2,4-diamino-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid and N-{4-[(2-amino-4-oxo-5-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-6-yl)thio]benzoyl}-L-glutamic acid as dual inhibitors of dihydrofol
AID57240Inhibitory activity against recombinant Escherichia coli dihydrofolate reductase2001Journal of medicinal chemistry, Jun-07, Volume: 44, Issue:12
Synthesis, antifolate, and antitumor activities of classical and nonclassical 2-amino-4-oxo-5-substituted-pyrrolo[2,3-d]pyrimidines.
AID1297307Inhibition of thymidylate synthase/GARFTase/AICARFTase in human SW620 cells assessed as growth inhibition coincubated with thymidine and adenosine2016European journal of medicinal chemistry, Jun-10, Volume: 115Design, synthesis and biological evaluation of 6-substituted pyrrolo[2,3-d]pyrimidines as dual inhibitors of TS and AICARFTase and as potential antitumor agents.
AID1062552Cytotoxicity against human KB cells expressing human RFC/FRalpha/PCFT after 96 hrs by CellTitre-Blue fluorescence assay2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID212843Binding affinity against TS1994Journal of medicinal chemistry, May-27, Volume: 37, Issue:11
Synthesis and antitumor activities of novel 6-5 fused ring heterocycle antifolates: N-[4-[omega-(2-amino-4-substituted-6,7-dihydrocyclopenta [d]pyrimidin-5-yl)alkyl]benzoyl]-L-glutamic acids.
AID621132Antiproliferative activity against human IGROV1 cells expressing human RFC, FRalpha and PCFT assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID391737Inhibition of Toxoplasma gondii DHFR2008Journal of medicinal chemistry, Sep-25, Volume: 51, Issue:18
Potent dual thymidylate synthase and dihydrofolate reductase inhibitors: classical and nonclassical 2-amino-4-oxo-5-arylthio-substituted-6-methylthieno[2,3-d]pyrimidine antifolates.
AID1709483Inhibition of human full length N-terminal His-tagged GARFTase expressed in Chinese Hamster MTXRII-OuaR2-4 R2 cells assessed as reduction in THF formation using 10-CHOTHF as substrate by spectrophotometric method2021Bioorganic & medicinal chemistry, 05-01, Volume: 37Discovery of 6-substituted thieno[2,3-d]pyrimidine analogs as dual inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis in folate receptor
AID621126Antiproliferative activity against chinese hamster D4 cells expressing human FRbeta assessed as reduction of viable cells after 96 hrs2011Journal of medicinal chemistry, Oct-27, Volume: 54, Issue:20
Synthesis, biological, and antitumor activity of a highly potent 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolate inhibitor with proton-coupled folate transporter and folate receptor selectivity over the reduced folate carrier that inhibits β-gl
AID240641Inhibitory concentration against human dihydrofolate reductase2005Bioorganic & medicinal chemistry letters, May-02, Volume: 15, Issue:9
Novel 2-amino-4-oxo-5-arylthio-substituted-pyrrolo[2,3-d]pyrimidines as nonclassical antifolate inhibitors of thymidylate synthase.
AID1513001Selectivity ratio of IC50 for human RFC2 expressed in human HeLa R1-11 cells to IC50 for human FR2 expressed in human HeLa R1-11 cells2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Fluorine-Substituted Pyrrolo[2,3- d]Pyrimidine Analogues with Tumor Targeting via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis.
AID212677Inhibitory concentration against isolated rat thymidylate synthase2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID1360121Antiproliferative activity against human A549 cells after 24 hrs by MTT assay2018European journal of medicinal chemistry, Jun-25, Volume: 154Design, synthesis and biological evaluation of N-phenyl-(2,4-dihydroxypyrimidine-5-sulfonamido)benzoyl hydrazide derivatives as thymidylate synthase (TS) inhibitors and as potential antitumor drugs.
AID341439Inhibition of Escherichia coli thymidylate synthase2008Journal of medicinal chemistry, Aug-14, Volume: 51, Issue:15
The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents.
AID55842Inhibitory concentration against isolated Pneumocystis carinii DHFR (Dihydrofolate reductase)2000Journal of medicinal chemistry, Oct-19, Volume: 43, Issue:21
Design, synthesis, and X-ray crystal structure of a potent dual inhibitor of thymidylate synthase and dihydrofolate reductase as an antitumor agent.
AID1062551Cytotoxicity against human KB cells expressing human RFC/FRalpha/PCFT after 96 hrs by CellTitre-Blue fluorescence assay in presence of folic acid2013Journal of medicinal chemistry, Dec-27, Volume: 56, Issue:24
Discovery of 5-substituted pyrrolo[2,3-d]pyrimidine antifolates as dual-acting inhibitors of glycinamide ribonucleotide formyltransferase and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis: implic
AID1252436Binding affinity to human PCFT expressed in Chinese hamster R2/PCFT4 cells assessed as intracellular drug level at 0.5 uM at 37 degC at pH 5.5 measured over 5 mins2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID56964Inhibitory activity against human dihydrofolate reductase2001Journal of medicinal chemistry, Jun-07, Volume: 44, Issue:12
Synthesis, antifolate, and antitumor activities of classical and nonclassical 2-amino-4-oxo-5-substituted-pyrrolo[2,3-d]pyrimidines.
AID1219356Brain efflux index in Bcrp1 deficient mouse assessed as transport of compound from ipsilateral cerebrum to the circulating blood across the blood-brain barrier at 10 nCi/ml, intracerebral microinjection after 30 mins in presence of 1 mM unlabelled compoun2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate.
AID1252422Selectivity ratio of IC50 for human RFC expressed in Chinese hamster PC43-10 cells assessed as cell growth inhibition to IC50 for human FRbeta expressed in Chinese hamster D4 cells assessed as cell growth inhibition2015Journal of medicinal chemistry, Sep-10, Volume: 58, Issue:17
6-Substituted Pyrrolo[2,3-d]pyrimidine Thienoyl Regioisomers as Targeted Antifolates for Folate Receptor α and the Proton-Coupled Folate Transporter in Human Tumors.
AID977610Experimentally measured binding affinity data (Ki) for protein-ligand complexes derived from PDB2001Journal of molecular biology, Nov-02, Volume: 313, Issue:4
Multi-targeted antifolates aimed at avoiding drug resistance form covalent closed inhibitory complexes with human and Escherichia coli thymidylate synthases.
AID1811Experimentally measured binding affinity data derived from PDB2001Journal of molecular biology, Nov-02, Volume: 313, Issue:4
Multi-targeted antifolates aimed at avoiding drug resistance form covalent closed inhibitory complexes with human and Escherichia coli thymidylate synthases.
AID1321673Antifungal activity against amphotericin-B resistant Candida albicans ATCC 90873 assessed as growth inhibition using alamar blue staining measured after 14 to 16 hrs by microdilution assay
AID1321696Binding affinity to cholesterol containing POPC large unilamellar vescicle in pH 7.4 phosphate buffer by SPR analysis
AID1321677Diffusion coefficient of the compound in 4:1 ratio of DMSO-d6/D2O solvent system at 1.2 mM and 25 degC by 1H NMR DOSY spectroscopy
AID1321691Binding affinity to sodium dodecyl sulphate-d25 micelles at 200 uM by 1H NMR DOSY spectroscopy
AID1321694Induction of membrane damage in POPC/cholesterol GUV model assessed as membrane curvature at 20 uM after 13 to 55 mins by TNM-DCCH staining based confocal microscopic analysis
AID1321679Aqueous diffusion coefficient of the compound in 98:2 ratio of D2O/H2O solvent system at 1.2 mM and 25 degC by 1H NMR DOSY spectroscopy
AID1321674Antifungal activity against Trichophyton assessed as growth inhibition using alamar blue staining measured after 14 to 16 hrs by microdilution assay
AID1321681Binding affinity to sodium dodecyl sulphate-d25 micelles assessed as reduction in compound 1H resonance line broadening at compound:Mn2+ ratio of 2:3 to 10:1 by 1H NMR DOSY spectroscopy
AID1321672Antifungal activity against wild-type Candida albicans ATCC 32354 assessed as growth inhibition using alamar blue staining measured after 14 to 16 hrs by microdilution assay
AID1321675Antifungal activity against Aspergillus assessed as growth inhibition using alamar blue staining measured after 14 to 16 hrs by microdilution assay
AID1321693Binding affinity to cholesterol free DMPC-d54/DHPC-d22 bicelles assessed as amide proton signals measured at 200 uM by 1H NMR spectroscopy
AID1321686Binding affinity to cholesterol containing DMPC-d54/DHPC-d22 bicelles assessed as fluorescence intensity measured at ratio of 2:10 Mn2+ to compound by 1H NMR spectroscopy relative to control
AID1321685Binding affinity to cholesterol containing DMPC-d54/DHPC-d22 bicelles assessed as fluorescence intensity measured at ratio of 1:10 Mn2+ to compound by 1H NMR spectroscopy relative to control
AID1321695Induction of membrane damage in POPC GUV model assessed as membrane curvature at 20 uM up to 55 mins by TNM-DCCH staining based confocal microscopic analysis
AID1321682Binding affinity to sodium dodecyl sulphate-d25 micelles assessed as increase in compound 1H signal intensity at compound:Mn2+ ratio of 2:3 to 10:1 by 1H NMR DOSY spectroscopy
AID1321678Diffusion coefficient of the compound in 4:1 ratio of DMSO-d6/D2O solvent system at 0.2 mM and 25 degC by 1H NMR DOSY spectroscopy
AID1321676Cytotoxicity against mouse P388 cells
AID1321687Binding affinity to cholesterol containing DMPC-d54/DHPC-d22 bicelles assessed as fluorescence intensity measured at ratio of 4:10 Mn2+ to compound by 1H NMR spectroscopy relative to control
AID1321688Binding affinity to DMPC-d54/DHPC-d22 bicelles assessed as fluorescence intensity measured at ratio of 4:10 Mn2+ to compound by 1H NMR spectroscopy relative to control
AID1321692Binding affinity to cholesterol free DMPC-d54/DHPC-d22 bicelles assessed as broadening of aromatic/olefinic signals at 6.5 to 7.6 ppm measured at 200 uM by 1H NMR spectroscopy
AID1321680Aqueous diffusion coefficient of the compound in 98:2 ratio of D2O/H2O solvent system at 0.2 mM and 25 degC by 1H NMR DOSY spectroscopy
AID1321683Binding affinity to POPC large unilamellar vesicle in pH 7.4 phosphate buffer by SPR analysis
AID1321684Binding affinity to cholesterol containing DMPC-d54/DHPC-d22 bicelles assessed as broadening of aromatic/olefinic signals at 6.5 to 7.6 ppm measured at 200 uM by 1H NMR spectroscopy
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (2,414)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's46 (1.91)18.2507
2000's521 (21.58)29.6817
2010's1424 (58.99)24.3611
2020's423 (17.52)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 84.76

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 Index84.76 (24.57)
Research Supply Index1.79 (2.92)
Research Growth Index4.51 (4.65)
Search Engine Demand Index142.89 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (84.76)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials657 (26.61%)5.53%
Trials0 (0.00%)5.53%
Reviews362 (14.66%)6.00%
Reviews0 (0.00%)6.00%
Case Studies288 (11.66%)4.05%
Case Studies0 (0.00%)4.05%
Observational28 (1.13%)0.25%
Observational0 (0.00%)0.25%
Other1,134 (45.93%)84.16%
Other5 (100.00%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (964)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Phase 1 Dose-Escalation Study to Examine the Safety and Tolerability of IC83/LY2603618 Administered After Pemetrexed 500 mg/m2 Every 21 Days in Patients With Cancer [NCT00415636]Phase 131 participants (Actual)Interventional2007-01-31Completed
A Single-Arm, Multicenter, Open-Label, Phase 2 Study of Pemetrexed/Cisplatin Chemotherapy for Patients With Metastatic/Recurrent Soft Tissue Sarcoma in 4- Independent Histologic Subtypes [NCT04605770]Phase 2164 participants (Anticipated)Interventional2020-12-22Recruiting
A Phase II/III Study of N-803 (ALT-803) Plus Pembrolizumab Versus Standard of Care in Participants With Stage IV or Recurrent Non-Small Cell Lung Cancer Previously Treated With Anti-PD-1 or Anti-PD-L1 Therapy (Lung-MAP Non-Match Sub-Study) [NCT05096663]Phase 2/Phase 382 participants (Actual)Interventional2022-03-15Active, not recruiting
A Phase 3, Randomized, Global Trial of Nivolumab and Epacadostat With Platinum Doublet Chemotherapy Versus Platinum Doublet Chemotherapy in First-line Treatment of Stage IV or Recurrent Non-Small Cell Lung Cancer (NSCLC) [NCT03348904]Phase 32 participants (Actual)Interventional2017-12-27Terminated(stopped due to Study halted prematurely and will not resume; participants are no longer being examined or receiving intervention.)
Phase II, Randomized, Open-Label Trial of Biweekly Pemetrexed Plus Gemcitabine vs. Pemetrexed or Pemetrexed Plus Carboplatin in Relapsed Non Small Cell Lung Cancer After Neoadjuvant or Adjuvant Chemotherapy [NCT00356525]Phase 241 participants (Actual)Interventional2006-09-30Terminated(stopped due to Stopped early due to low enrollment)
Phase I/II Trial of Gemcitabine/Pemetrexed Combination in Patients With Advanced Cutaneous T-Cell Lymphoma [NCT00369629]Phase 114 participants (Actual)Interventional2006-08-28Terminated(stopped due to This was planned as a phase I/II study originally, but due to a lack of funding, the phase II portion was never conducted.)
Trial of Pemetrexed in Combined With Cisplatin for the Treatment of Advanced Breast Cancer [NCT01143974]Phase 428 participants (Actual)Interventional2010-06-30Completed
Phase II Study of Pemetrexed Monotherapy in Patients With Platinum-resistant Squamous Cell Carcinoma of Head and Neck [NCT01333696]Phase 232 participants (Anticipated)Interventional2011-04-30Recruiting
A Phase I/Ib Study of MEK162, a MEK Inhibitor, in Combination With Carboplatin and Pemetrexed in Patients With Non-squamous Carcinoma of the Lung [NCT02185690]Phase 113 participants (Actual)Interventional2018-01-11Completed
A Randomized Phase III Trial of Chemo-Immunotherapy vs Immunotherapy Alone for the Vulnerable Older Adult With Advanced Non-Small Cell Lung Cancer: The ACHIEVE Study [NCT06096844]Phase 3304 participants (Anticipated)Interventional2024-02-16Not yet recruiting
A Randomized, Open-Label, Multicenter, Phase 2, Umbrella Study to Evaluate the Preliminary Efficacy, Safety, and Pharmacodynamics of Tislelizumab Monotherapy and Multiple Tislelizumab-based Immunotherapy Combinations With and Without Chemotherapy as Neoad [NCT05577702]Phase 2120 participants (Anticipated)Interventional2023-02-15Recruiting
A Phase 3, Open-Label, Randomized Study of Amivantamab and Lazertinib in Combination With Platinum-Based Chemotherapy Compared With Platinum-Based Chemotherapy in Patients With EGFR-Mutated Locally Advanced or Metastatic Non-Small Cell Lung Cancer After O [NCT04988295]Phase 3776 participants (Actual)Interventional2021-11-17Active, not recruiting
A Randomized, Open-label Phase 3 Study of Combination Amivantamab and Carboplatin-Pemetrexed Therapy, Compared With Carboplatin-Pemetrexed, in Patients With EGFR Exon 20ins Mutated Locally Advanced or Metastatic Non-Small Cell Lung Cancer [NCT04538664]Phase 3308 participants (Actual)Interventional2020-10-13Active, not recruiting
Randomized Phase III Trial of MEDI4736 (Durvalumab) as Concurrent and Consolidative Therapy or Consolidative Therapy Alone for Unresectable Stage 3 NSCLC [NCT04092283]Phase 3660 participants (Anticipated)Interventional2020-04-29Active, not recruiting
An Open-label Phase 1/1b Study to Evaluate the Safety and Pharmacokinetics of JNJ-73841937 (Lazertinib), a Third Generation EGFR-TKI, as Monotherapy or in Combinations With JNJ-61186372, a Human Bispecific EGFR and cMet Antibody in Participants With Advan [NCT04077463]Phase 1460 participants (Anticipated)Interventional2019-09-04Recruiting
A Phase 1, First-in-Human, Open-Label, Dose Escalation Study of JNJ-61186372, a Human Bispecific EGFR and cMet Antibody, in Subjects With Advanced Non-Small Cell Lung Cancer [NCT02609776]Phase 1751 participants (Actual)Interventional2016-05-24Active, not recruiting
A Phase I/Randomized Phase II Study of Cediranib (NSC#732208) Versus Placebo in Combination With Cisplatin and Pemetrexed in Chemonaive Patients With Malignant Pleural Mesothelioma [NCT01064648]Phase 1/Phase 2117 participants (Actual)Interventional2010-03-15Active, not recruiting
Open-label, Phase 2 Study of Tusamitamab Ravtansine (IBI126) Combined With Sintilimab and Tusamitamab Ravtansine (IBI126) Combined With Sintilimab Plus Platinum-based Chemotherapy and Pemetrexed in Subjects With CEACAM5 Positive Expression Advanced/Metast [NCT05849246]Phase 2130 participants (Anticipated)Interventional2023-05-30Not yet recruiting
Phase II Study of Concurrent Cisplatin/Pemetrexed and RT Followed by Docetaxel in Stage III NSCLC (Non Small Cell Lung Cancer) [NCT00301808]Phase 229 participants (Actual)Interventional2005-11-30Completed
Study of Atezolizumab in Combination With Carboplatin + Paclitaxel +Bevacizumab vs With Pemetrexed + Cisplatin or Carboplatin With Stage IV NON-SQUAMOUS NON-SMALL CELL LUNG CANCER With EGFR(+) or ALK(+) [NCT03991403]Phase 3228 participants (Actual)Interventional2019-08-27Active, not recruiting
Nivolumab Plus Cisplatin/Pemetrexed or Cisplatin/Gemcitabine as Induction in Resectable Non-Small Cell Lung Cancer [NCT03366766]Phase 214 participants (Actual)Interventional2017-12-20Completed
A Phase II Feasibility Study of Pleurectomy/Decortication With Intraoperative Intrathoracic/Intraperitoneal Heated Cisplatin With Sodium Thiosulfate Followed ny Adjuvant ALIMTA/Cisplatin [NCT00165503]Phase 216 participants (Actual)Interventional2004-04-30Terminated(stopped due to lack of acurral)
Pembrolizumab Plus Bevacizumab and Chemotherapy as First-Line Treatment for Advanced or Metastatic Non-Squamous NSCLC Patients With EGFR Exon 20 Insertion Mutation: An Open-Label, Single-Arm, Phase II Trial [NCT05751187]Phase 254 participants (Anticipated)Interventional2023-06-27Recruiting
A Pilot Window-of-opportunity Study of the Anti-PD-1 Antibody Pembrolizumab in Patients With Resectable Malignant Pleural Mesothelioma [NCT02707666]Phase 115 participants (Anticipated)Interventional2016-02-25Recruiting
Phase I Dose Escalation Trial of Biweekly Alimta (With Vitamin Supplementation) in Combination With Taxotere in Advanced Solid Tumor Patients [NCT01172028]Phase 133 participants (Actual)Interventional2005-09-30Completed
A Phase IIB/III Randomized, Double-blind, Placebo Controlled Study Comparing First Line Therapy With or Without TG4010 Immunotherapy Product in Patients With Stage IV Non-Small Cell Lung Cancer (NSCLC) [NCT01383148]Phase 2/Phase 3222 participants (Actual)Interventional2012-04-30Terminated
Induction Pemetrexed and Cisplatin Followed by Pemetrexed as Maintenance vs Carboplatin-paclitaxel and Bevacizumab Followed by Bevacizumab as Maintenance:Multicenter Randomized Phase III Study in Patients With Advanced Non-Squamous Non Small-cell Lung Can [NCT01303926]Phase 3118 participants (Anticipated)Interventional2010-01-31Active, not recruiting
Clinical and Radiographic Comparison of RetroMTA, OrthoMTA And Ferric Sulfate for Pulpotomy in Primary Molars [NCT03718676]32 participants (Actual)Interventional2017-06-02Completed
A Multicenter Phase II Trial of Neoadjuvant JS001, or JS001 in Combination With Chemotherapy in Resectable NSCLC. [NCT03623776]Phase 280 participants (Anticipated)Interventional2019-02-01Active, not recruiting
Individualized Pemetrexed Dosing in Patients With Non-small Cell Lung Cancer or Mesothelioma Based on Renal Function to Improve Treatment Response [NCT03655834]Phase 410 participants (Actual)Interventional2019-02-01Completed
Bevacizumab, Pemetrexed and Cisplatin, or Erlotinib and Bevacizumab for Advanced Non-Squamous NSCLC Stratified by EGFR Mutation Status. A Multicenter Phase II Trial Including Biopsy at Progression (BIO-PRO Trial). [NCT01116219]Phase 2149 participants (Anticipated)Interventional2010-06-30Completed
Atezolizumab in Combination With Bevacizumab, Carboplatin and Pemetrexed for EGFR-mutant Metastatic Non-small Cell Lung Cancer Patients After Failure of EGFR Tyrosine Kinase Inhibitors: a Single Arm Phase 2 Study [NCT03647956]Phase 240 participants (Anticipated)Interventional2018-10-01Recruiting
A Phase II Study of Pemetrexed in Recurrent Cervical Adenocarcinomas [NCT02868892]Phase 26 participants (Actual)Interventional2015-07-31Terminated(stopped due to departure of PI from institution and poor population for study participation)
A Phase Ib/II Multicenter, Randomized, Open Label Trial to Compare Tepotinib (MSC2156119J) Combined With Gefitinib Versus Chemotherapy as Second-Line Treatment in Subjects With MET Positive, Locally Advanced or Metastatic NSCLC Harboring EGFR Mutation and [NCT01982955]Phase 1/Phase 288 participants (Actual)Interventional2013-12-23Completed
A Multicenter Phase II Trial of Neoadjuvant IBI308, Bevacizumab, Plus Pemetrexed and Carboplatin Followed by Surgery in Patients With Unresectable Stage III Non-Small Cell Lung Cancer [NCT03872661]Phase 236 participants (Anticipated)Interventional2019-03-01Recruiting
A Phase 3 Study of Pembrolizumab in Combination With Pemetrexed/Platinum (Carboplatin or Cisplatin) Followed by Pembrolizumab and Maintenance Olaparib vs Maintenance Pemetrexed in the First-Line Treatment of Participants With Metastatic Nonsquamous Non-Sm [NCT03976323]Phase 31,005 participants (Actual)Interventional2019-06-28Active, not recruiting
Assessment of Bone Healing After MTA and PRF Application in Periapical Lesions By Using CBCT [NCT03743987]36 participants (Actual)Interventional2016-12-20Completed
A Phase 2 Study of Icotinib With Concurrent Radiotherapy vs. Pemetrexed+ Carboplatin With Concurrent Radiotherapy in Unresectable Stage III Non-small Cell Lung Cancer With Epidermal Growth Factor Receptor ( EGFR) Mutation [NCT02407366]Phase 280 participants (Anticipated)Interventional2014-12-31Recruiting
A Randomized Open-Label Phase III Trial of Pembrolizumab Versus Platinum Based Chemotherapy in 1L Subjects With PD-L1 Strong Metastatic Non-Small Cell Lung Cancer [NCT02142738]Phase 3305 participants (Actual)Interventional2014-08-25Completed
Comparative Clinical Trial of Erlotinib and Pemetrexed for Maintenance Treatment in Lung Adenocarcinoma [NCT02399566]Phase 4300 participants (Anticipated)Interventional2015-05-31Not yet recruiting
Exploiting Metformin Plus/Minus Cyclic Fasting Mimicking Diet (FMD) to Improve the Efficacy of First Line Chemo-immunotherapy in Advanced LKB1-inactive Lung Adenocarcinoma [NCT03709147]Phase 264 participants (Anticipated)Interventional2018-10-30Recruiting
A Phase II Trial of Pemetrexed and Cisplatin or Carboplatin in Combination With TTFields (150 kHz) as First-line Treatment in Malignant Pleural Mesothelioma [NCT02397928]Phase 282 participants (Actual)Interventional2015-02-28Completed
A Randomized, Double-blind, Multicenter Phase III Study of Anlotinib Hydrochloride Capsule Combined With Chemotherapy Versus Placebo Combined With Chemotherapy as First-line Treatment in Subjects With Advanced Non-squamous Cell Non-small Cell Lung Cancer [NCT04439890]Phase 3369 participants (Anticipated)Interventional2019-08-08Recruiting
SAMSUNG MEDICAL CENTER [NCT03110484]Phase 244 participants (Anticipated)Interventional2021-07-09Recruiting
Official Title: A Phase II Multicenter, Open Label, Non-randomized Study of Neoadjuvant and Adjuvant Treatment With IPH5201 and Durvalumab in Patients With Resectable, Early-stage (II to IIIA) Non-Small Cell Lung Cancer (MATISSE) [NCT05742607]Phase 270 participants (Anticipated)Interventional2023-06-23Recruiting
A Phase 3 Study to Evaluate Zimberelimab (AB122) Combined With AB154 in Front-Line, PD-L1-High, Locally Advanced or Metastatic Non-Small Cell Lung Cancer [NCT04736173]Phase 3750 participants (Anticipated)Interventional2021-02-08Recruiting
Phase 1b Dose-Finding Study of Niraparib, TSR-022, Bevacizumab, and Platinum-Based Doublet Chemotherapy in Combination With TSR-042 in Patients With Advanced or Metastatic Cancer [NCT03307785]Phase 158 participants (Actual)Interventional2017-10-12Active, not recruiting
A Phase 1/2, Open-Label, Safety, Tolerability, and Efficacy Study of Epacadostat in Combination With Pembrolizumab and Chemotherapy in Subjects With Advanced or Metastatic Solid Tumors (ECHO-207/KEYNOTE-723) [NCT03085914]Phase 1/Phase 270 participants (Actual)Interventional2017-05-02Completed
A Phase I Trial of Preoperative Carboplatin or Cisplatin and Pemetrexed With Thoracic Radiation Therapy Followed by Lobectomy in Resectable Stage III Patients With Non-Squamous Non Small Cell Lung Cancer (NSCLC) [NCT01373463]Phase 148 participants (Actual)Interventional2011-05-31Terminated(stopped due to Investigator left site)
The Safety and Efficacy of Rh-Endostatin (Endostar®) Continuous Intravenous Infusion in Combination With Docetaxel/Carboplatin or Pemetrexed/Carboplatin (DC/PC) Regimens for Untreated Stage IIIB/IV Non-small-cell Lung Cancer (NSCLC) [NCT03706703]Phase 250 participants (Anticipated)Interventional2018-10-01Recruiting
A Multicenter Phase II Trial of Carboplatin, Pemetrexed, and Bevacizumab Followed By Pemetrexed and Bevacizumab Maintenance Therapy in Patients With a Light or Never Smoking History [NCT01344824]Phase 238 participants (Actual)Interventional2010-03-31Completed
A Phase I/II Study of Anlotinib Combined With Platinum-based Chemotherapy as the First-line Treatment of Patients With Locally Advanced or Advanced Non-Small Cell Lung Cancer [NCT03636685]Phase 1/Phase 260 participants (Anticipated)Interventional2018-08-15Not yet recruiting
A Randomized Phase II Study of Anlotinib Combined With Pemetrexed and Cisplatin in First Treatment for Advanced Non-small Cell Lung Cancer With Epidermal Growth Factor Receptor Negative Mutations [NCT03671538]62 participants (Anticipated)Interventional2018-10-01Not yet recruiting
Chemotherapy Plus Pembrolizumab After Progression With Previous PD-1/PD-L1 Inhibitors in Patients With Advanced Non-small Cell Lung Cancer: Placebo-controlled Randomized Phase II Study [NCT03656094]Phase 298 participants (Anticipated)Interventional2018-11-01Recruiting
A Phase 2 Study of Pemetrexed Versus Pemetrexed Plus Erlotinib in Second-Line Treatment in Patients With Nonsquamous NSCLC [NCT00447057]Phase 2204 participants (Actual)Interventional2007-03-31Completed
Randomized Phase II Trial Evaluating the Optimal Sequencing of PD-1 Inhibition With Pembrolizumab (MK-3475) and Standard Platinum-based Chemotherapy in Patients With Chemotherapy Naive Stage IV Non-small Cell Lung Cancer [NCT02591615]Phase 291 participants (Actual)Interventional2016-03-31Completed
Pemetrexed Alone as Salvage Treatment in Metastatic Colorectal Cancer Patients Who Were Failed After Standard Chemotherapy: A Phase II Single Arm Prospective Study [NCT02588781]Phase 223 participants (Actual)Interventional2015-10-31Active, not recruiting
A Phase 1b Trial of LY2606368 in Combination With Chemotherapy or Targeted Agents in Advanced and/or Metastatic Tumors [NCT02124148]Phase 1167 participants (Actual)Interventional2014-06-18Completed
Phase III Randomized Trial of Pleurectomy/Decortication Plus Systemic Therapy With or Without Adjuvant Hemithoracic Intensity-Modulated Pleural Radiation Therapy (IMPRINT) for Malignant Pleural Mesothelioma (MPM) [NCT04158141]Phase 316 participants (Actual)Interventional2020-01-29Terminated(stopped due to Permanent Administrative Closure)
A Phase 1, Open-label, Dose-escalation and Dose-expansion Study to Evaluate the Safety, Tolerability Pharmacokinetics Immunogenicity, and Antitumor Activity of MEDI5752 in Subjects With Advanced Solid Tumors. [NCT03530397]Phase 1396 participants (Anticipated)Interventional2018-04-24Recruiting
A Phase III Multicenter, Randomized Study of Oral LDK378 Versus Standard Chemotherapy in Previously Untreated Adult Patients With ALK Rearranged (ALK-positive), Stage IIIB or IV, Non-squamous Non-small Cell Lung Cancer [NCT01828099]Phase 3376 participants (Actual)Interventional2013-07-09Active, not recruiting
A Phase III, Randomized, Double-blinded, Multicenter Study of AK105 Combined With Carboplatin and Pemetrexed vs Placebo Combined With Carboplatin and Pemetrexed as First-line Therapy in Patients With Metastatic Nonsquamous Non-small Cell Lung Cancer [NCT03866980]Phase 3164 participants (Actual)Interventional2018-11-27Active, not recruiting
A Pilot Study: Phase II Study of Histology-based Consolidation Chemotherapy Following Concurrent Chemo-radiotherapy for Inoperable Stage III Non-small Cell Lung Cancer [NCT01336543]Phase 20 participants (Actual)Interventional2011-03-31Withdrawn(stopped due to Low accrual, small patient population at center.)
Dose Climbing Trial of Anlotinib Plus Pemetrexed/Docetaxel in the Second-line Treatment of Advanced Gene Negative Non-squamous Non-small Cell Lung Cancer [NCT03566576]18 participants (Anticipated)Interventional2018-07-01Not yet recruiting
A Randomized, Multicenter, Open-Label Phase 3 Study of Pemetrexed-Cisplatin Chemotherapy Plus Necitumumab (IMC-11F8) Versus Pemetrexed-Cisplatin Chemotherapy Alone in the First-Line Treatment of Patients With Stage IV Nonsquamous Non-Small Cell Lung Cance [NCT00982111]Phase 3633 participants (Actual)Interventional2009-11-02Completed
The Efficacy and Safety of Osimertinib Plus Carboplatin and Pemetrexed Versus Osimertinib Monotherapy in Metastatic EGFRm NSCLC Patients With EGFRm Persistence in ctDNA at 3 Weeks After 1L Osimertinib: A Multicenter, Randomized Controlled Study [NCT04769388]Phase 2150 participants (Anticipated)Interventional2021-12-28Recruiting
Anlotinib-based Combination as First-line Treatment in Advanced Non-small Cell Lung Cancer: a Single Center, Three Arms and Exploratory Study [NCT03628521]Phase 180 participants (Anticipated)Interventional2018-07-20Recruiting
Study of Sintilimab Combined With Anlotinib and Platinum-Containing Dual-Agent Chemotherapy as First Line Therapy in Malignant Pleural Mesothelioma: A Single Arm, Open-label, Prospective Phase II Trial [NCT05188859]Phase 229 participants (Anticipated)Interventional2022-01-31Not yet recruiting
The Efficacy and Safety of Aumolertinib Combined Ommaya Reservoir Intrathecal Chemotherapy With Pemetrexed for Leptomeningeal Metastasis From EGFR-mutated NSCLC and Investigate the Efficacy Prognostic Value of Dynamic Changes of cfDNA [NCT05810350]Phase 240 participants (Anticipated)Interventional2023-01-01Recruiting
An Open-label, Randomized, Multi-center, Phase III Clinical Study of MRG002 Versus Investigator's Choice of Chemotherapy in the Treatment of Patients With HER2-positive Unresectable Locally Advanced or Metastatic Urothelial Cancer Previously Treated With [NCT05754853]Phase 3290 participants (Anticipated)Interventional2023-04-06Recruiting
Recombinant Human Endostatin in Combination With Platinum-Based Doublet Chemotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer With Negative Driver Gene: a Multicenter, Single-Arm Trial [NCT05574998]Phase 2100 participants (Anticipated)Interventional2021-02-01Recruiting
A Phase I/II Study of MK-3475 (SCH900475) in Combination With Chemotherapy or Immunotherapy in Patients With Locally Advanced or Metastatic Non-Small Cell Lung Carcinoma [NCT02039674]Phase 1/Phase 2267 participants (Actual)Interventional2014-02-21Completed
Randomized Phase II Trial of Single Agent Chemotherapy Plus Nivolumab or Single Agent Chemotherapy Alone in Patients With Advanced Squamous or Non-squamous NSCLC With Primary Resistance to Prior PD-1 or PDL-1 Inhibitor [NCT03041181]Phase 23 participants (Actual)Interventional2017-01-27Terminated(stopped due to Funder decision - lack of accrual)
A Phase II Trial of Oxaliplatin and Pemetrexed in Hormone Refractory Prostate Cancer [NCT01338792]Phase 247 participants (Actual)Interventional2006-06-30Completed
EGFR Tyrosine Kinase Inhibitor Combined With Concurrent or Sequential Chemotherapy for Advanced Lung Cancer Patients of Gradual Progression After First-line EGFR-TKI Therapy: a Randomized Controlled Study [NCT03544814]Phase 299 participants (Actual)Interventional2015-01-01Completed
Combination of Chemotherapy and Gefitinib as First-line Treatment of Patients With Advanced Lung Adenocarcinoma and Sensitive EGFR Mutations: a Randomised Controlled Trial [NCT02148380]121 participants (Actual)Interventional2014-05-31Completed
An Open-label, Phase 2 Basket Study of SEA-CD40 Combination Therapies in Advanced Malignancies [NCT04993677]Phase 277 participants (Actual)Interventional2021-10-06Active, not recruiting
A Phase III Randomized, Open-Label, Multi-Center, Global Study of MEDI4736 in Combination With Tremelimumab Therapy Versus Standard of Care Platinum-Based Chemotherapy in First-Line Treatment of Patients With Advanced or Metastatic Non Small-Cell Lung Can [NCT02542293]Phase 3953 participants (Actual)Interventional2015-11-03Active, not recruiting
A Randomized Phase 2 Trial of Doxorubicin Plus Pemetrexed Followed by Docetaxel, Versus Doxorubicin Plus Cyclophosphamide Followed by Docetaxel, as Neoadjuvant Treatment for Early Breast Cancer [NCT00149214]Phase 2257 participants (Actual)Interventional2005-09-30Completed
A Phase Ib, Open-Label, Dose-Escalation Study of the Safety and Pharmacology of GDC-0980 in Combination With Either Paclitaxel and Carboplatin (With or Without Bevacizumab) or Pemetrexed and Cisplatin in Patients With Solid Tumors [NCT01301716]Phase 175 participants (Actual)Interventional2011-09-30Completed
A Phase I Study of OSI-906 in Combination With Pemetrexed in Advanced Solid Tumor Malignancies [NCT01567384]Phase 10 participants (Actual)Interventional2012-05-31Withdrawn(stopped due to The sponsor withdrew support for the study before any patients were enrolled.)
Anlotinib Combined With Pemetrexed And Carboplatin Followed by Maintenance Therapy With Anlotinib Plus Pemetrexed as the First-line Treatment in Patients With Advanced Nonsquamous NSCLC [NCT03790228]Phase 143 participants (Anticipated)Interventional2019-03-22Recruiting
Phase II Study of SHR-1210(Anti-PD-1 Antibody) Combination With Apatinib Versus Pemetrexed and Carboplatin in Subjects With KRAS Mutant Stage IV Non-squamous Non-small Cell Lung Cancer [NCT03777124]Phase 2230 participants (Anticipated)Interventional2019-02-28Not yet recruiting
Study of Anlotinib Combined With Pemetrexed as the Second-line Treatment in Patients With Advanced Nonsquamous Non-Small-Cell Lung Cancer (ALTER-L025) [NCT03778138]Phase 251 participants (Anticipated)Interventional2019-01-01Recruiting
A Phase II Trial of Pemetrexed in Combination With Gemcitabine as First Line Treatment in Extensive-Stage Small Cell Lung Carcinoma [NCT00129974]Phase 21 participants (Actual)Interventional2005-08-31Terminated(stopped due to failure to accrue)
Combination Therapy With Ectiecinib, Pemetrexed and Platinum in Patients With Metastatic Non-squamous Non-small Cell Lung Cancer With EGFR Mutations Who Did Not Progress After Pemetrexed in Combination With Platinum-based Chemotherapy:a Single-arm, Open, [NCT03992885]Phase 370 participants (Anticipated)Interventional2019-07-01Recruiting
A Phase II Trial to Investigate Genetic Markers of Response to Pembrolizumab (MK-3475, SCH 900475) Combined With Chemotherapy as a First-line Treatment for Non-Small Cell Lung Cancer (KEYNOTE-782) [NCT03664024]Phase 2118 participants (Actual)Interventional2018-10-30Completed
Study of Pemetrexed Disodium Plus Cisplatin as First-line Therapy in Patients With Advanced Non-squamous Cell Lung Cancer: a Phase IIA Pharmacogenomic Trial [NCT01088906]Phase 257 participants (Actual)Interventional2010-01-31Terminated(stopped due to No safety reasons. Low recruitment.)
Randomized Phase III Study of Docetaxel or Pemetrexed With or Without Cetuximab in Patients With Recurrent or Progressive Non-Small Cell Lung Cancer After Platinum-Based Therapy [NCT00095199]Phase 3939 participants (Actual)Interventional2005-01-31Completed
Randomized, Phase II, Open-Label Controlled Study of Two Different Doses and Schedules of EMD 72000 (Matuzumab) in Combination With Pemetrexed, or Pemetrexed Alone, as Second-Line Treatment for Stage IIIB/IV Non-Small Cell Lung Cancer and Progressive Dise [NCT00111839]Phase 2150 participants (Actual)Interventional2005-05-31Completed
Phase III Open Label First Line Therapy Study of Tislelizumab With Chemotherapy Versus Chemotherapy in Untreated Advanced Non-Squamous Non-Small Cell Lung Cancer(NSCLC) [NCT03663205]Phase 3334 participants (Actual)Interventional2018-07-23Completed
Phase 2 Study of Pembrolizumab Plus Pemetrexed for Elderly Patients With Non-squamous Non-small Cell Lung Cancer With PD-L1 Tumor Proportion Score of Less Than 50%: CJLSG1901 [NCT04396457]Phase 250 participants (Anticipated)Interventional2020-05-25Active, not recruiting
A Phase II, Prospective, Single Arm Trial of Cadonilimab in Combination With Bevacizumab and Standard Chemotherapy as First Line Therapy in Unresectable Pleural Mesothelioma [NCT05930665]Phase 238 participants (Anticipated)Interventional2023-07-31Not yet recruiting
Phase II Trial of Standard Platinum Doublet Chemotherapy + Various Proton Beam Therapy (PBT) Doses in Order to Determine the Optimal Dose of PBT for Unresectable Stage 2/3 Non-Small Cell Lung Cancer [NCT03132532]Phase 218 participants (Actual)Interventional2017-07-31Active, not recruiting
A Phase III, Randomized, Open Label Trial of Nivolumab in Combination With Ipilimumab Versus Pemetrexed With Cisplatin or Carboplatin as First Line Therapy in Unresectable Pleural Mesothelioma [NCT02899299]Phase 3605 participants (Actual)Interventional2016-11-29Completed
Phase I Study in Patients With Tumours Requiring Arginine to Assess ADI-PEG 20 With Atezolizumab, Pemetrexed and Carboplatin (ADIAtezoPemCarbo) (iTRAP Study) [NCT03498222]Phase 10 participants (Actual)Interventional2018-06-01Withdrawn(stopped due to Funder and IMP manufacturer withdrew support)
A PHASE II STUDY Of ADJUVANT CHEMOTHERAPY After SURGERY For STAGE I Lung ADENOCARCINOMA PATIENTS With MICROPAPILLARY COMPONENT More Than Or EQUAL To 20% [NCT03351842]Phase 2460 participants (Anticipated)Interventional2017-09-01Recruiting
Almonertinib Alone Versus Almonertinib Plus Chemotherapy as First-line Treatment in Patients With Epidermal Growth Factor Receptor (EGFR) Mutation Positive With Concomitant Non-EGFR Driver Gene Mutation Positive, Locally Advanced or Metastatic Non-Small C [NCT04500704]Phase 3166 participants (Anticipated)Interventional2020-10-31Not yet recruiting
A Randomized, Open-label, Phase II Study of Maintaining Pan-ERBB Blockade Following Platinum-based Induction Chemotherapy in Patients With EGFR Mutated, Metastatic Non-small-cell Lung Cancer Progressing After Treatment With Afatinib as First EGFR-targetin [NCT02488694]Phase 24 participants (Actual)Interventional2015-11-30Terminated(stopped due to unsufficient recruitment)
Phase II Study of Tislelizumab Plus Pemetrexed in Patients With Relapsed or Refractory Primary Diffuse Large B-cell Lymphoma (DLBCL) of the Central Nervous System (CNS) [NCT05253118]Phase 228 participants (Anticipated)Interventional2022-08-18Recruiting
A Phase II Trial of GM-CSF Plus Maintenance Pembrolizumab +/- Pemetrexed After Completion of First Line Chemo-Immunotherapy in Advanced Non-Small Cell Lung Cancer Patients With PDL-1 of 1%-49% [NCT04856176]Phase 283 participants (Anticipated)Interventional2022-01-03Recruiting
A Phase III, Double-blind, Placebo-controlled, Multi-center International Study of Neoadjuvant/Adjuvant Durvalumab for the Treatment of Patients With Resectable Stages II and III Non-small Cell Lung Cancer (AEGEAN) [NCT03800134]Phase 3826 participants (Actual)Interventional2018-12-06Active, not recruiting
"A Study on theFuzhengTherapy Promoted Immune Reconstitution to Improve the Survival of Early-stage Lung Cancer After Surgical Operation" [NCT02603003]Phase 1218 participants (Actual)Interventional2015-06-30Completed
Individualized Pemetrexed Dosing in Patients With Non-small Cell Lung Cancer or Mesothelioma Based on Renal Function to Improve Treatment Response [NCT03655821]Phase 481 participants (Actual)Interventional2019-02-01Terminated(stopped due to 81 patients included, difficult inclusion and full inclusion would not change the results)
A Factorial Study Comparing Pemetrexed With Gemcitabine and Testing the Efficacy of the Addition of Cisplatin in Elderly Patients With Non Squamous Advanced, Metastatic or Recurrent NSCLC. [NCT01656551]Phase 3232 participants (Actual)Interventional2012-08-31Active, not recruiting
The Prognosis Study of Postoperative Non-small-cell Lung Cancer Patients Treated Precisely With the Integrated Traditional Chinese and Western Medicine Based on CTC Detection [NCT03269162]Phase 3144 participants (Anticipated)Interventional2016-09-30Active, not recruiting
A Study in Cancer Patients to Evaluate the Ability of LY2603618 to Act as an Inhibitor of CYP2D6 Using Desipramine as a Probe Substrate [NCT01358968]Phase 120 participants (Actual)Interventional2011-06-30Completed
A Phase 1/2 Study of Apatinib in Combination With AP(Pemetrexed/Cisplatin) or AC(Pemetrexed/Carboplatin) as First-line Chemotherapy for Advanced Epidermal Growth Factor Receptor(EGFR) Wild Type Non-squamous Non-small Cell Lung Cancer [NCT03201146]Phase 1/Phase 248 participants (Anticipated)Interventional2017-06-27Recruiting
A Multicenter, Open-Label, Phase Ib/II Study of AK119 and AK112 With or Without Chemotherapy in Patients With EGFR-mutant Locally Advanced or Metastatic Non-squamous Non-small Cell Lung Cancer Who Have Failed to Epidermal Growth Factor Receptor Tyrosine K [NCT05636267]Phase 1/Phase 2114 participants (Anticipated)Interventional2023-02-10Recruiting
A Phase 1/2, Open-Label, Multicenter Study to Investigate the Safety and Preliminary Efficacy of Combined Bempegaldesleukin (NKTR-214) and Pembrolizumab With or Without Chemotherapy in Patients With Locally Advanced or Metastatic Solid Tumors [NCT03138889]Phase 1/Phase 2162 participants (Actual)Interventional2017-06-09Terminated(stopped due to Sponsor decision)
Personalized Therapy for Esophagogastric Cancer Using Thymidylate Synthase Genetic Markers [NCT02296671]Phase 20 participants (Actual)Interventional2015-02-28Withdrawn(stopped due to Was unable to accrue any patients)
Neoadjuvant Icotinib With Chemotherapy for Resectable Stage II-IIIB N2 EGFR Mutation-positive Lung Adenocarcinoma: A Phase II Study [NCT05132985]Phase 245 participants (Anticipated)Interventional2022-01-01Not yet recruiting
Maintenance Pembrolizumab at Usual or Low doSE in Non-squamous Lung Cancer: a Non-inferiority Study [NCT05692999]Phase 31,166 participants (Anticipated)Interventional2023-03-20Recruiting
Tailored Second Line Treatment by EGFR Mutation in Patients With Advanced Lung Adenocarcinoma [NCT00903292]52 participants (Anticipated)Interventional2009-03-31Recruiting
A Randomized, Open-Label, Dose Escalation Study of Bevacizumab With Ambulatory Blood Pressure Monitoring in Previously Untreated Patients With Advanced Non-squamous Non-Small Cell Lung Cancer [NCT01063283]20 participants (Actual)Interventional2010-03-31Completed
A Single Armed Phase ⅡStudy of Alternating Icotinib and Chemotherapy for Advanced Non-small Cell Lung Cancer With EGFR Mutation [NCT02737774]Phase 260 participants (Anticipated)Interventional2016-04-13Active, not recruiting
Chemotherapy in KRAS Mutated Chemotherapy Naive Non-small Cell Lung Cancer Patients: a Phase III Study Comparing Cisplatin-pemetrexed With Carboplatin-paclitaxel-bevacizumab: NVALT 22 [NCT02743923]Phase 3203 participants (Actual)Interventional2016-04-30Active, not recruiting
Phase II Trial of Combination Pemetrexed (Alimta) and Carboplatin (Paraplatin) in Platinum Sensitive Recurrent Ovarian, Primary Peritoneal, and Fallopian Tube Carcinoma [NCT01001910]Phase 222 participants (Actual)Interventional2008-07-31Completed
A Phase I Study of INCB024360 (Epacadostat) Alone, INCB024360 in Combination With Pembrolizumab (MK-3475), and INCB024360 and Pembrolizumab in Combination With Chemotherapy in Patients With Advanced Solid Tumors (KEYNOTE-434) [NCT02862457]Phase 134 participants (Actual)Interventional2016-08-23Completed
An Open-Label, Phase 1B Study of NEO-PV-01 With Pembrolizumab Plus Chemotherapy in Patients With Advanced or Metastatic Nonsquamous Non-small Cell Lung Cancer [NCT03380871]Phase 138 participants (Actual)Interventional2018-05-04Completed
A Single-Arm Phase II Clinical Trial of Apatinib as the Maintenance Therapy in Advanced Lung Adenocarcinoma [NCT03376737]Phase 2120 participants (Anticipated)Interventional2017-10-12Recruiting
Local Consolidative Therapy (LCT) and Durvalumab (MEDI4736) for Oligoprogressive and Polyprogressive Stage III NSCLC After Chemoradiation and Anti-PD-L1 Therapy (ENDURE) [NCT04892953]Phase 251 participants (Anticipated)Interventional2021-07-07Recruiting
A Randomized, Open Label, Phase III Study of Overall Survival Comparing Pembrolizumab (MK-3475) Versus Platinum Based Chemotherapy in Treatment Naïve Subjects With PD-L1 Positive Advanced or Metastatic Non-Small Cell Lung Cancer (Keynote 042) [NCT03850444]Phase 3262 participants (Actual)Interventional2016-08-01Completed
A Phase II Trial to Assess the Safety, Immunological Activity of TroVax® Plus Pemetrexed/Cisplatin in Patients With Malignant Pleural Mesothelioma [NCT01569919]Phase 226 participants (Anticipated)Interventional2012-12-31Recruiting
BRIDGE Trial: Phase II Trial of durvalumaB and chemotheRapy Induction Followed by Durvalumab and Radiotherapy in larGe volumE Stage III NSCLC [NCT04765709]Phase 210 participants (Actual)Interventional2021-09-24Active, not recruiting
High-dose Pemetrexed to Treat Lung Adenocarcinoma With Brain Metastases [NCT02284490]Phase 225 participants (Anticipated)Interventional2014-11-30Not yet recruiting
A Multi-Regional, Randomized, Double-blind, Phase 3 Study of the Efficacy and Safety of Sintilimab Plus Chemotherapy vs Placebo Plus Chemotherapy Before Surgery and Sintilimab vs Placebo After Surgery for Resectable Non-small Cell Lung Cancer [NCT05116462]Phase 3800 participants (Anticipated)Interventional2021-11-11Not yet recruiting
A Randomized Phase II Trial Of Pemetrexed With Or Without PF-3512676 For The Treatment Of Patients With Locally Advanced Or Metastatic Non-Small Cell Lung Cancer After Failure Of One Prior Chemotherapy Regimen For Advanced Disease [NCT00321308]Phase 236 participants (Actual)Interventional2006-09-30Terminated(stopped due to See Termination Reason in Detailed Description.)
A Phase II Study of Pemetrexed in Children With Recurrent Malignancies [NCT00520936]Phase 272 participants (Actual)Interventional2007-09-30Completed
A Phase IB, Open-Label, Multi-Center Study to Determine the Efficacy and Safety of Durvalumab and/or Novel Oncology Therapies, With or Without Chemotherapy, for First-Line Stage IV Non-Small Cell Lung Cancer (NSCLC) (MAGELLAN) [NCT03819465]Phase 1175 participants (Actual)Interventional2018-12-27Active, not recruiting
Furmonertinib Combined With Cisplatin/Pemetrexed as Neoadjuvant Therapy in EGFR Mutated Stage IIIA-IIIB Resectable Non-small Cell Lung Cancer (FORESEE): a Prospective, Open-label, Single-arm, Phase 2 Study [NCT05430802]Phase 240 participants (Anticipated)Interventional2022-02-24Recruiting
Neoadjuvant Therapy With Toripalimab and JS004 Combined With Platinum-based Doublet Chemotherapy for Resectable or Potentially Resectable Stage III Non-small Cell Lung Cancer: A Randomised Controlled, Open-label, Phase 2 Trial [NCT05891080]Phase 2124 participants (Anticipated)Interventional2023-07-01Not yet recruiting
Pevonedistat as a Single Agent and in Combination With Chemotherapy in Patients With Malignant Mesothelioma [NCT03319537]Phase 1/Phase 29 participants (Actual)Interventional2017-10-05Completed
Clinical Study to Evaluate the Efficacy, Safety and Tolerability of Savolitinib + Osimertinib Versus Pemetrexed + Platinum in Treatment of Patients With NSCLC With MET Amplification [NCT05015608]Phase 3250 participants (Anticipated)Interventional2021-11-22Recruiting
Early Positron Emission Tomography as a Predictor of Response in Neoadjuvant Chemotherapy for Non-Small Cell Lung Cancer [NCT00227539]Phase 225 participants (Actual)Interventional2005-07-31Completed
Pemetrexed in Maintenance in Patients With Impaired Renal Function: Randomized Phase 4 Multicenter Study Comparing 2 Dose Calculation Strategies (PKAPIR) [NCT03607149]Phase 411 participants (Actual)Interventional2017-04-06Completed
A Phase 3, Double-Blind, Placebo-Controlled Study of Maintenance Pemetrexed Plus Best Supportive Care Versus Best Supportive Care Immediately Following Induction Treatment for Advanced Non-Small Cell Lung Cancer [NCT00102804]Phase 3663 participants (Actual)Interventional2005-03-31Completed
A Phase 2 Peri-Operative Trial of Fianlimab and Cemiplimab in Combination With Chemotherapy Versus Cemiplimab in Combination With Chemotherapy in Patients With Resectable Early Stage (Stage II to IIIB [N2]) NSCLC [NCT06161441]Phase 2180 participants (Anticipated)Interventional2024-03-05Not yet recruiting
Phase II Study of Pemetrexed and Pembrolizumab in Recurrent and/or Metastatic Salivary Gland Malignancies [NCT04895735]Phase 245 participants (Anticipated)Interventional2021-07-23Recruiting
A Phase 1/2, Dose Escalation, Dose Expansion, and Dose Optimization Study of the Safety, Tolerability, and Anti-tumor Activity of SAR444881 Administered Alone and in Combination With Pembrolizumab, Cetuximab and/or Chemotherapy in Participants With Advanc [NCT04717375]Phase 1/Phase 2456 participants (Anticipated)Interventional2021-04-11Recruiting
A Randomized, Double Blind, Phase 3 Study of Platinum-Based Chemotherapy With or Without INCMGA00012 in First-Line Metastatic Squamous and Nonsquamous Non-Small Cell Lung Cancer (POD1UM-304) [NCT04205812]Phase 3583 participants (Actual)Interventional2020-09-27Active, not recruiting
An Open-Label, Randomized Phase 3 Trial of Nivolumab, or Nivolumab Plus Ipilimumab, or Nivolumab Plus Platinum Doublet Chemotherapy Versus Platinum Doublet Chemotherapy in Subjects With Chemotherapy-Naïve Stage IV or Recurrent Non-Small Cell Lung Cancer ( [NCT02477826]Phase 32,748 participants (Actual)Interventional2015-08-05Active, not recruiting
Pemetrexed Monochemotherapy in Patients With Locally Advanced or Metastatic Non Small Cell Lung Cancer. A Pilot Study to Define the Best Dosing Schedule for a Planned Phase II Randomized Trial [NCT00370292]Phase 219 participants (Actual)Interventional2006-09-30Completed
Phase I Clinical Trial of Suberoylanilide Hydroxamic Acid (SAHA) in Combination With Pemetrexed and Cisplatin in Patients With Advanced Cancer [NCT00106626]Phase 152 participants (Actual)Interventional2005-08-31Completed
Study of Thalidomide With First-line Chemotherapy and as Maintenance Treatment of Advanced Nonsquamous NSCLC With Epidermal Growth Factor Receptor Wild-Type or Unknown Mutation Status: A Multicenter, Randomized, Prospective Clinical Trial [NCT03062800]Phase 2232 participants (Anticipated)Interventional2016-12-31Recruiting
Clinical Research of S-1 Versus Pemetrexed in the Maintenance Treatment of Advanced Non-squamous Non-small Cell Lung Cancer [NCT03700333]Phase 3120 participants (Anticipated)Interventional2018-10-20Not yet recruiting
A Phase II/III Randomized Study of Pembrolizumab in Patients With Advanced Malignant Pleural Mesothelioma [NCT02784171]Phase 2/Phase 3520 participants (Actual)Interventional2016-11-11Active, not recruiting
A MULTICENTER, OPEN-LABEL, PHASE 1B/2 STUDY TO EVALUATE SAFETY AND EFFICACY OF AVELUMAB (MSB0010718C) IN COMBINATION WITH CHEMOTHERAPY WITH OR WITHOUT OTHER ANTI-CANCER IMMUNOTHERAPIES AS FIRST-LINE TREATMENT IN PATIENTS WITH ADVANCED MALIGNANCIES [NCT03317496]Phase 1/Phase 267 participants (Actual)Interventional2017-12-21Terminated(stopped due to The study was terminated since there was no need for further safety or efficacy data to be collected. The participants having benefit from the investigational treatments have been moved to a continuation study (NCT05059522).)
ICARuS II (Intraperitoneal Chemotherapy After cytoReductive Surgery): A Multicenter, Randomized Phase II Trial of Normothermic Intraperitoneal Chemotherapy and Intravenous Chemotherapy After Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemother [NCT06057935]Phase 264 participants (Anticipated)Interventional2023-09-21Recruiting
A Phase 1a/1b Study of LY3537982 in Patients With KRAS G12C-Mutant Advanced Solid Tumors [NCT04956640]Phase 1400 participants (Anticipated)Interventional2021-07-19Recruiting
A Phase III Randomized, Open-Label, Multi-Center, Global Study of MEDI4736 in Combination With Tremelimumab Therapy or MEDI4736 Monotherapy Versus Standard of Care Platinum-Based Chemotherapy in First Line Treatment of Patients With Advanced or Metastatic [NCT02453282]Phase 31,118 participants (Actual)Interventional2015-07-21Active, not recruiting
A Biomarker-directed Phase 2 Platform Study in Patients With Advanced Non-Small Lung Cancer Whose Disease Has Progressed on First-Line Osimertinib Therapy. [NCT03944772]Phase 2250 participants (Anticipated)Interventional2019-06-25Active, not recruiting
Phase II Trial of Pemetrexed-Based Induction Chemotherapy Followed by Concomitant Chemoradiotherapy in Previously Irradiated Head and Neck Cancer Patients [NCT01172470]Phase 234 participants (Actual)Interventional2005-06-30Completed
The Prospective,Multicenter,Randomized Controlled Clinical Study of the Optimal Intervention Time of Radiotherapy for Oligometastatic Stage IV Non-small Cell Lung Cancer(NSCLC) [NCT02076477]Phase 3420 participants (Anticipated)Interventional2014-01-31Recruiting
Research on the Correlation Between Efficacy of Osimertinib and EGFR T790M Status and Ratio Via Digital Droplet PCR (ddPCR) From Peripheral Blood in NSCLC Patients [NCT05458726]Phase 280 participants (Anticipated)Interventional2020-09-15Recruiting
A Randomized,Open-label, Multi-Center, Phase II Clinical Trial to Assess the Efficacy and Safety of SHR-1210± SHR-1020 Versus Physician's Choice Chemotherapy in the Treatment of Recurrent or Metastatic Cervical Cancer Patients [NCT04680988]Phase 2194 participants (Actual)Interventional2021-04-05Active, not recruiting
Pemetrexed Disodium and Cisplatin Chemotherapy Combined With Synchronous Gefitinib vs Chemotherapy Alone as Adjuvent Therapy in Patient With Stage II-IIIA, Epidermal Growth Factor Receptor Mutant Expressing Lung Adenocarcinoma [NCT02518802]Phase 3220 participants (Anticipated)Interventional2015-01-31Recruiting
A Phase II Trial of AMG 102 in Combination With Pemetrexed and Cisplatin in Patients With Malignant Pleural Mesothelioma [NCT01105390]Phase 20 participants (Actual)Interventional2010-04-30Withdrawn(stopped due to withdrawn)
A Phase I Open Label Study of Continuous Oral Treatment With BIBF 1120 Together With Pemetrexed in Previously Treated Patients With Non-small Cell Lung Cancer [NCT02182102]Phase 126 participants (Actual)Interventional2005-09-30Completed
The Safety and Efficacy of Pemetrexed Rechallenge With Bevacizumab for Patients With Advanced Non-squamous Non-small Cell Lung Cancer [NCT02200354]Phase 220 participants (Actual)Interventional2014-07-31Terminated(stopped due to inclusion speed)
A Phase III, Randomized, Open-Label, Multi-center Study of SHR-1210(Anti-PD-1 Antibody) in Combination With Pemetrexed and Carboplatin as First Line Therapy in Subjects With Advanced/Metastatic Non-squamous Non-small Cell Lung Cancer [NCT03134872]Phase 3419 participants (Actual)Interventional2017-05-12Completed
Combining Genomics and Imageomics to Predict the Sensitivity of Neoadjuvant Pemetrexed and Cisplatin Chemotherapy in Patients With Lung Adenocarcinoma [NCT05185544]50 participants (Anticipated)Observational2022-01-01Not yet recruiting
Neoadjuvant Afatinib Combination With Chemotherapy for Stage Ⅱa-Ⅲb Non-small Cell Lung Cancer With Epidermal Growth Factor Receptor Activating Mutation [NCT04470076]Phase 230 participants (Anticipated)Interventional2020-07-10Not yet recruiting
A Randomized, Open-Label, Multicenter Phase 3 Study to Evaluate SKB264 Monotherapy Versus Pemetrexed in Combination With Platinum in Patients With Locally Advanced or Metastatic Non-squamous Non-small Cell Lung Cancer With EGFR Mutation Who Have Failed to [NCT05870319]Phase 3356 participants (Anticipated)Interventional2023-06-26Recruiting
Phase 3, Open-Label, Randomized, Controlled, Global Study of Telisotuzumab Vedotin (ABBV-399) Combined With Osimertinib vs Platinum-Based Chemotherapy in Subjects With c-Met Overexpressing (OE) EGFR Mutant, Locally Advanced/Metastatic Non-Squamous NSCLC A [NCT06093503]Phase 3250 participants (Anticipated)Interventional2024-05-31Not yet recruiting
A Single Arm, Phase 2 Study of Amivantamab, Lazertinib and Pemetrexed for First-line Treatment of Recurrent/Metastatic Non-small Cell Lung Cancers (NSCLCs) With EGFR Mutations [NCT05299125]Phase 249 participants (Anticipated)Interventional2023-05-24Recruiting
An Open-label, Dose-escalation, Phase I/II Study to Assess the Safety, the Tolerability, the Immunogenicity and the Preliminary Clinical Activity of the Therapeutic Cancer Vaccine, PDC*lung01, Associated or Not With Anti-PD-1 Treatment in Patients With No [NCT03970746]Phase 1/Phase 273 participants (Actual)Interventional2019-09-10Active, not recruiting
Phase I Study of TRC102 in Combination With Cisplatin and Pemetrexed in Patients With Advanced Solid Tumors / Phase II Study of TRC102 With Pemetrexed in Patients Refractory to Pemetrexed and Cisplatin or Carboplatin [NCT02535312]Phase 1/Phase 230 participants (Actual)Interventional2016-03-08Active, not recruiting
A Three Arm, Randomized, Double-Blind, Multicenter, Phase 3 Study of HLX10(Anti-PD-1 Antibody) in Combination With Carboplatin Plus (+) Pemetrexed With or Without HLX04(Avastin Biosimilar) Compared With Carboplatin+Pemetrexed in 1L Stage IIIB/IIIC or IV N [NCT03952403]Phase 3643 participants (Actual)Interventional2019-12-02Active, not recruiting
A Phase 1b, Open-Label, Dose Escalation Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of MK-0482 as Monotherapy and in Combination With Pembrolizumab in Participants With Advanced/Metastatic Solid Tumors. [NCT03918278]Phase 1230 participants (Anticipated)Interventional2019-06-19Active, not recruiting
A Randomized Phase I/II Study of Standard Chemotherapy (Cisplatin and Pemetrexed) With or Without Axitinib in Patients With Malignant Mesothelioma: Interim Biopsy Analysis to Determine Efficacy [NCT01211275]Phase 1/Phase 232 participants (Actual)Interventional2009-05-22Completed
A Phase I/II Study of First Line Vorinostat With Pemetrexed-cisplatin, in Patients With Malignant Pleural Mesothelioma [NCT01353482]Phase 1/Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to UCL CTC were informed by Merck Sharp & Dohme on 22.08.11 that support for the trial had been withdrawn in light of results from another trial with trial drug.)
Immunotherapy in Lung Cancer: Which Treatment After Immunotherapy Cessation: A Prospective Registry From the European Lung Cancer Working Party [NCT04465942]300 participants (Anticipated)Observational [Patient Registry]2020-06-26Recruiting
A Phase II Study of Pembrolizumab Plus Platinum and Pemetrexed as First Line Therapy in Advanced Non-squamous Non-small Cell Lung Cancer Patients With EGFR Exon 21 Point Mutation and Programmed Cell Death Ligand 1 Expression [NCT06142617]Phase 237 participants (Anticipated)Interventional2023-12-01Not yet recruiting
A Biomarker-Driven Protocol for Previously Treated ALK-Positive Non-Squamous NSCLC Patients: The NCI-NRG ALK Protocol [NCT03737994]Phase 210 participants (Actual)Interventional2019-07-25Active, not recruiting
A Phase III, Open-Label, Randomized Study of Atezolizumab (MPDL3280A, Anti-Pd-L1 Antibody) in Combination With Carboplatin or Cisplatin + Pemetrexed Compared With Carboplatin or Cisplatin + Pemetrexed in Patients Who Are Chemotherapy-Naive and Have Stage [NCT02657434]Phase 3578 participants (Actual)Interventional2016-04-30Completed
A Phase III Randomized Trial of Adjuvant Chemotherapy With or Without Bevacizumab for Patients With Completely Resected Stage IB (≥ 4 cm) - IIIA Non-small Cell Lung Cancer (NSCLC) [NCT00324805]Phase 31,501 participants (Actual)Interventional2007-06-01Active, not recruiting
A Phase 1b Combination Study of INCMGA00012 Plus Chemotherapy in Participants With Advanced Solid Tumors (POD1UM-105) [NCT03920839]Phase 10 participants (Actual)Interventional2019-07-15Withdrawn(stopped due to As of November 4, 2019 the study was halted prematurely and will not resume.)
Intrathecal-pemetrexed Combined With Concurrent Involved-field Radiotherapy for Leptomeningeal Metastasis From Solid Tumor: a Phase I/II Clinical Trial [NCT03507244]Phase 1/Phase 234 participants (Actual)Interventional2018-04-12Completed
A Phase 2 Open-label Single-arm Study to Evaluate the Combination of Pembrolizumab, Lenvatinib and Chemotherapy in Non-small Cell Lung Cancer (NSCLC) Harbouring Targetable Mutation and Failed Standard Tyrosine Kinase Inhibitors [NCT04989322]Phase 246 participants (Anticipated)Interventional2021-10-05Recruiting
Chemotherapy Combination With Local Radiotherapy and rhGM-CSF for Oligometastatic Stage IV NSCLC Patients Without Progression After First-line Chemotherapy: a Prospective Randomized Controlled Study [NCT03489616]45 participants (Anticipated)Interventional2018-01-15Recruiting
Phase I Clinical Trial of Ixabepilone and Pemetrexed in Advanced Solid Tumors [NCT01170871]Phase 10 participants (Actual)InterventionalWithdrawn(stopped due to Sponsor withdrew support)
A Randomized, Multicenter, Open-label Phase Ib/II Study of RO5083945 in Combination With Cisplatin and Gemcitabine/Pemetrexed Versus Cisplatin and Gemcitabine/Pemetrexed in Patients With Advanced or Recurrent Non Small Cell Lung Cancer Who Have Not Receiv [NCT01185847]Phase 290 participants (Actual)Interventional2010-11-30Completed
Phase II Study of Pemetrexed and Gemcitabine for Treatment Resistant Patients With Metastatic Colorectal Cancer and KRAS Mutations [NCT01109615]Phase 240 participants (Actual)Interventional2010-04-30Terminated(stopped due to Lacking effect of treatment)
An Open-Label, Phase I/Ib Dose Escalation Study to Assess the Safety and Tolerability of GSK1120212 in Combination With Docetaxel, Erlotinib, Pemetrexed, Pemetrexed + Carboplatin, Pemetrexed + Cisplatin, or Nab-Paclitaxel in Subjects With Advanced Solid T [NCT01192165]Phase 1169 participants (Actual)Interventional2010-09-14Completed
Phase 2 Study of Gefitinib Compared With Pemetrexed/Cisplatin in Advanced Non-Small [NCT01192243]Phase 268 participants (Anticipated)Interventional2009-12-31Recruiting
A Phase 1/1b Study to Evaluate the Safety and Tolerability of AB598 Monotherapy and Combination Therapy in Participants With Advanced Malignancies [NCT05891171]Phase 181 participants (Anticipated)Interventional2023-10-13Recruiting
A Phase III Randomized, Controlled, Open-label, Multicenter, Global Study of Capmatinib in Combination With Osimertinib Versus Platinum - Pemetrexed Based Doublet Chemotherapy in Patients With Locally Advanced or Metastatic NSCLC Harboring EGFR Activating [NCT04816214]Phase 36 participants (Actual)Interventional2021-09-22Terminated(stopped due to Novartis decided to terminate the study based on a business consideration and not related with any safety concerns. Randomized part was not initiated)
LIBRETTO-431: A Multicenter, Randomized, Open-Label, Phase 3 Trial Comparing Selpercatinib to Platinum-Based and Pemetrexed Therapy With or Without Pembrolizumab as Initial Treatment of Advanced or Metastatic RET Fusion-Positive Non-Small Cell Lung Cancer [NCT04194944]Phase 3261 participants (Actual)Interventional2020-02-17Active, not recruiting
Phase I Study of Cabozantinib in Combination With Pemetrexed in Advanced Non-squamous Non-small Cell Lung Cancer (NSCLC), Urothelial Cancer and Advanced Malignant Mesothelioma [NCT04173338]Phase 19 participants (Actual)Interventional2020-01-23Terminated(stopped due to Closed by IRB on 3/28/22)
Randomized Multicenter Study to Compare the Effectiveness and Safety of Erlotinib and Pemetrexed as Maintenance Therapy of Advanced Non-Squamous Non-Small Cell Lung Cancer [NCT03460678]Phase 49 participants (Actual)Interventional2018-02-28Terminated(stopped due to Difficulty recruiting)
A Multicenter, Open Clinical Trial of Using TS Gene Polymorphism to Predict Effect in Patients of Advanced Lung Adenocarcinoma to Pemetrexed Combining With Cisplatin Regiment as First-line Treatment [NCT00940069]Phase 260 participants (Actual)Interventional2009-03-31Completed
Efficacy and Safety of Anlotinib Combined With Platinum Plus Pemetrexed in T790M Mutation Negative Metastastic Non-squamous Non-small-cell Lung Cancer After Progression on First-line EGFR TKI: a Phase II, Muti-center, Single Arm Study [NCT03706287]Phase 1/Phase 262 participants (Anticipated)Interventional2018-12-06Recruiting
A Phase II, Open-Label, Multi-Cohort Study to Investigate the Preliminary Antitumor Activity, Safety, and Pharmacokinetics of the Anti-PD-1 Monoclonal Antibody BGB-A317 in Combination With Chemotherapy as First-Line Treatment in Chinese Subjects With Loca [NCT03432598]Phase 254 participants (Actual)Interventional2017-08-24Completed
A Study to Evaluate the Efficacy and Safety of Anlotinib Plus Pemetrexed as the 1-line Treatment of Patients With Platinum Intolerant Advanced Non-squamous NSCLC, With Pemetrexed Control. [NCT03768037]Phase 4106 participants (Anticipated)Interventional2018-11-26Recruiting
A Prospective, Single-center, One-arm Clinical Study of Apatinib Combined With Chemotherapy for Patients Who Progressed After First Line EGFR-TKI Treatment Without T790M Mutation [NCT03758677]Phase 230 participants (Anticipated)Interventional2020-08-01Not yet recruiting
Alflutinib Versus Alflutinib Plus Chemotherapy for Nonesmall Cell Lung Cancer With EGFR (T790M)- Associated Resistance to Initial EGFR Inhibitor Treatment: An Open-label, Randomised Phase 2 Clinical Trial [NCT05209256]Phase 2/Phase 390 participants (Anticipated)Interventional2022-03-01Not yet recruiting
International Multicenter Randomized Double-Blind Placebo-Controlled Clinical Trial Evaluating Efficacy and Safety Of BCD-100 in Combination With Pemetrexed+Cisplatin/Carboplatin Compared to Placebo in Combination With Pemetrexed+Cisplatin/Carboplatin as [NCT03912389]Phase 3292 participants (Anticipated)Interventional2019-06-01Recruiting
A Randomized Phase III Study of TaxoteRe Plus Cisplatin Versus AlImta Plus Cisplatin in 1st Line Non-squamous Cell Type Lung Cancer [NCT01282151]Phase 3148 participants (Actual)Interventional2011-07-31Terminated(stopped due to Difficulty in recruitment due to approval of maintenance pemetrexed treatment)
Phase I Trial of Haploidentical Natural Killer (NK) Cells in Combination With Pemetrexed in Patients With Stage IV Non-Small Cell Lung Cancer (NSCLC) [NCT03366064]Phase 15 participants (Actual)Interventional2017-11-09Completed
"An Open-label, Multi-center, Non-randomized Phase Ib Study to Investigate the Safety, Efficacy, and Pharmacokinetics of BAY 73-4506 Regorafenib, Administered in Combination With Pemetrexed and Cisplatin in Patients With Advanced Nonsquamous Non-Small Cel [NCT01187615]Phase 19 participants (Actual)Interventional2010-08-31Terminated
A Phase III Study to Investigate the Differential Influence of Prior Chemotherapy on the Efficacy of Erlotinib in Patients With Advanced Non-small Cell Lung Cancer (IIIB, IV) With or Without EGFR Gene Mutation [NCT01204307]Phase 2101 participants (Actual)Interventional2010-01-31Completed
A Single Arm, Phase II Clinical Trial of Orelabrutinib Combined With Pemetrexed in the Treatment for Patients With Relapsed/Refractory Central Nervous System Lymphoma [NCT05209620]Phase 230 participants (Anticipated)Interventional2021-12-21Recruiting
Efficacy and Safety of Pemetrexed Plus Cisplatin as Combination Chemotherapy for Post-operative Adenocarcinoma : Multi-center, Single Arm, Open-label, Phase Ⅱ Trial [NCT02498860]Phase 2106 participants (Actual)Interventional2015-09-01Completed
Pemetrexed Plus Apatinib Maintenance Treatment in Patients With Non-squamous Non-small Cell Lung Cancer Patients Who Have Not Progressed After 4 Cycles of Induction Chemotherapy of Pemetrexed in Combination With Platinum-based Regimen: A Prospective, Open [NCT03792503]Phase 420 participants (Anticipated)Interventional2019-02-28Not yet recruiting
Phase I Trial of Cisplatin and Pemetrexed in Combination With Panobinostat in Advanced Solid Tumors, With Emphasis on Non-Small Cell Lung Cancer [NCT01336842]Phase 123 participants (Actual)Interventional2011-04-30Completed
A Phase II Study of Pemetrexed Plus Carboplatin Combined With Radiation in Patients With Inoperable Locally Advanced Non-small Cell Lung Cancer [NCT00886678]Phase 263 participants (Anticipated)Interventional2008-07-31Active, not recruiting
Efficacy and Safety Evaluation of Percutaneous Ommaya Capsule Injection of Autologous Bi-dimensional Specific T Cells in the Treatment of Glioma and in Combination With Pemetrexed in the Treatment of Brain/Meningeal Metastases [NCT05459441]Early Phase 130 participants (Anticipated)Interventional2022-01-01Recruiting
Zanubrutinib With Pemetrexed for the Treatment of Relapsed/Refractory Primary and Secondary CNS Lymphomas: A Phase II Trial With a Safety Lead-In [NCT05681195]Phase 215 participants (Anticipated)Interventional2023-09-27Recruiting
Phase 1/2 Study of Pemetrexed Plus Cisplatin in Unresectable, Advanced Gastric Carcinoma. [NCT00320515]Phase 1/Phase 289 participants (Actual)Interventional2004-03-31Completed
A Randomized, Open-label, Phase II, 2-arm Multi-center Trial Comparing Maintenance Therapy With Pazopanib or Pemetrexed in Non-progressing Subjects With Metastatic Stage IVA and IVB Non-squamous Non-small Cell Lung Cancer (NSCLC) After Induction Therapy W [NCT01313663]Phase 220 participants (Actual)Interventional2011-02-28Terminated
A Multicenter Phase II Randomized Trial Of Immunotherapy Versus Chemotherapy Guided By Circulating Tumor DNA-Based Molecular Response On Patients With Metastatic NSCLC [NCT05715229]Phase 2108 participants (Anticipated)Interventional2023-09-29Recruiting
NASSIST (Neoadjuvant Chemoradiation +/- Immunotherapy Before Surgery for Superior Sulcus Tumors): A Randomized Phase II Trial of Trimodality +/- Atezolizumab in Resectable Superior Sulcus Non-Small Cell Lung Cancer [NCT04989283]Phase 20 participants (Actual)Interventional2021-09-09Withdrawn(stopped due to Due to no accrual)
A Feasibility Trial of Neoadjuvant Cisplatin-Pemetrexed With Atezolizumab in Combination and in Maintenance for Resectable Malignant Pleural Mesothelioma [NCT03228537]Phase 129 participants (Actual)Interventional2018-07-16Active, not recruiting
A Randomized Ph 3 Study Comparing First-Line Pemetrexed/Cisplatin Followed by Gefitinib With Gefitinib Alone in East Asian Never Smoker or Light Ex-Smoker Patients With Locally Advanced or Metastatic Nonsquamous NSCLC [NCT01017874]Phase 3236 participants (Actual)Interventional2009-11-30Completed
Phase 2 Study of Pemetrexed in Combination With Cisplatin and Cetuximab in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck [NCT01057589]Phase 266 participants (Actual)Interventional2010-02-28Completed
A Phase I/II Clinical Study of BBI608 in Combination With Pemetrexed and Cisplatin in Adult Patients With Malignant Pleural Mesothelioma [NCT02347917]Phase 1/Phase 228 participants (Actual)Interventional2015-02-28Completed
A Phase I Study of the Treatment of Recurrent Primary or Secondary CNS Lymphoma With ALIMTA (Pemetrexed), a Novel Anti-Folate [NCT00916630]Phase 118 participants (Actual)Interventional2009-01-31Completed
Randomized, Open-Label, Phase 3 Study of Pemetrexed Plus Carboplatin and Bevacizumab Followed by Maintenance Pemetrexed and Bevacizumab Versus Paclitaxel Plus Carboplatin and Bevacizumab Followed by Maintenance Bevacizumab in Patients With Stage IIIB or I [NCT00762034]Phase 3939 participants (Actual)Interventional2008-12-31Completed
An Open Label Phase Ib Dose Escalation Study to Evaluate the Safety, Tolerability, Pharmacokinetics, Immunogenicity and Maximum Tolerated Dose of Anetumab Ravtansine in Combination With Pemetrexed 500 mg/m2 and Cisplatin 75 mg/m2 in Subjects With Mesothel [NCT02639091]Phase 136 participants (Actual)Interventional2016-02-03Completed
A Phase I Open Label Study of GSK3359609 Administered Alone and in Combination With Anticancer Agents in Subjects With Selected Advanced Solid Tumors [NCT02723955]Phase 1829 participants (Actual)Interventional2016-06-23Completed
A Randomized Phase II Study to Assess the Efficacy of Pemetrexed or Sunitinib (NSC # 736511) or Pemetrexed Plus Sunitinib in the Second-Line Treatment of Advanced Non-small Cell Lung Cancer [NCT00698815]Phase 2130 participants (Actual)Interventional2008-04-15Completed
Phase 3 Study of Pemetrexed, Cisplatin, and Radiotherapy Followed by Consolidation Pemetrexed Versus Etoposide, Cisplatin, and Radiotherapy Followed by Consolidation Cytotoxic Chemotherapy of Choice in Patients With Unresectable, Locally Advanced, Stage I [NCT00686959]Phase 3598 participants (Actual)Interventional2008-09-30Completed
Pilot Study of Pemetrexed for the Treatment of Chordoma [NCT03955042]Phase 115 participants (Actual)Interventional2019-09-06Completed
A Phase 2 Double-Blind Randomized Study of Oral Enzastaurin HCl Versus Placebo Concurrently With Pemetrexed (Alimta®) as Second-Line Therapy in Patients With Advanced or Metastatic Non-Small Cell Lung Cancer [NCT00530621]Phase 2160 participants (Actual)Interventional2007-09-30Completed
Randomized Phase II Study of Pemetrexed Versus Pemetrexed and Carboplatin as Second Line Chemotherapy in Advanced Non-small-cell Lung Cancer (NSCLC). [NCT00786331]Phase 2230 participants (Anticipated)Interventional2007-07-31Recruiting
A Multicenter, Randomized, Phase II Study of Pemetrexed and Carboplatin With or Without Anlotinib Hydrochloride for Advanced or Locally-advanced Osimertinib-resistant Non-squamous Non-small Cell Lung Cancer (ALTER-L031) [NCT04136535]Phase 2105 participants (Anticipated)Interventional2019-10-18Not yet recruiting
A Phase II Study of Pemetrexed as Second-Line Treatment in Patients With Pancreatic Cancer Progressing Despite Therapy With Gemcitabine [NCT00864513]Phase 217 participants (Actual)Interventional2007-10-31Terminated(stopped due to At interim analysis the study did not meet the response criteria to continue)
A Randomized Phase II Study of Erlotinib Compared to Single Agent Chemotherapy-erlotinib Combination in Pretreated Patients With Advanced NSCLC (NVALT10 Study) [NCT00835471]Phase 2195 participants (Actual)Interventional2009-03-31Completed
Open-label Study of Bevacizumab Maintenance Therapy (AVASTIN®) With or Without Pemetrexed After First-line Chemotherapy With Bevacizumab-cisplatin-pemetrexed in Patients With Advanced, Metastatic or Recurrent Non-squamous Non-small Cell Lung Cancer (NSCLC [NCT00961415]Phase 3376 participants (Actual)Interventional2009-08-31Completed
Open-Label Single-Arm Phase 2 Study of Alimta in Patients With Recurrent or Metastatic Nasopharyngeal Carcinoma Who Have Had Prior Platinum Based Chemotherapy [NCT00630149]Phase 235 participants (Actual)Interventional2007-11-30Completed
Recombinant Human Endostatin Durative Transfusion Combined With Pemetrexed Plus Cisplatin or Carboplatin in the First-line Treatment of Advanced Lung Adenocarcinoma With Wild-type EGFR or ALK-negative [NCT02804646]Phase 4100 participants (Anticipated)Interventional2016-06-30Recruiting
A Phase I/II Study Evaluating the Safety, Tolerability, and Efficacy of GLS-012 and GLS-010 in Patients With Advanced Non-Small Cell Lung Cancer (Triumph-02) [NCT05978401]Phase 1/Phase 2152 participants (Anticipated)Interventional2023-08-10Not yet recruiting
A Phase III, Randomised, Open-Label Study of Savolitinib in Combination With Osimertinib Versus Platinum-Based Doublet Chemotherapy in Participants With EGFR Mutated, MET-Overexpressed and/or Amplified, Locally Advanced or Metastatic Non-Small Cell Lung C [NCT05261399]Phase 3324 participants (Anticipated)Interventional2022-08-03Recruiting
Pilot Study of Serial Plasma Genotyping to Guide the Adaptive Treatment of Advanced NSCLC Receiving First-line Pembrolizumab [NCT04166487]Phase 240 participants (Actual)Interventional2020-01-13Active, not recruiting
A Phase Ib Study of Selumetinib in Patients With Previously Treated or Untreated Advanced/Metastatic NSCLC Who Are Receiving Standard Chemotherapy Regimens. [NCT01783197]Phase 139 participants (Actual)Interventional2013-06-04Completed
A Randomized Phase II Study of Reolysin in Patients With Previously Treated Advanced or Metastatic, Non Small Cell Lung Cancer Receiving Standard Salvage Therapy. [NCT01708993]Phase 2166 participants (Actual)Interventional2012-12-10Completed
An Open-Label, Multicenter Study of IBI308 in Subjects With Selected Advanced Solid Tumors [NCT02937116]Phase 1233 participants (Actual)Interventional2016-10-19Completed
Randomized, Double-Blind, Phase 2/3 Study in Subjects With Malignant Pleural Mesotheliomato Assess ADI-PEG 20 With Pemetrexed and Cisplatin (ATOMIC-Meso Phase 2/3 Study) [NCT02709512]Phase 2/Phase 3249 participants (Actual)Interventional2017-08-01Completed
EGFR-TKI With Chemotherapy as First Line Treatment in Stage IIIB/IV NSCLC Patients With Both EGFR Mutation and BIM Deletion Polymorphism [NCT02859077]Phase 3100 participants (Anticipated)Interventional2016-08-31Not yet recruiting
Multicentric, Randomized, Phase III Trial Comparing 2 Strategies in Patients With Non-squamous Non-small Cell Lung Cancer With Asymptomatic Brain Metastases [NCT02162537]Phase 395 participants (Actual)Interventional2013-12-31Terminated(stopped due to Slow inclusions due in part to a change in practices. The first chemotherapy become a standard for patients with NSCL with asymptomatic brain metastases.)
A Phase 2 Randomized, Multicenter, Double-Blind Study of the Glutaminase Inhibitor Telaglenastat With Pembrolizumab and Chemotherapy Versus Placebo With Pembrolizumab and Chemotherapy in First-Line, Metastatic KEAP1/NRF2-Mutated, Nonsquamous, Non-Small Ce [NCT04265534]Phase 240 participants (Actual)Interventional2020-07-24Terminated(stopped due to Lack of Clinical Benefit)
A Randomized, Double-blinded, Phase III Study of Pemetrexed Plus Platinum Chemotherapy With or Without Sintilimab (IBI308) in First Line Advanced or Metastatic Non-squamous Non-small Cell Lung Cancer Subjects (Orient-11) [NCT03607539]Phase 3397 participants (Actual)Interventional2018-08-23Completed
Almonertinib Alone Versus Almonertinib Plus Chemotherapy as First-Line Treatment in Locally Advanced Or Metastatic NSCLC Patients With Concomitant EGFR and Tumor Suppressor Gene Mutation: A Multicenter, Open-Label, Randomized, Controlled Phase III Study ( [NCT04500717]Phase 3460 participants (Anticipated)Interventional2020-10-31Not yet recruiting
A Single-Arm Phase 2 Study to Investigate Bintrafusp Alfa With Platinum-Pemetrexed for TKI-Resistant EGFR-Mutant NSCLC [NCT04971187]Phase 23 participants (Actual)Interventional2021-06-30Terminated(stopped due to PI Request)
Four Versus Six Cycles of Pemetrexed/Platinum as a First Line Treatment of Malignant Pleural Mesothelioma; a Randomized Phase II Study [NCT02497053]Phase 270 participants (Anticipated)Interventional2015-06-30Recruiting
A Phase II Study to Evaluate Camrelizumab With Pemetrexed / Carboplatin in Patients With Brain Metastases of Driven Gene-negative, Non-squamous Non-small Cell Lung Cancer [NCT04211090]Phase 245 participants (Actual)Interventional2020-01-15Active, not recruiting
Chemotherapy in Combination With Erlotinib, or Sequential Chemotherapy for Erlotinib for Treatment, EGFR - TKI Resistance of EGFR Mutations in Patients With NSCLC Randomized Controlled Phase II Clinical Study [NCT02037997]Phase 280 participants (Anticipated)Interventional2013-12-31Recruiting
Master Protocol: A Phase 2, Open-label, Multi-arm Study of Tislelizumab in Combination With Investigational Agents With or Without Chemotherapy in Patients With Previously Untreated, Locally Advanced, Unresectable, or Metastatic Non-Small Cell Lung Cancer [NCT05635708]Phase 2200 participants (Anticipated)Interventional2023-03-07Recruiting
Phase I/II Clinical Trial of Intrathecal Pemetrexed as First Line Intrathecal Chemotherapy in Patients With Leptomeningeal Metastasis [NCT05289908]Phase 1/Phase 234 participants (Actual)Interventional2022-02-21Active, not recruiting
A Phase III, Randomized, Double-Blind Study of Bevacizumab, Carboplatin, and Paclitaxel or Pemetrexed With or Without Atezolizumab in Chemotherapy-Naïve Patients With Stage IV Non-Squamous Non-Small Cell Lung Cancer (IMpower151) [NCT04194203]Phase 3304 participants (Actual)Interventional2020-04-09Active, not recruiting
KEYMAKER-U01 Substudy 1: A Phase 2, Umbrella Study With Rolling Arms of Investigational Agents With Pembrolizumab in Combination With Chemotherapy in Treatment-Naive Patients With Advanced Non-small Cell Lung Cancer (NSCLC) [NCT04165070]Phase 2360 participants (Anticipated)Interventional2019-12-19Active, not recruiting
A Phase III, Randomized, Double-blind Trial of Platinum Doublet Chemotherapy +/-Pembrolizumab (MK-3475) as Neoadjuvant/Adjuvant Therapy for Participants With Resectable Stage II, IIIA, and Resectable IIIB (T3-4N2) Non-small Cell Lung Cancer (NSCLC) (KEYNO [NCT03425643]Phase 3797 participants (Actual)Interventional2018-04-24Active, not recruiting
A Phase Ib/II, Open-Label, Multicenter, Randomized Umbrella Study Evaluating The Efficacy And Safety Of Multiple Immunotherapy-Based Treatment Combinations In Patients With Metastatic Non-Small Cell Lung Cancer (Morpheus-Lung) [NCT03337698]Phase 1/Phase 2470 participants (Anticipated)Interventional2018-01-02Recruiting
A Phase 1 Study of RGX-104, a Small Molecule LXR Agonist, as a Single Agent and as Combination Therapy in Patients With Advanced Solid Malignancies and Expansion in Select Malignancies [NCT02922764]Phase 1150 participants (Anticipated)Interventional2016-11-30Recruiting
Combination of Induction Durvalumab and Tremelimumab Alone Versus Durvalumab and Tremelimumab With Chemotherapy for Potentially Resectable Pleural Mesothelioma [NCT05932199]Phase 1/Phase 252 participants (Anticipated)Interventional2023-09-30Not yet recruiting
Randomized Phase III Study of Maintenance Therapy With Bevacizumab, Pemetrexed, or a Combination of Bevacizumab and Pemetrexed Following Carboplatin, Paclitaxel and Bevacizumab for Advanced Non-Squamous NSCLC [NCT01107626]Phase 31,516 participants (Actual)Interventional2010-10-25Completed
Erlotinib Treatment Beyond Progression in EGFR Mutant or Patients Who Have Responded EGFR TKI in Stage IIIB/IV NSCLC [NCT02064491]Phase 218 participants (Actual)Interventional2014-02-28Completed
An Open-label, Randomized, Phase III, Multicenter Clinical Trial Comparing the Efficacy and Safety of Individualized Intraperitoneal and System Chemotherapy Versus System Chemotherapy as First-line Chemotherapy for Advanced Gastric Cancer [NCT03061058]Phase 3240 participants (Anticipated)Interventional2013-04-01Recruiting
A Phase III, Open-Label, Randomized Multicenter Study to Compare AC0010 and Pemetrexed/Cisplatin in Patients With Advanced NSCLC Who Have Progressed Following Prior EGFR TKI [NCT03058094]Phase 30 participants (Actual)Interventional2018-12-31Withdrawn(stopped due to Considiering to change the Chemotherapy into Gefitinib)
Efficacy and Safety of Maintenance Apatinib Combined With Pemetrexed After First - Line Induction Chemotherapy in Advanced Non-squamous Non-small Cell Lung Cancer: A Prospective, Open, Single Arm Clinical Study [NCT03190239]Phase 220 participants (Anticipated)Interventional2017-07-15Not yet recruiting
Penpulimab-based Combination Neoadjuvant/Adjuvant Therapy for Patients With Resectable Locally Advanced Non-small Cell Lung Cancer: a Phase II Clinical Study (ALTER-L043) [NCT04846634]Phase 290 participants (Anticipated)Interventional2021-08-31Not yet recruiting
A Randomized, Multicenter,Open-label Phase II Study is to Evaluate the Effects of Chemotherapy and Sintilimab or in Combination With Autologous Cytokine-induced Killer Cell Immunotherapy in Patients With Stage IV Non-small-cell Lung Cancer [NCT04836728]Phase 2156 participants (Anticipated)Interventional2021-04-01Not yet recruiting
A Phase 1b/2, Open-label, Randomized Study of Vudalimab in Combination With Chemotherapy or Pembrolizumab in Combination With Chemotherapy as First-line Treatment in Patients With Advanced Non-small Cell Lung Cancer [NCT06173505]Phase 1/Phase 2168 participants (Anticipated)Interventional2023-12-31Not yet recruiting
A Randomized, Open-Label, Phase 3 Study to Evaluate Zimberelimab and Domvanalimab in Combination With Chemotherapy Versus Pembrolizumab With Chemotherapy for the First-Line Treatment of Patients With Metastatic Non-Small Cell Lung Cancer With No Epidermal [NCT05502237]Phase 3720 participants (Anticipated)Interventional2022-10-12Recruiting
A Randomized, Double-blind, Multi-center, Phase III Study of AK112 or Placebo Combined With Pemetrexed and Carboplatin in Patients With EGFR-mutant Locally Advanced or Metastatic Non-squamous NSCLC Who Have Failed to EGFR-TKI Treatment [NCT05184712]Phase 3470 participants (Anticipated)Interventional2022-01-01Recruiting
Phase 2 Randomized Trial of Neoadjuvant or Palliative Chemotherapy With or Without Immunotherapy for Peritoneal Mesothelioma [NCT05001880]Phase 266 participants (Anticipated)Interventional2022-03-22Recruiting
Phase I Study of Osimertinib+Bevacizumab+Carboplatin and Pemetrexed for Untreated Patients With EGFR Mutation Advanced Non-squamous Non-Small Cell Lung Cancer With Concomitant Mutations. [NCT05507606]Phase 250 participants (Anticipated)Interventional2021-08-01Recruiting
An Open Label, Dose Escalation Followed by Dose Expansion, Safety and Tolerability Trial of CAN04, a Fully Humanized Monoclonal Antibody Against IL1RAP, in Subjects With Solid Malignant Tumors [NCT03267316]Phase 1/Phase 2167 participants (Actual)Interventional2017-09-19Active, not recruiting
"A Phase II Trial of Celecoxib Plus Chemotherapy [Docetaxel or Pemetrexed] in Patients With Previously Treated, COX Dependent Recurrent Non-Small Cell Lung Cancer" [NCT00520845]Phase 223 participants (Actual)Interventional2007-10-31Terminated(stopped due to slow accrual)
Phase II Study of Concurrent Carboplatin, Pemetrexed, and Radiotherapy for Limited Stage of Small Cell Lung Cancer [NCT00494026]Phase 24 participants (Actual)Interventional2007-09-30Terminated(stopped due to Study stopped early based on interim results of another trial, showing inferior activity of pemetrexed/carboplatin compared to etoposide/carboplatin in SCLC.)
Phase II Trial of Pemetrexed in Second Line Advanced/Metastatic Osteosarcomas [NCT00523419]Phase 232 participants (Actual)Interventional2007-09-30Completed
Phase II Trial of ALIMTA in Relapsed Small Cell Lung Cancer [NCT00191750]Phase 280 participants Interventional2004-07-31Completed
A Phase I, Open-Label, Dose-Escalation Study to Assess the Safety, Tolerability and Pharmacokinetics of AZD6918 Administered Daily as a Single Agent and in Combination Treatment in Adult Patients With Refractory Solid Malignancies [NCT00733031]Phase 175 participants (Anticipated)Interventional2008-08-31Terminated(stopped due to PK results demonstrate low and variable plasma concentrations so that achieving therapeutic concentrations is unlikely.)
Phase II Trial: Efficacy and Toxicity of Induction Pemetrexed (ALIMTA) and Oxaliplatin (ELOXATIN) in Patients With Locally Advanced Head and Neck Squamous Cell Carcinoma [NCT00503997]Phase 242 participants (Actual)Interventional2006-12-31Completed
Pemetrexed Plus Bevacizumab in Pretreated, Advanced or Metastatic Non Small Cell Lung Cancer (NSCLC) [NCT00741221]Phase 250 participants (Actual)Interventional2008-06-30Completed
A Phase 2, Open-Label, Parallel Cohort Study of Subcutaneous Amivantamab in Multiple Regimens in Patients With Advanced or Metastatic Solid Tumors Including EGFR-mutated Non-Small Cell Lung Cancer [NCT05498428]Phase 2390 participants (Anticipated)Interventional2022-11-11Recruiting
A Phase 3 Randomized, Placebo-controlled Study to Evaluate the Safety and Efficacy of Pemetrexed + Platinum Chemotherapy + Pembrolizumab (MK-3475) With or Without Lenvatinib (E7080/MK-7902) as First-line Intervention in Participants With Metastatic Nonsqu [NCT04716933]Phase 3201 participants (Actual)Interventional2019-11-05Active, not recruiting
Open-label, Phase 2 Study of Tusamitamab Ravtansine (SAR408701) Combined With Pembrolizumab and Tusamitamab Ravtansine (SAR408701) Combined With Pembrolizumab and Platinum-based Chemotherapy With or Without Pemetrexed in Patients With CEACAM5 Positive Exp [NCT04524689]Phase 2215 participants (Anticipated)Interventional2020-10-26Recruiting
A Phase III, Open-label, Randomized Study of Osimertinib With or Without Platinum Plus Pemetrexed Chemo, as First-line Treatment in Patients With Epidermal Growth Factor Receptor (EGFR) Mutation Positive, Locally Advanced or Metastatic Non-small Cell Lung [NCT04035486]Phase 3587 participants (Actual)Interventional2019-07-02Active, not recruiting
A Limited Access Phase II Trial of Pemetrexed (Alimta, LY231514) (NSC #698037) in Combination With Cisplatin (NSC #119875) in the Treatment of Advanced, Persistent, or Recurrent Carcinoma of the Cervix [NCT00691301]Phase 255 participants (Actual)Interventional2008-09-30Completed
Phase 2 Study of ALIMTA® (Pemetrexed) Plus Cisplatin as First-Line Treatment of Gastric Cancer [NCT00415168]Phase 253 participants (Actual)Interventional2006-12-31Completed
Phase II Prospective Study Evaluating the Role of Directed Cisplatin Based Chemo With Either Vinorelbine or Pemetrexed for the Adj Tx of Early Stage NSCLC in Patients Using Genomic Expression Profiles of Chemo Sensitivity to Guide Therapy [NCT00545948]Phase 231 participants (Actual)Interventional2007-12-31Terminated(stopped due to Study terminated due to reproducibility issues with genomics prediction model.)
Randomized Proteomic Stratified Phase III Study of Second-Line Erlotinib Versus Chemotherapy in Patients With Inoperable Non Small Cell Lung Cancer [NCT00989690]Phase 3275 participants (Anticipated)Interventional2008-02-29Recruiting
"A Phase 2 Trial of Pemetrexed and Cisplatin Followed Sequentially by Gefitinib Versus Pemetrexed and Cisplatin in Asian Never Smoker Patients With Advanced Non-Small Cell Lung Cancer" [NCT00409006]Phase 270 participants (Actual)Interventional2007-02-28Completed
A Phase 2 Study of Cisplatin + Pemetrexed + Avastin as First-Line Therapy in Patients With Advanced Non-Squamous, Non-Small Cell Lung Carcinoma [NCT00998166]Phase 24 participants (Actual)Interventional2007-06-30Terminated(stopped due to Poor enrollment)
Phase II Trial of Pemetrexed and Bevacizumab in Patients With Recurrent or Metastatic Head and Neck Cancer [NCT00222729]Phase 242 participants (Actual)Interventional2005-11-30Completed
A Phase 1 Dose-Finding Study Of The Anti-Angiogenesis Agent, AG-013736, In Combinations Of Paclitaxel/Carboplatin, Weekly Paclitaxel, Docetaxel, Capecitabine, Gemcitabine/Cisplatin and Pemetrexed/Cisplatin In Patients With Advanced Solid Tumors [NCT00454649]Phase 1102 participants (Actual)Interventional2005-12-31Completed
CHAMP - An Open-label, Randomised, Multicentre, Phase II Clinical Study of Panitumumab Plus Pemetrexed and Cisplatin (PemCisP) Versus PemCis in the First-line Treatment of Patients With Stage IIIB or IV Primary Nonsquamous Non-small Cell Lung Cancer, With [NCT01088620]Phase 2134 participants (Anticipated)Interventional2010-04-30Suspended(stopped due to the DSMB stopped the trial due to unacceptable side effects in the experimental arm which has not yet been verified)
A Phase 2 Clinical Trial:Low -Dose-bevacizumab and Pemetrexed Versus Treatment of Physician's Choice in Metastatic HER2-negative Breast Cancer Patients After Failure of Taxanes and Anthracycline-containing Regimens [NCT02829008]Phase 2120 participants (Anticipated)Interventional2016-04-30Active, not recruiting
A Phase II First-Line Study of a Combination of Pemetrexed, Carboplatin and Bevacizumab in Advanced Nonsquamous NSCLC Evaluating Efficacy and Tolerability in Elderly Patients (Age ≥ 70 Yrs) With Good Performance Status (PS < 2) [NCT00798603]Phase 265 participants (Actual)Interventional2008-12-31Completed
Pilot Study of Pembrolizumab Combined With Pemetrexed or Abemaciclib for the Treatment of Patients With High Grade Glioma [NCT04220892]Early Phase 10 participants (Actual)Interventional2020-07-08Withdrawn(stopped due to One of the study drugs will no longer be supplied by manufacturer and the pembrolizumab + abemaciclib study arm is removed due to toxicity seen in other trials.)
Role of Early 18F-FDG-PET/CT Scan in Predicting Mediastinal Downstaging With Neoadjuvant Chemotherapy in Resectable Stage III A NSCLC [NCT02607423]Phase 20 participants (Actual)Interventional2015-11-19Withdrawn(stopped due to The study failed to meet its accrual targets.)
A Phase II Trial of Pemetrexed (Alimta [Registered Trademark]) Combined With Sirolimus (Rapamycin, Rapamune [Registered Trademark]) in Subjects With Relapsed or Refractory NSCLC [NCT00923273]Phase 1/Phase 242 participants (Actual)Interventional2008-02-29Terminated(stopped due to Premature closure - investigator left the National Institutes of Health.)
A Phase II, Double-blind, Placebo Controlled, Randomised Study to Assess the Efficacy and Safety of ZD4054 (Zibotentan) in Combination With Pemetrexed (Alimta®) vs. Pemetrexed Alone in Patients With Non-small Cell Lung Cancer Who Have Failed One Prior Pla [NCT00745875]Phase 266 participants (Actual)Interventional2008-08-31Completed
A Randomized, Open-Label, Controlled, Multicenter Phase III Study of Camrelizumab Combined With Apatinib Mesylate or Camrelizumab Alone Versus Platinum-based Chemotherapy for First-line Treatment in Subjects With PD-L1 Positive Relapsed or Advanced NSCLC [NCT04203485]Phase 3762 participants (Anticipated)Interventional2020-06-15Not yet recruiting
A Single Arm,Open-label, Study of Fruquintinib Combined With Sintilimab and Chemotherapy in Patients With Unresectable or Metastatic Advanced Wild-type Genotype Non-squamous Non-small Cell Lung Cancer [NCT04956146]Phase 246 participants (Anticipated)Interventional2022-02-02Recruiting
Randomized Phase II Study of Durvalumab or Durvalumab Plus Chemotherapy in Kras Mutation Positive and PD-L1 High (≥ 50%) NSCLC Patients [NCT04470674]Phase 20 participants (Actual)Interventional2021-04-06Withdrawn(stopped due to Lack of Accrual)
A Phase I/II Study of VEGF-Antisense Oligonucleotide (VEGF-AS, Veglin) in Combination With Pemetrexed and Cisplatin for the Treatment of Advanced Malignant Mesothelioma [NCT00668499]Phase 1/Phase 20 participants (Actual)Interventional2008-04-30Withdrawn(stopped due to Sponsor withdrew support)
A Phase I Study of Pazopanib in Combination With Either Erlotinib or Pemetrexed in Patients With Advanced Solid Tumors [NCT00619424]Phase 158 participants (Actual)Interventional2007-11-15Completed
A Randomized, Double-blind, Phase 2 Study of BMS-986315 and Nivolumab in Combination With Chemotherapy Versus Nivolumab in Combination With Chemotherapy as First-line Treatment for Participants With Stage IV or Recurrent Non-small Cell Lung Cancer (NSCLC) [NCT06094296]Phase 2196 participants (Anticipated)Interventional2023-11-03Not yet recruiting
A Global, Randomised, Phase 3, Open-label Study of REGN2810 (ANTI-PD 1 Antibody) Versus Platinum Based Chemotherapy in First Line Treatment of Patients With Advanced or Metastatic PD L1+Non-small Cell Lung Cancer [NCT03088540]Phase 3712 participants (Actual)Interventional2017-05-29Active, not recruiting
MTA vs CEM Pulpotomy in Young Permanent Molars With a Diagnosis of Irreversible Pulpitis: A Randomized Clinical Trial [NCT04243733]40 participants (Anticipated)Interventional2018-10-10Recruiting
An Exploratory, Prospective Phase II Study to Investigate Progression-Free Survival, Response and Overall Survival Seen With Pemetrexed/Cisplatin and the Role of Thymidylate Synthase Expression [NCT00887549]Phase 270 participants (Actual)Interventional2009-04-30Completed
Evaluation of the Efficacy of Er,Cr:YSGG Laser in Partial Pulpotomy Therapy of Permanent Immature Molar Teeth With Deep Dentin Caries [NCT04010929]64 participants (Actual)Interventional2018-01-08Completed
A Phase I/II Study Of The Docetaxel/Pemetrexed Combination As First Line Treatment In Patients With Advanced/Metastatic NSCLC [NCT00684099]Phase 1/Phase 270 participants (Anticipated)Interventional2006-05-31Completed
A Phase II Study of Pemetrexed Plus Cisplatin With Concurrent Radiation Therapy Followed by Every-21-Day Docetaxel Consolidation in Patients With Inoperable Stage IIIA/B Squamous Cell Lung Cancer [NCT02787473]Phase 254 participants (Anticipated)Interventional2016-10-31Recruiting
A Phase 2 Study of Pemetrexed and Cisplatin Plus Cetuximab Followed by Pemetrexed and Cetuximab Maintenance Therapy in Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer (Stage IIIB or IV) Other Than Predominantly Squamous Cell Histol [NCT00867009]Phase 2113 participants (Actual)Interventional2009-04-30Completed
Phase II Study of Pemetrexed or Nab-paclitaxel With Pembrolizumab in Elderly (>/= 75 Years) Patients With Advanced Non-Small Cell Lung Cancer (NSCLC) [NCT04754815]Phase 20 participants (Actual)Interventional2021-04-01Withdrawn(stopped due to Logistical challenges primarily due to COVID led to delay in starting the study and now is not felt to be clinically relevant.)
Pembrolizumab + Platinum Doublets Without Radiation for Patients With PD-L1 ≥50% Locally Advanced Non-small Cell Lung Cancer: a Multicenter Prospective Single Arm Phase II Study [NCT04153734]Phase 221 participants (Anticipated)Interventional2019-12-01Not yet recruiting
Randomized, Double-Blind, Placebo Controlled, Phase 2 Study of Pemetrexed and Cisplatin Plus Enzastaurin Versus Pemetrexed and Cisplatin Plus Placebo in Chemonaive Patients With Advanced, Unresectable, or Metastatic (Stage IIIB or IV) Nonsquamous Non-Smal [NCT00538681]Phase 235 participants (Actual)Interventional2007-09-30Terminated(stopped due to Terminated due to lack of efficacy demonstrated in relevant participant population in other clinical trials.)
An Open-label, Randomized, Phase I/II Trial Investigating the Safety and Efficacy of IO102 in Combination With Pembrolizumab, With or Without Chemotherapy, as First-line Treatment for Patients With Metastatic Non-Small Cell Lung Cancer [NCT03562871]Phase 1/Phase 2109 participants (Actual)Interventional2018-08-22Completed
An Open-label, Multi-center Phase Ib/III Study Evaluating the Efficacy and Safety of IBI351 in Combination With Sintilimab ± Chemotherapy in Advanced Non-squamous Non-small Cell Lung Cancer Subjects With KRAS G12C Mutation [NCT05504278]Phase 1144 participants (Anticipated)Interventional2022-09-20Recruiting
QUILT 2.023: A Phase 3, Open-Label, 3-Cohort Randomized Study of N-803, in Combination With Current Standard of Care VS Standard of Care as First-Line Treatment for Patients With Advanced or Metastatic NSCLC. [NCT03520686]Phase 31,538 participants (Anticipated)Interventional2018-05-18Active, not recruiting
A Phase I/II Study of Concurrent Pemetrexed/Cisplatin/Radiation in Stage IIIA/B Non-Small Cell Lung Cancer [NCT00529100]Phase 1/Phase 249 participants (Actual)Interventional2005-12-31Completed
Phase II Combination of Pemetrexed and Oxaliplatin in Patients With Recurrent Non-Small Cell Lung Cancer After Failure to Platinum Based Adjuvant Chemotherapy [NCT00612677]Phase 21 participants (Actual)Interventional2007-06-30Terminated(stopped due to slow accrual - the 1 patient accrued did not go on treatment)
A Randomized Phase 3 Study Comparing Pemetrexed-Carboplatin With Docetaxel-Carboplatin as First-Line Treatment for Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer [NCT00520676]Phase 3260 participants (Actual)Interventional2007-10-31Completed
A Randomized, Open-label, Phase 3 Study of MK-2870 vs Chemotherapy (Docetaxel or Pemetrexed) in Previously Treated Advanced or Metastatic Nonsquamous Non-small Cell Lung Cancer (NSCLC) With EGFR Mutations or Other Genomic Alterations [NCT06074588]Phase 3556 participants (Anticipated)Interventional2023-11-12Recruiting
A Phase II, Randomized, Multicenter, Double-Blind, Controlled Study of Tobemstomig Plus Platinum-Based Chemotherapy Versus Pembrolizumab Plus Platinum-Based Chemotherapy in Patients With Previously Untreated Locally Advanced or Metastatic Non-Small Cell L [NCT05775289]Phase 2180 participants (Anticipated)Interventional2023-03-15Recruiting
A Phase 3 Randomized, Open-label Clinical Study to Evaluate the Pharmacokinetics and Safety of Subcutaneous Pembrolizumab Coformulated With Hyaluronidase (MK-3475A) Versus Intravenous Pembrolizumab, Administered With Chemotherapy, in the First-line Treatm [NCT05722015]Phase 3339 participants (Anticipated)Interventional2023-02-14Active, not recruiting
Open-label Phase 3 Study of MK-7684A (Coformulation of Vibostolimab With Pembrolizumab) in Combination With Concurrent Chemoradiotherapy Followed by MK-7684A Versus Concurrent Chemoradiotherapy Followed by Durvalumab in Participants With Unresectable, Loc [NCT05298423]Phase 3784 participants (Anticipated)Interventional2022-05-03Recruiting
Avelumab Master Protocol: An Open-label Continuation Study for Participants Continuing From Pfizer-sponsored Avelumab Clinical Studies. [NCT05059522]Phase 3262 participants (Anticipated)Interventional2021-09-29Recruiting
A Phase II, Open-Label, Multicenter Study Evaluating the Safety and Efficacy of Neoadjuvant and Adjuvant Tiragolumab Plus Atezolizumab, With or Without Platinum-Based Chemotherapy, in Patients With Previously Untreated Locally Advanced Resectable Stage II [NCT04832854]Phase 250 participants (Actual)Interventional2021-04-23Active, not recruiting
An Open Label, Multicenter Extension Study in Patients Previously Enrolled in a Genentech and/or F. Hoffmann-La Roche Ltd Sponsored Atezolizumab Study (IMbrella B) [NCT03768063]Phase 31,000 participants (Anticipated)Interventional2019-02-28Recruiting
A Phase 3, Open-Label, Randomized, Multi-Center Study of DZD9008 Versus Platinum-Based Doublet Chemotherapy as First-Line Treatment for Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer Harboring Epidermal Growth Factor Receptor Exon [NCT05668988]Phase 3320 participants (Anticipated)Interventional2022-12-13Recruiting
Phase I/II Study of BLU-451 in Advanced Cancers With EGFR Exon 20 Insertion Mutations [NCT05241873]Phase 1/Phase 2332 participants (Anticipated)Interventional2022-03-04Recruiting
An Open-label, Randomized, Multicenter, Phase 3 Study to Assess the Efficacy and Safety of Trastuzumab Deruxtecan as First-line Treatment of Unresectable, Locally Advanced, or Metastatic NSCLC Harboring HER2 Exon 19 or 20 Mutations (DESTINY-Lung04) [NCT05048797]Phase 3264 participants (Anticipated)Interventional2021-10-28Recruiting
A Phase I/Ib Trial of Pirfenidone Combined With Standard First-Line Chemotherapy in Advanced-Stage Lung NSCLC [NCT03177291]Phase 148 participants (Actual)Interventional2017-09-26Active, not recruiting
Safety and Efficacy of Anlotinib Hydrochloride Combined With Pemetrexed Plus Cisplatin/Carboplatin (AP) as First Line Treatment for Stage IIIB/IIIC/IV Non-squamous Non-small-cell Lung Cancer [NCT04012619]Phase 18 participants (Actual)Interventional2019-04-30Completed
Phase II Prospective Study Evaluating the Role of Personalized Chemotherapy Regimens for Chemo-Naive Select Stage IIIB and IV Non-Small Cell Lung Cancer (NSCLC) in Patients Using a Genomic Predictor of Platinum Resistance to Guide Therapy [NCT00509366]Phase 2101 participants (Actual)Interventional2007-05-31Terminated(stopped due to Study terminated due to reproducibility issues with genomics prediction model.)
A Randomized Phase 2 Study of Pemetrexed in Combination With Cisplatin or Carboplatin in the First Line Therapy of Advanced NSCLC [NCT00402051]Phase 2133 participants (Actual)Interventional2006-11-30Completed
Phase II Randomized Study of Pemetrexed With Sorafenib Versus Pemetrexed Alone as Second-Line Therapy in Patients With Advanced Non-Small Cell Lung Cancer [NCT00454194]Phase 2110 participants (Actual)Interventional2007-09-30Completed
A Randomized Phase II Trial of Pemetrexed/Gemcitabine/Bevacizumab or Pemetrexed/Carboplatin/Bevacizumab in the First-Line Treatment of Elderly Patients With Advanced Non-Small Cell Lung Cancer [NCT00456261]Phase 2110 participants (Actual)Interventional2007-03-31Completed
Phase 1, Dose Escalation Study Of CP-751,871 In Combination With Cisplatin And Gemcitabine In Previously Untreated Patients With Advanced Non-Small Cell Lung Cancer [NCT00560573]Phase 146 participants (Actual)Interventional2007-11-30Completed
Osimertinib Then Chemotherapy in EGFR-mutated Lung Cancer With Osimertinib Third-line Rechallenge [NCT04335292]Phase 2200 participants (Anticipated)Interventional2021-01-06Recruiting
A Phase II Study of ALIMTA® (Pemetrexed) and GEMZAR® (Gemcitabine) Every 14 Days Versus Pemetrexed and Gemcitabine Every 21 Days in Advanced Non-Small Cell Lung Cancer [NCT00383331]Phase 219 participants (Actual)Interventional2007-02-28Terminated(stopped due to Trial was stopped early due to low enrollment)
A Randomized Phase II Study of Two Chemotherapy Regimens, Pemetrexed-Carboplatin, and Gemcitabine-Vinorelbine, in Anthracycline and Taxanes Pretreated Advanced Breast Cancer Patients [NCT00325234]Phase 2135 participants (Actual)Interventional2006-06-30Completed
A Randomized Phase 2 Study of Pemetrexed in Combination With Cisplatin or Carboplatin as Adjuvant Chemotherapy in Patients With Completely Resected Stage Ib or II Non-Small Cell Lung Cancer [NCT00269152]Phase 2122 participants (Actual)Interventional2005-12-31Completed
A Phase II Study of Pemetrexed Chemotherapy in Poor-Risk Patients With Advanced Head and Neck Cancer [NCT00293579]Phase 25 participants (Actual)Interventional2006-02-28Completed
A Randomized, Double-Blind, Placebo-Controlled Trial of Tomivosertib in Combination With Anti-PD-(L)1 Therapy in Subjects With NSCLC as First Line Therapy or When Progressing on Single-Agent First-Line Anti PD (L)1 Therapy [NCT04622007]Phase 2180 participants (Anticipated)Interventional2021-06-02Recruiting
A First-in-Human, Open-label, Dose-escalation Trial With Expansion Cohorts to Evaluate Safety and Anti-tumor Activity of GEN1042 in Subjects With Malignant Solid Tumors [NCT04083599]Phase 1/Phase 21,287 participants (Anticipated)Interventional2019-09-17Recruiting
A Phase III, Randomized, Multi-Center, Open-Label, Comparative Global Study to Determine the Efficacy of Durvalumab or Durvalumab and Tremelimumab in Combination With Platinum-Based Chemotherapy for First-Line Treatment in Patients With Metastatic Non Sma [NCT03164616]Phase 31,186 participants (Actual)Interventional2017-06-01Active, not recruiting
Combining Afatinib and Concurrent Chemotherapy, Followed by Osimertinib and Concurrent Chemotherapy, in Untreated EGFR Positive NSCLC Tumors [NCT05298176]Phase 221 participants (Anticipated)Interventional2022-01-04Recruiting
An Open Label, Randomized Study of Neoadjuvant Nivolumab and Chemotherapy, With or Without Sub-ablative Stereotactic Body Radiation Therapy, for Resectable Stage IIA to IIIB Non-small Cell Lung Cancer [NCT05500092]Phase 252 participants (Anticipated)Interventional2023-01-25Recruiting
Phase I Study of Sorafenib, Pemetrexed, and Cisplatin for the Treatment of Advanced Solid Tumors. [NCT00703638]Phase 116 participants (Actual)Interventional2008-05-31Completed
Additional Chemotherapy for EGFRm Patients With the Continued Presence of Plasma ctDNA EGFRm at Week 3 After Start of Osimertinib 1st-line Treatment (PACE-LUNG) [NCT05281406]Phase 250 participants (Anticipated)Interventional2021-11-12Recruiting
A Randomized, Double-Blind, Phase III Study of Platinum+Pemetrexed Chemotherapy With or Without Pembrolizumab (MK-3475) in First Line Metastatic Non-squamous Non-small Cell Lung Cancer Subjects (KEYNOTE-189) [NCT03950674]Phase 340 participants (Actual)Interventional2016-02-22Completed
A Randomized, Double-Blind, Phase III Study of Platinum+Pemetrexed Chemotherapy With or Without Pembrolizumab (MK-3475) in First Line Metastatic Non-squamous Non-small Cell Lung Cancer Subjects (KEYNOTE-189) [NCT02578680]Phase 3616 participants (Actual)Interventional2016-01-15Completed
Biomarker Study Accompanying the AIO-TRK-0114 Study (MARBLE) [NCT02595840]Phase 24 participants (Actual)Interventional2015-11-25Terminated(stopped due to Futility)
A Phase II Study of Concurrent Pemetrexed and Radiation for Poor-Risk Stage III Non-Small Cell Lung Cancer: Hoosier Oncology Group LUN08-129 [NCT00732303]Phase 28 participants (Actual)Interventional2009-01-31Terminated(stopped due to Study terminated due to withdrawal of pharmaceutical funding)
A Phase II Trial of Paclitaxel, Bevacizumab and Pemetrexed in Patients With Untreated, Advanced Non-Small Cell Lung Cancer Using Web-Based Data Collection, Patient Self-Reporting of Adverse Effects and Automated Response Assessment [NCT00807573]Phase 244 participants (Actual)Interventional2008-12-31Completed
Multicenter, Randomized, Double-blind, Phase III Trial to Investigate the Efficacy and Safety of Oral BIBF 1120 Plus Standard Pemetrexed Therapy Compared to Placebo Plus Standard Pemetrexed Therapy in Patients With Stage IIIB/IV or Recurrent Non Small Cel [NCT00806819]Phase 3718 participants (Actual)Interventional2008-12-31Completed
An Open-Label Clinical Trial of MORAb-009 in Combination With Pemetrexed and Cisplatin in Subjects With Mesothelioma [NCT00738582]Phase 289 participants (Actual)Interventional2008-12-31Completed
A Phase II Study of Pemetrexed Plus Gemcitabine for Metastatic/Recurrent Head and Neck Cancer (HNSCC) [NCT00589667]Phase 226 participants (Actual)Interventional2006-09-30Completed
An Open-label, Multicentre, Randomised Phase II Study of Pazopanib in Combination With Pemetrexed in First-line Treatment of Subjects With Predominantly Non-squamous Cell Stage IIIBwet/IV Non-small Cell Lung Cancer [NCT00871403]Phase 2107 participants (Actual)Interventional2009-07-31Completed
Open Label Phase 1 Dose Finding Study of TRC102 in Combination With Pemetrexed in Patients With Advanced or Metastatic Solid Cancer for Whom Curative Therapy is Unavailable [NCT00692159]Phase 128 participants (Actual)Interventional2008-06-30Completed
MK-0646 IMPACT Study: MK-0646, Insulin Growth Factor 1 Receptor Antibody in Stage IIIB or IV Metastatic Non-Squamous Lung Cancer, Combined With Pemetrexed (Alimta) and Cisplatin, a Randomized Phase II Trial. [NCT00799240]Phase 227 participants (Actual)Interventional2009-06-30Completed
A Randomised Open-label Phase II Trial of BI 6727 Monotherapy and BI 6727 in Combination With Standard Dose Pemetrexed Compared to Pemetrexed Monotherapy in Second Line Non-small Cell Lung Cancer [NCT00824408]Phase 2143 participants (Actual)Interventional2009-03-31Completed
A Phase 1/2 Study of Aflibercept Administered in Combination With Pemetrexed and Cisplatin in Patients With Advanced Carcinoma [NCT00794417]Phase 1/Phase 260 participants (Actual)Interventional2008-11-30Terminated
A Phase II Study of a Combination of MTA (LY231514) and Gemcitabine in Patients With Metastatic Breast Cancer. [NCT00034489]Phase 20 participants InterventionalCompleted
A Phase II Study of Erlotinib and Chemotherapy for Patients With Stage IB-IIIA NSCLC With EGFR Mutations (ECON) [NCT00577707]Phase 29 participants (Actual)Interventional2007-11-30Completed
Phase I/II Study of Concurrent Cisplatin, Pemetrexed, and Radiotherapy for Limited Stage Small Cell Lung Cancer [NCT00447421]Phase 1/Phase 29 participants (Actual)Interventional2007-02-28Terminated(stopped due to Interim results of another trial showed inferior activity of treatment)
Stereotactic Radiosurgery and Systemic Dose Chemotherapy for Locally Advanced Lung Cancer (Protocol Number GK001) [NCT02568033]Early Phase 122 participants (Anticipated)Interventional2013-10-31Recruiting
Phase 1 Study Of Sunitinib (SU011248) In Combination With Pemetrexed In Patients With Advanced Solid Malignancies In Japan [NCT00732992]Phase 112 participants (Actual)Interventional2008-08-31Completed
Feasibility of Administering Adjuvant Chemotherapy of Pemetrexed Followed by Pemetrexed/Oxaliplatin Immediately Post-VATS in Patients With Completely Resected NSCLC [NCT00923637]Phase 275 participants (Anticipated)Interventional2009-06-30Recruiting
Systemic Chemotherapy Combined With Thoracic Cavity Perfusion of Recombinant Human Adenovirus Type 5 and Endostatin Injections Versus Cisplatin for Treatment Malignant Hydrothorax in Non Small Cell Lung Cancer (NSCLC) Patients: A Multi-center, Randomized, [NCT02579564]Phase 3134 participants (Anticipated)Interventional2016-10-31Not yet recruiting
A Multicenter, Double Blind, Randomized, Controlled Study of M7824 With Concurrent Chemoradiation Followed by M7824 Versus Concurrent Chemoradiation Plus Placebo Followed by Durvalumab in Participants With Unresectable Stage III Non-small Cell Lung Cancer [NCT03840902]Phase 2168 participants (Actual)Interventional2019-04-16Terminated(stopped due to Based on recommendations by an external IDMC, Sponsor decided to discontinue this clinical study due to a low likelihood of achieving superiority in the efficacy endpoints versus standard of care.)
A Randomized Parallel Group Phase III Trial of OSE2101 as 2nd or 3rd Line Compared With Standard Treatment (Docetaxel or Pemetrexed) in HLA-A2 Positive Patients With Advanced Non-Small-Cell Lung Cancer With Progressive Disease After Last Treatment With Im [NCT02654587]Phase 3363 participants (Anticipated)Interventional2016-02-29Active, not recruiting
0822GCC Randomized, Double-Blind, Placebo-Controlled Multicenter Phase 2 Study of the Efficacy and Safety of Apricoxib in Combination With Either Docetaxel or Pemetrexed in Non-Small Cell Lung Cancer Patients [NCT00771953]Phase 2109 participants (Actual)Interventional2008-11-30Completed
Pilot Study to Determine Therapeutic Response of Pemetrexed (Alimta) in Recurrent or Progressive Primary Central Nervous System Lymphoma (PCNSL) by Establishing the Radiographic Response Rate Using Modified Macdonald Criteria [NCT00712062]Phase 20 participants (Actual)Interventional2009-02-28Withdrawn(stopped due to Lack of accrual)
Efficacy and Safety of Immune Checkpoint (PD-1) Inhibitor Combined With Pemetrexed Intrathecal Injection in Patients With Leptomeningeal Metastases in NSCLC [NCT06132698]Phase 217 participants (Anticipated)Interventional2023-11-30Not yet recruiting
A Study of Pemetrexed Plus Carboplatin Followed by Maintenance Pemetrexed vs Paclitaxel Plus Carboplatin and Bevacizumab Followed by Maintenance Bevacizumab in Patients With Advanced NCSLC of Nonsquamous Histology [NCT00948675]Phase 3361 participants (Actual)Interventional2009-09-01Completed
A Randomized, Double-blind, Multi-center, Phase III Clinical Study Assessing the Efficacy and Safety of Sintilimab ± IBI305 Combined With Pemetrexed and Cisplatin in Patients With EGFR-mutant Locally Advanced or Metastatic Non-squamous Non-small Cell Lung [NCT03802240]Phase 3492 participants (Actual)Interventional2019-07-11Completed
Phase II Study of Neo-adjuvant Pemetrexed (ALIMTA) Plus Cisplatin Followed by Surgery and Radiation Therapy for Malignant Pleural Mesothelioma [NCT00895648]Phase 26 participants (Actual)Interventional2009-01-31Terminated(stopped due to low accrual due to competing study for same group of patients)
Efficacy and Safety of Toripalimab (JS001) Combined With Pemetrexed and Anlotinib for Patients With T790M Positive Non-Small Cell Lung Cancer After Osimertinib Resistance:a Phase II,Muti-center, Single Arm Study [NCT04316351]Phase 260 participants (Anticipated)Interventional2020-04-01Recruiting
[NCT02795884]Phase 30 participants (Actual)Interventional2016-06-30Withdrawn(stopped due to Because of reconsideration of using erlotinib(EGFR Tyrosine kinase inhibitor) as adjuvant aim)
Advanced Non-small Cell Lung Cancer With Chinese Medicine Comprehensive Treatment Plan [NCT02777788]Phase 2/Phase 3120 participants (Anticipated)Interventional2014-09-30Active, not recruiting
Phase I Study of Concomitant Pemetrexed and CDDP Plus Radiation Therapy in Patients With Locally Advanced or Metastatic Esophageal or Gastroesophageal (GEJ) Carcinomas [NCT00701857]Phase 110 participants (Actual)Interventional2008-02-29Completed
Randomized Phase II Study of Pemetrexed Alone vs Pemetrexed Plus Cisplatin in Patients With EGFR Mutation-positive Advanced NSCLC After First Line EGFR-TKIs Failure [NCT02725918]Phase 2150 participants (Anticipated)Interventional2016-04-01Recruiting
Identifying Genetic Predictors of Durable Clinical Benefit to Pembrolizumab in Advanced Non-small Cell Lung Cancer (NSCLC) Alone and in Combination With Chemotherapy. [NCT02710396]Phase 219 participants (Actual)Interventional2016-05-31Terminated(stopped due to Frontline pembrolizumab approved in NSCLC as monotherapy and in combination with chemotherapy representing a new standard of care.)
A Phase 2 Study of Pemetrexed and Erlotinib for Metastatic Colorectal Cancer Refractory to Standard Chemotherapy [NCT02723578]Phase 250 participants (Actual)Interventional2015-12-01Completed
Phase II Study of Gemcitabine and Pemetrexed in Primary Unknown Adenocarcinoma [NCT00191503]Phase 230 participants Interventional2005-01-31Completed
Phase II Study of First-Line Therapy With Pemetrexed and Gemcitabine in Patients With Advanced Non-Small Cell Lung Cancer [NCT00193414]Phase 272 participants (Actual)Interventional2005-05-31Completed
Post-marketing Clinical Trial of Induction Chemotherapy of Pemetrexed Plus Carboplatin Followed by Pemetrexed Maintenance Therapy for Advanced Nonsquamous Non-small Cell Lung Cancer [NCT01020786]Phase 4109 participants (Actual)Interventional2009-11-30Completed
A Phase II Trial With Pemetrexed Plus Cisplatin as First Line Chemotherapy for Advanced Non - Small Cell Lung Cancer (NSCLC) Patients With Measurable Asymptomatic Brain Metastasis (GFPC 07-01/METAL). [NCT00744900]Phase 245 participants (Actual)Interventional2008-09-30Completed
An Open Label Pilot Study of NovoTTF-100L in Combination With Pemetrexed (Alimta®) for Advanced Non-small Cell Lung Cancer [NCT00749346]Phase 1/Phase 242 participants (Anticipated)Interventional2008-05-31Completed
A Randomised Phase II Open-label Study With a Phase Ib Safety lead-in Cohort of ONCOS-102, an Immune-priming GM-CSF Coding Oncolytic Adenovirus, and Pemetrexed/Cisplatin in Patients With Unresectable Malignant Pleural Mesothelioma [NCT02879669]Phase 1/Phase 231 participants (Actual)Interventional2016-06-30Active, not recruiting
Pilot Study of the Feasibility of Intrapleural Photodynamic Therapy in a Multimodal Treatment Combining Extended Pleurectomy/Decortication, Adjuvant Chemotherapy and Prophylactic Radiotherapy in Patients With Malignant Pleural Mesothelioma [NCT02662504]Phase 26 participants (Actual)Interventional2016-01-16Completed
A Multicentre Randomised Phase III Trial Comparing Atezolizumab Plus Bevacizumab and Standard Chemotherapy Versus Bevacizumab and Standard Chemotherapy as First-line Treatment for Advanced Malignant Pleural Mesothelioma [NCT03762018]Phase 3401 participants (Actual)Interventional2019-04-30Active, not recruiting
Thymosin Alpha 1 Plus Maintenance Therapy With the Standard of Care (SoC) Chemotherapy Plus Cisplatin (or Carboplatin) in Patients With Metastatic Non-Small Cell Lung Cancer (NSCLC), EGFR Wild Type [NCT02906150]Phase 2140 participants (Anticipated)Interventional2016-09-30Not yet recruiting
A Randomized Phase 3 Trial of Alimta (Pemetrexed) and Carboplatin Versus Etoposide and Carboplatin in Extensive-Stage Small Cell Lung Cancer [NCT00363415]Phase 3908 participants (Actual)Interventional2006-08-31Completed
A Multi-center, Randomized, Double-blind, Phase III Trial of SHR-1210 in Combination With Famitinib or Placebo Plus Chemotherapy in Subjects With Non-squamous Non-small-cell Lung Cancer. [NCT04619433]Phase 3560 participants (Anticipated)Interventional2021-02-01Recruiting
An Exploratory Phase 2 Study of Pemetrexed/Cisplatin as Pre-operative Chemotherapy in the Treatment of Stage IIIAN2 Nonsquamous Non-Small Cell Lung Cancer [NCT01165021]Phase 219 participants (Actual)Interventional2010-11-30Completed
Phase III Study Comparing Maintenance With Pemetrexed or Gemcitabine to a Surveillance in Elderly Patients (70 Years Old and More) With a Advanced Non Small Cell Lung Cancer Controlled by Induction Chemotherapy. [NCT01850303]Phase 3632 participants (Actual)Interventional2013-05-16Completed
An Extended Feasibility Phase I/II Study of Methylenetetrahydrofolate and Pemetrexed Single Agent, Given as Neoadjuvant Treatment in Patients With Resectable Rectal Cancer [NCT01397305]Phase 1/Phase 224 participants (Actual)Interventional2011-04-14Completed
A Phase III, Double-Blinded, Multicenter, Randomized Study Evaluating the Efficacy and Safety of Neoadjuvant Treatment With Atezolizumab or Placebo in Combination With Platinum-Based Chemotherapy in Patients With Resectable Stage II, IIIA, or Select IIIB [NCT03456063]Phase 3453 participants (Actual)Interventional2018-04-24Active, not recruiting
A Randomized Phase 3 Trial of ALIMTA and Cisplatin Versus GEMZAR and Cisplatin in Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer [NCT00087711]Phase 31,713 participants (Actual)Interventional2004-07-31Completed
A Randomized Phase II Study Comparing Pemetrexed Plus Cisplatin With Gemcitabine Plus Cisplatin According to Thymidylate Synthase Expression in Non-squamous Non-small Cell Lung Cancer [NCT01401192]Phase 2304 participants (Anticipated)Interventional2011-07-31Recruiting
Adjuvant Chemotherapy Versus Observation in Fully Resected Stage I Lung Adenocarcinoma With High Risk of Post-operative Recurrence [NCT03380468]Phase 2/Phase 3300 participants (Anticipated)Interventional2018-01-01Recruiting
Pemetrexed and Platinum Use in the Neoadjuvant Setting for Resectable Stage II and IIIA Lung Adenocarcinoma [NCT02980991]Phase 280 participants (Actual)Interventional2015-12-31Completed
A Phase Ib, Open-Label, Dose-Escalation Study of the Safety and Pharmacology Of PI3-Kinase Inhibitor GDC-0941 In Combination With Either Paclitaxel And Carboplatin (With or Without Bevacizumab) or Pemetrexed, Cisplatin, And Bevacizumab in Patients With Ad [NCT00974584]Phase 165 participants (Actual)Interventional2009-10-31Completed
Phase 2 Randomized, Controlled, Open-label Study of Pemetrexed Versus Gefitinib in Patients With Locally Advanced or Metastatic Non Small Cell Lung Cancer Who Have Previously Received Platinum-Based Chemotherapy Without EGFR Mutations [NCT00891579]Phase 2161 participants (Actual)Interventional2009-02-28Completed
Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan [NCT01004601]179 participants (Actual)Interventional2005-03-31Completed
A Phase II Trial of Carboplatin, Bevacizumab and Pemetrexed in Advanced Non-Small Cell Lung Cancer [NCT00614822]Phase 250 participants (Actual)Interventional2007-11-30Completed
A Randomized Phase 2 Study Comparing Pemetrexed Plus Best Supportive Care With Best Supportive Care as Maintenance, Following First-Line Treatment With Pemetrexed-Cisplatin, in Patients With Advanced Non-Squamous Non-Small Cell Lung Cancer [NCT00606021]Phase 2106 participants (Actual)Interventional2008-01-31Completed
Pemetrexed With Simplified Folate and Dexamethasone Supplementation Versus Pemetrexed With Standard Supplementation as Second-line Chemotherapy for Patients With Non-squamous Non-small Cell Lung Cancer [NCT00609518]Phase 2111 participants (Actual)Interventional2008-02-29Completed
A Randomised, Open-label, Phase III Study of BIBW 2992 Versus Chemotherapy as First-line Treatment for Patients With Stage IIIB or IV Adenocarcinoma of the Lung Harbouring an EGFR Activating Mutation [NCT00949650]Phase 3345 participants (Actual)Interventional2009-08-14Completed
Pemetrexed and LBH589 in Previously-Treated Patients With Advanced Non-Small Cell Lung Cancer [NCT00907179]Phase 112 participants (Actual)Interventional2009-07-31Terminated(stopped due to Widespread use firstline Pemetrexed; slow recruitment; funding withdrawn..)
A Randomized Phase III Trial of Adjuvant Chemotherapy in Patients With Early Stage Non-Small Cell Lung Cancer Associated With Banking of Frozen Tumor Specimens and Collection of Gene Expression Profile Data [NCT00863512]Phase 334 participants (Actual)Interventional2009-03-31Terminated
Phase II Trial of Pemetrexed and Bevacizumab for Recurrent Ovarian and Primary Peritoneal Carcinoma [NCT00868192]Phase 238 participants (Actual)Interventional2008-05-31Completed
A Study to Evaluate the Safety and Feasibility of the Combined Use of Nivolumab With Pemetrexed for the Treatment of Advanced Squamous Cell Carcinoma of the Head and Neck [NCT04107103]Phase 220 participants (Anticipated)Interventional2020-03-19Recruiting
A Phase 3, Double-Blind, Placebo-Controlled Study of Maintenance Pemetrexed Plus Best Supportive Care Versus Best Supportive Care Immediately Following Induction Treatment With Pemetrexed + Cisplatin for Advanced Non-squamous Non-Small Cell Lung Cancer. [NCT00789373]Phase 3939 participants (Actual)Interventional2008-11-30Completed
Phase 2 Trial of Pemetrexed (Alimta™) Combined With Paclitaxel in Patients With Recurrent/Advanced Follicular, Papillary or Anaplastic Thyroid Cancer [NCT00786552]Phase 247 participants (Anticipated)Interventional2008-11-30Recruiting
A Randomized Phase 3 Multicenter Open-Label Study to Compare the Efficacy of TAK-788 as First-Line Treatment Versus Platinum-Based Chemotherapy in Patients With Non-Small Cell Lung Cancer With EGFR Exon 20 Insertion Mutations [NCT04129502]Phase 3354 participants (Actual)Interventional2020-01-10Active, not recruiting
Randomized Phase II Trial, Comparing Standard of Care Chemotherapy (Pemetrexed or Docetaxel) Plus Erlotinib to Standard of Care Chemotherapy (Pemetrexed or Docetaxel) Alone in EGFR TKI-Responsive Non-Small Cell Lung Cancer [NCT00660816]Phase 246 participants (Actual)Interventional2008-01-31Completed
Phase 1/1b Study Investigating Safety, Tolerability, PK and Antitumor Activity of Anti-TIGIT Monoclonal Antibody BGB-A1217 in Combination With Anti-PD-1 Monoclonal Antibody Tislelizumab in Patients With Advanced Solid Tumors [NCT04047862]Phase 1542 participants (Anticipated)Interventional2019-08-26Recruiting
A Phase 1 Open Label, Multi-Arm, Multicenter Study of MK-4830 as Monotherapy and in Combination With Pembrolizumab for Participants With Advanced Solid Tumors [NCT03564691]Phase 1442 participants (Anticipated)Interventional2018-07-11Active, not recruiting
A Phase III, Multi-center, Randomized Trial of Pemetrexed and Gefitinib in Never-smoker and Adenocarcinoma Patients With Non-small Cell Lung Cancer Previously Treated With Platinum-based Chemotherapy [NCT01066195]Phase 3129 participants (Anticipated)Interventional2008-05-31Enrolling by invitation
Randomized Phase 2 Study Of Cisplatin/Pemetrexed With Or Without Axitinib (AG-013736) As First-Line Treatment For Patients With Non-Squamous Non-Small Cell Lung Cancer [NCT00768755]Phase 1/Phase 2180 participants (Actual)Interventional2009-01-31Completed
A Phase 2 Study of ALIMTA in Solid Tumor Patients With Stable Third-Space Fluid [NCT00316225]Phase 231 participants (Actual)Interventional2006-12-31Completed
A Phase III Randomized, Open-Label, Multi-Center Study of Durvalumab (MEDI4736) Versus Standard of Care (SoC) Platinum-Based Chemotherapy as First Line Treatment in Patients With PD-L1-High Expression Advanced Non Small-Cell Lung Cancer [NCT03003962]Phase 3669 participants (Actual)Interventional2017-01-02Active, not recruiting
Nedaplatin or Cisplatin Combined With Pemetrexed in the First Line Treatment of Advanced Adenocarcinoma:A Prospective Multi-center Phase III Randomized Controlled Trial [NCT02607592]Phase 3293 participants (Anticipated)Interventional2015-08-31Recruiting
PD-1 Immune Checkpoint Inhibitors and Immune-Related Adverse Events: a Cohort Study [NCT04115410]4,724 participants (Anticipated)Observational2020-07-01Not yet recruiting
Clinical and Radiographic Evaluation of Premixed Verses Powder / Liquid Bioceramic Mineral Trioxide Aggregate in Indirect Pulp Capping of Immature Permanent Mandibular Molars: A Randomized Clinical Trial [NCT05597553]24 participants (Anticipated)Interventional2023-01-01Not yet recruiting
Efficacy and Safety of Rh-endostatin(Endostar)Combined With Platinum-based Doublet Chemotherapy and Pembrolizumab as First Line Therapy in Patients With Advanced or Metastatic Non-small-cell Lung Cancer [NCT04094909]Phase 2186 participants (Anticipated)Interventional2020-02-06Not yet recruiting
EGFR-TKI With/Without Chemotherapy in NSCLC Patients With Both EGFR Mutation and BIM Deletion Polymorphism [NCT03002844]Phase 250 participants (Anticipated)Interventional2016-12-31Not yet recruiting
Randomized Phase II Trial of Pemetrexed vs. Pemetrexed/Bevacizumab vs. Pemetrexed/Carboplatin/Bevacizumab in Patients With Stage IIIB/IV Non-Small-Cell Lung Cancer and ECOG Performance Status 2 [NCT00892710]Phase 2172 participants (Actual)Interventional2009-06-30Completed
Phase III Randomized Trial Comparing Overall Survival After Photon Versus Proton Chemoradiotherapy for Inoperable Stage II-IIIB NSCLC [NCT01993810]Phase 3330 participants (Actual)Interventional2014-02-03Active, not recruiting
A Phase II Study of a Combination of MTA (LY231514) and Gemcitabine in Patients With Metastatic Breast Cancer [NCT00006007]Phase 259 participants (Actual)Interventional2000-12-31Completed
Safety Confirmation Study of LY231514 Plus Cisplatin in Patients With Malignant Pleural Mesothelioma [NCT00386815]Phase 220 participants Interventional2006-10-31Completed
An Open-label, Multi-center, Phase II Umbrella Study to Assess Efficacy of Targeted Therapy or Immunotherapy Directed by Next Generation Sequencing (NGS) in Chinese Patients With Advanced NSCLC (TRUMP) [NCT03574402]Phase 2400 participants (Anticipated)Interventional2018-07-09Recruiting
A Pharmacokinetic Study of Pemetrexed in the Cerebrospinal Fluid of Patients With Leptomeningeal Metastases [NCT00424242]Early Phase 115 participants (Anticipated)Interventional2007-01-31Completed
MARVEL: Marker Validation of Erlotinib in Lung Cancer- A Phase III Biomarker Validation Study of Second-Line Therapy in Patients With Advanced Non-small Cell Lung Cancer (NSCLC) Randomized to Pemetrexed Versus Erlotinib [NCT00738881]Phase 323 participants (Actual)Interventional2008-10-31Terminated(stopped due to Slow accrual)
Phase I Study Of SU011248 In Combination With Pemetrexed, Pemetrexed/Cisplatin And Pemetrexed/Carboplatin In Patients With Advanced Solid Malignancies [NCT00528619]Phase 196 participants (Actual)Interventional2006-11-30Completed
A Phase 1 Dose-Escalation and Phase 2 Randomized, Open-Label Study of Nivolumab and Veliparib in Combination With Platinum Doublet Chemotherapy in Subjects With Metastatic or Advanced Non-Small Cell Lung Cancer (NSCLC) [NCT02944396]Phase 125 participants (Actual)Interventional2016-12-23Completed
Performance Status and Influencing Factors During Second-Line Treatment With Pemetrexed in Patients With Stage III/IVNon Small Cell Lung Cancer [NCT00540241]542 participants (Actual)Observational2007-09-30Completed
Phase II Study of Single-Agent Alimta in the Treatment of Patients With Advanced and Metastatic Hepatoma [NCT00191412]Phase 241 participants Interventional2005-01-31Completed
Protocol H6Q-MC-S034(a) Randomized, Double-Blind, Phase 2 Study of Pemetrexed + Carboplatin + Bevacizumab + Enzastaurin Versus Pemetrexed + Carboplatin + Bevacizumab + Placebo in Chemonaive Patients With Stage IIIB or IV Non-Small Cell Lung Cancer [NCT00533429]Phase 240 participants (Actual)Interventional2007-10-31Completed
NGR014: Randomized Phase II Study of NGR-hTNF in Combination With Standard Chemotherapy Versus Standard Chemotherapy Alone in Previously Untreated Patients With Advanced Non-small Cell Lung Cancer (NSCLC) [NCT00994097]Phase 2121 participants (Actual)Interventional2009-07-31Completed
Phase I Clinical Study of Autologous CIK Cell Immunotherapy Combination With PD-1 Inhibitor and Chemotherapy in the First-line Treatment of IIIB/IIIC/IV Non-Small Cell Lung Cancer [NCT03987867]Phase 130 participants (Anticipated)Interventional2019-06-01Recruiting
A Randomized Phase 3 Study of Pemetrexed in Combination With Cisplatin Versus Cisplatin Monotherapy in Patients With Recurrent or Metastatic Head and Neck Cancer [NCT00415194]Phase 3795 participants (Actual)Interventional2006-12-31Completed
A Phase II Evaluation of Pemetrexed (Alimta, LY231514l, IND # 40061) in the Treatment of Recurrent or Persistent Platinum Resistant Ovarian or Primary Peritoneal Carcinoma [NCT00461786]Phase 251 participants (Actual)Interventional2004-09-30Completed
Phase II Trial of Carboplatin and Pemetrexed Plus Bevacizumab in Patients With Advanced Non-Squamous Non-Small Cell Lung Cancer [NCT00234052]Phase 251 participants (Actual)Interventional2005-07-28Completed
A Randomized Phase II Study of ALIMTA® (Pemetrexed) and GEMZAR® (Gemcitabine) Every 14 Days Versus Pemetrexed and Gemcitabine Every 21 Days in Advanced Non-Small Cell Lung Cancer [NCT00407550]Phase 219 participants (Actual)Interventional2006-11-30Completed
Phase 1/2 Study of Biweekly ALIMTA Plus Cisplatin in Patients With Locally, Advanced, Non-Resectable or Metastatic Urothelial Cancer [NCT00374868]Phase 1/Phase 259 participants (Actual)Interventional2006-08-31Completed
A Phase II Trial of Preoperative Radiation and Chemotherapy (Pemetrexed and Carboplatin) for Locally Advanced Esophageal Cancer [NCT00268437]Phase 227 participants (Actual)Interventional2006-04-30Terminated(stopped due to Trial closed early because, during an interim analysis, the primary endpoint fell short.)
Open-Label Single-Arm Phase IV Study of Pemetrexed in Taiwanese Patients With Advanced Non-Small Cell Lung Cancer Who Have Had Prior Chemotherapy [NCT00380718]Phase 433 participants (Actual)Interventional2006-11-30Completed
A Phase II Evaluation of Pemetrexed in the Treatment of Recurrent or Persistent Endometrial Carcinoma [NCT00377520]Phase 227 participants (Actual)Interventional2006-09-30Completed
Phase II Trial of Pemetrexed in Patients With Selected Stage IIIB and IV Bronchioloalveolar Carcinoma (BAC) [NCT00265785]Phase 227 participants (Actual)Interventional2006-07-31Terminated(stopped due to Closed due to poor accrual)
INST 0601C: A Non-Randomized Phase II Protocol of Erlotinib for Patients With Newly Diagnosed, Advanced Non-Small Cell Carcinoma of the Lung [NCT00391586]Phase 245 participants (Actual)Interventional2006-07-31Terminated(stopped due to PI left institution.)
Phase 3 Study of Pemetrexed Versus Docetaxel in Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer Who Have Had Prior Chemotherapy [NCT00391274]Phase 3211 participants (Actual)Interventional2006-10-31Completed
Phase II Study of Lorlatinib in Combination With Chemotherapy in Participants With Metastatic Anaplastic Lymphoma Kinase Positive (ALK+) Non-small Cell Lung Cancer (NSCLC) Who Progressed on Single-agent Lorlatinib [NCT05948462]Phase 20 participants (Actual)Interventional2023-11-30Withdrawn(stopped due to Study withdrawn by pharmaceutical funding partner)
A Phase Ib/II, Open-Label, Multicenter Study Evaluating the Safety, Activity, and Pharmacokinetics of Divarasib in Combination With Other Anti-Cancer Therapies in Patients With Previously Untreated Advanced Or Metastatic Non-Small Cell Lung Cancer With a [NCT05789082]Phase 1/Phase 296 participants (Anticipated)Interventional2023-06-20Recruiting
A Randomized, Double-Blind, Phase 3 Study of MK-7684A in Combination With Chemotherapy Versus Pembrolizumab Plus Chemotherapy as First Line Treatment for Participants With Metastatic Non-Small Cell Lung Cancer [NCT05226598]Phase 3700 participants (Anticipated)Interventional2022-03-24Active, not recruiting
A Phase III, Open-Label, Randomized Study to Evaluate the Efficacy and Safety of Adjuvant Alectinib Versus Adjuvant Platinum-Based Chemotherapy in Patients With Completely Resected Stage IB (Tumors Equal to or Larger Than 4cm) to Stage IIIA Anaplastic Lym [NCT03456076]Phase 3257 participants (Actual)Interventional2018-08-16Active, not recruiting
Phase II Trial of Pemetrexed Plus Gemcitabine in Patients With Advanced Non-Clear Cell Renal Cell Cancer [NCT00491075]Phase 216 participants (Actual)Interventional2005-12-31Terminated(stopped due to Closed early for poor accrual.)
A Phase II Trial Pemetrexed Carboplatin as First Line Chemotherapy for Advanced Non-Small Cell Lung Cancer (NSCLC) in Elderly Patients [NCT00350792]Phase 262 participants (Actual)Interventional2006-08-31Completed
A Randomized, Open-label Phase II Study of Pemetrexed (Alimta) Plus Carboplatin With or Without Enzastaurin Hydrochloride, or Docetaxel Plus Carboplatin as First Line Treatment in Patients With Advanced Stage Non-small Cell Lung Cancer (NSCLC) [NCT00308750]Phase 2218 participants (Actual)Interventional2006-03-31Completed
A Phase 1 and 2 Clinical Trial of ALIMTA® (Pemetrexed) in Combination With Carboplatin in Patients With Recurrent Ovarian or Primary Peritoneal Cancer [NCT00489359]Phase 1/Phase 286 participants (Actual)Interventional2005-07-31Completed
A Phase Ib Clinical Trial to Evaluate the Safety and Efficacy of TQB2450 Injection Combined With AL2846 Capsules in Patients With Advanced Solid Tumors [NCT06116240]Phase 1135 participants (Anticipated)Interventional2022-09-02Active, not recruiting
A Phase II Study of Treatment of Brain Metastases From Non-Small Cell Lung Cancer With Concurrent Whole Brain Radiation Therapy and Pemetrexed [NCT00280748]Phase 210 participants (Actual)Interventional2005-05-31Terminated(stopped due to Slow accrual)
A Phase II Study to Evaluate Activity and Toxicity of Gemcitabine in Combination With Pemetrexed Long Term Infusion in the Treatment of Pretreated Metastatic Colorectal Cancer Patients [NCT01909830]Phase 218 participants (Actual)Interventional2012-07-31Completed
A Phase II/III, Randomized, Double-Blind, Placebo-Controlled Study of Tiragolumab in Combination With Atezolizumab Plus Pemetrexed and Carboplatin/Cisplatin Versus Pembrolizumab Plus Pemetrexed and Carboplatin/Cisplatin in Patients With Previously Untreat [NCT04619797]Phase 2/Phase 3542 participants (Actual)Interventional2020-12-14Active, not recruiting
A Phase 3 Randomized, Placebo-controlled Study to Evaluate the Safety and Efficacy of Pemetrexed + Platinum Chemotherapy + Pembrolizumab (MK-3475) With or Without Lenvatinib (E7080/MK-7902) as First-line Intervention in Participants With Metastatic Nonsqu [NCT03829319]Phase 3761 participants (Actual)Interventional2019-03-25Active, not recruiting
Phase II Trial of Pemetrexed Disodium and Carboplatin in Previously Untreated Extensive Stage Small Cell Lung Cancer [NCT00227565]Phase 250 participants (Actual)Interventional2006-02-28Completed
A Phase 2, Open-label, Randomized, Parallel Group, Controlled Study of Pemetrexed Maintenance With or Without ADXS11-001 Immunotherapy in Patients With Human Papillomavirus-Positive, Non-Squamous, Non-Small Cell Lung Carcinoma Following First-Line Inducti [NCT02531854]Phase 20 participants (Actual)Interventional2018-12-31Withdrawn(stopped due to Study was withdrawn per sponsor decision)
A Phase IB Study of Pembrolizumab in Combination With Pemetrexed and Oxaliplatin in Patients With Chemo-Refractory Metastatic Colorectal Cancer [NCT03626922]Phase 133 participants (Anticipated)Interventional2019-05-15Active, not recruiting
Multicenter Phase II Study of Pemetrexed/Cisplatin With or Without Bevacizumab in Patients With Brain Metastases From Non Squamous Non-small Cell Lung Cancer Harboring EGFR Wild Type [NCT01951482]Phase 2108 participants (Anticipated)Interventional2013-06-30Recruiting
A Study of Almonertinib With Chemotherapy as 1st Line Treatment in Patients With Mutated Epidermal Growth Factor Receptor Non-Small Cell Lung Cancer [NCT04646824]Phase 210 participants (Anticipated)Interventional2020-11-19Active, not recruiting
A Phase II, Open-Label, Multicenter, Prospective Clinical Study to Investigate the Efficacy and Safety of Tislelizumab Combined With Pemetrexed/ Carboplatin in Patients With Brain Metastases of Non-squamous Non-small Cell Lung Cancer [NCT04507217]Phase 236 participants (Actual)Interventional2020-09-15Completed
Pemetrexed for Previously Treated Patients With Metastatic Gastric Cancer: a Prospective Phase II Study [NCT01953419]Phase 234 participants (Actual)Interventional2010-09-30Completed
Phase I Study of Thoracic Radiotherapy and Concurrent Chemotherapy With Soy Isoflavones in Stage III NSCLC (Non-Small Cell Lung Cancer) Patients [NCT01958372]Phase 111 participants (Actual)Interventional2014-08-31Completed
A Phase IIIb, Randomized, Multicenter, Open-label Study to Assess the Efficacy of Durvalumab Plus Tremelimumab Versus Pembrolizumab in Combination With Platinum-Based Chemotherapy for First-Line Treatment in Metastatic Non-Small Cell Lung Cancer Patients [NCT06008093]Phase 3280 participants (Anticipated)Interventional2023-12-29Not yet recruiting
A Randomized, Double-Blind Phase 2/3 Study of Fianlimab (Anti-LAG-3 Antibody), Cemiplimab (Anti-PD-1 Antibody), and Chemotherapy Versus Cemiplimab and Chemotherapy in First-Line Treatment of Patients With Advanced Non-Small Cell Lung Cancer (NSCLC) Irresp [NCT05800015]Phase 2/Phase 3950 participants (Anticipated)Interventional2023-08-08Recruiting
A Phase 2 Platform Study Evaluating the Safety and Efficacy of Novel Treatment Combinations in Patients With Lung Cancer (VELOCITY-Lung) [NCT05633667]Phase 2397 participants (Anticipated)Interventional2023-03-16Recruiting
A Phase 2 Randomized Study of the BER Inhibitor TRC102 in Combination With Standard Pemetrexed-Platinum-Radiation in Stage III Non-Squamous Non-Small Cell Lung Cancer [NCT05198830]Phase 278 participants (Anticipated)Interventional2022-12-15Recruiting
A Phase I Trial Targeting Mitochondrial Metabolism With Papaverine in Combination With Chemoradiation for Stage II-III Non-Small Cell Lung Cancer [NCT05136846]Phase 128 participants (Anticipated)Interventional2021-12-06Recruiting
A Phase II, Randomized, Open-label Trial of Nivolumab in Combination With Ipilimumab Versus Pemetrexed With Cisplatin or Carboplatin as First-line Therapy for Unresectable Pleural Mesothelioma in Chinese Participants [NCT05136677]Phase 2100 participants (Anticipated)Interventional2022-01-25Recruiting
A Phase Ib Clinical Study to Evaluate the Safety and Efficacy of Pembrolizumab (MK-3475) in Combination With Cisplatin and Pemetrexed in Treatment-naive Participants With Advanced Malignant Pleural Mesothelioma (KEYNOTE-A17). [NCT04153565]Phase 119 participants (Actual)Interventional2019-12-09Completed
Comparison of Postoperative Adjuvant Chemotherapy With/Without Rh-endostatin: a Randomized, PhaseⅢ and Open Clinical Study of Non-small Cell Lung Cancer in PhaseⅠB [NCT02001168]Phase 3392 participants (Anticipated)Interventional2013-10-31Active, not recruiting
Maintenance Pemetrexed Therapy After Induction Chemotherapy Versus Pemetrexed at Progression in Advanced Non-Small-Cell Lung Cancer: A Randomized Phase III Study [NCT02004184]Phase 3230 participants (Actual)Interventional2013-12-31Terminated(stopped due to Poor enrollment due to the introduction of immunotherapy)
A Multicentre Phase II, Open-label, Non-randomized Study Evaluating Platinum-Pemetrexed-Atezolizumab (+/- Bevacizumab) for Patients With Stage IIIB/IV Non-squamous Non-small Cell Lung Cancer With EGFR Mutations, ALK Rearrangement or ROS1 Fusion Progressin [NCT04042558]Phase 2149 participants (Anticipated)Interventional2019-09-26Recruiting
Phase 2 Study of Pemetrexed and Cisplatin as Induction, Followed by Pemetrexed and Cisplatin With Concurrent Thoracic Radiotherapy, in Patients With Unresectable, Locally Advanced, Stage III, Nonsquamous Non-Small Cell Lung Cancer [NCT01000480]Phase 290 participants (Actual)Interventional2009-10-31Completed
EGFR-TKIs Combine Chemotherapy as First-line Therapy for Patients With Advanced EGFR Mutation-positive NSCLC [NCT02886195]Phase 3120 participants (Anticipated)Interventional2016-07-31Enrolling by invitation
A Phase II Randomized Trial Evaluating the Use of Proton Pump Inhibitors (PPIs) in Conjunction With Chemotherapy, in Patients With Recurrent Unresectable or Metastatic Cancers of the Head and Neck [NCT02013453]Phase 20 participants (Actual)Interventional2013-12-31Withdrawn(stopped due to Lack of funding)
A Phase 1/Randomized Phase 2 Study to Evaluate LY2603618 in Combination With Pemetrexed and Cisplatin in Patients With Stage IV Non-small Cell Lung Cancer [NCT01139775]Phase 1/Phase 276 participants (Actual)Interventional2011-02-28Completed
A Randomized, Double-Blind, Placebo-Controlled, Study of the Safety and Efficacy of Farletuzumab in Combination With a Platinum-Containing Doublet in Chemotherapy-Naive Subjects With Stage IV Adenocarcinoma of the Lung (FLAIR) [NCT01218516]Phase 2130 participants (Actual)Interventional2011-06-27Completed
BATTLE-FL: A Biomarker-Integrated Study in Patients With Advanced Non-Small Cell Lung Cancer Treated in the Front-Line (FL) Setting [NCT01263782]Phase 264 participants (Actual)Interventional2011-05-17Completed
Phase 3, Randomized, Open-label Study Of The Efficacy And Safety Of Pf-02341066 Versus Standard Of Care Chemotherapy (Pemetrexed Or Docetaxel) In Patients With Advanced Non-small Cell Lung Cancer (Nsclc) Harboring A Translocation Or Inversion Event Involv [NCT00932893]Phase 3347 participants (Actual)Interventional2009-09-30Completed
Phase I/II Study of Two Different Schedules of Pemetrexed (ALIMTA) and Erlotinib (TARCEVA) in Advanced Solid Tumors, With Emphasis on Non-Small Cell Lung Cancer (NSCLC) [NCT00387322]Phase 1/Phase 242 participants (Actual)Interventional2005-03-31Completed
Efficacy and Safety of Tislelizumab With Platinum Doublet Chemotherapy as Neoadjuvant Therapy for Participants With Initially Unresectable Stage III Non-small Cell Lung Cancer: A Single-arm, Phase II Trial [NCT05611879]Phase 230 participants (Anticipated)Interventional2023-03-12Recruiting
A Phase 1 Clinical Study to Evaluate the Bioavailability of Pembrolizumab Via Subcutaneous Injection of MK-3475A, a Formulation of Pembrolizumab With MK-5180, in Participants With Advanced Solid Tumors [NCT05017012]Phase 172 participants (Anticipated)Interventional2021-09-21Recruiting
Integration of Immunotherapy Into Adjuvant Therapy for Resected NSCLC: ALCHEMIST Chemo-IO (ACCIO) [NCT04267848]Phase 31,210 participants (Anticipated)Interventional2020-06-16Recruiting
EA5163/S1709 INSIGNA : A Randomized, Phase III Study of Firstline Immunotherapy Alone or in Combination With Chemotherapy in Induction/Maintenance or Postprogression in Advanced Nonsquamous Non-Small Cell Lung Cancer (NSCLC) With Immunobiomarker SIGNature [NCT03793179]Phase 3600 participants (Anticipated)Interventional2019-04-05Recruiting
Adjuvant Lung Cancer Enrichment Marker Identification and Sequencing Trial (ALCHEMIST) [NCT02194738]8,300 participants (Anticipated)Interventional2014-09-26Recruiting
Phase II Toxicity Study of Pleurectomy/Decortication, (Neo) Adjuvant Chemotherapy and Intensity Modulated Radiation Therapy to the Pleura in Patients With Locally Advanced Malignant Pleural Mesothelioma [NCT00715611]Phase 265 participants (Actual)Interventional2008-10-11Active, not recruiting
A Phase II, Randomized, Double-Blind, Placebo-Controlled Study Evaluating the Efficacy and Safety of Adjuvant Platinum-Doublet Chemotherapy, With or Without Atezolizumab, in Patients Who Are ctDNA Positive After Complete Surgical Resection of Stage IB to [NCT04611776]Phase 20 participants (Actual)Interventional2021-07-01Withdrawn(stopped due to The positive results from IMpower010 demonstrated benefit by adding atezolizumab as adjuvant therapy in early stage NSCLC. These results raised ethical concerns of enrolling pts to best supportive care over checkpoint inhibition in this setting.)
A Two Arm Phase I Trial of Sorafenib in Combination With Cisplatin/Etoposide or Carboplatin/Pemetrexed in Patients With Solid Tumors [NCT00573690]Phase 131 participants (Actual)Interventional2007-09-30Completed
Almonertinib Plus Pemetrexed and Carboplatin Versus Almonertinib Alone in Advanced NSCLC With EGFR T790M After First- or Second-generation TKIs Therapy: a Randomized, Controlled, Open-label, Phase 2 Study [NCT04592666]Phase 2226 participants (Anticipated)Interventional2020-10-09Not yet recruiting
Dynamic Positron Emission Tomography/Computed Tomography Evaluated the Response of Neoadjuvant Anti-programmed Cell Death Protein 1 Combination With Chemotherapy for Stage Ⅱa-Ⅲb Non-small Cell Lung Cancer [NCT04586465]Phase 223 participants (Anticipated)Interventional2020-10-10Recruiting
A Phase II Evaluation of Pemetrexed (Alimta) in the Treatment of Recurrent Carcinoma of the Cervix [NCT00190983]Phase 229 participants (Actual)Interventional2005-02-28Completed
A Phase 1/2 Dose-Escalating Study of ALIMTA and Cyclophosphamide Administered Every 21 Days in Patients With Locally Advanced or Metastatic Breast Cancer [NCT00190671]Phase 1/Phase 2103 participants (Actual)Interventional2005-06-30Completed
Open Multicenter Phase II Study in Second-Line Metastatic Colorectal Cancer Patients: Combination of ALIMTA and Irinotecan Administered Every Two-Weeks [NCT00191984]Phase 246 participants (Actual)Interventional2004-06-30Completed
Phase 2 Study of Pemetrexed in Combination With Carboplatin or Cisplatin and Cetuximab in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck [NCT01087970]Phase 269 participants (Actual)Interventional2010-08-31Completed
Randomized Phase II Study of Maintenance Pemetrexed Versus Observation for Patients With Malignant Pleural Mesothelioma Without Progression After First-Line Chemotherapy [NCT01085630]Phase 272 participants (Actual)Interventional2010-04-30Completed
Phase I/II Study of LY231514 Plus Cisplatin in Patients With Malignant Pleural Mesothelioma [NCT00251550]Phase 1/Phase 212 participants Interventional2005-10-31Completed
Efficacy of Targeted Therapy Combined Chemotherapy in Advanced EGFR Positive NSCLC Patients With Concurrent Driver Gene Mutations [NCT04552613]110 participants (Anticipated)Interventional2020-09-30Recruiting
A Phase I Study of Concurrent Pemetrexed/Cisplatin With Pleural Intensity Modulated Radiation Therapy for Patients With Unresectable Malignant Pleural Mesothelioma [NCT02639767]Phase 10 participants (Actual)Interventional2015-12-31Withdrawn(stopped due to Lack of accrual)
A Pilot Study of Camrelizumab With Chemotherapy in Adults With Medically Inoperable Early Stage Non-Small Cell Lung Cancer (NSCLC) [NCT04530227]Phase 230 participants (Anticipated)Interventional2020-09-25Recruiting
A Phase Ib, Open-label, Single-center Study to Assess the Safety of Cancer-immunotherapy Induction With Tremelimumab and Durvalumab Prior to Chemoradiotherapy and/or Resection in the Treatment of Locally Advanced NSCLC. [NCT04287894]Phase 134 participants (Anticipated)Interventional2018-12-28Recruiting
Phase III Prospective Randomized Trial of Primary Lung Tumor Stereotactic Body Radiation Therapy Followed by Concurrent Mediastinal Chemoradiation for Locally Advanced Non-Small Cell Lung Cancer [NCT05624996]Phase 3474 participants (Anticipated)Interventional2023-05-10Recruiting
A Randomized, Double-blind, Placebo-controlled, Phase 2 Study Evaluating Efficacy and Safety of Inupadenant in Combination With Carboplatin and Pemetrexed in Adults With Nonsquamous Non-small Cell Lung Cancer Who Have Progressed on Immunotherapy [NCT05403385]Phase 2192 participants (Anticipated)Interventional2022-08-26Recruiting
A Phase II Study of Neoadjuvant Sotorasib in Combination With Cisplatin or Carboplatin and Pemetrexed for Surgically Resectable Stage IIA-IIIB Non-Squamous Non-Small Cell Lung Cancer With a KRAS p.G12C Mutation [NCT05118854]Phase 227 participants (Anticipated)Interventional2022-03-30Recruiting
A Phase 2 Randomized Study of Osimertinib Versus Osimertinib Plus Chemotherapy for Patients With Metastatic EGFR-Mutant Lung Cancers That Have Detectable EGFR-Mutant cfDNA in Plasma After Initiation of Osimertinib [NCT04410796]Phase 2571 participants (Anticipated)Interventional2020-05-28Recruiting
A Phase III, Randomized, Placebo-controlled, Double-blind, Multi-center, International Study of Durvalumab Given Concurrently With Platinum-based Chemoradiation Therapy in Patients With Locally Advanced, Unresectable NSCLC (Stage III) (PACIFIC2) [NCT03519971]Phase 3328 participants (Actual)Interventional2018-03-29Active, not recruiting
AdvanTIG-306: A Randomized, Double-blind, Placebo-controlled, Phase III Study Evaluating the Efficacy and Safety of Ociperlimab (WCD118/BGB-A1217) Combined With Tislelizumab (VDT482/BGB-A317) Plus Platinum-based Doublet Chemotherapy Versus Placebo Combine [NCT05791097]Phase 30 participants (Actual)Interventional2023-07-28Withdrawn(stopped due to Business decision, not driven by safety concerns; no new safety signals have been observed in the ociperlimab program.)
A Phase III, Randomised, Open-label, Multicentre, Global Study of Datopotamab Deruxtecan (Dato-DXd) in Combination With Durvalumab and Carboplatin Versus Pembrolizumab in Combination With Platinum-based Chemotherapy for the First-line Treatment of Patient [NCT05687266]Phase 31,000 participants (Anticipated)Interventional2022-12-29Recruiting
A Pilot Study Evaluating Pemetrexed in ECOG Performance Status 3 Patients With Stage IV Non-squamous Non-small Cell Lung Cancer [NCT02426658]Phase 216 participants (Actual)Interventional2015-05-31Completed
"A Phase 1b/2 Study of Viagenpumatucel-L (HS-110) in Combination With Multiple Treatment Regimens in Patients With Non-Small Cell Lung Cancer (The DURGA Trial)" [NCT02439450]Phase 1/Phase 2121 participants (Actual)Interventional2015-04-15Completed
A Phase II Study of Neoadjuvant Pemetrexed, Carboplatin and Bevacizumab in Unresectable, Locally Advanced Lung Adenocarcinoma [NCT01588704]Phase 242 participants (Actual)Interventional2012-04-30Completed
A Phase III, Two-Arm, Parallel, Randomized, Multi-Center, Open-Label, Global Study to Determine the Efficacy of Volrustomig (MEDI5752) Plus Chemotherapy Versus Pembrolizumab Plus Chemotherapy for First-Line Treatment of Patients With Metastatic Non-Small [NCT05984277]Phase 3900 participants (Anticipated)Interventional2023-10-24Recruiting
Phase II Study of Induction Checkpoint Blockade for Untreated Stage I-IIIA Non-Small Cell Lung Cancers Amenable for Surgical Resection [NCT03158129]Phase 2101 participants (Actual)Interventional2017-06-09Active, not recruiting
Phase IB Study to Evaluate the Safety of Selinexor (KPT-330) in Combination With Multiple Standard Chemotherapy or Immunotherapy Agents in Patients With Advanced Malignancies [NCT02419495]Phase 1221 participants (Actual)Interventional2015-06-26Active, not recruiting
A Two Arm Phase I Dose Escalation Trial of Vinflunine With Erlotinib or Pemetrexed in Refractory Solid Tumors [NCT00320073]Phase 141 participants (Actual)Interventional2006-08-31Completed
A Phase 1 Trial of MK-7684 as Monotherapy and in Combination With Pembrolizumab in Subjects With Advanced Solid Tumors [NCT02964013]Phase 1492 participants (Anticipated)Interventional2016-12-13Active, not recruiting
A Multicenter Randomized Phase II Study of the Combination of Irinotecan/Cisplatin Versus Pemetrexed/Cisplatin as Second-line Treatment of Patients With Stage IIIB/IV Non-small Cell Lung Cancer (NSCLC) [NCT00614965]Phase 2124 participants (Anticipated)Interventional2006-11-30Completed
A Randomized Phase II Study to Evaluate Efficacy and Safety of DCVAC/LuCa Added to Chemotherapy With Carboplatin and Pemetrexed vs Chemotherapy Alone in Patients With Stage IV Non-small Cell Lung Cancer [NCT02669719]Phase 270 participants (Anticipated)Interventional2016-01-31Recruiting
BIBW 2992 Phase I Combination With Pemetrexed in Advanced Solid Tumours [NCT01169675]Phase 153 participants (Actual)Interventional2010-07-31Completed
A Phase I/II Study of Continuous, Concomitant Oral Treatment With BIBF 1120 and Pemetrexed - a Phase I, Open-label, Dose-escalation Study & a Phase II, 2 Arm, Randomized, Double-blind, Placebo-controlled Study in Japanese Patients With Stage IIIB/IV or Re [NCT00979576]Phase 119 participants (Actual)Interventional2009-10-31Terminated
A Single-Arm, Phase 2 Trial of Pemetrexed, Cisplatin,and Bevacizumab as Induction, Followed by Pemetrexed and Bevacizumab as Maintenance, in First-Line Treatment of Nonsquamous Advanced NSCLC [NCT01004250]Phase 2109 participants (Actual)Interventional2009-10-31Completed
An Open-label, Multicenter, Randomized, Phase 2 Study of a Recombinant Human Anti-VEGFR-2 Monoclonal Antibody, IMC-1121B in Combination With Platinum-based Chemotherapy Versus Platinum-based Chemotherapy Alone as First-line Treatment of Patients With Recu [NCT01160744]Phase 2280 participants (Actual)Interventional2010-09-30Completed
Open-label Study of Bevacizumab (Avastin®) in Combination With Pemetrexed or Pemetrexed and Carboplatin as First-line Treatment of Patients With Advanced or Recurrent Non-squamous Non-small Cell Lung Cancer [NCT00976456]Phase 3271 participants (Actual)Interventional2009-09-30Completed
A Randomized Phase II Study of S1 Plus Carboplatin Followed by Maintenance S1versus Pemetrexed Plus Carboplatin Followed by Maintenance Pemetrexed in Patients With EGFR Wild Type Stage IIIB or IV Nonsquamous Non-Small-Cell Lung Cancer [NCT02631460]Phase 2470 participants (Anticipated)Interventional2015-12-31Recruiting
A Phase 1b Study of LY573636-sodium in Combination With Alimta (Pemetrexed) in Patients With Solid Tumors [NCT01215916]Phase 139 participants (Actual)Interventional2008-02-29Completed
Phase 2 Pharmacological Study of Pemetrexed Administered With Cisplatin and a Vitamin Supplement in Patients With Nonresectable Pleural Mesothelioma [NCT00541073]Phase 260 participants (Anticipated)Interventional2007-06-30Completed
Phase II Randomized Controlled Trial of Neoadjuvant Pembrolizumab or Pembrolizumab With Histology-Specific Chemotherapy for Operable Stage IA3 to IIA Non-Small Cell Lung Cancer (NSCLC) [NCT04638582]Phase 244 participants (Anticipated)Interventional2022-08-28Recruiting
Multicenter Phase III Study of Gefitinib Mono-therapy or Gefitinib Combined With Chemotherapy in Patients With Brain Metastases From Non-small Cell Lung Cancer Harboring EGFR Mutation [NCT01951469]Phase 3160 participants (Anticipated)Interventional2016-01-31Recruiting
Phase I Study of PTK/ZK in Combination With Pemetrexed Disodium (ALIMTA) [NCT00390000]Phase 129 participants (Actual)Interventional2007-01-25Completed
A Phase II Evaluation Of Pemetrexed (ALIMTA, LY231514, IND #40061) In The Treatment Of Recurrent Or Persistent Endometrial Carcinoma [NCT00087100]Phase 251 participants (Anticipated)Interventional2006-05-31Completed
Phase II Study of the Combination of Bevacizumab Plus Pemetrexed and Carboplatin as First-line Therapy in Patients With Malignant Pleural Mesothelioma [NCT00407459]Phase 277 participants (Actual)Interventional2007-09-30Completed
A Phase I Study of Pemetrexed (LY231514, Alimta) in Children and Adolescents With Recurrent Solid Tumors [NCT00070473]Phase 133 participants (Actual)Interventional2003-10-31Completed
A Phase II Trial of Pemetrexed (ALIMTA®, LY231514, IND #40061) as Salvage Therapy for Failed Low Risk Gestational Trophoblastic Tumor [NCT00096187]Phase 255 participants (Anticipated)Interventional2005-07-31Terminated
Dacomitinib + Pemetrexed for Patients With Advanced Non-squamous Non-small Cell Lung Cancer (NSCLC): a Phase I Trial to Identify a Dose of Dacomitinib in Combination With Pemetrexed That is Safe and Tolerated as Determined by the Incidence of Dose Limitin [NCT01918761]Phase 15 participants (Actual)Interventional2013-07-30Terminated(stopped due to poor accrual)
Single Agent Alimta in Poor Performance Status in Patients With Non-Small Cell Lung Cancer (NSCLC) [NCT00508144]Phase 264 participants (Actual)Interventional2005-09-30Completed
Phase 1/2 Study of Pemetrexed (Alimta) Plus Carboplatin, or Pemetrexed Plus Cisplatin With Concurrent Radiation Therapy Followed by Every-21-Day Pemetrexed Consolidation in Patients With Favorable-Prognosis Inoperable Stage IIIA/B Non-Small-Cell Lung Canc [NCT00482014]Phase 1/Phase 2120 participants (Actual)Interventional2007-05-31Completed
Phase II Trial of Preoperative Pemetrexed and Carboplatin in Patients With Select Stage IB, II, and III Non-Squamous Non-Small-Cell Lung Cancer [NCT00906282]Phase 246 participants (Actual)Interventional2009-06-30Completed
A Randomized Phase 3 Study Comparing Pemetrexed Plus Cisplatin With Gemcitabine Plus Cisplatin as First-Line Treatment in Patients With Advanced Non-squamous Non-Small Cell Lung Cancer. [NCT01005680]Phase 3256 participants (Actual)Interventional2009-11-30Completed
Randomized Phase II Study of Pemetrexed and Cisplatin as Either Induction or Adjuvant Chemotherapy in Stage IB-II Non-Small Cell Lung Cancer (NSCLC) [NCT00389688]Phase 213 participants (Actual)Interventional2006-08-31Terminated(stopped due to low accrual)
Phase I Open-Label, Dose Escalation Study To Determine The Maximum Tolerated Dose And To Evaluate The Safety Profile Of Lenalidomide (Revlimid®, CC-5013) With Pemetrexed In Subjects With Advanced Non-Small Cell Lung Cancer [NCT00179699]Phase 140 participants Interventional2005-09-30Terminated
A Phase 1, Open-Label, Multicenter Study to Assess Safety, Tolerability, PK, and Efficacy of MK-1084 as Monotherapy and in Combination With Pembrolizumab in Subjects With KRAS G12C Mutant Advanced Solid Tumors [NCT05067283]Phase 1450 participants (Anticipated)Interventional2021-12-17Recruiting
A Phase 3, Randomized, Double-Blind, Multicenter Study of Talabostat and Pemetrexed vs. Pemetrexed and Placebo in Patients With Advanced (Stage IIIB/IV) Non-Small Cell Lung Cancer (NSCLC) After Failure of Platinum-Based Chemotherapy [NCT00290017]Phase 3400 participants (Anticipated)Interventional2006-02-28Terminated(stopped due to FDA Hold May 2007)
A Randomized, Double-blind, Placebo-controlled, Phase III Study Evaluating the Efficacy and Safety of Pembrolizumab Plus Platinum-based Doublet Chemotherapy With or Without Canakinumab as First Line Therapy for Locally Advanced or Metastatic Non-squamous [NCT03631199]Phase 3673 participants (Actual)Interventional2018-12-21Active, not recruiting
Phase 1/2 Study of Oral MKC-1 Administered Twice Daily for 14 Consecutive Days Every 3 Weeks in Combination With Pemetrexed [NCT00408226]Phase 1/Phase 227 participants (Actual)Interventional2006-10-31Completed
Phase III Study Comparing Osimertinib Monotherapy to Combination Therapy With Osimertinib,Carboplatin and Pemetrexed for Untreated Patients With Advanced Non-squamous Non-Small Cell Lung Cancer With Concurrent EGFR and TP53 Mutations [NCT04695925]Phase 3291 participants (Anticipated)Interventional2021-01-04Not yet recruiting
Phase II Study of Pemetrexed Plus Cisplatin in the Treatment of Patients With Extensive Small Cell Lung Cancer [NCT00475657]Phase 25 participants (Actual)Interventional2007-10-31Terminated(stopped due to Terminated due to lack of efficacy)
Phase I/II Clinical Trial of Bortezomib (Velcade) + Pemetrexed (Alimta) in Previously Treated Patients With Advanced Non-Small Cell Lung Cancer [NCT00516100]Phase 1/Phase 262 participants (Anticipated)Interventional2006-01-31Active, not recruiting
Advanced Metastatic Non-small Cell Lung Cancer Patients Aged or PS Score 2 Points for First Line Application Pemetrexed/Carboplatin Chemotherapy Regimens Sequential Pemetrexed Single Drug Maintenance Treatment of Clinical Research and Related Predictive B [NCT01860508]94 participants (Anticipated)Interventional2013-02-28Recruiting
ALIMTA Plus Gemcitabine as Front-line Chemotherapy for Patients With Locally Advanced or Metastatic Non-Small-Cell Lung Cancer: A Phase II Clinical Trial [NCT00061451]Phase 248 participants Interventional2002-12-31Completed
Clinical and Radiographic Evaluation of Pulpotomies in Primary Molars Using Tricalcium Silicate Cements. [NCT04902495]61 participants (Anticipated)Interventional2017-01-12Recruiting
An Open-label, Multicenter, Randomized Phase 2 Study Evaluating the Safety and Efficacy of Cisplatin and Pemetrexed With or Without Cixutumumab as First-Line Therapy in Patients With Advanced Nonsquamous Non-Small Cell Lung Carcinoma [NCT01232452]Phase 2172 participants (Actual)Interventional2011-04-30Completed
A Phase I-IIa Dose-Ranging Study of Pemetrexed (Alimta) Plus Cetuximab (Erbitux) in Patients With Recurrent Non-Small Cell Lung Cancer (NSCLC): Hoosier Oncology Group LUN04-79 [NCT00216203]Phase 1/Phase 236 participants (Actual)Interventional2005-05-31Completed
Individualized Pemetrexed Dosing in Patients With Non-small Cell Lung Cancer or Mesothelioma Based on Renal Function to Improve Treatment Response [NCT03656549]Phase 223 participants (Anticipated)Interventional2019-02-01Recruiting
Randomized, OpenLabel, Phase 3 Trial of Nivolumab Plus Ipilimumab or Nivolumab Plus Platinum Doublet Chemotherapy Versus Platinum Doublet Chemotherapy in Early Stage NSCLC [NCT02998528]Phase 3505 participants (Actual)Interventional2017-03-04Active, not recruiting
Phase I Trial of BKM120 in Combination With Carboplatin and Pemetrexed in Patients With Advanced Non-Squamous Non-Small Cell Lung Cancer (NSCLC) [NCT01723800]Phase 19 participants (Actual)Interventional2013-07-31Completed
Randomized Phase III Multicenter Trial of Customized Chemotherapy Versus Standard of Care for1st Line Treatment of Elderly Patients With Advanced Non-Small-Cell Lung Cancer [NCT03402048]Phase 3567 participants (Anticipated)Interventional2012-07-31Recruiting
A Prospective Phase 2 Study of PEmetrexed in Combination With Cisplatin in Patients With Advanced UrotheLIal CAnceR [NCT01490437]Phase 242 participants (Actual)Interventional2008-07-31Completed
A Phase II Randomized Trial Assessing the Combination of Gemcitabine and Pemetrexed in the First Line Treatment of Locally Advanced or Metastatic Non-Small Cell Lung Cancer [NCT00434135]Phase 2180 participants (Anticipated)Interventional2006-05-31Completed
A Multicenter Randomized Phase III Study of Pemetrexed Versus Erlotinib in Patients With Pretreated Advanced Non-Small-Cell Lung Cancer (NSCLC) [NCT00440414]Phase 3320 participants (Anticipated)Interventional2006-04-30Completed
A Phase II Study of Alimta as First Line Chemotherapy for Advanced or Metastatic Breast Cancer [NCT00106002]Phase 237 participants (Actual)Interventional2005-04-30Completed
A Multicenter Phase 2 Randomized Trial of Single-Agent ALIMTA or ALIMTA With Sequentially Administered GEMZAR as First-Line Chemotherapy in Elderly Patients or Patients Who Are Not Eligible for Platinum-Based Chemotherapy With Advanced NSCLC [NCT00489983]Phase 291 participants (Actual)Interventional2003-07-31Completed
A Phase 2 Trial of ALIMTA in Pretreated Patients With Unresectable or Metastatic Cancer of the Pancreas [NCT00490373]Phase 252 participants (Actual)Interventional2003-10-31Completed
Disposition of [14C]LY2603618 Following Intravenous Administration in Patients With Advanced and/or Metastatic Solid Tumors [NCT01296568]Phase 13 participants (Actual)Interventional2011-02-28Completed
A Randomized Multicenter Phase II Study Of Induction Therapy With Pemetrexed And Cisplatin Followed By Chemoradiation With Pemetrexed Versus Chemoradiation With Pemetrexed Followed By Consolidation Therapy With Pemetrexed And Cisplatin In Patients With St [NCT00497315]Phase 230 participants (Actual)Interventional2006-02-28Completed
Phase II Study of Pembrolizumab in Combination With Cisplatin or Carboplatin and Pemetrexed as Induction Chemo+Immunotherapy in Resectable Epithelioid and Biphasic Pleural Mesothelioma (CHIMERA Study) [NCT06155279]Phase 240 participants (Anticipated)Interventional2024-06-30Not yet recruiting
Phase II Trial of LP-300 in Combination With Carboplatin and Pemetrexed in Never Smoker Patients With Relapsed Advanced Primary Adenocarcinoma of the Lung After Treatment With Tyrosine Kinase Inhibitors (The HARMONIC Study) [NCT05456256]Phase 290 participants (Anticipated)Interventional2022-08-12Recruiting
A Phase 1 First-in-human Study of BMS-986406 as Monotherapy and Combination Therapies in Participants With Advanced Malignant Tumors [NCT05298592]Phase 1154 participants (Anticipated)Interventional2022-03-31Recruiting
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 3, Randomized Study of Nivolumab Plus Ipilimumab in Combination With Chemotherapy vs Chemotherapy Alone as First Line Therapy in Stage IV Non-Small Cell Lung Cancer [NCT03215706]Phase 3719 participants (Actual)Interventional2017-08-24Active, not recruiting
A Phase I Dose Escalation Study of Pemetrexed in Patients With Advanced Head and Neck Squamous Cell Cancer [NCT00507858]Phase 136 participants (Actual)Interventional2005-09-30Completed
Chemotherapy Plus Gefitinib Versus Gefitinib Alone as First-line Treatment for Patients With Advanced Lung Adenocarcinoma and Sensitive EGFR Mutations: a Randomized Controlled Trial [NCT02951637]Phase 2300 participants (Anticipated)Interventional2016-12-31Not yet recruiting
Feasibility Trial on Combination of Platinum Doublets and Hypofractionated Radiotherapy for Locally-advanced Stage and / or Inoperable Non-small Cell Lung Carcinoma [NCT02947113]Phase 20 participants (Actual)Interventional2017-11-30Withdrawn(stopped due to New studies available with immunotherapy so recruitment is no longer acceptable.)
Application of Detecting Circulating Tumor Cells in the Accurate Diagnosis and Treatment of Early Stage Lung Adenocarcinoma [NCT02951897]120 participants (Anticipated)Interventional2016-04-30Recruiting
[NCT02940990]Phase 250 participants (Anticipated)Interventional2016-11-30Not yet recruiting
A Single-arm, Two-stage Phase II Study of Lapatinib and Pemetrexed in the Second Line Treatment of Advanced or Metastatic Non-Small Cell Lung Cancer [NCT00528281]Phase 118 participants (Actual)Interventional2007-09-20Completed
Open-label, Multicenter, Randomized Phase II Trial of Treatment With Cisplatin and Pemetrexed or Cisplatin and Oral Vinorelbine in Chemotherapy Naïve Patients Affected by Stage IIIB-IV Non-Squamous Non-Small Cell Lung Cancer With High Thymidylate Synthase [NCT02919462]Phase 22 participants (Actual)Interventional2016-03-31Terminated(stopped due to low recruitement rate)
A Randomized, Phase 3, Open-label Study to Investigate the Pharmacokinetics and Safety of Subcutaneous Pembrolizumab Versus Intravenous Pembrolizumab, Administered With Platinum Doublet Chemotherapy, in the First-Line Treatment of Participants With Metast [NCT04956692]Phase 3531 participants (Actual)Interventional2021-08-05Active, not recruiting
A Phase 3, Randomized, Double-blind Study of Neoadjuvant Chemotherapy Plus Nivolumab Versus Neoadjuvant Chemotherapy Plus Placebo, Followed by Surgical Resection and Adjuvant Treatment With Nivolumab or Placebo for Participants With Resectable Stage II-II [NCT04025879]Phase 3452 participants (Anticipated)Interventional2019-11-05Active, not recruiting
Phase 1 Dose Escalation Study of LY2090314 in Patients With Advanced or Metastatic Cancer in Combination With Pemetrexed and Carboplatin [NCT01287520]Phase 141 participants (Actual)Interventional2007-11-30Completed
Concurrent Chemotherapy Based on Genetic Testing in Patients With High-Risk Salivary Gland Tumors [NCT02921984]Phase 120 participants (Actual)Interventional2013-09-30Completed
A Prospective, Single-arm, Single-center Clinical Trial of Envafolimab as First-line Replacement Maintenance in Advanced NSCLC [NCT05465733]Phase 225 participants (Anticipated)Interventional2022-08-01Not yet recruiting
Pilot Study Targeting Residual Hypermethylation in Early Stage Non-Small Cell Lung Cancer As Part of Adjuvant Therapy and Preventive Strategy [NCT01209520]6 participants (Actual)Interventional2009-07-31Completed
A Phase II Randomized, Multi-Center, Double-Blind, Global Study to Determine the Efficacy and Safety of Durvalumab Plus Olaparib Combination Therapy Compared With Durvalumab Monotherapy as Maintenance Therapy in Patients Whose Disease Has Not Progressed F [NCT03775486]Phase 2401 participants (Actual)Interventional2018-12-21Active, not recruiting
A Randomized, Phase 2 Trial of Lazertinib and Chemotherapy Combination in EGFR-mutant NSCLC Patients Without ctDNA Clearance After lead-in Lazertinib Monotherapy (CHAMELEON) [NCT06020989]Phase 2129 participants (Anticipated)Interventional2023-09-30Not yet recruiting
An Open-label, Multicenter, Phase Ib/II Study of AK104, Combined Chemotherapy as First-line Therapy to Treat Locally Advanced or Metastatic Non-small Cell Lung Carcinoma [NCT04647344]Phase 1/Phase 260 participants (Anticipated)Interventional2020-11-24Active, not recruiting
Study of Chinese Medicine Plus Chemotherapy Maintenance Versus Chemotherapy Maintenance in Advanced Non Small Cell Lung Cancer: A Randomized Double-blind Controlled Clinical Trial [NCT02900742]Phase 371 participants (Actual)Interventional2013-03-31Completed
Combination of Gefitinib With Chemotherapy or Anti-angiogenesis as 1st Line Treatment in Advanced NSCLC Patients Detected With Bim Deletion or Low EGFR Activating Mutation Abundance [NCT02930954]Phase 2180 participants (Anticipated)Interventional2016-11-30Not yet recruiting
A Phase 1b/2 Trial of AMG 386 in Combination With Pemetrexed and Carboplatin as First Line Treatment of Metastatic Non-Squamous Non-Small Cell Lung Cancer [NCT01666977]Phase 1/Phase 236 participants (Actual)Interventional2012-08-31Completed
A Phase 1 First-in-Human Dose Study of LY3023414 in Patients With Advanced Cancer [NCT01655225]Phase 1156 participants (Actual)Interventional2012-07-31Completed
An Open-label, Randomized Phase IIB/III Active Control Study of Second-line Tergenpumatucel-L (Hyper-Acute(R)-Lung ) Immunotherapy Versus Docetaxel in Progressive or Relapsed Non-Small Cell Lung Cancer [NCT01774578]Phase 2/Phase 3135 participants (Actual)Interventional2013-02-28Terminated
Personalized Escalation of Consolidation Treatment Following Chemoradiotherapy and Immunotherapy in Stage III NSCLC [NCT04585490]Phase 348 participants (Anticipated)Interventional2021-08-25Recruiting
A Phase II Two Cohorts Prospective Study to Evaluate the Efficacy and Safety of Tislelizumab Combined With Chemotherapy With or Without Bevacizumab in Non-squamous NSCLC With EGFR Sensitizing Mutation Who Failed EGFR TKI Therapy [NCT04405674]Phase 2120 participants (Anticipated)Interventional2020-07-15Recruiting
Phase II Study of a Triplet Combination of CBP501, Pemetrexed and Cisplatin as First Line Treatment in Patients With Stage IV Non-squamous Non Small Cell Lung Cancer (NSCLC) [NCT00942825]Phase 2195 participants (Actual)Interventional2009-04-30Completed
A Phase Ib, Open-label, Dose Escalation Trial Investigating Different Doses and Schedules of Sym004 in Combination With Platinum-doublets in Subjects With Stage IV Non-small Cell Lung Cancer [NCT02083679]Phase 115 participants (Actual)Interventional2014-07-31Terminated(stopped due to Sponsor the return rights of the compound to the collaboration partner for further clinical development)
A Phase II Study to Evaluate the Efficacy, Safety, and Tolerability of KN046 in Patients With Advanced Non-small Cell Lung Cancer [NCT03838848]Phase 2120 participants (Actual)Interventional2019-05-05Terminated(stopped due to Cohort A,B,C end enrollments. Cohort D and E were considered to have no significant clinical benefit at the SMC meeting and decided to terminate enrollment.)
Adjuvant Chemotherapy With Pemetrexed and Cisplatin vs. Vinorelbine and Cisplatin in NSCLC IB, IIA, IIB, T3N1: a Randomized Phase II Study [NCT00349089]Phase 2132 participants (Actual)Interventional2006-09-26Completed
Maintenance Immunotherapy With Autologous Cytokine-induced Killer Cells for Stage IIIb/IV Nonsquamous Non-small Cell Lung Cancer [NCT01481259]Phase 2/Phase 3120 participants (Anticipated)Interventional2010-01-31Recruiting
A Phase II Study of Pemetrexed in Patients With Advanced Neuroendocrine Tumors [NCT00424723]Phase 232 participants (Anticipated)Interventional2005-12-31Completed
Phase II Study of Carboplatin and Pemetrexed in Patients With Recurrent Platinum Sensitive Ovarian, Fallopian Tube or Primary Peritoneal Cancer [NCT00230542]Phase 244 participants (Actual)Interventional2005-09-30Completed
A Phase II Study of Eloxatin and Alimta in Combination With Bevacizumab in Advanced Non-Squamous NSCLC [NCT00251524]Phase 269 participants (Actual)Interventional2005-11-30Completed
A Phase 2 Study of ALIMTA Plus Doxorubicin Administered Every 21 Days in Patients With Advanced Breast Cancer [NCT00102219]Phase 277 participants (Actual)Interventional2004-10-31Completed
A Phase II Study of Pemetrexed Disodium (ALIMTA®) Plus Bevacizumab in Patients With Stage IIIB Pleural Effusion or Stage IV Non-Small Cell Lung Cancer (Second-Line Treatment) [NCT00268489]Phase 248 participants (Actual)Interventional2006-05-31Completed
A Two-Cohort Phase I/II Trial of PTK787 and Pemetrexed With or Without Cisplatin in Patients With Advanced Non-Small Cell Lung Cancers and Malignant Pleural Mesotheliomas [NCT00281125]Phase 1/Phase 220 participants Interventional2006-01-31Terminated(stopped due to Suspended due to data issues revealed at DSMB meeting. Planned amendment but was never submitted. Study was then closed.)
A Randomized Phase 3 Study of Two Doses of Alimta in Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer Who Have Failed a Prior Platinum-Containing Chemotherapy [NCT00078260]Phase 3589 participants (Actual)Interventional2003-12-31Completed
A Phase II Evaluation Of Pemetrexed (ALIMTA LY231514, IND #40061) In The Treatment Of Recurrent Or Persistent Platinum-Resistant Ovarian Or Primary Peritoneal Carcinoma [NCT00087087]Phase 251 participants (Anticipated)Interventional2004-07-31Completed
Cadonilimab (AK104) Plus Chemotherapy as First-line Treatment in Non-squamous Non-Small Cell Lung Cancer (NSCLC) Patients With Programmed Cell Death Ligand 1 (PD-L1) Negative:A Multi-center, Single-arm, Phase II Study [NCT06001151]Phase 249 participants (Anticipated)Interventional2023-08-07Recruiting
A Phase II Study to Evaluate the Efficacy, Safety and Tolerability of HLX26 (Anti-LAG-3 Monoclonal Antibody Injection) Combined With Serplulimab (Anti-PD-1 Humanized Monoclonal Antibody Injection) and Chemotherapy in Previously Untreated Advanced Non-smal [NCT05787613]Phase 260 participants (Anticipated)Interventional2023-07-10Recruiting
Older Non-Small Cell Lung Cancer Patients (>/= 70 Years of Age) Treated With First-Line MK-3475 (Pembrolizumab)+/- Chemotherapy (Oncologist's/Patient's Choice) [NCT04533451]Phase 2101 participants (Actual)Interventional2020-10-01Active, not recruiting
A Phase Ib/II, Open-Label Study of M7824 in Combination With Chemotherapy in Participants With Stage IV Non-small Cell Lung Cancer [NCT03840915]Phase 1/Phase 270 participants (Actual)Interventional2019-04-02Completed
A Phase II Trial to Evaluate Combination Therapy With Pemetrexed and Avelumab in Previously Treated Patients With MTAP-Deficient Advanced Urothelial Cancer [NCT03744793]Phase 218 participants (Actual)Interventional2019-04-11Active, not recruiting
A Phase 3 Trial of Balstilimab Versus Investigator Choice Chemotherapy in Patients With Recurrent Cervical Cancer After Platinum-Based Chemotherapy (BRAVA) [NCT04943627]Phase 30 participants (Actual)Interventional2021-08-02Withdrawn(stopped due to Strategic Business Decision)
Phase IV Randomized Trial of Pemetrexed Followed by Docetaxel or in Reverse Sequence in Non-small-cell Lung Cancer Patients Failed Previous Chemotherapy [NCT01442909]Phase 444 participants (Actual)Interventional2008-03-31Completed
Monitoring the Efficacy of Duvalizumab Combined With Neoadjuvant Chemotherapy for Ib-IIIb NSCLC by Sequencing of Immune Receptor Repertoire: a Prospective, One Arm Clinical Study [NCT04897386]Phase 230 participants (Anticipated)Interventional2021-06-01Not yet recruiting
A Phase II Evaluation Of Pemetrexed (ALIMTA, LY231517, IND #40061) In the Treatment Of Recurrent Carcinoma Of The Cervix [NCT00087113]Phase 20 participants Interventional2004-08-31Completed
A Phase Ib/II Clinical Trial of AK112 and AK104 With or Without Chemotherapy in Advanced Non-small Cell Lung Cancer [NCT05904379]Phase 1/Phase 2148 participants (Anticipated)Interventional2023-07-13Recruiting
Neoadjuvant Chemotherapy and Extrapleural Pneumonectomy of Malignant Pleural Mesothelioma (MPM) With or Without Hemithoracic Radiotherapy. A Randomized Multicenter Phase II Trial [NCT00334594]Phase 2153 participants (Actual)Interventional2005-11-14Completed
Binimetinib, Pemetrexed and Cisplatin, Followed by Maintenance With Binimetinib and Pemetrexed, in Patients With Advanced Non-small Cell Lung Cancer NSCLC With KRAS Mutations. A Multicenter Phase IB Trial. [NCT02964689]Phase 118 participants (Actual)Interventional2017-04-12Completed
A Phase 1, Open Label Study to Assess the Safety, Tolerability and Pharmacokinetics of AZD2171 and Selected Chemotherapy Regimens When Given in Combination to Patients With Advanced Solid Tumors [NCT00502567]Phase 1104 participants (Actual)Interventional2005-01-31Completed
Clinical Observation of the Effect of Neoadjuvant Anti-PD-1 Immunotherapy on Perioperative Analgesia and Postoperative Delirium in Patients With Non-small Cell Lung Cancer [NCT05273827]81 participants (Actual)Observational2022-03-22Active, not recruiting
A Phase II/III Multicenter Study Evaluating the Efficacy and Safety of Multiple Targeted Therapies as Treatments for Patients With Advanced or Metastatic Non-Small Cell Lung Cancer (NSCLC) Harboring Actionable Somatic Mutations Detected in Blood (B-FAST: [NCT03178552]Phase 2/Phase 31,000 participants (Anticipated)Interventional2017-09-22Recruiting
Random Open Exploratory Clinical Research of Sequential Gefitinib With Pemetrexed/Platinum Compare With Pemetrexed/Platinum Treatment for Advanced Non-small Cell Lung Cancer Exploratory Clinical Research [NCT01769066]Phase 2/Phase 3117 participants (Actual)Interventional2009-12-31Completed
A Phase II Trial to Evaluate Pemetrexed Clinical Responses in Relation to Tumor MTAP Gene Status in Patients With Previously Treated Metastatic Urothelial Carcinoma [NCT02693717]Phase 27 participants (Actual)Interventional2017-05-09Terminated(stopped due to The trial was closed due to the changing efficacy of treatments for metastatic urothelial cancer.)
Phase 2 Study of AZD2171 (NSC 732208) in Combination With Pemetrexed in Relapsed Non-Small Cell Lung Cancer (NOS: 10029514) [NCT00410904]Phase 260 participants (Actual)Interventional2006-10-31Completed
A Phase II Trial of Pemetrexed, Gemcitabine, and Bevacizumab Every Two Weeks in Chemotherapy-Naive Patients With Stages IIIB/IV Non- Squamous, Non-Small Cell Lung Cancer (NSCLC) [NCT00438204]Phase 239 participants (Actual)Interventional2006-05-31Terminated(stopped due to All data collection has completed.)
A Randomized, Phase II Study of Staggered, Chemo-Immunotherapy With Durvalumab, MEDI4736 Pemetrexed and Carboplatin (PC) for Metastatic Non-Squamous NSCLC [NCT04163432]Phase 284 participants (Anticipated)Interventional2020-06-16Recruiting
A Phase 1/2, Open-label, Multicenter Study of the Combination of NKTR-214 and Nivolumab or the Combination of NKTR-214, Nivolumab, and Other Anti-Cancer Therapies in Patients With Select Locally Advanced or Metastatic Solid Tumor Malignancies [NCT02983045]Phase 1/Phase 2557 participants (Actual)Interventional2016-12-19Completed
Phase II Study Evaluating the Interest of the Re-introduction of Pemetrexed and Platinum (Cisplatin or Carboplatin) With Prolonged Angiogenic Blocking by Bevacizumab in Non Squamous Non Small Cell Lung Cancer of Advanced Stage. [NCT01705184]Phase 2120 participants (Actual)Interventional2012-12-31Completed
A Phase II-III Randomized Trial Pemetrexed-Cisplatin Chemotherapy With or Without Bevacizumab (Avastin), 15 mg/kg, for Malignant Pleural Mesothelioma (MPM) [NCT00651456]Phase 2/Phase 3448 participants (Actual)Interventional2008-02-29Completed
A Phase II-III Randomized Trial Evaluating Maintenance Pembrolizumab (± Pemetrexed) Until Progression Versus Observation (± Pemetrexed) After 6 Months of Platinum-based Doublet Chemotherapy Plus Pembrolizumab Induction Treatment in Patients With Stage IV [NCT05255302]Phase 2/Phase 31,360 participants (Anticipated)Interventional2022-05-02Recruiting
A Randomised Phase 2 Trial of Pemetrexed and Gefitinib Versus Gefitinib as First Line Treatment for Patients With Stage IV Non-Squamous Non-Small Cell Lung Cancer With Activating Epidermal Growth Factor Receptor Mutations [NCT01469000]Phase 2195 participants (Actual)Interventional2012-02-29Completed
Randomized Phase II Study of Pemetrexed Versus Gefitinib in Previously Treated Patients With Advanced Non-small Cell Lung Cancer [NCT01783834]Phase 295 participants (Actual)Interventional2008-02-29Completed
A Phase IB Dose-Escalation Study of Pemetrexed and AUY922 in Previously-Treated Patients With Metastatic Non-Squamous, Non-Small Cell Lung Cancer [NCT01784640]Phase 113 participants (Actual)Interventional2014-01-31Completed
A Phase III, Open-Label, Randomized Study to Investigate the Efficacy and Safety of Atezolizumab (Anti-PD-L1 Antibody) Compared With Best Supportive Care Following Adjuvant Cisplatin-Based Chemotherapy in Patients With Completely Resected Stage IB-IIIA No [NCT02486718]Phase 31,280 participants (Actual)Interventional2015-10-31Active, not recruiting
A Randomized Phase II Trial of Erlotinib Versus Pemetrexed as Second-Line Therapy in Treating Patients With Advanced EGFR Wild-Type and EGFR FISH-Positive Lung Adenocarcinoma [NCT01565538]Phase 2123 participants (Actual)Interventional2008-12-31Completed
Phase II Trial of Lazertinib+Pemetrexed/Carboplatin in Patients With EGFR Sensitizing Mutation Positive Recurrent or Metastatic Non-Small Cell Lung Cancer Failed to Prior Lazertinib (LUCAS) [NCT05786430]Phase 287 participants (Anticipated)Interventional2023-03-31Not yet recruiting
A Phase 1 Study of MK-5890 as Monotherapy and in Combination With Pembrolizumab in Participants With Advanced Solid Tumors [NCT03396445]Phase 1202 participants (Anticipated)Interventional2018-02-18Active, not recruiting
Pemetrexed (Alimta) in Patients With Chemosensitive and Chemoresistant Relapsed Small Cell Lung Cancer: A Hoosier Oncology Group Phase II Study (LUN04-78) [NCT00216216]Phase 243 participants (Actual)Interventional2005-01-31Terminated(stopped due to The results from the interim analysis were not favorable to continue this trial.)
A Multi-arm Phase I Safety Study of Nivolumab in Combination With Gemcitabine/Cisplatin, Pemetrexed/Cisplatin, Carboplatin/Paclitaxel, Bevacizumab Maintenance, Erlotinib, Ipilimumab or as Monotherapy in Subjects With Stage IIIB/IV Non-small Cell Lung Canc [NCT01454102]Phase 1472 participants (Actual)Interventional2011-12-16Completed
Pemetrexed Plus Gemcitabine Or Carboplatin In Patients With Advanced Malignant Mesothelioma: A Randomized Phase II Trial [NCT00101283]Phase 232 participants (Actual)Interventional2006-02-23Completed
A Randomized, Double-Blind, Placebo-Controlled Phase 3 Study of INCMGA00012, an Anti-PD-1 Antibody, in Combination With Chemoradiation in Participants With Unresectable, Stage III Non-Small Cell Lung Cancer (POD1UM-301) [NCT04203511]Phase 30 participants (Actual)Interventional2020-07-31Withdrawn(stopped due to Sponsor Strategic/Business Decision)
Multicenter Phase II Study With Pemetrexed in Patients With Pre-Treated Metastatic Soft Tissue Sarcomas [NCT00427466]Phase 254 participants (Actual)Interventional2007-01-31Completed
A Phase I Study Investigating the Combination of Everolimus With Pemetrexed in Patients With Advanced Non Small Cell Lung Cancer (NSCLC) Previously Treated With Chemotherapy [NCT00434174]Phase 148 participants (Actual)Interventional2006-12-31Completed
A Phase I Study of ALIMTA Plus Oxaliplatin Administered Every Other Week in the Treatment of Patients With Metastatic Cancer [NCT00470405]Phase 125 participants (Actual)Interventional2004-05-31Completed
A Randomized (PhaseII), Double-blind, Multicenter Phase I/II Trial of Pemetrexed, Carboplatin Plus or Minus Sorafenib in the First-line Treatment of Patients With Stage IIIb or IV Non-Small Cell Lung Cancer [NCT00473486]Phase 1/Phase 212 participants (Actual)Interventional2007-05-31Terminated(stopped due to Sorafenib administered in the combination with pemetrexed-carboplatin appears to enhance thrombocytopenia compared to historical data.)
Efficacy and Safety of Platinum-based Chemotherapy + Bevacizumab + Durvalumab, and Salvage SBRT for IV Non-Small Cell Lung Cancer Patients With EGFR Mutations After Failure of First Line Osimertinib:A Multicenter, Prospective, Phase II Clinical Study [NCT04517526]Phase 260 participants (Anticipated)Interventional2020-11-01Not yet recruiting
A Randomized Phase 3 Study of Datopotamab Deruxtecan (Dato-DXd) and Pembrolizumab With or Without Platinum Chemotherapy in Subjects With No Prior Therapy for Advanced or Metastatic PD-L1 TPS <50% Non-squamous Non-small Cell Lung Cancer Without Actionable [NCT05555732]Phase 3975 participants (Anticipated)Interventional2023-01-11Recruiting
A Phase I, Open-Label Study to Assess the Safety and Tolerability of ZD6474 in Combination With Pemetrexed (Alimta) in Patients With Locally Advanced or Metastatic Non-small Cell Lung Cancer After Failure of Prior Chemotherapy. [NCT00506051]Phase 121 participants (Actual)Interventional2005-07-31Completed
A Randomized, Open Label, Phase II Study Comparing Pemetrexed Plus Cisplatin Followed by Pemetrexed Until Progression Versus Pemetrexed Alone Until Progression in Non-small Cell Lung Cancer Patients Who Have Progressed on First Line Epidermal Growth Facto [NCT03050437]Phase 296 participants (Anticipated)Interventional2013-03-31Recruiting
A Phase Ia/Ib Open-Label, Dose-Escalation Study of the Safety and Pharmacokinetics of Tiragolumab as a Single Agent and in Combination With Atezolizumab and/or Other Anti-Cancer Therapies in Patients With Locally Advanced or Metastatic Tumors [NCT02794571]Phase 1518 participants (Actual)Interventional2016-05-23Active, not recruiting
A Phase 1b Study of Ensartinib in Combination With Platinum-Based Chemotherapy and Bevacizumab in ALK-Positive Non-Small Cell Lung Cancer (NSCLC) [NCT04837716]Phase 112 participants (Anticipated)Interventional2021-03-18Active, not recruiting
A Phase I Study of Continuous Endostar Intravenous Infusion Combined With Pemetrexed and Carboplatin in Advanced NSCLC Patients [NCT01531790]Phase 119 participants (Actual)Interventional2011-09-30Completed
Sintilimab Plus Bevacizumab and Platinum-Based Doublet Chemotherapy as First-Line Treatment for Advanced Non-squamous Non-Small-Cell Lung Cancer With Negative Driver Gene: a Single-center, Single-Arm Trial [NCT05648071]Phase 360 participants (Anticipated)Interventional2021-12-01Recruiting
Beamion LUNG-2: A Phase III, Open-label, Randomized, Active-controlled, Multi-centre Trial Evaluating Orally Administered BI 1810631 Compared With Standard of Care as First-line Treatment in Patients With Unresectable, Locally Advanced or Metastatic Nonsq [NCT06151574]Phase 3270 participants (Anticipated)Interventional2024-01-15Not yet recruiting
A Two Steps Phase I Trial of Pazopanib or Pemetrexed in Combination With Crizotinib Followed by the Triplet, Crizotinib Plus Pazopanib Plus Pemetrexed in Patients With Advanced Malignancies [NCT01548144]Phase 1178 participants (Actual)Interventional2012-04-30Terminated(stopped due to PI request)
Afatinib Sequenced With Concurrent Chemotherapy and Radiation in EGFR-Mutant Non-Small Cell Lung Tumors: The ASCENT Trial [NCT01553942]Phase 230 participants (Anticipated)Interventional2012-04-30Recruiting
Phase I/II Study of Gemzar and Platinol Followed by Alimta and Gemzar in Patients With Advanced or Metastatic Bladder Cancer [NCT00101842]Phase 1/Phase 261 participants Interventional2004-12-31Completed
Phase II Trial of Pemetrexed for Advanced Chondrosarcomas [NCT00107419]Phase 275 participants (Actual)Interventional2005-09-30Completed
A Randomized, Double-Blind Phase 2 Study of Two Doses of Pemetrexed in the Treatment of Platinum-Resistant, Epithelial Ovarian or Primary Peritoneal Cancer [NCT00109096]Phase 2100 participants (Anticipated)Interventional2005-06-30Completed
Clinical Study of Neoadjuvant Anti-PD-1 Drug Toripalimab Combined With Chemotherapy in the Treatment of Locally Advanced Epithelial or Mixed Tissue Malignant Pleural Mesothelioma [NCT04713761]Phase 215 participants (Anticipated)Interventional2021-02-01Not yet recruiting
[NCT00034606]Phase 20 participants InterventionalCompleted
A Phase 3 Trial of ALIMTA (LY231514, Pemetrexed) Plus GEMZAR Versus GEMZAR in Patients With Unresectable or Metastatic Cancer of the Pancreas. [NCT00035035]Phase 30 participants InterventionalCompleted
A Phase II Clinical Trial of Tiragolumab in Combination With Carboplatin, Pemetrexed, and Atezolizumab in Patients With Non-squamous Non-small Cell Lung Cancer (NSCLC) and Untreated Brain Metastases [NCT05746481]Phase 235 participants (Anticipated)Interventional2023-08-08Recruiting
[NCT00055432]Phase 20 participants InterventionalTerminated
ALIMTA Plus Gemcitabine as Front-Line Chemotherapy for Patients With Malignant Pleural or Peritoneal Mesothelioma: A Phase II Clinical Trial [NCT00061477]Phase 248 participants Interventional2002-12-31Completed
Study of Two Doses of ALIMTA (Pemetrexed) as First Line Chemotherapy for Advanced Breast Cancer [NCT00065533]Phase 292 participants (Actual)Interventional2003-05-31Completed
A Phase II Study of Alimta and Carboplatin in the Treatment of Patients With Locally Advanced or Metastatic Breast Cancer [NCT00072865]Phase 250 participants Interventional2003-06-30Completed
A Phase 1/2 Study of the Highly Selective EGFR Inhibitor, BLU-701, in Patients With EGFR-Mutant Non-Small Cell Lung Cancer [NCT05153408]Phase 120 participants (Actual)Interventional2022-01-13Terminated(stopped due to Lack of efficacy)
A Prospective,Multi-center, Open-labeled Phase 2 Randomized and Comparative Clinical Study of First Line Intermittent and Maintenance of Icotinib in Combination With Pemetrexed/Carboplatin Compared With Icotinib Single Drug in ⅢB/IV Non Small Cell Lung Ca [NCT03151161]Phase 2118 participants (Anticipated)Interventional2015-12-31Not yet recruiting
A Phase II Trial of Alimta (Pemetrexed) in Patients With Recurrent Malignant Gliomas, Primary Central Nervous System Lymphoma, and Brain Metastases [NCT00276783]Phase 231 participants (Actual)Interventional2005-11-30Active, not recruiting
A Master Protocol of Phase 1/2 Studies of Nivolumab in Advanced NSCLC Using Nivolumab as Maintenance After Induction Chemotherapy or as First-line Treatment Alone or in Combination With Standard of Care Therapies (CheckMate 370: CHECKpoint Pathway and niv [NCT02574078]Phase 1/Phase 2341 participants (Actual)Interventional2015-11-23Completed
A Phase 1b Study of the Dual MDMX/MDM2 Inhibitor, ALRN-6924, for the Prevention of Chemotherapy-induced Myelosuppression [NCT04022876]Phase 135 participants (Actual)Interventional2019-09-03Terminated(stopped due to With a favorable safety profile the difference between treatment groups for the primary composite endpoint was not sufficient to generate statistically significant results with the targeted sample size)
[NCT00035061]Phase 20 participants InterventionalCompleted
A Phase I Study of Concurrent Pemetrexed, Cisplatin and Radiotherapy in Local Advanced Non-Small Cell Lung Cancer. [NCT00846443]Phase 112 participants (Anticipated)Interventional2009-01-31Recruiting
A Phase III, Open Label, Randomized Study of Atezolizumab (Anti-PD-L1 Antibody) Compared With a Platinum Agent (Cisplatin or Carboplatin) in Combination With Either Pemetrexed or Gemcitabine for PD-L1-Selected, Chemotherapy-Naive Patients With Stage IV No [NCT02409342]Phase 3572 participants (Actual)Interventional2015-07-20Completed
Induction Therapy With PD-1 Antibody Combined With Platinum-based Doublet Chemotherapy for Locally-advanced Non-small Cell Lung Cancer: A Randomised Controlled, Open-label, Phase 2 Trial [NCT05766800]Phase 2100 participants (Anticipated)Interventional2023-03-14Recruiting
A Phase II Clinical Trial Evaluating Three Schedules Of ALIMTA Plus Gemcitabine As Frontline Chemotherapy For Patients With Locally Advanced Or Metastatic Non-Small Cell Lung Cancer [NCT00022646]Phase 2157 participants (Actual)Interventional2001-08-31Completed
Phase I/II Trial Of Gemcitabine And ALIMTA In Patients With Measurable Or Evaluable, Unresectable Or Metastatic Biliary Tract Carcinoma (Intrahepatic, Extrahepatic, Ampulla Or Vater) And Gallbladder Carcinoma [NCT00059865]Phase 1/Phase 268 participants (Actual)Interventional2004-01-31Completed
ALIMTA Plus Gemcitabine as Front-Line Chemotherapy for Patients With Locally Advanced or Metastatic Non-Small Cell Lung Cancer: A Phase II Clinical Trial [NCT00061464]Phase 248 participants Interventional2003-02-28Completed
Phase 1/2 Dose-Escalating Study of Biweekly Alimta and Gemcitabine in Patients With Advanced Cancer [NCT00071136]Phase 1/Phase 248 participants Interventional2003-12-31Completed
A Phase II Study of Nivolumab in Combination With Carboplatin and Pemetrexed, or Nivolumab in Combination With Ipilimumab, in Patients With Advanced, EGFR-mutant or ALK-rearranged, Non-Small Cell Lung Cancer [NCT03256136]Phase 29 participants (Actual)Interventional2017-11-22Completed
[NCT00040625]0 participants Expanded AccessApproved for marketing
Phase I Trial of Cisplatin, Pemetrexed, and Imatinib Mesylate in Unresectable or Metastatic Malignant Mesothelioma [NCT00402766]Phase 119 participants (Actual)Interventional2006-08-31Completed
An Open-label, Randomized, Multicenter Trial of Intrathecal-pemetrexed Combined With Concurrent Involved-field Radiotherapy and Intrathecal-pemetrexed Alone in Patients With Leptomeningeal Metastasis From Solid Tumors [NCT05305885]100 participants (Anticipated)Interventional2022-08-19Recruiting
A Randomized Phase 2 Trial of ALIMTA Plus a Comparator Versus Leucovorin Modulated Fluorouracil Plus a Comparator in First Line Treatment of Locally Advanced or Metastatic Colorectal Cancer [NCT00079872]Phase 2120 participants Interventional2004-02-29Completed
A Randomized Phase 3 Trial Comparing ALIMTA Plus Best Supportive Care Versus Best Supportive Care Alone in Previously Treated Patients With Locally Advanced or Metastatic Malignant Pleural Mesothelioma [NCT00190762]Phase 3240 participants Interventional2001-10-31Completed
Open-Label Single-Arm Phase 2 Study of ALIMTA Plus Cisplatin in Korean Patients With Advanced Gastric Carcinoma [NCT00190801]Phase 250 participants Interventional2003-09-30Completed
Open-Label Single-Arm Phase 2 Study of ALIMTA in Patients With Advanced Non-Small Cell Lung Cancer Who Have Had Prior Chemotherapy [NCT00190840]Phase 2186 participants Interventional2003-09-30Completed
A Phase II Trial of Pemetrexed (Alimta) in the Treatment of Recurrent or Persistent Low Risk Gestational Trophoblastic Tumor [NCT00190918]Phase 250 participants (Anticipated)Interventional2006-07-31Completed
A Phase II Study of Biweekly Pemetrexed and Gemcitabine in Patients With Metastatic Breast Cancer [NCT00191347]Phase 230 participants Interventional2004-10-31Completed
Open-Label Single-Arm Phase II Study of ALIMTA in Combination With Oxaliplatin as First-Line Therapy in Advanced Gastric Carcinoma [NCT00192088]Phase 243 participants Interventional2004-05-31Completed
A Randomized Phase 2 Study Comparing Erlotinib-Pemetrexed, Pemetrexed Alone, and Erlotinib Alone, as Second-Line Treatment for Non-Smoker Patients With Locally Advanced or Metastatic Nonsquamous Non-Small Cell Lung Cancer [NCT00550173]Phase 2247 participants (Actual)Interventional2007-11-30Completed
Feasibility and Safety of Neoadjuvant Nivolumab and Chemotherapy for Resectable Malignant Pleural Mesothelioma [NCT04162015]Phase 135 participants (Anticipated)Interventional2019-11-12Recruiting
Adaptive-Dose to Mediastinum With Immunotherapy (Durvalumab MEDI4736) and Radiation in Locally-Advanced Non-Small Cell Lung Cancer [NCT04372927]Phase 21 participants (Actual)Interventional2021-12-10Terminated(stopped due to Terminated due to slow accrual)
Phase II Study of the Combination of Paclitaxel Poliglumex (CT-2103, Xyotax) and Pemetrexed (Alimta) for the Treatment of Patients With Advanced Non-small Cell Lung Cancer. [NCT00487669]Phase 214 participants (Actual)Interventional2006-10-31Completed
A Phase 2 Randomized Open-label Study of Erlotinib Plus Tivantinib (ARQ 197) Versus Single Agent Chemotherapy in Previously Treated KRAS Mutation Positive Subjects With Locally Advanced or Metastatic Non-Small Cell Lung Cancer [NCT01395758]Phase 296 participants (Actual)Interventional2011-07-31Completed
A Phase 2 Randomized Study of Relatlimab Plus Nivolumab in Combination With Chemotherapy vs. Nivolumab in Combination With Chemotherapy as First Line Treatment for Participants With Stage IV or Recurrent Non-small Cell Lung Cancer (NSCLC) [NCT04623775]Phase 2420 participants (Anticipated)Interventional2021-02-17Active, not recruiting
A Phase II Study of Pemetrexed and Gemcitabine Plus Bevacizumab as First Line Chemotherapy for Elderly Patients With Stage IIIB/IV Non-Small Cell Lung Cancer [NCT00517595]Phase 248 participants (Actual)Interventional2007-08-31Completed
Phase I Study of the Non-receptor Kinase Inhibitor Bosutinib in Combination With Pemetrexed in Patients With Selected Metastatic Solid Tumors [NCT03023319]Phase 16 participants (Actual)Interventional2019-12-10Completed
A Multicenter Phase II Trial of Neo-Adjuvant Pemetrexed (Alimta) Plus Cisplatin Followed by Surgery and Radiation for Pleural Mesothelioma [NCT00087698]Phase 277 participants (Actual)Interventional2003-09-30Completed
A Phase I Dose-Escalating Study of Induction Gemcitabine/Pemetrexed Followed by Pemetrexed and Concurrent Upper Abdominal Radiation Therapy in Patients With Locally Advanced Pancreatic Cancer [NCT00310050]Phase 14 participants (Actual)Interventional2005-10-31Terminated(stopped due to slow accrual)
Phase I/II Clinical Trial of Combined Pre-Irradiation With Pemetrexed and Erlotinib Followed by Maintenance Erlotinib for Recurrent and Second Primary Squamous Cell Carcinoma of the Head and Neck [NCT00573989]Phase 1/Phase 227 participants (Actual)Interventional2008-03-31Terminated(stopped due to Drug Supply Issue)
A Study of the Effect of Anlotinib, Pemetrexed or the Combination As Maintenance Therapy for Patients With Non-Squamous Non-Small Cell Lung Cancer. [NCT04453423]Phase 290 participants (Anticipated)Interventional2020-07-01Not yet recruiting
Association Between Proton Pump Inhibitors and the Incidence of Hematologic Toxicity of Pemetrexed: a Prospective, Multicenter, Observational and Longitudinal Study Among Patients Treated by a Pemetrexed-based Chemotherapy [NCT03537833]172 participants (Actual)Observational2018-05-02Completed
Phase I Open Label Trial of Alimta® Plus Cisplatin and Paclitaxel Given Intraperitoneally (IP) as First Line Treatment for Women With Stage III Ovarian Cancer [NCT00702299]Phase 115 participants (Actual)Interventional2007-09-30Completed
A Single-arm, Open-label, and Multicenter Phase Ⅱ Study Designed to Evaluate the Efficacy and Safety of Rulonilimab Combined With Chemotherapy in Patients With Advanced or Metastatic Non-small Cell Lung Cancer (NSCLC) [NCT05741021]Phase 284 participants (Anticipated)Interventional2023-03-01Not yet recruiting
A Phase Ib/II Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Initial Efficacy of Recombinant Humanized Anti-BTLA Monoclonal Antibody (JS004) Injection Combined With Toripalimab and With Standard Chemotherapy in Patients With Advanced Lun [NCT05664971]Phase 1/Phase 2240 participants (Anticipated)Interventional2023-02-09Recruiting
Alimta Plus Gemcitabine as Chemotherapy for Patients With Advanced Sarcoma: A Phase II Clinical [NCT00860015]Phase 212 participants (Actual)Interventional2005-08-31Completed
Use of Mineral Trioxide Aggregate in the Treatment of Traumatized Permanent Teeth With Necrotic Pulps and Chronic Periapical Lesions [NCT02625298]Phase 224 participants (Actual)Interventional2010-09-30Completed
Phase II, A Study of Lung-Sparing Combined Modality Protocol for the Treatment for Malignant Pleural Mesothelioma: The Columbia Protocol [NCT00859495]Phase 29 participants (Actual)Interventional2008-02-29Terminated
A Phase II Randomized, Open-Labelled, Multicenter Study of Safety & Efficacy of Combination Brigatinib and Carboplatin-Pemetrexed Therapy or Brigatinib Monotherapy as First-Line Treatment in Advanced ALK-Positive Non-Small Cell Lung Cancer [NCT05200481]Phase 2110 participants (Anticipated)Interventional2022-05-18Recruiting
A Phase II Study of Adjuvant Treatment With Cisplatin-based Chemotherapy Plus Concomitant Atezolizumab in Patients With Stage I (Tumors ≥ 4cm), IIA, IIB, and Select Stage III [Any T1-3 N1-2 and T4N0-2] Resected Non-small Cell Lung Cancer (NSCLC) and the C [NCT04367311]Phase 2100 participants (Anticipated)Interventional2020-05-22Recruiting
Phase II Multi-center Study of Pembrolizumab in Combination With Platinum-based Doublet Chemotherapy in NSCLC (Non-small Cell Lung Cancer) Patients With Targetable Genetic Alterations in Their Tumor Previously Treated With Appropriate Targeted Agents With [NCT03242915]Phase 233 participants (Actual)Interventional2017-10-03Active, not recruiting
S1300: A Randomized, Phase II Trial of Crizotinib Plus Pemetrexed Versus Pemetrexed Monotherapy in ALK-Positive Non-squamous NSCLC Patients Who Have Progressed Systemically After Previous Clinical Benefit From Crizotinib Monotherapy [NCT02134912]Phase 21 participants (Actual)Interventional2014-08-31Terminated(stopped due to science has moved forward and there is no intent to complete the study)
Phase 2 Study of Pemetrexed and Sorafenib for Treatment of Recurrent or Metastatic Triple Negative Breast Cancer [NCT02624700]Phase 213 participants (Actual)Interventional2016-01-28Terminated
Phase I Clinical Study of Tislelizumab in Combination With Bevacizumab and Pemetrexed for the First-line Treatment of Advanced Non-squamous Non-small Cell Lung Cancer in Elderly Patients [NCT05273814]Phase 130 participants (Anticipated)Interventional2022-08-01Not yet recruiting
A Phase 2 Study to Evaluate the Triplet Combination of Pemetrexed Plus AB928 (Etrumadenant) + AB122 (Zimberelimab) in Patients With Previously Treated Advanced or Metastatic MTAP Deficient Urothelial Carcinoma [NCT05335941]Phase 220 participants (Anticipated)Interventional2023-06-13Recruiting
Phase II Study of Alimta (Pemetrexed) Treatment of Advanced Thymoma and Thymic Carcinoma [NCT00198133]Phase 227 participants (Actual)Interventional2005-01-31Completed
A Phase II Feasibility Trial of Induction Chemotherapy Followed by Extrapleural Pneumonectomy and Postoperative Radiotherapy in Patients With Malignant Pleural Mesothelioma [NCT00227630]Phase 259 participants (Actual)Interventional2005-07-31Completed
Molecular Profiling and Safety Study of Operable Lung Cancer Patients Treated With Alimta Combined With Cisplatin as Neoadjuvant Chemotherapy [NCT00191308]Phase 230 participants (Actual)Interventional2005-05-31Completed
LY231514 500 mg/m2 and LY231514 1000 mg/m2 in Patients With Advanced Non-Small Cell Lung Cancer Who Were Previously Treated With Prior Systemic Anti Cancer Therapy: A Randomized Phase II Trial [NCT00191191]Phase 2226 participants (Actual)Interventional2004-10-31Completed
Pembrolizumab in Combination With Chemotherapy and Image-Guided Surgery for Malignant Pleural Mesothelioma (MPM) [NCT03760575]Phase 120 participants (Anticipated)Interventional2023-01-10Recruiting
Neoadjuvant PD-1 Antibody Plus Chemotherapy in Resectable Stage IIIA-N2 Non-Small-Cell Lung Cancer: A Randomized Phase II Study [NCT04422392]Phase 293 participants (Actual)Interventional2020-07-13Terminated(stopped due to since the CheckMate816 study is published in NEJM, enrolling patients becomes difficult)
An Open-label, Phase II Study of KN046 Evaluating the Efficacy and Safety of KN046 Plus Platinum-based Doublet Chemotherapy as First Line Therapy in Advanced Non-small Cell Lung Cancer Subjects. [NCT04054531]Phase 250 participants (Anticipated)Interventional2019-09-04Recruiting
A Phase III, Randomized, Double-blinded, Parallel Group, Multi-centre Study to Assess the Efficacy and Safety of ZD6474 (ZACTIMA™) in Combination With Pemetrexed (Alimta®) Versus Pemetrexed Alone in Patients With Locally-Advanced or Metastatic NSCLC [NCT00418886]Phase 3698 participants (Actual)Interventional2007-01-31Completed
A Phase I Study of Topotecan in Combination With Pemetrexed in Patients With Advanced Malignancies [NCT00315861]Phase 115 participants (Anticipated)Interventional2006-03-31Completed
A Phase I/II Trial of Temsirolimus and Pemetrexed in Recurrent/Refractory Non Small Cell Lung Cancer (NSCLC) [NCT00921310]Phase 1/Phase 212 participants (Actual)Interventional2009-09-30Terminated(stopped due to Withdrawal of funding from sponsor)
A Phase I Multicenter Study of Immunotherapy in Combination With Chemoradiation in Patients With Advanced Solid Tumors (CLOVER) [NCT03509012]Phase 1105 participants (Actual)Interventional2018-05-02Active, not recruiting
A Phase I Trial of Cetuximab (C225) and Pemetrexed With Concurrent Radiation in Head and Neck Cancer [NCT00291707]Phase 140 participants (Anticipated)Interventional2006-03-31Completed
A Randomized, Phase II, Multicenter, Double-Blind, Placebo-Controlled Study Evaluating the Efficacy and Safety of Metmab Vs. Placebo in Combination With Either Bevacizumab + Platinum + Paclitaxel or Pemetrexed + Platinum in Patients With Untreated Stage I [NCT01496742]Phase 2259 participants (Actual)Interventional2012-04-30Completed
A Phase II Study of Pemetrexed and Carboplatin in the Treatment of Esophageal Cancer [NCT00383266]Phase 29 participants (Actual)Interventional2006-10-31Terminated(stopped due to Low accrual)
A Randomized Placebo-controlled Phase II Study of Intercalated Administration of Pemetrexed/Cisplatin With Iressa® (Gefitinib) or Placebo as First-line Treatment of Stage IIIB/IV Lung Adenocarcinoma in Never-smokers [NCT01502202]Phase 2162 participants (Anticipated)Interventional2012-03-31Recruiting
Molecular and Genetic Changes in Patients With Resectable Non-Small Cell Lung Cancer (NSCLC) Following Neoadjuvant Chemotherapy With Cisplatin and Alimta - Phase II Study [NCT00248495]Phase 238 participants (Actual)Interventional2005-06-08Completed
Phase III Study Evaluating Two Strategies of Maintenance, One With Pemetrexed in Continuous Strategy and One According to the Response of Induction Chemotherapy, in Non Squamous Non Small Cell Lung Cancer of Advanced Stage [NCT01631136]Phase 3932 participants (Actual)Interventional2012-07-31Completed
A Phase II, Open-label, Multicentre, Randomised Study of Neoadjuvant and Adjuvant Treatment in Patients With Resectable, Early-stage (II to IIIB) Non-small Cell Lung Cancer (NeoCOAST-2) [NCT05061550]Phase 2350 participants (Anticipated)Interventional2022-04-14Recruiting
A Phase Ib Study of the Safety and Pharmacology of Atezolizumab (Anti-PD-L1 Antibody) Administered With Bevacizumab and/or Chemotherapy in Patients With Advanced Solid Tumors [NCT01633970]Phase 1240 participants (Actual)Interventional2012-07-11Completed
A Phase I Open-label Dose Escalation Study of Intravenous BI 2536 Together With Pemetrexed in Previously Treated Patients With Non-small-cell Lung Cancer [NCT02211833]Phase 141 participants (Actual)Interventional2006-10-31Completed
A Phase 1 Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of GS-5745 as Monotherapy and in Combination With Chemotherapy in Subjects With Advanced Solid Tumors [NCT01803282]Phase 1236 participants (Actual)Interventional2013-03-29Completed
Phase I Dose Escalation Study of Carboplatin, Pemetrexed and Exemestane in Post-menopausal Women With Metastatic Non-squamous NSCLC [NCT01664754]Phase 18 participants (Actual)Interventional2012-09-07Completed
A Phase 2 Multi-Center Randomized Trial to Assess Early Intervention With Adjuvant Nivolumab in Non-Small Cell Lung Cancer Participants With ctDNA-detected Minimal Residual Disease After Surgical Resection [NCT03770299]Phase 20 participants (Actual)Interventional2021-01-15Withdrawn(stopped due to Business objectives have changed)
Phase I Dose Escalation Study of Everolimus, Pemetrexed, Carboplatin, and Bevacizumab in Stage IV Non-Squamous Non-Small Cell Lung Cancer [NCT01700400]Phase 113 participants (Actual)Interventional2012-09-30Completed
Phase II Individualized Therapies Selection Study for Patients With Metastatic Colorectal Carcinoma According to the Genomic Expression Profile in Tumor Samples. [NCT01703910]Phase 229 participants (Actual)Interventional2012-11-30Completed
A Phase I-II Evaluation of the Safety and Efficacy of the Oral HSP90 Inhibitor Debio 0932 in Combination With Standard of Care in first-and Second-line Therapy of Patients With Stage IIIb or IV Non-small Cell Lung Cancer-the HALO Study (HSP90 Inhibition A [NCT01714037]Phase 182 participants (Actual)Interventional2012-08-31Terminated
Non-small Cell Lung Cancer: The Impact of Ethnic Origin on Patients Being Treated Second Line With Pemetrexed - An Observational Study [NCT00497770]434 participants (Actual)Observational2007-02-28Completed
Randomized Phase II Trial of Three-weekly Cisplatinum and Pemetrexed Versus Split-dose d1 and d8 Cisplatinum and Pemetrexed In Advanced and Inoperable Non-squamous Non-small-cell Lung Cancer (NSCLC) [NCT01742767]Phase 20 participants Interventional2012-11-30Recruiting
A Phase Ⅱ Randomized Controlled Trial to Compare Chemotherapy Sequenced by EGFR-TKIs and Chemotherapy Combined With EGFR-TKIs for Advanced or Metastatic NSCLC Patients Failed to EGFR-TKIs Therapy [NCT01746277]Phase 260 participants (Anticipated)Interventional2012-10-31Recruiting
Perioperative Immunotherpay Versus Adjuvant Immunotherapy for Resectable Non-small Cell Lung Cancer [NCT06109402]Phase 2160 participants (Anticipated)Interventional2023-12-20Not yet recruiting
A Phase 2 Trial of Combination Therapies With Adagrasib in Patients With Advanced Non-Small Cell Lung Cancer With KRAS G12C Mutation [NCT05609578]Phase 290 participants (Anticipated)Interventional2022-07-29Recruiting
SiCARIO (Split Course Adaptive Radioimmunotherapy) for the Treatment of Oligometastatic Non-Small Cell Lung Cancer (NSCLC) Using Biologically-Adaptive Radiotherapy - A Phase I/II Study [NCT05501665]Phase 1/Phase 225 participants (Anticipated)Interventional2023-05-09Recruiting
A Randomized Phase II/III Trial of Modern Immunotherapy Based Systemic Therapy With or Without SBRT for PD-L1-Negative, Advanced Non-Small Cell Lung Cancer [NCT04929041]Phase 2/Phase 3427 participants (Anticipated)Interventional2022-10-07Recruiting
A Phase III, Randomized, Double-Blind, Placebo-Controlled Study of Platinum Plus Pemetrexed Chemotherapy Plus Osimertinib Versus Platinum Plus Pemetrexed Chemotherapy Plus Placebo in Patients With EGFRm, Locally Advanced or Metastatic NSCLC Who Have Progr [NCT04765059]Phase 380 participants (Anticipated)Interventional2021-09-12Recruiting
A Phase 3 Study of Pembrolizumab (MK-3475) in Combination With Concurrent Chemoradiation Therapy Followed by Pembrolizumab With or Without Olaparib vs Concurrent Chemoradiation Therapy Followed by Durvalumab in Participants With Unresectable, Locally Adva [NCT04380636]Phase 3870 participants (Anticipated)Interventional2020-07-06Active, not recruiting
A Multi-center Phase II Randomized Study of Customized Neoadjuvant Therapy Versus Standard Chemotherapy in Non-small Cell Lung Cancer (NSLC) Patients With Resectable Stage IIIA (N2) Disease (CONTEST-TRIAL) [NCT01784549]Phase 2168 participants (Anticipated)Interventional2012-07-31Recruiting
A Phase II/III Clinical Trial to Evaluate the Efficacy and Safety of PM8002(Anti-PD-L1/VEGF) in Combination With Chemotherapy in Patients With EGFR-mutant Advanced Non-squamous NSCLC Who Have Failed to EGFR-TKI Treatment [NCT05756972]Phase 2/Phase 3374 participants (Anticipated)Interventional2023-06-26Recruiting
A Phase 1/2 Study Evaluating the Safety and Efficacy of ABT 751 in Combination With Pemetrexed Versus Pemetrexed Alone in Subjects With Advanced or Metastatic Non-Small Cell Lung Cancer [NCT00297089]Phase 1/Phase 2165 participants (Actual)Interventional2006-11-30Completed
Phase I Study of Pemetrexed and Sorafenib in Advanced Malignancy [NCT01450384]Phase 137 participants (Actual)Interventional2011-10-31Completed
A Pilot Study of Pembrolizumab and Single Agent Chemotherapy as First Line Treatment for Patients With Locally Advanced or Metastatic Non-small Cell Lung Cancer With Eastern Cooperative Oncology Group (ECOG) Performance Status of 2 [NCT04297605]Phase 128 participants (Anticipated)Interventional2020-05-15Recruiting
Neoadjuvant Chemotherapy for Non-metastatic Non-small Cell Lung Cancer [NCT01860040]Phase 21 participants (Actual)Interventional2013-04-30Terminated(stopped due to Poor accrual, no data to analyze)
Neoadjuvant of Sintilimab Combined With Chemotherapy for Resectable NSCLC(neoSCORE):A Prospective, Randomized, Open-Label, Single-Center Phase 2 Trial [NCT04459611]Phase 260 participants (Actual)Interventional2020-07-01Active, not recruiting
A Prospective, Single Center, Single Arm, Phase II Clinical Trial of Pyrotinib Combined With Pemetrexed Plus Carboplatin in the First-line Treatment of Patients With HER2 Mutant or Amplified Recurrent / Metastatic Non-small Cell Lung Cancer [NCT04706949]Phase 226 participants (Anticipated)Interventional2020-12-07Recruiting
A Phase I, Open Label, Multicentre Study to Assess the Safety, Tolerability, Pharmacokinetics and Preliminary Efficacy of Selumetinib (AZD6244; ARRY-142886) in Combination With First Line Chemotherapy Regimens in Patients With Non-Small Cell Lung Cancer ( [NCT01809210]Phase 155 participants (Actual)Interventional2013-04-04Completed
Atezolizumab/Carboplatin/Nab-Paclitaxel vs. Pembrolizumab/Platinum/Pemetrexed in Metastatic TTF-1 Negative Lung Adenocarcinoma [NCT05689671]Phase 4136 participants (Anticipated)Interventional2023-12-06Recruiting
A Phase 1b/2, Protocol Evaluating the Safety, Tolerability, Pharmacokinetics, and Efficacy of Sotorasib Monotherapy and in Combination With Other Anti-cancer Therapies in Subjects With Advanced Solid Tumors With KRAS p.G12C Mutation (CodeBreak 101) [NCT04185883]Phase 1/Phase 21,143 participants (Anticipated)Interventional2019-12-17Recruiting
A Randomized Double Blind Phase II Trial of Restorative Microbiota Therapy (RMT) in Combination With Durvalumab (MEDI4736) and Chemotherapy in Untreated Patients With Advanced or Metastatic Adenocarcinoma Non-Small Cell Lung Cancer [NCT04105270]Phase 282 participants (Anticipated)Interventional2022-11-30Recruiting
Phase II Study to Evaluate the Role of Postoperative Radiotherapy for Low Risk of Locoregional Recurrence Patients With Completely Resected Stage IIIA(N2) Non-Small Cell Lung Cancer [NCT02977169]Phase 2300 participants (Anticipated)Interventional2016-11-30Recruiting
Efficacy of Intrathecal Pemetrexed Combined With Tyrosine Kinase Inhibitor for Treating Leptomeningeal Metastasis in EGFR-Mutant NSCLC After Failure of Osimertinib [NCT05805631]Phase 223 participants (Anticipated)Interventional2023-05-01Not yet recruiting
A Randomized Phase II Study of Carboplatin and Pemetrexed w/ or w/o Selpercatinib in Participants With Non-Squamous RET Fusion-Positive Stage IV Non-Small Cell Lung Cancer and Progression of Disease on Prior RET Directed Therapy (Lung-MAP Sub-Study) [NCT05364645]Phase 274 participants (Anticipated)Interventional2022-07-25Recruiting
EORTC Randomized Phase II Study of Pleurectomy/ Decortication (P/D) Preceded or Followed by Chemotherapy in Patients With Early Stage Malignant Pleural Mesothelioma [NCT02436733]Phase 264 participants (Anticipated)Interventional2016-09-20Active, not recruiting
A Phase 1, Single Center, Dose-Escalation Study of SS1(dsFv)PE38 Administered Concurrently With Pemetrexed and Cisplatin in Subjects With Unresectable Malignant Epithelial Pleural Mesothelioma [NCT01445392]Phase 124 participants (Actual)Interventional2007-11-14Terminated
Apatinib Mesylate Combined With Pemetrexed in the Treatment of Pretreated Advanced Non-squamous Non-small Cell Lung Cancer :Single Arm Exploratory Study [NCT02974933]Phase 248 participants (Anticipated)Interventional2016-11-30Recruiting
A Phase II, Open, Randomised Study to Assess the Efficacy and Safety of AZD6244 Versus Pemetrexed (Alimta®) in Patients With Non-small Cell Lung Cancer, Who Have Failed One or Two Prior Chemotherapy Regimen [NCT00372788]Phase 288 participants (Actual)Interventional2006-08-31Completed
Phase 1b/2a Safety and Tolerability Study of Bemcentinib With Pembrolizumab/Carboplatin/Pemetrexed in Subjects With Untreated Advanced or Metastatic Non-squamous Non-small Cell Lung Cancer (NSCLC) Without/With a STK11 Mutation [NCT05469178]Phase 1/Phase 264 participants (Anticipated)Interventional2022-12-14Recruiting
A Phase II/III Study Comparing HX008 (a Humanized Monoclonal Antibody Against PD-1) Plus Chemotherapy With Pembrolizumab Plus Chemotherapy as the First-line Treatment in Participants With Advanced or Metastatic Nonsquamous Non-small Cell Lung Cancer. [NCT04750083]Phase 2/Phase 3700 participants (Anticipated)Interventional2020-09-25Recruiting
A Randomized Phase III Double Blind Trial Evaluating Selective COX-2 Inhibition in COX-2 Expressing Advanced Non-Small Cell Lung Cancer [NCT01041781]Phase 3313 participants (Actual)Interventional2010-02-28Terminated(stopped due to DSMB recommendation)
A Randomized Phase II Trial of Selumetinib in Patients Receiving Standard Pemetrexed and Platinum-based Chemotherapy for the Treatment of Advanced or Metastatic KRAS Wildtype or Unknown Non-Squamous Non-Small Cell Lung Cancer [NCT02337530]Phase 262 participants (Actual)Interventional2015-02-05Completed
Influence of Resin Cement Composition on the Longevity of Intraradicular Post in the Rehabilitation of Endodontically Treated Teeth - Randomized Clinical Trials [NCT03491527]20 participants (Anticipated)Interventional2018-05-01Not yet recruiting
Efficacy and Safety of Tislelizumab Combined With Bevacizumab and Platinum Plus Pemetrexed for Untreated EGFR+ and High PD-L1 Expression Non-squamous NSCLC :a Phase II, Single-center, Single Arm Study [NCT05394233]Phase 220 participants (Anticipated)Interventional2022-06-01Not yet recruiting
Neoadjuvant Osimertinib Plus Chemotherapy for EGFR-mutant Stage III N2 Non-squamous Non-small Cell Lung Cancer [NCT05011487]Phase 230 participants (Anticipated)Interventional2021-08-13Recruiting
Randomized Phase II Trial of Pemetrexed Plus Vinorelbine Versus Vinorelbine in Patients With Recurrent or Metastatic Breast Cancer Previously Treated With or Resistant to Anthracycline and Taxane [NCT03242616]Phase 2125 participants (Anticipated)Interventional2017-02-17Recruiting
A Phase I Study of Methoxyamine Combined With Chemo-Radiation for Locally Advanced Non-Squamous Non-Small Cell Lung Cancer [NCT02535325]Phase 117 participants (Actual)Interventional2015-09-30Completed
A Phase II Study of Carboplatin Plus Pemetrexed Plus Atezolizumab Plus Bevacizumab in Chemotherapy and Immunotherapy-naïve Patients With Stage IV Non-squamous Non-small Cell Lung Cancer: Big Ten Cancer Research Consortium BTCRC-LUN17-139 [NCT03713944]Phase 230 participants (Actual)Interventional2018-11-15Terminated(stopped due to Extreme toxicity, thromboembolic events)
A Randomized Phase II Study of Progression Free Survival Comparing Gemcitabine (1000 mg/m2 Infusion) Versus Carboplatin (AUC5 Infusion) Plus Alimta (500 mg/m2 Infusion) as First-line Chemotherapy in Elderly Patients With Locally Advanced (Stage IIIb) or M [NCT00754364]Phase 2108 participants (Actual)Interventional2008-10-31Terminated(stopped due to low enrollment rate)
A Phase I Pharmacokinetic Trial of LY231514 Administered Intravenously Every 3 Weeks in Advanced Cancer Patients With Varying Degrees of Renal Function [NCT00003706]Phase 150 participants (Actual)Interventional1998-04-30Completed
A Randomized Phase II Study of Radiation Therapy, Pemetrexed and Carboplatin With or Without Cetuximab in Stage III Non-Small Cell Lung Cancer [NCT00117962]Phase 2109 participants (Actual)Interventional2005-09-30Completed
Nivolumab With Chemotherapy in Pleural Mesothelioma After Surgery [NCT04177953]Phase 292 participants (Actual)Interventional2019-02-04Active, not recruiting
A Clinical and Molecular Risk-Directed Therapy for Newly Diagnosed Medulloblastoma [NCT01878617]Phase 2660 participants (Actual)Interventional2013-06-23Active, not recruiting
Intergroup Trial UNICANCER UC 0105-1305/ IFCT 1301: SAFIR02_Lung - Evaluation of the Efficacy of High Throughput Genome Analysis as a Therapeutic Decision Tool for Patients With Metastatic Non-small Cell Lung Cancer [NCT02117167]Phase 2999 participants (Actual)Interventional2014-04-23Active, not recruiting
Phase II Trial of Pemetrexed and Gemcitabine in Patients With Advanced Head and Neck Cancer (SCCHN) [NCT00394147]Phase 217 participants (Actual)Interventional2006-10-31Terminated(stopped due to stopped for lack of efficacy)
Sintilimab Combined With Anlotinib Hydrochloride and Standard Platinum-Containing Dual-Agent Chemotherapy in Non-Small Cell Lung Cancer (NSCLC) as First-Line Treatment: A Single-Arm, Prospective and Exploratory Clinical Study [NCT04846452]Phase 240 participants (Anticipated)Interventional2021-06-01Recruiting
Adjuvant Toripalimab Versus Placebo Combined With Chemotherapy for EGFR/ALK Mutation Negative Stage II-IIIB(N2) Non-small-cell Lung Cancer (LungMate-008): a Randomised, Double-blind, Controlled, Phase 3 Trial [NCT04772287]Phase 3341 participants (Anticipated)Interventional2021-03-31Not yet recruiting
A Phase 1/2 Study of NC318 in Combination With Chemotherapy for Subjects With Advanced or Metastatic Non-Small Cell Lung Cancer [NCT04430933]Phase 1/Phase 20 participants (Actual)Interventional2021-12-06Withdrawn(stopped due to Upon reviewing current available combo studies, the sponsor decided to prioritize different combo study.)
Randomized, Multicenter, Phase III, Open-Label Study of Alectinib Versus Pemetrexed or Docetaxel in Anaplastic Lymphoma Kinase-Positive Advanced Non Small Cell Lung Cancer Patients Previously Treated With Platinum-Based Chemotherapy and Crizotinib [NCT02604342]Phase 3119 participants (Actual)Interventional2015-11-03Completed
A Randomized, Open Label, Phase III Study of Overall Survival Comparing Pembrolizumab (MK-3475) Versus Platinum Based Chemotherapy in Treatment Naïve Subjects With PD-L1 Positive Advanced or Metastatic Non-Small Cell Lung Cancer (Keynote 042) [NCT02220894]Phase 31,274 participants (Actual)Interventional2014-10-30Completed
Alimta (Pemetrexed) and Cisplatin Treatment as Neoadjuvant Therapy in Non Small Cell Lung Cancer [NCT00259285]Phase 210 participants (Actual)Interventional2005-11-30Terminated(stopped due to Trial was stopped early due to low enrollment.)
Phase II Trial of Bevacizumab in Combination With Pemetrexed as Second Line Therapy in Patients With Stable Brain Metastases From Non-small Cell Lung Cancer (NSCLC) (Excluding Squamous Cell Carcinoma) [NCT00227019]Phase 216 participants (Actual)Interventional2006-03-31Completed
Phase II Study of Neoadjuvant Chemotherapy With Gemcitabine and Pemetrexed in Resectable Non-Small-Cell Lung Cancer (NSCLC) With Pharmacogenomic Correlates. [NCT00226577]Phase 252 participants (Actual)Interventional2004-02-29Completed
A Multicenter Phase II Trial of Pemetrexed Plus Carboplatin With or Without Apatinib in Patients With Advanced Non-small Cell Lung Cancer Without EGFR Mutation, ALK Gene Rearrangement, and ROS1 Gene Rearrangement [NCT03164694]Phase 2128 participants (Anticipated)Interventional2017-05-20Recruiting
Induction Therapy With Chemoimmunotherapy Followed by Surgery for Unresectable Stage III Non-small Cell Lung Cancer: a Single-center, Single-arm, Prospective Clinical Study [NCT04943029]Phase 230 participants (Anticipated)Interventional2021-08-20Recruiting
Phase II Trial of Lazertinib and Pemetrexed/Carboplatin Combination in Patients With EGFR Positive, Metastatic NSCLC With Asymptomatic or Mild Symptomatic Brain Metastases After Failure of Osimertinib [NCT05477615]Phase 228 participants (Anticipated)Interventional2022-08-31Not yet recruiting
A Single Arm, Open Label, Exploratory Study of Pemetrexed and S-1 in Combination With Bevacizumab in Patients Who Have Progressed After Standard Second Line Therapy [NCT03843853]Phase 20 participants (Actual)Interventional2019-05-01Withdrawn(stopped due to cooperation terminated)
QL1706 Combined With Platinum-based Chemotherapy Versus Placebo Combined With Platinum-based Chemotherapy as Adjuvant Therapy for Stage II-IIIB Non-small Cell Lung Cancer After Complete Surgical Resection: a Randomized, Double-blind, Multicenter Phase III [NCT05487391]Phase 3632 participants (Anticipated)Interventional2022-10-01Not yet recruiting
A Phase II Study of RRx-001 in Platinum Refractory/Resistant Small Cell Carcinoma, EGFR TKI Resistant EGFR+ T790M Negative Non-Small Cell Lung Cancer, High Grade Neuroendocrine Tumors and Resistant/Refractory Ovarian Cancer Prior to Re-administration of P [NCT02489903]Phase 2139 participants (Actual)Interventional2015-06-30Completed
DREAM3R: DuRvalumab (MEDI4736) With chEmotherapy as First Line treAtment in Advanced Pleural Mesothelioma - A Phase 3 Randomised Trial [NCT04334759]Phase 3214 participants (Actual)Interventional2021-02-18Active, not recruiting
A Dose-Finding Study Of Afatinib In Combination With Cisplatin Or Carboplatin + Pemetrexed In Patients With EGFR-Mutant Lung Cancers Undergoing Definitive Chemoradiation [NCT01836341]Phase 10 participants (Actual)Interventional2013-04-30Withdrawn
Phase III Trial of Single-Agent Pemetrexed (Alimta®) Versus the Combination of Carboplatin and Pemetrexed in Patients With Advanced Non-small-cell Lung Cancer and Performance Status of 2 [NCT01836575]Phase 3228 participants (Actual)Interventional2008-04-30Completed
A Randomized Phase II Study of Schedule-Modulated Concomitant Pemetrexed (Alimta) and Erlotinib (Tarceva) vs Single Agent Pemetrexed (Alimta®) in Patients With Progressive or Recurrent Non-small Cell Lung Cancer (NSCLC) [NCT00950365]Phase 279 participants (Actual)Interventional2006-04-30Completed
An Open-label, Safety and Tolerability Phase 1b Trial of CAN04, a Fully Humanized Anti-IL1RAP Monoclonal Antibody, and Pembrolizumab in Combination With and Without Carboplatin and Pemetrexed in Subjects With Solid Tumors [NCT04452214]Phase 119 participants (Actual)Interventional2020-09-24Completed
Phase 2 Study of Erlotinib Plus Pemetrexed/Cisplatin Treating Lung Adenocarcinoma With Brain Metastases [NCT01578668]Phase 269 participants (Actual)Interventional2012-01-31Completed
An Open Label, Randomized, Multicenter, Phase II Study to Compare Efficacy and Safety of Gefitinib/ Pemetrexed With Pemetrexed Alone as Maintenance Therapy in Patients With Advanced (Stage IV) EGFR Mutation Negative or T790M Single Mutation Nonsquamous NS [NCT01579630]Phase 2/Phase 352 participants (Actual)Interventional2011-03-31Active, not recruiting
A Randomized, Open-label, Phase III Study Comparing Pemetrexed With and Without Carboplatin in Elderly Patients With Advanced Non-Squamous Non-Small Cell Lung Cancer [NCT01593293]Phase 3266 participants (Anticipated)Interventional2012-03-31Recruiting
Phase Ib, Multicenter, Open Label Study of PDR001 in Combination With Platinum Doublet Chemotherapy and Other Immunooncology Agents in PD-L1 Unselected, Metastatic NSCLS Patients (ElevatION:NSCLC-101 Trial) [NCT03064854]Phase 1111 participants (Actual)Interventional2017-05-24Terminated(stopped due to Recruitment halted prematurely due to competitive landscape for lung cancer therapies)
A Randomized Open-Label Phase II Trial of Pemetrexed and a Platinum (Carboplatin or Cisplatin) With or Without Erlotinib in Patients With Non-Small Cell Lung Cancer Harboring Activating Epidermal Growth Factor Receptor Mutations and Acquired Resistance to [NCT01928160]Phase 20 participants (Actual)Interventional2014-06-30Withdrawn(stopped due to study not accruing)
Phase II Trial of Alimta (Pemetrexed) and Gemzar (Gemcitabine) in Metastatic Breast Cancer Patients Who Have Received Prior Taxane Therapy [NCT00063570]Phase 273 participants (Actual)Interventional2003-07-31Completed
Almonertinib Vs. Erlotinib/Chemotherapy for Neo-adjuVant Treatment of Stage IIIA-N2 EGFR-mutated NSCLC: a Multicenter, Open-label, Phase II Randomized Controlled Trial [NCT04455594]Phase 2168 participants (Anticipated)Interventional2020-10-31Not yet recruiting
A Phase III, Randomized, Open-Label Study of Pralsetinib Versus Standard of Care for First-Line Treatment of RET Fusion-Positive, Metastatic Non-Small Cell Lung Cancer [NCT04222972]Phase 3226 participants (Anticipated)Interventional2020-07-24Recruiting
A Phase II Study of Pemetrexed/Carboplatin/Radiotherapy and Bevacizumab in Patients With Unresectable Stage III Non-Small-Cell Lung Cancer [NCT00402883]Phase 25 participants (Actual)Interventional2006-11-30Terminated(stopped due to Terminated due to bevacizumab and chemoradiotherapy toxicity)
A Phase I Trial of Certolizumab in Combination With Chemotherapy for Patients With Stage IV Lung Adenocarcinomas [NCT02120807]Phase 130 participants (Actual)Interventional2014-04-15Completed
A Phase 1 Dose Escalation and Cohort Expansion Study of TSR-022, an Anti-TIM-3 Monoclonal Antibody, in Patients With Advanced Solid Tumors (AMBER) [NCT02817633]Phase 1475 participants (Anticipated)Interventional2016-07-08Recruiting
A Single-Blind Randomized Phase III Trial of MTA Plus Cisplatin Versus Cisplatin in Patients With Malignant Pleural Mesothelioma [NCT00005636]Phase 30 participants Interventional1999-11-30Completed
A Randomized, Double-Blind, Placebo-Controlled, Phase 3 Study to Compare the Efficacy and Safety of Neoadjuvant Treatment With Tislelizumab (BGB-A317, Anti-PD-1 Antibody) or Placebo Plus Platinum-Based Doublet Chemotherapy Followed By Adjuvant Tislelizuma [NCT04379635]Phase 3453 participants (Actual)Interventional2020-05-29Active, not recruiting
A Multicenter, Open-label, Randomized Phase II Study to Evaluate the Efficacy of AUY922 vs Pemetrexed or Docetaxel in NSCLC Patients With EGFR Mutations Who Have Progressed on Prior EGFR TKI Treatment [NCT01646125]Phase 259 participants (Actual)Interventional2012-11-23Terminated(stopped due to An interim analysis was conducted in May-2014. Upon review of the data, the committee recommended study termination due to futility.)
A Phase III Multicenter, Randomized, Open-Label Study Evaluating the Efficacy and Safety of Atezolizumab (MPDL3280A, Anti-PD-L1 Antibody) in Combination With Carboplatin+Nab-Paclitaxel for Chemotherapy-Naive Patients With Stage IV Non-Squamous Non-Small C [NCT02367781]Phase 3723 participants (Actual)Interventional2015-04-16Completed
A Pilot Trial of Platinum, Gemcitabine, or Pemetrexed Single- or Multi-Agent Therapy With Serial Tumor Specimen Collection in Patients With Advanced Non-Small-Cell Lung Cancer [NCT02145078]4 participants (Actual)Interventional2014-06-30Terminated(stopped due to Slow accrual.)
A Randomized, Double-Blind Phase 2 Study of Ruxolitinib or Placebo in Combination With Pemetrexed/Cisplatin and Pemetrexed Maintenance for Initial Treatment of Subjects With Nonsquamous Non-Small Cell Lung Cancer That Is Stage IIIB, Stage IV, or Recurrent [NCT02119650]Phase 276 participants (Actual)Interventional2014-02-11Terminated(stopped due to The study was terminated as other related studies of ruxolitinib did not provide sufficient efficacy to warrant continuation.)
A Phase II Study of AZD1775 Plus Pemetrexed and Carboplatin Followed by a Randomised Comparison of Pemetrexed and Carboplatin With or Without AZD1775 in Patients With Previously Untreated Stage IV Non-Squamous Non-Small-Cell Lung Cancer [NCT02087241]Phase 222 participants (Actual)Interventional2014-03-31Terminated(stopped due to The sponsor decided to terminate the study.)
An Open-label, Randomized, Phase IIIb Trial Evaluating the Efficacy and Safety of Standard of Care +/- Continuous Bevacizumab Treatment Beyond Progression of Disease (PD) in Patients With Advanced Non-squamous Non-small Cell Lung Cancer (NSCLC) After Firs [NCT01351415]Phase 3485 participants (Actual)Interventional2011-06-25Completed
A Phase Ib Multicenter, Open-label Study to Evaluate the Safety and Tolerability of Trastuzumab Deruxtecan (T-DXd) and Immunotherapy Agents With and Without Chemotherapy Agents in First-line Treatment of Patients With Advanced or Metastatic Non-squamous N [NCT04686305]Phase 1168 participants (Anticipated)Interventional2021-03-09Recruiting
An Open-Label, Randomized, Phase 3 Trial of Nivolumab Versus Investigator's Choice Chemotherapy as First-Line Therapy for Stage IV or Recurrent PD-L1+ Non-Small Cell Lung Cancer [NCT02041533]Phase 3541 participants (Actual)Interventional2014-03-27Completed
Phase II Trial of Motexafin Gadolinium and Pemetrexed (Alimta®) for Second Line Treatment in Patients With Non-Small Cell Lung Cancer [NCT00365183]Phase 274 participants (Actual)Interventional2006-06-30Terminated
A Phase I/II Multi-site Study of Rucaparib and Pembrolizumab Maintenance Therapy in Stage IV Non-Squamous Non-Small Cell Lung Cancer After Initial Therapy With Carboplatin, Pemetrexed, and Pembrolizumab [NCT03559049]Phase 1/Phase 225 participants (Actual)Interventional2018-12-24Active, not recruiting
An Open-label, Multicenter Study to Evaluate the Efficacy and Safety of HLX07 (Recombinant Humanized Anti-EGFR Monoclonal Antibody Injection) in Advanced nsqNSCLC Patients With High EGFR Expression [NCT05215925]Phase 260 participants (Anticipated)Interventional2023-02-01Not yet recruiting
A Randomized, Three-Arm, Open-Label Phase 3b Clinical Trial of Aumolertinib, Versus Aumolertinib With Chemotherapy, Versus Osimertinib for Patients With Metastatic NSCLC and an EGFR Mutation (TREBLE) [NCT05493501]Phase 38 participants (Actual)Interventional2022-12-14Terminated(stopped due to The study is being closed based on corporate changes at EQRx and is not related to any efficacy or safety issues with aumolertinib.)
Phase II Investigation of Use of CNS Active Pembrolizumab and Chemotherapy for Asymptomatic Brain Metastasis From Non-small Cell Lung Cancer (NSCLC) [NCT04964960]Phase 245 participants (Anticipated)Interventional2022-05-19Recruiting
A Phase 1/1b Study to Evaluate the Safety and Tolerability of Immunotherapy Combinations in Participants With Lung Cancer [NCT03846310]Phase 177 participants (Actual)Interventional2019-04-01Active, not recruiting
A Randomized, Double-Blind, Phase 3 Study of Pemetrexed + Platinum Chemotherapy With or Without Pembrolizumab (MK-3475) in TKI-resistant EGFR-mutated Tumors in Metastatic Non-squamous Non-small Cell Lung Cancer (NSCLC) Participants (KEYNOTE-789) [NCT03515837]Phase 3492 participants (Actual)Interventional2018-06-29Completed
A 2-Arm Phase 2 Double-Blind Randomized Study of Carboplatin, Pemetrexed Plus Placebo Versus Carboplatin, Pemetrexed Plus Truncated Demcizumab as First-Line Treatment in Subjects With Stage IV Non-Squamous Non-Small Cell Lung Cancer [NCT02259582]Phase 282 participants (Actual)Interventional2015-02-28Completed
Phase II Clinical Trial to Evaluate the Preliminary Efficacy, Safety and Pharmacokinetic Characteristics of PM8002 Injection Combined With Standard Chemotherapy in the First-line Treatment of Subjects With Inoperable Malignant Mesothelioma [NCT05918107]Phase 255 participants (Anticipated)Interventional2022-08-13Recruiting
First-in-Human Dose Study of IOA-244 Alone and in Combination With Pemetrexed/Cisplatin in Patients With Advanced or Metastatic Cancers [NCT04328844]Phase 1210 participants (Anticipated)Interventional2020-02-25Active, not recruiting
A Randomized, Double-Blind, Placebo-Controlled, Multicenter, Phase III Clinical Study on Toripalimab Combined With Platinum-Based Doublet Drug Chemotherapy for Resectable, Stage II-III, Non-Small Cell Lung Cancer [NCT04158440]Phase 3501 participants (Actual)Interventional2020-04-07Active, not recruiting
A Phase 2 Trial of Pembrolizumab (MK-3475) in Combination With Platinum Doublet Chemotherapy and Radiotherapy for Participants With Unresectable, Locally Advanced Stage III Non-Small Cell Lung Cancer (NSCLC) (KEYNOTE-799) [NCT03631784]Phase 2217 participants (Actual)Interventional2018-10-19Active, not recruiting
A Phase I Trial of LY231514 With Irinotecan Administered Intravenously Every 21 Days in Patients With Metastatic Cancer [NCT00003711]Phase 10 participants Interventional1997-09-30Completed
Alvopem® (Pemetrexed) Safety Assessment in Patients With Non-small Cell Lung Cancer and Malignant Pleural Mesothelioma [NCT04843007]199 participants (Actual)Observational2016-10-05Completed
a Clinical Trial Phase II Prospective, Single-arm Study of Recombinant Human Angioendostatin /PD-1 Mab Combined With First-line Chemotherapy in the Treatment of Driver Gene Negative Advanced Non-small Cell Lung Cancer [NCT05448781]Phase 238 participants (Anticipated)Interventional2022-07-20Not yet recruiting
Efficacy and Safety of Postoperative Adjuvant Chemotherapy Combined With Camrelizumab for Patients With ⅡA -ⅢA Non-small Cell Lung Cancer:a Phase II , Single-arm Clinical Study [NCT05825443]Phase 257 participants (Anticipated)Interventional2023-04-10Recruiting
From Liquid Biopsy to Cure: Using ctDNA Detection of Minimal Residual Disease to Identify Patients for Curative Therapy After Lung Cancer Resection [NCT04966663]Phase 266 participants (Anticipated)Interventional2022-03-28Recruiting
A First-in-Human Study of Repeat Dosing With REGN2810, a Monoclonal, Fully Human Antibody to Programmed Death - 1 (PD-1), as Single Therapy and in Combination With Other Anti-Cancer Therapies in Patients With Advanced Malignancies [NCT02383212]Phase 1398 participants (Actual)Interventional2015-02-02Completed
A Feasibility Study of Pemetrexed Single Agent and Folic Acid Given as Neoadjuvant Treatment in Patients With Resectable Rectal Cancer [NCT00330915]Phase 237 participants (Actual)Interventional2006-06-30Completed
A Phase II Trial of Cisplatin, Pemetrexed and Bevacizumab in Untreated Malignant Mesothelioma [NCT00295503]Phase 253 participants (Actual)Interventional2006-02-28Completed
Phase II Study of Dose Attenuated Chemotherapy in Patients With Lung Cancer and Age > 70 and/or Comorbidities [NCT05800587]Phase 2280 participants (Anticipated)Interventional2023-02-22Recruiting
Neoadjuvant Toripalimab Combined With Chemotherapy in Rare Mutations Stage IIB-IIIB NSCLC [NCT05800340]Phase 230 participants (Anticipated)Interventional2023-04-04Recruiting
A Randomized Phase 3 Multicenter Open-Label Study to Compare the Efficacy of YK-029A as First-Line Treatment Versus Platinum-Based Chemotherapy in Patients With Non-Small Cell Lung Cancer With EGFR Exon 20 Insertion Mutations [NCT05767892]Phase 3350 participants (Anticipated)Interventional2023-05-01Not yet recruiting
A Study of Nivolumab in Combination With Ipilimumab (Part 1); and Nivolumab Plus Ipilimumab in Combination With Chemotherapy (Part 2) as First Line Therapy in Stage IV Non-Small Cell Lung Cancer (NSCLC) [NCT02659059]Phase 2324 participants (Actual)Interventional2016-02-15Completed
Phase I/II Trial of Bevacizumab, Pemetrexed and Erlotinib in the First-Line Treatment of Elderly Patients With Advanced (Stage IIIB(With Malignant Pleural Effusion) or IV) Non-Squamous Non-Small Cell Lung Cancer (NSCLC) [NCT00351039]Phase 1/Phase 28 participants (Actual)Interventional2006-07-31Terminated(stopped due to slow accrual)
Phase I/II Study of Two Different Schedules of Bortezomib (VELCADE, PS-341) and Pemetrexed (ALIMTA) in Advanced Solid Tumors, With Emphasis on Non-Small Cell Lung Cancer (NSCLC) [NCT00389805]Phase 1/Phase 227 participants (Actual)Interventional2005-03-31Completed
Home Delivery of Pemetrexed as Maintenance Treatment in Patients Who Have Not Progressed After Induction Therapy for Advanced Nonsquamous Nonsmall Cell Lung Cancer: A Feasibility Study [NCT01473563]Phase 252 participants (Actual)Interventional2011-12-31Completed
A Phase III Randomised, Double Blind, Placebo Controlled, Parallel, Multicentre Study to Assess the Efficacy and Safety of Continuing IRESSA 250 mg in Addition to Chemotherapy Versus Chemotherapy Alone in Patients Who Have Epidermal Growth Factor Receptor [NCT01544179]Phase 3265 participants (Actual)Interventional2012-03-15Completed
A Single Arm, Prospective, and Exploratory Clinical Study of Camrelizumab Combined With Pemetrexed and Carboplatin in Advanced Non-small Cell Lung Cancer Patients With EGFR Mutation Who Failed EGFR-TKI Treatment [NCT04970043]58 participants (Anticipated)Interventional2021-07-31Not yet recruiting
A Randomized, Parallel Control, Exploratory Trial to Compare Apatinib Plus Chemotherapy Drug Versus Chemotherapy Drug as Second-line Treatment in Subjects With Advanced or Metastatic of the Non-small Cell Lung Cancer (NSCLC) [NCT03256721]Phase 237 participants (Actual)Interventional2017-08-16Terminated(stopped due to Due to the enrollment was slow and exceeded the expected enrollment time)
Fluzoparib Combined With Camrelizumab for Maintenance Treatment of Locally Advanced Non-small Cell Lung Cancer After Concurrent Chemotherapy and Radiotherapy. A Single-arm, Single-center, Phase II Clinical Study [NCT04828395]Phase 265 participants (Anticipated)Interventional2021-03-01Recruiting
A Phase II, Prospective, Single-center, Randomized, Controlled Study to Investigate the Efficacy and Safety of Sintilimab or Placebo in Combination With Chemotherapy as Second-line Treatment for Patients With Stage IV Nonsquamous Non-small Cell Lung Cance [NCT03863483]Phase 270 participants (Anticipated)Interventional2019-03-26Recruiting
A Phase II, Prospective, Single-center, Randomized, Controlled Study to Investigate the Efficacy and Safety of Sintilimab Compared With Docetaxel or Pemetrexed as Second-line Treatment for Patients With Stage IV Nonsquamous Non-small Cell Lung Cancer With [NCT03830411]Phase 276 participants (Anticipated)Interventional2019-03-13Recruiting
SUNRAY-01, A Global Pivotal Study in Participants With KRAS G12C-Mutant, Locally Advanced or Metastatic Non-Small Cell Lung Cancer Comparing First-Line Treatment of LY3537982 and Pembrolizumab vs Placebo and Pembrolizumab in Those With PD-L1 Expression ≥5 [NCT06119581]Phase 31,016 participants (Anticipated)Interventional2024-01-03Not yet recruiting
Phase II Pilot Study of Performance Status 2 vs. Performance Status 0-1 Non-Small Cell Lung Cancer Patients Treated With Chemo/Immunotherapy [NCT04253964]Phase 280 participants (Anticipated)Interventional2020-07-01Recruiting
A Phase 1b Study of LY2835219 in Combination With Multiple Single Agent Options for Patients With Stage IV NSCLC [NCT02079636]Phase 1142 participants (Actual)Interventional2014-03-28Completed
A Phase III, Open-label, Multicenter Trial of Avelumab (MSB0010718C) Versus Platinum-based Doublet as a First-line Treatment of Recurrent or Stage IV PD-L1+NSCLC [NCT02576574]Phase 31,214 participants (Actual)Interventional2015-10-29Active, not recruiting
Intrathecal Pemetrexed for Recurrent Leptomeningeal Metastasis From Non-small Cell Lung Cancer: A Prospective Pilot Clinical Trial [NCT03101579]Phase 113 participants (Actual)Interventional2017-03-01Completed
A Randomized, Double-blind, Phase III Trial to Compare the Efficacy and Safety of AK104 Combined With Chemotherapy to Tislelizumab Combined With Chemotherapy as First-line Treatment in PD-L1 TPS < 1% Non-small Cell Lung Cancer (NSCLC) [NCT05990127]Phase 3642 participants (Anticipated)Interventional2023-11-14Not yet recruiting
A Pilot Window of Opportunity Study Evaluating Durvalumab (MEDI4736) in Combination With Platinum Doublet Chemotherapy Followed by Evaluation of Durvalumab (MEDI4736) in Combination With Platinum Doublet Chemotherapy and Abequolixron (RGX-104) in Non-smal [NCT05911308]Phase 124 participants (Anticipated)Interventional2023-11-30Not yet recruiting
Nivolumab Plus Ipilimumab Plus Two Cycles of Platinum-based Chemotherapy as First Line Treatment for Stage IV/Recurrent Non-small Cell Lung Cancer (NSCLC) Patients With Synchronous Brain Metastases [NCT05012254]Phase 271 participants (Anticipated)Interventional2021-11-18Recruiting
A Phase II Trial of AK112 (PD1/VEGF Bispecific) in Combination With Chemotherapy in Patients With NSCLC [NCT04736823]Phase 2296 participants (Anticipated)Interventional2021-02-01Recruiting
A Phase II Trial of Neoadjuvant Treatment Carboplatin-Pemetrexed-Bevacizumab Plus Atezolizumab for the Treatment of Locally Advanced and Potentially Resectable NSCLC Patients With EGFR Mutations [NCT04512430]Phase 226 participants (Anticipated)Interventional2020-12-02Recruiting
Induction Durvalumab Followed by Chemoradiation and Consolidation Durvalumab (MEDI4736) for Stage III Non-small Cell Lung Cancer [NCT04364048]Phase 210 participants (Actual)Interventional2020-06-18Active, not recruiting
[NCT01454934]Phase 3540 participants (Actual)Interventional2011-12-09Completed
An Open-Label, Multicenter, Randomized Phase Ib/II Study of Eribulin Mesylate Administered in Combination With Pemetrexed Versus Pemetrexed Alone as Second Line Therapy in Patients With Stage IIIB or IV Nonsquamous Non Small Cell Lung Cancer [NCT01126736]Phase 1/Phase 298 participants (Actual)Interventional2010-06-10Completed
Phase II Trial Of Pemetrexed Disodium And Gemcitabine In Advanced Urothelial Cancer [NCT00053209]Phase 246 participants (Actual)Interventional2004-08-10Completed
Efficacy and Safety of Almonertinib Combined With or Without Chemotherapy as an Adjuvant Treatment for EGFR Mutation Positive Stage II-IIIA Non-small Cell Lung Carcinoma Following Complete Tumour Resection: A Multicenter, Randomized Controlled, Open-label [NCT04762459]Phase 3606 participants (Anticipated)Interventional2021-08-01Enrolling by invitation
Randomised, Controlled Study Comparing Chemotherapy Plus Intercalated EGFR-Tyrosine Kinase Inhibitors Combination Therapy With EGFR-Tyrosine Kinase Inhibitors Alone Therapy as First-line Treatment for Patients With Non-Small-Cell Lung Cancer [NCT02031601]Phase 4250 participants (Anticipated)Interventional2014-01-31Recruiting
Phase I/II Trial of Carboplatin, Bevacizumab and Pemetrexed in the First-Line Treatment of Patients With Malignant Pleural Mesothelioma (MPM) [NCT00604461]Phase 1/Phase 213 participants (Actual)Interventional2007-10-31Terminated(stopped due to Slow Accrual)
A Phase 1, Pharmacologic and Pharmacodynamic Study of MM-121 in Combination With Multiple Anticancer Therapies in Patients With Advanced Solid Tumors [NCT01447225]Phase 143 participants (Actual)Interventional2011-10-31Completed
Phase II Multicenter Randomized Trial Evaluating the Association of PIPAC and Systemic Chemotherapy Versus Systemic Chemotherapy Alone as 1st-line Treatment of Malignant Peritoneal Mesothelioma [NCT03875144]Phase 266 participants (Anticipated)Interventional2020-08-14Recruiting
Serial [18F]Fluorothymidine (FLT)PET/CT as a Biomarker of Therapeutic Response in Pemetrexed Therapy for Non-Small Cell Lung Cancer [NCT02274038]3 participants (Actual)Interventional2015-03-31Terminated(stopped due to Poor Accrual)
A Phase 3,Randomized,Open,Parallel Controlled, Multi-center Study of TQB2450 Injection Plus Chemotherapy Followed by TQB2450 Plus Anlotinib Versus Tislelizumab Plus Chemotherapy Followed by Tislelizumab as a First-line Treatment on Patient With Advanced N [NCT05346952]Phase 3390 participants (Anticipated)Interventional2022-01-25Recruiting
A Phase 2 Study to Evaluate LY2603618 in Combination With Pemetrexed in Patients With Advanced or Metastatic Non-small Cell Lung Cancer [NCT00988858]Phase 255 participants (Actual)Interventional2009-11-30Completed
A Randomized, Open-Label, Phase 2 Safety and Efficacy Trial of Ipilimumab Versus Pemetrexed in Subjects With Recurrent/Stage IV Non-Squamous, Non-Small Cell Lung Cancer Who Have Not Progressed After Four Cycles of a Platinum-Based First Line Chemotherapy [NCT01471197]Phase 29 participants (Actual)Interventional2012-07-31Terminated(stopped due to Administrative reasons)
A Randomized Phase 2, Double-blind, Placebo-controlled, Multi-center Study Comparing Pemetrexed in Combination With TH-302 vs. Pemetrexed in Combination With Placebo as Second-line Chemotherapy for Advanced Non-Squamous, Non-Small Cell Lung Cancer [NCT02093962]Phase 2265 participants (Actual)Interventional2014-06-30Terminated(stopped due to At the interim analysis, the futility boundary was not met and the study was stopped due to insufficient efficacy.)
An Open-label, Multi-center, Clinical Study to Evaluate Anti-PD-1 Antibody Therapies of Camrelizumab in Combination With Pemetrexed and Carboplatin as First-line Treatment in Patients With Advanced or Metastatic Non-Squamous Non-Small Cell Lung Cancer [NCT05841472]Phase 260 participants (Anticipated)Interventional2023-08-23Recruiting
A Phase III, Randomised, Controlled, Multi-center, 3-Arm Study of Neoadjuvant Osimertinib as Monotherapy or in Combination With Chemotherapy Versus Standard of Care Chemotherapy Alone for the Treatment of Patients With Epidermal Growth Factor Receptor Mut [NCT04351555]Phase 3328 participants (Anticipated)Interventional2020-12-16Recruiting
A Phase 2 Study of Neoadjuvant Pembrolizumab-Based Combination Immunotherapy in the Treatment of Early Stage Non-Small Cell Lung Cancer [NCT04061590]Phase 20 participants (Actual)Interventional2020-05-29Withdrawn(stopped due to Low Accrual)
Phase I/II Trial of Abraxane® (ABI-007) and Alimta® (Pemetrexed) in Advanced Solid Tumors With Emphasis on Non-Small Cell Lung Cancer (NSCLC) and Breast Cancer [NCT00470548]Phase 1/Phase 249 participants (Actual)Interventional2007-04-30Terminated(stopped due to Practice patterns with pemetrexed have evolved.)
Open-Label, Randomized Trial of Nivolumab (BMS-936558) Plus Pemetrexed/Platinum or Nivolumab Plus Ipilimumab (BMS-734016) vs Pemetrexed Plus Platinum in Stage IV or Recurrent Non-Small Cell Lung Cancer (NSCLC) Subjects With Epidermal Growth Factor Recepto [NCT02864251]Phase 3367 participants (Actual)Interventional2017-03-17Completed
A Phase I Study of MK-3475 Alone in Subjects With Advanced Solid Tumors and in Combination With Platinum-Doublet Chemotherapy or Immunotherapy in Subjects With Advanced Non-Small Cell Lung Cancer/Extensive-Disease Small Cell Lung Cancer. [NCT01840579]Phase 157 participants (Actual)Interventional2013-04-26Completed
Phase I/II Study of a Triplet Combination of CBP501, Pemetrexed and Cisplatin in Patients With Advanced Solid Tumors and in Chemotherapy-naïve Patients With Malignant Pleural Mesothelioma [NCT00700336]Phase 1/Phase 269 participants (Actual)Interventional2008-05-31Completed
Clinical, Radiographic, and Histologic Outcome of Sodium Hypoclorite as an Antibacterial Agent Prior to Calcium Hydroxide and Mineral Trioxide Aggregate Pulpotomies in Primary Teeth [NCT04270318]64 participants (Actual)Interventional2012-09-30Completed
A Randomized Phase II Study of Itraconazole and Pemetrexed in Patients With Previously Treated Non-Squamous Non-Small Cell Lung Cancer [NCT00769600]Phase 223 participants (Actual)Interventional2008-11-30Terminated(stopped due to Low accrual.)
A Randomized Phase II Trial of Cytotoxic Chemotherapy With or Without Epigenetic Priming in Patients With Advanced Non-Small Cell Lung Cancer [NCT01935947]Phase 217 participants (Actual)Interventional2013-05-31Terminated
Randomized, Double-blind, Placebo-controlled, Multi-center Study of Camrelizumab Combined With SRT/WBRT and Chemotherapy in Patients of NSCLC With Brain Metastases of Driven Gene-negative and Not Received Systemic Chemotherapy [NCT04768075]Phase 3200 participants (Anticipated)Interventional2021-03-05Not yet recruiting
TS Stratified Chemotherapy and VEGF Inhibition in Non-Squamous Non-Small Cell Lung Cancer - Stage IV [NCT01674738]Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to Decision of the Sponsor, as the funding of the study was no longer guaranteed.)
Integration of the PD-L1 Inhibitor Atezolizumab and WT1/DC Vaccination Into Platinum/Pemetrexed-based First-line Treatment for Epithelioid Malignant Pleural Mesothelioma [NCT05765084]Phase 1/Phase 215 participants (Anticipated)Interventional2023-02-24Recruiting
A Phase II Trial of Carboplatin, Pemetrexed, and Panitumumab in Patients With Advanced Non-Squamous K-ras Wild Type Non-Small-Cell Lung Cancer [NCT01042288]Phase 270 participants (Actual)Interventional2010-06-30Completed
Pemetrexed Plus Tarceva as Salvage Treatment in EGFR Overexpressed Metastatic Colorectal Cancer Patients Who Were Failed After Standard Chemotherapy: A Phase II Single Arm Prospective Study [NCT03086538]Phase 229 participants (Actual)Interventional2017-05-30Completed
A Prospective Study to Evaluate the Safety of Concurrent Durvalumab (MEDI4736) With Chemoradiation Therapy(CRT)Followed by Durvalumab for Chinese Unresectable Stage III Non Small Cell Lung Cancer(NSCLC) [NCT04982549]Phase 235 participants (Anticipated)Interventional2021-01-21Recruiting
Pembrolizumab With Standard Cytotoxic Chemotherapy in Treatment Naive Non-small Cell Lung Cancer Patients With Asymptomatic Brain Metastases [NCT04967417]Phase 250 participants (Anticipated)Interventional2021-10-31Not yet recruiting
A Phase 1B Study to Evaluate the Safety and Induction of Immune Response of CRS-207 in Combination With Pemetrexed and Cisplatin as Front-line Therapy in Adults With Malignant Pleural Mesothelioma [NCT01675765]Phase 160 participants (Actual)Interventional2014-09-03Completed
A Phase 3, Randomized Double-blind, Placebo-controlled, Multicenter Study of SHR-1701 in Combination With Bevacizumab and Chemotherapy in Advanced or Metastatic Non-squamous Non-small-cell Lung Cancer With EGFR Mutation After Failure of TKIs [NCT05132413]Phase 3561 participants (Anticipated)Interventional2021-12-30Not yet recruiting
Phase I Study of Cytoreductive Surgery and Hyperthermic Intraoperative Chemotherapy With Pemetrexed and Cisplatin for Malignant Pleural Mesotheliomas [NCT02838745]Phase 17 participants (Actual)Interventional2016-09-30Terminated(stopped due to Change in research plan.)
A Platform Study of RAS(ON) Inhibitor Combinations in Patients With RAS-Mutated Non-Small Cell Lung Cancer (NSCLC) [NCT06162221]Phase 1/Phase 2352 participants (Anticipated)Interventional2023-12-31Not yet recruiting
A Phase III, Randomized, Open-Label, Multicenter, Global Study of Volrustomig (MEDI5752) in Combination With Carboplatin Plus Pemetrexed Versus Platinum Plus Pemetrexed or Nivolumab Plus Ipilimumab in Participants With Unresectable Pleural Mesothelioma (e [NCT06097728]Phase 3600 participants (Anticipated)Interventional2023-11-09Recruiting
An Open-Label, Multicenter Extension Study in Participants Previously Enrolled in a Genentech and/or F. Hoffmann-La Roche Ltd Sponsored Atezolizumab Study (IMbrella C) [NCT05112965]Phase 3100 participants (Anticipated)Interventional2021-12-01Recruiting
A Phase II Randomized Study of Ramucirumab Plus MK3475 (Pembrolizumab) Versus Standard of Care for Patients Previously Treated With Immunotherapy for Stage IV or Recurrent Non-Small Cell Lung Cancer (Lung-MAP Non-Matched Sub-Study) [NCT03971474]Phase 2166 participants (Actual)Interventional2019-05-28Active, not recruiting
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
Maintenance Systemic Therapy Versus Local Consolidative Therapy (LCT) Plus Maintenance Systemic Therapy for Limited Metastatic Non-Small Cell Lung Cancer (NSCLC): A Randomized Phase II/III Trial [NCT03137771]Phase 2218 participants (Actual)Interventional2017-04-07Active, not recruiting
Single Cell Analysis of CXCL13+PD1+ CD8 T Cell in Association With Resistance to Pembrolizumab and Chemotherapy Neoadjuvant/Adjuvant of NSCLC [NCT05894889]Phase 270 participants (Anticipated)Interventional2023-10-01Not yet recruiting
"Neoadjuvant Tislelizumab and Platinum-Based Doublet Chemotherapy in Stage II-IIIB EGFR-Mutated Lung Adenocarcinoma With PD-L1 Positive Expression -- A Phase II Study (DuoVitality)" [NCT05527808]28 participants (Anticipated)Interventional2022-08-01Recruiting
LUME-Meso: Double Blind, Randomised, Multicentre, Phase II/III Study of Nintedanib in Combination With Pemetrexed / Cisplatin Followed by Continuing Nintedanib Monotherapy Versus Placebo in Combination With Pemetrexed / Cisplatin Followed by Continuing Pl [NCT01907100]Phase 2/Phase 3545 participants (Actual)Interventional2013-09-19Terminated
Multi-arm, Non-randomized, Open-Label Phase IB Study to Evaluate GSK3052230 in Combination With Paclitaxel and Carboplatin, or Docetaxel or as Single Agent in Subjects With Solid Malignancies and Deregulated FGF Pathway Signaling [NCT01868022]Phase 165 participants (Actual)Interventional2013-10-09Completed
Phase 2 Trial of Neoadjuvant Nivolumab + Platinum-based Chemotherapy + Certolizumab in Patients With Resectable Stages II-III Lung Cancers [NCT04991025]Phase 260 participants (Anticipated)Interventional2022-10-19Recruiting
A Safety and Feasibility Study of AGS-003-LNG for the Treatment of Stage 3 Non Small Cell Lung Cancer [NCT02662634]Phase 20 participants (Actual)Interventional2016-03-31Withdrawn
Phase II Trial of Neoadjuvant Platinum-based Chemotherapy for Patients With Resectable , Non-small Cell Lung Cancer With Switch to Chemotherapy Alternative in Nonresponders (NEOSCAN) [NCT01443078]Phase 242 participants (Actual)Interventional2011-10-31Completed
Phase II Randomized Trial of Radiation, Cetuximab and Pemetrexed With or Without Bevacizumab in Locally Advanced Head and Neck Cancer [NCT00703976]Phase 280 participants (Actual)Interventional2008-10-31Completed
An Open-Label, Phase 1/2 Study of ORIC-114 as a Single Agent or in Combination With Chemotherapy, in Patients With Advanced Solid Tumors Harboring an EGFR or HER2 Alteration [NCT05315700]Phase 1/Phase 2280 participants (Anticipated)Interventional2022-03-10Recruiting
Neoadjuvant Soft Tissue Ablation Utilizing Aliya™ Pulsed Electric Fields With Systemic Therapy in Early-Stage Resectable Non-Small Cell Lung Cancer (NSCLC) [NCT05583188]Phase 415 participants (Anticipated)Interventional2023-02-01Recruiting
A Single Arm, Open Label, Exploratory Study of Pemetrexed and TAS-102 in Combination With Bevacizumab in Patients Who Have Progressed After Standard Second Line Therapy [NCT04683965]Phase 227 participants (Anticipated)Interventional2021-01-01Active, not recruiting
Alternating Treatment Plans for Participants With Advanced Thoracic/Head & Neck Cancers (ATATcH) [NCT05358548]Phase 2150 participants (Anticipated)Interventional2022-04-28Recruiting
The Phase Three Trials of Pemetrexed/Cisplatin Intercalating Gefitinib vs Pemetrexed/Cisplatin Treating EGFR Wild NSCLC(Non Squamous Cell Carcinoma) [NCT03374280]Phase 2178 participants (Anticipated)Interventional2016-12-01Recruiting
A Phase III, Randomized, Multi-center Study to Determine the Efficacy of the Intercalating Combination Treatment of Chemotherapy and Gefitinib or Chemotherapy as Adjuvant Treatment in NSCLC With Common EGFR Mutations. [NCT03381066]Phase 3225 participants (Anticipated)Interventional2018-04-10Recruiting
Phase Ib Study to Evaluate the Efficacy, Safety and Tolerability of IBI188 Combination Therapy in Subjects With Advanced Malignancies [NCT04861948]Phase 19 participants (Actual)Interventional2021-05-25Terminated(stopped due to No signs of efficacy in solid tumors)
MEK114375: A Rollover Study to Provide Continued Treatment With GSK1120212 to Subjects With Solid Tumors or Leukemia [NCT01376310]Phase 2159 participants (Actual)Interventional2010-11-02Terminated(stopped due to Company Decision)
Emulation of a Comparative Effectiveness Study of Pembrolizumab and Chemotherapy vs. Chemotherapy for the First-line Treatment of Metastatic Non-small Cell Lung Cancer [NCT05908799]1,854 participants (Actual)Observational2023-06-05Completed
A Phase 3, Randomized, Open-label, Multicenter Study to Assess the Efficacy and Safety of HS-10241 Combined With Almonertinib Versus Pemetrexed Combined With Platinum in Metastatic or Locally Advanced NSCLC With MET Amplification After Failure of the Prio [NCT06110663]Phase 3314 participants (Anticipated)Interventional2023-12-30Not yet recruiting
A Phase II Clinical Study to Evaluate the Efficacy and Safety of SI-B001 in Combination With Chemotherapy in the Treatment of EGFR WT and ALK WT Recurrent and Metastatic Non-small Cell Lung Cancer [NCT05020457]Phase 260 participants (Anticipated)Interventional2021-12-07Recruiting
A Randomised Non-comparative Open Label Phase II Trial of Atezolizumab Plus Bevacizumab, With Carboplatin-paclitaxel or Pemetrexed, in EGFR-mutant Non-small Cell Lung Carcinoma With Acquired Resistance [NCT04245085]Phase 295 participants (Actual)Interventional2020-09-29Active, not recruiting
A Randomized, Open-Label, Phase 3 Trial of Tisotumab Vedotin vs Investigator's Choice Chemotherapy in Second- or Third-Line Recurrent or Metastatic Cervical Cancer [NCT04697628]Phase 3556 participants (Anticipated)Interventional2021-02-22Recruiting
PD-1 Inhibitor and Chemotherapy With Concurrent Irradiation at Varied Tumour Sites in Advanced Non-small Cell Lung Cancer [NCT03774732]Phase 3327 participants (Anticipated)Interventional2019-03-21Recruiting
Phase 2 Study of Platinum-Based Chemotherapy in Combination With Durvalumab (MEDI 4736) for NSCLC in Patients With a Poor Performance Status and the Elderly [NCT04262869]Phase 20 participants (Actual)Interventional2020-03-27Withdrawn(stopped due to Due to accrual issues)
Phase II Multicenter Trial of Neoadjuvant Cisplatin and Nab-paclitaxel for (N2) Defined Stage IIIA Non-Small Cell Lung Cancer (NSCLC) [NCT02276560]Phase 21 participants (Actual)Interventional2015-01-31Terminated(stopped due to This study was terminated due to lack of funding.)
A Phase 2 Trial of GFH018 and Toripalimab in Combination With Concurrent Chemoradiotherapy for Patients With Unresectable, Locally Advanced Stage III Non-Small Cell Lung Cancer (NSCLC) [NCT05386888]Phase 265 participants (Actual)Interventional2022-09-09Active, not recruiting
A Phase 1 Study to Investigate the Safety and Pharmacokinetics of Cemiplimab (Anti-PD-1) and Other Agents in Japanese Patients With Advanced Malignancies [NCT03233139]Phase 1145 participants (Anticipated)Interventional2017-06-21Recruiting
A Phase II, Single-Arm, Prospective Study of Neoadjuvant Icotinib With Chemotherapy for the Treatment of Patients With Epidermal Growth Factor Receptor Mutation Positive, Resectable for Stage II to IIIB(N2) Non-small Cell Lung Cancer [NCT05104788]Phase 227 participants (Anticipated)Interventional2021-10-25Recruiting
Cryoablation in Combination (or Not) With Pembrolizumab and Pemetrexed-carboplatin in First-line Treatment for Patients With Metastatic Lung Adenocarcinoma: A Randomized Phase III Study [NCT04339218]Phase 3214 participants (Anticipated)Interventional2020-08-28Recruiting
Phase II Study of Pemetrexed Plus Cisplatin in Patients With Refractory Soft Tissue Sarcoma [NCT03809637]Phase 237 participants (Actual)Interventional2017-01-10Active, not recruiting
A Phase 2, Multicenter, Randomized Study to Evaluate the Safety and Efficacy of Viagenpumatucel-L (HS-110) in Combination With Low Dose (Metronomic) Cyclophosphamide Versus Chemotherapy Alone in Patients With Non-Small Cell Lung Adenocarcinoma After Failu [NCT02117024]Phase 266 participants (Actual)Interventional2014-07-31Terminated(stopped due to Sponsor Decision; strategic - based on changing treatment landscape)
An Open Label, Multicenter, Phase Ib/II Study of SHR-1802 in Combination With Adebrelimab in Patients With Advanced Solid Tumors [NCT05794477]Phase 1/Phase 2132 participants (Anticipated)Interventional2023-04-28Recruiting
A Phase III, Multicenter, Randomized, Open-label Study of Oral LDK378 Versus Standard Chemotherapy in Adult Patients With ALK-rearranged (ALK-positive) Advanced Non-small Cell Lung Cancer Who Have Been Treated Previously With Chemotherapy (Platinum Double [NCT01828112]Phase 3231 participants (Actual)Interventional2013-06-28Completed
Predictive Biomarker for the Efficacy and Safety of the Combination of Chemotherapy and Tislelizumab in Non Small Cell Lung Cancer:a Multicentre Prospective Clinical Trial [NCT05244837]Phase 2100 participants (Anticipated)Interventional2020-12-22Recruiting
A Randomized, Double-blind, Placebo-controlled Study of the Safety and Efficacy of Amatuximab in Combination With Pemetrexed and Cisplatin in Subjects With Unresectable Malignant Pleural Mesothelioma [NCT02357147]Phase 2124 participants (Actual)Interventional2015-11-03Terminated(stopped due to Due to business reasons)
A Phase II Study of Lenvatinib (E7080/MK-7902) in Combination With Carboplatin Pemetrexed and Pembrolizumab (MK-3475) for Patients With Pretreated Advanced Non-squamous Non-small Cell Lung Cancer Harboring EGFR Mutations [NCT05258279]Phase 230 participants (Anticipated)Interventional2022-07-01Active, not recruiting
A Open-label, Single Center, Phase II Study of Surufatinib Combined With Toripalimab and Chemotherapy in Patients With Advanced Non-squamous Non-small-cell Lung Cancer [NCT05003037]Phase 2106 participants (Anticipated)Interventional2021-12-08Recruiting
A Randomized Phase II Trial Comparing Cetuximab With Concurrent Pemetrexed/Cetuximab Therapy for Non-Small Cell Lung Cancer Refractory to Primary Treatment [NCT00203931]Phase 255 participants (Actual)Interventional2005-03-31Terminated(stopped due to Slow accrual and evidence from other studies showing benefit of early initiation of pemetrexed after first-line therapy)
A Prospective Randomized Phase III Trial of Maintenance Pemetrexed Versus Observation in Patients With Recurrent or Metastatic Urothelial Carcinoma Who Completed First Line Platinum-based Chemotherapy Without Disease Progression [NCT03193788]Phase 374 participants (Anticipated)Interventional2017-01-31Recruiting
A Phase II Study of Pemetrexed (Alimta) as Second-Line Therapy for Hormone Refractory Prostate Cancer: Hoosier Oncology Group GU03-67 [NCT00216099]Phase 249 participants (Actual)Interventional2005-02-28Completed
Phase III Study to Evaluate the Optimal Timing of Postoperative Radiotherapy for High Risk of Locoregional Recurrence Patients With Completely Resected Stage IIIA(N2) Non-Small Cell Lung Cancer [NCT02974426]Phase 31,094 participants (Anticipated)Interventional2016-11-30Recruiting
FurmOnertinib Mesylate With or Without Chemotherapy +/- bevacizUmab as firSt Line Treatment in Advanced Non-small Cell Lung Cancer Patients With Uncleared Epidermal Growth Factor Receptor (EGFR) Mutation Positive Circulating Tumor Cell DNA [NCT05334277]Phase 2280 participants (Anticipated)Interventional2022-05-06Recruiting
TIGER-3: A Phase 3, Open-label, Multicenter, Randomized Study of Oral Rociletinib (CO-1686) Monotherapy Versus Single-agent Cytotoxic Chemotherapy in Patients With Mutant EGFR Non-small Cell Lung Cancer (NSCLC) After Failure of at Least 1 Previous EGFR-di [NCT02322281]Phase 3149 participants (Actual)Interventional2015-02-28Terminated(stopped due to Sponsor discontinued development of CO-1686 for NSCLC)
A Randomized, Open-Label, Multicenter, Phase 3 Trial Comparing Veliparib Plus Carboplatin and Paclitaxel Versus Investigator's Choice of Standard Chemotherapy in Subjects Receiving First Cytotoxic Chemotherapy for Metastatic or Advanced Non-Squamous Non-S [NCT02264990]Phase 3595 participants (Actual)Interventional2014-09-30Completed
A Randomized, Open-Label, Phase 3 Study of Cosibelimab (CK-301) in Combination With Platinum+Pemetrexed Chemotherapy in Subjects With First-Line Metastatic Non-squamous Non-Small Cell Lung Cancer [NCT04786964]Phase 325 participants (Actual)Interventional2021-12-08Terminated(stopped due to Regional political conflict)
PHASE 3, RANDOMIZED, OPEN-LABEL STUDY OF THE EFFICACY AND SAFETY OF CRIZOTINIB VERSUS PEMETREXED/CISPLATIN OR PEMETREXED/CARBOPLATIN IN PREVIOUSLY UNTREATED EAST ASIAN PATIENTS WITH NON-SQUAMOUS CARCINOMA OF THE LUNG HARBORING A TRANSLOCATION OR INVERSION [NCT01639001]Phase 3207 participants (Actual)Interventional2012-09-29Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00063570 (5) [back to overview]Overall Tumor Response
NCT00063570 (5) [back to overview]Time to Treatment Failure
NCT00063570 (5) [back to overview]Time to Progressive Disease
NCT00063570 (5) [back to overview]Overall Survival
NCT00063570 (5) [back to overview]Duration of (Confirmed) Complete Response or Partial Response
NCT00087698 (6) [back to overview]Pathological Complete Response
NCT00087698 (6) [back to overview]Time to Progressive Disease
NCT00087698 (6) [back to overview]Overall Tumor Response
NCT00087698 (6) [back to overview]The 1 and 2 Year Disease-Free Survival Rate (Percentage)
NCT00087698 (6) [back to overview]Time to Treatment Failure
NCT00087698 (6) [back to overview]Overall Survival Time
NCT00095199 (8) [back to overview]Duration of Overall Response (OR)
NCT00095199 (8) [back to overview]Overall Survival (OS)
NCT00095199 (8) [back to overview]Number of Participants With Common Toxicity Criteria (CTC) Grade 3 or 4 Toxicities
NCT00095199 (8) [back to overview]Percentage of Participants With Symptomatic Response (Symptom Response Rates) Using the Lung Cancer Subscale (LCS) Scores of Functional Assessment of Cancer Therapy for Participants With Lung Cancer (FACT-L)
NCT00095199 (8) [back to overview]Progression Free Survival (PFS)
NCT00095199 (8) [back to overview]Proportion of Randomized Participants With Best Overall Response (OR) of Partial Response (PR), Complete Response (CR), or Stable Disease (SD)
NCT00095199 (8) [back to overview]Proportion of Randomized Participants With the Best Overall Response (OR) of Partial Response (PR) or Complete Response (CR) (Overall Response Rate [ORR])
NCT00095199 (8) [back to overview]Time to Symptomatic Progression
NCT00101283 (3) [back to overview]Overall Survival
NCT00101283 (3) [back to overview]Best Overall Response by RECIST Criteria (Version 1.0)
NCT00101283 (3) [back to overview]Progression-Free Survival
NCT00102804 (7) [back to overview]Time to Worsening of Symptoms (TWS)
NCT00102804 (7) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00102804 (7) [back to overview]Maximum Improvement Over Baseline in Individual Symptom Scores and Quality of Life Using the LCSS
NCT00102804 (7) [back to overview]Time to Objective Progressive Disease (TPD)
NCT00102804 (7) [back to overview]Progression-Free Survival (PFS) Time
NCT00102804 (7) [back to overview]Percentage of Participants With a Complete Response (CR) or Partial Response (PR) (Objective Tumor Response Rate)
NCT00102804 (7) [back to overview]Overall Survival (OS) Time
NCT00106002 (5) [back to overview]Duration of Tumor Response
NCT00106002 (5) [back to overview]Overall Survival Time
NCT00106002 (5) [back to overview]Progression-Free Survival Time
NCT00106002 (5) [back to overview]Overall Tumor Response
NCT00106002 (5) [back to overview]Toxicity Profile: Adverse Events (Common Terminology Criteria for Adverse Events, Grade 3 and 4, Present in >5% of Participants)
NCT00106626 (2) [back to overview]Safety and Tolerability as Measured by the Number of Participants With Disease Progression
NCT00106626 (2) [back to overview]Maximum Tolerated Dose (MTD) Status as Determined by Number of Participants With Dose Limiting Toxicity (DLT) at Each Dose Level
NCT00111839 (5) [back to overview]Number of Participants With Objective Response Assessed by Independent Review Committee
NCT00111839 (5) [back to overview]Duration of Objective Response Assessed by Independent Review Committee
NCT00111839 (5) [back to overview]Overall Survival (OS)
NCT00111839 (5) [back to overview]Change From Baseline to Cycle 2 in Global Quality of Life (QoL), as Assessed Using Lung Cancer Symptom Scale (LCSS)
NCT00111839 (5) [back to overview]Progression-Free Survival (PFS)
NCT00117962 (4) [back to overview]Overall Survival
NCT00117962 (4) [back to overview]Number of Participants With Overall Tumor Response
NCT00117962 (4) [back to overview]Failure-free Survival
NCT00117962 (4) [back to overview]18 Month Survival
NCT00149214 (5) [back to overview]Number of Patients With Histologically Negative Axillary Lymph Node Status at Surgery
NCT00149214 (5) [back to overview]Disease-free Survival
NCT00149214 (5) [back to overview]Number of Participants With a Pathological Complete Response
NCT00149214 (5) [back to overview]Number of Participants With a Clinical Tumor Response After the Second Sequence of Chemotherapy
NCT00149214 (5) [back to overview]Number of Participants With a Clinical Tumor Response After the First Sequence of Chemotherapy
NCT00190671 (8) [back to overview]Pharmacokinetics - Maximum Observed Drug Concentration (Cmax)
NCT00190671 (8) [back to overview]Pharmacokinetics - Half-Life (t½)
NCT00190671 (8) [back to overview]Pharmacokinetics - Clearance (CL)
NCT00190671 (8) [back to overview]Pharmacokinetics - Area Under the Curve (AUC)
NCT00190671 (8) [back to overview]Time to Progressive Disease
NCT00190671 (8) [back to overview]Best Tumor Response
NCT00190671 (8) [back to overview]Pharmacokinetics - Volume of Distribution
NCT00190671 (8) [back to overview]Progression Free Survival
NCT00190983 (4) [back to overview]Tumor Response
NCT00190983 (4) [back to overview]Progression-Free Survival
NCT00190983 (4) [back to overview]Overall Survival
NCT00190983 (4) [back to overview]Duration of Response
NCT00191191 (5) [back to overview]Duration of Response
NCT00191191 (5) [back to overview]Best Overall Response
NCT00191191 (5) [back to overview]Change From Baseline to 3 Months in Quality of Life Questionnaire for Cancer Patients Treated With Anticancer Drugs (QOL-ACD)
NCT00191191 (5) [back to overview]Change From Baseline to 3 Months in Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) Lung Cancer Subscale (LCS)
NCT00191191 (5) [back to overview]Progression-Free Survival (PFS)
NCT00191308 (4) [back to overview]Overall Survival (OS)
NCT00191308 (4) [back to overview]Disease Free Survival (DFS)
NCT00191308 (4) [back to overview]Duration of Response
NCT00191308 (4) [back to overview]Percentage of Participants With Objective Tumor Response (Response Rate)
NCT00191984 (5) [back to overview]Overall Survival
NCT00191984 (5) [back to overview]Best Overall Tumor Response
NCT00191984 (5) [back to overview]Time to Treatment Failure
NCT00191984 (5) [back to overview]Progression-Free Survival (PFS)
NCT00191984 (5) [back to overview]Duration of Response
NCT00193414 (3) [back to overview]Progression-free Survival (PFS)
NCT00193414 (3) [back to overview]Overall Survival (OS)
NCT00193414 (3) [back to overview]Overall Response Rate
NCT00198133 (3) [back to overview]Duration of Remission
NCT00198133 (3) [back to overview]Grade 3/4 Treatment Related Adverse Events
NCT00198133 (3) [back to overview]Objective Response Rate (Complete and Partial Response)
NCT00203931 (5) [back to overview]Progression-free Survival Based on Serum Biomarker Status
NCT00203931 (5) [back to overview]Progression-free Survival Based on Rash Development
NCT00203931 (5) [back to overview]Progression-free Survival
NCT00203931 (5) [back to overview]Overall Survival
NCT00203931 (5) [back to overview]Objective Response Rate
NCT00216099 (8) [back to overview]Overall Survival
NCT00216099 (8) [back to overview]Rate of Clinical Benefit
NCT00216099 (8) [back to overview]Time to Progression
NCT00216099 (8) [back to overview]Safety and Tolerability
NCT00216099 (8) [back to overview]Time to Prostate-Specific Antigen (PSA)/Serological Progression
NCT00216099 (8) [back to overview]Best Overall PSA Response
NCT00216099 (8) [back to overview]OBJECTIVE Overall Response Rate
NCT00216099 (8) [back to overview]RFC1 G80A Genotype
NCT00216203 (4) [back to overview]Toxicity and Safety Profile
NCT00216203 (4) [back to overview]Median Survival Time
NCT00216203 (4) [back to overview]Time To Progression (TTP)
NCT00216203 (4) [back to overview]Maximum Tolerated Dose (MTD) of Pemetrexed in Combination With Cetuximab
NCT00222729 (4) [back to overview]Disease Control Rate (DCR)
NCT00222729 (4) [back to overview]Time-to-progression (TTP)
NCT00222729 (4) [back to overview]Objective Response Rate (ORR)
NCT00222729 (4) [back to overview]Overall Survival (OS)
NCT00226577 (5) [back to overview]Toxicity
NCT00226577 (5) [back to overview]Survival - Overall
NCT00226577 (5) [back to overview]Disease Response - Radiographic
NCT00226577 (5) [back to overview]Disease Response - Pathologic
NCT00226577 (5) [back to overview]Survival - Disease Free
NCT00227019 (3) [back to overview]Incidence of Central Nervous System (CNS) Hemorrhagic Events
NCT00227019 (3) [back to overview]Overall Survival (OS)
NCT00227019 (3) [back to overview]Progression-free Survival (PFS)
NCT00227539 (3) [back to overview]Safety of Neoadjuvant Chemotherapy
NCT00227539 (3) [back to overview]Positron Emission Tomography as a Predictor of Response Measured by the Decrease in Standard Uptake Variable (SUV) After 1 Course of Therapy
NCT00227539 (3) [back to overview]Efficacy of Neoadjuvant Chemotherapy as Measured by Radiologic Response Rate
NCT00234052 (7) [back to overview]Overall Survival Rate at 6, 12, 18, and 24 Months
NCT00234052 (7) [back to overview]Overall Survival Rate
NCT00234052 (7) [back to overview]Overall Response Rate
NCT00234052 (7) [back to overview]Duration of Response
NCT00234052 (7) [back to overview]Median Progression Free Survival
NCT00234052 (7) [back to overview]Toxicity of Carboplatin, Pemetrexed and Bevacizumab Combination Treatment
NCT00234052 (7) [back to overview]Progression Free Survival at 6, 12, 18, 24 Months
NCT00248495 (4) [back to overview]Percent Change in SUV Level Between Pre and Post Chemotherapy
NCT00248495 (4) [back to overview]Pathologically Complete Response
NCT00248495 (4) [back to overview]Overall Survival
NCT00248495 (4) [back to overview]Disease Free Survival
NCT00259285 (2) [back to overview]Treatment Response
NCT00259285 (2) [back to overview]Pathologic Remissions After Surgery
NCT00265785 (4) [back to overview]Number of Patients With Grade 3 Through 5 Adverse Events That Are Related to Study Drug
NCT00265785 (4) [back to overview]Response (Confirmed and Unconfirmed, Complete and Partial)
NCT00265785 (4) [back to overview]Progression-free Survival
NCT00265785 (4) [back to overview]Overall Survival
NCT00268437 (2) [back to overview]Pathologic Complete Response Rate
NCT00268437 (2) [back to overview]Overall Survival
NCT00269152 (5) [back to overview]The Feasibility of Post-Surgery Chemotherapy
NCT00269152 (5) [back to overview]Overall Survival at 6 Years
NCT00269152 (5) [back to overview]Grade III/IV Adverse Events
NCT00269152 (5) [back to overview]Overall Survival at 3 Years
NCT00269152 (5) [back to overview]3 Year Disease-Free Survival: Probability of Disease-Free Survival at 3 Years
NCT00280748 (4) [back to overview]Neurological Function by Radiation Oncology Group (RTOG) Neurological Function Classification
NCT00280748 (4) [back to overview]Neurological Function by Mini Mental State Examination
NCT00280748 (4) [back to overview]Estimate the Overall Survival of Patients Treated With This Regimen.
NCT00280748 (4) [back to overview]Number of Subjects Experiencing Adverse Events
NCT00293579 (4) [back to overview]Overall Response Rate
NCT00293579 (4) [back to overview]Impact of Pemetrexed Chemotherapy on Quality of Life
NCT00293579 (4) [back to overview]Toxicities of Pemetrexed,in Poor Risk Cases With Poor Performance Status and Advanced, Metastatic, or Recurrent Head and Neck Cancer
NCT00293579 (4) [back to overview]Overall Survival
NCT00295503 (3) [back to overview]Progression Free Survival Rate at 6 Months
NCT00295503 (3) [back to overview]Overall Survival
NCT00295503 (3) [back to overview]Response Rate
NCT00301808 (4) [back to overview]Safety Outcomes
NCT00301808 (4) [back to overview]Overall Survival
NCT00301808 (4) [back to overview]Probability of Overall Survival at One Year
NCT00301808 (4) [back to overview]Progression-free Survival
NCT00308750 (8) [back to overview]Duration of CR or PR (Duration of Response)
NCT00308750 (8) [back to overview]Change From Baseline in Total Functional Assessment of Cancer Therapy -Taxane (FACT-Taxane) Scale
NCT00308750 (8) [back to overview]Overall Survival (OS)
NCT00308750 (8) [back to overview]Time to Disease Progression
NCT00308750 (8) [back to overview]Number of Participants With Adverse Events (AEs) or Deaths
NCT00308750 (8) [back to overview]Time-to-Treatment Failure (TTF)
NCT00308750 (8) [back to overview]Change From Baseline in Total Functional Assessment of Cancer Therapy-Lung (FACT-L) Scale
NCT00308750 (8) [back to overview]Number of Participants With Complete Response (CR) or Partial Response (PR) [Tumor Response]
NCT00310050 (3) [back to overview]Patterns of Response
NCT00310050 (3) [back to overview]Qualitative Dose-limiting Toxicities of Pemetrexed in Combination With Radiation Therapy
NCT00310050 (3) [back to overview]Number of Participants That Survived
NCT00316225 (6) [back to overview]Pemetrexed Population Pharmacokinetics (PK): Clearance
NCT00316225 (6) [back to overview]Overall Tumor Response
NCT00316225 (6) [back to overview]Overview of Adverse Events
NCT00316225 (6) [back to overview]Pemetrexed Population Pharmacokinetics: Volume of Distribution
NCT00316225 (6) [back to overview]Discontinuations Due to Adverse Events
NCT00316225 (6) [back to overview]Number of Participants With Common Toxicity Criteria - National Cancer Institute Grade 3 and Grade 4 Toxicities
NCT00320515 (4) [back to overview]Objective Best Tumor Response
NCT00320515 (4) [back to overview]Progression Free Survival
NCT00320515 (4) [back to overview]Overall Survival
NCT00320515 (4) [back to overview]Duration of Response
NCT00324805 (2) [back to overview]Disease-free Survival
NCT00324805 (2) [back to overview]Overall Survival
NCT00325234 (6) [back to overview]Time To Treatment Failure (TTTF)
NCT00325234 (6) [back to overview]Time to Response
NCT00325234 (6) [back to overview]Time to Progressive Disease (PD)
NCT00325234 (6) [back to overview]Duration of Response (DOR)
NCT00325234 (6) [back to overview]Tumor Response Rate
NCT00325234 (6) [back to overview]Number of Participants With Adverse Events (AE)
NCT00330915 (4) [back to overview]Number of Participants With Complete Tumor Resection
NCT00330915 (4) [back to overview]Number of Participants Receiving Sphincter Saving Surgery
NCT00330915 (4) [back to overview]Pathological Complete Response (pCR)
NCT00330915 (4) [back to overview]Feasibility of Pemetrexed Prior to Surgery
NCT00350792 (5) [back to overview]Estimated Probability of One Year Progression-free Survival
NCT00350792 (5) [back to overview]Overall Survival
NCT00350792 (5) [back to overview]Percentage of Participants With a Complete or Partial Tumor Response (Overall Tumor Response)
NCT00350792 (5) [back to overview]Time to Treatment Failure
NCT00350792 (5) [back to overview]Time to Treatment Failure
NCT00351039 (1) [back to overview]Number of Participants With Grade 3 and Grade 4 Adverse Events
NCT00356525 (2) [back to overview]Objective Tumor Response
NCT00356525 (2) [back to overview]Overall Survival
NCT00363415 (5) [back to overview]Overall Survival
NCT00363415 (5) [back to overview]Progression Free Survival
NCT00363415 (5) [back to overview]Change From Baseline to Each Cycle in Functional Assessment of Cancer Therapy - Lung (FACT-L)
NCT00363415 (5) [back to overview]Number of Participants in Subgroups: LDH<=Upper Limit of Normal and History of Brain Metastases=Yes
NCT00363415 (5) [back to overview]Overall Survival (Subgroups)
NCT00369629 (1) [back to overview]Maximum Tolerated Dose as Measured by the Number of Dose Limiting Toxicities Seen in Cohort.
NCT00370292 (3) [back to overview]Mean Deoxycytidine Kinase (dCK) Expression Evaluated at Cycle 1, Cycle 2, and Cycle 3
NCT00370292 (3) [back to overview]Mean Human Equilibrative Nucleoside Transporter 1 (hENT) Expression Evaluated at Cycle 1, Cycle 2, and Cycle 3
NCT00370292 (3) [back to overview]Best Objective Tumor Response
NCT00374868 (8) [back to overview]Duration of Response
NCT00374868 (8) [back to overview]Duration of Stable Disease
NCT00374868 (8) [back to overview]Progression-Free Survival
NCT00374868 (8) [back to overview]Overall Survival
NCT00374868 (8) [back to overview]Time to Progressive Disease
NCT00374868 (8) [back to overview]Best Overall Tumor Response
NCT00374868 (8) [back to overview]Time to Treatment Failure (TTF)
NCT00374868 (8) [back to overview]Time to Response
NCT00377520 (2) [back to overview]Tumor Response
NCT00377520 (2) [back to overview]Number of Participants With Adverse Events by Grade (Measures of Toxicity)
NCT00380718 (7) [back to overview]Duration of Response
NCT00380718 (7) [back to overview]Proportion of Participants With a Complete or Partial Response (Objective Response Rate [ORR])
NCT00380718 (7) [back to overview]Overall Survival
NCT00380718 (7) [back to overview]Progression-Free Survival (PFS)
NCT00380718 (7) [back to overview]Proportion of Participants With a Best Overall Response of Complete Response (CR), Partial Response (PR), and Stable Disease (SD) (Disease Control Rate [DCR])
NCT00380718 (7) [back to overview]Time to Tumor Progression
NCT00380718 (7) [back to overview]Time to Treatment Failure
NCT00383266 (6) [back to overview]Overall Survival Rate
NCT00383266 (6) [back to overview]Overall Survival Rate
NCT00383266 (6) [back to overview]Toxicities
NCT00383266 (6) [back to overview]Overall Survival (OS)
NCT00383266 (6) [back to overview]Time to Disease Progression
NCT00383266 (6) [back to overview]Overall Response Rate (ORR)
NCT00383331 (3) [back to overview]Progression Free Survival
NCT00383331 (3) [back to overview]Overall Survival
NCT00383331 (3) [back to overview]Best Overall Tumor Response
NCT00389805 (5) [back to overview]Number of Patients With Grade ≥ 3 Toxicity (Phase I)
NCT00389805 (5) [back to overview]Number of Patients With Toxicity by NCI CTC v3.0 (Phase I)
NCT00389805 (5) [back to overview]Number of Patients Experiencing a Dose-limiting Toxicity (Phase I)
NCT00389805 (5) [back to overview]Number of Participants Who Experience Adverse Events (Phase I)
NCT00389805 (5) [back to overview]Maximum Tolerated Dose of Bortezomib in Combination With Pemetrexel (Phase I)
NCT00391274 (5) [back to overview]Pharmacology Toxicity
NCT00391274 (5) [back to overview]Overall Survival
NCT00391274 (5) [back to overview]Progression-Free Survival (PFS)
NCT00391274 (5) [back to overview]Number of Patients With Disease Progression
NCT00391274 (5) [back to overview]Overall Tumor Response
NCT00391586 (1) [back to overview]Toxicity Profile
NCT00394147 (3) [back to overview]Overall Survival
NCT00394147 (3) [back to overview]Time to Progression
NCT00394147 (3) [back to overview]Objective Response Rate
NCT00402051 (5) [back to overview]Percentage of Participants Surviving Progression-Free at 6 Months (Progression Free Survival [PFS] Rate)
NCT00402051 (5) [back to overview]Time to Treatment Failure (TTF)
NCT00402051 (5) [back to overview]Number of Participants With Tumor Response (as Basis for Response Rate)
NCT00402051 (5) [back to overview]Overall Survival
NCT00402051 (5) [back to overview]Pharmacology Toxicities
NCT00407550 (4) [back to overview]Number of Patients With Confirmed Responses
NCT00407550 (4) [back to overview]Adverse Event
NCT00407550 (4) [back to overview]Overall Survival
NCT00407550 (4) [back to overview]Progression-free Survival
NCT00409006 (4) [back to overview]Percentage of Participants Who Died During the Study
NCT00409006 (4) [back to overview]Number of Participants With Tumor Response
NCT00409006 (4) [back to overview]Duration of Response for Responders
NCT00409006 (4) [back to overview]Progression-Free Survival (PFS)
NCT00410904 (3) [back to overview]Response Rate (Complete and Partial) 2 Separate Cohorts of Relapsed NSCLC Cohort A: Pts Who Have Received Prior Chemo w/o Ever Having Received Bevacizumab. Cohort B: Pts Who Have Received Prior Bevacizumab.
NCT00410904 (3) [back to overview]Progression-free Survival
NCT00410904 (3) [back to overview]Overall Survival
NCT00415168 (7) [back to overview]Percentage of Participants With Objective Response (Objective Response Rate)
NCT00415168 (7) [back to overview]Number of Participants With Pharmacology Toxicity - Grade 3 or 4 Non-Laboratory Toxicity Possibly Related to Study Therapy
NCT00415168 (7) [back to overview]Number of Participants With Pharmacology Toxicity - Grade 3 or 4 Laboratory Toxicity Possibly Related to Study Therapy
NCT00415168 (7) [back to overview]Number of Participants Who Died During the Study
NCT00415168 (7) [back to overview]Progression Free Survival (PFS)
NCT00415168 (7) [back to overview]Overall Survival
NCT00415168 (7) [back to overview]Duration of Response
NCT00415194 (5) [back to overview]Time to Treatment Worsening in Functional Assessment of Cancer Therapy - Head and Neck Cancer (FACT-H&N) Total Score
NCT00415194 (5) [back to overview]Progression-free Survival (PFS)
NCT00415194 (5) [back to overview]Percent of Participants With a Tumor Response (Response Rate)
NCT00415194 (5) [back to overview]Overall Survival (OS)
NCT00415194 (5) [back to overview]Duration of Response (DoR)
NCT00415636 (4) [back to overview]Pharmacokinetic (PK) Parameter: Area Under the IC83/LY2603618 Plasma Concentration Versus Time Curve From Time Zero to Infinity (AUC[0-∞])
NCT00415636 (4) [back to overview]Pharmacokinetic (PK) Parameter: Maximum Observed Plasma Concentration (Cmax) of IC83/LY2603618
NCT00415636 (4) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00415636 (4) [back to overview]Percentage of Participants With Best Overall Response
NCT00418886 (10) [back to overview]Objective Response Rate (ORR)
NCT00418886 (10) [back to overview]Time to Deterioration of Disease-related Symptoms (TDS) by Lung Cancer Symptom Scale (LCSS) Total Score
NCT00418886 (10) [back to overview]Time to Deterioration of Disease-related Symptoms (TDS) by Average Symptom Burden Index (ASBI) Score
NCT00418886 (10) [back to overview]Progression-Free Survival (PFS) in the Overall Population
NCT00418886 (10) [back to overview]Overall Survival (OS)
NCT00418886 (10) [back to overview]Progression-Free Survival (PFS) in the Female Population
NCT00418886 (10) [back to overview]Longitudinal Analysis of Average Symptom Burden Index (ASBI) Score
NCT00418886 (10) [back to overview]Duration of Response (DoR)
NCT00418886 (10) [back to overview]Disease Control Rate (DCR)
NCT00418886 (10) [back to overview]Longitudinal Analysis of Lung Cancer Symptom Scale (LCSS) Total Score
NCT00438204 (5) [back to overview]Time to Treatment Failure
NCT00438204 (5) [back to overview]Overall Survival
NCT00438204 (5) [back to overview]Progression-free Survival (PFS)
NCT00438204 (5) [back to overview]Number of Participants With Grade 3 or Grade 4 Toxicity
NCT00438204 (5) [back to overview]Number of Participants With Response
NCT00447057 (7) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00447057 (7) [back to overview]Time to Treatment Failure (TTTF)
NCT00447057 (7) [back to overview]Percentage of Participants With Best Response of Complete Response (CR) or Partial Response (PR) (Response Rate)
NCT00447057 (7) [back to overview]Percentage of Participants With Best Response of Stable Disease (SD), Partial Response (PR) or Complete Response (CR) (Disease Control Rate)
NCT00447057 (7) [back to overview]Progression Free Survival (PFS)
NCT00447057 (7) [back to overview]Overall Survival (OS)
NCT00447057 (7) [back to overview]Percentage of Participants Surviving at 1 Year
NCT00454194 (6) [back to overview]Time to Treatment Failure
NCT00454194 (6) [back to overview]Confirmed Response Rate (Complete Response and Partial Response) as Assessed by Response Evaluation Criteria in Solid Tumors (RECIST)
NCT00454194 (6) [back to overview]Progression-free Survival
NCT00454194 (6) [back to overview]Overall Survival
NCT00454194 (6) [back to overview]Number of Participants With at Least One Grade 3 or Above Adverse Events Assessed by NCI CTCAE v4.0
NCT00454194 (6) [back to overview]Duration of Response
NCT00454649 (34) [back to overview]Plasma Clearance (CL) for Gemcitabine
NCT00454649 (34) [back to overview]Plasma Clearance (CL) for Paclitaxel
NCT00454649 (34) [back to overview]Plasma Clearance (CL) for Pemetrexed
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Axitinib (AG-013736)
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Capecitabine
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Cisplatin
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Docetaxel
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Carboplatin
NCT00454649 (34) [back to overview]Plasma Clearance (CL) for Cisplatin
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Gemcitabine
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Gemcitabine
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Paclitaxel
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Pemetrexed
NCT00454649 (34) [back to overview]Maximum Tolerated Dose (MTD) of Axitinib (AG-013736) in Combination With Chemotherapy
NCT00454649 (34) [back to overview]Percentage of Participants With Objective Response
NCT00454649 (34) [back to overview]Plasma Clearance (CL) for Carboplatin
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Docetaxel
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Cisplatin
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Carboplatin
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Pemetrexed
NCT00454649 (34) [back to overview]Plasma Decay Half Life (t1/2) for Paclitaxel
NCT00454649 (34) [back to overview]Plasma Clearance (CL) for Docetaxel
NCT00454649 (34) [back to overview]Apparent Oral Clearance (CL/F) for Axitinib (AG-013736)
NCT00454649 (34) [back to overview]Apparent Oral Clearance (CL/F) for Capecitabine
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Carboplatin
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Docetaxel
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Gemcitabine
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Paclitaxel
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Pemetrexed
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Time 24 Hours [AUC (0-24)] for Axitinib (AG-013736)
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Time 24 Hours [AUC (0-24)] for Capecitabine
NCT00454649 (34) [back to overview]Area Under the Curve From Time Zero to Time 8 Hours [AUC (0-8)] for Cisplatin
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Axitinib (AG-013736)
NCT00454649 (34) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Capecitabine
NCT00456261 (3) [back to overview]Overall Survival (OS), the Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Death
NCT00456261 (3) [back to overview]Time to Progression (TTP), the Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Worsening of Their Disease
NCT00456261 (3) [back to overview]Overall Response Rate (ORR), the Percentage of Patients Who Experience an Objective Benefit From Treatment
NCT00461786 (5) [back to overview]Duration of Response
NCT00461786 (5) [back to overview]Tumor Response
NCT00461786 (5) [back to overview]Number of Participants With Adverse Events by Grade
NCT00461786 (5) [back to overview]Progression-Free Survival
NCT00461786 (5) [back to overview]Overall Survival
NCT00470548 (7) [back to overview]Duration of Overall Survival
NCT00470548 (7) [back to overview]Number of Participants With Complete Response
NCT00470548 (7) [back to overview]Number of Participants With Disease Control
NCT00470548 (7) [back to overview]Number of Participants With Dose Limiting Toxicities
NCT00470548 (7) [back to overview]Number of Participants With Stable Disease
NCT00470548 (7) [back to overview]Number of Patients With Toxicities
NCT00470548 (7) [back to overview]Number of Participants With Partial Response
NCT00482014 (9) [back to overview]Phase 1 - Percentage of Participants With Complete Response or Partial Response (Response Rate)
NCT00482014 (9) [back to overview]Phase 1 - Pharmacology Toxicity: Number of Participants With Dose Limiting Toxicities (DLTs)
NCT00482014 (9) [back to overview]Phase 1 - Maximum Tolerated Dose (MTD) of Cisplatin
NCT00482014 (9) [back to overview]Phase 2 - Median Survival
NCT00482014 (9) [back to overview]Phase 2 - Survival Probability at 2 Years
NCT00482014 (9) [back to overview]Phase 2 - Percentage of Participants With Complete Response or Partial Response (Response Rate)
NCT00482014 (9) [back to overview]Phase 2 - Pharmacology Toxicity: Number of Participants With Adverse Events
NCT00482014 (9) [back to overview]Phase 1 - Maximum Tolerated Dose (MTD) of Carboplatin
NCT00482014 (9) [back to overview]Phase 2 - Time to Progression
NCT00487669 (3) [back to overview]To Evaluate the Overall Response Rate (Complete Plus Partial Responses by RECIST Criteria) to the Combination of Paclitaxel Poliglumex and Pemetrexed as Therapy in Patients With Advanced NSCLC.
NCT00487669 (3) [back to overview]Time to Progression
NCT00487669 (3) [back to overview]Overall Survival
NCT00489359 (12) [back to overview]Phase 1 - Number of Participants With Adverse Events (Toxicity)
NCT00489359 (12) [back to overview]Phase 1 - Number of Participants With Tumor Response
NCT00489359 (12) [back to overview]Phase 2 - Number of Participants With Adverse Events (Toxicity)
NCT00489359 (12) [back to overview]Phase 2 - Percentage of Participants With Overall Tumor Response (Response Rate)
NCT00489359 (12) [back to overview]Phase 2 - Progression-Free Survival
NCT00489359 (12) [back to overview]Phase 2 - Time to Disease Progression
NCT00489359 (12) [back to overview]Phase 2 - Time to Response (TTR)
NCT00489359 (12) [back to overview]Phase 1 - Number of Dose-Limiting Toxicities (DLTs)
NCT00489359 (12) [back to overview]Phase 1 - Recommended Area Under the Curve (AUC) Dose of Carboplatin for Phase 2
NCT00489359 (12) [back to overview]Phase 1 - Recommended Dose of Pemetrexed for Phase 2
NCT00489359 (12) [back to overview]Phase 2 - Time to Treatment Failure
NCT00489359 (12) [back to overview]Phase 2 - Duration of Response (DOR)
NCT00491075 (1) [back to overview]Overall Response
NCT00494026 (1) [back to overview]Pharmacology Toxicity
NCT00497770 (11) [back to overview]Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Drive or Use Public Transportation
NCT00497770 (11) [back to overview]Percentage of Participants With a Best Overall Disease Control Response (Disease Control Rate)
NCT00497770 (11) [back to overview]Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Prepare and Take Medications
NCT00497770 (11) [back to overview]Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Handle Personal Finances
NCT00497770 (11) [back to overview]Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Operate the Telephone
NCT00497770 (11) [back to overview]Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to do the Act of Shopping
NCT00497770 (11) [back to overview]Progression Free Survival
NCT00497770 (11) [back to overview]Overall Survival
NCT00497770 (11) [back to overview]Functional Status Based on Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Plan and Prepare Meals
NCT00497770 (11) [back to overview]Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Perform Housework Activities
NCT00497770 (11) [back to overview]Symptom Score Associated With Treatment as Measured by the M.D. Anderson Symptom Inventory - LC(MDASI-LC)
NCT00503997 (1) [back to overview]Patient Response to Treatment Measured by RECIST Criteria
NCT00508144 (1) [back to overview]Objective Response Rate (OR) Where OR=CR+PR: Number of Participants With Responses of Complete Response (CR) and Partial Response (PR)
NCT00509366 (3) [back to overview]1-year Progression Free Survival Rate in Chemo-naive Select Stage IIIB or Stage IV NSCLC Patients
NCT00509366 (3) [back to overview]Mean Change From Baseline to Follow-up Cycle in Quality of Life - Functional Assessment of Cancer Therapy-Lung (FACT-L)
NCT00509366 (3) [back to overview]Median Time to Progressive Disease
NCT00517595 (6) [back to overview]Overall Survival (OS)
NCT00517595 (6) [back to overview]Progression Free Survival (PFS) by Baseline Eastern Cooperative Oncology Group (ECOG) Performance Status
NCT00517595 (6) [back to overview]Progression Free Survival (PFS)
NCT00517595 (6) [back to overview]Time to Progression (TTP)
NCT00517595 (6) [back to overview]Overall Response
NCT00517595 (6) [back to overview]Overall Survival (OS) by Baseline Eastern Cooperative Oncology Group (ECOG) Performance Status
NCT00520676 (8) [back to overview]Progression-free Survival (PFS)
NCT00520676 (8) [back to overview]Survival Without Clinically Important Grade 3 or 4 Toxicity
NCT00520676 (8) [back to overview]Survival Without Grade 3 or 4 Toxicity
NCT00520676 (8) [back to overview]Survival Without Grade 4 Toxicity
NCT00520676 (8) [back to overview]Overall Survival (OS)
NCT00520676 (8) [back to overview]Percentage of Participants With Tumor Response (Response Rate)
NCT00520676 (8) [back to overview]Duration of Response
NCT00520676 (8) [back to overview]Number of Participants With Adverse Events (AEs)
NCT00520845 (3) [back to overview]Time to Progression
NCT00520845 (3) [back to overview]Overall Response Rate
NCT00520845 (3) [back to overview]Median Survival
NCT00520936 (2) [back to overview]Number of Patients With Adverse Events, Discontinuations, or Deaths Possibly Due to Study Drug
NCT00520936 (2) [back to overview]Percentage of Participants With Overall Tumor Response (Response Rate)
NCT00523419 (5) [back to overview]Duration of Response
NCT00523419 (5) [back to overview]Progression-Free Survival (PFS)
NCT00523419 (5) [back to overview]Overall Survival (OS) Time
NCT00523419 (5) [back to overview]Number of Participants With Adverse Events (Pharmacology Toxicity)
NCT00523419 (5) [back to overview]Percentage of Participants With Tumor Response
NCT00529100 (9) [back to overview]Phase 2: Percentage of Participants With Overall Survival (OS) at 1 Year
NCT00529100 (9) [back to overview]Phase 2: Time to Progressive Disease (PD)
NCT00529100 (9) [back to overview]Phase 1: Maximum Tolerated Dose (MTD) of Pemetrexed in Combination With Cisplatin and Radiation Therapy
NCT00529100 (9) [back to overview]Phase 1: Number of Participants With Adverse Events (AE; Toxicity)
NCT00529100 (9) [back to overview]Phase 2: Site of Progressive Disease (PD)
NCT00529100 (9) [back to overview]Phase 2: Percentage of Participants With Progression Free Survival (PFS)
NCT00529100 (9) [back to overview]Phase 2: Percentage of Participants With Overall Survival (OS) at 2 Years and 3 Years
NCT00529100 (9) [back to overview]Progression Free Survival (PFS)
NCT00529100 (9) [back to overview]Phase 2: Percentage of Participants With Objective Tumor Response (Response Rate)
NCT00530621 (8) [back to overview]Progression-Free Survival (PFS)
NCT00530621 (8) [back to overview]Number of Participants Who Died During the 30 Days After Treatment Discontinuation
NCT00530621 (8) [back to overview]Number of Participants Who Died During the Study Treatment
NCT00530621 (8) [back to overview]Tumor Biomarkers
NCT00530621 (8) [back to overview]Time-to-Worsening (TW) in Lung Cancer Symptom Scale (LCSS) - Health Related Quality of Life (HRQoL) Subscale
NCT00530621 (8) [back to overview]Duration of Disease Control (DDC)
NCT00530621 (8) [back to overview]Overall Survival (OS)
NCT00530621 (8) [back to overview]Percentage of Participants With Complete Response or Partial Response (Tumor Response Rate)
NCT00533429 (6) [back to overview]Duration of Response (DoR)
NCT00533429 (6) [back to overview]Overall Survival (OS)
NCT00533429 (6) [back to overview]Pharmacology Toxicity and Adverse Events (AEs)
NCT00533429 (6) [back to overview]Time to Progressive Disease (TTPD)
NCT00533429 (6) [back to overview]Progression-Free Survival (PFS)
NCT00533429 (6) [back to overview]Percentage of Participants With Complete Response (CR) or Partial Response (PR) (Response Rate)
NCT00538681 (2) [back to overview]Part 1: Evaluate Safety [Toxicity, Serious Adverse Events (SAEs) and Reasons for Participant's Discontinuation]
NCT00538681 (2) [back to overview]Part 2: To Evaluate the Safety and Toxicity Profile of Study Treatments
NCT00545948 (3) [back to overview]Percentage of Patients With Completely Resected NSCLC Tumors That Can Be Analyzed and Used to Direct Adjuvant Chemotherapy
NCT00545948 (3) [back to overview]2-Year Progression-Free Survival Rate in Patients With Completely Resected Stage IB, II, or IIIA NSCLC
NCT00545948 (3) [back to overview]2-Year Overall Survival in Patients Treated for NSCLC
NCT00550173 (8) [back to overview]Number of Participants With Adverse Events
NCT00550173 (8) [back to overview]Number of Participants With Mutated or Non-Mutated Epidermal Growth Factor Receptor (EGFR) Genotype Status
NCT00550173 (8) [back to overview]Overall Survival (OS)
NCT00550173 (8) [back to overview]Time to Worsening of Symptoms (TWS) on Lung Cancer Symptoms Scale (LCSS)
NCT00550173 (8) [back to overview]Percentage of Participants With a Tumor Response of Complete Response (CR) or Partial Response (PR) [Tumor Response Rate (TRR)]
NCT00550173 (8) [back to overview]Percentage of Participants With CR, PR, and Stable Disease (SD) - Disease Control Rate (DCR)
NCT00550173 (8) [back to overview]Progression-Free Survival (PFS)
NCT00550173 (8) [back to overview]Probability of OS at 12 Months
NCT00560573 (16) [back to overview]Recommended Phase 2 Dose (RP2D)
NCT00560573 (16) [back to overview]Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Figitumumab
NCT00560573 (16) [back to overview]Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Cisplatin
NCT00560573 (16) [back to overview]Percentage of Participants With Blood Anti-drug Antibody (ADA) Specific for Figitumumab
NCT00560573 (16) [back to overview]Progression-Free Survival (PFS)
NCT00560573 (16) [back to overview]Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Gemcitabine
NCT00560573 (16) [back to overview]Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Pemetrexed
NCT00560573 (16) [back to overview]Concentration at the End of Infusion (Cinf) for Figitumumab
NCT00560573 (16) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Cisplatin
NCT00560573 (16) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Gemcitabine
NCT00560573 (16) [back to overview]Maximum Observed Plasma Concentration (Cmax) for Pemetrexed
NCT00560573 (16) [back to overview]Maximum Tolerated Dose (MTD)
NCT00560573 (16) [back to overview]Minimum Observed Plasma Trough Concentration (Cmin) for Figitumumab
NCT00560573 (16) [back to overview]Number of Participants With Dose-limiting Toxicities (DLT)
NCT00560573 (16) [back to overview]Percentage of Participants With Objective Response or Prolonged Stabilization
NCT00560573 (16) [back to overview]Serum Total Circulating Insulin-like Growth Factor (IGF-1) Levels
NCT00573989 (10) [back to overview]Overall Survival
NCT00573989 (10) [back to overview]Median Overall Survival
NCT00573989 (10) [back to overview]Maximum Tolerated Dose of Erlotinib Hydrochloride (Phase I)
NCT00573989 (10) [back to overview]Progression-free Survival (PFS) at 1 Year (Phase II)
NCT00573989 (10) [back to overview]Change in Quality of Life- FACT H&N
NCT00573989 (10) [back to overview]Objective Tumor Response
NCT00573989 (10) [back to overview]Evaluation of Acute and Chronic Toxicity
NCT00573989 (10) [back to overview]Median Progression Free Survival
NCT00573989 (10) [back to overview]Change in Quality of Life: MDADI
NCT00573989 (10) [back to overview]Change in Quality of Life: PSS-HN
NCT00577707 (1) [back to overview]Number of Patients With Pathologic Complete Response Rate
NCT00589667 (2) [back to overview]Median Overall Survival
NCT00589667 (2) [back to overview]Overall Objective Response
NCT00604461 (2) [back to overview]Number of Participants With Partial Response (PR) of Target Lesions
NCT00604461 (2) [back to overview]Number of Months of Progression Free Survival (PFS)
NCT00606021 (6) [back to overview]Progression Free Survival During Overall Period (Induction Phase [IP] + Maintenance Phase [MP])
NCT00606021 (6) [back to overview]Tumor Response Rate and Disease Control Rate After Induction Phase (IP)
NCT00606021 (6) [back to overview]Number of Participants With Adverse Events (AEs) During Overall Period
NCT00606021 (6) [back to overview]Progression Free Survival During Maintenance Phase
NCT00606021 (6) [back to overview]Overall Survival During Overall Period (IP + MP)
NCT00606021 (6) [back to overview]Overall Survival During Maintenance Phase
NCT00609518 (4) [back to overview]Progression-free Survival (PFS)
NCT00609518 (4) [back to overview]Safety: Number of Participants With Drug-Related Grade 3 or 4 Toxicity
NCT00609518 (4) [back to overview]Overall Survival
NCT00609518 (4) [back to overview]Proportion of Participants With Best Overall Tumor Response (Response Rate)
NCT00614822 (4) [back to overview]Progression Free Survival
NCT00614822 (4) [back to overview]Overall Survival
NCT00614822 (4) [back to overview]Number of Participants With Complete and Partial Tumor Responses
NCT00614822 (4) [back to overview]Number of Participants With Adverse Events
NCT00660816 (4) [back to overview]Disease Stabilization Rate (e.g., Complete Response, Partial Response, and Stable Disease)
NCT00660816 (4) [back to overview]Overall Survival
NCT00660816 (4) [back to overview]Progression-free Survival
NCT00660816 (4) [back to overview]Response Rate
NCT00686959 (7) [back to overview]Survival Rates at 1, 2, and 3 Years
NCT00686959 (7) [back to overview]First Site of Disease Failure in Terms of Relapse
NCT00686959 (7) [back to overview]Adverse Events: The Number of Deaths Per Treatment Group
NCT00686959 (7) [back to overview]Progression-free Survival (PFS)
NCT00686959 (7) [back to overview]Overall Survival
NCT00686959 (7) [back to overview]Objective Response Rate (Complete Response [CR] + Partial Response [PR])
NCT00686959 (7) [back to overview]Percentage of Participants With a Post Baseline Swallowing Diary Score >=4
NCT00691301 (4) [back to overview]Progression-free Survival
NCT00691301 (4) [back to overview]Duration of Overall Survival
NCT00691301 (4) [back to overview]Frequency and Severity of Observed Adverse Effects
NCT00691301 (4) [back to overview]Patients With Objective Tumor Response Rate (Complete Response [CR] or Partial Response [PR]) Using RECIST Version 1.0
NCT00698815 (4) [back to overview]18 Week Progression-free Survival (PFS) Rate
NCT00698815 (4) [back to overview]Overall Response Rate
NCT00698815 (4) [back to overview]PFS
NCT00698815 (4) [back to overview]Overall Survival (OS)
NCT00700336 (1) [back to overview]4M PFS Rate of Patients With Previously Untreated, Unresectable Malignant Pleural Mesothelioma (MPM) Treated With CBP501, Pemetrexed and Cisplatin
NCT00702299 (6) [back to overview]Maximum-tolerated Dose of Pemetrexed With a Day 2 i.p. Cisplatin (75 mg/m2) and Day 8 i.p. Paclitaxel (60 mg/m2)
NCT00702299 (6) [back to overview]Overall Survival
NCT00702299 (6) [back to overview]Pharmacokinetics (Mean Cmax, ug/mL)for Different Dosages of Pemetrexed
NCT00702299 (6) [back to overview]Patients That Completed at Least 6 Courses of Therapy of Pemetrexed Along With Day 2 i.p. Cisplatin (75 mg/m2) and Day 8 i.p. Paclitaxel (60 mg/m2)at the Determined Maximum Tolerated Dose
NCT00702299 (6) [back to overview]Patients Experienced Grade >=3 Toxicity at Dose Level 5 (1,000 mg/m2 IP Pemetrexed)
NCT00702299 (6) [back to overview]Progression-free Survival at 18 Months as Assessed by Cancer Antigen 125
NCT00703976 (2) [back to overview]2-year Progression-free Survival (PFS)
NCT00703976 (2) [back to overview]2-year Overall Survival (OS)
NCT00732303 (1) [back to overview]Assess Safety and Toxicity
NCT00732992 (11) [back to overview]Maximum Concentration of Pemetrexed Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1
NCT00732992 (11) [back to overview]Terminal Phase Elimination Half-Life (T1/2) of Pemetrexed Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1
NCT00732992 (11) [back to overview]"Sunitinib Relative Dose Intensity in the Sunitinib 37.5 mg/Day Continuous Daily Dosing Treatment Arm"
NCT00732992 (11) [back to overview]AUC 0-24 of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1
NCT00732992 (11) [back to overview]"Sunitinib Relative Dose Intensity in the Sunitinib 50 mg/Day Schedule-2/1 Treatment Arm"
NCT00732992 (11) [back to overview]Number of Participants With Adverse Events
NCT00732992 (11) [back to overview]Summary of Best Overall Response According to Response Evaluation Criteria in Solid Tumors (RECIST): Number of Participants
NCT00732992 (11) [back to overview]Tmax of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1
NCT00732992 (11) [back to overview]Trough and Maximum Concentration of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1
NCT00732992 (11) [back to overview]Trough Concentrations of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) After Coadministration of Sunitinib 50 mg/Day and Pemetrexed 500 mg/m^2 (Cycle 1 Day 1), Followed by Sunitinib 50 mg/Day on Schedule-2/1 at Cycle 1 Day 14 or 15
NCT00732992 (11) [back to overview]AUC0-∞ of Pemetrexed Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1
NCT00738582 (7) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs) as a Measure of Safety and Tolerability of Amatuximab
NCT00738582 (7) [back to overview]Number of Participants With Progression Free Survival (PFS) Responders and Non-responders at Month 6
NCT00738582 (7) [back to overview]Time to Tumor Response (TTR)
NCT00738582 (7) [back to overview]Overall Survival (OS)
NCT00738582 (7) [back to overview]Overall Response Rate (ORR)
NCT00738582 (7) [back to overview]Duration of Response (DR)
NCT00738582 (7) [back to overview]Overall Progression Free Survival
NCT00738881 (1) [back to overview]Progression-free Survival (PFS)
NCT00745875 (2) [back to overview]Time to Death
NCT00745875 (2) [back to overview]Progression-free Survival
NCT00762034 (28) [back to overview]Time to Progressive Disease
NCT00762034 (28) [back to overview]Translational Research: Overall Survival (OS) Based on Nuclear Thyroid Transcription Factor-1 (TTF-1) Expression Regardless of Study Treatment
NCT00762034 (28) [back to overview]Change From Baseline in Participant Reported Outcomes as Assessed by the Functional Assessment of Cancer Therapy - Lung (FACT-L)
NCT00762034 (28) [back to overview]Change From Baseline in Participant Reported Outcomes as Assessed by the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group- Neurotoxicity (FACT/GOG-Ntx)
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Time Curve From Zero to Infinity (AUC(0-∞)) for Total (Bound and Unbound) Platinum and Unbound Platinum
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Elimination Half-life (t1/2) for Total (Bound and Unbound) Platinum and Unbound Platinum
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) for Total (Bound and Unbound) Platinum and Unbound Platinum
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Platinum Clearance (CL) for Total (Bound and Unbound) and Unbound Forms
NCT00762034 (28) [back to overview]Translational Research: Number of Participants With Epidermal Growth Factor Receptor (EGFR) Mutations
NCT00762034 (28) [back to overview]Translational Research: Overall Survival (OS) Based on Cytoplasmic and Membrane Folate Receptor Alpha (FR-α) Expression
NCT00762034 (28) [back to overview]Translational Research: Overall Survival (OS) Based on Cytoplasmic and Nuclear Thymidylate Synthase (TS) Expression
NCT00762034 (28) [back to overview]Safety and Toxicity Profile of Study Treatments
NCT00762034 (28) [back to overview]Change From Baseline in Participant Reported Outcomes as Assessed by the Functional Assessment of Cancer Therapy - General (FACT-G)
NCT00762034 (28) [back to overview]Duration of Hospitalizations Per Participant
NCT00762034 (28) [back to overview]Number of Participants Receiving Concomitant Medication
NCT00762034 (28) [back to overview]Number of Participants Who Received a Transfusion
NCT00762034 (28) [back to overview]Overall Survival
NCT00762034 (28) [back to overview]Percentage of Participants With a Complete Response (CR) and Partial Response (PR) (Overall Response Rate)
NCT00762034 (28) [back to overview]Percentage of Participants With a Complete Response (CR), Partial Response (PR), and Stable Disease (SD) (Disease Control Rate)
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Time Curve From Zero to Infinity (AUC(0-∞)) Bevacizumab
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Time Curve From Zero to Infinity (AUC(0-∞)) for Pemetrexed
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Bevacizumab Clearance (CL)
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Elimination Half-life (t1/2) for Bevacizumab
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Elimination Half-life (t1/2) for Pemetrexed
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) for Bevacizumab
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) for Pemetrexed
NCT00762034 (28) [back to overview]Pharmacokinetics (PK): Pemetrexed Clearance (CL)
NCT00762034 (28) [back to overview]Progression Free Survival Time
NCT00768755 (6) [back to overview]Change From Baseline in Monroe Dunaway (MD) Anderson Symptom Inventory (MDASI) Symptom Severity Score
NCT00768755 (6) [back to overview]Change From Baseline in Monroe Dunaway (MD) Anderson Symptom Inventory (MDASI) Symptom Interference Score
NCT00768755 (6) [back to overview]Duration of Response (DR)
NCT00768755 (6) [back to overview]Overall Survival (OS)
NCT00768755 (6) [back to overview]Percentage of Participants With Objective Response (OR)
NCT00768755 (6) [back to overview]Progression-Free Survival (PFS)
NCT00769600 (3) [back to overview]RECIST Response
NCT00769600 (3) [back to overview]Progression Free Survival as Measured by Number of Days Without Disease Progression
NCT00769600 (3) [back to overview]Overall Survival
NCT00771953 (1) [back to overview]Progression Free Survival
NCT00789373 (12) [back to overview]Percentage of Participants With Independently-Assessed Objective Tumor Response (Response Rate) During Maintenance Phase Up to Primary Data Cut-Off
NCT00789373 (12) [back to overview]Percentage of Participants With Serious Adverse Events During Maintenance Phase
NCT00789373 (12) [back to overview]Change From Baseline in the EuroQol Instrument (EQ-5D) Index Score
NCT00789373 (12) [back to overview]Percentage of Participants With a Non-Serious Adverse Event (AE) During Maintenance Phase
NCT00789373 (12) [back to overview]Overall Survival (OS)
NCT00789373 (12) [back to overview]Percentage of Participants With Objective Tumor Response (Response Rate) During Maintenance Phase of Study up to Primary Data Cut-Off
NCT00789373 (12) [back to overview]Change From Baseline in EuroQol Instrument (EQ-5D) Visual Analog Scale (VAS)
NCT00789373 (12) [back to overview]Change From Baseline in EuroQol Instrument (EQ-5D) Visual Analog Scale (VAS)
NCT00789373 (12) [back to overview]Change From Baseline in the EuroQol Instrument (EQ-5D) Index Score
NCT00789373 (12) [back to overview]Percentage of Participants With Hospitalizations Due to Adverse Events or Requiring Transfusion (Resource Utilization)
NCT00789373 (12) [back to overview]Independently-assessed Objective Progression-free Survival (PFS)
NCT00789373 (12) [back to overview]Investigator-assessed Objective Progression-free Survival (PFS)
NCT00794417 (14) [back to overview]Phase 1 and 2: Terminal Half-Life (t1/2) of Aflibercept
NCT00794417 (14) [back to overview]Phase 1 and 2: Number of Participants With Treatment Emergent Adverse Events (TEAEs)
NCT00794417 (14) [back to overview]Phase 1 and 2: Number of Participants With Positive Anti-drug Antibody (ADA) of Aflibercept
NCT00794417 (14) [back to overview]Phase 1 and 2: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-inf) of Pemetrexed
NCT00794417 (14) [back to overview]Phase 1 and 2: Terminal Half-Life (t1/2) of Pemetrexed
NCT00794417 (14) [back to overview]Phase 1 and 2: Maximum Observed Plasma Concentration (Cmax) of Aflibercept and Pemetrexed
NCT00794417 (14) [back to overview]Phase 1 and 2: Number of Participants With All Grade Hematology Abnormalities
NCT00794417 (14) [back to overview]Phase 2: Progression-free Survival (PFS)
NCT00794417 (14) [back to overview]Phase 2: Objective Response Rate
NCT00794417 (14) [back to overview]Phase 1: Recommended Dose of Aflibercept for Phase 2
NCT00794417 (14) [back to overview]Phase 1 and 2: Total Body Clearance of Pemetrexed
NCT00794417 (14) [back to overview]Phase 1 and 2: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-inf) of Aflibercept
NCT00794417 (14) [back to overview]Phase 1 and 2: Total Body Clearance of Aflibercept
NCT00794417 (14) [back to overview]Phase 1 and 2: Number of Participants With All Grade Glucose Abnormalities
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 3 in Fatigue Assessed by Treatment-specific Adverse Events Scale
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 5 in Fatigue Assessed by Treatment-specific Adverse Events Scale
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 3 in Neuropathy Assessed by Treatment-specific Adverse Events Scale
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 3 in Overall Quality of Life Assessed by Linear Analogue Self Assessment (LASA)
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 3 in Nausea Assessed by Treatment-specific Adverse Events Scale
NCT00798603 (17) [back to overview]Number of Grade 3 or Higher Adverse Events Occurring in >=10% of Patients
NCT00798603 (17) [back to overview]Proportion of Confirmed Tumor Response Defined as an Objective Status of Complete Response or Partial Response on Two Consecutive Evaluations
NCT00798603 (17) [back to overview]Time to Treatment Failure
NCT00798603 (17) [back to overview]Progression-free Survival at 6 Months
NCT00798603 (17) [back to overview]Progression-free Survival
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 3 in Quality of Life (QOL) as Assessed by the Lung Cancer Symptom Scale
NCT00798603 (17) [back to overview]Overall Survival
NCT00798603 (17) [back to overview]Duration of Response
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 5 in Quality of Life (QOL) as Assessed by the Lung Cancer Symptom Scale
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 5 in Overall Quality of Life Assessed by Linear Analogue Self Assessment (LASA)
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 5 in Neuropathy Assessed by Treatment-specific Adverse Events Scale
NCT00798603 (17) [back to overview]Change From Baseline to Cycle 5 in Nausea Assessed by Treatment-specific Adverse Events Scale
NCT00806819 (14) [back to overview]Overall Survival (Key Secondary Endpoint)
NCT00806819 (14) [back to overview]Duration of Disease Control
NCT00806819 (14) [back to overview]Incidence and Intensity of Adverse Events
NCT00806819 (14) [back to overview]Objective Tumor Response
NCT00806819 (14) [back to overview]Duration of Confirmed Objective Tumour Response
NCT00806819 (14) [back to overview]Progression Free Survival (PFS) as Assessed by Central Independent Review
NCT00806819 (14) [back to overview]Clinical Improvement.
NCT00806819 (14) [back to overview]Follow-up Analysis of Progression Free Survival (PFS) as Assessed by Central Independent Review
NCT00806819 (14) [back to overview]Follow-up Analysis of Progression Free Survival (PFS) as Assessed by Investigator
NCT00806819 (14) [back to overview]Quality of Life (QoL)
NCT00806819 (14) [back to overview]Time to Confirmed Objective Tumour Response
NCT00806819 (14) [back to overview]Dose Normalised Predose Plasma Concentration at Steady State (Cpre,ss,Norm) of Nintedanib and of Its Metabolites BIBF 1202 and BIBF 1202 Glucuronide
NCT00806819 (14) [back to overview]Disease Control
NCT00806819 (14) [back to overview]Change From Baseline in Tumour Size
NCT00807573 (1) [back to overview]Objective Response Rate (CR + PR by RECIST) Paclitaxel, Pemetrexed, and Bevacizumab in Patients With Advanced Non-Small Lung Cancer Who Have Received no Prior Treatment for Metastatic Disease.
NCT00824408 (13) [back to overview]Duration of Overall Response
NCT00824408 (13) [back to overview]Objective Tumor Response, Defined as Complete Response (CR), and Partial Response (PR), Evaluated According to RECIST Criteria.
NCT00824408 (13) [back to overview]Cmax of Volasertib
NCT00824408 (13) [back to overview]CL of Pemetrexed
NCT00824408 (13) [back to overview]Cmax of Pemetrexed
NCT00824408 (13) [back to overview]Occurence of DLT
NCT00824408 (13) [back to overview]Frequency of Patients With Possible Clinically Significant Abnormalities
NCT00824408 (13) [back to overview]Occurrence and Intensity of AEs Graded According to CTCAE.
NCT00824408 (13) [back to overview]Vss of Volasertib
NCT00824408 (13) [back to overview]Overall Survival (OS)
NCT00824408 (13) [back to overview]Progression Free Survival (PFS) Time From the Date of Randomization to Date of Disease Progression or Death, Whichever Occurred First.
NCT00824408 (13) [back to overview]Total Clearance (CL) of Volasertib
NCT00824408 (13) [back to overview]Vss of Pemetrexed
NCT00859495 (1) [back to overview]Number of Subjects Who Were Able to Complete Trimodal Therapy (Combination of Surgery, Intrapleural and Systemic Chemotherapy and P-32 Radiotherapy).
NCT00860015 (1) [back to overview]Tumor Best Response Rate
NCT00864513 (4) [back to overview]Objective Response
NCT00864513 (4) [back to overview]Number of Participants With Adverse Events
NCT00864513 (4) [back to overview]CA 19-9 Response
NCT00864513 (4) [back to overview]Progression-free Survival
NCT00867009 (4) [back to overview]The Percentage of Participants Still Living at One Year (One Year Survival Rate)
NCT00867009 (4) [back to overview]Progression-free Survival (PFS)
NCT00867009 (4) [back to overview]Percentage of Participants With a Tumor Response (Objective Tumor Response Rate)
NCT00867009 (4) [back to overview]The Percentage of Participants With Complete Response (CR), Partial Response (PR) or Stable Disease (SD) (Disease Control Rate [DCR])
NCT00868192 (10) [back to overview]Overall Survival (OS)
NCT00868192 (10) [back to overview]Overall Response Rate
NCT00868192 (10) [back to overview]Overall Survival (OS)
NCT00868192 (10) [back to overview]Progression-free Survival (PFS)
NCT00868192 (10) [back to overview]Progression-free Survival (PFS)
NCT00868192 (10) [back to overview]CA-125 Response
NCT00868192 (10) [back to overview]Distribution of Overall Survival (OS)
NCT00868192 (10) [back to overview]Distribution of Progression-free Survival (PFS)
NCT00868192 (10) [back to overview]Toxicity Associated With Bevacizumab and Pemetrexed
NCT00868192 (10) [back to overview]Frequency of Clinical Response
NCT00871403 (3) [back to overview]Best Overall Response, Assessed as the Number of Participants With the Indicated Tumor Response: Investigator Assessed Only
NCT00871403 (3) [back to overview]Progression-free Survival (PFS)
NCT00871403 (3) [back to overview]Percentage of Participants With a Complete Response or a Partial Response
NCT00887549 (4) [back to overview]Percentage of Participants With Concordance Between Local and Central Histological Diagnosis
NCT00887549 (4) [back to overview]Percentage of Participants With Tumor Response (Tumor Response Rate)
NCT00887549 (4) [back to overview]Progression Free Survival (PFS)
NCT00887549 (4) [back to overview]Percentage of Participants Surviving at 18 Months (Overall Survival Rate)
NCT00892710 (6) [back to overview]Overall Survival (OS)
NCT00892710 (6) [back to overview]Time to Treatment Failure (TTTF)
NCT00892710 (6) [back to overview]6-month and 12-month Overall Survival Probability
NCT00892710 (6) [back to overview]Time to Progression (TTP)
NCT00892710 (6) [back to overview]Progression Free Survival (PFS)
NCT00892710 (6) [back to overview]Overall Response Rate (ORR), the Number of Patients Who Experience an Objective Benefit From Treatment
NCT00906282 (5) [back to overview]Rate of Residual Disease as an Assessment of Pathological Partial Response (pPR)
NCT00906282 (5) [back to overview]3-Year Overall Survival Rate
NCT00906282 (5) [back to overview]Objective Tumor Response
NCT00906282 (5) [back to overview]Complete Resection Rate
NCT00906282 (5) [back to overview]Pathologic Response Rate
NCT00921310 (6) [back to overview]Phase I Only: Maximum Tolerated Dose (MTD) of Pemetrexed That Could be Administered Weekly in Combination With Temsirolimus
NCT00921310 (6) [back to overview]Phase I Only: Maximum Tolerated Dose (MTD) of Temsirolimus That Could be Administered Weekly in Combination With Pemetrexed
NCT00921310 (6) [back to overview]Phase I Only: Number of Participants Who Experience Dose-limiting Toxicities (DLT) of Temsirolimus and Pemetrexed
NCT00921310 (6) [back to overview]Phase 2 Only: Progression-free Survival (PFS)
NCT00921310 (6) [back to overview]Phase I and Phase II: Overall Response Rate (Complete Response + Partial Response)
NCT00921310 (6) [back to overview]Phase 2 Only: Survival Rate
NCT00923273 (4) [back to overview]Phase II: Clinical Response Rate
NCT00923273 (4) [back to overview]Number of Participants With Serious and Non-Serious Adverse Events
NCT00923273 (4) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Sirolimus
NCT00923273 (4) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of Pemetrexed
NCT00932893 (15) [back to overview]Percentage of Participants With Objective Response (OR)
NCT00932893 (15) [back to overview]Progression-Free Survival (PFS)
NCT00932893 (15) [back to overview]Time to Deterioration (TTD) in Participant Reported Pain, Dyspnea, and Cough
NCT00932893 (15) [back to overview]Percentage of Participants With Disease Control at Week 12
NCT00932893 (15) [back to overview]Time to Tumor Response (TTR)
NCT00932893 (15) [back to overview]European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 (EORTC QLQ-C30)
NCT00932893 (15) [back to overview]European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire-Supplement Module for Lung Cancer (EORTC QLQ-LC13)
NCT00932893 (15) [back to overview]European Quality of Life - 5 Dimensional (EQ-5D) Visual Analog Scale (VAS)
NCT00932893 (15) [back to overview]Number of Participants With Categorical Maximum QTcF for Crizotinib
NCT00932893 (15) [back to overview]Duration of Response (DR)
NCT00932893 (15) [back to overview]Overall Survival (OS)
NCT00932893 (15) [back to overview]Overall Survival Probability at Months 6 and 12
NCT00932893 (15) [back to overview]Plasma Concentration of Crizotinib
NCT00932893 (15) [back to overview]Plasma Concentration of Soluble c-Met Ectodomain and Hepatocyte Growth Factor Scatter Proteins
NCT00932893 (15) [back to overview]Percentage of Participants With Disease Control at Week 6
NCT00942825 (1) [back to overview]The Primary Efficacy Endpoint is Progression Free Survival, Analyzed in the Treated Population. PFS is Assessed From Randomization Until Either Tumor Progression, as Per RECIST Criteria, or Until Death Due to Any Reason.
NCT00948675 (5) [back to overview]Percentage of Participants With Complete Response or Partial Response (Overall Tumor Response Rate)
NCT00948675 (5) [back to overview]Overall Survival (OS)
NCT00948675 (5) [back to overview]Disease Control Rates Defined as Complete Response (CR), Partial Response (PR), and Stable Disease (SD)
NCT00948675 (5) [back to overview]Progression Free Survival (PFS)
NCT00948675 (5) [back to overview]Progression Free Survival Without Grade 4 Toxicity (G4PFS) as Measured by the Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0
NCT00949650 (13) [back to overview]Trough Plasma Concentrations of Afatinib at Day 29
NCT00949650 (13) [back to overview]Trough Plasma Concentrations of Afatinib at Day 22
NCT00949650 (13) [back to overview]Progression-Free Survival (PFS) Time
NCT00949650 (13) [back to overview]Percentage of Patients With Objective Response (OR)
NCT00949650 (13) [back to overview]Percentage of Participants With Disease Control (DC)
NCT00949650 (13) [back to overview]HRQOL: Time to Deterioration in Pain
NCT00949650 (13) [back to overview]HRQOL: Time to Deterioration in Dyspnoea
NCT00949650 (13) [back to overview]Health Related Quality of Life (HRQOL): Time to Deterioration in Coughing
NCT00949650 (13) [back to overview]Overall Survival (OS) Time
NCT00949650 (13) [back to overview]Trough Plasma Concentrations of Afatinib at Day 43
NCT00949650 (13) [back to overview]Eastern Cooperative Oncology Group (ECOG) Performance Status (PS)
NCT00949650 (13) [back to overview]Change From Baseline in Body Weight
NCT00949650 (13) [back to overview]Tumour Shrinkage
NCT00950365 (3) [back to overview]PFS (Progression Free Survival)
NCT00950365 (3) [back to overview]Overall Survival
NCT00950365 (3) [back to overview]Objective Response Rate (CR +PR) Evaluated Using RECIST
NCT00961415 (8) [back to overview]Number of Participants With Marked Laboratory Abnormalities
NCT00961415 (8) [back to overview]Duration of Response During Maintenance Treatment Phase
NCT00961415 (8) [back to overview]Duration of Disease Control During Maintenance Treatment Phase
NCT00961415 (8) [back to overview]Overall Survival During Maintenance Treatment Phase
NCT00961415 (8) [back to overview]Progression Free Survival During Maintenance Treatment Phase
NCT00961415 (8) [back to overview]Best Overall Response Rate During Maintenance Treatment Phase
NCT00961415 (8) [back to overview]Incidence of Adverse Events and Serious Adverse Event
NCT00961415 (8) [back to overview]Quality of Life
NCT00976456 (2) [back to overview]Overall Survival
NCT00976456 (2) [back to overview]Progression Free Survival
NCT00979576 (10) [back to overview]AUC0-inf of Pemetrexed
NCT00979576 (10) [back to overview]Adverse Events According to Common Terminology Criteria for Adverse Events (CTCAE), Version 3.0 for All Courses
NCT00979576 (10) [back to overview]Duration of Disease Control
NCT00979576 (10) [back to overview]Dose Limiting Toxicities
NCT00979576 (10) [back to overview]AUC0-inf of Nintedanib
NCT00979576 (10) [back to overview]Disease Control Rate
NCT00979576 (10) [back to overview]Cmax of Nintedanib
NCT00979576 (10) [back to overview]Overall Response Rate
NCT00979576 (10) [back to overview]Number of Participants With Clinically Relevant Abnormalities in Laboratory Parameters
NCT00979576 (10) [back to overview]Cmax of Pemetrexed
NCT00982111 (10) [back to overview]Pharmacokinetics (PK): Minimum Concentration (Cmin) of Necitumumab
NCT00982111 (10) [back to overview]Overall Survival Time (OS)
NCT00982111 (10) [back to overview]Progression-Free Survival (PFS)
NCT00982111 (10) [back to overview]Time to Treatment Failure (TTF)
NCT00982111 (10) [back to overview]Percentage of Participants Who Achieve Best Overall Tumor Response of Complete Response (CR) or Partial Response (PR) (Objective Tumor Response Rate [ORR])
NCT00982111 (10) [back to overview]Percentage of Participants With EGFR Measured by IHC
NCT00982111 (10) [back to overview]Number of Participants With Serum Anti-Necitumumab Antibody Assessment (Immunogenicity)
NCT00982111 (10) [back to overview]Epidermal Growth Factor Hormone (EGFR) Protein Expression Measured by Immunohistochemistry (IHC)
NCT00982111 (10) [back to overview]Mean Change From Baseline in Patient Reported Outcomes (PRO) Using the European Quality of Life-5 Dimensions (EQ-5D)
NCT00982111 (10) [back to overview]Mean Change From Baseline in PRO as Measured Using the Lung Cancer Symptom Scale (LCSS)
NCT00988858 (9) [back to overview]Duration of Response
NCT00988858 (9) [back to overview]Overall Tumor Response - Percentage of Participants Achieving Complete Response (CR) or Partial Response (PR) [Overall Response Rate (ORR)]
NCT00988858 (9) [back to overview]Percentage of Participants Who Achieved a Best Response of Complete Response (CR), Partial Response (PR), or Stable Disease (SD) (Clinical Benefit Rate)
NCT00988858 (9) [back to overview]Progression-free Survival (PFS)
NCT00988858 (9) [back to overview]Pharmacokinetics (PK): Maximum Plasma Concentration (Cmax) of LY2603618
NCT00988858 (9) [back to overview]PK: Area Under the Plasma Concentration vs. Time Curve From Time Zero to Infinity [AUC(0-∞)] of LY2603618
NCT00988858 (9) [back to overview]Change in Symptom Burden Scores of Lung Cancer Symptom Scale (LCSS)
NCT00988858 (9) [back to overview]PK: Maximum Plasma Concentration (Cmax) of Pemetrexed
NCT00988858 (9) [back to overview]PK: Area Under the Plasma Concentration vs. Time Curve From Time Zero to Infinity [AUC(0-∞)] of Pemetrexed
NCT01000480 (3) [back to overview]Number of Participants With an Objective Tumor Response
NCT01000480 (3) [back to overview]Overall Survival
NCT01000480 (3) [back to overview]1 Year Progression Free Survival
NCT01001910 (3) [back to overview]Progression-free Interval
NCT01001910 (3) [back to overview]Incidence of Toxicities
NCT01001910 (3) [back to overview]Overall Survival (OS)
NCT01004250 (5) [back to overview]Percentage of Participants With Confirmed Complete Response or Partial Response During the Maintenance Therapy Only
NCT01004250 (5) [back to overview]Percentage of Participants With Confirmed Complete Response or Partial Response During Study Treatment (Induction and Maintenance)
NCT01004250 (5) [back to overview]Overall Survival
NCT01004250 (5) [back to overview]Progression-Free Survival
NCT01004250 (5) [back to overview]Percentage of Participants With Confirmed Response Complete or Partial Response During the Induction Treatment Only
NCT01005680 (9) [back to overview]Time to Treatment Failure (TtTF)
NCT01005680 (9) [back to overview]Time to Progressive Disease (TtPD)
NCT01005680 (9) [back to overview]Tumor Response Rate
NCT01005680 (9) [back to overview]Survival Without Toxicity (SWT)
NCT01005680 (9) [back to overview]Risk/Benefit Ratio
NCT01005680 (9) [back to overview]Progression Free Survival (PFS)
NCT01005680 (9) [back to overview]Overall Survival (OS)
NCT01005680 (9) [back to overview]Duration of Response (DoR)
NCT01005680 (9) [back to overview]Disease Control Rate (DCR)
NCT01017874 (7) [back to overview]Time to Progressive Disease (TtPD)
NCT01017874 (7) [back to overview]Time to Worsening of Health-Related Quality of Life (TWQ) Using the Participant-Rated Lung Cancer Symptom Scale (LCSS)
NCT01017874 (7) [back to overview]Percentage of Participants With Complete Response (CR) or Partial Response (PR) [Tumor Response Rate (TRR)]
NCT01017874 (7) [back to overview]Percentage of Participants With Complete Response (CR), Partial Response (PR) or Stable Disease (SD) [Disease Control Rate (DCR)]
NCT01017874 (7) [back to overview]Progression Free Survival (PFS)
NCT01017874 (7) [back to overview]Overall Survival (OS)
NCT01017874 (7) [back to overview]Duration of Tumor Response
NCT01020786 (9) [back to overview]Overall Survival (OS) During the Maintenance Therapy Period
NCT01020786 (9) [back to overview]Percentage of Participants Who Achieved a Complete Response (CR) or Partial Response (PR) During the Induction and Maintenance Therapy Periods
NCT01020786 (9) [back to overview]Percentage of Participants Who Achieved a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) During the Maintenance Therapy Period
NCT01020786 (9) [back to overview]Progression Free Survival (PFS) During the Maintenance Therapy Period
NCT01020786 (9) [back to overview]Percentage of Participants Who Achieve a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) During the Induction and Maintenance Therapy Periods
NCT01020786 (9) [back to overview]Overall Survival (OS) During the Induction and Maintenance Therapy Periods
NCT01020786 (9) [back to overview]Percentage of Participants Who Achieve a Complete Response (CR) or a Partial Response (PR) During the Induction Therapy Period
NCT01020786 (9) [back to overview]Percentage of Participants Who Observe a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) During the Induction Therapy Period
NCT01020786 (9) [back to overview]Progression Free Survival (PFS) During the Induction and Maintenance Therapy Periods
NCT01041781 (7) [back to overview]Incidence of Toxicities as Assessed by NCI CTCAE v. 4.0
NCT01041781 (7) [back to overview]Prognostic Value of Urinary Prostaglandin Metabolites (PGE-M) Levels for Worse PFS for Patients Who Had Baseline Urinary PGE-M Above/Below the Median Quartile (Q2)
NCT01041781 (7) [back to overview]Prognostic Value of Urinary Prostaglandin Metabolites (PGE-M) Levels for Worse PFS for Patients Who Had Baseline Urinary PGE-M Above/Below the Third Quartile (Q3)
NCT01041781 (7) [back to overview]Progression-free Survival
NCT01041781 (7) [back to overview]Prognostic Value of Urinary Prostaglandin Metabolites (PGE-M) Levels for Worse PFS for Patients Who Had Baseline Urinary PGE-M Above/Below the First Quartile (Q1)
NCT01041781 (7) [back to overview]Response Rate
NCT01041781 (7) [back to overview]Overall Survival
NCT01042288 (5) [back to overview]Median Overall Survival (OS)
NCT01042288 (5) [back to overview]Median Progression-free Survival (PFS)
NCT01042288 (5) [back to overview]Median Time to Progression (TTP)
NCT01042288 (5) [back to overview]Frequency of Adverse Events and Severity as a Measure of Toxicity
NCT01042288 (5) [back to overview]Objective Response Rate
NCT01057589 (6) [back to overview]Progression Free Survival (PFS)
NCT01057589 (6) [back to overview]Percent of Participants With a Partial Response (PR) or a Complete Response (CR)
NCT01057589 (6) [back to overview]Change From Baseline in Performance Status Scale for Head and Neck Cancer Patients (PSS-HNC)
NCT01057589 (6) [back to overview]Change From Baseline in Participant Reported European-Quality of Life 5 Dimension Instrument (EQ-5D) Visual Analog Scale (VAS) at End of Triplet Combination Therapy and End of Maintenance Therapy
NCT01057589 (6) [back to overview]Overall Survival (OS)
NCT01057589 (6) [back to overview]Change From Baseline in Participant Reported EQ-5D Utility Score at End of Triplet Combination Therapy and End of Maintenance Therapy
NCT01063283 (2) [back to overview]Response Rate
NCT01063283 (2) [back to overview]Change in 24 Hour Diastolic Blood Pressure (DBP)
NCT01064648 (9) [back to overview]Response Rate by Modified RECIST (Phase II)
NCT01064648 (9) [back to overview]Overall Survival (Phase II)
NCT01064648 (9) [back to overview]Maximum Tolerated Dose of Cediranib in Combination With Cisplatin and Pemetrexed (Phase I)
NCT01064648 (9) [back to overview]Disease Control Rate by RECIST 1.1 (Phase II)
NCT01064648 (9) [back to overview](Phase I) Number of Patients With Gr 3 Through 5 Adverse Events That Are Related to Study Drugs
NCT01064648 (9) [back to overview](Phase II) Number of Patients With Gr 3 Through 5 Adverse Events That Are Related to Study Drugs
NCT01064648 (9) [back to overview]Progression-free Survival (Phase II)
NCT01064648 (9) [back to overview]Disease Control Rate by Modified RECIST (Phase II)
NCT01064648 (9) [back to overview]Response Rate by RECIST1.1 (Phase II)
NCT01085630 (4) [back to overview]Overall Survival
NCT01085630 (4) [back to overview]Progression-free Survival
NCT01085630 (4) [back to overview]Toxicity, Assessed Using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 (v4)
NCT01085630 (4) [back to overview]Response Rate
NCT01087970 (6) [back to overview]Overall Survival (OS)
NCT01087970 (6) [back to overview]Percentage of Participants Having a Confirmed Partial Response (PR) or Complete Response (CR)
NCT01087970 (6) [back to overview]Progression-Free Survival (PFS)
NCT01087970 (6) [back to overview]Change From Baseline in Performance Status Scale for Head and Neck Cancer (PSS-HNC)
NCT01087970 (6) [back to overview]Number of Participants Who Died While on Treatment and Died During 30-Day Post-Treatment Discontinuation Follow-Up (FU)
NCT01087970 (6) [back to overview]Change From Baseline in Participant-Reported European-Quality of Life-5 Dimension Instrument-3 Levels (EQ-5D-3L)
NCT01107626 (3) [back to overview]Response Rate
NCT01107626 (3) [back to overview]Overall Survival
NCT01107626 (3) [back to overview]Progression-free Survival
NCT01126736 (4) [back to overview]Phase 2: Percentage of Participants Who Experienced TEAEs
NCT01126736 (4) [back to overview]Phase 1b: Percentage of Participants With Grade 3 or Higher Treatment Emergent Adverse Events (TEAEs)
NCT01126736 (4) [back to overview]Phase 2: Progression-free Survival (PFS)
NCT01126736 (4) [back to overview]Phase 1b: Number of Participants With Dose-Limiting Toxicity (DLTs)
NCT01139775 (15) [back to overview]Phase 1: Document Any Antitumor Activity Per Radiological Scans and/or Tumor Markers
NCT01139775 (15) [back to overview]Deaths
NCT01139775 (15) [back to overview]Phase 2: Progression-Free Survival Time
NCT01139775 (15) [back to overview]Phase 2: Pharmacokinetic: Cmax (LY2603618)
NCT01139775 (15) [back to overview]Phase 1: Pharmacokinetic: Area Under the Plasma Concentration Versus Time Curve (AUC) (LY2603618)
NCT01139775 (15) [back to overview]Phase 2: Overall Survival
NCT01139775 (15) [back to overview]Phase 1: Pharmacokinetic: AUC (Pemetrexed and Cisplatin)
NCT01139775 (15) [back to overview]Phase 1: Pharmacokinetic: Cmax (Pemetrexed and Cisplatin)
NCT01139775 (15) [back to overview]Phase 2: Clinical Benefit Rate: Percentage of Participant Who Achieved a Response of Stable Disease (SD), Partial Response (PR), or Complete Response (CR)
NCT01139775 (15) [back to overview]Phase 2: Change in Tumor Size
NCT01139775 (15) [back to overview]Phase 1: Recommended Phase 2 Dose of LY2603618
NCT01139775 (15) [back to overview]Phase 1: Pharmacokinetic: Maximum Plasma Concentration (Cmax) (LY2603618)
NCT01139775 (15) [back to overview]Phase 2: Change From Baseline to Long-term Follow up in Lung Cancer Symptom Scale (LCSS)
NCT01139775 (15) [back to overview]Phase 2: Pharmacokinetic: AUC (LY2603618)
NCT01139775 (15) [back to overview]Phase 2: Overall Tumor Response Rate: Percentage of Participants Who Achieved a Confirmed Best Response of Completed Response (CR) or Partial Response (PR)
NCT01160744 (7) [back to overview]Change in Tumor Size (CTS)
NCT01160744 (7) [back to overview]Duration of Response (DOR)
NCT01160744 (7) [back to overview]Overall Survival (OS)
NCT01160744 (7) [back to overview]Percentage of Participants With Best Overall Response of Complete Response (CR) or Partial Response (PR) [Objective Response Rate (ORR)]
NCT01160744 (7) [back to overview]Percentage of Participants With CR, PR, or Stable Disease (SD) [Disease Control Rate (DCR)]
NCT01160744 (7) [back to overview]Progression-Free Survival (PFS)
NCT01160744 (7) [back to overview]Number of Participants Reporting Treatment-Emergent Adverse Events (TEAEs) and Who Died
NCT01165021 (5) [back to overview]Overall Survival (OS)
NCT01165021 (5) [back to overview]Percentage of Participants With Complete Response (CR) or Partial Response (PR) [Overall Response Rate (ORR)]
NCT01165021 (5) [back to overview]Percentage of Participants With No Viable Tumor Cells in Resected Lung Tissue [Pathological Complete Remission (pCR)]
NCT01165021 (5) [back to overview]Progression-Free Survival (PFS)
NCT01165021 (5) [back to overview]Percentage of Participants Who Exhibit a Downward Shift in Tumor Extent From Stage IIIAN2 to Stages IIIA, II, I, or Stage 0
NCT01169675 (6) [back to overview]Disease Control
NCT01169675 (6) [back to overview]Objective Response (OR)
NCT01169675 (6) [back to overview]Tumour Shrinkage
NCT01169675 (6) [back to overview]Progression Free Survival (PFS)
NCT01169675 (6) [back to overview]Investigator Defined Dose Limiting Toxicity (DLT) During First Course of Treatment, Treated Set
NCT01169675 (6) [back to overview]Investigator Defined Dose Limiting Toxicity (DLT) During All Courses of Treatment, Treated Set
NCT01215916 (4) [back to overview]Percentage of Participants With a Tumor Response
NCT01215916 (4) [back to overview]Number of Participants With Clinically Significant Effects
NCT01215916 (4) [back to overview]Pharmacokinetics, Area Under the Curve (AUC) of LY573636
NCT01215916 (4) [back to overview]Pharmacokinetics, Concentration Maximum (Cmax) of LY573636
NCT01218516 (5) [back to overview]Progression-free Survival (PFS)
NCT01218516 (5) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Treatment-Emergent Serious Adverse Events (SAEs)
NCT01218516 (5) [back to overview]Overall Response Rate (ORR)
NCT01218516 (5) [back to overview]Overall Survival (OS)
NCT01218516 (5) [back to overview]Duration of Response (DR)
NCT01232452 (10) [back to overview]Pharmacokinetics (PK): Maximum Serum Concentration (Cmax) of Cixutumumab, Cycle 1 (First Infusion) and Cycle 4 (Fourth Infusion)
NCT01232452 (10) [back to overview]Time to Worsening of Symptoms as Measured by Lung Cancer Symptom Scale (LCSS) Score
NCT01232452 (10) [back to overview]PK: Area Under the Concentration Time Curve (AUC[0-inf]) of Cixutumumab, Cycle 1 (i.e. First Infusion)
NCT01232452 (10) [back to overview]Percentage of Participants Achieving an Objective Response Rate (ORR)
NCT01232452 (10) [back to overview]Overall Survival (OS)
NCT01232452 (10) [back to overview]Duration of Response (DOR)
NCT01232452 (10) [back to overview]Change in Tumor Size (CTS)
NCT01232452 (10) [back to overview]Time to Progressive Disease (TTPS)
NCT01232452 (10) [back to overview]Progression-free Survival (PFS)
NCT01232452 (10) [back to overview]PK: Area Under the Concentration Time Curve During 1 Dosing Interval (i.e. 504 hr, AUC(0-tau) of Cixutumumab, Cycle 4 (i.e. Fourth Infusion)
NCT01263782 (2) [back to overview]Overall Response Rate
NCT01263782 (2) [back to overview]Progression Free Survival
NCT01287520 (10) [back to overview]PK Parameter: Maximum Plasma Concentration (Cmax) of LY2090314
NCT01287520 (10) [back to overview]PK Parameter: AUC0-∞ of Free Carboplatin (Carb)
NCT01287520 (10) [back to overview]PK Parameter: Cmax of Free Carboplatin
NCT01287520 (10) [back to overview]Pharmacokinetic (PK) Parameter: Area Under the Concentration-Time Curve From Time 0 Hour to Infinity (AUC0-∞) of Pemetrexed (Pem)
NCT01287520 (10) [back to overview]Pharmacokinetic (PK) Parameter: Area Under the Concentration-Time Curve From Time 0 Hour to Infinity (AUC0-∞) of LY2090314
NCT01287520 (10) [back to overview]PK Parameter: Maximum Plasma Concentration (Cmax) of Pemetrexed (Pem)
NCT01287520 (10) [back to overview]PK Parameter: AUC0-∞ of LY2090314 Coadministered With Pemetrexed (Pem) and Carboplatin (Carb)
NCT01287520 (10) [back to overview]Number of Participants With Best Overall Tumor Response
NCT01287520 (10) [back to overview]Recommended LY2090314 Dose for Phase 2 Studies (Maximum Tolerated Dose [MTD])
NCT01287520 (10) [back to overview]PK Parameter: Maximum Plasma Concentration (Cmax) of LY2090314 Coadministered With Pemetrexed (Pem) and Carboplatin (Carb)
NCT01296568 (10) [back to overview]Plasma Pharmacokinetics of LY2603618: Area Under the Concentration Time Curve From Time Zero to Infinity [AUC(0-infinity)]
NCT01296568 (10) [back to overview]Relative Abundance of LY2603618 and the Metabolites of LY2603618 in Feces
NCT01296568 (10) [back to overview]Plasma Pharmacokinetics of LY2603618: Maximum Observed Drug Concentration (Cmax)
NCT01296568 (10) [back to overview]Plasma Pharmacokinetics of LY2603618: Area Under the Concentration Time Curve From Time Zero to Time t [AUC(0-tlast)]
NCT01296568 (10) [back to overview]Relative Abundance of LY2603618 and the Metabolites of LY2603618 in Urine
NCT01296568 (10) [back to overview]The Number of Participants With a Tumor Response
NCT01296568 (10) [back to overview]Plasma Pharmacokinetics of Radioactivity: Area Under the Concentration Time Curve From Time Zero to Infinity [AUC(0-infinity)]
NCT01296568 (10) [back to overview]Plasma Pharmacokinetics of Radioactivity: Area Under the Concentration Time Curve From Time Zero to Time t [AUC(0-tlast)]
NCT01296568 (10) [back to overview]Plasma Pharmacokinetics of Radioactivity: Maximum Observed Drug Concentration (Cmax)
NCT01296568 (10) [back to overview]Urinary and Fecal Excretion of LY2603618 Radioactivity Over Time Expressed as a Percentage of the Total Radioactive Dose Administered
NCT01313663 (14) [back to overview]Number of Participants With the Indicated Changes From Baseline Value in Lactate Dehydrogenase (LDH)
NCT01313663 (14) [back to overview]Number of Participants With Any On-therapy AE (Serious or Non-serious) Leading to Dose Reductions (DRs) or Interruptions/Delays in the Study
NCT01313663 (14) [back to overview]Number of Participants With Any Non-serious On-therapy Adverse Event (AE: Occurring in >=5% Participants in Any Treatment Arm) and Serious Adverse Event (SAE)
NCT01313663 (14) [back to overview]Number of Participants With a Change From Baseline Grade to Grade 3 and 4 for the Indicated Clinical Laboratory Parameters
NCT01313663 (14) [back to overview]Number of Participants (Par.) With the Indicated Best Overall Response
NCT01313663 (14) [back to overview]Time on Study Treatment (Pazopanib), as a Measure of Extent of Exposure
NCT01313663 (14) [back to overview]Number of Participants With Any AE (Serious or Non-serious) Leading to Withdrawal From Study Treatment
NCT01313663 (14) [back to overview]Average Dose of Pemetrexed for All Cycles, as a Measure of Extent of Exposure
NCT01313663 (14) [back to overview]Mean Daily Dose, as a Measure of Extent of Exposure
NCT01313663 (14) [back to overview]Number of Participants With the Indicated Worst-case Change From Baseline in Blood Pressure
NCT01313663 (14) [back to overview]Progression Free Survival (PFS)
NCT01313663 (14) [back to overview]Mean Number of Pemetrexed Dosing Cycles, as a Measure of Extent of Exposure
NCT01313663 (14) [back to overview]Number of Participants With a Increase From Baseline in Bazett's QTc at the Indicated Time Points
NCT01313663 (14) [back to overview]Number of Participants With the Indicated Grade Changes From Baseline Grade in Aspartate Aminotransferase (AST), Alanine Aminotransferase (ALT), Alkaline Phosphatase (Alk. Phos.), and Total Bilirubin (TB)
NCT01338792 (3) [back to overview]Time to Disease Progression and Overall Survival
NCT01338792 (3) [back to overview]Best Overall Response
NCT01338792 (3) [back to overview]Number of Participants With Serious Adverse Events (SAEs)
NCT01344824 (3) [back to overview]Overall Survival
NCT01344824 (3) [back to overview]Subjects Experiencing Toxicity
NCT01344824 (3) [back to overview]Progression-free Survival
NCT01351415 (8) [back to overview]Overall Survival (OS)
NCT01351415 (8) [back to overview]Percentage of Participants With Disease Control According to RECIST v1.1
NCT01351415 (8) [back to overview]Percentage of Participants With Objective Response According to RECIST v1.1
NCT01351415 (8) [back to overview]Percentage of Participants Who Are Alive at Month 6, 12, and 18
NCT01351415 (8) [back to overview]Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT01351415 (8) [back to overview]Progression-free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1)
NCT01351415 (8) [back to overview]Time to Progression (TTP) According to RECIST v1.1
NCT01351415 (8) [back to overview]Duration of Response (DoR) According to RECIST v1.1
NCT01358968 (7) [back to overview]Number of Participants With a Tumor Response
NCT01358968 (7) [back to overview]Plasma Pharmacokinetics of LY2603618: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to Infinity [AUC(0-∞)]
NCT01358968 (7) [back to overview]Plasma Pharmacokinetics of LY2603618: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to the Last Observed Plasma Concentration of LY2603618 [AUC(0-tlast)]
NCT01358968 (7) [back to overview]Plasma Pharmacokinetics of LY2603618: the Maximum Concentration of LY2603618 in the Plasma (Cmax)
NCT01358968 (7) [back to overview]Plasma Pharmacokinetics of Desipramine: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to Infinity [AUC(0-∞)]
NCT01358968 (7) [back to overview]Plasma Pharmacokinetics of Desipramine: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to the Last Observed Plasma Concentration of Desipramine [AUC(0-tlast)]
NCT01358968 (7) [back to overview]Plasma Pharmacokinetics of Desipramine: the Maximum Concentration of Desipramine in the Plasma (Cmax)
NCT01376310 (1) [back to overview]Number of Participants With Adverse Events
NCT01395758 (4) [back to overview]ORR Among Subjects in the Crossover Period Treated With Erlotinib Plus Tivantinib
NCT01395758 (4) [back to overview]Overall Survival (OS) Among All Eligible Subjects (ITT Population) Treated With Erlotinib Plus Tivantinib Compared to Chemotherapy.
NCT01395758 (4) [back to overview]Objective Response Rate (ORR) Among All Eligible Subjects (ITT Population) Treated With Erlotinib Plus Tivantinib Compared to Chemotherapy.
NCT01395758 (4) [back to overview]Progression-free Survival (PFS) Among Subjects With KRAS Mutation Positive NSCLC (ITT Population) Treated With Erlotinib Plus Tivantinib Compared to Single Agent Chemotherapy.
NCT01443078 (2) [back to overview]PERCIST Partial Metabolic Response
NCT01443078 (2) [back to overview]Pathologic Response Rate
NCT01447225 (11) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: MM-121 Doses
NCT01447225 (11) [back to overview]Immunogenicity
NCT01447225 (11) [back to overview]Objective Response Rate
NCT01447225 (11) [back to overview]Pharmacokinetics
NCT01447225 (11) [back to overview]Pharmacokinetics (AUClast)
NCT01447225 (11) [back to overview]To Characterize Dose-limiting Toxicities (DLTs) Associated With the Combination of MM-121 With Anticancer Therapies
NCT01447225 (11) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Carboplatin
NCT01447225 (11) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Cabazitaxel
NCT01447225 (11) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Gemcitabine
NCT01447225 (11) [back to overview]To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Pemetrexed
NCT01447225 (11) [back to overview]To Evaluate the Safety and Tolerability of Escalating Doses of the MM-121 Anticancer Therapies
NCT01454102 (6) [back to overview]Number of Participants With Abnormalities in Selected Hepatic Clinical Laboratory Tests
NCT01454102 (6) [back to overview]Number of Participants With Abnormalities in Selected Thyroid Clinical Laboratory Tests
NCT01454102 (6) [back to overview]Objective Response Rate (ORR)
NCT01454102 (6) [back to overview]Progression-Free Survival Rate (PFSR) at Week 24
NCT01454102 (6) [back to overview]Number of Participants Who Experienced Selected Adverse Events
NCT01454102 (6) [back to overview]Number of Participants Who Experienced Serious Adverse Events (SAE), Adverse Events (AE) Leading to Discontinuation, or Death
NCT01454934 (3) [back to overview]Overall Survival (OS)
NCT01454934 (3) [back to overview]Progression Free Survival (PFS) by Response Evaluation Criteria in Solid Tumors (RECIST)
NCT01454934 (3) [back to overview]Objective Response Rate (ORR)
NCT01469000 (7) [back to overview]Time to Worsening of Symptom (TWS) as Per Lung Cancer Symptom Scale (LCSS)
NCT01469000 (7) [back to overview]Time To Progressive Disease (TTPD)
NCT01469000 (7) [back to overview]Overall Survival (OS)
NCT01469000 (7) [back to overview]Duration of Response (DoR)
NCT01469000 (7) [back to overview]Progression Free Survival (PFS)
NCT01469000 (7) [back to overview]Percentage of Participants Achieving Complete Response (CR) or Partial Response (PR) (Overall Response Rate [ORR])
NCT01469000 (7) [back to overview]Percentage of Participants With CR, PR, and Stable Disease (SD) (Disease Control Rate [DCR])
NCT01471197 (3) [back to overview]Number of Participants Who Died Within 30 Days and 31 Days After Last Dose - All Treated Participants
NCT01471197 (3) [back to overview]Number of Participants With Deaths, Adverse Events (AEs), Serious AEs (SAEs) and AEs Leading to Discontinuation - All Treated Participants
NCT01471197 (3) [back to overview]Overall Survival of Participants During the Study - All Treated Participants
NCT01473563 (16) [back to overview]Participant Satisfaction: Preferences Regarding Home and/or Hospital Treatment
NCT01473563 (16) [back to overview]Participant Satisfaction: Regarding the Study Nurse
NCT01473563 (16) [back to overview]Physician Satisfaction: Distant Management of Participant
NCT01473563 (16) [back to overview]Resource Utilization: Number of Participants With an Unplanned Use of Healthcare Resources
NCT01473563 (16) [back to overview]Overall Survival (OS) at 6 Months
NCT01473563 (16) [back to overview]Number of Participants Who Had Treatment-Emergent Adverse Events (TEAEs), Serious Adverse Events (SAEs), or Died
NCT01473563 (16) [back to overview]Change From Baseline in the EQ-5D Index Score
NCT01473563 (16) [back to overview]Time to Treatment Failure (TTF)
NCT01473563 (16) [back to overview]Resource Utilization: Duration of Health Care Visits
NCT01473563 (16) [back to overview]Percentage of Participants Who Adhered to Treatment Administration at Home
NCT01473563 (16) [back to overview]Participant Satisfaction: Chemotherapy at Home
NCT01473563 (16) [back to overview]Resource Utilization: Unplanned Health Care Visits, Consultations, and Diagnostic Services
NCT01473563 (16) [back to overview]Resource Utilization: Distances Traveled
NCT01473563 (16) [back to overview]Participant Satisfaction: Chemotherapy at Hospital
NCT01473563 (16) [back to overview]Change From Baseline in the European Quality of Life Instrument (EQ-5D) Visual Analogue Scale (VAS)
NCT01473563 (16) [back to overview]Maximum Improvement Over Baseline in Individual Lung Cancer Symptoms Scale (LCSS) Item Scores
NCT01544179 (12) [back to overview]Median Overall Survival (OS) at Time of PFS Analysis
NCT01544179 (12) [back to overview]Median Progression-Free Survival (Site Read, Investigator Assessment)
NCT01544179 (12) [back to overview]Objective Response Rate (ORR) (Site Read Data)
NCT01544179 (12) [back to overview]Progression-Free Survival (Site Read, Investigator Assessment)
NCT01544179 (12) [back to overview]Time to Worsening in FACT-L Total Score
NCT01544179 (12) [back to overview]Time to Worsening in Lung Cancer Subscale
NCT01544179 (12) [back to overview]Overall Survival (OS)
NCT01544179 (12) [back to overview]Time to Worsening in Trial Outcome Index
NCT01544179 (12) [back to overview]Disease Control Rate (DCR)
NCT01544179 (12) [back to overview]Improvement in FACT-L Total Score
NCT01544179 (12) [back to overview]Improvement in Trial Outcome Index
NCT01544179 (12) [back to overview]Improvement in Lung Cancer Subscale
NCT01565538 (3) [back to overview]Overall Survival
NCT01565538 (3) [back to overview]Progression-Free Survival
NCT01565538 (3) [back to overview]Best Tumor Response
NCT01639001 (21) [back to overview]Percentage of Participants With Visual Disturbance as Assessed by Visual Symptom Assessment Questionnaire (VSAQ-ALK)
NCT01639001 (21) [back to overview]Time to Deterioration (TTD) in Participant Reported Pain, Dyspnea, or Cough Assessed Using Quality of Life Questionnaire Supplement Module for Lung Cancer (QLQ-LC13)
NCT01639001 (21) [back to overview]Percentage of Participants With Visual Disturbance as Assessed by Visual Symptom Assessment Questionnaire (VSAQ-ALK)
NCT01639001 (21) [back to overview]Percentage of Participants With Treatment-Emergent AEs (Treatment Related)
NCT01639001 (21) [back to overview]Percentage of Participants With Treatment-Emergent Adverse Events (AEs; All Causalities)
NCT01639001 (21) [back to overview]Estimate of the Percentage of Participants Surviving at 1 Year and at 18 Months
NCT01639001 (21) [back to overview]Change From Baseline Scores in QLQ-C30 Symptoms as Assessed by the EORTC-QLQ-C30
NCT01639001 (21) [back to overview]Change From Baseline in Lung Cancer Symptom Scores as Assessed by the EORTC Quality of Life Questionnaire-Lung Cancer 13 (QLQ-LC13)
NCT01639001 (21) [back to overview]Change From Baseline in Functioning and Global Quality of Life (QOL) as Assessed by the European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC-QLQ-C30)
NCT01639001 (21) [back to overview]Agreement Between Central Laboratory ALK FISH and ALK IHC Test Results - Molecular Profiling Evaluable
NCT01639001 (21) [back to overview]Time to Tumor Response (TTR) Based on IRR
NCT01639001 (21) [back to overview]Time to Progression (TTP) Based on IRR
NCT01639001 (21) [back to overview]Change From Baseline in General Health Status as Assessed by EuroQol 5D (EQ-5D)-Visual Analog Scale (VAS)
NCT01639001 (21) [back to overview]Change From Baseline in General Health Status as Assessed by EQ-5D-Index
NCT01639001 (21) [back to overview]Progression-Free Survival (PFS) Based on IRR by Treatment Arm
NCT01639001 (21) [back to overview]Duration of Response (DR) Based on IRR
NCT01639001 (21) [back to overview]Extracranial Time to Progression (EC-TTP) Based on IRR
NCT01639001 (21) [back to overview]Intracranial Time to Progression (IC-TTP) Based on IRR
NCT01639001 (21) [back to overview]Objective Response Rate (ORR) - Percentage of Participants With Objective Response Based on IRR
NCT01639001 (21) [back to overview]Overall Survival (OS)
NCT01639001 (21) [back to overview]Percentage of Participants With Disease Control at 12 Weeks Based on IRR
NCT01646125 (2) [back to overview]Progression Free Survival (PFS)
NCT01646125 (2) [back to overview]Overall Response Rate (ORR)
NCT01675765 (2) [back to overview]Number of Subjects Reporting Adverse Events
NCT01675765 (2) [back to overview]Objective Tumor Response
NCT01803282 (2) [back to overview]Percentage of Participants Experiencing Treatment-Emergent Adverse Events
NCT01803282 (2) [back to overview]Percentage of Participants Experiencing Laboratory Abnormalities
NCT01809210 (9) [back to overview]Cmax,ss
NCT01809210 (9) [back to overview]CL/F
NCT01809210 (9) [back to overview]Best Percentage Change From Baseline in Target Lesion Size
NCT01809210 (9) [back to overview]Dose Limiting Toxicity (DLT) Events in Chemotherapy in Combination With Selumetinib
NCT01809210 (9) [back to overview]Best Objective Response
NCT01809210 (9) [back to overview]AUC (0-tau)
NCT01809210 (9) [back to overview]Percentage Change From Baseline at 6 Weeks in Target Lesion Size
NCT01809210 (9) [back to overview]Objective Response Rate (ORR)
NCT01809210 (9) [back to overview]Tmax,ss
NCT01828099 (1) [back to overview]Progression Free Survival (PFS) by Blinded Independent Review Committee (BIRC)
NCT01828112 (1) [back to overview]Progression Free Survival (PFS) Blinded Independent Review Committee Per Blinded Independent Review Committee (BIRC)
NCT01840579 (42) [back to overview]Number of Participants Who Experienced Dose-limiting Toxicities (DLTs)
NCT01840579 (42) [back to overview]Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 8: Parts A, B, C, and E
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 6: Part A
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 4: Parts A and D
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 18: Part A
NCT01840579 (42) [back to overview]Number of Participants Who Experienced at Least One Adverse Event (AE)
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 2: Part A
NCT01840579 (42) [back to overview]Volume of Distribution (Vz) of Pembrolizumab Over Time for Part A Cycle 1
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 9: Part A
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 8: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 7: Parts A, B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 6: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 5: Parts A, B, C, and E
NCT01840579 (42) [back to overview]Area Under the Concentration Time Curve From 0-28 Days (AUC 0-28) for Part A Cycle 1
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 4: Part D
NCT01840579 (42) [back to overview]Clearance (CL) of Pembrolizumab Over Time for Part A Cycle 1
NCT01840579 (42) [back to overview]Area Under the Concentration Time Curve From 0-Infinity (AUC 0-inf) for Part A Cycle 1
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 1: Parts A, B, C, D, and E
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 10: Part A
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 12: Part A
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 14: Part A
NCT01840579 (42) [back to overview]Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 16: Part A
NCT01840579 (42) [back to overview]Time to Maximum Serum Concentration (Tmax) of Pembrolizumab for Cycle 8: Parts B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 3: Parts A, B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 3: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 27: Parts B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 23: Parts B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 2: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 19: Parts B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 17: Part A
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 16: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 15: Parts A, B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 14: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 13: Part A
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 12: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 11: Parts A, B, C, and E
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 10: Part D
NCT01840579 (42) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab for Cycle 1: Parts A, B, C, D, and E
NCT01840579 (42) [back to overview]Time to Maximum Serum Concentration (Tmax) of Pembrolizumab for Cycle 4: Part D
NCT01840579 (42) [back to overview]Time to Maximum Serum Concentration (Tmax) of Pembrolizumab for Cycle 1: Parts A, B, C, D, and E
NCT01840579 (42) [back to overview]Terminal Half-Life (t1/2) of Pembrolizumab Over Time for Part A Cycle 1
NCT01868022 (23) [back to overview]Number of Participants With Non-serious Adverse Events (AEs) and Serious AEs (SAEs)
NCT01868022 (23) [back to overview]Number of Participants With the Abnormal Urinalysis Findings
NCT01868022 (23) [back to overview]Number of Participants With Clinically Significant Findings for 12-lead Electrocardiogram (ECG)
NCT01868022 (23) [back to overview]Change From Baseline in Heart Rate
NCT01868022 (23) [back to overview]Change From Baseline in Temperature
NCT01868022 (23) [back to overview]Number of Participants With Dose Delays
NCT01868022 (23) [back to overview]Number of Participants With Dose Reduction
NCT01868022 (23) [back to overview]Number of Participants With Dose-Limiting Toxicities (DLT)
NCT01868022 (23) [back to overview]Number of Participants With Maximum Tolerated Dose (MTD) or Maximum Feasible Dose (MFD)
NCT01868022 (23) [back to overview]Number of Participants With Overall Response Rate (ORR)
NCT01868022 (23) [back to overview]Number of Participants Withdrew Due to AEs
NCT01868022 (23) [back to overview]Progression Free Survival (PFS) as Assessed by Investigator
NCT01868022 (23) [back to overview]Treatment Duration With GSK3052230
NCT01868022 (23) [back to overview]Change From Baseline in Forced Vital Capacity (FVC) in of Arm C Participants With Malignant Pleural Mesothelioma (MPM)
NCT01868022 (23) [back to overview]Change From Baseline in Forced Vital Capacity (FVC) in of Arm C Participants With Malignant Pleural Mesothelioma (MPM)
NCT01868022 (23) [back to overview]Change From Baseline in Forced Vital Capacity (FVC) in of Arm C Participants With Malignant Pleural Mesothelioma (MPM)
NCT01868022 (23) [back to overview]Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)
NCT01868022 (23) [back to overview]Number of Participants With Abnormal Echocardiogram (ECHO) Findings
NCT01868022 (23) [back to overview]Number of Participants With Hematology Change From Baseline With Respect to the Normal Range
NCT01868022 (23) [back to overview]Number of Participants With Best Response
NCT01868022 (23) [back to overview]Number of Participants With Clinical Chemistry Changes From Baseline With Respect to the Normal Range
NCT01868022 (23) [back to overview]Number of Participants With Clinical Chemistry Changes From Baseline With Respect to the Normal Range
NCT01868022 (23) [back to overview]Number of Participants With Clinical Chemistry Changes From Baseline With Respect to the Normal Range
NCT01907100 (4) [back to overview]Disease Control According to Modified RECIST- Investigator Assessment
NCT01907100 (4) [back to overview]Objective Response According to Modified RECIST- Investigator Assessment
NCT01907100 (4) [back to overview]Overall Survival (OS)
NCT01907100 (4) [back to overview]Progression-Free Survival (PFS)
NCT01918761 (4) [back to overview]Overall Survival
NCT01918761 (4) [back to overview]Progression-free Survival
NCT01918761 (4) [back to overview]Dose Limiting Toxicities (DLTs)
NCT01918761 (4) [back to overview]Overall Response Rate
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Clinically Significant Abnormalities in 12-Lead Electrocardiograms (ECG) Findings
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Clinically Significant Abnormalities in Vital Signs
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status Score of 2 or Higher Than 2
NCT01982955 (44) [back to overview]Phase 1b: Percentage of Participants With Disease Control Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
NCT01982955 (44) [back to overview]Phase 1b: Percentage of Participants With Objective Response Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
NCT01982955 (44) [back to overview]Phase 2 (Non-Randomized Part Only): Percentage of Participants With Disease Control Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
NCT01982955 (44) [back to overview]Phase 2 (Non-Randomized Part Only): Percentage of Participants With Objective Response Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
NCT01982955 (44) [back to overview]Phase 2 (Non-Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by Independent Review Committee (IRC)
NCT01982955 (44) [back to overview]Phase 2 (Non-Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by Investigator
NCT01982955 (44) [back to overview]Phase 2 (Randomized Part Only): Percentage of Participants With Disease Control Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
NCT01982955 (44) [back to overview]Phase 2 (Randomized Part Only): Percentage of Participants With Objective Response Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria
NCT01982955 (44) [back to overview]Phase 2 (Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by Independent Review Committee (IRC)
NCT01982955 (44) [back to overview]Phase 2 (Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by the Investigator
NCT01982955 (44) [back to overview]Phase 2: (Non-Randomized Part Only): Overall Survival (OS) Time
NCT01982955 (44) [back to overview]Phase 2: (Randomized Part Only): Overall Survival (OS) Time
NCT01982955 (44) [back to overview]Phase 2: Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life (EORTC QLQ-C30) Global Health Status Scale Score at End of Treatment (EOT)
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Clinically Significant Abnormalities in 12-Lead Electrocardiograms (ECG) Findings
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Clinically Significant Abnormalities in Vital Signs
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status Score of 2 or Higher Than 2
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) Leading to Permanent Treatment Discontinuation
NCT01982955 (44) [back to overview]Phase 2: Time-to-Symptom Progression (TTSP)
NCT01982955 (44) [back to overview]Phase 1b: Apparent Terminal Elimination Rate Constant Lambda(z) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Apparent Terminal Half-Life (t1/2) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Apparent Total Body Clearance From Plasma (CL/F) of Tepotinib and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Apparent Volume of Distribution (Vz/F) During the Terminal Phase of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Apparent Volume of Distribution During the Steady State (Vss/F) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Area Under the Plasma Concentration Versus Time Curve From Time Zero to the Last Sampling Time AUC (0-t) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC 0-infinity) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Area Under the Plasma Concentration-Time Curve Within 1 Dosing Interval (AUC 0-tau) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Average Observed Plasma Concentration (Cavg) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Maximum Observed Plasma Concentration (Cmax) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Minimum Observed Plasma Concentration (Cmin) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Death and Reasons
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Grade 3/4 Treatment-Emergent Adverse Events (TEAEs) and Grade 3/4 Treatment-Related TEAEs According to National Cancer Institute Common Toxicity Criteria for Adverse Events (NCI-CTCAE) Version 4.03
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Laboratory Test Abnormalities of Grade 3 or Higher Severity Based on NCI-CTCAE Version 4.03 Reported as Treatment-Emergent Adverse Events (TEAEs)
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious TEAEs
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) Leading to Permanent Treatment Discontinuation
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants With Treatment-Related Treatment-Emergent Adverse Events (TEAEs) and Treatment-Related Serious TEAEs According to National Cancer Institute Common Toxicity Criteria for Adverse Events Version 4.03
NCT01982955 (44) [back to overview]Phase 1b: Time to Reach Maximum Plasma Concentration (Tmax) of Tepotinib, Its Metabolites and Gefitinib
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Death and Reasons
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Greater Than or Equal to (>=) Grade 3 Treatment-Emergent Adverse Events (TEAEs) and >= Grade 3 Treatment-Related TEAEs According to National Cancer Institute Common Toxicity Criteria for Adverse Events Version 4.03
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Laboratory Test Abnormalities of Grade 3 or Higher Severity Based on NCI-CTCAE Version 4.03 Reported as Treatment-Emergent Adverse Events (TEAEs)
NCT01982955 (44) [back to overview]Phase 2: Number of Participants With Treatment-Emergent Adverse Events (TEAEs), Serious TEAEs, Treatment-Related TEAEs and Treatment-Related Serious TEAEs According to National Cancer Institute Common Toxicity Criteria for Adverse Events Version 4.03
NCT01982955 (44) [back to overview]Phase 1b: Number of Participants Experiencing at Least One Dose Limiting Toxicity (DLT)
NCT02039674 (6) [back to overview]Part 2 Cohorts G+ and G-: Duration of Response (DOR)
NCT02039674 (6) [back to overview]Part 2 Cohorts G+ and G-: Progression-Free Survival (PFS)
NCT02039674 (6) [back to overview]Part 2 Cohorts G+ and G-: Overall Survival (OS)
NCT02039674 (6) [back to overview]All Cohorts: Number of Participants Who Experienced a Dose-limiting Toxicity (DLT)
NCT02039674 (6) [back to overview]Part 2 Cohorts D4 and H: Objective Response Rate (ORR)
NCT02039674 (6) [back to overview]Part 2 Cohorts G+ and G-: Objective Response Rate (ORR)
NCT02041533 (6) [back to overview]Disease-related Symptom Improvement Rate by Week 12
NCT02041533 (6) [back to overview]Progression-Free Survival in Participants With PD-L1 Expression >= 5%
NCT02041533 (6) [back to overview]Progression-Free Survival in All Randomized Participants
NCT02041533 (6) [back to overview]Overall Survival in Participants With PD-L1 Expression >= 5%
NCT02041533 (6) [back to overview]Overall Survival in All Randomized Participants
NCT02041533 (6) [back to overview]Objective Response Rate (ORR) in Participants With PD-L1 Expression >= 5%
NCT02079636 (15) [back to overview]Pharmacokinetics (PK): Maximum Concentration (Cmax) of Abemaciclib on Day 1 and at Steady State (Cycle 2 Day 1) in Part A , B, C, D and E
NCT02079636 (15) [back to overview]Number of Participants With Dose-Limiting Toxicities (DLT) or DLT-equivalent in Part A, B, C, D and E
NCT02079636 (15) [back to overview]Number of Participants Achieving Complete Response (CR) or Partial Response (PR) (Overall Response Rate [ORR]) in Part A, B, C, D and E
NCT02079636 (15) [back to overview]PK: Area Under the Concentration Curve (AUC) of Pemetrexed at Steady State in Part A
NCT02079636 (15) [back to overview]Progression Free Survival Time in Part A, B, C, D and E
NCT02079636 (15) [back to overview]PK: Maximum Concentration (Cmax) of Ramucirumab at 1 Hour Post-End-of-Infusion in Part C
NCT02079636 (15) [back to overview]PK: Maximum Concentration (Cmax) of LY3023414 in Part D
NCT02079636 (15) [back to overview]Change From Baseline in MD Anderson Symptom Inventory Scale-Lung Cancer (MDASI-LC) in Part A, B, C, D and E
NCT02079636 (15) [back to overview]PK: Area Under the Concentration Curve (AUC) of LY3023414 in Part D
NCT02079636 (15) [back to overview]PK: Dose-normalized Maximum Concentration (Cmax) of Active Gemcitabine Metabolite: 2',2'-Difluorodeoxyuridine (dFdU) on Day 1 and at Steady State (Cycle 2 Day 1) in Part B
NCT02079636 (15) [back to overview]PK: Area Under the Concentration Curve (AUC) of Active Gemcitabine Metabolite: 2',2'-Difluorodeoxyuridine (dFdU) on Day 1 and at Steady State (Cycle 2 Day 1) in Part B
NCT02079636 (15) [back to overview]PK: Area Under the Concentration Curve (AUC) of Abemaciclib in Part A, B, C, D and E
NCT02079636 (15) [back to overview]Pharmacokinetics (PK): Maximum Concentration (Cmax) of Abemaciclib on Day 1 and at Steady State (Cycle 2 Day 1) in Part A , B, C, D and E
NCT02079636 (15) [back to overview]PK: Area Under the Concentration Curve (AUC) of Abemaciclib in Part A, B, C, D and E
NCT02079636 (15) [back to overview]Pharmacokinetics: Maximum Concentration (Cmax) of Pemetrexed at Steady State in Part A
NCT02083679 (2) [back to overview]Number of Subjects With Dose Limiting Toxicities (DLTs)
NCT02083679 (2) [back to overview]Number of Subjects With Treatment-emergent Adverse (TEAEs), Serious TEAEs, TEAEs Leading to Discontinuation and TEAEs Leading to Death
NCT02087241 (2) [back to overview]Assess the Objective Response Rates in Each Arm
NCT02087241 (2) [back to overview]Assess the Disease Control Rate in Each Treatment Arm
NCT02093962 (1) [back to overview]Overall Survival
NCT02117024 (6) [back to overview]Progression-Free Survival (PFS)
NCT02117024 (6) [back to overview]Overall Survival (OS)
NCT02117024 (6) [back to overview]Frequency of Adverse Events: Number of Participants With Treatment-Emergent Adverse Events (TEAE)
NCT02117024 (6) [back to overview]Survival at 6 Months
NCT02117024 (6) [back to overview]Survival at 12 Months
NCT02117024 (6) [back to overview]Time to Progression (TTP)
NCT02119650 (5) [back to overview]Progression-free Survival (PFS)
NCT02119650 (5) [back to overview]Duration of Response
NCT02119650 (5) [back to overview]Overall Survival (OS)
NCT02119650 (5) [back to overview]Objective Response Rate (ORR)
NCT02119650 (5) [back to overview]Participants With Treatment-emergent Adverse Events (TEAEs)
NCT02142738 (3) [back to overview]Objective Response Rate (ORR)
NCT02142738 (3) [back to overview]Overall Survival (OS) Rate
NCT02142738 (3) [back to overview]Progression Free Survival (PFS) Rate at Month 6
NCT02145078 (2) [back to overview]Overall Survival (OS)
NCT02145078 (2) [back to overview]Progression-free Survival (PFS)
NCT02220894 (11) [back to overview]Number of Participants Who Experienced At Least One Adverse Event (AE)
NCT02220894 (11) [back to overview]Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%
NCT02220894 (11) [back to overview]Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%
NCT02220894 (11) [back to overview]Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%
NCT02220894 (11) [back to overview]Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥20%
NCT02220894 (11) [back to overview]Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥50%
NCT02220894 (11) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%
NCT02220894 (11) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%
NCT02220894 (11) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%
NCT02220894 (11) [back to overview]Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥1%
NCT02220894 (11) [back to overview]Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)
NCT02264990 (6) [back to overview]Progression Free Survival (PFS) in All Participants
NCT02264990 (6) [back to overview]Overall Survival (OS) in the Lung Subtype Panel Positive Subgroup
NCT02264990 (6) [back to overview]Overall Survival in All Participants
NCT02264990 (6) [back to overview]Objective Response Rate (ORR) in the Lung Subtype Panel Positive Subgroup
NCT02264990 (6) [back to overview]Objective Response Rate (ORR) in All Participants
NCT02264990 (6) [back to overview]Progression Free Survival (PFS) in the Lung Subtype Panel Positive Subgroup
NCT02274038 (1) [back to overview]Overall Survival
NCT02322281 (5) [back to overview]Duration of Response (DOR) According to RECIST Version 1.1 as Determined by Investigator Assessment
NCT02322281 (5) [back to overview]Overall Survival (OS)
NCT02322281 (5) [back to overview]Percentage of Participants With Confirmed Response
NCT02322281 (5) [back to overview]Progression Free Survival (PFS) According to RECIST Version 1.1 as Determined by Investigator Review (invPFS)
NCT02322281 (5) [back to overview]Plasma PK for Patients Treated With Rociletinib Based on Sparse Sampling
NCT02337530 (1) [back to overview]Progression Free Survival
NCT02347917 (11) [back to overview]Overall Survival(OS)
NCT02347917 (11) [back to overview]Phase 1 Part: Number of Participants With Dose-limiting Toxicities (DLTs)
NCT02347917 (11) [back to overview]Phase 2 Part: Progression-free Survival (PFS)
NCT02347917 (11) [back to overview]Phase 1 Part: Area Under the Concentration-time Curve
NCT02347917 (11) [back to overview]Phase 1 Part: Maximum Observed Concentration (Cmax) and Minimum Observed Concentration (Cmin) of BBI608 When Administered With Pem and CDDP
NCT02347917 (11) [back to overview]Phase 1 Part: Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) and Adverse Drug Reactions (ADRs)
NCT02347917 (11) [back to overview]Respiratory Function Tests (Forced Expiratory Volume in the First Second [FEV1])
NCT02347917 (11) [back to overview]Respiratory Function Tests (Forced Expiratory Volume in the First Second [FEV1])
NCT02347917 (11) [back to overview]Response Rate (RR) and Disease Control Rate (DCR)
NCT02347917 (11) [back to overview]Respiratory Function Tests (Vital Capacity [VC] and Forced Vital Capacity [FVC])
NCT02347917 (11) [back to overview]Respiratory Function Tests (Vital Capacity [VC] and Forced Vital Capacity [FVC])
NCT02357147 (1) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT02367781 (19) [back to overview]Percentage of Participants With Adverse Events
NCT02367781 (19) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms Score Using the Symptoms in Lung Cancer (SILC) Scale
NCT02367781 (19) [back to overview]Overall Survival (OS) in the ITT-WT Population
NCT02367781 (19) [back to overview]OS as Determined by the Investigator Using Recist v1.1 in the ITT Population
NCT02367781 (19) [back to overview]Plasma Concentrations of Nab-Paclitaxel Reported as Total Paclitaxel
NCT02367781 (19) [back to overview]Plasma Concentrations of Carboplatin
NCT02367781 (19) [back to overview]PFS as Determined by the Investigator Using Recist v1.1 in the ITT Population, PD-L1 Expression Population, and PD-L1 Expression WT Population
NCT02367781 (19) [back to overview]Percentage of Participants With Anti-Therapeutic Antibodies (ATAs) to Atezolizumab
NCT02367781 (19) [back to overview]Percentage of Participants With an Objective Response (OR) (Complete Response [CR] or Partial Response [PR]) as Determined by the Investigator Using RECIST v1.1 in the ITT Population, PD-L1 Expression Population, and PD-L1 Expression WT Population
NCT02367781 (19) [back to overview]OS as Determined by the Investigator Using RECIST v1.1 in the PD-L1 Expression Population and PD-L1 Expression WT Population
NCT02367781 (19) [back to overview]Minimum Observed Serum Concentration (Cmin) of Atezolizumab Prior to Infusion in Atezolizumab+Carboplain+Nab-Paclitaxel
NCT02367781 (19) [back to overview]Maximum Observed Serum Concentration (Cmax) of Atezolizumab for Patients in Atezolizumab+Carboplatin+Nab-Paclitaxel Arm
NCT02367781 (19) [back to overview]Time to Deterioration (TTD) in Patient-Reported Lung Cancer Symptoms in the ITT-WT Population
NCT02367781 (19) [back to overview]Event Free Rate (%) at Year 1 and 2 in PD-L1 Expression Population and PD-L1 Expression WT Population
NCT02367781 (19) [back to overview]Event Free Rate (%) at Year 1 and 2 in ITT-WT Population and ITT Population
NCT02367781 (19) [back to overview]Duration of Response (DOR) as Determined by the Investigator Using RECIST v1.1 in ITT-WT Population, ITT Population, and PD-L1 Expression Population and PD-L1 Expression WT Population
NCT02367781 (19) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms Score Using the Symptoms in Lung Cancer (SILC) Scale
NCT02367781 (19) [back to overview]Progression-Free Survival (PFS) as Determined by the Investigator Using Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) in the ITT-WT Population
NCT02367781 (19) [back to overview]Percentage of Participants With an Objective Response (OR) (Complete Response [CR] or Partial Response [PR]) as Determined by the Investigator Using RECIST v1.1 in the ITT-WT Population
NCT02409342 (24) [back to overview]Duration of Response (DOR) in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]TTD as Assessed Using EORTC QLQ Supplementary Lung Cancer Module (EORTC QLQ-LC13) in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Change From Baseline in Patient-reported Lung Cancer Symptoms Score as Assessed by the SILC Scale Symptom Score in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Time to Deterioration (TTD) in Patient-reported Lung Cancer Symptoms Score as Assessed by the Symptoms in Lung Cancer (SILC) Scale Symptom Score in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Overall Survival (OS) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations
NCT02409342 (24) [back to overview]Percentage of Participants Who Are Alive at 1 Year in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations
NCT02409342 (24) [back to overview]Percentage of Participants Who Are Alive at 2 Years in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations
NCT02409342 (24) [back to overview]Percentage of Participants With Anti-therapeutic Antibodies (ATAs)
NCT02409342 (24) [back to overview]Change From Baseline in Patient-reported Lung Cancer Symptoms Score as Assessed by the SILC Scale Symptom Score in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Progression-free Survival (PFS) in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Percentage of Participants With Objective Response (ORR) in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Percentage of Participants With at Least One Adverse Event
NCT02409342 (24) [back to overview]Percentage of Participants Who Are Alive at 2 Years in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Percentage of Participants Who Are Alive at 1 Year in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Progression-free Survival (PFS) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations
NCT02409342 (24) [back to overview]OS in Participants With PD-L1 Expression
NCT02409342 (24) [back to overview]Minimum Observed Serum Concentration (Cmin) of Atezolizumab
NCT02409342 (24) [back to overview]Investigator-Assessed PFS in Participants With PD-L1 Expression According to RECIST v1.1
NCT02409342 (24) [back to overview]Investigator-Assessed PFS in Participants With bTMB According to RECIST v1.1
NCT02409342 (24) [back to overview]OS in Participants With Blood Tumor Mutational Burden (bTMB)
NCT02409342 (24) [back to overview]Overall Survival (OS) in the TC3 or IC3-WT Populations
NCT02409342 (24) [back to overview]Maximum Observed Serum Concentration (Cmax) of Atezolizumab
NCT02409342 (24) [back to overview]Percentage of Participants With Objective Response (ORR) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations
NCT02409342 (24) [back to overview]Duration of Response (DOR) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations
NCT02426658 (5) [back to overview]Overall Survival
NCT02426658 (5) [back to overview]Time to Tumor Progression
NCT02426658 (5) [back to overview]Change in Quality of Life (QOL), Assessed by the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (QLQ-C30) and QLQ-Lung Cancer 13-item (LC13)
NCT02426658 (5) [back to overview]Incidence of Hematologic Toxicity, Graded According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0
NCT02426658 (5) [back to overview]Response Rate
NCT02439450 (1) [back to overview]Phase 1b: Frequency of Treatment Emergent Adverse Events (TEAEs) as Assessed by CTCAE v4.03.
NCT02453282 (29) [back to overview]DoR; PD-L1 (TC >=1%) Analysis Set Population
NCT02453282 (29) [back to overview]DoR; FAS Population
NCT02453282 (29) [back to overview]Ctrough_ss of Tremelimumab
NCT02453282 (29) [back to overview]Cmax_ss of Tremelimumab
NCT02453282 (29) [back to overview]PFS; PD-L1 (TC >=25%) Analysis Set Population, Durvalumab Monotherapy Vs SoC Chemotherapy and Durvalumab + Tremelimumab Vs Durvalumab Monotherapy
NCT02453282 (29) [back to overview]Serum Concentrations of Tremelimumab
NCT02453282 (29) [back to overview]Serum Concentrations of Durvalumab
NCT02453282 (29) [back to overview]Number of Participants With Anti-Drug Antibody (ADA) Response to Durvalumab
NCT02453282 (29) [back to overview]Number of Participants With ADA Response to Tremelimumab
NCT02453282 (29) [back to overview]Change From Baseline in Disease-Related Symptoms as Assessed by European Organisation for Research and Treatment of Cancer Quality of Life Questionnaires (EORTC QLQ) at 12 Months
NCT02453282 (29) [back to overview]Trough Serum Concentration at Steady State (Ctrough_ss) of Durvalumab
NCT02453282 (29) [back to overview]Time From Randomization to Second Progression (PFS2); PD-L1 (TC >=25%) Analysis Set Population
NCT02453282 (29) [back to overview]Progression-Free Survival (PFS); PD-L1 (TC >=25%) Analysis Set Population, Durvalumab + Tremelimumab Vs SoC Chemotherapy
NCT02453282 (29) [back to overview]PFS2; PD-L1 (TC >=1%) Analysis Set Population
NCT02453282 (29) [back to overview]PFS2; FAS Population
NCT02453282 (29) [back to overview]PFS; PD-L1 (TC >=1%) Analysis Set Population
NCT02453282 (29) [back to overview]PFS; FAS Population
NCT02453282 (29) [back to overview]Percentage of Participants APF12; PD-L1 (TC >=1%) Analysis Set Population
NCT02453282 (29) [back to overview]Percentage of Participants APF12; FAS Population
NCT02453282 (29) [back to overview]Percentage of Participants Alive and Progression Free at 12 Months (APF12); PD-L1 (TC >=25%) Analysis Set Population
NCT02453282 (29) [back to overview]Overall Survival (OS); PD-L1 (TC >=25%) Analysis Set Population, Durvalumab Monotherapy Vs SoC Chemotherapy and Durvalumab + Tremelimumab Vs SoC Chemotherapy
NCT02453282 (29) [back to overview]OS; PD-L1 (TC >=25%) Analysis Set Population, Durvalumab + Tremelimumab Vs Durvalumab Monotherapy
NCT02453282 (29) [back to overview]OS; PD-L1 (TC >=1%) Analysis Set Population
NCT02453282 (29) [back to overview]OS; FAS Population
NCT02453282 (29) [back to overview]ORR; PD-L1 (TC >=1%) Analysis Set Population
NCT02453282 (29) [back to overview]ORR; FAS Population
NCT02453282 (29) [back to overview]Objective Response Rate (ORR); PD-L1 (TC >=25%) Analysis Set Population
NCT02453282 (29) [back to overview]Maximum Serum Concentration at Steady State (Cmax_ss) of Durvalumab
NCT02453282 (29) [back to overview]Duration of Response (DoR); PD-L1 (TC >=25%) Analysis Set Population
NCT02542293 (20) [back to overview]Duration of Response (DoR); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]DoR; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets
NCT02542293 (20) [back to overview]APF12; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets
NCT02542293 (20) [back to overview]Objective Response Rate (ORR); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, bTMB ≥12 Mut/Mb, tTMB ≥14 Mut/Mb, tTMB ≥12 Mut/Mb, tTMB ≥10 Mut/Mb, and tTMB ≥8 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]OS; China Cohort: China Programmed Cell Death Ligand 1 (PD-L1) Negative NSCLC Analysis Set
NCT02542293 (20) [back to overview]Serum Concentrations of Durvalumab
NCT02542293 (20) [back to overview]Time From Randomization to Second Progression or Death (PFS2); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]Serum Concentrations of Tremelimumab
NCT02542293 (20) [back to overview]Overall Survival (OS); Global Cohort: Blood Tumor Mutational Burden (bTMB) ≥20 Mutations Per Megabase (Mut/Mb) Analysis Set
NCT02542293 (20) [back to overview]Number of Participants With ADA Response to Tremelimumab
NCT02542293 (20) [back to overview]Number of Participants With Anti-Drug Antibody (ADA) Response to Durvalumab
NCT02542293 (20) [back to overview]ORR; Global and China Cohorts: PD-L1-Negative NSCLC, FAS, PD-L1 TC ≥25%, and PD-L1 TC ≥50% Analysis Sets
NCT02542293 (20) [back to overview]OS at Months 12, 18 and 24; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets
NCT02542293 (20) [back to overview]OS at Months 12, 18 and 24; Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]OS; Global and China Cohorts: FAS, PD-L1 Tumor Cell (TC) ≥25%, and PD-L1 TC ≥50% Analysis Sets
NCT02542293 (20) [back to overview]OS; Global Cohort: bTMB ≥16 Mut/Mb, bTMB ≥12 Mut/Mb, PD-L1-Negative NSCLC, bTMB <20 Mut/Mb, bTMB Non-Evaluable Population, tTMB ≥14 Mut/Mb, tTMB ≥12 Mut/Mb, tTMB ≥10 Mut/Mb, and tTMB ≥8 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]Alive and Progression-Free at 12 Months (APF12); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]PFS; Global and China Cohorts: PD-L1-Negative NSCLC, FAS, PD-L1 TC ≥25%, and PD-L1 TC ≥50% Analysis Sets
NCT02542293 (20) [back to overview]Progression-Free Survival (PFS); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, bTMB ≥12 Mut/Mb, tTMB ≥14 Mut/Mb, tTMB ≥12 Mut/Mb, tTMB ≥10 Mut/Mb, and tTMB ≥8 Mut/Mb Analysis Sets
NCT02542293 (20) [back to overview]PFS2; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets
NCT02574078 (7) [back to overview]Percentage of Participants With Treatment-related Adverse Events (AEs) Leading to Both Study Drugs Discontinuation, Group E Only
NCT02574078 (7) [back to overview]Overall Survival (OS), Groups A-C Only
NCT02574078 (7) [back to overview]Overall Survival (OS), Group D Only
NCT02574078 (7) [back to overview]Objective Response Rate (ORR), Groups A-E
NCT02574078 (7) [back to overview]Duration of Response (DOR), Groups A-D Only
NCT02574078 (7) [back to overview]Progression-Free Survival (PFS), Group E Only
NCT02574078 (7) [back to overview]Progression-Free Survival (PFS), Groups A-D Only
NCT02576574 (32) [back to overview]Overall Survival (OS) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)
NCT02576574 (32) [back to overview]Overall Survival (OS) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Full Analysis Set (FAS)
NCT02576574 (32) [back to overview]Overall Survival (OS) in High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)
NCT02576574 (32) [back to overview]Overall Survival (OS) in High Programmed Death Ligand 1 (PD-L1) + Full Analysis Set (FAS)
NCT02576574 (32) [back to overview]Overall Survival (OS) in Full Analysis Set (FAS)
NCT02576574 (32) [back to overview]Duration of Response (DOR) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)
NCT02576574 (32) [back to overview]Duration of Response (DOR) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1)+ Full Analysis Set (FAS)
NCT02576574 (32) [back to overview]Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer 13 (EORTC QLQ-LC13) at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ Modified HRQoL Analysis Set
NCT02576574 (32) [back to overview]Change From Baseline in European Quality Of Life 5-dimensions (EQ-5D-5L) Visual Analog Scale (VAS) in High Programmed Death Ligand 1 (PD-L1)+ Modified HRQoL Analysis Set
NCT02576574 (32) [back to overview]Number of Participants With At Least One Positive Anti-Drug Antibodies (ADAs) and Neutralizing Antibodies (NAbs) for Avelumab
NCT02576574 (32) [back to overview]Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Heart Rate Increase/Decrease
NCT02576574 (32) [back to overview]Overall Survival (OS) in Modified Full Analysis Set (mFAS)
NCT02576574 (32) [back to overview]Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Body Temperature Increase
NCT02576574 (32) [back to overview]Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance: Baseline Score Versus (Vs) Worst Post-baseline Score
NCT02576574 (32) [back to overview]Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer 13 (EORTC QLQ-LC13) at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ HRQoL Analysis Set
NCT02576574 (32) [back to overview]Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)
NCT02576574 (32) [back to overview]Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Full Analysis Set (FAS)
NCT02576574 (32) [back to overview]Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1) + Modified Full Analysis Set (mFAS)
NCT02576574 (32) [back to overview]Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1) + Full Analysis Set (FAS)
NCT02576574 (32) [back to overview]Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in Moderate and High PD-L1+ Modified Full Analysis Set
NCT02576574 (32) [back to overview]Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Systolic Blood Pressure Increase/Decrease and Maximal Diastolic Blood Pressure Increase/Decrease
NCT02576574 (32) [back to overview]Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Weight Increase/Decrease
NCT02576574 (32) [back to overview]Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in Moderate and High PD-L1+ Full Analysis Set
NCT02576574 (32) [back to overview]Change From Baseline in European Quality Of Life 5-dimensions (EQ-5D-5L) Visual Analog Scale (VAS) in High PD-L1+ Health-related Quality of Life (HRQoL) Analysis Set at End of Treatment
NCT02576574 (32) [back to overview]Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life (EORTC QLQ-C30) Global Health Status at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ Modified HRQoL Analysis Set
NCT02576574 (32) [back to overview]Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life (EORTC QLQ-C30) Global Health Status at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ HRQoL Analysis Set
NCT02576574 (32) [back to overview]Number of Participants With Potentially Clinically Significant Abnormalities (PCSA) in Electrocardiogram (ECG) Parameters
NCT02576574 (32) [back to overview]Number of Participants With Shift From Baseline to Greater Than or Equal to (>=) Grade 3 in Laboratory Parameter Values Based on National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE), Version 4.03
NCT02576574 (32) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and AEs of Special Interest (AESIs)
NCT02576574 (32) [back to overview]Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High PD-L1+ Modified Full Analysis Set
NCT02576574 (32) [back to overview]Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High PD-L1+ Full Analysis Set
NCT02576574 (32) [back to overview]Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Respiration Rate Increase/Decrease
NCT02578680 (6) [back to overview]Number of Participants Who Experienced an Adverse Event (AE)
NCT02578680 (6) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 as Assessed by Blinded Central Imaging
NCT02578680 (6) [back to overview]Number of Participants Who Discontinued Any Study Drug Due to an AE
NCT02578680 (6) [back to overview]Duration of Response (DOR) Per RECIST 1.1 as Assessed by Blinded Central Imaging
NCT02578680 (6) [back to overview]Overall Response Rate (ORR) Per RECIST 1.1 as Assessed by Blinded Central Imaging
NCT02578680 (6) [back to overview]Overall Survival (OS)
NCT02591615 (3) [back to overview]Overall Response Rate (ORR) Per RECIST 1.1
NCT02591615 (3) [back to overview]Incidence of Treatment-Emergent Adverse Events (Safety and Tolerability)
NCT02591615 (3) [back to overview]Compare Progression-Free Survival (PFS) Per RECIST 1.1
NCT02604342 (27) [back to overview]Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC30 Score for ITT Population
NCT02604342 (27) [back to overview]Plasma Concentration of Alectinib
NCT02604342 (27) [back to overview]Duration of Response for Lesions in the CNS (C-DOR) Using RECIST Version 1.1 as Assessed by IRC
NCT02604342 (27) [back to overview]Overall Survival (OS)
NCT02604342 (27) [back to overview]Percentage of Participants With Adverse Events (AEs)
NCT02604342 (27) [back to overview]Percentage of Participants With CNS Objective Response Rate (ORR) With Measurable CNS Metastases at Baseline Using RECIST Version 1.1 as Assessed By IRC
NCT02604342 (27) [back to overview]Percentage of Participants With Disease Control in C-ITT Population Using RECIST Version 1.1 as Assessed by IRC
NCT02604342 (27) [back to overview]Percentage of Participants With ORR in C-ITT Population Using RECIST Version 1.1 as Assessed by IRC
NCT02604342 (27) [back to overview]PFS Using RECIST Version 1.1 as Assessed by IRC
NCT02604342 (27) [back to overview]Plasma Concentration of Alectinib Metabolite
NCT02604342 (27) [back to overview]Progression-Free Survival (PFS) Using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 as Assessed by Investigator
NCT02604342 (27) [back to overview]Time to CNS Progression in C-ITT Population Using RECIST Version 1.1 as Assessed by IRC
NCT02604342 (27) [back to overview]TTD in Composite of Three Symptoms (Cough, Dyspnea, and Chest Pain) Using EORTC QLQ-LC13 Score for C-ITT Population
NCT02604342 (27) [back to overview]TTD in Composite of Three Symptoms (Cough, Dyspnea, and Chest Pain) Using EORTC QLQ-LC13 Score for ITT Population
NCT02604342 (27) [back to overview]Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (EORTC QLQ-C30) Over Time
NCT02604342 (27) [back to overview]Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (EORTC QLQ-C30) Over Time
NCT02604342 (27) [back to overview]Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer-13 (EORTC QLQ-LC13) Over Time
NCT02604342 (27) [back to overview]Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer-13 (EORTC QLQ-LC13) Over Time
NCT02604342 (27) [back to overview]Compliance of European Quality of Life (EuroQoL) 5 Dimension 5 Levels (EQ-5D-5L) Questionnaire Over Time
NCT02604342 (27) [back to overview]Compliance of European Quality of Life (EuroQoL) 5 Dimension 5 Levels (EQ-5D-5L) Questionnaire Over Time
NCT02604342 (27) [back to overview]Duration of Response (DOR) Using RECIST Version 1.1 as Assessed by Investigator and IRC
NCT02604342 (27) [back to overview]Percentage of Participants With Disease Control Using RECIST Version 1.1 as Assessed by Investigator and IRC
NCT02604342 (27) [back to overview]Percentage of Participants With Objective Response of CR or PR Using RECIST Version 1.1 as Assessed by Investigator and IRC
NCT02604342 (27) [back to overview]PFS in C-ITT Population Using RECIST Version 1.1 as Assessed by Investigator and IRC
NCT02604342 (27) [back to overview]Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC13 Score for C-ITT Population
NCT02604342 (27) [back to overview]Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC13 Score for ITT Population
NCT02604342 (27) [back to overview]Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC30 Score for C-ITT Population
NCT02624700 (3) [back to overview]The 2-year Survival Rate After Initial Study Treatment.
NCT02624700 (3) [back to overview]Number of Participants at Risk and Affected by Adverse Events (AEs)
NCT02624700 (3) [back to overview]The Duration of Progression-free Survival (PFS).
NCT02625298 (2) [back to overview]Changes Between Initial and Post Treatment Dimensions of Periapical Lesions
NCT02625298 (2) [back to overview]Presence of Clinical Symptoms
NCT02657434 (18) [back to overview]Percentage of Participants With Anti-Therapeutic Antibodies (ATAs) of Atezolizumab
NCT02657434 (18) [back to overview]Percentage of Participants With an Objective Response (Complete Response [CR] or Partial Response [PR]) Assessed by the Investigator Using RECIST V1.1
NCT02657434 (18) [back to overview]Minimum Observed Serum Atezolizumab Concentration (Cmin)
NCT02657434 (18) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by EORTC Quality-of-Life Lung Cancer Module (QLQ-LC13) Symptom Score
NCT02657434 (18) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality-of-Life Questionnaire-Core 30 (QLQ-C30) Symptom Score
NCT02657434 (18) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality-of-Life Questionnaire-Core 30 (QLQ-C30) Symptom Score
NCT02657434 (18) [back to overview]Duration of Response (DOR) as Determined by the Investigator Using RECIST v1.1
NCT02657434 (18) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms as Reported Using the Symptoms in Lung Cancer (SILC) Scale Score
NCT02657434 (18) [back to overview]Maximum Observed Serum Atezolizumab Concentration (Cmax)
NCT02657434 (18) [back to overview]Overall Survival (OS)
NCT02657434 (18) [back to overview]Overall Survival Rate at Year 1
NCT02657434 (18) [back to overview]Overall Survival Rate Year 2
NCT02657434 (18) [back to overview]Progression Free Survival (PFS) as Assessed by the Investigator Using Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1)
NCT02657434 (18) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by EORTC Quality-of-Life Lung Cancer Module (QLQ-LC13) Symptom Score
NCT02657434 (18) [back to overview]Plasma Concentrations for Pemetrexed in Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed)
NCT02657434 (18) [back to overview]Change From Baseline in Patient-Reported Lung Cancer Symptoms as Reported Using the Symptoms in Lung Cancer (SILC) Scale Score
NCT02657434 (18) [back to overview]Plasma Concentrations for Cisplatin in Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed)
NCT02657434 (18) [back to overview]Plasma Concentrations for Carboplatin in Arm A(Atezolizumab + Carboplatin or Cisplatin + Pemetrexed)
NCT02659059 (17) [back to overview]Number of Participants With Laboratory Abnormalities in Hepatic Tests - Part 2
NCT02659059 (17) [back to overview]Progression Free Survival (PFS) - Part 1
NCT02659059 (17) [back to overview]Overall Survival (OS) - Part 2
NCT02659059 (17) [back to overview]Overall Survival (OS) - Part 1
NCT02659059 (17) [back to overview]Objective Response Rate (ORR) - Part 2
NCT02659059 (17) [back to overview]Objective Response Rate (ORR) - Part 1
NCT02659059 (17) [back to overview]Number of Participants With Dose Limiting Toxicities (DLTs) - Part 2
NCT02659059 (17) [back to overview]Number of Participants With Adverse Events (AEs) - Part 2
NCT02659059 (17) [back to overview]Progression Free Survival (PFS) by Tumor Mutation Burden (TMB) Levels - Part 1
NCT02659059 (17) [back to overview]Progression Free Survival (PFS) - Part 2
NCT02659059 (17) [back to overview]Progression Free Survival (PFS) by PD-L1 Expression Levels - Part 1
NCT02659059 (17) [back to overview]Overall Survival (OS) by Tumor Mutation Burden (TMB) Levels - Part 1
NCT02659059 (17) [back to overview]Overall Survival (OS) by PD-L1 Expression Levels - Part 1
NCT02659059 (17) [back to overview]Objective Response Rate (ORR) by Tumor Mutation Burden (TMB) Levels - Part 1
NCT02659059 (17) [back to overview]Objective Response Rate (ORR) by PD-L1 Positive and Negative Levels - Part 1
NCT02659059 (17) [back to overview]Objective Response Rate (ORR) by PD-L1 Expression Levels-Part 1
NCT02659059 (17) [back to overview]Number of Participants With Laboratory Abnormalities in Thyroid Tests - Part 2
NCT02709512 (4) [back to overview]Overall Survival
NCT02709512 (4) [back to overview]Overall Survival Phase 3 Interim Analysis
NCT02709512 (4) [back to overview]Progression Free Survival
NCT02709512 (4) [back to overview]Response Rate
NCT02710396 (4) [back to overview]Number of Subjects With NSCLC Who Achieved DCB
NCT02710396 (4) [back to overview]Objective Response Rate (ORR)
NCT02710396 (4) [back to overview]Progression Free Survival (PFS)
NCT02710396 (4) [back to overview]Overall Survival (OS)
NCT02862457 (17) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab in Part B Cycles 1, 2, 4, 6, and 8
NCT02862457 (17) [back to overview]Maximum Concentration (Cmax) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Maximum Concentration (Cmax) of Pembrolizumab in Part B Cycle 1
NCT02862457 (17) [back to overview]Trough Concentration (Ctrough) of Pembrolizumab in Part A Cycles 1, 2, 4, 6, and 8
NCT02862457 (17) [back to overview]Trough Concentration (Ctrough) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Trough Concentration (Ctrough) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Time to Maximum Concentration (Tmax) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Maximum Concentration (Cmax) of Pembrolizumab in Part A Cycle 1
NCT02862457 (17) [back to overview]Time to Maximum Concentration (Tmax) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Terminal Half-Life (t1/2) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Terminal Half-Life (t1/2) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Maximum Concentration (Cmax) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Area Under the Concentration-Time Curve From Zero to the Time of the Last Measurable Concentration (AUC0-t) of Epacadostat in Part A
NCT02862457 (17) [back to overview]Number of Participants Who Experienced At Least One Adverse Event (AE)
NCT02862457 (17) [back to overview]Number of Participants Who Discontinued Study Treatment Due to An Adverse Event (AE)
NCT02862457 (17) [back to overview]Number of Participants Experiencing Dose-Limiting Toxicities (DLTs) According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (NCI-CTCAE v.4.0)
NCT02862457 (17) [back to overview]Area Under the Concentration-Time Curve From Zero to the Time of the Last Measurable Concentration (AUC0-t) of Epacadostat in Part A
NCT02864251 (6) [back to overview]Overall Survival (OS)
NCT02864251 (6) [back to overview]Objective Response Rate (ORR) by Blinded Independent Centralized Review (BICR)
NCT02864251 (6) [back to overview]Duration of Response (DOR) by Blinded Independent Centralized Review (BICR)
NCT02864251 (6) [back to overview]12 Month Progression Free Survival Rates (PFSR) by Blinded Independent Centralized Review (BICR)
NCT02864251 (6) [back to overview]9 Month Progression Free Survival Rates (PFSR) by Blinded Independent Centralized Review (BICR)
NCT02864251 (6) [back to overview]Progression Free Survival (PFS) by Blinded Independent Centralized Review (BICR)
NCT02899299 (7) [back to overview]Progression Free Survival (PFS)
NCT02899299 (7) [back to overview]Objective Response Rate (ORR) According to PD-L1 Expression Level
NCT02899299 (7) [back to overview]Disease Control Rate (DCR)
NCT02899299 (7) [back to overview]Objective Response Rate (ORR)
NCT02899299 (7) [back to overview]Overall Survival (OS)
NCT02899299 (7) [back to overview]Progression Free Survival (PFS) According to PD-L1 Expression Level
NCT02899299 (7) [back to overview]Overall Survival (OS) According to PD-L1 Expression Level
NCT02937116 (13) [back to overview]Area Under the Concentration-time Curve From Zero Time (Predose) to the Time of the Last Measurable Concentration (AUC0-t)
NCT02937116 (13) [back to overview]The Half-life (t1/2) of IBI308 in Plasma After Single Dose Administration
NCT02937116 (13) [back to overview]Time to Maximum Concentration (Tmax) of Sintilimab in Solid Tumor Participants
NCT02937116 (13) [back to overview]Clearance of IBI308 in Plasma After Single Dose Administration
NCT02937116 (13) [back to overview]DOR According to RECIST 1.1 as Assessed by Investigator
NCT02937116 (13) [back to overview]Maximum Concentration (Cmax) of Sintilimab in Solid Tumor Participants
NCT02937116 (13) [back to overview]TTR According to RECIST 1.1 as Assessed by Investigator
NCT02937116 (13) [back to overview]Number of Participants Experiencing Dose-limiting Toxicities (DLTs)
NCT02937116 (13) [back to overview]Volume of Distribution of IBI308 in Plasma After Single Dose Administration
NCT02937116 (13) [back to overview]PFS According to RECIST 1.1 as Assessed by Investigator
NCT02937116 (13) [back to overview]OS for Participants
NCT02937116 (13) [back to overview]Objective Response Rate (ORR) According to RECIST 1.1 as Assessed by Independent Review Committee by Investigator
NCT02937116 (13) [back to overview]Number of All Study Participants Who Demonstrate a Tumor Response
NCT02983045 (3) [back to overview]Part 3 Schedule Finding: Incidence of Dose-limiting Toxicity (DLT) During the DLT Evaluation Window
NCT02983045 (3) [back to overview]Part 2 and Part 4: Objective Response Rate (ORR) Per RECIST 1.1 at Recommended Phase 2 Dose (RP2D)
NCT02983045 (3) [back to overview]Part 1 Dose Escalation: Incidence of Dose-limiting Toxicity (DLT) During the DLT Evaluation Window
NCT02998528 (4) [back to overview]Major Pathologic Response (MPR) Rate
NCT02998528 (4) [back to overview]Event-Free Survival (EFS)
NCT02998528 (4) [back to overview]Time to Death or Distant Metastases (TTDM)
NCT02998528 (4) [back to overview]Pathologic Complete Response (pCR) Rate
NCT03003962 (44) [back to overview]OS at 24 Months in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]Alive and Progression-Free at 12 Months (APF12)
NCT03003962 (44) [back to overview]DoR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >=50% LREM Analysis Set
NCT03003962 (44) [back to overview]DoR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >=50% Analysis Set
NCT03003962 (44) [back to overview]DoR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >=25% LREM Analysis Set
NCT03003962 (44) [back to overview]APF12 in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]APF12 in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]APF12 in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]Time to Deterioration of EORTC QLQ-LC13 in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]Time to Deterioration of EORTC QLQ-LC13
NCT03003962 (44) [back to overview]Time to Deterioration of EORTC QLQ-C30 in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]Time to Deterioration of EORTC QLQ-C30
NCT03003962 (44) [back to overview]Percentage of Participants With Antidrug Antibody (ADA) Response to Durvalumab
NCT03003962 (44) [back to overview]Percentage of Participants With ADA Response to Durvalumab in LREM Analysis Set
NCT03003962 (44) [back to overview]Number of Participants With ECOG Performance Status in PD-L1 TC >=25% LREM Analysis Set
NCT03003962 (44) [back to overview]Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status
NCT03003962 (44) [back to overview]Change From Baseline in European Organization for Research and Treatment of Cancer (EORTC) 30-Item Core Quality of Life Questionnaire Version 3 (QLQ-C30)
NCT03003962 (44) [back to overview]Change From Baseline in EORTC QLQ-LC13 in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]Change From Baseline in EORTC QLQ-C30 in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]Change From Baseline in EORTC 13-Item Lung Cancer Quality of Life Questionnaire (QLQ-LC13)
NCT03003962 (44) [back to overview]Time From Randomization to Second Progression (PFS2)
NCT03003962 (44) [back to overview]Progression Free Survival (PFS) Based on Investigator Assessment According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1)
NCT03003962 (44) [back to overview]Duration of Response (DoR) as Per RECIST 1.1 Using Investigator Assessment
NCT03003962 (44) [back to overview]Objective Response Rate (ORR) as Per RECIST 1.1 Using Investigator Assessment
NCT03003962 (44) [back to overview]ORR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]ORR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]ORR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]OS at 18 Months
NCT03003962 (44) [back to overview]OS at 18 Months in PD-L1 TC > = 25% LREM Analysis Set
NCT03003962 (44) [back to overview]OS at 18 Months in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]OS at 18 Months in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]OS at 24 Months
NCT03003962 (44) [back to overview]OS at 24 Months in PD-L1 TC > = 25% LREM Analysis Set
NCT03003962 (44) [back to overview]OS at 24 Months in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]PFS2 in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]PFS2 in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]PFS2 in PD-L1 TC >= 25% LREM Analysis Set
NCT03003962 (44) [back to overview]PFS Based on Investigator Assessment According to RECIST 1.1 in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]PFS Based on Investigator Assessment According to RECIST 1.1 in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]PFS Based on Investigator Assessment According to RECIST 1.1 in LREM Analysis Set
NCT03003962 (44) [back to overview]Overall Survival (OS)
NCT03003962 (44) [back to overview]OS in PD-L1 TC >= 50% LREM Analysis Set
NCT03003962 (44) [back to overview]OS in PD-L1 TC >= 50% Analysis Set
NCT03003962 (44) [back to overview]OS in Participants With LREM
NCT03041181 (1) [back to overview]Number of Participants With Grade 3 or Grade 4 Adverse Events
NCT03085914 (3) [back to overview]Phases 1 and 2: Objective Response Rate (ORR)
NCT03085914 (3) [back to overview]Phases 1 & 2: Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious TEAEs
NCT03085914 (3) [back to overview]Phases 1 and 2: Number of Participants With Dose Limiting Toxicities (DLTs)
NCT03138889 (4) [back to overview]Objective Response Rate (ORR) Per Blinded Independent Central Review (BICR) by RECIST 1.1 of NKTR-214 Plus Pembrolizumab for Dose Expansion Cohorts 2 and 3.
NCT03138889 (4) [back to overview]Objective Response Rate (ORR) Per Investigator's Assessment by RECIST 1.1 of NKTR-214 at a Dose of 0.006 mg/kg With Pembrolizumab and Platinum-based Chemotherapy for Dose Expansion Cohorts 4+5.
NCT03138889 (4) [back to overview]Number of Participants Experiencing Dose-Limiting Toxicities in Dose Optimization Cohort 1a
NCT03138889 (4) [back to overview]Number of Participants With Treatment-Emergent Adverse Events [Safety and Tolerability] for Dose Optimization Cohort 1a.
NCT03164616 (13) [back to overview]PK of Tremelimumab; Peak and Trough Serum Concentrations
NCT03164616 (13) [back to overview]Pharmacokinetics (PK) of Durvalumab; Peak and Trough Serum Concentrations
NCT03164616 (13) [back to overview]Number of Patients With Anti-Drug Antibody (ADA) Response to Durvalumab
NCT03164616 (13) [back to overview]Number of Patients With ADA Response to Tremelimumab
NCT03164616 (13) [back to overview]Time From Randomization to Second Progression (PFS2)
NCT03164616 (13) [back to overview]Progression-Free Survival (PFS); D + SoC Compared With SoC Alone
NCT03164616 (13) [back to overview]PFS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC
NCT03164616 (13) [back to overview]Overall Survival (OS); D + SoC Compared With SoC Alone
NCT03164616 (13) [back to overview]OS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC
NCT03164616 (13) [back to overview]Duration of Response (DoR)
NCT03164616 (13) [back to overview]Time to Deterioration of PRO Symptoms, Assessed Using EORTC QLQ-Lung Cancer Module 13 (QLQ-LC13)
NCT03164616 (13) [back to overview]Time to Deterioration of Global Health Status / Health-Related Quality of Life (HRQoL) and Patient Reported Outcome (PRO) Symptoms, Assessed Using European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ)
NCT03164616 (13) [back to overview]Objective Response Rate (ORR)
NCT03215706 (8) [back to overview]Objective Response Rate (ORR) by BICR by PD-LI Tumor Cell Expression
NCT03215706 (8) [back to overview]Duration of Response (DoR)
NCT03215706 (8) [back to overview]Objective Response Rate (ORR) by BICR
NCT03215706 (8) [back to overview]Overall Survival (OS)
NCT03215706 (8) [back to overview]Progression Free Survival (PFS) by BICR
NCT03215706 (8) [back to overview]Time to Response (TTR)
NCT03215706 (8) [back to overview]OS by PD-L1 Tumor Cell Expression
NCT03215706 (8) [back to overview]PFS by BICR by PD-L1 Tumor Cell Expression
NCT03256136 (5) [back to overview]Duration Of Response
NCT03256136 (5) [back to overview]Progression Free Survival (PFS)
NCT03256136 (5) [back to overview]Disease Control Rate (DCR), Presented in Numbers of Participants
NCT03256136 (5) [back to overview]Overall Survival (OS)
NCT03256136 (5) [back to overview]Objective Response Rate (ORR), Presented in Numbers of Participants
NCT03307785 (158) [back to overview]Part A: Area Under the Plasma Concentration From Time Zero to Infinity (AUC[0-infinity]) of Niraparib
NCT03307785 (158) [back to overview]Part A: AUC at Steady State (AUCss) of Niraparib
NCT03307785 (158) [back to overview]Part A: AUC(0-infinity) of TSR-042
NCT03307785 (158) [back to overview]Part A: Cmax of TSR-042
NCT03307785 (158) [back to overview]Part A: Ctau of TSR-042
NCT03307785 (158) [back to overview]Part A: Disease Control Rate
NCT03307785 (158) [back to overview]Part A: Duration of Response
NCT03307785 (158) [back to overview]Part A: Maximum Observed Plasma (Cmax) of Niraparib
NCT03307785 (158) [back to overview]Part A: Number of Participants With Dose-limiting Toxicity (DLT)
NCT03307785 (158) [back to overview]Part A: Number of Participants With Positive Anti-TSR-042 Antibodies
NCT03307785 (158) [back to overview]Part A: Objective Response Rate
NCT03307785 (158) [back to overview]Part A: Observed Concentration at the End of the Dosing Interval (Ctau) of Niraparib
NCT03307785 (158) [back to overview]Part A: Progression-free Survival
NCT03307785 (158) [back to overview]Part A: Time to Reach Maximum Plasma Concentration (Tmax) of Niraparib
NCT03307785 (158) [back to overview]Part A: Tmax of TSR-042
NCT03307785 (158) [back to overview]Part B: AUC(0-infinity) of TSR-042
NCT03307785 (158) [back to overview]Part B: Cmax of TSR-042
NCT03307785 (158) [back to overview]Part B: Ctau of TSR-042
NCT03307785 (158) [back to overview]Part B: Disease Control Rate
NCT03307785 (158) [back to overview]Part B: Number of Participants With DLT
NCT03307785 (158) [back to overview]Part B: Number of Participants With Positive Anti-TSR-042 Antibodies
NCT03307785 (158) [back to overview]Part B: Objective Response Rate
NCT03307785 (158) [back to overview]Part F: Number of Participants With Non-serious TEAEs, STEAEs and AESIs
NCT03307785 (158) [back to overview]Part E: Number of Participants With Non-serious TEAEs, STEAEs and AESIs
NCT03307785 (158) [back to overview]Part D: Vz of TSR-042
NCT03307785 (158) [back to overview]Part D: Vss of TSR-042
NCT03307785 (158) [back to overview]Part D: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part D: Number of Participants With Non-serious TEAEs, STEAEs and AESIs
NCT03307785 (158) [back to overview]Part D: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part D: CL of TSR-042
NCT03307785 (158) [back to overview]Part D: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part D: AUC0-t of TSR-042
NCT03307785 (158) [back to overview]Part C: Vz/F of Niraparib
NCT03307785 (158) [back to overview]Part C: Vz of TSR-042
NCT03307785 (158) [back to overview]Part C: Vss of TSR-042
NCT03307785 (158) [back to overview]Part C: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part C: Tmax,ss of Niraparib
NCT03307785 (158) [back to overview]Part C: Number of Participants With Non-serious TEAEs, STEAEs and AESIs
NCT03307785 (158) [back to overview]Part C: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part C: Ctau,ss of Niraparib
NCT03307785 (158) [back to overview]Part C: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part C: Cmax,ss of Niraparib
NCT03307785 (158) [back to overview]Part C: CL/F of Niraparib
NCT03307785 (158) [back to overview]Part C: CL of TSR-042
NCT03307785 (158) [back to overview]Part C: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part C: AUC0-t of TSR-042
NCT03307785 (158) [back to overview]Part C: AUC0-t of Niraparib
NCT03307785 (158) [back to overview]Part B: Vz of TSR-042
NCT03307785 (158) [back to overview]Part B: Vss of TSR-042
NCT03307785 (158) [back to overview]Part B: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part B: Number of Participants With Non-serious TEAEs, STEAEs and AESIs
NCT03307785 (158) [back to overview]Part B: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part B: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part B: CL of TSR-042
NCT03307785 (158) [back to overview]Part B: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part B: AUC0-t of TSR-042
NCT03307785 (158) [back to overview]Part A: Volume of Distribution After Oral Administration (Vz/F) of Niraparib
NCT03307785 (158) [back to overview]Part A: Volume of Distribution After Oral Administration (Vz/F) of Niraparib
NCT03307785 (158) [back to overview]Part A: Volume of Distribution After Intravenous Administration (Vz) of of TSR-042
NCT03307785 (158) [back to overview]Part A: Volume of Distribution After Intravenous Administration (Vz) of of TSR-042
NCT03307785 (158) [back to overview]Part C: Ctau of TSR-042
NCT03307785 (158) [back to overview]Part C: Ctau of Niraparib
NCT03307785 (158) [back to overview]Part C: Cmax of TSR-042
NCT03307785 (158) [back to overview]Part C: Cmax of Niraparib
NCT03307785 (158) [back to overview]Part F: AUC0-t of TSR-042
NCT03307785 (158) [back to overview]Part F: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part F: CL of TSR-022
NCT03307785 (158) [back to overview]Part F: CL of TSR-042
NCT03307785 (158) [back to overview]Part F: Cmax of TSR-022
NCT03307785 (158) [back to overview]Part F: Cmax of TSR-042
NCT03307785 (158) [back to overview]Part F: Cmax,ss of TSR-022
NCT03307785 (158) [back to overview]Part C: AUCss of Niraparib
NCT03307785 (158) [back to overview]Part C: AUC(0-infinity) of TSR-042
NCT03307785 (158) [back to overview]Part C: AUC(0-infinity) of Niraparib
NCT03307785 (158) [back to overview]Part B: Tmax of TSR-042
NCT03307785 (158) [back to overview]Part B: Progression-free Survival
NCT03307785 (158) [back to overview]Part A: Tmax at Steady State (Tmax,ss) of Niraparib
NCT03307785 (158) [back to overview]Part A: Tmax at Steady State (Tmax,ss) of Niraparib
NCT03307785 (158) [back to overview]Part A: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part A: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part A: Volume of Distribution After Intravenous Administration (Vss) of of TSR-042
NCT03307785 (158) [back to overview]Part A: Volume of Distribution After Intravenous Administration (Vss) of of TSR-042
NCT03307785 (158) [back to overview]Part F: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part F: Ctau of TSR-022
NCT03307785 (158) [back to overview]Part F: Ctau of TSR-042
NCT03307785 (158) [back to overview]Part F: Ctau,ss of TSR-022
NCT03307785 (158) [back to overview]Part F: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part F: AUCss of TSR-022
NCT03307785 (158) [back to overview]Part F: Disease Control Rate
NCT03307785 (158) [back to overview]Part F: Duration of Response
NCT03307785 (158) [back to overview]Part F: Number of Participants With DLT
NCT03307785 (158) [back to overview]Part F: Number of Participants With Positive Anti-TSR-022 Antibodies
NCT03307785 (158) [back to overview]Part F: Number of Participants With Positive Anti-TSR-042 Antibodies
NCT03307785 (158) [back to overview]Part F: Objective Response Rate
NCT03307785 (158) [back to overview]Part F: AUC0-t of TSR-022
NCT03307785 (158) [back to overview]Part F: AUC(0-infinity) of TSR-042
NCT03307785 (158) [back to overview]Part F: AUC(0-infinity) of TSR-022
NCT03307785 (158) [back to overview]Part E: Vz of TSR-042
NCT03307785 (158) [back to overview]Part E: Vss of TSR-042
NCT03307785 (158) [back to overview]Part E: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part E: Tmax of TSR-042
NCT03307785 (158) [back to overview]Part E: Progression-free Survival
NCT03307785 (158) [back to overview]Part E: Objective Response Rate
NCT03307785 (158) [back to overview]Part E: Number of Participants With Positive Anti-TSR-042 Antibodies
NCT03307785 (158) [back to overview]Part E: Number of Participants With DLT
NCT03307785 (158) [back to overview]Part E: Duration of Response
NCT03307785 (158) [back to overview]Part E: Disease Control Rate
NCT03307785 (158) [back to overview]Part E: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part E: Ctau of TSR-042
NCT03307785 (158) [back to overview]Part E: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part E: Cmax of TSR-042
NCT03307785 (158) [back to overview]Part E: CL of TSR-042
NCT03307785 (158) [back to overview]Part E: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part E: AUC0-t of TSR-042
NCT03307785 (158) [back to overview]Part E: AUC(0-infinity) of TSR-042
NCT03307785 (158) [back to overview]Part D: Tmax of TSR-042
NCT03307785 (158) [back to overview]Part F: Progression-free Survival
NCT03307785 (158) [back to overview]Part F: Tmax of TSR-022
NCT03307785 (158) [back to overview]Part F: Tmax of TSR-042
NCT03307785 (158) [back to overview]Part F: Tmax,ss of TSR-022
NCT03307785 (158) [back to overview]Part F: Tmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part F: Vss of TSR-022
NCT03307785 (158) [back to overview]Part F: Vss of TSR-042
NCT03307785 (158) [back to overview]Part F: Vz of TSR-022
NCT03307785 (158) [back to overview]Part F: Vz of TSR-042
NCT03307785 (158) [back to overview]Part A: Area Under the Plasma Concentration From Time Zero to t (AUC[0-t]) of Niraparib
NCT03307785 (158) [back to overview]Part A: AUC(0-t) of TSR-042
NCT03307785 (158) [back to overview]Part A: AUC(0-t) of TSR-042
NCT03307785 (158) [back to overview]Part A: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part A: AUCss of TSR-042
NCT03307785 (158) [back to overview]Part A: Clearance After Intravenous Administration (CL) of TSR-042
NCT03307785 (158) [back to overview]Part A: Clearance After Intravenous Administration (CL) of TSR-042
NCT03307785 (158) [back to overview]Part A: Clearance After Oral Administration (CL/F) of Niraparib
NCT03307785 (158) [back to overview]Part A: Clearance After Oral Administration (CL/F) of Niraparib
NCT03307785 (158) [back to overview]Part A: Cmax at Steady State (Cmax,ss) of Niraparib
NCT03307785 (158) [back to overview]Part D: Progression-free Survival
NCT03307785 (158) [back to overview]Part D: Objective Response Rate
NCT03307785 (158) [back to overview]Part D: Number of Participants With Positive Anti-TSR-042 Antibodies
NCT03307785 (158) [back to overview]Part D: Number of Participants With DLT
NCT03307785 (158) [back to overview]Part D: Disease Control Rate
NCT03307785 (158) [back to overview]Part D: Ctau of TSR-042
NCT03307785 (158) [back to overview]Part D: Cmax of TSR-042
NCT03307785 (158) [back to overview]Part D: AUC(0-infinity) of TSR-042
NCT03307785 (158) [back to overview]Part C: Tmax of TSR-042
NCT03307785 (158) [back to overview]Part A: Cmax at Steady State (Cmax,ss) of Niraparib
NCT03307785 (158) [back to overview]Part A: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part A: Cmax,ss of TSR-042
NCT03307785 (158) [back to overview]Part A: Ctau at Steady State (Ctau,ss) of Niraparib
NCT03307785 (158) [back to overview]Part A: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part A: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part A: Number of Participants With Non-serious Treatment-emergent Adverse Events (TEAEs), Serious TEAEs (STEAEs) and Adverse Events of Special Interest (AESIs)
NCT03307785 (158) [back to overview]Part D: Ctau,ss of TSR-042
NCT03307785 (158) [back to overview]Part C: Tmax of Niraparib
NCT03307785 (158) [back to overview]Part C: Progression-free Survival
NCT03307785 (158) [back to overview]Part C: Objective Response Rate
NCT03307785 (158) [back to overview]Part C: Number of Participants With Positive Anti-TSR-042 Antibodies
NCT03307785 (158) [back to overview]Part C: Number of Participants With DLT
NCT03307785 (158) [back to overview]Part C: Disease Control Rate
NCT03317496 (13) [back to overview]Absolute Value of Tumor Mutational Burden (TMB) in Tumor Tissue
NCT03317496 (13) [back to overview]Duration of Response (DOR) as Per RECIST v 1.1 by Investigator Assessment
NCT03317496 (13) [back to overview]Number of Participants With Grade 3 or Higher TEAEs Based on National Cancer Institute Common Toxicity Criteria for Adverse Events (NCI CTCAE) v 4.03
NCT03317496 (13) [back to overview]Number of Participants With Treatment Related TEAEs
NCT03317496 (13) [back to overview]Overall Survival (OS)
NCT03317496 (13) [back to overview]Percentage of Participants With Confirmed Objective Response (OR) as Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1 by Investigator Assessment
NCT03317496 (13) [back to overview]Phase 1b Lead-in: Number of Participants With Dose-Limiting Toxicities (DLT)
NCT03317496 (13) [back to overview]Progression Free Survival (PFS) as Per RECIST v 1.1 by Investigator Assessment
NCT03317496 (13) [back to overview]Time-to-Tumor Response (TTR) as Per RECIST v 1.1 by Investigator Assessment
NCT03317496 (13) [back to overview]Number of Participants With Grade 3 or Higher Laboratory Abnormalities by CTCAE Grade
NCT03317496 (13) [back to overview]Number of Participants With Programmed Death-Ligand 1 (PD-L1) Expression
NCT03317496 (13) [back to overview]Number of Participants With Treatment Emergent Adverse Events (TEAEs) and Serious TEAEs
NCT03317496 (13) [back to overview]Serum Concentration of Avelumab
NCT03607539 (6) [back to overview]Time to Response (TTR) by IRRC Assessment
NCT03607539 (6) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 as Assessed by Independent Radiographic Review Committee (IRRC)
NCT03607539 (6) [back to overview]Duration of Response (DOR) by IRRC Assessment
NCT03607539 (6) [back to overview]Overall Survival (OS)
NCT03607539 (6) [back to overview]Objective Response Rate (ORR) by IRRC Assessment
NCT03607539 (6) [back to overview]Disease Control Rate (DCR) by IRRC Assessment
NCT03631784 (4) [back to overview]Number of Participants Who Discontinued From Study Treatment Due to an AE
NCT03631784 (4) [back to overview]Percentage of Participants Who Developed Grade 3 or Higher Pneumonitis
NCT03631784 (4) [back to overview]Number of Participants Who Experienced an Adverse Event (AE)
NCT03631784 (4) [back to overview]Overall Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1)
NCT03664024 (6) [back to overview]Percentage of Participants Discontinuing Study Intervention Due to an AE.
NCT03664024 (6) [back to overview]Percentage of Participants Who Experienced One or More Adverse Events (AEs)
NCT03664024 (6) [back to overview]Overall Survival (OS)
NCT03664024 (6) [back to overview]Objective Response Rate
NCT03664024 (6) [back to overview]Progression Free Survival (PFS)
NCT03664024 (6) [back to overview]Tumor Mutation Burden (TMB) in Cell-free Circulating Tumor Deoxyribonucleic Acid (ctDNA)
NCT03713944 (5) [back to overview]Number of Participants With Adverse Events
NCT03713944 (5) [back to overview]Disease Control Rate
NCT03713944 (5) [back to overview]Overall Response Rate
NCT03713944 (5) [back to overview]Overall Survival
NCT03713944 (5) [back to overview]Progression Free Survival
NCT03737994 (12) [back to overview]Objective Response Rate (ORR), Per Investigator Assessment Using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 Criteria
NCT03737994 (12) [back to overview]Progression-free Survival (PFS), Per Investigator Assessment Using RECIST v1.1 Criteria
NCT03737994 (12) [back to overview]Overall Survival (OS)
NCT03737994 (12) [back to overview]Progression-free Survival (PFS), Per Investigator Assessment Using RECIST v1.1 Criteria
NCT03737994 (12) [back to overview]Objective Response Rate (ORR), Per Investigator Assessment Using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 Criteria
NCT03737994 (12) [back to overview]Progression-free Survival (PFS), Per Investigator Assessment Using RECIST v1.1 Criteria
NCT03737994 (12) [back to overview]Objective Response Rate (ORR), Per Investigator Assessment Using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 Criteria
NCT03737994 (12) [back to overview]Duration of Overall Response, Per Investigator Assessment Using RECIST v1.1
NCT03737994 (12) [back to overview]Duration of Overall Response, Per Investigator Assessment Using RECIST v1.1
NCT03737994 (12) [back to overview]Number of Participants by Highest Grade Adverse Event Reported
NCT03737994 (12) [back to overview]Overall Survival (OS)
NCT03737994 (12) [back to overview]Overall Survival (OS)
NCT03775486 (11) [back to overview]Progression-free Survival
NCT03775486 (11) [back to overview]Time to Deterioration in European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-Lung Cancer (LC)13
NCT03775486 (11) [back to overview]Time to Deterioration in EORTC Quality of Life Questionnaire (QLQ) QLQ-C30
NCT03775486 (11) [back to overview]Progression-free Survival in Homologous Recombination Repair Related Gene Mutation (HRRm) Population
NCT03775486 (11) [back to overview]Presence of Anti-drug Antibodies (ADAs) for Durvalumab
NCT03775486 (11) [back to overview]Overall Survival
NCT03775486 (11) [back to overview]Change From Baseline in European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-Lung Cancer (LC)13
NCT03775486 (11) [back to overview]Duration of Response
NCT03775486 (11) [back to overview]Concentration of Durvalumab
NCT03775486 (11) [back to overview]Change From Baseline in EORTC Quality of Life Questionnaire (QLQ) QLQ-C30
NCT03775486 (11) [back to overview]Number of Participants With Treatment-Related Adverse Events
NCT03840915 (7) [back to overview]Number of Participants With Positive Antidrug Antibodies (ADA)
NCT03840915 (7) [back to overview]Number of Participants With Dose-Limiting Toxicities (DLTs)
NCT03840915 (7) [back to overview]Duration of Response (DOR)
NCT03840915 (7) [back to overview]Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious TEAEs
NCT03840915 (7) [back to overview]Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Investigator (IRC)
NCT03840915 (7) [back to overview]Overall Survival (OS)
NCT03840915 (7) [back to overview]Progression-Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors (RECIST Version 1.1) Assessed by Investigator
NCT03850444 (11) [back to overview]Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)
NCT03850444 (11) [back to overview]Number of Participants Who Experienced At Least One Adverse Event (AE)
NCT03850444 (11) [back to overview]Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%
NCT03850444 (11) [back to overview]Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%
NCT03850444 (11) [back to overview]Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%
NCT03850444 (11) [back to overview]Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥1%
NCT03850444 (11) [back to overview]Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥20%
NCT03850444 (11) [back to overview]Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥50%
NCT03850444 (11) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%
NCT03850444 (11) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%
NCT03850444 (11) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%
NCT03950674 (7) [back to overview]Overall Response Rate (ORR) Per RECIST 1.1 as Assessed by Blinded Central Imaging
NCT03950674 (7) [back to overview]Duration of Response (DOR) Per RECIST 1.1 as Assessed by Blinded Central Imaging
NCT03950674 (7) [back to overview]Number of Participants Who Experienced an Adverse Event (AE)
NCT03950674 (7) [back to overview]Overall Survival (OS)
NCT03950674 (7) [back to overview]Progression-Free Survival (PFS) as Assessed by Investigator Immune-related RECIST (irRECIST) Response Criteria
NCT03950674 (7) [back to overview]Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 as Assessed by Blinded Central Imaging
NCT03950674 (7) [back to overview]Number of Participants Who Discontinued Any Study Drug Due to an AE
NCT03971474 (8) [back to overview]Disease Control Rate (DCR)
NCT03971474 (8) [back to overview]Duration of Response (DOR)
NCT03971474 (8) [back to overview]Investigator Assessed-progression-free Survival (IA-PFS)
NCT03971474 (8) [back to overview]Overall Survival (OS), Subgroup Analysis by Stratification Factors
NCT03971474 (8) [back to overview]Number of Participants With Gr 3 Through 5 Adverse Events That Are Related to Study Drugs
NCT03971474 (8) [back to overview]Investigator-Assessed Progression-Free Survival (IA-PFS), Subgroup Analysis by Stratification Factors
NCT03971474 (8) [back to overview]Response Rate (RR)
NCT03971474 (8) [back to overview]Overall Survival (OS)
NCT04012619 (1) [back to overview]the Maximum Tolerated Dose (MTD)
NCT04372927 (4) [back to overview]Response Rate
NCT04372927 (4) [back to overview]Frequency and Severity of Pneumonitis
NCT04372927 (4) [back to overview]Frequency of Adverse Events
NCT04372927 (4) [back to overview]Overall Survival (OS)
NCT04816214 (15) [back to overview]Run-in Part: Number of Participants With Dose Limiting Toxicities (DLTs)
NCT04816214 (15) [back to overview]Run-in Part: Maximum Plasma Concentration (Cmax) of Osimertinib and Its Metabolites (AZ5104 and AZ7550)
NCT04816214 (15) [back to overview]Run-in Part: Disease Control Rate (DCR) as Per Investigator Assessment
NCT04816214 (15) [back to overview]Run-in Part: Duration of Response (DOR) as Per Investigator Assessment
NCT04816214 (15) [back to overview]Run-in Part: Time to Response (TTR) as Per Investigator Assessment
NCT04816214 (15) [back to overview]Run-in Part: Median Duration of Exposure to Each Study Drug
NCT04816214 (15) [back to overview]Run-in Part: Number of Participants With at Least One Dose Interruption and Dose Reduction of Each Study Drug
NCT04816214 (15) [back to overview]Run-in Part: Time to Maximum Plasma Concentration (Tmax) of Capmatinib
NCT04816214 (15) [back to overview]Run-in Part: Time to Maximum Plasma Concentration (Tmax) of Osimertinib and Its Metabolites (AZ5104 and AZ7550)
NCT04816214 (15) [back to overview]Run-in Part: Maximum Plasma Concentration (Cmax) of Capmatinib
NCT04816214 (15) [back to overview]Run-in Part: Progression-Free Survival (PFS) as Per Investigator Assessment
NCT04816214 (15) [back to overview]Run-in Part: Dose Intensity of Each Study Drug
NCT04816214 (15) [back to overview]Run-in Part: Overall Response Rate (ORR) as Per Investigator Assessment
NCT04816214 (15) [back to overview]Run-in Part: Area Under the Curve to the Last Measurable Concentration (AUClast) of Osimertinib and Its Metabolites (AZ5104 and AZ7550)
NCT04816214 (15) [back to overview]Run-in Part: Area Under the Curve to the Last Measurable Concentration (AUClast) of Capmatinib

Overall Tumor Response

Best overall (confirmed) response recorded from start of treatment until disease progression/recurrence, start of other anti-tumor therapy/intervention, or end of trial, whichever comes first. Response must be confirmed at least 6 weeks from previous scans. Best overall response assignment depends on both measurement and confirmation criteria. (NCT00063570)
Timeframe: Every 6 weeks from start of treatment until documented disease progression or for 6 months from last dose of study drug, whichever occurs first. After 6 months, clinical assessment every 12 weeks and radiologic test performed as clinically indicated

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
21-Day Schedule051060
Bi-Weekly Schedule2826142

[back to top]

Time to Treatment Failure

Time to treatment failure is calculated as (Date of First Disease Progression, Death as a Result of any Cause, or Early Discontinuation of Treatment Due to Adverse Event or Physician Perception of Lack of Efficacy or Patient and Physician Perception of Lack of Efficacy, whichever Comes First - First Dose Date + 1)/ (365.25/12) (NCT00063570)
Timeframe: Every 6 weeks from start of treatment until documented disease progression or for 6 months from last dose of study drug, whichever occurs first. After 6 months, clinical assessment every 12 weeks and radiologic test performed as clinically indicated

Interventionmonths (Median)
Bi-Weekly Schedule2.79
21-Day Schedule2.56

[back to top]

Time to Progressive Disease

Time to progressive disease is calculated as (Date of First Disease Progression or Death Due to Disease under Study whichever Comes First - First Dose Date + 1)/(365.25/12). (NCT00063570)
Timeframe: Every 6 weeks from start of treatment until documented disease progression or for 6 months from last dose of study drug, whichever occurs first. After 6 months, clinical assessment every 12 weeks and radiologic test performed as clinically indicated

Interventionmonths (Median)
Bi-Weekly Schedule3.19
21-Day Schedule4.01

[back to top]

Overall Survival

Overall survival time is calculated as (Date of Death as a Result of any Cause - First Dose Date + 1)/ (365.25/12). (NCT00063570)
Timeframe: Every 6 weeks from start of treatment until documented disease progression or for 6 months from last dose of study drug, whichever occurs first. After 6 months, clinical assessment every 12 weeks and radiologic test performed as clinically indicated

Interventionmonths (Median)
Bi-Weekly Schedule13.44
21-Day Schedule16.20

[back to top]

Duration of (Confirmed) Complete Response or Partial Response

Duration of response is calculated as (Date of First Disease Progression or Death as a Result of any Cause whichever Comes First - Date of First Objective Status Assessment of Confirmed CR or PR + 1)/(365.25/12). (NCT00063570)
Timeframe: Every 6 weeks from start of treatment until documented disease progression or for 6 months from last dose of study drug, whichever occurs first. After 6 months, clinical assessment every 12 weeks and radiologic test performed as clinically indicated

Interventionmonths (Median)
Bi-Weekly Schedule5.85
21-Day Schedule4.17

[back to top]

Pathological Complete Response

"Number of participants with results of pathological review that indicated a complete response. Pathological complete response should be evaluated at the time of surgery (Extrapleural Pneumonectomy [EPP]).~Resected tissue or pleural fluid should be sent for pathological and histological evaluation." (NCT00087698)
Timeframe: Surgery (at least 3 weeks post last dose of chemotherapy, up to a maximum interval of 8 weeks)

Interventionparticipants (Number)
Pemetrexed3

[back to top]

Time to Progressive Disease

Number of months between the first dose date and the date of first disease progression or death as a result of any cause, whichever comes first. (NCT00087698)
Timeframe: baseline to measured progressive disease

Interventionmonths (Mean)
Pemetrexed14.98

[back to top]

Overall Tumor Response

The frequency of best overall tumor response summarized by response category. The best (unconfirmed) response recorded from the start of chemotherapy treatment until disease progression/recurrence, start of any further anti-tumor therapy, or time of surgery whichever comes first. (NCT00087698)
Timeframe: baseline to measured progressive disease

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown/Unavailable
Pemetrexed12436511

[back to top]

The 1 and 2 Year Disease-Free Survival Rate (Percentage)

Kaplan-Meier estimates of the percentage of participants still alive at 1-year and 2-years, based upon the total number of participants who had surgery. (NCT00087698)
Timeframe: 1 year and 2 years

Interventionpercentage of participants (Mean)
1-Year Disease-Free Rate2-Year Disease-Free Rate
Pemetrexed61.4033.24

[back to top]

Time to Treatment Failure

Time to relapse (treatment failure) is measured in months and calculated as (Date of first surgery - Date of first relapse after surgery + 1)/(365.25/12). Time to relapse will be censored at the date of the last visit or start date of further anti-tumor therapy or intervention, whichever comes first. (NCT00087698)
Timeframe: baseline to stopping treatment

Interventionmonths (Mean)
Pemetrexed18.30

[back to top]

Overall Survival Time

Number of months between the first dose date and the date of death as a result of any cause. Overall survival time calculated as (Date of death - First dose date + 1)/(365.25/12). (NCT00087698)
Timeframe: baseline to date of death from any cause

Interventionmonths (Mean)
Pemetrexed21.85

[back to top]

Duration of Overall Response (OR)

The duration of response, in participants with best OR of complete response (CR) or partial response (PR), was measured from the date criteria are met for CR/PR (not confirmation date, whichever was first recorded), until the first occurrence date that the criteria of progressive disease (PD) was met, or death. Participants who were alive and without progression were censored at the date of their last independent review committee (IRC) tumor assessment. The tumor response and progression were assessed by the IRC in the Pemetrexed group and by the investigator in the Docetaxel group. (NCT00095199)
Timeframe: Time of first occurrence of either (PR) or (CR) to the first date of progressive disease or death up to 32.5 months

Interventionmonths (Median)
Cetuximab & Pemetrexed4.17
Pemetrexed6.93
Cetuximab & Docetaxel5.36
Docetaxel5.39

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to death. Participants without a date of death were censored on the last date participants were known to be alive, or lost to follow-up. (NCT00095199)
Timeframe: Randomization to the date of death from any cause up to 72.8 months

Interventionmonths (Median)
Cetuximab & Pemetrexed6.93
Pemetrexed7.79
Cetuximab & Docetaxel5.75
Docetaxel8.15

[back to top]

Number of Participants With Common Toxicity Criteria (CTC) Grade 3 or 4 Toxicities

National Cancer Institutes-Common Toxicity Criteria version 3.0 was used by investigators to assess participant toxicities. Mapping of investigator verbatim terms to CTCAE terms was done by the sponsor/designee using CTCAE v4.0. Participants reported had grade 3 or 4 toxicities (or both potentially). Grade 3 AEs: severe or medically significant but not immediately life-threatening;hospitalization or prolongation of hospitalization indicated; disabling; limiting self care activities of daily living. Grade 4 AEs: life-threatening consequences; urgent intervention indicated. (NCT00095199)
Timeframe: Time from first dose to 30 days after last dose of study therapy up to 28.3 months for Cetuximab & Pemetrexed (versus Pemetrexed alone) and up to 54.3 months for Cetuximab + Docetaxel (versus Docetaxel alone)

Interventionparticipants (Number)
Cetuximab & Pemetrexed179
Pemetrexed143
Cetuximab + Docetaxel112
Docetaxel97

[back to top]

Percentage of Participants With Symptomatic Response (Symptom Response Rates) Using the Lung Cancer Subscale (LCS) Scores of Functional Assessment of Cancer Therapy for Participants With Lung Cancer (FACT-L)

The FACT-LCS is a set of 7 questions to inventory problems specific to lung cancer symptoms. Participants rate each item on a 5-point Likert-type scale from 0 (not at all) to 4 (very much). Scores range from 0-28 and higher score indicates fewer symptoms. Symptom response (improvement) was defined as ≥2 point increase from baseline in the 7-item LCS score that was maintained for 2 consecutive assessments at least 3 weeks, and not >5 weeks apart for participants, whose baseline LCS score was ≤26. Symptom response rate was the percentage of participants with symptomatic response. (NCT00095199)
Timeframe: At baseline, every 3 weeks and 30 days after end of therapy up to 50 months

Interventionpercentage of participants (Number)
Cetuximab & Pemetrexed17.1
Pemetrexed22.9
Cetuximab & Docetaxel17.3
Docetaxel13.5

[back to top]

Progression Free Survival (PFS)

PFS was defined as the time from randomization until the date of progressive disease (PD) or death from any cause. Participants who were alive and without progression were censored at the date of their last tumor assessment. PFS was assessed by the independent review committee (IRC) in the Pemetrexed group (Cetuximab & Pemetrexed versus Pemetrexed) and by the investigator in the Docetaxel group (Cetuximab & Docetaxel versus Docetaxel). (NCT00095199)
Timeframe: Randomization to progression of disease or death due to any cause up to 59.6 months

Interventionmonths (Median)
Cetuximab & Pemetrexed2.89
Pemetrexed2.76
Cetuximab & Docetaxel2.37
Docetaxel1.54

[back to top]

Proportion of Randomized Participants With Best Overall Response (OR) of Partial Response (PR), Complete Response (CR), or Stable Disease (SD)

The disease control rate (DCR) was the proportion of randomized participants with a best OR of CR, PR or SD according to modified World Health Organization (WHO) guidelines. It was calculated as the total number of participants with CR, PR or SD divided by the total number of participants randomized in that arm. The tumor response was assessed by the independent review committee (IRC) in the Pemetrexed group and by the investigator in the Docetaxel group. (NCT00095199)
Timeframe: Randomization to progression of disease or death due to any cause up to 59.6 months

Interventionproportion of participants (Number)
Cetuximab & Pemetrexed0.522
Pemetrexed0.480
Cetuximab & Docetaxel0.335
Docetaxel0.253

[back to top]

Proportion of Randomized Participants With the Best Overall Response (OR) of Partial Response (PR) or Complete Response (CR) (Overall Response Rate [ORR])

The best overall response rate (ORR) was the proportion of randomized participants with a best OR of CR or PR, according to modified World Health Organization (WHO) guidelines. It was calculated as the total number of participants with CR or PR divided by the total number of participants treated in that arm. Participants with no post-baseline evaluation were considered as non-responders. The tumor response was assessed by the independent review committee (IRC) in the Pemetrexed group and by the investigator in the Docetaxel group. (NCT00095199)
Timeframe: Randomization until progression of disease or death from any cause up to 59.6 months

Interventionproportion of participants (Number)
Cetuximab & Pemetrexed0.066
Pemetrexed0.043
Cetuximab & Docetaxel0.078
Docetaxel0.066

[back to top]

Time to Symptomatic Progression

The FACT-LCS (see description in Outcome measure 5) inventories problems specific to lung cancer symptoms. Using this Scale, Symptom progression = a ≥ 2 point decrease from baseline in LCS score maintained for 2 consecutive assessments ≥3 weeks, and <5 weeks, apart. The symptom progression date = the first of 2 consecutive assessments with a ≥2 point decline. Time to symptomatic progression = the time from randomization to the symptom progression date. For participants with no symptom progression, time to symptomatic progression was censored the date of last symptom assessment. (NCT00095199)
Timeframe: Randomization until symptomatic progression up to 48.3 months

Interventionmonths (Median)
Cetuximab & PemetrexedNA
PemetrexedNA
Cetuximab & DocetaxelNA
DocetaxelNA

[back to top]

Overall Survival

Time from randomization to death. Patients alive at last follow-up were censored. (NCT00101283)
Timeframe: Assessed every 3 months for 2 years, then every 6 months for 1 year

InterventionMonths (Median)
Pemetrexed/Carboplatin13.0
Pemetrexed/Gemcitabine6.0

[back to top]

Best Overall Response by RECIST Criteria (Version 1.0)

Number of eligible, treated participants in each response category by RECIST criteria. Response categories represent best response for each patient prior to progression. (NCT00101283)
Timeframe: Assessed every 2 cycles (6 weeks) while on treatment, then every 3 months for 2 years, then every 6 months for 1 year until disease progression

,
Interventioneligible, treated participants (Number)
Partial ResponseStable DiseaseProgressionUnevaluable
Pemetrexed/Carboplatin3751
Pemetrexed/Gemcitabine0652

[back to top]

Progression-Free Survival

Time from randomization to the earlier of disease progression or death. Patients alive and progression-free at last follow-up were censored. (NCT00101283)
Timeframe: Assessed every 3 months for 2 years, then every 6 months for 1 year

InterventionMonths (Median)
Pemetrexed/Carboplatin4.1
Pemetrexed/Gemcitabine3.3

[back to top]

Time to Worsening of Symptoms (TWS)

TWS was the elapsed time from the date of randomization to the first date of worsening [defined as a 15-millimeter (mm) increase from baseline based on a 100-mm scale] of each symptom and summary item in the Lung Cancer Symptom Scale (LCSS). The participant-reported LCSS was a 9-item questionnaire. Six items were symptom-specific measures for lung cancer (loss of appetite, fatigue, cough, dyspnea, hemoptysis, and pain), and 3 summation items described total symptomatic distress, interference with activity level, and global quality of life. Participant (pt) responses to each item were measured using visual analogue scales (VAS) from 0 (for best outcome) to 100 (for worst outcome). TWS was censored at the date of the last LCSS assessment for pts who were not known to have LCSS worsening. (NCT00102804)
Timeframe: Randomization to worsening of each LCSS item (up to 39 months)

,
Interventionmonths (Median)
Loss of appetiteFatigueCoughDyspneaHemoptysisPainSymptomatic distressInterference with activity levelGlobal quality of life
Pemetrexed and BSC3.783.066.055.36NA6.114.216.515.75
Placebo and BSC4.403.094.674.40NA4.633.783.983.71

[back to top]

Number of Participants With Adverse Events (AEs)

Clinically significant events were defined as serious adverse events (SAEs) and other non-serious AEs regardless of causality. A summary of serious and other non-serious AEs, regardless of causality, is located in the Reported Adverse Events module. (NCT00102804)
Timeframe: Baseline to study completion (up to 41 Months)

,
Interventionparticipants (Number)
SAEsOther Non-Serious AEs
Pemetrexed and BSC83386
Placebo and BSC31178

[back to top]

Maximum Improvement Over Baseline in Individual Symptom Scores and Quality of Life Using the LCSS

The participant-reported LCSS was a 9-item questionnaire. Six items were symptom-specific measures for lung cancer (loss of appetite, fatigue, cough, dyspnea, hemoptysis, and pain), and 3 summation items described total symptomatic distress, interference with activity level, and global quality of life. Participant responses to each item were measured using VAS from 0 (for best outcome) to 100 (for worst outcome). The average symptom burden index (ASBI) was the mean of the 6 symptom-specific items. The LCSS total score was the mean of the 9 items. (NCT00102804)
Timeframe: Baseline through 30 days post discontinuation of study treatment (up to 39 Months)

,
Interventionunits on a scale (Mean)
Loss of appetite (n=403, 197)Fatigue (n=403, 197)Cough (n=402, 197)Dyspnea (n=400, 196)Pain (n=401, 197)Hemoptysis (n=402, 196)Symptom distress (n=401, 196)Interference with activity level (n=400, 197)Global quality of life (n=401, 195)ASBI (n=392, 195)Total LCSS (n=388, 193)
Pemetrexed and BSC7.310.27.67.65.41.56.510.810.73.74.07
Placebo and BSC10.610.46.75.44.32.18.29.310.53.84.04

[back to top]

Time to Objective Progressive Disease (TPD)

TPD was the elapsed time from the date of randomization to the first date of objective PD. TPD was censored at the date of the participant's last tumor assessment for participants who were not known to have PD as of the data-inclusion cut-off date for analysis or who died without objective PD. PD, defined using RECIST v1.0, was at least a 20% increase in the sum of LD of target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of 1 or more new lesions. (NCT00102804)
Timeframe: Randomization to measured PD (up to 41 months)

Interventionmonths (Median)
Pemetrexed and BSC4.27
Placebo and BSC2.60

[back to top]

Progression-Free Survival (PFS) Time

PFS time was the elapsed time from the date of randomization to the first date of objective progression of disease or death from any cause. PFS was censored at the date of the participant's last tumor assessment for participants who were not known to have died or to have PD as of the data-inclusion cut-off date for analysis. PD, defined using Response Evaluation Criteria in Solid Tumors version 1.0 (RECIST v1.0), was at least a 20% increase in the sum of longest diameter (LD) of target lesions taking as references the smallest sum LD recorded since the treatment started or the appearance of 1 or more new lesions. (NCT00102804)
Timeframe: Randomization to measured PD or death from any cause (up to 41 months)

Interventionmonths (Median)
Pemetrexed and BSC4.27
Placebo and BSC2.60

[back to top]

Percentage of Participants With a Complete Response (CR) or Partial Response (PR) (Objective Tumor Response Rate)

Response was defined using RECIST v1.0 criteria. CR was defined as the disappearance of all target lesions. PR was defined either A) at least a 30% decrease in the sum of the LD of target lesions taking as reference the baseline sum LDs or B) complete disappearance of target lesions, with persistence (but not worsening) of 1 or more nontarget lesions. In either case, no new lesions may have appeared. The percentage of participants with CR or PR=(Number of participants with CR or PR)/(Number of participants assessed)*100. (NCT00102804)
Timeframe: Baseline to measured PD (up to 41 months)

Interventionpercentage of participants (Number)
Pemetrexed and BSC6.8
Placebo and BSC1.8

[back to top]

Overall Survival (OS) Time

OS time was the elapsed time from the date of randomization to the date of death from any cause. OS was censored at the last date of contact for participants who were not known to have died as of the data-inclusion cut-off date for analysis. (NCT00102804)
Timeframe: Randomization to date of death from any cause (up to 41 months)

Interventionmonths (Median)
Pemetrexed and BSC13.37
Placebo and BSC10.58

[back to top]

Duration of Tumor Response

Defined as time from first observation of complete response or partial response to the first observation of progressive disease or death due to any cause. (NCT00106002)
Timeframe: every 3 cycles (approximately 6-7 weeks) or until patient has disease progression

Interventionmonths (Median)
Pemetrexed7.3

[back to top]

Overall Survival Time

Defined as the time from date of first dose to time of death due to any cause. (NCT00106002)
Timeframe: every 14 day cycle, during 30-days post-therapy follow-up, and every 6 months during the long-term follow-up

Interventionmonths (Median)
Pemetrexed18.9

[back to top]

Progression-Free Survival Time

Defined as the time from date of first dose to the first observation of disease progression, or death due to any cause. (NCT00106002)
Timeframe: every 3 cycles (approximately 6-7 weeks) or until patient has disease progression

Interventionmonths (Median)
Pemetrexed4.1

[back to top]

Overall Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment." (NCT00106002)
Timeframe: every 3 cycles (approximately 6-7 weeks) or until patient has disease progression

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseBest Response Not Evaluable
Pemetrexed181493

[back to top]

Toxicity Profile: Adverse Events (Common Terminology Criteria for Adverse Events, Grade 3 and 4, Present in >5% of Participants)

Participants rated for toxicity prior to each cycle using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events v3.0 (CTCAE). Grades range from 0 (no AE or within normal limits) to 5 (death related to AE). (NCT00106002)
Timeframe: every 14 day cycle, and during 30-days post-therapy follow-up and long-term follow-up

Interventionparticipants (Number)
Anaemia - Grade 3Anaemia - Grade 4Leukopenia - Grade 3Leukopenia - Grade 4Neutropenia - Grade 3Neutropenia - Grade 4Constipation - Grade 3Constipation - Grade 4Nausea - Grade 3Nausea - Grade 4Asthenia - Grade 3Asthenia - Grade 4Fatigue - Grade 3Fatigue - Grade 4Cellulitis - Grade 3Cellulitis - Grade 4Dehydration - Grade 3Dehydration - Grade 4Hyperglycaemia - Grade 3Hyperglycaemia - Grade 4Dyspnoea - Grade 3Dyspnoea - Grade 4Pleural effusion - Grade 3Pleural effusion - Grade 4
Pemetrexed504285203030502020324020

[back to top]

Safety and Tolerability as Measured by the Number of Participants With Disease Progression

Number of participants with disease progression (protocol-mandated reason for discontinuation). Disease progression was determined by the principle investigator. (NCT00106626)
Timeframe: Any time during 8 cycle treatment period through 30 days after.

InterventionParticipants (Number)
Cohort A - Vorinostat BID + Pemetrexed + Cisplatin4
Cohort B - Vorinostat QD + Pemetrexed + Cisplatin1
Cohort C - Vorinostat BID + Pemetrexed9
Cohort D - Vorinostat QD + Pemetrexed10

[back to top]

Maximum Tolerated Dose (MTD) Status as Determined by Number of Participants With Dose Limiting Toxicity (DLT) at Each Dose Level

MTD was determined by the occurrence of DLTs during the first treatment cycle. DLT describes side effects of a drug or other treatment that are serious enough to prevent an increase in dose or level of that treatment. The dose level is equal to the MTD if < 2 patients experience a DLT and is also the highest tolerated dose level in the cohort. (NCT00106626)
Timeframe: Cycle 1 (21 days)

,,,,,,,,
InterventionParticipants (Number)
Number (#) of DLT# Participants treated at a dose level # Participants treated at a dose level =MTD# Participants treated at a dose level >MTD
Dose Level A.11060
Dose Level A.23007
Dose Level B.10030
Dose Level B.22006
Dose Level C.11700
Dose Level C.20400
Dose Level C.30030
Dose Level D.11090
Dose Level D.22007

[back to top]

Number of Participants With Objective Response Assessed by Independent Review Committee

Objective response was defined as having a complete response (CR) or a partial response (PR). Response assessment was performed using modified World Health Organization (WHO) criteria. Complete response: disappearance of all index and non-index lesions, without appearance of any new lesion. PR: greater than (>) 50 percent (%) decrease from baseline in sum of product of diameters of index lesions, without appearance of any new lesion. (NCT00111839)
Timeframe: Baseline up to PD or death due to any cause (up to approximately 2 years)

Interventionparticipants (Number)
Pemetrexed Alone2
Pemetrexed Plus Matuzumab 800 mg Per Week8
Pemetrexed Plus Matuzumab 1600 mg Every 3 Weeks1

[back to top]

Duration of Objective Response Assessed by Independent Review Committee

Objective response was defined as having a CR or a PR. Response assessment was performed using modified WHO criteria. CR: disappearance of all index and non-index lesions, without appearance of any new lesion. PR: >50% decrease from baseline in sum of product of diameters of index lesions, without appearance of any new lesion. Duration of objective response was defined as time from first appearance of CR or PR to time of PD (>25% increase in one or more lesions, or appearance new lesions) or death. Duration of objective response was to be assessed using Kaplan-Meier analysis. (NCT00111839)
Timeframe: From first documented objective response to PD or death due to any cause (up to approximately 3.5 years)

Interventionmonths (Median)
Pemetrexed AloneNA
Pemetrexed Plus Matuzumab 800 mg Per WeekNA
Pemetrexed Plus Matuzumab 1600 mg Every 3 WeeksNA

[back to top]

Overall Survival (OS)

OS was defined as the duration from randomization to death (due to any cause). OS was estimated using Kaplan-Meier analysis. (NCT00111839)
Timeframe: Baseline up to PD or death due to any cause (up to approximately 3.5 years)

Interventionmonths (Median)
Pemetrexed Alone7.9
Pemetrexed Plus Matuzumab 800 mg Per Week12.4
Pemetrexed Plus Matuzumab 1600 mg Every 3 Weeks5.9

[back to top]

Change From Baseline to Cycle 2 in Global Quality of Life (QoL), as Assessed Using Lung Cancer Symptom Scale (LCSS)

The LCSS consisted of 9 items: 6 items focused on lung cancer symptoms (loss of appetite, fatigue, cough, dyspnea, hemoptysis, and pain] and 3 items were global items (symptom distress, interference with activity level, and global QoL). The global QoL item scores are reported here. The total global QoL item score ranged from 0 (worse QoL) to 100 (best QoL). (NCT00111839)
Timeframe: Baseline, Cycle 2 (Cycle length = 3 weeks)

,,
Interventionunits on a scale (Mean)
BaselineChange at Cycle 2
Pemetrexed Alone35.93.5
Pemetrexed Plus Matuzumab 1600 mg Every 3 Weeks35.815.7
Pemetrexed Plus Matuzumab 800 mg Per Week31.10.8

[back to top]

Progression-Free Survival (PFS)

PFS was defined as the time from randomization to the first documentation of disease progression (PD) or to death due to any cause, whichever occurred first. PD: >25% increase in one or more lesions, or appearance new lesions. PFS was estimated using Kaplan-Meier analysis. (NCT00111839)
Timeframe: Baseline up to PD or death due to any cause (up to approximately 3.5 years)

Interventionmonths (Median)
Pemetrexed Alone2.7
Pemetrexed Plus Matuzumab 800 mg Per Week2.3
Pemetrexed Plus Matuzumab 1600 mg Every 3 Weeks2.5

[back to top]

Overall Survival

Overall survival (OS) is defined as the time from patient randomization (arm assignment) to death from any cause. The median OS with 95% CI was estimated using the Kaplan-Meier method. (NCT00117962)
Timeframe: Time from randomization to death (up to 4 years)

Interventionmonths (Median)
Std Tx + Pemetrexed21.2
Std Tx + Pemetrexed and Cetuximab25.2

[back to top]

Number of Participants With Overall Tumor Response

"Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria:~Complete Response (CR): disappearance of all target lesions;~Partial Response (PR) 30% decrease in sum of longest diameter of target lesions;~Progressive Disease (PD): 20% increase in sum of longest diameter of target lesions;~Stable Disease (SD): small changes that do not meet above criteria.~Overall tumor response is the total number of CR and PRs." (NCT00117962)
Timeframe: Duration of study until progression (up to 4 years)

Interventionparticipants (Number)
Std Tx + Pemetrexed37
Std Tx + Pemetrexed and Cetuximab38

[back to top]

Failure-free Survival

Failure-free survival (FFS) is the time from randomization to a failure event, defined as disease progression or death from any cause (which ever occurred first). The median FFS with 95% CI was estimated using the Kaplan-Meier method, (NCT00117962)
Timeframe: Time from randomization to failure (up to 4 years)

Interventionmonths (Median)
Std Tx + Pemetrexed12.6
Std Tx + Pemetrexed and Cetuximab12.3

[back to top]

18 Month Survival

Percentage of participants who were alive at 18 months. The 18 month survival, with 95% CI, was estimated using the Kaplan-Meier method. (NCT00117962)
Timeframe: 18 months (from randomization)

Interventionpercentage of participants (Number)
Std Tx + Pemetrexed58
Std Tx + Pemetrexed and Cetuximab54

[back to top]

Number of Patients With Histologically Negative Axillary Lymph Node Status at Surgery

Histologically negative is defined as no malignant cells present in the axillary lymph nodes during surgery. (NCT00149214)
Timeframe: surgery after eight 21-day cycles of chemotherapy

Interventionparticipants (Number)
Pemetrexed Plus Doxorubicin, Followed by Docetaxel64
Cyclophosphamide Plus Doxorubicin, Followed by Docetaxel63

[back to top]

Disease-free Survival

Disease-free survival is defined as the time from date of study enrollment (randomization) to first date of progressive disease (PD) or death from any cause. PD per Response Evaluation Criteria In Solid Tumors (RECIST) criteria is at least a 20% increase in the sum of longest diameter (LD) of target lesions taking as references the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. For patients not known to have died as of the data cut-off date and who do not have progressive disease, disease-free survival was censored at the last contact date. (NCT00149214)
Timeframe: baseline through post surgery, follow-up for 3 years post-surgery (up to 5.2 years after randomization)

Interventionmonths (Median)
Pemetrexed Plus Doxorubicin, Followed by DocetaxelNA
Cyclophosphamide Plus Doxorubicin, Followed by DocetaxelNA

[back to top]

Number of Participants With a Pathological Complete Response

pathological assessment of tissue removed during surgery to determine if tumor tissue is still present after chemotherapy (NCT00149214)
Timeframe: surgery after eight 21-day cycles of chemotherapy

,
Interventionparticipants (Number)
Pathological Complete ResponseTumor Cells Still PresentNot evaluable
Cyclophosphamide Plus Doxorubicin, Followed by Docetaxel24896
Pemetrexed Plus Doxorubicin, Followed by Docetaxel21997

[back to top]

Number of Participants With a Clinical Tumor Response After the Second Sequence of Chemotherapy

The number of participants with a clinical tumor response based on measurement of tumor size after the second sequence of chemotherapy, without a second confirmatory tumor measurement required, per protocol. (NCT00149214)
Timeframe: Cycles 5-8 (21-day cycles)

,
Interventionparticipants (Number)
Complete Tumor ResponsePartial Tumor ResponseStable DiseaseProgressive DiseaseUnknownNot Done
Cyclophosphamide Plus Doxorubicin, Followed by Docetaxel2160241103
Pemetrexed Plus Doxorubicin, Followed by Docetaxel195935297

[back to top]

Number of Participants With a Clinical Tumor Response After the First Sequence of Chemotherapy

The number of participants with a clinical tumor response based on measurement of tumor size after the first sequence of chemotherapy, without a second confirmatory tumor measurement, per protocol. (NCT00149214)
Timeframe: Cycles 1-4 (21-day cycles)

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknownNot Done
Cyclophosphamide Plus Doxorubicin, Followed by Docetaxel943442174
Pemetrexed Plus Doxorubicin, Followed by Docetaxel845493179

[back to top]

Pharmacokinetics - Maximum Observed Drug Concentration (Cmax)

(NCT00190671)
Timeframe: cycle 1 (Day 1: <1 min prior to end of pemetrexed infusion; 1/2, 1, 1.5, 2, 3, 4, 6, 8, 24, 48, 72 hours after start of pemetrexed infusion)

Interventionmicrograms per milliliters (Geometric Mean)
Pemetrexed 600mg/m2125
Pemetrexed 1800mg/m2369

[back to top]

Pharmacokinetics - Half-Life (t½)

The half-life associated with the terminal elimination rate constant. (NCT00190671)
Timeframe: cycle 1 (Day 1: <1 min prior to end of pemetrexed infusion; 1/2, 1, 1.5, 2, 3, 4, 6, 8, 24, 48, 72 hours after start of pemetrexed infusion)

Interventionhours (Geometric Mean)
Pemetrexed 600mg/m24.19
Pemetrexed 1800mg/m23.79

[back to top]

Pharmacokinetics - Clearance (CL)

Total body clearance of drug calculated after intravenous administration. (NCT00190671)
Timeframe: cycle 1 (Day 1: <1 min prior to end of pemetrexed infusion; 1/2, 1, 1.5, 2, 3, 4, 6, 8, 24, 48, 72 hours after start of pemetrexed infusion)

Interventionmilliliters per minute (Geometric Mean)
Pemetrexed 600mg/m245.9
Pemetrexed 1800mg/m251.8

[back to top]

Pharmacokinetics - Area Under the Curve (AUC)

Area under the gemcitabine concentration-time curve from zero to last quantifiable concentration [AUC(0-t)] was calculated by combination of linear and logarithmic trapezoidal methods. Linear trapezoidal method was employed up to tmax (time to reach maximal concentration), and then log trapezoidal method was used for those data after tmax. (NCT00190671)
Timeframe: cycle 1 (Day 1: <1 min prior to end of pemetrexed infusion; 1/2, 1, 1.5, 2, 3, 4, 6, 8, 24, 48, 72 hours after start of pemetrexed infusion)

Interventionhour times microgram per milliliter (Geometric Mean)
Pemetrexed 600mg/m2375
Pemetrexed 1800mg/m21050

[back to top]

Time to Progressive Disease

Time to progressive disease was defined as the time from the date of the first treatment dose to the first date of progressive disease or death from study disease. (NCT00190671)
Timeframe: baseline to measured progressive disease

Interventionmonths (Median)
Pemetrexed 1800mg/m26.56

[back to top]

Best Tumor Response

Tumor response was assessed using radiological imaging, which was repeated every 6 weeks prior to every other cycle. Confirmation of response was to occur no less than 4 weeks (28 days) after the first evidence of response. (NCT00190671)
Timeframe: baseline to measured progressive disease

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknownNot Assessed
Pemetrexed 1800mg/m202026852
Pemetrexed 600mg/m208181330

[back to top]

Pharmacokinetics - Volume of Distribution

Central volume (V1) and peripheral volume (V2) of distribution. (NCT00190671)
Timeframe: cycle 1 (Day 1: <1 min prior to end of pemetrexed infusion; 1/2, 1, 1.5, 2, 3, 4, 6, 8, 24, 48, 72 hours after start of pemetrexed infusion)

,
InterventionLiters (Geometric Mean)
Central Volume of Distribution (V1)Peripheral Volume of Distribution (V2)
Pemetrexed 1800mg/m28.143.33
Pemetrexed 600mg/m27.463.58

[back to top]

Progression Free Survival

Defined as date of first treatment dose to first date of progressive disease or death from any cause. For patients not known to have died as of data cutoff date and who did not have progressive disease, the progression free survival date was censored at last contact date. (NCT00190671)
Timeframe: baseline to measured progressive disease

Interventionmonths (Median)
Pemetrexed 1800mg/m26.26

[back to top]

Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment." (NCT00190983)
Timeframe: baseline to measured progressive disease (up to 5 years)

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseDisease Progression
Pemetrexed04167

[back to top]

Progression-Free Survival

The period from study entry until disease progression, death or date of last contact. (NCT00190983)
Timeframe: baseline until documented tumor progression (up to 5 years)

Interventionmonths (Median)
Pemetrexed3.1

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00190983)
Timeframe: baseline until death from any cause (up to 5 years)

Interventionmonths (Median)
Pemetrexed7.4

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. (NCT00190983)
Timeframe: time of initial response until documented tumor progression (up to 5 years)

Interventionmonths (Median)
Pemetrexed4.4

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression. (NCT00191191)
Timeframe: time of response to progressive disease (up to 3.2 years)

Interventionmonths (Median)
Pemetrexed 500 mg/m24.9
Pemetrexed 1000 mg/m23.0

[back to top]

Best Overall Response

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria as determined by the Case Judgment Committee. Best overall response was defined as the most favorable overall response recorded for each patient during the observation period. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. (NCT00191191)
Timeframe: baseline to measured progressive disease (up to 3.2 years)

,
Interventionparticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)
Pemetrexed 1000 mg/m20163458
Pemetrexed 500 mg/m20204048

[back to top]

Change From Baseline to 3 Months in Quality of Life Questionnaire for Cancer Patients Treated With Anticancer Drugs (QOL-ACD)

20-items assessed quality of life in patients undergoing chemotherapy. Scores range from 1 (not at all/very poor) to 5 (very much/very well). Face scale scores (patient circles number of the face that best fits his/her feelings) range from 1 (sad face) to 5 (smiling face). Item scores were grouped according to Functional (daily activity: 5 items), Physical (5 items), Emotional (psychological condition: 4 items), Social Attitude (5 items), and Face Scale (1 item). Score of subscales were converted to scores with range from 0 to 100. Higher scores represent higher QOL. (NCT00191191)
Timeframe: Baseline (pre-dose), 3 Months after first dose of Cycle 1

,
Interventionunits on a scale (Mean)
Functional Baseline (n=107, n=107)Functional Change from Baseline (n=58, n=60)Physical Baseline (n=107, n=107)Physical Change from Baseline (n=58, n=60)Emotional Baseline (n=107, n=107)Emotional Change from Baseline (n=58, n=60)Sociality Baseline (n=107, n=107)Sociality Change from Baseline (n=58, n=60)Face Scale Baseline (n=107, n=107)Face Scale Change from Baseline (n=58, n=60)
Pemetrexed 1000 mg/m275.0-3.773.9-3.562.10.346.11.659.3-2.1
Pemetrexed 500 mg/m277.0-1.578.9-1.667.2-1.745.91.659.3-1.3

[back to top]

Change From Baseline to 3 Months in Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) Lung Cancer Subscale (LCS)

FACT-L LCS measured health-related quality of life (HR-QL) related to additional concerns of lung cancer. Original LCS subscale scores range from 0 to 28, but the scores were converted to scores with a range of 0 to 100 in this study. Higher scores represent better HR-QL. (NCT00191191)
Timeframe: Baseline (pre-dose), 3 Months after first dose of Cycle 1

,
Interventionunits on a scale (Mean)
Baseline (n=107, n=107)Change from Baseline (n=58, n=60)
Pemetrexed 1000 mg/m269.60.4
Pemetrexed 500 mg/m271.53.0

[back to top]

Progression-Free Survival (PFS)

PFS was defined as time from the scheduled date of the first treatment cycle until the date of confirmation of progressive disease on the overall response rating. For patients who died before confirmation of progressive disease, the number of days until the date of death (from any cause) was handled as progression-free survival. (NCT00191191)
Timeframe: baseline to measured progressive disease (up to 3.2 years)

Interventionmonths (Median)
Pemetrexed 500 mg/m23.0
Pemetrexed 1000 mg/m22.5

[back to top]

Overall Survival (OS)

OS was defined as the time from treatment start to death from any cause. For participants who were alive, OS was censored at the last contact date. (NCT00191308)
Timeframe: Treatment start to death from any cause (up to 47.6 months)

Interventionmonths (Median)
Pemetrexed + CisplatinNA

[back to top]

Disease Free Survival (DFS)

DFS was the time from date of first dose to first observation of progressive disease (PD) or death due to any cause. PD=20% increase in sum of longest diameter of target lesions. If a participant was not known to have died or have PD, DFS was censored at the date of the last objective progression-free disease assessment. (NCT00191308)
Timeframe: Treatment start to disease progression or death from any cause (up to 45.5 months)

Interventionmonths (Median)
Pemetrexed + Cisplatin43.7

[back to top]

Duration of Response

The duration of response was defined as the time from complete response (CR) or partial response (PR) to disease progression. Complete Response (CR)=disappearance of all target lesions; Partial Response (PR)=30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD)=20% increase in sum of longest diameter of target lesions. (NCT00191308)
Timeframe: Time of response to disease progression (up to 44.4 months)

Interventionmonths (Median)
Pemetrexed + Cisplatin42.5

[back to top]

Percentage of Participants With Objective Tumor Response (Response Rate)

Tumor response to treatment using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response (CR)=disappearance of all target lesions; Partial Response (PR)=30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD)=20% increase in sum of longest diameter of target lesions; Stable Disease (SD)=small changes that do not meet above criteria. Response rate was estimated as the total number of CR or PR, divided by the total number of participants treated. (NCT00191308)
Timeframe: Treatment start to disease progression or surgery (4-8 weeks after last dose of pemetrexed)

Interventionpercentage of participants (Number)
Pemetrexed + Cisplatin34.5

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00191984)
Timeframe: baseline to date of death from any cause (up to 2 years follow-up)

Interventiondays (Median)
Pemetrexed + Irinotecan422

[back to top]

Best Overall Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment.~Complete response (CR) = disappearance of all target lesions. Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions.~Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions. Stable disease (SD) = small changes that do not meet above criteria." (NCT00191984)
Timeframe: baseline to measured progressive disease (up to 2 years follow-up)

Interventionparticipants (Number)
Partial ResponseStable DiseaseProgressive DiseaseUnknown
Pemetrexed + Irinotecan618155

[back to top]

Time to Treatment Failure

Defined as the time from study enrollment to the first observation of disease progression, death as a result of any cause, or early discontinuation of treatment. Time to treatment failure was censored at the date of the last follow-up visit for patients who did not discontinue early, who were still alive, and who have not progressed. (NCT00191984)
Timeframe: baseline to stopping treatment (up to 2 years follow-up)

Interventiondays (Median)
Pemetrexed + Irinotecan66

[back to top]

Progression-Free Survival (PFS)

Defined as the time from study enrollment to the first date of disease progression or death as a result of any cause. PFS was censored at the date of the last follow-up visit for participants who were still alive and who had not progressed. (NCT00191984)
Timeframe: baseline to measured progressive disease or death (up to 2 years follow-up)

Interventiondays (Median)
Pemetrexed + Irinotecan123

[back to top]

Duration of Response

"The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. Response was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment.~Complete response (CR) = disappearance of all target lesions. Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions." (NCT00191984)
Timeframe: time of response to progressive disease or death (up to 2 years follow-up)

Interventiondays (Median)
Pemetrexed + Irinotecan236

[back to top]

Progression-free Survival (PFS)

PFS was defined as the interval between the start date of treatment and the date of occurrence of progressive disease or death from any cause. (NCT00193414)
Timeframe: 18 months

InterventionMonths (Median)
Intervention6.2

[back to top]

Overall Survival (OS)

OS was measured from the date of study entry until the date of death. (NCT00193414)
Timeframe: 18 months

InterventionMonths (Median)
Intervention8.5

[back to top]

Overall Response Rate

Overall response rate is the percentage of patients with complete response or partial response per RECIST v.1 Criteria. Complete response (CR) = Disappearance of all target lesions, disappearance of all nontarget lesions for at least 4 weeks. Partial Response (PR) = At least a 30% decrease in the sum of the longest diameter of target lesions, taking as reference the baseline sum of longest diameters. (NCT00193414)
Timeframe: 18 months

InterventionPercentage of participants (Number)
Intervention26

[back to top]

Duration of Remission

Will be examined using Kaplan-Meier estimates. Time from earliest confirmed remission criteria until death or progression will be calculated. If a patient continued to be in remission at the end of the study, they will be censored at their last evaluation in the analysis. (NCT00198133)
Timeframe: Time from the date of remission until progression or death, assessed up to 3 years

Interventionmonths (Median)
Thymoma4.0
Thymic Carcinoma3.8

[back to top] [back to top]

Objective Response Rate (Complete and Partial Response)

The percent of patients having an objective response (complete or partial response) will be estimated with a 95% exact binomial confidence interval for the percent of patients receiving drug. RECIST v1.0 will be used. At least a 30% decrease in the sum of the longest diameter of target lesions in reference to the baseline longest diameter will need to take place to be considered an objective response. (NCT00198133)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Thymoma26.7
Thymic Carcinoma9.1

[back to top]

Progression-free Survival Based on Serum Biomarker Status

Progression-free survival in this analysis looking at the association between a serum proteomic biomarker and progression-free survival will be defined as the time from the start of treatment until progression (documented according to Response Evaluation Criteria in Solid Tumors [RECIST] criteria and defined as at least a 20% increase in the sum of the longest diameter of target lesions) or death from any cause, whichever comes first. (NCT00203931)
Timeframe: up to 5 years

Interventionmonths (Median)
Poor Prognosis1.57
Good Prognosis1.64

[back to top]

Progression-free Survival Based on Rash Development

Progression-free survival in this landmark analysis looking at the utility of early rash in predicting progression-free survival will be defined as the time from day 22 of study therapy until progression (documented according to Response Evaluation Criteria in Solid Tumors [RECIST] criteria and defined as at least a 20% increase in the sum of the longest diameter of target lesions) or death from any cause, whichever comes first. Patients last known to be alive and progression-free were censored at the date of the last scan without evidence of progression. (NCT00203931)
Timeframe: up to 5 years

Interventionmonths (Median)
Early Rash0.9
No Early Rash0.9

[back to top]

Progression-free Survival

Progression-free survival will be defined as the time from the start of treatment until progression (documented according to Response Evaluation Criteria in Solid Tumors [RECIST] criteria and defined as at least a 20% increase in the sum of the longest diameter of target lesions) or death from any cause, whichever comes first. (NCT00203931)
Timeframe: Up to 5 years

Interventionmonths (Median)
Cetuximab1.6
Cetuximab and Pemetrexed2.3

[back to top]

Overall Survival

Overall survival will be defined as the time from the start of treatment until death from any cause. (NCT00203931)
Timeframe: Up to 5 years

Interventionmonths (Median)
Cetuximab3.5
Cetuximab and Pemetrexed10.3

[back to top]

Objective Response Rate

Objective response (complete response [CR] + partial response [PR]) will be evaluated using RECIST criteria. CR is the disappearance of all target lesions. PR requires at least a 30% decrease in the sum of the longest diameter of target lesions. (NCT00203931)
Timeframe: up to 2 years

Interventionpercentage of participants (Number)
Cetuximab0
Cetuximab and Pemetrexed13.0

[back to top]

Overall Survival

(NCT00216099)
Timeframe: From study enrollment until death (for life)

Interventionmonths (Median)
Pemetrexed 500mg/m^214

[back to top]

Rate of Clinical Benefit

"A clinical benefit is defined as an improvement for at least 3 consecutive weeks in at least one of the following parameters without any sustained worsening in any other:~> 50% reduction in analgesic consumption or > 50% reduction in pain intensity or > 20 point gain in performance status." (NCT00216099)
Timeframe: Any time among evaluable subjects (for life)

Interventionpercentage of participants (Number)
Pemetrexed 500mg/m^233

[back to top]

Time to Progression

Progression per Response Evaluation Criteria in Solid Tumors (RECIST) or Prostate-Specific Antigen (PSA) Progression RECIST PD=at least a 20% increase in the sum of the longest diameter (LD) of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions or Appearance of one or more new lesions and/or unequivocal progression of existing non-target lesions PSA progression=increase in PSA to >50% above lowest level recorded on study. Two consecutive increases required at least 4 weeks apart, but time to progression will be determined at time of first PSA showing increase > 50% above baseline *Note, upper confidence interval was not reached* (NCT00216099)
Timeframe: Study enrollment until progression per RECIST or PSA (for life)

Interventionmonths (Median)
Pemetrexed 500mg/m^25

[back to top]

Safety and Tolerability

Safety and Tolerability was evaluated by reporting the percentage of patient who experienced grade 3 or 4 toxicities using Common Terminology Criteria for Adverse Events CTCAE v3.0 criteria. CTCAE grades the severity of an adverse event from 1-5 where 1=least severe and 5=death. (NCT00216099)
Timeframe: 18 months

Interventionpercentage of participants (Number)
Grade 3Grade 4
Pemetrexed 500mg/m^242.98.2

[back to top]

Time to Prostate-Specific Antigen (PSA)/Serological Progression

Serological Progression (sPD) - increase in PSA to >50% above lowest level recorded on study. Two consecutive increases required at least 4 weeks apart, but time to progression will be determined at time of first PSA showing increase > 50% above baseline (NCT00216099)
Timeframe: From study enrollment to progression per PSA criteria (for life)

Interventionmonths (Median)
Pemetrexed 500mg/m^22

[back to top]

Best Overall PSA Response

"Best overall Prostate-Specific Antigen (PSA) response~PSA response is defined by a greater than or equal to 50% decline in PSA confirmed by a second PSA value at least 4 weeks after the first PSA response timepoint PSA Stable Disease is defined as less than a 50% decline in PSA and less than a 50% increase in PSA from baseline PSA progression is defined as greater than or equal to a 50% increase in PSA compared to baseline" (NCT00216099)
Timeframe: Start of treatment until disease progression/recurrence (for life)

Interventionpercentage of participatns (Number)
>50% decline in PSAStable PSAPSA progression
Pemetrexed 500mg/m^282065

[back to top]

OBJECTIVE Overall Response Rate

"Response Evaluation Criteria in Solid Tumors (RECIST). Objective overall response rate is defined as Complete Response (CR) + Partial Response (PR)~Per RECIST:~CR= Disappearance of all target and non-target lesions and normalization of tumor marker level PR= Disappearance of all target lesions and persistence of non-target lesion(s) or maintenance of tumor marker level above normal limits OR at least a 30% decrease in the sum of the longest diameter, taking as reference the baseline sum longest diameter and disappearance of all non-target lesions or persistence of non-target lesion(s) or maintenance of tumor marker level above normal limits" (NCT00216099)
Timeframe: Start of treatment until disease progression/recurrence (for life)

Interventionpercentage of participants (Number)
Participants with PR with meas. dis. per RECISTparticipants with SD maintained at 12 weeks
Pemetrexed 500mg/m^2839

[back to top]

RFC1 G80A Genotype

Samples for RFC1 G80A pharmacogenetic analysis were collected at screening (NCT00216099)
Timeframe: Screening

Interventionparticipants (Number)
A/A GenotypeA/G GenotypeG/G Genotype
Pemetrexed 500mg/m^262218

[back to top]

Toxicity and Safety Profile

(NCT00216203)
Timeframe: 12 months

Interventionpercentage of particpants (Number)
Acne-like rash: Phase I - Any GradeAcne-like rash: Phase I - Grade 3/4Acne-like rash: Phase II - Any GradeAcne-like rash: Phase II - Grade 3/4ALT elevation: Phase I - Any GradeALT elevation: Phase I - Grade 3/4ALT elevation: Phase II - Any GradeALT elevation: Phase II - Grade 3/4Anemia: Phase I - Any GradeAnemia: Phase I - Grade 3/4Anemia: Phase II - Any GradeAnemia: Phase II - Grade 3/4Thrombocytopenia: Phase I - Any GradeThrombocytopenia: Phase I - Grade 3/4Thrombocytopenia: Phase II - Any GradeThrombocytopenia: Phase II - Grade 3/4Neutropenia: Phase I - Any GradeNeutropenia: Phase I - Grade 3/4Neutropenia: Phase II - Any GradeNeutropenia: Phase II - Grade 3/4Febrile Neutropenia: Phase I - Any GradeFebrile Neutropenia: Phase I - Grade 3/4Febrile Neutropenia: Phase II - Any GradeFebrile Neutropenia: Phase II - Grade 3/4Dizziness: Phase I - Any GradeDizziness: Phase I - Grade 3/4Dizziness: Phase II - Any GradeDizziness: Phase II - Grade 3/4Dyspnea: Phase I - Any GradeDyspnea: Phase I - Grade 3/4Dyspnea: Phase II - Any GradeDyspnea: Phase II - Grade 3/4Cough: Phase I - Any GradeCough: Phase I - Grade 3/4Cough: Phase II - Any GradeCough: Phase II - Grade 3/4Fatigue: Phase I - Any GradeFatigue: Phase I - Grade 3/4Fatigue: Phase II - Any GradeFatigue: Phase II - Grade 3/4Anorexia: Phase I - Any GradeAnorexia: Phase I - Grade 3/4Anorexia: Phase II - Any GradeAnorexia: Phase II - Grade 3/4Mucositis: Phase I - Any GradeMucositis: Phase I - Grade 3/4Mucositis: Phase II - Any GradeMucositis: Phase II - Grade 3/4Nausea: Phase I - Any GradeNausea: Phase I - Grade 3/4Nausea: Phase II - Any GradeNausea: Phase II - Grade 3/4Diarrhea: Phase I - Any GradeDiarrhea: Phase I - Grade 3/4Diarrhea: Phase II - Any GradeDiarrhea: Phase II - Grade 3/4Constipation: Phase I - Any GradeConstipation: Phase I - Grade 3/4Constipation: Phase II - Any GradeConstipation: Phase II - Grade 3/4Fever(no neutropenia): Phase I - Any GradeFever(no neutropenia): Phase I - Grade 3/4Fever(no neutropenia): Phase II - Any GradeFever(no neutropenia): Phase II - Grade 3/4Headache: Phase I - Any GradeHeadache: Phase I - Grade 3/4Headache: Phase II - Any GradeHeadache: Phase II - Grade 3/4Hypomagnesmia: Phase I - Any GradeHypomagnesmia: Phase I - Grade 3/4Hypomagnesmia: Phase II - Any GradeHypomagnesmia: Phase II - Grade 3/4Vomiting: Phase I - Any GradeVomiting: Phase I - Grade 3/4Vomiting: Phase II - Any GradeVomiting: Phase II - Grade 3/4
Pemetrexed + Cetuximab58.316.75723.833.316.7000014.308.3014.308.304.84.88.38.3008.38.3194.8258.333.3025028.6033.30764.833.30194.841.70384.833.3028.6033.3033.39.516.7023.84.88.30194.850023.8000194.816.7023.84.8

[back to top]

Median Survival Time

(NCT00216203)
Timeframe: 24 Months

Interventionweeks (Median)
Pemetrexed + Cetuximab42.0

[back to top]

Time To Progression (TTP)

The primary objective of the phase II portion is to estimate the time to progression of this combination, evaluated per RECIST criteria where 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 (NCT00216203)
Timeframe: 24 Months

InterventionWeeks (Median)
Pemetrexed + Cetuximab14.6

[back to top]

Maximum Tolerated Dose (MTD) of Pemetrexed in Combination With Cetuximab

The primary objective of the phase I portion of this study is to define the maximum tolerated dose (MTD) of the combination of pemetrexed and cetuximab (NCT00216203)
Timeframe: 12 months

Interventionmg/m^2 every 21 days (Number)
Investigational Treatment750

[back to top]

Disease Control Rate (DCR)

(NCT00222729)
Timeframe: Up to 36 months

Interventionpercentage (Median)
Pemetrexed + Bevacizumab86

[back to top]

Time-to-progression (TTP)

TTP was calculated from treatment initiation to disease progression or last follow-up. (NCT00222729)
Timeframe: Up to 36 months

Interventionmonths (Median)
Pemetrexed + Bevacizumab5

[back to top]

Objective Response Rate (ORR)

(NCT00222729)
Timeframe: Up to 36 months

Interventionpercentage (Median)
Pemetrexed + Bevacizumab30

[back to top]

Overall Survival (OS)

(NCT00222729)
Timeframe: Up to 36 months

Interventionmonths (Median)
Pemetrexed + Bevacizumab11.3

[back to top]

Toxicity

Number of participants with toxicity ≥ Grade 3 after gemcitabine plus pemetrexed induction chemotherapy. (NCT00226577)
Timeframe: 06/20/2008 Index date for patients enrolled between 04/2004 and 04/2006

Interventionparticipants (Number)
Pre-Surgery Chemotherapy18

[back to top]

Survival - Overall

Median range of number of participants with Overall Survival. Overall survival (OS) will be defined as the period of time from the first day of drug treatment to the date of death of the patient. Patients taken off study will be followed quarterly until death for survival data. (NCT00226577)
Timeframe: 06/20/2008 Index date for patients enrolled between 04/2004 and 04/2006

Interventionmonths (Median)
Pre-Surgery Chemotherapy27.8

[back to top]

Disease Response - Radiographic

"Number of participants with partial or Complete Response. Complete response (CR) is defined as the total disappearance of all malignant and evaluable clinical evidence of cancer without the development of any new malignant lesions documented on the post chemotherapy chest CT and PET scan.~Partial response (PR) (measurable disease only): When compared with pre-treatment measurements, a reduction of >30% in the sum of the largest diameters of all measurable lesions and absence of new lesions." (NCT00226577)
Timeframe: 06/20/2008 Index date for patients enrolled between 04/2004 and 04/2006

Interventionparticipants (Number)
Complete RemissionPartial Remission
Pre-Surgery Chemotherapy116

[back to top]

Disease Response - Pathologic

Number of participants with Pathologic Complete Response. Pathologic complete response (pCR) is defined by a surgical pathology specimen, which consists of equal to or more than 95% fibrosis and necrosis. (NCT00226577)
Timeframe: 06/20/2008 Index date for patients enrolled between 04/2004 and 04/2006

Interventionparticipants (Number)
Pre-Surgery Chemotherapy13

[back to top]

Survival - Disease Free

Disease-free survival (DFS) is defined as the period of time from surgery to the time when disease recurrence is clearly documented. A histologic confirmation is required in equivalent cases. (NCT00226577)
Timeframe: 06/20/2008 Index date for patients enrolled between 04/2004 and 04/2006

Interventionmonths (Median)
Pre-Surgery Chemotherapy33.7

[back to top]

Incidence of Central Nervous System (CNS) Hemorrhagic Events

Number of events of brain or central nervous system (CNS) bleeding (NCT00227019)
Timeframe: 18 months

InterventionCNS hemorrhagic events (Number)
Bevacizumab + Pemetrexed0

[back to top]

Overall Survival (OS)

"Overall Survival (OS) is defined as the duration of time from start of treatment to deat.~Kaplan-Meier survival curves for OS were generated with IBM SPSS Statistics version 19.0 (SPSS, Inc, Chicago, IL)." (NCT00227019)
Timeframe: 18 months

Interventionmonths (Median)
Bevacizumab + Pemetrexed14.8

[back to top]

Progression-free Survival (PFS)

"Progression-free survival (PFS) is defined as the duration of time from start of treatment to time of documented disease progression or death.~Kaplan-Meier survival curves for PFS were generated with IBM SPSS Statistics version 19.0 (SPSS, Inc, Chicago, IL)." (NCT00227019)
Timeframe: 18 months

Interventionmonths (Median)
Bevacizumab + Pemetrexed7.2

[back to top]

Safety of Neoadjuvant Chemotherapy

The number of patients that experienced a grade 3 or higher adverse event. (NCT00227539)
Timeframe: Up to 4 weeks after last dose of chemotherapy

InterventionParticipants (Count of Participants)
Neoadjuvant Therapy, PET Scan and Surgery10

[back to top]

Positron Emission Tomography as a Predictor of Response Measured by the Decrease in Standard Uptake Variable (SUV) After 1 Course of Therapy

Number of Participants with Decrease in Standard Uptake Variable (SUV) After 1 Course of Therapy (NCT00227539)
Timeframe: Between days 18 and 22 prior to second chemotherapy infusion

InterventionParticipants (Count of Participants)
Neoadjuvant Therapy, PET Scan and Surgery11

[back to top]

Efficacy of Neoadjuvant Chemotherapy as Measured by Radiologic Response Rate

The number of patients that had either a CR, PR or SD after the completion of 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; Progression, as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions; Stable Disease (SD), neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for disease progression. (NCT00227539)
Timeframe: Up to 4 weeks after last dose of chemotherapy

InterventionParticipants (Count of Participants)
Neoadjuvant Therapy, PET Scan and Surgery20

[back to top]

Overall Survival Rate at 6, 12, 18, and 24 Months

"Overall Survival (OS) Rate of carboplatin, pemetrexed and bevacizumab combination treatment will be defined as the percentage of patients with documentation of status of alive at the following timepoints from registration to the study:~6 months 12 months 18 months 24 months" (NCT00234052)
Timeframe: 6, 12,18, and 24 months from treatment initiation

Interventionpercentage of patients alive (Number)
6 Months12 Months18 Months24 Months
Treatment With Carboplatin + Pemetrexed + Bevacizumab86613826

[back to top]

Overall Survival Rate

Overall Survival (OS) Rate of carboplatin, pemetrexed and bevacizumab combination treatment is defined from the time of registration to the study until death from any cause. Patients that are lost to follow up will be censored from last documentation of survival status. (NCT00234052)
Timeframe: During treatment and then every 3 months x 2 years, then every 6 months x 3 years or until death.

InterventionMonths (Median)
Treatment With Carboplatin + Pemetrexed + Bevacizumab14.1

[back to top]

Overall Response Rate

"Overall Response Rate (ORR) of patients treated with carboplatin, pemetrexed, and bevacizumab combination is defined as the number of patients who's best response is a Complete Response (CR) plus Partial Response (PR)as recorded from the start of treatment until disease progression as assessed by RECIST 1.0.~CR=Disappearance of all target lesions for a minimum of 4 weeks. PR=At least a 30% decrease in the sum of the longest diameter (LD) of target lesions for a minimum of 4 weeks, taking as reference the baseline sum LD. No simultaneous increase in the size of any lesion or the appearance of a new lesion may occur." (NCT00234052)
Timeframe: Every two cycles until disease progression. Median follow up of 13 months (range 0.8 to 34.4 months)

InterventionParticipants (Count of Participants)
Treatment With Carboplatin + Pemetrexed + Bevacizumab27

[back to top]

Duration of Response

Duration of Response for patients treated with the combination of carboplatin, pemetrexed and bevacizumab is measured from the time measurement criteria are met for Complete Response or Partial Response (whichever is first recorded) until the first date of documented progressive disease. (NCT00234052)
Timeframe: From documentation of response, every two cycles (1 cycle = 21 days) until progressive disease with range of cycles completed 1-51.

InterventionMonths (Median)
Treatment With Carboplatin + Pemetrexed + Bevacizumab7.7

[back to top]

Median Progression Free Survival

Progression Free Survival (PFS) in patients treated with the combination of carboplatin, pemetrexed and bevacizumab is defined as the time from registration to the time of documented disease progression or death from any cause. Patients that were lost to follow up or withdrew consent were censored at that point. (NCT00234052)
Timeframe: Approximately every 3 weeks until disease progression or death. Median follow up of 13 months (range 0.8 to 34.4 months)

InterventionMonths (Median)
Treatment With Carboplatin + Pemetrexed + Bevacizumab7.8

[back to top]

Toxicity of Carboplatin, Pemetrexed and Bevacizumab Combination Treatment

"To characterize the toxicity profile of carboplatin, pemetrexed and bevacizumab combination treatment.~Toxicity data will be collected from initiation of treatment, every cycle, until 30 days post last treatment. Adverse events will be graded according to the National Cancer Institute's Common Toxicity Criteria for adverse events version 3.0 (CTCAE v3.0). Only toxicity determined to be a least possibility related to at least one study drug and grade 3 or 4 was collected for this outcome measure.~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" (NCT00234052)
Timeframe: From treatment initiation, at the beginning of each cycle where one cycle equals 21 days until 30 days post treatment (range of cycles 1-51)

InterventionParticipants (Count of Participants)
AnemiaThrombocytopeniaNeutropeniaVenous ThrombosisFatigueDiverticulitisInfectionProteinuriaArterial ThrombosisIncreased Creatinine Levels
Treatment With Carboplatin + Pemetrexed + Bevacizumab3423445111

[back to top]

Progression Free Survival at 6, 12, 18, 24 Months

"Progression Free Survival (PFS) Rate of carboplatin, pemetrexed and bevacizumab combination treatment is defined as the percentage of patients without progression at the following time points from registration to the study:~6 months 12 months 18 months 24 months." (NCT00234052)
Timeframe: 6, 12,18, and 24 months from treatment initiation

Interventionpercentage of patients with PFS (Number)
6 Months12 Months18 Months24 Months
Treatment With Carboplatin + Pemetrexed + Bevacizumab59342719

[back to top]

Percent Change in SUV Level Between Pre and Post Chemotherapy

Percent change of PET/SUV levels between baseline and post-chemotherapy. (NCT00248495)
Timeframe: Baseline and post-chemotherapy

InterventionPercentage change in SUV level (Mean)
Neoadjuvant Chemotherapy-36.1

[back to top]

Pathologically Complete Response

Pathologic Complete Response is defined by a surgical pathology specimen, which is free of all gross and microscopic evidence of viable tumor. (NCT00248495)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Neoadjuvant Chemotherapy0

[back to top]

Overall Survival

Overall survival was defined as time from date of treatment initiation until date of death due to any cause. (NCT00248495)
Timeframe: Every 6 months until the time of death up to 126 months

Interventionmonths (Median)
Neoadjuvant Chemotherapy100.7

[back to top]

Disease Free Survival

Progressive disease is defined as at least a 20% increase in the sum of the longest diameter of target lesions or the appearance of new lesions. Disease Free Survival was defined as time from date of treatment initiation until date of first documented progression or date of death from any cause, whichever came first. (NCT00248495)
Timeframe: At least every 3 months after the completion of adjuvant therapy for two years and thereafter every 6 months for 3 years and then yearly up to 126 months

Interventionmonths (Median)
Neoadjuvant Chemotherapy34.5

[back to top]

Treatment Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment." (NCT00259285)
Timeframe: every 21 day cycle (3 cycles) and 3-4 weeks after last cycle

Interventionparticipants (Number)
Complete ResponsePartial ResponseIncomplete Response/Stable DiseaseProgressive DiseaseNot Evaluable
Pemetrexed + Cisplatin05410

[back to top]

Pathologic Remissions After Surgery

The status of the pathological response was evaluated on the basis of the original results of the histopathological examination of the tumour samples resected. A complete pathological response was defined as the absence of any viable tumour cell in the tumour samples obtained for histological examination. (NCT00259285)
Timeframe: surgical tumor resection (3-4 weeks after completing three 21-day cycles of therapy)

Interventionparticipants (Number)
Complete Remission - YesComplete Remission - No
Pemetrexed + Cisplatin52

[back to top] [back to top]

Response (Confirmed and Unconfirmed, Complete and Partial)

Complete response (CR) is complete disappearance of all measurable and non-measurable disease. No new lesions. No disease related symptoms. Normalization of markers and other abnormal lab values. Partial Response (PR) is greater than or equal to 30% decrease under baseline of the sum of longest diameters of all target measurable lesions. No unequivocal progression of non-measurable disease. No new lesions. Confirmation of CR or PR means a repeat scan at least 4 weeks apart documented before progression or symptomatic deterioration. (NCT00265785)
Timeframe: Assessed at weeks 7 and 13 while on treatment. After off treatment prior to disease progression, disease assessment takes place every 3 months for a maximum of 3 years.

Interventionpercentage of participants (Number)
Premetrexed29

[back to top]

Progression-free Survival

Progression is defined as any one or more of the following: 20% increase in the sum of longest diameters of target measurable lesions over smallest sum observed (over baseline if no decrease during therapy) using the same techniques as baseline; Unequivocal progression of non-measurable disease in the opinion of the treating physician (an explanation must be provided); Appearance of any new lesion/site; Death due to disease without prior documentation of progression and without symptomatic deterioration. Symptomatic deterioration is defined as globabl deterioration of health status requiring discontinuation of treatment without objective evidence of progression. Progression-free survival is measured from date of registration to date of first documentation of progression or symptomatic deterioration, or death due to any cause. Patients last known to be alive and progression-free are censored at date of last contact. (NCT00265785)
Timeframe: 0 - 3 years

Interventionmonths (Median)
Premetrexed6

[back to top]

Overall Survival

Measured from date of registration to date of death due to any cause. Patients last known to be alive are censored at date of last contact. (NCT00265785)
Timeframe: 0 - 3 years

Interventionmonths (Median)
Premetrexed25

[back to top]

Pathologic Complete Response Rate

The proportion of pathologic complete responses will be estimated by the number of pathologic complete responses divided by the total number of evaluable patients. Ninety-five percent confidence intervals for the true pathologic complete response rate will be calculated. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for Measurable disease is defined as at least one lesion whose longest diameter can be accurately measured as ≥2.0 cm with conventional techniques or as ≥1.0 cm with spiral CT. Lesions on chest x-ray are acceptable as measurable lesions when they are clearly defined and surrounded by aerated lung. However, CT is preferable. (NCT00268437)
Timeframe: Baseline to time of surgery (around 10 - 18 weeks post-baseline)

Interventionpercentage of participants (Number)
Pemetrexed/Carboplatin23

[back to top]

Overall Survival

Time from registration to death due to any cause. (NCT00268437)
Timeframe: From baseline to 4 years

Interventionmonths (Median)
Pemetrexed/Carboplatin17.8

[back to top]

The Feasibility of Post-Surgery Chemotherapy

Feasibility was measured by completion of 4 treatment cycles without remaining toxicities >=Grade 3 at 30 days after last infusion. (NCT00269152)
Timeframe: every 21-day cycle for 4 cycles up to 30 days after last infusion

,
Interventionparticipants (Number)
"Participants feasible"Non-feasible = Early DiscontinuationNon-feasible = Lost to Follow-upNon-feasible = Remaining Grade 3/4 ToxicityNon-feasible = Underdosage (<95% intended dose)
Pemetrexed + Carboplatin2762319
Pemetrexed + Cisplatin3818145

[back to top]

Overall Survival at 6 Years

For each treatment arm, the Kaplan-Meier technique was used to estimate the 6 year survival rate. Results are presented as probability (%) of survival at 6 years. Overall survival is the duration from enrollment to death. For participants not known to have died, overall survival was censored at the last known alive date. (NCT00269152)
Timeframe: Baseline to date of death from any cause assessed at 6 years

Interventionpercent probability of survival (%) (Number)
Pemetrexed + Cisplatin72.6
Pemetrexed + Carboplatin83.2

[back to top]

Grade III/IV Adverse Events

Number of participants experiencing Grade III/IV hematologic and non-hematologic adverse events possibly related to study drug or protocol procedures in this study. (NCT00269152)
Timeframe: every 21-day cycle for 4 cycles

,
Interventionparticipants (Number)
NeutropeniaAnaemiaThrombocytopeniaFebrile neutropeniaLeukopeniaLymphopeniaNeutrophil count decreasedHaemoglobin count decreasedPlatelet count decreasedWhite blood cell count decreasedAstheniaNauseaVomitingFatigueCatheter related infectionGamma-glutamyltransferase increasedAnorexiaHyperglycaemiaHyperkalaemiaPsychotic disorder
Pemetrexed + Carboplatin63321062122002000000
Pemetrexed + Cisplatin90000110104330111111

[back to top]

Overall Survival at 3 Years

For each treatment arm, the Kaplan-Meier technique was used to estimate the 3 year survival rate. Results are presented as probability (%) of survival at 3 years. Overall survival is the duration from enrollment to death. For participants not known to have died, overall survival was censored at the last known alive date. (NCT00269152)
Timeframe: baseline to date of death from any cause, assessed at 3 years

Interventionpercent probability of survival (%) (Number)
Pemetrexed + Cisplatin82.0
Pemetrexed + Carboplatin83.2

[back to top]

3 Year Disease-Free Survival: Probability of Disease-Free Survival at 3 Years

For each treatment arm, the Kaplan-Meier technique was used to estimate the 3 year disease-free rate. Disease-free survival is defined as the time from enrollment to the first observation of disease progression, or death due to any cause. For participants not known to have died and to have had recurrent disease, disease-free survival was censored at the date of the last participant contact with No Recurrence status. Results are presented as probability (%) of disease-free survival at 3 years. (NCT00269152)
Timeframe: length of time disease free, assessed at 3 years

Interventionprobability of disease-free survival (%) (Number)
Pemetrexed + Cisplatin61.2
Pemetrexed + Carboplatin67.3

[back to top]

Neurological Function by Radiation Oncology Group (RTOG) Neurological Function Classification

"A classification score defined as follows:~Able to work or to perform normal activities: neurological findings minor or absent~Able to carry out normal activities with minimal difficulties. Neurological impairment does not require nursing care or hospitalization~Seriously limited in performing normal activities. Requiring nursing care or hospitalization. Patients confined to bed or wheelchair or have significant intellectual impairment~Unable to perform even minimal normal activities. Requiring hospitalization and constant nursing care and feeding. Patients unable to communicate or in coma.A higher score indicates worse function." (NCT00280748)
Timeframe: At Baseline, 30 days, and at end of treatment (maximum 5 months).

Interventionscores on a scale (Mean)
Pre-TreatmentCycle 2End of Treatment
Single Arm Study1.61.92.2

[back to top]

Neurological Function by Mini Mental State Examination

The Mini Mental State Examination is a 30-point questionnaire that is used to measure cognitive impairment. Score totals range from normal cognition (24-30 points), mild impairment (19-23 points), moderate impairment (10-18 points), to severe impairment (≤9 points). (NCT00280748)
Timeframe: Baseline (pre-treatment), 30 days (Cycle 2 Day 1), and maximum 5 months (end of treatment).

Interventionscore on the scale (Mean)
Pre-TreatmentCycle 2 Day 1End of treatment
Single Arm Study2828.227.7

[back to top]

Estimate the Overall Survival of Patients Treated With This Regimen.

Patients were followed for survival from start of treatment until death from any cause (up to 4 years) (NCT00280748)
Timeframe: 4 years

Interventionmonths (Median)
Single Arm Study6.0

[back to top]

Number of Subjects Experiencing Adverse Events

"Toxicities was assessed using Common Terminology Criteria for Adverse Events (CTCAE) grading scale. Only toxicities with attribution to chemotherapy of definite or probable are considered, as determined by treating physician." (NCT00280748)
Timeframe: maximum 5 months

InterventionParticipants (Count of Participants)
AnemiaAlpeciaNauseaAnorexiaPruritic rash on headCrampsAcute bronchitisBlurred visionDry skinFatigue
Single Arm Study2261111112

[back to top]

Overall Response Rate

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT00293579)
Timeframe: Up to 3 years

Interventionpatients (Number)
Pemetrexed0

[back to top]

Impact of Pemetrexed Chemotherapy on Quality of Life

Quality of life assessements conducted at BL (baseline assessment) and EoT (End of treatment). University of Washington QOL (UW-QOL) questionnaire tests 9 specific areas relating to head and neck cancer. A composite score is calculated by adding together the 9 domain scores to give a scale from 0 (for poor health) to 900 (good health). (NCT00293579)
Timeframe: Baseline, End of Treatment [up to 3 years]

InterventionUnits on a scale (Mean)
Baseline scoreEnd of Treatment score
Pemetrexed524335.5

[back to top]

Toxicities of Pemetrexed,in Poor Risk Cases With Poor Performance Status and Advanced, Metastatic, or Recurrent Head and Neck Cancer

Drug induced toxicities were assessed and graded according to the revised NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00293579)
Timeframe: Up to 3 years

Interventionpercent of patients (Number)
fatigueHypercalcemiaAnemia
Pemetrexed802020

[back to top]

Overall Survival

Overall survival was measured from the time of initial study entry to death due to any cause. (NCT00293579)
Timeframe: Up to 2 years

Interventionmonths (Median)
Pemetrexed4.4

[back to top]

Progression Free Survival Rate at 6 Months

This is the percentage of patients alive and progression-free at 6 months from initiation of treatment. (NCT00295503)
Timeframe: patients progression free at 6 months

Interventionpercentage of participants (Number)
Cisplatin, Pemetrexed, Bevacizumab56

[back to top]

Overall Survival

overall survival was measured from time of initiation of treatment to death from any cause (NCT00295503)
Timeframe: From date of randomization until the date of first documented progression or date of death from any cause, whichever came first, assessed up 100 months

Interventionmonths (Median)
Cisplatin, Pemetrexed and Bevacizumab14.8

[back to top]

Response Rate

response was assessed by the RECIST criteria (version 1.0). Per those criteria, progression is defined 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. (NCT00295503)
Timeframe: from time of enrollment to time of best response or death from any cause, whichever came first up to 100 months

Interventionpercentage of participants (Number)
Cisplatin, Pemetrexed, Bevacizumab40

[back to top]

Safety Outcomes

Toxicity: total number of SAEs and other AEs (NCT00301808)
Timeframe: 72 hours after 2nd and 3rd cycles: 30 days after completion of study treatment; Every 2 months thereafter; then once a year

InterventionAdverse event (Number)
Serious (grade 3 or 4)other (grade 0, 1, or 2)
Cisplatin, Docetaxel & Radiation Therapy20245

[back to top]

Overall Survival

Overall survival using Kaplan-Meier estimates (NCT00301808)
Timeframe: Date of registration to the date of death

Interventionmonths (Median)
Cisplatin, Docetaxel & Radiation Therapy33.7

[back to top]

Probability of Overall Survival at One Year

Overall Survival at one year using Kaplan-Meier product-limit analysis (NCT00301808)
Timeframe: at 1 year

Interventionprobability of overall survival at 1 yr. (Number)
Cisplatin, Docetaxel & Radiation Therapy0.66

[back to top]

Progression-free Survival

Progression-free survival using Kaplan-Meier estimates (NCT00301808)
Timeframe: Approximately 3 weeks after the last cycle of cisplatin/pemetrexed or completion of radiation whichever is the later.

Interventionmonths (Median)
Cisplatin, Docetaxel & Radiation Therapy16.9

[back to top]

Duration of CR or PR (Duration of Response)

The duration of a CR or PR was defined as the time from first objective status assessment of CR or PR to the first time of progression or death due to any cause. Response was defined using RECIST, version 1.0 criteria. CR was defined as the disappearance of all target lesions. PR was defined as having at least a 30% decrease in sum of longest diameter of target lesions. (NCT00308750)
Timeframe: Date of first response to the date of progression or death due to any cause up to 22.3 months

Interventionmonths (Median)
Enzastaurin/Pemetrexed/Carboplatin7.4
Pemetrexed/Carboplatin9.3
Docetaxel/Carboplatin5.8

[back to top]

Change From Baseline in Total Functional Assessment of Cancer Therapy -Taxane (FACT-Taxane) Scale

"The FACT-Taxane version 4 scale is used to assess HRQoL in participants receiving taxane chemotherapy. The FACT-taxane has 5 subscales: PWB, SFWB, and FWB subscales which include 7 items each, EWB subscale which includes 6 items, and a taxane subscale which include 16 items and has two domains (neurotoxicity and taxane). Total FACT-Taxane is the sum of all the 5 subscales. Each item is scored from 0 to 4 giving a total overall score from 0 worst quality of life to 172 best quality of life. The LS mean was calculated using an ANCOVA model adjusted for change scores and baseline scores." (NCT00308750)
Timeframe: Baseline, Cycle 1 (Week 3), Cycle 2 (Week 6), Cycle 3 (Week 9), Cycle 4 (Week 12), Cycle 5 (Week 15) and Cycle 6 (Week 18) [21-day cycle each]

,,
Interventionunits on a scale (Least Squares Mean)
Cycle 1 (Week 3)Cycle 2 (Week 6)Cycle 3 (Week 9)Cycle 4 (Week 12)Cycle 5 (Week 15)Cycle 6 (Week 18)
Docetaxel/Carboplatin-2.17-3.13-4.40-2.58-7.74-0.34
Enzastaurin/Pemetrexed/Carboplatin-3.16-6.22-1.89-2.28-2.522.77
Pemetrexed/Carboplatin-0.78-5.66-1.52-0.77-3.11-2.81

[back to top]

Overall Survival (OS)

OS was the duration from the date of randomization to the date of death from any cause. For participants who were alive, OS was censored at the date of last follow-up visit or at the date of last contact. (NCT00308750)
Timeframe: Baseline to date of death from any cause up to 35 months

Interventionmonths (Median)
Enzastaurin/Pemetrexed/Carboplatin7.2
Pemetrexed/Carboplatin12.7
Docetaxel/Carboplatin9.2

[back to top]

Time to Disease Progression

Time to disease progression was defined as the time from randomization to the first date of documented disease progression or death if the participant dies due to disease progression. Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Progressive disease (PD) was defined as having at least a 20% increase in sum of the longest diameter of target lesions. For participants who have not had documented disease progression, time to disease progression was censored at the date of death or date of last visit. For participants who received other anti-tumor therapy prior to disease progression, time to disease progression was censored at the first available date of other anti-tumor therapy. (NCT00308750)
Timeframe: Baseline to measured PD up to 22.3 months

Interventionmonths (Median)
Enzastaurin/Pemetrexed/Carboplatin4.6
Pemetrexed/Carboplatin6.0
Docetaxel/Carboplatin4.1

[back to top]

Number of Participants With Adverse Events (AEs) or Deaths

Data presented are the number of participants who experienced 1 or more AEs or any serious AEs (SAEs) regardless of causality, or deaths during the study including 30 days after treatment discontinuation. A summary of SAEs and other non-serious AEs is located in the Reported Adverse Events section of this report. (NCT00308750)
Timeframe: Baseline through study completion up to 6 cycles (21-day cycle each) and 30-day safety follow-up

,,
InterventionParticipants (Count of Participants)
AEsSAEsDeaths Due to AEs
Docetaxel/Carboplatin69264
Enzastaurin/Pemetrexed/Carboplatin63353
Pemetrexed/Carboplatin70205

[back to top]

Time-to-Treatment Failure (TTF)

TTF was defined as the time from randomization to the first observation of PD, death due to any cause, or early discontinuation of treatment. Response was defined using RECIST, version 1.0 criteria. PD was defined as having at least a 20% increase in sum of longest diameter of target lesions. TTF was censored at the date of the last follow-up visit for participants who did not discontinue early, who were still alive, and who have not progressed. (NCT00308750)
Timeframe: Baseline to stopping treatment up to 14.1 months

Interventionmonths (Median)
Enzastaurin/Pemetrexed/Carboplatin2.6
Pemetrexed/Carboplatin3.8
Docetaxel/Carboplatin2.6

[back to top]

Change From Baseline in Total Functional Assessment of Cancer Therapy-Lung (FACT-L) Scale

"The FACT-L version 4 scale is used to assess health-related quality of life (HRQoL) in participants with lung cancer. The FACT-L has 5 subscales: Physical Well-Being (PWB), Social and Family Well-Being (SFWB) and Functional Well-Being (FWB) subscales which include 7 items each, Emotional Well-Being (EWB) subscale which includes 6 items, and a Lung-Cancer Specific (LCS) subscale which include 7 items. Total FACT-L is the sum of all 5 subscales. Each item is scored from 0 to 4 giving a total overall score from 0 equal to worst quality of life to 136 equal to best quality of life. The Least Square (LS) mean was calculated using an analysis of covariance (ANCOVA) model adjusted for change scores and baseline scores." (NCT00308750)
Timeframe: Baseline, Cycle 1 (Week 3), Cycle 2 (Week 6), Cycle 3 (Week 9), Cycle 4 (Week 12), Cycle 5 (Week 15) and Cycle 6 (Week 18) [21-day cycle each]

,,
Interventionunits on a scale (Least Squares Mean)
Cycle 1 (Week 3)Cycle 2 (Week 6)Cycle 3 (Week 9)Cycle 4 (Week 12)Cycle 5 (Week 15)Cycle 6 (Week 18)
Docetaxel/Carboplatin-3.33-2.16-1.93-0.81-4.693.38
Enzastaurin/Pemetrexed/Carboplatin-3.98-3.250.390.311.897.38
Pemetrexed/Carboplatin1.12-1.540.643.54-0.26-0.83

[back to top]

Number of Participants With Complete Response (CR) or Partial Response (PR) [Tumor Response]

Response was defined using RECIST, version 1.0 criteria. Participants with a best response of CR or PR were considered to have had a tumor response. CR was defined as the disappearance of all target lesions. PR was defined as having at least a 30% decrease in sum of longest diameter of target lesions. (NCT00308750)
Timeframe: Baseline to measured PD up to 22.3 months

InterventionParticipants (Count of Participants)
Enzastaurin/Pemetrexed/Carboplatin9
Pemetrexed/Carboplatin16
Docetaxel/Carboplatin19

[back to top]

Patterns of Response

(NCT00310050)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Partial ResponseStable disease
Pemetrexed in Combination With Concomitant Radiotherapy12

[back to top]

Qualitative Dose-limiting Toxicities of Pemetrexed in Combination With Radiation Therapy

Toxicity will be determined using the revised NCI Common Toxicity Criteria (CTC) version 3.0 for Toxicity and Adverse Event Reporting. Number of events with grade 1-5 will be reported. (NCT00310050)
Timeframe: 42 days

Interventionevents (Number)
Grade 1Grade 2Grade 3Grade 4Grade 5
Pemetrexed in Combination With Concomitant Radiotherapy227300

[back to top]

Number of Participants That Survived

(NCT00310050)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Pemetrexed in Combination With Concomitant Radiotherapy2

[back to top]

Pemetrexed Population Pharmacokinetics (PK): Clearance

Clearance (CL) can be defined as the volume of plasma which is completely cleared of drug (pemetrexed) per unit time. Total body clearance is calculated after intravenous administration of the drug (pemetrexed) and is measured by taking plasma samples at various timepoints and measuring the amount of pemetrexed in the plasma. (NCT00316225)
Timeframe: Cycle 1 and Cycle 2: before the end of infusion (approximately 9.5 minutes), 2 hours, 9-10 hours, 24-48 hours, 480-528 hours (20 to 22 days) after start of pemetrexed infusion

Interventionmilliliter per minute (mL/min) (Mean)
Pemetrexed85.6

[back to top]

Overall Tumor Response

"Overall tumor response was determined using Response Evaluation Criteria In Solid Tumors (RECIST), which defines when cancer patients improve (respond), stay the same (stabilize), or worsen (progression) during treatments.~CR (complete response) = disappearance of all target lesions. PR (partial response) = 30% decrease in the sum of the longest diameter of target lesions.~PD (progressive disease) = 20% increase in the sum of the longest diameter of target lesions.~SD (stable disease) = small changes that do not meet above criteria." (NCT00316225)
Timeframe: baseline, up to 18 weeks

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Pemetrexed028129

[back to top]

Overview of Adverse Events

Any untoward medical occurrence in a patient who received study drug was considered an adverse event (AE), without regard to possibility of causal relationship. Treatment-emergent adverse events (TEAE): those which occurred or worsened after baseline. An adverse event resulting in any of the following outcomes, or deemed to be significant for any other reason, was considered to be a serious adverse event (SAE): death; initial or prolonged inpatient hospitalization; a life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly. (NCT00316225)
Timeframe: baseline, up to 18 weeks

Interventionparticipants (Number)
TEAE - All, regardless of causalityTEAE - Possibly related to study drugSAE - All, regardless of causalitySAE - Possibly related to study drugDiscontinuations Due to SAEs (including death)Discontinuations - Possibly related to study drugDiscontinuations Due to Nonserious AEsDeaths - On StudyDeaths - Possibly related to study drugDeaths - Within 30 days of Study Discontinuation
Pemetrexed2823121410200

[back to top]

Pemetrexed Population Pharmacokinetics: Volume of Distribution

Volume of distribution is the theoretical size of the compartment necessary to account for total drug amount in the body if it were present throughout the body in the same concentration found in plasma. Volume of distribution is defined as distribution of pemetrexed in the body and is determined by volume of distribution = dose/drug concentration. By knowing dose and measuring concentration of pemetrexed in plasma, volume was calculated. Central volume (V1) was determined by dose/peak serum level of pemetrexed. Peripheral volume (V2) is sum of all tissue spaces outside the central compartment. (NCT00316225)
Timeframe: Cycle 1 and Cycle 2: before the end of infusion (approximately 9.5 minutes), 2 hours, 9-10 hours, 24-48 hours, 480-528 hours (20 to 22 days) after start of pemetrexed infusion

InterventionLiters (L) (Mean)
Central Volume of DistributionPeripheral Volume of Distribution (V2)Peripheral Volume of Distribution (V3)
Pemetrexed6.618.911.26

[back to top]

Discontinuations Due to Adverse Events

Adverse events were coded using the Medical Dictionary for Regulatory Activities, Version 11.0. (NCT00316225)
Timeframe: baseline, up to 18 weeks

Interventionparticipants (Number)
Atrial fibrillationFebrile neutropenia (possibly related)Pneumonia (resulted in death)Respiratory failure (resulted in death)
Pemetrexed1111

[back to top]

Number of Participants With Common Toxicity Criteria - National Cancer Institute Grade 3 and Grade 4 Toxicities

"Number of participants with laboratory and non-laboratory toxicities possibly related to study drug, which were graded using the Common Terminology Criteria for Adverse Events version 3.0 (CTCAE v3.0) for defining and grading specific adverse events. Grades range from 0 (none) to 5 (death). Grade 3 is severe and Grade 4 is life-threatening.~NOS = Not otherwise specified." (NCT00316225)
Timeframe: baseline, up to 18 weeks

Interventionparticipants (Number)
LeukocytopeniaNeutropenia/GranulocytopeniaPlateletsAscites (Non-Malignant)Febrile NeutropeniaPain Pulmonary/Upper Respiratory-Chest/Thorax NOSPleural Effusion (Non-Malignant)
Pemetrexed1111111

[back to top]

Objective Best Tumor Response

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. (NCT00320515)
Timeframe: baseline to measured progressive disease (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until disease progression, or up to 12 months after enrollment)

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Pemetrexed + Cisplatin11522246

[back to top]

Progression Free Survival

The period from study entry until disease progression or death on study, whichever occurred first. (NCT00320515)
Timeframe: baseline to measured progressive disease or death (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until disease progression, or up to 12 months after enrollment)

Interventionmonths (Median)
Pemetrexed + Cisplatin4.9

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00320515)
Timeframe: baseline to date of death from any cause (Survival follow-up were performed every 2 cycles during therapy and approximately every 3 months during post-therapy until death or up to 12 months after enrollment)

Interventionmonths (Median)
Pemetrexed + Cisplatin11.8

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. (NCT00320515)
Timeframe: time of response to progressive disease or death (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until disease progression, or up to 12 months after enrollment)

Interventionmonths (Median)
Pemetrexed + Cisplatin5.4

[back to top]

Disease-free Survival

Disease-free survival (DFS) was defined as the time from randomization to an event. Events include disease recurrence, new primary of lung cancer, second primaries or death, whichever occurred first; however, it should be noted that patients with new primaries at other non-lung sites should have continued followup for recurrence of the original cancer. Patients that have not had an event reported at analysis were censored at their last date of disease assessment. (NCT00324805)
Timeframe: From registration to death, up to 10 years

Interventionmonths (Median)
Arm I (Chemotherapy)42.9
Arm II (Chemotherapy, Bevacizumab)40.6

[back to top]

Overall Survival

Overall survival (OS) was defined as the time from randomization to death from any cause, and patients who were thought to be alive at the time of final analysis were censored at the last date of contact. The study failed to meet its primary endpoint. (NCT00324805)
Timeframe: From registration to death, up to 10 years

Interventionmonths (Median)
Arm I (Chemotherapy)NA
Arm II (Chemotherapy, Bevacizumab)85.8

[back to top]

Time To Treatment Failure (TTTF)

TTTF is defined as the time from date of study enrollment to the first documented date of death, PD, or study treatment discontinuation due to adverse event (AE). For participants not known to have discontinued as of the data cut-off date, TTTF is censored at the last contact date. For participants who discontinued for reasons other than death, PD, or AE, TTTF is censored at the date of discontinuation. (NCT00325234)
Timeframe: Baseline to end of treatment (up to 21.9 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin4.8
Gemcitabine/Vinorelbine5.1

[back to top]

Time to Response

Time to response (Complete Response(CR) or Partial Response (PR) is defined as the time from the date of study enrollment to the first date when the measurement criteria are met for complete response or partial response (whichever status is recorded first). CR=Disappearance of target lesions lesions. PR=≥30% size decrease of lesions. (NCT00325234)
Timeframe: Baseline to response (up to 7.8 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin1.8
Gemcitabine/Vinorelbine1.8

[back to top]

Time to Progressive Disease (PD)

Time to PD is defined as the time from the date of study enrollment to the first documented date of PD or death from study disease. For participants who die from causes other than study disease and without PD, time to PD was censored at the date of death. For participants not known to have died as of the data cut-off date and do not have PD, time to PD was censored at the last contact date. For participants who received subsequent chemotherapy (after discontinuation from the study chemotherapy) prior to disease progression, time to PD was censored at the date of subsequent chemotherapy. (NCT00325234)
Timeframe: Baseline to measured PD (up to 25.1 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin5.1
Gemcitabine/Vinorelbine5.6

[back to top]

Duration of Response (DOR)

DOR-RECIST criteria of (Complete Response [CR =Disappearance of lesions] or Partial Response [PR=≥30% size decrease of lesions]) is defined as time from the date when measurement criteria are met for CR or PR until the date of first observation of progressive disease (PD) or death from study disease. For participants who die from causes other than study disease and without PD, DOR will be censored at the date of death. For participants who have not died as of the data cut-off date who are without PD, DOR was censored at last contact date. (NCT00325234)
Timeframe: Time of response to progressive disease (up to 19 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin7.7
Gemcitabine/Vinorelbine7.5

[back to top]

Tumor Response Rate

Participants with best overall response determined from complete response (CR) or partial response (PR) according to Response Criteria in Solid Tumors (RECIST) criteria. For CR or PR, best response must be confirmed. A second assessment performed at 28 days. Two determinations of CR before progression required for rate to=CR. Evaluations include: CR=Disappearance of lesions. PR=≥30% size decrease of lesions. Progressive Disease (PD)=≥20% size increase of lesions. Stable Disease (SD)=Not enough shrinkage for PR nor enough increase for PD. Overall Response Rate=PR+CR/Qualified Participants*100. (NCT00325234)
Timeframe: Baseline up to 30 days of follow-up after 21 cycles of treatment

,
Interventionpercentage of participants (Number)
Overall ResponseComplete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Gemcitabine/Vinorelbine29.53.326.234.427.98.2
Pemetrexed/Carboplatin26.60.026.635.926.610.9

[back to top]

Number of Participants With Adverse Events (AE)

A listing of adverse events is presented in the Reported Adverse Event Module. (NCT00325234)
Timeframe: every cycle up to twenty-one 21-day cycles (plus 30 days of follow-up)

,
Interventionparticipants (Number)
Adverse EventsSerious Adverse Events
Gemcitabine/Vinorelbine6622
Pemetrexed/Carboplatin6418

[back to top]

Number of Participants With Complete Tumor Resection

(NCT00330915)
Timeframe: surgery following 3 cycles (21-day cycles) of chemotherapy

Interventionparticipants (Number)
Pemetrexed37

[back to top]

Number of Participants Receiving Sphincter Saving Surgery

(NCT00330915)
Timeframe: surgery following 3 cycles (21-day cycles) of chemotherapy

Interventionparticipants (Number)
Surgery - YesSurgery - No
Pemetrexed2512

[back to top]

Pathological Complete Response (pCR)

Pathological complete response was defined as the absence of any tumor cells. (NCT00330915)
Timeframe: surgery following 3 cycles (21-day cycles) of chemotherapy

Interventionparticipants (Number)
Complete Response - YesComplete Response - NoComplete Response - Not Determined
Pemetrexed0136

[back to top]

Feasibility of Pemetrexed Prior to Surgery

Feasibility was defined as the ability to receive the total planned dose of Pemetrexed administered over a period of no more than 9 weeks permitting scheduling conflict. A ±5 percent variance in the calculated total dose was allowed. (NCT00330915)
Timeframe: 3 cycles (21-day cycles)

Interventionparticipants (Number)
Feasibility - YesFeasibility - No
Pemetrexed334

[back to top]

Estimated Probability of One Year Progression-free Survival

Progression free survival (PFS) is the duration from enrollment until first disease progression or death. For patients not known to have died as of the data cut-off date and who do not have progressive disease, PFS is censored at the last radiological assessment date. (NCT00350792)
Timeframe: baseline to measured progressive disease or death, 1 year

Interventionpercentage of patients (Median)
Pemetrexed + Carboplatin13.1

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00350792)
Timeframe: baseline to date of death from any cause (up to 14.5 months)

Interventionmonths (Median)
Pemetrexed + Carboplatin10.4

[back to top]

Percentage of Participants With a Complete or Partial Tumor Response (Overall Tumor Response)

Tumor response is defined as the percentage of patients with either a complete response or a partial response. Response was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response=disappearance of all target lesions and Partial Response=30% decrease in sum of longest diameter of target lesions. (NCT00350792)
Timeframe: baseline to measured objective tumor response (up to six 21-day cycles)

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin28.6

[back to top]

Time to Treatment Failure

Defined as the time from study enrollment to the first observation of disease progression, death as a result of any cause, or early discontinuation of treatment. Time to treatment failure was censored at the date of the last follow-up visit for patients who did not discontinue early, who were still alive, and who have not progressed. (NCT00350792)
Timeframe: baseline to stopping treatment (up to six 21-day cycles)

Interventionweeks (Median)
Pemetrexed + Carboplatin16.0

[back to top]

Time to Treatment Failure

Defined as the time from study enrollment to the first observation of disease progression, death as a result of any cause, or early discontinuation of treatment. Time to treatment failure was censored at the date of the last follow-up visit for patients who did not discontinue early, who were still alive, and who have not progressed. (NCT00350792)
Timeframe: baseline to stopping treatment (up to six 21-day cycles)

Interventionweeks (Mean)
Pemetrexed + Carboplatin12.95

[back to top]

Number of Participants With Grade 3 and Grade 4 Adverse Events

By Safety, the intent was to capture, tabulate, list all of the grade 3 and 4 adverse effects seen by this protocol. For each toxicity, we followed the Common Toxicity Criteria(NCI CTC)Version 2.0 Toxicity scale guidelines. (NCT00351039)
Timeframe: 26 Months

InterventionParticipants (Number)
Experimental: Bevacizumab, Erlotinib, Pemetrexed8

[back to top]

Objective Tumor Response

Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. (NCT00356525)
Timeframe: baseline to time of response (up to 17.5 months)

,,,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Less Than One Year: Pemetrexed07322
Less Than One Year: Pemetrexed + Gemcitabine00920
One Year or Greater: Pemetrexed + Carboplatin02500
One Year or Greater: Pemetrexed + Gemcitabine01611

[back to top]

Overall Survival

Overall survival is the number of participants who were alive when the trial was terminated. (NCT00356525)
Timeframe: baseline to trial termination (17.5 months)

Interventionparticipants alive (Number)
Less Than One Year: Pemetrexed6
Less Than One Year: Pemetrexed + Gemcitabine4
One Year or Greater: Pemetrexed + Carboplatin6
One Year or Greater: Pemetrexed + Gemcitabine7

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00363415)
Timeframe: baseline to date of death from any cause (up to 19.6 months)

Interventionmonths (Median)
Pemetrexed + Carboplatin8.1
Etoposide + Carboplatin10.6

[back to top]

Progression Free Survival

The period from study entry until disease progression, death or date of last contact. (NCT00363415)
Timeframe: baseline to measured progressive disease (up to 14.7 months)

Interventionmonths (Median)
Pemetrexed + Carboplatin3.8
Etoposide + Carboplatin5.4

[back to top]

Change From Baseline to Each Cycle in Functional Assessment of Cancer Therapy - Lung (FACT-L)

FACT-L measures following domains of health-related quality of life (HR-QL): physical well-being, social/family well-being, emotional well-being, functional well-being, and additional concerns of lung cancer. Total scores range from 0 to 136, with higher scores representing better HR-QL. A clinically meaningful change is considered to be 5 points. (NCT00363415)
Timeframe: baseline and 6 cycles (21-day cycles)

,
Interventionunits on a scale (Mean)
Baseline (n=384, n=383)Cycle 1 Change from Baseline (n=270, n=275)Cycle 2 Change from Baseline (n=283, n=310)Cycle 3 Change from Baseline (n=225, n=277)Cycle 4 Change from Baseline (n=199, n=259)Cycle 5 Change from Baseline (n=140, n=203)Cycle 6 Change from Baseline (n=98, n=146)
Etoposide + Carboplatin87.791.551.731.701.691.943.73
Pemetrexed + Carboplatin87.42-0.220.170.06-0.140.270.34

[back to top]

Number of Participants in Subgroups: LDH<=Upper Limit of Normal and History of Brain Metastases=Yes

Number of participants with Low Density Lipoprotein <=upper limit of normal and the number of participants with a history of brain metastases. This post-hoc outcome replaces the one for Overall Survival (Subgroups: LDH<=Upper Limit of Normal and History of Brain Metastases=Yes). (NCT00363415)
Timeframe: baseline to date of death due to any cause (up to 19.6 months)

,
Interventionparticipants (Number)
LDH: <=Upper Limit of NormalHistory of Brain Metastases: Yes
Etoposide + Carboplatin16941
Pemetrexed + Carboplatin16743

[back to top]

Overall Survival (Subgroups)

The effects of individual baseline factors (sex, race, Eastern Cooperative Oncology Group (ECOG) performance, region, lactate dehydrogenase (LDH), age, number of metastatic sites, and history of brain metastases) on overall survival are reported. For two subgroups - LDH<=upper limit of normal and brain metastases=yes, the upper limits of the 95% confidence interval were not calculable for the etoposide+carboplatin group - instead the number of participants in these two subgroups are presented as a post-hoc outcome measure. (NCT00363415)
Timeframe: baseline to date of death from any cause (up to 19.6 months)

,
Interventionmonths (Median)
Sex: Male (n=325, n=330)Sex: Female (n=128, n=125)Race: Caucasian (n=391, n=379)Race: Non-Caucasian (n=62, n=76)ECOG: 0 or 1 (n=398, n=398)ECOG: 2 (n=54, n=55)Region: United States (n=92, n=83)Region: European Union (n=279, n=278)Region: Intercontinential Region (n=82, n=94)LDH: >Upper Limit of Normal (n=276, n=273)Age: <= 65 years (n=267, n=275)Age: >65 years (n=186, n=180)Number Metastatic Sites: <=2 (n=172, n=204)Number Metastatic Sites: >=3 (n=273, n=246)History of Brain Metastases: No (n=410, n=412)
Etoposide + Carboplatin10.411.611.28.711.35.211.311.29.99.311.59.711.510.310.6
Pemetrexed + Carboplatin8.28.18.27.18.56.28.18.57.27.28.47.710.07.78.2

[back to top]

Maximum Tolerated Dose as Measured by the Number of Dose Limiting Toxicities Seen in Cohort.

"Only dose limiting toxicities (DLT) were collected. DLTs were graded according to the National Cancer Institute's Common Toxicity Criteria for adverse events version 3.0 (CTCAE v3.0) 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~The occurrence of any of the following toxicities during the first treatment cycle constitutes DLT in this study:~Grade 3 and/or 4 non-hematologic toxicity other than grade 3 nausea or vomiting.~Grade 3 and/or 4 unexpected non-hematologic toxicities. Grade 4 vomiting despite maximal antiemetic support. Grade 4 neutropenia and fever during first cycle. Grade 4 neutropenia on Day 1 of 2nd treatment cycle despite growth factor support or grade 4 thrombocytopenia on Day 1 of 2nd treatment cycle." (NCT00369629)
Timeframe: From the day that the first treatment is given through the first 28 day period for each patient.

,,
InterventionDLT (Number)
Perforated sigmoid diverticulitisFebrile neutropenia
Cohort 110
Cohort 200
Cohort 301

[back to top]

Mean Deoxycytidine Kinase (dCK) Expression Evaluated at Cycle 1, Cycle 2, and Cycle 3

dCK and hENT expression (see Outcome #2) on normal lymphocytes were measured after Pemetrexed administration to evaluate if there was reproducible timing of maximum dCK expression, and to assess proper time interval between pemetrexed and gemcitabine for treatment of patients with advanced Non-Small Cell Lung Cancer (NSCLC). Values are calculated as ratio between thereshold cycles (number of polymerase chain reaction [PCR] cycles) with respect to a reference gene; in this case glyceraldehyde 3-phosphate dehydrogenase (GAPDH). (NCT00370292)
Timeframe: pre-dose, 1, 2, 4, 6, 24, and 48 hours post-dose (3 cycles)

,,
InterventionmRNA relative values (ratio with GAPDH) (Mean)
Pre-Dose1 Hour Post-Dose2 Hours Post-Dose4 Hours Post-Dose6 Hours Post-Dose24 Hours Post-Dose48 Hours Post-Dose
dCK - Cycle 10.920.960.960.930.920.960.97
dCK - Cycle 20.920.950.960.940.910.970.96
dCK - Cycle 30.920.960.970.920.910.960.97

[back to top]

Mean Human Equilibrative Nucleoside Transporter 1 (hENT) Expression Evaluated at Cycle 1, Cycle 2, and Cycle 3

dCK (see Outcome #1)and hENT expression (see Outcome #2) on normal lymphocytes were measured after Pemetrexed administration to evaluate if there was reproducible timing of maximum dCK expression, and to assess proper time interval between pemetrexed and gemcitabine for treatment of patients with advanced Non-Small Cell Lung Cancer (NSCLC). Values are calculated as ratio between thereshold cycles (number of polymerase chain reaction [PCR] cycles) with respect to a reference gene; in this case glyceraldehyde 3-phosphate dehydrogenase (GAPDH). (NCT00370292)
Timeframe: pre-dose, 1, 2, 4, 6, 24, and 48 hours post-dose (3 cycles)

,,
InterventionmRNA relative values (ratio with GAPDH) (Mean)
Pre-Dose1 Hour Post-Dose2 Hours Post-Dose4 Hours Post-Dose6 Hours Post-Dose24 Hours Post-Dose48 Hours Post-Dose
hENT - Cycle 10.860.880.880.860.860.890.90
hENT - Cycle 20.860.890.900.880.860.910.91
hENT - Cycle 30.870.900.900.860.860.910.91

[back to top]

Best Objective Tumor Response

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. (NCT00370292)
Timeframe: baseline to measured response (every 14 days for 6 cycles)

,
Interventionparticipants (Number)
Partial ResponseStable DiseaseProgressive DiseaseEarly DeathUnconfirmed Stable Disease
Pemetrexed - After Amendment04003
Pemetrexed - Before Amendment12315

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. Complete response (CR) = disappearance of all target lesions. Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions. (NCT00374868)
Timeframe: time of response to progressive disease (up to 620 days)

Interventiondays (Mean)
Pemetrexed + Cisplatin184.100

[back to top]

Duration of Stable Disease

Defined as time from study enrollment to the first progression of disease, complete response, partial response, or death from any cause. Complete response (CR) = disappearance of all target lesions. Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions. Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions. Stable disease (SD) = small changes that do not meet above criteria. (NCT00374868)
Timeframe: time of no response or progression (up to 620 days)

Interventiondays (Median)
Pemetrexed + Cisplatin80

[back to top]

Progression-Free Survival

Defined as the time from study enrollment until disease progression or death from any cause. (NCT00374868)
Timeframe: baseline to measured progressive disease (up to 620 days)

Interventiondays (Median)
Pemetrexed + Cisplatin203

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00374868)
Timeframe: baseline to date of death from any cause (up to 620 days)

Interventiondays (Mean)
Pemetrexed + Cisplatin264.190

[back to top]

Time to Progressive Disease

Defined as the time from study enrollment to the first date of disease progression. Time to disease progression was censored at the date of death if death was due to other cause. Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions. (NCT00374868)
Timeframe: baseline to measured progressive disease (up to 620 days)

Interventiondays (Mean)
Pemetrexed + Cisplatin260.182

[back to top]

Best Overall Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment. Complete response (CR) = disappearance of all target lesions. Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions. Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions. Stable disease (SD) = small changes that do not meet above criteria." (NCT00374868)
Timeframe: baseline to measured progressive disease (up to 620 days)

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot EvaluatedNot PerformedMissing
Pemetrexed + Cisplatin213131333

[back to top]

Time to Treatment Failure (TTF)

Time from study enrollment to first observation of disease progression, death from any cause, or early discontinuation of treatment (including toxicity or if patient had stable disease after 4 cycles). TTF was censored at date of last follow-up visit for patients who did not discontinue early, who were still alive, and who had not progressed. (NCT00374868)
Timeframe: baseline to stopping treatment (up to 620 days)

Interventiondays (Median)
Pemetrexed + Cisplatin133.5

[back to top]

Time to Response

Defined as time from study enrollment to the first Complete Response or Partial Response (using RECIST criteria). Complete response (CR) = disappearance of all target lesions. Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions. (NCT00374868)
Timeframe: baseline to response (up to 620 days)

Interventiondays (Mean)
Pemetrexed + Cisplatin111.816

[back to top]

Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment. Complete response (CR) = disappearance of all target lesions; Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions; Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions; Stable disease (SD) = small changes that do not meet above criteria." (NCT00377520)
Timeframe: baseline to measured progressive disease (up to 24 months)

Interventionparticipants (Number)
Completed ResponsePartial ResponseStable DiseaseProgressive DiseaseInevaluable
Pemetrexed0112112

[back to top]

Number of Participants With Adverse Events by Grade (Measures of Toxicity)

Adverse events were graded using the Common Terminology Criteria for Adverse Events version 3.0 (CTCAE v3.0) for defining and grading specific adverse events. A grading (severity) scale is provided for each adverse event term. Grades range from 0 (none) to 5 (death). The worst grade event per cycle is reported. (NCT00377520)
Timeframe: every 21-day cycle (up to 24 months)

Interventionparticipants (Number)
Leukopenia - Grade 1Leukopenia - Grade 2Leukopenia - Grade 3Leukopenia - Grade 4Thrombocytopenia - Grade 1Thrombocytopenia - Grade 2Thrombocytopenia - Grade 3Thrombocytopenia - Grade 4Neutropenia - Grade 1Neutropenia - Grade 2Neutropenia - Grade 3Neutropenia - Grade 4Anemia - Grade 1Anemia - Grade 2Anemia - Grade 3Anemia - Grade 4Coagulation - Grade 1Coagulation - Grade 2Coagulation - Grade 3Coagulation - Grade 4Nausea/vomiting - Grade 1Nausea/vomiting - Grade 2Nausea/vomiting - Grade 3Nausea/vomiting - Grade 4Gastrointestinal - Grade 1Gastrointestinal - Grade 2Gastrointestinal - Grade 3Gastrointestinal - Grade 4Alopecia - Grade 1Alopecia - Grade 2Dermatologic - Grade 1Dermatologic - Grade 2Dermatologic - Grade 3Dermatologic - Grade 4Alkaline phosphatase - Grade 1Alkaline phosphatase - Grade 2Alkaline phosphatase - Grade 3Alkaline phosphatase - Grade 4Serum glutamic-oxaloacetic transaminase - Grade 1Serum glutamic-oxaloacetic transaminase - Grade 2Serum glutamic-oxaloacetic transaminase - Grade 3Serum glutamic-oxaloacetic transaminase - Grade 4Neurologic - Grade 1Neurologic - Grade 2Neurologic - Grade 3Neurologic - Grade 4Infection - Grade 1Infection - Grade 2Infection - Grade 3Infection - Grade 4Pulmonary - Grade 1Pulmonary - Grade 2Pulmonary - Grade 3Pulmonary - Grade 4Metabolic - Grade 1Metabolic - Grade 2Metabolic - Grade 3Metabolic - Grade 4Lymphatics - Grade 1Lymphatics - Grade 2Lymphatics - Grade 3Lymphatics - Grade 4Pain - Grade 1Pain - Grade 2Pain - Grade 3Pain - Grade 4Constitutional - Grade 1Constitutional - Grade 2Constitutional - Grade 3Constitutional - Grade 4Renal - Grade 1Renal - Grade 2Renal - Grade 3Renal - Grade 4Musculoskeletal - Grade 1Musculoskeletal - Grade 2Musculoskeletal - Grade 3Musculoskeletal - Grade 4Ocular - Grade 1Ocular - Grade 2Ocular - Grade 3Ocular - Grade 4Cardiovascular - Grade 1Cardiovascular - Grade 2Cardiovascular - Grade 3Cardiovascular - Grade 4Vascular - Grade 1Vascular - Grade 2Vascular - Grade 3Vascular - Grade 4Endocrine - Grade 1Endocrine - Grade 2Endocrine - Grade 3Endocrine - Grade 4
Pemetrexed34827140239391150001052206630104210400030006301042020007100301032304850100011002100110000100100

[back to top]

Duration of Response

Duration of overall tumor response was measured from the time of first documentation of complete response or partial response (whichever status was first recorded) until the date of progression-free survival, with censoring defined as: disease had not progressed, treatment was discontinued due to undocumented progression or toxicity/other reason, onset of new anti-tumor therapy or otherwise experienced death/progression after more than one missed (assessment) visit. (NCT00380718)
Timeframe: time of response to measured progressive disease or death from any cause (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until documented disease progression, or up to 18 months after enrollment)

Interventionmonths (Median)
Pemetrexed6.6

[back to top]

Proportion of Participants With a Complete or Partial Response (Objective Response Rate [ORR])

"The objective response rate (ORR) was defined as the proportion of participants who achieved a best response of either complete response (CR) or partial response (PR) (responders) based on the RECIST criteria. ORR=(CR+PR)/Number of Participants. The RECIST define when cancer patients improve (respond), stay the same (stabilize), or worsen (progression) during treatments." (NCT00380718)
Timeframe: baseline to measured progressive disease (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until documented disease progression, or up to 18 months after enrollment)

Interventionproportion of responders (Mean)
Pemetrexed0.182

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death from any cause. For patients who are alive, overall survival is censored at the last follow-up visit. (NCT00380718)
Timeframe: baseline to date of death from any cause (includes post-treatment follow-up of up to 18 months post-Last Patient Entered Treatment)

Interventionmonths (Median)
Pemetrexed20.2

[back to top]

Progression-Free Survival (PFS)

Time to PFS was defined as the time from the date of enrollment to the date of the first of the following events: objective disease progression or death due to any cause. Survival time frame includes post-treatment follow-up of up to 18 months post-Last Patient Entered Treatment. Patients were censored if their disease had not progressed, treatment was discontinued due to an undocumented progression or toxicity/other reason, onset of new anti-tumor therapy or otherwise experienced death/progression after more than one missed (assessment) visit. (NCT00380718)
Timeframe: baseline to measured progressive disease or death from any cause (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until documented disease progression, or up to 18 months after enrollment)

Interventionmonths (Median)
Pemetrexed6.9

[back to top]

Proportion of Participants With a Best Overall Response of Complete Response (CR), Partial Response (PR), and Stable Disease (SD) (Disease Control Rate [DCR])

DCR was defined as the proportion of best overall response of CR, PR, and SD. DCR=(CR+PR+SD)/Number of participants. Response was evaluated using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. (NCT00380718)
Timeframe: baseline to measured progressive disease (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until documented disease progression, or up to 18 months after enrollment)

Interventionproportion of participants (Mean)
Pemetrexed0.545

[back to top]

Time to Tumor Progression

Time to documented tumor progression was defined as the time from the date of enrollment to the first date of documented disease progression. Time to documented disease progression was censored at the date of death for participants who had not had documented disease progression. Otherwise, the censoring rules were the same as for Progression-Free Survival. (NCT00380718)
Timeframe: baseline to measured progressive disease (Tumor assessments were performed every 2 cycles during therapy and 6-8 weeks during post-therapy until documented disease progression, or up to 18 months after enrollment)

Interventionmonths (Median)
Pemetrexed6.9

[back to top]

Time to Treatment Failure

Time to treatment failure was define as the time from the date of enrollment to the date of the first of the following events: objective disease progression, death due to any cause, treatment discontinuation for undocumented progression, early treatment discontinuation for toxicity or other reason, or new anticancer treatment started. Time to treatment failure for participants who were still participating in the study without treatment failure at the time of analysis were treated as censored at the date of the last tumor assessment. (NCT00380718)
Timeframe: baseline to early treatment discontinuation or measured progressive disease or death from any cause (assessments every 2 cycles during therapy and 6-8 weeks during post-therapy until documented disease progression, or up to 18 months after enrollment)

Interventionmonths (Median)
Pemetrexed2.9

[back to top]

Overall Survival Rate

(NCT00383266)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin22.2

[back to top]

Overall Survival Rate

(NCT00383266)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin0

[back to top]

Toxicities

(NCT00383266)
Timeframe: 30 days following completion of treatment (maximum number of cycles = 6)

Interventionparticipants (Number)
Cardiac ischemia/infarctionPancreatitisTumor painChest/thorax painHemoglobinChest painDry eye syndromeAbdominal painConstipationDiarrheaDysphagiaHiccoughsMucositisNauseaOral cavity painStomatitisVomitingChillsEdemaFatigueFeverSweatingColitis (c. difficile)Infection without neutropeniaALTASTAlkaline phosphtaseLeukocytes (WBC)Low CO2LymphopeniaNeutrophilsPlateletsWeight lossAnorexiaDehydrationHypercalcemiaHyperglycemiaHyperkalemiaHypernatremiaHypoalbuminemiaHypocalcemiaHypoglycemiaHypokalemiaHyponatremiaLipaseBack painHerniaLimb painDizzinessHeadacheNeuropathy - motorNeuropathy - sensoryInsomniaMood alteration - anxietyCreatinineCreatinine clearanceAllergic rhintisCoughDyspneaHemorrhage - noseAlopeciaDry skinOther: skinPuritisRashSkin breakdown/decubitous ulcerHot flash
Pemetrexed + Carboplatin1111811233112611421515114223262112113333111111113315421124311211111

[back to top]

Overall Survival (OS)

OS is defined as the time from initiation of treatment to the date of any reason death while those living subjects will be censored at the last assessment date. (NCT00383266)
Timeframe: Until patient's death (median follow-up 293 days -- range (63-632 days))

Interventionmonths (Median)
Pemetrexed + Carboplatin9.6

[back to top]

Time to Disease Progression

-Progressive disease=at least a 20% increase in the sum of the LD of the target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions (NCT00383266)
Timeframe: Until patient progresses (median follow-up 293 days -- range (63-632 days)

Interventionmonths (Median)
Pemetrexed + Carboplatin3.6

[back to top]

Overall Response Rate (ORR)

"Overall response rate = complete response (CR) + partial response (PR) using RECIST.~CR=disappearance of all target lesions and disappearance of all non-target lesions and normalization of tumor marker level~PR=at least a 30% decrease in the sum of the longest diameter (LD) of the target lesions taking as reference the baseline sum LD" (NCT00383266)
Timeframe: Until patient progresses or dies (median follow-up 293 days -- range (63-632 days)

Interventionpercentage of participants (Number)
Complete responsePartial response
Pemetrexed + Carboplatin033

[back to top]

Progression Free Survival

baseline to measured progressive disease (NCT00383331)
Timeframe: baseline and every 14 or 21 day cycle (6-9 cycles), every 6 weeks post-therapy follow-up

Interventionmonths (Median)
21-Day Cycle1.66
14-Day Cycle5.04

[back to top]

Overall Survival

Survival time is defined as the time from date of randomization to death due to any cause. (NCT00383331)
Timeframe: baseline and every 14 or 21 day cycle (6-9 cycles), every 6 weeks post-therapy follow-up

Interventionmonths (Median)
21-Day Cycle5.1
14-Day Cycle8.1

[back to top]

Best Overall Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment." (NCT00383331)
Timeframe: baseline and every 14 or 21 day cycle (6-9 cycles), every 6 weeks post-therapy follow-up

,
Interventionparticipants (Number)
Complete ResponsePartial Response
14-Day Cycle00
21-Day Cycle02

[back to top]

Number of Patients With Grade ≥ 3 Toxicity (Phase I)

Grade 3/4 toxicity occurring in a patient within 1 cycle. (NCT00389805)
Timeframe: First cycle of treatment (3 weeks)

,
Interventionparticipants (Number)
NeutropeniaAnemiaThrombocytopeniaIncreased transaminasesFatigue
Arm A121012
Arm B10100

[back to top]

Number of Patients With Toxicity by NCI CTC v3.0 (Phase I)

Adverse events possibly related to treatment, graded according to the NCI CTCAE v3.0. (NCT00389805)
Timeframe: Up to 36 months

,
Interventionparticipants (Number)
NeutropeniaAnemiaThrombocytopeniaIncreased transaminasesAnorexiaConstipationCoughDehydrationDiarrheaDizzinessEdemaFatigueFeverInfectionNausea +/- vomitingNeuropathyRashRenal impairmentWeakness
Arm A12101245542541536104650
Arm B10156341344113466512

[back to top]

Number of Patients Experiencing a Dose-limiting Toxicity (Phase I)

Grade 4 thrombocytopenia or grade 3 thrombocytopenia associated with bleeding, requirement for transfusion or lasting >7 days; febrile neutropenia; grade 3 neutropenia associated with infection; any other grade >/=3 non-hematologic toxicity considered by the investigator to be related to study drug. (NCT00389805)
Timeframe: Up to 36 months

InterventionParticipants (Count of Participants)
Arm A3
Arm B0

[back to top]

Number of Participants Who Experience Adverse Events (Phase I)

Number of participants with treatment-related adverse events as assessed by CTCAE v3.0 (Phase I). (NCT00389805)
Timeframe: Throughout the entire study (up to 36 months).

InterventionParticipants (Count of Participants)
Arm A15
Arm B12

[back to top]

Maximum Tolerated Dose of Bortezomib in Combination With Pemetrexel (Phase I)

(NCT00389805)
Timeframe: Up to 36 months

InterventionMg/m^2 (Number)
Arm A1.3
Arm B1.6

[back to top]

Pharmacology Toxicity

Maximum common terminology criteria (CTC) Grade 3 or 4 toxicities possibly related to study drug are reported. The worst grade event per cycle is reported. Grades range from 0 (none) to 5 (death). Grade 3 events are severe and Grade 4 events are life-threatening. (NCT00391274)
Timeframe: first dose of study drug up to 24 months

,
Interventionparticipants (Number)
Serum Glutamic Pyruvic TransaminaseHemoglobinLeukocytesLymphopeniaNeutrophils/GranulocytesPlateletsHypokalemiaAnorexiaConstipationDiarrheaEnteritisFatigue (Asthenia, Lethargy, Malaise)Febrile NeutropeniaInfection (Clinical/Microbiological: Neutrophils)Infection (Unknown: Pneumonia)Mucositis/StomatitisNeuropathy: MotorPain: Thorax Not Otherwise Specified (NOS)Pericardial Effusion (Non-Malignant)Pulmonary/Upper Respiratory - OtherRash/Desquamation
Docetaxel03210290101515411011000
Pemetrexed174157010003200100111

[back to top]

Overall Survival

Overall survival was defined as the time from the date of study enrollment to the date of death due to any cause. Survival time was censored at the date of last contact for patients who were still alive or lost to follow-up. An amendment allowed for the collection of overall survival on an additional 43 survival events. At the time the original record was released, it was not possible to provide results with the 95% Confidence Interval (CI) since the upper limit was not calculable. The median and 95% CIs are now reported. (NCT00391274)
Timeframe: baseline to date of death from any cause (up to 24 months after study enrollment); amendment (up to 30 months after study enrollment)

,
Interventionmonths (Median)
Overall Survival (up to 24 months)Overall Survival (up to 30 months)
Docetaxel12.211.5
Pemetrexed11.711.4

[back to top]

Progression-Free Survival (PFS)

Progression-free survival (PFS) time was defined as the time from the date of study enrollment to the date of the first of the following events: objective disease progression or death due to any cause. For patients who were alive and had not progressed, PFS was censored at the last contact. (NCT00391274)
Timeframe: baseline to measured progressive disease (up to 24 months after study enrollment)

Interventionmonths (Median)
Pemetrexed2.8
Docetaxel3.1

[back to top]

Number of Patients With Disease Progression

Number of patients who have died or have had progression of disease. This outcome substitutes for the outcome on Duration of Response. (NCT00391274)
Timeframe: time of response to progressive disease (up to 12 months)

Interventionparticipants (Number)
Pemetrexed4
Docetaxel1

[back to top]

Overall Tumor Response

"Response based on Response Evaluation Criteria In Solid Tumors (RECIST), which define when cancer patients improve (respond), stay the same (stabilize), or worsen (progression) during treatments. CR (complete response) = disappearance of all target lesions; PR (partial response) = 30% decrease in the sum of the longest diameter of target lesions; PD (progressive disease) = 20% increase in the sum of the longest diameter of target lesions; SD (stable disease) = small changes that do not meet above criteria." (NCT00391274)
Timeframe: baseline to measured tumor response (up to 24 months after study enrollment)

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseEarly Death from Malignant DiseaseEarly Death from Other CausesUnknown
Docetaxel044636615
Pemetrexed0103349714

[back to top]

Toxicity Profile

Toxicities are assessed according to the National Cancer Institute's Common Terminology Criteria for Adverse Events, version 3.0. Toxicities are reported as the number of patients who experienced grade 3 or grade 4 adverse events after receiving at least one dose of on-study treatment. (NCT00391586)
Timeframe: 28 days after last on-study treatment

Interventionparticipants (Number)
AcneAnorexiaConfusionDehydrationDiarrheaDyspneaFatigueNasal hemorrhageInsomniaKidney painLymphocyte count decreasedMuscle weaknessNeutrophil count decreasedDesquamating rashSyncopeThrombosis (clotting)
Erlotinib Followed by Chemotherapy1111238111112111

[back to top]

Overall Survival

(NCT00394147)
Timeframe: 1 year

Interventionmonths (Median)
Pemetrexed and Gemcitabine5.9

[back to top]

Time to 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. (NCT00394147)
Timeframe: 1 year

Interventionmonths (Median)
Pemetrexed and Gemcitabine5.9

[back to top]

Objective Response Rate

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT00394147)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Pemetrexed and Gemcitabine2

[back to top]

Percentage of Participants Surviving Progression-Free at 6 Months (Progression Free Survival [PFS] Rate)

For this study, we used the exponential distribution (assumption done for the calculation of the sample size) to estimate the PFS rate. The PFS rate (%) and the 95% confidence intervals were calculated based on the following formula: exp(-6 λ) ± 1.96 * exp(-6 λ) * (-6 λ)/√r. Where λ was calculated based on the Maximum-Likelihood estimator for ln(λ) as given by (Collett 2003): ln(λ) = ln[ r / ∑ti ] with r = number of patients with events up to 6 months, ti = survival time of patient i (i=1,…,n), event or censored up to 6 months, and n= total number of patients per treatment group. (NCT00402051)
Timeframe: Randomization to Month 6

Interventionpercentage (Number)
Pemetrexed + Cisplatin52.8
Pemetrexed + Carboplatin39.3

[back to top]

Time to Treatment Failure (TTF)

Defined as time from randomization to the first date of disease progression, death due to any cause, or early discontinuation of treatment (any reason), whichever occurred first (NCT00402051)
Timeframe: Randomization to stopping of treatment, progression, death or initiation of further chemotherapy, whichever occurs first (up to 1 year)

Interventionmonths (Median)
Pemetrexed + Cisplatin3.0
Pemetrexed + Carboplatin3.4

[back to top]

Number of Participants With Tumor Response (as Basis for Response Rate)

"Best overall response was evaluated using RECIST Criteria which define when cancer patients improve (respond), stay the same (stabilize), or worsen (progression) during treatment. CR: complete response, disappearance of all target lesions; PR: partial response, 30% decrease in sum of the longest diameter of target lesions; PD: progressive disease, 20% increase in sum of the longest diameter of target lesions; SD: stable disease, small changes not meeting above criteria. Response Rate: number of participants with response(CR+PR)per total population, multiplied by 100 to give a percentage." (NCT00402051)
Timeframe: Every 6 weeks for 6 months during the treatment period, and every 3 months during the follow-up period, until disease progression

,
Interventionparticipants (Number)
Partial ResponseStable DiseaseDisease ProgressionUnknown/Not Done
Pemetrexed + Carboplatin1332155
Pemetrexed + Cisplatin213176

[back to top]

Overall Survival

Defined as the time from randomization to the date of death from any cause. (NCT00402051)
Timeframe: Randomization to date of death from any cause (up to 1 year)

Interventionmonths (Median)
Pemetrexed + Cisplatin11.7
Pemetrexed + Carboplatin8.9

[back to top]

Pharmacology Toxicities

Number of patients experiencing Grade 3 or 4 hematologic and non-hematologic adverse events (AEs) possibly related to study drug or protocol procedures in this study (a subset of those listed in the AE Module). AEs were graded using the Common Terminology Criteria for Adverse Events version 3.0 (CTCAE v3.0) for defining and grading specific adverse events. A grading (severity) scale is provided for each adverse event term. Grades range from 0 (none) to 5 (death). Grade 3 AEs are severe and undesirable; Grade 4 AEs are life-threatening or disabling. (NCT00402051)
Timeframe: Every 21-day cycle for up to 6 cycles

,
Interventionparticipants (Number)
Any Grade 3/4 ToxicityGrade 3/4 LeucopeniaGrade 3/4 NeutropeniaGrade 3/4 AnemiaGrade 3/4 ThrombocytopeniaGrade 3/4 NauseaGrade 3/4 VomitingGrade 3/4 FatigueGrade 3/4 AnorexiaGrade 3/4 Urinary Tract Infection
Pemetrexed + Carboplatin36121771151222
Pemetrexed + Cisplatin298115232210

[back to top]

Number of Patients With Confirmed Responses

"Confirmed tumor response (complete and partial) as measured by RECIST(Response Evaluation Criteria In Solid Tumors) criteria on 2 consecutive evaluations at least 6 weeks apart.~>~> Confirmed tumor response is at least a 30% decrease in the sum of the longest diameter of target lesions and no new lesions." (NCT00407550)
Timeframe: Two consecutive evaluations at least 6 weeks apart (up to 2 years)

Interventionparticipants (Number)
Gemzar x22
Gemzar x10

[back to top]

Adverse Event

Number of patients that experienced adverse events (grade 4 or more) as measured by NCI CTCAE (Common Terminology Criteria for Adverse Events) v3.0 (NCT00407550)
Timeframe: Gemzar x2 Arm every 21 days, Gemzar x1 Arm every 14 days (up to 2 years)

InterventionParticipants (Count of Participants)
Gemzar x26
Gemzar x14

[back to top]

Overall Survival

Overall survival time was defined as the number of months from registration to the date of death or last follow-up (NCT00407550)
Timeframe: Death or last follow-up (up to 2 years)

Interventionmonths (Median)
Gemzar x25.1
Gemzar x18.1

[back to top]

Progression-free Survival

Progression-free survival was defined as the number of months from registration to the date of disease progression or death, with patients who are alive and progression free being censored on the date of their last evaluation. (NCT00407550)
Timeframe: Time from registration to progression or death (up to 2 years)

Interventionmonths (Median)
Gemzar x21.66
Gemzar x15.04

[back to top]

Percentage of Participants Who Died During the Study

This outcome measure takes the place of the outcome measure for Overall Survival, which could not be reported since the median value could not be calculated. (NCT00409006)
Timeframe: Baseline up to 31 months

InterventionPercentage of Participants (Number)
Pemetrexed/Cisplatin/Gefitinib35.9
Pemetrexed/Cisplatin19.4

[back to top]

Number of Participants With Tumor Response

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes not meeting above criteria. Responder is a participant exhibiting a best overall study response of CR or PR. (NCT00409006)
Timeframe: Baseline to measured response or death, 12 weeks up to 31 months

InterventionParticipants (Number)
Pemetrexed/Cisplatin/Gefitinib18
Pemetrexed/Cisplatin11

[back to top]

Duration of Response for Responders

The duration of a complete response (CR; the disappearance of all target lesions) or partial response (PR; at least a 30% decrease in the sum of the longest diameter of target lesions) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. A responder is a patient exhibiting a best overall study response of CR or PR. (NCT00409006)
Timeframe: Time of response to progressive disease or death, 12 weeks up to 31 months

InterventionMonths (Median)
Pemetrexed/Cisplatin/Gefitinib12.29
Pemetrexed/Cisplatin4.14

[back to top]

Progression-Free Survival (PFS)

Defined as the time from randomization to the first observation of disease progression, or death due to any cause. (NCT00409006)
Timeframe: Baseline to first observation of disease progression or death, 12 weeks up to 31 months

InterventionMonths (Median)
Pemetrexed/Cisplatin/Gefitinib9.95
Pemetrexed/Cisplatin6.83

[back to top]

Response Rate (Complete and Partial) 2 Separate Cohorts of Relapsed NSCLC Cohort A: Pts Who Have Received Prior Chemo w/o Ever Having Received Bevacizumab. Cohort B: Pts Who Have Received Prior Bevacizumab.

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v 1.0) for target lesions and assessed by MRI or CT:~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~Overall Response (OR) = CR + PR, the best response recorded from the start of the treatment until disease progression/recurrence (taking as reference for PD the smallest measurements recorded since the treatment started)" (NCT00410904)
Timeframe: Up to 4 years

Interventionparticipants (Number)
Arm I - Cohort A, no Prior Bevacizumab (Avastin)10
Arm I - Cohort B, Prior Bevacizumab (Avastin)4

[back to top]

Progression-free Survival

Estimated with the standard Kaplan-Meier method, from which summary statistics of interest (median, 1-year rate, etc.) will be derived. Both point and 95% confidence interval estimates of all PFS and OS statistics will be calculated. 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 (NCT00410904)
Timeframe: The duration of time from start of treatment to time of progression, assessed up to 4 years

Interventionmonths (Median)
Arm I5.6

[back to top]

Overall Survival

Estimated with the standard Kaplan-Meier method, from which summary statistics of interest (median, 1-year rate, etc.) will be derived. Both point and 95% confidence interval estimates of all PFS and OS statistics will be calculated. (NCT00410904)
Timeframe: The time from start of treatment to time of death, assessed up to 4 years

Interventionmonths (Median)
Arm I11.5

[back to top]

Percentage of Participants With Objective Response (Objective Response Rate)

Tumor responder is defined as participants exhibiting a best overall study response of complete response (CR; disappearance of all target lesions) or partial response (PR; 30% decrease in sum of longest diameter of target lesions). Non-responders are those who did not meet the above criteria. (NCT00415168)
Timeframe: Baseline to time of response up to six or eight 21-day cycles of treatment

Interventionpercentage of responders (Number)
ResponderNon-Responder
Pemetrexed + Cisplatin32.167.9

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

Number of Participants Who Died During the Study

(NCT00415168)
Timeframe: During study drug therapy up to six or eight 21-day cycles or treatment; maximum duration of study follow-up was 17.4 months

Interventionparticipants (Number)
Study DiseaseCardiac Failure AcuteDeath (General Disorders/Administration Site Cond)Intestinal HemorrhagePancytopeniaSudden DeathMulti-Organ Failure
Pemetrexed + Cisplatin18111131

[back to top]

Progression Free Survival (PFS)

Defined as time from baseline to the date of disease progression or death on study, whichever occurs first. The PFS 1 definition from the United States Food and Drug Administration (FDA) draft guidance on clinical endpoints was used (FDA 2005). (NCT00415168)
Timeframe: Baseline to measured progressive disease or death up to six or eight 21-day cycles of treatment; maximum duration of study follow-up was 17.4 months

Interventionmonths (Median)
Pemetrexed + Cisplatin3.7

[back to top]

Overall Survival

Defined as the time from baseline to date of death due to any cause. Survival time is censored at the date of last contact for patients who are still alive or lost to follow up. (NCT00415168)
Timeframe: Baseline to date of death from any cause up to six or eight 21-day cycles of treatment; maximum duration of study follow-up was 17.4 months

Interventionmonths (Median)
Pemetrexed + Cisplatin8.8

[back to top]

Duration of Response

Measured from the time of first documentation of CR or PR (whichever status is first recorded) until the date of time to disease progression. (NCT00415168)
Timeframe: Time of response to progressive disease up to six or eight 21-day cycles of treatment; maximum duration of study follow-up was 17.4 months

Interventionmonths (Median)
Pemetrexed + Cisplatin3.8

[back to top]

Time to Treatment Worsening in Functional Assessment of Cancer Therapy - Head and Neck Cancer (FACT-H&N) Total Score

FACT-H&N consists of 39 items with 5-point rating scale from 0 (not at all) to 4 (very much). FACT-H&N Total score ranges from 0 to 148. Higher score represents a better quality of life. Time to worsening was defined as the first date of worsening in the FACT H&N Total score that was considered at least the prospectively defined minimally important difference (MID) as compared with participant's baseline score, or date of death from any cause. The MID for FACT H&N Total score was a decrease of 12 points. (NCT00415194)
Timeframe: Baseline (

InterventionMonths (Median)
Pemetrexed/Cisplatin3.29
Placebo/Cisplatin2.89

[back to top]

Progression-free Survival (PFS)

Objective PFS is defined as the time from date of randomization to date of objectively determined progressive disease (PD) or death from any cause, whichever comes first. PD was defined by Response Evaluation Criteria in Solid Tumors (RECIST). PD=at least a 20% increase in sum of longest diameter of target lesions. For participants who are not known to have died as of the data-inclusion cut-off date, and who do not have progressive disease, PFS will be censored at the date of the last objective progression-free disease assessment prior to the date of any subsequent systemic anticancer therapy. (NCT00415194)
Timeframe: baseline to measured progressive disease up to 33 months

Interventionmonths (Median)
Pemetrexed/Cisplatin3.61
Placebo/Cisplatin2.79

[back to top]

Percent of Participants With a Tumor Response (Response Rate)

Tumor Response is evaluated as CR (Complete Response) or PR (Partial Response) per Response Evaluation Criteria in Solid Tumors (RECIST criteria). Possible evaluations include: CR: Disappearance of all target lesions. PR: At least a 30% decrease in the size of target lesions. Response rate (%) = (number of participants with CR+PR/number of participants)*100 (NCT00415194)
Timeframe: Baseline to progressive disease or discontinuation of study treatment up to 11 months

InterventionPercentage of participants (Number)
Pemetrexed/Cisplatin12.1
Placebo/Cisplatin8.1

[back to top]

Overall Survival (OS)

OS duration is defined as the time from the date of randomization to the date of death from any cause. For each participant who is not known to have died as of the data-inclusion cut-off date, OS duration will be censored at the date of the participant's last contact prior to that cut-off date. (NCT00415194)
Timeframe: Baseline to date of death from any cause up to 36 months

InterventionMonths (Median)
Pemetrexed/Cisplatin7.33
Placebo/Cisplatin6.28

[back to top]

Duration of Response (DoR)

DoR is time from first observation of complete response (CR) or partial response (PR) to first observation of PD or death. Response is objective status of CR or PR using RECIST criteria. CR is disappearance of lesions. PR is >30% decrease in size of lesions. Responder is any participant with CR or PR. PD is at least 20% increase in sum of longest diameter of target lesions. For participants alive as of data-inclusion cut-off date and who do not have PD, DoR will be censored at date of last objective progression-free disease assessment before date of any subsequent systemic anticancer therapy. (NCT00415194)
Timeframe: time of response to progressive disease up to 24 months

InterventionMonths (Median)
Pemetrexed/Cisplatin5.29
Placebo/Cisplatin4.37

[back to top]

Pharmacokinetic (PK) Parameter: Area Under the IC83/LY2603618 Plasma Concentration Versus Time Curve From Time Zero to Infinity (AUC[0-∞])

AUC[0-∞] was calculated from the plasma concentration data of LY2603618 versus time profiles. (NCT00415636)
Timeframe: Day 1 and Day 9 of Cycle 1

,,,,,
Interventionnanograms*hour per milliliter (ng*h/mL) (Mean)
Day 1Day 9
LY2603618 105 mg/m^27640052000
LY2603618 150 mg/m^26190055000
LY2603618 195 mg/m^2122000119000
LY2603618 40 mg/m^2 (1-hour Infusion)1030011000
LY2603618 40 mg/m^2 (4.5-hour Infusion)75807540
LY2603618 70 mg/m^2951010900

[back to top]

Pharmacokinetic (PK) Parameter: Maximum Observed Plasma Concentration (Cmax) of IC83/LY2603618

Cmax was estimated from the plasma concentration data of LY2603618 versus time profiles. (NCT00415636)
Timeframe: Day 1 and Day 9 of Cycle 1

,,,,,
Interventionnanograms per millimeter (ng/mL) (Mean)
Day 1Day 9
LY2603618 105 mg/m^242204540
LY2603618 150 mg/m^242303560
LY2603618 195 mg/m^268407580
LY2603618 40 mg/m^2 (1-hour Infusion)15602010
LY2603618 40 mg/m^2 (4.5-hour Infusion)578614
LY2603618 70 mg/m^223102510

[back to top]

Number of Participants With Adverse Events (AEs)

Summary tables of serious AEs (SAEs) and all other non-serious adverse events (AEs) are located in the Reported Adverse Event Module. (NCT00415636)
Timeframe: baseline up to 24 months

,,,,,
InterventionParticipants (Count of Participants)
Other Non-Serious Adverse Events (AEs)Serious Adverse Events (SAEs)
LY2603618 105 mg/m^2138
LY2603618 150 mg/m^264
LY2603618 195 mg/m^232
LY2603618 40 mg/m^2 (1-hour Infusion)30
LY2603618 40 mg/m^2 (4.5-hour Infusion)31
LY2603618 70 mg/m^232

[back to top]

Percentage of Participants With Best Overall Response

Percentage of participants with tumor response (best confirmed overall response) assessed as complete response (CR) or partial response (PR) to treatment according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria. CR=disappearance of all target lesions; PR=30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD)=20% increase in sum of longest diameter of target lesions; Stable Disease (SD) =small changes that do not meet above criteria. Best Overall Response (%)=number of participants with CR+PR/number of participants in treatment arm * 100. (NCT00415636)
Timeframe: baseline up to 24 months

,,,,,
InterventionPercentage of participants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Not DeterminedBest Overall Response (CR + PR)
LY2603618 105 mg/m^20102070010
LY2603618 150 mg/m^200752500
LY2603618 195 mg/m^200505000
LY2603618 40 mg/m^2 (1 Hour)0066.733.300
LY2603618 40 mg/m^2 (4.5 Hours)00010000
LY2603618 70 mg/m^200505000

[back to top]

Objective Response Rate (ORR)

"The ORR is the number of patients that are responders ie those patients with a confirmed best objective response of complete response (CR) or partial response (PR) as defined by RECIST criteria.~The categories for best objective response are CR, PR, stable disease (SD)>= 6 weeks, progressive disease (PD) or NE." (NCT00418886)
Timeframe: Each patient was assessed for objective response from the sequence of RECIST scan data up to data cut off. RECIST tumour assessments carried out every 6 weeks (+/- 3 days) from randomisation until objective progression

InterventionPercentage of Participants (Number)
Vandetanib Plus Pemetrexed19.1
Placebo Plus Pemetrexed7.9

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

Progression-Free Survival (PFS) in the Overall Population

Median time (in weeks) from randomisation until objective disease progression or death (by any cause in the absence of objective progression) provided death is within 3 months from the last evaluable RECIST assessment (NCT00418886)
Timeframe: RECIST tumour assessments carried out every 6 weeks (+/- 3 days) from randomisation until objective progression

Interventionweeks (Median)
Vandetanib Plus Pemetrexed17.6
Placebo Plus Pemetrexed11.9

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from date of randomization until death. Any patient not known to have died at the time of analysis will be censored based on the last recorded date on which the patient was known to be alive (ie their status must be known at the censored date and should not be lost to follow up or unknown). (NCT00418886)
Timeframe: Time to death in months

Interventionmonths (Median)
Vandetanib Plus Pemetrexed10.5
Placebo Plus Pemetrexed9.2

[back to top]

Progression-Free Survival (PFS) in the Female Population

Median time (in weeks) from randomisation until objective disease progression or death (by any cause in the absence of objective progression) provided death is within 3 months from the last evaluable RECIST assessment (NCT00418886)
Timeframe: RECIST tumour assessments carried out every 6 weeks (+/- 3 days) from randomisation until objective progression

Interventionweeks (Median)
Vandetanib Plus Pemetrexed17.9
Placebo Plus Pemetrexed13.0

[back to top]

Longitudinal Analysis of Average Symptom Burden Index (ASBI) Score

"The longitudinal data analysis will include all non-missing visit scores and the model will include only the first 12 weeks of data. ASBI is an average of the six symptom visual analogue patient scales from none (0 mm) to as much as it could be (100 mm)." (NCT00418886)
Timeframe: ASBI is a score taken from the Lung Cancer Symptom Scale (LCSS) questionnaires administered every 3 weeks after randomisation

Interventionmms on a visual analogue scale (Least Squares Mean)
Vandetanib Plus Pemetrexed21.7
Placebo Plus Pemetrexed24.3

[back to top]

Duration of Response (DoR)

Response is defined as a confirmed best objective response of CR or PR. Duration of response is defined as time from the date of first documented response until date of documented progression or death in the absence of disease progression (provided death is within 3 months of last RECIST assessment) (NCT00418886)
Timeframe: RECIST tumour assessments carried out every 6 weeks (+/- 3 days) from randomisation until objective progression

Interventionweeks (Median)
Vandetanib Plus Pemetrexed24.1
Placebo Plus Pemetrexed24.4

[back to top]

Disease Control Rate (DCR)

Disease control rate is defined as the number of patients who achieved disease control at 6 weeks following randomisation. Disease control at 6 weeks is defined as a best objective response of complete response (CR), partial response (PR) or stable disease (SD) >= 6 weeks (NCT00418886)
Timeframe: RECIST tumour assessments carried out every 6 weeks (+/- 3 days) from randomisation until objective progression

InterventionPercentage of Participants (Number)
Vandetanib Plus Pemetrexed56.6
Placebo Plus Pemetrexed45.7

[back to top]

Longitudinal Analysis of Lung Cancer Symptom Scale (LCSS) Total Score

"The longitudinal data analysis will include all non-missing visit scores and the model will include only the first 12 weeks of data. LCSS total score is an average of all nine visual analogue patient scales from none (0 mm) to as much as it could be (100 mm)" (NCT00418886)
Timeframe: LCSS questionnaires are to be administered every 3 weeks after randomisation

Interventionmms on a visual analogue scale (Least Squares Mean)
Vandetanib Plus Pemetrexed25.7
Placebo Plus Pemetrexed28.3

[back to top]

Time to Treatment Failure

Time to treatment failure using the Kaplan-Meier method. (NCT00438204)
Timeframe: Every 8 weeks, for up to 54 months

Interventionmonths (Median)
Bevacizumab, Gemcitabine Hydrochloride6.2

[back to top]

Overall Survival

Overall survival using the Kaplan-Meier method. (NCT00438204)
Timeframe: Every 8 weeks, for up to 54 months

Interventionmonths (Median)
Bevacizumab, Gemcitabine Hydrochloride17.5

[back to top]

Progression-free Survival (PFS)

RECIST criteria for tumor progression of 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. (Note: the appearance of one or more new lesions is also considered progressions). (NCT00438204)
Timeframe: Up to 12 months

Interventionmonths (Median)
Bevacizumab, Gemcitabine Hydrochloride6.1

[back to top]

Number of Participants With Grade 3 or Grade 4 Toxicity

Grade 3/4 toxicity according to the NCI Common Toxicity Criteria v3.0 . (NCT00438204)
Timeframe: Every two weeks, for up to 54 months

InterventionParticipants (Count of Participants)
NeutropeniaLeukopeniaAnemiaThrombocytopeniaFebrile NeutropeniaElevated ALT/ASTAcute renal insufficiencyAnorexiaThrombosis/embolismDehydrationFatigueHyperglycemiaHypertensionNausea/vomitingBowel PerforationDyspneaDiverticulitisAtaxia
Bevacizumab, Gemcitabine Hydrochloride1131114123179211421

[back to top]

Number of Participants With Response

The rate of 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. (NCT00438204)
Timeframe: Every 8 weeks, for up to 54 months

InterventionParticipants (Count of Participants)
Complete responsePartial responseStable diseaseProgressive diseaseNot response evaluable
Bevacizumab, Gemcitabine Hydrochloride11512101

[back to top]

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. (NCT00447057)
Timeframe: Baseline up to 42.2 months

,
Interventionparticipants (Number)
serious adverse eventsother adverse events
Pemetrexed (Nonsquamous and Squamous)4391
Pemetrexed + Erlotinib (Nonsquamous and Squamous)5398

[back to top]

Time to Treatment Failure (TTTF)

"Defined as the time from randomization to death from any cause, first observation of PD, or study treatment discontinuation due to any reason other than protocol complete or satisfactory response. For participants who discontinued due to protocol complete or satisfactory response, or for participants not known to have discontinued as of the data cut-off date, TTTF was censored at the last contact date." (NCT00447057)
Timeframe: "Baseline to first date among death from any cause, PD, or study treatment discontinuation for any reason other than protocol complete or satisfactory response. Maximum follow-up was from Baseline to 32.2 months"

Interventionmonths (Median)
Pemetrexed (Nonsquamous)2.4
Pemetrexed + Erlotinib (Nonsquamous)3.0

[back to top]

Percentage of Participants With Best Response of Complete Response (CR) or Partial Response (PR) (Response Rate)

"CR: Disappearance of all tumor lesions; PR: Either a) at least a 30% decrease in sum of LD of target lesions or b) complete disappearance of target lesions, with persistence (but not worsening) ≥1 nontarget lesions. In either case, no new lesions may have appeared.~SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD taking as references the smallest sum LD.~PD: ≤20% increase in the sum of LD of target lesions. Response Rate (%) = (CR+PR)/number of participants in arm*100." (NCT00447057)
Timeframe: Baseline to measured progressive disease. Maximum follow-up was from Baseline to 34 months

Interventionpercentage of participants (Number)
Pemetrexed (Nonsquamous)10.8
Pemetrexed + Erlotinib (Nonsquamous)17.1

[back to top]

Percentage of Participants With Best Response of Stable Disease (SD), Partial Response (PR) or Complete Response (CR) (Disease Control Rate)

"Per RECIST:~CR: Disappearance of all target lesions; PR: Either a) ≤30% decrease in sum of longest diameter (LD) of target lesions or b) complete disappearance of target lesions, with persistence (but not worsening) of ≥1 nontarget lesions. In either case, no new lesions may have appeared.~SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD taking as references the smallest sum LD.~PD: ≤20% increase in sum of LD of target lesions. Disease Control Rate (%) = (SD+PR+CR)/number of participants in arm*100." (NCT00447057)
Timeframe: Baseline to measured PD. Maximum follow-up was from Baseline to 34 months

Interventionpercentage of participants (Number)
Pemetrexed (Nonsquamous)51.8
Pemetrexed + Erlotinib (Nonsquamous)55.3

[back to top]

Progression Free Survival (PFS)

PFS per Response Evaluation Criteria in Solid Tumors (RECIST) 1.0 criteria using computed tomography (CT) or magnetic resonance imaging (MRI) for objective determination of progressive disease (PD: ≤20% increase in sum of longest diameter of target lesion). For participants alive as of data cut-off date who did not have PD, PFS was censored at date of last CT/MRI. For participants who received subsequent systemic anticancer therapy (after study discontinuation) prior to PD or death, PFS censored at date of last CT/MRI prior to initiation of post discontinuation systemic anticancer therapy. (NCT00447057)
Timeframe: Baseline to date of measured PD or death from any cause. Maximum follow-up was from baseline to 32.2 months

Interventionmonths (Median)
Pemetrexed (Nonsquamous)2.9
Pemetrexed + Erlotinib (Nonsquamous)3.2

[back to top]

Overall Survival (OS)

OS time is the duration from randomization to the date of death from any cause. For each participant who was not known to have died as of the data inclusion cut-off date, OS was censored at the date of last contact. (NCT00447057)
Timeframe: Baseline to date of death from any cause. Maximum follow-up was from Baseline to 42.6 months.

Interventionmonths (Median)
Pemetrexed (Nonsquamous)7.8
Pemetrexed + Erlotinib (Nonsquamous)11.8

[back to top]

Percentage of Participants Surviving at 1 Year

Overall Survival (OS) rate at 1 year from the date of randomization was determined using the distribution of OS times and was estimated using the Kaplan-Meier method. (NCT00447057)
Timeframe: Baseline to date of death from any cause up to 1 year

InterventionPercentage participants with OS ≥1 year (Median)
Pemetrexed (Nonsquamous)34.1
Pemetrexed + Erlotinib (Nonsquamous)49.4

[back to top]

Time to Treatment Failure

Time to treatment failure was defined as the time from date of randomization to the date at which the patient was removed from the treatment due to progression, toxicity, refusal or other medical problems. (NCT00454194)
Timeframe: Up to 5 years

Interventionmonths (Median)
Arm I (Pemetrexed + Sorafenib)2.0
Arm II (Pemetrexed)3.1

[back to top]

Confirmed Response Rate (Complete Response and Partial Response) as Assessed by Response Evaluation Criteria in Solid Tumors (RECIST)

"A confirmed tumor response was defined as a complete response (CR) or partial response (PR) noted as the objective status on 2 consecutive evaluations at least 6 weeks apart.~Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria:~Complete Response (CR): disappearance of all target lesions;~Partial Response (PR) 30% decrease in sum of longest diameter of target lesions;~Progressive Disease (PD): 20% increase in sum of longest diameter of target lesions;~Stable Disease (SD): small changes that do not meet above criteria." (NCT00454194)
Timeframe: Up to 5 years

,
Interventionpercentage of participants (Number)
Confirmed Response (Partial Response)Stable DiseaseProgressionNot Assessed
Arm I (Pemetrexed + Sorafenib)12.838.323.425.5
Arm II (Pemetrexed)9.845.135.39.8

[back to top]

Progression-free Survival

The progression-free survival (PFS) was defined as the time from date of randomization to the documentation of disease progression or death as a result of any cause, whichever comes first. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST), 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. (NCT00454194)
Timeframe: Time from randomization to the disease progression or death (up to 5 years)

Interventionmonths (Median)
Arm I (Pemetrexed + Sorafenib)3.4
Arm II (Pemetrexed)4.1

[back to top]

Overall Survival

Overall survival was defined as the time from study enrollment (randomization) to the time of death from any cause or last follow-up. (NCT00454194)
Timeframe: Time from randomization to death or last follow-up (up to 5 years)

Interventionmonths (Median)
Arm I (Pemetrexed + Sorafenib)9.3
Arm II (Pemetrexed)10.4

[back to top]

Number of Participants With at Least One Grade 3 or Above Adverse Events Assessed by NCI CTCAE v4.0

Adverse events were assessed by Common Terminology Criteria for Adverse Events (CTCAE) v4.0. Grading: Grade 1=Mild, Grade 2=Moderate, Grade 3=Severe, Grade 4=Life-threatening, Grade 5=Death. The maximum grade for each type of adverse events were recorded for each patient. (NCT00454194)
Timeframe: Up to 3 years

Interventionparticipants (Number)
Arm I (Pemetrexed + Sorafenib)43
Arm II (Pemetrexed)27

[back to top]

Duration of Response

"Duration of response was defined as the time from the date at which the patient's earliest best objective status was first noted to be either a complete response (CR) or partial response (PR) to the earliest date progression was documented.~Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria:~Complete Response (CR): disappearance of all target lesions;~Partial Response (PR) 30% decrease in sum of longest diameter of target lesions;" (NCT00454194)
Timeframe: Up to 5 years

Interventionmonths (Median)
Arm I (Pemetrexed + Sorafenib)7.4
Arm II (Pemetrexed)8.5

[back to top]

Plasma Clearance (CL) for Gemcitabine

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 0.75, 1, 1.25, 1.5, 2, 3, 4 hr after start of infusion on Day 1 of Cycle 2 for cohort 8

InterventionL/hr (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)224.36

[back to top]

Plasma Clearance (CL) for Paclitaxel

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. (NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 3.25, 3.5, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3; 0 (pre-dose), 0.5, 1, 2, 3, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 4

InterventionL/hr (Mean)
Axitinib + Paclitaxel (Cohort 4)30.48
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)21.61

[back to top]

Plasma Clearance (CL) for Pemetrexed

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. (NCT00454649)
Timeframe: 0 (pre-dose), 10 minutes (end of infusion), 0.5, 1, 1.5, 2, 4, 6, 8 hr after end of infusion on Day 1 of Cycle 2 for cohort 9

InterventionL/hr (Mean)
Axitinib + Pemetrexed + Cisplatin (Cohort 9)7.26

[back to top]

Plasma Decay Half Life (t1/2) for Axitinib (AG-013736)

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 4, 6, 8 hr post-dose on Day -1 for cohort 1, 2, 3, 5 and 8; on Day 22 of Cycle 1 for cohort 4; on Day 18 of Cycle 1 for cohorts 6 and 7; 0 (pre-dose), 1.2, 2.2, 3.2, 4.2, 6.2, 8.2 hr post-dose on Day -1 for cohort 9

Interventionhr (Mean)
Axitinib + Paclitaxel + Carboplatin (Cohort 1)2.75
Axitinib + Paclitaxel + Carboplatin (Cohort 2)2.90
Axitinib + Paclitaxel + Carboplatin (Cohort 3)2.80
Axitinib + Paclitaxel (Cohort 4)1.45
Axitinib + Docetaxel (Cohort 5)4.07
Axitinib + Capecitabine (Cohort 6)3.85
Axitinib + Capecitabine (Cohort 7)3.64
Axitinib + Gemcitabine + Cisplatin (Cohort 8)2.68
Axitinib + Pemetrexed + Cisplatin (Cohort 9)5.02

[back to top]

Plasma Decay Half Life (t1/2) for Capecitabine

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 4, 6, 8 hr post-dose on Day 1 of Cycle 2 for cohort 6 and 7

Interventionhr (Mean)
Axitinib + Capecitabine (Cohort 6)0.85
Axitinib + Capecitabine (Cohort 7)1.44

[back to top]

Plasma Decay Half Life (t1/2) for Cisplatin

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 3, 5, 7 hr after start of infusion on Day 1 of Cycle 2 for cohort 8 and 9

Interventionhr (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)2.61
Axitinib + Pemetrexed + Cisplatin (Cohort 9)3.91

[back to top]

Plasma Decay Half Life (t1/2) for Docetaxel

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 3, 4, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 5

Interventionhr (Mean)
Axitinib + Docetaxel (Cohort 5)11.49

[back to top]

Plasma Decay Half Life (t1/2) for Carboplatin

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 5 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3

Interventionhr (Mean)
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)2.62

[back to top]

Plasma Clearance (CL) for Cisplatin

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. (NCT00454649)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 3, 5, 7 hr after start of infusion on Day 1 of Cycle 2 for cohort 8 and 9

InterventionL/hr (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)46.31
Axitinib + Pemetrexed + Cisplatin (Cohort 9)46.80

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Gemcitabine

(NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 0.75, 1, 1.25, 1.5, 2, 3, 4 hr after start of infusion on Day 1 of Cycle 2 for cohort 8

Interventionng/mL (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)20635.29

[back to top]

Plasma Decay Half Life (t1/2) for Gemcitabine

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 0.75, 1, 1.25, 1.5, 2, 3, 4 hr after start of infusion on Day 1 of Cycle 2 for cohort 8

Interventionhr (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)0.29

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Paclitaxel

(NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 3.25, 3.5, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3; 0 (pre-dose), 0.5, 1, 2, 3, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 4

Interventionng/mL (Mean)
Axitinib + Paclitaxel (Cohort 4)3698.33
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)6105.00

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Pemetrexed

(NCT00454649)
Timeframe: 0 (pre-dose), 10 minutes (end of infusion), 0.5, 1, 1.5, 2, 4, 6, 8 hr after end of infusion on Day 1 of Cycle 2 for cohort 9

Interventionng/mL (Mean)
Axitinib + Pemetrexed + Cisplatin (Cohort 9)83925.00

[back to top]

Maximum Tolerated Dose (MTD) of Axitinib (AG-013736) in Combination With Chemotherapy

MTD defined as the dose level at which more than 1 out of 6 participants experienced a dose limiting toxicity (DLT). DLT included grade (Gr) 4 neutropenia or thrombocytopenia, greater than or equal to (>=) Gr 3 nonhematological toxicities or >=0.5 teaspoon/day hemoptysis or >=2 gram /24 hours proteinuria or inability to resume background chemotherapy or axitinib (AG-013736) dosing within 14 days of stopping due to treatment related toxicity. (NCT00454649)
Timeframe: Baseline to withdrawal from study or Day 21 of Cycle 1 [all cohorts except cohort 4 (Day 28 of Cycle 1)]

Interventionmg BID (Number)
Axitinib + Paclitaxel + Carboplatin (Cohort 1)5
Axitinib + Paclitaxel + Carboplatin (Cohort 2)5
Axitinib + Paclitaxel + Carboplatin (Cohort 3)5
Axitinib + Paclitaxel (Cohort 4)5
Axitinib + Docetaxel (Cohort 5)NA
Axitinib + Capecitabine (Cohort 6)5
Axitinib + Capecitabine (Cohort 7)5
Axitinib + Gemcitabine + Cisplatin (Cohort 8)5
Axitinib + Pemetrexed + Cisplatin (Cohort 9)5

[back to top]

Percentage of Participants With Objective Response

Percentage of participants with objective response based assessment of confirmed complete response (CR) or confirmed partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.0. Confirmed response are those that persist on repeat imaging study at least 4 weeks after initial documentation of response. CR are defined as the disappearance of all lesions (target and/or non target). PR are those with at least 30% decrease in the sum of the longest dimensions of the target lesions taking as a reference the baseline sum longest dimensions. (NCT00454649)
Timeframe: Baseline and thereafter every 2 cycles up to disease progression or discontinuation from study or up to 155 weeks

InterventionPercentage of Participants (Number)
Axitinib + Paclitaxel + Carboplatin (Cohort 1)100.0
Axitinib + Paclitaxel + Carboplatin (Cohort 2)0
Axitinib + Paclitaxel + Carboplatin (Cohort 3)35.0
Axitinib + Paclitaxel (Cohort 4)66.7
Axitinib + Docetaxel (Cohort 5)50.0
Axitinib + Capecitabine (Cohort 6)11.1
Axitinib + Capecitabine (Cohort 7)11.8
Axitinib + Gemcitabine + Cisplatin (Cohort 8)23.8
Axitinib + Pemetrexed + Cisplatin (Cohort 9)0

[back to top]

Plasma Clearance (CL) for Carboplatin

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 5 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3

InterventionL/hr (Mean)
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)12.57

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Docetaxel

(NCT00454649)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 3, 4, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 5

Interventionng/mL (Mean)
Axitinib + Docetaxel (Cohort 5)3130.00

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Cisplatin

(NCT00454649)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 3, 5, 7 hr after start of infusion on Day 1 of Cycle 2 for cohort 8 and 9

Interventionng/mL (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)1680.54
Axitinib + Pemetrexed + Cisplatin (Cohort 9)1176.00

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Carboplatin

(NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 5 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3

Interventionng/mL (Mean)
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)23383.33

[back to top]

Plasma Decay Half Life (t1/2) for Pemetrexed

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 10 minutes (end of infusion), 0.5, 1, 1.5, 2, 4, 6, 8 hr after end of infusion on Day 1 of Cycle 2 for cohort 9

Interventionhr (Mean)
Axitinib + Pemetrexed + Cisplatin (Cohort 9)2.77

[back to top]

Plasma Decay Half Life (t1/2) for Paclitaxel

t1/2 is the time measured for the plasma concentration to decrease by one half. (NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 3.25, 3.5, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3; 0 (pre-dose), 0.5, 1, 2, 3, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 4

Interventionhr (Mean)
Axitinib + Paclitaxel (Cohort 4)12.51
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)8.36

[back to top]

Plasma Clearance (CL) for Docetaxel

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the plasma. (NCT00454649)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 3, 4, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 5

InterventionL/hr (Mean)
Axitinib + Docetaxel (Cohort 5)42.96

[back to top]

Apparent Oral Clearance (CL/F) for Axitinib (AG-013736)

Clearance (CL) of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes and F is the absolute oral bioavailability. Apparent oral clearance(CL/F) is obtained following oral administration. (NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 4, 6, 8 hr post-dose on Day -1 for cohort 1, 2, 3, 5 and 8; on Day 22 of Cycle 1 for cohort 4; on Day 18 of Cycle 1 for cohorts 6 and 7; 0 (pre-dose), 1.2, 2.2, 3.2, 4.2, 6.2, 8.2 hr post-dose on Day -1 for cohort 9

InterventionLiter/hour (L/hr) (Mean)
Axitinib + Paclitaxel + Carboplatin (Cohort 1)49.39
Axitinib + Paclitaxel + Carboplatin (Cohort 2)40.72
Axitinib + Paclitaxel + Carboplatin (Cohort 3)29.73
Axitinib + Paclitaxel (Cohort 4)65.69
Axitinib + Docetaxel (Cohort 5)14.35
Axitinib + Capecitabine (Cohort 6)26.64
Axitinib + Capecitabine (Cohort 7)83.46
Axitinib + Gemcitabine + Cisplatin (Cohort 8)50.05
Axitinib + Pemetrexed + Cisplatin (Cohort 9)25.10

[back to top]

Apparent Oral Clearance (CL/F) for Capecitabine

Clearance (CL) of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes and F is the absolute oral bioavailability. Apparent oral clearance(CL/F) is obtained following oral administration. (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 4, 6, 8 hr post-dose on Day 1 of Cycle 2 for cohort 6 and 7

InterventionLiter/hr (Mean)
Axitinib + Capecitabine (Cohort 6)209.05
Axitinib + Capecitabine (Cohort 7)314.12

[back to top]

Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Carboplatin

AUC (0-∞) = Area under the plasma concentration versus time curve (AUC) from time zero (pre-dose) to extrapolated infinite time (0-∞). It is obtained from AUC (0-t) plus AUC (t-∞). (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 5 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3

Interventionng*hr/mL (Mean)
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)55580.26

[back to top]

Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Docetaxel

AUC (0-∞) = Area under the plasma concentration versus time curve (AUC) from time zero (pre-dose) to extrapolated infinite time (0-∞). It is obtained from AUC (0-t) plus AUC (t-∞). (NCT00454649)
Timeframe: 0 (pre-dose), 0.5, 1, 1.5, 2, 3, 4, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 5

Interventionng*hr/mL (Mean)
Axitinib + Docetaxel (Cohort 5)3478.49

[back to top]

Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Gemcitabine

AUC (0-∞) = Area under the plasma concentration versus time curve (AUC) from time zero (pre-dose) to extrapolated infinite time (0-∞). It is obtained from AUC (0-t) plus AUC (t-∞). (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 0.75, 1, 1.25, 1.5, 2, 3, 4 hr after start of infusion on Day 1 of Cycle 2 for cohort 8

Interventionng*hr/mL (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)10991.16

[back to top]

Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Paclitaxel

AUC (0-∞) = Area under the plasma concentration versus time curve (AUC) from time zero (pre-dose) to extrapolated infinite time (0-∞). It is obtained from AUC (0-t) plus AUC (t-∞). (NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 3.25, 3.5, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 1-3; 0 (pre-dose), 0.5, 1, 2, 3, 4, 5, 6, 8, 24, 30 hr after start of infusion on Day 1 of Cycle 2 for cohort 4

Interventionng*hr/mL (Mean)
Axitinib + Paclitaxel (Cohort 4)5683.55
Axitinib + Paclitaxel + Carboplatin (Combined Cohort 1, 2, 3)19959.91

[back to top]

Area Under the Curve From Time Zero to Extrapolated Infinite Time [AUC (0-∞)] for Pemetrexed

AUC (0-∞) = Area under the plasma concentration versus time curve (AUC) from time zero (pre-dose) to extrapolated infinite time (0-∞). It is obtained from AUC (0-t) plus AUC (t-∞). (NCT00454649)
Timeframe: 0 (pre-dose), 10 minutes (end of infusion), 0.5, 1, 1.5, 2, 4, 6, 8 hr after end of infusion on Day 1 of Cycle 2 for cohort 9

Interventionng*hr/mL (Mean)
Axitinib + Pemetrexed + Cisplatin (Cohort 9)133032.97

[back to top]

Area Under the Curve From Time Zero to Time 24 Hours [AUC (0-24)] for Axitinib (AG-013736)

AUC (0-24) = Area under the plasma concentration versus time curve from time zero (pre-dose) to time 24 hours (0-24). (NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 4, 6, 8 hr post-dose on Day -1 for cohort 1, 2, 3, 5 and 8; on Day 22 of Cycle 1 for cohort 4; on Day 18 of Cycle 1 for cohorts 6 and 7; 0 (pre-dose), 1.2, 2.2, 3.2, 4.2, 6.2, 8.2 hr post-dose on Day -1 for cohort 9

Interventionnanogram*hour/milliliter (ng*hr/mL) (Mean)
Axitinib + Paclitaxel + Carboplatin (Cohort 1)61.58
Axitinib + Paclitaxel + Carboplatin (Cohort 2)242.41
Axitinib + Paclitaxel + Carboplatin (Cohort 3)475.18
Axitinib + Paclitaxel (Cohort 4)154.43
Axitinib + Docetaxel (Cohort 5)780.99
Axitinib + Capecitabine (Cohort 6)365.95
Axitinib + Capecitabine (Cohort 7)449.99
Axitinib + Gemcitabine + Cisplatin (Cohort 8)416.30
Axitinib + Pemetrexed + Cisplatin (Cohort 9)420.64

[back to top]

Area Under the Curve From Time Zero to Time 24 Hours [AUC (0-24)] for Capecitabine

AUC (0-24) = Area under the plasma concentration versus time curve from time zero (pre-dose) to time 24 hours (0-24). (NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 4, 6, 8 hr post-dose on Day 1 of Cycle 2 for cohort 6 and 7

Interventionng*hr/mL (Mean)
Axitinib + Capecitabine (Cohort 6)20534.52
Axitinib + Capecitabine (Cohort 7)22163.88

[back to top]

Area Under the Curve From Time Zero to Time 8 Hours [AUC (0-8)] for Cisplatin

AUC (0-8) = Area under the plasma concentration versus time curve from time zero (pre-dose) to time 8 hours (0-8). (NCT00454649)
Timeframe: Pre-dose, 0.5, 1, 1.5, 2, 3, 5, 7 hr after start of infusion on Day 1 of Cycle 2 for cohort 8 and 9

Interventionng*hr/mL (Mean)
Axitinib + Gemcitabine + Cisplatin (Cohort 8)2932.43
Axitinib + Pemetrexed + Cisplatin (Cohort 9)2703.92

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Axitinib (AG-013736)

(NCT00454649)
Timeframe: 0 (pre-dose), 1, 2, 3, 4, 6, 8 hr post-dose on Day -1 for cohort 1, 2, 3, 5 and 8; on Day 22 of Cycle 1 for cohort 4; on Day 18 of Cycle 1 for cohorts 6 and 7; 0 (pre-dose), 1.2, 2.2, 3.2, 4.2, 6.2, 8.2 hr post-dose on Day -1 for cohort 9

Interventionng/mL (Mean)
Axitinib + Paclitaxel + Carboplatin (Cohort 1)5.97
Axitinib + Paclitaxel + Carboplatin (Cohort 2)23.36
Axitinib + Paclitaxel + Carboplatin (Cohort 3)42.58
Axitinib + Paclitaxel (Cohort 4)44.58
Axitinib + Docetaxel (Cohort 5)67.96
Axitinib + Capecitabine (Cohort 6)37.51
Axitinib + Capecitabine (Cohort 7)43.97
Axitinib + Gemcitabine + Cisplatin (Cohort 8)40.97
Axitinib + Pemetrexed + Cisplatin (Cohort 9)31.53

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Capecitabine

(NCT00454649)
Timeframe: 0 (pre-dose), 0.25, 0.5, 1, 2, 3, 4, 6, 8 hr post-dose on Day 1 of Cycle 2 for cohort 6 and 7

Interventionng/mL (Mean)
Axitinib + Capecitabine (Cohort 6)10808.00
Axitinib + Capecitabine (Cohort 7)10588.38

[back to top]

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

OS is defined as the time from the date of study entry until the date of death due to any cause. In the absence of confirmation of death or lack of data beyond follow-up period, the survival time was censored to the last date the participant was known to be alive. (NCT00456261)
Timeframe: From date of study entry until the date of death from any cause or the date the patient was last known alive, up to 18 months

Interventionmonths (Number)
Bevacizumab/Pemetrexed/Gemcitabine7.5
Bevacizumab/Pemetrexed/Carboplatin14.8

[back to top]

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

Time to Progression (TTP) is defined as the interval between the date of treatment initiation and the date of progressive disease. Progression is defined using the Response Evaluation Criteria in Solid Tumors (RECIST v1.0). Progressive Disease (PD): At least a 20% increase in the sum of the longest diameter (LD) of target lesions, taking as reference the smallest sum LD recorded since the treatment started or unequivocal progression of non-target lesions or the appearance of one or more new lesions. (NCT00456261)
Timeframe: From the date of treatment initiation until the date of first documented PD or date of last study contact or date of other therapy begins up to 18 months

Interventionmonths (Median)
Bevacizumab/Pemetrexed/Gemcitabine4.7
Bevacizumab/Pemetrexed/Carboplatin10.2

[back to top]

Overall Response Rate (ORR), the Percentage of Patients Who Experience an Objective Benefit From Treatment

Overall response rate (ORR) is defined as the percentage of patients who have a partial or complete response to therapy. Responses were assessed by the Response Evaluation Criteria in Solid Tumors (RECIST; version 1.0). Complete Response: Disappearance of all target lesions, and disappearance of all non-target lesions. Partial Response: At least a 30% decrease in the sum of the longest diameter of target lesions (taking as reference the baseline sum of longest diameters) (NCT00456261)
Timeframe: From date of treatment initiation to end of study treatment up to 18 months

Interventionpercentage of patients (Number)
Bevacizumab/Pemetrexed/Gemcitabine35
Bevacizumab/Pemetrexed/Carboplatin35

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. (NCT00461786)
Timeframe: time of initial response until documented tumor progression (up to 44 months)

Interventionmonths (Median)
Pemetrexed8.4

[back to top]

Tumor Response

"Best response recorded from the start of treatment until disease progression/recurrence using Response Evaluation Criteria In Solid Tumors (RECIST) criteria that defines when participants improve (respond), stay the same (stable), or worsen (progression) during treatment. Complete response (CR) = disappearance of all target lesions; Partial response (PR) = 30% decrease in the sum of the longest diameter of target lesions; Progressive disease (PD) = 20% increase in the sum of the longest diameter of target lesions; Stable disease (SD) = small changes that do not meet above criteria." (NCT00461786)
Timeframe: baseline to measured progressive disease (up to 44 months)

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseInevaluable
Pemetrexed1917183

[back to top]

Number of Participants With Adverse Events by Grade

Adverse events were graded using the Common Terminology Criteria for Adverse Events version 3.0 (CTCAE v3.0) for defining and grading specific adverse events. A grading (severity) scale is provided for each adverse event term. Grades range from 0 (none) to 5 (death). The worst grade event per cycle is reported. (NCT00461786)
Timeframe: every 21-day cycle up to 5 year follow-up

Interventionparticipants (Number)
Leukopenia - Grade 1Leukopenia - Grade 2Leukopenia - Grade 3Leukopenia - Grade 4Thrombocytopenia - Grade 1Thrombocytopenia - Grade 2Thrombocytopenia - Grade 3Thrombocytopenia - Grade 4Neutropenia - Grade 1Neutropenia - Grade 2Neutropenia - Grade 3Neutropenia - Grade 4Anemia - Grade 1Anemia - Grade 2Anemia - Grade 3Anemia - Grade 4Transfusion - Grade 1Transfusion - Grade 2Transfusion - Grade 3Transfusion - Grade 4Coagulation - Grade 1Coagulation - Grade 2Coagulation - Grade 3Coagulation - Grade 4Gastrointestinal - Grade 1Gastrointestinal - Grade 2Gastrointestinal - Grade 3Gastrointestinal - Grade 4Nausea/vomiting - Grade 1Nausea/vomiting - Grade 2Nausea/vomiting - Grade 3Nausea/vomiting - Grade 4Genitourinary - Grade 1Genitourinary - Grade 2Genitourinary - Grade 3Genitourinary - Grade 4Hepatic - Grade 1Hepatic - Grade 2Hepatic - Grade 3Hepatic - Grade 4Alopecia - Grade 1Alopecia - Grade 2Dermatologic - Grade 1Dermatologic - Grade 2Dermatologic - Grade 3Dermatologic - Grade 4Neurologic - Grade 1Neurologic - Grade 2Neurologic - Grade 3Neurologic - Grade 4SGOT - Grade 1SGOT - Grade 2SGOT - Grade 3SGOT - Grade 4Alkaline phosphatase - Grade 1Alkaline phosphatase - Grade 2Alkaline phosphatase - Grade 3Alkaline phosphatase - Grade 4Ocular - Grade 1Ocular - Grade 2Ocular - Grade 3Ocular - Grade 4Hemorrhage - Grade 1Hemorrhage - Grade 2Hemorrhage - Grade 3Hemorrhage - Grade 4Pulmonary - Grade 1Pulmonary - Grade 2Pulmonary - Grade 3Pulmonary - Grade 4Lymphopenia - Grade 1Lymphopenia - Grade 2Lymphopenia - Grade 3Lymphopenia - Grade 4Constitutional - Grade 1Constitutional - Grade 2Constitutional - Grade 3Constitutional - Grade 4Metabolic - Grade 1Metabolic - Grade 2Metabolic - Grade 3Metabolic - Grade 4Cardiovascular - Grade 1Cardiovascular - Grade 2Cardiovascular - Grade 3Cardiovascular - Grade 4Pain - Grade 1Pain - Grade 2Pain - Grade 3Pain - Grade 4Infection - Grade 1Infection - Grade 2Infection - Grade 3Infection - Grade 4Auditory - Grade 1Auditory - Grade 2Auditory - Grade 3Auditory - Grade 4Lymphatics - Grade 1Lymphatics - Grade 2Lymphatics - Grade 3Lymphatics - Grade 4Endocrine - Grade 1Endocrine - Grade 2Endocrine - Grade 3Endocrine - Grade 4Allergy - Grade 1Allergy - Grade 2Allergy - Grade 3Allergy - Grade 4
Pemetrexed111311115524961191417700100010014144015740402022107510103012141940033002310201041001000171452209403000861125500210410120000100

[back to top]

Progression-Free Survival

Defined as the time from date of first dose to the first observation of disease progression, or death due to any cause. (NCT00461786)
Timeframe: baseline until documented tumor progression (up to 44 months)

Interventionmonths (Median)
Pemetrexed3.0

[back to top]

Overall Survival

Overall survival is the duration from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00461786)
Timeframe: baseline until death from any cause up to 5-year follow-up

Interventionmonths (Median)
Pemetrexed11.4

[back to top]

Duration of Overall Survival

From time of enrollment to the first observation of disease progression or death. (NCT00470548)
Timeframe: Up to 2 years

Interventionmonths (Median)
Phase I: Abraxane and Pemetrexed13.5
Phase II: Abraxane and Pemetrexed4.5

[back to top]

Number of Participants With Complete Response

Per RECIST criteria, complete response (CR) is defined as the disappearance of all target lesions. (NCT00470548)
Timeframe: Up to 2 years

Interventionparticipants (Number)
Phase I: Abraxane and Pemetrexed0
Phase II: Abraxane and Pemetrexed0

[back to top]

Number of Participants With Disease Control

Disease control is complete response plus partial response plus stable disease from the start of treatment to death or disease progression. (NCT00470548)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Phase I: Abraxane and Pemetrexed7
Phase II: Abraxane and Pemetrexed17

[back to top]

Number of Participants With Dose Limiting Toxicities

Dose limiting toxicity (DLT) was defined as any of the following occurring during the first cycle: Grade 4 thrombocytopenia, or grade 3 thrombocytopenia associated with bleeding, requirement for transfusion, febrile neutropenia, neutropenia with documented infection. Non-hematologic DLT included any other ≥ grade 3 non-hematologic toxicity that was clinically significant and considered by the investigator to be related to study drug. Alopecia and grade 3 allergic reaction/hypersensitivity with infusion were not considered DLTs. (NCT00470548)
Timeframe: Up to21 days

Interventionparticipants (Number)
Phase I: Dose Level 10
Phase I: Dose Level 20
Phase I: Dose Level 30

[back to top]

Number of Participants With Stable Disease

Stable Disease is measured from the start of the treatment until the criteria for disease progression are met. 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 (NCT00470548)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Phase I: Abraxane and Pemetrexed7
Phase II: Abraxane and Pemetrexed12

[back to top]

Number of Patients With Toxicities

Toxicities was evaluated based on the standard NCI CTCAE Version 3.0 grading criteria. Attributable grade ≥ 3 hematologic and non-hematologic toxicities are presented here. (NCT00470548)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Phase II: Pemetrexed and Abraxane15

[back to top]

Number of Participants With Partial Response

At least a 30% decrease in the sum of the longest diameter of target lesions (NCT00470548)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Phase I: Abraxane and Pemetrexed0
Phase II: Abraxane and Pemetrexed5

[back to top]

Phase 1 - Percentage of Participants With Complete Response or Partial Response (Response Rate)

Response rate is the percentage of participants with complete response (CR) or partial response (PR), as assessed according to the Response Evaluation Criteria In Solid Tumors (RECIST) guidelines. CR is disappearance of all target and non-target lesions; PR is ≥30% decrease in sum of longest diameter of target lesions. Response rate is calculated as a total number of participants with CR or PR divided by the total number of participants treated multiplied by 100. (NCT00482014)
Timeframe: Phase 1 enrollment to the end of the study treatment up to Week 11

,
Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Pemetrexed + Carboplatin Treatment Group011.1
Pemetrexed + Cisplatin Treatment045.5

[back to top]

Phase 1 - Pharmacology Toxicity: Number of Participants With Dose Limiting Toxicities (DLTs)

Phase 1 pharmacology toxicity was defined as the number of participants experiencing dose limiting toxicities (DLTs). DLT was defined as any of the following events occurring during the entire radiation therapy (RT) course: Grade 4 neutropenia (<0.5 x 10^9 cells per liter) >7 days, febrile neutropenia, ≥Grade 3 neutropenia with fever >38.5 degrees Celsius (°C), Grade 4 thrombocytopenia, Grade 3 thrombocytopenia with ≥Grade 2 bleeding, ≥Grade 3 nonhematologic toxicity (excluding nausea, vomiting, and transaminase elevations), and ≥Grade 3 pulmonary or esophageal toxicity (radiation-related pneumonitis or esophagitis). Grade 5 events are the events leading to the death. (NCT00482014)
Timeframe: Phase 1 enrollment up to Week 11

,
Interventionparticipants (Number)
Grade 5 Pneumocystis jiroveci pneumoniaGrade 5 Hemoptysis (pemetrexed+Cisplatin30 mg/m² )
Pemetrexed + Carboplatin Treatment Group10
Pemetrexed + Cisplatin Treatment01

[back to top]

Phase 1 - Maximum Tolerated Dose (MTD) of Cisplatin

MTD was defined as a dose at which the occurrence of at least 2 dose-limiting toxicities (DLTs) was observed. DLT was defined as any of the following events occurring during the entire radiation therapy (RT) course, including a 2-week recovery period following completion of RT: Grade 4 neutropenia (<0.5 x 10^9 cells per liter) lasting >7 days, febrile neutropenia; ≥Grade 3 neutropenia with fever >38.5 degrees Celsius (°C), Grade 4 thrombocytopenia, Grade 3 thrombocytopenia with ≥Grade 2 bleeding, ≥Grade 3 nonhematologic toxicity (excluding nausea, vomiting, and transaminase elevations) and ≥Grade 3 pulmonary or esophageal toxicity (radiation-related pneumonitis or esophagitis). (NCT00482014)
Timeframe: Phase 1 enrollment to the end of study treatment up to Week 11

Interventionmilligrams/meter squared (mg/m²) (Number)
Pemetrexed + Cisplatin Treatment GroupNA

[back to top]

Phase 2 - Median Survival

(NCT00482014)
Timeframe: Phase 2 randomization to death as the result of any cause up to 30.0 month

Interventionmonths (Median)
Pemetrexed + Carboplatin Treatment Group18.7
Pemetrexed + Cisplatin Treatment Group27.0

[back to top]

Phase 2 - Survival Probability at 2 Years

(NCT00482014)
Timeframe: Phase 2 randomization up to 2 years

Interventionpercentage survival (Mean)
Pemetrexed + Carboplatin Treatment Group45.4
Pemetrexed + Cisplatin Treatment Group58.4

[back to top]

Phase 2 - Percentage of Participants With Complete Response or Partial Response (Response Rate)

Response rate is the percentage of participants with complete response (CR) or partial response (PR), as assessed according to the Response Evaluation Criteria In Solid Tumors (RECIST) guidelines. CR is disappearance of all target and non-target lesions; PR is ≥30% decrease in sum of longest diameter of target lesions. Response rate is calculated as a total number of participants with CR or PR divided by the total number of participants treated multiplied by 100. (NCT00482014)
Timeframe: Phase 2 randomization to the end of the treatment up to 30.0 months

,
Interventionpercentage of participants (Number)
Complete Response (CR)Partial Response (PR)
Pemetrexed + Carboplatin Treatment Group6.545.7
Pemetrexed + Cisplatin Treatment Group3.842.3

[back to top]

Phase 2 - Pharmacology Toxicity: Number of Participants With Adverse Events

Phase 2 pharmacology toxicity was defined as the number of participants who experienced serious adverse events or all other nonserious adverse events during the study. A summary of serious adverse events and other nonserious adverse events is located in the Reported Adverse Events section. (NCT00482014)
Timeframe: Phase 2 randomization to the end of the study treatment up to 30.0 months

,
Interventionparticipants (Number)
Serious Adverse EventsOther Nonserious Adverse Events
Pemetrexed + Carboplatin Treatment Group2144
Pemetrexed + Cisplatin Treatment Group1951

[back to top]

Phase 1 - Maximum Tolerated Dose (MTD) of Carboplatin

MTD was defined as a dose at which the occurrence of at least 2 dose-limiting toxicities (DLTs) was observed. DLT was defined as any of the following events occurring during the entire radiation therapy (RT) course, including a 2-week recovery period following completion of RT: Grade 4 neutropenia (<0.5 x 10^9 cells per liter) lasting >7 days, febrile neutropenia; ≥Grade 3 neutropenia with fever >38.5 degrees Celsius (°C), Grade 4 thrombocytopenia, Grade 3 thrombocytopenia with ≥Grade 2 bleeding, ≥Grade 3 nonhematologic toxicity (excluding nausea, vomiting, and transaminase elevations) and ≥Grade 3 pulmonary or esophageal toxicity (radiation-related pneumonitis or esophagitis). (NCT00482014)
Timeframe: Phase 1 enrollment to the end of study treatment up to Week 11

Interventionmilligram/milliliter*minute (mg/mL*min) (Number)
Pemetrexed + Carboplatin Treatment GroupNA

[back to top]

Phase 2 - Time to Progression

Time to disease progression was measured from randomization of Study Phase 2 to the first observation of disease progression according to the Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Disease progression is ≥20% increase in sum of longest diameter of target lesions and/or a new lesion. (NCT00482014)
Timeframe: Phase 2 randomization to measured disease progression up to 24 months

Interventionmonths (Median)
Pemetrexed + Carboplatin Treatment Group8.8
Pemetrexed + Cisplatin Treatment Group13.1

[back to top]

To Evaluate the Overall Response Rate (Complete Plus Partial Responses by RECIST Criteria) to the Combination of Paclitaxel Poliglumex and Pemetrexed as Therapy in Patients With Advanced NSCLC.

(NCT00487669)
Timeframe: CT or MRI scans of the chest will be obtained after every 2 cycles (6-week intervals +/- 7 days)

,
Interventionparticipants (Number)
Stable DiseaseProgressive Disease
Level 151
Level 242

[back to top]

Time to Progression

(NCT00487669)
Timeframe: time from study entry until the first documented sign of progression

Interventionmonths (Median)
Level 13.8
Level 22.8

[back to top]

Overall Survival

(NCT00487669)
Timeframe: time from study entry until death

Interventionmonths (Median)
Level 17.7
Level 28.5

[back to top]

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

A listing of adverse events is located in the Reported Adverse Event module. (NCT00489359)
Timeframe: baseline measured to progressive disease (up to 18 months)

InterventionParticipants (Number)
Serious Adverse EventsOther Adverse Events
Pemetrexed/Carboplatin Phase 1219

[back to top]

Phase 1 - Number of Participants With Tumor Response

Patients were analyzed by Cancer Antigen-125 (CA-125) response criteria and RECIST guidelines. Possible evaluations include: Complete Response (CR): Disappearance of all target lesions. Partial Response (PR): At least a 30% decrease in the size of target lesions. Progressive Disease (PD): At least a 20% increase in the size of target lesions. Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT00489359)
Timeframe: baseline measured to progressive disease (up to 18 months)

InterventionParticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)
Pemetrexed/Carboplatin Phase 112412

[back to top]

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

A listing of adverse events is located in the Reported Adverse Event module. (NCT00489359)
Timeframe: baseline through end of Phase 2 (up to 31 months)

Interventionparticipants (Number)
Serious Adverse EventsOther Adverse Events
Pemetrexed/Carboplatin Phase 21563

[back to top]

Phase 2 - Percentage of Participants With Overall Tumor Response (Response Rate)

"Response is defined as CR (Complete Response) or PR (Partial Response) per Response Evaluation Criteria in Solid Tumor (RECIST criteria). Possible evaluations include: CR: Disappearance of all target lesions. PR: At least a 30% decrease in the size of target lesions.~Response rate (%) = (number of patients with CR+PR/number of patients in Phase 2)*100" (NCT00489359)
Timeframe: baseline to measured progressive disease (PD) (up to 18 months)

Interventionpercentage of participants (Number)
Pemetrexed/Carboplatin Phase 232.8

[back to top]

Phase 2 - Progression-Free Survival

Progression-free survival (PFS) is defined as the time from the date of study enrollment to the date of objectively determined PD or death from any cause, whichever comes first. For patients who are still alive at the time of analysis, and who do not have PD, PFS will be censored at the date of the last objective progression-free disease assessment. (NCT00489359)
Timeframe: baseline to measured progressive disease (up to 31 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin Phase 29.4

[back to top]

Phase 2 - Time to Disease Progression

Time to objective progressive disease (TTPD) is defined as the time from the date of study enrollment to the date of objectively determined Progressive Disease (PD). For patients who die without objective PD (including death from study disease), TTPD will be censored at the date of the last objective progression-free disease assessment. For patients who are still alive at the time of analysis, and who do not have PD, TTPD will be censored at the date of the last objective progression-free disease assessment. (NCT00489359)
Timeframe: baseline to measured progressive disease (up to 31 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin Phase 29.5

[back to top]

Phase 2 - Time to Response (TTR)

Response is defined as CR (Complete Response) or PR (Partial Response) per RECIST criteria. Possible evaluations include: Complete Response (CR): Disappearance of all target lesions. Partial Response (PR): At least a 30% decrease in the size of target lesions. Progressive Disease (PD): At least a 20% increase in the size of target lesions. Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT00489359)
Timeframe: First treatment to response (up to 31 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin Phase 21.8

[back to top]

Phase 1 - Number of Dose-Limiting Toxicities (DLTs)

"The following toxicities were considered DLT: CTCAE Grade 4 neutropenia (absolute neutrophil count [ANC] <0.5 × 10^9/L lasting ≥7 days. Febrile neutropenia (ANC <1.0 × 10^9/L, fever 38.5°C, and no documented infection). CTCAE Grade 4 thrombocytopenia (platelets <25.0 × 10^9/L).~Any hemorrhage with CTCAE Grade ≥3 thrombocytopenia (50.0 × 10^9/L). CTCAE Grade ≥3 nonhematologic toxicity (excluding nausea, vomiting, or CTCAE Grade 3 alanine transaminase (ALT) or aspartate aminotransferase (AST) that returned to baseline prior to next treatment).~Treatment delay more than 1 week due to toxicity." (NCT00489359)
Timeframe: baseline through end of Phase 1 (up to 18 months)

InterventionDLT events (Number)
Number of DLT EventsDLT Event: Grade 4 Neutropenia
Pemetrexed/Carboplatin Phase 111

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

Phase 2 - Time to Treatment Failure

Time to treatment failure (TTTF) is defined as the time from the date of study enrollment to the date of the first observation of disease progression, death from any cause, or early discontinuation of treatment (any reason). For patients who are alive, progression-free, and have not discontinued early at the time of analysis, TTTF will be censored at the date of the last objective progression-free disease assessment. (NCT00489359)
Timeframe: First treatment to discontinuation of study drug, progressive disease, or death (up to 31 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin Phase 27.1

[back to top]

Phase 2 - Duration of Response (DOR)

Duration of response is defined as the time from first observation of Complete Response or Partial Response to the first observation of Progressive Disease or death from any cause. For patients who are still alive at the time of analysis, and who do not have Progressive Disease, duration of response will be censored at the date of the last objective progression-free disease assessment. (NCT00489359)
Timeframe: time of response to progressive disease (up to 31 months)

InterventionMonths (Median)
Pemetrexed/Carboplatin Phase 29.1

[back to top]

Overall Response

Number of participants with complete or partial response. Response Evaluation Criteria in Solid Tumors (RECIST) of Complete Response: disappearance all target lesions; Partial Response: >30% decrease in sum of longest diameter (LD) of target lesions, reference baseline sum LD; Progressive Disease: >20% increase sum of LD of target lesions, reference smallest sum LD recorded since treatment started or appearance of 1 or > new lesions; Stable Disease: Insufficient shrinkage for partial response, or insufficient increase for progressive disease, reference smallest sum LD since treatment started. (NCT00491075)
Timeframe: Baseline to 8 weeks (after 4 cycles) protocol response at 16 weeks

Interventionpercentage of participants (Number)
Pemetrexed + Gemcitabine6.7

[back to top]

Pharmacology Toxicity

Radiation Therapy Oncology Group (RTOG) criteria were used for assessing toxicity. Toxicity grade reflected the most severe degree occurring during the evaluated period, not an average. When two criteria were available for similar toxicities, the one resulting in the more severe grade was used. Toxiccity grades range from 0 to 5. Toxicity grade = 5 if that toxicity caused the death of the patient. (NCT00494026)
Timeframe: every 21-day cycle for 4 cycles

Interventionparticipants (Number)
Cycle 3: Grade 2 Hematologic White Blood CellCycle 3: Grade 1 PlateletsCycle 3: Grade 3 NeutrophilsCycle 3: Grade 2 HemoglobinCycle 3: Grade 2 HematocritCycle 4: Grade 3 Hematologic White Blood CellCycle 4: Grade 3 PlateletsCycle 4: Grade 2 NeutrophilsCycle 4: Grade 3 HemoglobinCycle 4: Grade 3 Hematocrit
Pemetrexed + Carboplatin1111111111

[back to top]

Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Drive or Use Public Transportation

The daily activities assessed: ability to drive or use public transportation. OARS-IADL assesses the participant's ability to drive or use public transportation at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American63.132.30.04.629.220.00.050.8
Asian American62.229.72.75.445.90.00.054.1
Caucasian62.229.60.77.623.414.81.060.9
Hispanic50.050.00.00.021.47.10.071.4

[back to top]

Percentage of Participants With a Best Overall Disease Control Response (Disease Control Rate)

Disease Control Rate [DCR] is the percentage of participants with Complete Response (CR), Partial Response (PR), Stable Disease (SD), and SD or Incomplete Response (SI). Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. CR=disappearance of all target lesions; PR=30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD)=20% increase in sum of longest diameter of target lesions; SD=small changes not meeting above criteria; SI=persistence of 1 or more non-target lesion(s) and/or maintenance of tumor marker level above normal limits. (NCT00497770)
Timeframe: baseline to measured progressive disease (up to 20 cycles [14 months])

Interventionpercentage of participants (Number)
Caucasian43.7
African American45.0
Asian American53.33
Hispanic34.6

[back to top]

Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Prepare and Take Medications

The daily activities assessed: ability to prepare and take medications. OARS-IADL assesses the participant's ability to prepare and take medications at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American73.821.50.04.636.910.81.54.6
Asian American86.55.42.75.445.90.00.05.4
Caucasian79.312.80.77.230.97.60.77.2
Hispanic85.77.17.10.025.00.03.60.0

[back to top]

Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Handle Personal Finances

The daily activities assessed: ability to handle personal finances. OARS-IADL assesses the participant's ability to handle personal finances at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American67.726.20.06.238.510.80.050.8
Asian American83.88.12.75.445.90.00.054.1
Caucasian78.611.52.37.631.95.61.361.2
Hispanic71.417.910.70.025.03.60.071.4

[back to top]

Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Operate the Telephone

The daily activities assessed: ability to operate the telephone. OARS-IADL assesses the participant's ability to operate the telephone at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American90.84.60.04.641.57.70.050.8
Asian American86.55.42.75.445.90.00.054.1
Caucasian86.55.60.07.934.93.01.360.9
Hispanic92.93.60.03.625.03.60.071.4

[back to top]

Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to do the Act of Shopping

The daily activities assessed: ability to do the act of shopping. OARS-IADL assesses the participant's ability to do the act of shopping at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American58.532.34.64.624.621.53.150.8
Asian American59.529.75.45.440.55.40.054.1
Caucasian57.229.95.67.220.114.14.661.2
Hispanic50.035.714.30.014.314.30.071.4

[back to top]

Progression Free Survival

Time from start of second line therapy until death, disease progression, or last contact expressed in months. Participants lost to follow-up (that were alive at last contact) were treated as censored using Kaplan-Meier survival analysis method. (NCT00497770)
Timeframe: baseline to measured progressive disease or death (up to 20 cycles [14 months])

Interventionmonths (Median)
Caucasian2.66
African American2.99
Asian American5.08
Hispanic2.20

[back to top]

Overall Survival

Overall survival is the duration (months) from enrollment to death. For patients who are alive, overall survival is censored at the last contact. (NCT00497770)
Timeframe: baseline to date of death from any cause (up to 20 cycles [14 months])

Interventionmonths (Median)
Caucasian6.77
African American6.90
Asian American16.0
Hispanic5.35

[back to top]

Functional Status Based on Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Plan and Prepare Meals

The daily activities assessed: ability to plan and prepare meals. OARS-IADL assesses the participant's ability to plan and prepare meals at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American63.123.19.24.626.213.89.250.8
Asian American70.313.510.85.437.88.10.054.1
Caucasian63.224.34.97.625.39.54.360.9
Hispanic71.410.717.90.014.310.73.671.4

[back to top]

Functional Status Based on the Older Americans Resources and Services Instrumental Activities of Daily Living Scale(OARS-IADL), Ability to Perform Housework Activities

The daily activities assessed: ability to perform housework activities. OARS-IADL assesses the participant's ability to perform housework activities at baseline and end of treatment. Responses: ability to do the activity without help, some help, unable, or not done (missing). The percent of participants responding in the different levels of functional ability are provided. (NCT00497770)
Timeframe: beginning and at end of pemetrexed treatment (up to 20 cycles [14 months])

,,,
Interventionpercentage of participants (Number)
Without Help-baselineSome Help-baselineUnable-baselineNot Done (missing)-baselineWithout Help-end of treatmentSome Help-end of treatmentUnable-end of treatmentNot Done (missing)-end of treatment
African American44.633.816.94.621.518.59.250.8
Asian American54.132.48.15.427.016.22.754.1
Caucasian44.138.29.58.215.815.57.261.5
Hispanic42.932.125.00.010.714.33.671.4

[back to top]

Symptom Score Associated With Treatment as Measured by the M.D. Anderson Symptom Inventory - LC(MDASI-LC)

The symptom score assessed: pain, fatigue, nausea, sleep, distress, shortness of breath, memory, appetite, drowsy, dry mouth, sadness, vomiting, numbness, cough, constipation, general activity, mood, work, relationships with other people, walking, enjoyment. Scores for each item range from 0 to 10, where 0 equaled no symptoms and 10 equaled worst possible symptoms. Assessed at baseline and end of treatment. (NCT00497770)
Timeframe: Baseline and end of treatment (up to 20 cycles [14 months])

,,,
Interventionparticipants (Mean)
Pain-baseline (n=281, 62, 35, 28)Pain-end of treatment (EOT) (n=118, 31, 17, 8)Fatigue-baseline (n=279, 62, 35, 28)Fatigue-EOT (n=119, 32, 17, 8)Nausea-baseline (n=280, 62, 35, 28)Nausea-EOT (n=118, 30, 17, 8)Sleep-baseline (n=280, 62, 34, 28)Sleep-EOT (n=118, 31, 17, 8)Distress (Upset)-baseline (n=277, 62, 34, 28)Distress (Upset)-EOT (n=119, 32, 17, 8)Shortness of Breath-baseline (n=282, 61, 35, 28)Shortness of Breath-EOT (n=118, 32, 17, 8)Memory-baseline (n=278, 62, 35, 28)Memory-EOT (n=119, 32, 17, 8)Appetite-baseline (n=281, 62, 35, 28)Appetite-EOT (n=119, 32, 16, 8)Drowsy-baseline (n=280, 65, 32, 28)Drowsy-EOT (n=119, 32, 17, 8)Dry Mouth-baseline (n=279, 62, 35, 28)Dry Mouth-EOT (n=119, 32, 17, 8)Sadness-baseline (n=278, 61, 35, 28)Sadness-EOT (n=119, 32, 17, 8)Vomiting-baseline (n=282, 62, 35, 28)Vomiting-EOT (n=119, 32, 17, 8)Numbness-baseline (n=280, 61, 33, 28)Numbness-EOT (n=119, 32, 17, 8)Cough-baseline (n=281, 61, 35, 28)Cough-EOT (n=119, 31, 16, 8)Constipation-baseline (n=281, 62 34, 28)Constipation-EOT (n=119, 32 17, 8)General Activity-baseline (n=280, 62, 35, 28)General Activity-EOT (n=119,32,16,8)Mood-baseline (n=279, 61, 34, 28)Mood-EOT (n=119, 32, 16, 8)Work-baseline (n=278, 62, 35, 28)Work-EOT (n=118, 32, 16, 8)Relationships with Others-baseline(n=282,61,35,28)Relationships with Others-EOT (n=118, 31, 16, 8)Walking-baseline (n=280, 62, 35, 28)Walking-EOT (n=119, 32, 16, 8)Enjoyment-baseline (n=282, 61, 35, 28)Enjoyment-EOT (n=119, 32, 16, 8)
African American3.735.164.584.911.551.673.664.163.003.223.705.062.712.813.114.723.323.692.732.442.572.810.971.063.343.842.983.942.372.883.564.533.111.884.234.632.412.714.004.443.514.00
Asian American3.891.824.143.711.341.183.212.942.762.412.691.822.091.823.372.382.631.593.292.712.742.060.660.882.762.472.631.632.792.472.462.882.441.892.512.812.001.502.802.942.832.81
Caucasian3.373.524.634.981.391.763.243.582.893.053.583.762.402.472.842.873.203.302.892.912.402.390.740.972.422.342.892.502.472.553.713.892.852.073.724.051.832.173.844.123.333.39
Hispanic3.751.885.044.002.430.754.113.134.073.634.292.882.500.634.864.004.363.133.362.254.323.881.250.132.390.632.613.133.541.504.463.633.861.864.864.253.072.754.214.004.394.00

[back to top]

Patient Response to Treatment Measured by RECIST Criteria

RECIST response categories: Progressive disease (PD): >=20% increase in sum of longest diameter (LD) of target lesion(s), taking as reference smallest sum LD recorded since treatment started. Complete response (CR): disappearance of all target lesions. Partial response (PR): >=30% decrease in sum of LD of target lesion(s), taking as reference baseline sum LD. Stable disease (SD): neither sufficient shrinkage to qualify as PR nor sufficient increase to qualify as PD. (NCT00503997)
Timeframe: at 8 weeks

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot Evaluable
Pemetrexed/Oxaliplatin1181922

[back to top]

Objective Response Rate (OR) Where OR=CR+PR: Number of Participants With Responses of Complete Response (CR) and Partial Response (PR)

"Complete Response (CR): Complete disappearance of all measurable & non-measurable disease; No new lesions; No disease related symptoms; Normalization of markers & other abnormal lab values.~Partial Response (PR): Applies only to those with at least one measurable lesion. >/= 30% decrease under baseline of sum of longest diameters of all target measurable lesions. No unequivocal progression of non-measurable disease. No new lesions. All target measurable lesions assessed using same techniques as baseline.~Progression: 20% increase in sum of longest diameters of target measurable lesions over smallest sum observed (over baseline if no decrease during therapy) using same techniques as baseline. Unequivocal progression of non-measurable disease in opinion of treating physician.~Evaluated for symptoms 1-2 times per week while receiving treatment then 2 weeks after stopping study treatment (expected 4 cycles)." (NCT00508144)
Timeframe: Evaluated with 3 week treatment cycles, up to 4 cycles or 12 weeks

InterventionParticipants (Count of Participants)
Alimta3

[back to top]

1-year Progression Free Survival Rate in Chemo-naive Select Stage IIIB or Stage IV NSCLC Patients

One-year progression-free survival was defined from the time from initiation of study treatment to the first date of disease progression or death as a result of any cause. Progression was defined as at least a 20% increase in the sum of the longest diameter (LD) of target lesions taking as references the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. Time was censored at the date of the last follow-up visit for patients who were still alive and have not progressed. The one-year progression free survival rate is a percentage, representing the fraction of treated patients who, after one-year, are disease free or alive. (NCT00509366)
Timeframe: 1 year

Interventionpercentage of treated patients (Number)
Treatment19.15

[back to top]

Mean Change From Baseline to Follow-up Cycle in Quality of Life - Functional Assessment of Cancer Therapy-Lung (FACT-L)

The outcome measure is mean change in the Trial Outcome Index (TOI) between baseline and each follow-up assessment measured by the Functional Assessment of Cancer Therapy-Lung (FACT-L). The FACT-L instrument consists of 34 items to assess physical (PWB), social and family (SWB), emotional (EWB), functional well-being (FWB) and additional lung specific concerns (LCS). Using a 5-point Likert type scale, responses to individual items range from 0 (not at all) to 4 (Very Much) with higher scores indicating better quality of life. The TOI is the sum of PWB (7 items), FWB (7 items) and LCS scores (7 items), which each have a possible range between 0 and 28. Therefore, TOI ranges from 0 to 84. (NCT00509366)
Timeframe: Baseline, Every 21 days for a maximum of 6 cycles

Interventionunits on a scale (Mean)
Change from baseline to Cycle 1Change from baseline to Cycle 2Change from baseline to Cycle 3Change from baseline to Cycle 4Change from baseline to Cycle 5Change from baseline to Cycle 6
Treatment0.15-1.04-1.28-2.62-6.14-0.72

[back to top]

Median Time to Progressive Disease

Median time to progressive disease was defined as the time from enrollment to the the time at which 50% of patients had experienced disease progression. Enrollment is defined as having successful genomic analysis and start of chemotherapy. Time was censored at date of death for patients who have not had documented disease progression, at first available date of other anti-tumor therapy for patients who were either administered other anti-tumor therapy prior to documented disease progression or administered other anti-tumor therapy without documented disease progression, and at last date of followup if neither non-protocol therapy was administered nor progression documented. (NCT00509366)
Timeframe: 1 Year

Interventionmonths (Number)
Treatment4.60

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from treatment start until death from any cause. The median overall survival time is used to measure OS. (NCT00517595)
Timeframe: OS was measured from day 1 of treatment until time of death, assessed up to 20 months.

InterventionMonths (Median)
Pemetrexed, Gemcitabine, and Bevacizumab8.78

[back to top]

Progression Free Survival (PFS) by Baseline Eastern Cooperative Oncology Group (ECOG) Performance Status

PFS is defined as the duration of time from start of treatment to time of progression or death, whichever comes first. The median progression free survival is the parameter used to describe PFS. ECOG performance status describes how daily living activities of the patient are affected by disease. ECOG of 0 means the patient is fully active without restriction. ECOG of 1 means the patient is restricted in physically strenuous activity but is able to carry out light work. The investigator assigned the ECOG score at baseline (i.e., before the patient started study treatment). (NCT00517595)
Timeframe: PFS was measured from day 1 of treatment until time of progression (assessed every 8 weeks) or death, whichever came first, assessed up to 15 months.

InterventionMonths (Median)
Baseline ECOG performance status 0 (N=18)Baseline ECOG performance status 1 (N=30)
Pemetrexed, Gemcitabine, and Bevacizumab7.574.67

[back to top]

Progression Free Survival (PFS)

PFS is defined as the duration of time from start of treatment to time of progression or death, whichever comes first. Progression is defined per RECIST criteria v1.0 as a measurable increase in the smallest diameter of any target lesion, progression of existing non-target lesions, or the appearance of 1 or more new lesions. The median progression free survival is the parameter used to describe PFS. (NCT00517595)
Timeframe: PFS was measured from day 1 of treatment until time of progression (assessed every 8 weeks) or death, whichever came first, assessed up to 15 months.

InterventionMonths (Median)
Pemetrexed, Gemcitabine, and Bevacizumab4.90

[back to top]

Time to Progression (TTP)

Time to progression is defined as the time from treatment start until objective tumor progression. Progression is defined per RECIST criteria v1.0 as a measurable increase in the smallest diameter of any target lesion, progression of existing non-target lesions, or the appearance of 1 or more new lesions. The median time to progression is the parameter used to describe TTP. (NCT00517595)
Timeframe: TTP was measured from day 1 of treatment until time of progression (assessed every 8 weeks), assessed up to 15 months.

InterventionMonths (Median)
Pemetrexed, Gemcitabine, and Bevacizumab5.56

[back to top]

Overall Response

Response was evaluated via changes from baseline in radiological tumor measurements performed after every 4th treatment cycle and at the end of treatment or time of progression. Response was evaluated using RECIST version 1.0 guidelines, where complete response (CR) is the disappearance of all target lesions; partial response (PR) is >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (SD) is neither sufficient shrinkage in sum of LD of target lesions to be PR nor increase of >=20%; Progressive Disease (PD) is the increase in existing lesions or new lesions. (NCT00517595)
Timeframe: Response to treatment was assessed after every 8 weeks of treatment, up to 50 weeks.

InterventionParticipants (Number)
Complete response (CR)Partial response (PR)Stable disease (SD)Progressive disease (PD)Not evaluable (NE)
Pemetrexed, Gemcitabine, and Bevacizumab0172164

[back to top]

Overall Survival (OS) by Baseline Eastern Cooperative Oncology Group (ECOG) Performance Status

Overall survival is defined as the time from treatment start until death from any cause. The median overall survival time is used to measure OS. ECOG performance status describes how daily living activities of the patient are affected by disease. ECOG of 0 means the patient is fully active without restriction. ECOG of 1 means the patient is restricted in physically strenuous activity but is able to carry out light work. The investigator assigned the ECOG score at baseline (i.e., before the patient started study treatment). (NCT00517595)
Timeframe: OS was measured from day 1 of treatment until time of death, assessed up to 20 months.

InterventionMonths (Median)
Baseline ECOG performance status 0 (N=18)Baseline ECOG performance status 1 (N=30)
Pemetrexed, Gemcitabine, and Bevacizumab19.876.71

[back to top]

Progression-free Survival (PFS)

Defined as the time from date of first dose to the first observation of disease progression (PD), or death due to any cause. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

Interventionmonths (Median)
Pemetrexed Plus Carboplatin5.8
Docetaxel Plus Carboplatin6.0

[back to top]

Survival Without Clinically Important Grade 3 or 4 Toxicity

Survival without Grade 3 or 4 toxicity is the time from date of randomization to the first date of the following clinically important Grade 3 or 4 TEAEs graded by the Common Terminology Criteria for Adverse Events [CTCAE], version 3.0: neutropenia (lasting >5 days), febrile neutropenia, documented infections related to neutropenia, anemia, thrombocytopenia, fatigue, nausea, vomiting, diarrhea, stomatitis, and neurosensory events; or death due to any cause. Participants who were alive without experiencing Grade 3 or 4 toxicity were censored for this analysis at the date of last contact. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

Interventionmonths (Median)
Pemetrexed Plus Carboplatin3.6
Docetaxel Plus Carboplatin1.3

[back to top]

Survival Without Grade 3 or 4 Toxicity

"Defined as the time from date of randomization to first date of a Grade 3 or 4 treatment-emergent adverse event (TEAE; as graded by the National Cancer Institute Common Terminology Criteria for Adverse Events [CTCAE], version 3.0) or death due to any cause. Grade 3 TEAE: Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated. Grade 4 TEAE: Life-threatening consequences; urgent intervention indicated.~Participants who were alive without experiencing Grade 3 or 4 toxicity were censored at the date of last contact." (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

Interventionmonths (Median)
Pemetrexed Plus Carboplatin3.2
Docetaxel Plus Carboplatin0.7

[back to top]

Survival Without Grade 4 Toxicity

Survival without Grade 4 toxicity is the time from the date of randomization to the first date of a Grade 4 TEAE or death due to any cause. Participants who are alive without experiencing Grade 4 toxicity will be censored for this analysis at the date of last contact. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 4 toxicity) have been observed (up to 33.3 months).

Interventionparticipants (Median)
Pemetrexed Plus Carboplatin12.2
Docetaxel Plus Carboplatin2.0

[back to top]

Overall Survival (OS)

OS is the duration from enrollment to death. For participants who are alive, OS is censored at the last contact. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

Interventionmonths (Median)
Pemetrexed Plus Carboplatin14.9
Docetaxel Plus Carboplatin14.7

[back to top]

Percentage of Participants With Tumor Response (Response Rate)

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response (CR)=disappearance of all target lesions; Partial Response (PR)=at least a 30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD)=at least a 20% increase in sum of longest diameter of target lesions; Stable Disease (SD)=small changes not meeting above criteria. Response rate (%)=Number of participants with CR+PR/Number of participants analyzed *100. Disease Control rate=Number of participants with SD+PR+CR/Number of participants analyzed *100. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

,
Interventionpercentage of participants (Number)
Tumor Response RateDisease Control RateComplete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Docetaxel Plus Carboplatin23.164.4023.141.317.318.3
Pemetrexed Plus Carboplatin34.074.50.933.040.616.09.4

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death as a result of any cause. Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. CR=disappearance of all target lesions; PR=at least a 30% decrease in sum of longest diameter of target lesions. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

Interventionmonths (Median)
Pemetrexed Plus Carboplatin5.5
Docetaxel Plus Carboplatin5.4

[back to top]

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. (NCT00520676)
Timeframe: Baseline to until 218 events (defined as death or Grade 3 or 4 toxicity) have been observed (up to 33.3 months).

,
Interventionparticipants (Number)
Non-Serious Adverse Events (AEs)Serious Adverse Events (SAEs)
Docetaxel Plus Carboplatin10035
Pemetrexed Plus Carboplatin9428

[back to top]

Time to Progression

Estimated probable duration from on-study date to date of disease progression, using the Kaplan-Meier method with censoring (see analysis population description for additional details). Disease progression is defined under RECIST v1.1 as >=20% increase in sum of longest diameters of target lesions, unequivocal progression of non-target lesions, or appearance of new lesions. (NCT00520845)
Timeframe: 2 years from date of registration

Interventiondays (Median)
Treatment Arm89

[back to top]

Overall Response Rate

Overall response rate is measured by complete response + partial response. Number of patients in each response category, per RECIST v1.1, summarized as follows for target lesion criteria (see RECIST v1.1 for additional details): complete response (CR),disappearance of target lesions; partial response (PR), >=30% decrease in sum of longest diameter of target lesions; progressive disease (PD), >=20% increase in sum of LD of target lesions or appearance of new lesions; stable disease (SD), insufficient change in target lesions or new lesions to qualify as either PD or SD. Patients are categorized according to the best response achieved prior to occurrence of progressive disease, where best response hierarchy is CR>PR>SD>PD. (NCT00520845)
Timeframe: On-treatment date to date of disease progression (assessed at 6 weeks up to 2 years)

Interventionparticipants (Number)
Treatment Arm0

[back to top]

Median Survival

Estimated probable duration of life from on-study date to date of death from any cause, using the Kaplan-Meier method with censoring (see analysis population description for additional details) (NCT00520845)
Timeframe: 2 years from date of registration

Interventiondays (Median)
Treatment Arm184

[back to top]

Number of Patients With Adverse Events, Discontinuations, or Deaths Possibly Due to Study Drug

AdEERS= Adverse Event Expedited Reporting System; AE = adverse event. Patients may be counted in more than 1 category. Includes events that were considered possibly related to study drug (PRSD) as judged by the investigator. (NCT00520936)
Timeframe: every cycle (up to 2 years and 7 months)

,,,,,,,
InterventionParticipants (Number)
>=1 AdEERs possibly related to study drugDiscontinued due to AE possibly related to drugDied on therapy possibly related to study drugDied within 31 days of last dose of drug PRSD
Ependymoma2000
Ewing's Sarcoma/Peripheral Primitive Neuroectodermal Tumors2100
Medulloblastoma/Supratentorial Primitive Neuroectodermal Tumor2200
Neuroblastoma (Measureable Disease)2000
Neuroblastoma (Metaiodobenzylguanidine Positive Evaluable)3000
Non-Brainstem High-Grade Glioma2000
Osteosarcoma3000
Rhabdomyosarcoma0000

[back to top]

Percentage of Participants With Overall Tumor Response (Response Rate)

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response = disappearance of all target lesions. Partial Response = 30% decrease in sum of longest diameter of target lesions. Response rate (percent [%])= (number of participants with complete response (CR) or partial response (PR) in stratum/number of participants in stratum)*100. (NCT00520936)
Timeframe: baseline to measured progressive disease (up to 1 year)

InterventionPercentage of Participants (Number)
Osteosarcoma0
Ewing's Sarcoma/Peripheral Primitive Neuroectodermal Tumors0
Rhabdomyosarcoma0
Neuroblastoma (Measureable Disease)0
Neuroblastoma (Metaiodobenzylguanidine Positive Evaluable)0
Ependymoma0
Medulloblastoma/Supratentorial Primitive Neuroectodermal Tumor0
Non-Brainstem High-Grade Glioma0

[back to top]

Duration of Response

The duration of a complete response (CR) or partial response (PR) was defined as the time from the first objective status assessment of CR or PR to the first date of progression or death as a result of any cause: CR was achieved if all tumor lesions disappeared; PR was achieved if there was >=30% decrease in sum of the longest diameter (LD) of target lesions (reference: baseline sum LDs) or complete disappearance of target lesions with persistence (but not worsening) of >=1 nontarget lesions and no appearance of new lesions. (NCT00523419)
Timeframe: Baseline to 31 months

Interventionmonths (Number)
Pemetrexed9.5

[back to top]

Progression-Free Survival (PFS)

PFS was from date of study enrollment to first date of objectively determined progressive disease (PD) or death from any cause. For participants who did not die as of data cut-off date and who did not have objective PD, PFS was censored at date of last objective progression-free disease assessment. For participants who received subsequent systemic anticancer therapy (after discontinuation from study drug) before objectively determined disease progression or death, PFS was censored at date of last objective progression-free disease assessment, before post-discontinuation chemotherapy. (NCT00523419)
Timeframe: Baseline to 10.4 months

Interventionmonths (Median)
Pemetrexed1.4

[back to top]

Overall Survival (OS) Time

OS was the duration from enrollment to death. For participants who lived, OS was censored at the last contact. (NCT00523419)
Timeframe: Baseline to 27.6 months

Interventionmonths (Median)
Pemetrexed5.5

[back to top]

Number of Participants With Adverse Events (Pharmacology Toxicity)

Pharmacology toxicity was defined as serious and non-serious adverse events. Summaries of these adverse events are located in the Reported Adverse Event Section. (NCT00523419)
Timeframe: Baseline to 21 months

Interventionparticipants (Number)
Serious Adverse EventsAll Other Nonserious Adverse Events
Pemetrexed1126

[back to top]

Percentage of Participants With Tumor Response

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria: Complete Response (CR) = disappearance of all target lesions; Partial Response (PR) = at least a 30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD) = at least a 20% increase in sum of longest diameter of target lesions; Stable Disease (SD) = small changes that do not meet above criteria. Tumor Response Rate(%) = sum of number of PR + CR observed/number of participants qualified for tumor response analysis * 100. (NCT00523419)
Timeframe: Baseline to 21 months

Interventionpercentage of participants (Number)
Response RateComplete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Unknown
Pemetrexed3.103.115.668.812.5

[back to top]

Phase 2: Percentage of Participants With Overall Survival (OS) at 1 Year

OS was defined as the time from date of enrollment to death due to any cause. (NCT00529100)
Timeframe: Baseline to date of death from any cause (up to 1 year)

Interventionpercentage of participants (Number)
Pemetrexed/Cisplatin/Radiation Phase 279.0

[back to top]

Phase 2: Time to Progressive Disease (PD)

Time to PD was defined as the time from study enrollment to the first date of objective disease progression as defined by Response Evaluation Criteria in Solid Tumors (RECIST 1.0) as at least a 20% increase in the sum of the longest diameter (LD) of target lesions as references the smallest sum LD recorded since treatment started or the appearance of 1 or more new lesions. Time to PD was censored at the date of death if death was due to other cause. For participants not known to have died as of the data cut-off date and who did not have PD, time to PD was censored at the last progression-free disease assessment. For participants who received subsequent cancer therapy (after discontinuation from the study therapy) before PD, time to PD was censored at the date of subsequent cancer therapy initiation. (NCT00529100)
Timeframe: Baseline to measured PD (up to 3 years)

Interventionmonths (Median)
Pemetrexed/Cisplatin/Radiation Phase 213.7

[back to top]

Phase 1: Maximum Tolerated Dose (MTD) of Pemetrexed in Combination With Cisplatin and Radiation Therapy

Recommended Phase 2 MTD was highest dose at which no more than 1 of 6 participants experienced dose level toxicity (DLT). DLT=(1) Grade 3/4 dysphagia/esophagitis, leukopenia, thrombocytopenia, febrile neutropenia, fatigue/malaise, pneumonitis, dermatitis, persistent elevation of bilirubin/alkaline phosphatase/aspartate aminotransferase only if resulting in delay of radiotherapy >1 week, delay of pemetrexed/cisplatin Cycle 2 >2 weeks, or delay of pemetrexed/cisplatin Cycle 3 past 5 weeks after radiotherapy; (2) other Grade 3 or 4 toxicity possibly related to concurrent treatment administration. (NCT00529100)
Timeframe: Baseline to measured progressive disease (PD; up to 1 year)

Interventionmilligrams per square meter (mg/m^2) (Number)
Concurrent Phase MTD: PemetrexedConcurrent Phase MTD: CisplatinConsolidation Phase MTD: PemetrexedConsolidation Phase MTD: Cisplatin
Pemetrexed/Cisplatin/Radiation Phase 15002050075

[back to top]

Phase 1: Number of Participants With Adverse Events (AE; Toxicity)

A listing of AEs is located in the Reported Adverse Event module. (NCT00529100)
Timeframe: Baseline to measured PD (up to 1 year)

Interventionparticipants (Number)
Serious Adverse Events (SAEs)Other Non-serious Adverse Events (AEs)
Pemetrexed/Cisplatin/Radiation Phase 1510

[back to top]

Phase 2: Site of Progressive Disease (PD)

Summarized participants with local (progression within the sites of initial disease)/regional (disease progression adjacent to but not within the site of initial disease at the start of treatment), distant (disease progression that is blood borne to other parts of the body, including outside the chest or involving the contralateral lung), and local + distant sites of disease. Objective PD is defined by Response Evaluation Criteria in Solid Tumors (RECIST 1.0) as at least a 20% increase in the sum of the longest diameter (LD) of target lesions as references the smallest sum LD recorded since treatment started or the appearance of 1 or more new lesions. (NCT00529100)
Timeframe: Baseline to measured PD (up to 3 years)

Interventionparticipants (Number)
Local/RegionalDistantLocal + DistantUnknown
Pemetrexed/Cisplatin/Radiation Phase 281711

[back to top]

Phase 2: Percentage of Participants With Progression Free Survival (PFS)

The percentage of participants not known to have died as of the data cut-off date or last contact and who did not have PD. (NCT00529100)
Timeframe: Baseline and 1 year and 2 years and 3 years

Interventionpercentage of participants (Number)
1 Year2 Years3 Years
Pemetrexed/Cisplatin/Radiation Phase 248.730.820.2

[back to top]

Phase 2: Percentage of Participants With Overall Survival (OS) at 2 Years and 3 Years

OS was defined as the time from date of enrollment to death due to any cause. (NCT00529100)
Timeframe: Baseline and 2 years and 3 years

Interventionpercentage of participants (Number)
2 years3 years
Pemetrexed/Cisplatin/Radiation Phase 256.446.2

[back to top]

Progression Free Survival (PFS)

PFS was defined as the period from study entry until PD, death, or date of last contact. For participants not known to have died as of the data cut-off date and who did not have PD, the PFS date was censored at the last contact date (contacts considered in the determination of last progression free disease assessment). (NCT00529100)
Timeframe: Baseline to measured PD (up to 36 months)

Interventionmonths (Median)
Pemetrexed/Cisplatin/Radiation Phase 211.8

[back to top]

Phase 2: Percentage of Participants With Objective Tumor Response (Response Rate)

Response using Response Evaluation Criteria In Solid Tumors (RECIST 1.0). Complete Response (CR)=disappearance of all target lesions; Partial Response (PR)=30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD)=20% increase in sum of longest diameter of target lesions; Stable Disease (SD)=small changes that do not meet above criteria. Objective response rate (%)=number of objective responders divided by the number of participants with measurable disease * 100, where objective responders are those participants who have met criteria either for CR or PR. (NCT00529100)
Timeframe: Baseline to measured PD (up to 3 years)

Interventionpercentage of participants (Number)
Complete ResponsePartial Response
Pemetrexed/Cisplatin/Radiation Phase 2045.95

[back to top]

Progression-Free Survival (PFS)

PFS was defined as the time from date of randomization to the first documented observation of disease progression or death from any cause. Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Progressive disease (PD) was defined as having at least a 20% increase in sum of the longest diameter of target lesions or the appearance of new lesions. PFS was censored at the date of the last objective progression-free disease assessment for participants who did not experience PD or death at the data inclusion cut-off date. (NCT00530621)
Timeframe: Baseline to measured progressive disease up to 9.92 months

Interventionmonths (Median)
Pemetrexed + Enzastaurin2.96
Pemetrexed + Placebo3.02

[back to top]

Number of Participants Who Died During the 30 Days After Treatment Discontinuation

Reported are the deaths due to study disease and adverse events (AEs) that occurred during the 30 days after treatment discontinuation. (NCT00530621)
Timeframe: End of study treatment [16 Cycles (21-day cycles, except Cycle 1 [28 days])] through 30 days after treatment discontinuation

,
InterventionParticipants (Count of Participants)
Study DiseaseAEs
Pemetrexed + Enzastaurin52
Pemetrexed + Placebo100

[back to top]

Number of Participants Who Died During the Study Treatment

Reported are the deaths due to study disease and adverse events (AEs) that occurred while on study treatment. (NCT00530621)
Timeframe: Baseline through study completion [up to 16 Cycles (21-day cycles, except Cycle 1 [28 days])]

,
InterventionParticipants (Count of Participants)
Study DiseaseAEs
Pemetrexed + Enzastaurin21
Pemetrexed + Placebo33

[back to top]

Tumor Biomarkers

Protein expression was measured using an Immunohistochemistry (IHC) assay from the tumor tissue samples. IHC histo-scores (H-scores) were determined separately for each of the 3 biomarkers: folate receptor alpha (FR alpha) in cytoplasm and apical membrane, thymidylate synthase (TS) in cytoplasm and nucleus, and thyroid transcription factor-1 (TTF1) in the nucleus. Tumor tissue samples were to be scored using a 0 (negative, no staining) to 3+ (brightest staining) scoring system for cytoplasmic and nuclear staining. IHC H-score was calculated using the formula: 1 * (percentage of cells stained 1+) + 2 * (percentage of cells stained 2+) + 3 * (percentage of cells stained 3+), giving a minimum score of 0 to a maximum score of 300. The maximum score indicates the strongest expression. (NCT00530621)
Timeframe: Tumor samples collected at baseline

,,
InterventionH-scores (Mean)
FR Alpha in CytoplasmFR Alpha in Apical MembraneTS in CytoplasmTS in NucleusTTF-1 in Nucleus
Pemetrexed + Enzastaurin55.969.9616.385.9247.88
Pemetrexed + Placebo24.5018.2522.308.0032.35
Total (Pemetrexed + Enzastaurin and Pemetrexed + Placebo)41.9813.6418.966.8341.13

[back to top] [back to top]

Duration of Disease Control (DDC)

DDC was defined as the time from randomization to the first documented observation of disease progression or death from any cause and was limited to the participants with a best tumor response of complete response (CR), partial response (PR), or stable disease (SD). Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Progressive disease (PD) was defined as having at least a 20% increase in sum of the longest diameter of target lesions or the appearance of new lesions. CR was defined as the disappearance of all target lesions. PR was defined as having at least a 30% decrease in sum of longest diameter of target lesions. SD was defined as small changes that did not meet the above criteria. DDC was censored at the date of the last objective progression-free disease assessment for participants who did not experience PD or death. (NCT00530621)
Timeframe: Baseline to measured progressive disease up to 9.92 months

Interventionmonths (Median)
Pemetrexed + Enzastaurin5.32
Pemetrexed + Placebo6.31

[back to top]

Overall Survival (OS)

OS was defined as the duration from the date of randomization to the date of death from any cause. For participants who were alive at the time of the data inclusion cutoff, OS was censored at the date the participant was last known to be alive. (NCT00530621)
Timeframe: Baseline to date of death from any cause up to 12.32 months

Interventionmonths (Median)
Pemetrexed + Enzastaurin9.63
Pemetrexed + Placebo7.39

[back to top]

Percentage of Participants With Complete Response or Partial Response (Tumor Response Rate)

Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Participants with a best response of complete response (CR) or partial response (PR) were considered to have had a tumor response. CR was defined as the disappearance of all target lesions. PR was defined as having at least a 30% decrease in sum of longest diameter of target lesions. Percentage of participants was calculated as the total number of participants affected divided by the number of participants analyzed then multiplied by 100. (NCT00530621)
Timeframe: Baseline to measured progressive disease up to 9.92 months

Interventionpercentage of participants (Number)
Pemetrexed + Enzastaurin3.9
Pemetrexed + Placebo2.6

[back to top]

Duration of Response (DoR)

The duration of a complete response (CR) or partial response (PR) was defined as the time from first objective status assessment of CR or PR to the first time of progression or death from any cause. Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST v1.0) criteria. CR was defined as the disappearance of all target lesions and PR was defined as having at least a 30% decrease in sum of longest diameter (LD) of target lesions taking as reference the baseline sum of LDs. For participants who died without progressive disease or who were alive, DoR was censored at the last contact of progression free assessment. For participants who received subsequent anticancer therapy (after discontinuation from all study treatment) prior to PD, DoR was censored at the date of last progression-free assessment prior to the initiation of post-discontinuation anticancer therapy. Due to early study closure, DoR was not analyzed. (NCT00533429)
Timeframe: Time of response to disease progression or death from any cause up to 12.2 months

InterventionMonths (Median)
Pemetrexed + Carboplatin + Bevacizumab + Enzastaurin4.7
Pemetrexed + Carboplatin + Bevacizumab + Placebo3.5

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of randomization to the date of death from any cause. For participants who were not known to have died as of the data cutoff, OS was censored at the last contact date. (NCT00533429)
Timeframe: Randomization to date of death up to 14.3 months

Interventionmonths (Median)
Pemetrexed + Carboplatin + Bevacizumab + Enzastaurin9.1
Pemetrexed + Carboplatin + Bevacizumab + Placebo7.6

[back to top]

Pharmacology Toxicity and Adverse Events (AEs)

Clinically significant events were defined as serious AEs (SAEs) and other non-serious AEs. Participants who died due to progressive disease (PD) or an AE while on treatment and or died during the 30 day post-treatment follow-up are included. A summary of SAEs and other non-serious AEs, regardless of causality, is located in the Reported Adverse Events module. (NCT00533429)
Timeframe: Randomization up to 14.3 months and 30-day follow-up

,
InterventionParticipants (Count of Participants)
Non-serious AEsSerious AEsDeaths Due to PDDeaths Due to AEsDeaths within 30-days after treatment
Pemetrexed + Carboplatin + Bevacizumab + Enzastaurin196010
Pemetrexed + Carboplatin + Bevacizumab + Placebo207010

[back to top]

Time to Progressive Disease (TTPD)

TTPD was defined as the time from the date of randomization until the first date of objectively determined progressive disease (PD). For participants who died without objective PD (including death from study disease), TTPD was censored at the date of the last objective progression-free disease assessment. For participants not known to have died as of the data cutoff and did not have PD, TTPD was censored at the date of the last objective progression-free disease assessment. (NCT00533429)
Timeframe: Randomization to measured PD or death from any cause up to 12.2 months

InterventionMonths (Median)
Enzastaurin + Pemetrexed + Carboplatin + Bevacizumab4.3
Pemetrexed + Carboplatin + Bevacizumab + Placebo4.8

[back to top]

Progression-Free Survival (PFS)

PFS was defined as the time from date of randomization to the first observation of progressive disease (PD) or death due to any cause. For participants not known to have died as of the data cutoff date and who did not have objective PD, PFS was censored at the date of the last objective progression-free assessment. For participants who received subsequent anticancer therapy (after discontinuation from all study treatment) prior to objective disease progression or death, PFS was censored at the date of the last objective progression-free assessment prior to the initiation of post-discontinuation anticancer therapy. (NCT00533429)
Timeframe: Randomization to measured PD or death from any cause up to 12.2 months

Interventionmonths (Number)
Pemetrexed + Carboplatin + Bevacizumab + Enzastaurin3.5
Pemetrexed + Carboplatin + Bevacizumab + Placebo4.3

[back to top]

Percentage of Participants With Complete Response (CR) or Partial Response (PR) (Response Rate)

"Tumor response rate was defined as number of participants with overall best response of CR or PR over number of protocol qualified participants using the Response Evaluation Criteria in Solid Tumors (RECIST v1.0) criteria. CR was defined as the disappearance of all tumor lesions. PR was defined as at least a 30% decrease in the sum of the longest diameter (LD) of target lesions taking as reference the baseline sum of LDs or complete disappearance of target lesions, with persistence (but not worsening) of 1 or more non-target lesions. In either case, no new lesions may have appeared.~Percentage of participants was calculated as: (number of participants with CR or PR/ number of participants qualified for tumor response analysis) × 100." (NCT00533429)
Timeframe: Randomization to measured progressive disease or death from any cause up to 12.2 months

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin + Bevacizumab + Enzastaurin20.0
Pemetrexed + Carboplatin + Bevacizumab + Placebo30.0

[back to top]

Part 1: Evaluate Safety [Toxicity, Serious Adverse Events (SAEs) and Reasons for Participant's Discontinuation]

Presented are data that evaluates safety based on toxicity using Common Terminology Criteria for Adverse Events (CTCAE v3.0), SAEs, and discontinuations due to SAEs or other non-serious adverse events (AE's) of study participants. A summary of SAEs and other non-serious AEs regardless of causality is located in the Reported Adverse Event module. (NCT00538681)
Timeframe: Cycle 1 (28-day cycle), Cycles 2, 3, 4, 5, and 6 (21-day cycles) and 30-day follow up

,
InterventionParticipants (Count of Participants)
SAEsAEsDiscontinued due to AE
Part 1- Cohort 1591
Part 1- Cohort 2041

[back to top]

Part 2: To Evaluate the Safety and Toxicity Profile of Study Treatments

The safety and toxicity profile for Part 2 was defined as serious adverse events (SAEs) and other non-serious adverse events (AEs). A summary of SAEs and other non-serious AEs regardless of causality is located in the Reported Adverse Event module. (NCT00538681)
Timeframe: Cycle 1 (28-day cycle), Cycles 2, 3, 4, 5, and 6 (21-day cycles) and 30-day follow-up

,
InterventionParticipants (Count of Participants)
SAEsAEs
Part 2- Enzastaurin28
Part 2- Placebo48

[back to top]

Percentage of Patients With Completely Resected NSCLC Tumors That Can Be Analyzed and Used to Direct Adjuvant Chemotherapy

The percentage of patients with completely resected NSCLC tumors who had successful genomic analysis and assigned to treatment among patients. All 31 patients enrolled in the study had completely resected tumors. These tumors included a mixture of squamous and non-squamous histologies as indicated the original protocol. However, an amendment dated January 25, 2010 limited eligibility to patients with non-squamous disease. Given that only 5 patients were accrued into the study after this amendment, results reported will consider all histologies. (NCT00545948)
Timeframe: 4 years

InterventionPercentage of participants (Number)
All Registered Patients77.4

[back to top]

2-Year Progression-Free Survival Rate in Patients With Completely Resected Stage IB, II, or IIIA NSCLC

Progression-free survival time was defined as the time from initiation of study treatment to the first date of disease progression or death as a result of any cause. Progression was defined as at least a 20% increase in the sum of the longest diameter (LD) of target lesions taking as references the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. Time was censored at the date of the last follow-up visit for patients who were still alive and have not progressed. The two-year progression free survival rate is a percentage, representing the fraction of treated patients who, after two years, are disease free or alive. (NCT00545948)
Timeframe: 2 years

Interventionpercentage of treated patients (Number)
Treatment63.64

[back to top]

2-Year Overall Survival in Patients Treated for NSCLC

Overall survival time was defined as the time from initiation of study treatment to the date of death as a result of any cause. Time was censored at the date of the last follow-up visit for patients who were still alive. The two-year overall survival rate is a percentage, representing the fraction of treated patients who, after two years, are alive (NCT00545948)
Timeframe: 2 years

Interventionpercentage of treated patients (Number)
Treatment81.81

[back to top]

Number of Participants With Adverse Events

A summary of serious and all other non-serious adverse events (AEs), which include AEs reported for pharmacological toxicity, is located in the Reported Adverse Event module. (NCT00550173)
Timeframe: Randomization up to 39 months

,,
Interventionparticipants (Number)
Serious Adverse EventsOther Non-Serious Adverse Events
Erlotinib1876
Pemetrexed2267
Pemetrexed + Erlotinib2572

[back to top]

Number of Participants With Mutated or Non-Mutated Epidermal Growth Factor Receptor (EGFR) Genotype Status

EGFR mutation status was defined as: participants with any mutations detected were categorized as mutated and participants without any mutations detected were categorized as non-mutated. (NCT00550173)
Timeframe: Randomization to date of PD or death up to 38 months

,,
Interventionparticipants (Number)
MutatedNon-mutatedMutation status unknown (treated as missing)
Erlotinib862
Pemetrexed933
Pemetrexed + Erlotinib7105

[back to top]

Overall Survival (OS)

OS is defined as the time from randomization to the date of death from any cause. (NCT00550173)
Timeframe: Baseline to date of death from any cause up to 45.5 months

Interventionmonths (Median)
Pemetrexed + Erlotinib20.5
Erlotinib22.8
Pemetrexed17.7

[back to top]

Time to Worsening of Symptoms (TWS) on Lung Cancer Symptoms Scale (LCSS)

TWS assessed using the LCSS a participant rated lung cancer instrument which consisted of 9 disease related symptoms and quality of life (QoL) items, with 6 subscales related to major lung cancer symptoms (appetite, cough, fatigue, dyspnea, hemoptysis, and pain) and 3 summation items related to QoL (activity status, symptomatic distress, and overall QoL). Each item is marked on a visual analog scale (VAS) 0 (low symptoms/QoL items) to 100 (high symptoms/QoL items). The mean of the 6 subscales is used to calculate the average symptom burden index. TWS was measured from the date of study enrollment to the first date of a worsening in any 1 of the 6 LCSS symptom-specific items (as defined by a VAS 15-mm increase from baseline in the patient-reported score for any of these 6 items). (NCT00550173)
Timeframe: Randomization to first date of worsening of any of 6 LCSS symptom specific items or up to 12.4 months

Interventionmonths (Median)
Pemetrexed + Erlotinib1.0
Erlotinib0.8
Pemetrexed1.5

[back to top]

Percentage of Participants With a Tumor Response of Complete Response (CR) or Partial Response (PR) [Tumor Response Rate (TRR)]

TRR was defined as the number of responders (complete or partial) divided by the number of participants qualified for tumor response, as assessed using the RECIST version 1.0 guideline, multiplied by 100. RECIST guidelines: CR was defined as the disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeter (mm) and normalization of tumor marker level of non-target lesions; PR was defined as at least a 30% decrease in sum of longest diameter of target lesions. (NCT00550173)
Timeframe: Randomization to measured disease progression up to 38 months

Interventionpercentage of participants (Number)
Pemetrexed + Erlotinib44.7
Erlotinib29.3
Pemetrexed10.0

[back to top]

Percentage of Participants With CR, PR, and Stable Disease (SD) - Disease Control Rate (DCR)

DCR was defined as the percentage of participants with CR, PR, or SD divided by the number of randomized and treated participants as assessed using the RECIST criteria. CR was defined as the disappearance of all target lesions; PR was defined as 1) at least a 30% decrease in sum of longest diameter of target lesions or 2) complete disappearance of target lesions, with persistence (but not worsening) of 1 or more non-target lesions; PD was defined as at least a 20% increase in sum of longest diameter of target lesions; SD was defined as small changes that did not meet the above criteria. (NCT00550173)
Timeframe: Randomization to disease progression up to 38 months

Interventionpercentage of participants (Number)
Pemetrexed + Erlotinib64.5
Erlotinib52.4
Pemetrexed56.3

[back to top]

Progression-Free Survival (PFS)

PFS is defined as the time from randomization to the first date of progressive disease (PD; either objectively determined or clinical progression) or death from any cause. PD was defined as at least a 20% increase in sum of longest diameter of target lesions as assessed by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.0 guidelines. Time to disease progression was censored at the date of death. (NCT00550173)
Timeframe: Randomization to measured PD up to 38 months

Interventionmonths (Median)
Pemetrexed + Erlotinib7.4
Erlotinib3.8
Pemetrexed4.4

[back to top]

Probability of OS at 12 Months

OS time is censored at the date of last contact for participants who were still alive or lost to follow-up. (NCT00550173)
Timeframe: Month 12

Interventionpercent chance of survival (Number)
Pemetrexed + Erlotinib40.2
Erlotinib26.2
Pemetrexed18.1

[back to top] [back to top]

Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Figitumumab

Area under the plasma concentration time-curve from zero to the last measured concentration (AUClast). Figitumumab PK data was analyzed using noncompartmental methods (NCT00560573)
Timeframe: 0 (pre-dose), 1, 24, 72, 168, 336, 504 hr in Cycle 1 for dose escation and 0 (pre-dose), 1, 24, 72, 168, 336, 504 hr in Cycle 4 for expansion

Interventionmg*hr/L (Mean)
Figitumumab 6 mg/kg26020
Figitumumab 10 mg/kg21500
Figitumumab 20 mg/kg Dose Escalation84100
Figitumumab 20 mg/kg Expansion121200

[back to top]

Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Cisplatin

Area under the plasma concentration time-curve from zero to the last measured concentration (AUClast). Cisplatin PK data was analyzed using noncompartmental methods. Plasma exposure parameters for cisplatin were analyzed in the absence (Cycle 1) and presence (Cycle 2) of figitumumab (NCT00560573)
Timeframe: 0 (pre-dose), 1.917, 2.5, 3, 4, 5, 24 hr on Cycle 1, Day 1 and Cycle 2, Day 1 for cisplatin 75 mg/m^2 and 0 (pre-dose), 0.917, 1.5, 2, 3, 4, 23 hr on Cycle 1, Day 1 and Cycle 2, Day 1 for cisplatin 80 mg/m^2

Interventionng*hr/L (Mean)
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 1)38.23
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 2)45.18
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 1)37.12
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 2)48.29

[back to top]

Percentage of Participants With Blood Anti-drug Antibody (ADA) Specific for Figitumumab

Percentage of participants with positive total or neutralizing anti-drug antibody (ADA) for figitumumab (NCT00560573)
Timeframe: 30 min prior to figitumumab infusion in Cycle 1 and Cycle 4, end of study, fourth follow up visit (approximately 150 days after last dose)

,,,,,
InterventionPercentage of participants (Number)
C1D1 - NegativeC1D1 - PositiveC1D1 - Not determinedC4D1 - NegativeC4D1 - PositiveC4D1 - Not determinedEnd of Study - NegativeEnd of Study - PositiveEnd of Study - Not determinedFollow Up - NegativeFollow Up - PositiveFollow Up - Not determined
Figitumumab 10 mg/kg33.3066.7001000010000100
Figitumumab 20 mg/kg Dose Escalation50.0050.050.0050.050.0050.000100
Figitumumab 20 mg/kg Pemetrexed Expansion1000053.8046.246.2053.800100
Figitumumab 20 mg/kg RP2D Expansion 1.0 Infusion80.0020.030.0070.020.0080.000100
Figitumumab 20 mg/kg RP2D Expansion 2.5 Infusion1000071.4028.642.9051.714.3085.7
Figitumumab 6 mg/kg50.0050.016.7083.383.3016.700100

[back to top]

Progression-Free Survival (PFS)

Time from the date of enrollment to date of documented disease progression, or death due to any cause (NCT00560573)
Timeframe: Screening, from Cycle 2 onwards CT scan done within 7-10 days prior to next cycle (approximately Day 15 of each cycle), follow-up (30 days after last study treatment dose)

Interventionmonths (Median)
Overall Population5.7
Figitumumab 20 mg/kg With Gemcitabine and Cisplatin6.5
Figitumumab 20 mg/kg Expansion With Pemetrexed5.4

[back to top]

Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Gemcitabine

Area under the plasma concentration time-curve from zero to the last measured concentration (AUClast). Gemcitabine PK data was analyzed using noncompartmental methods. Plasma exposure parameters for gemcitabine were analyzed in the absence (Cycle 1) and presence (Cycle 2) of figitumumab (NCT00560573)
Timeframe: 0 (pre-dose), 0.417, 1, 1.5, 2.5, 3.5 hr on Cycle 1, Day 1 and Cycle 2, Day 8

Interventionng*hr/L (Mean)
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 1)6.665
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 2)12.53

[back to top]

Area Under the Curve From Time Zero to Last Quantifiable Concentration (AUClast) for Pemetrexed

Area under the plasma concentration time-curve from zero to the last measured concentration (AUClast). Pemetrexed PK data was analyzed using noncompartmental methods. Plasma exposure parameters for pemetrexed were analyzed in the absence (Cycle 1) and presence (Cycle 2) of figitumumab (NCT00560573)
Timeframe: 0, 0.167, 1.167, 2.167, 4.167, 6.167, 24.167 hr on Cycle 1, Day 1 and Cycle 2, Day 1 for pemetrexed 500 mg/m^2

Interventionng*hr/L (Mean)
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 1)132.2
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 2)161.6

[back to top]

Concentration at the End of Infusion (Cinf) for Figitumumab

Figitumumab pharmacokinetic (PK) data was analyzed using noncompartmental methods (NCT00560573)
Timeframe: Cycle 1 for dose escalation and Cycle 4 for dose expansion

Interventionmg/liter (L) (Mean)
Figitumumab 6 mg/kg120.4
Figitumumab 10 mg/kg137.0
Figitumumab 20 mg/kg Dose Escalation435.0
Figitumumab 20 mg/kg Expansion513.4

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Cisplatin

Cisplatin PK data was analyzed using noncompartmental methods. Plasma exposure parameters for cisplatin were analyzed in the absence (Cycle 1) and presence of (Cycle 2) figitumumab (NCT00560573)
Timeframe: 0 (pre-dose), 1.917, 2.5, 3, 4, 5, 24 hr on Cycle 1, Day 1 and Cycle 2, Day 1 for cisplatin 75 mg/m^2 and 0 (pre-dose), 0.917, 1.5, 2, 3, 4, 23 hr on Cycle 1, Day 1 and Cycle 2, Day 1 for cisplatin 80 mg/m^2

Interventionnanogram (ng)/mL (Mean)
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 1)3.816
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 2)3.880
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 1)2.845
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 2)3.490

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Gemcitabine

Gemcitabine PK data was analyzed using noncompartmental methods. Plasma exposure parameters for gemcitabine were analyzed in the absence (Cycle) 1 and presence (Cycle 2) of figitumumab (NCT00560573)
Timeframe: 0 (pre-dose), 0.417, 1, 1.5, 2.5, 3.5 hr on Cycle 1, Day 1 and Cycle 2, Day 8

Interventionng/L (Mean)
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 1)12.85
Cisplatin 80 mg/m^2/Gemcitabine Expansion (Cycle 2)23.97

[back to top]

Maximum Observed Plasma Concentration (Cmax) for Pemetrexed

Pemetrexed PK data was analyzed using noncompartmental methods. Plasma exposure parameters for pemetrexed were analyzed in the absence (Cycle 1) and presence (Cycle 2) of figitumumab (NCT00560573)
Timeframe: 0, 0.167, 1.167, 2.167, 4.167, 6.167, 24.167 hr on Cycle 1, Day 1 and Cycle 2, Day 1 for pemetrexed 500 mg/m^2

Interventionng/mL (Mean)
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 1)72.96
Cisplatin 75 mg/m^2/Pemetrexed Expansion (Cycle 2)93.13

[back to top]

Maximum Tolerated Dose (MTD)

The MTD was defined as the highest dose level below the maximum administered dose which caused 0 or 1 out of 6 participants to experience a DLT in that given cohort at Cycle 1 (NCT00560573)
Timeframe: Cycle 1, up to Day 21

Interventionmg/kg (Number)
Overall Population20

[back to top]

Minimum Observed Plasma Trough Concentration (Cmin) for Figitumumab

Concentration at the end of Cycle 4 (NCT00560573)
Timeframe: 0 (pre-dose) in Cycle 5 Day 1

Interventionmg/L (Mean)
Figitumumab 20 mg/kg Expansion113.6

[back to top]

Number of Participants With Dose-limiting Toxicities (DLT)

Cycle 1 figitumumab attributed: Grade (Gr) 4 neutropenia (absolute neutrophil count <500 cells/cubic millimeter [mm^3]) >=7 days, febrile neutropenia (Gr 3, fever >=38.5 degrees Celsius), neutropenic infection (Gr 3 neutropenia, infection); Gr 4 thrombocytopenia (platelet <25,000 cells/mm^3), Gr 3 thrombocytopenia >=7 days/bleeding; other Gr 3 not blood/bone marrow Common Terminology Criteria for Adverse Events bar gastrointestinal toxicity, treatment-managed hyperglycemia/fatigue, hypersensitivity; Gr 3-4 hyperglycemia despite treatment; fail to adequately recover to continue study treatment (NCT00560573)
Timeframe: Start of treatment up to end of Cycle 1, Day 21

Interventionparticipants (Number)
Figitumumab 6 mg/kg0
Figitumumab 10 mg/kg0
Figitumumab 20 mg/kg Dose Escalation1

[back to top]

Percentage of Participants With Objective Response or Prolonged Stabilization

Percentage of participants with a confirmed complete response (CR), confirmed partial response (PR), or stable disease (SD) for at least 12 weeks on study according to Response Evaluation Criteria in Solid Tumors (RECIST). Participants with non measurable disease were considered having a clinical benefit response only in the case of achievement of CR. Participants who developed early progressive disease post dosing and prior to response evaluation were considered to have progressed on study. Confirmed responses were those that persisted on repeat imaging >= 4 weeks after initial response (NCT00560573)
Timeframe: Screening, from Cycle 2 onwards computerized tomography (CT) scan done within 7-10 days prior to next cycle (approximately Day 15 of each cycle), follow-up (30 days after last study treatment dose)

InterventionPercentage of participants (Number)
Overall Population53.3
Figitumumab 20 mg/kg With Gemcitabine and Cisplatin56.5
Figitumumab 20 mg/kg Expansion With Pemetrexed46.2

[back to top]

Serum Total Circulating Insulin-like Growth Factor (IGF-1) Levels

To monitor serum total IGF-1 levels as a potential pharmacodynamic response to figitumumab treatment (NCT00560573)
Timeframe: Baseline, Day 8, end of study

Interventionng/mL (Mean)
BaselineEnd of study
Overall Population124.88543.63

[back to top]

Overall Survival

Overall survival of participants reported after 2 years. (NCT00573989)
Timeframe: 1 and 2 years

InterventionParticipants (Count of Participants)
1 year2 years
Erlotinib97

[back to top]

Median Overall Survival

Median Overall Survival of participants reported after 2 years. (NCT00573989)
Timeframe: up to 5 years

Interventionyears (Median)
Erlotinib1.01

[back to top]

Maximum Tolerated Dose of Erlotinib Hydrochloride (Phase I)

Dose at which 100% of participants tolerated the dose (NCT00573989)
Timeframe: 56 Days

Interventionmg (Number)
Erlotinib125

[back to top]

Progression-free Survival (PFS) at 1 Year (Phase II)

Determine Progression Free Survival at 1 year defined as the percentage of patients who are alive at 1 year after beginning of their concurrent re-irradiation and chemotherapy without loco-regional progression of their disease as measured by CT scan or MRI. (NCT00573989)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Erlotinib5

[back to top]

Change in Quality of Life- FACT H&N

The Functional Assessment of Cancer Therapy-Head and Neck (FACT H&N) consists of 27 core items which assess patient function in four domains: Physical, Social/Family, Emotional, and Functional well-being, which is further supplemented by 12 site specific items to assess for head and neck related symptoms. Each item is rated on a 0 to 4 Likert type scale, and then combined to produce subscale scores for each domain. Score range is 0-156. Higher scores denotes better outcomes (NCT00573989)
Timeframe: baseline and 12 months

Interventionunits on a scale (Mean)
baseline12 months
Erlotinib84.8788.61

[back to top]

Objective Tumor Response

Objective Tumor Response reported on participants at 1 year (complete, partial, progression, or stable response). (NCT00573989)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
CompletePartialProgressionStable
Erlotinib7542

[back to top]

Evaluation of Acute and Chronic Toxicity

Evaluate acute and chronic toxicity of the combined re-irradiation with radiosensitizing drugs: Pemetrexed and Erlotinib. Adverse events with Common Toxicity Criteria grades of 4 and 5 are reported for phase I and II. (NCT00573989)
Timeframe: 1 year

Interventionevents (Number)
phase Iphase II
Erlotinib186

[back to top]

Median Progression Free Survival

Median Progression Free Survival of participants reported after 2 years. (NCT00573989)
Timeframe: 2 years

Interventionyears (Median)
Erlotinib.71

[back to top]

Change in Quality of Life: MDADI

The M.D. Anderson Dysphagia Inventory (MDADI) was used to assess effects of dysphagia on the quality of life of patients with head and neck cancer. It incorporates 3 domains (emotional, functional, and physical) as well as 1 global question. Each subscale with five possible responses scored on a scale of 1 to 5 (strongly agree, agree, no opinion, disagree and strongly disagree). Scores range from 0 (extremely low functioning) to 100 (higher functioning). Higher MDADI score represents better day-to-day functioning and better quality of life. (NCT00573989)
Timeframe: baseline and 12 months

Interventionunits on a scale (Mean)
baseline- global12 months- globalbaseline- emotion12 months- emotionbaseline- function12 months- functionbaseline- physical12 months- physical
Erlotinib64.4490.070.3778.3365.7884.060.073.75

[back to top]

Change in Quality of Life: PSS-HN

The Performance Status Scale for Head & Neck Cancer Patients (PSS-HN) is s designed to evaluate performance in areas of functioning most likely affected by head and neck cancer and its treatment, specifically Normalcy of Diet, Eating in Public, and Understandability of Speech. Each subscale is rated from 0 to 100, with higher scores indicating better performance (NCT00573989)
Timeframe: baseline and 6 months

Interventionunits on a scale (Mean)
Eating BaselineEating 6 monthsSpeech BaselineSpeech 6 monthsDiet BaselineDiet 6 months
Erlotinib56.2543.7581.2575.044.4425.0

[back to top]

Number of Patients With Pathologic Complete Response Rate

Complete Response (CR): Disappearance of all clinical evidence of tumor. Partial Response (PR): A 50% or greater decrease in the sum of the products of measured lesions. No simultaneous increase in the size of any lesion or the appearance of new lesions may occur. Non-measurable lesions must remain stable or regress for this category. Minor Response (MR): A > 25% and < 50% decrease in the sum of the products of measured lesions. No simultaneous increase in the size of any lesion or the appearance of new lesions may occur. Non-measurable lesions must remain stable or regress for this category. Stable Disease (SD): A less than 25% decrease. This includes a decrease of less than 25% in the sum of the products of the measured lesions, and any increase of less than 25% in the sum of the products of the measured lesions. There may be no appearance of new disease sites for this category. Progressive Disease (PD): A ≥25% increase in one or more lesions, or appearance of new lesions. (NCT00577707)
Timeframe: Patients will undergo a CT scan of chest every 3 months for year 1 and every 4 months for year 2. In years 3 and 4, a chest CT or chest x-ray every 6 months.

Interventionparticipants (Number)
Minor Response (MR)Partial Response (PR)Stable Disease (SD)Progressive Disease (POD)
Patients With Stage IB-IIIA NSCLC With EGFR Mutations1122

[back to top]

Median Overall Survival

To determine the median overall survival for patients with recurrent or metastatic Head and Neck Squamous Cell Carcinoma treated with pemetrexed and gemcitabine. (NCT00589667)
Timeframe: 2 years

Interventionmonths (Median)
Pemetrexed Plus Gemcitabine8.8

[back to top]

Overall Objective Response

"To determine the objective radiologic response rate of pemetrexed and gemcitabine in patients with recurrent or metastatic Head and Neck Squamouse Cell Carcinoma.~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." (NCT00589667)
Timeframe: 2 years

Interventionparticipants (Number)
Partial ResponseStable DiseaseProgression of DiseaseResponse not available
Pemetrexed Plus Gemcitabine41821

[back to top]

Number of Participants With Partial Response (PR) of Target Lesions

Tumor response was assessed in 12 patients who had at least one follow-up computed tomography (CT) scan. Response Evaluation Criteria in Solid Tumors (RECIST) definition of Partial Response: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters. (NCT00604461)
Timeframe: Up to 12 Months

Interventionparticipants (Number)
Dose Escalation Followed by Maintenance Therapy4

[back to top]

Number of Months of Progression Free Survival (PFS)

The PFS is defined as the duration of time from the start of treatment to time of progression or death, whichever occurs first. (NCT00604461)
Timeframe: 2 Years, 9 Months

InterventionMonths (Median)
Dose Escalation Followed by Maintenance Therapy7.8

[back to top]

Progression Free Survival During Overall Period (Induction Phase [IP] + Maintenance Phase [MP])

Progression-free survival in overall period is defined as the time from the date of first dose of study drug during IP until the date of PD or death from any cause. PD was determined using RECIST criteria. PD is ≥20% increase in sum of longest diameter of target lesions. PD in overall period uses the screening lesion assessment prior to the induction phase as the baseline assessment. (NCT00606021)
Timeframe: First dose of study drug during IP to PD or date of death from any cause up to 33.6 months

Interventionmonths (Median)
Pemetrexed Plus Best Supportive Care (Maintenance Phase)6.2
Best Supportive Care (Maintenance Phase)6.0

[back to top]

Tumor Response Rate and Disease Control Rate After Induction Phase (IP)

Tumor response rate (%) is the number of responders (participants with best response of CR or PR) divided by the number of participants qualified for tumor response according to RECIST criteria multiplied by 100. Disease control rate is percentage of participants with a best response of stable disease [SD], PR, or CR. CR=disappearance of all target lesions; PR=30% decrease in sum of longest diameter of target lesions; PD is≥20% increase in sum of longest diameter of target lesions. SD= neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD. (NCT00606021)
Timeframe: Randomization to measured PD up to 31.4 months

,
Interventionpercentage of participants (Number)
Best Overall Response RateCRPRSDDisease Control RatePDEarly Death From Malignant DiseaseEarly Death From ToxicityEarly Death From other causeUnknown
Best Supportive Care (Maintenance Phase)00044.444.437.000018.5
Pemetrexed Plus Best Supportive Care (Maintenance Phase)00057.157.132.17.103.60

[back to top]

Number of Participants With Adverse Events (AEs) During Overall Period

The list of serious adverse events (SAEs) and other non-serious adverse events (AEs) are in Adverse Events Section. (NCT00606021)
Timeframe: First dose of study drug during IP through overall study completion (up to 34.3) months

,,
Interventionparticipants (Number)
Adverse Events (AE)Serious Adverse Events (SAE)
Best Supportive Care (Maintenance Phase)161
Pemetrexed Plus Best Supportive Care (Maintenance Phase)151
Pemetrexed Plus Cisplatin (Induction Phase)7413

[back to top]

Progression Free Survival During Maintenance Phase

Progression free survival is defined as the time from randomization until the date of progression of disease (PD) or death from any cause. PD was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. PD is ≥20% increase in sum of longest diameter of target lesions. PD in maintenance phase uses the last lesion assessment prior to randomization as the baseline assessment. (NCT00606021)
Timeframe: Randomization to progression of disease (PD) or date of death from any cause up to 30.9 months

Interventionmonths (Median)
Pemetrexed Plus Best Supportive Care (Maintenance Phase)3.2
Best Supportive Care (Maintenance Phase)3.2

[back to top]

Overall Survival During Overall Period (IP + MP)

Overall survival in overall period is defined as the time from first dose of study drug during IP to death. Participants who were alive were censored at the last contact. (NCT00606021)
Timeframe: First dose of study drug during IP to PD or date of death from any cause up to 34.1 months

Interventionmonths (Median)
Pemetrexed Plus Best Supportive Care (Maintenance Phase)15.4
Best Supportive Care (Maintenance Phase)16.4

[back to top]

Overall Survival During Maintenance Phase

Overall survival in maintenance phase is defined as the time from randomization to death. Participants who were alive were censored at the last contact. (NCT00606021)
Timeframe: Randomization to PD or date of death from any cause up to 31.3 months

Interventionmonths (Median)
Pemetrexed Plus Best Supportive Care (Maintenance Phase)12.2
Best Supportive Care (Maintenance Phase)11.8

[back to top]

Progression-free Survival (PFS)

Defined as the time from date of first dose to the first observation of disease progression, or death due to any cause. For patients who are alive and have not progressed, PFS is censored at the date of last radiological assessment. (NCT00609518)
Timeframe: Randomization (≤4 weeks from baseline visit) to 12 months after randomization

Interventionmonths (Median)
Standard Vitamin and Steroid Schedule + Pemetrexed3.7
Simplified Vitamin and Steroid Schedule + Pemetrexed3.8

[back to top] [back to top]

Overall Survival

Overall survival is the duration from randomization to death. For patients who are alive, overall survival is censored at the date of last contact. (NCT00609518)
Timeframe: Randomization (≤4 weeks from baseline visit) to 12 months after randomization

Interventionmonths (Median)
Standard Vitamin and Steroid Schedule + Pemetrexed8.2
Simplified Vitamin and Steroid Schedule + Pemetrexed9.2

[back to top]

Proportion of Participants With Best Overall Tumor Response (Response Rate)

Response defined per Response Evaluation Criteria In Solid Tumors (RECIST) criteria: Complete Response (CR)=disappearance of all target lesions; Partial Response (PR)=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. Best Overall Tumor Response is complete response plus partial response. (NCT00609518)
Timeframe: Baseline until disease progression, new therapy initiated, or death from any cause, up to 12 months after enrollment.

Interventionproportion of patients (Mean)
Standard Vitamin and Steroid Schedule + Pemetrexed0.118
Simplified Vitamin and Steroid Schedule + Pemetrexed0.064

[back to top]

Progression Free Survival

"From date of randomization until the date of first documented progression or date of death from any cause, whichever came first, assessed up to 1 year)." (NCT00614822)
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 1 year)."

Interventionweeks (Median)
Carboplatin, Bevacicumab, Premetrexed28

[back to top]

Overall Survival

Overal survival was defined as time between the date of treatment assignment and the date of death (NCT00614822)
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 1 year)."

Interventionweeks (Median)
One Arm for Study49

[back to top]

Number of Participants With Complete and Partial Tumor Responses

A complete response (CR),was disappearance of all target lesions on CT scan and absence of appearance of any new lesion was required. Partial response (PR) was assessed by at least a 30% decrease in the sum of the longest diameter (LD) of target lesions without appearance of any new lesions. Progressive disease (PD) was defined as at least a 20% increase in the sum of the LD of target lesions or the appearance of one or more new lesions. Patients were assessed to have stable disease if neither sufficient decrease to qualify for PR nor sufficient increase to qualify for PD, without appearance of new lesions. Patients who received one or more cycles were evaluable for response. (NCT00614822)
Timeframe: Patients were enrolled over a 24 month period for treatment visits. After end of treatment visits, subjects were seen or contacted every 3 months for survival data. Median follow up was 49 weeks (6 weeks to death.

InterventionParticipants (Count of Participants)
Complete ResponsePartial Response
One Arm for Study127

[back to top]

Number of Participants With Adverse Events

Measured by adverse events such as grade 4 toxicities, hospitalizations for toxicities, fever and neutropenia events, and clinically significant bleeding/thrombotic events. (NCT00614822)
Timeframe: Subjects were seen or contacted every 3 months with medain follow up of 49 weeks.

InterventionParticipants (Count of Participants)
ThrombocytopeniaRespiratory distressGrade 3 diarrheafatigue
One Arm for Study1111

[back to top]

Disease Stabilization Rate (e.g., Complete Response, Partial Response, and Stable Disease)

Estimated based on number of evaluable patients with complete response, partial response or stable disease (NCT00660816)
Timeframe: 36 months after enrollment of last patient

Interventionparticipants (Number)
Active Comparator15
Experimental16

[back to top]

Overall Survival

Measured from the date of randomization to the date of death, whichever occurs first and censored at the date of last followed for those survivors (NCT00660816)
Timeframe: 36 months after enrollment of last patient

InterventionMonths (Median)
Active Comparator16.4
Experimental14.2

[back to top]

Progression-free Survival

From the date of randomization to the date of disease progression or the date of death, whichever occurs first and censored at the date of last followed for those survivors without disease progression. (NCT00660816)
Timeframe: 18 months after enrollment of last patient

InterventionMonths (Median)
Active Comparator5.5
Experimental4.4

[back to top]

Response Rate

Estimated based on the number of responses by excluding the dropouts who are not evaluable for response using a binomial distribution (NCT00660816)
Timeframe: 36 months after enrollment of last evaluable patient

Interventionparticipants (Number)
Active Comparator2
Experimental3

[back to top]

Survival Rates at 1, 2, and 3 Years

The probability that survival time is at least 1, 2, or 3 years was summarized using Kaplan-Meier estimates. (NCT00686959)
Timeframe: Baseline to Date of Death from Any Cause (Up to 71.4 Months)

,
Interventionprobability of survival (Number)
1 year (12 months)2 years (24 months)3 years (36 months)
Arm A: Pemetrexed + Cisplatin and TRT0.760.520.40
Arm B: Etoposide + Cisplatin and TRT0.770.520.37

[back to top]

First Site of Disease Failure in Terms of Relapse

The percentage of participants with first sites of disease failure in terms of relapse within the radiation treatment field, inside the thorax, (outside of the radiation field), or distant disease are presented. Results were summarized using Kaplan-Meier estimates. Some participants relapsed in more than 1 location/site and appear in more than a single category. (NCT00686959)
Timeframe: Baseline to Relapse (Up to 66.6 Months)

,
Interventionpercentage of participants (Number)
Relapsed within the radiation treatment fieldRelapsed inside thorax, outside of radiation fieldRelapsed distant disease
Arm A: Pemetrexed + Cisplatin and TRT37.320.550.0
Arm B: Etoposide + Cisplatin and TRT45.816.345.8

[back to top]

Adverse Events: The Number of Deaths Per Treatment Group

The number of deaths that occurred while on study drug, the number of deaths due to adverse events (AEs) while on study drug, and the number of deaths due to the study disease (that is, disease progression) while on study drug are presented. In addition, the number of deaths within 30 days of treatment discontinuation, the number of deaths due to AEs within 30 days of treatment discontinuation, and the number of deaths due to study disease within 30 days of treatment discontinuation are presented. For both the deaths due to AEs that occurred on study and for deaths due to AEs that occurred within 30 days of treatment discontinuation, the causality (events assess as possibly related [poss related] to study drug per investigator judgement) is also presented. A summary of serious and other non-serious adverse events regardless of causality is located in the Reported Adverse Events module. (NCT00686959)
Timeframe: Baseline through 30 Days Post Study

,
Interventionparticipants (Number)
On study drug (total)On study drug: Due to AEOn study drug: Due to AE poss relatedOn study drug: Due to study diseaseWithin 30 Days of Discontinuation (disc) (total)Within 30 Days of Disc: Due to AEWithin 30 Days of Disc: Due to AE poss relatedWithin 30 Days of Disc: Due to study disease
Arm A: Pemetrexed + Cisplatin and TRT12105210525
Arm B: Etoposide + Cisplatin and TRT64306402

[back to top]

Progression-free Survival (PFS)

Progression-free survival (PFS) time is from baseline to the first date of documented objective progressive disease (PD) or death from any cause. For participants who were not known to have died or to have had objective PD as of the data inclusion cut-off date for a particular analysis, PFS was censored at the date of the last objective progression-free disease assessments. For participants who took any subsequent systemic anticancer therapy prior to progression or death, PFS was censored at the date of the last objective progression-free disease assessment prior to the start date of any subsequent systemic anticancer therapy. PFS time was summarized using Kaplan-Meier estimates. (NCT00686959)
Timeframe: Baseline to Measured Progressive Disease or Death from Any Cause (Up to 66.6 Months)

Interventionmonths (Median)
Arm A: Pemetrexed + Cisplatin and TRT11.37
Arm B: Etoposide + Cisplatin and TRT9.76

[back to top]

Overall Survival

Overall survival (OS) time is from baseline to the date of death from any cause. For participants not known to have died as of the data cut-off date, OS time was censored at the last contact date the participant was known to be alive prior to the data cut-off date. OS was summarized using Kaplan-Meier estimates. (NCT00686959)
Timeframe: Baseline to Date of Death from Any Cause (Up to 71.4 Months)

Interventionmonths (Median)
Arm A: Pemetrexed + Cisplatin and TRT26.81
Arm B: Etoposide + Cisplatin and TRT24.97

[back to top]

Objective Response Rate (Complete Response [CR] + Partial Response [PR])

Overall response rate (ORR) is the best response of CR or PR as classified by the investigators according to the Response Evaluation Criteria in Solid Tumors (RECIST, v1.1) guidelines. CR is defined as the disappearance of all target and non-target lesions, normalization of tumor marker level of non-target lesions, and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeter (mm). PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of non-target lesions or appearance of new lesions. Overall response rate is calculated as a total number of participants with CR or PR divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT00686959)
Timeframe: Baseline to Measured Progressive Disease (Up to 7 Months)

Interventionpercentage of participants (Number)
Arm A: Pemetrexed + Cisplatin and TRT35.9
Arm B: Etoposide + Cisplatin and TRT33.0

[back to top]

Percentage of Participants With a Post Baseline Swallowing Diary Score >=4

Participants were provided with a swallowing diary to record issues with swallowing using a 5-point categorical scale: (1) no problems; (2) mild soreness; (3) swallowing solids with some difficulty; (4) inability to swallow solids; and (5) inability to swallow liquids. Participants rated swallowing over the previous 24 hours. The percentage of participants was calculated by dividing the number of with a post baseline swallowing diary score >=4 by total number of participants analyzed, multiplied by 100. No adjustments were made for the number of available assessments nor were any interpolation of missing assessments made. (NCT00686959)
Timeframe: Baseline through 30 Days Post Study

Interventionpercentage of participants (Number)
Arm A: Pemetrexed + Cisplatin and TRT33.8
Arm B: Etoposide + Cisplatin and TRT29.0

[back to top]

Progression-free Survival

Duration of progression-free survival in months. (NCT00691301)
Timeframe: From enrollment onto the study until the onset of disease progression or death, up to 5 years

Interventionmonths (Median)
Pemetrexed and Cisplatin5.6

[back to top]

Duration of Overall Survival

Overall survival is defined as the duration of time from study entry to time of death or the date of last contact. (NCT00691301)
Timeframe: Every cycle during treatment, then every 3 months for the first 2 years, then every six months for the next three years and then annually, up to 5 years.

Interventionmonths (Median)
Pemetrexed and Cisplatin12.3

[back to top]

Frequency and Severity of Observed Adverse Effects

All eligible and evaluable patients (NCT00691301)
Timeframe: every 21 days during study treatment and up to 30 days after the last cycle of treatment.

,,,,,
InterventionParticipants (Count of Participants)
LeukopeniaThrombocytopeniaNeutropeniaAnemiaOther hematologicAllergy/immunologyAuditory/earCardiacCoagulationConstitutionalDermatologicEndocrineNauseaVomitingGastrointestinalGenitourinary/renalHemorrhageInfectionLymphaticsMetabolicMusculoskeletalNeurosensoryOther neurologicalOcular/visualPainPulmonarySexual/reproductiveSyndromesVascular
Grade 0163220249493947536245371944344374210503142441837535250
Grade 1 (CTCAE v 3.0)811517030501220119111567051931586119110
Grade 2 (CTCAE v 3.0)1551022301420239022172440117100632136013
Grade 3 (CTCAE v 3.0)12312611101121067101260101212122001
Grade 4 (CTCAE v 3.0)33771100010000101005000000000
Grade 5 (CTCAE v 3.0)00000000000000000000000000000

[back to top]

Patients With Objective Tumor Response Rate (Complete Response [CR] or Partial Response [PR]) Using RECIST Version 1.0

RECIST 1.0 defines complete response as the disappearance of all target lesions and non-target lesions and no evidence of new lesions documented by two disease assessments at least 4 weeks apart. Partial response is defined as at least a 30% decrease in the sum of longest dimensions (LD) of all target measurable lesions taking as reference the baseline sum of LD. There can be no unequivocal progression of non-target lesions and no new lesions. Documentation by two disease assessments at least 4 weeks apart is required. In the case where the ONLY target lesion is a solitary pelvic mass measured by physical exam, which is not radiographically measurable, a 50% decrease in the LD is required. These patients will have their response classified according to the definitions stated above. Complete and partial responses are included in the objective tumor response rate. (NCT00691301)
Timeframe: CT scan or MRI if used to follow lesion for measurable disease every other cycle until disease progression or study withdrawal; and at any other time if clinically indicated, up to 5 years.

Interventionparticipants (Number)
Complete ResponsePartial Response
Pemetrexed and Cisplatin116

[back to top]

18 Week Progression-free Survival (PFS) Rate

The 18 week progression-free survival rate was defined as the proportion of patients that were alive and progression-free 18 weeks after registration into the study. Disease progression was assessed per modified RECIST criteria, and defined as at least a 20% increase in the sum of the longest diameters of target lesions, in either primary or nodal lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, or the appearance of new lesions. Kaplan-Meier estimate of 18-week progression-free survival was calculated. (NCT00698815)
Timeframe: At 18 weeks

Interventionpercentage of participants (Number)
Arm I (Pemetrexed)54
Arm II (Sunitinib)37
Arm III (Pemetrexed and Sunitinib)48

[back to top]

Overall Response Rate

"The proportion of patients who respond (completely or partially) to each combination regimen will be estimated. An exact binomial confidence interval will be computed for these estimates.~Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria: Complete Response (CR): disappearance of all target lesions; Partial Response (PR) 30% decrease in sum of longest diameter of target lesions." (NCT00698815)
Timeframe: Duration of treatment (up to 3 years)

Interventionpercentage of participants (Number)
Arm I (Pemetrexed)14
Arm II (Sunitinib)17
Arm III (Pemetrexed and Sunitinib)22

[back to top]

PFS

PFS was defined as the time from randomization until disease progression or death, whichever occurs first. The median PFS with 95% CI was estimated using the Kaplan-Meier method. Progression is defined as in the primary outcome measure. (NCT00698815)
Timeframe: Time from randomization to disease progression and death of any cause, whichever comes first (up to 3 years)

Interventionmonths (Median)
Arm I (Pemetrexed)4.9
Arm II (Sunitinib)3.3
Arm III (Pemetrexed and Sunitinib)3.7

[back to top]

Overall Survival (OS)

OS is defined as the time from patient randomization to death from any cause. The median OS with 95% CI was estimated using the Kaplan-Meier method. (NCT00698815)
Timeframe: Time from randomization to death (up to 3 years)

Interventionmonths (Median)
Arm I (Pemetrexed)10.5
Arm II (Sunitinib)8.0
Arm III (Pemetrexed and Sunitinib)6.7

[back to top]

4M PFS Rate of Patients With Previously Untreated, Unresectable Malignant Pleural Mesothelioma (MPM) Treated With CBP501, Pemetrexed and Cisplatin

Planned: Forty-two patients were to be treated in Arm A. If ≥ 23 patients (>54%) were free of progression and death at 4 months, then the study regimen would be considered for further evaluation in this indication. (NCT00700336)
Timeframe: End of study

,
Interventionparticipants (Number)
4M PFS by Independent Image Review4M PFS by Site assess
Pemetrexed and Cisplatin913
Pemetrexed, Cisplatin, and CBP5012527

[back to top]

Maximum-tolerated Dose of Pemetrexed With a Day 2 i.p. Cisplatin (75 mg/m2) and Day 8 i.p. Paclitaxel (60 mg/m2)

If none of the initial 3 patients on a dose level experienced a dose-limiting toxicity (DLT) after the first cycle of therapy, then the dose was escalated to the next level. If 2 or more patients on any dose level experienced a DLT, then the maximum tolerated dose would be determined to be the next lower dose level. (NCT00702299)
Timeframe: 18 months

Interventionmg/m2 (Number)
Receiving Treatment500

[back to top]

Overall Survival

(NCT00702299)
Timeframe: Average Length of follow-up 788 days

InterventionDays (Median)
Receiving Treatment680

[back to top]

Pharmacokinetics (Mean Cmax, ug/mL)for Different Dosages of Pemetrexed

Cmax levels were found through plasma collected between 0.5 to 4 hours and at 24 hours after initiation of intraperitoneal administration (NCT00702299)
Timeframe: 18 months

Interventionug/mL (Mean)
Pemetrexed Dose 500mg/m225.1
Pemetrexed Dose 750 mg/m239.3
Pemetrexed Dose 1,000mg/m238.7

[back to top]

Patients That Completed at Least 6 Courses of Therapy of Pemetrexed Along With Day 2 i.p. Cisplatin (75 mg/m2) and Day 8 i.p. Paclitaxel (60 mg/m2)at the Determined Maximum Tolerated Dose

If none of the initial 3 patients on a dose level experienced a dose-limiting toxicity (DLT) after the first cycle of therapy, then the dose was escalated to the next level. If 2 or more patients on any dose level experienced a DLT, then the Maximum Tolerance Dose (MTD) would be determined to be the next lower dose level. (NCT00702299)
Timeframe: 18 months

Intervention% of participants (Number)
Receiving Treatment80.0

[back to top]

Patients Experienced Grade >=3 Toxicity at Dose Level 5 (1,000 mg/m2 IP Pemetrexed)

Toxicity was assessed by NCI Common Toxicity Criteria for Adverse Effects v3.0 (NCT00702299)
Timeframe: 18 months

Interventionparticipants (Number)
Receiving Treatment2

[back to top]

Progression-free Survival at 18 Months as Assessed by Cancer Antigen 125

Progression was evaluated with posttreatment CT scans and measured changes in cancer antigen 125 levels 6 months after the initiation of the treatment regimen, or within one month after discontinuation of treatment if stopped early. Cancer antigen 125 response in evaluable patients (N=13) was analyzed using the modified Gynecologic Cancer Intergroup (GCIG) criteria. There was one evaluable patient by Response Evaluation Criteria in Solid Tumors(RECIST) criteria (NCT00702299)
Timeframe: 18 months

Intervention% of participants (Number)
Receiving Treatment78.6

[back to top]

2-year Progression-free Survival (PFS)

Two-year PFS is an estimated percentage of participants without disease progression (locoregional or distant) at two years after the start of study treatment. Progression was defined using Response Evaluation Criteria In Solid Tumors (RECIST v1.0), as: at least a 20% (and at least 5 millimeters) increase in the sum of the diameters of target lesions, or the appearance of one or more new lesions. (NCT00703976)
Timeframe: 18 months to patient accrual and 2 years of follow-up after closing accrual.

Interventionpercentage of participants (Number)
Cetuximab, Pemetrexed and Radiation Therapy79
Cetuximab, Pemetrexed, Radiation Therapy Plus Bevacizumab75

[back to top]

2-year Overall Survival (OS)

Two-year OS is an estimated percentage of participants still living at two years after the start of study treatment. (NCT00703976)
Timeframe: 2 years of follow-up after closing accrual

Interventionpercentage of participants (Number)
All Participants (Overall Study)88
Cetuximab, Pemetrexed and Radiation Therapy91
Cetuximab, Pemetrexed, Radiation Therapy Plus Bevacizumab87

[back to top]

Assess Safety and Toxicity

- To determine the toxicities of pemetrexed and concurrent definitive radiation in patients with poor risk stage III NSCLC. (NCT00732303)
Timeframe: 24 months

Interventionparticipants (Number)
Grade 1 and Grade 2 FatigueGrade 1 and 2 AnorexiaGrade 1 and 2 CoughGrade 3 and 4 FatigueGrade 3 and 4 AnorexiaGrade 3 and 4 Neutropenia
Single Arm Assignment766433

[back to top]

Maximum Concentration of Pemetrexed Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1

(NCT00732992)
Timeframe: Cycle 2 Day 1: Pre-dose, 10 minutes after the start of infusion (immediately before the end of infusion), and 1, 2, 4, 6, 8, 10, and 24 hours post-dose

Interventionmicrogram/mL (Mean)
Sunitinib 37.5 mg/Day Continuous Daily Dosing163

[back to top]

Terminal Phase Elimination Half-Life (T1/2) of Pemetrexed Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1

"Terminal phase elimination half-life was calculated as natural logarithm of 2 (ln2) divided by the rate constant for terminal phase (kel)." (NCT00732992)
Timeframe: Cycle 2 Day 1: Pre-dose, 10 minutes after the start of infusion (immediately before the end of infusion), and 1, 2, 4, 6, 8, 10, and 24 hours post-dose

Interventionhours (Mean)
Sunitinib 37.5 mg/Day Continuous Daily Dosing2.754

[back to top]

"Sunitinib Relative Dose Intensity in the Sunitinib 37.5 mg/Day Continuous Daily Dosing Treatment Arm"

Relative dose intensity was defined as percentage of total dose administered over total planned dose in the given period. (NCT00732992)
Timeframe: Up to Cycle 5 (end of study)

Interventionpercent of total planned dose (Median)
Cycle 1 (n=6)Cycle 2 (n=6)Cycle 3 (n=4)Cycle 4 (n=4)Cycle 5 (n=1)
Sunitinib 37.5 mg/Day Continuous Daily Dosing92.054.846.055.644.4

[back to top]

AUC 0-24 of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1

AUC0-24 = Area under the plasma concentration versus time curve to 24 hours post dose was calculated using the linear/logarithmic trapezoidal method. SU012662 is an active metabolite of sunitinib. (NCT00732992)
Timeframe: Cycle 2 Day 1: Pre-dose and 2, 4, 6, 8, 10, and 24 hours post-dose

Interventionnanogram*hour/mL (Mean)
SunitinibSU012662Total Drug
Sunitinib 37.5 mg/Day Continuous Daily Dosing11906751866

[back to top]

"Sunitinib Relative Dose Intensity in the Sunitinib 50 mg/Day Schedule-2/1 Treatment Arm"

Relative dose intensity was defined as percentage of total dose administered over total planned dose in the given period. (NCT00732992)
Timeframe: Up to Cycle 6

Interventionpercent of total planned dose (Median)
Cycle 1 (n=6)Cycle 2 (n=5)Cycle 3 (n=4)Cycle 4 (n=3)Cycle 5 (n=3)Cycle 6 (n=3)
Sunitinib 50 mg/Day Schedule-2/187.580.453.650.050.050.0

[back to top]

Number of Participants With Adverse Events

Number of participants with any adverse events, adverse events graded as Common Terminology Criteria for Adverse Events Version 3.0 (CTCAE) Grade 3 or higher , dose limiting toxicities (DLT), serious adverse events, adverse events resulted in discontinuation. (NCT00732992)
Timeframe: End of study (up to individual discontinuation)

,
InterventionParticipants (Number)
Any adverse eventsAny dose limiting toxicitiesAny serious adverse eventsAny Grade-3 or -4 adverse eventsAny Grade-5 adverse events (= death)Discontinuation due to adverse events
Sunitinib 37.5 mg/Day Continuous Daily Dosing602601
Sunitinib 50 mg/Day Schedule-2/1601301

[back to top]

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

Complete response (CR): disappearance of all target lesions; Partial response (PR): >=30% decrease in the sum of the longest dimensions (SLD) of the target lesions taking as a reference the baseline SLD; Progressive disease (PD): >=20% increase in the SLD of the target lesions taking as a reference the smallest SLD recorded since the treatment started, or the appearance of >=1 new lesions; Stable disease (SD): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD taking as a reference the smallest SLD since the treatment started. (NCT00732992)
Timeframe: End of study (Up to individual study discontinuation)

,
InterventionParticipants (Number)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)Not Evaluable (NE)Objective Response (CR+PR)Stable Disease (SD) >=184 daysClinical Benefit (CR+PR+SD >=184 days)
Sunitinib 37.5 mg/Day Continuous Daily Dosing01210101
Sunitinib 50 mg/Day Schedule-2/100310011

[back to top]

Tmax of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1

Tmax = Time to maximum plasma concentration. SU012662 is an active metabolite of sunitinib. (NCT00732992)
Timeframe: Cycle 2 Day 1: Pre-dose and 2, 4, 6, 8, 10, and 24 hours post-dose

Interventionhours (Median)
SunitinibSU012662Total Drug
Sunitinib 37.5 mg/Day Continuous Daily Dosing444

[back to top]

Trough and Maximum Concentration of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1

Trough concentration was defined as observed concentration at 24 hours post dose. SU012662 is an active metabolite of sunitinib. (NCT00732992)
Timeframe: Cycle 2 Day 1: Pre-dose and 2, 4, 6, 8, 10, and 24 hours post-dose

Interventionnanogram/mL (Mean)
Sunitinib: Trough concentrationSunitinib: Maximum concentrationSU012662: Trough concentrationSU012662: Maximum concentrationTotal drug: Trough concentrationTotal drug: Maximum concentration
Sunitinib 37.5 mg/Day Continuous Daily Dosing45.659.925.131.670.691.5

[back to top]

Trough Concentrations of Sunitinib, SU012662, and Total Drug (Sunitinib + SU012662) After Coadministration of Sunitinib 50 mg/Day and Pemetrexed 500 mg/m^2 (Cycle 1 Day 1), Followed by Sunitinib 50 mg/Day on Schedule-2/1 at Cycle 1 Day 14 or 15

Trough concentration was defined as observed concentration at 24 hours post dose. SU012662 is an active metabolite of sunitinib. (NCT00732992)
Timeframe: Cycle 1 Day 14 (or 15): approximately 24 hours after the previous dose

Interventionnanogram/mL (Mean)
SunitinibSU012662Total Drug
Sunitinib 50 mg/Day Schedule-2/178.538.2117

[back to top]

AUC0-∞ of Pemetrexed Following Continuous Daily Dosing of Sunitinib 37.5 mg/Day in Combination With Pemetrexed 500 mg/m^2 at Cycle 2 Day 1

AUC0-∞ = Area under the plasma concentration versus time curve from zero time to infinity was calculated as the sum of AUClast and (Ct*/kel), where Ct* was the estimated concentration at the time of the last quantifiable concentration, kel was terminal phase rate constant that is estimated as the absolute value of the slope of a linear regression during the terminal phase of the natural-logarithm (ln) transformed concentration-time profile. (NCT00732992)
Timeframe: Cycle 2 Day 1: Pre-dose, 10 minutes after the start of infusion (immediately before the end of infusion), and 1, 2, 4, 6, 8, 10, and 24 hours post-dose

Interventionmicrogram*hour/mL (Mean)
Sunitinib 37.5 mg/Day Continuous Daily Dosing191

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs) as a Measure of Safety and Tolerability of Amatuximab

An adverse event (AE) was any untoward medical occurrence (example; any unfavorable and unintended sign including abnormal laboratory findings, symptom or disease) in a participant or clinical investigation participant after providing written informed consent for participation in the study until the end of study visit. TEAEs were defined as an AE that developed or worsened in severity during the on-treatment period (from first dose of amatuximab to 30 days after last dose of amatuximab). SAE was defined as any adverse event (appearance of [or worsening of any pre-existing]) undesirable sign, symptom or medical conditions which is fatal or life-threatening or results in persistent or significant disability/incapacity or constitutes a congenital anomaly/birth defect or requires inpatient hospitalization or prolongation of existing hospitalization or is medically significant. (NCT00738582)
Timeframe: From date of first dose of study drug up to 30 days after the last dose of study treatment, up to approximately 5 years

,
InterventionParticipants (Count of Participants)
Participants with at least 1 TEAEParticipants with at least 1 SAE
Amatuximab5215
Amatuximab/Pemetrexed/Cisplatin8938

[back to top]

Number of Participants With Progression Free Survival (PFS) Responders and Non-responders at Month 6

"Number of participants with PFS responders and non-responders at Month 6 was reported. PFS was defined as the time from the date of the first dose of amatuximab to the date of disease progression or death due to any cause, as determined by independent radiologist based on the modified Response Evaluation Criteria in Solid Tumors (RECIST) utilizing the total tumor measurement (performed by computerized tomography (CT)/magnetic resonance imaging (MRI)) which includes the pleural unidimensional measure plus the total of the target lesion(s) measurement. A response, in terms of PFS, was defined to be at least a 6-month stabilization of disease. Progressive disease (PD) as measured by Modified RECIST was defined as an increase of at least 20 percent (%) in the total tumor measurement over the nadir measurement, or the appearance of one or more new lesions." (NCT00738582)
Timeframe: Month 6

InterventionParticipants (Count of Participants)
PFS RespondersPFS non-responders
All Participants2651

[back to top]

Time to Tumor Response (TTR)

TTR was derived for those participants with objective evidence of CR or PR. TTR was defined as the time from the date of the first dose of amatuximab to first documentation of objective tumor response. CR was defined as the disappearance of all target lesions with no evidence of tumor elsewhere, and PR was defined as at least a 30% reduction in the total tumor measurement. (NCT00738582)
Timeframe: From the date of the first dose to first documentation of objective response, up to approximately 5 years

InterventionMonths (Median)
All Participants2.3

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of the first dose of amatuximab to the date of death. (NCT00738582)
Timeframe: From the date of first dose to the date of death, up to approximately 5 years

InterventionMonths (Median)
All Participants14.8

[back to top]

Overall Response Rate (ORR)

ORR, defined as the percentage of participants with objective evidence of complete response (CR) or partial response (PR) as determined by independent radiologist based on the modified RECIST utilizing the total tumor measurement (performed by CT/ MRI) which includes the pleural unidimensional measure plus the total of the target lesion(s) measurement. Tumor assessments performed up to the initiation of further anticancer therapy were considered. CR was defined as the disappearance of all target lesions with no evidence of tumor elsewhere, and PR was defined as at least a 30% reduction in the total tumor measurement. A confirmed response required a repeat observation on two occasions 4 weeks apart. ORR = CR + PR. (NCT00738582)
Timeframe: From the date of first dose until evidence of CR or PR, up to approximately 5 years

InterventionPercentage of participants (Number)
All Participants34.5

[back to top]

Duration of Response (DR)

DR was derived for those participants who achieved a PFS Response and had an objective evidence of CR or PR. DR was defined as the time (in months) from first documentation of objective response (CR or PR) to the first documentation of disease progression or death [as determined by independent radiologist based on the modified RECIST utilizing the total tumor measurement (performed by CT/ MRI) which includes the pleural unidimensional measure plus the total of the target lesion(s) measurement]. Tumor assessments performed up to the initiation of further anticancer therapy were considered. CR was defined as the disappearance of all target lesions with no evidence of tumor elsewhere, and PR was defined as at least a 30% reduction in the total tumor measurement. PD as measured by Modified RECIST was defined as an increase of at least 20% in the total tumor measurement over the nadir measurement, or the appearance of one or more new lesions. (NCT00738582)
Timeframe: From the first documentation of objective response (CR or PR) to the first documentation of disease progression, up to approximately 5 years

InterventionMonths (Median)
All Participants9.2

[back to top]

Overall Progression Free Survival

Overall PFS was defined as the time from the date of first dose of amatuximab to the date of disease progression or death due to any cause. In the absence of confirmation of death, the survival time was censored at the date of the last follow-up contact. Tumor assessments performed up to the initiation of further anticancer therapy were considered. PD as measured by Modified RECIST was defined as an increase of at least 20% in the total tumor measurement over the nadir measurement, or the appearance of one or more new lesions. (NCT00738582)
Timeframe: From the date of first dose of amatuximab to the date of disease progression, up to approximately 5 years

InterventionMonths (Median)
All Participants6.3

[back to top]

Progression-free Survival (PFS)

Estimated using the method of Kaplan-Meier survival curves to compare PFS between the erlotinib and pemetrexed arms using an intent-to-treat (ITT) analysis. Due to the small sample size (21 of the required 954 patients ~2%), analyses within the FISH(+) and FISH(-) groups were not performed, and no formal analyses for the primary or the secondary efficacy outcomes were performed. (NCT00738881)
Timeframe: Time from randomization to the first date of documented disease progression or death, assessed up to 5 years

Interventionmonths (Median)
Arm I2
Arm II3.1

[back to top]

Time to Death

Median time (in days) from randomisation until death using the Kaplan-Meier method (Calculator for survival probability) (NCT00745875)
Timeframe: Patients were followed up for survival every week for the first 3 weeks then every 3 weeks whilst on study medication until the data cut-off (17th January 2010).

InterventionDays (Median)
Placebo + Pemetrexed193
ZD4054 + Pemetrexed146

[back to top]

Progression-free Survival

Median time (in days) from randomisation until disease progression/death using the Kaplan-Meier method (NCT00745875)
Timeframe: Tumour assessments for progression were performed at screening, every 3 weeks, Mandatory Tumour Assessment Visit (19 August 2009 ± 3 days), treatment discontinuation

InterventionDays (Median)
Placebo + Pemetrexed87
ZD4054 + Pemetrexed110

[back to top]

Time to Progressive Disease

Time to progressive disease was defined as the time from randomization to the first date of objective disease progression. Participants were censored at date of last PFS assessment prior to the cutoff date or the date of initiation of subsequent systemic anticancer therapy, whichever was earlier. (NCT00762034)
Timeframe: Baseline to measured progressive disease (up to 37.06 months)

Interventionmonths (Median)
Pem/Carbo/Bev7.03
Pac/Carbo/Bev6.04

[back to top]

Translational Research: Overall Survival (OS) Based on Nuclear Thyroid Transcription Factor-1 (TTF-1) Expression Regardless of Study Treatment

Nuclear Thyroid Transcription Factor-1 (TTF-1) expression was measured using an Immunohistochemistry (IHC) assay which were scored using a 0 (negative, no staining) to 3+ (brightest staining) scoring system, and H score is a calculated using formula: 1x(percentage of cells stained 1+) + 2x(percentage of cells stained 2+) + 3x(percentage of cells stained 3+). TTF-1 Positive have an H score >0 and TTF-1 Negative have an H score=0. Overall survival (OS) is the duration from date of randomization to date of death from any cause. Participants were censored at the date they were last known to be alive. (NCT00762034)
Timeframe: Baseline to date of death from any cause (up to 37.06 months)

Interventionmonths (Median)
TTF-1 Positive (H Score > 0)14.9
TTF-1 Negative (H Score = 0)8.7

[back to top]

Change From Baseline in Participant Reported Outcomes as Assessed by the Functional Assessment of Cancer Therapy - Lung (FACT-L)

"FACT-L is a valid instrument used to measure quality of life (QOL) in participants with cancer consisting of the 27-item FACT-General (G) and 9-item lung cancer subscale (LCS). FACT-G is organized into subscales: physical well-being (PWB)-7 items; social/family well-being (SWB)-7 items; emotional well-being (EWB)-6 items; functional well-being (FWB)-7 items. Each item uses a 5 point rating scale (0=not at all and 4=equals very much). FACT-L Total Score=4 subscales + LCS and ranges from 0 to 144. Trial Outcome Index-Lung (TOI-L)=PWB+FWB+LCS and ranges from 0 to 92. Higher scores indicate better QOL. Least squares mean (LSmean) change is calculated using the linear-mixed model (LMM) analysis controlled for treatment, baseline value, time point and treatment by time point interaction." (NCT00762034)
Timeframe: Baseline, up to first 10 cycles (4 induction and 6 maintenance cycles, cycle=21 days)

,
Interventionunits on a scale (Least Squares Mean)
FACT-L Total Score (n=397, 392)Trial Outcome Index-Lung (TOI-L) (n=396, 394)
Pac/Carbo/Bev1.66-0.40
Pem/Carbo/Bev1.88-0.38

[back to top]

Change From Baseline in Participant Reported Outcomes as Assessed by the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group- Neurotoxicity (FACT/GOG-Ntx)

"FACT/GOG-Ntx is a validated instrument used to measure quality of life (QOL) in participants with cancer and neurotoxicity (Ntx) consisting of 27-item FACT-General (G) and 11-item Ntx subscale. FACT-G is organized into domain subscales: physical well-being (PWB)-7 items; social/family well-being (SWB)-7 items; emotional well-being (EWB)-6 items; functional well-being (FWB)-7 items; each uses a 5 point rating scale (0=not at all and 4=equals very much). FACT/GOG-Ntx Total Score=sum 5 subscales and ranges from 0-152. Ntx Trial Outcome Index (TOI-Ntx)=PWB+FWB+NTX and range from 0-100. For all FACT scales, higher scores indicate better QOL. Least squares mean (LSmean) change is calculated using the linear-mixed model (LMM) analysis controlled for treatment, baseline value, time point and treatment by time point interaction." (NCT00762034)
Timeframe: Baseline, up to first 10 cycles (4 induction and 6 maintenance cycles, cycle=21 days)

,
Interventionunits on a scale (Least Squares Mean)
FACT/GOG-Ntx Total Score (n=393, 389)Ntx Trial Outcome Index (TOI-Ntx)(n=393, 390)
Pac/Carbo/Bev-5.48-7.60
Pem/Carbo/Bev-0.60-2.79

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Time Curve From Zero to Infinity (AUC(0-∞)) for Total (Bound and Unbound) Platinum and Unbound Platinum

Platinum is a metabolite of Carboplatin (Carbo) and is found in the blood as both a bound and unbound form. (NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.25, 0.5, 0.67, 1.42, 2.17, 4, 6, 8, 24, 48, and 72 hours post-dose)

,
Interventionmicrogram*hour per milliliter (μg•hr/mL) (Geometric Mean)
Total (Bound and Unbound)Unbound (n=17, 13)
Pac/Carbo/Bev18262.9
Pem/Carbo/Bev16055.7

[back to top]

Pharmacokinetics (PK): Elimination Half-life (t1/2) for Total (Bound and Unbound) Platinum and Unbound Platinum

Platinum is a metabolite of Carboplatin (Carbo) and is found in the blood as both a bound and unbound form. (NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.25, 0.5, 0.67, 1.42, 2.17, 4, 6, 8, 24, 48, and 72 hours post-dose)

,
Interventionhours (hr) (Geometric Mean)
Total (Bound and Unbound)Unbound (n=17, 13)
Pac/Carbo/Bev86.41.95
Pem/Carbo/Bev65.62.03

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) for Total (Bound and Unbound) Platinum and Unbound Platinum

Platinum is a metabolite of Carboplatin (Carbo) and is found in the blood as both a bound and unbound form. (NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.25, 0.5, 0.67, 1.42, 2.17, 4, 6, 8, 24, 48, and 72 hours post-dose)

,
Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Total (Bound and Unbound)Unbound (n=18, 15)
Pac/Carbo/Bev17.817.1
Pem/Carbo/Bev18.421.1

[back to top]

Pharmacokinetics (PK): Platinum Clearance (CL) for Total (Bound and Unbound) and Unbound Forms

Platinum is a metabolite of Carboplatin (Carbo) and is found in the blood as both a bound and unbound form. (NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.25, 0.5, 0.67, 1.42, 2.17, 4, 6, 8, 24, 48, and 72 hours post-dose)

,
Interventionliters per hour (L/hr) (Geometric Mean)
Total (Bound and Unbound)Unbound (n=17, 13)
Pac/Carbo/Bev1.875.36
Pem/Carbo/Bev2.025.81

[back to top]

Translational Research: Number of Participants With Epidermal Growth Factor Receptor (EGFR) Mutations

Epidermal Growth Factor Receptor (EGFR) mutations were measured by polymerase chain reaction (PCR). (NCT00762034)
Timeframe: Baseline

Interventionparticipants (Number)
EGFR mutation positiveEGFR mutation negative
Pem or Pac Plus Carbo/Bev11121

[back to top]

Translational Research: Overall Survival (OS) Based on Cytoplasmic and Membrane Folate Receptor Alpha (FR-α) Expression

Cytoplasmic and membrane Folate Receptor Alpha (FR-α) expression was measured using an Immunohistochemistry (IHC) assay which were scored using a 0 (negative, no staining) to 3+ (brightest staining) scoring system for cytoplasmic or membrane staining, and H score is a calculated using formula: 1x(percentage of cells stained 1+) + 2x(percentage of cells stained 2+) + 3x(percentage of cells stained 3+). FR-α Positive have an H score >0 and FR-α Negative have an H score=0. Overall survival (OS) is the duration from date of randomization to date of death from any cause. Participants were censored at the date they were last known to be alive. (NCT00762034)
Timeframe: Baseline to date of death from any cause (up to 37.06 months)

,
Interventionmonths (Median)
FR-α Cytoplasm Positive (H score > 0; n=64, 53)FR-α Cytoplasm Negative (H score = 0; n=34, 29)FR-α Membrane Positive (H score > 0; n=39, 22)FR-α Membrane Negative (H score = 0; n=59, 60)
Pac/Carbo/Bev14.311.215.511.3
Pem/Carbo/Bev14.412.019.212.9

[back to top]

Translational Research: Overall Survival (OS) Based on Cytoplasmic and Nuclear Thymidylate Synthase (TS) Expression

Cytoplasmic and nuclear Thymidylate Synthase (TS) expression was measured using an Immunohistochemistry (IHC) assay which were scored using a 0 (negative, no staining) to 3+ (brightest staining) scoring system for cytoplasmic and nuclear staining, and H score was calculated using formula: 1x(percentage of cells stained 1+) + 2x(percentage of cells stained 2+) + 3x(percentage of cells stained 3+). TS Positive have an H score >0 and TS Negative have an H score=0. Overall survival (OS) is the duration from date of randomization to date of death from any cause. Participants were censored at the date they were last known to be alive. (NCT00762034)
Timeframe: Baseline to date of death from any cause (up to 37.06 months)

,
Interventionmonths (Median)
TS Cytoplasm Positive (H score > 0; n=90, 83)TS Cytoplasm Negative (H score = 0; n=10, 6)TS Nucleus Positive (H score > 0; n=68, 51)TS Nucleus Negative (H score = 0; n=32, 38)
Pac/Carbo/Bev12.411.612.412.4
Pem/Carbo/Bev12.918.212.719.2

[back to top]

Safety and Toxicity Profile of Study Treatments

Safety and toxicity profile was defined as serious and other non-serious adverse events. A summary of serious and all other non-serious adverse events is located in the Reported Adverse Event module. (NCT00762034)
Timeframe: Baseline to study endpoint (up to 37.06 months)

,,,
Interventionparticipants (Number)
Serious Adverse Events (SAEs)Other Adverse Events (AEs)
Pac/Carbo/Bev; Induction Phase123431
Pac/Carbo/Bev; Maintenance Phase68296
Pem/Carbo/Bev; Induction Phase111432
Pem/Carbo/Bev; Maintenance Phase83288

[back to top]

Change From Baseline in Participant Reported Outcomes as Assessed by the Functional Assessment of Cancer Therapy - General (FACT-G)

"The FACT-G is a validated instrument used to measure quality of life (QOL) in participants with cancer consisting of the 27-item questionnaire and is organized into subscales, each designed to assess a QOL domain: physical well-being (PWB)-7 items; social/family well-being (SWB)-7 items; emotional well-being (EWB)-6 items; functional well-being (FWB)-7 items. Each item uses a 5 point rating scale (0=not at all and 4=equals very much). FACT-G Total is the sum of the scores of all 4 subscales and ranges from 0 to 108. Higher scores indicate better QOL. Least squares mean (LSmean) change is calculated using the linear-mixed model (LMM) analysis controlled for treatment, baseline value, time point and treatment by time point interaction." (NCT00762034)
Timeframe: Baseline, up to first 10 cycles (4 induction and 6 maintenance cycles, cycle=21 days)

Interventionunits on a scale (Least Squares Mean)
Pem/Carbo/Bev0.51
Pac/Carbo/Bev0.18

[back to top]

Duration of Hospitalizations Per Participant

Length of hospitalization in participants hospitalized during the study or within 30 days of discontinuation regardless of whether the hospitalization was or was not due to study drug. (NCT00762034)
Timeframe: Baseline to study endpoint (up to 37.06 months)

Interventiondays (Mean)
Pem/Carbo/Bev9.4
Pac/Carbo/Bev8.0

[back to top]

Number of Participants Receiving Concomitant Medication

(NCT00762034)
Timeframe: Baseline to study endpoint (up to 37.06 months)

Interventionparticipants (Number)
Pem/Carbo/Bev406
Pac/Carbo/Bev421

[back to top]

Number of Participants Who Received a Transfusion

(NCT00762034)
Timeframe: Baseline to study endpoint (up to 37.06 months)

Interventionparticipants (Number)
Pem/Carbo/Bev116
Pac/Carbo/Bev44

[back to top]

Overall Survival

Overall survival (OS) is the duration from date of randomization to date of death from any cause. Participants were censored at the date they were last known to be alive. (NCT00762034)
Timeframe: Baseline to date of death from any cause (up to 37.06 months)

Interventionmonths (Median)
Pem/Carbo/Bev12.55
Pac/Carbo/Bev13.40

[back to top]

Percentage of Participants With a Complete Response (CR) and Partial Response (PR) (Overall Response Rate)

Overall Response Rate (ORR) is the number of participants with a Complete Response (CR) and Partial Response (PR) divided by the total number of randomized participants per arm, then multiplied by 100. Response is based on the Response Evaluation Criteria In Solid Tumors (RECIST 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 compared to baseline or the complete disappearance of target lesions, with persistence of 1 or more nontarget lesion(s) and no new lesions. (NCT00762034)
Timeframe: Baseline to measured progressive disease (up to 37.06 months)

Interventionpercentage of participants (Number)
Pem/Carbo/Bev34.1
Pac/Carbo/Bev33.0

[back to top]

Percentage of Participants With a Complete Response (CR), Partial Response (PR), and Stable Disease (SD) (Disease Control Rate)

Disease Control Rate (DCR) is the number of participants with a Complete Response (CR), Partial Response (PR), and Stable Disease (SD) divided by the total number of randomized participants per arm, then multiplied by 100. Response is based on the Response Evaluation Criteria In Solid Tumors (RECIST 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 compared with baseline or the complete disappearance of target lesions, with persistence of 1 or more nontarget lesion(s) and no new lesions. Progressive Disease (PD) was defined as at least 20% increase in sum of longest diameter of target lesions compared with the smallest sum of the longest diameter recorded since the start of treatment or the appearance of 1 or more new lesion(s). Stable Disease (SD) was defined as small changes that did not meet above criteria. (NCT00762034)
Timeframe: Baseline to measured progressive disease (up to 37.06 months)

Interventionpercentage of participants (Number)
Pem/Carbo/Bev65.9
Pac/Carbo/Bev69.8

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Time Curve From Zero to Infinity (AUC(0-∞)) Bevacizumab

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.75, 1.5, 3, 5, 7, 24, 48, 72, 168, 336, and 503 hours post-dose)

Interventionmicrogram*day per milliliter (μg•day/mL) (Geometric Mean)
Pem/Carbo/Bev3070
Pac/Carbo/Bev3160

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Time Curve From Zero to Infinity (AUC(0-∞)) for Pemetrexed

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.17, 0.33, 0.58, 0.83, 1, 1.75, 2.5, 4. 6. 8, and 24 hours post-dose)

Interventionmicrogram*hour per milliliter (μg•hr/mL) (Geometric Mean)
Pem/Carbo/Bev203

[back to top]

Pharmacokinetics (PK): Bevacizumab Clearance (CL)

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.75, 1.5, 3, 5, 7, 24, 48, 72, 168, 336, and 503 hours post-dose)

Interventionliters per day (L/day) (Geometric Mean)
Pem/Carbo/Bev0.341
Pac/Carbo/Bev0.376

[back to top]

Pharmacokinetics (PK): Elimination Half-life (t1/2) for Bevacizumab

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.75, 1.5, 3, 5, 7, 24, 48, 72, 168, 336, and 503 hours post-dose)

Interventiondays (Geometric Mean)
Pem/Carbo/Bev14.8
Pac/Carbo/Bev12.8

[back to top]

Pharmacokinetics (PK): Elimination Half-life (t1/2) for Pemetrexed

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.17, 0.33, 0.58, 0.83, 1, 1.75, 2.5, 4. 6. 8, and 24 hours post-dose)

Interventionhours (hr) (Geometric Mean)
Pem/Carbo/Bev2.88

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) for Bevacizumab

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.75, 1.5, 3, 5, 7, 24, 48, 72, 168, 336, and 503 hours post-dose)

Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Pem/Carbo/Bev276
Pac/Carbo/Bev302

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) for Pemetrexed

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.17, 0.33, 0.58, 0.83, 1, 1.75, 2.5, 4. 6. 8, and 24 hours post-dose)

Interventionmicrograms per milliliter (μg/mL) (Geometric Mean)
Pem/Carbo/Bev122

[back to top]

Pharmacokinetics (PK): Pemetrexed Clearance (CL)

(NCT00762034)
Timeframe: Cycle 1 (pre-dose, 0.17, 0.33, 0.58, 0.83, 1, 1.75, 2.5, 4. 6. 8, and 24 hours post-dose)

Interventionmilliliters per minute (mL/min) (Geometric Mean)
Pem/Carbo/Bev72.1

[back to top]

Progression Free Survival Time

Progression free survival (PFS) is defined as the time from date of randomization to the date of objective disease progression or death due to any cause. Participants were censored at date of last PFS assessment prior to the cutoff date or the date of initiation of subsequent systemic anticancer therapy, whichever was earlier. (NCT00762034)
Timeframe: Baseline to measured progressive disease or date of death from any cause (up to 33.54 months)

Interventionmonths (Median)
Pem/Carbo/Bev6.04
Pac/Carbo/Bev5.55

[back to top]

Change From Baseline in Monroe Dunaway (MD) Anderson Symptom Inventory (MDASI) Symptom Severity Score

Symptom severity score is comprised of average of 13 MDASI core items (pain, fatigue, nausea, disturbed sleep, distressed, shortness of breath, remembering things, lack of appetite, drowsy, dry mouth, sadness, vomiting, numbness or tingling) and ranges from 0 to 10. Participants were asked to rate severity of each symptom at their worst in last 24 hours; each item rated from 0 to 10, with 0 = symptom not present and 10 = as bad as you can imagine. Lower scores indicated better outcome. (NCT00768755)
Timeframe: Phase 2 baseline (Cycle1/Day1), Cycle1/Day8, then Day 1 and 8 of each cycle of chemotherapy (C) up to CycleC6, Day 1 of each cycle of single-agent phase (A) up to CycleA8 and end of treatment (EOT)

,,
Interventionunits on a scale (Mean)
Phase 2 baseline (CycleC1/Day 1), (n= 55, 55, 53)Change at CycleC1/Day 8, (n= 49, 49, 50)Change at CycleC2/Day1 (n= 51, 51, 41)Change at CycleC2/Day8 (n= 39, 38, 34)Change at CycleC3/Day1 (n= 45, 47, 39)Change at CycleC3/Day8 (n= 38, 35, 32)Change at CycleC4/Day1 (n= 43, 43, 38)Change at CycleC4/Day8 (n= 33, 33, 27)Change at CycleC5/Day1 (n= 35, 34, 29)Change at CycleC5/Day8 (n= 29, 26, 25)Change at CycleC6/Day1 (n= 27, 28, 28)Change at CycleC6/Day8 (n= 25, 25, 21)Change at CycleA1/Day1 (n= 35, 33, 21)Change at CycleA2/Day1 (n= 32, 30, 14)Change at CycleA3/Day1 (n= 27, 26, 10)Change at CycleA4/Day1 (n= 22, 25, 0)Change at CycleA5/Day1 (n= 17, 23, 12)Change at CycleA6/Day1 (n= 17, 16, 10)Change at CycleA7/Day1 (n= 15, 15, 0)Change at CycleA8/Day1 (n= 15, 14, 10)Change at CycleA9/Day1 (n= 13, 11, 0)Change at CycleA10/Day1 (n= 0, 10, 0)Change at EOT (n= 40, 35, 31)
Axitinib (Continuous) + Pemetrexed/Cisplatin (Phase 2)1.751.550.121.520.571.330.851.370.591.190.671.440.630.360.250.030.190.10-0.26-0.28-0.46NA0.66
Axitinib (Modified) + Pemetrexed/Cisplatin (Phase 2)2.090.36-0.050.390.290.870.480.900.480.820.420.850.24-0.010.010.07-0.12-0.23-0.330.300.340.611.02
Pemetrexed/Cisplatin (Phase 2)1.800.950.390.690.321.150.371.480.721.160.671.280.680.140.12NA0.300.14NA0.02NANA0.99

[back to top]

Change From Baseline in Monroe Dunaway (MD) Anderson Symptom Inventory (MDASI) Symptom Interference Score

Symptom interference score is comprised of average of 6 function items from MDASI core (general activity, mood, work, relations with others, walking, and enjoyment of life) and ranges from 0 to 10. Participants were asked to rate how much symptoms have interfered in last 24 hours; each item rated from 0 to 10, with 0 = did not interfere and 10 = interfered completely. Lower scores indicated better outcome. (NCT00768755)
Timeframe: Phase 2 baseline (Cycle1/Day1), Cycle1/Day8, then Day 1 and 8 of each cycle of chemotherapy (C) up to CycleC6, Day 1 of each cycle of single-agent phase (A) up to CycleA8 and EOT

,,
Interventionunits on a scale (Mean)
Phase 2 baseline (CycleC1/Day 1), (n= 55, 55, 53)Change at CycleC1/Day 8, (n= 49, 49, 50)Change at CycleC2/Day1 (n= 51, 51, 41)Change at CycleC2/Day8 (n= 39, 38, 34)Change at CycleC3/Day1 (n= 45, 47, 39)Change at CycleC3/Day8 (n= 38, 35, 32)Change at CycleC4/Day1 (n= 43, 43, 38)Change at CycleC4/Day8 (n= 33, 33, 27)Change at CycleC5/Day1 (n= 35, 34, 29)Change at CycleC5/Day8 (n= 29, 26, 25)Change at CycleC6/Day1 (n= 27, 28, 28)Change at CycleC6/Day8 (n= 25, 25, 21)Change at CycleA1/Day1 (n= 35, 33, 21)Change at CycleA2/Day1 (n= 32, 30, 14)Change at CycleA3/Day1 (n= 27, 26, 10)Change at CycleA4/Day1 (n= 22, 25, 0)Change at CycleA5/Day1 (n= 17, 23, 12)Change at CycleA6/Day1 (n= 17, 16, 10)Change at CycleA7/Day1 (n= 15, 15, 0)Change at CycleA8/Day1 (n= 15, 14, 10)Change at CycleA9/Day1 (n= 13, 11, 0)Change at CycleA10/Day1 (n= 0, 10, 0)Change at EOT (n= 40, 35, 31)
Axitinib (Continuous) + Pemetrexed/Cisplatin (Phase 2)2.361.480.272.000.641.540.721.390.140.780.181.000.490.250.020.330.360.160.12-0.07-0.13NA0.69
Axitinib (Modified) + Pemetrexed/Cisplatin (Phase 2)2.97-0.03-0.240.03-0.210.280.111.08-0.050.880.171.040.390.16-0.200.230.120.280.000.301.051.030.69
Pemetrexed/Cisplatin (Phase 2)2.640.810.300.520.030.410.281.200.490.760.661.200.960.100.27NA0.400.12NA0.42NANA0.91

[back to top]

Duration of Response (DR)

Time in months from the first documentation of objective tumor response to objective tumor progression or death due to any cause, whichever occurs first. Duration of tumor response was calculated as (the date of the first documentation of objective tumor progression or death due to cause minus the date of the first CR or PR that was subsequently confirmed plus 1) divided by 30.4. DR was calculated for the subgroup of participants with a confirmed objective tumor response. (NCT00768755)
Timeframe: Phase 2 baseline until the date of first documented progression or discontinuation from the study due to any cause or initiation of subsequent anticancer therapy, assessed every 6 weeks up to 84 weeks

Interventionmonths (Median)
Axitinib (Continuous) + Pemetrexed/Cisplatin (Phase 2)7.82
Axitinib (Modified) + Pemetrexed/Cisplatin (Phase 2)6.73
Pemetrexed/Cisplatin (Phase 2)7.10

[back to top]

Overall Survival (OS)

Time in months from the date of randomization to date of death due to any cause. OS was calculated as (the death date minus the date of randomization plus 1) divided by 30.4. Death was determined from AE data (where outcome was death) or from follow-up contact data (where the participant current status was death). (NCT00768755)
Timeframe: Baseline until death or collected bimonthly following discontinuation of study treatment until at least 1 year after randomization of the last participant

Interventionmonths (Median)
Axitinib (Continuous) + Pemetrexed/Cisplatin (Phase 2)17.0
Axitinib (Modified) + Pemetrexed/Cisplatin (Phase 2)14.7
Pemetrexed/Cisplatin (Phase 2)15.9

[back to top]

Percentage of Participants With Objective Response (OR)

Percentage of participants with OR based assessment of confirmed complete response (CR)/confirmed partial response (PR) according to Response Evaluation Criteria in Solid Tumors(RECIST).Confirmed responses: those persist on repeat imaging study at least 4 weeks after initial documentation of response.CR: disappearance of all lesions (target/non target) and no appearance of new lesions.PR: those with at least 30 % decrease in sum of longest dimensions of target lesions taking as reference baseline sum longest dimensions,without progression of non target lesions and no appearance of new lesions. (NCT00768755)
Timeframe: Phase 2 baseline until the date of first documented progression or discontinuation from the study due to any cause or initiation of subsequent anticancer therapy, assessed every 6 weeks up to 84 weeks

Interventionpercentage of participants (Number)
Axitinib (Continuous) + Pemetrexed/Cisplatin (Phase 2)45.5
Axitinib (Modified) + Pemetrexed/Cisplatin (Phase 2)39.7
Pemetrexed/Cisplatin (Phase 2)26.3

[back to top]

Progression-Free Survival (PFS)

"Time in months from the date of randomization to first documentation of objective tumor progression or death due to any cause. PFS was calculated as (first event date minus minus the date of randomization plus 1) divided by 30.4. Tumor progression was determined from oncologic assessment data (where data meet the criteria for progressive disease [PD]); death was determined from AE data (where the outcome was Death) or from the end of study data." (NCT00768755)
Timeframe: Phase 2 baseline until the date of first documented progression or death due to any cause or initiation of subsequent anticancer therapy, assessed every 6 weeks up to 84 weeks

Interventionmonths (Median)
Axitinib (Continuous) + Pemetrexed/Cisplatin (Phase 2)8.0
Axitinib (Modified) + Pemetrexed/Cisplatin (Phase 2)8.1
Pemetrexed/Cisplatin (Phase 2)7.1

[back to top]

RECIST Response

Number of participants with partial response (PR), stable disease (SD) and progressive disease (PD) as defined by Response Evaluation Criteria in Solid Tumors (RECIST) (NCT00769600)
Timeframe: Up to 3 years

,
InterventionParticipants (Count of Participants)
PRSDPDInevaluable
Itraconazole Open Label Added to Standard of Care Pemetrexed3822
Single Agent Pemetrexed0521

[back to top]

Progression Free Survival as Measured by Number of Days Without Disease Progression

(NCT00769600)
Timeframe: 1 year

Interventiondays (Median)
Itraconazole Open Label Added to Standard of Care Pemetrexed168
Single Agent Pemetrexed84

[back to top]

Overall Survival

Median number of days alive (NCT00769600)
Timeframe: up to 3 years

Interventiondays (Median)
Arm A971
Arm B242

[back to top]

Progression Free Survival

For determining progression-free survival, progression was determined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0). Progression was defined 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. (NCT00771953)
Timeframe: From the date of randomization until the first date that recurrent or progressive disease is objectively documented.

Interventiondays (Median)
Apricoxib Plus Docetaxel75
Placebo Plus Docetaxel97
Apricoxib Plus Pemetrexed103
Placebo Plus Pemetrexed98

[back to top]

Percentage of Participants With Independently-Assessed Objective Tumor Response (Response Rate) During Maintenance Phase Up to Primary Data Cut-Off

Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response (CR)=disappearance of all target lesions; Partial Response (PR) is at least a 30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD) is at least a 20% increase in sum of longest diameter of target lesions; Stable Disease (SD)=no change or small changes that do not meet the above criteria for CR, PR, or PD. Response Rate = (CR+PR)/Participants in Arm*100. Disease Control Rate=(CR+PR+SD)/Number of Participants in Arm*100. (NCT00789373)
Timeframe: Date of randomization to date of measured PD (up to 19.3 months)

,
Interventionpercentage of participants (Number)
Response RateDisease Control Rate
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed46.298.1
Pemetrexed + Cisplatin Followed by Placebo42.294.4

[back to top]

Percentage of Participants With Serious Adverse Events During Maintenance Phase

A summary of serious adverse events is located in the Reported Adverse Event Module. (NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 49.7 months)

,
Interventionpercentage of participants (Number)
Serious Adverse Events: up to 19.3 MonthsSerious Adverse Events: up to 49.7 Months
Pemetrexed Plus Cisplatin Followed by Maintenance Pemetrexed18.926.2
Pemetrexed Plus Cisplatin Followed by Placebo12.220.0

[back to top]

Change From Baseline in the EuroQol Instrument (EQ-5D) Index Score

The EQ-5D is a generic instrument that describes health status in 5 attributes (mobility, self-care, pain/discomfort, anxiety/depression, usual activities) using a three level scale (no problem, some problems, and major problems). These combinations of attributes are converted into a weighted health-state Index Score according to the United Kingdom (UK) population-based algorithm. The possible values for the Index Score range from -0.59 (severe problems in all 5 dimensions) to 1.0 (no problem in any dimension). (NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 19.3 months)

Interventionunits on a scale (Mean)
BaselineCycle 1 Day 1Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 5 Day 1Cycle 6 Day 1Cycle 7 Day 1Cycle 8 Day 1Cycle 9 Day 1Cycle 10 Day 1Cycle 11 Day 1Cycle 12 Day 1Cycle 13 Day 1Cycle 14 Day 1Cycle 15 Day 1Cycle 16 Day 1Cycle 17 Day 1Cycle 18 Day 130 Day Post-Study Visit
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed0.770.010.00.0-0.010.01-0.020.010.01-0.030.0-0.02-0.06-0.010.03-0.07-0.010.320.45-0.13

[back to top]

Percentage of Participants With a Non-Serious Adverse Event (AE) During Maintenance Phase

A summary of non-serious AEs is located in the Reported Adverse Event Module. (NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 49.7 months)

,
Interventionpercentage of participants (Number)
Non-Serious AEs at 2% Threshold: up to 19.3 MonthNon-Serious AEs at 5% Threshold: up to 49.7 Months
Pemetrexed Plus Cisplatin Followed by Maintenance Pemetrexed59.975.5
Pemetrexed Plus Cisplatin Followed by Placebo50.662.2

[back to top]

Overall Survival (OS)

OS is the duration from enrollment to death. For patients who are alive, OS is censored at the last contact. (NCT00789373)
Timeframe: Date of randomization to the date of death from any cause up to 39.5 months

Interventionmonths (Median)
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed13.86
Pemetrexed + Cisplatin Followed by Placebo11.01

[back to top]

Percentage of Participants With Objective Tumor Response (Response Rate) During Maintenance Phase of Study up to Primary Data Cut-Off

Analysis for combined phases was not performed since response was calculated separately for each phase of study. Response using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response (CR)=disappearance of all target lesions; Partial Response(PR)is at least a 30% decrease in sum of longest diameter of target lesions; Progressive Disease(PD) is at least a 20% increase in sum of longest diameter of target lesions; Stable Disease(SD)=no change or small changes that do not meet the above criteria for CR, PR, or PD. (NCT00789373)
Timeframe: Baseline to date of measured progressive disease (up to 19.3 months)

Interventionpercentage of participants (Number)
Pemetrexed Plus Cisplatin Followed by Maintenance Pemetrexed46.2
Pemetrexed + Cisplatin Followed by Placebo42.2

[back to top]

Change From Baseline in EuroQol Instrument (EQ-5D) Visual Analog Scale (VAS)

Patients indicate their present health state through completion of the VAS. Possible scores range from 0 (worst imaginable health state) to 100 (best imaginable health state). (NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 19.3 months)

Interventionunits on a scale (Mean)
BaselineCycle 1 Day 1Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 5 Day 1Cycle 6 Day 1Cycle 7 Day 1Cycle 8 Day 1Cycle 9 Day 1Cycle 10 Day 1Cycle 11 Day 1Cycle 12 Day 1Cycle 13 Day 1Cycle 14 Day 1Cycle 15 Day 1Cycle 16 Day 130 days post-study
Pemetrexed + Cisplatin Followed by Placebo71.01.423.154.906.155.995.763.987.587.616.230.944.6310.014.012.015.0-3.92

[back to top]

Change From Baseline in EuroQol Instrument (EQ-5D) Visual Analog Scale (VAS)

Patients indicate their present health state through completion of the VAS. Possible scores range from 0 (worst imaginable health state) to 100 (best imaginable health state). (NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 19.3 months)

Interventionunits on a scale (Mean)
BaselineCycle 1 Day 1Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 5 Day 1Cycle 6 Day 1Cycle 7 Day 1Cycle 8 Day 1Cycle 9 Day 1Cycle 10 Day 1Cycle 11 Day 1Cycle 12 Day 1Cycle 13 Day 1Cycle 14 Day 1Cycle 15 Day 1Cycle 16 Day 1Cycle 17 Day 1Cycle 18 Day 130 days post-study
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed71.11.651.241.820.691.553.012.74.124.195.142.582.116.293.648.405.8315.75.0-4.77

[back to top]

Change From Baseline in the EuroQol Instrument (EQ-5D) Index Score

The EQ-5D is a generic instrument that describes health status in 5 attributes (mobility, self-care, pain/discomfort, anxiety/depression, usual activities) using a three level scale (no problem, some problems, and major problems). These combinations of attributes are converted into a weighted health-state Index Score according to the United Kingdom (UK) population-based algorithm. The possible values for the Index Score range from -0.59 (severe problems in all 5 dimensions) to 1.0 (no problem in any dimension). (NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 19.3 months)

Interventionunits on a scale (Mean)
BaselineCycle 1 Day 1Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 5 Day 1Cycle 6 Day 1Cycle 7 Day 1Cycle 8 Day 1Cycle 9 Day 1Cycle 10 Day 1Cycle 11 Day 1Cycle 12 Day 1Cycle 13 Day 1Cycle 14 Day 1Cycle 15 Day 1Cycle 16 Day 130 Day Post-Study Visit
Pemetrexed + Cisplatin Followed by Placebo0.79-0.010.010.030.020.010.040.010.050.060.080.040.060.00.030.010.0-0.09

[back to top]

Percentage of Participants With Hospitalizations Due to Adverse Events or Requiring Transfusion (Resource Utilization)

(NCT00789373)
Timeframe: Baseline randomization through 30-day post-discontinuation visit (up to 19.3 months)

,
Interventionpercentage of participants (Number)
Hospitalization due to Drug-related Adverse EventTransfusions Packed Red Blood CellsTransfusions Whole BloodTransfusions PlateletsTransfusions Fresh Frozen Plasma
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed8.412.31.41.40
Pemetrexed + Cisplatin Followed by Placebo3.34.40.60.60.6

[back to top]

Independently-assessed Objective Progression-free Survival (PFS)

To further evaluate the robustness of the PFS analysis, Lilly established an independent review of PFS to assess the potential for investigator bias in the determination of objective PD. PFS was measured from the date of randomization to the first date of objectively determined PD or death. For patients alive as of the data cutoff date and who did not have PD, PFS was censored at the date of the last objective tumor assessment. PD was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. PD = 20% increase in sum of longest diameter of target lesions. (NCT00789373)
Timeframe: Date of randomization to first date of measured PD or date of death from any cause (up to 19.3 months)

Interventionmonths (Median)
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed3.94
Pemetrexed + Cisplatin Followed by Placebo2.60

[back to top]

Investigator-assessed Objective Progression-free Survival (PFS)

Investigator-assessed objective PFS was measured from the date of randomization to the first date of objectively determined progressive disease (PD) or death from any cause. For patients not known to have died as of the data cutoff date and who did not have objective PD, PFS was censored at the date of last objective tumor assessment. PD was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. PD = 20% increase in sum of longest diameter of target lesions. (NCT00789373)
Timeframe: Date of randomization to the date of measured PD or date of death from any cause (up to 19.3 months)

Interventionmonths (Median)
Pemetrexed + Cisplatin Followed by Maintenance Pemetrexed4.11
Pemetrexed + Cisplatin Followed by Placebo2.83

[back to top]

Phase 1 and 2: Terminal Half-Life (t1/2) of Aflibercept

Terminal half-life was defined as the time required for the plasma concentration of drug to decrease 50 percent in the final stage of its elimination. (NCT00794417)
Timeframe: Phase 1 and 2: Pre-dose up to Day 22 post-dose

InterventionDays (Mean)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin3.16
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin5.53
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin3.72
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin4.62

[back to top]

Phase 1 and 2: Number of Participants With Treatment Emergent Adverse Events (TEAEs)

An adverse event (AE) was defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (for example, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug. A treatment-emergent adverse event (TEAE) was defined as an adverse event with an onset that occurs after receiving study drug. Any TEAE included participants with both serious and non-serious AEs. (NCT00794417)
Timeframe: Phase 1: Baseline up to 751 Days; Phase 2: Baseline (Day 421) up to 972 Days

InterventionParticipants (Count of Participants)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin4
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin7
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin7
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin42

[back to top]

Phase 1 and 2: Number of Participants With Positive Anti-drug Antibody (ADA) of Aflibercept

Serum samples were analyzed by a validated electrochemiluminescence immunoassay to detect the presence of ADA. (NCT00794417)
Timeframe: Phase 1: Baseline up to 315 Days; Phase 2: Baseline (Day 421) up to Day 739

InterventionParticipants (Count of Participants)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin0
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin0
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin1
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin2

[back to top]

Phase 1 and 2: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-inf) of Pemetrexed

The AUC0-inf was estimated by determining the total area under the curve of the concentration versus time curve extrapolated to infinity. (NCT00794417)
Timeframe: Phase 1 and 2: Pre-dose up to Day 1 post-dose, Day 2 post-dose (only in Phase 1)

InterventionHour*milligrams per liter (mg/L) (Mean)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin151
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin151
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin162
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin148

[back to top]

Phase 1 and 2: Terminal Half-Life (t1/2) of Pemetrexed

Terminal half-life was defined as the time required for the plasma concentration of drug to decrease 50 percent in the final stage of its elimination. (NCT00794417)
Timeframe: Phase 1 and 2: Pre-dose up to Day 1 post-dose, Day 2 post-dose (only in Phase 1)

InterventionHours (Mean)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin1.47
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin1.63
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin1.73
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin1.48

[back to top]

Phase 1 and 2: Maximum Observed Plasma Concentration (Cmax) of Aflibercept and Pemetrexed

Cmax is the maximum observed plasma concentration obtained directly from the concentration versus time curve. (NCT00794417)
Timeframe: Phase 1 and 2: Aflibercept: Pre-dose up to Day 22 post-dose; Pemetrexed: Pre-dose up to Day 1 post-dose, Day 2 post-dose (only in Phase 1)

,,,
Interventionmg/L (Mean)
AfliberceptPemetrexed
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin53.6124
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin68.6112
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin148113
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin10476.8

[back to top]

Phase 1 and 2: Number of Participants With All Grade Hematology Abnormalities

(NCT00794417)
Timeframe: Phase 1: Baseline up to 751 Days; Phase 2: Baseline (Day 421) up to 972 Days

,,,
InterventionParticipants (Number)
Absolute Neutrophil Count (ANC)HemoglobinPlatelet Count
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin232
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin474
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin762
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin13168

[back to top]

Phase 2: Progression-free Survival (PFS)

PFS was defined as the time in days from the date of first study drug administration to the date of first documentation of tumor progression or death from any cause, whichever occurs first, as assessed by the modified RECIST. Median time of PFS was estimated using Kaplan-Meier method. (NCT00794417)
Timeframe: Phase 2: Baseline (Day 421) up to end of study (Day 972)

InterventionDays (Median)
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin149

[back to top]

Phase 2: Objective Response Rate

Objective response rate was defined as the percentage of participants who achieved complete response (CR) or partial response (PR) as assessed by modified Response Evaluation Criteria in Solid Tumors (RECIST). CR was defined as disappearance of all target lesions and PR was defined as at least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking the Baseline sum LD as reference. (NCT00794417)
Timeframe: Phase 2: Baseline (Day 421) up to end of study (Day 972)

InterventionPercentage of Participants (Number)
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin23.8

[back to top] [back to top]

Phase 1 and 2: Total Body Clearance of Pemetrexed

Clearance of a drug was a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. (NCT00794417)
Timeframe: Phase 1 and 2: Pre-dose up to Day 1 post-dose, Day 2 post-dose (only in Phase 1)

InterventionLiter/hour/m^2 (Mean)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin3.40
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin3.47
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin3.10
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin3.49

[back to top]

Phase 1 and 2: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-inf) of Aflibercept

The AUC0-inf was estimated by determining the total area under the curve of the concentration versus time curve extrapolated to infinity. (NCT00794417)
Timeframe: Phase 1 and 2: Pre-dose up to Day 22 post-dose

InterventionDay*milligrams per liter (mg/L) (Mean)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin201
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin330
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin442
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin402

[back to top]

Phase 1 and 2: Total Body Clearance of Aflibercept

Clearance of a drug was a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. (NCT00794417)
Timeframe: Phase 1 and 2: Pre-dose up to Day 22 post-dose

InterventionLiter/Day/kg (Mean)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin0.011
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin0.016
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin0.016
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin0.016

[back to top]

Phase 1 and 2: Number of Participants With All Grade Glucose Abnormalities

(NCT00794417)
Timeframe: Phase 1: Baseline up to 751 Days; Phase 2: Baseline (Day 421) up to 972 Days

,,,
InterventionParticipants (Number)
Hyperglycemia (Non-Fasting)Hyperglycemia (Fasting)Hypoglycemia (Non-Fasting)Hypoglycemia (Fasting)
Phase 1: Aflibercept 2 mg/kg and Pemetrexed and Cisplatin4100
Phase 1: Aflibercept 4 mg/kg and Pemetrexed and Cisplatin6300
Phase 1: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin7600
Phase 2: Aflibercept 6 mg/kg and Pemetrexed and Cisplatin37650

[back to top]

Change From Baseline to Cycle 3 in Fatigue Assessed by Treatment-specific Adverse Events Scale

The single-item fatigue question was on a 10-points scale with 0=no fatigue and 10=as bad as you can imagine. The item scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 3 was calculated by subtracting the baseline scores from the scores at cycle 3. (NCT00798603)
Timeframe: Baseline and Cycle 3

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Change From Baseline to Cycle 5 in Fatigue Assessed by Treatment-specific Adverse Events Scale

The single-item fatigue question was on a 10-points scale with 0=no fatigue and 10=as bad as you can imagine. The item scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 5 was calculated by subtracting the baseline scores from the scores at cycle 5. (NCT00798603)
Timeframe: Baseline and Cycle 5

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab-10

[back to top]

Change From Baseline to Cycle 3 in Neuropathy Assessed by Treatment-specific Adverse Events Scale

The single-item neuropathy question was on a 10-points scale with 0=no numbness or tingling in fingers and toes and 10=worst numbness or tingling in fingers and toes imaginable. The item scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 3 was calculated by subtracting the baseline scores from the scores at cycle 3. (NCT00798603)
Timeframe: Baseline and Cycle 3

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Change From Baseline to Cycle 3 in Overall Quality of Life Assessed by Linear Analogue Self Assessment (LASA)

The overall quality of life (QOL) question was on a 10-points scale with 0=as bad as it can be and 10=as good as it can be. The QOL scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 3 was calculated by subtracting the baseline scores from the scores at cycle 3. (NCT00798603)
Timeframe: Baseline and Cycle 3

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Change From Baseline to Cycle 3 in Nausea Assessed by Treatment-specific Adverse Events Scale

The single-item nausea question was on a 10-points scale with 0=no nausea and 10=as bad as you can imagine. The item scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 3 was calculated by subtracting the baseline scores from the scores at cycle 3. (NCT00798603)
Timeframe: Baseline and Cycle 3

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Number of Grade 3 or Higher Adverse Events Occurring in >=10% of Patients

Adverse events were assessed by Common Terminology Criteria for Adverse Events (CTCAE) v3.0. Grading: Grade 1=Mild, Grade 2=Moderate, Grade 3=Severe, Grade 4=Life-threatening, Grade 5=Death. (NCT00798603)
Timeframe: Up to 2.5 years

Interventionparticpants (Number)
FatigueHypertensionNeutropeniaThrombocytopenia
Pemetrexed + Carboplatin + Bevacizumab1671811

[back to top]

Proportion of Confirmed Tumor Response Defined as an Objective Status of Complete Response or Partial Response on Two Consecutive Evaluations

"Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria:~Complete Response (CR): disappearance of all target and non-target lesions and no new lesions.~Partial Response (PR): disappearance of all target lesions, persistence of one or more non-target lesions, and no new lesions; or at least a 30% decrease in the sum of the longest diameter (LD) of target lesions taking as reference the baseline sum LD, no appearance of one/more new lesions, unequivocal progression of existing non-target lesions, and no new lesions." (NCT00798603)
Timeframe: Duration of study until progression (up to 5 years)

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin + Bevacizumab40

[back to top]

Time to Treatment Failure

Time to treatment failure was defined to be the time from date of registration to the date at which the patient is removed from the treatment due to progression, toxicity, refusal or death from any cause. (NCT00798603)
Timeframe: Up to 5 years

Interventionmonths (Median)
Pemetrexed + Carboplatin + Bevacizumab4.89

[back to top]

Progression-free Survival at 6 Months

Estimated using the Binomial point estimator (number of successes divided by the total number of evaluable patients). A patient is classified as a success if alive and progression-free at 6 months. (NCT00798603)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin + Bevacizumab60

[back to top]

Progression-free Survival

Progression-free survival was defined as the time from study enrollment to the first date of disease progression or death as a result of any cause, whichever occurs first. Progression-free survival will be censored at the date of the last contact for patients who are still alive and who have not had disease progression. (NCT00798603)
Timeframe: Up to 5 years

Interventionmonths (Median)
Pemetrexed + Carboplatin + Bevacizumab7

[back to top]

Change From Baseline to Cycle 3 in Quality of Life (QOL) as Assessed by the Lung Cancer Symptom Scale

Lung Cancer Symptom Scale (LCSS) consist of 9 items that assess the symptoms of lung cancer during the past days on a 10-points scale with 0 as no symptoms and 10 as worse symptoms. The individual item score was translated onto a 0-100 point scale with lower values indicating worse symptoms. An average of the aggregate score of all 9 items was used for a total score. Change from baseline to cycle 3 was calculated by subtracting the baseline scores from the scores at cycle 3. (NCT00798603)
Timeframe: Baseline and Cycle 3

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab1.1

[back to top]

Overall Survival

Overall survival was defined as the time from study enrollment to the time of death from any cause. Overall survival will be censored at the date of the last follow-up visit for patients who are still alive or lost to follow-up. (NCT00798603)
Timeframe: Up to 5 years

Interventionmonths (Median)
Pemetrexed + Carboplatin + Bevacizumab13.7

[back to top]

Duration of Response

Duration of response for responders was defined as the time from the date of the first objective status assessment of a confirmed CR or PR to the first date of disease progression. Duration of response will be censored at the date of last post-therapy follow-up visit for responders who have not had disease progression. Duration of response will be calculated for all evaluable patients who have achieved an objective confirmed response. (NCT00798603)
Timeframe: Up to 5 years

Interventionmonths (Median)
Pemetrexed + Carboplatin + Bevacizumab8.8

[back to top]

Change From Baseline to Cycle 5 in Quality of Life (QOL) as Assessed by the Lung Cancer Symptom Scale

Lung Cancer Symptom Scale (LCSS) consist of 9 items that assess the symptoms of lung cancer during the past days on a 10-points scale with 0 as no symptoms and 10 as worse symptoms. The individual item score was translated onto a 0-100 point scale with lower values indicating worse symptoms. An average of the aggregate score of all 9 items was used for a total score. Change from baseline to cycle 5 was calculated by subtracting the baseline scores from the scores at cycle 5. (NCT00798603)
Timeframe: Baseline and Cycle 5

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab1.1

[back to top]

Change From Baseline to Cycle 5 in Overall Quality of Life Assessed by Linear Analogue Self Assessment (LASA)

The overall quality of life (QOL) question was on a 10-points scale with 0=as bad as it can be and 10=as good as it can be. The QOL scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 5 was calculated by subtracting the baseline scores from the scores at cycle 5. (NCT00798603)
Timeframe: Baseline and Cycle 5

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Change From Baseline to Cycle 5 in Neuropathy Assessed by Treatment-specific Adverse Events Scale

The single-item neuropathy question was on a 10-points scale with 0=no numbness or tingling in fingers and toes and 10=worst numbness or tingling in fingers and toes imaginable. The item scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 5 was calculated by subtracting the baseline scores from the scores at cycle 5. (NCT00798603)
Timeframe: Baseline and Cycle 5

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Change From Baseline to Cycle 5 in Nausea Assessed by Treatment-specific Adverse Events Scale

The single-item nausea question was on a 10-points scale with 0=no nausea and 10=as bad as you can imagine. The item scores was translated onto a 0 to 100 point scale, with lower values indicating worse symptoms. Change from baseline to cycle 5 was calculated by subtracting the baseline scores from the scores at cycle 5. (NCT00798603)
Timeframe: Baseline and Cycle 5

Interventionunits on a scale (Median)
Pemetrexed + Carboplatin + Bevacizumab0

[back to top]

Overall Survival (Key Secondary Endpoint)

Overall Survival (OS) defined as the duration from randomisation to death (irrespective of the reason of death). Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve. (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

Interventionmonths (Median)
Nintedanib Plus Pemetrexed12.0
Placebo Plus Pemetrexed12.7

[back to top]

Duration of Disease Control

"The duration of disease control was defined as the time from randomisation to the date of disease progression or death (which ever occurs first) for patients with disease control. Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve.~This endpoint was analysed based on the central independent reviewer as well as the investigator." (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
InterventionMonths (Median)
Central independent review (N=215, 192)Investigator assessment (N=233, 217)
Nintedanib Plus Pemetrexed7.46.9
Placebo Plus Pemetrexed6.86.8

[back to top]

Incidence and Intensity of Adverse Events

"Incidence and intensity of adverse events according to the Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. The worst CTCAE grade per patient is reported and MedDRA version 15.1 used.~Serious signs and symptoms of progressive disease were reported as an adverse event in analysis of this endpoint." (NCT00806819)
Timeframe: From the first drug administration until 28 days after the last drug administration, up to 36 months

,
Intervention% of participants (Number)
CTCAE grade 1CTCAE grade 2CTCAE grade 3CTCAE grade 4CTCAE grade 5
Nintedanib Plus Pemetrexed4.922.246.112.49.8
Placebo Plus Pemetrexed9.230.534.57.812.0

[back to top]

Objective Tumor Response

Confirmed objective response is defined as confirmed Complete Response (CR) and Partial Response (PR) and evaluated according to the modified RECIST criteria version 1.0. This endpoint was analysed based on the central independent reviewer as well as the investigator (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
Intervention% of participants (Number)
Central independent reviewerInvestigator assessment
Nintedanib Plus Pemetrexed9.115.0
Placebo Plus Pemetrexed8.313.3

[back to top]

Duration of Confirmed Objective Tumour Response

"The duration of objective response is the time from first documented (CR) or (PR) to the time of progression or death and evaluated according to the modified RECIST criteria version 1.0. Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve.~This endpoint was analysed based on the central independent reviewer as well as the investigator." (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
InterventionMonths (Median)
central independent reviewer (N=32, 30)Investigator assessment (N=53, 48)
Nintedanib Plus Pemetrexed6.96.5
Placebo Plus Pemetrexed4.47.2

[back to top]

Progression Free Survival (PFS) as Assessed by Central Independent Review

"Progression Free Survival (PFS) as assessed by central independent review according to the modified RECIST (version 1.0) criteria. Progression free survival (PFS) is defined as the duration of time from date of randomisation to date of progression or death (whatever occurs earlier).~Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve." (NCT00806819)
Timeframe: From randomisation until cut-off date 9 July 2012

Interventionmonths (Median)
Nintedanib Plus Pemetrexed4.4
Placebo Plus Pemetrexed3.6

[back to top]

Clinical Improvement.

"Clinical improvement was defined as the time from randomisation to deterioration in body weight and/or Eastern Cooperative Oncology group performance score (ECOG PS) whichever occurred first.~Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve." (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

InterventionMonths (Median)
Nintedanib Plus Pemetrexed7.2
Placebo Plus Pemetrexed7.5

[back to top]

Follow-up Analysis of Progression Free Survival (PFS) as Assessed by Central Independent Review

Follow-up analysis was conducted at the time of overall survival analysis. Progression Free Survival (PFS) as assessed by central independent review according to the modified RECIST (version 1.0) criteria. Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve. (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

InterventionMonths (Median)
Nintedanib Plus Pemetrexed4.4
Placebo Plus Pemetrexed3.4

[back to top]

Follow-up Analysis of Progression Free Survival (PFS) as Assessed by Investigator

Follow-up analysis was conducted at the time of overall survival analysis. Progression Free Survival (PFS) as assessed by investigator according to the modified RECIST (version 1.0) criteria. Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve. (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

InterventionMonths (Median)
Nintedanib Plus Pemetrexed5.3
Placebo Plus Pemetrexed4.3

[back to top]

Quality of Life (QoL)

"QoL was measured by standardised questionnaires (EQ-5D, EORTC QLQ-C30, EORTC QLQ-LC13). The EORTC QLQ-C30 comprises of 30 questions, using both multi-item scales and single-item measures. EORTC LC-13 comprises of 13 questions incorporating 1 multi-item scale and a series of single items. The following were the main points of interest: Time to deterioration of cough (QLQ-LC13 question 1), Time to deterioration of dyspnoea (QLQ-LC13, composite of questions 3 to 5), Time to deterioration of pain (QLQ- C30, composite of questions 9 and 19). Time to deterioration of cough, dyspnoea and pain was defined as the time to a 10-point increase from the baseline score.~Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve." (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
InterventionMonths (Median)
Time to deterioration of coughTime to deterioration of dyspnoeaTime to deterioration of pain
Nintedanib Plus Pemetrexed6.02.42.8
Placebo Plus Pemetrexed4.32.02.7

[back to top]

Time to Confirmed Objective Tumour Response

"Time to confirmed objective response is defined as time from randomisation to the date of first documented (CR) or (PR) and evaluated according to the modified RECIST criteria version 1.0. Median, 25th and 75th percentiles are calculated from an unadjusted Kaplan-Meier curve.~This endpoint was analysed based on the central independent reviewer as well as the investigator." (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
InterventionMonths (Median)
Central independent review (N=32, 30)Investigator assessment (N=53, 48)
Nintedanib Plus Pemetrexed2.62.6
Placebo Plus Pemetrexed2.72.8

[back to top]

Dose Normalised Predose Plasma Concentration at Steady State (Cpre,ss,Norm) of Nintedanib and of Its Metabolites BIBF 1202 and BIBF 1202 Glucuronide

Geometric mean of dose normalised predose plasma concentration (Cpre,ss,norm) of nintedanib and of its metabolites BIBF 1202 and BIBF 1202 glucuronide evaluated at steady state based on course 2 and 3. If only one value was available and valid, then this value was used for calculation of Cpre,ss,norm. (NCT00806819)
Timeframe: Before the administration of nintedanib or placebo and between a window of 30 mins to an hour after administration of trial drug during Course 2 and between 1 and 3 hours after administration of trial drug during Course 3

,
Interventionng/mL/mg (Geometric Mean)
Nintedanib BIBF 1120 (N=188, 39)Nintedanib BIBF 1202 (N=188, 40)Nintedanib BIBF 1202 glucuronide (N=184, 39)
Nintedanib 150 mg Bid Plus Pemetrexed0.1030.1511.72
Nintedanib Plus Pemetrexed0.08830.1311.40

[back to top]

Disease Control

"Disease control was defined as a best overall response of Complete Response (CR), Partial Response (PR), or Stable Disease (SD) and evaluated according to the modified RECIST criteria version 1.0.~This endpoint was analysed based on the central independent reviewer as well as the investigator." (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
Intervention% of participants (Number)
Central independent review (N=215, 192)Investigator assessment (N=233, 217)
Nintedanib Plus Pemetrexed60.966.0
Placebo Plus Pemetrexed53.360.3

[back to top]

Change From Baseline in Tumour Size

Percentage change from baseline in tumour size is defined as decrease in the sum of the longest diameter of the target lesion. Presented means are in fact adjusted best means percentage changes generated from ANOVA model adjusted for baseline ECOG PS (0 vs. 1), tumour histology (adenocarcinoma vs. non-adenocarcinoma), brain metastases at baseline (yes vs no) and prior treatment with bevacizumab (yes vs no) This endpoint was analysed based on the central independent reviewer as well as the investigator. (NCT00806819)
Timeframe: From randomisation until data cut-off (15 February 2013), Up to 30 months

,
Interventionpercentage of change in tumor size in mm (Mean)
Central independent review (N=298, 305)Investigator assessment (N=322, 325)
Nintedanib Plus Pemetrexed-10.10-15.60
Placebo Plus Pemetrexed-7.53-11.28

[back to top]

Objective Response Rate (CR + PR by RECIST) Paclitaxel, Pemetrexed, and Bevacizumab in Patients With Advanced Non-Small Lung Cancer Who Have Received no Prior Treatment for Metastatic Disease.

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. (NCT00807573)
Timeframe: 2 years

Interventionparticipants (Number)
Partial ResponseStable Disease
Paclitaxel, Bevacizumab & Pemetrexed2715

[back to top]

Duration of Overall Response

The duration of overall response was measured from the time measurement criteria were met for CR or PR (whichever was first recorded) until the first date that recurrent or progressive disease (PD) was objectively documented (taking as reference for PD the smallest measurements recorded since treatment began). The duration of overall CR was measured from the time measurement criteria were first met for CR until the first date that recurrent disease was objectively documented. Duration of disease control is presented here. (NCT00824408)
Timeframe: From the time measurement criteria were met for CR or PR (whichever was first recorded) until the first date that recurrent or progressive disease was objectively documented

Interventionweeks (Median)
Randomization Phase: Volasertib 300 mg23.0
Randomization Phase: Volasertib 300 mg + Pemetrexed 500 mg/m219.6
Randomization Phase: Pemetrexed 500 mg/m223.4

[back to top]

Objective Tumor Response, Defined as Complete Response (CR), and Partial Response (PR), Evaluated According to RECIST Criteria.

Objective tumor response, defined as complete response (CR), and partial response (PR), evaluated according to RECIST criteria. Evaluation of 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, taking as reference the baseline sum longest diameter. Evaluation of nontarget lesions: Complete Response (CR): disappearance of all nontarget lesions. (NCT00824408)
Timeframe: From first drug infusion until 21 days after last drug infusion, up to 1100 days

,,,,
Interventionpercentage of participants (Number)
Complete response (CR)Partial response (PR)
Randomization Phase: Pemetrexed 500 mg/m20.010.6
Randomization Phase: Volasertib 300 mg0.08.1
Randomization Phase: Volasertib 300 mg + Pemetrexed 500 mg/m20.021.3
Run-in Phase: Volasertib 250 mg + Pemetrexed 500 mg/m20.016.7
Run-in Phase: Volasertib 300 mg + Pemetrexed 500 mg/m20.050.0

[back to top]

Cmax of Volasertib

Cmax - maximum measured concentration of volasertib in plasma. (NCT00824408)
Timeframe: 5 minutes (min) before the start of Volasertib infusion and 1 hour (h), 2h, 4h, 24h, 168h and 336h after the start of Volasertib infusion

Interventionng/mL (Geometric Mean)
Volasertib 250 mg + Pemetrexed 500 mg/m2554
Volasertib 300 mg + Pemetrexed 500 mg/m2635
Volasertib 300 mg565

[back to top]

CL of Pemetrexed

CL - total clearance of pemetrexed in plasma after IV administration (NCT00824408)
Timeframe: 5 minutes before pemetrexed infusion, at the end of the infusion and 1.5 hours (h), 2.5h, 4.5h and 25.5h after the end of pemetrexed infusion

InterventionmL/min (Geometric Mean)
Volasertib 250 mg + Pemetrexed 500 mg/m254.4
Volasertib 300 mg + Pemetrexed 500 mg/m269.1

[back to top]

Cmax of Pemetrexed

Cmax - maximum measured concentration of pemetrexed in plasma (NCT00824408)
Timeframe: 5 minutes before pemetrexed infusion, at the end of the infusion and 1.5 hours (h), 2.5h, 4.5h and 25.5h after the end of pemetrexed infusion

Interventionng/mL (Geometric Mean)
Volasertib 250 mg + Pemetrexed 500 mg/m2131000
Volasertib 300 mg + Pemetrexed 500 mg/m2115000

[back to top]

Occurence of DLT

"Occurence of Dose-limiting toxicity (DLT). A DLT was defined as one or more of the following:~treatment-related CTCAE Grade 3 or 4 nonhematological toxicity (except emesis or diarrhea responding to supportive treatment).~treatment-related CTCAE Grade 4 neutropenia for ≥7 days and/or complicated by infection.~CTCAE Grade 4 thrombocytopenia." (NCT00824408)
Timeframe: Patients were treated for repeated 21-day treatment cycles until disease progression or intolerability of the trial drug, whichever occurred first.

Interventionparticipants (Number)
Run-in Phase: Volasertib 250 mg + Pemetrexed 500 mg/m21
Run-in Phase: Volasertib 300 mg + Pemetrexed 500 mg/m21

[back to top]

Frequency of Patients With Possible Clinically Significant Abnormalities

Frequency of patients with possible clinically significant abnormalities (NCT00824408)
Timeframe: From first drug infusion until 21 days after last drug infusion, up to 1100 days

,,,,
Interventionparticipants (Number)
Haemoglobin - LowWhite blood cell ct. - LowWhite blood cell ct. - HighPlatelets - LowPlatelets - HighNeutrophils - LowLymphocytes - LowAST/GOT, SGOT - HighALT/GPT, SGPT - HighAlkaline phosphatase - HighCreatinine - HighBilirubin, total - High
Randomization Phase: Pemetrexed 500 mg/m21412123122236501
Randomization Phase: Volasertib 300 mg1615183131600001
Randomization Phase: Volasertib 300 mg + Pemetrexed 500 mg/m221250302322610031
Run-in Phase: Volasertib 250 mg + Pemetrexed 500 mg/m2140004400001
Run-in Phase: Volasertib 300 mg + Pemetrexed 500 mg/m2220112411000

[back to top]

Occurrence and Intensity of AEs Graded According to CTCAE.

All patients were carefully monitored during and after each treatment cycle. Adverse events (AEs) were recorded and were graded according to the National Cancer Institute - Common Terminology Criteria for Adverse Events (CTCAE). (NCT00824408)
Timeframe: From first drug infusion until 21 days after last drug infusion, up to 1100 days

,,,,
Interventionparticipants (Number)
CTCAE Grade 1CTCAE Grade 2CTCAE Grade 3CTCAE Grade 4CTCAE Grade 5
Randomization Phase: Pemetrexed 500 mg/m26181550
Randomization Phase: Volasertib 300 mg612863
Randomization Phase: Volasertib 300 mg + Pemetrexed 500 mg/m22181952
Run-in Phase: Volasertib 250 mg + Pemetrexed 500 mg/m200330
Run-in Phase: Volasertib 300 mg + Pemetrexed 500 mg/m202310

[back to top]

Vss of Volasertib

Vss - apparent volume of distribution at steady state following IV administration of volasertib (NCT00824408)
Timeframe: 5 minutes (min) before the start of Volasertib infusion and 1 hour (h), 2h, 4h, 24h, 168h and 336h after the start of Volasertib infusion

InterventionLitres (Geometric Mean)
Volasertib 250 mg + Pemetrexed 500 mg/m26730
Volasertib 300 mg + Pemetrexed 500 mg/m26750
Volasertib 300 mg6230

[back to top]

Overall Survival (OS)

Overall survival (OS) was defined as the duration of time from randomization to time of death. (NCT00824408)
Timeframe: From randomization until time of death

Interventionmonths (Median)
Randomization Phase: Volasertib 300 mg22.9
Randomization Phase: Volasertib 300 mg + Pemetrexed 500 mg/m217.1
Randomization Phase: Pemetrexed 500 mg/m217.4

[back to top]

Progression Free Survival (PFS) Time From the Date of Randomization to Date of Disease Progression or Death, Whichever Occurred First.

"Disease progression was defined according to the Response Evaluation Criteria in Solid Tumours (RECIST)) criteria. Progression-free survival time was calculated as the duration from the date of randomization to the date of disease progression or death, whichever occured first. For patients with known date of progression (or death): PFS [days] = min (date of progression, date of death) - date of randomization + 1 day. For patients without progression or death, PFS was censored at the last imaging date that showed no disease progression: PFS [days, censored] = date of last imaging showing no progression - date randomization + 1 day.~The number of participants analysed displays the number of patients with an event (progression)." (NCT00824408)
Timeframe: From randomization until disease progression or death

Interventionmonths (Median)
Randomization Phase: Volasertib 300 mg1.4
Randomization Phase: Volasertib 300 mg + Pemetrexed 500 mg/m23.3
Randomization Phase: Pemetrexed 500 mg/m25.3

[back to top]

Total Clearance (CL) of Volasertib

CL - total clearance of volasertib in plasma after IV administration (NCT00824408)
Timeframe: 5 minutes (min) before the start of Volasertib infusion and 1 hour (h), 2h, 4h, 24h, 168h and 336h after the start of Volasertib infusion

InterventionmL/min (Geometric Mean)
Volasertib 250 mg + Pemetrexed 500 mg/m2782
Volasertib 300 mg + Pemetrexed 500 mg/m2882
Volasertib 300 mg867

[back to top]

Vss of Pemetrexed

Vss - apparent volume of distribution at steady state following IV administration of pemetrexed (NCT00824408)
Timeframe: 5 minutes before pemetrexed infusion, at the end of the infusion and 1.5 hours (h), 2.5h, 4.5h and 25.5h after the end of pemetrexed infusion

InterventionLitres (Geometric Mean)
Volasertib 250 mg + Pemetrexed 500 mg/m29.40
Volasertib 300 mg + Pemetrexed 500 mg/m213.1

[back to top]

Number of Subjects Who Were Able to Complete Trimodal Therapy (Combination of Surgery, Intrapleural and Systemic Chemotherapy and P-32 Radiotherapy).

To determine the feasibility of multimodal lung sparing regimen. Intrapleural chemotherapy (12 weeks) will be administered within two weeks after surgery. Approximately 30 days post last dose of chemotherapy (+/- 14 days), the patient should be reassessed for resolution of any treatment-related toxicity which may have occurred during the course of study participation. (NCT00859495)
Timeframe: 20 weeks

InterventionParticipants (Count of Participants)
Multimodal Lung Sparing Regimen9

[back to top]

Tumor Best Response Rate

The best response rate will include patients with both Complete Response and Partial Response after 2 months of treatment. 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. (NCT00860015)
Timeframe: Two months

InterventionParticipants (Count of Participants)
Alimta/Gemcitabine0

[back to top]

Objective Response

Evaluation of tumor extent by CT scans, according to RECIST criteria (a 20% decrease in the sum of the longest unidimensional measurements of existing disease), version 1.0 (NCT00864513)
Timeframe: Within two months of the completion of the last dose of chemotherapy

Interventionparticipants (Number)
Chemotherapy0

[back to top]

Number of Participants With Adverse Events

Toxicity by National Cancer Institute Common Toxicity Criteria Adverse Event Version 3.0 (NCT00864513)
Timeframe: 30 days after last dose of study drug

Interventionparticipants (Number)
Chemotherapy2

[back to top]

CA 19-9 Response

CA 19-9 was evaluatd every three weeks, before the next study treatment. Approximately 30% of patients are not expected to have detectable CA 19-9, based on Lews-Y antigen. CA 19-9 response is defined as more than 50% decrease from baseline. (NCT00864513)
Timeframe: Within two months of the last dose of chemotherapy

Interventionparticipants (Number)
Chemotherapy2

[back to top]

Progression-free Survival

Number of days from first dose of study treatment until the date of progression, as measured by worsening disease (new site of disease, or increase in existing disease) or death. (NCT00864513)
Timeframe: 6 months after last patient enrolled

Interventiondays (Median)
Chemotherapy59

[back to top]

The Percentage of Participants Still Living at One Year (One Year Survival Rate)

The one year survival rate is presented as percentage (%) of participants still living at one year and is the number of participants that are still alive at one year divided by the number of participants in the protocol qualified (PQ) population, which is then multiplied by 100. (NCT00867009)
Timeframe: One year

Interventionpercentage of participants (Number)
Pem/Cis + Cet45

[back to top]

Progression-free Survival (PFS)

PFS is measured from study entry until disease progression, death or date of last contact. Progressive disease (PD) was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. PD = 20% increase in sum of longest diameter of target lesions or the appearance of 1 or more new lesions and/or unequivocal progression of existing non-target lesions. For participants not known to have died or have had objective PD as of the data cutoff date, PFS was censored at the date of the last objective progression-free disease assessment. (NCT00867009)
Timeframe: From start of treatment until documented disease progression or death from any cause (up to 18.9 months)

Interventionmonths (Median)
Pem/Cis + Cet5.82

[back to top]

Percentage of Participants With a Tumor Response (Objective Tumor Response Rate)

Response was assessed using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response (CR)=disappearance of all target lesions; Partial Response (PR)=30% decrease in sum of longest diameter of target lesions. Tumor response is presented as a percentage (%) and is the number of participants with a CR plus PR divided by the number of participants in the protocol qualified (PQ) population, then multiplied by 100. (NCT00867009)
Timeframe: From start of treatment until documented best response. (up to 18.9 months)

Interventionpercentage of participants (Number)
Pem/Cis + Cet38.5

[back to top]

The Percentage of Participants With Complete Response (CR), Partial Response (PR) or Stable Disease (SD) (Disease Control Rate [DCR])

The DCR is presented as percentage (%) and is the number of participants with a best tumor response of CR, PR, or SD divided by the number of participants in the protocol qualified (PQ) population, then multiplied by 100. Best tumor response of CR, PR, or SD was determined from the sequence of tumor response assessments. Tumor response was assessed using RECIST criteria. CR=disappearance of all target lesions; PR=30% decrease in sum of longest diameter of target lesions; SD=small changes that do not meet above criteria. (NCT00867009)
Timeframe: From start of treatment until documented best tumor response (up to 18.9 months)

Interventionpercentage of participants (Number)
Pem/Cis + Cet59.6

[back to top]

Overall Survival (OS)

OS = observed length of time from entry into the study to death or date of last contact (NCT00868192)
Timeframe: Median follow-up was 25.7 months (range 3.0-47.2 months)

Interventionmonths (Median)
Pemetrexed and Bevacizumab25.7

[back to top]

Overall Response Rate

"Overall response rate = complete response + partial response~Complete response = disappearance of all target and non-target lesions and no evidence of new lesions documented by two disease assessments at least 4 weeks apart.~Partial response = at least a 30% decrease in the sum of the longest dimensions (LD) of all target measurable lesions taking as reference the baseline sum of LD. There can be non unequivocal progression of non-target lesions and no new lesions." (NCT00868192)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Pemetrexed and Bevacizumab41

[back to top]

Overall Survival (OS)

OS = observed length of time from entry into the study to death or date of last contact (NCT00868192)
Timeframe: 12 months

Interventionpercentage of participants (Number)
Pemetrexed and Bevacizumab79

[back to top]

Progression-free Survival (PFS)

PFS = Period from study entry until disease progression, death, or date of last contact (NCT00868192)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Pemetrexed and Bevacizumab56

[back to top]

Progression-free Survival (PFS)

PFS = Period from study entry until disease progression, death, or date of last contact (NCT00868192)
Timeframe: Median follow-up was 25.7 months (range 3.0-47.2 months)

Interventionmonths (Median)
Pemetrexed and Bevacizumab7.9

[back to top]

CA-125 Response

A CA-125 response was defined as at least a 50% reduction in CA-125 levels from a pretreatment sample following guidelines described by the Gynecological Cancer Intergroup. (NCT00868192)
Timeframe: 6 months

Interventionparticipants (Number)
50% CA-125 response75% CA-125 responseNo CA-125 response
Pemetrexed and Bevacizumab1782

[back to top]

Distribution of Overall Survival (OS)

OS = observed length of time from entry into the study to death or date of last contact (NCT00868192)
Timeframe: Median follow-up was 25.7 months (range 3.0-47.2 months)

Interventionmonths (Median)
Platinum-free interval of <6 monthsPlatinum-free interval of 6-12 monthsPlatinum-free interval of >12 months
Pemetrexed and Bevacizumab16.724.928.0

[back to top]

Distribution of Progression-free Survival (PFS)

PFS = Period from study entry until disease progression, death, or date of last contact (NCT00868192)
Timeframe: Median follow-up was 25.7 months (range 3.0-47.2 months)

Interventionmonths (Median)
Platinum-free interval of <6 monthsPlatinum-free interval of 6-12 monthsPlatinum-free interval of >12 months
Pemetrexed and Bevacizumab6.74.716.8

[back to top]

Toxicity Associated With Bevacizumab and Pemetrexed

Detailed serious adverse events and other adverse events are shown in the adverse event module of the results. (NCT00868192)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Grade 3/4 hematologic toxicityMost common non-hematologic toxicity - fatigueGrade 3 renal toxicityGastrointestinal toxicitySubsequently developed hematologic malignancies
Pemetrexed and Bevacizumab53946916

[back to top]

Frequency of Clinical Response

As measured by RECIST criteria (NCT00868192)
Timeframe: 6 months

Interventionparticipants (Number)
Complete responsePartial responseStable diseaseProgressive disease
Pemetrexed and Bevacizumab014182

[back to top]

Best Overall Response, Assessed as the Number of Participants With the Indicated Tumor Response: Investigator Assessed Only

Tumor response was assessed by the Investigator according to the RECIST, version 1.0. A participant was defined as a responder if he/she sustained a complete response (CR; the disappearance of all target lesions) or partial response (PR; >=30% decrease in the sum of the longest diameter of target lesions) for at least 4 weeks at any time during randomized treatment. Stable disease is defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum longest diameter since the treatment started. (NCT00871403)
Timeframe: Randomization until response or progressive disease (up to 85 weeks)

,
Interventionparticipants (Number)
Complete responsePartial responseStable diseaseProgressive diseaseUnknown
Cisplatin 75 mg/m^2 Plus Pemetrexed 500 mg/m^20121454
Pazopanib 800 mg Plus Pemetrexed 500 mg/m^201413629

[back to top]

Progression-free Survival (PFS)

PFS is defined as the interval between the date of randomization (date on which the investigator evaluated the participant and first determined he/she had disease progression) and the first occurrence of progressive disease (PD) or death from any cause. Per Response Evaluation Criteria in Solid Tumors (RECIST), version 1, PD is defined as 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 >=1 new lesion). (NCT00871403)
Timeframe: Randomization until progression or death (up to 85 weeks)

Interventionweeks (Median)
Pazopanib 800 mg Plus Pemetrexed 500 mg/m^225.0
Cisplatin 75 mg/m^2 Plus Pemetrexed 500 mg/m^222.9

[back to top]

Percentage of Participants With a Complete Response or a Partial Response

The percentage of participants with a complete response or a partial response was evaluated. (NCT00871403)
Timeframe: Randomization until response or progressive disease (up to 85 weeks)

Interventionpercentage of participants (Number)
Pazopanib 800 mg Plus Pemetrexed 500 mg/m^214
Cisplatin 75 mg/m^2 Plus Pemetrexed 500 mg/m^212

[back to top]

Percentage of Participants With Concordance Between Local and Central Histological Diagnosis

A centralized pathology review on all enrolled participants was performed to confirm the histological diagnosis performed at the site. Upon review of the local diagnosis obtained at the respective site, the central reviewer established whether or not there was an agreement between the local and central diagnosis. The percentage of participants with concordance was defined as the number of participants for which there was an agreement divided by the number of treated participants (concordance rate) multiplied by 100. (NCT00887549)
Timeframe: Baseline

Interventionpercentage of participants (Number)
Pemetrexed78.6

[back to top]

Percentage of Participants With Tumor Response (Tumor Response Rate)

Tumor response was assessed using Response Evaluation Criteria In Solid Tumors (RECIST 1.0) criteria. Complete Response=disappearance of all target lesions; Partial Response=30% decrease in sum of longest diameter of target lesions; Progressive Disease=20% increase in sum of longest diameter of target lesions; Stable Disease=small changes that do not meet above criteria. Percentage of participants with tumor response was determined by the number of participants with PR or CR (confirmed or not) divided by the total number of treated participants multiplied by 100. (NCT00887549)
Timeframe: Baseline to disease progression (up to 20 months)

Interventionpercentage of participants (Number)
Pemetrexed30

[back to top]

Progression Free Survival (PFS)

PFS is time from first dose to first observation of disease progression/death (any cause). PFS is reported for participants with thymidylate synthase (TS) scores. For participants not known to have died by the data cut-off date and who do not have progressive disease, PFS will be censored at date of last objective progression-free disease assessment. For participants who receive systemic anticancer therapy after study drug discontinuation and prior to disease progression/death, PFS will be censored at date of last objective progression-free disease assessment prior to chemotherapy. (NCT00887549)
Timeframe: Baseline to measured progressive disease with follow-up every 6 weeks until progression of disease (up to 18 months after the last participant commenced induction therapy)

Interventionmonths (Median)
Pemetrexed5.5

[back to top]

Percentage of Participants Surviving at 18 Months (Overall Survival Rate)

The percentage of participants surviving at 18 months was defined as the number of treated participants who had not died prior to 18 months from the date of their first dose divided by the total number of treated participants multiplied by 100. For participants who are alive, overall survival was censored at the last contact. (NCT00887549)
Timeframe: Baseline to date of death (up to 24.5 months)

Interventionpercentage of participants (Number)
Pemetrexed37.1

[back to top]

Overall Survival (OS)

The Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Death (NCT00892710)
Timeframe: 18 months

Interventionmonths (Median)
Pemetrexed7.7
Pemetrexed/Bevacizumab8.6
Pemetrexed/Bevacizumab/Carboplatin8.7

[back to top]

Time to Treatment Failure (TTTF)

Defined as the Length of Time, in Months, that Patients were Alive from the Date of First Treatment Until Treatment Discontinuation for Any Reason. (NCT00892710)
Timeframe: 18 months

Interventionmonths (Median)
Pemetrexed2.4
Pemetrexed/Bevacizumab3.1
Pemetrexed/Bevacizumab/Carboplatin3.3

[back to top]

6-month and 12-month Overall Survival Probability

Overall Survival = The Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Death (NCT00892710)
Timeframe: 12 months

,,
Interventionprobability out of 1 (Number)
6-month OS probability12-month OS probability
Pemetrexed0.520.3
Pemetrexed/Bevacizumab0.610.32
Pemetrexed/Bevacizumab/Carboplatin0.570.44

[back to top]

Time to Progression (TTP)

The Length of Time, in Months, That Patients Remain Alive From Their First Date of Protocol Treatment Until Worsening of Their Disease. 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. (NCT00892710)
Timeframe: 18 months

Interventionmonths (Median)
Pemetrexed3.5
Pemetrexed/Bevacizumab5.3
Pemetrexed/Bevacizumab/Carboplatin5.7

[back to top]

Progression Free Survival (PFS)

The Length of Time, in Months, That Patients Were Alive From Their First Date of Protocol Treatment Until Worsening of Their Disease. 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. (NCT00892710)
Timeframe: 18 months

Interventionmonths (Median)
Pemetrexed2.8
Pemetrexed/Bevacizumab4.0
Pemetrexed/Bevacizumab/Carboplatin4.8

[back to top]

Overall Response Rate (ORR), the Number of Patients Who Experience an Objective Benefit From Treatment

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions and assessed by MRI or CT: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT00892710)
Timeframe: 18 months

Interventionparticipants (Number)
Pemetrexed7
Pemetrexed/Bevacizumab18
Pemetrexed/Bevacizumab/Carboplatin24

[back to top]

Rate of Residual Disease as an Assessment of Pathological Partial Response (pPR)

pPR was further assessed by the amount of residual tumor measured at surgery: microscopic residual disease = less than 1 centimeter (<1 cm); macroscopic residual disease = 1 centimeter or greater (≥1 cm). (NCT00906282)
Timeframe: At 15-18 weeks

Interventioncentimeters (Median)
Pemetrexed/Carboplatin/Surgery2.5

[back to top]

3-Year Overall Survival Rate

The percentage of patients who were alive at 3 years from time of first study treatment until date of death from any cause. Overall survival is shown for the Intent-to-Treat population. (NCT00906282)
Timeframe: 36 months

Interventionpercentage of participants (Number)
Pemetrexed/Carboplatin/Surgery45

[back to top]

Objective Tumor Response

Objective Tumor Response defined as the percent of patients who completed up to 4 cycles of pre-operative chemotherapy and achieved a complete response (CR) or partial response (PR) assessed by Response Evaluation in Solid Tumors (RECIST) 1.0. Patients with stable disease (SD) or response to treatment were deemed surgical candidates. [CR=disappearance of all target tumors; PR= ≥30% decrease in the sum of the longest diameters of target tumors. 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.] (NCT00906282)
Timeframe: At 6 and 12 weeks

Interventionpercentage of participants (Number)
PRSD
Pemetrexed/Carboplatin/Surgery4148

[back to top]

Complete Resection Rate

The percent of patients who had surgical resection listed by procedure type: lobectomy or pneumonectomy, or resection of adjacent chest wall or mediastinal structures when appropriate. Surgery followed standard guidelines for resection of non-small-cell lung cancer (NSCLC). (NCT00906282)
Timeframe: At weeks 15-18

Interventionpercentage of patients (Number)
LobectomyWedge ResectionPneumonectomyBilobectomy
Pemetrexed/Carboplatin/Surgery7015114

[back to top]

Pathologic Response Rate

Percent of patients having a pathological complete or partial response (pCR or pPR) at surgery. pCR defined as complete removal of all tumor. pPR defined as residual viable tumor demonstrated in the resected specimen. (NCT00906282)
Timeframe: weeks 15 -18

Interventionpercentage of participants (Number)
pPRpCR
Pemetrexed/Carboplatin/Surgery1000

[back to top]

Phase I Only: Maximum Tolerated Dose (MTD) of Pemetrexed That Could be Administered Weekly in Combination With Temsirolimus

The starting dose and schedule of pemetrexed will be 500 mg/m2 given every 3 weeks and the starting dose for temsirolimus will be 15 mg given weekly for 3 weeks, to complete 1 cycle. In subsequent cohorts, dose will be escalated or de-escalated. Enrollment to each cohort is based on toxicity experienced at that dose level. The maximum tolerated dose (MTD) is defined as the dose level immediately below the dose level at which 2 patients of a cohort experience dose-limiting toxicity during the first cycle. Six patients will be enrolled at the maximum tolerated dose to ensure that no more than 2 DLTs occur at the MTD. Dose escalations will proceed until the MTD has been reached. (NCT00921310)
Timeframe: Completion of first cycle by all enrolled patients in Phase I portion of study

Interventionmg/m^2 (Number)
Phase I (Premetrexed & Temsirolimus)375

[back to top]

Phase I Only: Maximum Tolerated Dose (MTD) of Temsirolimus That Could be Administered Weekly in Combination With Pemetrexed

The starting dose and schedule of pemetrexed will be 500 mg/m2 given every 3 weeks and the starting dose for temsirolimus will be 15 mg given weekly for 3 weeks, to complete 1 cycle. In subsequent cohorts, dose will be escalated or de-escalated. Enrollment to each cohort is based on toxicity experienced at that dose level. The maximum tolerated dose (MTD) is defined as the dose level immediately below the dose level at which 2 patients of a cohort experience dose-limiting toxicity during the first cycle. Six patients will be enrolled at the maximum tolerated dose to ensure that no more than 2 DLTs occur at the MTD. Dose escalations will proceed until the MTD has been reached. (NCT00921310)
Timeframe: Completion of first cycle by all enrolled patients in Phase I portion of study

Interventionmg (Number)
Phase I (Premetrexed & Temsirolimus)15

[back to top]

Phase I Only: Number of Participants Who Experience Dose-limiting Toxicities (DLT) of Temsirolimus and Pemetrexed

"DLT will be defined as occurring within the first cycle of Phase I only and will be graded according to the Common Terminology Criteria for Adverse Events v 3.0 (CTCAE)~Any grade 3 or higher hematologic toxicity with the exception of anemia.~Any grade 3 or higher non-hematologic toxicity related to study therapy (except alopecia).~Grade 3 or 4 pneumonitis or esophagitis.~Treatment delay of temsirolimus for more than 14 consecutive days due to study-related toxicity.~Treatment delay of pemetrexed therapy for more than 14 consecutive days because of study-related toxicity." (NCT00921310)
Timeframe: Completion of first cycle (approximately 21 days)

Interventionparticipants (Number)
Phase I Dose Level 1 (Premetrexed & Temsirolimus)2
Phase 1 Dose Level -1 (Pemetrexed & Temsirolimus)0

[back to top]

Phase 2 Only: Progression-free Survival (PFS)

PFS is defined as the duration of time from start of treatment to time of progression. (NCT00921310)
Timeframe: 2 years from completion of treatment

Interventiondays (Number)
Patient APatient B
Phase 2 (Pemetrexed & Temsirolimus)523167

[back to top]

Phase I and Phase II: Overall Response Rate (Complete Response + Partial Response)

"Only those patients who have measurable disease present at baseline, have received at least one cycle of therapy, and have had their disease re-evaluated will be considered evaluable for response.~Complete response (CR)-disappearance of all target lesions and disappearance of all non-target lesions and normalization of tumor marker level.~Partial response (PR)-at least a 30% decrease in the sum of the longest diameter (LD) of the target lesions taking as reference the baseline sum LD" (NCT00921310)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Phase I (Premetrexed & Temsirolimus)0
Phase 2 (Pemetrexed & Temsirolimus)33.3

[back to top]

Phase 2 Only: Survival Rate

(NCT00921310)
Timeframe: 1 year after start of treatment

Interventionpercentage of participants (Number)
Phase 2 (Pemetrexed & Temsirolimus)50

[back to top]

Phase II: Clinical Response Rate

Clinical response is assessed by the Response Evaluation Criteria in Solid Tumors (RECIST). Complete response (CR) is 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. Stable disease (SD) is 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. Progressive disease (PD) is 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. (NCT00923273)
Timeframe: 21 weeks

InterventionParticipants (Count of Participants)
Complete responsePartial responseStable diseaseProgressive diseaseNot evaluable
Treatment Level 4: 10mg Load051327

[back to top]

Number of Participants With Serious and Non-Serious Adverse Events

Here is the count of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria for Adverse Events (CTCAE v3.0). 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. (NCT00923273)
Timeframe: Date treatment consent signed to date off study, approximately 45 months

,,,,
InterventionParticipants (Count of Participants)
Subjects from phase I8subjects prior peme & 12 subjects peme naive
Treatment Level 1: 3mg Load40
Treatment Level 2: 6mg Load30
Treatment Level 3: 6mg Load30
Treatment Level 4: 10 mg Load720
Treatment Level 5: 15mg Load50

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Sirolimus

The phase I component of the study are to determine the safety and tolerability of sirolimus in human subjects with non small cell lung cancer (NSCLC), and to determine the maximum tolerated dose. (NCT00923273)
Timeframe: 5 weeks

Interventionmg/m^2 (Number)
Treatment Level 1: 3mg Load10
Treatment Level 2: 6mg Load10
Treatment Level 3: 6mg Load10
Treatment Level 4: 10mg Load10
Treatment Level 5: 15mg Load10

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of Pemetrexed

The phase I component of the study are to determine the safety and tolerability of pemetrexed in human subjects with non small cell lung cancer (NSCLC), and to determine the maximum tolerated dose. (NCT00923273)
Timeframe: 5 weeks

Interventionmg/m^2 (Number)
Treatment Level 1: 3mg Load500
Treatment Level 2: 6mg Load500
Treatment Level 3: 6mg Load500
Treatment Level 4: 10mg Load500
Treatment Level 5: 15mg Load500

[back to top]

Percentage of Participants With Objective Response (OR)

Percentage of participants with objective response based on assessment of complete response (CR) or partial response (PR) according to RECIST v1.1. CR: disappearance of all target and non-target lesions and normalization of tumor marker level, all lymph nodes must be non-pathological in size (<10 millimeter [mm] short axis). PR: at least 30 percent (%) decrease in sum of diameters of target lesions, taking as reference the baseline sum diameters persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits. Objective response is based on independent radiology review. (NCT00932893)
Timeframe: Randomization until PD or initiation of antitumor therapy in the absence of PD or death, assessed every 6 weeks (up to 112 weeks)

Interventionpercentage of participants (Number)
Crizotinib65.3
Chemotherapy19.5

[back to top]

Progression-Free Survival (PFS)

PFS: Time in months from randomization to first documentation of objective disease progression as determined by independent radiology review or to death due to any cause, whichever occurred first. PFS was calculated as (first event date minus the date of randomization plus 1) divided by 30.4. Progression is defined using Response Evaluation Criteria in Solid Tumors Criteria version 1.1 (RECIST v1.1), as at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of diameters of target lesions, taking as reference the smallest sum on study and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT00932893)
Timeframe: Randomization until progressive disease (PD) or initiation of antitumor therapy in the absence of PD or death, assessed every 6 weeks (up to 112 weeks)

Interventionmonths (Median)
Crizotinib7.7
Chemotherapy3.0

[back to top]

Time to Deterioration (TTD) in Participant Reported Pain, Dyspnea, and Cough

TTD in pain (pain in chest from European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire-Supplement Module for Lung Cancer [EORTC QLQ-LC13]), dyspnea (from EORTC QLQ-LC13), or cough (from EORTC QLQ-LC13) symptoms was defined as the time from randomization to the earliest time the participant's score showed a 10 point or higher increase from baseline in any of the three symptoms from the instrument. The transformed score of pain, dyspnea, and cough symptom scales of EORTC QLQ-LC13 range from 0 to 100, greater scores = higher symptom severity. (NCT00932893)
Timeframe: Baseline up to end of treatment (up to 112 weeks)

Interventionmonths (Median)
Crizotinib4.5
Chemotherapy1.4

[back to top]

Percentage of Participants With Disease Control at Week 12

Disease control: participants with CR, PR, or SD according to RECIST v1.1. CR: disappearance of all target and non-target lesions and normalization of tumor marker level, all lymph nodes must be non-pathological in size (<10 mm short axis). PR: at least 30 % decrease in sum of diameters of target lesions, taking as reference the baseline sum diameters persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits. 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. PD: at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of diameters of target lesions, taking as reference the smallest sum on study and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT00932893)
Timeframe: Week 12

Interventionpercentage of participants (Number)
Crizotinib64.2
Chemotherapy38.5

[back to top]

Time to Tumor Response (TTR)

Time from date of randomization to first documentation of objective tumor response. TTR was calculated for the subgroup of participants with objective tumor response. Objective tumor response was defined as CR or PR according to RECIST v1.1. CR: disappearance of all target and non-target lesions and normalization of tumor marker level, all lymph nodes must be non-pathological in size (<10 mm short axis). PR: at least 30 % decrease in sum of diameters of target lesions, taking as reference the baseline sum diameters persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits. (NCT00932893)
Timeframe: Randomization until PD or initiation of antitumor therapy in the absence of PD or death, assessed every 6 weeks (up to 112 weeks)

Interventionweeks (Median)
Crizotinib6.3
Chemotherapy12.6

[back to top]

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

EORTC QLQ-C30: included global health status/quality of life (QoL), functional scales (physical, role, cognitive, emotional, and social), symptom scales (fatigue, pain, nausea/vomiting), and single items (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 averaged, transformed to 0-100 scale; higher score for Global Qol/functional scales=better level of QoL/functioning or higher score for symptom scale=greater degree of symptoms. (NCT00932893)
Timeframe: Baseline, Day (D) 1 of each cycle (C) until disease progression, end of treatment (EOT, up to 112 weeks)

,
Interventionunits on a scale (Mean)
Global QoL: Baseline (n=165, 162)Global QoL: C2D1 (n=154, 133)Global QoL: C3D1 (n=154, 106)Global QoL: C4D1 (n=135, 90)Global QoL: C5D1 (n=123, 74)Global QoL: C6D1 (n=120, 70)Global QoL: C7D1 (n=114, 52)Global QoL: C8D1 (n=110, 43)Global QoL: C9D1 (n=101, 37)Global QoL: C10D1 (n=94, 33)Global QoL: C11D1 (n=83, 25)Global QoL: C12D1 (n=76, 23)Global QoL: C13D1 (n=74, 21)Global QoL: C14D1 (n=66, 19)Global QoL: C15D1 (n=62, 16)Global QoL: C16D1 (n=53, 12)Global QoL: C17D1 (n=47, 11)Global QoL: C18D1 (n=44, 11)Global QoL: C19D1 (n=40, 9)Global QoL: C20D1 (n=35, 8)Global QoL: C21D1 (n=30, 8)Global QoL: C22D1 (n=24, 7)Global QoL: C23D1 (n=23, 4)Global QoL: C24D1 (n=20, 3)Global QoL: C25D1 (n=18, 3)Global QoL: C26D1 (n=14, 3)Global QoL: C27D1 (n=14, 2)Global QoL: C28D1 (n=11, 2)Global QoL: C29D1 (n=8, 2)Global QoL: C30D1 (n=8, 1)Global QoL: C31D1 (n=7, 0)Global QoL: C32D1 (n=6, 0)Global QoL: C33D1 (n=6, 0)Global QoL: C34D1 (n=5, 0)Global QoL: C35D1 (n=4, 0)Global QoL: C36D1 (n=1, 0)Global QoL: C37D1 (n=1, 0)Physical Functioning: Baseline (n=165, 163)Physical Functioning: C2D1 (n=155, 133)Physical Functioning: C3D1 (n=154, 106)Physical Functioning: C4D1 (n=135, 91)Physical Functioning: C5D1 (n=123, 74)Physical Functioning: C6D1 (n=120, 70)Physical Functioning: C7D1 (n=115, 52)Physical Functioning: C8D1 (n=111, 43)Physical Functioning: C9D1 (n=101, 37)Physical Functioning: C10D1 (n=94, 33)Physical Functioning: C11D1 (n=84, 25)Physical Functioning: C12D1 (n=76, 23)Physical Functioning: C13D1 (n=74, 21)Physical Functioning: C14D1 (n=66, 19)Physical Functioning: C15D1 (n=62, 16)Physical Functioning: C16D1 (n=53, 12)Physical Functioning: C17D1 (n=47, 11)Physical Functioning: C18D1 (n=45, 11)Physical Functioning: C19D1 (n=40, 9)Physical Functioning: C20D1 (n=35, 8)Physical Functioning: C21D1 (n=30, 8)Physical Functioning: C22D1 (n=24, 7)Physical Functioning: C23D1 (n=23, 4)Physical Functioning: C24D1 (n=20, 3)Physical Functioning: C25D1 (n=18, 3)Physical Functioning: C26D1 (n=14, 3)Physical Functioning: C27D1 (n=14, 2)Physical Functioning: C28D1 (n=11, 2)Physical Functioning: C29D1 (n=8, 2)Physical Functioning: C30D1 (n=8, 1)Physical Functioning: C31D1 (n=7, 0)Physical Functioning: C32D1 (n=6, 0)Physical Functioning: C33D1 (n=6, 0)Physical Functioning: C34D1 (n=5, 0)Physical Functioning: C35D1 (n=4, 0)Physical Functioning: C36D1 (n=1, 0)Physical Functioning: C37D1 (n=1, 0)Role Functioning: Baseline (n=165, 163)Role Functioning: C2D1 (n=155, 133)Role Functioning: C3D1 (n=154, 106)Role Functioning: C4D1 (n=134, 91)Role Functioning: C5D1 (n=123, 74)Role Functioning: C6D1 (n=120, 70)Role Functioning: C7D1 (n=115, 52)Role Functioning: C8D1 (n=111, 43)Role Functioning: C9D1 (n=101, 37)Role Functioning: C10D1 (n=94, 33)Role Functioning: C11D1 (n=84, 25)Role Functioning: C12D1 (n=76, 23)Role Functioning: C13D1 (n=74, 21)Role Functioning: C14D1 (n=66, 19)Role Functioning: C15D1 (n=62, 16)Role Functioning: C16D1 (n=53, 12)Role Functioning: C17D1 (n=47, 11)Role Functioning: C18D1 (n=45, 11)Role Functioning: C19D1 (n=40, 9)Role Functioning: C20D1 (n=35, 8)Role Functioning: C21D1 (n=30, 8)Role Functioning: C22D1 (n=24, 7)Role Functioning: C23D1 (n=23, 4)Role Functioning: C24D1 (n=20, 3)Role Functioning: C25D1 (n=18, 3)Role Functioning: C26D1 (n=14, 3)Role Functioning: C27D1 (n=14, 2)Role Functioning: C28D1 (n=11, 2)Role Functioning: C29D1 (n=8, 2)Role Functioning: C30D1 (n=8, 1)Role Functioning: C31D1 (n=7, 0)Role Functioning: C32D1 (n=6, 0)Role Functioning: C33D1 (n=6, 0)Role Functioning: C34D1 (n=5, 0)Role Functioning: C35D1 (n=4, 0)Role Functioning: C36D1 (n=1, 0)Role Functioning: C37D1 (n=1, 0)Emotional Functioning: Baseline (n=165, 162)Emotional Functioning: C2D1 (n=155, 133)Emotional Functioning: C3D1 (n=154, 106)Emotional Functioning: C4D1 (n=135, 90)Emotional Functioning: C5D1 (n=123, 74)Emotional Functioning: C6D1 (n=120, 70)Emotional Functioning: C7D1 (n=115, 52)Emotional Functioning: C8D1 (n=111, 43)Emotional Functioning: C9D1 (n=101, 37)Emotional Functioning: C10D1 (n=94, 33)Emotional Functioning: C11D1 (n=84,25)Emotional Functioning: C12D1 (n=76, 23)Emotional Functioning: C13D1 (n=74, 21)Emotional Functioning: C14D1 (n=66, 19)Emotional Functioning: C15D1 (n=62, 16)Emotional Functioning: C16D1 (n=53, 12)Emotional Functioning: C17D1 (n=47, 11)Emotional Functioning: C18D1 (n=44, 11)Emotional Functioning: C19D1 (n=40, 9)Emotional Functioning: C20D1 (n=35, 8)Emotional Functioning: C21D1 (n=30, 8)Emotional Functioning: C22D1 (n=24, 7)Emotional Functioning: C23D1 (n=23, 4)Emotional Functioning: C24D1 (n=20, 3)Emotional Functioning: C25D1 (n=18, 3)Emotional Functioning: C26D1 (n=14, 3)Emotional Functioning: C27D1 (n=14, 2)Emotional Functioning: C28D1 (n=11, 2)Emotional Functioning: C29D1 (n=8, 2)Emotional Functioning: C30D1 (n=8, 1)Emotional Functioning: C31D1 (n=7, 0)Emotional Functioning: C32D1 (n=6, 0)Emotional Functioning: C33D1 (n=6, 0)Emotional Functioning: C34D1 (n=5, 0)Emotional Functioning: C35D1 (n=4, 0)Emotional Functioning: C36D1 (n=1, 0)Emotional Functioning: C37D1 (n=1, 0)Cognitive Functioning: Baseline (n=165, 162)Cognitive Functioning: C2D1 (n=155, 133)Cognitive Functioning: C3D1 (n=154, 106)Cognitive Functioning: C4D1 (n=135, 90)Cognitive Functioning: C5D1 (n=123, 74)Cognitive Functioning: C6D1 (n=120, 70)Cognitive Functioning: C7D1 (n=115, 52)Cognitive Functioning: C8D1 (n=111, 43)Cognitive Functioning: C9D1 (n=101, 37)Cognitive Functioning: C10D1 (n=94, 33)Cognitive Functioning: C11D1 (n=84, 25)Cognitive Functioning: C12D1 (n=76, 23)Cognitive Functioning: C13D1 (n=74, 21)Cognitive Functioning: C14D1 (n=66, 19)Cognitive Functioning: C15D1 (n=62, 16)Cognitive Functioning: C16D1 (n=53, 12)Cognitive Functioning: C17D1 (n=47, 11)Cognitive Functioning: C18D1 (n=44, 11)Cognitive Functioning: C19D1 (n=40, 9)Cognitive Functioning: C20D1 (n=35, 8)Cognitive Functioning: C21D1 (n=30, 8)Cognitive Functioning: C22D1 (n=24, 7)Cognitive Functioning: C23D1 (n=23, 4)Cognitive Functioning: C24D1 (n=20, 3)Cognitive Functioning: C25D1 (n=18, 3)Cognitive Functioning: C26D1 (n=14, 3)Cognitive Functioning: C27D1 (n=14, 2)Cognitive Functioning: C28D1 (n=11, 2)Cognitive Functioning: C29D1 (n=8, 2)Cognitive Functioning: C30D1 (n=8, 1)Cognitive Functioning: C31D1 (n=7, 0)Cognitive Functioning: C32D1 (n=6, 0)Cognitive Functioning: C33D1 (n=6, 0)Cognitive Functioning: C34D1 (n=5, 0)Cognitive Functioning: C35D1 (n=4, 0)Cognitive Functioning: C36D1 (n=1, 0)Cognitive Functioning: C37D1 (n=1, 0)Social Functioning: Baseline (n=165, 162)Social Functioning: C2D1 (n=155, 133)Social Functioning: C3D1 (n=154, 106)Social Functioning: C4D1 (n=135, 90)Social Functioning: C5D1 (n=123, 74)Social Functioning: C6D1 (n=120, 70)Social Functioning: C7D1 (n=115, 52)Social Functioning: C8D1 (n=111, 43)Social Functioning: C9D1 (n=101, 37)Social Functioning: C10D1 (n=94, 33)Social Functioning: C11D1 (n=84, 25)Social Functioning: C12D1 (n=76, 23)Social Functioning: C13D1 (n=74, 21)Social Functioning: C14D1 (n=66, 19)Social Functioning: C15D1 (n=62, 16)Social Functioning: C16D1 (n=53, 12)Social Functioning: C17D1 (n=47, 11)Social Functioning: C18D1 (n=44, 11)Social Functioning: C19D1 (n=40, 9)Social Functioning: C20D1 (n=35, 8)Social Functioning: C21D1 (n=30, 8)Social Functioning: C22D1 (n=24, 7)Social Functioning: C23D1 (n=23, 4)Social Functioning: C24D1 (n=20, 3)Social Functioning: C25D1 (n=18, 3)Social Functioning: C26D1 (n=14, 3)Social Functioning: C27D1 (n=14, 2)Social Functioning: C28D1 (n=11, 2)Social Functioning: C29D1 (n=8, 2)Social Functioning: C30D1 (n=8, 1)Social Functioning: C31D1 (n=7, 0)Social Functioning: C32D1 (n=6, 0)Social Functioning: C33D1 (n=6, 0)Social Functioning: C34D1 (n=5, 0)Social Functioning: C35D1 (n=4, 0)Social Functioning: C36D1 (n=1, 0)Social Functioning: C37D1 (n=1, 0)Fatigue: Baseline (n=165, 163)Fatigue: C2D1 (n=155, 133)Fatigue: C3D1 (n=154, 106)Fatigue: C4D1 (n=135, 91)Fatigue: C5D1 (n=123, 74)Fatigue: C6D1 (n=120, 70)Fatigue: C7D1 (n=115, 52)Fatigue: C8D1 (n=111, 43)Fatigue: C9D1 (n=101, 37)Fatigue: C10D1 (n=94, 33)Fatigue: C11D1 (n=84, 25)Fatigue: C12D1 (n=76, 23)Fatigue: C13D1 (n=74, 21)Fatigue: C14D1 (n=66, 19)Fatigue: C15D1 (n=62, 16)Fatigue: C16D1 (n=53, 12)Fatigue: C17D1 (n=47, 11)Fatigue: C18D1 (n=45, 11)Fatigue: C19D1 (n=40, 9)Fatigue: C20D1 (n=35, 8)Fatigue: C21D1 (n=30, 8)Fatigue: C22D1 (n=24, 7)Fatigue: C23D1 (n=23, 4)Fatigue: C24D1 (n=20, 3)Fatigue: C25D1 (n=18, 3)Fatigue: C26D1 (n=14, 3)Fatigue: C27D1 (n=14, 2)Fatigue: C28D1 (n=11, 2)Fatigue: C29D1 (n=8, 2)Fatigue: C30D1 (n=8, 1)Fatigue: C31D1 (n=7, 0)Fatigue: C32D1 (n=6, 0)Fatigue: C33D1 (n=6, 0)Fatigue: C34D1 (n=5, 0)Fatigue: C35D1 (n=4, 0)Fatigue: C36D1 (n=1, 0)Fatigue: C37D1 (n=1, 0)Nausea and Vomiting: Baseline (n=165, 163)Nausea and Vomiting: C2D1 (n=155, 133)Nausea and Vomiting: C3D1 (n=154, 106)Nausea and Vomiting: C4D1 (n=135, 91)Nausea and Vomiting: C5D1 (n=123, 74)Nausea and Vomiting: C6D1 (n=120, 70)Nausea and Vomiting: C7D1 (n=115, 52)Nausea and Vomiting: C8D1 (n=111, 43)Nausea and Vomiting: C9D1 (n=101, 37)Nausea and Vomiting: C10D1 (n=94, 33)Nausea and Vomiting: C11D1 (n=84, 25)Nausea and Vomiting: C12D1 (n=76, 23)Nausea and Vomiting: C13D1 (n=74, 21)Nausea and Vomiting: C14D1 (n=66, 19)Nausea and Vomiting: C15D1 (n=62, 16)Nausea and Vomiting: C16D1 (n=53, 12)Nausea and Vomiting: C17D1 (n=47, 11)Nausea and Vomiting: C18D1 (n=45, 11)Nausea and Vomiting: C19D1 (n=40, 9)Nausea and Vomiting: C20D1 (n=35, 8)Nausea and Vomiting: C21D1 (n=30, 8)Nausea and Vomiting: C22D1 (n=24, 7)Nausea and Vomiting: C23D1 (n=23, 4)Nausea and Vomiting: C24D1 (n=20, 3)Nausea and Vomiting: C25D1 (n=18, 3)Nausea and Vomiting: C26D1 (n=14, 3)Nausea and Vomiting: C27D1 (n=14, 2)Nausea and Vomiting: C28D1 (n=11, 2)Nausea and Vomiting: C29D1 (n=8, 2)Nausea and Vomiting: C30D1 (n=8, 1)Nausea and Vomiting: C31D1 (n=7, 0)Nausea and Vomiting: C32D1 (n=6, 0)Nausea and Vomiting: C33D1 (n=6, 0)Nausea and Vomiting: C34D1 (n=5, 0)Nausea and Vomiting: C35D1 (n=4, 0)Nausea and Vomiting: C36D1 (n=1, 0)Nausea and Vomiting: C37D1 (n=1, 0)Pain: Baseline (n=165, 163)Pain: C2D1 (n=155, 133)Pain: C3D1 (n=154, 106)Pain: C4D1 (n=135, 91)Pain: C5D1 (n=123, 74)Pain: C6D1 (n=120, 70)Pain: C7D1 (n=115, 52)Pain: C8D1 (n=111, 43)Pain: C9D1 (n=101, 37)Pain: C10D1 (n=94, 33)Pain: C11D1 (n=84, 25)Pain: C12D1 (n=76, 23)Pain: C13D1 (n=74, 21)Pain: C14D1 (n=66, 19)Pain: C15D1 (n=62, 16)Pain: C16D1 (n=53, 12)Pain: C17D1 (n=47, 11)Pain: C18D1 (n=45, 11)Pain: C19D1 (n=40, 9)Pain: C20D1 (n=35, 8)Pain: C21D1 (n=30, 8)Pain: C22D1 (n=24, 7)Pain: C23D1 (n=23, 4)Pain: C24D1 (n=20, 3)Pain: C25D1 (n=18, 3)Pain: C26D1 (n=14, 3)Pain: C27D1 (n=14, 2)Pain: C28D1 (n=11, 2)Pain: C29D1 (n=8, 2)Pain: C30D1 (n=8, 1)Pain: C31D1 (n=7, 0)Pain: C32D1 (n=6, 0)Pain: C33D1 (n=6, 0)Pain: C34D1 (n=5, 0)Pain: C35D1 (n=4, 0)Pain: C36D1 (n=1, 0)Pain: C37D1 (n=1, 0)Dyspnea: Baseline (n=165, 163)Dyspnea: C2D1 (n=155, 133)Dyspnea: C3D1 (n=154, 106)Dyspnea: C4D1 (n=135, 91)Dyspnea: C5D1 (n=123, 74)Dyspnea: C6D1 (n=120, 70)Dyspnea: C7D1 (n=115, 52)Dyspnea: C8D1 (n=111, 43)Dyspnea: C9D1 (n=101, 37)Dyspnea: C10D1 (n=94, 33)Dyspnea: C11D1 (n=84, 25)Dyspnea: C12D1 (n=76, 23)Dyspnea: C13D1 (n=74, 21)Dyspnea: C14D1 (n=66, 19)Dyspnea: C15D1 (n=62, 16)Dyspnea: C16D1 (n=53, 12)Dyspnea: C17D1 (n=46, 11)Dyspnea: C18D1 (n=45, 11)Dyspnea: C19D1 (n=40, 9)Dyspnea: C20D1 (n=35, 8)Dyspnea: C21D1 (n=30, 8)Dyspnea: C22D1 (n=24, 7)Dyspnea: C23D1 (n=23, 4)Dyspnea: C24D1 (n=20, 3)Dyspnea: C25D1 (n=18, 3)Dyspnea: C26D1 (n=14, 3)Dyspnea: C27D1 (n=14, 2)Dyspnea: C28D1 (n=11, 2)Dyspnea: C29D1 (n=8, 2)Dyspnea: C30D1 (n=8, 1)Dyspnea: C31D1 (n=7, 0)Dyspnea: C32D1 (n=6, 0)Dyspnea: C33D1 (n=6, 0)Dyspnea: C34D1 (n=5, 0)Dyspnea: C35D1 (n=4, 0)Dyspnea: C36D1 (n=1, 0)Dyspnea: C37D1 (n=1, 0)Insomnia : Baseline (n=164, 163)Insomnia : C2D1 (n=155, 133)Insomnia : C3D1 (n=154, 106)Insomnia : C4D1 (n=135, 91)Insomnia : C5D1 (n=123, 74)Insomnia : C6D1 (n=120, 70)Insomnia : C7D1 (n=114, 52)Insomnia : C8D1 (n=111, 43)Insomnia : C9D1 (n=100, 37)Insomnia : C10D1 (n=94, 33)Insomnia : C11D1 (n=84, 25)Insomnia : C12D1 (n=76, 23)Insomnia : C13D1 (n=74, 21)Insomnia : C14D1 (n=66, 19)Insomnia : C15D1 (n=62, 16)Insomnia : C16D1 (n=53, 11)Insomnia : C17D1 (n=47, 11)Insomnia : C18D1 (n=45, 11)Insomnia : C19D1 (n=40, 9)Insomnia : C20D1 (n=35, 8)Insomnia : C21D1 (n=30, 8)Insomnia : C22D1 (n=24, 7)Insomnia : C23D1 (n=23, 4)Insomnia : C24D1 (n=20, 3)Insomnia : C25D1 (n=18, 3)Insomnia : C26D1 (n=14, 3)Insomnia : C27D1 (n=14, 2)Insomnia : C28D1 (n=11, 2)Insomnia : C29D1 (n=8, 2)Insomnia : C30D1 (n=8, 1)Insomnia : C31D1 (n=7, 0)Insomnia : C32D1 (n=6, 0)Insomnia : C33D1 (n=6, 0)Insomnia : C34D1 (n=5, 0)Insomnia : C35D1 (n=4, 0)Insomnia : C36D1 (n=1, 0)Insomnia : C37D1 (n=1, 0)Appetite loss : Baseline(n=165, 163)Appetite loss : C2/D1(n=155, 133)Appetite loss : C3/D1(n=154, 106)Appetite loss : C4/D1(n=135, 91)Appetite loss : C5/D1(n=123, 74)Appetite loss : C6/D1(n=120, 70)Appetite loss : C7/D1(n=115, 52)Appetite loss : C8/D1(n=111, 43)Appetite loss : C9/D1(n=101, 37)Appetite loss : C10/D1(n=94, 33)Appetite loss : C11/D1(n=84, 25)Appetite loss : C12/D1(n=76, 23)Appetite loss : C13/D1(n=74, 21)Appetite loss : C15/D1(n=62, 16)Appetite loss : C16/D1(n=53, 12)Appetite loss : C17/D1(n=47, 11)Appetite loss : C18/D1(n=45, 11)Appetite loss : C19/D1(n=40, 9)Appetite loss : C20/D1(n=35, 8)Appetite loss : C21/D1(n=30, 8)Appetite loss : C22/D1(n=24, 7)Appetite loss : C23/D1(n=23, 4)Appetite loss : C24/D1(n=20, 3)Appetite loss : C25/D1(n=18, 3)Appetite loss : C26/D1(n=14, 3)Appetite loss : C27/D1(n=14, 2)Appetite loss : C28/D1(n=11, 2)Appetite loss : C29/D1(n=8, 2)Appetite loss : C30/D1(n=8, 1)Appetite loss : C31/D1(n=7, 0)Constipation: Baseline (n=164, 162)Constipation: C2D1 (n=155, 133)Constipation: C3D1 (n=154, 106)Constipation: C4D1 (n=134, 90)Constipation: C5D1 (n=123, 74)Constipation: C6D1 (n=120, 70)Constipation: C7D1 (n=115, 52)Constipation: C8D1 (n=111, 43)Constipation: C9D1 (n=101, 37)Constipation: C10D1 (n=94, 33)Constipation: C11D1 (n=84, 25)Constipation: C12D1 (n=76, 23)Constipation: C13D1 (n=74, 21)Constipation: C14D1 (n=66, 19)Constipation: C15D1 (n=62, 16)Constipation: C16D1 (n=53, 12)Constipation: C17D1 (n=47, 11)Constipation: C18D1 (n=44, 11)Constipation: C19D1 (n=40, 9)Constipation: C20D1 (n=35, 8)Constipation: C21D1 (n=30, 8)Constipation: C22D1 (n=24, 7)Constipation: C23D1 (n=23, 4)Constipation: C24D1 (n=20, 3)Constipation: C25D1 (n=18, 3)Constipation: C26D1 (n=14, 3)Constipation: C27D1 (n=14, 2)Constipation: C28D1 (n=11, 2)Constipation: C29D1 (n=8, 2)Constipation: C30D1 (n=8, 1)Constipation: C31D1 (n=7, 0)Constipation: C32D1 (n=6, 0)Constipation: C33D1 (n=6, 0)Constipation: C34D1 (n=5, 0)Constipation: C35D1 (n=4, 0)Constipation: C36D1 (n=1, 0)Constipation: C37D1 (n=1, 0)Constipation: EOT (n=49, 90)Diarrhea: Baseline (n=165, 162)Diarrhea: C2D1 (n=155, 132)Diarrhea: C3D1 (n=153, 106)Diarrhea: C4D1 (n=134, 90)Diarrhea: C5D1 (n=123, 74)Diarrhea: C6D1 (n=120, 70)Diarrhea: C7D1 (n=115, 52)Diarrhea: C8D1 (n=111, 43)Diarrhea: C9D1 (n=101, 37)Diarrhea: C10D1 (n=94, 33)Diarrhea: C11D1 (n=84, 25)Diarrhea: C12D1 (n=76, 23)Diarrhea: C13D1 (n=74, 21)Diarrhea: C14D1 (n=66, 19)Diarrhea: C15D1 (n=62, 16)Diarrhea: C16D1 (n=52, 12)Diarrhea: C17D1 (n=47, 11)Diarrhea: C18D1 (n=44, 11)Diarrhea: C19D1 (n=40, 9)Diarrhea: C20D1 (n=35, 8)Diarrhea: C21D1 (n=30, 8)Diarrhea: C22D1 (n=24, 7)Diarrhea: C23D1 (n=23, 4)Diarrhea: C24D1 (n=20, 3)Diarrhea: C25D1 (n=18, 3)Diarrhea: C26D1 (n=14, 3)Diarrhea: C27D1 (n=14, 2)Diarrhea: C28D1 (n=11, 2)Diarrhea: C29D1 (n=8, 2)Diarrhea: C30D1 (n=8, 1)Diarrhea: C31D1 (n=7, 0)Diarrhea: C32D1 (n=6, 0)Diarrhea: C33D1 (n=6, 0)Diarrhea: C34D1 (n=5, 0)Diarrhea: C35D1 (n=4, 0)Diarrhea: C36D1 (n=1, 0)Diarrhea: C37D1 (n=1, 0)Diarrhea: EOT (n=49, 90)Financial Difficulties: Baseline (n=165, 161)Financial Difficulties: C2D1 (n=155, 133)Financial Difficulties: C3D1 (n=154, 105)Financial Difficulties: C4D1 (n=135, 90)Financial Difficulties: C5D1 (n=123, 74)Financial Difficulties: C6D1 (n=120, 70)Financial Difficulties: C7D1 (n=115, 52)Financial Difficulties: C8D1 (n=111, 43)Financial Difficulties: C9D1 (n=101, 36)Financial Difficulties: C10D1 (n=94, 33)Financial Difficulties: C11D1 (n=84, 25)Financial Difficulties: C12D1 (n=76, 23)Financial Difficulties: C13D1 (n=74, 21)Financial Difficulties: C14D1 (n=66, 19)Financial Difficulties: C15D1 (n=62, 16)Financial Difficulties: C16D1 (n=53, 12)Financial Difficulties: C17D1 (n=47, 11)Financial Difficulties: C18D1 (n=44, 11)Financial Difficulties: C19D1 (n=40, 9)Financial Difficulties: C20D1 (n=35, 8)Financial Difficulties: C21D1 (n=30, 8)Financial Difficulties: C22D1 (n=24, 7)Financial Difficulties: C23D1 (n=23, 4)Financial Difficulties: C24D1 (n=20, 3)Financial Difficulties: C25D1 (n=18, 3)Financial Difficulties: C26D1 (n=14, 3)Financial Difficulties: C27D1 (n=14, 2)Financial Difficulties: C28D1 (n=11, 2)Financial Difficulties: C29D1 (n=8, 2)Financial Difficulties: C30D1 (n=8, 1)Financial Difficulties: C31D1 (n=7, 0)Financial Difficulties: C32D1 (n=6, 0)Financial Difficulties: C33D1 (n=6, 0)Financial Difficulties: C34D1 (n=5, 0)Financial Difficulties: C35D1 (n=4, 0)Financial Difficulties: C36D1 (n=1, 0)Financial Difficulties: C37D1 (n=1, 0)Financial Difficulties: EOT (n=49, 90)Appetite loss: C32D1 (n=6, 0)Appetite loss: C33D1 (n=6, 0)Appetite loss: C34D1 (n=5, 0)Appetite loss: C35D1 (n=4, 0)Appetite loss: C36D1 (n=1, 0)Appetite loss: C37D1 (n=1, 0)Appetite loss: EOT (n=49, 90)Global Qol: EOT (n=49, 90)Physical Functioning: EOT (n=49, 90)Role Functioning: EOT (n=49, 90)Emotional Functioning: EOT (n=49, 90)Cognitive Functioning: EOT (n=49, 90)Social Function: EOT (n=49, 90)Fatigue: EOT (n=49, 90)Nausea and Vomiting: EOT (n=49, 90)Pain: EOT (n=49, 90)Dyspnea: EOT (n=49, 90)Insomnia: EOT (n=49, 90)
Chemotherapy58.158.159.461.164.167.566.566.966.467.471.065.666.765.866.163.269.770.572.260.463.564.350.055.661.155.641.754.241.733.3NANANANANANANA75.873.575.576.678.680.780.381.682.381.883.279.778.480.082.981.181.882.481.579.277.578.160.064.460.062.250.060.046.726.7NANANANANANANA66.664.765.367.269.669.572.175.276.675.375.372.568.368.477.169.471.271.274.170.866.767.937.544.444.433.325.025.025.00.0NANANANANANANA73.777.777.980.579.881.281.981.682.481.885.084.880.681.682.377.879.575.881.582.380.278.670.866.772.272.258.358.354.258.3NANANANANANANA83.684.583.882.682.783.885.382.284.780.885.380.479.481.684.472.275.875.870.472.972.969.058.361.155.661.141.741.733.316.7NANANANANANANA67.169.572.571.774.576.477.977.580.276.378.771.071.469.374.065.369.765.264.870.864.665.541.761.150.050.033.333.333.30.0NANANANANANANA36.139.539.434.131.531.027.626.628.528.622.226.127.528.124.324.123.227.324.727.825.727.047.240.744.444.455.638.950.055.6NANANANANANANA11.712.79.98.19.06.97.78.99.58.18.07.27.17.06.26.910.63.09.310.48.314.345.833.327.833.333.325.025.033.3NANANANANANANA28.025.723.319.020.519.521.517.423.024.222.025.427.823.722.931.924.224.224.122.929.233.333.327.838.938.958.350.041.783.3NANANANANANANA32.533.633.329.329.326.228.222.523.424.225.330.423.829.820.819.424.224.225.933.333.328.641.744.433.333.350.050.050.066.7NANANANANANANA27.827.123.623.825.723.321.821.722.524.222.723.225.424.629.221.215.221.222.220.829.231.050.022.244.422.250.050.033.033.3NANANANANANANA23.324.321.719.419.412.912.810.914.414.114.714.517.512.516.715.212.118.520.825.014.350.033.344.433.333.333.333.3100.0NA16.914.016.014.820.315.719.916.316.219.221.321.722.224.618.722.224.215.222.220.816.719.025.022.233.344.450.050.050.033.3NANANANANANANA18.17.811.17.98.55.97.19.69.37.27.18.08.712.78.84.25.612.112.17.44.28.311.925.011.122.211.116.716.716.70.0NANANANANANANA10.027.322.320.321.919.418.618.615.516.715.217.318.817.517.516.730.633.315.225.925.029.228.633.333.344.444.466.766.766.7100.0NANANANANANANA24.1NANANANANANA28.146.466.254.174.580.059.446.915.733.740.030.4
Crizotinib57.264.565.268.468.568.369.568.768.767.069.567.966.871.569.069.767.665.769.264.365.070.867.067.167.160.758.966.759.464.664.359.766.768.360.466.766.776.379.282.383.884.586.286.786.587.787.987.288.189.389.087.889.386.688.488.387.688.490.688.085.087.480.578.681.885.082.583.888.990.085.391.786.786.769.373.874.477.978.280.081.780.480.981.681.381.680.283.880.684.077.780.082.181.980.084.776.880.887.075.069.077.377.177.171.486.188.996.791.7100.0100.074.583.183.384.183.085.386.483.984.487.286.185.384.984.586.385.885.686.487.383.884.583.784.187.981.573.270.881.186.584.476.287.590.391.783.358.358.385.685.587.088.188.987.586.787.888.389.287.989.588.588.987.687.485.188.387.585.785.686.185.585.886.181.076.278.887.583.381.083.386.190.095.8100.083.368.075.978.579.479.481.882.382.080.481.981.982.081.881.381.582.781.982.684.682.982.882.684.484.289.878.685.783.383.385.473.883.394.496.791.766.766.738.331.430.827.124.724.623.923.222.921.422.121.923.321.720.822.223.421.520.824.423.923.625.124.424.730.235.728.330.626.428.625.918.515.627.833.311.18.415.213.99.99.210.17.18.86.98.58.36.48.65.85.95.37.87.08.87.47.82.85.47.53.710.711.93.010.46.314.311.111.113.312.50.00.023.913.913.713.011.510.69.611.311.88.710.58.610.18.19.49.410.611.111.311.911.76.99.14.24.617.913.14.52.14.24.85.62.83.38.30.00.031.121.521.617.316.015.817.415.813.917.717.514.514.015.215.613.814.514.114.210.513.313.99.418.311.114.311.912.14.28.39.516.711.113.316.70.00.022.615.316.213.313.614.29.412.512.313.513.913.610.811.111.313.813.512.613.315.212.212.517.411.720.421.416.79.18.312.50.05.65.613.316.70.00.024.421.118.414.612.713.610.412.812.212.413.511.09.58.68.88.511.18.37.611.16.99.45.01.911.97.115.212.58.314.314.828.627.122.921.421.721.718.618.816.021.019.721.221.722.619.522.022.725.022.924.418.111.621.722.226.235.721.229.229.233.333.338.940.033.30.00.028.69.718.121.623.124.422.220.618.617.317.721.017.517.617.716.116.019.922.721.721.022.223.615.921.720.419.023.815.220.812.514.311.111.16.716.733.333.318.428.521.919.017.516.815.614.816.817.816.317.517.116.214.114.015.717.718.917.516.213.318.117.420.011.114.314.315.28.34.29.522.216.713.38.333.366.717.011.111.113.316.70.00.021.856.180.771.875.283.778.232.016.323.823.818.4

[back to top]

European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire-Supplement Module for Lung Cancer (EORTC QLQ-LC13)

QLQ-LC13 consisted of 13 questions relating to disease symptoms specific to lung cancer and treatment side effects typical of treatment with chemotherapy and radiotherapy. The 13 questions comprised 1 multi-item scale for dyspnea and 10 single-item symptoms and side effects (coughing, hemoptysis, sore mouth, dysphagia, peripheral neuropathy, alopecia, chest pain, arm pain, other pain, and medicine for pain). Recall period: past week; response range: 1 'Not at All' to 4 'Very Much'. Scores averaged, transformed to 0-100 scale; higher symptom score = greater degree of symptoms. (NCT00932893)
Timeframe: Baseline, Day 1 of each cycle until disease progression, end of treatment (up to 112 weeks)

,
Interventionunits on a scale (Mean)
Dyspnea: Baseline (n=164, 162)Dyspnea: C2D1 (n=155, 132)Dyspnea: C3D1 (n=153, 106)Dyspnea: C4D1 (n=135, 91)Dyspnea: C5D1 (n=123, 74)Dyspnea: C6D1 (n=119, 70)Dyspnea: C7D1 (n=115, 52)Dyspnea: C8D1 (n=111, 43)Dyspnea: C9D1 (n=101, 37)Dyspnea: C10D1 (n=94, 33)Dyspnea: C11D1 (n=84, 25)Dyspnea: C12D1 (n=76, 23)Dyspnea: C13D1 (n=74, 21)Dyspnea: C14D1 (n=66, 19)Dyspnea: C15D1 (n=62, 16)Dyspnea: C16D1 (n=53, 12)Dyspnea: C17D1 (n=46, 11)Dyspnea: C18D1 (n=45, 11)Dyspnea: C19D1 (n=40, 9)Dyspnea: C20D1 (n=35, 8)Dyspnea: C21D1 (n=30, 8)Dyspnea: C22D1 (n=24, 7)Dyspnea: C23D1 (n=23, 4)Dyspnea: C24D1 (n=20, 3)Dyspnea: C25D1 (n=18, 3)Dyspnea: C26D1 (n=14, 3)Dyspnea: C27D1 (n=14, 2)Dyspnea: C28D1 (n=11, 2)Dyspnea: C29D1 (n=8, 2)Dyspnea: C30D1 (n=8, 1)Dyspnea: C31D1 (n=7, 0)Dyspnea: C32D1 (n=6, 0)Dyspnea: C33D1 (n=6, 0)Dyspnea: C34D1 (n=5, 0)Dyspnea: C35D1 (n=4, 0)Dyspnea: C36D1 (n=1, 0)Dyspnea: C37D1 (n=1, 0)Dyspnea: EOT (n=49, 90)Coughing: Baseline (n=164, 162)Coughing: C2D1 (n=155, 132)Coughing: C3D1 (n=153, 106)Coughing: C4D1 (n=135, 91)Coughing: C5D1 (n=123, 74)Coughing: C6D1 (n=119, 70)Coughing: C7D1 (n=115, 52)Coughing: C8D1 (n=111, 43)Coughing: C9D1 (n=101, 37)Coughing: C10D1 (n=94, 33)Coughing: C11D1 (n=83, 25)Coughing: C12D1 (n=76, 23)Coughing: C13D1 (n=74, 21)Coughing: C14D1 (n=66, 19)Coughing: C15D1 (n=62, 16)Coughing: C16D1 (n=53, 12)Coughing: C17D1 (n=47, 11)Coughing: C18D1 (n=45, 11)Coughing: C19D1 (n=40, 9)Coughing: C20D1 (n=35, 8)Coughing: C21D1 (n=30, 8)Coughing: C22D1 (n=24, 7)Coughing: C23D1 (n=23, 4)Coughing: C24D1 (n=20, 3)Coughing: C25D1 (n=18, 3)Coughing: C26D1 (n=14, 3)Coughing: C27D1 (n=14, 2)Coughing: C28D1 (n=11, 2)Coughing: C29D1 (n=8, 2)Coughing: C30D1 (n=8, 1)Coughing: C31D1 (n=7, 0)Coughing: C32D1 (n=6, 0)Coughing: C33D1 (n=6, 0)Coughing: C34D1 (n=5, 0)Coughing: C35D1 (n=4, 0)Coughing: C36D1 (n=1, 0)Coughing: C37D1 (n=1, 0)Coughing: EOT (n=49, 90)Hemoptysis: Baseline (n=164, 162)Hemoptysis: C2D1 (n=155, 132)Hemoptysis: C3D1 (n=153, 106)Hemoptysis: C4D1 (n=135, 91)Hemoptysis: C5D1 (n=123, 74)Hemoptysis: C6D1 (n=119, 70)Hemoptysis: C7D1 (n=115, 52)Hemoptysis: C8D1 (n=111, 43)Hemoptysis: C9D1 (n=101, 37)Hemoptysis: C10D1 (n=94, 33)Hemoptysis: C11D1 (n=83, 25)Hemoptysis: C12D1 (n=76, 23)Hemoptysis: C13D1 (n=74, 21)Hemoptysis: C14D1 (n=66, 19)Hemoptysis: C15D1 (n=62, 16)Hemoptysis: C16D1 (n=53, 12)Hemoptysis: C17D1 (n=47, 11)Hemoptysis: C18D1 (n=45, 11)Hemoptysis: C19D1 (n=40, 9)Hemoptysis: C20D1 (n=35, 8)Hemoptysis: C21D1 (n=30, 8)Hemoptysis: C22D1 (n=24, 7)Hemoptysis: C23D1 (n=23, 4)Hemoptysis: C24D1 (n=20, 3)Hemoptysis: C25D1 (n=18, 3)Hemoptysis: C26D1 (n=14, 3)Hemoptysis: C27D1 (n=14, 2)Hemoptysis: C28D1 (n=11, 2)Hemoptysis: C29D1 (n=8, 2)Hemoptysis: C30D1 (n=8, 1)Hemoptysis: C31D1 (n=7, 0)Hemoptysis: C32D1 (n=6, 0)Hemoptysis: C33D1 (n=6, 0)Hemoptysis: C34D1 (n=5, 0)Hemoptysis: C35D1 (n=4, 0)Hemoptysis: C36D1 (n=1, 0)Hemoptysis: C37D1 (n=1, 0)Hemoptysis: EOT (n=49, 90)Sore Mouth: Baseline (n=164, 162)Sore Mouth: C2D1 (n=155, 132)Sore Mouth: C3D1 (n=153, 106)Sore Mouth: C4D1 (n=135, 91)Sore Mouth: C5D1 (n=123, 73)Sore Mouth: C6D1 (n=119, 70)Sore Mouth: C7D1 (n=115, 52)Sore Mouth: C8D1 (n=111, 43)Sore Mouth: C9D1 (n=101, 37)Sore Mouth: C10D1 (n=94, 33)Sore Mouth: C11D1 (n=84, 25)Sore Mouth: C12D1 (n=76, 23)Sore Mouth: C13D1 (n=74, 21)Sore Mouth: C14D1 (n=66, 19)Sore Mouth: C15D1 (n=62, 16)Sore Mouth: C16D1 (n=53, 12)Sore Mouth: C17D1 (n=47, 11)Sore Mouth: C18D1 (n=45, 11)Sore Mouth: C19D1 (n=40, 9)Sore Mouth: C20D1 (n=35, 8)Sore Mouth: C21D1 (n=30, 8)Sore Mouth: C22D1 (n=24, 7)Sore Mouth: C23D1 (n=23, 4)Sore Mouth: C24D1 (n=20, 3)Sore Mouth: C25D1 (n=18, 3)Sore Mouth: C26D1 (n=14, 3)Sore Mouth: C27D1 (n=14, 2)Sore Mouth: C28D1 (n=11, 2)Sore Mouth: C29D1 (n=8, 2)Sore Mouth: C30D1 (n=8, 1)Sore Mouth: C31D1 (n=7, 0)Sore Mouth: C32D1 (n=6, 0)Sore Mouth: C33D1 (n=6, 0)Sore Mouth: C34D1 (n=5, 0)Sore Mouth: C35D1 (n=4, 0)Sore Mouth: C36D1 (n=1, 0)Sore Mouth: C37D1 (n=1, 0)Sore Mouth: EOT (n=49, 90)Dysphagia: Baseline (n=164, 162)Dysphagia: C2D1 (n=155, 132)Dysphagia: C3D1 (n=153, 106)Dysphagia: C4D1 (n=135, 91)Dysphagia: C5D1 (n=123, 74)Dysphagia: C6D1 (n=119, 70)Dysphagia: C7D1 (n=115, 52)Dysphagia: C8D1 (n=111, 43)Dysphagia: C9D1 (n=101, 37)Dysphagia: C10D1 (n=94, 33)Dysphagia: C11D1 (n=84, 25)Dysphagia: C12D1 (n=76, 23)Dysphagia: C13D1 (n=74, 21)Dysphagia: C14D1 (n=66, 19)Dysphagia: C15D1 (n=62, 16)Dysphagia: C16D1 (n=53, 12)Dysphagia: C17D1 (n=47, 11)Dysphagia: C18D1 (n=45, 11)Dysphagia: C19D1 (n=40, 9)Dysphagia: C20D1 (n=35, 8)Dysphagia: C21D1 (n=30, 8)Dysphagia: C22D1 (n=24, 7)Dysphagia: C23D1 (n=23, 4)Dysphagia: C24D1 (n=20, 3)Dysphagia: C25D1 (n=18, 3)Dysphagia: C26D1 (n=14, 3)Dysphagia: C27D1 (n=14, 2)Dysphagia: C28D1 (n=11, 2)Dysphagia: C29D1 (n=8, 2)Dysphagia: C30D1 (n=8, 1)Dysphagia: C31D1 (n=7, 0)Dysphagia: C32D1 (n=6, 0)Dysphagia: C33D1 (n=6, 0)Dysphagia: C34D1 (n=5, 0)Dysphagia: C35D1 (n=4, 0)Dysphagia: C36D1 (n=1, 0)Dysphagia: C37D1 (n=1, 0)Dysphagia: EOT (n=49, 90)Peripheral Neuropathy: Baseline (n=164, 162)Peripheral Neuropathy: C2D1 (n=155, 132)Peripheral Neuropathy: C3D1 (n=153, 106)Peripheral Neuropathy: C4D1 (n=134, 91)Peripheral Neuropathy: C5D1 (n=123, 74)Peripheral Neuropathy: C6D1 (n=119, 70)Peripheral Neuropathy: C7D1 (n=115, 52)Peripheral Neuropathy: C8D1 (n=111, 43)Peripheral Neuropathy: C9D1 (n=101, 37)Peripheral Neuropathy: C10D1 (n=94, 33)Peripheral Neuropathy: C11D1 (n=84, 25)Peripheral Neuropathy: C12D1 (n=76, 23)Peripheral Neuropathy: C13D1 (n=74, 21)Peripheral Neuropathy: C14D1 (n=66, 19)Peripheral Neuropathy: C15D1 (n=62, 16)Peripheral Neuropathy: C16D1 (n=52, 12)Peripheral Neuropathy: C17D1 (n=47, 11)Peripheral Neuropathy: C18D1 (n=45, 11)Peripheral Neuropathy: C19D1 (n=40, 9)Peripheral Neuropathy: C20D1 (n=35, 8)Peripheral Neuropathy: C21D1 (n=30, 8)Peripheral Neuropathy: C22D1 (n=24, 7)Peripheral Neuropathy: C23D1 (n=23, 4)Peripheral Neuropathy: C24D1 (n=20, 3)Peripheral Neuropathy: C25D1 (n=18, 3)Peripheral Neuropathy: C26D1 (n=14, 3)Peripheral Neuropathy: C27D1 (n=14, 2)Peripheral Neuropathy: C28D1 (n=11, 2)Peripheral Neuropathy: C29D1 (n=8, 2)Peripheral Neuropathy: C30D1 (n=8, 1)Peripheral Neuropathy: C31D1 (n=7, 0)Peripheral Neuropathy: C32D1 (n=6, 0)Peripheral Neuropathy: C33D1 (n=6, 0)Peripheral Neuropathy: C34D1 (n=5, 0)Peripheral Neuropathy: C35D1 (n=4, 0)Peripheral Neuropathy: C36D1 (n=1, 0)Peripheral Neuropathy: C37D1 (n=1, 0)Peripheral Neuropathy: EOT (n=49, 90)Alopecia: Baseline (n=163, 162)Alopecia: C2D1 (n=155, 132)Alopecia: C3D1 (n=153, 106)Alopecia: C4D1 (n=135, 91)Alopecia: C5D1 (n=123, 74)Alopecia: C6D1 (n=118, 70)Alopecia: C7D1 (n=115, 52)Alopecia: C8D1 (n=111, 43)Alopecia: C9D1 (n=101, 37)Alopecia: C10D1 (n=94, 33)Alopecia: C11D1 (n=84, 25)Alopecia: C12D1 (n=76, 23)Alopecia: C13D1 (n=74, 21)Alopecia: C14D1 (n=65, 19)Alopecia: C15D1 (n=62, 16)Alopecia: C16D1 (n=53, 12)Alopecia: C17D1 (n=46, 11)Alopecia: C18D1 (n=45, 11)Alopecia: C19D1 (n=40, 9)Alopecia: C20D1 (n=35, 8)Alopecia: C21D1 (n=30, 8)Alopecia: C22D1 (n=24, 7)Alopecia: C23D1 (n=23, 3)Alopecia: C24D1 (n=20, 3)Alopecia: C25D1 (n=18, 3)Alopecia: C26D1 (n=14, 3)Alopecia: C27D1 (n=14, 2)Alopecia: C28D1 (n=11, 2)Alopecia: C29D1 (n=8, 2)Alopecia: C30D1 (n=8, 1)Alopecia: C31D1 (n=7, 0)Alopecia: C32D1 (n=6, 0)Alopecia: C33D1 (n=6, 0)Alopecia: C34D1 (n=5, 0)Alopecia: C35D1 (n=4, 0)Alopecia: C36D1 (n=1, 0)Alopecia: C37D1 (n=1, 0)Alopecia: EOT (n=49, 90)Pain in Chest: Baseline (n=163, 160)Pain in Chest: C2D1 (n=155, 132)Pain in Chest: C3D1 (n=153, 106)Pain in Chest: C4D1 (n=135, 91)Pain in Chest: C5D1 (n=123, 74)Pain in Chest: C6D1 (n=119, 70)Pain in Chest: C7D1 (n=115, 52)Pain in Chest: C8D1 (n=111, 43)Pain in Chest: C9D1 (n=101, 37)Pain in Chest: C10D1 (n=94, 33)Pain in Chest: C11D1 (n=84, 25)Pain in Chest: C12D1 (n=76, 23)Pain in Chest: C13D1 (n=74, 21)Pain in Chest: C14D1 (n=66, 19)Pain in Chest: C15D1 (n=62, 16)Pain in Chest: C16D1 (n=53, 12)Pain in Chest: C17D1 (n=46, 11)Pain in Chest: C18D1 (n=45, 11)Pain in Chest: C19D1 (n=40, 9)Pain in Chest: C20D1 (n=35, 8)Pain in Chest: C21D1 (n=30, 8)Pain in Chest: C22D1 (n=24, 7)Pain in Chest: C23D1 (n=23, 4)Pain in Chest: C24D1 (n=20, 3)Pain in Chest: C25D1 (n=18, 3)Pain in Chest: C26D1 (n=14, 3)Pain in Chest: C27D1 (n=14, 2)Pain in Chest: C28D1 (n=11, 2)Pain in Chest: C29D1 (n=8, 2)Pain in Chest: C30D1 (n=8, 1)Pain in Chest: C31D1 (n=7, 0)Pain in Chest: C32D1 (n=6, 0)Pain in Chest: C33D1 (n=6, 0)Pain in Chest: C34D1 (n=5, 0)Pain in Chest: C35D1 (n=4, 0)Pain in Chest: C36D1 (n=1, 0)Pain in Chest: C37D1 (n=1, 0)Pain in Chest: EOT (n=49, 90)Pain in Arm or Shoulder: Baseline (n=164, 161)Pain in Arm or Shoulder: C2D1 (n=155, 132)Pain in Arm or Shoulder: C3D1 (n=153, 105)Pain in Arm or Shoulder: C4D1 (n=135, 91)Pain in Arm or Shoulder: C5D1 (n=123, 74)Pain in Arm or Shoulder: C6D1 (n=119, 70)Pain in Arm or Shoulder: C7D1 (n=115, 52)Pain in Arm or Shoulder: C8D1 (n=111, 43)Pain in Arm or Shoulder: C9D1 (n=101, 37)Pain in Arm or Shoulder: C10D1 (n=94, 33)Pain in Arm or Shoulder: C11D1 (n=84, 25)Pain in Arm or Shoulder: C12D1 (n=76, 23)Pain in Arm or Shoulder: C13D1 (n=74, 21)Pain in Arm or Shoulder: C14D1 (n=66, 19)Pain in Arm or Shoulder: C15D1 (n=62, 16)Pain in Arm or Shoulder: C16D1 (n=53, 12)Pain in Arm or Shoulder: C17D1 (n=47, 11)Pain in Arm or Shoulder: C18D1 (n=45, 11)Pain in Arm or Shoulder: C19D1 (n=40, 9)Pain in Arm or Shoulder: C20D1 (n=35, 8)Pain in Arm or Shoulder: C21D1 (n=30, 8)Pain in Arm or Shoulder: C22D1 (n=24, 7)Pain in Arm or Shoulder: C23D1 (n=23, 4)Pain in Arm or Shoulder: C24D1 (n=20, 3)Pain in Arm or Shoulder: C25D1 (n=18, 3)Pain in Arm or Shoulder: C26D1 (n=14, 3)Pain in Arm or Shoulder: C27D1 (n=14, 2)Pain in Arm or Shoulder: C28D1 (n=11, 2)Pain in Arm or Shoulder: C29D1 (n=8, 2)Pain in Arm or Shoulder: C30D1 (n=8, 1)Pain in Arm or Shoulder: C31D1 (n=7, 0)Pain in Arm or Shoulder: C32D1 (n=6, 0)Pain in Arm or Shoulder: C33D1 (n=6, 0)Pain in Arm or Shoulder: C34D1 (n=5, 0)Pain in Arm or Shoulder: C35D1 (n=4, 0)Pain in Arm or Shoulder: C36D1 (n=1, 0)Pain in Arm or Shoulder: C37D1 (n=1, 0)Pain in Arm or Shoulder: EOT (n=48, 90)Pain in Other Parts: Baseline (n=163, 158)Pain in Other Parts: C2D1 (n=153, 125)Pain in Other Parts: C3D1 (n=152, 104)Pain in Other Parts: C4D1 (n=134, 90)Pain in Other Parts: C5D1 (n=123, 73)Pain in Other Parts: C6D1 (n=118, 68)Pain in Other Parts: C7D1 (n=115, 51)Pain in Other Parts: C8D1 (n=111, 42)Pain in Other Parts: C9D1 (n=100, 37)Pain in Other Parts: C10D1 (n=92, 33)Pain in Other Parts: C11D1 (n=83, 25)Pain in Other Parts: C12D1 (n=74, 23)Pain in Other Parts: C13D1 (n=74, 21)Pain in Other Parts: C14D1 (n=66, 19)Pain in Other Parts: C15D1 (n=61, 16)Pain in Other Parts: C16D1 (n=53, 12)Pain in Other Parts: C17D1 (n=46, 11)Pain in Other Parts: C18D1 (n=45, 11)Pain in Other Parts: C19D1 (n=40, 9)Pain in Other Parts: C20D1 (n=35, 8)Pain in Other Parts: C21D1 (n=30, 8)Pain in Other Parts: C22D1 (n=24, 7)Pain in Other Parts: C23D1 (n=23, 4)Pain in Other Parts: C24D1 (n=20, 3)Pain in Other Parts: C25D1 (n=18, 3)Pain in Other Parts: C26D1 (n=14, 3)Pain in Other Parts: C27D1 (n=14, 2)Pain in Other Parts: C28D1 (n=11, 2)Pain in Other Parts: C29D1 (n=8, 2)Pain in Other Parts: C30D1 (n=8, 1)Pain in Other Parts: C31D1 (n=7, 0)Pain in Other Parts: C32D1 (n=6, 0)Pain in Other Parts: C33D1 (n=6, 0)Pain in Other Parts: C34D1 (n=5, 0)Pain in Other Parts: C35D1 (n=4, 0)Pain in Other Parts: C36D1 (n=1, 0)Pain in Other Parts: C37D1 (n=1, 0)Pain in Other Parts: EOT (n=49, 90)
Chemotherapy26.928.628.027.524.825.224.422.019.521.520.422.726.526.322.922.230.326.323.534.725.027.052.848.140.744.455.661.150.077.8NANANANANANANA35.642.234.832.427.125.730.026.322.525.220.218.723.227.024.620.822.227.318.214.829.233.328.633.344.444.433.350.050.033.333.3NANANANANANANA37.43.73.52.21.50.93.81.32.32.73.04.05.84.80.00.05.69.13.03.78.34.29.50.00.00.00.00.00.00.00.0NANANANANANANA4.86.49.19.410.39.610.57.710.910.812.19.38.79.57.014.68.36.112.17.412.54.27.116.70.00.011.116.70.016.70.0NANANANANANANA8.18.69.89.78.17.710.05.16.27.26.14.07.26.37.04.22.86.13.00.08.38.30.08.30.011.111.116.716.716.733.3NANANANANANANA8.517.721.518.221.621.218.121.824.029.727.324.033.333.329.825.025.027.330.322.233.329.228.641.744.444.422.266.733.350.033.3NANANANANANANA21.916.936.630.524.923.923.319.918.616.214.116.020.312.715.88.313.918.218.218.525.016.714.322.233.333.344.433.350.033.333.3NANANANANANANA33.324.023.723.319.817.116.216.714.017.118.220.023.223.822.818.716.721.224.218.529.225.026.241.733.333.333.350.050.050.066.7NANANANANANANA28.519.519.917.514.313.117.117.320.223.425.324.029.025.429.825.016.721.230.318.525.029.221.433.344.433.322.250.033.366.733.3NANANANANANANA21.931.425.921.221.520.518.116.322.224.328.324.024.631.724.625.038.924.233.325.929.229.240.541.711.144.433.350.033.316.733.3NANANANANANANA29.6
Crizotinib27.217.617.916.216.915.215.614.713.915.415.113.712.814.813.212.612.314.114.413.712.610.613.814.415.414.315.111.112.59.715.918.518.520.016.722.211.121.838.223.023.519.818.414.615.913.213.514.212.914.514.013.111.813.86.413.312.510.513.38.311.68.35.62.47.118.28.320.819.016.711.120.033.333.333.325.92.41.70.91.00.30.60.00.90.30.42.40.92.30.51.10.60.01.50.01.00.00.01.40.00.00.00.00.00.00.00.00.00.00.00.00.00.00.75.58.07.08.15.14.85.23.04.86.44.83.94.54.55.46.33.55.24.27.64.45.65.16.70.04.87.16.18.38.39.50.00.00.00.00.00.03.47.18.27.66.26.07.04.13.25.04.33.23.54.13.03.84.42.83.74.24.83.32.82.95.00.09.52.46.18.30.04.85.60.06.78.30.00.04.814.018.117.615.915.213.713.010.811.211.311.111.410.410.68.610.312.111.99.212.411.18.38.73.39.316.79.515.212.516.719.011.111.120.025.00.00.010.217.49.57.68.96.04.24.93.94.04.64.04.46.33.64.86.35.13.75.83.87.82.81.46.79.37.116.76.116.712.59.50.00.00.00.00.00.06.818.89.57.07.46.86.27.55.15.96.76.35.36.37.65.93.87.25.95.05.75.64.23.61.75.67.14.89.10.04.29.50.00.06.78.30.00.017.016.39.08.16.96.26.76.46.95.67.46.06.65.97.65.45.03.58.16.79.57.82.80.73.31.97.14.80.00.00.00.011.10.00.00.00.00.011.123.115.011.412.714.410.711.310.510.79.812.412.210.811.69.810.713.810.414.213.321.112.55.16.79.323.816.76.14.212.59.55.60.06.70.00.00.018.4

[back to top]

European Quality of Life - 5 Dimensional (EQ-5D) Visual Analog Scale (VAS)

EQ-5D: participant rated questionnaire to assess health-related quality of life in terms of a single index value. The VAS component rates current health state on a scale from 0 (worst imaginable health state) to 100 (best imaginable health state); higher scores indicate a better health state. (NCT00932893)
Timeframe: Baseline, Day 1 of each cycle until disease progression, end of treatment (up to 112 weeks)

,
Interventionunits on a scale (Mean)
Baseline (n=164, 161)C2D1 (n=153, 131)C3D1 (n=153, 105)C4D1 (n=135, 90)C5D1 (n=122, 74)C6D1 (n=120, 70)C7D1 (n=115, 52)C8D1 (n=110, 43)C9D1 (n=101, 37)C10D1 (n=94, 33)C11D1 (n=84, 25)C12D1 (n=77, 23)C13D1 (n=73, 21)C14D1 (n=66, 19)C15D1 (n=62, 16)C16D1 (n=53, 12)C17D1 (n=47, 11)C18D1 (n=45, 11)C19D1 (n=40, 9)C20D1 (n=35, 8)C21D1 (n=30, 8)C22D1 (n=24, 7)C23D1 (n=23, 4)C24D1 (n=20, 3)C25D1 (n=18, 3)C26D1 (n=14, 3)C27D1 (n=14, 2)C28D1 (n=11, 2)C29D1 (n=8, 2)C30D1 (n=8, 1)C31D1 (n=7, 0)C32D1 (n=6, 0)C33D1 (n=6, 0)C34D1 (n=5, 0)C35D1 (n=4, 0)C36D1 (n=1, 0)C37D1 (n=1, 0)EOT (n=49, 90)
Chemotherapy66.7666.3365.8469.1368.1269.7170.6372.3072.2774.2777.2474.8373.0074.1177.4479.0081.7378.9178.0076.7573.6372.2154.0062.0063.6763.0050.0055.0045.0040.00NANANANANANANA58.34
Crizotinib64.0969.1973.1373.7875.2775.7977.0274.7274.4575.4976.3276.9576.3878.7777.7175.3275.0975.8776.8572.6674.1377.5475.4871.4075.6172.1466.5772.3671.3869.5068.5765.8367.5068.0072.2590.0085.0068.33

[back to top]

Number of Participants With Categorical Maximum QTcF for Crizotinib

QT interval corrected using Fridericia's formula (QTcF): QT interval (time corresponding to the beginning of depolarization to re-polarization of the ventricles) divided by cube root of RR interval. Maximum QTcF was categorized as less than (<) 450 milliseconds (msec), 450 msec to <480 msec, 480 msec to <500 msec, and more than or equal to (>=) 500 msec. A participant is reported only once under the maximum QTcF interval observed at any of the time-points. Only participants receiving crizotinib were to be analyzed for this outcome measure as per planned analysis. (NCT00932893)
Timeframe: Pre-dose on Day 1 of Cycle 1, 2 to 6 hours post-dose on Day 1 of Cycle 1, 2

Interventionparticipants (Number)
<450 msec450 msec to <480 msec480 msec to <500 msec>=500 msec
Crizotinib137918

[back to top]

Duration of Response (DR)

Time in weeks from the first documentation of objective tumor response to objective tumor progression or death due to any cause. Duration of tumor response was calculated as (the date of the first documentation of objective tumor progression or death due to any cause minus the date of the first CR or PR that was subsequently confirmed plus 1) divided by 7.02. DR was calculated for the subgroup of participants with a confirmed objective tumor response. (NCT00932893)
Timeframe: Randomization until PD or initiation of antitumor therapy in the absence of PD or death, assessed every 6 weeks (up to 112 weeks)

Interventionweeks (Median)
Crizotinib32.1
Chemotherapy24.4

[back to top]

Overall Survival (OS)

OS: Time in months from randomization to date of death due to any cause. OS was calculated as (the death date minus the date of randomization plus 1) divided by 30.4. (NCT00932893)
Timeframe: Randomization until death (up to 4.5 years)

Interventionmonths (Median)
Crizotinib21.7
Chemotherapy21.9

[back to top]

Overall Survival Probability at Months 6 and 12

Overall survival probability at Month 6 and 12 was defined as the probability of survival at 6 and 12 months respectively, after the randomization of study treatment. The survival probability was estimated using the Kaplan-Meier method. (NCT00932893)
Timeframe: Month 6, 12

,
Interventionpercent chance of survival (Number)
Month 6Month 12
Chemotherapy83.866.7
Crizotinib86.670.4

[back to top]

Plasma Concentration of Crizotinib

Only participants receiving crizotinib were to be analyzed for this outcome measure as per planned analysis. (NCT00932893)
Timeframe: Pre-dose on Cycle 1 Day 1, Cycle 1 Day 15, and Day 1 of Cycles 2, 3, 5

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Cycle 1 Day 1 (n=15)Cycle 1 Day 15 (n=92)Cycle 2 Day 1 (n=62)Cycle 3 Day 1 (n=61)Cycle 5 Day 1 (n=47)
CrizotinibNA298293306291

[back to top]

Plasma Concentration of Soluble c-Met Ectodomain and Hepatocyte Growth Factor Scatter Proteins

Descriptive statistics (absolute value and change from baseline as measured by ratio to baseline) for each best overall response category (CR, PR, SD, PD or combined) have been used to summarize the data from optional soluble c-Met ectodomain assays for crizotinib treated patients. (NCT00932893)
Timeframe: Pre-dose on Day 1 of Cycle 1, 2 to 6 hours post-dose on Day 1 of Cycle 2, end of treatment (up to 112 weeks)

Interventionnanogram per milliliter (ng/mL) (Mean)
Baseline (N = 81)Cycle 2 Day 1 6-hour post dose (N = 69)End of treatment (N = 40)
Overall Values1428.31683.01751.8

[back to top]

Percentage of Participants With Disease Control at Week 6

Disease control: participants with CR, PR, or stable disease (SD) according to RECIST v1.1. CR: disappearance of all target and non-target lesions and normalization of tumor marker level, all lymph nodes must be non-pathological in size (<10 mm short axis). PR: at least 30 % decrease in sum of diameters of target lesions, taking as reference the baseline sum diameters persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease (PD), taking as reference the smallest sum diameters while on study. PD: at least a 20% increase (including an absolute increase of at least 5 mm) in the sum of diameters of target lesions, taking as reference the smallest sum on study and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. Disease control is based on independent radiology review. (NCT00932893)
Timeframe: Week 6

Interventionpercentage of participants (Number)
Crizotinib81.5
Chemotherapy55.2

[back to top]

The Primary Efficacy Endpoint is Progression Free Survival, Analyzed in the Treated Population. PFS is Assessed From Randomization Until Either Tumor Progression, as Per RECIST Criteria, or Until Death Due to Any Reason.

(NCT00942825)
Timeframe: 15 June 2009 to 30 September 2012

Interventiondays (Median)
A CBP501 +Cisplatin + Pemetrexed140
B Cisplatin + Pemetrexed165

[back to top]

Percentage of Participants With Complete Response or Partial Response (Overall Tumor Response Rate)

Overall Response rate (ORR) was the percentage of participants with a confirmed complete response (CR) or partial response (PR), as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST) criteria version 1.0. CR was the disappearance of all target and non-target lesions; PR was a ≥30% decrease in sum of longest diameter of target lesions without new lesion and progression of non-target lesions. ORR was calculated as a total number of participants with CR or PR from the start of study treatment until disease progression or recurrence divided by the total number of participants treated, then multiplied by 100. (NCT00948675)
Timeframe: Baseline to date of objective progressive disease up to 39.49 months

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin23.6
Paclitaxel + Carboplatin + Bevacizumab27.4

[back to top]

Overall Survival (OS)

OS was defined as the duration from the date of randomization to the date of death from any cause. For participants who were alive at the time of the data inclusion cutoff, OS was censored at the last date the participant was known to be alive. (NCT00948675)
Timeframe: Randomization to date of death from any cause up to 39.49 months

Interventionmonths (Median)
Pemetrexed + Carboplatin10.51
Paclitaxel + Carboplatin + Bevacizumab11.66

[back to top]

Disease Control Rates Defined as Complete Response (CR), Partial Response (PR), and Stable Disease (SD)

Disease control rate was the percentage of participants with a confirmed CR, PR or SD, as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST) criteria version 1.0. CR was the disappearance of all target and non-target lesions; PR was a ≥30% decrease in sum of longest diameter of target lesions without new lesion and progression of non-target lesion; SD was neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease. Disease control rate was calculated as a total number of participants with CR or PR or SD divided by the total number of participants treated, then multiplied by 100. (NCT00948675)
Timeframe: Baseline to date of objective progressive disease up to 39.49 months

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin59.9
Paclitaxel + Carboplatin + Bevacizumab57.0

[back to top]

Progression Free Survival (PFS)

PFS was defined as the duration from the date of randomization to the date of progressive disease (PD) or death from any cause. PD was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria version 1.0. PD was ≥20% increase in sum of longest diameter of target lesions or the appearance of new lesions. For participants who had no PD or death at the time of the data inclusion cutoff, PFS was censored at their last objective progression-free disease assessment prior to the cutoff date or the date of initiation of subsequent systemic anticancer therapy. (NCT00948675)
Timeframe: Randomization to measured progressive disease up to 39.49 months

Interventionmonths (Median)
Pemetrexed + Carboplatin4.44
Paclitaxel + Carboplatin + Bevacizumab5.45

[back to top]

Progression Free Survival Without Grade 4 Toxicity (G4PFS) as Measured by the Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0

G4PFS was defined as the duration from the date of randomization to the earliest occurrence date of one of the following three events: Common Terminology Criteria (CTC) grade 4 adverse events (G4AEs), or progressive disease (PD) or death from any cause, whichever occurred earlier. PD was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria version 1.0. PD is ≥20% increase in sum of longest diameter of target lesions or the appearance of new lesions. For participants who had no G4AEs, or PD, or death at the time of the data inclusion cutoff, PFS was censored at their last objective progression-free disease assessment prior to the cutoff date or the date of initiation of subsequent systemic anticancer therapy. (NCT00948675)
Timeframe: Randomization to measured progressive disease or treatment discontinuation up to 39.49 months

Interventionmonths (Median)
Pemetrexed + Carboplatin3.91
Paclitaxel + Carboplatin + Bevacizumab2.86

[back to top]

Trough Plasma Concentrations of Afatinib at Day 29

Trough plasma concentrations of Afatinib at day 29 (course 2, visit 2) after multiple daily dosing of 40 mg Afatinib and after dose escalation to 50 mg or dose reduction to 30 mg or 20 mg. (NCT00949650)
Timeframe: Day 29.

Interventionng/mL. (Geometric Mean)
Afatinib 30 mg28.0
Afatinib 40 mg25.8
Afatinib 50 mg29.6

[back to top]

Trough Plasma Concentrations of Afatinib at Day 22

Trough plasma concentrations of Afatinib at Day 22 (course 2, visit 1) after multiple daily dosing of 40 mg Afatinib and after dose escalation to 50 mg or dose reduction to 30 mg or 20 mg. (NCT00949650)
Timeframe: Day 22.

Interventionng/mL. (Geometric Mean)
Afatinib 30 mg21.8
Afatinib 40 mg28.0
Afatinib 50 mg29.9

[back to top]

Progression-Free Survival (PFS) Time

PFS was defined as time from randomisation to disease progression or death whichever occured first. Assessed by central independent review according to the Response Evaluation Criteria in Solid Tumours (RECIST 1.1). Median time results from unstratified Kaplan-Meier estimates. (NCT00949650)
Timeframe: Tumour assessments were performed at Screening, Week 6, Week 12, Week 18 and then every 12-18 weeks until disease progression

InterventionMonths. (Median)
Afatinib 40 mg11.17
Pemetrexed/Cisplatin Chemotherapy6.90

[back to top]

Percentage of Patients With Objective Response (OR)

OR was defined as Complete Response (CR) or Partial Response (PR). Assessed by central independent review according to RECIST 1.1. (NCT00949650)
Timeframe: Tumour assessments were performed at Screening, Week 6, Week 12, Week 18 and then every 12-18 weeks until disease progression

InterventionPercentage of patients with OR. (Number)
Afatinib 40 mg56.5
Pemetrexed/Cisplatin Chemotherapy22.6

[back to top]

Percentage of Participants With Disease Control (DC)

DC was defined as a patient with OR or Stable Disease (SD). Assessed by central independent review according to the RECIST 1.1. (NCT00949650)
Timeframe: Tumour assessments were performed at Screening, Week 6, Week 12, Week 18 and then every 12-18 weeks until disease progression

InterventionPercentage of participants with DC. (Number)
Afatinib 40 mg90.4
Pemetrexed/Cisplatin Chemotherapy80.9

[back to top]

HRQOL: Time to Deterioration in Pain

HRQOL was measured by EORTC QLQ-C30 and its lung cancer specific module QLQ-LC13. Analysis for pain is based on composite of QLQ-C30 questions 9 and 19. Time to deterioration was defined as the time from randomisation to a score increased (worsened) by at least 10 points from baseline (0-100 point scale). Patients were considered deteriorated at time of death. Median time results from unstratified Kaplan-Meier estimates. (NCT00949650)
Timeframe: Throughout the trial until progression (every 3 weeks).

InterventionMonths. (Median)
Afatinib 40 mg4.17
Pemetrexed/Cisplatin Chemotherapy3.09

[back to top]

HRQOL: Time to Deterioration in Dyspnoea

HRQOL was measured by EORTC QLQ-C30 and its lung cancer specific module QLQ-LC13. Analysis for dyspnoea is based on composite of QLQ-LC13 questions 3-5. Time to deterioration was defined as the time from randomisation to a score increased (worsened) by at least 10 points from baseline (0-100 point scale). Patients were considered deteriorated at time of death. Median time results from unstratified Kaplan-Meier estimates. (NCT00949650)
Timeframe: Throughout the trial until progression (every 3 weeks).

InterventionMonths. (Median)
Afatinib 40 mg10.41
Pemetrexed/Cisplatin Chemotherapy2.86

[back to top] [back to top]

Overall Survival (OS) Time

OS was defined as time from randomisation to death. (NCT00949650)
Timeframe: From randomisation to cut-off date (17MAR2017).

InterventionMonths. (Median)
Afatinib 40 mg28.16
Pemetrexed/Cisplatin Chemotherapy28.22

[back to top]

Trough Plasma Concentrations of Afatinib at Day 43

Trough plasma concentrations of Afatinib at Day 43 (course 3, visit 1) after multiple daily dosing of 40 mg Afatinib and after dose escalation to 50 mg or dose reduction to 30 mg or 20 mg. (NCT00949650)
Timeframe: Day 43.

Interventionng/mL. (Geometric Mean)
Afatinib 20 mg24.4
Afatinib 30 mg24.7
Afatinib 40 mg23.5
Afatinib 50 mg27.5

[back to top]

Eastern Cooperative Oncology Group (ECOG) Performance Status (PS)

"ECOG PS measured on 6 point scale to assess participant's performance status. 0=Fully active, able to carry on all pre-disease activities without restriction.~Restricted in physically strenuous activity, but ambulatory and able to carry out light or sedentary work.~Ambulatory (>50 percent of waking hours), capable of all self-care, unable to carry out any work activities.~Capable of only limited self-care, confined to bed or chair more than 50 percent of waking hours.~Completely disabled, cannot carry on any self-care, totally confined to bed or chair.~Dead." (NCT00949650)
Timeframe: Throughout the trial until progression (every 3 weeks), up to 28 months.

,
InterventionParticipants (Number)
ECOG PS 0 (last value)ECOG PS 1 (last value)ECOG PS 2 (last value)
Afatinib 40 mg921380
Pemetrexed/Cisplatin Chemotherapy41731

[back to top]

Change From Baseline in Body Weight

Because the PFS was longer for patients in the Afatinib arm than for patients in the chemotherapy arm, the period of data collection for ECOG status and body weight continued for a longer time in the Afatinib arm. (NCT00949650)
Timeframe: Baseline and throughout the trial until progression (every 3 weeks), up to 28 months.

,
InterventionKg. (Mean)
Change from baseline at lowest valueChange from baseline at last value
Afatinib 40 mg-3.95-1.19
Pemetrexed/Cisplatin Chemotherapy-2.68-0.29

[back to top]

Tumour Shrinkage

Tumour shrinkage was calculated as the minimum Sum of Diameters (SoD) of target lesions from all post-baseline tumour assessments, as read by the central independent review. The mean of these minimum values were presented after adjusting for baseline SoD, EGFR mutation group and race. (NCT00949650)
Timeframe: Tumour assessments were performed at Screening, Week 6, Week 12, Week 18 and then every 12-18 weeks until disease progression

Interventionmm. (Mean)
Afatinib 40 mg33.19
Pemetrexed/Cisplatin Chemotherapy43.00

[back to top]

PFS (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 (NCT00950365)
Timeframe: Time from randomization until documented tumor progression or death from any cause, assessed up to 12 months

Interventionmonths (Median)
Arm A (Pemetrexed)8
Arm B (Pemetrexed Disodium, Erlotinib Hydrochloride)20

[back to top]

Overall Survival

Time to event endpoints will be analyzed using standard survival analytic methods, including the Kaplan-Meier approach for estimating the survival distributions. (NCT00950365)
Timeframe: Time from the date of randomization to date of death due to any cause, assessed up to 12 months

InterventionParticipants (Count of Participants)
Arm A (Pemetrexed)25
Arm B (Pemetrexed Disodium, Erlotinib Hydrochloride)50

[back to top]

Objective Response Rate (CR +PR) Evaluated Using RECIST

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR~Response rates in each arm will be summarized by computing proportions and corresponding 95% confidence intervals." (NCT00950365)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Arm A (Pemetrexed)12
Arm B (Pemetrexed Disodium, Erlotinib Hydrochloride)28

[back to top]

Number of Participants With Marked Laboratory Abnormalities

Marked laboratory abnormalities were defined as those values that were outside the reference range and showed a clinically relevant change from Baseline. The reference range for Platelets was 100-550 (10^9/L), for White blood cells (WBC) was 3.0-18.0 (10^9/L), for Lymphocytes was 0.70-7.60 (10^9/L), and Neutrophil 1.50-9.25 (10^9/L ). (NCT00961415)
Timeframe: Up to 21 months

,,
InterventionParticipants (Number)
Alanine amino transferase (ALT)Aspartate amino transferase (AST)Alkaline phosphataseHemoglobinInternational normalized ratio (INR)LymphocytesNeutrophilsPlateletsSerum creatinineWBCActivated partial thromboplastin time (aPTT)
Bevacizumab +Pemetrexed Maintenance Trt Arm B26262630030303026300
Bevacizumab Maintenance Trt Arm A18181828128282818281
No Maintenance Trt8882502525258250

[back to top]

Duration of Response During Maintenance Treatment Phase

Duration of response is defined as the time in months from the initial start of response PR or better to the earlier of documented PD or death due to any cause. Participants who had neither progressed nor died at the date of clinical cutoff, who withdrew from the study, were lost to follow-up, or were without documented disease progression were censored at the date of the last available tumor assessment. The analysis was based on all participants with measurable disease at baseline who achieved response.Tumor assessment was done before Cycle 3, at Cycle 2 of maintenance therapy and every nine weeks thereafter. (NCT00961415)
Timeframe: Up to 21 months

InterventionMonths (Median)
Bevacizumab Maintenance Trt Arm A5.7
Bevacizumab +Pemetrexed Maintenance Trt Arm B9.2

[back to top]

Duration of Disease Control During Maintenance Treatment Phase

Duration of disease control is defined as the time in months from randomization to the earlier of documented PD or death due to any cause. Tumor assessment was done before Cycle 3, at Cycle 2 of maintenance therapy and every nine weeks thereafter. (NCT00961415)
Timeframe: Up to 21 months

InterventionMonths (Median)
Bevacizumab Maintenance Trt Arm A4.9
Bevacizumab +Pemetrexed Maintenance Trt Arm B7.8

[back to top]

Overall Survival During Maintenance Treatment Phase

Overall survival (OS) is assessed from the date of first induction treatment until the date of death. Tumor assessment was done before Cycle 3, at Cycle 2 of maintenance therapy and every nine weeks thereafter. (NCT00961415)
Timeframe: Up to 21 months

InterventionMonths (Median)
Bevacizumab Maintenance Trt Arm A15.7
Bevacizumab +Pemetrexed Maintenance Trt Arm BNA

[back to top]

Progression Free Survival During Maintenance Treatment Phase

Progression free survival (PFS) is defined as the time from randomization to the date of documented disease progression according to Response Evaluation Criteria in Solid Tumors (RECIST v1.1) , or the date of occurrence of a second primary cancer, or date of death from any cause, whichever comes first. Progression is defined using (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions, the appearance of new lesions and increase of at least 5 mm in the sum of diameters of target lesions.Tumor assessment was done before Cycle 3, at Cycle 2 of maintenance therapy and every nine weeks thereafter. (NCT00961415)
Timeframe: Up to 21 months

InterventionMonths (Median)
Bevacizumab Maintenance Trt Arm A6.6
Bevacizumab +Pemetrexed Maintenance Trt Arm B10.2

[back to top]

Best Overall Response Rate During Maintenance Treatment Phase

The best overall response rate (BORR) is defined as the percentage of participants having achieved confirmed Complete Response (CR) and Partial Response (PR) as the best overall response. CR was defined as complete disappearance of all target lesions and non-target disease. PR was defined as a greater than or equal to (≥) 30% decrease under baseline of the sum of diameters of all target lesions. Stable disease (SD) is defined as steady state of disease with neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease (PD).Tumor assessment was done before Cycle 3, at Cycle 2 of maintenance therapy and every nine weeks thereafter. (NCT00961415)
Timeframe: Up to 21 months

,
Interventionpercentage of participants (Number)
Partial response (PR)Stable disease (SD)
Bevacizumab +Pemetrexed Maintenance Trt Arm B55.544.5
Bevacizumab Maintenance Trt Arm A5050

[back to top]

Incidence of Adverse Events and Serious Adverse Event

An adverse events (AE) is any untoward medical occurrence in a participant or clinical investigation participant administered a pharmaceutical product and that does not necessarily have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of a medicinal (investigational) product, whether or not considered related to the medicinal (investigational) product. An serious adverse event (SAE) is any experience that suggests a significant hazard, contraindication, side effect, or precaution. (NCT00961415)
Timeframe: Up to 21 months

,,
InterventionParticipants (Number)
AESAE
Bevacizumab +Pemetrexed Maintenance Trt Arm B12342
Bevacizumab Maintenance Trt Arm A11626
No Maintenance Trt11970

[back to top]

Quality of Life

European Organization for Research and Treatment of Cancer Quality-of-Life Questionnaire-Cancer 30 (EORTC QLQ-C30): included functional scales (physical, role, cognitive, emotional, and social), global health status, symptom scales (fatigue, pain, nausea/vomiting) and single items (dyspnoea, appetite loss, insomnia, constipation/diarrhea and financial difficulties). The European Organization for Research and Treatment of Cancer Quality-of-Life Questionnaire-Lung Cancer 13 [EORTC QLQ-LC13]consisted of 1 multi-item scale and 9 single items that assessed the specific symptoms (dyspnea, cough, hemoptysis, and site specific pain), side effects (sore mouth, dysphagia, neuropathy, and alopecia), and pain medication use of lung cancer participants receiving chemotherapy. Scale score range: 0 to 100. Higher symptom score = greater degree of symptom severity. QOL was assessed using Pre-Induction Baseline (Pre-ind BL), Maintenance (MTC), End of study (EOS) cycles. (NCT00961415)
Timeframe: Up to 21 months

,
InterventionScore on scale (Mean)
[Pre-ind BL] Global Health Status Scale n=116,121[Pre-ind BL] Physical Functional Scale (n=118,121)[Pre-ind BL] Role Functional Scale (n=118,121)[Pre-ind BL] Emotional Functional Scale n=117,121[Pre-ind BL] Cognitive Functional Scale n=117,121[Pre-ind BL]] Social Functional Scale (n=116,120)[Pre-ind BL] Fatigue Symptom Scale (n=118,121)[Pre-ind BL] Nausea & Vomiting Scale n=118,121[Pre-ind BL] Pain Symptom Scale (n=118,121)[Pre-ind BL] Dyspnea Symptom Scale (n=118,121)[Pre-ind BL] Insomnia Symptom Scale n=118,121[Pre-ind BL] Appetite Loss Symptom Scale n=118,121[Pre-ind BL]] Constipation Symptom Scale n=118,121[Pre-ind BL] Diarrhea Symptom Scale (n=117,121)[Pre-ind BL]Financial Difficulties Scale n=114,120[MTC Cycle 3]Global Health Status (n=69,87)[MTC Cycle 3]Physical Functional Scale (n=69,87)[MTC Cycle 3]Role Functional Scale (n=69,87)[MTC Cycle 3]Emotional Functional Scale (n=69,87)[MTC Cycle 3]Cognitive Functional Scale (n=69,87)[MTC Cycle 3]Social Functional Scale (n=69,87)[MTC Cycle 3]Fatigue Symptom Scale (n=69,87)[MTC Cycle 3]Nausea and Vomiting Scale (n=69,87)[MTC Cycle 3]Pain Symptom Scale (n=69,87)[MTC Cycle 3]Dyspnea Symptom Scale (n=69,87)MTC Cycle 3]Appetite Loss Symptom Scale (n=69,87)[MTC Cycle 3]Insomnia Symptom Scale (n=69,87)[MTC Cycle 3]Constipation Symptom Scale (n=69,87)[MTC Cycle 3]Diarrhea Symptom Scale (n=69,87)[MTC Cycle 3]Financial Difficulties Scale (n=68,86[MTC Cycle 5] Global Health Status (n=51,77)[MTC Cycle 5] Physical Functional Scale (n=51,77)[MTC Cycle 5] Role Functional Scale (n=51,77)[MTC Cycle 5] Emotional Functional Scale (n=51,77)[MTC Cycle 5] Cognitive Functional Scale (n=51,77)[MTC Cycle 5] Social Functional Scale (n=51,77)[MTC Cycle 5] Fatigue Symptom Scale (n=51,77)[MTC Cycle 5] Nausea and Vomiting Scale (n=51,77)[MTC Cycle 5] Pain Symptom Scale (n=51,77)[MTC Cycle 5] Dyspnea Symptom Scale (n=51,77)[MTC Cycle 5] Insomnia Symptom Scale( n=51,77)MTC Cycle 5] Appetite Loss Symptom Scale (n=51,77)[MTC Cycle 5] Constipation Symptom Scale (n=51,77)[MTC Cycle 5] Diarrhea Symptom Scale (n=51,77)[MTC Cycle 5] Financial Difficulties Scale n=50,77[MTC Cycle 7] Global Health Status Scale (n=38,64)[MTC Cycle 7] Physical Functional Scale (n=38,64)[MTC Cycle 7]Role Functional Scale (n=38,64)[MTC Cycle 7]Emotional Functional Scale (n=38,64)[MTC Cycle 7]Cognitive Functional Scale (n=38,64)[MTC Cycle 7]Social Functional Scale (n=38,64)[MTC Cycle 7]Fatigue Symptom Scale (n=38,64)[MTC Cycle 7]Nausea & Vomiting Scale (n=38,64)[MTC Cycle 7]Pain Symptom Scale (n=38,64)[MTC Cycle 7]Dyspnea Symptom Scale (n=38,64)[MTC Cycle 7]Insomnia Symptom Scale (n=38,64)[MTC Cycle 7]Appetite Loss Scale (n=38,64)[MTC Cycle 7]Constipation Symptom Scale (n=38,64)[MTC Cycle 7]Diarrhea Symptom Scale (n=38,64)[MTC Cycle 7]Financial Difficulties Scale n=38,64[MTC Cycle 9 ]Global Health Status Scale (n=33,50)[MTC Cycle 9 ]Physical Functional Scale (n=33,50)[MTC Cycle 9 ]Role Functional Scale (n=33,50)[MTC Cycle 9 ]Emotional Functional Scale (n=33,50)[MTC Cycle 9 ]Cognitive Functional Scale (n=33,50)[MTC Cycle 9 ]Social Functional Scale (n=33,50)[MTC Cycle 9 ]Fatigue Symptom Scale (n=33,50)[MTC Cycle 9 ]Nausea & Vomiting Symptom (n=33,50)[MTC Cycle 9 ]Pain Symptom Scale (n=33,50)[MTC Cycle 9 ]Dyspnea Symptom Scale (n=32,50)[MTC Cycle 9 ]Insomnia Symptom Scale (n=33,49)[MTC Cycle 9 ]Appetite Loss Scale (n=33,50)[MTC Cycle 9 ]Constipation Symptom Scale (n=33,50)[MTC Cycle 9 ]Diarrhea Symptom Scale (n=33,50)[MTC Cycle 9]Financial Difficulties Scale n=33,50[MTC Cycle 11 ] Global Health Status Scale n=25,37[MTC Cycle 11 ]Physical Functional Scale (n=25,37)[MTC Cycle 11 ]Role Functional Scale (n=25,37)[MTC Cycle 11 ]Emotional Functional Scale n=25,37[MTC Cycle 11 ]Cognitive Functional Scale n=25,37[MTC Cycle 11 ]Social Functional Scale (n=25,37)[MTC Cycle 11 ]Fatigue Symptom Scale (n=25,37)[MTC Cycle 11 ]Nausea & Vomiting Scale (n=25,37)[MTC Cycle 11 ]Pain Symptom Scale (n=25,37)[MTC Cycle 11 ]Dyspnea Symptom Scale (n=25,37)[MTC Cycle 11 ]Insomnia Symptom Scale (n=25,37)[MTC Cycle 11 ]Appetite Loss Scale (n=25,37)MTC Cycle 11 ]Constipation Symptom Scale( n=25,37)[MTC Cycle 11 ]Diarrhea Symptom Scale ( n=25,37)[MTC Cycle 11]Financial Difficulties Scale n=24,37[EOS]Global Health Status Scale (n=123,127)[EOS]Physical Functional Scale (n=125,127)[EOS]Role Functional Scale (n=125,127)[EOS]Emotional Functional Scale (n=124,127)[EOS]Cognitive Functional Scale ( n=124,127)[EOS]Social Functional Scale (n=124,127)[EOS]Fatigue Symptom Scale (n=125,127)[EOS]Nausea & Vomiting Scale (n=125,127)[EOS]Pain Symptom Scale (n=125,127)[EOS]Dyspnea Symptom Scale (n=124,127)[EOS]Insomnia Symptom Scale( n=125,127)[EOS]Appetite Loss Symptom Scale (n=125,127)[EOS]Constipation Symptom Scale (n=125,127)[EOS]Diarrhea Symptom Scale(n=124,127)[EOS]Financial Difficulties Scale (n=123,126)
Bevacizumab +Pemetrexed Maintenance Trt Arm B59.779.471.169.888.078.731.54.324.730.633.918.79.15.217.258.676.069.978.482.673.034.58.817.429.918.419.511.15.719.462.978.468.878.784.677.934.510.020.135.922.119.917.32.617.761.374.770.877.581.072.133.79.619.83728.621.417.28.920.361.274.969.77581.771.735.112.018.735.330.623.316.07.322.059.981.173.980.084.277.931.29.519.828.818.91817.17.218.057.272.564.377.383.572.637.612.321.134.122.626.012.16.816.9
Bevacizumab Maintenance Trt Arm A57.974.868.471.786.079.333.37.236.729.733.319.218.67.117.363.377.174.282.584.382.132.17.223.926.113.519.310.13.418.163.278.774.578.881.085.332.29.223.525.521.610.59.210.516.061.275.676.881.677.282.932.38.321.528.116.712.3148.814.060.680.875.882.383.383.828.35.125.825.023.210.113.17.115.257.779.277.378.381.384.732.46.729.330.720.09.36.71.315.355.170.263.574.479.373.341.014.430.431.527.226.917.69.716.8

[back to top]

Overall Survival

Overall survival (defined as the number of days from the day of first treatment to death (from any cause), or until the last day if we know that the patient is alive). (NCT00976456)
Timeframe: 42 months

Interventionmonths (Median)
Bevacizumab + Pemetrexed11.6
Bevacizumab + Pemetrexed + Carboplatin14.4

[back to top]

Progression Free Survival

Progression free survival (defined as the number of days from the day of the first treatment until day of death (from any cause) or progression, whichever occurs earlier, or until the day of the last response assessment, if no progression or death (from any cause) is observed during the study). (NCT00976456)
Timeframe: 42 months

Interventionmonths (Median)
Bevacizumab + Pemetrexed4.8
Bevacizumab + Pemetrexed + Carboplatin6.8

[back to top]

AUC0-inf of Pemetrexed

Area under the concentration-time curve of pemetrexed in plasma over the time interval from 0 extrapolated to infinity (AUC0-inf) (NCT00979576)
Timeframe: 5 minutes (min) before pemetrexed administration and 10min, 40min, 1 hour (h), 2h, 4h, 6h, 23h 55min, 47h 55min after pemetrexed administration in cycles 1 and 2

Interventionng*h/mL (Geometric Mean)
Cycle 1Cycle 2 (N=11)
Pemetrexed 500 mg/m2194000200000

[back to top]

Adverse Events According to Common Terminology Criteria for Adverse Events (CTCAE), Version 3.0 for All Courses

"Number of patients with adverse events according to worst Common Terminology Criteria for Adverse Events (CTCAE), version 3.0 for all courses.~CTCAE grades are: 1 (mild AE), 2 (moderate AE), 3 (severe AE), 4 (life-threatening or disabling AE) or 5 (death related to AE)." (NCT00979576)
Timeframe: Between first administration of pemetrexed and 28 days after last administration of pemetrexed and/or BIBF 1120, up to 1020 days

,,
Interventionparticipants (Number)
Grade 1Grade 2Grade 3Grade 4Grade 5
BIBF 1120 100 mg + Pemetrexed 500 mg/m^202100
BIBF 1120 150 mg + Pemetrexed 500 mg/m^201500
BIBF 1120 200 mg + Pemetrexed 500 mg/m^212510

[back to top]

Duration of Disease Control

Duration of disease control was defined as the time period from the first study drug administration to the progressive disease (PD) or death of patients, whichever occurred earlier. (NCT00979576)
Timeframe: From first study drug administration until PD or death, up to 1003 days

InterventionDays (Median)
BIBF 1120 100 mg + Pemetrexed 500 mg/m^2248.5
BIBF 1120 150 mg + Pemetrexed 500 mg/m^2228.5
BIBF 1120 200 mg + Pemetrexed 500 mg/m^2149.0

[back to top]

Dose Limiting Toxicities

Number of participants with dose limiting toxicity (DLT) in combination therapy of BIBF 1120 and pemetrexed during the first course (NCT00979576)
Timeframe: During the first course, 21 days

InterventionParticipants (Number)
BIBF 1120 100 mg + Pemetrexed 500 mg/m^20
BIBF 1120 150 mg + Pemetrexed 500 mg/m^21
BIBF 1120 200 mg + Pemetrexed 500 mg/m^22

[back to top]

AUC0-inf of Nintedanib

Area under the concentration-time curve of nintedanib in plasma over the time interval from 0 extrapolated to infinity (AUC0-inf) (NCT00979576)
Timeframe: 5 minutes (min) before nintedanib administration and 1h, 2h, 3h, 4h, 6h, 7h, 10h and 23h 55min after nintedanib administration in cycle 1

Interventionng*h/mL (Geometric Mean)
BIBF 1120 100 mg + Pemetrexed 500 mg/m^2105
BIBF 1120 150 mg + Pemetrexed 500 mg/m^2232
BIBF 1120 200 mg + Pemetrexed 500 mg/m^2323

[back to top]

Disease Control Rate

Number of participants with complete response (CR), partial response (PR) or stable disease (SD) according to the Response Evaluation Criteria In Solid Tumors (RECIST) 1.0 (NCT00979576)
Timeframe: Every 6 weeks after start of study treatment until end of treatment, up to 992 days

InterventionParticipants (Number)
BIBF 1120 100 mg + Pemetrexed 500 mg/m^22
BIBF 1120 150 mg + Pemetrexed 500 mg/m^24
BIBF 1120 200 mg + Pemetrexed 500 mg/m^26

[back to top]

Cmax of Nintedanib

Maximum measured concentration of nintedanib in plasma (Cmax) (NCT00979576)
Timeframe: 5 minutes (min) before nintedanib administration and 1h, 2h, 3h, 4h, 6h, 7h, 10h and 23h 55min after nintedanib administration in cycle 1

Interventionng/mL (Geometric Mean)
BIBF 1120 100 mg + Pemetrexed 500 mg/m^220.5
BIBF 1120 150 mg + Pemetrexed 500 mg/m^237.9
BIBF 1120 200 mg + Pemetrexed 500 mg/m^255.1

[back to top]

Overall Response Rate

Number of participants with complete response (CR) or partial response (PR) according to the Response Evaluation Criteria In Solid Tumors (RECIST) 1.0 (NCT00979576)
Timeframe: Every 6 weeks after start of study treatment until end of treatment, up to 992 days

,,
InterventionParticipants (Number)
Complete responsePartial response
BIBF 1120 100 mg + Pemetrexed 500 mg/m^200
BIBF 1120 150 mg + Pemetrexed 500 mg/m^201
BIBF 1120 200 mg + Pemetrexed 500 mg/m^201

[back to top]

Number of Participants With Clinically Relevant Abnormalities in Laboratory Parameters

Number of participants with clinically relevant abnormalities in laboratory parameters reported as adverse events which occurred in >= 20% of patients (NCT00979576)
Timeframe: Between first administration of pemetrexed and 28 days after last administration of pemetrexed and/or BIBF 1120, up to 1020 days

,,
Interventionparticipants (Number)
Alanine aminotransferase increasedGamma-glutamyltransferase increasedAspartate aminotransferase increasedLymphocyte count decreasedNeutrophil count decreasedWhite blood cell count decreasedBlood alkaline phosphatase increasedBlood albumin decreasedHaemoglobin decreased
BIBF 1120 100 mg + Pemetrexed 500 mg/m^2222222001
BIBF 1120 150 mg + Pemetrexed 500 mg/m^2655132111
BIBF 1120 200 mg + Pemetrexed 500 mg/m^2887243433

[back to top]

Cmax of Pemetrexed

Maximum measured concentration of pemetrexed in plasma (Cmax) (NCT00979576)
Timeframe: 5 minutes (min) before pemetrexed administration and 10min, 40min, 1 hour (h), 2h, 4h, 6h, 23h 55min, 47h 55min after pemetrexed administration in cycles 1 and 2

Interventionng/mL (Geometric Mean)
Cycle 1Cycle 2 (N=11)
Pemetrexed 500 mg/m2139000149000

[back to top]

Pharmacokinetics (PK): Minimum Concentration (Cmin) of Necitumumab

(NCT00982111)
Timeframe: Predose Day 1 of Cycle 2,3,4,5 and 6 Prior to Necitumumab Infusion, Up to 23 Weeks

Interventionmicrograms/milliliter (ug/ml) (Geometric Mean)
Predose Cycle 2 Day 1Predose Cycle 3 Day 1Predose Cycle 4 Day 1Predose Cycle 5 Day 1Predose Cycle 6 Day 1
Necitumumab + Pemetrexed + Cisplatin57.580.8110115119

[back to top]

Overall Survival Time (OS)

OS is defined as the time from randomization to death from any cause. Participants who do not die at the end of the extended follow-up period, or were lost to follow-up during the study, were censored at the last date they were known to be alive. OS was estimated using the Kaplan-Meier method. (NCT00982111)
Timeframe: Randomization to Death from Any Cause (Up to 31.6 Months)

InterventionMonths (Median)
Necitumumab + Pemetrexed + Cisplatin11.3
Pemetrexed + Cisplatin11.5

[back to top]

Progression-Free Survival (PFS)

PFS is defined as the time from randomization until the first radiographic documentation of measured progressive disease as defined by RECIST (Version 1.0), or death from any cause. Participants who die without a reported prior progression will be considered to have progressed on the day of their death. Participants who did not progress or were lost to follow-up were censored at the day of their last radiographic tumor assessment. If no baseline or postbaseline radiologic assessment was available, the participant was censored at the date of randomization. If death or PD occurs after two or more consecutive missing radiographic visits, censoring occurred at the date of the last radiographic visit prior to the missed visits. (NCT00982111)
Timeframe: Randomization to Measured Progressive Disease or Death from Any Cause (Up to 30.4 Months)

InterventionMonths (Median)
Necitumumab + Pemetrexed + Cisplatin5.6
Pemetrexed + Cisplatin5.6

[back to top]

Time to Treatment Failure (TTF)

TTF was defined as the time from study enrollment/randomization to the first observation of measured progressive disease, death from any cause, or early discontinuation of treatment or initiation of new anti-cancer therapies. Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria. Progressive Disease (PD) was defined as having at least a 20% increase in sum of longest diameter of target lesions. Time to treatment failure was censored at the date of the last follow-up visit for participants who did not discontinue early, who were still alive, and who have not progressed. (NCT00982111)
Timeframe: Randomization to Measured Progressive Disease, Death from Any Cause, Discontinuation of Treatment or Initiation of New Anticancer Therapy (Up to 30.4 Months)

InterventionMonths (Median)
Necitumumab + Pemetrexed + Cisplatin3.5
Pemetrexed + Cisplatin4.3

[back to top]

Percentage of Participants Who Achieve Best Overall Tumor Response of Complete Response (CR) or Partial Response (PR) (Objective Tumor Response Rate [ORR])

ORR is confirmed best overall tumor response of CR or PR. According to RECIST v1.0, CR was defined as the disappearance of all target and non-target lesions; PR defined as a >30% decrease in the sum of the longest diameters (LD) of the target lesions, taking as reference the baseline sum of the LD. Percentage of participants was calculated as: (total number of participants with CR or PR from start of the treatment until disease progression or recurrence)/total number of participants treated) * 100. (NCT00982111)
Timeframe: Baseline to Measured Progressive Disease (Up to 30.4 Months)

Interventionpercentage of participants (Number)
Necitumumab + Pemetrexed + Cisplatin31.1
Pemetrexed + Cisplatin32.1

[back to top]

Percentage of Participants With EGFR Measured by IHC

EGFR IHC H-score = weighted sum of % 1+ cells, twice % 2+ cells, and three times % 3+ cells. IHC H-score criteria assesses participants with a low EGFR expression defined by a H-score cutoff value of < 200 and participants with a high EGFR expression defined by a H-score of cutoff value of >=200. (NCT00982111)
Timeframe: Baseline

,
Interventionpercentage of participants (Number)
H-score <200H-score >=200
Necitumumab + Pemetrexed + Cisplatin58.841.2
Pemetrexed + Cisplatin59.640.4

[back to top]

Number of Participants With Serum Anti-Necitumumab Antibody Assessment (Immunogenicity)

A participant was considered to have an anti-Necitumumab antibody response if anti-drug antibodies (ADA) were confirmed positive. Treatment emergent antibodies were defined as any anti-Necitumumab antibody titer equal to or greater than 4-fold the participant's baseline titer. (NCT00982111)
Timeframe: Baseline to Study Completion (Up to 31.6 Months)

Interventionparticipants (Number)
1 Positive TiterAntibodies Detected
Necitumumab + Pemextrexed + Cisplatin3718

[back to top]

Epidermal Growth Factor Hormone (EGFR) Protein Expression Measured by Immunohistochemistry (IHC)

EGFR IHC H-score = weighted sum of % 1+ cells, twice % 2+ cells, and three times % 3+ cells. IHC H-score criteria assesses participants with a low EGFR expression defined by a H-score cutoff value of < 200 and participants with a high EGFR expression defined by a H-score of cutoff value of >=200. (NCT00982111)
Timeframe: Baseline

,
InterventionH-Score (Mean)
H-score <200H-score >=200
Necitumumab + Pemetrexed + Cisplatin69.06259.35
Pemetrexed + Cisplatin66.23256.26

[back to top]

Mean Change From Baseline in Patient Reported Outcomes (PRO) Using the European Quality of Life-5 Dimensions (EQ-5D)

The EQ-5D is a generic, multidimensional, health-related, quality-of-life instrument. The profile allows participants to rate their health state in 5 health domains: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression using a three level scale 1-3 (no problem, some problems, and major problems). These combinations of attributes were converted into a weighted health-state Index Score according to the United Kingdom (UK) population-based algorithm. The possible values for the Index Score ranged from -0.59 (severe problems in all 5 dimensions) to 1.0 (no problem in any dimension). (NCT00982111)
Timeframe: Baseline, Cycle 6 (Cycle = 3 weeks)

Interventionunits on a scale (Mean)
Necitumumab + Pemetrexed + Cisplatin0.0419
Pemetrexed + Cisplatin0.0478

[back to top]

Mean Change From Baseline in PRO as Measured Using the Lung Cancer Symptom Scale (LCSS)

The LCSS consisted of 9 items: 6 items focused on lung cancer symptoms [loss of appetite, fatigue, cough, dyspnea (shortness of breath), hemoptysis (blood in sputum), and pain] and 3 items were global items (symptom distress, interference with activity level, and global quality of life). Participant responses to each item were measured using visual analogue scales (VAS) with 100-mm lines. A higher score for any item represented a higher level of symptoms/problems. Scores for each of the reported categories ranged from 0 (for best outcome) to 100 (for worst outcome). The Average Symptom Burden Index (ASBI) was the mean of the 6 symptom items of the LCSS, and the Total LCSS was the mean of all 9 LCSS items. ASBI and Total LCSS were not computed for a participant if he/she had 1 or more missing values for the 6 and 9 items, respectively. (NCT00982111)
Timeframe: Baseline, Cycle 6 (Cycle =3 Weeks)

,
Interventionmillimeter (mm) (Mean)
Loss of AppetiteFatigueCoughDyspneaHemoptysisPainOverall SymptomsQuality of LifeInterferenceAverage Symptom Burden Index (ASBI)LCSS Total Score
Necitumumab + Pemetrexed + Cisplatin4.64.5-9.1-2.8-1.1-4.2-3.12.53.2-0.90.1
Pemetrexed + Cisplatin0.61.6-10.3-1.5-1.1-7.1-7.4-3.3-4.0-3.1-4.3

[back to top]

Duration of Response

Duration of Response is defined as the time from the first observation of CR or PR to the first observation of progressive disease (PD) or death from any cause. A response is defined as a confirmed objective status of CR or PR. For participants who are not known to have died as of the data inclusion cut-off date and who do not have PD, the duration will be censored at the date of the last objective progression free disease assessment prior to the date of any subsequent anticancer therapy. (NCT00988858)
Timeframe: First Observation of CR or PR until Progressive Disease or Death Due to Any Cause (Up to 23 Months)

Interventionmonths (Median)
LY2603618 and Pemetrexed8.7

[back to top]

Overall Tumor Response - Percentage of Participants Achieving Complete Response (CR) or Partial Response (PR) [Overall Response Rate (ORR)]

Overall response rate is the best response of complete response (CR) or partial response (PR) as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST v1.1). CR is a disappearance of all target and non-target lesions and normalization of tumor marker level. PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of not-target lesions or appearance of new lesions. Overall response rate is calculated as a total number of participants with CR or PR divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT00988858)
Timeframe: Baseline until Progressive Disease or Study Discontinuation (Up to 23 Months)

Interventionpercentage of participants (Number)
LY2603618 and Pemetrexed9.1

[back to top]

Percentage of Participants Who Achieved a Best Response of Complete Response (CR), Partial Response (PR), or Stable Disease (SD) (Clinical Benefit Rate)

Clinical benefit rate is the best response CR, PR, or stable disease (SD) as classified by the investigators according to the RECIST v1.1. CR is a disappearance of all target and non-target lesions and normalization of tumor marker level. PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of not-target lesions or appearance of new lesions. SD is neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum diameter since treatment started. Clinical benefit rate is calculated as a total number of participants with CR, PR, or SD divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT00988858)
Timeframe: Baseline until Progressive Disease or Study Discontinuation (Up to 23 Months)

Interventionpercentage of participants (Number)
LY2603618 and Pemetrexed45.5

[back to top]

Progression-free Survival (PFS)

Progression-free survival (PFS) time was defined as the time from the date of randomization to the first date of progressive disease (symptomatic or objective) or death due to any cause, whichever occurred first. For participants who were not known to have died or progressed as of the data-inclusion cutoff date, PFS time was censored at the date of the last objective progression-free disease assessment prior to the date of any subsequent systematic anticancer therapy. PFS was summarized using Kaplan-Meier estimates. (NCT00988858)
Timeframe: Baseline to Progressive Disease or Death Due to Any Cause (Up to 27.1 Months)

Interventionmonths (Median)
LY2603618 and Pemetrexed2.3

[back to top]

Pharmacokinetics (PK): Maximum Plasma Concentration (Cmax) of LY2603618

(NCT00988858)
Timeframe: Day 2 and Day 3 of Cycle 1 and Cycle 2: Prior to End of Infusion (EOI); EOI + 1-2 hr; EOI + 4-6 hr; EOI + 20-28 hr; anytime on Day 8 of Cycle 1 and Cycle 2

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Day 2/Cycle 1 (n=41)Day 2/Cycle 2 (n=48)
LY2603618 and Pemetrexed34303560

[back to top]

PK: Area Under the Plasma Concentration vs. Time Curve From Time Zero to Infinity [AUC(0-∞)] of LY2603618

(NCT00988858)
Timeframe: Day 2 and Day 3 of Cycle 1 and Cycle 2: Prior to End of Infusion (EOI); EOI + 1-2 hr; EOI + 4-6 hr; EOI + 20-28 hr; anytime on Day 8 of Cycle 1 and Cycle 2

Interventionnanograms*hour per milliliter (ng*h/mL) (Geometric Mean)
Day 2/Cycle 1 (n=41)Day 2/Cycle 2 (n=48)
LY2603618 and Pemetrexed3800041500

[back to top]

Change in Symptom Burden Scores of Lung Cancer Symptom Scale (LCSS)

The LCSS participants scale is a 9-item questionnaire. Six questions are symptom-specific measures for lung cancer (appetite, fatigue, cough, dyspnea, hemoptysis and pain), and 3 summation items describe total symptomatic distress, activity status, and overall quality of life. Participant responses were measured using visual analogue scales (VAS) with 100-milliliter (mm) lines. Scores range from 0 (for best outcome) to 100 (for worst outcome). The Average Symptom Burden Index (ASBI) was calculated as the mean of 6 symptom-specific questions from the LCSS. (NCT00988858)
Timeframe: Baseline until End of Study (Up to 27.1 Months)

Interventionparticipants (Number)
ImprovedWorsenedStableUnknown
LY2603618 and Pemetrexed1261810

[back to top]

PK: Maximum Plasma Concentration (Cmax) of Pemetrexed

(NCT00988858)
Timeframe: Day 1 and Day 2 of Cycle 1 and Cycle 2: Prior to End of Infusion (EOI); EOI + 1-2 hour (hr); EOI + 4-6- hr; EOI + 20-28 hr

Interventionmicrogram per milliliter (μg/mL) (Geometric Mean)
Day 1/Cycle 1 (n=40)Day 1/Cycle 2 (n=43)
LY2603618 and Pemetrexed10296.8

[back to top]

PK: Area Under the Plasma Concentration vs. Time Curve From Time Zero to Infinity [AUC(0-∞)] of Pemetrexed

(NCT00988858)
Timeframe: Day 1 and Day 2 of Cycle 1 and Cycle 2: Prior to End of Infusion (EOI); EOI + 1-2 hour (hr); EOI + 4-6- hr; EOI + 20-28 hr

Interventionmicrogram*hour per milliliter (µg*hr/mL) (Geometric Mean)
Day 1/Cycle 1(n=40)Day 1/Cycle 2(n=43)
LY2603618 and Pemetrexed193202

[back to top]

Number of Participants With an Objective Tumor Response

Participants with confirmed complete response (CR), confirmed partial response (PR), stable disease (SD), or progressive disease (PD) according to Response Evaluation Criteria In Solid Tumors (RECIST, version 1.0) criteria, as well as participants with a not evaluable/tumor response unknown. CR: disappearance of all tumor lesions. PR: either a) at least a 30% decrease in sum of longest diameter (LD) of target lesions taking as a reference baseline sum LDs, or b) complete disappearance of target lesions, with persistence (not worsening) of 1 or more nontarget lesions. In either case, no new lesions appeared. SD: small changes that did not meet above criteria. PD: at least a 20% increase in sum of LD of target lesions taking as reference smallest sum LD recorded since treatment started or appearance of 1 or more new lesions. Participants who discontinued study treatment (for reasons other than progression) before entering concurrent phase were considered to have non-evaluable response. (NCT01000480)
Timeframe: Date of first dose through end of follow-up [up to 30 weeks (1 cycle=21 days)]

Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseDisease ProgressionNot evaluable/Response unknown
Pemetrexed, Cisplatin, and Thoracic Radiotherapy94516128

[back to top]

Overall Survival

Overall survival (OS) was the duration from enrollment to death due to any cause. Participants who were alive were censored at the last contact. (NCT01000480)
Timeframe: Date of first dose to date of death (up to 35.4 months)

Interventionmonths (Median)
Pemetrexed, Cisplatin, and Thoracic Radiotherapy26.2

[back to top]

1 Year Progression Free Survival

Progression free survival (PFS) was defined as the time from study enrollment to the first observation of progressive disease (PD) or death from any cause. For participants not known to have died as of the data cut-off date and who did not have objective PD, PFS was censored at the date of the last objective progression-free disease assessment. For participants who received subsequent systemic anticancer therapy (after discontinuation from the study drug) prior to objectively determined PD or death, PFS was censored at the date of the last objective progression-free disease assessment prior to start of postdiscontinuation chemotherapy. If a participant did not have a complete baseline disease assessment, then PFS was censored at the enrollment date, regardless whether or not objectively determined PD or death had been observed for the participant. (NCT01000480)
Timeframe: Date of first dose to date of objectively determined PD or death [every cycle up to 4 cycles and then every 3 months up to 1 year (1 cycle=21 days)]

Interventionpercentage of participants (Number)
Pemetrexed, Cisplatin, and Thoracic Radiotherapy53.7

[back to top]

Progression-free Interval

Kaplan-Meier curve will be used to examine all the time-to-event data points in analyzing progression free interval. (NCT01001910)
Timeframe: Time from the first day of treatment to the day that progression is first noted, assessed up to 4.5 years

Interventionmonths (Median)
Treatment (Pemetrexed Disodium, Carboplatin)6.8

[back to top]

Incidence of Toxicities

Secondary outcome included detailed measurement of adverse events from treatment assessed according to National Cancer Institute (NCI) Common Toxicity Criteria (CTC) version 3.0. (NCT01001910)
Timeframe: 4.5 years

InterventionFrequency of event (Number)
Hematologic AE Any GradeNon-Hematologic AE Any Grade
Treatment (Pemetrexed Disodium, Carboplatin)24060

[back to top]

Overall Survival (OS)

Kaplan-Meier method will be used t analyze the time-to-event data including overall survival (OS) and progression-free survival (PFS) (NCT01001910)
Timeframe: First day of treatment on protocol to the date of death, or for living patients the last date of contact, assessed up to 4.5 years

Interventionmonths (Median)
PFSOS
Treatment (Pemetrexed Disodium, Carboplatin)6.850.3

[back to top]

Percentage of Participants With Confirmed Complete Response or Partial Response During the Maintenance Therapy Only

CR and PR defined per RECIST Guidelines, Version 1.0. CR is disappearance of all tumor lesions. PR is either a) at least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LDs, or b) complete disappearance of target lesions, with persistence (but not worsening) of 1 or more nontarget lesions. In either case, no new lesions may have appeared. (NCT01004250)
Timeframe: From the start of the maintenance to the first date of objectively determined PD during the maintenance therapy (assessment during maintenance treatment completed at every other cycle till PD and at 30 day follow-up)(Cycle 5 up to 104.1 Weeks)

InterventionPercentage of Participants (Number)
Study Treament11.1

[back to top]

Percentage of Participants With Confirmed Complete Response or Partial Response During Study Treatment (Induction and Maintenance)

Overall Response Rate (ORR) is defined as the percentage of participants whose best response is complete response (CR) or partial response (PR) per RECIST Guidelines, Version 1.0. CR is disappearance of all tumor lesions. PR is either a) at least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LDs, or b) complete disappearance of target lesions, with persistence (but not worsening) of 1 or more nontarget lesions. In either case, no new lesions may have appeared. (NCT01004250)
Timeframe: From enrollment to objectively determined PD (assessment during study treatment completed at every other cycle till PD and at 30 day follow-up)(Baseline up to 104.1 Weeks)

InterventionPercentage of Participants (Number)
Study Treatment42.2

[back to top]

Overall Survival

Overall Survival (OS) is defined as the time from the date of study enrollment to the date of death from any cause. For participants not known to have died as of the data cut-off date, OS will be censored at the last contact date. (NCT01004250)
Timeframe: From enrollment to the date of death from any cause (every cycle during study treatment, every 6 weeks during follow-up period until PD, and then at least every 3 Months) (Baseline up to 36.3 Months)

InterventionMonths (Median)
Study Treatment14.7

[back to top]

Progression-Free Survival

Progression-Free Survival (PFS) is defined as the time from the date of study enrollment to the first date of objectively determined PD or death from any cause. PD is defined using Response Evaluation Criteria in Solid Tumours (RECIST) Guidelines (Version 1.0), as at least a 20% increase in the sum of longest diameter (LD) of target lesions, taking as references the smallest sum LD recorded since the treatment started or the appearance of 1 or more new lesions. For participants not known to have died as of the data cut-off date and who do not have objective PD, PFS will be censored at the date of the last objective progression-free disease assessment. For participants who receive subsequent systemic anticancer therapy, PFS will be censored at the date of the last objective progression-free disease assessment prior to post-discontinuation systemic therapy. (NCT01004250)
Timeframe: From enrollment to the first date of objectively determined Progressive Disease (PD) or death from any cause (every other cycle during study treatment and then every 6 weeks during follow-up period)(Baseline up to 36.1 Months)

InterventionMonths (Median)
Study Treatment6.9

[back to top]

Percentage of Participants With Confirmed Response Complete or Partial Response During the Induction Treatment Only

CR and PR defined per RECIST Guidelines, Version 1.0. CR is disappearance of all tumor lesions. PR is either a) at least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LDs, or b) complete disappearance of target lesions, with persistence (but not worsening) of 1 or more nontarget lesions. In either case, no new lesions may have appeared. (NCT01004250)
Timeframe: From the time of study enrollment to the first date of objectively determined PD during the induction therapy (assessment during study treatment completed at every other cycle up to four cycles) (Baseline up to 4 cycles)

InterventionPercentage of Participants (Number)
Study Treatment34.9

[back to top]

Time to Treatment Failure (TtTF)

TtTF was defined as date of randomization until the date of discontinuation of study treatment due to adverse event, progressive disease (PD), or death from any cause. PD assessed using Response Evaluation Criteria in Solid Tumor (RECIST v1.0) criteria and 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 ever recorded since study treatment started, or the appearance of 1 or more new lesions. Participants who discontinued study treatment for any other reason were censored at the date of discontinuation of study treatment. Participants still on study drug at data-inclusion cut-off date were censored at the cut-off date. (NCT01005680)
Timeframe: Randomization until date of discontinuation of study treatment due to adverse events, PD, or death from any cause up to 6.3 months post-randomization

Interventionmonths (Median)
Pemetrexed Plus Cisplatin (PC)NA
Gemcitabine Plus Cisplatin (GC)NA

[back to top]

Time to Progressive Disease (TtPD)

TtPD defined as the time from study randomization to the first date of progressive disease (PD). PD assessed using Response Evaluation Criteria in Solid Tumor (RECIST v1.0) criteria and 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 ever recorded since study treatment started, or the appearance of 1 or more new lesions. Participants who were not known to have PD or who died without PD were censored at the date of last of last tumor assessment. (NCT01005680)
Timeframe: Randomization to first date of PD up to 23.7 months post-randomization

Interventionmonths (Median)
Pemetrexed Plus Cisplatin (PC)5.82
Gemcitabine Plus Cisplatin (GC)5.82

[back to top]

Tumor Response Rate

Tumor response rate was the percentage of participants with confirmed best tumor response of complete response (CR) or partial response (PR) using Response Evaluation Criteria in Solid Tumor (RECIST v1.0) 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. Progressive disease (PD) assessed using RECIST v1.0 criteria and 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 ever recorded since study treatment started, or the appearance of 1 or more new lesions. (NCT01005680)
Timeframe: Randomization until date of objective PD or death from any cause up to 35.8 months post-randomization

Interventionpercentage of participants (Number)
Pemetrexed Plus Cisplatin (PC)24.8
Gemcitabine Plus Cisplatin (GC)20.7

[back to top]

Survival Without Toxicity (SWT)

SWT was defined as the time from randomization to a study-drug related toxicity. Toxicity was defined as Common Terminology Criteria for Adverse Events (CTCAE v3.0) Grade 3 or 4 or death. Participants who do not have a CTCAE Grade 3 or higher toxicity and are alive will be censored at the date of last contact. (NCT01005680)
Timeframe: Randomization to date of toxicity or date of death up to 34.6 months post-randomization

Interventionmonths (Median)
Pemetrexed Plus Cisplatin (PC)5.85
Gemcitabine Plus Cisplatin (GC)2.56

[back to top]

Risk/Benefit Ratio

Risk/benefit ratio was calculated as the percentage of participants who experienced a study-drug related toxicity of Common Terminology Criteria for Adverse Events (CTCAE v3.0) Cancer Therapy Evaluation Program (CTEP) Grade 3 or higher, divided by the Kaplan-Meier estimated percentage of participants surviving one year. (NCT01005680)
Timeframe: Randomization to date of death from any cause up to 35.8 months post-randomization

Interventionratio (Number)
Pemetrexed Plus Cisplatin (PC)0.70
Gemcitabine Plus Cisplatin (GC)0.83

[back to top]

Progression Free Survival (PFS)

PFS was defined as the date of randomization to date of first observation of clinical or objective progressive disease (PD) or death due to any cause. PD assessed using Response Evaluation Criteria in Solid Tumor (RECIST v1.0) criteria and 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 ever recorded since study treatment started, or the appearance of one or more new lesions. Participants who were not known to have died or had PD were censored at the date of last contact. (NCT01005680)
Timeframe: Randomization to first date of Progressive Disease (PD) or death from any cause up to 33.0 months post-randomization

Interventionmonths (Median)
Pemetrexed Plus Cisplatin (PC)5.88
Gemcitabine Plus Cisplatin (GC)5.85

[back to top]

Overall Survival (OS)

OS was defined as the duration from date of randomization to date of death from any cause. Participants who were alive were censored at the date of last contact. (NCT01005680)
Timeframe: Randomization to date of death from any cause up to 35.8 months post-randomization

Interventionmonths (Median)
Pemetrexed Plus Cisplatin (PC)17.54
Gemcitabine Plus Cisplatin (GC)15.51

[back to top]

Duration of Response (DoR)

DoR was defined as the time from first objective status assessment of complete response (CR) or partial response (PR) to the first time progressive disease (PD) or death as a result of any cause. Response using Response Evaluation Criteria in Solid Tumor (RECIST v1.0) criteria. CR was defined as the disappearance of all target lesions. PR was defined as having at least a 30% decrease in sum of longest diameter of target lesions. Participants who are not known to have died or to have PD were censored at the date of last contact. PD assessed using RECIST v1.0 and 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 ever recorded since study treatment started, or the appearance of 1 or more new lesions (NCT01005680)
Timeframe: Date of first response to the date of (PD) or death from any cause up to 22.9 months post-randomization

Interventionmonths (Median)
Pemetrexed Plus Cisplatin (PC)4.53
Gemcitabine Plus Cisplatin (GC)4.98

[back to top]

Disease Control Rate (DCR)

DCR was the percentage of participants with Complete Response (CR), Partial Response (PR), and Stable Disease (SD). Response determined using Response Evaluation Criteria In Solid Tumors (RECIST v1.0) criteria. CR was defined as the disappearance of all target lesions; PR was defined as at least a 30% decrease in sum of longest diameter of target lesions; progressive disease (PD) was 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 ever recorded since study treatment started, or the appearance of 1 or more new lesions; SD was defined as small changes that did not meet the above criteria. (NCT01005680)
Timeframe: Randomization to date of objective PD or death from any cause up to 35.8 months post-randomization

Interventionpercentage of participants (Number)
Pemetrexed Plus Cisplatin (PC)78.5
Gemcitabine Plus Cisplatin (GC)75.9

[back to top]

Time to Progressive Disease (TtPD)

TtPD was defined as the time from randomization to the first date of objectively determined progressive disease (PD). For participants who were not known to have had objective progression of disease as of the data-inclusion cut-off date for a particular analysis, or who had died without objective progression of disease, TtPD was censored at the date of the participant's last objective progression-free disease assessment prior to cut-off date. (NCT01017874)
Timeframe: Randomization to the first date of measured PD up to 37.32 months

Interventionmonths (Median)
Pemetrexed + Cisplatin + Gefitinib8.61
Gefitinib9.69

[back to top] [back to top]

Percentage of Participants With Complete Response (CR) or Partial Response (PR) [Tumor Response Rate (TRR)]

TRR was defined as the percentage of randomized participants having a best overall study response of CR or PR using Response Evaluation Criteria in Solid Tumors (RECIST v1.0) criteria. CR was defined as the disappearance of all target lesions; PR was defined as at least a 30% decrease in sum of longest diameter (LD) of target lesions taking as reference the baseline sum LDs or complete disappearance of target lesions, with persistence (but not worsening) of 1 or more non-target lesions and the appearance of no new lesions. (NCT01017874)
Timeframe: Randomization up to 37.52 months

Interventionpercentage of participants (Number)
Pemetrexed + Cisplatin + Gefitinib41.5
Gefitinib47.5

[back to top]

Percentage of Participants With Complete Response (CR), Partial Response (PR) or Stable Disease (SD) [Disease Control Rate (DCR)]

DCR was defined as the percentage of randomized participants with overall response of CR, PR or SD using Response Evaluation Criteria in Solid Tumors (RECIST v1.0) criteria. CR was defined as the disappearance of all tumor lesions; PR was defined as at least a 30% decrease in sum of longest diameter (LD) of target lesions taking as reference the baseline sum LDs or complete disappearance of target lesions, with persistence (but not worsening) of 1 or more non-target lesions and no new lesions having appeared; SD defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease (PD) taking as reference the smallest sum LD. PD defined as at least 20% increase in the sum of LD of target, lesions taking as reference, the smallest sum LD recorded since the treatment started or the appearance of 1 or more new lesions or progression of nontarget lesions. (NCT01017874)
Timeframe: Randomization up to 37.52 months

Interventionpercentage of participants (Number)
Pemetrexed + Cisplatin + Gefitinib71.2
Gefitinib64.4

[back to top]

Progression Free Survival (PFS)

PFS was defined as the time from date of randomization to the objective disease progression or death due to any cause. Response was defined using Response Evaluation Criteria in Solid Tumors (RECIST v1.0) criteria. Progressive disease (PD) was defined as at least a 20% increase in the sum of longest diameter (LD) of target lesions taking as references the smallest sum LD recorded since the treatment started or the appearance of 1 or more new lesions and/or unequivocal progression of existing nontarget lesions. Participants who did not have a complete baseline disease assessment were censored at the date of randomization, regardless if PD was objectively determined or if participant died or if a participant was not known to have died or have objective PD at the data inclusion cutoff date. PFS was censored at the last complete objective progression-free disease assessment date. (NCT01017874)
Timeframe: Randomization to the first date of measured PD or death up to 37.32 months

Interventionmonths (Median)
Pemetrexed + Cisplatin + Gefitinib8.38
Gefitinib9.63

[back to top]

Overall Survival (OS)

OS was the duration from randomization to the date of death from any cause. For participants who were not known to have died as of the data-inclusion cut-off date for a particular analysis, OS was censored at the date of last contact prior to the data inclusion cutoff date (contacts considered in the determination of last contact date included adverse event date, lesion assessment date, visit date, and last known alive date). (NCT01017874)
Timeframe: Randomization up to date of death from any cause up to 57.13 months

Interventionmonths (Median)
Pemetrexed + Cisplatin + Gefitinib26.87
Gefitinib27.86

[back to top]

Duration of Tumor Response

The duration of a complete response (CR) or partial response (PR) using Response Evaluation Criteria in Solid Tumors (RECIST v1.0) criteria was defined as the time from first objective status assessment of CR or PR to the first time of objective disease progression or death as a result of any cause. CR was defined as the disappearance of all tumor lesions. PR was defined as at least a 30% decrease in sum of longest diameter (LD) of target lesions taking as reference the baseline sum of LDs or complete disappearance of target lesions, with persistence (but not worsening) of 1 or more non-target lesions and no new lesions having appeared. Participants who were not known to have died or had objective progression of disease as of the data-inclusion cut-off date were censored at the date of the participant's last complete objective progression-free disease assessment prior to that cut-off date. (NCT01017874)
Timeframe: Date of initial response to the date of measured PD or death up to 34.43 months

Interventionmonths (Median)
Pemetrexed + Cisplatin + Gefitinib12.09
Gefitinib11.93

[back to top]

Overall Survival (OS) During the Maintenance Therapy Period

OS was defined as the duration from the date of the first dose of the maintenance therapy to the date of death from any cause and was calculated by subtracting the induction therapy period from OS. Participants receiving any subsequent systemic anticancer therapy before objective progression or death were censored at date of last objective progression-free disease assessment before starting subsequent systemic anticancer therapy. For participants who were alive, OS was censored at the last contact. (NCT01020786)
Timeframe: From the start of maintenance therapy in Cycle 5 (21-day cycle) until the date of measured progressive disease (PD) or death from any cause (up to 26.3 months)

Interventionmonths (Median)
Overall Survival (OS) at primary endpointOverall Survival (OS) at final endpoint
Pemetrexed + CarboplatinNANA

[back to top]

Percentage of Participants Who Achieved a Complete Response (CR) or Partial Response (PR) During the Induction and Maintenance Therapy Periods

"Calculated as the percentage of participants who achieved a confirmed CR or PR. Tumor response was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) guideline version 1.0, which define when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments. CR = disappearance of all target lesions. PR = 30% decrease in the sum of the longest diameter of target lesions. Progressive Disease (PD) = 20% increase in the sum of the longest diameter of target lesions. Stable Disease (SD) = small changes that do not meet above criteria." (NCT01020786)
Timeframe: Enrollment to date of progressive disease (up to 18 months)

Interventionpercentage of participants (Number)
CRPR
Pemetrexed + Carboplatin0.034.9

[back to top]

Percentage of Participants Who Achieved a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) During the Maintenance Therapy Period

"Percentage of participants who achieved confirmed CR (disappearance of all target lesions), PR (30% decrease in sum of longest diameter of target lesions), or SD (small changes that do not meet above criteria). Response derived from target lesion assessments performed before maintenance therapy (as baseline), during maintenance therapy (as post-baseline), and non-target lesion assessments performed during maintenance therapy according to RECIST guideline version 1.0, defines when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments." (NCT01020786)
Timeframe: From the start of maintenance therapy in Cycle 5 (21-day cycle) until the date of measured progressive disease (PD) or death from any cause (up to 18 months)

Interventionpercentage of participants (Number)
CR+PRCR+PR+SD
Pemetrexed + Carboplatin3.348.3

[back to top]

Progression Free Survival (PFS) During the Maintenance Therapy Period

"Measured from the date of the first dose of the maintenance therapy. Calculated by subtracting induction therapy period from PFS. Tumor response assessed using Response Evaluation Criteria in Solid Tumors (RECIST) guideline version 1.0; define when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments. Participants receiving any subsequent systemic anticancer therapy before objective progression or death were censored at date of last objective progression-free disease assessment before starting subsequent systemic anticancer therapy." (NCT01020786)
Timeframe: From the start of maintenance therapy in Cycle 5 (21-day cycle) until the date of measured progressive disease (PD) or death from any cause (up to 24.4 months)

Interventionmonths (Median)
PFS at primary endpointPFS at final endpoint
Pemetrexed + Carboplatin3.93.9

[back to top]

Percentage of Participants Who Achieve a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) During the Induction and Maintenance Therapy Periods

"Calculated as the percentage of participants who achieved a confirmed CR, PR, or SD. Tumor response was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) guideline version 1.0, which define when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments. CR = disappearance of all target lesions. PR = 30% decrease in the sum of the longest diameter of target lesions. Progressive Disease (PD) = 20% increase in the sum of the longest diameter of target lesions. SD = small changes that do not meet above criteria." (NCT01020786)
Timeframe: Enrollment to date of progressive disease (up to 18 months)

Interventionpercentage of participants (Number)
CR+PRCR+PR+SD
Pemetrexed + Carboplatin34.972.5

[back to top]

Overall Survival (OS) During the Induction and Maintenance Therapy Periods

OS was defined as the time from the enrollment date to the date of death from any cause. For participants who were alive, OS was censored at the last contact. (NCT01020786)
Timeframe: Enrollment to the date of death from any cause (up to 30.8 months)

Interventionmonths (Median)
Overall Survival (OS) at primary endpointOverall Survival (OS) at final endpoint
Pemetrexed + CarboplatinNA20.2

[back to top]

Percentage of Participants Who Achieve a Complete Response (CR) or a Partial Response (PR) During the Induction Therapy Period

"Calculated as percentage of participants who achieved a CR or PR (confirmed or not). Tumor response was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) guideline version 1.0, which define when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments. CR = disappearance of all target lesions. PR = 30% decrease in sum of the longest diameter of target lesions. Progressive Disease (PD) = 20% increase in the sum of longest diameter of target lesions. Stable Disease (SD) = small changes that do not meet above criteria." (NCT01020786)
Timeframe: Enrollment to date of PD, or end of induction period up to Cycle 4 (21-day cycle)

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin38.5

[back to top]

Percentage of Participants Who Observe a Complete Response (CR), Partial Response (PR), or Stable Disease (SD) During the Induction Therapy Period

"Calculated as the percentage of participants who achieved a CR, PR, or SD (confirmed or not). Tumor response was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) guideline version 1.0, which define when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments. CR = disappearance of all target lesions. PR = 30% decrease in the sum of the longest diameter of target lesions. Progressive Disease (PD) = 20% increase in the sum of the longest diameter of target lesions. SD = small changes that do not meet above criteria." (NCT01020786)
Timeframe: Enrollment to the date of PD, or end of induction period up to Cycle 4 (21-day cycle)

Interventionpercentage of participants (Number)
Pemetrexed + Carboplatin81.7

[back to top]

Progression Free Survival (PFS) During the Induction and Maintenance Therapy Periods

"PFS defined as time from enrollment date to first date of objective progression of disease or of death from any cause. Tumor response was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) guideline version 1.0, which define when cancer participants improve (respond), stay the same (stabilize), or worsen (progression) during treatments. Participants receiving any subsequent systemic anticancer therapy before objective progression or death were censored at date of last objective progression-free disease assessment before starting subsequent systemic anticancer therapy." (NCT01020786)
Timeframe: Enrollment to the date of progressive disease (PD) or the date of death from any cause (up to 18 months)

Interventionmonths (Median)
Pemetrexed + Carboplatin5.6

[back to top]

Incidence of Toxicities as Assessed by NCI CTCAE v. 4.0

The overall toxicity rates (percentages) for grade 3 or higher adverse events considered at least possibly related to treatment (NCT01041781)
Timeframe: Up to 5 years

Interventionpercentage of patients (Number)
Arm I (Arm A: Celecoxib + Standard Chemotherapy)61.04
Arm II (Arm B: Placebo + Standard Chemotherapy)55.06

[back to top]

Prognostic Value of Urinary Prostaglandin Metabolites (PGE-M) Levels for Worse PFS for Patients Who Had Baseline Urinary PGE-M Above/Below the Median Quartile (Q2)

prognostic value of urinary prostaglandin metabolites (PGE-M) levels for worse PFS for patients who had baseline urinary PGE-M above/below the median quartile (Q2, 15.38). Progression free survival (PFS) is defined as the time from the date of randomization to the date of disease progression or death resulting from any cause, whichever comes first. Progression is defined according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01041781)
Timeframe: Up to 5 years

Interventionmonths (Median)
PGE-M < Q26.2
PGE-M >= Q24.2

[back to top]

Prognostic Value of Urinary Prostaglandin Metabolites (PGE-M) Levels for Worse PFS for Patients Who Had Baseline Urinary PGE-M Above/Below the Third Quartile (Q3)

Prognostic value of urinary prostaglandin metabolites (PGE-M) levels for worse PFS for patients who had baseline urinary PGE-M above/below the median quartile (Q3, 27.86). Progression free survival (PFS) is defined as the time from the date of randomization to the date of disease progression or death resulting from any cause, whichever comes first. Progression is defined according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01041781)
Timeframe: Up to 5 years

Interventionmonths (Median)
PGE-M < Q36.0
PGE-M >= Q33.0

[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 according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01041781)
Timeframe: Time between randomization and disease relapse or death from any cause, assessed up to 5 years

Interventionmonths (Median)
Arm I (Arm A: Celecoxib + Standard Chemotherapy)5.16
Arm II (Arm B: Placebo + Standard Chemotherapy)5.26

[back to top]

Prognostic Value of Urinary Prostaglandin Metabolites (PGE-M) Levels for Worse PFS for Patients Who Had Baseline Urinary PGE-M Above/Below the First Quartile (Q1)

Prognostic value of urinary prostaglandin metabolites (PGE-M) levels for worse PFS for patients who had baseline urinary PGE-M above/below the first quartile (Q1, 10.09). Progression free survival (PFS) is defined as the time from the date of randomization to the date of disease progression or death resulting from any cause, whichever comes first. Progression is defined according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01041781)
Timeframe: Up to 5 years

Interventionmonths (Median)
PGE-M < Q17.7
PGE-M >= Q14.9

[back to top]

Response Rate

The response rate (percentage) is the percent of patients whose best response was Complete Response (CR) or Partial Response (PR) as defined by RECIST 1.1 criteria. Percentage of successes will be estimated by 100 times the number of successes divided by the total number of evaluable patients. Response rates (including complete and partial response) will be tested using Fisher's exact test (NCT01041781)
Timeframe: Up to 5 years

Interventionpercentage of patients (Number)
Arm I (Arm A: Celecoxib + Standard Chemotherapy)40
Arm II (Arm B: Placebo + Standard Chemotherapy)35

[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. (NCT01041781)
Timeframe: Time between randomization and death from any cause, assessed up to 5 years

Interventionmonths (Median)
Arm I (Arm A: Celecoxib + Standard Chemotherapy)11.4
Arm II (Arm B: Placebo + Standard Chemotherapy)12.5

[back to top]

Median Overall Survival (OS)

Defined as the time between Day 1-Cycle 1 to the date of death from any cause. (NCT01042288)
Timeframe: 18 months

Interventionmonths (Median)
Carboplatin/Pemetrexed/Panitumumab17.02

[back to top]

Median Progression-free Survival (PFS)

Defined as the time between Day 1-Cycle 1 and date of first documented disease progression or death. (NCT01042288)
Timeframe: Assessments by clinical evaluation, radiographic status, and date of disease progression, estimated 18 months

Interventionmonths (Median)
Carboplatin/Pemetrexed/Panitumumab5.75

[back to top]

Median Time to Progression (TTP)

Defined as the time between Day 1-Cycle 1 and date of first documented disease progression assessed using Response Evaluation Criteria in Solid Tumors (RECISTS) v1.1. (NCT01042288)
Timeframe: 18 months

Interventionmonths (Median)
Carboplatin/Pemetrexed/Panitumumab6.11

[back to top]

Frequency of Adverse Events and Severity as a Measure of Toxicity

Assessed using NCI CTCAE v4.0 (NCT01042288)
Timeframe: Every 3 weeks (1 cycle) for 6 cycles, then every 7 weeks thereafter

Interventionparticipants (Number)
RashNauseaFatigueHypomagnesemiaNeutrophil Count DecreasedPlatelet Count DecreasedAnemiaMucositisDry SkinAnorexiaDiarrheaConstipationWhite Blood Cell DecreasedPruritusVomitingDehydrationDysgeusiaAlopecia
Carboplatin/Pemetrexed/Panitumumab553933303029282322191917161515131211

[back to top]

Objective Response Rate

(NCT01042288)
Timeframe: Projected 18 months

Interventionpercentage of evaluated participants (Number)
Carboplatin/Pemetrexed/Panitumumab53

[back to top]

Progression Free Survival (PFS)

PFS based on Response Evaluation Criteria in Solid Tumors (RECIST) Guidelines defined as the time from the date of first dose of study drug to first documented objective progressive disease (PD) or death from any cause. PD is defined as at least a 20% increase in the sum of longest diameter (LD) of target lesions taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions. (NCT01057589)
Timeframe: Baseline to date of PD or death up to 18.7 months

Interventionmonths (Median)
Pemetrexed Cisplatin Cetuximab4.4

[back to top]

Percent of Participants With a Partial Response (PR) or a Complete Response (CR)

CR and PR based on RECIST Guidelines: CR is defined as the disappearance of all tumor lesions; PR is defined as at least a 30% decrease in the sum of the LD of target lesions taking as reference the baseline sum LDs or the complete disappearance of target lesions, with persistence (but not worsening) of one or more nontarget lesions and the appearance of no new lesions. PD is defined as at least a 20% increase in the sum of 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. (NCT01057589)
Timeframe: Date of first response to PD (up to 18.7 months)

Interventionpercentage of participants (Number)
Pemetrexed Cisplatin Cetuximab29.3

[back to top]

Change From Baseline in Performance Status Scale for Head and Neck Cancer Patients (PSS-HNC)

PSS-HNC is a clinician-rated instrument designed to measure speaking and eating disabilities of participants with head and neck cancer and consists of 3 subscales: Normalcy of Diet (NOD) subscale measures the ability of the participants to eat a normal diet, scale ranged from 0 (non-oral feeding) to 100 (unrestricted diet); Understandability of Speech (UOS)subscale measured the degree a clinician was able to understand the participant's speech, subscale ranged from 0 (never understandable) to 100 (always understandable); Eating in Public (EIP) subscale, rating based on clinician question to the participant to report who he/she eats with and in what setting, subscale ranged from 0 (always eats alone) to 100 (no restriction of place, food, or companion). Change from baseline: negative value represents a decrease in function and a positive value represents an increase in function. (NCT01057589)
Timeframe: Baseline, Triplet Combination Therapy Cycles 2, 4, 6 (cycle = 21 days) and optional Maintenance Therapy Cycles 1, 3, 5 and 7 (cycle = 21 days)

Interventionunits on a scale (Mean)
NOD-Change at Triplet Cycle 2 (n=53)NOD-Change at Triplet Cycle 4 (n=41)NOD-Change at Triplet Cycle 6 (n=24)NOD-Change at Maintenance Cycle 1 (n=23)NOD-Change at Maintenance Cycle 3 (n=10)NOD-Change at Maintenance Cycle 5 (n=6)NOD-Change at Maintenance Cycle 7 (n=5)EIP-Change at Triplet Cycle 2 (n=46)EIP-Change at Triplet Cycle 4 (n=37)EIP-Change at Triplet Cycle 6 (n=22)EIP-Change at Maintenance Cycle 1 (n=23)EIP-Change at Maintenance Cycle 3 (n=10)EIP-Change at Maintenance Cycle 5 (n=6)EIP-Change at Maintenance Cycle 7 (n=5)UOS-Change at Triplet Cycle 2 (n=53)UOS-Change at Triplet Cycle 4 (n=41)UOS-Change at Triplet Cycle 6 (n=24)UOS-Change at Maintenance Cycle 1 (n=23)UOS-Change at Maintenance Cycle 3 (n=10)UOS-Change at Maintenance Cycle 5 (n=6)UOS-Change at Maintenance Cycle 7 (n=5)
Pemetrexed Cisplatin Cetuximab-0.43.93.30.4-1.08.3-8.02.7-6.8-3.4-8.7-10.0-4.2-15.00.5-4.3-10.4-1.1-2.5-12.5-15.0

[back to top]

Change From Baseline in Participant Reported European-Quality of Life 5 Dimension Instrument (EQ-5D) Visual Analog Scale (VAS) at End of Triplet Combination Therapy and End of Maintenance Therapy

Vertical VAS - a 20 millimeter (mm), fractionated scale in the form of a thermometer with endpoints of 0 (worst imaginable health state) and 100 (best imaginable health state). Participants used the EQ-5D VAS scale to rate their overall health on the day the questionnaire was administered. Possible change values range from -100 (best imaginable health at baseline changed to worst possible health at visit) to 100 (worst possible health at baseline changed to best possible health at visit). (NCT01057589)
Timeframe: Baseline, End of Triplet Combination Therapy (up to Cycle 6 [4.2 months]), End of Maintenance Therapy (up to 18.7 months)

Interventionunits on a scale (Mean)
Change at End of Triplet Therapy (n=23)Change at End of Maintenance Therapy (n=11)
Pemetrexed Cisplatin Cetuximab-1.2-10.6

[back to top]

Overall Survival (OS)

OS defined as the time from the date of first dose of study drug to the date to death from any cause. (NCT01057589)
Timeframe: Baseline to date of death up to 18.7 months

Interventionmonths (Median)
Pemetrexed Cisplatin Cetuximab9.7

[back to top]

Change From Baseline in Participant Reported EQ-5D Utility Score at End of Triplet Combination Therapy and End of Maintenance Therapy

EQ-5D Index is derived by converting the Descriptive System (participant is required to rate health by checking 1 [no limitation], 2 [some limitation] or 3 [severe or complete limitation] in 5 dimensions [mobility, self-care, usual activities, pain/comfort and anxiety/depression]) to a single summary index. A utility value assigned to each individual's health state based on the absence or presence of moderate or severe problems in the 5 dimensions. A regression equation defines a utility value for these health states. The possible values for health utility ranged from -0.59 (severe problems in all 5 dimensions) to 1 (no problem in all dimensions) on a scale where 0 represents death and 1 represents the best possible health state. Possible change values range from -1.59 (no problems at baseline to severe problems at visit) to 1.59 (severe problems at baseline to no problems at visit). (NCT01057589)
Timeframe: Baseline, End of Triplet Combination Therapy (up to 6 cycles [4.2 months]) , End of Maintenance Therapy (up to 18.7 months)

Interventionunits on a scale (Mean)
Change at End of Triplet Therapy (n=29)Change at End of Maintenance Therapy (n=15)
Pemetrexed Cisplatin Cetuximab0.05-0.02

[back to top]

Response Rate

Percentage of patients with a complete or partial 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. (NCT01063283)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Group A3
Group B2

[back to top]

Change in 24 Hour Diastolic Blood Pressure (DBP)

The change for each patient was calculated as mean 24 hour DBP during cycle 2 - mean 24 hour DBP during cycle 1 (NCT01063283)
Timeframe: 2 cycles

InterventionmmHg (Mean)
Group A3
Group B3

[back to top]

Response Rate by Modified RECIST (Phase II)

"Per modified RECIST for Pleural Tumors. In addition to RECIST1.1, for modified RECIST, measurements based on the sum of 6 CT cuts in the pleural perpendicular to the chest wall are applied to standard RECIST criterial (sum of 6 = one univariate diameter). Complete Response (CR), Disappearance of all measurable and non-measurable disease; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR.~All target measurable lesions must be assessed using the same techniques as baseline." (NCT01064648)
Timeframe: Disease assessment will be repeated every 6 weeks until disease progression, up to 3 years.Best response is documented for as long as the patient remains on protocol treatment.

Interventionpercentage of analyzed participants (Number)
Phase II Cisplatin-pemetrexed Cediranib 20mg50
Phase II Cisplatin-pemetrexed Placebo20

[back to top]

Overall Survival (Phase II)

From date of registration to date of death due to any cause. Patients last known to be alive are censored at date of last contact. (NCT01064648)
Timeframe: From date of registration to death.Disease assessment will be repeated every 6 weeks until disease progression. After progression, follow up will occur every 6 months for the first two years and then at the end of the third year after registration.

Interventionmonth (Median)
Phase II Cisplatin-pemetrexed Cediranib 20mg10
Phase II Cisplatin-pemetrexed Placebo8.5

[back to top]

Maximum Tolerated Dose of Cediranib in Combination With Cisplatin and Pemetrexed (Phase I)

"MTD was determined by testing dose-de-escalation to 20mg PO daily on dose de-escalation cohort 1 to 2 with 3 to 6 patients each. MTD reflects the highest dose of drug that did not cause a Dose-Limiting Toxicity (DLT) in > 33% of participants. Toxicities will be graded according to the CTEP Active Version of the NCI Common Terminology Criteria for Adverse Events. Dose-limiting toxicities (DLT) apply only during Cycle 1 and must be drug-related (i.e. possibly, probably or definitely related to one of the 3 study drugs). The following events occurring in the first cycle of treatment are considered dose limiting.~Febrile neutropenia~Grade 4 neutrophil count decrease for more than 7 days' duration~Grade 4 platelet count decrease~Grade 3 or 4 non-hematologic toxicity (excluding alopecia)" (NCT01064648)
Timeframe: Weekly during first cycle (1cycle = 21 days). Then will be reported every cycle while patient is on treatment.

Interventionmg (Number)
All Phase I Participants20

[back to top]

Disease Control Rate by RECIST 1.1 (Phase II)

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1): Complete Response (CR), Disappearance of all measurable and non-measurable disease; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (STA): Does not qualify for CR, PR, Progression or Symptomatic Deterioration. Disease Control Rate (DCR) = CR + PR + STA~All target measurable lesions must be assessed using the same techniques as baseline." (NCT01064648)
Timeframe: Disease assessment will be repeated every 6 weeks until disease progression, up to 3 years.Disease control rate is documented for as long as the patient remains on protocol treatment.

Interventionpercentage of analyzed participants (Number)
Phase II Cisplatin-pemetrexed Cediranib 20mg74
Phase II Cisplatin-pemetrexed Placebo80

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

Progression-free Survival (Phase II)

From date of registration to date of first documentation of progression or symptomatic deterioration, or death due to any cause. Patients last known to be alive and progression free are censored at date of last contact. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v 1.1), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesions, or the appearance of new lesions. (NCT01064648)
Timeframe: From date of registration to date of first documentation of progression or symptomatic deterioration, or death due to any cause, whichever came first, assessed up to 5 years.Disease assessment will be repeated every 6 weeks until disease progression.

Interventionmonth (Median)
Phase II Cisplatin-pemetrexed Cediranib 20mg7.2
Phase II Cisplatin-pemetrexed Placebo5.6

[back to top]

Disease Control Rate by Modified RECIST (Phase II)

"Per modified RECIST for Pleural Tumors. In addition to RECIST1.1, for modified RECIST, measurements based on the sum of 6 CT cuts in the pleural perpendicular to the chest wall are applied to standard RECIST criterial (sum of 6 = one univariate diameter). Complete Response (CR), Disappearance of all measurable and non-measurable disease; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Stable Disease (STA), does not qualify for CR, PR, Progression or Symptomatic Deterioration. Disease Control Rate (DCR) = CR + PR + STA.~All target measurable lesions must be assessed using the same techniques as baseline." (NCT01064648)
Timeframe: Disease assessment will be repeated every 6 weeks until disease progression, up to 3 years.Disease control is documented for as long as the patient remains on protocol treatment.

Interventionpercentage of analyzed participants (Number)
Phase II Cisplatin-pemetrexed Cediranib 20mg75
Phase II Cisplatin-pemetrexed Placebo83

[back to top]

Response Rate by RECIST1.1 (Phase II)

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1): Complete Response (CR), Disappearance of all measurable and non-measurable disease; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR.~All target measurable lesions must be assessed using the same techniques as baseline." (NCT01064648)
Timeframe: Disease assessment will be repeated every 6 weeks until disease progression, up to 3 years. Best response is documented for as long as the patient remains on protocol treatment.

Interventionpercentage of analyzed participants (Number)
Phase II Cisplatin-pemetrexed Cediranib 20mg26
Phase II Cisplatin-pemetrexed Placebo15

[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. (NCT01085630)
Timeframe: Baseline up to 3 years

Interventionmonths (Median)
Arm A (Observation)11.8
Arm B (Pemetrexed)16.3

[back to top]

Progression-free Survival

Progression free survival (PFS) is defined as the time from the date of randomization to the date of disease progression or death resulting from any cause, whichever comes first. Progression is defined according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The median and 95% confidence intervals are estimated using the Kaplan-Meier estimator. (NCT01085630)
Timeframe: Baseline up to 3 years

Interventionmonths (Median)
Arm A (Observation)3.0
Arm B (Pemetrexed)3.4

[back to top]

Toxicity, Assessed Using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 (v4)

The number of patients reporting grade 3 or higher adverse events considered at least possibly related to study treatment as graded by the NCI's Common Toxicity Criteria (CTCAE) Version 4 are reported here. A complete list of all reported adverse events is reported in the Adverse Events section of this report. (NCT01085630)
Timeframe: Baseline up to 3 years

InterventionParticipants (Count of Participants)
Grade 3 Adverse EventGrade 4 Adverse Event
Arm B (Pemetrexed)92

[back to top]

Response Rate

The response rate (percentage) is the percent of patients whose best response was Complete Response (CR) or Partial Response (PR) as defined by RECIST 1.1 criteria. Percentage of successes will be estimated by 100 times the number of successes divided by the total number of evaluable patients. Response rates (including complete and partial response) will be tested using Fisher's exact test (NCT01085630)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Arm A (Observation)0
Arm B (Pemetrexed)11.1

[back to top]

Overall Survival (OS)

OS is defined as the duration from the date of enrollment to the date of death from any cause. For participants who were alive at the time of the data inclusion cutoff, OS was censored at the date of last contact prior to that cut-off date. (NCT01087970)
Timeframe: From enrollment to the date of death from any cause up to 26.4 months (assessment completed during trial period at least every 3 months)

Interventionmonths (Median)
Cetuximab + Pemetrexed + Carboplatin/Cisplatin11.1

[back to top]

Percentage of Participants Having a Confirmed Partial Response (PR) or Complete Response (CR)

PR or CR is classified by the investigators according to RECIST criteria version 1.0. PR is a ≥30% decrease in sum of longest diameter of target lesions without new lesion and progression of non-target lesions; CR is the disappearance of all target and non-target lesions. Percentage of participants having a PR or CR is calculated as a total number of participants with PR or CR from enrollment until disease progression or recurrence divided by the total number of participants treated, then multiplied by 100. (NCT01087970)
Timeframe: From enrollment to objectively determined progressive disease up to 15.3 months (tumor assessments performed every other cycle during study treatment until progressive disease)

Interventionpercentage of participants (Number)
Cetuximab + Pemetrexed + Carboplatin/Cisplatin25.9

[back to top]

Progression-Free Survival (PFS)

PFS was defined as the duration from the date of enrollment to the first date of documented objective progressive disease (PD) or death from any cause. PD was determined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria version 1.0. PD is ≥20% increase in sum of longest diameter of target lesions or the appearance of new lesions. For participants who were not known to have died or to have had objective PD at the time of the data inclusion cutoff, PFS was censored at their last objective progression-free disease assessment prior to the cutoff date or the date of initiation of subsequent systemic anticancer therapy. (NCT01087970)
Timeframe: From enrollment to measured progressive disease up to 15.3 months (tumor assessments performed every other cycle during study treatment, and then every 6 weeks during follow-up)

Interventionmonths (Median)
Cetuximab + Pemetrexed + Carboplatin/Cisplatin5.1

[back to top]

Change From Baseline in Performance Status Scale for Head and Neck Cancer (PSS-HNC)

PSS-HNC is a clinician-rated instrument designed to measure speaking and eating disabilities of participants with head and neck cancer and consists of 3 subscales: normalcy of diet (NOD) subscale measures the ability of the participants to eat a normal diet, subscale ranges from 0 (non-oral feeding) to 100 (unrestricted diet); understandability of speech (UOS) subscale measures the degree a clinician is able to understand the participant's speech, subscale ranges from 0 (never understandable) to 100 (always understandable); eating in public (EIP) subscale, rating based on the participant's response to the questions of whom he/she eats with and in what setting, subscale ranges from 0 (always eats alone) to 100 (no restriction of place, food, or companion). Change from baseline: negative value represents a decrease in function and a positive value represents an increase in function. (NCT01087970)
Timeframe: Baseline, Day 1 of Cycles 2, 4, 6, cetuximab monotherapy Cycles 2 and 4 (21-day cycle)

Interventionunits on a scale (Mean)
Change in NOD at Cycle 2 (n=52)Change in UOS at Cycle 2 (n=52)change in EIP at Cycle 2 (n=52)Change in NOD at Cycle 4 (n=34)Change in UOS at Cycle 4 (n=35)Change in EIP at Cycle 4 (n=34)Change in NOD at Cycle 6 (n=28)Change in UOS at Cycle 6 (n=28)Change in EIP at Cycle 6 (n=28)Change in NOD at monotherapy Cycle 2 (n=16)Change in UOS at monotherapy Cycle 2 (n=16)Change in EIP at monotherapy Cycle 2 (n=17)Change in NOD at monotherapy Cycle 4 (n=9)Change in UOS at monotherapy Cycle 4 (n=9)Change in EIP at monotherapy Cycle 4 (n=9)
Cetuximab + Pemetrexed + Carboplatin/Cisplatin4.62-2.402.883.24-0.71-6.629.29-1.79-1.7911.88-4.69-4.418.892.782.78

[back to top]

Number of Participants Who Died While on Treatment and Died During 30-Day Post-Treatment Discontinuation Follow-Up (FU)

Presented are the number of participants who died due to adverse events (AEs) while on treatment and participants who died due to progressive disease (PD) during the 30-day post-treatment discontinuation FU. (NCT01087970)
Timeframe: From enrollment to 30 days post-treatment discontinuation up to 26.4 months

Interventionparticipants (Number)
Due to AE while on treatmentDue to PD during 30-day FU
Cetuximab + Pemetrexed + Carboplatin/Cisplatin35

[back to top]

Change From Baseline in Participant-Reported European-Quality of Life-5 Dimension Instrument-3 Levels (EQ-5D-3L)

EQ-5D-3L is a descriptive system of health-related quality of life states consisting of 5 dimensions (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression) and each of which has 3 levels of severity (no problems/some or moderate problems/extreme problems) within a particular EQ-5D dimension. A regression equation defines a utility value for these health states to generate an index score. The possible values for index score range from -0.594 (severe problems in all 5 dimensions) to 1 (no problem in all dimensions) on a scale where 1 represents the best possible health state. The EQ-5D Visual Analog Scale (VAS) is used to record a participant's rating for his/her current health-related quality of life state on the day of questionnaire administration and is captured on a scale of 0 (worst imaginable health state) to 100 (best imaginable health state). (NCT01087970)
Timeframe: Baseline, Day 1 of Cycles 2, 4, 6, cetuximab monotherapy Cycles 2 and 4 (21-day cycle)

Interventionunits on a scale (Mean)
Change in Index Score at Cycle 2 (n=43)Change in Index Score at Cycle 4 (n=29)Change in Index Score at Cycle 6 (n=24)Change in Index Score at monotherapy Cycle2 (n=14)Change in Index Score at monotherapy Cycle 4 (n=7)Change in VAS Score at Cycle 2 (n=41)Change in VAS Score at Cycle 4 (n=28)Change in VAS Score at Cycle 6 (n=21)Change in VAS Score at monotherapy Cycle 2 (n=14)Change in VAS Score at monotherapy Cycle 4 (n=7)
Cetuximab + Pemetrexed + Carboplatin/Cisplatin0.0380.070-0.0630.0160.0593.8542.1070.333-1.5000.571

[back to top]

Response Rate

"Response is evaluated based on RECIST criteria v1.1 and defined as either complete response or partial response. Complete response is defined as disappearance of all lesions. Partial response is defined as at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the current step's baseline sum diameters.~The primary analysis is among patients who were randomized to the maintenance therapy. Patients who received induction therapy only and did not participate in the randomization part of the study were not included in this analysis." (NCT01107626)
Timeframe: Assessed every 6 weeks during induction therapy and every 3 cycles during maintenance therapy; after discontinuation of study therapy, assessed every 3 months for 2 years and every 6 months for years 3-5

Interventionproportion of participants (Number)
Arm A (Induction Then Maintenance With Bevacizumab)0.125
Arm B (Induction Then Maintenance With Pemetrexed0.187
Arm C (Induction Then Maintenance With Bevacizumab and Pemetrexed)0.212

[back to top]

Overall Survival

"Overall survival is defined as the time from randomization to death or date last known alive.~The primary analysis is among patients who were randomized to the maintenance therapy. Patients who received induction therapy only and did not participate in the randomization part of the study were not included in this analysis." (NCT01107626)
Timeframe: Assessed every 6 weeks during induction therapy and every 3 cycles during maintenance therapy; after discontinuation of study therapy, assessed every 3 months for 2 years and every 6 months for years 3-5

Interventionmonths (Median)
Arm A (Induction Then Maintenance With Bevacizumab)14.4
Arm B (Induction Then Maintenance With Pemetrexed15.9
Arm C (Induction Then Maintenance With Bevacizumab and Pemetrexed)16.4

[back to top]

Progression-free Survival

"Progression-free survival is defined as the time from randomization to progression or death, whichever occurs first. Progression is evaluated based on RECIST criteria and defined as appearance of one or more new lesions, unequivocal progression of existing non-target lesions, or at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on current step. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm.~The primary analysis is among patients who were randomized to the maintenance therapy. Patients who received induction therapy only and did not participate in the randomization part of the study were not included in this analysis." (NCT01107626)
Timeframe: Assessed every 6 weeks during induction therapy and every 3 cycles during maintenance therapy; after discontinuation of study therapy, assessed every 3 months for 2 years and every 6 months for years 3-5

Interventionmonths (Median)
Arm A (Induction Then Maintenance With Bevacizumab)4.2
Arm B (Induction Then Maintenance With Pemetrexed5.1
Arm C (Induction Then Maintenance With Bevacizumab and Pemetrexed)7.5

[back to top]

Phase 2: Percentage of Participants Who Experienced TEAEs

Safety assessment included monitoring and recording all AE including all CTCAE version 4.0 grades (for both increasing and decreasing severity), and SAE; regular monitoring of hematology, blood chemistry, and urine values; periodic measurement of vital signs and ECGs; and performance of physical examinations. A TEAE was defined as an AE that had on onset date, or a worsening in severity from Baseline (pretreatment), on or after the first dose of study drug up to the end of the study. (NCT01126736)
Timeframe: From date of first dose up to 30 days after the last dose of study drug in Phase 2, up to approximately 3 years 6 months

Interventionpercentage of participants (Number)
Phase 2, Arm 1: 0.9 mg/m^2 Eribulin Plus 500 mg/m^2 Pemetrexed97.6
Phase 2, Arm 2: 500 mg/m^2 Pemetrexed94.9

[back to top]

Phase 1b: Percentage of Participants With Grade 3 or Higher Treatment Emergent Adverse Events (TEAEs)

Safety assessment included monitoring and recording all AE including all Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 grades (for both increasing and decreasing severity), and serious adverse events (SAE); regular monitoring of hematology, blood chemistry, and urine values; periodic measurement of vital signs and electrocardiograms (ECGs); and performance of physical examinations. A TEAE was defined as an AE that had on onset date, or a worsening in severity from Baseline (pretreatment), on or after the first dose of study drug up to the end of the study. As per CTCAE, Grade 1 scales as Mild; Grade 2 scales as Moderate; Grade 3 scales as severe or medically significant but not immediately life threatening; Grade 4 scales as life-threatening consequences; and Grade 5 scales as death related to AE. (NCT01126736)
Timeframe: From date of first dose up to 30 days after the last dose of study drug in Phase 1b, up to approximately 1 year 2 months

Interventionpercentage of participants (Number)
Phase 1b, Arm 1 - Cohort 1: 0.9 mg/m^2 Eribulin Plus Pemetrexed100.0
Phase 1b, Arm 1 - Cohort 2: 1.4 mg/m^2 Eribulin Plus Pemetrexed83.3
Phase 1b, Arm 2 - Cohort 1: 0.7 mg/m^2 Eribulin Plus Pemetrexed100.0

[back to top]

Phase 2: Progression-free Survival (PFS)

PFS was defined as the time from the date of randomization until the earlier of the following two events: the date of progressive disease (PD) or the date of death. Progressive disease was defined as at least a 20 percent (%) increase in the sum of the longest diameter of target lesions (taking as reference the smallest sum on study), recorded since the treatment started or the appearance of 1 or more new lesions based on investigator assessments according to Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1). If a participant did not progress, they were censored at the date of last tumor assessment, last known alive date, or until the start of a next line of therapy, whichever occurred first. PFS was estimated using Kaplan-Meier method and presented with 2-sided 95% confidence interval. (NCT01126736)
Timeframe: From the date of randomization until the earlier of the following two events: the date of PD or the date of death (Up to approximately 3 years 5 months)

Interventionweeks (Median)
Phase 2, Arm 1: 0.9 mg/m^2 Eribulin Plus 500 mg/m^2 Pemetrexed18.1
Phase 2, Arm 2: 500 mg/m^2 Pemetrexed22.0

[back to top]

Phase 1b: Number of Participants With Dose-Limiting Toxicity (DLTs)

DLT were defined as clinically significant adverse events (AE) occurring less than or equal to (<=) 21 days after treatment. Events as: Non-hematological: 1) Grade greater than or equal to (>=) 3 peripheral neuropathy; 2) Grade >=3 nausea, vomiting despite optimal antiemetic treatment; 3) Any nonhematologic toxicity of Grade >=3, with exceptions as alopecia, single laboratory values out of normal range, hypersensitivity reaction. Hematological:1) Grade 4 neutropenia lasting >7 days; 2) Febrile neutropenia as fever >=38.5 degree Celsius with absolute neutrophil count less than (<)1.0*10^9 per liter(/L); 3) Grade 3 thrombocytopenia with nontraumatic bleeding requiring platelet transfusion; 4) Grade 4 thrombocytopenia with/without nontraumatic bleeding. Other 1) Study drug related death; 2) Toxicity that dose escalation committee believed to be DLT that was not covered by above DLT criteria. (NCT01126736)
Timeframe: Cycle 1 (cycle length=21 days)

,,
InterventionParticipants (Count of Participants)
Alanine transaminase (ALT) increased (grade 3)Aspartate transaminase (AST) increased (grade 3)Febrile neutropenia (grade 4)Neutropenia (grade 4)Pneumonia (grade 4)Thrombocytopenia (grade 4)
Phase 1b, Arm 1 - Cohort 1: 0.9 mg/m^2 Eribulin Plus Pemetrexed000000
Phase 1b, Arm 1 - Cohort 2: 1.4 mg/m^2 Eribulin Plus Pemetrexed111101
Phase 1b, Arm 2 - Cohort 1: 0.7 mg/m^2 Eribulin Plus Pemetrexed100010

[back to top]

Phase 1: Document Any Antitumor Activity Per Radiological Scans and/or Tumor Markers

Overall response rate is presented. Overall response rate is defined as the percentage of participants with a best response of CR or PR as classified by the investigators according to RECIST, v1.1 criteria. CR is defined as the disappearance of all target and non-target lesions, normalization of tumor marker level of non-target lesions, and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of non-target lesions or appearance of new lesions. Overall response rate is calculated as a total number of participants with CR or PR divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT01139775)
Timeframe: Baseline through end of Phase 1

Interventionpercentage of participants (Number)
130 mg185 mg240 mg275 mg
Phase 1: Pemetrexed + Cisplatin + LY2603618066.725.00

[back to top]

Deaths

Deaths that occurred during the study are presented. A summary of serious and other non-serious adverse events regardless of causality is located in the Reported Adverse Events module. (NCT01139775)
Timeframe: Randomization through 12 months after the last participant was randomized

,,
InterventionParticipants (Count of Participants)
Total deathsDeaths while on treatmentDeath within 30 days of last dose of study drugDeaths during follow-up period
Phase 1: Pemetrexed + Cisplatin + LY26036180000
Phase 2: Pemetrexed + Cisplatin151014
Phase 2: Pemetrexed + Cisplatin + LY2603618213117

[back to top]

Phase 2: Progression-Free Survival Time

Progression-free survival (PFS) time is defined as the time from the date of randomization to the first date of documented objective progressive disease (PD) or death from any cause. For participants who were not known to have had objective PD as of the data inclusion cut-off date for a particular analysis, PFS was censored at the date of the last objective progression-free disease assessments. For participants who took any subsequent systemic anticancer therapy prior to progression, PFS was censored at the date of the last objective progression-free disease assessment prior to the start date of any subsequent systemic anticancer therapy. PFS time was summarized using Kaplan-Meier estimates. (NCT01139775)
Timeframe: Randomization up to first date of PD or death from any cause (up to 6 months after the last participant entered treatment)

Interventionmonths (Median)
Phase 2: Pemetrexed + Cisplatin + LY26036184.7
Phase 2: Pemetrexed + Cisplatin1.5

[back to top]

Phase 2: Pharmacokinetic: Cmax (LY2603618)

(NCT01139775)
Timeframe: Cycle 1/Day 2 - predose, immediately prior to the end of the LY2603618 infusion, and 2-6, 24-48, and 72-96 hours postdose

Interventionng/mL (Geometric Mean)
Phase 2: Pemetrexed + Cisplatin + LY26036184130

[back to top]

Phase 1: Pharmacokinetic: Area Under the Plasma Concentration Versus Time Curve (AUC) (LY2603618)

AUC from time zero to 24 hours (AUC[0-24]), AUC from time zero to the last time point with a measurable concentration (AUC[0-tlast]), and AUC from time zero to infinity (AUC[0-∞]) values are reported for each LY2603618 dose level on Cycle 1 /Day 2 and Cycle 2 /Day 2. The number of pharmacokinetic observations (n) used in the analysis is presented for each dose level and time point. (NCT01139775)
Timeframe: Cycle 1/Day 2 - immediately prior to end of LY2603618 infusion and 1, 3, 6, 24, 48, 72, and 144 hours postdose; Cycle 2/Day 2 - predose, immediately prior to end of LY2603618 infusion, and 1, 3, 6, 24, 48, 72, and 144 hours postdose

Interventionng*h/mL (Geometric Mean)
130 mg, Cycle 1/Day 2, AUC(0-24)130 mg, Cycle 2/Day 2, AUC(0-24)130 mg, Cycle 1/Day 2, AUC(0-tlast)130 mg, Cycle 2/Day 2, AUC(0-tlast)130 mg, Cycle 1/Day 2, AUC(0-∞)130 mg, Cycle 2/Day 2, AUC(0-∞)185 mg, Cycle 1/Day 2, AUC(0-24)185 mg, Cycle 2/Day 2, AUC(0-24)185 mg, Cycle 1/Day 2, AUC(0-tlast)185 mg, Cycle 2/Day 2, AUC(0-tlast)185 mg, Cycle 1/Day 2, AUC(0-∞)185 mg, Cycle 2/Day 2, AUC(0-∞)240 mg, Cycle 1/Day 2, AUC(0-24)240 mg, Cycle 2/Day 2, AUC(0-24)240 mg, Cycle 1/Day 2, AUC(0-tlast)240 mg, Cycle 2/Day 2, AUC(0-tlast)240 mg, Cycle 1/Day 2, AUC(0-∞)240 mg, Cycle 2/Day 2, AUC(0-∞)275 mg, Cycle 1/Day 2, AUC(0-24)275 mg, Cycle 2/Day 2, AUC(0-24)275 mg, Cycle 1/Day 2, AUC(0-tlast)275 mg, Cycle 2/Day 2, AUC(0-tlast)275 mg, Cycle 1/Day 2, AUC(0-∞)275 mg, Cycle 2/Day 2, AUC(0-∞)
Phase 1: Pemetrexed + Cisplatin + LY26036188700978010200113001020011300138001250018300148001840015700262002210032200273003230027500289002350038100308003830030900

[back to top]

Phase 2: Overall Survival

Overall survival (OS) time is defined as the time from the date of randomization to the date of death from any cause. For participants not known to have died as of the data cut-off date, OS time was censored at the last contact date the participant was known to be alive prior to the data cut-off date. OS was summarized using Kaplan-Meier estimates. (NCT01139775)
Timeframe: Randomization to the date of death from any cause through the time of study discontinuation (approximately 12 months after last participant was randomized)

Interventionmonths (Median)
Phase 2: Pemetrexed + Cisplatin + LY260361812.9
Phase 2: Pemetrexed + Cisplatin6.6

[back to top]

Phase 1: Pharmacokinetic: AUC (Pemetrexed and Cisplatin)

AUC(0-tlast) and AUC(0-∞) values are reported for pemetrexed and t-platinum from cisplatin. The number of pharmacokinetic observations (n) used in the analysis is presented for each drug. (NCT01139775)
Timeframe: Pemetrexed: Cycle 1/Day 1 - immediately prior to end of pemetrexed infusion and 1, 2, 6 and 24 hours postdose. Cisplatin: Cycle 1/Day 1 - immediately prior to end of cisplatin infusion and 0.5, 1, 2, 6, 24, 72, 96, and 168 hours postdose.

Interventionng*h/mL (Geometric Mean)
Pemetrexed, AUC(0-tlast)Pemetrexed, AUC (0-∞)T-platinum from cisplatin, AUC (0-tlast)T-platinum from cisplatin, AUC (0-∞)
Phase 1: Pemetrexed + Cisplatin + LY2603618159000160000163000269000

[back to top]

Phase 1: Pharmacokinetic: Cmax (Pemetrexed and Cisplatin)

Cmax for pemetrexed and total platinum (t-platinum) from cisplatin is reported. The number of pharmacokinetic observations (n) used in the analysis is presented for each drug. (NCT01139775)
Timeframe: Pemetrexed: Cycle 1/Day 1 - immediately prior to end of pemetrexed infusion and 1, 2, 6 and 24 hours postdose. Cisplatin: Cycle 1/Day 1 - immediately prior to end of cisplatin infusion and 0.5, 1, 2, 6, 24, 72, 96, and 168 hours postdose.

Interventionng/mL (Geometric Mean)
PemetrexedT-platinum from cisplatin
Phase 1: Pemetrexed + Cisplatin + LY2603618883003710

[back to top]

Phase 2: Clinical Benefit Rate: Percentage of Participant Who Achieved a Response of Stable Disease (SD), Partial Response (PR), or Complete Response (CR)

Clinical benefit rate is the best response CR, PR, or SD as classified by the investigators according to the RECIST, v1.1 guidelines. CR is defined as the disappearance of all target and non-target lesions, normalization of tumor marker level of non-target lesions, and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of non-target lesions or appearance of new lesions. SD is neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum diameter since treatment started. Clinical benefit rate is calculated as a total number of participants with CR, PR, or SD divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT01139775)
Timeframe: Randomization until date of disease progression or death (up to 6 months after the last participant was randomized)

Interventionpercentage of participants (Number)
Phase 2: Pemetrexed + Cisplatin + LY260361869.2
Phase 2: Pemetrexed + Cisplatin47.8

[back to top]

Phase 2: Change in Tumor Size

Change in tumor size was based on tumor measurements collected according to RECIST, v1.1 guidelines. Tumor size is the sum of the tumor measurements (longest diameters) of target lesions at each tumor evaluation. Change in tumor size was defined as the change in log tumor size from baseline evaluation to the evaluation at the end of Cycle 2. (NCT01139775)
Timeframe: Baseline, end of Cycle 2

Interventioncentimeters (Mean)
Phase 2: Pemetrexed + Cisplatin + LY2603618-0.30
Phase 2: Pemetrexed + Cisplatin-0.14

[back to top] [back to top]

Phase 1: Pharmacokinetic: Maximum Plasma Concentration (Cmax) (LY2603618)

Cmax is reported for each LY2603618 dose level on Cycle 1 /Day 2 and Cycle 2 /Day 2. The number of pharmacokinetic observations (n) used in the analysis is presented for each dose level and time point. (NCT01139775)
Timeframe: Cycle 1/Day 2 - immediately prior to end of LY2603618 infusion, and 1, 3, 6, 24, 48, 72, and 144 hours postdose; Cycle 2/Day 2 - predose, immediately prior to end of LY2603618 infusion, and 1, 3, 6, 24, 48, 72, and 144 hours postdose

Interventionng/mL (Geometric Mean)
130 mg, Cycle 1/Day 2130 mg, Cycle 2/Day 2185 mg, Cycle 1/Day 2185 mg, Cycle 2/Day 2240 mg, Cycle 1/Day 2240 mg, Cycle 2/Day 2275 mg, Cycle 1/Day 2275 mg, Cycle 2/Day 2
Phase 1: Pemetrexed + Cisplatin + LY260361818101730220021903470275041303620

[back to top]

Phase 2: Change From Baseline to Long-term Follow up in Lung Cancer Symptom Scale (LCSS)

"Health-related quality of life and participant symptoms were assessed using the LCSS (patient scale). However, improper implementation of questionnaires at the site level reduced the sponsor's ability to accurately evaluate the impacted data. Therefore, the LCSS data should be interpreted with caution.~The LCSS is a 9-item questionnaire. Six questions are symptom-specific measures for lung cancer (appetite, fatigue, cough, dyspnea, hemoptysis, and pain), and 3 summation items describe total symptomatic distress, activity status, and overall quality of life. Participant responses were measured using visual analogue scales (VAS) with 100-milliliter (mm) lines. Scores range from 0 (for best outcome) to 100 (for worst outcome). The Average Symptom Burden Index (ASBI) was calculated as the mean of 6 symptom-specific questions from the LCSS. The total LCSS score was calculated as the mean of 9 questions from the LCSS." (NCT01139775)
Timeframe: Randomization to the end of study (approximately 12 months after the last participant entered treatment)

,
Interventionunits on a scale (Mean)
Total LCSSASBI
Phase 2: Pemetrexed + Cisplatin-11.7-12.6
Phase 2: Pemetrexed + Cisplatin + LY2603618-10.7-11.6

[back to top]

Phase 2: Pharmacokinetic: AUC (LY2603618)

AUC (0-24), AUC(0-tlast), and AUC(0-∞) values are reported for LY2603618. The number of pharmacokinetic observations (n) used in the analysis is presented. (NCT01139775)
Timeframe: Cycle 1/Day 2 - predose, immediately prior to the end of the LY2603618 infusion, and 2-6, 24-48, and 72-96 hours postdose

Interventionng*h/mL (Geometric Mean)
AUC (0-24)AUC (0-tlast)AUC (0-∞)
Phase 2: Pemetrexed + Cisplatin + LY2603618314003930041100

[back to top]

Phase 2: Overall Tumor Response Rate: Percentage of Participants Who Achieved a Confirmed Best Response of Completed Response (CR) or Partial Response (PR)

Overall response rate is the best response of CR or PR as classified by the investigators according to the Response Evaluation Criteria in Solid Tumors (RECIST, v1.1) guidelines. CR is defined as the disappearance of all target and non-target lesions, normalization of tumor marker level of non-target lesions, and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeter (mm). PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of non-target lesions or appearance of new lesions. Overall response rate is calculated as a total number of participants with CR or PR divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT01139775)
Timeframe: Randomization until date of disease progression (up to 6 months after the last participant was randomized)

Interventionpercentage of participants (Number)
Phase 2: Pemetrexed + Cisplatin + LY260361843.6
Phase 2: Pemetrexed + Cisplatin21.7

[back to top]

Change in Tumor Size (CTS)

CTS was defined as the log ratio of tumor size at 6 weeks to tumor size at baseline. CTS at 6 weeks=Log (Sum of Target Lesion Measurements at 6 Weeks)-Log (Sum of Target Lesion Measurements at Baseline). (NCT01160744)
Timeframe: Baseline, 6 weeks

Interventionlog ratio (Mean)
Pem + Carb or Cis (Non-Squamous)-0.2
Ram + Pem + Carb or Cis (Non-Squamous)-0.2
Gem + Carb or Cis (Squamous)-0.3
Ram + Gem + Carb or Cis (Squamous)-0.4

[back to top]

Duration of Response (DOR)

DOR was measured from the time criteria were met for the first objectively recorded CR or PR until the first date criteria for PD were met or death. Response was defined using RECIST v 1.1 criteria. CR was defined as the disappearance of all lesions, pathological lymph node reduction in short axis to <10 mm, and normalization of tumor marker level of non-target lesions. PR was defined as ≥30% decrease SOD of target lesions taking as reference the baseline sum diameter. PD was defined as ≥20% increase in SOD of target lesions and short axes of target lymph nodes, taking as reference the smallest sum of the longest diameters recorded since treatment started and an absolute increase in sum diameter of ≥5 mm; appearance of ≥1 new lesions and/or unequivocal progression of existing non-target lesions. Participants who did not relapse were censored at the day of their last objective tumor assessment. (NCT01160744)
Timeframe: Time of first response (CR or PR) until PD or death (up to 24 months)

Interventionmonths (Median)
Pem + Carb or Cis (Non-Squamous)4.5
Ram + Pem + Carb or Cis (Non-Squamous)5.5
Gem + Carb or Cis (Squamous)4.3
Ram + Gem + Carb or Cis (Squamous)4.3

[back to top]

Overall Survival (OS)

OS was defined as the time from the date of randomization to the date of death from any cause. If the participant was alive at the end of the follow-up period or was lost to follow-up, OS was censored on the last date the participant was known to be alive. (NCT01160744)
Timeframe: Randomization to the date of death from any cause (up to 31.3 months)

Interventionmonths (Median)
Pem + Carb or Cis (Non-Squamous)10.4
Ram + Pem + Carb or Cis (Non-Squamous)13.9
Gem + Carb or Cis (Squamous)11.3
Ram + Gem + Carb or Cis (Squamous)10.4

[back to top]

Percentage of Participants With Best Overall Response of Complete Response (CR) or Partial Response (PR) [Objective Response Rate (ORR)]

Best overall response of CR or PR was defined using RECIST v 1.1 criteria. CR was defined as the disappearance of all lesions, pathological lymph node reduction in short axis to <10 mm, and normalization of tumor marker levels of non-target lesions. PR was defined as ≥30% decrease in SOD of target lesions taking as reference the baseline sum diameter. PD was defined as ≥20% increase in SOD of target lesions and short axes of target lymph nodes, taking as reference the smallest sum of the longest diameters recorded since treatment started and an absolute increase in sum diameter of ≥5 mm; appearance of ≥1 new lesions and/or unequivocal progression of existing non-target lesions. Participants who had no post baseline tumor assessments were considered non-responders and included in the denominator when calculating response rate. Percentage of participants=(number of participants with CR+PR/total number of participants)*100. (NCT01160744)
Timeframe: Day 1, Cycle 1 (3-week cycles) and every 6 weeks thereafter to PD (up to 24 months)

Interventionpercentage of participants (Number)
Pem + Carb or Cis (Non-Squamous)38.0
Ram + Pem + Carb or Cis (Non-Squamous)49.3
Gem + Carb or Cis (Squamous)24.6
Ram + Gem + Carb or Cis (Squamous)46.5

[back to top]

Percentage of Participants With CR, PR, or Stable Disease (SD) [Disease Control Rate (DCR)]

DCR: percentage of participants with CR, PR, or SD using RECIST v 1.1 criteria. CR: disappearance of all lesions, pathological lymph node reduction in short axis to <10 mm, and normalization of tumor marker levels of non-target lesions. PR: ≥30% decrease in SOD of target lesions taking as reference baseline sum diameter. PD: ≥20% increase in SOD of target lesions and short axes of target lymph nodes, taking as reference smallest sum of longest diameters recorded since treatment started and an absolute increase in sum diameter ≥5 mm; appearance of ≥1 new lesions and/or unequivocal progression of existing non-target lesions. SD: neither sufficient shrinkage to qualify for PR nor increase to qualify for PD. Participants who had no post baseline tumor assessments were considered non-responders and included in the denominator when calculating response rate. Percentage of participants=(number of participants with CR+PR+SD/total number of participants)*100. (NCT01160744)
Timeframe: Day 1, Cycle 1 (3-week cycles) and every 6 weeks thereafter to PD (up to 24 months)

Interventionpercentage of participants (Number)
Pem + Carb or Cis (Non-Squamous)70.4
Ram + Pem + Carb or Cis (Non-Squamous)85.5
Gem + Carb or Cis (Squamous)66.7
Ram + Gem + Carb or Cis (Squamous)73.2

[back to top]

Progression-Free Survival (PFS)

PFS was the time from randomization to the first objective progression as defined by Response Evaluation Criteria in Solid Tumors, Version 1.1 (RECIST v 1.1) or death from any cause, whichever occurred first. Progressive disease (PD) was defined as ≥20% increase in sum of diameter (SOD) of target lesions and short axes of target lymph nodes, taking as reference the smallest sum of the longest diameters recorded since treatment started and an absolute increase in sum diameter of ≥5 millimeters (mm); appearance of ≥1 new lesions and/or unequivocal progression of existing non-target lesions. Participants alive and without disease progression were censored at the time of the last objective tumor assessment. Participants who did not progress and were lost to follow-up were censored at their last radiographic assessment. If no baseline or post baseline radiologic assessments were available, participants were censored at date of randomization. (NCT01160744)
Timeframe: Randomization to PD or death (up to 24 months)

Interventionmonths (Median)
Pem + Carb or Cis (Non-Squamous)5.6
Ram + Pem + Carb or Cis (Non-Squamous)7.2
Gem + Carb or Cis (Squamous)5.4
Ram + Gem + Carb or Cis (Squamous)5.6

[back to top]

Number of Participants Reporting Treatment-Emergent Adverse Events (TEAEs) and Who Died

Data presented are the number of participants with at least 1 treatment-emergent adverse event (TEAE) and treatment-emergent serious adverse event (SAE), as well as, the number of participants who died during the study. TEAEs were defined as serious and other non-serious AEs that occurred or worsened after study treatment (regardless of causality). A summary of SAEs and other non-serious adverse events, regardless of causality, is located in the Reported Adverse Events module. (NCT01160744)
Timeframe: Day 1, Cycle 1 (3-week cycles) Up to 3 Years

,,,
InterventionParticipants (Count of Participants)
Treatment-Emergent SAETreatment-Emergent Adverse EventDeaths
Gem + Carb or Cis (Squamous)296355
Pem + Carb or Cis (Non-Squamous)386851
Ram + Gem + Carb or Cis (Squamous)397156
Ram + Pem + Carb or Cis (Non-Squamous)446755

[back to top]

Overall Survival (OS)

OS was defined as duration from the date of study enrollment to the date of death from any cause. Participants not known to have died as of the data inclusion cut-off date were censored at the date of last contact. The last contact for participants in post-discontinuation was the last date participant was known to be alive. (NCT01165021)
Timeframe: Enrollment until the date of death from any cause up to 64 months

Interventionmonths (Median)
Pemetrexed + Cisplatin34.6

[back to top]

Percentage of Participants With Complete Response (CR) or Partial Response (PR) [Overall Response Rate (ORR)]

Response was defined using Response Evaluation Criteria In Solid Tumors (RECIST v1.1) criteria. CR was defined as the disappearance of all target and non-target lesions and all target and non-target lymph nodes were non-pathological or normal in size [<10 millimeter (mm) short axis]. PR was defined as having at least a 30% decrease in sum of longest diameter of target lesions taking as reference the baseline sum diameters. ORR calculated as: (sum of the number of participants with PRs and CRs) divided by (number of evaluable participants) multiplied by 100. (NCT01165021)
Timeframe: From study enrollment until disease progression or recurrence up to completion of 3 cycles (21-day cycles) of chemotherapy

Interventionpercentage of participants (Number)
Pemetrexed + Cisplatin41.2

[back to top]

Percentage of Participants With No Viable Tumor Cells in Resected Lung Tissue [Pathological Complete Remission (pCR)]

pCR after the participant has undergone surgery was calculated as: (total number of participants with pCR) divided by (the total number of participants in pathological response population) multiplied by 100. (NCT01165021)
Timeframe: At the time of surgery (within 3 to 6 weeks of Day 1 of Cycle 3 [21-day cycles] of chemotherapy)

Interventionpercentage of participants (Number)
Pemetrexed + Cisplatin93.3

[back to top]

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. For participants not known to have died or did not have objective progressive disease (PD) as of the data inclusion cut-off date, PFS was censored at the date of the last objective progression-free disease assessment. PD was defined using RECIST v1.1 criteria as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (including the baseline sum if that is the smallest). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered progression. (NCT01165021)
Timeframe: Enrollment until the first date of objectively determined PD or death up to 64 months

Interventionmonths (Median)
Pemetrexed + Cisplatin12.4

[back to top]

Percentage of Participants Who Exhibit a Downward Shift in Tumor Extent From Stage IIIAN2 to Stages IIIA, II, I, or Stage 0

Tumor downstaging compared to baseline (Stage IIIAN2) were those participants who exhibited a downward shift in tumor extent from Stage IIIAN2 to Stages IIIA, II, I, or 0 were reported. Downstaging was based on radiological examination. Stage IIIAN2 was locally advanced and/or involved lymph nodes, metastasis in ipsilateral mediastinal and or subcarinal lymph nodes, tumors were ≤2 centimeters (cm) up to 5 cm in greatest dimension; Stage IIIA was locally advanced and/or involved lymph nodes, tumor extension was restricted to the affected lung; Stage II was locally advanced and/or involved lymph nodes; Stage I was small localized cancers, usually curable; Stage 0 the cancer did not spread beyond the inner lining of the lung. Missing responses were also reported. Percentage of participants calculated as: (number of participants with a downward shift in extent of their tumor) divided by (total number of evaluable participants) multiplied by 100. (NCT01165021)
Timeframe: From study enrollment until disease progression or recurrence up to completion of 3 cycles (21-day cycles) of chemotherapy

Interventionpercentage of participants (Number)
No Change or Worsening of Tumor StageChange to Stage IIIAChange to Stage IIChange to Stage IMissing
Pemetrexed + Cisplatin29.429.411.817.611.8

[back to top]

Disease Control

Disease Control is defined as complete response, partial response, or stable disease according to the response evaluation criteria in solid tumours (RECIST) version 1.1. (NCT01169675)
Timeframe: Every 6 weeks before week 48 and every 12 weeks after week 48 until progression

Interventionparticipants (Number)
Continuous Afatinib 30 mg6
Continuous Afatinib 40 mg1
Pulsed Afatinib 50 mg3
Pulsed Afatinib 60 mg6
Pulsed Afatinib 70 mg2

[back to top]

Objective Response (OR)

"Objective Response is defined as complete response or partial response according to the response evaluation criteria in solid tumours (RECIST) version 1.1.~Complete Response (CR): disappearance of all non-target lesions and normalization of tumor marker level; Partial Response (PR): at least 30% decrease of the sum of longest diameter (LD) of target lesions; Progressive Disease (PD): at least a 20% increase in the sum of LD of target lesions together with an absolute increase in the sum of LD of at least 5 millimeters; Stable Disease (SD): neither sufficient shrinkage to qualify for PR, nor sufficient increase to qualify for PD." (NCT01169675)
Timeframe: Every 6 weeks before week 48 and every 12 weeks after week 48 until progression

Interventionparticipants (Number)
Continuous Afatinib 30 mg2
Continuous Afatinib 40 mg0
Pulsed Afatinib 50 mg0
Pulsed Afatinib 60 mg2
Pulsed Afatinib 70 mg0

[back to top]

Tumour Shrinkage

Tumour shrinkage is defined as the maximum percentage decrease from baseline in the sum of the longest diameters of target lesions. (NCT01169675)
Timeframe: Every 6 weeks before week 48 and every 12 weeks after week 48 until progression

,,,,
Interventionparticipants (Number)
Patients with any decrease in tumour sizePatients having >30% decrease in tumour size
Continuous Afatinib 30 mg102
Continuous Afatinib 40 mg20
Pulsed Afatinib 50 mg30
Pulsed Afatinib 60 mg52
Pulsed Afatinib 70 mg10

[back to top]

Progression Free Survival (PFS)

PFS was defined as the time from the first treatment to the occurence of tumour progression or death, whichever came first. It was assessed according to RECIST version 1.1 criteria. (NCT01169675)
Timeframe: Every 6 weeks before week 48 and every 12 weeks after week 48 until progression

Interventionmonths (Median)
Continuous Afatinib 30 mg2.49
Continuous Afatinib 40 mg2.52
Pulsed Afatinib 50 mg2.56
Pulsed Afatinib 60 mg2.52
Pulsed Afatinib 70 mg2.69

[back to top]

Investigator Defined Dose Limiting Toxicity (DLT) During First Course of Treatment, Treated Set

Occurence of DLT during the first course of treatment to determine the maximum tolerated dose (MTD) of Afatinib at two different dose schedules in combination with the standard established dose of pemetrexed (500 mg/m2). (NCT01169675)
Timeframe: DLT were assessed during the first cycle (days 1-21)

Interventionparticipants (Number)
Continuous Afatinib 30 mg6
Continuous Afatinib 40 mg2
Pulsed Afatinib 50 mg1
Pulsed Afatinib 60 mg6
Pulsed Afatinib 70 mg4

[back to top]

Investigator Defined Dose Limiting Toxicity (DLT) During All Courses of Treatment, Treated Set

Occurence of DLT during all courses of treatment with Afatinib at two different dose schedules in combination with the standard established dose of pemetrexed (500 mg/m2). (NCT01169675)
Timeframe: DLT were assessed during all cycles of treatment

Interventionparticipants (Number)
Continuous Afatinib 30 mg11
Continuous Afatinib 40 mg2
Pulsed Afatinib 50 mg3
Pulsed Afatinib 60 mg11
Pulsed Afatinib 70 mg4

[back to top]

Percentage of Participants With a Tumor Response

Tumor response was evaluated using Response Evaluation Criteria In Solid Tumors (RECIST) criteria and confirmed by repeat assessment. Complete Response (CR) was defined as the disappearance of all target lesions and the normalization of tumor marker levels for non-target lesions; Partial Response (PR) was defined as at least a 30% decrease in the sum of the longest diameter of target lesions. Percentage of participants with a tumor response = (number of participants with CR or PR/number of enrolled participants)*100. (NCT01215916)
Timeframe: Baseline to progressive disease (up to 1 year of treatment plus 30-day follow-up)

Interventionpercentage of participants (Number)
Pemetrexed Followed by LY5736360.0
LY573636 Followed by Pemetrexed13.3
LY573636 and Pemetrexed on Day 10.0

[back to top]

Number of Participants With Clinically Significant Effects

Clinically significant effects were defined as serious and other non-serious adverse events (AEs) regardless of causality. A summary of serious and all other non-serious AEs is located in the Reported Adverse Events module. (NCT01215916)
Timeframe: Baseline to end of study (up to 1 year of treatment plus 30-day follow-up)

,,,,,,,,
InterventionParticipants (Count of Participants)
Serious AEsNon-Serious AEs
LY573636, 300 µg/mL on Day1 Plus Pemetrexed, 375 mg/m2 on Day423
LY573636, 300µg/mL Plus Pemetrexed, 375mg/m2 on Day 111
LY573636, 320 µg/mL on Day1 Plus Pemetrexed, 375 mg/m2 on Day446
LY573636, 320 µg/mL on Day1 Plus Pemetrexed, 500 mg/m2 on Day436
LY573636, 340 µg/mL Plus Pemetrexed, 500 mg/m2 on Day 145
Pemetrexed, 375 mg/m2 on Day1 Plus LY573636, 300 µg/mL on Day434
Pemetrexed, 375 mg/m2 on Day1 Plus LY573636, 320 µg/mL on Day449
Pemetrexed, 375 mg/m2 on Day1 Plus LY573636, 340 µg/mL on Day422
Pemetrexed, 500 mg/m2 on Day1 Plus LY573636, 320 µg/mL on Day433

[back to top]

Pharmacokinetics, Area Under the Curve (AUC) of LY573636

Area under the concentration-time curve above the albumin corrected threshold (AUCalb) is provided for LY573636, which has been found to be highly bound to albumin. AUCalb is a surrogate measure of exposure to unbound (free) LY573636. (NCT01215916)
Timeframe: Cycles 1 and 2 on Day 4 (prior to and at the end of LY573636 infusion, 2 and 4 hours post LY573636 infusion), Day 8 (anytime), Day 15 (anytime)

Interventionhour*micrograms per milliliter (h*µg/mL) (Geometric Mean)
Cycle 1Cycle 2
All Enrolled Participants132.78368.813

[back to top]

Pharmacokinetics, Concentration Maximum (Cmax) of LY573636

(NCT01215916)
Timeframe: Cycles 1 and 2 on Day 4 (prior to and at the end of LY573636 infusion, 2 and 4 hours post LY573636 infusion), Day 8 (anytime), Day 15 (anytime)

Interventionmicrograms per milliliter (µg/mL) (Geometric Mean)
Cycle 1Cycle 2
All Enrolled Participants264.285225.588

[back to top]

Progression-free Survival (PFS)

PFS was defined as the time from the date of randomization to the date of the first observation of investigator-assessed (radiology review) progression based on Response Evaluation Criteria In Solid Tumors (RECIST) v.1.1 or other protocol-approved measures of disease progression (e.g., new occurrence of positive fluid cytology, newly diagnosed evidence of disease progression from histologic samples, PET-positive metastases, or new bone or brain metastases), or date of death, whatever the cause. Disease progression as assessed by the investigator per RECIST v1.0 was defined as at least a 20% increase in sum of longest diameters (RECIST definition) compared to baseline (or lowest sum while on study if less than baseline), or any new lesions (measurable or nonmeasurable). (NCT01218516)
Timeframe: From date of first administration of study drug up to 6 month follow-up from randomization of the last participant, i.e., cut-off date 15 Dec 2012 for primary analysis and cut-off date of 1 Nov 2013 or up to approximately 28 months for final analysis

,
InterventionMonths (Median)
Per Primary Analysis Cut-Off DatePer Final Analysis Cut-Off Date
Combination Therapy: Farletuzumab + Chemotherapy4.74.7
Combination Therapy: Placebo + Chemotherapy5.85.9

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Treatment-Emergent Serious Adverse Events (SAEs)

An adverse event (AE) was defined as any untoward medical occurrence in a clinical investigation participant administered with an investigational product. A serious adverse event (SAE) was defined as any untoward medical occurrence that at any dose; resulted in death, was life-threatening (i.e., the participant was at a risk of death at the time of the event; this did not include an event that hypothetically might have caused death if it had been more severe), required inpatient hospitalization or prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity or substantial disruption of the ability to conduct normal life functions, or was a congenital abnormality/birth defect. In this study, TEAEs (defined as an AE that started/increased in severity on/after the first dose of study medication up to 30 days after the final dose of study medication) were assessed. (NCT01218516)
Timeframe: For each participant, from the first dose till 30 days after the last dose or cut-off date of 1 Nov 2013, i.e., up to approximately 28 months for final analysis

,,,
InterventionParticipants (Count of Participants)
TEAEsTreatment emergent SAEs
Combination Therapy: Farletuzumab + Chemotherapy6327
Combination Therapy: Placebo + Chemotherapy6027
Monotherapy: Farletuzumab257
Monotherapy: Placebo306

[back to top]

Overall Response Rate (ORR)

ORR, defined as the percentage of participants who had best overall response (BOR) of complete response (CR) or partial response (PR) as determined by investigator's radiologic assessments using RECIST 1.1 for target lesions and assessed by Magnetic resonance imaging (MRI) and computerized tomography (CT) scan (for double blind treatment period i.e. Randomization Phase). CR was defined as disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) had to have reduction in short axis to less than 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. ORR = CR + PR. (NCT01218516)
Timeframe: From Day 1 until documented radiographic progression, other protocol-approved measures of disease progression, withdrawal by participant, death due to any cause, or cut-off date of 1 Nov 2013, i.e., up to approximately 28 months for final analysis

InterventionPercentage of participants (Number)
Combination Therapy: Placebo + Chemotherapy37.3
Combination Therapy: Farletuzumab + Chemotherapy41.3

[back to top]

Overall Survival (OS)

OS was defined as the time (in months) from the date of randomization to the date of death, regardless of cause. (NCT01218516)
Timeframe: From the date of randomization to the date of death due to any cause or up to cut-off date of 1 Nov 2013 (up to approximately 28 months) for final analysis

InterventionMonths (Median)
Combination Therapy: Placebo + Chemotherapy10.5
Combination Therapy: Farletuzumab + Chemotherapy14.1

[back to top]

Duration of Response (DR)

DR was derived for those participants with objective evidence of CR or PR. DR was defined as the time (in months) from first documentation of objective response (CR or PR) to the first documentation of disease progression (ie, objective tumor progression as assessed by investigator's radiology review or other protocol-approved measures of disease progression) or death due to any cause. CR was defined as disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) had to have reduction in short axis to less than 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. (NCT01218516)
Timeframe: From the first documentation of objective response (CR or PR) to the first documentation of disease progression, death due to any cause, or cut-off date of 1 Nov 2013, i.e., up to approximately 28 months for final analysis

InterventionMonths (Median)
Combination Therapy: Placebo + Chemotherapy6.7
Combination Therapy: Farletuzumab + Chemotherapy4.1

[back to top]

Pharmacokinetics (PK): Maximum Serum Concentration (Cmax) of Cixutumumab, Cycle 1 (First Infusion) and Cycle 4 (Fourth Infusion)

(NCT01232452)
Timeframe: First Infusion: [Prior to Infusion (of Cycle1): 1, 72, 168, 336 hours(hrs) and 504 hrs (i.e. Prior to Infusion of Cycle 2)] and Fourth Infusion; [Prior to Infusion (of Cycle 4),1,24,72,120,168,240,336 hrs and 504 hrs (i.e. Prior to Infusion of Cycle 5)]

Interventionmicrogram/milliliter (ug/mL) (Geometric Mean)
First InfusionFourth Infusion
Pemetrexed + Cisplatin + Cixutumumab481556

[back to top]

Time to Worsening of Symptoms as Measured by Lung Cancer Symptom Scale (LCSS) Score

TTPS was defined as the time from the date of randomization until the date of worsening of symptoms as measured by Lung Cancer Symptom Scale (LCSS) score. Symptomatic progression was defined as an increase (worsening) of the Average Symptomatic Burden Index (ASBI) that is, the mean of the six major lung cancer specific symptom scores of the LCSS patient scale - ranging from 0 to 100 where higher score indicates worst outcome). For each participant, the maximum improvement over baseline score was calculated for each of the 9 LCSS items, ASBI and LCSS total score. Participants without event are censored at the date of the last LCSS assessment. TTPS was estimated using the Kaplan-Meier method. (NCT01232452)
Timeframe: Time to worsening of symptoms as measured by LCSS score Up to 18.3 Months

InterventionMonths (Median)
Pemetrexed + Cisplatin + Cixutumumab2.14
Pemetrexed + Cisplatin4.21

[back to top]

PK: Area Under the Concentration Time Curve (AUC[0-inf]) of Cixutumumab, Cycle 1 (i.e. First Infusion)

(NCT01232452)
Timeframe: Prior to Infusion (of Cycle 1), 1, 72, 168, 336 hrs and 504 hrs (i.e. Prior to Infusion of Cycle 2)

Interventionmicrogram*hour/milliliter (ug*hr/mL) (Geometric Mean)
Pemetrexed + Cisplatin + Cixutumumab73200

[back to top]

Percentage of Participants Achieving an Objective Response Rate (ORR)

The ORR is the percentage of all participants with Partial Response (PR) or Complete Response (CR) according to RECIST v1.1. Disease progression was defined as at least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, and/or the appearance of one or more new lesion(s), and/or unequivocal progression of existing nontarget lesions. ORR is confirmed best overall tumor response of CR and PR. CR was defined as the disappearance of all target and non-target lesions; PR defined as a >30% decrease in the sum of the longest diameters (LD) of the target lesions, taking as reference the baseline sum of the LD. (NCT01232452)
Timeframe: Randomization to Disease Progression Up to 18.3 Months

Interventionpercentage of participants (Number)
Pemetrexed + Cisplatin + Cixutumumab37.9
Pemetrexed + Cisplatin30.6

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from the date of randomization to the date of death from any cause. If the participant is alive at the end of the follow-up period or is lost to follow-up, OS will be censored on the last date the participant is known to be alive. OS was estimated using the Kaplan-Meier method. (NCT01232452)
Timeframe: Randomization Date to Death From Any Cause Up to 20 Months

InterventionMonths (Median)
Pemetrexed + Cisplatin + Cixutumumab10.68
Pemetrexed + Cisplatin10.38

[back to top]

Duration of Response (DOR)

Duration of response is measured from the time measurement criteria are first met for CR/PR (whichever is first recorded) until the first date that the criteria for PD is met, or death, is objectively documented. DOR was estimated using the Kaplan-Meier method. Disease progression was assessed via RECIST version 1.1, and defined as at least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, and/or the appearance of one or more new lesion(s), and/or unequivocal progression of existing non-target lesions (NCT01232452)
Timeframe: Time from Response to Disease Progression or Death from Any Cause Up to 20 Months

InterventionMonths (Median)
Pemetrexed + Cisplatin + Cixutumumab4.90
Pemetrexed + Cisplatin3.91

[back to top]

Change in Tumor Size (CTS)

CTS was measured by percentage change of tumor size at the end of Cycle 2 comparing to baseline tumor size. (NCT01232452)
Timeframe: Change from baseline measurement to the end of Cycle 2, average of 42 days

InterventionPercent Change (Mean)
Pemetrexed + Cisplatin + Cixutumumab-23.88
Pemetrexed + Cisplatin-16.04

[back to top]

Time to Progressive Disease (TTPS)

TTPS was defined as the time from the date of randomization until the date of disease progression. Disease progression was assessed via RECIST version 1.1, and defined as at least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, and/or the appearance of one or more new lesion(s), and/or unequivocal progression of existing non-target lesions.TTPS was estimated using the Kaplan-Meier method. (NCT01232452)
Timeframe: Randomization Date to Disease Progression Up to 18.3 Months

InterventionMonths (Median)
Pemetrexed + Cisplatin + Cixutumumab6.05
Pemetrexed + Cisplatin6.05

[back to top]

Progression-free Survival (PFS)

PFS was defined as the time from date of randomization until the date of disease progression, or death from any cause, whichever was first. Disease progression was assessed via Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1, and defined as at least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, and/or the appearance of one or more new lesion(s), and/or unequivocal progression of existing nontarget lesions. Participants without documentation for disease progression or death were censored at the date of last tumor assessment. The PFS was estimated following the Kaplan-Meier method. (NCT01232452)
Timeframe: Randomization Date to Disease Progression or Death From Any Cause Up to 18.3 Months

Interventionmonths (Median)
Pemetrexed + Cisplatin + Cixutumumab5.45
Pemetrexed + Cisplatin5.22

[back to top]

PK: Area Under the Concentration Time Curve During 1 Dosing Interval (i.e. 504 hr, AUC(0-tau) of Cixutumumab, Cycle 4 (i.e. Fourth Infusion)

(NCT01232452)
Timeframe: Prior to Infusion (of Cycle 4), 1, 24, 72, 120, 168, 240, 336 hrs and 504 hrs (i.e. Prior to Infusion of Cycle 5)

Intervention(ug*hr/mL) (Geometric Mean)
Pemetrexed + Cisplatin + Cixutumumab79700

[back to top]

Overall Response Rate

Tumor response was assessed every two cycles of completed therapy. Responses will be based on a comparison to the pretreatment tumor evaluation. 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 (NCT01263782)
Timeframe: From treatment start to every two cycles of completed therapy.

,,,
InterventionParticipants (Count of Participants)
No change/Stable DiseasePartial RemissionProgressive Disease
Carboplatin + Pemetrexed + Bevacizumab Followed by Maintenance330
Carboplatin + Pemetrexed + Cetuximab Followed by Maintenance P210
Carboplatin + Pemetrexed + Cixutumumab Followed by Maintenance320
Carboplatin + Pemetrexed x 4 Cycles Followed by Maintenance Pe501

[back to top]

Progression Free Survival

It is defined as from treatment start to the time of progression or death, whichever occurred first, or to the time of last contact. 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. (NCT01263782)
Timeframe: From treatment start to the time of progression or death, whichever occurred first, or to the time of last contact, assessed up to 5 years

Interventionmonth (Median)
Carboplatin + Pemetrexed x 4 Cycles Followed by Maintenance Pe5.2
Carboplatin + Pemetrexed + Bevacizumab Followed by Maintenance14.5
Carboplatin + Pemetrexed + Cetuximab Followed by Maintenance P20.6
Carboplatin + Pemetrexed + Cixutumumab Followed by Maintenance8

[back to top]

PK Parameter: Maximum Plasma Concentration (Cmax) of LY2090314

(NCT01287520)
Timeframe: Cycle 1 Day 1 of a 28-day cycle

Interventionnanograms per milliliter (Geometric Mean)
LY 10/Carb 5 or Carb 6/Pem 500 (Cohorts 1 and 2)122
LY 20/Carb 6/Pem 500 (Cohort 3)246
LY 40/Carb 6/Pem 500 (Cohorts 4 and 9 )603
LY 80/Carb 6/Pem 500 (Cohorts 5 and 7898
LY 120/Carb 6/Pem 500 (Cohort 6)1700
LY 60/Carb 6/Pem 500 + R50 (Cohort 8)881

[back to top]

PK Parameter: AUC0-∞ of Free Carboplatin (Carb)

AUC0-∞ of free Carb was calculated from the area under the concentration versus time curves of Carb given as a single dose with Pem (doublet therapy) and when co-administered with Pem and LY2090314 (triplet therapy). (NCT01287520)
Timeframe: Cycle 1 Day 8 of 28-day cycle and Cycle 2 up to Cycle 9: Day 1 of 21-day cycle

Interventionhours*nanograms per milliliter per mg (Geometric Mean)
Carboplatin (Doublet Therapy)81.6
Carboplatin (Triplet Therapy)88.1

[back to top]

PK Parameter: Cmax of Free Carboplatin

Cmax of free Carb given as a single dose with Pem (doublet therapy) and when co-administered with Pem and LY2090314 (triplet therapy). (NCT01287520)
Timeframe: Cycle 1, Day 8 of 28-day cycle and Cycle 2 up to Cycle 9: Day 1 of 21-day cycle

Interventionnanograms/milliliter/milligram (Geometric Mean)
Carboplatin (Doublet Therapy)24.0
Carboplatin (Triplet Therapy)25.5

[back to top]

Pharmacokinetic (PK) Parameter: Area Under the Concentration-Time Curve From Time 0 Hour to Infinity (AUC0-∞) of Pemetrexed (Pem)

AUC0-∞ was calculated from the area under the concentration versus time curves of Pem given as a single dose with Carb (doublet therapy) and when co-administered with Carb and LY2090314 (triplet therapy). (NCT01287520)
Timeframe: Cycle 1 Day 8 of 28-day cycle and Cycle 2 up to Cycle 10 Day 1 of 21-day cycle

Interventionhours*nanograms/milliliter/ milligram (Geometric Mean)
Pemetrexed (Doublet Therapy)212
Pemetrexed (Triplet Therapy)202

[back to top]

Pharmacokinetic (PK) Parameter: Area Under the Concentration-Time Curve From Time 0 Hour to Infinity (AUC0-∞) of LY2090314

AUC0-∞ was calculated from the area under the concentration versus time curve from time 0 to infinity of LY2090314 when administered alone. (NCT01287520)
Timeframe: Cycle 1 Day 1 of a 28 day cycle

Interventionnanograms*hour per milliliter (Geometric Mean)
LY 10/Carb 5 or Carb 6/Pem 500 (Cohorts 1 and 2)216
LY 20/Carb 6/Pem 500 (Cohort 3)427
LY 40/Carb 6/Pem 500 (Cohorts 4 and 9)976
LY 80/Carb 6/Pem 500 (Cohorts 5 and 7)1870
LY 120/Carb 6/Pem 500 (Cohort 6)3310
LY 60/Carb 6/Pem 500 + R50 (Cohort 8)1600

[back to top]

PK Parameter: Maximum Plasma Concentration (Cmax) of Pemetrexed (Pem)

Cmax of Pem given as a single dose with Carb (doublet therapy) and when coadministered with Carb and LY2090314 (triplet therapy). (NCT01287520)
Timeframe: Cycle 1 Day 8 of 28-day cycle and Cycle 2 up to Cycle 10 Day 1 of 21-day cycle

Interventionnanogram per milliliter per milligram (Geometric Mean)
Pemetrexed (Doublet Therapy)109
Pemetrexed (Triplet Therapy)108

[back to top]

PK Parameter: AUC0-∞ of LY2090314 Coadministered With Pemetrexed (Pem) and Carboplatin (Carb)

AUC0-∞ was calculated from the area under the concentration versus time curves of LY2090314 from time zero to infinity when coadministered with Pem and Carb. (NCT01287520)
Timeframe: Cycle 1 Day 8 of a 28-day cycle or Cycle 2 Day 1 of a 21-day cycle

Interventionnanograms*hour per milliliter (Geometric Mean)
LY 10/Carb 5 or Carb 6/Pem 500 (Cohorts 1 and 2)192
LY 20/Carb 6/Pem 500 (Cohort 3)404
LY 40/Carb 6/Pem 500 (Cohorts 4 and 9)938
LY 80/Carb 6/Pem 500 (Cohorts 5 and 7)1830
LY 120/Carb 6/Pem 500 (Cohort 6)2190
LY 60/Carb 6/Pem 500 + R50 (Cohort 8)1570

[back to top]

Number of Participants With Best Overall Tumor Response

Best overall observed tumor response at any point during the study until disease progression/recurrence defined using Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete Response (CR) was defined as the disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeter (mm) and normalization of tumor marker level of non-target lesions; Partial Response (PR) was defined as at least 30% decrease in sum of longest diameter of target lesions; Progressive Disease (PD) was defined as at least 20% increase in sum of longest diameter of target lesions and minimum 5 mm increase over nadir; Stable Disease (SD) was defined as small changes that did not meet above criteria. (NCT01287520)
Timeframe: Baseline up to Cycle 9 (Cycle 1 was 28 days, Cycles 2 to 9 were 21 days)

,,,,,,,,
Interventionparticipants (Number)
CRPRSDPDUnknown (discontinued before response assessment)
LY 10/Carb 5/Pem 500 (Cohort 1)00201
LY 10/Carb 6/Pem 500 (Cohort 2)00421
LY 120/Carb 6/Pem 500 (Cohort 6)00000
LY 20/Carb 6/Pem 500 (Cohort 3)01211
LY 40/Carb 6/Pem 500 (Cohort 4)01230
LY 40/Carb 6/Pem 500 + R50 (Cohort 9)00221
LY 60/Carb 6/Pem 500 + R50 (Cohort 8)01120
LY 80/Carb 6/Pem 500 (Cohort 5)00012
LY 80/Carb 6/Pem 500 + R50 (Cohort 7)01100

[back to top] [back to top]

PK Parameter: Maximum Plasma Concentration (Cmax) of LY2090314 Coadministered With Pemetrexed (Pem) and Carboplatin (Carb)

(NCT01287520)
Timeframe: Cycle 1 Day 8 of a 28-day cycle or Cycle 2 Day 1 of a 21-day cycle

Interventionnanograms per milliliter (Geometric Mean)
LY 10/Carb 5 or Carb 6/Pem 500 (Cohorts 1 and 2)106
LY 20/Carb 6/Pem 500 (Cohort 3)271
LY 40/Carb 6/Pem 500 (Cohorts 4 and 9)657
LY 80/Carb 6/Pem 500 (Cohorts 5 and 7)1150
LY 120/Carb 6/Pem 500 (Cohort 6)768
LY 60/Carb 6/Pem 500 + R50 (Cohort 8)1040

[back to top]

Plasma Pharmacokinetics of LY2603618: Area Under the Concentration Time Curve From Time Zero to Infinity [AUC(0-infinity)]

Plasma LY2603618 AUC(0-infinity) following a single dose on Day 1. (NCT01296568)
Timeframe: 0, 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72 and 96 hours post-dose

Interventionnanograms*hours per milliliter (ng*h/mL) (Geometric Mean)
[^14C]LY260361822300

[back to top]

Relative Abundance of LY2603618 and the Metabolites of LY2603618 in Feces

Relative abundance was expressed as the percentage of the dose of study drug administered and calculated as %=[amount of LY2603618 or its metabolites excreted/amount of radioactive dose administered]*100. (NCT01296568)
Timeframe: Day 1 through 7 days postdose

Interventionpercentage of [^14C]LY2603618 (Mean)
LY2603618 (parent)Metabolites
[^14C]LY26036185.661.4

[back to top]

Plasma Pharmacokinetics of LY2603618: Maximum Observed Drug Concentration (Cmax)

Plasma LY2603618 Cmax following a single dose on Day 1. (NCT01296568)
Timeframe: 0, 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72 and 96 hours post-dose

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
[^14C]LY26036184750

[back to top]

Plasma Pharmacokinetics of LY2603618: Area Under the Concentration Time Curve From Time Zero to Time t [AUC(0-tlast)]

Plasma LY2603618 AUC(0-tlast) where tlast is the last time point with a measurable concentration following a single dose on Day 1. (NCT01296568)
Timeframe: 0, 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72 and 96 hours post-dose

Interventionnanograms*hours per milliliter (ng*h/mL) (Geometric Mean)
[^14C]LY260361822200

[back to top]

Relative Abundance of LY2603618 and the Metabolites of LY2603618 in Urine

Relative abundance was expressed as the percentage of the dose of study drug administered and calculated as %=[amount of LY2603618 or its metabolites excreted/amount of radioactive dose administered]*100. (NCT01296568)
Timeframe: Day 1 through 7 days postdose

Interventionpercentage of [^14C]LY2603618 (Mean)
LY2603618 (parent)Metabolites
[^14C]LY26036183.33.0

[back to top]

The Number of Participants With a Tumor Response

Tumor responses were followed and measured according to Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Complete response was defined as the disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 millimeter (mm) and normalization of tumor marker level of non-target lesions. Partial response was defined as at least a 30% decrease in sum of longest diameter of target lesions. Progressive disease was defined as at least 20% increase in sum of longest diameter of target lesions and minimum 5 mm increase over nadir; Stable disease was defined as small changes that did not meet above criteria. (NCT01296568)
Timeframe: Baseline through study completion [Cycle 5 (28 days/cycle) and 21-day safety follow-up]

InterventionParticipants (Count of Participants)
Complete responsePartial responseProgressive diseaseStable disease
Entire Study Population0020

[back to top]

Plasma Pharmacokinetics of Radioactivity: Area Under the Concentration Time Curve From Time Zero to Infinity [AUC(0-infinity)]

Plasma radioactivity AUC(0-infinity) [nanogram equivalents*hours per milliliter (ng Eq*h/mL)] following a single dose on Day 1. (NCT01296568)
Timeframe: 0, 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72 and 96 hours post-dose

Interventionng Eq*h/mL (Geometric Mean)
[^14C]LY260361832500

[back to top]

Plasma Pharmacokinetics of Radioactivity: Area Under the Concentration Time Curve From Time Zero to Time t [AUC(0-tlast)]

Plasma radioactivity AUC(0-tlast) [nanogram equivalents*hours per milliliter (ng Eq*h/mL)] where tlast is the last time point with a measurable concentration following a single dose on Day 1. (NCT01296568)
Timeframe: 0, 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72 and 96 hours post-dose

Interventionng Eq*h/mL (Geometric Mean)
[^14C]LY260361827700

[back to top]

Plasma Pharmacokinetics of Radioactivity: Maximum Observed Drug Concentration (Cmax)

Plasma radioactivity Cmax [nanogram equivalents per milliliter (ng Eq/mL)] following a single dose on Day 1. (NCT01296568)
Timeframe: 0, 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 72 and 96 hours post-dose

Interventionng Eq/mL (Geometric Mean)
[^14C]LY26036185660

[back to top]

Urinary and Fecal Excretion of LY2603618 Radioactivity Over Time Expressed as a Percentage of the Total Radioactive Dose Administered

Urinary and fecal excretion samples from each participant were measured by liquid scintillation counting. The radioactive counts detected in urine and fecal samples were each divided by the theoretical radioactive count in the total radioactive dose administered and multiplied by 100% to arrive at a percentage of total radioactive dose excreted in urine and feces. (NCT01296568)
Timeframe: 0 to 6 hours, 6 to 12, 12 to 24, 24 to 48, 48 to 72 and 72 to 96 hours post-dose

Interventionpercentage of total dose (Mean)
FecesUrine
[^14C]LY260361872.211.0

[back to top]

Number of Participants With the Indicated Changes From Baseline Value in Lactate Dehydrogenase (LDH)

"Change from Baseline in the laboratory parameter LDH was assessed as decrease to low, change to normal of no change, and increase to high. Participants with missing Baseline values were assumed to have a normal Baseline value. There is no standard normal range for LDH." (NCT01313663)
Timeframe: From the time of the first dose of study treatment until 28 days following discontinuation of study treatment (up to Study Week 55)

,
Interventionparticipants (Number)
Decrease to lowChange to normal/No changeIncrease to high
Pazopanib 800 mg073
Pemetrexed 500 mg/m^2045

[back to top]

Number of Participants With Any On-therapy AE (Serious or Non-serious) Leading to Dose Reductions (DRs) or Interruptions/Delays in the Study

An AE is defined as any untoward medical occurrence in a participant or clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An SAE is any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, or results in a congenital anomaly/birth defect. In addition, all Grade 4 laboratory abnormalities and other medically important events that require medical or surgical intervention to prevent one of the outcomes listed previously are considered to be SAEs. Refer to the general Adverse AE/SAE module for a complete list of AEs/SAEs. Management of AEs may require DRs/interruptions in study treatment. If necessary, the pazopanib dose should be reduced stepwise by 200 mg at each step. DRs for pemetrexed were 50-75% of prior dose based on the toxicity leading to DR. (NCT01313663)
Timeframe: From the time the first dose of study treatment was administered until discontinuation of treatment (up to Study Week 55)

,
Interventionparticipants (Number)
Dose reductionsDose interruptions/delays
Pazopanib 800 mg23
Pemetrexed 500 mg/m^211

[back to top]

Number of Participants With Any Non-serious On-therapy Adverse Event (AE: Occurring in >=5% Participants in Any Treatment Arm) and Serious Adverse Event (SAE)

An AE is defined as any untoward medical occurrence in a participant or clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered to be related to the medicinal product. An SAE is any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, or results in a congenital anomaly/birth defect. In addition, all Grade 4 laboratory abnormalities and other medically important events that require medical or surgical intervention to prevent one of the outcomes listed previously are considered to be SAEs. Refer to the general Adverse AE/SAE module for a complete list of AEs and SAEs. (NCT01313663)
Timeframe: From the time the first dose of study treatment was administered until 28 days following discontinuation of study treatment regardless of initiation of a new cancer therapy or transfer to hospice (up to Study Week 55)

,
Interventionparticipants (Number)
Non-serious AEsSAEs
Pazopanib 800 mg92
Pemetrexed 500 mg/m^292

[back to top]

Number of Participants With a Change From Baseline Grade to Grade 3 and 4 for the Indicated Clinical Laboratory Parameters

Hematology and clinical chemistry data were summarized according to National Cancer Institutes (NCI) CTCAE grade, version 4.0. Grade 1, Mild; Grade 2, Moderate; Grade 3, Severe; Grade 4, Life-threatening or disabling; Grade 5, Death. Data are presented for only those parameters for which an increase to Grade 3 or Grade 4 occurred. Participants with missing Baseline grades were assumed to have a Baseline grade of 0. Lymphocyte count increased: Grade 3, <500 - 200/millimeters cubed (mm^3); <0.5 - 0.2x 10e9/Liters (L); Grade 4, <200/mm^3; <0.2x 10e9/L. Lymphocyte count decreased: Grade 3, >20000/mm^3; Grade 4, NA. Hyperglycemia; Grade 3, >250 - 500 milligrams per deciliter (mg/dL); >13.9 - 27.8 millimoles per Liter (mmol/L); hospitalization indicated; Grade 4, >500 mg/dL; >27.8 mmol/L; life-threatening consequences. Hypophosphatemia (inorganic phosphorus): Grade 3, <2.0 - 1.0 mg/dL, <0.6 - 0.3 mmol/L; Grade 4, <1.0 mg/dL, <0.3 mmol/L, life-threatening consequences. (NCT01313663)
Timeframe: From the time of the first dose of study treatment until 28 days following discontinuation of study treatment (up to Study Week 55)

,
Interventionparticipants (Number)
Lymphocytes, Grade 3Hyperglycemia, Grade 3Phosphorus inorganic, Grade 3
Pazopanib 800 mg111
Pemetrexed 500 mg/m^2200

[back to top]

Number of Participants (Par.) With the Indicated Best Overall Response

A par. was defined as a responder if s/he sustained a CR (The disappearance of all target lesions. Any pathological lymph node must be less than 10 millimeters [mm] in the short axis) or PR (At least a 30% decrease in the sum of the diameters of target lesions, taking as a reference, the baseline sum of the diameters) that was confirmed after >=28 days. Response was evaluated by an investigator per RECIST, version 1.1. A par. without a post-Baseline assessment was considered a non-responder. Stable disease (SD) is defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease (PD; at least a 20% increase in the sum of the diameters of target lesions, taking as a reference, the smallest sum of diameters recorded since the treatment started). To qualify as a best response of SD, a response of SD had to be observed >=12 weeks after randomization. A par. who was not evaluable had no scans at all or did not have a confirmatory scan. (NCT01313663)
Timeframe: From randomization until the time of the first documented evidence of a confirmed complete response (CR) or partial response (PR) (average of 10 weeks)

,
Interventionparticipants (Number)
CRPRSDPDNot evaluable
Pazopanib 800 mg00245
Pemetrexed 500 mg/m^200243

[back to top]

Time on Study Treatment (Pazopanib), as a Measure of Extent of Exposure

"Time on study treatment, as a measure of extent of exposure, was assessed in all participants who received pazopanib. Time on study treatment was not measured in participants receiving pemetrexed. For these participants, extent of exposure was measured as the mean number of dosing cycles and dose intensity. See the outcome measures entitled Mean number of dosing cycles, as a measure of extent of exposure and Average dose of pemetrexed for all cycles, as a measure of extent of exposure, respectively, for pemetrexed data." (NCT01313663)
Timeframe: From the first day to the last day of treatment (average of 8 weeks)

Interventionmonths (Mean)
Pazopanib 800 mg1.8

[back to top]

Number of Participants With Any AE (Serious or Non-serious) Leading to Withdrawal From Study Treatment

An AE is defined as any untoward medical occurrence in a subject or clinical investigation subject, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An SAE is any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, or results in a congenital anomaly/birth defect. In addition, all Grade 4 laboratory abnormalities and other medically important events that require medical or surgical intervention to prevent one of the outcomes listed previously are considered to be SAEs. Refer to the general Adverse AE/SAE module for a complete list of AEs and SAEs. A participant cold have been withdrawn fom study treatment due to an SAE or AE. (NCT01313663)
Timeframe: From the time the first dose of study treatment was administered until withdrawal from study treatment (up to Study Week 55)

Interventionparticipants (Number)
Pemetrexed 500 mg/m^22
Pazopanib 800 mg2

[back to top]

Average Dose of Pemetrexed for All Cycles, as a Measure of Extent of Exposure

"The average dose of pemetrexed for all cycles, as a measure of extent of exposure, was assessed in all participants who received pemetrexed. The average dose was not measured in participants receiving pazopanib. For these participants, extent of exposure was measured as the time on study treatment and mean daily dose. See the outcome measures entitled Time on study treatment (pazopanib), as a measure of extent of exposure and Mean daily dose, as a measure of extent of exposure, respectively, for pazopanib data." (NCT01313663)
Timeframe: From the time the first dose of study treatment was administered until discontinuation of the study or death (average of 16 weeks)

Interventionmilligrams per meters squared (m^2) (Mean)
Pemetrexed 500 mg/m^2499.54

[back to top]

Mean Daily Dose, as a Measure of Extent of Exposure

"Mean daily dose, as a measure of extent of exposure, was assessed in all participants who received pazopanib. Mean daily dose was not measured in participants receiving pemetrexed. For these participants, extent of exposure was measured as the mean number of dosing cycles and dose intensity. See the outcome measures entitled Mean number of dosing cycles, as a measure of extent of exposure and Average dose of pemetrexed for all cycles, as a measure of extent of exposure, respectively, for pemetrexed data." (NCT01313663)
Timeframe: From the first day to the last day of treatment (average of 8 weeks)

Interventionmilligrams (Mean)
Pazopanib 800 mg751.3

[back to top]

Number of Participants With the Indicated Worst-case Change From Baseline in Blood Pressure

Systolic and diastolic blood pressure (BP) were measured. Categories correspond to the following Common Terminology Criteria for Adverse Events (CTCAE) grades: normal, <120/80 millimeters of mercury (mmHg); prehypertension, 120-139/80-89 mmHg, warranting intervention in participants with high risk; stage I hypertension, 140-159/90-99 mmHg, warranting intervention; and stage II hypertension >/=160/100, warranting immediate attentive intervention to prevent acute symptoms. An increase is defined as an increase in CTCAE grade relative to Baseline grade. Participants with a missing Baseline value were assumed to have a Baseline value of <120 for systolic BP (SBP) and <80 for diastolic BP (DBP). (NCT01313663)
Timeframe: From the time of the first dose of study treatment until 28 days following discontinuation of study treatment (up to Study Week 55)

,
Interventionparticipants (Number)
SBP, increase to 120-139 mmHgSBP, increase to 140-159 mmHgSBP, increase to >=160 mmHgDBP, increase to 80-89 mmHgDBP, increase to 90-99 mmHgDBP, increase to >=100 mmHg
Pazopanib 800 mg134350
Pemetrexed 500 mg/m^2231210

[back to top]

Progression Free Survival (PFS)

PFS is defined as the interval between the date of randomization and the first documented sign of investigator-assessed (per Response Evaluation Criteria in Solid Tumors [RECIST] version 1.1) disease progression (PD) or death, whichever occurs first. The date of documented PD is the date of lesion evaluation in the case of radiological PD and the date of symptomatic cancer progression in the case of symptomatic progression (radiological confirmation is required). PD is defined as at least a 20% increase in the sum of the diameters of target lesions, taking as a reference, the smallest sum of diameters recorded since the treatment started. If the participant received subsequent anti-cancer therapy prior to the date of documented progression or death, PFS was to be censored at the last adequate assessment (LAA) prior to the initiation of therapy. Otherwise, if the participant did not have a documented date of progression or death, PFS was to be censored at the date of the LAA. (NCT01313663)
Timeframe: From randomization until the first documented sign of investigator-assessed disease progression or death, whichever occurred first (average of 10 study weeks)

Interventionweeks (Median)
Pemetrexed 500 mg/m^210.3
Pazopanib 800 mg12.7

[back to top]

Mean Number of Pemetrexed Dosing Cycles, as a Measure of Extent of Exposure

"Duration of therapy/time on study treatment, measured as the mean number of pemetrexed dosing cycles as a measure of extent of exposure, was assessed in all participants who received pemetrexed. The mean number of dosing cycles was not measured in participants receiving pazopanib. For these participants, extent of exposure was measured as the time on study treatment and mean daily dose. See the outcome measures entitled Time on study treatment (pazopanib), as a measure of extent of exposure and Mean daily dose, as a measure of extent of exposure, respectively, for pazopanib data." (NCT01313663)
Timeframe: From the time the first dose of study treatment was administered until discontinuation of the study or death (average of 16 weeks)

Interventionnumber of cycles (Mean)
Pemetrexed 500 mg/m^25.2

[back to top]

Number of Participants With a Increase From Baseline in Bazett's QTc at the Indicated Time Points

The QT interval is a measure of the time between the start of the Q wave and the end of the T wave in the heart's electrical cycle. In clinical studies with pazopanib, events of QT prolongation have occurred. (NCT01313663)
Timeframe: Baseline; Week 6; Week 15; every 9 weeks in the first 6 months; every 12 weeks in the next 6 months; and, after 1 year, every 6 months (up to Study Week 55)

Interventionparticipants (Number)
Pemetrexed 500 mg/m^20
Pazopanib 800 mg0

[back to top]

Number of Participants With the Indicated Grade Changes From Baseline Grade in Aspartate Aminotransferase (AST), Alanine Aminotransferase (ALT), Alkaline Phosphatase (Alk. Phos.), and Total Bilirubin (TB)

The laboratory parameters AST, ALT, Alk. Phos., and TB were summarized according to NCI CTCAE grade, version 4.0. Grade 1, Mild; Grade 2, Moderate; Grade 3, Severe; Grade 4, Life-threatening or disabling; Grade 5, Death. Data are presented for any grade increase, increase to Grade 3, and increase to Grade 4. Participants with missing Baseline grades were assumed to have a Baseline grade of 0. AST/ALT: Grade 1, >upper limit of normal (ULN) - 3.0x ULN; Grade 2, >3.0 to 5.0x ULN; Grade 3, >5.0 - 20.0x ULN; Grade 4, >20.0x ULN; Grade 5, not available (NA). Alk. Phos.: Grade 1, >ULN - 2.5x ULN; Grade 2, >2.5 - 5.0x ULN; Grade 3, >5.0 - 20.0x ULN; Grade 4, >20.0x ULN; Grade 5, NA. TB: Grade 1, >ULN - >1.5x ULN; Grade 2, >1.5 - 3.0x ULN; Grade 3, >3.0 - 10.0x ULN; Grade 4, >10.0x ULN; Grade 5, NA. (NCT01313663)
Timeframe: From the time of the first dose of study treatment until 28 days following discontinuation of study treatment (up to Study Week 55)

,
Interventionparticipants (Number)
AST, any GradeAST, Grade 3AST, Grade 4ALT, any GradeALT, Grade 3ALT, Grade 4Alk. Phos., any GradeAlk. Phos., Grade 3Alk. Phos., Grade 4TB, any GradeTB, Grade 3TB, Grade 4
Pazopanib 800 mg000100100000
Pemetrexed 500 mg/m^2100100000000

[back to top]

Time to Disease Progression and Overall Survival

Progression-free survival was defined as the time from the first infusion of study treatment to the date of radiographic disease progression according to RECIST 1.0, or until two consecutive PSA rises occurred with an absolute increase of 5 ng/mL and a 50% relative increase over baseline. For patients without documented disease progression, the date of death or last follow-up without disease progression was used. (NCT01338792)
Timeframe: Baseline, after every 2 courses, and then every 6 months after off-study (RECIST) until progression; or baseline, day 1 of each course, at the final evaluation, and then every 6 months after off-study (PSA) until progression

InterventionMonths (Mean)
Median SurvivalMedian Time to ProgressionMedian Progression Free Survival
Treatment (Chemotherapy and Enzyme Inhibitor)12.05.85.4

[back to top]

Best Overall Response

For patients with measurable disease, the RECIST 1.0 criteria was used to determine response. Complete Response = disappearance of all target lesions, Partial Response = greater or equal to 30% decrease in sum of longest diameter or target lesions, Stable Disease = <30% decrease or <20% increase, Progressive Disease = greater or equal to 20% increase in longest diameter of target lesions. For patients who do not have measurable disease by RECIST, the response was based on PSA response defined by Prostate Cancer Working Group criteria (1999) as 50% reduction in PSA confirmed on a second measurement at least 4 weeks later. (NCT01338792)
Timeframe: RECIST evaluation: Baseline, after every 2 courses, and then every 6 months after off-study, up to 1 year. PSA evaluation: baseline, day 1 of each course, final evaluation, and then every 6 months after off-study, up to 1 year

InterventionParticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseInevaluable
Treatment (Chemotherapy and Enzyme Inhibitor)0142184

[back to top]

Number of Participants With Serious Adverse Events (SAEs)

Safety evaluation according to the NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT01338792)
Timeframe: Baseline, days 1 and 7 of each course, and at last evaluation, up to 1 year

InterventionParticipants (Number)
Treatment (Chemotherapy and Enzyme Inhibitor)35

[back to top]

Overall Survival

Time of enrollment to date of death. (NCT01344824)
Timeframe: 1400 days

Interventionmonths (Median)
Single Arm Trial20.3

[back to top]

Subjects Experiencing Toxicity

Toxicity will be evaluated using CTCAE criteria, version 3, all grade 3 and 4 events. (NCT01344824)
Timeframe: 90 days

Interventionparticipants (Number)
DiarrheaDyspnea (shortness of breath)Fatigue (asthenia, lethargy, malaise)HemoglobinHypertensionLeukocytes (total white blood cell count)LymphopeniaNauseaNeutrophils/granulocytes (ANC/AGC)Joint painPlateletsVomiting
Bevacizumab, Carboplatin, and Pemetrexed Disodium, With Option326662427222

[back to top]

Progression-free Survival

Documented radiographic response per Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by imaging: 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. criteria each year, until subject death (NCT01344824)
Timeframe: 1400 days

Interventionmonths (Median)
Single Arm Trial12.6

[back to top]

Overall Survival (OS)

Overall survival (OS) was defined as the time from the date of randomization at first progression of disease to the date of death, regardless of the cause of death. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

InterventionMonths (Median)
Bevacizumab + Standard of Care11.86
Standard of Care10.22

[back to top]

Percentage of Participants With Disease Control According to RECIST v1.1

The disease control rate is defined as CR or PR or stable disease (SD) assessed according to the RECIST v.1.1 criteria with baseline tumour assessment as the reference. SD was defined as neither sufficient shrinkage to qualify for a PR nor sufficient increase to qualify for PD, taking as reference the smallest sum of the longest diameter since treatment started for target lesions and the persistence of 1 or more non-target lesions. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

InterventionPercentage of Participants (Number)
Bevacizumab + Standard of Care80.2
Standard of Care77.0

[back to top]

Percentage of Participants With Objective Response According to RECIST v1.1

The objective response is defined as complete response (CR) or partial response (PR) assessed according to the RECIST v.1.1 criteria with baseline tumour assessment as the reference. CR was defined as disappearance of all target and non-target lesions and (if applicable) normalization of tumor marker levels. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. PR was defined as greater than or equal to (≥) 30 percent (%) decrease in sum of longest diameter (LD) of target lesions in reference to Baseline sum LD. Response was to be confirmed ≥4 weeks after the initial assessment of CR or PR. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

InterventionPercentage of Participants (Number)
Bevacizumab + Standard of Care8.6
Standard of Care6.3

[back to top]

Percentage of Participants Who Are Alive at Month 6, 12, and 18

Percentage of participants who were alive at Month 6, 12 and 18 were reported. (NCT01351415)
Timeframe: Month 6, 12, 18

,
InterventionPercentage of Participants (Number)
Month 6Month 12Month 18
Bevacizumab + Standard of Care0.80.50.4
Standard of Care0.70.40.3

[back to top]

Percentage of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

An 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 and laboratory or clinical tests that resulted in a change in treatment or discontinuation from study drug were reported as adverse events. A SAE was any experience that: resulted in death, was life-threatening, required in-patient hospitalization or prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity, was a congenital anomaly/birth defect or was medically significant. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

,
InterventionPercentage of Participants (Number)
AEsSAEs
Bevacizumab + Standard of Care97.551.9
Standard of Care96.137.1

[back to top]

Progression-free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1)

PFS was defined as the time from start of treatment to the first event of death or PD. Tumor response was assessed by the IRF according to RECIST v1.1. Disease progression or PD was defined as ≥20% increase in sum LD in reference to the smallest on-study sum LD, or the appearance of new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. PFS2 is defined as the time between randomization at PD1 and the date of PD2 or death, whichever occurs first. PFS3 is defined as the time between PD2 and the date of PD3 or death, whichever occurs first. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

,
InterventionMonths (Median)
PFS 2PFS 3
Bevacizumab + Standard of Care5.454.01
Standard of Care3.982.60

[back to top]

Time to Progression (TTP) According to RECIST v1.1

The time to progression was defined as the time from baseline until disease progression as determined by the RECIST v1.1. TTP2 is defined as the interval between the day of randomization at PD1 and PD2. TTP3 is defined as the interval between the day of PD2 and PD3. PD was defined as ≥20% increase in sum LD in reference to the smallest on-study sum LD, or the appearance of new lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

,
InterventionMonths (Median)
TTP2TTP3
Bevacizumab + Standard of Care5.554.07
Standard of Care4.212.73

[back to top]

Duration of Response (DoR) According to RECIST v1.1

Duration of response is defined as the time that measurement criteria are met for objective response (CR/PR) (whichever status is recorded first) until the first date of progression or death is documented. CR was defined as disappearance of all target and non-target lesions and (if applicable) normalization of tumor marker levels. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to less than < 10 mm. PR was defined as greater than or equal to ≥30 % decrease in sum of longest diameter of target lesions in reference to baseline sum longest diameter. (NCT01351415)
Timeframe: Up to data cut-off date 24 June 2016 (approximately 5 years)

InterventionMonths (Median)
Bevacizumab + Standard of Care7.46
Standard of Care6.24

[back to top]

Number of Participants With a Tumor Response

Tumor response is complete response (CR) + partial response (PR), as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST) criteria (Version 1.1). CR is the disappearance of all target and non-target lesions and PR is a ≥30% decrease in sum of longest diameter of target lesions. Number of participants with a tumor response is the total number of participants with CR or PR. (NCT01358968)
Timeframe: Baseline to study completion up to 11 cycles of 21-day cycles

InterventionParticipants (Count of Participants)
LY2603618 + Gemcitabine (Continued Access)1
LY2603618 + Pemetrexed (Continued Access)0

[back to top]

Plasma Pharmacokinetics of LY2603618: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to Infinity [AUC(0-∞)]

(NCT01358968)
Timeframe: Period 2 only: Predose, 1, 2, 4, 6, 8, 12, 24, 48, 72, 96, 120 and 144 hours post dose

Interventionnanograms*hour/milliliter (ng*h/mL) (Geometric Mean)
LY260361853700

[back to top]

Plasma Pharmacokinetics of LY2603618: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to the Last Observed Plasma Concentration of LY2603618 [AUC(0-tlast)]

(NCT01358968)
Timeframe: Period 2 only: Predose 1, 2, 4, 6, 8, 12, 24, 48, 72, 96, 120 and 144 hours post dose

Interventionnanograms*hour/milliliter (ng*h/mL) (Geometric Mean)
LY260361853000

[back to top]

Plasma Pharmacokinetics of LY2603618: the Maximum Concentration of LY2603618 in the Plasma (Cmax)

(NCT01358968)
Timeframe: Period 2 only: Predose, 1, 2, 4, 6, 8, 12, 24, 48, 72, 96, 120 and 144 hours post dose

Interventionnanograms/milliliter (ng/mL) (Geometric Mean)
LY26036185950

[back to top]

Plasma Pharmacokinetics of Desipramine: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to Infinity [AUC(0-∞)]

(NCT01358968)
Timeframe: Periods 1 and 2: Predose, 0.5, 1, 2, 4, 8, 12, 24, 48, 72, 96, 120, 144 and 168 hours post dose

Interventionnanograms*hour/milliliter (ng*h/mL) (Geometric Mean)
Period 1Period 2
Desipramine655687

[back to top]

Plasma Pharmacokinetics of Desipramine: the Area Under the Plasma Concentration Versus Time Curve From Time Zero to the Last Observed Plasma Concentration of Desipramine [AUC(0-tlast)]

(NCT01358968)
Timeframe: Periods 1 and 2: Predose, 0.5, 1, 2, 4, 8, 12, 24, 48, 72, 96, 120, 144 and 168 hours post dose

Interventionnanogram*hour/milliliter (ng*h/mL) (Geometric Mean)
Period 1Period 2
Desipramine619653

[back to top]

Plasma Pharmacokinetics of Desipramine: the Maximum Concentration of Desipramine in the Plasma (Cmax)

(NCT01358968)
Timeframe: Periods 1 and 2: Predose, 0.5, 1, 2, 4, 8, 12, 24, 48, 72, 96, 120, 144 and 168 hours post dose

Interventionnanograms/milliliter (ng/mL) (Geometric Mean)
Period 1Period 2
Desipramine21.222.8

[back to top]

Number of Participants With Adverse Events

Number of participants with adverse events as a measure of safety and tolerability (NCT01376310)
Timeframe: Until 30 days after the last dose of study treatment. Subjects may have continued to receive study treatment until disease progression, death, unacceptable toxicity or until locally commercially available. The maximum duration of exposure was 76 months.

,
InterventionParticipants (Count of Participants)
Adverse EventsTreatment-Related Adverse EventsSerious Adverse EventsTreatment-Related Serious Adverse Events
Cohort A (GSK1120212 < 24 Weeks)119101268
Cohort B (GSK1120212 >= 24 Weeks)3026134

[back to top]

ORR Among Subjects in the Crossover Period Treated With Erlotinib Plus Tivantinib

Per RECIST v1.1, CR = disappearance of all lesions and PR = at least 30% decrease in the sum of diameters of target lesions. ORR = (CR+PR)/# subjects. (NCT01395758)
Timeframe: Date of randomization to the date of death from any cause or to the date that the subject discontinues from the study, assessed up to 24 months.

InterventionParticipants (Count of Participants)
Tivantinib Plus Erlotinib Crossover Period2

[back to top]

Overall Survival (OS) Among All Eligible Subjects (ITT Population) Treated With Erlotinib Plus Tivantinib Compared to Chemotherapy.

OS is calculated from the date of randomization until death from any cause. (NCT01395758)
Timeframe: Date of randomization to the date of death from any cause, assessed up to 24 months

Interventionmonths (Median)
Tivantinib Plus Erlotinib Arm6.8
Chemotherapy Arm8.5

[back to top]

Objective Response Rate (ORR) Among All Eligible Subjects (ITT Population) Treated With Erlotinib Plus Tivantinib Compared to Chemotherapy.

Per RECIST v1.1, Complete Response (CR) = disappearance of all lesions and Partial Response (PR) = at least 30% decrease in the sum of diameters of target lesions. ORR = (CR+PR)/# subjects. (NCT01395758)
Timeframe: Date of randomization to the date of death from any cause or to the date that the subject discontinues from the study, assessed up to 24 months

InterventionParticipants (Count of Participants)
Tivantinib Plus Erlotinib Arm0
Chemotherapy Arm4

[back to top]

Progression-free Survival (PFS) Among Subjects With KRAS Mutation Positive NSCLC (ITT Population) Treated With Erlotinib Plus Tivantinib Compared to Single Agent Chemotherapy.

Progression is defined using Response Evaluation Criteria in Solid Tumors (RECIST v 1.1) criteria 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 also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions or progression of existing non-target lesions are also considered progression. (NCT01395758)
Timeframe: Date of randomization until disease progression per RECIST (v 1.1) or death from any cause, whichever came first, assessed up to 24 months.

Interventionweeks (Median)
Tivantinib Plus Erlotinib Arm7.3
Chemotherapy Arm18.6

[back to top]

PERCIST Partial Metabolic Response

"The primary endpoint was partial metabolic response after 2 cycles of switch therapy as assessed by PERCIST (SUVmax decrease ≥30% using the pre-switch scan as new baseline)." (NCT01443078)
Timeframe: 2 years

Interventionparticipants (Number)
All Patients6

[back to top]

Pathologic Response Rate

The percentage of patients with a major pathologic response (NCT01443078)
Timeframe: 2 years

Interventionpercentage of patients (Number)
All Patients17

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: MM-121 Doses

"Using a 3+3 dose escalation model, the maximum tolerated dose of each therapy combination was determined by assessing dose-limiting toxicities in each cohort. If 3 patients were treated and passed the observation window, escalation to the next cohort was initiated. If a DLT was reported, 3-4 additional patients were enrolled and observed. If a DLT was observed in expanded cohort, this dose was considered to be the maximum tolerated dose. The maximum tolerated dose was defined at the cohort in which two dose-limiting toxicities were observed, or as the highest target dose tested in the absence of DLTs. The determined MTD was considered the Recommended Phase 2 Dose.~Dose Levels (3 week cycles) MM-121 doses tested: 20 mg/kg IV one-time loading dose then 12 mg/kg IV QW (20/12 mg/kg); 40/20 mg/kg Gemcitabine doses tested: 1000 mg/m2 Day 1 and 8 Pemetrexed doses tested: 500 mg/m2 Day 1 Carboplatin doses tested: 5 or 6 AUC Day 1 Cabazitaxel doses tested: 20 or 25 mg/m2 Day 1 of 3" (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

,,,
Interventionmg/kg (Number)
one-time loading dosemaintenance dose
MM-121 Plus Cabazitaxel4020
MM-121 Plus Carboplatin4020
MM-121 Plus Gemcitabine4020
MM-121 Plus Pemetrexed4020

[back to top]

Immunogenicity

Samples were collected to determine the presence of an immunologic reaction to MM-121 (i.e. human anti-human antibodies). (NCT01447225)
Timeframe: Samples were collected for all patients pre-dose on all cycles for duration of treatment, the longest of which was 88.1 weeks, and a collection was made post-infusion in any case of infusion reaction

Intervention (Number)
MM-121 Plus Gemcitabine: Cohort 1NA
MM-121 Plus Gemcitabine: Cohort 2NA
MM-121 Plus Carboplatin: Cohort 1NA
MM-121 Plus Carboplatin: Cohort 2NA
MM-121 Plus Carboplatin: Cohort 3NA
MM-121 Plus Pemetrexed: Cohort 1NA
MM-121 Plus Pemetrexed: Cohort 2NA
MM-121 Plus Cabazitaxel: Cohort 1NA
MM-121 Plus Cabazitaxel: Cohort 2NA
MM-121 Plus Cabazitaxel: Cohort 3NA

[back to top]

Objective Response Rate

To determine the number of patients reporting an objective response using RECIST v 1.1 where a Partial Response (PR) is defined as >20% decrease in tumor burden from baseline and a Complete Response (CR) is defined as complete disappearance from tumor burden from baseline. Objective Response is presented as the total # patients with PR or CR. (NCT01447225)
Timeframe: patients were assessed for response during their time on study, the longest of which was 88.1 weeks

Interventionparticipants with objective response (Number)
MM-121 Plus Gemcitabine: Cohort 10
MM-121 Plus Gemcitabine: Cohort 22
MM-121 Plus Carboplatin: Cohort 10
MM-121 Plus Carboplatin: Cohort 20
MM-121 Plus Carboplatin: Cohort 30
MM-121 Plus Pemetrexed: Cohort 10
MM-121 Plus Pemetrexed: Cohort 21
MM-121 Plus Cabazitaxel: Cohort 11
MM-121 Plus Cabazitaxel: Cohort 22
MM-121 Plus Cabazitaxel: Cohort 31

[back to top]

Pharmacokinetics

Pharmacokinetic (PK) evaluation was performed on plasma samples obtained weekly for the first cycle of the study and then on day 1 of each additional cycle to assess pre-treatment trough concentrations of MM-121. Non-compartmental analysis (NCA) was performed to calculate standard PK parameters, including the maximum observed concentration (Cmax). Serum levels of MM-121 were measured at a central lab using an enzyme-linked immunosorbent assay (ELISA). Data is presented per dose level of MM-121 (12 mg/kg, 20 mg/kg, or 40/20 mg/kg). (NCT01447225)
Timeframe: Collections taken at Cycle 1, Week 1 for all patients at start of the infusion (pretreatment), at the end of the infusion, and at 2, 4, 24 and 48 hours after the start of the MM-121 infusion

Interventionug/mL (Geometric Mean)
MM-121 + Gemcitabine: 20/12 mg/kg560
MM-121 + Carboplatin: 20/12 mg/kg554.8
MM-121 + Pemetrexed: 20/12 mg/kg900.7
MM-121 + Cisplatin: 20/12 mg/kg677.1
MM-121 + Gemcitabine: 40/20 mg/kg1033.4
MM-121 + Carboplatin: 40/20 mg/kg1107.2
MM-121 + Pemetrexed: 40/20 mg/kg1100.8
MM-121 + Cisplatin: 40/20 mg/kg1087.9

[back to top]

Pharmacokinetics (AUClast)

Pharmacokinetic (PK) evaluation was performed on plasma samples obtained weekly for the first cycle of the study and then on day 1 of each additional cycle to assess pre-treatment trough concentrations of MM-121. Non-compartmental analysis (NCA) was performed to calculate standard PK parameters, including the AUClast. Serum levels of MM-121 were measured at a central lab using an enzyme-linked immunosorbent assay (ELISA). Data is presented per dose level of MM-121 (12 mg/kg, 20 mg/kg, or 40/20 mg/kg) and per study part (Part 1 or Part 2). (NCT01447225)
Timeframe: Collections taken at Cycle 1, Week 1 for all patients at the start of the infusion (pretreatment), at the end of the infusion, and at 2, 4, 24 and 48 hours after the start of the MM-121 infusion

Interventionhr* ug/mL (Geometric Mean)
MM-121 + Gemcitabine: 20/12 mg/kg39666.4
MM-121 + Carboplatin: 20/12 mg/kg49749.6
MM-121 + Pemetrexed: 20/12 mg/kg59984.1
MM-121 + Cisplatin: 20/12 mg/kg51995.1
MM-121 + Gemcitabine: 40/20 mg/kg72132.1
MM-121 + Carboplatin: 40/20 mg/kg100309.9
MM-121 + Pemetrexed: 40/20 mg/kg92732.9
MM-121 + Cisplatin: 40/20 mg/kg98142.6

[back to top]

To Characterize Dose-limiting Toxicities (DLTs) Associated With the Combination of MM-121 With Anticancer Therapies

To establish the safety of escalating doses of MM-121 administered in combination with multiple anti-cancer therapies in order to determine the recommended phase 2 dose. Dose-escalation conducted using standard 3+3 model to determine maximum tolerated dose. Reports of Dose-Limiting Toxicities (DLTs) were assessed to determine the MTD to be used for the expansion cohort. DLTs were not measured in the Expansion Cohort. (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

Interventionparticipants reporting DLTs (Number)
MM-121 Plus Gemcitabine: Cohort 10
MM-121 Plus Gemcitabine: Cohort 21
MM-121 Plus Carboplatin: Cohort 11
MM-121 Plus Carboplatin: Cohort 20
MM-121 Plus Carboplatin: Cohort 30
MM-121 Plus Pemetrexed: Cohort 10
MM-121 Plus Pemetrexed: Cohort 21
MM-121 Plus Cabazitaxel: Cohort 10
MM-121 Plus Cabazitaxel: Cohort 20
MM-121 Plus Cabazitaxel: Cohort 30

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Carboplatin

"Maximum Tolerated Dose reported in Target AUC, as calculated by the Calvert Formula~Using a 3+3 dose escalation model, the maximum tolerated dose of each therapy combination was determined by assessing dose-limiting toxicities in each cohort. If 3 patients were treated and passed the observation window, escalation to the next cohort was initiated. If a DLT was reported, 3-4 additional patients were enrolled and observed. If a DLT was observed in expanded cohort, this dose was considered to be the maximum tolerated dose. The maximum tolerated dose was defined at the cohort in which two dose-limiting toxicities were observed, or as the highest target dose tested in the absence of DLTs. The determined MTD was considered the Recommended Phase 2 Dose.~Dose Levels (3 week cycles) MM-121 doses tested: 20 mg/kg IV one-time loading dose then 12 mg/kg IV QW (20/12 mg/kg); 40/20 mg/kg Carboplatin doses tested: 5 or 6 AUC Day 1" (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

Interventiontarget AUC (mg*min/mL) (Number)
MM-121 + Carboplatin5

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Cabazitaxel

"Using a 3+3 dose escalation model, the maximum tolerated dose of each therapy combination was determined by assessing dose-limiting toxicities in each cohort. If 3 patients were treated and passed the observation window, escalation to the next cohort was initiated. If a DLT was reported, 3-4 additional patients were enrolled and observed. If a DLT was observed in expanded cohort, this dose was considered to be the maximum tolerated dose. The maximum tolerated dose was defined at the cohort in which two dose-limiting toxicities were observed, or as the highest target dose tested in the absence of DLTs. The determined MTD was considered the Recommended Phase 2 Dose.~Dose Levels (3 week cycles) MM-121 doses tested: 20 mg/kg IV one-time loading dose then 12 mg/kg IV QW (20/12 mg/kg); 40/20 mg/kg Cabazitaxel doses tested: 20 or 25 mg/m2 Day 1 of 3" (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

Interventionmg/m2 (Number)
MM-121 + Cabazitaxel25

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Gemcitabine

"Using a 3+3 dose escalation model, the maximum tolerated dose of each therapy combination was determined by assessing dose-limiting toxicities in each cohort. If 3 patients were treated and passed the observation window, escalation to the next cohort was initiated. If a DLT was reported, 3-4 additional patients were enrolled and observed. If a DLT was observed in expanded cohort, this dose was considered to be the maximum tolerated dose. The maximum tolerated dose was defined at the cohort in which two dose-limiting toxicities were observed, or as the highest target dose tested in the absence of DLTs. The determined MTD was considered the Recommended Phase 2 Dose.~Dose Levels (3 week cycles) MM-121 doses tested: 20 mg/kg IV one-time loading dose then 12 mg/kg IV QW (20/12 mg/kg); 40/20 mg/kg Gemcitabine doses tested: 1000 mg/m2 Day 1 and 8" (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

Interventionmg/m2 (Number)
MM-121 + Gemcitabine1000

[back to top]

To Determine the Maximum Tolerated Dose (MTD) of MM-121 in Combination With Anticancer Therapies: Pemetrexed

"Using a 3+3 dose escalation model, the maximum tolerated dose of each therapy combination was determined by assessing dose-limiting toxicities in each cohort. If 3 patients were treated and passed the observation window, escalation to the next cohort was initiated. If a DLT was reported, 3-4 additional patients were enrolled and observed. If a DLT was observed in expanded cohort, this dose was considered to be the maximum tolerated dose. The maximum tolerated dose was defined at the cohort in which two dose-limiting toxicities were observed, or as the highest target dose tested in the absence of DLTs. The determined MTD was considered the Recommended Phase 2 Dose.~Dose Levels (3 week cycles) MM-121 doses tested: 20 mg/kg IV one-time loading dose then 12 mg/kg IV QW (20/12 mg/kg); 40/20 mg/kg Pemetrexed doses tested: 500 mg/m2 Day 1" (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

Interventionmg/m2 (Number)
MM-121 + Pemetrexed500

[back to top]

To Evaluate the Safety and Tolerability of Escalating Doses of the MM-121 Anticancer Therapies

Safety and tolerability data presented in detail in the adverse events and serious adverse events section of the results posting (NCT01447225)
Timeframe: From date of first dose to 30 days after termination, the longest 88.1 weeks

Interventionparticipants reporting adverse events (Number)
MM-121 Plus Gemcitabine: Cohort 13
MM-121 Plus Gemcitabine: Cohort 28
MM-121 Plus Carboplatin: Cohort 15
MM-121 Plus Carboplatin: Cohort 23
MM-121 Plus Carboplatin: Cohort 33
MM-121 Plus Pemetrexed: Cohort 13
MM-121 Plus Pemetrexed: Cohort 27
MM-121 Plus Cabazitaxel: Cohort 14
MM-121 Plus Cabazitaxel: Cohort 23
MM-121 Plus Cabazitaxel: Cohort 34

[back to top]

Number of Participants With Abnormalities in Selected Hepatic Clinical Laboratory Tests

"The number of subjects with selected hepatic laboratory abnormalities is reported.~AST= aspartate aminotransferase; ALT= alanine aminotransferase; ULN= upper limit of normal." (NCT01454102)
Timeframe: From first dose to 30 days following last dose of study drug (assessed up to July 2016, approximately 55 months)

,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
ALT OR AST > 3XULNALT OR AST > 5XULNALT OR AST > 10XULNALT OR AST > 20XULNTOTAL BILIRUBIN > 2XULNAST or ALT>3XULN with Bilirubin>2XULN within 1 dayAST or ALT>3XULN with Bilirubin>2XULN within 30day
Arm A: Nivolumab + Gemcitabine + Cisplatin0000000
Arm B: Nivolumab + Pemetrexed + Cisplatin0000000
Arm C10: Nivolumab + Paclitaxel + Carboplatin2000000
Arm C5: Nivolumab + Paclitaxel + Carboplatin2000000
Arm D: Nivolumab + Bevacizumab Maintenance1000000
Arm E: Nivolumab + Erlotinib4210111
Arm F: Nivolumab2200111
Arm GH: Nivolumab + Ipilimumab6421000
Arm IJ: Nivolumab + Ipilimumab0000000
Arm K: Nivolumab in Squamous Histology Subjects (NSCLC)0000000
Arm L: Nivolumab in Non-squamous Histology Subjects (NSCLC)0000000
Arm M: Nivolumab0000000
Arm N: Nivolumab + Ipilimumab1111000
Arm O: Nivolumab + Ipilimumab2100000
Arm P: Nivolumab + Ipilimumab1000000
Arm Q: Nivolumab + Ipilimumab0000000

[back to top]

Number of Participants With Abnormalities in Selected Thyroid Clinical Laboratory Tests

"The number of subjects with selected thyroid laboratory abnormalities is reported. FT3 and FT4 test abnormalities were considered for a 2-week window after the abnormal TSH test date.~TSH= thyroid-stimulating hormone; FT3= Free T3; FT4= Free T4; LLN= lower limit of normal; ULN= upper limit of normal" (NCT01454102)
Timeframe: From first dose to 30 days following last dose of study drug (assessed up to July 2016, approximately 55 months)

,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
TSH > ULNTSH > ULN WITH TSH <= ULN AT BASELINETSH > ULN WITH >=1 FT3/FT4 TEST VALUE < LLNTSH > ULN WITH ALL FT3/FT4 TEST VALUES >= LLNTSH > ULN WITH FT3/FT4 TEST MISSINGTSH < LLNTSH < LLN WITH TSH >= LLN AT BASELINETSH < LLN WITH >=1 FT3/FT4 TEST VALUE > ULNTSH < LLN WITH ALL FT3/FT4 TEST VALUES <= ULNTSH < LLN WITH FT3/FT4 TEST MISSING
Arm A: Nivolumab + Gemcitabine + Cisplatin2100221101
Arm B: Nivolumab + Pemetrexed + Cisplatin3212073133
Arm C10: Nivolumab + Paclitaxel + Carboplatin1101051014
Arm C5: Nivolumab + Paclitaxel + Carboplatin5420375205
Arm D: Nivolumab + Bevacizumab Maintenance8410721011
Arm E: Nivolumab + Erlotinib12870577403
Arm F: Nivolumab2113341499414
Arm GH: Nivolumab + Ipilimumab10105141212633
Arm IJ: Nivolumab + Ipilimumab7652099513
Arm K: Nivolumab in Squamous Histology Subjects (NSCLC)5532033210
Arm L: Nivolumab in Non-squamous Histology Subjects (NSCLC)5302310100
Arm M: Nivolumab2100264105
Arm N: Nivolumab + Ipilimumab10761399621
Arm O: Nivolumab + Ipilimumab129714109721
Arm P: Nivolumab + Ipilimumab11550666114
Arm Q: Nivolumab + Ipilimumab11842599117

[back to top]

Objective Response Rate (ORR)

ORR was defined as the percentage of all treated participants who achieved a Best Overall Response (BOR) of Complete Response (CR) or Partial Response (PR) using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 criteria as per investigator assessment. This proportion was multiplied by 100 and expressed as a percentage. BOR was defined as the best response designation recorded between the date of randomization and the date of progression, or the date of subsequent anticancer therapy, whichever occurred first. CR or PR determinations included in the BOR assessment were confirmed by a second scan at least 4 weeks after the criteria for responses were first met. For participants without progression or subsequent therapy, all available response designations contributed to the BOR determination. For participants who continued treatment beyond progression, the BOR was determined based on response designations recorded up to the time of the initial progression. (NCT01454102)
Timeframe: From first dose until date of progression or subsequent anti-cancer therapy (assessed up to July 2016, approximately 55 months)

InterventionPercentage of participants (Number)
Arm A: Nivolumab + Gemcitabine + Cisplatin41.7
Arm B: Nivolumab + Pemetrexed + Cisplatin46.7
Arm C10: Nivolumab + Paclitaxel + Carboplatin46.7
Arm D: Nivolumab + Bevacizumab Maintenance16.7
Arm E: Nivolumab + Erlotinib19.0
Arm F: Nivolumab23.1
Arm GH: Nivolumab + Ipilimumab20.8
Arm IJ: Nivolumab + Ipilimumab24.0
Arm K: Nivolumab in Squamous Histology Subjects (NSCLC)0
Arm L: Nivolumab in Non-squamous Histology Subjects (NSCLC)15.4
Arm M: Nivolumab8.3
Arm N: Nivolumab + Ipilimumab22.6
Arm O: Nivolumab + Ipilimumab32.5
Arm P: Nivolumab + Ipilimumab47.4
Arm Q: Nivolumab + Ipilimumab38.5
Arm C5: Nivolumab + Paclitaxel + Carboplatin50.0

[back to top]

Progression-Free Survival Rate (PFSR) at Week 24

"Progression-Free Survival (PFS) was defined as the time from the date of first dose of study medication to the date of first disease progression or death, if death occurred within 100 days of the final dose of study drug. Among participants without previous RECIST-defined progression, participants who died beyond 100 days and those who remained alive were censored at the last tumor assessment date (before subsequent therapy).~PFSR at week 24 was defined as the proportion of subjects remaining progression free and surviving at 24 weeks. The proportion was calculated by the product-limit method (Kaplan-Meier estimate), which takes into account censored data, and expressed as a percentage." (NCT01454102)
Timeframe: 24 weeks

InterventionPercentage of participants (Number)
Arm A: Nivolumab + Gemcitabine + Cisplatin50.5
Arm B: Nivolumab + Pemetrexed + Cisplatin68.4
Arm C10: Nivolumab + Paclitaxel + Carboplatin34.3
Arm D: Nivolumab + Bevacizumab Maintenance58.3
Arm E: Nivolumab + Erlotinib50.6
Arm F: Nivolumab39.7
Arm GH: Nivolumab + Ipilimumab42.8
Arm IJ: Nivolumab + Ipilimumab37.3
Arm K: Nivolumab in Squamous Histology Subjects (NSCLC)50.0
Arm L: Nivolumab in Non-squamous Histology Subjects (NSCLC)20.5
Arm M: Nivolumab8.3
Arm N: Nivolumab + Ipilimumab49.1
Arm O: Nivolumab + Ipilimumab48.0
Arm P: Nivolumab + Ipilimumab72.4
Arm Q: Nivolumab + Ipilimumab39.5
Arm C5: Nivolumab + Paclitaxel + Carboplatin59.3

[back to top]

Number of Participants Who Experienced Selected Adverse Events

"The number of participants who experienced an AE of interest due to any cause is presented. Endocrine, Gastrointestinal, Hepatic, Pulmonary, Renal, Skin, and~Hypersensitivity/Infusion select AEs were identified that are potentially associated with the use of nivolumab, based on the following 4 guiding principles:~AEs that may differ in type, frequency, or severity from AEs caused by non-immunotherapies~AEs that may require immunosuppression (eg, corticosteroids) as part of their management~AEs whose early recognition and management may mitigate severe toxicity~AEs for which multiple event terms may be used to describe a single type of AE, thereby necessitating the pooling of terms for full characterization." (NCT01454102)
Timeframe: From first dose to 30 days after the last dose of study drug (assessed up to July 2016, approximately 55 months)

,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
EndorcrineGastrointestinalHepaticPulmonaryRenalSkinHypersensitivity/Infusion Reactions
Arm A: Nivolumab + Gemcitabine + Cisplatin3402131
Arm B: Nivolumab + Pemetrexed + Cisplatin2622696
Arm C10: Nivolumab + Paclitaxel + Carboplatin0810177
Arm C5: Nivolumab + Paclitaxel + Carboplatin1601471
Arm D: Nivolumab + Bevacizumab Maintenance2202151
Arm E: Nivolumab + Erlotinib410412162
Arm F: Nivolumab816530274
Arm GH: Nivolumab + Ipilimumab815732151
Arm IJ: Nivolumab + Ipilimumab612220143
Arm K: Nivolumab in Squamous Histology Subjects (NSCLC)3521342
Arm L: Nivolumab in Non-squamous Histology Subjects (NSCLC)2102020
Arm M: Nivolumab0100020
Arm N: Nivolumab + Ipilimumab412432201
Arm O: Nivolumab + Ipilimumab15131332201
Arm P: Nivolumab + Ipilimumab811156213
Arm Q: Nivolumab + Ipilimumab1213234201

[back to top]

Number of Participants Who Experienced Serious Adverse Events (SAE), Adverse Events (AE) Leading to Discontinuation, or Death

AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. SAE=a medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency/abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. Treatment-related=having certain, probable, possible, or missing relationship to study drug. Grade (Gr) 1=Mild, Gr 2=Moderate, Gr 3=Severe, Gr 4= Potentially Life-threatening or disabling. (NCT01454102)
Timeframe: From first dose to 30 days after the last dose of study drug (assessed up to July 2016, approximately 55 months)

,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
SAEsAEs leading to discontinuationDeath
Arm A: Nivolumab + Gemcitabine + Cisplatin420
Arm B: Nivolumab + Pemetrexed + Cisplatin1050
Arm C10: Nivolumab + Paclitaxel + Carboplatin810
Arm C5: Nivolumab + Paclitaxel + Carboplatin920
Arm D: Nivolumab + Bevacizumab Maintenance330
Arm E: Nivolumab + Erlotinib1130
Arm F: Nivolumab2392
Arm GH: Nivolumab + Ipilimumab18112
Arm IJ: Nivolumab + Ipilimumab17133
Arm K: Nivolumab in Squamous Histology Subjects (NSCLC)330
Arm L: Nivolumab in Non-squamous Histology Subjects (NSCLC)441
Arm M: Nivolumab411
Arm N: Nivolumab + Ipilimumab1231
Arm O: Nivolumab + Ipilimumab2141
Arm P: Nivolumab + Ipilimumab25104
Arm Q: Nivolumab + Ipilimumab2582

[back to top]

Overall Survival (OS)

The OS was defined as the time in months from the date of randomization to the date of death, regardless of cause. In the absence of confirmation of death, the participants were censored either at the date that participant was last known to be alive or the date of study cut-off, whichever was earlier. The two treatment arms were compared using the log-rank test, stratified by histology, TPC option, and geographic region; and the treatment difference between eribulin mesylate and TPC was tested at a significance level of 0.05 (2-sided). Kaplan-Meier (K-M) survival probabilities for each arm were plotted over time. The treatment effect was estimated by fitting a Cox Proportional Hazards model to the OS times including treatment arm as a factor and histology, TPC option and geographic region as strata. (NCT01454934)
Timeframe: Randomization (Day 1) until date of death from any cause, or 37 months

Interventionmonths (Median)
Arm A: Eribulin Mesylate9.5
Arm B: Vinorelbine, Gemcitabine, Docetaxel, or Pemetrexed9.5

[back to top]

Progression Free Survival (PFS) by Response Evaluation Criteria in Solid Tumors (RECIST)

PFS was defined as the time from the date of randomization to the date of first documentation of disease progression, or date of death, whichever occurred first. The difference in PFS (based on the tumor response evaluation as determined by the investigator) between eribulin mesylate and TPC was evaluated using the log rank test, stratified by histology, TPC option, and geographic region, tested at an alpha level of 0.05 (2-sided). PFS censoring rules will be defined in the SAP and follow Federal Department of Agriculture (FDA) guidance. (NCT01454934)
Timeframe: Randomization (Day 1) until date of disease progression or death (whichever occurred first), or 37 months

Interventionmonths (Median)
Arm A: Eribulin Mesylate3.0
Arm B: Vinorelbine, Gemcitabine, Docetaxel, or Pemetrexed2.8

[back to top]

Objective Response Rate (ORR)

The ORR was defined as the proportion of participants with best overall response of complete response (CR) or partial response (PR) per RECIST criteria. The ORR was estimated by study arm based on the tumor response evaluation as determined by the investigator, according to RECIST 1.1. Participants with unknown response were treated as non-responders. The statistical difference in ORR between treatment arms was evaluated using the Cochran-Mantel-Haenszel (CMH) chi-square test with histology, TPC option, and geographic region as strata, tested at an alpha level of 0.05 (2-sided). The 95 percent confidence interval (CI) was calculated using Clopper Pearson method. (NCT01454934)
Timeframe: Randomization (Day 1) to CR or PR

Interventionpercentage of participants (Number)
Arm A: Eribulin Mesylate12.2
Arm B: Vinorelbine, Gemcitabine, Docetaxel, or Pemetrexed15.2

[back to top]

Time to Worsening of Symptom (TWS) as Per Lung Cancer Symptom Scale (LCSS)

TWS was the elapsed time from the date of randomization to the first date of worsening in any one of the 6 LCSS symptoms. The LCSS (patient and observer) were administered at baseline and at the end of each 21-day cycle, until disease progression, and at short-term follow-up. Content for the patient version -collected using a visual analog scale (VAS) anchored at 0 and 100, respectively representing the best and worst outcome- included 6 disease-specific measures: (appetite, fatigue, cough, dyspnea, hemoptysis and pain) and 3 summary consequences of lung cancer (overall symptom burden, diminished normal activities, and lowered quality-of-life).The observer version included only the 6 disease-specific measures and the responses were collected using a 5-point ordinal scale that was reverse coded with 0 and 100 respectively representing the worst and best outcome.TWS was censored at the date of the last LCSS assessment for participants who were not known to have LCSS worsening. (NCT01469000)
Timeframe: Baseline to Progressive Disease (Up To 50 Months )

,
Interventionmonths (Median)
a. Loss of Appetite (Patient-rated)b. Fatigue (Patient-rated)c. Cough (Patient-rated)d. Dyspnea (Patient-rated)e. Hemoptysis (Patient-rated)f. Pain (Patient-rated)g. Overall Symptoms (Patient-rated)h. Interference (Patient-rated)i. Quality of Life (Patient-rated)j. Loss of Appetite (Observer-rated)k. Fatigue (Observer-rated)l. Cough (Observer-rated)m. Dyspnea (Observer-rated)n. Hemoptysis (Observer-rated)o. Pain (Observer-rated)
250 mg Gefitinib7.667.66NA24.87NANA30.4624.7110.6134.1012.98NANANA10.51
250 mg Gefitinib/500 mg Pemetrexed5.398.5118.7618.76NA34.8310.125.7213.408.617.5241.92NANA34.83

[back to top]

Time To Progressive Disease (TTPD)

TTPD is defined as time from the date of randomization to the first date of disease progression. For each participant who is not known to have had a progression of disease as of the data-inclusion cut-off date for a particular analysis, or who has died without progression of disease, TTPD will be censored for that analysis at the date of the participant's last tumor assessment prior to that cut-off date. TTPD was analyzed twice: (1) excluding clinical progressions of disease (that is,those not defined according to the RECIST version 1.1 criteria ), and (2) including clinical progressions. Progressive disease (PD) was defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study. Also, the sum must also demonstrate an absolute increase of at least 5 millimeter (mm). The appearance of one or more new lesions is also considered progression. (NCT01469000)
Timeframe: Randomization to Progressive Disease (Up To 58.78 Months)

Interventionmonths (Median)
250 mg Gefitinib/500 mg Pemetrexed16.23
250 mg Gefitinib10.94

[back to top]

Overall Survival (OS)

OS is defined as the time from randomization to the date of death from any cause. Survival time is censored at the date of last contact for participants who are still alive or lost to follow up. (NCT01469000)
Timeframe: Randomization to Date of Death Due to Any Cause (Up To 67.12 Months)

InterventionMonths (Median)
250 mg Gefitinib/500 mg Pemetrexed43.43
250 mg Gefitinib36.76

[back to top]

Duration of Response (DoR)

DoR was defined as the time from the date of the first CR or PR to the first date of Progressive Disease (PD) ( RECIST 1.1 Criteria) or death from any cause.CR is the disappearance of all target lesions. Any pathological lymph nodes must have reduction in short axis to <10 mm.Tumor marker results must have normalized. PR is defined as at least a 30% decrease in the sum of diameter of target lesions, taking as reference the baseline sum diameters. PD was defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study. Also,the sum must also demonstrate an absolute increase of at least 5 mm.The appearance of one or more new lesions is also considered progression. Participants not known to have died or to have had progression of disease as of the data-inclusion cut-off date for a particular analysis,duration of tumor response was censored at the date of the participants last tumor assessment prior to that cut-off date. (NCT01469000)
Timeframe: First Observation of CR or PR to Progressive Disease or Death Due to Any Cause (Up To 57.36 Months)

InterventionMonths (Median)
250 mg Gefitinib/500 mg Pemetrexed15.44
250 mg Gefitinib11.30

[back to top]

Progression Free Survival (PFS)

PFS is defined as the time from randomization to the first date of objectively determined progressive disease or death from any cause,whichever is earlier.The censoring is taken in the following order: If a participant didn't have a complete baseline disease assessment,then the PFS time was censored at the enrollment date, regardless of whether or not objectively determined disease progression or death has been observed for the participant;otherwise,if a participant is not known to have died or have objective progression as of the data inclusion cutoff date for the analysis,the PFS time will be censored at the last complete objective progression-free disease assessment date.Progressive disease (PD) was defined as at least a 20% increase in the sum of the diameters of target lesions,taking as reference the smallest sum on study. Also,the sum must also demonstrate an absolute increase of at least 5 millimeter (mm).The appearance of one or more new lesions is also considered progression. (NCT01469000)
Timeframe: Randomization to Progressive Disease or Death Due to Any Cause (Up to 58.78 Months)

InterventionMonths (Median)
250 mg Gefitinib/500 mg Pemetrexed16.23
250 mg Gefitinib11.07

[back to top]

Percentage of Participants Achieving Complete Response (CR) or Partial Response (PR) (Overall Response Rate [ORR])

ORR is the best response of complete response (CR) or partial response (PR) as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST v1.1). Complete Response (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. Tumor marker results must have normalized. Partial Response (PR) is defined at least a 30% decrease in the sum of diameter of target lesions, taking as reference the baseline sum diameters. (NCT01469000)
Timeframe: Randomization to Progressive Disease (Up to 57.36 Months)

Interventionpercentage of participants (Number)
250 mg Gefitinib/500 mg Pemetrexed80.2
250 mg Gefitinib73.8

[back to top]

Percentage of Participants With CR, PR, and Stable Disease (SD) (Disease Control Rate [DCR])

Disease control rate is the percentage of participants with a confirmed CR, PR or SD as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST version 1.1) criteria. 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. Tumor marker results must have normalized. PR is defined at least a 30% decrease in the sum of diameter of target lesions, taking as reference the baseline sum diameters. SD is defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. (NCT01469000)
Timeframe: Randomization to Progressive Disease (Up To 57.36 Months)

Interventionpercentage of participants (Number)
250 mg Gefitinib/500 mg Pemetrexed92.9
250 mg Gefitinib93.8

[back to top]

Number of Participants Who Died Within 30 Days and 31 Days After Last Dose - All Treated Participants

Due to study termination, the categories presented below are deaths occurring within 30 days of last dose and deaths occurring within 32 days of last dose. If the study had not been terminated early, the categories presented would have been 30 days and 90 days after last dose. (NCT01471197)
Timeframe: Day 1 of Treatment to Date of Death, up to last patient, last visit, approximately 7 months after study started.

,
Interventionparticipants (Number)
Death within 30 Days of Last DoseDeath within 32 Days of Last Dose
Ipilimumab 10 mg/kg11
Pemetrexed 500 mg/m^200

[back to top]

Number of Participants With Deaths, Adverse Events (AEs), Serious AEs (SAEs) and AEs Leading to Discontinuation - All Treated Participants

AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. SAE=a medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency/abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. Treatment-related=having certain, probable, possible, or missing relationship to study drug. Grade (Gr) 1=Mild, Gr 2=Moderate, Gr 3=Severe, Gr 4= Potentially Life-threatening or disabling. Participants were evaluated from Day 1 (first day of treatment with study drug) to the date of the last participant, last visit of the study. (NCT01471197)
Timeframe: Day 1 to Date of last patient, last visit, approximately 7 months after study started.

,
Interventionparticipants (Number)
DeathsSAEsAEsAEs leading to discontinuation
Ipilimumab 10 mg/kg2561
Pemetrexed 500 mg/m^20120

[back to top]

Overall Survival of Participants During the Study - All Treated Participants

Overall survival (OS) was defined as the time from the date of randomization until the date of death. For those participants who did not die by the time the study was terminated and last patient, last visit occurred, OS was censored (+) on the last date the participant was known to be alive. OS is presented below in increasing monthly categories of survival. OS analysis was to be performed when a total of approximately 132 deaths were observed but due to the early termination of the study, statistical analyses were not performed. (NCT01471197)
Timeframe: Date of Randomization to date of death, up to last patient, last visit, approximately 7 months after study started

,
Interventionparticipants (Number)
Overall Survival 0.9 monthsOverall Survival 1.8 monthsOverall Survival 3.9 months+(censored)Overall Survival 4.4 months+(censored)Overall Survival 4.6 months+(censored)Overall Survival 5.4 months+(censored)
Ipilimumab 10 mg/kg111210
Pemetrexed 500 mg/m^2000002

[back to top]

Participant Satisfaction: Preferences Regarding Home and/or Hospital Treatment

"Participants were asked to evaluate their preferences regarding home and/or hospital treatment delivery in this study by answering 2 questions (Q). Q15: Do you prefer having your chemotherapy at home or at the hospital, or are you indifferent? Choices included: Home, Hospital, or Indifferent. Q16: Would you recommend having chemotherapy at home to someone else in your same situation? Choices included: Yes, No, or Not sure." (NCT01473563)
Timeframe: The first evaluation completed at either Cycle 4, Day 1 (21 days/cycle) or 30 days post treatment discontinuation

Interventionparticipants (Number)
Q15, HomeQ15, HospitalQ15, IndifferentQ16, YesQ16, NoQ16, Not sure
Pemetrexed33053700

[back to top]

Participant Satisfaction: Regarding the Study Nurse

"Participants were asked 7 questions (Q) about their study nurse for home treatment. Q8: Was the nurse an easy person to talk to?, Q9: When the nurse came, did you feel he/she had enough time to do the required things?, Q10: Do you think the nurse had time to discuss things with you?, Q11: Did you feel that the nurse knew enough about you and your illness? Choices for Q8 through Q11 included: Yes or No. Q12: Were you able to get all the information you wanted about your illness or treatment? Choices included: Yes, No, or Uncertain. Q13: Would you say that the nurse gave… Choices included: a lot of reassurance and support, some reassurance and support, or hardly any reassurance and support. Q14: How would you rate your overall satisfaction with the nursing staff during chemotherapy at home? Choices included: Very dissatisfied, Somewhat dissatisfied, Neither satisfied nor dissatisfied, Somewhat satisfied, or Very satisfied." (NCT01473563)
Timeframe: The first evaluation completed at either Cycle 4, Day 1 (21 days/cycle) or 30 days post treatment discontinuation

Interventionparticipants (Number)
Q8, YesQ8, NoQ9, YesQ9, NoQ10, YesQ10, NoQ11, YesQ11, NoQ12, YesQ12, NoQ12, UncertainQ13, Hardly any reassurance and supportQ13, Some reassurance and supportQ13, A lot of reassurance and supportQ14, Very dissatisfiedQ14, Somewhat dissatisfiedQ14, Neither satisfied nor dissatisfiedQ14, Somewhat satisfiedQ14, Very satisfied
Pemetrexed37036137136034030235301133

[back to top]

Physician Satisfaction: Distant Management of Participant

"The physician was asked, How would you rate your overall satisfaction with the distant management of the participant during chemotherapy at home? Choices included: Very dissatisfied, Somewhat dissatisfied, Neither satisfied nor dissatisfied, Somewhat satisfied, or Very satisfied." (NCT01473563)
Timeframe: 30 days post treatment discontinuation

Interventioninvestigators (Number)
Very DissatisfiedSomewhat DissatisfiedNeither Satisfied Nor DissatisfiedSomewhat SatisfiedVery Satisfied
Pemetrexed210820

[back to top]

Resource Utilization: Number of Participants With an Unplanned Use of Healthcare Resources

The number of participants who had at least 1 unplanned use of health care resources [accident and emergency department (dept.), specialists [oncologist, pulmonologist, etcetera (etc.)], general practitioner (GP) or family doctor, or diagnostic procedures] during the study is reported. (NCT01473563)
Timeframe: Cycle 1, Day 1 through last day of cycle when participant reverted to hospital administration or discontinued (up to Cycle 19, 21 days/cycle)

Interventionparticipants (Number)
Pemetrexed29

[back to top]

Overall Survival (OS) at 6 Months

The percentage of participants who were alive at Month 6 was calculated as a cumulative percentage by Kaplan-Meier survival analyses approach. For participants not known to have died as of the cut-off date, OS was censored as the last contact date (known alive). (NCT01473563)
Timeframe: Cycle 1, Day 1 to the date of death from any cause (up to Month 6)

Interventionpercentage of participants (Number)
Pemetrexed73

[back to top]

Number of Participants Who Had Treatment-Emergent Adverse Events (TEAEs), Serious Adverse Events (SAEs), or Died

The number of participants who had at least 1 TEAE or serious TEAE (regardless of causality) is reported along with the number of participants who died (due to any cause) while on therapy or during treatment discontinuation follow-up (up to 6 months). TEAEs started on or after the date and time of first dose of study drug, or started prior to study drug but worsened after study drug started. Clinically significant events were defined as SAEs and other non-serious adverse events (AEs). A summary of SAEs and other non-serious AEs is located in the Reported Adverse Events module. (NCT01473563)
Timeframe: First dose of study drug (Cycle 1, Day 1) through study completion [up to Cycle 19 (21 days/cycle) or treatment discontinuation, plus up to 6 months post treatment discontinuation]

Interventionparticipants (Number)
At least 1 TEAEAt least 1 Serious TEAEDeath, AE (fell, multiple injuries)Death, Study Drug Toxicity (atypical pneumonia)Death, Study Disease
Pemetrexed51211126

[back to top]

Change From Baseline in the EQ-5D Index Score

The EQ-5D scale was used to provide an estimate of the health state utility in this population. The EQ-5D scale includes a 5-dimensional descriptive system that measures each of the health state attributes: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression according to a 3-point scale (no problem, some problems, and major problems) and a VAS that allows participants to rate their present health condition from 0 (worst imaginable health state) to 100 (best imaginable health state). The change from baseline EQ-5D Index score is reported and the EQ-5D Index score was calculated by converting health state scores into a weighted health state index according to a United Kingdom population-based algorithm. The possible values for the EQ-5D Index score range from -0.59 (severe problems in all 5 dimensions) to 1.0 (no problem in any dimension), on a scale where 1 represents the best possible health state. (NCT01473563)
Timeframe: Baseline, Day 1 of Cycles 2 and 4 (21 days/cycle) and 30 days post treatment discontinuation

Interventionunits on a scale (Mean)
Cycle 2 (n=38)Cycle 4 (n=23)30 days post treatment discontinuation (n=26)
Pemetrexed0.030.08-0.9

[back to top]

Time to Treatment Failure (TTF)

The time from the date of the first dose of study treatment (Cycle 1, Day 1) to the date of death from any cause, PD (clinical and objective), or discontinuation of pemetrexed due to toxicity. Response was defined using RECIST, v1.1 criteria. PD was defined as having at least a 20% increase in the sum of the longest diameter of target lesions and at a minimum 5 mm increase above nadir. TTF was censored at the date of the last visit for participants who did not discontinue pemetrexed, who were still alive, and who had not progressed. (NCT01473563)
Timeframe: Cycle 1, Day 1 to first event (up to Cycle 19, 21 days/cycle)

Interventionmonths (Median)
Pemetrexed3.0

[back to top]

Resource Utilization: Duration of Health Care Visits

The duration of the health care visit in the home setting is reported. The visit started when the nurse arrived and included the entire treatment process. The visit ended when the nurse left the home setting. Due to the limited number of participants with evaluable data, results are reported for Cycles 2 through 4. (NCT01473563)
Timeframe: Cycle 2, Day 1 through last day of cycle when participant reverted to hospital administration or discontinued (up to Cycle 4, 21 days/cycle)

Interventionhours (Mean)
Cycle 2 (n=42)Cycle 3 (n=35)Cycle 4 (n=28)
Pemetrexed1.671.661.57

[back to top]

Percentage of Participants Who Adhered to Treatment Administration at Home

Participants were considered adherent from the time of the first dose in Cycle 1 (hospital administration) until either the last day of the cycle when the participant reverted to pemetrexed hospital administration or the last day of the cycle when the participant discontinued study treatment or the study for reasons related to the home setting. The percentage of participants who adhered to treatment administration at home was estimated by a Kaplan-Meier survival analyses approach. Participants who died or discontinued the study and treatment without reverting to hospital administration were censored at the time of discontinuation. (NCT01473563)
Timeframe: Cycle 1, Day 1 through Cycle 19, Day 1 and Cycle 19, Day 1 (21 days/cycle)

Interventionpercentage of participants (Number)
Cycle 1, Hospital DeliveryCycle 2, Home DeliveryCycle 3, Home DeliveryCycle 4, Home DeliveryCycle 5, Home DeliveryCycle 6, Home DeliveryCycle 7, Home DeliveryCycle 8, Home DeliveryCycle 9, Home DeliveryCycle 10, Home DeliveryCycle 11, Home DeliveryCycle 12, Home DeliveryCycle 13, Home DeliveryCycle 14, Home DeliveryCycle 15, Home DeliveryCycle 16, Home DeliveryCycle 17, Home DeliveryCycle 18, Home DeliveryCycle 19, Home Delivery
Pemetrexed10098.098.098.098.098.090.790.790.790.790.790.790.790.790.790.790.790.790.7

[back to top]

Participant Satisfaction: Chemotherapy at Home

"Participants were asked to evaluate their home treatment experiences in this study by answering 4 questions (Q). Q5: What do you do consider advantages of having chemotherapy at home? Choose all that apply. Choices included: No need to travel, Not having to wait for treatment, Personalized service, More privacy, and Other. Q6:What do you consider disadvantages of having chemotherapy at home? Choose all that apply. Choices included: Lack of other patients' support, Extra burden for family/friends, Safety concerns, Need to rely on 1 medical specialist, and Other. Q7: How would you rate your overall satisfaction with chemotherapy at home? Choices included: Very dissatisfied, Somewhat dissatisfied, Neither satisfied nor dissatisfied, Somewhat satisfied, or Very satisfied." (NCT01473563)
Timeframe: The first evaluation completed at either Cycle 4, Day 1 (21 days/cycle) or 30 days post treatment discontinuation

Interventionparticipants (Number)
Q5, No need to travelQ5, Not having to wait for treatmentQ5, Personalized serviceQ5, More privacyQ5, OtherQ6, Lack of other patients' supportQ6, Extra burden for family/friendsQ6, Safety concernsQ6, Need to rely on 1 medical specialistQ6, OtherQ7, Very dissatisfiedQ7, Somewhat dissatisfiedQ7, Neither satisfied nor dissatisfiedQ7, Somewhat satisfiedQ7, Very Satisfied
Pemetrexed372728202514719102130

[back to top]

Resource Utilization: Unplanned Health Care Visits, Consultations, and Diagnostic Services

The unplanned use of any 1 of the following 4 resources is reported, as well as the unplanned use of each resource: accident and emergency dept., specialists (oncologist, pulmonologist etc.), GP or family doctor, and diagnostic procedures. Results are reported as the number of participants with an unplanned resource use (visit) for a specified number of times. (NCT01473563)
Timeframe: Cycle 1, Day 1 through last day of cycle when participant reverted to hospital administration or discontinued (up to Cycle 19, 21 days/cycle)

Interventionparticipants (Number)
1 Unplanned Visit, Any Resource2 Unplanned Visits, Any Resource3 Unplanned Visits, Any Resource4 Unplanned Visits, Any Resource5 Unplanned Visits, Any Resource9 Unplanned Visits, Any Resource10 Unplanned Visits, Any Resource13 Unplanned Visits, Any Resource1 Unplanned Visit, Accident and Emergency Dept.2 Unplanned Visits, Accident and Emergency Dept.3 Unplanned Visits, Accident and Emergency Dept.1 Unplanned Visit, Specialist2 Unplanned Visits, Specialist5 Unplanned Visits, Specialist1 Unplanned Visit, GP or Family Doctor2 Unplanned Visits, GP or Family Doctor3 Unplanned Visits, GP or Family Doctor4 Unplanned Visits, GP or Family Doctor1 Unplanned Visit, Diagnostic procedures2 Unplanned Visits, Diagnostic procedures3 Unplanned Visits, Diagnostic procedures4 Unplanned Visits, Diagnostic procedures
Pemetrexed8652311172293163236231

[back to top]

Resource Utilization: Distances Traveled

The distance traveled is reported by region (Great Britain and Sweden) and includes the distance traveled by the participant from his/her home to the hospital (Cycle 1) and other cycles where the homecare nurse traveled from the hospital to the participant's home. Due to the limited number of participants with evaluable data, results are reported for Cycles 1 through 4. (NCT01473563)
Timeframe: Cycle 1, Day 1 through last day of cycle when participant reverted to hospital administration or discontinued (up to Cycle 4, 21 days/cycle)

Interventionkilometers (km) (Mean)
Cycle 1, Home to Hospital, Great Britain (n=25)Cycle 1, Home to Hospital, Sweden (n=7)Cycle 2, Hospital to Home, Great Britain (n=24)Cycle 2, Hospital to Home, Sweden (n=7)Cycle 3, Hospital to Home, Great Britain (n=11)Cycle 3, Hospital to Home, Sweden (n=4)Cycle 4, Hospital to Home, Great Britain (n=7)Cycle 4, Hospital to Home, Sweden (n=2)
Pemetrexed19.730.715.523.923.817.511.724.5

[back to top]

Participant Satisfaction: Chemotherapy at Hospital

"Participants were asked to evaluate their hospital experiences in this study by answering 4 questions (Q). Q1: What do you consider advantages of having chemotherapy at the hospital? Choose all that apply. Choices included: Support from other patients, Access to other medical specialists, Access to more technical services, Safer in case something goes wrong, and Other. Q2: What do you consider disadvantages of having chemotherapy at the hospital? Choose all that apply. Choices included: Need to travel, Having to wait for treatment, Not having a personalized treatment, Lack of privacy on the ward, and Other. Q3: How would you rate your overall satisfaction with chemotherapy at the hospital? and Q4: How would you rate your overall satisfaction with the nursing staff during chemotherapy at the hospital? Choices for Q3 and Q4 included: Very dissatisfied, Somewhat dissatisfied, Neither satisfied nor dissatisfied, Somewhat satisfied, or Very satisfied." (NCT01473563)
Timeframe: The first evaluation completed at either Cycle 4, Day 1 (21 days/cycle) or 30 days post treatment discontinuation

Interventionparticipants (Number)
Q1, Support from other patientsQ1, Access to other medical specialistsQ1, Access to more technical servicesQ1, Safer in case something goes wrongQ1, OtherQ2, Need to travelQ2, Having to wait for treatmentQ2, Not having a personalized treatmentQ2, Lack of privacy on the wardQ2, OtherQ3, Very dissatisfiedQ3, Somewhat dissatisfiedQ3, Neither satisfied nor dissatisfiedQ3, Somewhat satisfiedQ3, Very satisfiedQ4, Very dissatisfiedQ4, Somewhat dissatisfiedQ4, Neither satisfied nor dissatisfiedQ4, Somewhat satisfiedQ4, Very satisfied
Pemetrexed8191219336275710331121300332

[back to top]

Change From Baseline in the European Quality of Life Instrument (EQ-5D) Visual Analogue Scale (VAS)

The EQ-5D scale was used to provide an estimate of the health state utility in this population. The EQ-5D scale includes a 5-dimensional descriptive system that measures each of the health state attributes: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression according to a 3-point scale (no problem, some problems, and major problems) and a VAS that allows participants to rate their present health condition from 0 (worst imaginable health state) to 100 (best imaginable health state). The change from baseline in EQ-5D VAS is reported. (NCT01473563)
Timeframe: Baseline, Day 1 of Cycles 2 and 4 (21 days/cycle) and 30 days post treatment discontinuation

Interventionunits on a scale (Mean)
Cycle 2 (n=34)Cycle 4 (n=20)30 days post treatment discontinuation (n=22)
Pemetrexed3.07.7-0.9

[back to top]

Maximum Improvement Over Baseline in Individual Lung Cancer Symptoms Scale (LCSS) Item Scores

LCSS is a 9-item questionnaire; 6 items are symptom-specific measures for lung cancer (loss of appetite, fatigue, cough, dyspnea, hemoptysis, and pain), and 3 summation items describe overall symptomatic distress, interference with activity level, and overall quality of life during the past 24 hours. Participant responses were measured using a VAS with 100-millimeter (mm) lines. Scores ranged from 0 mm (no symptoms and no impact on activities, quality of life) to 100 mm (symptoms as bad as they could be, impacting activities and quality of life). (NCT01473563)
Timeframe: Baseline, Day 1 of each cycle (up to Cycle 19, 21 days/cycle), and 30 days post treatment discontinuation

Interventionmm (Mean)
Loss of Appetite (n=43)Fatigue (n=43)Cough (n=41)Dyspnea (n=41)Hemoptysis (n=43)Pain (n=40)Overall Symptomatic Distress (n=43)Interference With Activity Level (n=43)Overall Quality of Life (n=41)
Pemetrexed16.324.59.217.20.411.88.315.312.2

[back to top]

Median Overall Survival (OS) at Time of PFS Analysis

(NCT01544179)
Timeframe: Baseline and then every 6 weeks after randomization until objective disease progression. OS is then assessed 8 weekly following PFS progression up to PFS analysis data cut off.

InterventionMonths (Median)
Gefitinib14.8
Placebo17.2

[back to top]

Median Progression-Free Survival (Site Read, Investigator Assessment)

PFS is the time from randomisation until the date of objective disease progression as defined by Response Evaluation Criteria In Solid Tumours (RECIST version 1.1) or death (by any cause in the absence of progression). Progression is defined using RECIST (v1.1), 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 also demonstrate an absolute increase of at least 5mm. (NCT01544179)
Timeframe: Radiologic evaluations were carried out every 6 weeks from randomization until documented progression, withdrawal of consent, loss to follow up, death or the primary data cut off (DCO) for the analysis, assessed up to 50 weeks

InterventionMonths (Median)
Gefitinib5.4
Placebo5.4

[back to top]

Objective Response Rate (ORR) (Site Read Data)

ORR rate is defined as the number (%) of subjects with at least one visit response of Complete Response (CR) or Partial Response (PR) , as defined by Response Evaluation Criteria in Solid Tumours (RECIST v1.1) for target lesions and assessed by CT or MRI. CR, Disappearance of all target lesions; PR, ≥30% decrease in the sum of the longest diameter of target lesions. Data obtained up until progression, or last evaluable assessment in the absence of progression, was included in the assessment of ORR. (NCT01544179)
Timeframe: Radiologic evaluations were carried out every 6 weeks from randomization until documented progression, withdrawal of consent, loss to follow up, death or the primary data cut off (DCO) for the analysis.

InterventionPercentage of Participants (Number)
Gefitinib31.6
Placebo34.1

[back to top]

Progression-Free Survival (Site Read, Investigator Assessment)

PFS is the time from randomisation until the date of objective disease progression as defined by Response Evaluation Criteria In Solid Tumours (RECIST version 1.1) or death (by any cause in the absence of progression). Progression is defined using RECIST (v1.1), 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 also demonstrate an absolute increase of at least 5mm. (NCT01544179)
Timeframe: Radiologic evaluations were carried out every 6 weeks from randomization until documented progression, withdrawal of consent, loss to follow up, death or the primary data cut off (DCO) for the analysis, assessed up to 50 weeks

InterventionPatients with a progression event (Number)
Gefitinib98
Placebo107

[back to top]

Time to Worsening in FACT-L Total Score

A worsening is defined as a change from baseline of ≤ -6 (0-136 score range). Measured by the Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) questionnaire (NCT01544179)
Timeframe: At visits 2-8, then every 6 weeks until progression, at progression or treatment discontinuation, and every 8 weeks after progression until PFS analysis data cut off.

InterventionWeeks (Median)
Gefitinib12.0
Placebo8.9

[back to top]

Time to Worsening in Lung Cancer Subscale

A worsening is defined as a change from baseline of ≤ -2 (0-28 score range). Measured by the Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) questionnaire (NCT01544179)
Timeframe: At visits 2-8, then every 6 weeks until progression, at progression or treatment discontinuation, and every 8 weeks after progression until PFS analysis data cut off.

InterventionWeeks (Median)
Gefitinib14.6
Placebo9.1

[back to top]

Overall Survival (OS)

OS is the time from the date of randomisation until death due to any cause. Any subject not known to have died at the time of analysis will be censored based on the last recorded date on which the subject was known to be alive. (NCT01544179)
Timeframe: Following progression survival data was collected every 8 weeks until documentation of death, withdrawal of consent, loss to follow-up or the final data cut-off, whichever occurs first.

InterventionNumber of patients with an OS event (Number)
Gefitinib50
Placebo37

[back to top]

Time to Worsening in Trial Outcome Index

A worsening is defined as a change from baseline of ≤ -6 (0-84 score range). Measured by the Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) questionnaire (NCT01544179)
Timeframe: At visits 2-8, then every 6 weeks until progression, at progression or treatment discontinuation, and every 8 weeks after progression until PFS analysis data cut off.

InterventionWeeks (Median)
Gefitinib12.1
Placebo9.4

[back to top]

Disease Control Rate (DCR)

DCR is the percentage of patients who achieve disease control at 6 weeks following randomisation. DCR is defined as a Best Objective Response (BOR) of Complete Response, Partial Response or Stable Disease, as defined by Response Evaluation Criteria in Solid Tumours (RECIST v1.1) for target lesions and assessed by CT or MRI. CR, Disappearance of all target lesions; PR, ≥30% decrease in the sum of the longest diameter of target lesions; SD, neither sufficient shrinkage to qualify for PR not sufficient increase to qualify for Progressive Disease (PD); PD, ≥20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study, and the sum must have shown an absolute increase of ≥5mm (NCT01544179)
Timeframe: Radiologic evaluations were carried out every 6 weeks from randomization until documented progression, withdrawal of consent, loss to follow up, death or the primary data cut off (DCO) for the analysis.

InterventionPercentage of Participants (Number)
Gefitinib84.2
Placebo78.8

[back to top]

Improvement in FACT-L Total Score

An improvement is defined as a change from baseline of ≥ +6 (0-136 score range). Measured by the Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) questionnaire (NCT01544179)
Timeframe: At visits 2-8, then every 6 weeks until progression, at progression or treatment discontinuation, and every 8 weeks after progression until PFS analysis data cut off.

InterventionNumber of patients improving (Number)
Gefitinib44
Placebo49

[back to top]

Improvement in Trial Outcome Index

An improvement is defined as a change from baseline of ≥ +6 (0-84 score range). Measured by the Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) questionnaire. (NCT01544179)
Timeframe: At visits 2-8, then every 6 weeks until progression, at progression or treatment discontinuation, and every 8 weeks after progression until PFS analysis data cut off.

InterventionNumber of participants improving (Number)
Gefitinib36
Placebo39

[back to top]

Improvement in Lung Cancer Subscale

An improvement is defined as a change from baseline of ≥ +2 (0-28 score range). Measured by the Functional Assessment of Cancer Therapy for Lung Cancer (FACT-L) questionnaire (NCT01544179)
Timeframe: At visits 2-8, then every 6 weeks until progression, at progression or treatment discontinuation, and every 8 weeks after progression until PFS analysis data cut off.

InterventionNumber of participants improving (Number)
Gefitinib54
Placebo55

[back to top]

Overall Survival

(NCT01565538)
Timeframe: From date of randomization until the date of death from any cause, assessed until at least 12 months after randomization.

Interventionmonths (Median)
Erlotinib11.7
Pemetrexed13.4

[back to top]

Progression-Free Survival

Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT01565538)
Timeframe: From the date of randomization to the date of tumour progression or death from any cause, assessed until at least 12 months after randomization.

Interventionmonths (Median)
Erlotinib4.1
Pemetrexed3.9

[back to top]

Best Tumor Response

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by CT: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT01565538)
Timeframe: From the date of randomization, assessed every 6 weeks, until at least 12 months after randomization.

,
Interventionparticipants (Number)
Objective responseNo objective response
Erlotinib1249
Pemetrexed557

[back to top]

Percentage of Participants With Visual Disturbance as Assessed by Visual Symptom Assessment Questionnaire (VSAQ-ALK)

"VSAQ-ALK is a self-report measure that was developed to assess the problems of visual disturbances and symptoms may include the appearance of overlapping shadows and after images; shimmering, flashing or trailing lights; strings, streamers, or floaters; as well as hazy or blurry vision. The participants answered Yes to the first question (Q1) of VSAQ-ALK Have you experienced any visual disturbances? were considered to have experienced visual disturbance and were instructed to complete the rest of the questionnaire. The percentage of participants who responded to Q1 of VSAQ-ALK as Yes and as No during each study cycle was calculated as (n/N*)*100 where N* was the number of participants who had completed Q1." (NCT01639001)
Timeframe: Cycle 1 Day 1 to end of treatment or withdrawal, no later than 4 weeks (+/- 1 week) from last dose of study medication or when the decision was taken to withdraw from the study (whichever was sooner, assessed up to Cycle 86)

InterventionPercentage of participants (Number)
Baseline: Cycle 1/Day 1 (Answer to Q1: Yes)Baseline: Cycle 1/Day 1 (Answer to Q1: No)Cycle 2/Day 1 (Answer to Q1: Yes)Cycle 2/Day 1 (Answer to Q1: No)Cycle 3/Day 1 (Answer to Q1: Yes)Cycle 3/Day 1 (Answer to Q1: No)Cycle 4/Day 1 (Answer to Q1: Yes)Cycle 4/Day 1 (Answer to Q1: No)Cycle 5/Day 1 (Answer to Q1: Yes)Cycle 5/Day 1 (Answer to Q1: No)Cycle 6/Day 1 (Answer to Q1: Yes)Cycle 6/Day 1 (Answer to Q1: No)Cycle 7/Day 1 (Answer to Q1: Yes)Cycle 7/Day 1 (Answer to Q1: No)Cycle 8/Day 1 (Answer to Q1: Yes)Cycle 8/Day 1 (Answer to Q1: No)Cycle 9/Day 1 (Answer to Q1: Yes)Cycle 9/Day 1 (Answer to Q1: No)Cycle 10/Day 1 (Answer to Q1: Yes)Cycle 10/Day 1 (Answer to Q1: No)Cycle 11/Day 1 (Answer to Q1: Yes)Cycle 11/Day 1 (Answer to Q1: No)Cycle 12/Day 1 (Answer to Q1: Yes)Cycle 12/Day 1 (Answer to Q1: No)Cycle 13/Day 1 (Answer to Q1: Yes)Cycle 13/Day 1 (Answer to Q1: No)Cycle 14/Day 1 (Answer to Q1: Yes)Cycle 14/Day 1 (Answer to Q1: No)Cycle 15/Day 1 (Answer to Q1: Yes)Cycle 15/Day 1 (Answer to Q1: No)Cycle 16/Day 1 (Answer to Q1: Yes)Cycle 16/Day 1 (Answer to Q1: No)Cycle 17/Day 1 (Answer to Q1: Yes)Cycle 17/Day 1 (Answer to Q1: No)Cycle 18/Day 1 (Answer to Q1: Yes)Cycle 18/Day 1 (Answer to Q1: No)Cycle 19/Day 1 (Answer to Q1: Yes)Cycle 19/Day 1 (Answer to Q1: No)Cycle 20/Day 1 (Answer to Q1: Yes)Cycle 20/Day 1 (Answer to Q1: No)Cycle 21/Day 1 (Answer to Q1: Yes)Cycle 21/Day 1 (Answer to Q1: No)Cycle 22/Day 1 (Answer to Q1: Yes)Cycle 22/Day 1 (Answer to Q1: No)Cycle 23/Day 1 (Answer to Q1: Yes)Cycle 23/Day 1 (Answer to Q1: No)Cycle 24/Day 1 (Answer to Q1: Yes)Cycle 24/Day 1 (Answer to Q1: No)Cycle 25/Day 1 (Answer to Q1: Yes)Cycle 25/Day 1 (Answer to Q1: No)Cycle 26/Day 1 (Answer to Q1: Yes)Cycle 26/Day 1 (Answer to Q1: No)Cycle 27/Day 1 (Answer to Q1: Yes)Cycle 27/Day 1 (Answer to Q1: No)Cycle 28/Day 1 (Answer to Q1: Yes)Cycle 28/Day 1 (Answer to Q1: No)Cycle 29/Day 1 (Answer to Q1: Yes)Cycle 29/Day 1 (Answer to Q1: No)Cycle 30/Day 1 (Answer to Q1: Yes)Cycle 30/Day 1 (Answer to Q1: No)Cycle 31/Day 1 (Answer to Q1: Yes)Cycle 31/Day 1 (Answer to Q1: No)Cycle 32/Day 1 (Answer to Q1: Yes)Cycle 32/Day 1 (Answer to Q1: No)Cycle 33/Day 1 (Answer to Q1: Yes)Cycle 33/Day 1 (Answer to Q1: No)Cycle 34/Day 1 (Answer to Q1: Yes)Cycle 34/Day 1 (Answer to Q1: No)Cycle 35/Day 1 (Answer to Q1: Yes)Cycle 35/Day 1 (Answer to Q1: No)Cycle 36/Day 1 (Answer to Q1: Yes)Cycle 36/Day 1 (Answer to Q1: No)Cycle 37/Day 1 (Answer to Q1: Yes)Cycle 37/Day 1 (Answer to Q1: No)Cycle 38/Day 1 (Answer to Q1: Yes)Cycle 38/Day 1 (Answer to Q1: No)Cycle 39/Day 1 (Answer to Q1: Yes)Cycle 39/Day 1 (Answer to Q1: No)Cycle 40/Day 1 (Answer to Q1: Yes)Cycle 40/Day 1 (Answer to Q1: No)Cycle 41/Day 1 (Answer to Q1: Yes)Cycle 41/Day 1 (Answer to Q1: No)Cycle 42/Day 1 (Answer to Q1: Yes)Cycle 42/Day 1 (Answer to Q1: No)Cycle 43/Day 1 (Answer to Q1: Yes)Cycle 43/Day 1 (Answer to Q1: No)Cycle 44/Day 1 (Answer to Q1: Yes)Cycle 44/Day 1 (Answer to Q1: No)Cycle 45/Day 1 (Answer to Q1: Yes)Cycle 45/Day 1 (Answer to Q1: No)Cycle 46/Day 1 (Answer to Q1: Yes)Cycle 46/Day 1 (Answer to Q1: No)Cycle 47/Day 1 (Answer to Q1: Yes)Cycle 47/Day 1 (Answer to Q1: No)Cycle 48/Day 1 (Answer to Q1: Yes)Cycle 48/Day 1 (Answer to Q1: No)Cycle 49/Day 1 (Answer to Q1: Yes)Cycle 49/Day 1 (Answer to Q1: No)Cycle 50/Day 1 (Answer to Q1: Yes)Cycle 50/Day 1 (Answer to Q1: No)Cycle 51/Day 1 (Answer to Q1: Yes)Cycle 51/Day 1 (Answer to Q1: No)Cycle 52/Day 1 (Answer to Q1: Yes)Cycle 52/Day 1 (Answer to Q1: No)Cycle 53/Day 1 (Answer to Q1: Yes)Cycle 53/Day 1 (Answer to Q1: No)Cycle 54/Day 1 (Answer to Q1: Yes)Cycle 54/Day 1 (Answer to Q1: No)Cycle 55/Day 1 (Answer to Q1: Yes)Cycle 55/Day 1 (Answer to Q1: No)Cycle 56/Day 1 (Answer to Q1: Yes)Cycle 56/Day 1 (Answer to Q1: No)Cycle 57/Day 1 (Answer to Q1: Yes)Cycle 57/Day 1 (Answer to Q1: No)Cycle 58/Day 1 (Answer to Q1: Yes)Cycle 58/Day 1 (Answer to Q1: No)Cycle 59/Day 1 (Answer to Q1: Yes)Cycle 59/Day 1 (Answer to Q1: No)Cycle 60/Day 1 (Answer to Q1: Yes)Cycle 60/Day 1 (Answer to Q1: No)Cycle 61/Day 1 (Answer to Q1: Yes)Cycle 61/Day 1 (Answer to Q1: No)Cycle 62/Day 1 (Answer to Q1: Yes)Cycle 62/Day 1 (Answer to Q1: No)Cycle 63/Day 1 (Answer to Q1: Yes)Cycle 63/Day 1 (Answer to Q1: No)Cycle 64/Day 1 (Answer to Q1: Yes)Cycle 64/Day 1 (Answer to Q1: No)Cycle 65/Day 1 (Answer to Q1: Yes)Cycle 65/Day 1 (Answer to Q1: No)Cycle 66/Day 1 (Answer to Q1: Yes)Cycle 66/Day 1 (Answer to Q1: No)Cycle 67/Day 1 (Answer to Q1: Yes)Cycle 67/Day 1 (Answer to Q1: No)Cycle 68/Day 1 (Answer to Q1: Yes)Cycle 68/Day 1 (Answer to Q1: No)Cycle 69/Day 1 (Answer to Q1: Yes)Cycle 69/Day 1 (Answer to Q1: No)Cycle 70/Day 1 (Answer to Q1: Yes)Cycle 70/Day 1 (Answer to Q1: No)Cycle 71/Day 1 (Answer to Q1: Yes)Cycle 71/Day 1 (Answer to Q1: No)Cycle 72/Day 1 (Answer to Q1: Yes)Cycle 72/Day 1 (Answer to Q1: No)Cycle 73/Day 1 (Answer to Q1: Yes)Cycle 73/Day 1 (No)Cycle 74/Day 1 (Answer to Q1: Yes)Cycle 74/Day 1 (Answer to Q1: No)Cycle 75/Day 1 (Answer to Q1: Yes)Cycle 75/Day 1 (Answer to Q1: No)Cycle 76/Day 1 (Answer to Q1: Yes)Cycle 76/Day 1 (Answer to Q1: No)Cycle 77/Day 1 (Answer to Q1: Yes)Cycle 77/Day 1 (Answer to Q1: No)Cycle 78/Day 1 (Answer to Q1: Yes)Cycle 78/Day 1 (Answer to Q1: No)Cycle 79/Day 1 (Answer to Q1: Yes)Cycle 79/Day 1 (Answer to Q1: No)Cycle 80/Day 1 (Answer to Q1: Yes)Cycle 80/Day 1 (Answer to Q1: No)Cycle 81/Day 1 (Answer to Q1: Yes)Cycle 81/Day 1 (Answer to Q1: No)Cycle 82/Day 1 (Answer to Q1: Yes)Cycle 82/Day 1 Answer to Q1: (No)Cycle 83/Day 1 (Answer to Q1: Yes)Cycle 83/Day 1 (Answer to Q1: No)Cycle 84/Day 1 (Answer to Q1: Yes)Cycle 84/Day 1 (Answer to Q1: No)Cycle 86/Day 1 (Answer to Q1: Yes)Cycle 86/Day 1 (Answer to Q1: No)
Crizotinib6.893.251.049.046.153.943.956.135.864.237.462.636.064.028.171.924.175.923.976.123.576.521.378.825.374.727.073.028.271.828.671.428.671.428.471.628.171.927.972.130.569.529.170.928.671.431.568.532.167.928.871.228.072.030.070.030.070.032.068.024.575.525.075.028.371.727.372.728.671.426.873.227.073.024.375.730.369.725.075.032.367.725.874.224.175.930.070.025.075.027.672.426.973.129.670.429.270.829.270.826.173.929.270.825.075.036.463.629.470.638.961.120.080.035.364.721.478.637.562.523.176.937.562.521.478.635.764.321.478.630.869.225.075.030.869.225.075.030.869.230.070.022.277.822.277.825.075.022.277.816.783.337.562.520.080.020.080.00.0100.00.0100.00.0100.00.0100.00.0100.00.0100.0

[back to top]

Time to Deterioration (TTD) in Participant Reported Pain, Dyspnea, or Cough Assessed Using Quality of Life Questionnaire Supplement Module for Lung Cancer (QLQ-LC13)

The QLQ-LC13 consisted of 1 multi-item scale and 9 single items that assessed the specific symptoms (dyspnea, cough, hemoptysis, and site specific pain), side effects (sore mouth, dysphagia, neuropathy, and alopecia), and pain medication use of lung cancer participants receiving chemotherapy. The QLQ-LC13 Coughing, Dyspnoea and Pain in chest each ranged from 0-100 with higher scores indicating a high level of symptomatology/problems. TTD in pain in chest, dyspnea, or cough from the QLQ-LC13 was a composite endpoint defined as the time from randomization to the earliest time the participant's scale scores showed a 10 point or greater increase after baseline in any of the 3 symptoms. (NCT01639001)
Timeframe: From Baseline to deterioration while on study treatment. For participants with no deterioration, the data was censored at the last date when QLQ-LC13 assessment for pain, dyspnea, or cough was completed (assessed up to 33 months)

InterventionMonths (Median)
Crizotinib2.8
Chemotherapy0.3

[back to top]

Percentage of Participants With Visual Disturbance as Assessed by Visual Symptom Assessment Questionnaire (VSAQ-ALK)

"VSAQ-ALK is a self-report measure that was developed to assess the problems of visual disturbances and symptoms may include the appearance of overlapping shadows and after images; shimmering, flashing or trailing lights; strings, streamers, or floaters; as well as hazy or blurry vision. The participants answered Yes to the first question (Q1) of VSAQ-ALK Have you experienced any visual disturbances? were considered to have experienced visual disturbance and were instructed to complete the rest of the questionnaire. The percentage of participants who responded to Q1 of VSAQ-ALK as Yes and as No during each study cycle was calculated as (n/N*)*100 where N* was the number of participants who had completed Q1." (NCT01639001)
Timeframe: Cycle 1 Day 1 to end of treatment or withdrawal, no later than 4 weeks (+/- 1 week) from last dose of study medication or when the decision was taken to withdraw from the study (whichever was sooner, assessed up to Cycle 86)

InterventionPercentage of participants (Number)
Baseline: Cycle 1/Day 1 (Answer to Q1: Yes)Baseline: Cycle 1/Day 1 (Answer to Q1: No)Cycle 2/Day 1 (Answer to Q1: Yes)Cycle 2/Day 1 (Answer to Q1: No)Cycle 3/Day 1 (Answer to Q1: Yes)Cycle 3/Day 1 (Answer to Q1: No)Cycle 4/Day 1 (Answer to Q1: Yes)Cycle 4/Day 1 (Answer to Q1: No)Cycle 5/Day 1 (Answer to Q1: Yes)Cycle 5/Day 1 (Answer to Q1: No)Cycle 6/Day 1 (Answer to Q1: Yes)Cycle 6/Day 1 (Answer to Q1: No)
Chemotherapy7.192.913.486.611.188.914.885.213.586.510.189.9

[back to top]

Percentage of Participants With Treatment-Emergent AEs (Treatment Related)

An AE was an untoward medical occurrence in a participant who received study treatment without regard to possibility of causal relationship. Serious adverse event (SAE) was an AE resulting in any of the following outcomes: death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. Grade 3 and 4 AEs in below table indicated severe AE and life-threatening consequences respectively; Grade 5 indicated death due to AE. (NCT01639001)
Timeframe: From the first dose of study medication up to 28 days after the last dose of study medication or up to the time that a participant died or received subsequent anticancer treatment (maximum duration between first and last dose: 324.4 weeks)

,
InterventionPercentage of participants (Number)
AEsSerious AEsGrade 3/4 AEsGrade 5 AEsAEs associated with permanent discontinuationAEs associated with dose reductionAEs associated with temporary discontinuation
Chemotherapy96.03.040.601.07.931.7
Crizotinib98.18.743.31.95.813.528.8

[back to top]

Percentage of Participants With Treatment-Emergent Adverse Events (AEs; All Causalities)

An AE was an untoward medical occurrence in a participant who received study treatment without regard to possibility of causal relationship. Serious adverse event (SAE) was an AE resulting in any of the following outcomes: death, initial or prolonged inpatient hospitalization, life-threatening experience, persistent or significant disability/incapacity, congenital anomaly. Treatment-emergent AEs were those with initial onset or that worsen in severity after the first dose of study medication. Grade 3 and 4 AEs in below table indicated severe AE and life-threatening consequences respectively; Grade 5 indicated death due to AE. (NCT01639001)
Timeframe: From the first dose of study medication up to 28 days after the last dose of study medication or up to the time that a participant died or received subsequent anticancer treatment (maximum duration between first and last dose: 324.4 weeks)

,
InterventionPercentage of participants (Number)
AEsSAEsGrade 3/4 AEsGrade 5 AEsAEs associated with permanent discontinuationAEs associated with dose reductionAEs associated with temporary discontinuation
Chemotherapy99.012.952.52.04.07.937.6
Crizotinib99.044.258.722.126.914.439.4

[back to top]

Estimate of the Percentage of Participants Surviving at 1 Year and at 18 Months

Probability of survival 1 year and 18 month after randomization. The probability of survival at 1 year was estimated using the Kaplan Meier method and a 2-sided 95% CI for the log [-log(1-year survival probability)] was calculated using a normal approximation and then back transformed to give a CI for the 1-year survival probability itself. The probability of survival at 18 months was estimated similarly. (NCT01639001)
Timeframe: From randomization to 1 year and from randomization to 18 months

,
InterventionPercentage of paricipants (Number)
Up to 1 yearUp to 18 months
Chemotherapy79.572.1
Crizotinib79.371.2

[back to top]

Change From Baseline Scores in QLQ-C30 Symptoms as Assessed by the EORTC-QLQ-C30

EORTC QLQ-C30: included functional scales (physical, role, cognitive, emotional, and social), global health status, symptom scales (fatigue, pain, nausea/vomiting) and single items (dyspnoea, appetite loss, insomnia, constipation/diarrhea and financial difficulties). The QLQ-C30 Appetite loss, Constipation, Diarrhea, Dyspnea, Fatigue, Financial difficulties, Insomnia, Nausea/vomiting, and Pain each ranged from 0-100 with higher scores indicating a high level of symptomatology/problems. (NCT01639001)
Timeframe: From Baseline to deterioration while on study treatment. For participants with no deterioration, the data was censored at the last date when QLQ-LC13 assessment for pain, dyspnea, or cough was completed (assessed up to 33 months)

,
InterventionUnits on a scale (Mean)
QLQ-C30 Appetite lossQLQ-C30 ConstipationQLQ-C30 DiarrheaQLQ-C30 DyspneaQLQ-C30 FatigueQLQ-C30 Financial DifficultiesQLQ-C30 InsomniaQLQ-C30 Nausea and VomitingQLQ-C30 Pain
Chemotherapy4.44652.6341-0.4791-0.19032.60280.3826-1.60606.5986-0.6956
Crizotinib-1.59677.136715.3294-7.9353-3.8888-3.2339-8.38164.0796-9.1305

[back to top]

Change From Baseline in Lung Cancer Symptom Scores as Assessed by the EORTC Quality of Life Questionnaire-Lung Cancer 13 (QLQ-LC13)

The QLQ-LC13 consisted of 1 multi-item scale and 9 single items that assessed the specific symptoms (dyspnea, cough, hemoptysis, and site specific pain), side effects (sore mouth, dysphagia, neuropathy, and alopecia), and pain medication use of lung cancer participants receiving chemotherapy. The QLQ-LC13 Alopecia, Coughing, Dysphagia, Dyspnoea, Haemoptysis, Pain in arm or shoulder, Pain in chest, Pain in other parts, Peripheral neuropathy, and Sore mouth each ranged from 0-100 with higher scores indicating a high level of symptomatology/problems. (NCT01639001)
Timeframe: From Baseline to deterioration while on study treatment. For participants with no deterioration, the data was censored at the last date when QLQ-LC13 assessment for pain, dyspnea, or cough was completed (assessed up to 33 months)

,
InterventionUnits on a scale (Mean)
QLQ-LC13 AlopeciaQLQ-LC13 CoughingQLQ-LC13 DysphagiaQLQ-LC13 DyspnoeaQLQ-LC13 HaemoptysisQLQ-LC13 Pain in Arm or ShoulderQLQ-LC13 Pain in ChestQLQ-LC13 Pain in Other PartsQLQ-LC13 Peripheral NeuropathyQLQ-LC13 Sore Mouth
Chemotherapy2.7710-10.27481.0535-0.4371-3.0513-2.5927-4.1328-0.25731.85193.9114
Crizotinib-2.0837-17.27040.5354-9.0842-4.3017-6.8289-8.3565-4.84750.17871.5134

[back to top]

Change From Baseline in Functioning and Global Quality of Life (QOL) as Assessed by the European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC-QLQ-C30)

EORTC QLQ-C30: included functional scales (physical, role, cognitive, emotional, and social), global health status, symptom scales (fatigue, pain, nausea/vomiting) and single items (dyspnoea, appetite loss, insomnia, constipation/diarrhea and financial difficulties). The QLQ-C30 Global QOL, Physical Functioning, Role Functioning, Cognitive Functioning, Emotional Functioning, and Social Functioning each ranged from 0-100 with higher scores indicating a better level of functioning or better quality of life. (NCT01639001)
Timeframe: From Baseline to deterioration while on study treatment. For participants with no deterioration, the data was censored at the last date when QLQ-LC13 assessment for pain, dyspnea, or cough was completed (assessed up to 33 months)

,
InterventionUnits on a scale (Mean)
QLQ-C30 Global QoLQLQ-C30 Cognitive FunctioningQLQ-C30 Emotional FunctioningQLQ-C30 Physical FunctioningQLQ-C30 Role FunctioningQLQ-C30 Social Functioning
Chemotherapy-2.3619-4.80262.0557-2.9562-5.7570-4.7228
Crizotinib5.0891-1.18963.70773.77051.05380.8712

[back to top]

Agreement Between Central Laboratory ALK FISH and ALK IHC Test Results - Molecular Profiling Evaluable

Agreement between central laboratory anaplastic lymphoma kinase (ALK) fluorescence in situ hybridization (FISH) and ALK immunohistochemistry (IHC) test results is based on analysis of participants in the Molecular Profiling (MP) evaluable population that have an ALK IHC result and an ALK FISH result of either positive or negative only. This MP evaluable population included participants who screen failed, which their ALK test results were negative based on FISH test. Tumor tissue samples from these screen failure participants were consented and kept. These samples served as a part of negative sample set for evaluation of IHC test and/or polymerase chain reaction (PCR) to determine ALK fusion events. Participants with FISH results of uninformative and assay not performed and IHC results of valid IHC status not available were excluded from the analysis of agreement between central laboratory ALK FISH and ALK IHC test results. (NCT01639001)
Timeframe: During the screening (less than or equal to 28 days prior to dosing)

,
InterventionParticipants (Count of Participants)
Participants with Positive ALK IHC StatusParticipants with Negative ALK IHC Status
Participants With Negative ALK FISH Status31502
Participants With Positive ALK FISH Status21820

[back to top]

Time to Tumor Response (TTR) Based on IRR

TTR was defined as the time from randomization to first documentation of objective tumor response (CR or PR) as determined by the IRR. For participants proceeding from PR to CR, the onset of PR was taken as the onset of response. TTR was calculated for the subgroup of participants with objective tumor response. (NCT01639001)
Timeframe: Randomization to first documentation of objective tumor response (up to 33 months).

InterventionWeeks (Median)
Crizotinib6.3
Chemotherapy12.1

[back to top]

Time to Progression (TTP) Based on IRR

TTP was defined as the time from the date of randomization to the date of the first documentation of objective tumor progression, as determined by IRR. If tumor progression data included more than 1 date, the first date was used. TTP (in months) was calculated as (first event date - randomization date +1)/30.44. (NCT01639001)
Timeframe: Randomization to objective progression, death or last tumor assessment without progression before any additional anti-cancer therapy (whichever occurred first, up to 33 months)

InterventionMonths (Median)
Crizotinib12.0
Chemotherapy6.9

[back to top]

Change From Baseline in General Health Status as Assessed by EuroQol 5D (EQ-5D)-Visual Analog Scale (VAS)

EQ-5D is a standardized, participant-administered measure of health outcome. It provides a descriptive profile for 5 dimensions (mobility, self-care, usual activities, pain/discomfort, anxiety/depression), using 3 levels (no, moderate, or extreme problems) and a visual analog scale (VAS). The VAS component rates current health state on a scale from 0 (worst imaginable health state) to 100 (best imaginable health state); higher scores indicate a better health state. (NCT01639001)
Timeframe: From Baseline to deterioration while on study treatment. For participants with no deterioration, the data was censored at the last date when QLQ-LC13 assessment for pain, dyspnea, or cough was completed (assessed up to 33 months)

InterventionUnits on a scale (Mean)
Crizotinib3.4209
Chemotherapy-0.4927

[back to top]

Change From Baseline in General Health Status as Assessed by EQ-5D-Index

EQ-5D is a standardized, participant-administered measure of health outcome. It provides a descriptive profile for 5 dimensions (mobility, self-care, usual activities, pain/discomfort, anxiety/depression), using 3 levels (no, moderate, or extreme problems) and a visual analog scale (VAS). EQ-5D summary index is obtained with a formula that weights each level of the 5 dimensions. The index-based score is interpreted along a continuum of 0 (death) to 1 (perfect health). (NCT01639001)
Timeframe: From Baseline to deterioration while on study treatment. For participants with no deterioration, the data was censored at the last date when QLQ-LC13 assessment for pain, dyspnea, or cough was completed (assessed up to 33 months)

InterventionUnits on a scale (Mean)
Crizotinib0.0502
Chemotherapy0.0077

[back to top]

Progression-Free Survival (PFS) Based on IRR by Treatment Arm

PFS was defined as the time from the date of randomization to the date of the first documentation of objective tumor progression (by IRR) or death on study due to any cause, whichever occured first. If tumor progression data included more than 1 date, the first date was used. PFS (in months) was calculated as (first event date - randomization date +1)/30.44. Progression is defined using RECIST v1.1, as at least a 20% increase (including an absolute increase of at least 5 millimeters) in the sum of diameters of target lesions, taking as reference the smallest sum on study and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT01639001)
Timeframe: Randomization to objective progression, death or last tumor assessment without progression before any additional anti-cancer therapy (whichever occurred first, assessed up to 33 months)

InterventionMonths (Median)
Crizotinib11.1
Chemotherapy6.8

[back to top]

Duration of Response (DR) Based on IRR

DR was defined as the time from the first documentation of objective tumor response (CR or PR), as determined by the IRR, to the first documentation of objective tumor progression or to death due to any cause, whichever occurred first. If tumor progression data included more than 1 date, the first date was used. DR (in weeks) was calculated as (first date of PD or death - first date of CR or PR +1)/7. DR was only calculated for the subgroup of participants with an objective tumor response. (NCT01639001)
Timeframe: From objective response to date of progression, death or last tumor assessment without progression before any additional anti-cancer therapy (whichever occurred first, up to 33 months)

InterventionWeeks (Median)
Crizotinib44.4
Chemotherapy18.1

[back to top]

Extracranial Time to Progression (EC-TTP) Based on IRR

EC-TTP was defined similarly to TTP, but only considering extracranial disease (excluding intracranial disease) and the progression was determined based on either new extracranial lesions or progression of existing extracranial lesions. (NCT01639001)
Timeframe: Randomization to objective extracranial progression or last tumor assessment without progression before any additional anti-cancer therapy (whichever occurred first, up to 33 months)

InterventionMonths (Median)
Crizotinib18.0
Chemotherapy7.0

[back to top]

Intracranial Time to Progression (IC-TTP) Based on IRR

IC-TTP was defined similarly to TTP, but only considering intracranial disease (excluding extracranial disease) and the progression was determined based on either new brain metastases or progression of existing brain metastases. (NCT01639001)
Timeframe: Randomization to objective intracranial progression or last tumor assessment without progression before any additional anti-cancer therapy (whichever occurred first, up to 33 months)

InterventionMonths (Median)
CrizotinibNA
Chemotherapy16.0

[back to top]

Objective Response Rate (ORR) - Percentage of Participants With Objective Response Based on IRR

Percentage of participants with objective response of complete response (CR) or partial response (PR) according to RECIST version 1.1. CR was defined as complete disappearance of all target lesions and non-target disease, with the exception of nodal disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). No new lesions and disappearance of all non-target lesions. PR was defined as >=30% decrease under baseline of the sum of diameters of all target lesions. The short axis was used in the sum for target nodes, while the longest diameter was used in the sum for all other target lesions. No unequivocal progression of non-target disease. No new lesions. (NCT01639001)
Timeframe: Randomization to objective progression, death or last tumor assessment without progression before any additional anti-cancer therapy (assessed up to 33 months)

InterventionPercentage of participants (Number)
Crizotinib87.5
Chemotherapy45.6

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to the date of death due to any cause. OS (in months) was calculated as (date of death - date of randomization +1)/30.44. For participants who were lost to follow-up or withdrew consent, the OS was censored on the last date that participants were known to be alive. (NCT01639001)
Timeframe: From randomization to death or last date known as alive for those who were lost to follow-up or withdrew consent (assessed up to 64 months).

InterventionMonths (Median)
Crizotinib33.7
Chemotherapy32.9

[back to top]

Percentage of Participants With Disease Control at 12 Weeks Based on IRR

Disease Control Rate (DCR) at 12 weeks is defined as the percent of participants with CR, PR or stable disease (SD) at 12 weeks according to RECIST version 1.1 as determined by the IRR. The best response of SD can be assigned if SD criteria were met at least once after randomization at a minimum interval of 6 weeks. CR was defined as complete disappearance of all target lesions and non-target disease, with the exception of nodal disease. All nodes, both target and non-target, must decrease to normal (short axis <10 mm). No new lesions and disappearance of all non-target lesions. PR was defined as >=30% decrease under baseline of the sum of diameters of all target lesions. The short axis was used in the sum for target nodes, while the longest diameter was used in the sum for all other target lesions. No unequivocal progression of non-target disease. No new lesions. (NCT01639001)
Timeframe: From randomization to Week 12

InterventionPercentage of participants (Number)
Crizotinib82.7
Chemotherapy73.8

[back to top]

Progression Free Survival (PFS)

Compared PFS between the treatment of AUY922 to comparators Pemetrexed or Docetaxel. Progression-free survival (PFS) based on local investigator assessment per RECIST 1.1 was the time from date of randomization/start of treatment to the date of event defined as the first documented progression or death due to any cause. If a patient had not had an event, progression-free survival is censored at the date of last adequate tumor assessment. Progression was defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions (NCT01646125)
Timeframe: 16 months

InterventionMonths (Median)
AUY922 Arm1.5
Chemotherapy Arm2.3

[back to top]

Overall Response Rate (ORR)

ORR was to be compared between treatment arms. The ORR was to be based on local investigator assessment per Response Evaluation Criteria In Solid Tumors Criteria 1.1 (RECIST 1.1). Per this criteria 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.This outcome measure was originally planned to be analyzed up to 24 months. The DMC recommendation at the IA was to stop the study for futility. As a result, collection of all the efficacy assessments was stopped at that time. (NCT01646125)
Timeframe: 16 months

,
InterventionParticipants (Number)
Complete Response (CR)Partial Response (PR)ORR (CR + PR)
AUY922 Arm033
Chemotherapy Arm022

[back to top]

Number of Subjects Reporting Adverse Events

Count of subjects with incidences of adverse events. (NCT01675765)
Timeframe: From first study dose until 28 days after the final dose (an average of 44 weeks)

InterventionParticipants (Count of Participants)
Immunotherapy Plus Chemotherapy38
Immunotherapy With Cyclophosphamide Plus Chemotherapy22

[back to top]

Objective Tumor Response

Objective tumor response was measured using modified Response Evaluation Criteria in Solid Tumors (mRECIST) for assessment of response in malignant pleural mesothelioma (MPM). Per mRECIST for target lesions and assessed by 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, those who fulfilled the criteria for neither PR nor PD; Progressive Disease (PD), >=20% increase in the sum of the longest diameter of target lesions. (NCT01675765)
Timeframe: Baseline to measured disease progression or death (up to 12 months or longer)

,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseNot Assessable
Immunotherapy Plus Chemotherapy1191411
Immunotherapy With Cyclophosphamide Plus Chemotherapy011820

[back to top]

Percentage of Participants Experiencing Treatment-Emergent Adverse Events

(NCT01803282)
Timeframe: Part A: First dose date up to 32 weeks plus 30 days; Part B: First dose date up to 181 weeks plus 30 days

Interventionpercentage of participants (Number)
Part A: ADX 200 mg100.0
Part A: ADX 600 mg100.0
Part A: ADX 1800 mg83.3
Part B: PAC, ADX 800 mg100.0
Part B: LAC, ADX 1200 mg100.0
Part B: LSC, ADX 1200 mg100.0
Part B: EGC, ADX 800 mg100.0
Part B: FL CRC, ADX 800 mg+BEV 5 mg/kg100.0
Part B: FL CRC, ADX 800 mg+BEV 10 mg/kg100.0
Part B: SL CRC, ADX 800 mg+BEV 5 mg/kg100.0
Part B: SL CRC, ADX 800 mg+BEV 10 mg/kg100.0
Part B: BRCA, ADX 800 mg100.0

[back to top]

Percentage of Participants Experiencing Laboratory Abnormalities

Treatment-emergent laboratory abnormalities were defined as values that increase at least one toxicity grade from baseline. Participants with any laboratory abnormality were reported. (NCT01803282)
Timeframe: Part A: First dose date up to 32 weeks plus 30 days; Part B: First dose date up to 181 weeks plus 30 days

,,,,,,,,,,,
Interventionpercentage of participants (Number)
Any Laboratory abnormalities: HematologyAny Laboratory abnormalities: Serum ChemistryAny Laboratory abnormalities: Coagulation
Part A: ADX 1800 mg33.383.333.3
Part A: ADX 200 mg0.0100.025.0
Part A: ADX 600 mg33.366.70.0
Part B: BRCA, ADX 800 mg86.766.740.0
Part B: EGC, ADX 800 mg87.587.535.0
Part B: FL CRC, ADX 800 mg+BEV 10 mg/kg90.981.836.4
Part B: FL CRC, ADX 800 mg+BEV 5 mg/kg84.495.637.8
Part B: LAC, ADX 1200 mg90.060.010.0
Part B: LSC, ADX 1200 mg80.070.010.0
Part B: PAC, ADX 800 mg97.288.938.9
Part B: SL CRC, ADX 800 mg+BEV 10 mg/kg100.087.537.5
Part B: SL CRC, ADX 800 mg+BEV 5 mg/kg95.586.445.5

[back to top]

Cmax,ss

Maximum plasma concentration at steady state (NCT01809210)
Timeframe: Cycle 2 Day1, pre-dose, 0.5, 1, 1.5, 2, 4, 8, 10 hours post dose

Interventionng/mL (Geometric Mean)
Cohort 1 sel50, Gem, Cis476.7
Cohort 2 sel50, Gem, Carb1222
Cohort 3 sel75, Gem, Cis1487
Cohort 4 sel150, Pem, Carb1615
Cohort 5 sel75, Pem, Carb1375
Cohort 6 sel75, Pem, Cis1364
Cohort 7 sel100, Pem, Carb2309

[back to top]

CL/F

Apparent oral plasma clearance (NCT01809210)
Timeframe: Cycle 2 Day1, pre-dose, 0.5, 1, 1.5, 2, 4, 8, 10 hours post dose

InterventionL/h (Mean)
Cohort 1 sel50, Gem, CisNA
Cohort 2 sel50, Gem, Carb14.72
Cohort 3 sel75, Gem, Cis22.64
Cohort 4 sel150, Pem, Carb10.72
Cohort 5 sel75, Pem, Carb18.82
Cohort 6 sel75, Pem, Cis19.08
Cohort 7 sel100, Pem, Carb21.02

[back to top]

Best Percentage Change From Baseline in Target Lesion Size

The best percentage change in tumour size a patient has had during their time in the study up until RECIST progression or last valuable assessment in the absence of RECIST progression. Percentage change was derived at each visit by the percentage change in the sum of the diameters of target lesions (NCT01809210)
Timeframe: Screening, week 6 and week 12

Intervention% change (Mean)
Cohort 1 sel50, Gem, Cis-11.9
Cohort 2 sel50, Gem, Carb-34.6
Cohort 3 sel75, Gem, Cis-41.3
Cohort 4 sel150, Pem, Carb-28.3
Cohort 5 sel75, Pem, Carb-34.7
Cohort 6 sel75, Pem, Cis-24.4
Cohort 7 sel100, Pem, Carb-25.4

[back to top]

Dose Limiting Toxicity (DLT) Events in Chemotherapy in Combination With Selumetinib

Any toxicity not attributable to the disease or disease-related processess under investigation, considered related to the combination of chemotherapy plus selumetinib, which occurs within the timeframe and is dose limiting (NCT01809210)
Timeframe: The first dose on Cycle 1 Day 1 up to the time before dosing on Cycle 2 Day 1, assessed up to 3 weeks

,,,,,,
Interventionparticipants (Number)
Evaluable patientsEvaluable patients with a DLT Event
Cohort 1 sel50, Gem, Cis30
Cohort 2 sel50, Gem, Carb72
Cohort 3 sel75, Gem, Cis41
Cohort 4 sel150, Pem, Carb30
Cohort 5 sel75, Pem, Carb60
Cohort 6 sel75, Pem, Cis121
Cohort 7 sel100, Pem, Carb61

[back to top]

Best Objective Response

The best response a patient has had during their time in the study up until RECIST progression or last valuable assessment in the absence of RECIST progression. Per Response Evaluation Criteria In Solid Tumours (RECIST version 1.1) for target lesions (TL) and 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 nor sufficient increase to qualify for Progressive Disease (PD); Progressive Disease (PD), >=20% increase in the sum of the longest diameter of target lesions, the sum must also demonstrate an absolute increase of >=5mm; Complete Response (CR), disappearance of all target lesions, any pathological lymph nodes selected as TLs must have a reduction in short axis to <10mm (NCT01809210)
Timeframe: Screening, week 6 and week 12

,,,,,,
Interventionparticipants (Number)
Complete responsePartial responseUnconfirmed complete or partial responseStable diseaseRECIST progressionDeathIncomplete post-baseline assessments
Cohort 1 sel50, Gem, Cis0100200
Cohort 2 sel50, Gem, Carb0231003
Cohort 3 sel75, Gem, Cis0202102
Cohort 4 sel150, Pem, Carb0102000
Cohort 5 sel75, Pem, Carb0123000
Cohort 6 sel75, Pem, Cis0227112
Cohort 7 sel100, Pem, Carb0226101

[back to top]

AUC (0-tau)

Area under the concentration time curve (AUC) over a dosing interval at steady state (0-tau) (NCT01809210)
Timeframe: Cycle 2 Day1, pre-dose, 0.5, 1, 1.5, 2, 4, 8, 10 hours post dose

Interventionh*ng/mL (Geometric Mean)
Cohort 1 sel50, Gem, CisNA
Cohort 2 sel50, Gem, Carb3571
Cohort 3 sel75, Gem, Cis3339
Cohort 4 sel150, Pem, Carb4813
Cohort 5 sel75, Pem, Carb4366
Cohort 6 sel75, Pem, Cis4116
Cohort 7 sel100, Pem, Carb5202

[back to top]

Percentage Change From Baseline at 6 Weeks in Target Lesion Size

The percentage change in the sum of the diameters of target lesions (NCT01809210)
Timeframe: Week 6

Intervention% change (Mean)
Cohort 1 sel50, Gem, Cis-7.5
Cohort 2 sel50, Gem, Carb-29.3
Cohort 3 sel75, Gem, Cis-10.4
Cohort 4 sel150, Pem, Carb-14.7
Cohort 5 sel75, Pem, Carb-24.4
Cohort 6 sel75, Pem, Cis-18.9
Cohort 7 sel100, Pem, Carb-18.4

[back to top]

Objective Response Rate (ORR)

The number of patients who had at least 1 confirmed visit response of Complete Response (CR) or Partial Response (PR) prior to any evidence of progression. Per Response Evaluation Criteria In Solid Tumours (RECIST version 1.1) for target lesions (TL) and assessed by MRI or CT: Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Complete Response (CR), disappearance of all target lesions, any pathological lymph nodes selected as TLs must have a reduction in short axis to <10mm; Objective Response Rate (ORR) = CR + PR (NCT01809210)
Timeframe: Up until progression or last evaluable assessment in the absence of progression, up to 9 months

Interventionparticipants (Number)
Cohort 1 sel50, Gem, Cis1
Cohort 2 sel50, Gem, Carb2
Cohort 3 sel75, Gem, Cis2
Cohort 4 sel150, Pem, Carb1
Cohort 5 sel75, Pem, Carb1
Cohort 6 sel75, Pem, Cis2
Cohort 7 sel100, Pem, Carb2

[back to top]

Tmax,ss

Time to reach maximum plasma concentration at steady state (NCT01809210)
Timeframe: Cycle 2 Day1, pre-dose, 0.5, 1, 1.5, 2, 4, 8, 10 hours post dose

Interventionh (Median)
Cohort 1 sel50, Gem, Cis1.00
Cohort 2 sel50, Gem, Carb1.25
Cohort 3 sel75, Gem, Cis1.00
Cohort 4 sel150, Pem, Carb1.00
Cohort 5 sel75, Pem, Carb1.75
Cohort 6 sel75, Pem, Cis1.48
Cohort 7 sel100, Pem, Carb1.50

[back to top]

Progression Free Survival (PFS) by Blinded Independent Review Committee (BIRC)

PFS defined as time from date of randomization to date of first documented disease (as assessed by Blinded Independent Review Committee (BIRC) per RECIST 1.1) or date of death due to any cause (NCT01828099)
Timeframe: from the date of randomization to the date of first radiologically documented disease progression or death due to any cause (assessed every 6 weeks up to approximately 34 months)

Interventionmonths (Median)
Ceritinib16.6
Chemotherapy8.1

[back to top]

Progression Free Survival (PFS) Blinded Independent Review Committee Per Blinded Independent Review Committee (BIRC)

PFS is defined as the time from the date of randomization to the date of the first radiologically documented disease progression or death due to any cause. (NCT01828112)
Timeframe: 'from the date of randomization to the date of first radiologically documented disease progression or death due to any cause up to approximately 24 months

InterventionPercentage of participants (Median)
Ceritinib5.4
Chemotherapy1.6

[back to top]

Number of Participants Who Experienced Dose-limiting Toxicities (DLTs)

"The following toxicities graded per the Common Terminology Criteria for Adverse Events version 4.0 (CTCAE v.4.0) were considered DLTs if judged by the investigator to be related to either study drug:~Grade (G) 4 neutropenia lasting >7 days~Grade 3 and Grade 4 febrile neutropenia~Grade 4 thrombocytopenia (<25,000/mm^3)~Grade 4 anemia~Grade 4 non-hematologic toxicity (not laboratory)~Grade 3 non-hematologic toxicity (not laboratory) lasting >3 days despite optimal supportive care~Any Grade 3 non-hematologic laboratory value if medical intervention is required to treat the participant or the abnormality persists for >7 days.~(Part D only) Missing the second dose of pembrolizumab (Cycle1 Day 22) due to drug-related adverse event" (NCT01840579)
Timeframe: Cycle 1 (first dose and up to 4 weeks in Part A, 3 weeks in Parts B, C, E, and 6 weeks in Part D)

InterventionParticipants (Count of Participants)
Part A: Pembrolizumab 2 mg/kg0
Part A: Pembrolizumab 10 mg/kg0
Part B: Pembrolizumab+Cisplatin/Pemetrexed1
Part B: Pembrolizumab+Carboplatin/Pemetrexed0
Part C: Pembrolizumab+Carboplatin/Paclitaxel2
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel0
Part D: Pembrolizumab+Ipilimumab0
Part E: Pembrolizumab+Cisplatin/Etoposide3
Part E: Pembrolizumab+Carboplatin/Etoposide0
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF0

[back to top]

Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)

An adverse event is defined as any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An adverse event can therefore be any unfavourable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a preexisting condition that is temporally associated with the use of the Sponsor's product, is also an adverse event. (NCT01840579)
Timeframe: Up to approximately 37.9 months

InterventionParticipants (Count of Participants)
Part A: Pembrolizumab 2 mg/kg0
Part A: Pembrolizumab 10 mg/kg0
Part B: Pembrolizumab+Cisplatin/Pemetrexed1
Part B: Pembrolizumab+Carboplatin/Pemetrexed5
Part C: Pembrolizumab+Carboplatin/Paclitaxel4
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel3
Part D: Pembrolizumab+Ipilimumab2
Part E: Pembrolizumab+Cisplatin/Etoposide1
Part E: Pembrolizumab+Carboplatin/Etoposide1
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF0

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 8: Parts A, B, C, and E

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Parts A, B, C and E on Day 1 of Cycle 8 at pre-dose and 0-30 minutes post-dose. Cycle 8 length for Part A was 14 days. Cycle 8 length for Parts B, C, and E was 21 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 8 was only planned for Parts A, B, C, and E. (NCT01840579)
Timeframe: Cycle 8 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg115
Part A: Pembrolizumab 10 mg/kg298
Part B: Pembrolizumab+Cisplatin/Pemetrexed124.47
Part B: Pembrolizumab+Carboplatin/Pemetrexed95.51
Part C: Pembrolizumab+Carboplatin/Paclitaxel122.00
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel66.62
Part E: Pembrolizumab+Cisplatin/Etoposide150.00
Part E: Pembrolizumab+Carboplatin/Etoposide131.78

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 6: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 6 at pre-dose and 0-30 minutes post-dose. Cycle 6 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 6 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 6 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg115
Part A: Pembrolizumab 10 mg/kg286

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 4: Parts A and D

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 4 at pre-dose and 0-30 minutes post-dose and for Part D on Day 22 of Cycle 4 at pre-dose (Day 1 of the following cycle prior to pembrolizumab infusion) and 0-30 minutes post-dose. Cycle 4 length for Part A was 14 days. Cycle 4 length for Part D was 42 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 4 was only planned for Parts A and D. (NCT01840579)
Timeframe: Cycle 4 Day 1 pre- and post-dose (Part A) and Day 22 pre-dose (Day 1 of the following cycle prior to pembrolizumab infusion) and post-dose (Part D)

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg93.0
Part A: Pembrolizumab 10 mg/kg367
Part D: Pembrolizumab+Ipilimumab86.21

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 18: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 18 at pre-dose and 0-30 minutes post-dose. Cycle 18 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 18 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 18 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg329

[back to top]

Number of Participants Who Experienced at Least One Adverse Event (AE)

An adverse event is defined as any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An adverse event can therefore be any unfavourable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a preexisting condition that is temporally associated with the use of the Sponsor's product, is also an adverse event. (NCT01840579)
Timeframe: Up to approximately 51.3 months

InterventionParticipants (Count of Participants)
Part A: Pembrolizumab 2 mg/kg3
Part A: Pembrolizumab 10 mg/kg7
Part B: Pembrolizumab+Cisplatin/Pemetrexed6
Part B: Pembrolizumab+Carboplatin/Pemetrexed6
Part C: Pembrolizumab+Carboplatin/Paclitaxel8
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel6
Part D: Pembrolizumab+Ipilimumab5
Part E: Pembrolizumab+Cisplatin/Etoposide6
Part E: Pembrolizumab+Carboplatin/Etoposide6
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF3

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 2: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 2 at pre-dose and 0-30 minutes post-dose. Cycle 2 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 2 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 2 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg82.7
Part A: Pembrolizumab 10 mg/kg322

[back to top]

Volume of Distribution (Vz) of Pembrolizumab Over Time for Part A Cycle 1

Vz was the volume of distribution during the terminal phase. Blood sampling was taken at the following timepoints: Part A Cycle 1 Day 1 at pre-dose, 0-30 minutes post-dose, and at 6, 24, 48, 168, 336, 504 hours after completion of pembrolizumab infusion and Day 1 of Cycle 2 prior to pembrolizumab infusion. Cycle 1 length A was 28 days. Vz is reported as geometric mean with a percent coefficient of variation. Per protocol, analysis for Vz was only planned for Part A Cycle 1. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Part A (Up to approximately 28 days)

InterventionmL/kg (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg65.3
Part A: Pembrolizumab 10 mg/kg76.5

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 9: Part A

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 9 Ctrough was taken for Part A on Day 1 of Cycle 10 at pre-dose (prior to Cycle 10 infusion). Cycle 9 length for Part A was 14 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 9 Ctrough was only planned for Part A. (NCT01840579)
Timeframe: Cycle 10 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg68.4
Part A: Pembrolizumab 10 mg/kg125

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 8: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 8 Ctrough was taken for Part D on Day 22 of Cycle 8 at pre-dose (prior to Cycle 9 infusion). Cycle 8 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 8 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 8 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab35.20

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 7: Parts A, B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 7 Ctrough was taken for Parts A, B, C, and E on Day 1 of Cycle 8 at pre-dose (prior to Cycle 8 infusion). Cycle 7 length for Part A was 14 days. Cycle 7 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 7 Ctrough was only planned for Parts A, B, C, and E. (NCT01840579)
Timeframe: Cycle 8 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg54.9
Part A: Pembrolizumab 10 mg/kg134
Part B: Pembrolizumab+Cisplatin/Pemetrexed39.33
Part B: Pembrolizumab+Carboplatin/Pemetrexed37.44
Part C: Pembrolizumab+Carboplatin/Paclitaxel42.70
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel32.44
Part E: Pembrolizumab+Cisplatin/Etoposide54.0
Part E: Pembrolizumab+Carboplatin/Etoposide48.92
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF43.10

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 6: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 6 Ctrough was taken for Part D on Day 22 of Cycle 6 at pre-dose (prior to Cycle 7 infusion). Cycle 6 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 6 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 6 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab40.10

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 5: Parts A, B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 5 Ctrough was taken for Parts A, B, C, and E on Day 1 of Cycle 6 at pre-dose (prior to Cycle 6 infusion). Cycle 5 length for Part A was 14 days. Cycle 5 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 5 Ctrough was only planned for Parts A, B, C, and E. (NCT01840579)
Timeframe: Cycle 6 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg46.5
Part A: Pembrolizumab 10 mg/kg38.3
Part B: Pembrolizumab+Cisplatin/Pemetrexed37.70
Part B: Pembrolizumab+Carboplatin/Pemetrexed23.84
Part C: Pembrolizumab+Carboplatin/Paclitaxel35.33
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel33.91
Part E: Pembrolizumab+Cisplatin/Etoposide47.82
Part E: Pembrolizumab+Carboplatin/Etoposide38.63
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF46.47

[back to top]

Area Under the Concentration Time Curve From 0-28 Days (AUC 0-28) for Part A Cycle 1

AUC 0-28 was the AUC of pembrolizumab from time zero to 28 days after dosing. Blood sampling was taken at the following timepoints: Part A Cycle 1 Day 1 at pre-dose, 0-30 minutes post-dose, and at 6, 24, 48, 168, 336, 504 hours after completion of pembrolizumab infusion and Day 1 of Cycle 2 pre-dose. Cycle 1 length was 28 days. AUC 0-28 is reported as geometric mean with a percent coefficient of variation. Per protocol, analysis for AUC 0-28 was only planned for Part A Cycle 1. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Part A (Up to approximately 28 days)

Interventionµg•day/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg507
Part A: Pembrolizumab 10 mg/kg2219

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 4: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 4 Ctrough was taken for Part D on Day 22 of Cycle 4 at pre-dose (prior to Cycle 5 infusion). Cycle 4 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 4 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 4 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab28.08

[back to top]

Clearance (CL) of Pembrolizumab Over Time for Part A Cycle 1

CL was the volume of plasma from which pembrolizumab was eliminated per unit time. Blood sampling was taken at the following timepoints: Part A Cycle 1 Day 1 at pre-dose, 0-30 minutes post-dose, and at 6, 24, 48, 168, 336, 504 hours after completion of pembrolizumab infusion and Day 1 of Cycle 2 prior to pembrolizumab infusion. Cycle 1 length A was 28 days. CL is reported as geometric mean with a percent coefficient of variation. Per protocol, analysis for CL was only planned for Part A Cycle 1. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Part A (Up to approximately 28 days)

InterventionmL/day/kg (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg2.46
Part A: Pembrolizumab 10 mg/kg2.93

[back to top]

Area Under the Concentration Time Curve From 0-Infinity (AUC 0-inf) for Part A Cycle 1

AUC 0-inf was the AUC of pembrolizumab from time zero to infinity after dosing. Blood sampling was taken at the following timepoints: Part A Cycle 1 Day 1 at pre-dose, 0-30 minutes post-dose, and at 6, 24, 48, 168, 336, 504 hours after completion of pembrolizumab infusion and Day 1 of Cycle 2 pre-dose. Cycle 1 length was 28 days. AUC 0-inf is reported as geometric mean with a percent coefficient of variation. Per protocol, analysis for AUC 0-inf was only planned for Part A Cycle 1. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Part A (Up to approximately 28 days)

Interventionµg•day/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg812
Part A: Pembrolizumab 10 mg/kg3410

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 1: Parts A, B, C, D, and E

Cmax was the maximum observed concentration of pembrolizumab in serum. For Part A, samples were collected on Day 1 at pre-dose, post-dose (to +30 min), 6 hour (hr) (±30 min), and 24hr; Days 2, 3, 8, 15, and 22 (±2 hr for Day 2 to Day 22) after completion of infusion. For Parts B, C, and E, samples were collected on Day 1 at pre-dose, post-dose (to +30 min), and 24hr; Day 5 (96hr was preferred, 72hr or 120hr were also acceptable, ±2 hr), and Day 15 (±24 hr) after completion of infusion. For Part D, samples were collected on Day 1 at pre-dose, post-dose (to +30 min), and 24hr; Day 5 (96hr was preferred, 72hr or 120hr were also acceptable, ±2 hr), Day 15, and Day 22 (±24 hr) after completion of infusion. Cycle 1 length for Part A was 28 days. Cycle 1 length for Parts B, C, and E was 21 days. Cycle 1 length for Part D was 42 days. Cmax is reported as geometric mean with a percent coefficient of variation. Per protocol, analysis for Cmax was planned for Parts A, B, C, D, and E. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Parts A, B, C, D, and E (up to approximately 22 days)

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg47.4
Part A: Pembrolizumab 10 mg/kg250
Part B: Pembrolizumab+Cisplatin/Pemetrexed52.51
Part B: Pembrolizumab+Carboplatin/Pemetrexed47.46
Part C: Pembrolizumab+Carboplatin/Paclitaxel56.39
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel53.79
Part D: Pembrolizumab+Ipilimumab72.71
Part E: Pembrolizumab+Cisplatin/Etoposide74.33
Part E: Pembrolizumab+Carboplatin/Etoposide84.55
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF88.65

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 10: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 10 at pre-dose and 0-30 minutes post-dose. Cycle 10 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 10 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 10 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg133
Part A: Pembrolizumab 10 mg/kg266

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 12: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 12 at pre-dose and 0-30 minutes post-dose. Cycle 12 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 12 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 12 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg357

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 14: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 14 at pre-dose and 0-30 minutes post-dose. Cycle 14 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 14 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 14 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg335

[back to top]

Maximum Serum Concentration (Cmax) of Pembrolizumab for Cycle 16: Part A

Cmax was the maximum observed concentration of pembrolizumab in serum. Blood sampling was taken for Part A on Day 1 of Cycle 16 at pre-dose and 0-30 minutes post-dose. Cycle 16 length for Part A was 14 days. Cmax is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cmax in Cycle 16 was only planned for Part A. (NCT01840579)
Timeframe: Cycle 16 Day 1 pre- and post-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg348

[back to top]

Time to Maximum Serum Concentration (Tmax) of Pembrolizumab for Cycle 8: Parts B, C, and E

Tmax was the time required to reach the maximum concentration of pembrolizumab in serum. Blood sampling was taken for Parts B, C, and E on Day 1 of Cycle 8 at pre-dose and 0-30 minutes post-dose. Cycle 8 for Parts B, C, and E was 21 days. Tmax is reported as median and full range. Per protocol, analysis for Tmax in Cycle 8was planned for Parts B, C, and E. (NCT01840579)
Timeframe: Cycle 8 Day 1 pre- and post-dose

Interventiondays (Median)
Part B: Pembrolizumab+Cisplatin/Pemetrexed0.024
Part B: Pembrolizumab+Carboplatin/Pemetrexed0.026
Part C: Pembrolizumab+Carboplatin/Paclitaxel0.028
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel0.027
Part E: Pembrolizumab+Cisplatin/Etoposide0.029
Part E: Pembrolizumab+Carboplatin/Etoposide0.026

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 3: Parts A, B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 3 Ctrough was taken for Parts A, B, C, and E on Day 1 of Cycle 4 at pre-dose (prior to Cycle 4 infusion). Cycle 3 length for Part A was 14 days. Cycle 3 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). (NCT01840579)
Timeframe: Cycle 4 Day 1 at Pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg37.2
Part A: Pembrolizumab 10 mg/kg83.0
Part B: Pembrolizumab+Cisplatin/Pemetrexed32.71
Part B: Pembrolizumab+Carboplatin/Pemetrexed25.38
Part C: Pembrolizumab+Carboplatin/Paclitaxel24.80
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel23.63
Part E: Pembrolizumab+Cisplatin/Etoposide32.20
Part E: Pembrolizumab+Carboplatin/Etoposide30.43
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF34.31

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 3: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 3 Ctrough was taken for Part D on Day 22 of Cycle 3 at pre-dose (prior to Cycle 4 infusion). Cycle 3 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). (NCT01840579)
Timeframe: Cycle 3 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab30.30

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 27: Parts B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling was taken for Parts B, C, and E on Day 1 of Cycle 28 at pre-dose (prior to Cycle 28 infusion). Cycle 27 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 27 Ctrough was only planned for Parts B, C, and E. (NCT01840579)
Timeframe: Cycle 28 Day 1 Pre-dose

Interventionµg/mL (Geometric Mean)
Part B: Pembrolizumab+Cisplatin/Pemetrexed53.90
Part B: Pembrolizumab+Carboplatin/Pemetrexed68.20

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 23: Parts B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling was taken for Parts B, C, and E on Day 1 of Cycle 24 at pre-dose (prior to Cycle 24 infusion). Cycle 23 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 23 Ctrough was only planned for Parts B, C, and E. (NCT01840579)
Timeframe: Cycle 24 Day 1 Pre-dose

Interventionµg/mL (Geometric Mean)
Part B: Pembrolizumab+Cisplatin/Pemetrexed42.40
Part B: Pembrolizumab+Carboplatin/Pemetrexed51.72

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 2: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 2 Ctrough was taken for Part D on Day 22 of Cycle 2 at pre-dose (prior to Cycle 3 infusion). Cycle 2 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 2 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 2 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab20.10

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 19: Parts B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling was taken for Parts B, C, and E on Day 1 of Cycle 20 at pre-dose (prior to Cycle 20 infusion). Cycle 19 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 19 Ctrough was only planned for Parts B, C, and E. (NCT01840579)
Timeframe: Cycle 20 Day 1 Pre-dose

Interventionµg/mL (Geometric Mean)
Part B: Pembrolizumab+Cisplatin/Pemetrexed48.50
Part B: Pembrolizumab+Carboplatin/Pemetrexed50.98

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 17: Part A

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling was taken for Part A on Day 1 of Cycle 18 at pre-dose (prior to Cycle 18 infusion). Cycle 17 length for Part A was 14 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 17 Ctrough was planned for Part A. (NCT01840579)
Timeframe: Cycle 18 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg125

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 16: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 16 Ctrough was taken for Part D on Day 22 of Cycle 16 at pre-dose (prior to Cycle 17 infusion). Cycle 16 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 16 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 16 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab30.70

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 15: Parts A, B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling was taken for Parts A, B, C, and E on Day 1 of Cycle 16 at pre-dose (prior to Cycle 16 infusion). Cycle 15 length for Part A was 14 days. Cycle 15 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 15 Ctrough was only planned for Parts A, B, C, and E. (NCT01840579)
Timeframe: Cycle 16 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg199
Part B: Pembrolizumab+Cisplatin/Pemetrexed47.50
Part B: Pembrolizumab+Carboplatin/Pemetrexed56.27
Part E: Pembrolizumab+Cisplatin/Etoposide52.50

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 14: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 14 Ctrough was taken for Part D on Day 22 of Cycle 14 at pre-dose (prior to Cycle 15 infusion). Cycle 14 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 14 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 14 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab28.40

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 13: Part A

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 13 Ctrough was taken for Part A on Day 1 of Cycle 14 at pre-dose (prior to Cycle 14 infusion). Cycle 13 length for Part A was 14 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 13 Ctrough was only planned for Part A. (NCT01840579)
Timeframe: Cycle 14 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg147

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 12: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 12 Ctrough was taken for Part D on Day 22 of Cycle 12 at pre-dose (prior to Cycle 13 infusion). Cycle 12 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 12 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 12 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab30.90

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 11: Parts A, B, C, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 11 Ctrough was taken for Parts A, B, C, and E on Day 1 of Cycle 12 at pre-dose (prior to Cycle 12 infusion). Cycle 11 length for Part A was 14 days. Cycle 11 length for Parts B, C, and E was 21 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 11 Ctrough was only planned for Parts A, B, C, and E. (NCT01840579)
Timeframe: Cycle 12 Day 1 Pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 10 mg/kg159
Part B: Pembrolizumab+Cisplatin/Pemetrexed39.30
Part B: Pembrolizumab+Carboplatin/Pemetrexed45.00
Part C: Pembrolizumab+Carboplatin/Paclitaxel32.60
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel57.80
Part E: Pembrolizumab+Cisplatin/Etoposide52.20

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 10: Part D

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 10 Ctrough was taken for Part D on Day 22 of Cycle 10 at pre-dose (prior to Cycle 11 infusion). Cycle 10 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 10 Ctrough was only planned for Part D. (NCT01840579)
Timeframe: Cycle 10 Day 22 pre-dose

Interventionµg/mL (Geometric Mean)
Part D: Pembrolizumab+Ipilimumab36.30

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab for Cycle 1: Parts A, B, C, D, and E

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose. Blood sampling for Cycle 1 Ctrough was taken for Parts A, B, C, D, and E on Day 1 of Cycle 2 at pre-dose (prior to Cycle 2 infusion). Cycle 1 length for Part A was 28 days. Cycle 1 length for Parts B, C, and E was 21 days. Cycle 1 length for Part D was 42 days. Ctrough is reported as geometric mean with a percent coefficient of variation (%CV). Per protocol, analysis for Cycle 1 Ctrough was planned for Parts A, B, C, D, and E. (NCT01840579)
Timeframe: Cycle 2 Day 1 pre-dose

Interventionµg/mL (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg11.2
Part A: Pembrolizumab 10 mg/kg47.9
Part B: Pembrolizumab+Cisplatin/Pemetrexed19.88
Part B: Pembrolizumab+Carboplatin/Pemetrexed15.79
Part C: Pembrolizumab+Carboplatin/Paclitaxel9.45
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel10.96
Part D: Pembrolizumab+Ipilimumab8.26
Part E: Pembrolizumab+Cisplatin/Etoposide14.57
Part E: Pembrolizumab+Carboplatin/Etoposide15.30
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF17.88

[back to top]

Time to Maximum Serum Concentration (Tmax) of Pembrolizumab for Cycle 4: Part D

Tmax was the time required to reach the maximum concentration of pembrolizumab in serum. Blood sampling was taken for Part D on Day 22 of Cycle 4 at pre-dose (Day 1 of the following cycle prior to pembrolizumab infusion) and 0-30 minutes post-dose. Cycle length for Part D was 42 days. Tmax is reported as median and full range. Per protocol, analysis for Tmax in Cycle 4 was only planned for Part D. (NCT01840579)
Timeframe: Cycle 4 Day 22 pre-dose (Day 1 of the following cycle prior to pembrolizumab infusion) and post-dose

Interventiondays (Median)
Part D: Pembrolizumab+Ipilimumab0.026

[back to top]

Time to Maximum Serum Concentration (Tmax) of Pembrolizumab for Cycle 1: Parts A, B, C, D, and E

Tmax was the time required to reach the maximum concentration of pembrolizumab in serum. For Part A, samples were collected on Day 1 at pre-dose, post-dose (to +30 min), 6 hour (hr) (±30 min), and 24hr; Days 2, 3, 8, 15, and 22 (±2 hr for Day 2 to Day 22) after completion of infusion. For Parts B, C, and E, samples were collected on Day 1 at pre-dose, post-dose (to +30 min), and 24hr; Day 5 (96hr was preferred, 72hr or 120hr were also acceptable, ±2 hr), and Day 15 (±24 hr) after completion of infusion. For Part D, samples were collected on Day 1 at pre-dose, post-dose (to +30 min), and 24hr; Day 5 (96hr was preferred, 72hr or 120hr were also acceptable, ±2 hr), Day 15, and Day 22 (±24 hr) after completion of infusion. Cycle 1 length for Part A was 28 days. Cycle 1 length for Parts B, C, and E was 21 days. Cycle 1 length for Part D was 42 days. Tmax is reported as median and full range. Per protocol, analysis for Tmax in Cycle 1 was planned for Parts A, B, C, D, and E. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Parts A, B, C, D, and E (up to approximately 22 days)

Interventiondays (Median)
Part A: Pembrolizumab 2 mg/kg0.223
Part A: Pembrolizumab 10 mg/kg0.00903
Part B: Pembrolizumab+Cisplatin/Pemetrexed1.98
Part B: Pembrolizumab+Carboplatin/Pemetrexed2.52
Part C: Pembrolizumab+Carboplatin/Paclitaxel0.028
Part C: Pembrolizumab+Carboplatin/Nab-paclitaxel0.027
Part D: Pembrolizumab+Ipilimumab0.026
Part E: Pembrolizumab+Cisplatin/Etoposide0.026
Part E: Pembrolizumab+Carboplatin/Etoposide0.028
Part E: Pembrolizumab+Cisplatin/Etoposide+G-CSF0.031

[back to top]

Terminal Half-Life (t1/2) of Pembrolizumab Over Time for Part A Cycle 1

t1/2 was the time required to divide the pembrolizumab concentration by two after reaching pseudo-equilibrium. Blood sampling was taken at the following timepoints: Part A Cycle 1 Day 1 at pre-dose, 0-30 minutes post-dose, and at 6, 24, 48, 168, 336, 504 hours after completion of pembrolizumab infusion and pre-dose Day 1 of the following cycle prior to pembrolizumab infusion. Cycle 1 length A was 28 days. t1/2 is reported as geometric mean with a percent coefficient of variation. Per protocol, analysis for t1/2 was only planned for Part A Cycle 1. (NCT01840579)
Timeframe: At designated timepoints in Cycle 1 for Part A (Up to approximately 28 days)

Interventiondays (Geometric Mean)
Part A: Pembrolizumab 2 mg/kg18.4
Part A: Pembrolizumab 10 mg/kg18.1

[back to top]

Number of Participants With Non-serious Adverse Events (AEs) and Serious AEs (SAEs)

An AE is any untoward medical occurrence in a clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. Any untoward event resulting in death, life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, congenital anomaly/birth defect, protocol-specific events including drug-induced liver injury with hyperbilirubinaemia, any new primary cancers, cardiac toxicity including Left Ventricular Ejection Fraction (LVEF) changes or treatment emergent cardiac valve toxicity and treatment emergent acute anterior uveitis were categorized as SAE. Participants having non-serious AE or SAE were included in the analysis. The All Treated Subjects Population comprised of all participants who received at least one dose of study treatment. (NCT01868022)
Timeframe: Median of 28.5 weeks

,,,,,,,,
InterventionParticipants (Count of Participants)
Non-serious AEsSAEs
10 mg/kg GSK3052230 + Docetaxel: Arm B33
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A33
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C30
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C245
20 mg/kg GSK3052230 + Docetaxel: Arm B31
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A145
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C84
5 mg/kg GSK3052230 + Docetaxel: Arm B32
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A30

[back to top]

Number of Participants With the Abnormal Urinalysis Findings

Urinalysis parameters included urine protein, urine glucose, urine ketones and occult blood were assessed. Dipstick test was performed for routine urinalysis. Abnormal values such as trace, 1+, 2+, 3+, 4+, >1000, >=1000, and >10 have been reported. (NCT01868022)
Timeframe: Up to Cycle 16 (each cycle was of 21 days)

,,,,,,,,
InterventionParticipants (Count of Participants)
Cycle 1 Day 1, Urine glucose, >1000Cycle 1 Day 1, Urine glucose, 2+Cycle 1 Day 1, Urine glucose, traceCycle 2 Day 1, Urine glucose, traceCycle 4 Day 1, Urine glucose, >=1000Cycle 4 Day 1, Urine glucose, 4+Cycle 1 Day 1, Urine ketones, traceCycle 4 Day 1, Urine ketones, traceCycle 28, Day 1, Urine ketones, traceCycle 1 Day 1, Occult Blood, 1+Cycle 1 Day 1, Occult Blood, 3+Cycle 2 Day 1, Occult Blood, 1+Cycle 2 Day 1, Occult Blood, traceCycle 4 Day 1, Occult Blood, traceCycle 4 Day 1, Occult Blood, 1+Cycle 4 Day 1, Occult Blood, 2+Cycle 8 Day 1, Occult Blood, 1+Cycle 1 Day 1, Urine protein, 1+Cycle 1 Day 1, Urine protein, traceCycle 2, Day 1, Urine protein, traceCycle 2, Day 1, Urine protein, 2+Cycle 4, Day 1, Urine protein, traceCycle 8, Day 1, Urine protein, traceCycle 12, Day 1, Urine protein, traceCycle 16, Day 1, Urine protein, trace
10 mg/kg GSK3052230 + Docetaxel: Arm B0000000000000000001100000
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0000000000000000001000001
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0000000000000000000000000
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0000001000110001101002310
20 mg/kg GSK3052230 + Docetaxel: Arm B0000000000000000000000000
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A1000101102000000012100000
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0011012011011100020111000
5 mg/kg GSK3052230 + Docetaxel: Arm B0100000000000000000000000
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0000000000000100000000000

[back to top]

Number of Participants With Clinically Significant Findings for 12-lead Electrocardiogram (ECG)

A single 12-lead ECG was performed at the specified timepoints during the study where the participant was instructed to be in semi-recumbent position for 5 minutes before obtaining the ECG. An ECG machine that automatically calculated the heart rate and measures like the PR, QRS, QT, and corrected QT intervals. Number of participants with worst-case post-Baseline abnormal clinically significant findings and abnormal not clinically significant findings in ECG results has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

,,,,,,,,
InterventionParticipants (Count of Participants)
Abnormal not clinically significantAbnormal clinically significant
10 mg/kg GSK3052230 + Docetaxel: Arm B20
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A20
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C10
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C123
20 mg/kg GSK3052230 + Docetaxel: Arm B30
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A81
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C23
5 mg/kg GSK3052230 + Docetaxel: Arm B20
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A00

[back to top]

Change From Baseline in Heart Rate

Heart rate was measured in a semi-supine position after 5 minutes of rest. Heart rate was measured before start of first chemotherapy infusion and within 20 minutes before start of GSK3052230 infusion on Day 1 of every cycle and Baseline. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. The worst-case post-Baseline values has been presented. (NCT01868022)
Timeframe: Baseline and up to Median of 28.5 weeks

InterventionBeats per minute (bpm) (Mean)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A-0.3
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A3.2
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A8.0
5 mg/kg GSK3052230 + Docetaxel: Arm B28.0
10 mg/kg GSK3052230 + Docetaxel: Arm B10.5
20 mg/kg GSK3052230 + Docetaxel: Arm B0.6
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C-5.0
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C2.8
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C24.4

[back to top]

Change From Baseline in Temperature

Temperature was measured in a semi-supine position after 5 minutes of rest. Temperature was measured before start of first chemotherapy infusion and within 20 minutes before start of GSK3052230 infusion on Day 1 of every cycle and Baseline. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. The worst-case post-Baseline values has been presented. (NCT01868022)
Timeframe: Baseline and up to Median of 28.5 weeks

InterventionDegree Celsius (Mean)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A-0.30
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A-0.38
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0.35
5 mg/kg GSK3052230 + Docetaxel: Arm B0.10
10 mg/kg GSK3052230 + Docetaxel: Arm B1.03
20 mg/kg GSK3052230 + Docetaxel: Arm B-0.03
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C-0.23
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C-0.16
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0.12

[back to top]

Number of Participants With Dose Delays

Dose reduction and delays were done due to toxicity, or in the interest of participant's safety per investigator discretion. Requirement for more than 2 dose reductions resulted in permanent discontinuation of chemotherapy. Participants with dose delay has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionParticipants (Count of Participants)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A2
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A4
5 mg/kg GSK3052230 + Docetaxel: Arm B2
10 mg/kg GSK3052230 + Docetaxel: Arm B1
20 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C2
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C12
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C1

[back to top]

Number of Participants With Dose Reduction

Dose reduction and delays were done due to toxicity, or in the interest of participant's safety per investigator discretion. Requirement for more than 2 dose reductions resulted in permanent discontinuation of chemotherapy. Participants with dose reduction has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionParticipants (Count of Participants)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A1
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A3
5 mg/kg GSK3052230 + Docetaxel: Arm B1
10 mg/kg GSK3052230 + Docetaxel: Arm B0
20 mg/kg GSK3052230 + Docetaxel: Arm B2
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C2
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0

[back to top]

Number of Participants With Dose-Limiting Toxicities (DLT)

DLT is defined as toxicities due to GSK3052230 or due to the combination of GSK3052230 with chemotherapy within Cycle 1 (first 21 days of period on study) that are unlikely to be due to another cause, such as the known effects of cytotoxics chemotherapy alone, disease progression, or accident, and protocol-specified criteria. Clinically significant toxicities that persist or occur beyond Cycle 1 that the investigator and GlaxoSmithKline (GSK) medical monitor consider dose-limiting may also be designated a DLT for the purpose of establishing Maximum tolerated dose (MTD). Number of participants with DLTs has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionParticipants (Count of Participants)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
5 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Docetaxel: Arm B0
20 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C1
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C3

[back to top]

Number of Participants With Maximum Tolerated Dose (MTD) or Maximum Feasible Dose (MFD)

The MTD is defined as the highest dose level tested at which < 33 percent of participants experience a DLT. In cases when MTD is not reached dose was described as the MFD. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionParticipants (Count of Participants)
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A14
20 mg/kg GSK3052230 + Docetaxel: Arm B3
15 mg/kg GSK3052230 + Pemetrexed + Carboplatin: Arm C25

[back to top]

Number of Participants With Overall Response Rate (ORR)

Overall Response Rate (ORR) is defined as the percentage of participants achieving a confirmed Complete response (CR) or Partial response (PR) from the start of treatment until disease progression as per RECIST version 1.1 or modified RECIST for participants in Arm C. This was determined based on Investigator assessments of response. 95% confidence intervals (CI) are calculated based on the unconditional exact method. ORR as per RECIST vesrion 1.1 for Arm A and B has been reported. ORR as per RECIST version 1.1 and modified RECIST version 1.1 for Arm C has been reported. The study population used for decision-making at the interim analyses during the dose expansion cohorts of the study arms is termed as the All Evaluable Participants Population. NA indicates 0 participants met ORR criteria therefore no dispersion. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionParticipants (Count of Participants)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A1
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A2
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A6
5 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Docetaxel: Arm B0
20 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C2
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C11
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C1

[back to top]

Number of Participants Withdrew Due to AEs

An AE can be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. The AEs leading to permanent discontinuation from the study has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionParticipants (Count of Participants)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0
5 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Docetaxel: Arm B1
20 mg/kg GSK3052230 + Docetaxel: Arm B0
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C2
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C1
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C3

[back to top]

Progression Free Survival (PFS) as Assessed by Investigator

PFS is defined as the interval between first dose of GSK3052230 and the earliest date of disease progression or death due to any cause by investigator assessment per RECIST 1.1 (for Arm A and B participants) or modified RECIST (for Arm C participants). For participants who do not progress or die, PFS was censored at the time of last radiological scan. Participants who discontinued study with no post-treatment tumor assessment were censored at date of first dose of study drug. Mean and 95 percent CI has been reported. NA indicates that data were not available as only 1 participant had event, other two censored therefore there is no confidence interval. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionMonths (Median)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A4.1
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm ANA
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A5.5
5 mg/kg GSK3052230 + Docetaxel: Arm B4.6
10 mg/kg GSK3052230 + Docetaxel: Arm B9.5
20 mg/kg GSK3052230 + Docetaxel: Arm B5.1
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C4.6
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C7.4
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C4.1

[back to top]

Treatment Duration With GSK3052230

The number of participants administered study treatment were summarized according to the duration of therapy. The extent of treatment exposure is calculated as the number of cycles administered. The duration of exposure to study treatment is calculated from first day to last day of treatment plus 1 day. Median and full range (minimum and maximum) has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

InterventionCycles (Median)
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A7.0
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A15.0
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A8.0
5 mg/kg GSK3052230 + Docetaxel: Arm B6.0
10 mg/kg GSK3052230 + Docetaxel: Arm B4.0
20 mg/kg GSK3052230 + Docetaxel: Arm B6.0
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C6.0
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C11.0
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C3.0

[back to top]

Change From Baseline in Forced Vital Capacity (FVC) in of Arm C Participants With Malignant Pleural Mesothelioma (MPM)

FVC is the total amount of air exhaled during the Forced Expiratory Volume test. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Assessment of FVC was done on Day 1 of every odd cycle for Arm C participants with MPM. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). NA indicates that data were not available as standard deviation could not be calculated for a single participant. (NCT01868022)
Timeframe: Up to 31 cycles (each cycle was of 21 days)

InterventionLiters (Mean)
Cycle 3,Day 1, n=3, 24, 4Cycle 4,Day 1, n=1, 4, 1Cycle 5,Day 1, n=1, 23, 3Cycle 6,Day 1, n=1, 5, 2Cycle 7,Day 1, n=1, 15, 2Cycle 8,Day 1, n=0, 3, 1Cycle 9,Day 1, n=0, 15, 1Cycle 10,Day 1, n=0, 3, 0Cycle 11,Day 1, n=0, 12, 2Cycle 12,Day 1, n=0, 2, 1Cycle 13,Day 1, n=0, 8, 2Cycle 14,Day 1, n=0, 3, 1Cycle 15,Day 1, n=0, 6, 1Cycle 17,Day 1, n=0, 4, 1Cycle 19,Day 1, n=0, 1, 1Cycle 21,Day 1, n=0, 1, 1Cycle 23,Day 1, n=0, 1, 1
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0.2800.4130.2380.4640.3990.2630.3530.0630.3470.2250.389-0.2070.2470.158-0.02-0.27-0.62

[back to top]

Change From Baseline in Forced Vital Capacity (FVC) in of Arm C Participants With Malignant Pleural Mesothelioma (MPM)

FVC is the total amount of air exhaled during the Forced Expiratory Volume test. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Assessment of FVC was done on Day 1 of every odd cycle for Arm C participants with MPM. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). NA indicates that data were not available as standard deviation could not be calculated for a single participant. (NCT01868022)
Timeframe: Up to 31 cycles (each cycle was of 21 days)

InterventionLiters (Mean)
Cycle 3,Day 1, n=3, 24, 4Cycle 4,Day 1, n=1, 4, 1Cycle 5,Day 1, n=1, 23, 3Cycle 6,Day 1, n=1, 5, 2Cycle 7,Day 1, n=1, 15, 2Cycle 8,Day 1, n=0, 3, 1Cycle 9,Day 1, n=0, 15, 1Cycle 11,Day 1, n=0, 12, 2Cycle 12,Day 1, n=0, 2, 1Cycle 13,Day 1, n=0, 8, 2Cycle 14,Day 1, n=0, 3, 1Cycle 15,Day 1, n=0, 6, 1Cycle 17,Day 1, n=0, 4, 1Cycle 19,Day 1, n=0, 1, 1Cycle 21,Day 1, n=0, 1, 1Cycle 23,Day 1, n=0, 1, 1Cycle 25,Day 1, n=0, 0, 1Cycle 27,Day 1, n=0, 0, 1Cycle 31,Day 1, n=0, 0, 1
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0.1380.6000.5870.1100.5750.6401.1300.5100.5800.5550.910-0.0300.5600.6200.5600.5100.4300.2200.270

[back to top]

Change From Baseline in Forced Vital Capacity (FVC) in of Arm C Participants With Malignant Pleural Mesothelioma (MPM)

FVC is the total amount of air exhaled during the Forced Expiratory Volume test. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Assessment of FVC was done on Day 1 of every odd cycle for Arm C participants with MPM. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). NA indicates that data were not available as standard deviation could not be calculated for a single participant. (NCT01868022)
Timeframe: Up to 31 cycles (each cycle was of 21 days)

InterventionLiters (Mean)
Cycle 3,Day 1, n=3, 24, 4Cycle 4,Day 1, n=1, 4, 1Cycle 5,Day 1, n=1, 23, 3Cycle 6,Day 1, n=1, 5, 2Cycle 7,Day 1, n=1, 15, 2
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0.303-0.3400.2400.3100.530

[back to top]

Change From Baseline in Systolic Blood Pressure (SBP) and Diastolic Blood Pressure (DBP)

Blood pressure was measured in a semi-supine position after 5 minutes of rest. Blood pressure was measured before start of first chemotherapy infusion and within 20 minutes before start of GSK3052230 infusion on Day 1 of every cycle and Baseline. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. The worst-case post-Baseline values has been presented. NA indicates that data were not available as standard deviation could not be calculated for a single participant. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01868022)
Timeframe: Baseline and up to Median of 28.5 weeks

,,,,,,,,
InterventionMillimeters of mercury (mmHg) (Mean)
DBP, n=3, 2, 9, 3, 2, 2, 3, 21, 5SBP, n=3, 1, 13, 2, 2, 2, 3, 21, 4
10 mg/kg GSK3052230 + Docetaxel: Arm B6.017.5
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A0.5-14.0
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C2.33.0
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C12.812.5
20 mg/kg GSK3052230 + Docetaxel: Arm B3.56.0
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A2.09.6
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C10.618.7
5 mg/kg GSK3052230 + Docetaxel: Arm B-0.610.5
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A6.3-12.3

[back to top]

Number of Participants With Abnormal Echocardiogram (ECHO) Findings

Echocardiography scans were obtained at given time points using an echocardiogram and the findings for left ventricular ejection fraction (LVEF) were obtained. LVEF values at end of treatment (EOT) were recorded as no change or any increase and any decrease values. Only those participants available at the specified time points were analyzed. (NCT01868022)
Timeframe: Median of 28.5 weeks

,,,,,
InterventionParticipants (Count of Participants)
No change or any increaseAny Decrease
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C23
20 mg/kg GSK3052230 + Docetaxel: Arm B11
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A11
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C20
5 mg/kg GSK3052230 + Docetaxel: Arm B01
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A01

[back to top]

Number of Participants With Hematology Change From Baseline With Respect to the Normal Range

Hematology parameters included platelet Count, red blood cell (RBC) Count, hemoglobin, absolute white blood cell (WBC) Count, absolute neutrophils (Neu), absolute lymphocytes (Lym), absolute monocytes (Mono), absolute eosinophils (Eos), absolute basophils (Baso). Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Hematology parameters with worst-case change from Baseline with respect to normal range only has been presented. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01868022)
Timeframe: Median of 28.5 weeks

,,,,,,,,
InterventionParticipants (Count of Participants)
Baso, decrease to low, n=3,3,14,3,3,3,3,22,8Baso, normal or no change, n=3,3,14,3,3,3,3,22,8Baso, increase to high, n=3,3,14,3,3,3,3,22,8Eos, decrease to low, n=3,3,14,3,3,3,3,24,8Eos, normal or no change, n=3,3,14,3,3,3,3,24,8Eos, increase to high, n=3,3,14,3,3,3,3,24,8Mono, decrease to low, n=3,3,14,3,3,3,3,25,8Mono, normal or no change, n=3,3,14,3,3,3,3,25,8Mono, increase to high, n=3,3,14,3,3,3,3,25,8RBC, decrease to low, n=3,3,14,3,3,3,3,25,8RBC, normal or no change, n=3,3,14,3,3,3,3,25,8RBC, increase to high, n=3,3,14,3,3,3,3,25,8
10 mg/kg GSK3052230 + Docetaxel: Arm B111130203121
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A030030203210
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C030030220120
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C51348145612101793
20 mg/kg GSK3052230 + Docetaxel: Arm B030030102210
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A194111211451041
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C161351335341
5 mg/kg GSK3052230 + Docetaxel: Arm B030030021030
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A030030310300

[back to top]

Number of Participants With Best Response

Best response defined as complete response (CR:disappearance of all target. Any pathological lymph nodes < 10 millimeter [mm] in the short axis) or partial response (PR at least a 30 percent decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters), stable disease (SD neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for progressive disease) or progressive disease (PR at least a 20 percent increase in the sum of the diameters of target lesions, taking as a reference, the smallest sum of diameters recorded since the treatment started. In addition, the sum must have an absolute increase from nadir of 5 mm) according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 or modified RECIST. Best response as per RECIST version 1.1 for Arm A and B participants has been reported. Best response according to RECIST version 1.1 or modified RECIST for Arm C participants has been reported. (NCT01868022)
Timeframe: Median of 28.5 weeks

,,,,,,,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable disease, discontinuedStable disease, ongoingProgressive disease
10 mg/kg GSK3052230 + Docetaxel: Arm B00200
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A02100
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C02100
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C0111021
20 mg/kg GSK3052230 + Docetaxel: Arm B00200
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A06502
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C01201
5 mg/kg GSK3052230 + Docetaxel: Arm B00201
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A01101

[back to top]

Number of Participants With Clinical Chemistry Changes From Baseline With Respect to the Normal Range

Clinical chemistry parameters included potassium, sodium, chloride (Cl), total carbon dioxide (CO2), total and ionized calcium, magnesium, phosphate, albumin, glucose (fasting), Blood urea nitrogen (BUN), creatinine (Cr), uric acid, creatinine clearance, aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma glutamyl transferase (GGT), alkaline phosphatase, Total bilirubin (T. Bil), and Direct bilirubin (D. Bil), total T3 and T4, free T4, amylase, lipase, prothrombin time, partial thromboplastin time, international normalized ratio, and fibrinogen. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Clinical chemistry parameters with change from Baseline with respect to normal range only has been presented. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01868022)
Timeframe: Median of 28.5 weeks

,,
InterventionParticipants (Count of Participants)
D.Bil, decrease to low, n=2,2,8,1,2,2,1,12,6D.Bil, normal or no change, n=2,2,8,1,2,2,1,12,6D.Bil, increase to high, n=2,2,8,1,2,2,1,12,6Cl, decrease to low, n=3,3,14,3,3,3,3,25,8Cl, normal or no change, n=3,3,14,3,3,3,3,25,8Cl, increase to high, n=3,3,14,3,3,3,3,25,8CO2, decrease to low, n=3,3,14,3,3,3,3,25,8CO2, normal or no change, n=3,3,14,3,3,3,3,25,8CO2, increase to high, n=3,3,14,3,3,3,3,25,8Cr CL, decrease to low, n=3,3,9,2,1,3,3,15,4Cr CL, normal or no change, n=3,3,9,2,1,3,3,15,4Cr CL, increase to high, n=3,3,9,2,1,3,3,15,4Urea/BUN, decrease to low, n=3,2,13,3,2,3,3,25,8Urea/BUN,normal or no change,n=3,2,13,3,2,3,3,25,8Urea/BUN, increase to high, n=3,2,13,3,2,3,3,25,8
20 mg/kg GSK3052230 + Docetaxel: Arm B110120012030021
5 mg/kg GSK3052230 + Docetaxel: Arm B010030120011111
5 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A020120021120120

[back to top]

Number of Participants With Clinical Chemistry Changes From Baseline With Respect to the Normal Range

Clinical chemistry parameters included potassium, sodium, chloride (Cl), total carbon dioxide (CO2), total and ionized calcium, magnesium, phosphate, albumin, glucose (fasting), Blood urea nitrogen (BUN), creatinine (Cr), uric acid, creatinine clearance, aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma glutamyl transferase (GGT), alkaline phosphatase, Total bilirubin (T. Bil), and Direct bilirubin (D. Bil), total T3 and T4, free T4, amylase, lipase, prothrombin time, partial thromboplastin time, international normalized ratio, and fibrinogen. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Clinical chemistry parameters with change from Baseline with respect to normal range only has been presented. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01868022)
Timeframe: Median of 28.5 weeks

,,
InterventionParticipants (Count of Participants)
D.Bil, decrease to low, n=2,2,8,1,2,2,1,12,6D.Bil, normal or no change, n=2,2,8,1,2,2,1,12,6D.Bil, increase to high, n=2,2,8,1,2,2,1,12,6Cl, decrease to low, n=3,3,14,3,3,3,3,25,8Cl, normal or no change, n=3,3,14,3,3,3,3,25,8Cl, increase to high, n=3,3,14,3,3,3,3,25,8CO2, decrease to low, n=3,3,14,3,3,3,3,25,8CO2, normal or no change, n=3,3,14,3,3,3,3,25,8CO2, increase to high, n=3,3,14,3,3,3,3,25,8Cr CL, decrease to low, n=3,3,9,2,1,3,3,15,4Cr CL, normal or no change, n=3,3,9,2,1,3,3,15,4Cr CL, increase to high, n=3,3,9,2,1,3,3,15,4T4 free, decrease to low, n=0,1,6,0,1,0,2,16,3T4 free, normal or no change,=0,1,6,0,1,0,2,16,3T4 free, increase to high, n=0,1,6,0,1,0,2,16,3Total T3, decrease to low, n=0,1,1,0,1,0,1,6,3Total T3, normal or no change, n=0,1,1,0,1,0,1,6,3Total T3, increase to high, n=0,1,1,0,1,0,1,6,3Urea/BUN, decrease to low, n=3,2,13,3,2,3,3,25,8Urea/BUN,normal or no change,n=3,2,13,3,2,3,3,25,8Urea/BUN, increase to high, n=3,2,13,3,2,3,3,25,8
10 mg/kg GSK3052230 + Docetaxel: Arm B020210030010010010020
10 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A011102112210010010020
20 mg/kg GSK3052230 + Paclitaxel + Carboplatin: Arm A071581386180150010085

[back to top]

Number of Participants With Clinical Chemistry Changes From Baseline With Respect to the Normal Range

Clinical chemistry parameters included potassium, sodium, chloride (Cl), total carbon dioxide (CO2), total and ionized calcium, magnesium, phosphate, albumin, glucose (fasting), Blood urea nitrogen (BUN), creatinine (Cr), uric acid, creatinine clearance, aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma glutamyl transferase (GGT), alkaline phosphatase, Total bilirubin (T. Bil), and Direct bilirubin (D. Bil), total T3 and T4, free T4, amylase, lipase, prothrombin time, partial thromboplastin time, international normalized ratio, and fibrinogen. Baseline is defined as the most recent, non-missing value prior to or on the first study GSK3052230 treatment dose date. Change from Baseline is calculated as visit value minus Baseline value. Clinical chemistry parameters with change from Baseline with respect to normal range only has been presented. Only those participants available at the specified time points were analyzed (represented by n=X in the category titles). (NCT01868022)
Timeframe: Median of 28.5 weeks

,,
InterventionParticipants (Count of Participants)
D.Bil, decrease to low, n=2,2,8,1,2,2,1,12,6D.Bil, normal or no change, n=2,2,8,1,2,2,1,12,6D.Bil, increase to high, n=2,2,8,1,2,2,1,12,6Cl, decrease to low, n=3,3,14,3,3,3,3,25,8Cl, normal or no change, n=3,3,14,3,3,3,3,25,8Cl, increase to high, n=3,3,14,3,3,3,3,25,8CO2, decrease to low, n=3,3,14,3,3,3,3,25,8CO2, normal or no change, n=3,3,14,3,3,3,3,25,8CO2, increase to high, n=3,3,14,3,3,3,3,25,8Cr CL, decrease to low, n=3,3,9,2,1,3,3,15,4Cr CL, normal or no change, n=3,3,9,2,1,3,3,15,4Cr CL, increase to high, n=3,3,9,2,1,3,3,15,4T4 free, decrease to low, n=0,1,6,0,1,0,2,16,3T4 free, normal or no change,=0,1,6,0,1,0,2,16,3T4 free, increase to high, n=0,1,6,0,1,0,2,16,3T4 total, decrease to low, n=0,0,0,0,0,0,1,4,1T4 total, normal or no change, n=0,0,0,0,0,0,1,4,1T4 total, increase to high, n=0,0,0,0,0,0,1,4,1Total T3, decrease to low, n=0,1,1,0,1,0,1,6,3Total T3, normal or no change, n=0,1,1,0,1,0,1,6,3Total T3, increase to high, n=0,1,1,0,1,0,1,6,3Urea/BUN, decrease to low, n=3,2,13,3,2,3,3,25,8Urea/BUN,normal or no change,n=3,2,13,3,2,3,3,25,8Urea/BUN, increase to high, n=3,2,13,3,2,3,3,25,8
10 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C010210120120020010010012
15 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C07591522177961112311232141110
20 mg/kg GSK3052230 + Pemetrexed + Cisplatin: Arm C051352053040120001210134

[back to top]

Disease Control According to Modified RECIST- Investigator Assessment

"Disease control (best overall response of confirmed CR or PR, or Stable Disease (SD) that lasted ≥36 days) according to modified RECIST.~Percentage of Patients with Disease control is presented. This endpoint was only evaluated for Phase III part." (NCT01907100)
Timeframe: Tumour imaging was to be performed every 6 weeks until disease progression, death or start of subsequent anti-cancer therapy, whichever occurred earlier; up to 54 months

InterventionPercentage of participants (Number)
Placebo92.6
Nintedanib90.8

[back to top]

Objective Response According to Modified RECIST- Investigator Assessment

"Objective response (best overall tumour response of confirmed complete response [CR] or confirmed partial response [PR]).~Complete Response: disappearance of all target lesions Partial Response: at least a 30 % decrease in the total tumour measurement of target lesions, taking as reference the baseline total tumour measurement.~Percentage of Patients with confirmed objective response is presented. This endpoint was only evaluated for Phase III part." (NCT01907100)
Timeframe: Tumour imaging was to be performed every 6 weeks until disease progression, death or start of subsequent anti-cancer therapy, whichever occurred earlier; up to 54 months

InterventionPercentage of participants (Number)
Placebo42.8
Nintedanib45.0

[back to top]

Overall Survival (OS)

"Overall survival was defined as the duration of time from randomization to time of death.~This is the key secondary endpoint of the trial." (NCT01907100)
Timeframe: From randomization until the earliest of disease progression, death or (Phase II: cut-off date of 4-March-2016; up to 889 days) (Phase III: cut-off date of 16-March-2018; up to 31 months)

,
InterventionMonths (Median)
Phase IIPhase III
Nintedanib18.3014.36
Placebo14.4616.07

[back to top]

Progression-Free Survival (PFS)

This outcome measure presents progression-free survival. Disease progression was defined according to the modified Response Evaluation Criteria in Solid Tumours (RECIST) criteria. Progression-free survival time was calculated as the duration from the date of randomization to the date of disease progression or death, whichever occurred first. For patients with known date of progression (or death): PFS (days) = min (date of progression, date of death) - date of randomization + 1 day. For patients without progression or death, PFS was censored at the last imaging date that showed no disease progression: PFS (days, censored) = date of last imaging showing no progression - date randomization + 1 day. (NCT01907100)
Timeframe: From randomization until the earliest of disease progression, death or (Phase II: cut-off date of 4-March-2016; up to 889 days) (Phase III: cut-off date of 16-March-2018; up to 31 months)

,
InterventionMonths (Median)
Phase IIPhase III
Nintedanib9.366.77
Placebo5.726.97

[back to top]

Overall Survival

Overall survival (OS) defined as time from start of Dacomitinib to date of death from any cause. Patients without recorded death were censored at the date the patient was last known to be alive. Patients were followed up for survival for 24 month after end of Treatment. (NCT01918761)
Timeframe: until date of death. The study was suspended after 36 months.

Interventionmonths (Median)
Dacomitinib, Pemetrexed9.6

[back to top]

Progression-free Survival

Progression-free survival (PFS) defined as time from start of Dacomitinib to date of progression or date of death from any cause, whichever occurred first. Patients without recorded progression or death were censored at the last date they were known to have not progressed. Patients were followed up for progression-free survival for 24 month after end of Treatment. (NCT01918761)
Timeframe: Up to progression or death due to any cause. The study was suspended after 36 months

Interventionmonths (Median)
Dacomitinib, Pemetrexed4.7

[back to top]

Dose Limiting Toxicities (DLTs)

The primary objective of this study is to determine the maximal tolerated dose (MTD) of the combination pemetrexed + dacomitinib by the incidence of dose limiting toxicities (DLTs). (NCT01918761)
Timeframe: From start of treatment to end of treatment or death, whichever occurs first. The study was suspended after 36 months.

Interventionpercentage of participants (Number)
Dacomitinib, Pemetrexed40

[back to top]

Overall Response Rate

Overall Response Rate (ORR) is defined as the proportion of patients with complete Response (CR) or partial Response (PR). (NCT01918761)
Timeframe: Until progression of disease (PD) or 24 month after end of treatment for participants with no PD. The study was suspended after 36 months

Interventionpercentage of participants (Number)
Dacomitinib, Pemetrexed0

[back to top]

Phase 1b: Number of Participants With Clinically Significant Abnormalities in 12-Lead Electrocardiograms (ECG) Findings

ECG parameters included heart rhythm, pulse rate intervals, QRS, QT intervals, RR intervals and corrected QT(QTc) intervals. Clinical significance was determined by the investigator. Number of participants with clinically significant abnormalities in 12-lead ECG were reported. (NCT01982955)
Timeframe: Up to 175 weeks

InterventionParticipants (Count of Participants)
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg0
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg0

[back to top]

Phase 1b: Number of Participants With Clinically Significant Abnormalities in Vital Signs

Vital signs assessment included blood pressure, heart rate, respiratory rate and body temperature. Number of Participants with any clinically significant abnormalities in vital signs were reported. Clinical significance was determined by the investigator. (NCT01982955)
Timeframe: Up to 175 weeks

InterventionParticipants (Count of Participants)
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg0
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg0

[back to top]

Phase 1b: Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status Score of 2 or Higher Than 2

ECOG PS score is widely used by doctors and researchers to assess how a participants' disease is progressing, and is used to assess how the disease affects the daily living abilities of the participant, and determine appropriate treatment and prognosis. The score ranges from Grade 0 to Grade 5, where Grade 0 = Fully active, able to carry on all pre-disease performance without restriction, Grade 1 = Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature (like light house work, office work), Grade 2 = Ambulatory and capable of all self-care but unable to carry out any work activities, Grade 3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours and Grade 4 = Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair, Grade 5 = Death. Number of participants with ECOG performance status score of 2 or higher than 2 were reported. (NCT01982955)
Timeframe: Up to 175 weeks

InterventionParticipants (Count of Participants)
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg1
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg9

[back to top]

Phase 1b: Percentage of Participants With Disease Control Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria

Disease control defined as CR, PR, or stable disease(SD) as the best overall response according to local radiological assessments from the date of randomization/the first administration of the study treatment to the first observation of PD. CR:disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. PR:at least 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD:an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5mm. SD:as any cases that do not qualify for either PR or PD at minimum interval of 42 days after randomization/start of study treatment (NCT01982955)
Timeframe: Up to 328 weeks

InterventionPercentage of Participants (Number)
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg50.0
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg58.3

[back to top]

Phase 1b: Percentage of Participants With Objective Response Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria

Objective response (OR) was defined as the percentage of participants who had achieved complete response (CR) or partial response (PR) as the best overall response according to local radiological assessments from randomization/the first administration of the study treatment to the first observation of disease progression (PD). CR: defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR: defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionPercentage of Participants (Number)
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg33.3
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg33.3

[back to top]

Phase 2 (Non-Randomized Part Only): Percentage of Participants With Disease Control Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria

Disease control defined as CR, PR, or stable disease(SD) as the best overall response according to local radiological assessments from the date of randomization/the first administration of the study treatment to the first observation of PD. CR:disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. PR:at least 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD:an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5mm. SD:as any cases that do not qualify for either PR/PD at minimum interval of 42 days after randomization/start of study treatment. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionPercentage of Participants (Number)
Phase 2: Single-arm Cohort (MET+ T790M Positive)40

[back to top]

Phase 2 (Non-Randomized Part Only): Percentage of Participants With Objective Response Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria

Objective response (OR) was defined as the percentage of participants who had achieved complete response (CR) or partial response (PR) as the best overall response according to local radiological assessments from randomization/the first administration of the study treatment to the first observation of disease progression (PD). CR: defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR: defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionPercentage of Participants (Number)
Phase 2: Single-arm Cohort (MET+ T790M Positive)0

[back to top]

Phase 2 (Non-Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by Independent Review Committee (IRC)

Progression-free survival (assessed by Independent Review Committee) time was defined as the time in months from randomization to either first observation of radiologically confirmed progression disease by the IRC or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. PFS was measured using Kaplan-Meier (KM) estimates. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Single-arm Cohort (MET+ T790M Positive)2.63

[back to top]

Phase 2 (Non-Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by Investigator

Progression-free survival (assessed by Investigator) time was defined as the time in months from randomization to either first observation of radiologically confirmed progression disease by the Investigator or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. PFS was measured using Kaplan-Meier (KM) estimates. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Single-arm Cohort (MET+ T790M Positive)1.41

[back to top]

Phase 2 (Randomized Part Only): Percentage of Participants With Disease Control Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria

Disease control defined as CR, PR, or stable disease(SD) as the best overall response according to local radiological assessments from the date of randomization/the first administration of the study treatment to the first observation of PD. CR:disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. PR:at least 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD:an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5mm. SD:as any cases that do not qualify for either PR/PD at minimum interval of 42 days after randomization/start of study treatment. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionPercentage of Participants (Number)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)83.9
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)70.8

[back to top]

Phase 2 (Randomized Part Only): Percentage of Participants With Objective Response Based on Tumor Response Assessment According to Response Evaluation Criteria In Solid Tumors Version 1.1 (RECIST v1.1) Criteria

Objective response (OR) was defined as the percentage of participants who had achieved complete response (CR) or partial response (PR) as the best overall response according to local radiological assessments from randomization/the first administration of the study treatment to the first observation of disease progression (PD). CR: defined as disappearance of all target and non-target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. PR: defined as at least a 30% decrease in the sum of diameters of target lesions taking as reference the baseline sum diameters. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionPercentage of Participants (Number)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)45.2
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)33.3

[back to top]

Phase 2 (Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by Independent Review Committee (IRC)

Progression-free survival (assessed by Independent Review Committee) time was defined as the time in months from randomization to either first observation of radiologically confirmed progression disease (PD) by the IRC or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. PFS was measured using Kaplan-Meier (KM) estimates. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)10.15
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)4.34

[back to top]

Phase 2 (Randomized Part Only): Progression-free Survival (PFS) Based on Tumor Assessment by the Investigator

Progression-free survival (assessed by the Investigator) time was defined as the time in months from randomization to either first observation of radiologically confirmed progression disease (PD) by the investigator or occurrence of death due to any cause within 84 days of either randomization or the last tumor assessment. PD is defined as at least a 20 percent (%) increase in the sum of diameters of target lesions, taking as reference the smallest sum on study; and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 millimeter (mm). PFS was measured using Kaplan-Meier (KM) estimates. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)4.86
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)4.37

[back to top]

Phase 2: (Non-Randomized Part Only): Overall Survival (OS) Time

Overall survival time was measured as time in months between the date of randomization and the date of death. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Single-arm Cohort (MET+ T790M Positive)25.86

[back to top]

Phase 2: (Randomized Part Only): Overall Survival (OS) Time

Overall survival time was measured as time in months between the date of randomization and the date of death. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)17.25
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)19.48

[back to top]

Phase 2: Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life (EORTC QLQ-C30) Global Health Status Scale Score at End of Treatment (EOT)

EORTC QLQ-C30 is a 30-question tool used to assess the overall quality of life (QoL) in cancer participants. It consisted of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, role, cognitive, emotional, social), and 9 symptom scales/items (Fatigue, nausea and vomiting, pain, dyspnoea, sleep disturbance, appetite loss, constipation, diarrhea, financial impact. The EORTC QLQ-C30 GHS/QoL score ranges from 0 to 100; High score indicates better GHS/QoL. Score 0 represents: very poor physical condition and QoL. Score 100 represents: excellent overall physical condition and QoL. (NCT01982955)
Timeframe: Baseline and EOT (up to 110 weeks)

InterventionUnits on a Scale (Mean)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)-16.29
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)-2.78
Phase 2: Single-arm Cohort (MET+ T790M Positive)-24.19

[back to top]

Phase 2: Number of Participants With Clinically Significant Abnormalities in 12-Lead Electrocardiograms (ECG) Findings

ECG parameters included heart rhythm, pulse rate intervals, QRS, QT intervals, RR intervals and corrected QT(QTc) intervals. Clinical significance was determined by the investigator. Number of participants with clinically significant abnormalities in 12-lead ECG were reported. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionParticipants (Count of Participants)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)2
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)1
Phase 2: Single-arm Cohort (MET+ T790M Positive)0

[back to top]

Phase 2: Number of Participants With Clinically Significant Abnormalities in Vital Signs

Vital signs assessment included blood pressure, heart rate, respiratory rate and body temperature. Number of Participants with any clinically significant abnormalities in vital signs were reported. Clinical significance was determined by the investigator. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionParticipants (Count of Participants)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)0
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)0
Phase 2: Single-arm Cohort (MET+ T790M Positive)0

[back to top]

Phase 2: Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status Score of 2 or Higher Than 2

ECOG PS score is widely used by doctors and researchers to assess how a participants' disease is progressing, and is used to assess how the disease affects the daily living abilities of the participant, and determine appropriate treatment and prognosis. The score ranges from Grade 0 to Grade 5, where Grade 0 = Fully active, able to carry on all pre-disease performance without restriction, Grade 1 = Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature (like light house work, office work), Grade 2 = Ambulatory and capable of all self-care but unable to carry out any work activities, Grade 3 = Capable of only limited self-care, confined to bed or chair more than 50% of waking hours and Grade 4 = Completely disabled. Cannot carry on any self-care. Totally confined to bed or chair, Grade 5 = Death. Number of participants with ECOG performance status score of 2 or higher than 2 were reported. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionParticipants (Count of Participants)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)6
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)1
Phase 2: Single-arm Cohort (MET+ T790M Positive)1

[back to top]

Phase 2: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) Leading to Permanent Treatment Discontinuation

An adverse event (AE) was defined as any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. A serious AE was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. Term TEAE is defined as AEs starting/worsening after first intake of the study drug. TEAEs included both Serious TEAEs and non-serious TEAEs. Number of participants with TEAEs leading to permanent treatment discontinuation were reported. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionParticipants (Count of Participants)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)3
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)1
Phase 2: Single-arm Cohort (MET+ T790M Positive)2

[back to top]

Phase 2: Time-to-Symptom Progression (TTSP)

TTSP was measured from randomization to symptomatic progression by lung cancer symptom scale (LCSS) used to measure symptom changes relevant to quality of life (QoL).It consisted of 9 items focused on cancer symptoms (loss of appetite, fatigue, cough, shortness of breath, blood in sputum, pain, symptoms of cancer, illness affecting normal activity, QoL).For each symptom score distance from left boundary to point where participant has marked line was measured in millimeters (mm).Total scale length was 100 mm. Symptomatic progression was defined as increase/worsening of average symptomatic burden index (ASBI) (mean of 6 major lung cancer specific symptom scores);Worsening defined as 10% increase of scale breadth from baseline. Score 0 indicate no/minimum symptoms;100 indicates maximum level of symptoms. (NCT01982955)
Timeframe: Up to 328 weeks

InterventionMonths (Median)
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)5.75
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)7.95
Phase 2: Single-arm Cohort (MET+ T790M Positive)2.63

[back to top]

Phase 1b: Apparent Terminal Elimination Rate Constant Lambda(z) of Tepotinib, Its Metabolites and Gefitinib

Lambda(z) was determined from the terminal slope of the log-transformed plasma concentration curve using linear regression method. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
Intervention1 per hour (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mgNANANANANANANANA
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mgNANANANANANANANA

[back to top]

Phase 1b: Apparent Terminal Half-Life (t1/2) of Tepotinib, Its Metabolites and Gefitinib

Apparent terminal half-life was defined as the time required for the plasma concentration of drug to decrease 50 percent in the final stage of its elimination. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
InterventionHours (Median)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mgNANANANANANANANA
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mgNANANANANANANANA

[back to top]

Phase 1b: Apparent Total Body Clearance From Plasma (CL/F) of Tepotinib and Gefitinib

The CL/f is a measure of the rate at which it was metabolized or eliminated by normal biological processes. Clearance obtained after oral dose was influenced by the fraction of the dose absorbed. The CL/F from plasma was calculated using the formula: Dose divided by area under the concentration time curve from time zero to infinity (AUC0-inf). (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 15 of Cycle 1 (each Cycle is 21 days)

,
Interventionliter per hour (L/h) (Geometric Mean)
TepotinibGefitinib
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg17.332.5
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg20.335.3

[back to top]

Phase 1b: Apparent Volume of Distribution (Vz/F) During the Terminal Phase of Tepotinib, Its Metabolites and Gefitinib

The Vz/f was defined as the theoretical volume in which the total amount of required to uniformly distribute to produce the desired plasma concentration. Apparent volume of distribution after oral dose (Vz/F) was influenced by the fraction absorbed. The Vz/f was calculated by dividing the dose with area under the concentration time curve from time zero to infinity multiplied with terminal elimination rate constant Lambda(z). Vz/f=Dose/AUC(0-inf) multiply Lambda(z). (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
InterventionLiter (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mgNANANANANANANANA
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mgNANANANANANANANA

[back to top]

Phase 1b: Apparent Volume of Distribution During the Steady State (Vss/F) of Tepotinib, Its Metabolites and Gefitinib

Volume of distribution was defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired plasma concentration of a drug. Vss/f after oral dose was influenced by the fraction absorbed. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
InterventionLiter (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mgNANANANANANANANA
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mgNANANANANANANANA

[back to top]

Phase 1b: Area Under the Plasma Concentration Versus Time Curve From Time Zero to the Last Sampling Time AUC (0-t) of Tepotinib, Its Metabolites and Gefitinib

Area under the plasma concentration versus time curve from time zero to the last sampling time t at which the concentration is at or above the lower limit of quantification (LLLQ). AUC(0-t) was calculated according to the mixed log-linear trapezoidal rule. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
Interventionnanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A Day15 of Cycle 1MSC2571107A Day 1 of Cycle 1MSC2571107A Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg62801560016804420248872NA7690
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg92102220017707530324188029307080

[back to top]

Phase 1b: Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC 0-infinity) of Tepotinib, Its Metabolites and Gefitinib

The AUC(0-inf) was estimated by determining the total area under the curve of the concentration versus time curve extrapolated to infinity. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
Interventionng*h/mL (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mgNANANANANANANANA
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mgNANANANANANANANA

[back to top]

Phase 1b: Area Under the Plasma Concentration-Time Curve Within 1 Dosing Interval (AUC 0-tau) of Tepotinib, Its Metabolites and Gefitinib

AUC (0-tau) is the area under the plasma concentration time curve within 1 dosing interval. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
Interventionng*h/mL (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg62801560016804420248872NA7690
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg92102220017707530324188029307080

[back to top]

Phase 1b: Average Observed Plasma Concentration (Cavg) of Tepotinib, Its Metabolites and Gefitinib

Cavg is the average plasma concentration within 1 dosing interval obtained directly from the concentration versus time curve. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 15 of Cycle 1 (each Cycle is 21 days)

,
Interventionng/mL (Geometric Mean)
TepotinibMSC2571109AMSC2571107AGefitinib
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg65418536.4321
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg92431478.3295

[back to top]

Phase 1b: Maximum Observed Plasma Concentration (Cmax) of Tepotinib, Its Metabolites and Gefitinib

Cmax is the maximum observed plasma concentration obtained directly from the concentration versus time curve. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
InterventionNanogram per Milliliter (ng/mL) (Geometric Mean)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg37576313228016.844.9NA432
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg575105014944424.394.9215366

[back to top]

Phase 1b: Minimum Observed Plasma Concentration (Cmin) of Tepotinib, Its Metabolites and Gefitinib

Cmin is minimum observed plasma concentration obtained directly from the concentration versus time curve. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 15 of Cycle 1 (each Cycle is 21 days)

,
Interventionng/mL (Geometric Mean)
TepotinibMSC2571109AMSC2571107AGeftinib
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg53415632.8231
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg73527068.7190

[back to top]

Phase 1b: Number of Participants With Death and Reasons

Number of participants with death due to progressive disease (PD), adverse event (AE) related to study treatment, AE not related to study treatment were reported. An AE was defined as any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. Number of participants with deaths due to PD, AE related to study treatment, AE not related to study treatment were reported. (NCT01982955)
Timeframe: Up to 175 weeks

,
InterventionParticipants (Count of Participants)
Death due to PDDeath due to AE related to study treatmentDeath due to AE not related to study treatment
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg001
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg300

[back to top] [back to top]

Phase 1b: Number of Participants With Laboratory Test Abnormalities of Grade 3 or Higher Severity Based on NCI-CTCAE Version 4.03 Reported as Treatment-Emergent Adverse Events (TEAEs)

The laboratory measurements included hematology and coagulation, biochemistry and urinalysis. (NCT01982955)
Timeframe: Up to 175 weeks

,
InterventionParticipants (Count of Participants)
Amylase increasedLipase increasedNeutrophil count decreasedHyperglycemiaHypocalcemiaHyponatremiaHypoproteinemia
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg2100101
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg2212020

[back to top]

Phase 1b: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious TEAEs

An AE was defined as any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. A serious AE was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. The term TEAE is defined as AEs starting or worsening after the first intake of the study drug. TEAEs include both Serious TEAEs and non-serious TEAEs. Number of Participants with TEAEs and serious TEAEs were reported. (NCT01982955)
Timeframe: Up to 175 weeks

,
InterventionParticipants (Count of Participants)
Any TEAEsAny Serious TEAEs
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg64
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg127

[back to top]

Phase 1b: Number of Participants With Treatment-Emergent Adverse Events (TEAEs) Leading to Permanent Treatment Discontinuation

An adverse event (AE) was defined as any unfavourable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. A serious AE was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. Term TEAE is defined as AEs starting/worsening after first intake of the study drug. TEAEs included both Serious TEAEs and non-serious TEAEs. Number of participants with TEAEs leading to permanent treatment discontinuation were reported. (NCT01982955)
Timeframe: Up to 175 weeks

,
InterventionParticipants (Count of Participants)
TEAE Leading Permanent Tepotinib DiscontinuationTEAE Leading Permanent Gefitinib Discontinuation
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg00
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg21

[back to top] [back to top]

Phase 1b: Time to Reach Maximum Plasma Concentration (Tmax) of Tepotinib, Its Metabolites and Gefitinib

Tmax is time to reach maximum observed plasma concentration obtained directly from the concentration versus time curve. (NCT01982955)
Timeframe: Pre dose, 0.25, 0.5, 1, 2, 4, 8, 10 and 24 hours post dose on Day 1 and 15 of Cycle 1 (each Cycle is 21 days)

,
InterventionHours (Median)
Tepotinib: Day 1 of Cycle 1Tepotinib: Day 15 of Cycle 1MSC2571109A: Day 1 of Cycle 1MSC2571109A: Day 15 of Cycle 1MSC2571107A: Day 1 of Cycle 1MSC2571107A: Day 15 of Cycle 1Gefitinib: Day 1 of Cycle 1Gefitinib: Day 15 of Cycle 1
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg8.006.0024.000.0024.000.13NA4.00
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg9.019.0024.000.0024.000.258.008.00

[back to top]

Phase 2: Number of Participants With Death and Reasons

An AE was defined as any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with the use of study drug, whether or not considered related to the study drug. PD defined as an increase of at least 20% in the sum of the diameters of target lesions, taking as reference the smallest sum of the diameters of target lesions recorded since treatment started and/or unequivocal progression of existing non-target lesions and/or the presence of new lesions. The sum must also demonstrate an absolute increase of at least 5 mm. Number of participants with deaths due to progression disease (PD), AE related to study treatment, unknown reason was reported. (NCT01982955)
Timeframe: Up to 328 weeks

,,
InterventionParticipants (Count of Participants)
Death due to disease progressionDeath due to AE related to study treatmentDeath due to unknown reason
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)1505
Phase 2: Single-arm Cohort (MET+ T790M Positive)802
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)2102

[back to top] [back to top]

Phase 2: Number of Participants With Laboratory Test Abnormalities of Grade 3 or Higher Severity Based on NCI-CTCAE Version 4.03 Reported as Treatment-Emergent Adverse Events (TEAEs)

The laboratory measurements included hematology and coagulation, biochemistry and urinalysis. (NCT01982955)
Timeframe: Up to 328 weeks

,,
InterventionParticipants (Count of Participants)
AnaemiaNeutropeniaAlanine aminotransferase increasedAmylase increasedGamma-glutamyltransferase increasedLipase increasedNeutrophil count decreasedWhite blood cell count decreasedHyponatremiaHypokalemiahypophosphatemiaHypoalbuminemia
Phase 2: Pemetrexed and Cisplatin/Carboplatin (MET + T790 Negative)710212323210
Phase 2: Single-arm Cohort (MET+ T790M Positive)000201000000
Phase 2: Tepotinib 500 mg + Gefitinib 250 mg (MET + T790 Negative)001705211001

[back to top] [back to top]

Phase 1b: Number of Participants Experiencing at Least One Dose Limiting Toxicity (DLT)

Dose limiting toxicity (DLT) using National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) Version 4.0 was defined as toxicities at any dose level and judged to be related to the study treatment by investigator and/or the sponsor. DLTs included Grade 4 neutropenia for more than 7 days; Grade greater than or equal to (>=) 3 febrile neutropenia for more than 1 day; Grade 4 thrombocytopenia or Grade 3 thrombocytopenia with non-traumatic bleeding; Grade >= 3 uncontrolled nausea/vomiting and/or diarrhea despite adequate and optimal treatment and Grade >= 3 any non-hematological adverse event (AE), except the aforementioned gastrointestinal events and alopecia. Number of participants who experienced DLT during Phase 1b were reported. (NCT01982955)
Timeframe: Day 1 to Day 21 of Cycle 1 (each cycle is 21 days)

InterventionParticipants (Count of Participants)
Phase 1b: Tepotinib 300 mg + Gefitinib 250 mg0
Phase 1b: Tepotinib 500 mg + Gefitinib 250 mg0

[back to top]

Part 2 Cohorts G+ and G-: Duration of Response (DOR)

For participants who demonstrated a confirmed response (Complete Response [CR]: Disappearance of all target lesions or Partial Response [PR]: At least a 30% decrease in the sum of diameters of target lesions) per RECIST 1.1. Per RECIST 1.1, DOR was defined as the time from first documented evidence of CR or PR until disease progression or death. DOR was assessed by BICR. (NCT02039674)
Timeframe: Up to approximately 2 years

InterventionMonths (Median)
Part 2 Cohort G+ (Pembro 200 mg+Pe+C)NA
Part 2 Cohort G- (Placebo+Pe+C)NA

[back to top]

Part 2 Cohorts G+ and G-: Progression-Free Survival (PFS)

PFS was defined as the time from randomization to the first documented disease progression, or death due to any cause, whichever occurred first. Per RECIST 1.1, progressive disease was defined as at least a 20% increase in the sum of diameters of target lesions. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions was also considered progression. PFS was assessed by BICR. (NCT02039674)
Timeframe: Up to approximately 2 years

InterventionMonths (Median)
Part 2 Cohort G+ (Pembro 200 mg+Pe+C)24.5
Part 2 Cohort G- (Placebo+Pe+C)9.9

[back to top]

Part 2 Cohorts G+ and G-: Overall Survival (OS)

OS was defined as the time from randomization to death due to any cause. (NCT02039674)
Timeframe: Up to approximately 2 years

InterventionMonths (Median)
Part 2 Cohort G+ (Pembro 200 mg+Pe+C)34.5
Part 2 Cohort G- (Placebo+Pe+C)21.1

[back to top]

All Cohorts: Number of Participants Who Experienced a Dose-limiting Toxicity (DLT)

DLTs were assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events version 4. A DLT was defined as any of the following events: Grade 4 non-hematologic toxicity (not laboratory); Grade 4 hematologic toxicity lasting ≥7 days; Grade 3 non-hematologic toxicity (not laboratory, specifically nausea, vomiting and diarrhea) lasting >3 days despite optimal supportive care; Any Grade 3 or Grade 4 non-hematologic laboratory value requiring treatment or hospitalization, or persisting for >1 week; Febrile neutropenia Grade 3 or Grade 4; Qualifying thrombocytopenia <25,000/mm^3; Prolonged delay (>2 weeks) in initiating Cycle 2 due to treatment-related toxicity; Missing >10% of erlotinib or gefitinib doses as a result of adverse events (AEs) during the DLT window of observation; or Grade 5 toxicity. (NCT02039674)
Timeframe: Cycle 1 (Up to 21 days)

InterventionParticipants (Count of Participants)
Part1 Cohort A2 (Pembro 2 mg/kg+Paclitaxel [Pa]+Carboplatin [C])0
Part1 Cohort A10 (Pembro10mg/kg+Pa+C)0
Part 1 Cohort B2 (Pembro 2mg/kg+Pa+C+Bevacizumab [B])0
Part 1 Cohort B10 (Pembro 10 mg/kg+Pa+C+B)0
Part 1 Cohort C2 (Pembro 2 mg/kg+Pemetrexed [Pe]+C)0
Part 1 Cohort C10 (Pembro 10 mg/kg+Pe+C)0
Part 1 Cohort D1 (Pembro 10mg/kg+Ipilimumab [I])0
Part 1 Cohort D2 (Pembro 10 mg/kg+I)0
Part 1 Cohort D4 (Pembro 2 mg/kg+I)0
Part 1 Cohort E (Pembro 2 mg/kg+Erlotinib)0
Part 1 Cohort F (Pembro 2 mg/kg+Gefitinib)0
Part 2 Cohort G+ (Pembro 200 mg+Pe+C)0
Part 2 Cohort G- (Placebo+Pe+C)0
Part 2 Cohort H (Pembro 2 mg/kg+I)0

[back to top]

Part 2 Cohorts D4 and H: Objective Response Rate (ORR)

For participants who demonstrated a confirmed response (Complete Response [CR]: Disappearance of all target lesions or Partial Response [PR]: At least a 30% decrease in the sum of diameters of target lesions) per RECIST 1.1. Per RECIST 1.1, DOR was defined as the time from first documented evidence of CR or PR until disease progression or death. DOR was assessed by BICR. (NCT02039674)
Timeframe: Up to approximately 2 years

InterventionPercentage of Participants (Number)
Part 2 Cohorts D4 & H (Pembro 2mg/kg+I)29.5

[back to top]

Part 2 Cohorts G+ and G-: Objective Response Rate (ORR)

ORR was defined as the percentage of participants in the analysis population 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) per Response Criteria in Solid Tumors version 1.1 (RECIST 1.1) as assessed by blinded independent central review (BICR). (NCT02039674)
Timeframe: Up to approximately 2 years

InterventionPercentage of Participants (Number)
Part 2 Cohort G+ (Pembro 200 mg+Pe+C)55.0
Part 2 Cohort G- (Placebo+Pe+C)28.6

[back to top] [back to top]

Progression-Free Survival in Participants With PD-L1 Expression >= 5%

Progression-Free Survival (PFS) was defined as the time between the date of randomization and the first date of documented tumor progression, as determined by the Independent Radiology Review Committee (IRRC) per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1, or death due to any cause, whichever occurs first. Participants who die without a reported progression were considered to have progressed on the date of their death. Participants who did not progress or die were censored on the date of their last evaluable tumor assessment. Participants who did not have any on-study tumor assessments and did not die were censored on the day they were randomized. Participants who received subsequent anti-cancer therapy prior to documented progression were censored at the last evaluable tumor assessment prior to the initiation of new therapy. (NCT02041533)
Timeframe: From date of randomization until date of documented tumor progression (assessed up to August 2016, approximately 28 months)

InterventionMonths (Median)
Nivolumab4.21
Investigator Choice of Chemotherapy5.88

[back to top]

Progression-Free Survival in All Randomized Participants

Progression-Free Survival (PFS) was defined as the time between the date of randomization and the first date of documented tumor progression, as determined by the Independent Radiology Review Committee (IRRC) per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1, or death due to any cause, whichever occurs first. Participants who die without a reported progression were considered to have progressed on the date of their death. Participants who did not progress or die were censored on the date of their last evaluable tumor assessment. Participants who did not have any on-study tumor assessments and did not die were censored on the day they were randomized. Participants who received subsequent anti-cancer therapy prior to documented progression were censored at the last evaluable tumor assessment prior to the initiation of new therapy. (NCT02041533)
Timeframe: From date of randomization until date of documented tumor progression (assessed up to August 2016, approximately 28 months)

InterventionMonths (Median)
Nivolumab4.21
Investigator Choice of Chemotherapy5.82

[back to top]

Overall Survival in Participants With PD-L1 Expression >= 5%

Overall Survival (OS) was defined as the time from randomization to the date of death. A participant who had not died was censored at the last known alive date. OS was censored at the date of randomization for participants who were randomized but had no follow-up. (NCT02041533)
Timeframe: From date of randomization to date of death (up to approximately 89 months)

InterventionMonths (Median)
Nivolumab14.36
Investigator Choice of Chemotherapy13.21

[back to top]

Overall Survival in All Randomized Participants

Overall Survival (OS) was defined as the time from randomization to the date of death. A participant who had not died was censored at the last known alive date. OS was censored at the date of randomization for participants who were randomized but had no follow-up. (NCT02041533)
Timeframe: From date of randomization to date of death (up to approximately 89 months)

InterventionMonths (Median)
Nivolumab13.73
Investigator Choice of Chemotherapy13.80

[back to top]

Objective Response Rate (ORR) in Participants With PD-L1 Expression >= 5%

ORR was defined as the proportion of randomized participants who achieved a Best Overall Response (BOR) of CR or PR using the RECIST v1.1 criteria per Independent Radiology Review Committee (IRRC) assessment. BOR was defined as the best response designation recorded between the date of randomization and the date of objectively documented progression or start of subsequent anti-cancer therapy, whichever occurred first. For participants without documented progression or subsequent therapy, all available response designations contributed to the BOR assessment. For participants who continued treatment beyond progression, BOR was determined from response designations recorded up to the time of initial progression. CR= Disappearance of all evidence of disease, confirmed by PET scan; PR= Regression of measureable disease and no new sites; Stable Disease (SD)= Failure to attain CR/PR or PD; Progressive Disease (PD)= Any new lesion or increase by >=50% of previously involved sites from nadir. (NCT02041533)
Timeframe: From date of randomization until date of documented tumor progression or subsequent anti-cancer therapy, whichever occurs first (assessed up to August 2016, approximately 28 months)

InterventionPercentage of participants (Number)
Nivolumab26.1
Investigator Choice of Chemotherapy33.5

[back to top]

Pharmacokinetics (PK): Maximum Concentration (Cmax) of Abemaciclib on Day 1 and at Steady State (Cycle 2 Day 1) in Part A , B, C, D and E

Cmax of Abemaciclib on day 1 and at steady State (Cycle 2 Day 1) Part A , B, C, D and E was evaluated. (NCT02079636)
Timeframe: Cycle 1 Day 1 (C1D1) pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; Cycle 2 Day 1 (C2D1) pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,,,,,,,,,
Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Day 1Steady State
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed212509
Part A:150 mg Abemaciclib + 500 mg/m^2 Pemetrexed114NA
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine80.6NA
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine201417
Part C: 150 mg Abemaciclib + 8 mg/kg or 10 mg/kg Ramucirumab140322
Part C: 200 mg Abemaciclib + 10mg/kg Ramucirumab Day 1195228
Part D: 100 mg Abemaciclib + 100 mg LY302341486.2227
Part D: 150 mg Abemaciclib + 100 or 150 or 200 mg LY3023414159305
Part E: 100 mg Abemaciclib + 200 mg Pembrolizumab125270
Part E: 150 mg Abemaciclib + 200 mg Pembrolizumab114240

[back to top]

Number of Participants With Dose-Limiting Toxicities (DLT) or DLT-equivalent in Part A, B, C, D and E

A DLT defined as adverse event(AE) occurring between Day 1 and Day 21 of Cycle 1 that was considered at least possibly related to either abemaciclib or the combination therapy and fulfilled a criteria selected (using the National Cancer Institute Common Terminology Criteria for Adverse Events,version 4.0 [NCI-CTCAE v 4.0] [NCI 2009]):Grade(Gr)≥3 nonhematological toxicity,Gr4 thrombocytopenia lasting at least 5 days and/or complicated with bleeding,Gr≥3 febrile neutropenia(ntr) and for Part D participants (pts): Gr3 hyperglycemia (fasting) of <5 days, Gr3 hypertriglyceridemia or hyperlipidemia without optimal treatment.A DLT-equivalent defined as AE that would have met the criteria for DLT if it had occurred during Cycle 1 for pts enrolled in dose-escalation phase,but that occurs between 1)Day 1 and Day 21 of Cycle 2 and beyond for a participant enrolled in dose-escalation phase 2) at any time for a participant in dose-expansion phase. (NCT02079636)
Timeframe: Baseline through study completion (Up To 15 Months)

InterventionParticipants (Count of Participants)
Part A:150 Milligram(mg) Abemaciclib + 500 mg/m^2 Pemetrexed1
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed4
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine0
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine5
Part C:150 mg Abemaciclib+10 mg/kg Ramucirumab Day 11
Part C: 200 mg Abemaciclib + 10 mg/kg Ramucirumab Day 14
Part C: 150 mg Abemaciclib + 8 mg/kg Ramucirumab Days 1 and 81
Part C: 150 mg Abemaciclib + 10 mg/kg Ramucirumab Days 1 and 83
Part D: 100 mg Abemaciclib + 100 mg LY30234141
Part D: 150 mg Abemaciclib + 100 mg LY30234141
Part D: 150 mg Abemaciclib + 150 mg LY30234141
Part D: 200 mg Abemaciclib + 150 mg LY30234141
Part D: 150 mg Abemaciclib + 200 mg LY30234142
Part E: 100 mg Abemaciclib + 200 mg Pembrolizumab0
Part E: 150 mg Abemaciclib + 200 mg Pembrolizumab0

[back to top]

Number of Participants Achieving Complete Response (CR) or Partial Response (PR) (Overall Response Rate [ORR]) in Part A, B, C, D and E

ORR is the best response of CR or PR as classified by the investigators according to the Response Evaluation Criteria In Solid Tumors (RECIST v1.1). CR is a disappearance of all target and non-target lesions and normalization of tumor marker level. PR is an at least 30% decrease in the sum of the diameters of target lesions (taking as reference the baseline sum diameter) without progression of not-target lesions or appearance of new lesions. Overall response rate is calculated as a total number of participants with CR or PR divided by the total number of participants with at least 1 measurable lesion, multiplied by 100. (NCT02079636)
Timeframe: Baseline through study completion (Up To 15 Months)

InterventionParticipants (Count of Participants)
Part A:150 Milligram(mg) Abemaciclib + 500 mg/m^2 Pemetrexed0
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed1
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine0
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine1
Part C:150 mg Abemaciclib+10mg/kg Ramucirumab Day11
Part C: 200 mg Abemaciclib + 10 mg/kg Ramucirumab Day 11
Part C: 150 mg Abemaciclib + 8 mg/kg Ramucirumab Days 1 and 80
Part C: 150 mg Abemaciclib + 10 mg/kg Ramucirumab Days 1 and 80
Part D: 100 mg Abemaciclib + 100 mg LY30234140
Part D: 150 mg Abemaciclib + 100 mg LY30234140
Part D: 150 mg Abemaciclib + 150 mg LY30234141
Part D: 200 mg Abemaciclib + 150 mg LY30234140
Part D: 150 mg Abemaciclib + 200 mg LY30234140
Part E: 100 mg Abemaciclib + 200 mg Pembrolizumab0
Part E: 150 mg Abemaciclib + 200 mg Pembrolizumab2

[back to top]

PK: Area Under the Concentration Curve (AUC) of Pemetrexed at Steady State in Part A

Area under the plasma concentration versus time curve from time zero to infinity (AUC[0-∞]) of Pemetrexed in Part A was evaluated. (NCT02079636)
Timeframe: C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

Interventionmicrogram*hour per milliliter (μg*hr/mL) (Geometric Mean)
Part A:150 mg Abemaciclib + 500 mg/m^2 Pemetrexed201
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed198

[back to top]

Progression Free Survival Time in Part A, B, C, D and E

Progression free survival (PFS) defined as the time from the date of randomization to the first evidence of disease progression as defined by response evaluation criteria in solid tumors (RECIST) v1.1 or death from any cause. Progressive Disease (PD) was at least a 20% increase in the sum of the diameters of target lesions, with reference being the smallest sum on study and an absolute increase of at least 5 mm, or unequivocal progression of non-target lesions, or 1 or more new lesions. If a participant does not have a complete baseline disease assessment, then the PFS time was censored at the date of randomization, regardless of whether or not objectively determined disease progression or death has been observed for the participant. If a participant was not known to have died or have objective progression as of the data inclusion cutoff date for the analysis, the PFS time was censored at the last adequate tumor assessment date. PFS time was summarized using Kaplan-Meier estimates. (NCT02079636)
Timeframe: Date of first dose until first documented progression or death (Up To 15 Months)

InterventionMonths (Median)
Part A:Abemaciclib + 500 mg/m^2 Pemetrexed5.55
Part B: Abemaciclib + 1250 mg/m^2 Gemcitabine1.58
Part C:Abemaciclib+ 8/10mg/kg Ramucirumab Day1/84.83
Part D: Abemaciclib + 100 mg/150 mg/ 200 mg LY30234141.87
Part E: 100 mg/150 mg Abemaciclib + 200 mg Pembrolizumab4.11

[back to top]

PK: Maximum Concentration (Cmax) of Ramucirumab at 1 Hour Post-End-of-Infusion in Part C

Cmax of ramucirumab at 1 hour post-end-of-Infusion in Part C was evaluated. (NCT02079636)
Timeframe: C1D1 and C2D1: 1 hour post-end-of-infusion

,,,
Interventionmicrogram per milliliter (μg/mL) (Geometric Mean)
C1D1C2D1
Part C: 150 mg Abemaciclib + 10 mg/kg Ramucirumab Day 1226NA
Part C: 150 mg Abemaciclib + 10 mg/kg Ramucirumab Days 1 and 8193NA
Part C: 150 mg Abemaciclib + 8 mg/kg Ramucirumab Days 1 and 8174221
Part C: 200 mg Abemaciclib + 10mg/kg Ramucirumab Day1221226

[back to top]

PK: Maximum Concentration (Cmax) of LY3023414 in Part D

Cmax of LY3023414 in Part D was evaluated. (NCT02079636)
Timeframe: C1D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,,
Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Day 1Steady State
Part D: 100 or 150 mg Abemaciclib + 100 mg LY3023414298438
Part D: 150 mg Abemaciclib + 200 mg LY3023414578NA
Part D: 150 or 200 mg Abemaciclib + 150 mg LY3023414454491

[back to top]

Change From Baseline in MD Anderson Symptom Inventory Scale-Lung Cancer (MDASI-LC) in Part A, B, C, D and E

"The MDASI-LC is a self-reported lung cancer instrument included 22 items covered by one of the following dimensions: Mean core symptom severity (Core items 1-13: pain, fatigue, nausea, disturbed sleep, distress, shortness of breath, remembering things, lack of appetite, drowsy, dry mouth, sad, vomiting, numbness/tingling), Lung cancer symptoms (3 items: coughing, constipation, sore throat), Mean symptom severity (13 core items plus 3 lung items) and Interference with mood or functional status (6 items: general activity, mood, work, relations with other people, walking, enjoyment of life). The mean of all symptom subscale items was calculated where 0 equals not present and 10 equals as bad as you can imagine. A change from baseline with negative values indicate improvement, positive values indicate worsening." (NCT02079636)
Timeframe: Baseline, through study completion (Up To 15 Months)

,,,,,,,,,,,,,,
Interventionunits on a scale (Mean)
Mean core symptom severityMean symptom severityMean lung cancer symptomMean interference severity
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed0.890.830.710.49
Part A:150 Milligram(mg) Abemaciclib + 500 mg/m^2 Pemetrexed-0.16-0.051.222.32
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine-0.54-0.441.670.00
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine-0.08-0.030.190.95
Part C: 150 mg Abemaciclib + 10 mg/kg Ramucirumab Days 1 and 8-1.39-1.013.01-3.08
Part C: 150 mg Abemaciclib + 8 mg/kg Ramucirumab Days 1 and 8-0.64-0.61-0.78-1.00
Part C: 200 mg Abemaciclib + 10mg/kg Ramucirumab Day10.610.721.130.15
Part C:150 mg Abemaciclib+10mg/kg Ramucirumab Day1-0.35-0.41-0.67-1.33
Part D: 100 mg Abemaciclib + 100 mg LY30234140.610.640.780.72
Part D: 150 mg Abemaciclib + 100 mg LY30234141.891.861.401.47
Part D: 150 mg Abemaciclib + 150 mg LY30234141.601.602.191.81
Part D: 150 mg Abemaciclib + 200 mg LY30234140.280.190.67-0.38
Part D: 200 mg Abemaciclib + 150 mg LY3023414-0.80-0.73-0.44-0.22
Part E: 100 mg Abemaciclib + 200 mg Pembrolizumab0.910.991.671.44
Part E: 150 mg Abemaciclib + 200 mg Pembrolizumab0.880.750.231.00

[back to top]

PK: Area Under the Concentration Curve (AUC) of LY3023414 in Part D

Area under the plasma concentration versus time curve from time zero to infinity (AUC) of LY3023414 in Part D was evaluated. For Day 1, AUC is defined as AUC from time zero to infinity (AUC[0-∞]), for steady state, AUC is defined as AUC from time zero to the end of the dosing interval, tau (AUC[0-tau ]) (NCT02079636)
Timeframe: C1D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,,
Interventionnanogram*hour/mL (ng*h/mL) (Geometric Mean)
Day 1Steady State
Part D: 100 or 150 mg Abemaciclib + 100 mg LY302341410051303
Part D: 150 mg Abemaciclib + 200 mg LY30234143809NA
Part D: 150 or 200 mg Abemaciclib + 150 mg LY302341417511293

[back to top]

PK: Dose-normalized Maximum Concentration (Cmax) of Active Gemcitabine Metabolite: 2',2'-Difluorodeoxyuridine (dFdU) on Day 1 and at Steady State (Cycle 2 Day 1) in Part B

Cmax of active gemcitabine metabolite (dFdU) on day 1 and at steady state (Cycle 2 Day 1) dose-normalized to 1250 mg/m^2 in Part B was evaluated. (NCT02079636)
Timeframe: C1D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,,
Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Day 1Steady State
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine35600NA
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine4960038700
Part B: 200 mg or MTD Abemaciclib + 1250 mg/m^2 Gemcitabine4190035800

[back to top]

PK: Area Under the Concentration Curve (AUC) of Active Gemcitabine Metabolite: 2',2'-Difluorodeoxyuridine (dFdU) on Day 1 and at Steady State (Cycle 2 Day 1) in Part B

Area under the plasma concentration time curve from time zero to 12 hours (AUC[0-12h]) of active gemcitabine metabolite (dFdU) in Part B was evaluated (NCT02079636)
Timeframe: C1D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,,
Interventionnanogram*hour/mL (ng*h/mL) (Geometric Mean)
Day 1Steady State
200 mg or MTD Abemaciclib + 1250 mg/m^2 Gemcitabine198000210000
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine175000NA
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine246000208000

[back to top]

PK: Area Under the Concentration Curve (AUC) of Abemaciclib in Part A, B, C, D and E

Area under the concentration time curve from zero to 8 hours AUC(0-8h) of abemaciclib in Part A, B, C, D and E was evaluated. (NCT02079636)
Timeframe: C1D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,,,,,,,,
Interventionnanogram*hour/mL (ng*h/mL) (Geometric Mean)
Day 1Steady State
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed9793710
Part B: 200 mg Abemaciclib + 1250 mg/m^2 Gemcitabine8862690
Part C: 150 mg Abemaciclib + 8 mg/kg or 10 mg/kg Ramucirumab7971720
Part C: 200 mg Abemaciclib + 10mg/kg Ramucirumab Day110301840
Part D: 100 mg Abemaciclib + 100 mg LY3023414394NA
Part D: 150 mg Abemaciclib + 100 or 150 or 200 mg LY30234147191550
Part A:150 Milligram(mg) Abemaciclib + 500 mg/m^2 Pemetrexed533NA
Part B: 150 mg Abemaciclib + 1250 mg/m^2 Gemcitabine393NA
Part E: 150 mg Abemaciclib + 200 mg Pembrolizumab5181400

[back to top]

Pharmacokinetics (PK): Maximum Concentration (Cmax) of Abemaciclib on Day 1 and at Steady State (Cycle 2 Day 1) in Part A , B, C, D and E

Cmax of Abemaciclib on day 1 and at steady State (Cycle 2 Day 1) Part A , B, C, D and E was evaluated. (NCT02079636)
Timeframe: Cycle 1 Day 1 (C1D1) pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; Cycle 2 Day 1 (C2D1) pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Day 1
Part D: 200 mg Abemaciclib + 150 mg LY3023414225

[back to top]

PK: Area Under the Concentration Curve (AUC) of Abemaciclib in Part A, B, C, D and E

Area under the concentration time curve from zero to 8 hours AUC(0-8h) of abemaciclib in Part A, B, C, D and E was evaluated. (NCT02079636)
Timeframe: C1D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose; C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

,
Interventionnanogram*hour/mL (ng*h/mL) (Geometric Mean)
Day 1
Part E: 100 mg Abemaciclib + 200 mg PembrolizumabNA
Part D: 200 mg Abemaciclib + 150 mg LY3023414967

[back to top]

Pharmacokinetics: Maximum Concentration (Cmax) of Pemetrexed at Steady State in Part A

Cmax of pemetrexed at steady state in Part A was evaluated. (NCT02079636)
Timeframe: C2D1 pre-dose and 1, 2, 4, 6, 8, 10 h post-dose

Interventionmicrogram per milliliter (μg/mL) (Geometric Mean)
Part A:150 mg Abemaciclib + 500 mg/m^2 Pemetrexed98.1
Part A: 200 mg Abemaciclib + 500 mg/m^2 Pemetrexed93.5

[back to top]

Number of Subjects With Dose Limiting Toxicities (DLTs)

DLT: any National Cancer Institute Common Toxicity Criteria for Adverse Events Version 4.03 Grade 4 hematologic or Grade 3/4 non-hematologic toxicities that occurred during DLT observation period and were considered by Investigator to be at least possibly related to trial treatment, and were confirmed by Safety Monitoring Committee (SMC), with exception of Grade 4 neutropenia for not >5 days; Grade 4 lymphocytopenia/ thrombocytopenia for not >5 days; fatigue/headache lasting < 7 days; nausea/vomiting/diarrhoea lasting not >3 days; asymptomatic Grade 3 increase in liver function tests that resolve to baseline within 7 days; Mucositis >= Grade 3 lasting < 7 days; Grade 3 hyperglycemia that resolves in < 7 days; any laboratory values >Grade 3 without any clinical correlate (resolve within 5 days); Grade 3 skin toxicities that resolve to Grade 2 within 7 days; Grade 3/4 hypomagnesemia that resolves within 5 days. Subjects with DLTs presented based on investigator and SMC decision. (NCT02083679)
Timeframe: Day 1 to Day 21 of Cycle 1

,,,
InterventionSubjects (Number)
InvestigatorSMC
Part 1: Sym004 6 mg/kg + Carboplatin/Paclitaxel10
Part 1: Sym004 6 mg/kg + Cisplatin/Gemcitabine22
Part 1: Sym004 6 mg/kg + Cisplatin/Pemetrexed11
Part 1: Sym004 6/12 mg/kg + Carboplatin/Paclitaxel00

[back to top]

Number of Subjects With Treatment-emergent Adverse (TEAEs), Serious TEAEs, TEAEs Leading to Discontinuation and TEAEs Leading to Death

An adverse event (AE) was defined as any untoward medical occurrence in a subject or clinical investigation subject administered a pharmaceutical product, which does not necessarily have a causal relationship with this treatment. An AE was any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease temporally associated with use of a medicinal product, whether or not considered related to the medicinal product. A serious adverse event (SAE) was an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged inpatient hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. AEs were considered treatment emergent if they started on or after the day of first administration of the first trial treatment given (Sym004 or one of the individual Platinum-Doublet therapies) or if they worsened after receiving first dose of treatment. (NCT02083679)
Timeframe: Day 1 up to 28 days after last dose of study drug (up to 53 weeks)

,,,
Interventionsubjects (Number)
TEAEsSerious TEAEsTEAE leading to DiscontinuationTEAEs Leading to Death
Part 1: Sym004 6 mg/kg + Carboplatin/Paclitaxel3300
Part 1: Sym004 6 mg/kg + Cisplatin/Gemcitabine3310
Part 1: Sym004 6 mg/kg + Cisplatin/Pemetrexed6530
Part 1: Sym004 6/12 mg/kg + Carboplatin/Paclitaxel3320

[back to top]

Assess the Objective Response Rates in Each Arm

The objective response rate is defined as the number of the subjects with a confirmed best overall response of CR or PR divided by the number of subjects in the Full Analysis Set (FAS) for whom measureable disease is present at baseline (NCT02087241)
Timeframe: Up to a maximum of 4 treatment cycles (treatment cycles will be repeated every 21 days)

InterventionPercentage of Participants (Number)
Cohort 166.7
Cohort 233.3
Cohort 30
Cohort A14.3

[back to top]

Assess the Disease Control Rate in Each Treatment Arm

the disease control rate is defined as the percentage of FAS subjects with a best overall response of CR, PR or SD). (NCT02087241)
Timeframe: Up to a maximum of 4 treatment cycles (treatment cycles will be repeated every 21 days)

InterventionPercentage of Participants (Number)
Cohort 166.7
Cohort 266.7
Cohort 30
Cohort A85.7

[back to top]

Overall Survival

To assess the efficacy of pemetrexed in combination with TH-302 as determined by overall survival in patients with advanced non-squamous NSCLC in the second-line chemotherapy setting compared with pemetrexed in combination with placebo (NCT02093962)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
TH-302 and Pemetrexed51
Placebo and Pemetrexed49

[back to top]

Progression-Free Survival (PFS)

Evaluate immune-related PFS (irPFS) and PFS by RECIST (Response Evaluation Criteria for Solid Tumors) (NCT02117024)
Timeframe: Up to 3 years

,
InterventionDays (Median)
immune-related PFS (irPFS)Progression Free Survival (PFS)
Chemotherapy Alone190.0190.0
Viagenpumatucel-L Plus Metronomic Cyclophosphamide76.070.0

[back to top]

Overall Survival (OS)

"Overall survival (OS) calculated as the duration of survival from the date of randomization to the date of death from any cause, or was censored on the date the patient was last known to be alive.~Survival time was calculated from the randomization date up to the date of death,or censored on the date that the patient was last known to be alive (last available visit date) utilizing Kaplan-Meier Estimate of Overall Survival Ending Events" (NCT02117024)
Timeframe: Up to 3 years

InterventionDays (Median)
Viagenpumatucel-L Plus Metronomic Cyclophosphamide176
Chemotherapy Alone372

[back to top]

Frequency of Adverse Events: Number of Participants With Treatment-Emergent Adverse Events (TEAE)

Evaluate the safety of the combination of viagenpumatucel-L and low-dose cyclophosphamide by frequency of Treatment-Emergent Adverse Events (NCT02117024)
Timeframe: Up to 3 years

,
InterventionParticipants (Count of Participants)
At least one TEAEAt least one severe TEAEAt least one treatment-related TEAEAt least one SAEFatal TEAEAt least one TEAE Leading to Tx DiscontinuationAt least one TEAE Leading to a Dose Reduction
Chemotherapy Alone2011158022
Viagenpumatucel-L Plus Metronomic Cyclophosphamide41253217770

[back to top]

Survival at 6 Months

Evaluate the proportion of patients who are alive at 6 months following randomization (NCT02117024)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Viagenpumatucel-L Plus Metronomic Cyclophosphamide21
Chemotherapy Alone17

[back to top]

Survival at 12 Months

Evaluate the proportion of patients who are alive at 12 months following randomization (NCT02117024)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Viagenpumatucel-L Plus Metronomic Cyclophosphamide8
Chemotherapy Alone11

[back to top]

Time to Progression (TTP)

Evaluate immune-related TTP (irTTP) and also TTP (Time to Progression) by RECIST (NCT02117024)
Timeframe: Up to 3 years

,
InterventionDays (Median)
immune-related TTP (irTTP)Time to Progression (TTP)
Chemotherapy Alone71.073.5
Viagenpumatucel-L Plus Metronomic Cyclophosphamide67.067.5

[back to top]

Progression-free Survival (PFS)

PFS is defined as the time from randomization until the earliest date of disease progression determined by investigator assessment of objective radiographic disease assessments per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1, or death due to any cause if sooner. Progressive Disease (PD) is defined using Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum Longest Diameter (LD) recorded since the treatment started or the appearance of one or more new lesions, unequivocal progression of non-target lesions or increase in disease burden for subjects with only nonmeasurable disease. (NCT02119650)
Timeframe: Randomization to disease progression, or death due to any cause if sooner; up to 16 months or to the data cutoff 11FEB2016.

InterventionMonths (Median)
Double-Blind Treatment: Ruxolitinib + Pemetrexed/CisplatinNA
Double-Blind Treatment: Placebo Plus Pemetrexed/CisplatinNA

[back to top]

Duration of Response

For objective responders, the duration of response is defined as the difference of the end of response and the start of response. The start of a response was the first visit where the subject achieves PR or better based on RECIST v1.1 criteria. The end of response was the first visit after PD based on RECIST v1.1 criteria. (NCT02119650)
Timeframe: From the start of response to the end of response; up to 16 months or to the data cutoff 11FEB2016.

Interventionweeks (Median)
Double-Blind Treatment: Ruxolitinib + Pemetrexed/Cisplatin20.14
Double-Blind Treatment: Placebo Plus Pemetrexed/Cisplatin12.14

[back to top]

Overall Survival (OS)

Overall survival is defined as the time from randomization to death due to any cause. Participants without death observed at the time of the analysis were censored at last date known to be alive. The median overall survival time was estimated using the Kaplan-Meier method. Overall survival was compared between treatment groups using log-rank test. (NCT02119650)
Timeframe: Randomization until death due to any cause; up to 16 months or data cutoff 11FEB2016.

Interventionmonths (Median)
Double-Blind Treatment: Ruxolitinib + Pemetrexed/Cisplatin7.5
Double-Blind Treatment: Placebo Plus Pemetrexed/Cisplatin5.9

[back to top]

Objective Response Rate (ORR)

Objective response rate determined by radiographic disease assessments per RECIST (v1.1), by investigator assessment and was defined as the percentage of participants with Complete Response (CR) or Partial Response (PR) by Response Evaluation Criteria in Solid Tumours (RECIST) at any post baseline visit. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST) for target lesions and assessed by computed tomography (CT) and/or magnetic resonance imaging (MRI) : Complete Response (CR), Disappearance of all target and non-target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions with no worsening of non-target lesions and no new lesions; Overall Response (OR) = CR + PR. (NCT02119650)
Timeframe: Baseline through end of study; up to 16 months or to the data cutoff 11FEB2016.

,
InterventionParticipants (Count of Participants)
Overall ResponseComplete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnable to EvaluateNot Assessed
Double-Blind Treatment: Placebo Plus Pemetrexed/Cisplatin1301354312
Double-Blind Treatment: Ruxolitinib + Pemetrexed/Cisplatin1201246215

[back to top]

Participants With Treatment-emergent Adverse Events (TEAEs)

A treatment-emergent AE was defined as an event occurring (or worsening of any pre-existing) after exposure to at least 1 dose of study drug. A treatment-related AE was defined as an event with a definite, probable, or possible causality to study medication. A serious AE is an event resulting in death, hospitalization, persistent or significant disability/incapacity, or is life threatening, a congenital anomaly/birth defect or requires medical or surgical intervention to prevent 1 of the outcomes above. The intensity of an AE was graded according to the National Cancer Institute common terminology criteria for adverse events (NCI-CTCAE) version 4.03: Grade 1 (Mild); Grade 2 (Moderate); Grade 3 (Severe); Grade 4 (life-threatening). (NCT02119650)
Timeframe: Baseline through approximately 30 days post treatment discontinuation; up to 16 months or to the data cutoff 11FEB2016.

,
InterventionParticipants (Count of Participants)
Participants who had any TEAEsParticipants who had treatment-related TEAEsParticipants with any serious TEAEParticipants who had Grade 3 or higher TEAEsParticipants with a fatal TEAETEAEs related to reference therapyParticipants who were hospitalized due to TEAEsParticipants who discontinued drug due to TEAEsParticipants who interrupted drug due to TEAEsDiscontinued reference therapy due to TEAEsInterrupted reference therapy due to TEAEsParticipants given concomitant meds due to TEAEsProcedure/nondrug therapy due to TEAEs
Double-Blind Treatment: Placebo Plus Pemetrexed/Cisplatin3628162243515415773212
Double-Blind Treatment: Ruxolitinib + Pemetrexed/Cisplatin3916192543016211483617

[back to top]

Objective Response Rate (ORR)

ORR was defined as the percentage of participants in the analysis population who experienced 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) and was assessed using RECIST 1.1 based on BICR evaluation. ORR was assessed from enrollment/treatment initiation of a participant through data cutoff of 09-May-2016. The ORR is presented for each treatment group. (NCT02142738)
Timeframe: Up to ~1.6 years

InterventionPercentage of Participants (Number)
Pembrolizumab44.8
SOC Chemotherapy27.8

[back to top]

Overall Survival (OS) Rate

OS was defined as the time from randomization to death due to any cause. Participants without documented death at the time of the analysis were censored at the date of the last follow-up. The data cutoff was 10-July-2017. The median OS rate at 12 months is presented. (NCT02142738)
Timeframe: 12 months

InterventionPercentage of Participants (Number)
Pembrolizumab70.3
SOC Chemotherapy54.8

[back to top]

Progression Free Survival (PFS) Rate at Month 6

PFS was defined as the time from randomization to documented disease progression per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1) or death due to any cause, whichever occurred first and was based on blinded independent central radiologists' (BICR) review. Progressive Disease (PD) was defined as ≥20% increase in the sum of diameters of target lesions and an absolute increase of ≥5 mm. (Note: the appearance of one or more new lesions was also considered progression). Participants were evaluated every 9 weeks with radiographic imaging to assess their response to treatment. The data cutoff was 09-May-2016. The PFS rate at Month 6 was calculated. (NCT02142738)
Timeframe: Month 6

InterventionPercentage of Participants (Number)
Pembrolizumab62.1
SOC Chemotherapy50.3

[back to top]

Overall Survival (OS)

Since, the study was canceled due to slow accrual, no biomarkers were measured; therefore, no analysis with biomarkers. Only the OS will be calculated. (NCT02145078)
Timeframe: From the date of protocol-specified treatment initiation to the date of death or last observation, assessed up to 12 months

Interventionmonths (Median)
Treatment (Chemotherapy Regimen)2.0

[back to top]

Progression-free Survival (PFS)

Each biomarker evaluated using its expression level (continuous variable) and dichotomous form (based on a median cutoff). A univariable Cox regression model will be used to assess the relationship of expression levels of each biomarker to PFS. In addition, for the dichotomous variables, the sample will be divided into those above and below the median for each biomarker. PFS probabilities for each group will be estimated using the Kaplan-Meier method, with standard errors based on Greenwood's formula. Log rank tests will be used to determine the level of significance between survival curves. Since, the study was canceled due to slow accrual, no biomarkers were measured; therefore, no analysis with biomarkers. Only the PFS will be calculated. (NCT02145078)
Timeframe: From the date of protocol-specified treatment initiation to the date of progression, death, or last observation, assessed up to 12 months

Interventiondays (Median)
Treatment (Chemotherapy Regimen)42

[back to top]

Number of Participants Who Experienced At Least One Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a participant administered a study treatment and which does not necessarily have to have a causal relationship with this treatment. An AE could therefore be any unfavorable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the study treatment or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a pre-existing condition that was temporally associated with the use of study treatment, was also an AE. The number of participants who experienced at least one AE is presented. (NCT02220894)
Timeframe: Up to approximately 38 months

InterventionParticipants (Count of Participants)
Pembrolizumab608
Chemotherapy (SOC Treatment)606

[back to top]

Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%

ORR was determined for participants with a TPS of ≥1%. ORR was determined per RECIST 1.1 and was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was ORR in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The percentage of participants who had a TPS ≥1% and who experienced a CR or PR is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionPercentage of participants (Number)
Pembrolizumab27.2
Chemotherapy (SOC Treatment)26.5

[back to top]

Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%

ORR was determined for participants with a TPS of ≥20%. ORR was determined per RECIST 1.1 and was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was ORR in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The percentage of participants who had a TPS ≥20% and who experienced a CR or PR is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionPercentage of participants (Number)
Pembrolizumab33.2
Chemotherapy (SOC Treatment)28.9

[back to top]

Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%

ORR was determined for participants with a TPS of ≥50%. ORR was determined per RECIST 1.1 and was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was ORR in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The percentage of participants who had a TPS ≥50% and who experienced a CR or PR is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionPercentage of participants (Number)
Pembrolizumab39.1
Chemotherapy (SOC Treatment)32.0

[back to top]

Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥20%

OS was determined for participants with a TPS of ≥20% and was defined as the time from randomization to death due to any cause. Participants without documented death at the time of the interim analysis were censored at the date of the last follow-up. The OS was calculated using the product-limit (Kaplan-Meier) method for censored data. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was OS in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The OS for participants with a TPS ≥20% is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionMonths (Median)
Pembrolizumab18.0
Chemotherapy (SOC Treatment)13.0

[back to top]

Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥50%

OS was determined for participants with a TPS of ≥50% and was defined as the time from randomization to death due to any cause. Participants without documented death at the time of the interim analysis were censored at the date of the last follow-up. The OS was calculated using the product-limit (Kaplan-Meier) method for censored data. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was OS in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The OS for participants with a TPS ≥50% is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionMonths (Median)
Pembrolizumab20.0
Chemotherapy (SOC Treatment)12.2

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%

PFS was determined for participants with a TPS of ≥1% and was defined as the time from randomization to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. The appearance of one or more new lesions was also considered PD. The PFS per RECIST 1.1 was calculated using the product-limit (Kaplan-Meier) method for censored data. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was PFS in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The PFS for participants with a TPS ≥1% is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionMonths (Median)
Pembrolizumab5.4
Chemotherapy (SOC Treatment)6.6

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%

PFS was determined for participants with a TPS of ≥20% and was defined as the time from randomization to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. The appearance of one or more new lesions was also considered PD. The PFS per RECIST 1.1 was calculated using the product-limit (Kaplan-Meier) method for censored data. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was PFS in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The PFS for participants with a TPS ≥20% is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionMonths (Median)
Pembrolizumab6.2
Chemotherapy (SOC Treatment)6.7

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%

PFS was determined for participants with a TPS of ≥50% and was defined as the time from randomization to the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. The appearance of one or more new lesions was also considered PD. The PFS per RECIST 1.1 was calculated using the product-limit (Kaplan-Meier) method for censored data. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was PFS in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The PFS for participants with a TPS ≥50% is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionMonths (Median)
Pembrolizumab6.5
Chemotherapy (SOC Treatment)6.4

[back to top]

Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥1%

OS was determined for participants with a TPS of ≥1% and was defined as the time from randomization to death due to any cause. Participants without documented death at the time of the interim analysis were censored at the date of the last follow-up. The OS was calculated using the product-limit (Kaplan-Meier) method for censored data. The efficacy hypothesis was analyzed using a sequential testing strategy that involved testing a hypothesis only if the superiority of pembrolizumab over chemotherapy was established for all the preceding hypotheses. The order of testing was OS in participants with TPS≥50%, then with TPS≥20%, and finally with TPS≥1%. The OS for participants with a TPS ≥1% is presented. (NCT02220894)
Timeframe: Up to approximately 44 months

InterventionMonths (Median)
Pembrolizumab16.4
Chemotherapy (SOC Treatment)12.1

[back to top]

Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a participant administered a study treatment and which did not necessarily have to have a causal relationship with this treatment. An AE could therefore be any unfavorable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the study treatment or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a pre-existing condition that was temporally associated with the use of study treatment, was also an AE. The number of participants who discontinued study treatment due to an AE is presented. (NCT02220894)
Timeframe: Up to approximately 35 months

InterventionParticipants (Count of Participants)
Pembrolizumab126
Chemotherapy (SOC Treatment)93

[back to top]

Progression Free Survival (PFS) in All Participants

"Progression-free survival is defined as the time from the date of randomization to the date of disease progression (PD) per RECIST version 1.1 or death (all causes of mortality), whichever occurred first.~PD: At least a 20% increase in the size of target lesions, taking as reference the smallest size recorded since the treatment started (Baseline or after) with an absolute increase of at least 5 mm, the appearance of one or more new lesions, or unequivocal progression of existing non-target lesions.~PFS was estimated using Kaplan-Meier methodology. Participants who did not have an event of disease progression or had not died on or before the cut-off date were censored at the date of their last disease progression assessment on or before the cut-off date. Any PD and death occurring > 26 weeks and > 12 weeks after the previous assessment, respectively, were excluded and patients were censored at last assessment before PD or death." (NCT02264990)
Timeframe: From randomization up to the data cut-off date of 15 July 2019; the median follow-up time was 45.4 and 44.6 months in all participants for the investigator's choice chemotherapy and veliparib + C/P arms, respectively.

Interventionmonths (Median)
Investigator's Choice Chemotherapy6.7
Veliparib + Carboplatin + Paclitaxel5.9

[back to top]

Overall Survival (OS) in the Lung Subtype Panel Positive Subgroup

Overall survival is defined as the time from the date that the participant was randomized to the date of the participant's death. Overall survival was estimated using Kaplan-Meier methodology. Participants still alive at the data cut-off date were censored at the date they were last known to be alive. (NCT02264990)
Timeframe: From randomization up to the data cut-off date of 15 July 2019; median follow-up time was 44.5 and 45.3 months in LSP+ participants for the investigator's choice chemotherapy and veliparib + C/P arms, respectively.

Interventionmonths (Median)
Investigator's Choice Chemotherapy9.2
Veliparib + Carboplatin + Paclitaxel11.2

[back to top]

Overall Survival in All Participants

Overall survival is defined as the time from the date that the participant was randomized to the date of the participant's death. OS was estimated using Kaplan-Meier methodology. Participants still alive at the data cut-off date were censored at the date they were last known to be alive. (NCT02264990)
Timeframe: From randomization up to the data cut-off date of 15 July 2019; the median OS follow-up time was 45.4 and 44.6 months in all participants for the investigator's choice chemotherapy and veliparib + C/P arms, respectively.

Interventionmonths (Median)
Investigator's Choice Chemotherapy12.1
Veliparib + Carboplatin + Paclitaxel12.1

[back to top]

Objective Response Rate (ORR) in the Lung Subtype Panel Positive Subgroup

"Objective response rate is defined as the percentage of participants with a complete response (CR) or partial response (PR) per Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1 criteria. Response must have been confirmed at a consecutive assessment 28 days or more after the assessment at which response was first observed.~CR: The disappearance of all target and non-target lesions and no new lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the Baseline sum diameters, persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits, or any new lesions." (NCT02264990)
Timeframe: Assessed on Day 1 of Cycles 3 and 5 then every 9 weeks for 1 year or until maintenance therapy was discontinued, then every 12 weeks until radiographic progression or death; median time on follow-up was 5.2 and 6.3 months in each group, respectively.

Interventionpercentage of participants (Number)
Investigator's Choice Chemotherapy30.0
Veliparib + Carboplatin + Paclitaxel22.5

[back to top]

Objective Response Rate (ORR) in All Participants

"Objective response rate is defined as the percentage of participants with a complete response (CR) or partial response (PR) per RECIST version 1.1 criteria. Response must have been confirmed at a consecutive assessment 28 days or more after the assessment at which response was first observed.~CR: The disappearance of all target and non-target lesions and no new lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR: At least a 30% decrease in the sum of diameters of target lesions, taking as reference the Baseline sum diameters, persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits, or any new lesions." (NCT02264990)
Timeframe: Assessed on Day 1 of Cycles 3 and 5 then every 9 weeks for 1 year or until maintenance therapy was discontinued, then every 12 weeks until radiographic progression or death; median time on follow-up was 6.7 and 5.9 months in each group, respectively.

Interventionpercentage of participants (Number)
Investigator's Choice Chemotherapy29.0
Veliparib + Carboplatin + Paclitaxel26.2

[back to top]

Progression Free Survival (PFS) in the Lung Subtype Panel Positive Subgroup

"Progression-free survival is defined as the time from the date of randomization to the date of disease progression (PD) per Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1 or death (all causes of mortality), whichever occurred first.~PD: At least a 20% increase in the size of target lesions, taking as reference the smallest size recorded since the treatment started (Baseline or after) with an absolute increase of at least 5 mm, the appearance of one or more new lesions, or unequivocal progression of existing non-target lesions.~PFS was estimated using Kaplan-Meier methodology. Participants who did not have an event of disease progression or had not died on or before the cutoff date were censored at the date of their last disease progression assessment on or before the cut-off date. Any PD and death occurring > 26 weeks and > 12 weeks after the previous assessment, respectively, were excluded and patients were censored at last assessment before PD or death." (NCT02264990)
Timeframe: From randomization up to the data cut-off date of 15 July 2019; the median follow-up time was 44.5 and 45.3 months in LSP+ participants for the investigator's choice chemotherapy and veliparib + C/P arms, respectively.

Interventionmonths (Median)
Investigator's Choice Chemotherapy5.2
Veliparib + Carboplatin + Paclitaxel6.3

[back to top]

Overall Survival

Overall survival from enrollment to 12 months from enrollment measured in months (NCT02274038)
Timeframe: 1 year from study enrollment

Interventionmonths (Mean)
18F-thymidine (FLT) PET/CT4.1

[back to top]

Duration of Response (DOR) According to RECIST Version 1.1 as Determined by Investigator Assessment

DOR in patients with confirmed response per investigator. The DOR for complete response (CR) and partial response (PR) was measured from date that any of these best responses is first recorded until first date that progressive disease (PD) is objectively documented. For patients who continue treatment post-progression, the first date of progression was used for the analysis. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions, defined by and assessed as: CR is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10mm. PR is at least a 30% decrease in sum of the longest diameter of target lesions, taking as reference the baseline sum of longest diameter. Overall Response is the best response from start of the treatment until disease progression/recurrence (taking as reference for progressive disease the smallest measurements recorded since the treatment started). (NCT02322281)
Timeframe: Cycle 1 Day 1 to End of Treatment, up to approximately 35 months

InterventionDays (Median)
Rociletinib 500 mg BID335.0
Rociletinib 625 mg BID275.0
Chemotherapy206.0

[back to top]

Overall Survival (OS)

OS was calculated as 1+ the number of days from randomization to death due to any cause. Patients without a documented date of death were censored on the date the patient was last known to be alive. (NCT02322281)
Timeframe: Cycle 1 Day 1 to date of death, assessed up to 3 years

InterventionDays (Median)
Rociletinib 500 mg BID665
Rociletinib 625 mg BID541
Chemotherapy348

[back to top]

Percentage of Participants With Confirmed Response

Percentage of patients with a best overall confirmed response of partial response (PR) or complete response (CR) recorded from the start of the treatment until disease progression or recurrence. Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1) for target lesions, defined by and assessed as: Complete Response (CR), is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm. Partial Response (PR),at least a 30% decrease in the sum of the longest diameter of target lesions, taking as reference the baseline sum of longest diameter. Overall Response (OR),is the best response recorded from the start of the treatment until disease progression/recurrence (taking as reference for progressive disease the smallest measurements recorded since the treatment started). The patient's best response assignment was dependent on the achievement of both measurement and confirmation criteria. (NCT02322281)
Timeframe: Cycle 1 Day 1 to End of Treatment, up to approximately 35 months. This Time Frame includes the cross-over period, however, participants who crossed over to rociletinib were not analyzed for best overall confirmed response.

Interventionpercentage of participants (Number)
Rociletinib 500 mg BID17.0
Rociletinib 625 mg BID18.2
Chemotherapy8.2

[back to top]

Progression Free Survival (PFS) According to RECIST Version 1.1 as Determined by Investigator Review (invPFS)

PFS was calculated as 1+ the number of days from the date of randomization to documented radiographic progression as determined by the investigator, or death due to any cause, whichever occurs first. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.1), as at least a 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm.The appearance of one or more new lesions is also considered progression. (NCT02322281)
Timeframe: Cycle 1 Day 1 to End of Treatment, up to approximately 35 months. This Time Frame includes the cross-over period, however, participants who crossed over to rociletinib were not analyzed for PFS.

InterventionDays (Median)
Rociletinib 500 mg BID125.0
Rociletinib 625 mg BID166.0
Chemotherapy77.0

[back to top]

Plasma PK for Patients Treated With Rociletinib Based on Sparse Sampling

Blood samples were drawn for PK analysis at 21 ± 3 day intervals for the first 6 months (Day 1 of Cycles 2 to 7 inclusive). The sample could be taken predose or postdose. Plasma concentrations are presented for Rociletinib and 3 metabolites (M460, M502, M544). (NCT02322281)
Timeframe: Cycles 2 Day 1 to Cycle 7 Day 1, or approximately 6 months

,
InterventionPlasma concentration (ng/mL) (Median)
RociletinibM460M502M544
Rociletinib 500 mg BID80.420.0573.0765.0
Rociletinib 625 mg BID207.0555.03260.0525.0

[back to top]

Progression Free Survival

Defined as the time from randomization to the first objective documentation of disease progression, defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions, or death due to any cause with patients who had not progressed or died at the time of final analysis censored on the date of the last tumour assessment. (NCT02337530)
Timeframe: 3 years

Interventionmonths (Median)
Intermittent Oral Selumatinib With Pemetrexed and Platinum7.2
Continuous Oral Selumatinib With Pemetrexed and Platinum6.9
Pemetrexed and Platinum Alone4.0

[back to top]

Overall Survival(OS)

OS was defined as the time from BBI608 administration to death from any cause. Participants alive at final observation or lost to follow-up were censored at their last contact (i.e., visit or telephone) date. (NCT02347917)
Timeframe: From BBI608 administration to death from any cause, up to 31 months

Interventionmonth (Median)
NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)19.81
MPM: BBI608 + Pem +CDDP (Phase 1 Part)30.1
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)NA
MPM: BBI608 + Pem + CDDP (Phase 2 Part)12.14
MPM: BBI608 + Pem + CDDP (Phase 2 Part Including 1 Participant With MPM in Phase 1 Part)12.75

[back to top]

Phase 1 Part: Number of Participants With Dose-limiting Toxicities (DLTs)

"DLT was defined as an adverse event meeting any of the following that occurred during the DLT evaluation period in any participants given BBI608 with the causal relationship to BBI608 assessed as Definite, Probable, or Possible. The severity of adverse events was graded according to the CTCAE v4.0-JCOG.~Grade 4 neutropenia persisting for ≥ 7 days~Grade ≥ 3 febrile neutropenia persisting for ≥ 5 days~Grade 3 thrombocytopenia requiring platelet transfusions, grade 4 thrombocytopenia~Grade ≥ 3 non-hematotoxicity except the following:~Inappetence, nausea, vomiting and electrolyte abnormality which, within 3 days of onset, improved to grade ≤ 2 or resolved after appropriate treatment~Diarrhoea and fatigue which, within 5 days of onset, improved to grade ≤ 2 or resolved after appropriate treatment~Other clinically significant signs in the opinion of the investigator" (NCT02347917)
Timeframe: From Day 1 of Cycle 1 to Day 24 pre-dose examination (23 days)

InterventionParticipants (Count of Participants)
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)0

[back to top]

Phase 2 Part: Progression-free Survival (PFS)

PFS was defined as the time from BBI608 administration to documented PD (as assessed according to the mRECIST or RECIST 1.1) or death, whichever is earlier. The result of imaging assessment by the imaging assessment committee was used for phase 2 part. (NCT02347917)
Timeframe: From BBI608 administration to documented PD or death, whichever is earlier, about 17 months

Interventionmonth (Median)
MPM: BBI608 + Pem + CDDP (Phase 2 Part)5.59
MPM: BBI608 + Pem + CDDP (Phase 2 Part Including 1 Participant With MPM in Phase 1 Part)5.59

[back to top]

Phase 1 Part: Area Under the Concentration-time Curve

AUC0-12: Area under the concentration-time curve from time zero to 12 hours, AUC0-24: Area under the concentration-time curve from time zero to 24 hours, AUC0-inf: Area under the concentration-time curve from time zero to infinity (NCT02347917)
Timeframe: Cycle 1 Day 1 and Day 23

Interventionh*ng/mL (Mean)
Day 1: AUC0-12Day 1: AUC0-24Day 1: AUC0-infDay 23: AUC0-12Day 23: AUC0-24Day 23: AUC0-inf
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)1814.32223.82413.53797.06103.710762.7

[back to top]

Phase 1 Part: Maximum Observed Concentration (Cmax) and Minimum Observed Concentration (Cmin) of BBI608 When Administered With Pem and CDDP

(NCT02347917)
Timeframe: Cycle 1 Day 1 (Cmax only) and Day 23

Interventionng/mL (Mean)
Day 1: CmaxDay 23: CmaxDay 23: Cmin
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)307.5473.7196.1

[back to top]

Phase 1 Part: Number of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs) and Adverse Drug Reactions (ADRs)

"An AE is any untoward medical occurrence in a study subject administered an investigational drug and which does not necessarily have a causal relationship with this treatment.~A SAE was an AE that met one or more of the following criteria:~Results in death~Is life-threatening~Requires hospitalization or prolongation of existing hospitalization~Results in persistent or significant disability or incapacity~Is a congenital anomaly or birth defect~Is an important medical event that may jeopardize the subject or may require a medical or surgical intervention to prevent one of the outcomes listed above. Examples of such medical events include allergic bronchospasm requiring intensive treatment in an emergency room or at home, blood dyscrasias or convulsions that do not result in inpatient hospitalization.~An ADR was defined as adverse events assessed to be related to the investigational drug" (NCT02347917)
Timeframe: Between initial dosing of the investigational drug and final evaluation in the follow-up observation period, about 17 months

InterventionParticipants (Count of Participants)
Any AEsAEs leading to deathSerious AEsAEs leading to drug withdrawn (BBI608)AEs leading to drug interrupted (BBI608)AEs leading to dose reduced (BBI608)Any ADRsADRs leading to deathSerious ADRsADRs leading to drug withdrawn (BBI608)ADRs leading to drug interrupted (BBI608)ADRs leading to dose reduced (BBI608)
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)400110400110

[back to top]

Respiratory Function Tests (Forced Expiratory Volume in the First Second [FEV1])

(NCT02347917)
Timeframe: Every 6 weeks from the first dose of BBI608 until Week 30, and then every 9 weeks from Week 31 [Actually up to Week 111]

,
InterventionL (Median)
The change from baseline in FEV1: 6 weeksThe change from baseline in FEV1: 12 weeksThe change from baseline in FEV1: 18 weeksThe change from baseline in FEV1: 24 weeksThe change from baseline in FEV1: 30 weeksThe change from baseline in FEV1: 39 weeksThe change from baseline in FEV1: 48 weeksThe change from baseline in FEV1: 57 weeksThe change from baseline in FEV1: 66 weeksThe change from baseline in FEV1: 75 weeksThe change from baseline in FEV1: 84 weeksThe change from baseline in FEV1: 93 weeksThe change from baseline in FEV1: 102 weeksThe change from baseline in FEV1: 111 weeks
MPM: BBI608 + Pem + CDDP (Phase 2 Part Including 1 Participant With MPM in Phase 1 Part)0.050-0.130-0.110-0.040-0.030-0.0500.1150.0100.085-0.045-0.130-0.215NANA
MPM: BBI608 + Pem + CDDP (Phase 2 Part)0.050-0.130-0.110-0.040-0.030-0.0500.1150.0100.085-0.045-0.130-0.215NANA

[back to top]

Respiratory Function Tests (Forced Expiratory Volume in the First Second [FEV1])

(NCT02347917)
Timeframe: Every 6 weeks from the first dose of BBI608 until Week 30, and then every 9 weeks from Week 31 [Actually up to Week 111]

,
InterventionL (Median)
The change from baseline in FEV1: 6 weeksThe change from baseline in FEV1: 12 weeksThe change from baseline in FEV1: 18 weeksThe change from baseline in FEV1: 24 weeksThe change from baseline in FEV1: 30 weeksThe change from baseline in FEV1: 39 weeksThe change from baseline in FEV1: 48 weeksThe change from baseline in FEV1: 57 weeksThe change from baseline in FEV1: 66 weeks
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)0.1600.0700.000-0.010-0.055-0.140NANANA
NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)0.1600.0700.000-0.010-0.055-0.140NANANA

[back to top]

Response Rate (RR) and Disease Control Rate (DCR)

"Response rate (RR): Proportion of subjects whose best overall response is CR or PR.~Disease control rate (DCR): Proportion of subjects whose best overall response is CR, PR or SD.~The result of imaging assessment by study site was used for phase 1 part, and the result of imaging assessment by the imaging assessment committee was used for phase 2 part." (NCT02347917)
Timeframe: From BBI608 administration to death from any cause, about 17 months

,,,,
InterventionParticipants (Count of Participants)
Response rate (RR)Disease control rate (DCR)
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)03
MPM: BBI608 + Pem + CDDP (Phase 2 Part Including 1 Participant With MPM in Phase 1 Part)819
MPM: BBI608 + Pem + CDDP (Phase 2 Part)819
MPM: BBI608 + Pem +CDDP (Phase 1 Part)00
NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)03

[back to top]

Respiratory Function Tests (Vital Capacity [VC] and Forced Vital Capacity [FVC])

(NCT02347917)
Timeframe: Every 6 weeks from the first dose of BBI608 until Week 30, and then every 9 weeks from Week 31 [Actually up to Week 111]

,
InterventionmL (Median)
The change from baseline in VC: 6 weeksThe change from baseline in VC: 12 weeksThe change from baseline in VC: 18 weeksThe change from baseline in VC: 24 weeksThe change from baseline in VC: 30 weeksThe change from baseline in VC: 39 weeksThe change from baseline in VC: 48 weeksThe change from baseline in VC: 57 weeksThe change from baseline in VC: 66 weeksThe change from baseline in VC: 75 weeksThe change from baseline in VC: 84 weeksThe change from baseline in VC: 93 weeksThe change from baseline in VC: 102 weeksThe change from baseline in VC: 111 weeksThe change from baseline in FVC: 6 weeksThe change from baseline in FVC: 12 weeksThe change from baseline in FVC: 18 weeksThe change from baseline in FVC: 24 weeksThe change from baseline in FVC: 30 weeksThe change from baseline in FVC: 39 weeksThe change from baseline in FVC: 48 weeksThe change from baseline in FVC: 57 weeksThe change from baseline in FVC: 66 weeksThe change from baseline in FVC: 75 weeksThe change from baseline in FVC: 84 weeksThe change from baseline in FVC: 93 weeksThe change from baseline in FVC: 102 weeksThe change from baseline in FVC: 111 weeks
MPM: BBI608 + Pem + CDDP (Phase 2 Part)30.00.0-70.0-50.020.030.015.0-20.0300.0175.0145.0-205.0NANA35.010.0-140.0-50.0150.0-35.070.060.0300.050.010.0-230.0NANA
MPM: BBI608 + Pem + CDDP (Phase 2 Part Including 1 Participant With MPM in Phase 1 Part)30.00.0-70.0-50.020.030.015.0-20.0300.0175.0145.0-205.0NANA35.010.0-140.0-50.0150.0-35.070.060.0300.050.010.0-230.0NANA

[back to top]

Respiratory Function Tests (Vital Capacity [VC] and Forced Vital Capacity [FVC])

(NCT02347917)
Timeframe: Every 6 weeks from the first dose of BBI608 until Week 30, and then every 9 weeks from Week 31 [Actually up to Week 111]

,
InterventionmL (Median)
The change from baseline in VC: 6 weeksThe change from baseline in VC: 12 weeksThe change from baseline in VC: 18 weeksThe change from baseline in VC: 24 weeksThe change from baseline in VC: 30 weeksThe change from baseline in VC: 39 weeksThe change from baseline in VC: 48 weeksThe change from baseline in VC: 57 weeksThe change from baseline in VC: 66 weeksThe change from baseline in FVC: 6 weeksThe change from baseline in FVC: 12 weeksThe change from baseline in FVC: 18 weeksThe change from baseline in FVC: 24 weeksThe change from baseline in FVC: 30 weeksThe change from baseline in FVC: 39 weeksThe change from baseline in FVC: 48 weeksThe change from baseline in FVC: 57 weeksThe change from baseline in FVC: 66 weeks
MPM or NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)70.0110.0-60.0-290.0-240.0-305.0NANANA0.080.0-20.0-155.0-265.0-345.0NANANA
NSCLC: BBI608 + Pem + CDDP (Phase 1 Part)70.0110.0-60.0-290.0-240.0-305.0NANANA0.080.0-20.0-155.0-265.0-345.0NANANA

[back to top]

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

AEs included both non-SAEs and SAEs and the same participant can have both SAEs and as well non-SAEs. (NCT02357147)
Timeframe: Baseline up to 3 years

,,,
InterventionParticipants (Count of Participants)
AEsSAEs
Combination Treatment Phase: Placebo + Pemetrexed + Cisplatin5211
Combination Treatment Phase:Amatuximab + Pemetrexed +Cisplatin5015
Maintenance Treatment Phase: Amatuximab191
Maintenance Treatment Phase: Placebo214

[back to top]

Percentage of Participants With Adverse Events

Percentage of participants with at least one adverse event. Adverse event onset date before cross over. (NCT02367781)
Timeframe: Up to approximately 69 months after first patient enrolled

InterventionPercentage of participants (Number)
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)99.6
Arm B (Nab-Paclitaxel+Carboplatin)98.7

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms Score Using the Symptoms in Lung Cancer (SILC) Scale

Change from baseline per SILC scale will be analyzed for each lung cancer symptoms scores. SILC questionnaire comprises 3 individual symptoms & are scored at individual symptom level, thus have a dyspnea score, chest pain score, & cough score. There are a total of 9 questions in SILC questionnaire, each question has a minimum value of 0 & maximum value of 4. Each individual symptom score is calculated as average of responses for symptom items. 'Chest pain' score is mean of question 1 & 2, 'Cough' score is mean of question 3 & 4 and 'Dyspnea' score is mean of question 5 to 9 in SILC questionnaire. An increase in score is suggestive of a worsening in symptomology. A score change of ≥0.3 points for dyspnea & cough symptom scores is considered to be clinically significant; whereas a score change of≥0.5 points for chest pain score is considered to be clinically significant. (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

InterventionUnits on a scale (Mean)
Chest Pain, Week 1Chest Pain, Week 2Chest Pain, Week 3Chest Pain, Week 4Chest Pain, Week 5Chest Pain, Week 6Chest Pain, Week 7Chest Pain, Week 8Chest Pain, Week 9Chest Pain, Week 10Chest Pain, Week 11Chest Pain, Week 12Chest Pain, Week 13Chest Pain, Week 14Chest Pain, Week 15Chest Pain, Week 16Chest Pain, Week 17Chest Pain, Week 18Chest Pain, Week 19Chest Pain, Week 20Chest Pain, Week 21Chest Pain, Week 22Chest Pain, Week 23Chest Pain, Week 24Chest Pain, Week 25Chest Pain, Week 26Chest Pain, Week 27Chest Pain, Week 28Chest Pain, Week 29Chest Pain, Week 30Chest Pain, Week 31Chest Pain, Week 32Chest Pain, Week 33Chest Pain, Week 34Chest Pain, Week 35Chest Pain, Week 36Chest Pain, Week 37Chest Pain, Week 38Chest Pain, Week 39Chest Pain, Week 40Chest Pain, Week 41Chest Pain, Week 42Chest Pain, Week 43Chest Pain, Week 44Chest Pain, Week 45Chest Pain, Week 46Chest Pain, Week 47Chest Pain, Week 48Chest Pain, Week 49Chest Pain, Week 50Chest Pain, Week 51Chest Pain, Week 52Chest Pain, Week 53Chest Pain, Week 54Chest Pain, Week 55Chest Pain, Week 56Chest Pain, Week 57Chest Pain, Week 58Chest Pain, Week 59Chest Pain, Week 60Chest Pain, Week 61Chest Pain, Week 62Chest Pain, Week 63Chest Pain, Week 64Chest Pain, Week 65Chest Pain, Week 66Chest Pain, Week 67Chest Pain, Week 68Chest Pain, Week 69Chest Pain, Week 70Chest Pain, Week 71Chest Pain, Week 72Chest Pain, Week 73Chest Pain, Week 74Chest Pain, Week 75Chest Pain, Week 76Chest Pain, Week 77Chest Pain, Week 78Chest Pain, Week 79Chest Pain, Week 80Chest Pain, Week 81Chest Pain, Week 82Chest Pain, Week 83Chest Pain, Week 84Chest Pain, Week 85Chest Pain, Week 86Chest Pain, Week 87Chest Pain, Week 88Chest Pain, Week 89Chest Pain, Week 90Chest Pain, Week 91Chest Pain, Week 92Chest Pain, Week 93Chest Pain, Week 94Chest Pain, Week 95Chest Pain, Week 96Chest Pain, Week 97Chest Pain, Week 98Chest Pain, Week 99Chest Pain, Week 100Chest Pain, Week 101Chest Pain, Week 102Chest Pain, Week 103Chest Pain, Week 104Chest Pain, Week 105Chest Pain, Week 106Chest Pain, Week 107Chest Pain, Week 108Chest Pain, Week 109Chest Pain, Week 110Chest Pain, Week 111Chest Pain, Week 112Chest Pain, Week 113Chest Pain, Week 114Chest Pain, Week 115Chest Pain, Week 116Chest Pain, Week 117Chest Pain, Week 118Chest Pain, Week 119Chest Pain, Week 120Chest Pain, Week 121Chest Pain, Week 122Chest Pain, Week 123Chest Pain, Week 124Chest Pain, Week 125Chest Pain, Survival Follow-Up Month 1Chest Pain, Survival Follow-Up Month 2Chest Pain, Survival Follow-Up Month 3Chest Pain, Survival Follow-Up Month 4Chest Pain, Survival Follow-Up Month 5Chest Pain, Survival Follow-Up Month 6Cough, Week 1Cough, Week 2Cough, Week 3Cough, Week 4Cough, Week 5Cough, Week 6Cough, Week 7Cough, Week 8Cough, Week 9Cough, Week 10Cough, Week 11Cough, Week 12Cough, Week 13Cough, Week 14Cough, Week 15Cough, Week 16Cough, Week 17Cough, Week 18Cough, Week 19Cough, Week 20Cough, Week 21Cough, Week 22Cough, Week 23Cough, Week 24Cough, Week 25Cough, Week 26Cough, Week 27Cough, Week 28Cough, Week 29Cough, Week 30Cough, Week 31Cough, Week 32Cough, Week 33Cough, Week 34Cough, Week 35Cough, Week 36Cough, Week 37Cough, Week 38Cough, Week 39Cough, Week 40Cough, Week 41Cough, Week 42Cough, Week 43Cough, Week 44Cough, Week 45Cough, Week 46Cough, Week 47Cough, Week 48Cough, Week 49Cough, Week 50Cough, Week 51Cough, Week 52Cough, Week 53Cough, Week 54Cough, Week 55Cough, Week 56Cough, Week 57Cough, Week 58Cough, Week 59Cough, Week 60Cough, Week 61Cough, Week 62Cough, Week 63Cough, Week 64Cough, Week 65Cough, Week 66Cough, Week 67Cough, Week 68Cough, Week 69Cough, Week 70Cough, Week 71Cough, Week 72Cough, Week 73Cough, Week 74Cough, Week 75Cough, Week 76Cough, Week 77Cough, Week 78Cough, Week 79Cough, Week 80Cough, Week 81Cough, Week 82Cough, Week 83Cough, Week 84Cough, Week 85Cough, Week 86Cough, Week 87Cough, Week 88Cough, Week 89Cough, Week 90Cough, Week 91Cough, Week 92Cough, Week 93Cough, Week 94Cough, Week 95Cough, Week 96Cough, Week 97Cough, Week 98Cough, Week 99Cough, Week 100Cough, Week 101Cough, Week 102Cough, Week 103Cough, Week 104Cough, Week 105Cough, Week 106Cough, Week 107Cough, Week 108Cough, Week 109Cough, Week 110Cough, Week 111Cough, Week 112Cough, Week 113Cough, Week 114Cough, Week 115Cough, Week 116Cough, Week 117Cough, Week 118Cough, Week 119Cough, Week 120Cough, Week 121Cough, Week 122Cough, Week 123Cough, Week 124Cough, Week 125Cough, Survival Follow-Up Month 1Cough, Survival Follow-Up Month 2Cough, Survival Follow-Up Month 3Cough, Survival Follow-Up Month 4Cough, Survival Follow-Up Month 5Cough, Survival Follow-Up Month 6Dyspnoea, Week 1Dyspnoea, Week 2Dyspnoea, Week 3Dyspnoea, Week 4Dyspnoea, Week 5Dyspnoea, Week 6Dyspnoea, Week 7Dyspnoea, Week 8Dyspnoea, Week 9Dyspnoea, Week 10Dyspnoea, Week 11Dyspnoea, Week 12Dyspnoea, Week 13Dyspnoea, Week 14Dyspnoea, Week 15Dyspnoea, Week 16Dyspnoea, Week 17Dyspnoea, Week 18Dyspnoea, Week 19Dyspnoea, Week 20Dyspnoea, Week 21Dyspnoea, Week 22Dyspnoea, Week 23Dyspnoea, Week 24Dyspnoea, Week 25Dyspnoea, Week 26Dyspnoea, Week 27Dyspnoea, Week 28Dyspnoea, Week 29Dyspnoea, Week 30Dyspnoea, Week 31Dyspnoea, Week 32Dyspnoea, Week 33Dyspnoea, Week 34Dyspnoea, Week 35Dyspnoea, Week 36Dyspnoea, Week 37Dyspnoea, Week 38Dyspnoea, Week 39Dyspnoea, Week 40Dyspnoea, Week 41Dyspnoea, Week 42Dyspnoea, Week 43Dyspnoea, Week 44Dyspnoea, Week 45Dyspnoea, Week 46Dyspnoea, Week 47Dyspnoea, Week 48Dyspnoea, Week 49Dyspnoea, Week 50Dyspnoea, Week 51Dyspnoea, Week 52Dyspnoea, Week 53Dyspnoea, Week 54Dyspnoea, Week 55Dyspnoea, Week 56Dyspnoea, Week 57Dyspnoea, Week 58Dyspnoea, Week 59Dyspnoea, Week 60Dyspnoea, Week 61Dyspnoea, Week 62Dyspnoea, Week 63Dyspnoea, Week 64Dyspnoea, Week 65Dyspnoea, Week 66Dyspnoea, Week 67Dyspnoea, Week 68Dyspnoea, Week 69Dyspnoea, Week 70Dyspnoea, Week 71Dyspnoea, Week 72Dyspnoea, Week 73Dyspnoea, Week 74Dyspnoea, Week 75Dyspnoea, Week 76Dyspnoea, Week 77Dyspnoea, Week 78Dyspnoea, Week 79Dyspnoea, Week 80Dyspnoea, Week 81Dyspnoea, Week 82Dyspnoea, Week 83Dyspnoea, Week 84Dyspnoea, Week 85Dyspnoea, Week 86Dyspnoea, Week 87Dyspnoea, Week 88Dyspnoea, Week 89Dyspnoea, Week 90Dyspnoea, Week 91Dyspnoea, Week 92Dyspnoea, Week 93Dyspnoea, Week 94Dyspnoea, Week 95Dyspnoea, Week 96Dyspnoea, Week 97Dyspnoea, Week 98Dyspnoea, Week 99Dyspnoea, Week 100Dyspnoea, Week 101Dyspnoea, Week 102Dyspnoea, Week 103Dyspnoea, Week 104Dyspnoea, Week 105Dyspnoea, Week 106Dyspnoea, Week 107Dyspnoea, Week 108Dyspnoea, Week 109Dyspnoea, Week 110Dyspnoea, Week 111Dyspnoea, Week 112Dyspnoea, Week 113Dyspnoea, Week 114Dyspnoea, Week 115Dyspnoea, Week 116Dyspnoea, Week 117Dyspnoea, Week 118Dyspnoea, Week 119Dyspnoea, Week 120Dyspnoea, Week 121Dyspnoea, Week 122Dyspnoea, Week 123Dyspnoea, Week 124Dyspnoea, Week 125Dyspnoea, Survival Follow-Up Month 1Dyspnoea, Survival Follow-Up Month 2Dyspnoea, Survival Follow-Up Month 3Dyspnoea, Survival Follow-Up Month 4Dyspnoea, Survival Follow-Up Month 5Dyspnoea, Survival Follow-Up Month 6
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)0.19-0.02-0.05-0.11-0.12-0.24-0.23-0.21-0.18-0.10-0.11-0.15-0.26-0.28-0.26-0.33-0.33-0.28-0.28-0.26-0.25-0.28-0.24-0.21-0.20-0.17-0.22-0.20-0.27-0.15-0.16-0.19-0.18-0.18-0.10-0.21-0.18-0.32-0.28-0.19-0.25-0.16-0.24-0.24-0.14-0.15-0.22-0.14-0.22-0.18-0.13-0.15-0.20-0.22-0.34-0.19-0.19-0.32-0.25-0.27-0.28-0.16-0.12-0.15-0.31-0.25-0.18-0.15-0.13-0.14-0.10-0.24-0.25-0.08-0.210.03-0.06-0.04-0.11-0.18-0.59-0.39-0.34-0.20-0.44-0.38-0.53-0.46-0.55-0.18-0.32-0.40-0.18-0.30-0.05-0.17-0.19-0.25-0.21-0.50-0.36-0.75-0.33-0.60-1.00-1.00-1.00-0.75-0.60-0.42-0.50-0.130.130.000.250.500.25-0.25-0.250.000.000.00-0.750.500.500.01-0.070.15-0.28-0.11-0.370.080.020.02-0.06-0.09-0.15-0.11-0.13-0.15-0.20-0.15-0.17-0.24-0.23-0.27-0.37-0.32-0.33-0.33-0.37-0.37-0.37-0.30-0.38-0.49-0.43-0.41-0.52-0.43-0.43-0.32-0.30-0.19-0.35-0.46-0.35-0.46-0.38-0.27-0.38-0.44-0.44-0.39-0.38-0.30-0.25-0.44-0.29-0.38-0.39-0.30-0.32-0.37-0.41-0.40-0.26-0.35-0.33-0.31-0.42-0.35-0.23-0.22-0.19-0.23-0.29-0.48-0.34-0.34-0.26-0.23-0.48-0.43-0.42-0.32-0.31-0.40-0.52-0.08-0.48-0.44-0.39-0.25-0.30-0.32-0.54-0.33-0.46-0.36-0.29-0.27-0.35-0.55-0.400.05-0.28-0.31-0.10-0.43-0.58-0.50-0.50-0.75-0.80-0.63-0.63-0.63-0.50-0.50-0.50-0.30-0.63-0.25-0.330.25-0.25-0.50-0.250.00-0.25-0.50-0.50-0.750.500.50-0.21-0.07-0.14-0.39-0.25-0.230.130.100.220.230.260.270.290.320.380.410.500.470.320.340.290.220.280.230.260.240.260.210.180.220.210.170.200.160.160.240.120.200.180.210.220.210.160.180.310.310.240.260.170.220.260.290.200.320.240.320.240.270.240.280.260.260.370.190.320.350.330.450.440.360.270.320.280.350.440.330.500.170.410.300.260.230.200.310.320.30-0.120.020.130.190.05-0.07-0.110.060.090.370.330.200.350.500.480.510.330.380.110.270.200.350.470.48-0.20-0.20-0.30-0.150.160.33-0.040.200.200.400.800.600.300.300.500.300.600.60-0.20-0.200.200.410.360.270.020.13-0.09

[back to top]

Overall Survival (OS) in the ITT-WT Population

OS is defined as the time between the date of randomization and date of death from any cause in the ITT-WT population. (NCT02367781)
Timeframe: Up to approximately 35 months after first patient enrolled

InterventionMonths (Median)
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)18.6
Arm B (Nab-Paclitaxel+Carboplatin)13.9

[back to top]

OS as Determined by the Investigator Using Recist v1.1 in the ITT Population

OS is defined as the time between the date of randomization and date of death from any cause in the ITT population. (NCT02367781)
Timeframe: Up to approximately 41 months after first subject enrolled

InterventionMonths (Median)
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)17.0
Arm B (Nab-Paclitaxel+Carboplatin)13.5

[back to top]

Plasma Concentrations of Nab-Paclitaxel Reported as Total Paclitaxel

(NCT02367781)
Timeframe: Predose (same day of treatment administration), 5-10 minutes before end of nab-paclitaxel infusion, 1 hour after nab-paclitaxel infusion (infusion duration=30 minutes) on Day 1 of Cycle 1 and 3 (1 Cycle=21 days) (up to approximately 35 months)

,
Interventionng/mL (Mean)
Cycle 1 Day 1 Pre-doseCycle 1 Day 1 Before End of InfusionCycle 1 Day 1 Post InfusionCycle 3 Day 1 Pre-doseCycle 3 Day 1 Before End of InfusionCycle 3 Day 1 Post Infusion
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)NA3520307NA4480357
Arm B (Nab-Paclitaxel+Carboplatin Crossover)NA2530417NA2030447

[back to top]

Plasma Concentrations of Carboplatin

(NCT02367781)
Timeframe: Predose (same day of treatment administration), 5-10 minutes before end of carboplatin infusion, 1 hour after carboplatin infusion (infusion duration=15 to 30 minutes) on Day 1 of Cycle 1 and 3 (1 Cycle=21 days) (up to approximately 35 months)

,
Interventionng/mL (Mean)
Cycle 1 Day 1 Pre-doseCycle 1 Day 1 Before End of InfusionCycle 1 Day 1 Post InfusionCycle 3 Day 1 Pre-doseCycle 3 Day 1 Before End of InfusionCycle 3 Day 1 Post Infusion
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)NA20,50011,90016915,30011,400
Arm B (Nab-Paclitaxel+Carboplatin Crossover)NA17,00012,40016017,80013,400

[back to top]

PFS as Determined by the Investigator Using Recist v1.1 in the ITT Population, PD-L1 Expression Population, and PD-L1 Expression WT Population

PFS is defined as the time between the date of randomization and the date of first documented disease progression as determined by the investigator according to RECIST v1.1 or death from any cause, whichever occurs first. The ITT population was defined as all randomized participants, regardless of receipt of the assigned treatment. The PD-L1 expression population is defined as one of the following: PD-L1 IHC TC1/2/3 or IC1/2/3 population, defined as ITT participants with PD-L1 IHC TC1/2/3 or IC1/2/3 expression in baseline tumor tissue; PD-L1 IHC TC2/3 or IC2/3 population, defined as ITT participants with PD-L1 IHC TC2/3 or IC2/3 expression in baseline tumor tissue; PD-L1 IHC TC3 or IC3 population, defined as ITT participants with PD-L1 IHC TC3 or IC3 expression in baseline tumor tissue. The PD-L1 expression WT population is defined as the PD-L1 expression population excluding participants with an activating EGFR mutation or ALK translocation. (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

,
InterventionMonths (Median)
ITT PopulationTC1/2/3 or IC1/2/3 ITT PopulationTC1/2/3 or IC1/2/3-WT ITT Population
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)7.07.57.5
Arm B (Nab-Paclitaxel+Carboplatin)5.65.75.9

[back to top]

Percentage of Participants With Anti-Therapeutic Antibodies (ATAs) to Atezolizumab

Baseline prevalence and post-baseline incidence of anti-drug antibodies (ADA) to Atezolizumab in the Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed) and Arm B Carboplatin+nab-paclitaxel Crossover Participants (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

,
InterventionPerecentage of participants (Number)
BaselinePost-baseline
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)3.122.4
Arm B (Nab-Paclitaxel+Carboplatin)4.823.5

[back to top]

Percentage of Participants With an Objective Response (OR) (Complete Response [CR] or Partial Response [PR]) as Determined by the Investigator Using RECIST v1.1 in the ITT Population, PD-L1 Expression Population, and PD-L1 Expression WT Population

ORR (confirmation not required) is defined as proportion of participants with an objective response, either CR or PR, with the use of RECIST v1.1, as determined by investigator in ITT population, PD-L1 Expression population, and PD-L1 Expression WT population. ITT population was defined as all randomized participants, regardless of receipt of the assigned treatment. PD-L1 expression population is defined as one of the following: PD-L1 IHC TC1/2/3 or IC1/2/3 population, defined as ITT participants with PD-L1 IHC TC1/2/3 or IC1/2/3 expression in baseline tumor tissue; PD-L1 IHC TC2/3 or IC2/3 population, defined as ITT participants with PD-L1 IHC TC2/3 or IC2/3 expression in baseline tumor tissue; PD-L1 IHC TC3 or IC3 population, defined as ITT participants with PD-L1 IHC TC3 or IC3 expression in baseline tumor tissue. PD-L1 expression WT population is defined as PD-L1 expression population excluding participants with an activating EGFR mutation or ALK translocation. (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

,
InterventionPercentage of participants (Number)
ITT PopulationTC1/2/3 or IC1/2/3 ITT WT PopulationTC1/2/3 or IC1/2/3 ITT Population
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)59.165.664.6
Arm B (Nab-Paclitaxel+Carboplatin)42.246.245.0

[back to top]

OS as Determined by the Investigator Using RECIST v1.1 in the PD-L1 Expression Population and PD-L1 Expression WT Population

OS is defined as the time between the date of randomization and date of death from any cause in the PD-L1 Expression Population and PD-L1 Expression WT Population. The PD-L1 expression population is defined as one of the following: PD-L1 IHC TC1/2/3 or IC1/2/3 population, defined as ITT participants with PD-L1 IHC TC1/2/3 or IC1/2/3 expression in baseline tumor tissue; PD-L1 IHC TC2/3 or IC2/3 population, defined as ITT participants with PD-L1 IHC TC2/3 or IC2/3 expression in baseline tumor tissue; PD-L1 IHC TC3 or IC3 population, defined as ITT participants with PD-L1 IHC TC3 or IC3 expression in baseline tumor tissue. The PD-L1 expression WT population is defined as the PD-L1 expression population excluding participants with an activating EGFR mutation or ALK translocation. (NCT02367781)
Timeframe: Up to approximately 35 months after first patient enrolled

,
InterventionMonths (Median)
TC1/2/3 or IC1/2/3 ITT PopulationTC1/2/3 or IC1/2/3 WT ITT Population
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)21.221.2
Arm B (Nab-Paclitaxel+Carboplatin)16.916.9

[back to top]

Minimum Observed Serum Concentration (Cmin) of Atezolizumab Prior to Infusion in Atezolizumab+Carboplain+Nab-Paclitaxel

Predose samples will be collected on the same day of treatment administration. (NCT02367781)
Timeframe: Cycle 1 Day 21, Cycle 2 Day 21, Cycle 3 Day 21, and Cycle 7 Day 21 (Cycle length = 21 days)

Interventionmcg/mL (Mean)
Cycle 1 Day 21Cycle 2 Day 21Cycle 3 Day 21Cycle 7 Day 21
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)70.9111134218

[back to top]

Maximum Observed Serum Concentration (Cmax) of Atezolizumab for Patients in Atezolizumab+Carboplatin+Nab-Paclitaxel Arm

Predose samples will be collected on the same day of treatment administration. The infusion duration of atezolizumab will be of 30-60 minutes. (NCT02367781)
Timeframe: Cycle 1 Day 1 and Cycle 3 Day 1 (Cycle length = 21 days)

Interventionmcg/mL (Mean)
Cycle 1 Day 1Cycle 3 Day 1
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)392454

[back to top]

Time to Deterioration (TTD) in Patient-Reported Lung Cancer Symptoms in the ITT-WT Population

Defined as time from randomization to confirmed deterioration (10-point change) on the combined European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire-Core (EORTC QLQ-C30) and supplemental lung cancer module (EORTC QLQ-LC13) symptom subscales. (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

InterventionMonths (Median)
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)2.2
Arm B (Nab-Paclitaxel+Carboplatin)1.9

[back to top]

Event Free Rate (%) at Year 1 and 2 in PD-L1 Expression Population and PD-L1 Expression WT Population

The OS rate at the 1- and 2-year landmark time points after randomization in the PD-L1 Expression Population and PD-L1 Expression WT Population. The PD-L1 expression population is defined as one of the following: PD-L1 IHC TC1/2/3 or IC1/2/3 population, defined as ITT participants with PD-L1 IHC TC1/2/3 or IC1/2/3 expression in baseline tumor tissue; PD-L1 IHC TC2/3 or IC2/3 population, defined as ITT participants with PD-L1 IHC TC2/3 or IC2/3 expression in baseline tumor tissue; PD-L1 IHC TC3 or IC3 population, defined as ITT participants with PD-L1 IHC TC3 or IC3 expression in baseline tumor tissue. The PD-L1 expression WT population is defined as the PD-L1 expression population excluding participants with an activating EGFR mutation or ALK translocation. (NCT02367781)
Timeframe: Up to 35 months after first patient enrolled, years 1 and 2 reported

,
InterventionPercentage of participants (Number)
Event Free Rate (%) at Year 1 TC1/2/3 or IC1/2/3 ITTEvent Free Rate (%) at Year 2 TC1/2/3 or IC1/2/3 ITTEvent Free Rate (%) at Year 1 TC1/2/3 or IC1/2/3 ITT WTEvent Free Rate (%) at Year 2 TC1/2/3 or IC1/2/3 ITT WT
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)68.5644.6368.8444.02
Arm B (Nab-Paclitaxel+Carboplatin)61.8635.9862.5135.33

[back to top]

Event Free Rate (%) at Year 1 and 2 in ITT-WT Population and ITT Population

The OS rate at the 1- and 2-year landmark time points after randomization. (NCT02367781)
Timeframe: Up to 41 months after first patient enrolled, years 1 and 2 reported

,
InterventionPercentage of participants (Number)
Event Free Rate (%) at Year 1 ITT WTEvent Free Rate (%) at Year 2 ITT WTEvent Free Rate (%) at Year 1 ITTEvent Free Rate (%) at Year 2 ITT
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)62.0240.4361.6539.73
Arm B (Nab-Paclitaxel+Carboplatin)54.5632.3654.4732.21

[back to top]

Duration of Response (DOR) as Determined by the Investigator Using RECIST v1.1 in ITT-WT Population, ITT Population, and PD-L1 Expression Population and PD-L1 Expression WT Population

DOR,defined for participants with objective response (OR) as time from 1st documented OR to documented disease progression as determined by investigator using RECIST v1.1,or death from any cause,whichever occurs 1st.ITT defined as all randomized participants,regardless of receipt of assigned treatment.ITT-WT defined as ITT population excluding participants with activating EGFR mutation or ALK translocation.PD-L1 expression population is defined as one of following:PD-L1 IHC TC1/2/3 or IC1/2/3 population,defined as ITT participants with PD-L1 IHC TC1/2/3 or IC1/2/3 expression in baseline tumor tissue;PD-L1 IHC TC2/3 or IC2/3 population, defined as ITT participants with PD-L1 IHC TC2/3 or IC2/3 expression in baseline tumor tissue;PD-L1 IHC TC3 or IC3 population,defined as ITT participants with PD-L1 IHC TC3 or IC3 expression in baseline tumor tissue.PD-L1 expression WT is defined as PD-L1 expression population excluding participants with activating EGFR mutation or ALK translocation. (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

,
InterventionMonths (Median)
ITT PopulationITT-WT PopulationTC1/2/3 or IC1/2/3 ITT PopulationTC1/2/3 or IC1/2/3 ITT WT Population
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)6.26.77.27.2
Arm B (Nab-Paclitaxel+Carboplatin)5.45.45.05.0

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms Score Using the Symptoms in Lung Cancer (SILC) Scale

Change from baseline per SILC scale will be analyzed for each lung cancer symptoms scores. SILC questionnaire comprises 3 individual symptoms & are scored at individual symptom level, thus have a dyspnea score, chest pain score, & cough score. There are a total of 9 questions in SILC questionnaire, each question has a minimum value of 0 & maximum value of 4. Each individual symptom score is calculated as average of responses for symptom items. 'Chest pain' score is mean of question 1 & 2, 'Cough' score is mean of question 3 & 4 and 'Dyspnea' score is mean of question 5 to 9 in SILC questionnaire. An increase in score is suggestive of a worsening in symptomology. A score change of ≥0.3 points for dyspnea & cough symptom scores is considered to be clinically significant; whereas a score change of≥0.5 points for chest pain score is considered to be clinically significant. (NCT02367781)
Timeframe: Up to approximately 35 months after first subject enrolled

InterventionUnits on a scale (Mean)
Chest Pain, Week 1Chest Pain, Week 2Chest Pain, Week 3Chest Pain, Week 4Chest Pain, Week 5Chest Pain, Week 6Chest Pain, Week 7Chest Pain, Week 8Chest Pain, Week 9Chest Pain, Week 10Chest Pain, Week 11Chest Pain, Week 12Chest Pain, Week 13Chest Pain, Week 14Chest Pain, Week 15Chest Pain, Week 16Chest Pain, Week 17Chest Pain, Week 18Chest Pain, Week 19Chest Pain, Week 20Chest Pain, Week 21Chest Pain, Week 22Chest Pain, Week 23Chest Pain, Week 24Chest Pain, Week 25Chest Pain, Week 26Chest Pain, Week 27Chest Pain, Week 28Chest Pain, Week 29Chest Pain, Week 30Chest Pain, Week 31Chest Pain, Week 32Chest Pain, Week 33Chest Pain, Week 34Chest Pain, Week 35Chest Pain, Week 36Chest Pain, Week 37Chest Pain, Week 38Chest Pain, Week 39Chest Pain, Week 40Chest Pain, Week 41Chest Pain, Week 42Chest Pain, Week 43Chest Pain, Week 44Chest Pain, Week 45Chest Pain, Week 46Chest Pain, Week 47Chest Pain, Week 48Chest Pain, Week 49Chest Pain, Week 50Chest Pain, Week 51Chest Pain, Week 52Chest Pain, Week 53Chest Pain, Week 54Chest Pain, Week 55Chest Pain, Week 56Chest Pain, Week 57Chest Pain, Week 58Chest Pain, Week 59Chest Pain, Week 60Chest Pain, Week 61Chest Pain, Week 62Chest Pain, Week 63Chest Pain, Week 64Chest Pain, Week 65Chest Pain, Week 66Chest Pain, Week 67Chest Pain, Week 68Chest Pain, Week 69Chest Pain, Week 70Chest Pain, Week 71Chest Pain, Week 72Chest Pain, Week 73Chest Pain, Week 74Chest Pain, Week 75Chest Pain, Week 76Chest Pain, Week 77Chest Pain, Week 78Chest Pain, Week 79Chest Pain, Week 80Chest Pain, Week 81Chest Pain, Week 82Chest Pain, Week 83Chest Pain, Week 84Chest Pain, Week 85Chest Pain, Week 86Chest Pain, Week 87Chest Pain, Week 88Chest Pain, Week 89Chest Pain, Week 90Chest Pain, Week 91Chest Pain, Week 92Chest Pain, Week 93Chest Pain, Survival Follow-Up Month 1Chest Pain, Survival Follow-Up Month 2Chest Pain, Survival Follow-Up Month 3Chest Pain, Survival Follow-Up Month 4Chest Pain, Survival Follow-Up Month 5Chest Pain, Survival Follow-Up Month 6Cough, Week 1Cough, Week 2Cough, Week 3Cough, Week 4Cough, Week 5Cough, Week 6Cough, Week 7Cough, Week 8Cough, Week 9Cough, Week 10Cough, Week 11Cough, Week 12Cough, Week 13Cough, Week 14Cough, Week 15Cough, Week 16Cough, Week 17Cough, Week 18Cough, Week 19Cough, Week 20Cough, Week 21Cough, Week 22Cough, Week 23Cough, Week 24Cough, Week 25Cough, Week 26Cough, Week 27Cough, Week 28Cough, Week 29Cough, Week 30Cough, Week 31Cough, Week 32Cough, Week 33Cough, Week 34Cough, Week 35Cough, Week 36Cough, Week 37Cough, Week 38Cough, Week 39Cough, Week 40Cough, Week 41Cough, Week 42Cough, Week 43Cough, Week 44Cough, Week 45Cough, Week 46Cough, Week 47Cough, Week 48Cough, Week 49Cough, Week 50Cough, Week 51Cough, Week 52Cough, Week 53Cough, Week 54Cough, Week 55Cough, Week 56Cough, Week 57Cough, Week 58Cough, Week 59Cough, Week 60Cough, Week 61Cough, Week 62Cough, Week 63Cough, Week 64Cough, Week 65Cough, Week 66Cough, Week 67Cough, Week 68Cough, Week 69Cough, Week 70Cough, Week 71Cough, Week 72Cough, Week 73Cough, Week 74Cough, Week 75Cough, Week 76Cough, Week 77Cough, Week 78Cough, Week 79Cough, Week 80Cough, Week 81Cough, Week 82Cough, Week 83Cough, Week 84Cough, Week 85Cough, Week 86Cough, Week 87Cough, Week 88Cough, Week 89Cough, Week 90Cough, Week 91Cough, Week 92Cough, Week 93Cough, Survival Follow-Up Month 1Cough, Survival Follow-Up Month 2Cough, Survival Follow-Up Month 3Cough, Survival Follow-Up Month 4Cough, Survival Follow-Up Month 5Cough, Survival Follow-Up Month 6Dyspnoea, Week 1Dyspnoea, Week 2Dyspnoea, Week 3Dyspnoea, Week 4Dyspnoea, Week 5Dyspnoea, Week 6Dyspnoea, Week 7Dyspnoea, Week 8Dyspnoea, Week 9Dyspnoea, Week 10Dyspnoea, Week 11Dyspnoea, Week 12Dyspnoea, Week 13Dyspnoea, Week 14Dyspnoea, Week 15Dyspnoea, Week 16Dyspnoea, Week 17Dyspnoea, Week 18Dyspnoea, Week 19Dyspnoea, Week 20Dyspnoea, Week 21Dyspnoea, Week 22Dyspnoea, Week 23Dyspnoea, Week 24Dyspnoea, Week 25Dyspnoea, Week 26Dyspnoea, Week 27Dyspnoea, Week 28Dyspnoea, Week 29Dyspnoea, Week 30Dyspnoea, Week 31Dyspnoea, Week 32Dyspnoea, Week 33Dyspnoea, Week 34Dyspnoea, Week 35Dyspnoea, Week 36Dyspnoea, Week 37Dyspnoea, Week 38Dyspnoea, Week 39Dyspnoea, Week 40Dyspnoea, Week 41Dyspnoea, Week 42Dyspnoea, Week 43Dyspnoea, Week 44Dyspnoea, Week 45Dyspnoea, Week 46Dyspnoea, Week 47Dyspnoea, Week 48Dyspnoea, Week 49Dyspnoea, Week 50Dyspnoea, Week 51Dyspnoea, Week 52Dyspnoea, Week 53Dyspnoea, Week 54Dyspnoea, Week 55Dyspnoea, Week 56Dyspnoea, Week 57Dyspnoea, Week 58Dyspnoea, Week 59Dyspnoea, Week 60Dyspnoea, Week 61Dyspnoea, Week 62Dyspnoea, Week 63Dyspnoea, Week 64Dyspnoea, Week 65Dyspnoea, Week 66Dyspnoea, Week 67Dyspnoea, Week 68Dyspnoea, Week 69Dyspnoea, Week 70Dyspnoea, Week 71Dyspnoea, Week 72Dyspnoea, Week 73Dyspnoea, Week 74Dyspnoea, Week 75Dyspnoea, Week 76Dyspnoea, Week 77Dyspnoea, Week 78Dyspnoea, Week 79Dyspnoea, Week 80Dyspnoea, Week 81Dyspnoea, Week 82Dyspnoea, Week 83Dyspnoea, Week 84Dyspnoea, Week 85Dyspnoea, Week 86Dyspnoea, Week 87Dyspnoea, Week 88Dyspnoea, Week 89Dyspnoea, Week 90Dyspnoea, Week 91Dyspnoea, Week 92Dyspnoea, Week 93Dyspnoea, Survival Follow-Up Month 1Dyspnoea, Survival Follow-Up Month 2Dyspnoea, Survival Follow-Up Month 3Dyspnoea, Survival Follow-Up Month 4Dyspnoea, Survival Follow-Up Month 5Dyspnoea, Survival Follow-Up Month 6
Arm B (Nab-Paclitaxel+Carboplatin)0.140.030.010.010.000.030.03-0.14-0.01-0.070.01-0.10-0.03-0.17-0.19-0.16-0.14-0.07-0.16-0.22-0.32-0.11-0.19-0.43-0.24-0.13-0.15-0.07-0.04-0.28-0.12-0.30-0.18-0.170.05-0.35-0.10-0.05-0.22-0.39-0.19-0.41-0.220.00-0.07-0.07-0.15-0.18-0.72-0.19-0.50-0.36-0.33-0.21-0.30-0.33-0.40-0.50-0.63-0.50-0.20-0.40-0.100.380.170.250.13-0.25-0.170.000.00-1.00-1.50-1.50-1.50-1.50-1.50-1.50-0.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.50-1.500.22-0.16-0.08-0.07-0.020.020.040.04-0.09-0.10-0.09-0.08-0.06-0.21-0.13-0.07-0.15-0.11-0.04-0.18-0.05-0.25-0.21-0.33-0.40-0.31-0.33-0.24-0.41-0.54-0.47-0.34-0.48-0.43-0.46-0.46-0.38-0.12-0.52-0.330.00-0.20-0.45-0.03-0.17-0.50-0.22-0.41-0.13-0.16-0.14-0.03-0.120.14-0.56-0.19-0.40-0.21-0.17-0.070.100.170.000.130.130.380.800.500.500.000.830.630.380.130.170.170.33-1.50-0.50-0.50-0.50-0.50-0.500.001.001.00-0.501.000.501.001.500.000.000.000.000.000.001.000.00-0.01-0.31-0.13-0.45-0.27-0.290.230.310.320.450.420.510.600.530.620.750.600.740.760.610.710.670.680.620.540.540.390.410.410.310.200.300.300.220.330.270.260.380.190.300.460.340.100.510.260.040.180.080.410.290.310.470.450.290.000.430.060.290.300.400.440.530.240.350.350.500.600.720.56-0.500.070.500.400.400.730.730.600.60-1.80-1.00-1.00-1.00-1.50-1.00-1.00-1.00-0.80-0.80-1.00-1.00-0.80-1.00-1.00-1.00-1.00-1.00-1.00-0.80-0.800.600.460.610.450.480.54

[back to top]

Progression-Free Survival (PFS) as Determined by the Investigator Using Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) in the ITT-WT Population

PFS is defined as the time between the date of randomization and the date of first documented disease progression as determined by the investigator according to RECIST v1.1 or death from any cause, whichever occurs first in the ITT-WT population. (NCT02367781)
Timeframe: Up to approximately 35 months after first patient enrolled

InterventionMonths (Median)
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)7.0
Arm B (Nab-Paclitaxel+Carboplatin)5.5

[back to top]

Percentage of Participants With an Objective Response (OR) (Complete Response [CR] or Partial Response [PR]) as Determined by the Investigator Using RECIST v1.1 in the ITT-WT Population

ORR (confirmation not required) is defined as the proportion of participants with an objective response, either CR or PR, with the use of RECIST v1.1, as determined by the investigator in the ITT-WT population. (NCT02367781)
Timeframe: Up to approximately 41 months after first subject enrolled

InterventionPercentage of participants (Number)
Arm A (Atezolizumab+Nab-Paclitaxel+Carboplatin)60.2
Arm B (Nab-Paclitaxel+Carboplatin)41.0

[back to top]

Duration of Response (DOR) in the TC3 or IC3-WT Populations

DOR is defined as the time from the first occurrence of a documented objective response to the time of disease progression, as determined by the investigator with use of RECIST v1.1, or death from any cause, whichever occurs first. (NCT02409342)
Timeframe: From first occurrence of a complete response or partial response, whichever occurs first, until first date that progressive disease or death is documented, whichever occurs first until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionMonths (Number)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)6.7
AtezolizumabNA

[back to top]

TTD as Assessed Using EORTC QLQ Supplementary Lung Cancer Module (EORTC QLQ-LC13) in the TC3 or IC3-WT Populations

TTD in patient-reported lung cancer symptoms, defined as time from randomization to deterioration (10-point change) in any of the following symptom subscales (cough, dyspnea [multi-item scale], and chest pain), whichever occurs first, as measured by the EORTC QLQ-LC13. EORTC QLQ-LC13 module incorporates one multi-item scale to assess dyspnea and a series of single items assessing pain, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. (NCT02409342)
Timeframe: Baseline until data cut-off on 10 September 2018 (up to approximately 38 months)

,
InterventionMonths (Median)
CoughDyspneaChest pain
AtezolizumabNA11.1NA
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)NA11.8NA

[back to top]

Change From Baseline in Patient-reported Lung Cancer Symptoms Score as Assessed by the SILC Scale Symptom Score in the TC3 or IC3-WT Populations

Change from baseline in each of the patient-reported lung cancer symptoms with use of the SILC scale. The SILC scale is a nine-item content valid self-report measure of lung cancer symptoms. It measures severity of cough, dyspnea, and chest pain with a total symptom severity score. Each SILC symptom scale (dyspnea, cough, chest pain) score was calculated as the average of the component items (range 0 to 4). An increase in score suggested worsening in symptomatology. A symptom score change of 0.3 points for the dyspnea and cough scores was considered to be clinically significant; whereas a symptom score change of 0.5 points for the chest pain score was considered to be clinically significant. (NCT02409342)
Timeframe: Baseline until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionScore on a scale (Mean)
Chest Pain, Week 1Chest Pain, Week 2Chest Pain, Week 3Chest Pain, Week 4Chest Pain, Week 5Chest Pain, Week 6Chest Pain, Week 7Chest Pain, Week 8Chest Pain, Week 9Chest Pain, Week 10Chest Pain, Week 11Chest Pain, Week 12Chest Pain, Week 13Chest Pain, Week 14Chest Pain, Week 15Chest Pain, Week 16Chest Pain, Week 17Chest Pain, Week 18Chest Pain, Week 19Chest Pain, Week 20Chest Pain, Week 21Chest Pain, Week 22Chest Pain, Week 23Chest Pain, Week 24Chest Pain, Week 25Chest Pain, Week 26Chest Pain, Week 27Chest Pain, Week 28Chest Pain, Week 29Chest Pain, Week 30Chest Pain, Week 31Chest Pain, Week 32Chest Pain, Week 33Chest Pain, Week 34Chest Pain, Week 35Chest Pain, Week 36Chest Pain, Week 37Chest Pain, Week 38Chest Pain, Week 39Chest Pain, Week 40Chest Pain, Week 41Chest Pain, Week 42Chest Pain, Week 43Chest Pain, Week 44Chest Pain, Week 45Chest Pain, Week 46Chest Pain, Week 47Chest Pain, Week 48Chest Pain, Week 49Chest Pain, Week 50Chest Pain, Week 51Chest Pain, Week 52Chest Pain, Week 53Chest Pain, Week 54Chest Pain, Week 55Chest Pain, Week 56Chest Pain, Week 57Chest Pain, Week 58Chest Pain, Week 59Chest Pain, Week 60Chest Pain, Week 61Chest Pain, Week 62Chest Pain, Week 63Chest Pain, Week 64Chest Pain, Week 65Chest Pain, Week 66Chest Pain, Week 67Chest Pain, Week 68Chest Pain, Week 69Chest Pain, Week 70Chest Pain, Week 71Chest Pain, Week 72Chest Pain, Week 73Chest Pain, Week 74Chest Pain, Week 75Chest Pain, Week 76Chest Pain, Week 77Chest Pain, Week 78Chest Pain, Week 79Chest Pain, Week 80Chest Pain, Week 81Chest Pain, Week 82Chest Pain, Week 83Chest Pain, Week 84Chest Pain, Week 85Chest Pain, Week 86Chest Pain, Week 87Cough, Week 1Cough, Week 2Cough, Week 3Cough, Week 4Cough, Week 5Cough, Week 6Cough, Week 7Cough, Week 8Cough, Week 9Cough, Week 10Cough, Week 11Cough, Week 12Cough, Week 13Cough, Week 14Cough, Week 15Cough, Week 16Cough, Week 17Cough, Week 18Cough, Week 19Cough, Week 20Cough, Week 21Cough, Week 22Cough, Week 23Cough, Week 24Cough, Week 25Cough, Week 26Cough, Week 27Cough, Week 28Cough, Week 29Cough, Week 30Cough, Week 31Cough, Week 32Cough, Week 33Cough, Week 34Cough, Week 35Cough, Week 36Cough, Week 37Cough, Week 38Cough, Week 39Cough, Week 40Cough, Week 41Cough, Week 42Cough, Week 43Cough, Week 44Cough, Week 45Cough, Week 46Cough, Week 47Cough, Week 48Cough, Week 49Cough, Week 50Cough, Week 51Cough, Week 52Cough, Week 53Cough, Week 54Cough, Week 55Cough, Week 56Cough, Week 57Cough, Week 58Cough, Week 59Cough, Week 60Cough, Week 61Cough, Week 62Cough, Week 63Cough, Week 64Cough, Week 65Cough, Week 66Cough, Week 67Cough, Week 68Cough, Week 69Cough, Week 70Cough, Week 71Cough, Week 72Cough, Week 73Cough, Week 74Cough, Week 75Cough, Week 76Cough, Week 77Cough, Week 78Cough, Week 79Cough, Week 80Cough, Week 81Cough, Week 82Cough, Week 83Cough, Week 84Cough, Week 85Cough, Week 86Cough, Week 87Dyspnoea, Week 1Dyspnoea, Week 2Dyspnoea, Week 3Dyspnoea, Week 4Dyspnoea, Week 5Dyspnoea, Week 6Dyspnoea, Week 7Dyspnoea, Week 8Dyspnoea, Week 9Dyspnoea, Week 10Dyspnoea, Week 11Dyspnoea, Week 12Dyspnoea, Week 13Dyspnoea, Week 14Dyspnoea, Week 15Dyspnoea, Week 16Dyspnoea, Week 17Dyspnoea, Week 18Dyspnoea, Week 19Dyspnoea, Week 20Dyspnoea, Week 21Dyspnoea, Week 22Dyspnoea, Week 23Dyspnoea, Week 24Dyspnoea, Week 25Dyspnoea, Week 26Dyspnoea, Week 27Dyspnoea, Week 28Dyspnoea, Week 29Dyspnoea, Week 30Dyspnoea, Week 31Dyspnoea, Week 32Dyspnoea, Week 33Dyspnoea, Week 34Dyspnoea, Week 35Dyspnoea, Week 36Dyspnoea, Week 37Dyspnoea, Week 38Dyspnoea, Week 39Dyspnoea, Week 40Dyspnoea, Week 41Dyspnoea, Week 42Dyspnoea, Week 43Dyspnoea, Week 44Dyspnoea, Week 45Dyspnoea, Week 46Dyspnoea, Week 47Dyspnoea, Week 48Dyspnoea, Week 49Dyspnoea, Week 50Dyspnoea, Week 51Dyspnoea, Week 52Dyspnoea, Week 53Dyspnoea, Week 54Dyspnoea, Week 55Dyspnoea, Week 56Dyspnoea, Week 57Dyspnoea, Week 58Dyspnoea, Week 59Dyspnoea, Week 60Dyspnoea, Week 61Dyspnoea, Week 62Dyspnoea, Week 63Dyspnoea, Week 64Dyspnoea, Week 65Dyspnoea, Week 66Dyspnoea, Week 67Dyspnoea, Week 68Dyspnoea, Week 69Dyspnoea, Week 70Dyspnoea, Week 71Dyspnoea, Week 72Dyspnoea, Week 73Dyspnoea, Week 74Dyspnoea, Week 75Dyspnoea, Week 76Dyspnoea, Week 77Dyspnoea, Week 78Dyspnoea, Week 79Dyspnoea, Week 80Dyspnoea, Week 81Dyspnoea, Week 82Dyspnoea, Week 83Dyspnoea, Week 84Dyspnoea, Week 85Dyspnoea, Week 86Dyspnoea, Week 87
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)0.570.420.250.150.270.300.230.170.270.300.260.220.070.070.100.000.220.110.110.200.030.300.00-0.090.270.120.050.00-0.250.210.180.410.000.400.000.060.310.230.220.430.360.250.300.140.630.300.500.501.000.800.880.750.701.131.170.380.671.171.000.501.000.830.500.000.000.001.000.750.501.000.501.001.500.500.750.500.501.000.500.750.750.000.000.500.000.000.000.13-0.01-0.03-0.02-0.01-0.18-0.20-0.10-0.24-0.070.02-0.08-0.09-0.20-0.05-0.10-0.040.02-0.230.00-0.17-0.20-0.22-0.13-0.45-0.46-0.50-0.45-0.50-0.88-0.86-0.73-0.77-0.80-0.14-0.17-0.250.05-0.50-0.360.210.170.100.360.250.100.500.170.670.300.130.630.700.500.670.500.500.670.330.750.500.330.000.000.500.000.250.250.250.250.00-0.500.000.000.000.250.000.250.000.250.250.000.000.500.000.000.000.420.330.250.430.550.500.630.500.460.430.360.390.430.260.360.180.480.390.290.600.270.550.340.360.220.320.350.350.200.180.180.220.200.160.110.110.280.400.270.570.310.300.400.490.650.560.300.601.400.241.051.100.761.101.000.400.530.800.601.200.730.670.300.200.600.400.500.200.300.300.100.000.000.200.300.300.400.300.100.500.400.600.600.800.600.400.40

[back to top]

Time to Deterioration (TTD) in Patient-reported Lung Cancer Symptoms Score as Assessed by the Symptoms in Lung Cancer (SILC) Scale Symptom Score in the TC3 or IC3-WT Populations

TTD in each of the patient-reported lung cancer symptoms with use of the SILC scale. The SILC scale is a nine-item content valid self-report measure of lung cancer symptoms. It measures severity of cough, dyspnea, and chest pain with a total symptom severity score. Each SILC symptom scale (dyspnea, cough, chest pain) score was calculated as the average of the component items (range 0 to 4). An increase in score suggested worsening in symptomatology. A symptom score change of 0.3 points for the dyspnea and cough scores was considered to be clinically significant; whereas a symptom score change of 0.5 points for the chest pain score was considered to be clinically significant. (NCT02409342)
Timeframe: Baseline until data cut-off on 10 September 2018 (up to approximately 38 months)

,
InterventionMonths (Median)
CoughDyspneaChest pain
Atezolizumab3.51.31.7
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)3.41.01.1

[back to top]

Overall Survival (OS) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations

OS is defined as the time from randomization to death from any cause. (NCT02409342)
Timeframe: From randomization to death from any cause until data cut-off on 4 February 2020 (up to approximately 54.5 months)

,
InterventionMonths (Median)
TC2/3 or IC2/3-WT PopulationTC1/2/3 or IC1/2/3-WT Population
Atezolizumab19.918.9
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)16.114.7

[back to top]

Percentage of Participants Who Are Alive at 1 Year in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations

(NCT02409342)
Timeframe: Baseline to 1 year or death, whichever occurs first until clinical cut-off date on 4 February 2020 (up to approximately 54.5 months)

,
InterventionPercentage of participants (Number)
1-Year TC2/3 or IC2/3-WT Population1-Year TC1/2/3 or IC1/2/3-WT Population
Atezolizumab63.3959.95
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)58.6554.89

[back to top]

Percentage of Participants Who Are Alive at 2 Years in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations

(NCT02409342)
Timeframe: Baseline to 2 years or death, whichever occurs first until clinical cut-off date on 4 February 2020 (up to approximately 54.5 months)

,
InterventionPercentage of participants (Number)
2-Years TC2/3 or IC2/3-WT Population2-Years TC1/2/3 or IC1/2/3-WT Population
Atezolizumab44.1541.76
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)35.4230.82

[back to top]

Percentage of Participants With Anti-therapeutic Antibodies (ATAs)

(NCT02409342)
Timeframe: Baseline until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionPercentage of participants (Number)
Baseline evaluable participantsPost-baseline evaluable participants
Atezolizumab1.424.3

[back to top]

Change From Baseline in Patient-reported Lung Cancer Symptoms Score as Assessed by the SILC Scale Symptom Score in the TC3 or IC3-WT Populations

Change from baseline in each of the patient-reported lung cancer symptoms with use of the SILC scale. The SILC scale is a nine-item content valid self-report measure of lung cancer symptoms. It measures severity of cough, dyspnea, and chest pain with a total symptom severity score. Each SILC symptom scale (dyspnea, cough, chest pain) score was calculated as the average of the component items (range 0 to 4). An increase in score suggested worsening in symptomatology. A symptom score change of 0.3 points for the dyspnea and cough scores was considered to be clinically significant; whereas a symptom score change of 0.5 points for the chest pain score was considered to be clinically significant. (NCT02409342)
Timeframe: Baseline until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionScore on a scale (Mean)
Chest Pain, Week 1Chest Pain, Week 2Chest Pain, Week 3Chest Pain, Week 4Chest Pain, Week 5Chest Pain, Week 6Chest Pain, Week 7Chest Pain, Week 8Chest Pain, Week 9Chest Pain, Week 10Chest Pain, Week 11Chest Pain, Week 12Chest Pain, Week 13Chest Pain, Week 14Chest Pain, Week 15Chest Pain, Week 16Chest Pain, Week 17Chest Pain, Week 18Chest Pain, Week 19Chest Pain, Week 20Chest Pain, Week 21Chest Pain, Week 22Chest Pain, Week 23Chest Pain, Week 24Chest Pain, Week 25Chest Pain, Week 26Chest Pain, Week 27Chest Pain, Week 28Chest Pain, Week 29Chest Pain, Week 30Chest Pain, Week 31Chest Pain, Week 32Chest Pain, Week 33Chest Pain, Week 34Chest Pain, Week 35Chest Pain, Week 36Chest Pain, Week 37Chest Pain, Week 38Chest Pain, Week 39Chest Pain, Week 40Chest Pain, Week 41Chest Pain, Week 42Chest Pain, Week 43Chest Pain, Week 44Chest Pain, Week 45Chest Pain, Week 46Chest Pain, Week 47Chest Pain, Week 48Chest Pain, Week 49Chest Pain, Week 50Chest Pain, Week 51Chest Pain, Week 52Chest Pain, Week 53Chest Pain, Week 54Chest Pain, Week 55Chest Pain, Week 56Chest Pain, Week 57Chest Pain, Week 58Chest Pain, Week 59Chest Pain, Week 60Chest Pain, Week 61Chest Pain, Week 62Chest Pain, Week 63Chest Pain, Week 64Chest Pain, Week 65Chest Pain, Week 66Chest Pain, Week 67Chest Pain, Week 68Chest Pain, Week 69Chest Pain, Week 70Chest Pain, Week 71Chest Pain, Week 72Chest Pain, Week 73Chest Pain, Week 74Chest Pain, Week 75Chest Pain, Week 76Chest Pain, Week 77Chest Pain, Week 78Chest Pain, Week 79Chest Pain, Week 80Chest Pain, Week 81Chest Pain, Week 82Chest Pain, Week 83Chest Pain, Week 84Chest Pain, Week 85Chest Pain, Week 86Chest Pain, Week 87Chest Pain, Week 88Chest Pain, Week 89Chest Pain, Week 90Chest Pain, Week 91Chest Pain, Week 92Chest Pain, Week 93Chest Pain, Week 94Chest Pain, Week 95Chest Pain, Week 96Chest Pain, Week 97Chest Pain, Week 98Chest Pain, Week 99Chest Pain, Week 100Chest Pain, Week 101Chest Pain, Week 102Chest Pain, Week 103Chest Pain, Week 104Chest Pain, Week 105Chest Pain, Week 106Chest Pain, Week 107Chest Pain, Week 108Chest Pain, Week 109Chest Pain, Week 110Chest Pain, Week 111Chest Pain, Week 112Chest Pain, Week 113Chest Pain, Week 114Chest Pain, Week 115Chest Pain, Week 116Chest Pain, Week 117Chest Pain, Week 118Chest Pain, Week 119Chest Pain, Week 120Chest Pain, Week 121Chest Pain, Week 122Chest Pain, Week 123Chest Pain, Week 124Chest Pain, Week 125Chest Pain, Week 126Chest Pain, Week 127Chest Pain, Week 128Chest Pain, Week 129Chest Pain, Week 130Chest Pain, Week 131Cough, Week 1Cough, Week 2Cough, Week 3Cough, Week 4Cough, Week 5Cough, Week 6Cough, Week 7Cough, Week 8Cough, Week 9Cough, Week 10Cough, Week 11Cough, Week 12Cough, Week 13Cough, Week 14Cough, Week 15Cough, Week 16Cough, Week 17Cough, Week 18Cough, Week 19Cough, Week 20Cough, Week 21Cough, Week 22Cough, Week 23Cough, Week 24Cough, Week 25Cough, Week 26Cough, Week 27Cough, Week 28Cough, Week 29Cough, Week 30Cough, Week 31Cough, Week 32Cough, Week 33Cough, Week 34Cough, Week 35Cough, Week 36Cough, Week 37Cough, Week 38Cough, Week 39Cough, Week 40Cough, Week 41Cough, Week 42Cough, Week 43Cough, Week 44Cough, Week 45Cough, Week 46Cough, Week 47Cough, Week 48Cough, Week 49Cough, Week 50Cough, Week 51Cough, Week 52Cough, Week 53Cough, Week 54Cough, Week 55Cough, Week 56Cough, Week 57Cough, Week 58Cough, Week 59Cough, Week 60Cough, Week 61Cough, Week 62Cough, Week 63Cough, Week 64Cough, Week 65Cough, Week 66Cough, Week 67Cough, Week 68Cough, Week 69Cough, Week 70Cough, Week 71Cough, Week 72Cough, Week 73Cough, Week 74Cough, Week 75Cough, Week 76Cough, Week 77Cough, Week 78Cough, Week 79Cough, Week 80Cough, Week 81Cough, Week 82Cough, Week 83Cough, Week 84Cough, Week 85Cough, Week 86Cough, Week 87Cough, Week 88Cough, Week 89Cough, Week 90Cough, Week 91Cough, Week 92Cough, Week 93Cough, Week 94Cough, Week 95Cough, Week 96Cough, Week 97Cough, Week 98Cough, Week 99Cough, Week 100Cough, Week 101Cough, Week 102Cough, Week 103Cough, Week 104Cough, Week 105Cough, Week 106Cough, Week 107Cough, Week 108Cough, Week 109Cough, Week 110Cough, Week 111Cough, Week 112Cough, Week 113Cough, Week 114Cough, Week 115Cough, Week 116Cough, Week 117Cough, Week 118Cough, Week 119Cough, Week 120Cough, Week 121Cough, Week 122Cough, Week 123Cough, Week 124Cough, Week 125Cough, Week 126Cough, Week 127Cough, Week 128Cough, Week 129Cough, Week 130Cough, Week 131Dyspnoea, Week 1Dyspnoea, Week 2Dyspnoea, Week 3Dyspnoea, Week 4Dyspnoea, Week 5Dyspnoea, Week 6Dyspnoea, Week 7Dyspnoea, Week 8Dyspnoea, Week 9Dyspnoea, Week 10Dyspnoea, Week 11Dyspnoea, Week 12Dyspnoea, Week 13Dyspnoea, Week 14Dyspnoea, Week 15Dyspnoea, Week 16Dyspnoea, Week 17Dyspnoea, Week 18Dyspnoea, Week 19Dyspnoea, Week 20Dyspnoea, Week 21Dyspnoea, Week 22Dyspnoea, Week 23Dyspnoea, Week 24Dyspnoea, Week 25Dyspnoea, Week 26Dyspnoea, Week 27Dyspnoea, Week 28Dyspnoea, Week 29Dyspnoea, Week 30Dyspnoea, Week 31Dyspnoea, Week 32Dyspnoea, Week 33Dyspnoea, Week 34Dyspnoea, Week 35Dyspnoea, Week 36Dyspnoea, Week 37Dyspnoea, Week 38Dyspnoea, Week 39Dyspnoea, Week 40Dyspnoea, Week 41Dyspnoea, Week 42Dyspnoea, Week 43Dyspnoea, Week 44Dyspnoea, Week 45Dyspnoea, Week 46Dyspnoea, Week 47Dyspnoea, Week 48Dyspnoea, Week 49Dyspnoea, Week 50Dyspnoea, Week 51Dyspnoea, Week 52Dyspnoea, Week 53Dyspnoea, Week 54Dyspnoea, Week 55Dyspnoea, Week 56Dyspnoea, Week 57Dyspnoea, Week 58Dyspnoea, Week 59Dyspnoea, Week 60Dyspnoea, Week 61Dyspnoea, Week 62Dyspnoea, Week 63Dyspnoea, Week 64Dyspnoea, Week 65Dyspnoea, Week 66Dyspnoea, Week 67Dyspnoea, Week 68Dyspnoea, Week 69Dyspnoea, Week 70Dyspnoea, Week 71Dyspnoea, Week 72Dyspnoea, Week 73Dyspnoea, Week 74Dyspnoea, Week 75Dyspnoea, Week 76Dyspnoea, Week 77Dyspnoea, Week 78Dyspnoea, Week 79Dyspnoea, Week 80Dyspnoea, Week 81Dyspnoea, Week 82Dyspnoea, Week 83Dyspnoea, Week 84Dyspnoea, Week 85Dyspnoea, Week 86Dyspnoea, Week 87Dyspnoea, Week 88Dyspnoea, Week 89Dyspnoea, Week 90Dyspnoea, Week 91Dyspnoea, Week 92Dyspnoea, Week 93Dyspnoea, Week 94Dyspnoea, Week 95Dyspnoea, Week 96Dyspnoea, Week 97Dyspnoea, Week 98Dyspnoea, Week 99Dyspnoea, Week 100Dyspnoea, Week 101Dyspnoea, Week 102Dyspnoea, Week 103Dyspnoea, Week 104Dyspnoea, Week 105Dyspnoea, Week 106Dyspnoea, Week 107Dyspnoea, Week 108Dyspnoea, Week 109Dyspnoea, Week 110Dyspnoea, Week 111Dyspnoea, Week 112Dyspnoea, Week 113Dyspnoea, Week 114Dyspnoea, Week 115Dyspnoea, Week 116Dyspnoea, Week 117Dyspnoea, Week 118Dyspnoea, Week 119Dyspnoea, Week 120Dyspnoea, Week 121Dyspnoea, Week 122Dyspnoea, Week 123Dyspnoea, Week 124Dyspnoea, Week 125Dyspnoea, Week 126Dyspnoea, Week 127Dyspnoea, Week 128Dyspnoea, Week 129Dyspnoea, Week 130Dyspnoea, Week 131
Atezolizumab0.310.330.430.430.280.250.300.200.330.250.210.330.300.270.450.190.220.260.230.230.110.190.330.180.290.110.200.230.180.320.330.350.040.310.280.080.390.310.140.300.300.330.180.420.310.370.310.170.470.200.310.540.290.250.150.290.080.190.000.380.360.430.290.400.350.45-0.090.390.500.180.450.420.270.500.330.000.190.210.310.720.080.080.500.430.380.210.290.43-0.200.07-0.070.330.500.200.330.330.500.330.880.500.830.500.631.001.000.501.001.501.752.001.501.501.502.001.752.002.002.001.752.502.003.002.503.003.003.002.003.003.002.002.000.100.19-0.01-0.030.00-0.18-0.06-0.13-0.06-0.10-0.07-0.09-0.20-0.07-0.19-0.19-0.21-0.26-0.27-0.33-0.19-0.170.05-0.120.14-0.03-0.050.080.050.120.05-0.04-0.29-0.02-0.09-0.36-0.04-0.07-0.20-0.390.05-0.200.03-0.11-0.260.08-0.030.230.260.000.060.270.380.070.120.140.150.080.06-0.040.000.14-0.040.350.120.45-0.090.610.500.230.550.420.640.560.670.310.560.360.630.330.580.250.670.290.380.570.640.640.500.430.500.670.790.300.670.580.570.670.250.600.500.500.250.000.33-0.500.000.000.500.500.500.000.500.500.500.500.500.500.502.002.002.002.002.002.002.002.002.002.002.002.000.120.290.250.280.210.210.320.250.340.490.240.350.230.260.390.260.160.210.330.230.400.220.380.420.500.280.370.450.380.540.410.580.360.280.27-0.020.420.500.250.440.530.520.410.310.530.370.390.390.470.530.590.720.400.630.720.610.450.600.180.570.540.660.770.660.940.670.330.820.740.750.840.800.670.890.710.530.650.400.550.690.530.530.870.890.830.660.661.200.560.740.510.871.200.480.871.131.260.800.700.881.130.960.551.071.070.401.131.501.301.501.601.501.501.601.401.301.501.301.502.202.402.202.202.202.202.202.202.202.202.202.20

[back to top]

Progression-free Survival (PFS) in the TC3 or IC3-WT Populations

PFS is defined as the time from randomization to the first occurrence of disease progression, as determined by the investigator with use of RECIST v1.1, or death from any cause, whichever occurs first. PFS could not be formally tested. (NCT02409342)
Timeframe: From randomization to the first occurrence of disease progression or death from any cause, whichever occurs first until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionMonths (Number)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)5.0
Atezolizumab8.1

[back to top]

Percentage of Participants With Objective Response (ORR) in the TC3 or IC3-WT Populations

Objective response (partial response plus complete response) as determined by the investigator according to RECIST v1.1. (NCT02409342)
Timeframe: Every 6 weeks for 48 weeks following Day 1, thereafter every 9 weeks after completion of Week 48 tumor assessment, regardless of treatment delays, until radiographic disease progression until data cut-off on 10 Sep 2018 (up to approximately 38 months)

InterventionPercentage of participants (Number)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)28.6
Atezolizumab38.3

[back to top]

Percentage of Participants With at Least One Adverse Event

Percentage of participants with at least one adverse event. (NCT02409342)
Timeframe: Baseline up to until data cut-off on 8 March 2022 (up to approximately 79.5 months)

InterventionPercentage of participants (Number)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)95.1
Atezolizumab92.3

[back to top]

Percentage of Participants Who Are Alive at 2 Years in the TC3 or IC3-WT Populations

(NCT02409342)
Timeframe: Baseline to 2 years or death, whichever occurs first until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionPercentage of participants (Number)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)24.79
Atezolizumab45.49

[back to top]

Percentage of Participants Who Are Alive at 1 Year in the TC3 or IC3-WT Populations

(NCT02409342)
Timeframe: Baseline to 1 year or death, whichever occurs first until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionPercentage of participants (Number)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)50.64
Atezolizumab64.90

[back to top]

Progression-free Survival (PFS) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations

PFS is defined as the time from randomization to the first occurrence of disease progression, as determined by the investigator with use of RECIST v1.1, or death from any cause, whichever occurs first. PFS could not be formally tested. (NCT02409342)
Timeframe: From randomization to the first occurrence of disease progression or death from any cause, whichever occurs first until data cut-off on 4 February 2020 (up to approximately 54.5 months)

,
InterventionMonths (Number)
TC2/3 or IC2/3-WT PopulationTC1/2/3 or IC1/2/3-WT Population
Atezolizumab7.35.8
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)5.55.6

[back to top]

OS in Participants With PD-L1 Expression

OS is defined as the time from randomization to death from any cause. (NCT02409342)
Timeframe: From randomization to death from any cause until data cut-off on 10 September 2018 (up to approximately 38 months)

,
InterventionMonths (Median)
SP263 >=50%-WT PopulationSP263 >=25%-WT PopulationSP263 =1%-WT Population
Atezolizumab19.518.217.8
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)16.112.614.0

[back to top]

Minimum Observed Serum Concentration (Cmin) of Atezolizumab

(NCT02409342)
Timeframe: Prior to infusion (0 hour) on Day 1 of Cycles 2, 3, 4, 8, 16, and every eighth cycle thereafter, and at treatment discontinuation until data cut-off on 10 September 2018 (up to approximately 38 months) (cycle duration = 21 days)

Interventionmicrograms per milliliter (μg/ mL) (Mean)
Cycle 2 Day 1Cycle 3 Day 1Cycle 4 Day 1Cycle 8 Day 1Cycle 16 Day 1Cycle 24 Day 1Cycle 32 Day 1Cycle 40 Day 1Cycle 48 Day 1Treatment Discontinuation Visit
Atezolizumab76.7121154201213245276252555121

[back to top]

Investigator-Assessed PFS in Participants With PD-L1 Expression According to RECIST v1.1

Investigator-assessed PFS according to RECIST v1.1 in the PD-L1 (defined with SP263 IHC assay) (NCT02409342)
Timeframe: From randomization to the first occurrence of disease progression or death from any cause, whichever occurs first until data cut-off on 10 September 2018 (up to approximately 38 months)

,
InterventionMonths (Median)
SP263 >=50%-WT PopulationSP263 >=25%-WT PopulationSP263 >=1%-WT Population
Atezolizumab7.06.96.8
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)4.94.95.4

[back to top]

Investigator-Assessed PFS in Participants With bTMB According to RECIST v1.1

PFS according to RECIST v1.1 in the bTMB subpopulations. (NCT02409342)
Timeframe: From randomization to the first occurrence of disease progression or death from any cause, whichever occurs first until data cut-off on 10 September 2018 (up to approximately 38 months)

,
InterventionMonths (Median)
bTMB >=10-WT PopulationbTMB >=16-WT PopulationbTMB >=20-WT Population
Atezolizumab5.56.86.8
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)4.34.45.2

[back to top]

OS in Participants With Blood Tumor Mutational Burden (bTMB)

OS is defined as the time from randomization to death from any cause. (NCT02409342)
Timeframe: From randomization to death from any cause until data cut-off on 10 September 2018 (up to approximately 38 months)

,
InterventionMonths (Median)
bTMB >=10-WT PopulationbTMB >=16-WT PopulationbTMB >=20-WT Population
Atezolizumab11.213.917.2
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)10.38.510.5

[back to top]

Overall Survival (OS) in the TC3 or IC3-WT Populations

OS is defined as the time from randomization to death from any cause. (NCT02409342)
Timeframe: From randomization to death from any cause until data cut-off on 10 September 2018 (up to approximately 38 months)

InterventionMonths (Median)
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)13.1
Atezolizumab20.2

[back to top]

Maximum Observed Serum Concentration (Cmax) of Atezolizumab

(NCT02409342)
Timeframe: 0 hour (predose) and 30 minutes after atezolizumab infusion on Day 1 (infusion duration = up to 1 hour)

Interventionmicrograms per milliliter (μg/ mL) (Mean)
Atezolizumab411

[back to top]

Percentage of Participants With Objective Response (ORR) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations

Objective response (partial response plus complete response) as determined by the investigator according to RECIST v1.1. (NCT02409342)
Timeframe: Every 6 weeks for 48 weeks following Day 1, thereafter every 9 weeks after completion of Week 48 tumor assessment, regardless of treatment delays, until radiographic disease progression until data cut-off on 4 Feb 2020 (up to approximately 54.5 months)

,
InterventionPercentage of participants (Number)
TC2/3 or IC2/3-WT PopulationTC1/2/3 or IC1/2/3-WT Population
Atezolizumab33.731.4
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)32.132.1

[back to top]

Duration of Response (DOR) in the TC2/3 or IC2/3-WT and TC1/2/3 or IC1/2/3-WT Populations

DOR is defined as the time from the first occurrence of a documented objective response to the time of disease progression, as determined by the investigator with use of RECIST v1.1, or death from any cause, whichever occurs first. (NCT02409342)
Timeframe: From first occurrence of a complete response or partial response, whichever occurs first, until first date that progressive disease or death is documented, whichever occurs first until data cut-off on 4 February 2020 (up to approximately 54.5 months)

,
InterventionMonths (Number)
TC2/3 or IC2/3-WT PopulationTC1/2/3 or IC1/2/3-WT Population
Atezolizumab38.926.3
Chemotherapy (Carboplatin/ Cisplatin) + (Pemetrexed/ Gemcitabine)5.85.7

[back to top]

Overall Survival

Examined by estimating a Kaplan-Meier survival curve using all patients enrolled. (NCT02426658)
Timeframe: The duration of time from the start of treatment to date of death or date of last contact, assessed up to 2 years

Interventionmonths (Median)
Treatment (Pemetrexed Disodium)2.4

[back to top]

Time to Tumor Progression

It will be determined whether each patient has a progression (or dies) before or after 12 weeks. A 95% exact (Clopper Pearson) confidence interval will then be around the proportion with PFS greater than or equal to 12 weeks. If this confidence interval includes 50% then that would provide evidence that the therapy is potentially promising. If the upper bound of the confidence interval does not include 50% then this would indicate that the treatment may not be promising for patients. In addition, a Kaplan Meier survival curve will be constructed to describe the time to progression data. (NCT02426658)
Timeframe: The duration of time from the start of treatment to the time of progression, death, or date of last contact, assessed up to 2 years

Interventionmonths (Median)
Treatment (Pemetrexed Disodium)2.0

[back to top]

Change in Quality of Life (QOL), Assessed by the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (QLQ-C30) and QLQ-Lung Cancer 13-item (LC13)

Quality of life will be assessed at each treatment time (i.e. every three weeks). A longitudinal mixed models analysis will be used to look at QOL over the time course. A paired t-test will also be calculated to see if the average change is more than 0 (worsening) versus a two-sided alternative that the difference is 0 or better. Score range from 0-100 (1 = not at all, 2 = a little, 3 = quite a bit, or 4 = very much). The higher the score, the greater the change in the quality of life for the worse. (NCT02426658)
Timeframe: Baseline to 12 weeks

Interventionscore on a scale (Mean)
Baseline12 weeks
Treatment (Pemetrexed Disodium)51.282158.3333

[back to top]

Incidence of Hematologic Toxicity, Graded According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0

The number and type of toxicities observed during this protocol will be estimated, focusing on unexpected grade 3 or higher toxicities. No formal statistical tests will be done on these estimates. (NCT02426658)
Timeframe: Up to 30 days

InterventionParticipants (Count of Participants)
AnemiaCD4 lymphocyte decreasedFebrile neutropeniaLeukocytosisLymphocyte count decreasedNeutrophil count decreasedPlatelet count decreasedWhite blood cell count decreased
Treatment (Pemetrexed Disodium)133119111010

[back to top]

Response Rate

Response rate will be estimated every 6 weeks for patients, and these estimates will be presented with confidence intervals. (NCT02426658)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Partial responseStable diseaseProgressive disease
Treatment (Pemetrexed Disodium)162

[back to top]

Phase 1b: Frequency of Treatment Emergent Adverse Events (TEAEs) as Assessed by CTCAE v4.03.

The number and percent of patients with a given TEAE will be summarized overall and by system organ class and preferred term by treatment group. The number and percent of patients with TEAEs will be tabulated by maximum severity. (NCT02439450)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Arm 5: Viagenpumatucel-L + Nivolumab CPI Naive47
Arm 6: Viagenpumatucel-L + Pembrolizumab66
Arm 5: Viagenpumatucel-L + Nivolumab CPI Progressor2
Arm 6: Viagenpumatucel-L + Pembrolizumab + Pemetrexed4

[back to top]

DoR; PD-L1 (TC >=1%) Analysis Set Population

The DoR per RECIST 1.1 using BICR assessments was defined as the time from the date of first documented response (CR or PR) until the first date of documented progression or death in the absence of disease progression (ie, date of PFS event or censoring - date of first response + 1). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab MonotherapyNA
Durvalumab + TremelimumabNA
SoC Chemotherapy4.4

[back to top]

DoR; FAS Population

The DoR per RECIST 1.1 using BICR assessments was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (ie, date of PFS event or censoring - date of first response + 1). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab MonotherapyNA
Durvalumab + TremelimumabNA
SoC Chemotherapy4.3

[back to top]

Ctrough_ss of Tremelimumab

Blood samples were collected to determine the Ctrough_ss of tremelimumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. (NCT02453282)
Timeframe: Pre-dose at Week 12.

Interventionmcg/mL (Mean)
Durvalumab + Tremelimumab4.9

[back to top]

Cmax_ss of Tremelimumab

Blood samples were collected to determine the Cmax_ss of tremelimumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. (NCT02453282)
Timeframe: Within 1 hour after end of infusion on infusion day at Week 12.

Interventionmcg/mL (Mean)
Durvalumab + Tremelimumab24.8

[back to top]

PFS; PD-L1 (TC >=25%) Analysis Set Population, Durvalumab Monotherapy Vs SoC Chemotherapy and Durvalumab + Tremelimumab Vs Durvalumab Monotherapy

The PFS per RECIST 1.1 using BICR assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). The PD was defined as at least a 20% increase in the sum of diameters of TLs and an absolute increase of at least 5 mm, taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy4.7
Durvalumab + Tremelimumab3.9
SoC Chemotherapy5.4

[back to top]

Serum Concentrations of Tremelimumab

Blood samples were collected to determine the serum concentration of tremelimumab. (NCT02453282)
Timeframe: Pre-dose and within 1 hour after end of infusion at Week 0 and 12, and at follow-up Month 3.

Interventionmcg/mL (Mean)
At Week 0: Pre-infusionAt Week 0: End of infusionAt Week 12: Pre-infusionAt Week 12: End of infusionAt follow-up Month 3
Durvalumab + TremelimumabNA22.64.924.80.5

[back to top]

Serum Concentrations of Durvalumab

Blood samples were collected to determine the serum concentration of durvalumab. (NCT02453282)
Timeframe: Pre-dose and within 1 hour after end of infusion at Week 0, 12 and 24, and at follow-up Month 3.

,
Interventionmicrogram per milliliter (mcg/mL) (Mean)
At Week 0: Pre-infusionAt Week 0: End of infusionAt Week 12: Pre-infusionAt Week 12: End of infusionAt Week 24: Pre-infusionAt Week 24: End of infusionAt follow-up Month 3
Durvalumab + TremelimumabNA444.3140.8506.1197.0553.241.4
Durvalumab MonotherapyNA484.5139.5625.3163.0598.249.3

[back to top]

Number of Participants With Anti-Drug Antibody (ADA) Response to Durvalumab

Blood samples were measured for the presence of ADAs and ADA-neutralizing antibodies (nAb) for durvalumab using validated assays. Tiered analysis was performed to include screening, confirmatory, and titer assay components, and positive-negative cut points previously statistically determined from drug-naïve validation samples were employed. Immunogenicity results were analyzed by summarizing the number of participants who developed detectable ADAs against durvalumab. Persistently positive is defined as having at least 2 post-baseline ADA positive measurements with at least 16 weeks (112 days) between the first and last positive measurements, or an ADA positive result at the last available assessment. Transiently positive is defined as having at least one post-baseline ADA positive measurement and not fulfilling the conditions for persistently positive. (NCT02453282)
Timeframe: At Weeks 0, 12, and 24; 3 and 6 months after last dose of study treatment.

,
InterventionParticipants (Count of Participants)
ADA positive at any timeTreatment-emergent ADA positiveADA positive at baseline and post-baselineADA positive at post-baseline onlyADA positive at baseline onlyTreatment-boosted ADAPersistent positiveTransient positivenAb positive at any visit
Durvalumab + Tremelimumab1481850901
Durvalumab Monotherapy1783860832

[back to top]

Number of Participants With ADA Response to Tremelimumab

Blood samples were measured for the presence of ADAs and ADA-nAb for tremelimumab using validated assays. Tiered analysis was performed to include screening, confirmatory, and titer assay components, and positive-negative cut points previously statistically determined from drug-naïve validation samples were employed. Immunogenicity results were analyzed by summarizing the number of participants who developed detectable ADAs against tremelimumab. Persistently positive is defined as having at least 2 post-baseline ADA positive measurements with at least 16 weeks (112 days) between the first and last positive measurements, or an ADA positive result at the last available assessment. Transiently positive is defined as having at least one post-baseline ADA positive measurement and not fulfilling the conditions for persistently positive. (NCT02453282)
Timeframe: At Weeks 0 and 12; 3 and 6 months after last dose of study treatment.

InterventionParticipants (Count of Participants)
ADA positive at any timeTreatment-emergent ADA positiveADA positive at baseline and post-baselineADA positive at post-baseline onlyADA positive at baseline onlyTreatment-boosted ADAPersistent positiveTransient positivenAb positive at any visit
Durvalumab + Tremelimumab33281284025425

[back to top] [back to top]

Trough Serum Concentration at Steady State (Ctrough_ss) of Durvalumab

Blood samples were collected to determine the Ctrough_ss of durvalumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. (NCT02453282)
Timeframe: Pre-dose at Week 12.

Interventionmcg/mL (Mean)
Durvalumab Monotherapy139.5
Durvalumab + Tremelimumab140.8

[back to top]

Time From Randomization to Second Progression (PFS2); PD-L1 (TC >=25%) Analysis Set Population

The PFS2 was defined as the time from the date of randomization to the earliest of the progression events (subsequent to that used for the primary variable PFS and excluding any confirmation of progression scans performed for first progression) or death (ie, date of PFS2 event or censoring - date of randomization + 1). The second progression event was determined by local standard clinical practice which may have included any of the following: objective radiological imaging, symptomatic progression, or death. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to Week 48, then every 8 weeks thereafter until 1st progression. Disease then assessed per local practice until 2nd progression. Assessed up to data cut-off date (maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy12.7
Durvalumab + Tremelimumab10.9
SoC Chemotherapy10.4

[back to top]

Progression-Free Survival (PFS); PD-L1 (TC >=25%) Analysis Set Population, Durvalumab + Tremelimumab Vs SoC Chemotherapy

The PFS per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST 1.1) using blinded independent central review (BICR) assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). Progressive disease (PD) was defined as at least a 20% increase in the sum of diameters of target lesions (TLs) and an absolute increase of at least 5 millimeter (mm), taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab + Tremelimumab3.9
SoC Chemotherapy5.4

[back to top]

PFS2; PD-L1 (TC >=1%) Analysis Set Population

The PFS2 was defined as the time from the date of randomization to the earliest of the progression events (subsequent to that used for the primary variable PFS and excluding any confirmation of progression scans performed for first progression) or death (ie, date of PFS2 event or censoring - date of randomization + 1). The second progression event was determined by local standard clinical practice which may have included any of the following: objective radiological imaging, symptomatic progression, or death. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to Week 48, then every 8 weeks thereafter until 1st progression. Disease then assessed per local practice until 2nd progression. Assessed up to data cut-off date (maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy10.6
Durvalumab + Tremelimumab9.4
SoC Chemotherapy10.5

[back to top]

PFS2; FAS Population

The PFS2 was defined as the time from the date of randomization to the earliest of the progression events (subsequent to that used for the primary variable PFS and excluding any confirmation of progression scans performed for first progression) or death (ie, date of PFS2 event or censoring - date of randomization + 1). The second progression event was determined by local standard clinical practice which may have included any of the following: objective radiological imaging, symptomatic progression, or death. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to Week 48, then every 8 weeks thereafter until 1st progression. Disease then assessed per local practice until 2nd progression. Assessed up to data cut-off date (maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy9.3
Durvalumab + Tremelimumab9.8
SoC Chemotherapy10.1

[back to top]

PFS; PD-L1 (TC >=1%) Analysis Set Population

The PFS per RECIST 1.1 using BICR assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). The PD was defined as at least a 20% increase in the sum of diameters of TLs and an absolute increase of at least 5 mm, taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy3.6
Durvalumab + Tremelimumab2.8
SoC Chemotherapy5.5

[back to top]

PFS; FAS Population

The PFS per RECIST 1.1 using BICR assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). The PD was defined as at least a 20% increase in the sum of diameters of TLs and an absolute increase of at least 5 mm, taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy2.8
Durvalumab + Tremelimumab2.9
SoC Chemotherapy5.4

[back to top]

Percentage of Participants APF12; PD-L1 (TC >=1%) Analysis Set Population

The APF12 was defined as the percentage of participants who were alive and progression free per RECIST v1.1 using BICR assessments at 12 months after randomization. The PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 12 months.

Interventionpercentage of participants (Number)
Durvalumab Monotherapy27.0
Durvalumab + Tremelimumab20.4
SoC Chemotherapy14.9

[back to top]

Percentage of Participants APF12; FAS Population

The APF12 was defined as the percentage of participants who were alive and progression free per RECIST v1.1 using BICR assessments at 12 months after randomization. The PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 12 months.

Interventionpercentage of participants (Number)
Durvalumab Monotherapy22.5
Durvalumab + Tremelimumab19.8
SoC Chemotherapy13.8

[back to top]

Percentage of Participants Alive and Progression Free at 12 Months (APF12); PD-L1 (TC >=25%) Analysis Set Population

The APF12 was defined as the percentage of participants who were alive and progression free per RECIST v1.1 using BICR assessments at 12 months after randomization. The PFS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 12 months.

Interventionpercentage of participants (Number)
Durvalumab Monotherapy32.3
Durvalumab + Tremelimumab25.8
SoC Chemotherapy14.3

[back to top]

Overall Survival (OS); PD-L1 (TC >=25%) Analysis Set Population, Durvalumab Monotherapy Vs SoC Chemotherapy and Durvalumab + Tremelimumab Vs SoC Chemotherapy

The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: From baseline (Day 1, Week 0) until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy16.3
Durvalumab + Tremelimumab11.9
SoC Chemotherapy12.9

[back to top]

OS; PD-L1 (TC >=25%) Analysis Set Population, Durvalumab + Tremelimumab Vs Durvalumab Monotherapy

The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: From baseline until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy16.3
Durvalumab + Tremelimumab11.9
SoC Chemotherapy12.9

[back to top]

OS; PD-L1 (TC >=1%) Analysis Set Population

The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: From baseline until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy14.6
Durvalumab + Tremelimumab10.9
SoC Chemotherapy12.3

[back to top]

OS; FAS Population

The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT02453282)
Timeframe: From baseline until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab Monotherapy12.3
Durvalumab + Tremelimumab11.2
SoC Chemotherapy11.8

[back to top]

ORR; PD-L1 (TC >=1%) Analysis Set Population

The ORR per RECIST 1.1 using BICR assessments was defined as the percentage of participants with at least 1 visit response of CR or PR. The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionpercentage of participants (Number)
Durvalumab Monotherapy26.5
Durvalumab + Tremelimumab25.3
SoC Chemotherapy33.6

[back to top]

ORR; FAS Population

The ORR per RECIST 1.1 using BICR assessments was defined as the percentage of participants with at least 1 visit response of CR or PR. The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionpercentage of participants (Number)
Durvalumab Monotherapy22.2
Durvalumab + Tremelimumab24.7
SoC Chemotherapy30.1

[back to top]

Objective Response Rate (ORR); PD-L1 (TC >=25%) Analysis Set Population

The ORR per RECIST 1.1 using BICR assessments was defined as the percentage of participants with at least 1 visit response of Complete Response (CR) or Partial Response (PR). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionpercentage of participants (Number)
Durvalumab Monotherapy35.6
Durvalumab + Tremelimumab34.4
SoC Chemotherapy37.7

[back to top]

Maximum Serum Concentration at Steady State (Cmax_ss) of Durvalumab

Blood samples were collected to determine the Cmax_ss of durvalumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. (NCT02453282)
Timeframe: Within 1 hour after end of infusion on infusion day at Week 12.

Interventionmcg/mL (Mean)
Durvalumab Monotherapy625.3
Durvalumab + Tremelimumab506.1

[back to top]

Duration of Response (DoR); PD-L1 (TC >=25%) Analysis Set Population

The DoR per RECIST 1.1 using BICR assessments was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (ie, date of PFS event or censoring - date of first response + 1). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). (NCT02453282)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).

Interventionmonths (Median)
Durvalumab MonotherapyNA
Durvalumab + TremelimumabNA
SoC Chemotherapy4.4

[back to top]

Duration of Response (DoR); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets

"DoR (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of first documented response (CR or PR) until the first date of documented progression or death in the absence of PD (ie, date of PFS event or censoring - date of first response + 1).~bTMB ≥20 mut/Mb, bTMB ≥16 mut/Mb and bTMB ≥12 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥20 mut/Mb, ≥16 mut/Mb and ≥12 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global cohort DCO date (approximately 44 months).

,
Interventionmonths (Median)
bTMB ≥20 mut/Mb analysis setbTMB ≥16 mut/Mb analysis setbTMB ≥12 mut/Mb analysis set
Global: Durvalumab + Tremelimumab11.610.611.5
Global: SoC Chemotherapy4.24.34.3

[back to top]

DoR; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets

"DoR (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of first documented response (CR or PR) until the first date of documented progression or death in the absence of PD (ie, date of PFS event or censoring - date of first response + 1).~PD-L1-negative analysis set included the subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by the Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~Global Cohort: The FAS included all randomized participants prior to the end of global recruitment.~China Cohort: The China FAS included all randomized participants in the China cohort and were used for all China only efficacy analyses." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global or China cohort DCO dates, as applicable (approximately 44 months) for each cohort.

,,,
Interventionmonths (Median)
PD-L1-negative NSCLC analysis setFAS
China: Durvalumab + Tremelimumab10.512.9
China: SoC Chemotherapy6.16.1
Global: Durvalumab + Tremelimumab10.211.1
Global: SoC Chemotherapy4.94.9

[back to top]

APF12; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets

"The APF12 was defined as the percentage of patients who were alive and progression free at 12 months from randomization (ie, PFS rate at 12 months).~PD-L1-negative analysis set included the subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by the Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~Global Cohort: The FAS included all randomized participants prior to the end of global recruitment.~China Cohort: The China FAS included all randomized participants in the China cohort and were used for all China only efficacy analyses." (NCT02542293)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 12 months.

,,,
Interventionpercentage of participants (Number)
PD-L1-negative NSCLC analysis setFAS
China: Durvalumab + Tremelimumab15.623.9
China: SoC Chemotherapy11.316.6
Global: Durvalumab + Tremelimumab18.220.2
Global: SoC Chemotherapy12.114.9

[back to top]

Objective Response Rate (ORR); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, bTMB ≥12 Mut/Mb, tTMB ≥14 Mut/Mb, tTMB ≥12 Mut/Mb, tTMB ≥10 Mut/Mb, and tTMB ≥8 Mut/Mb Analysis Sets

"The ORR (per RECIST 1.1 using Investigator assessments) was defined as the percentage of participants with at least 1 visit response of complete response (CR) or partial response (PR) prior to PD.~bTMB ≥20 mut/Mb, bTMB ≥16 mut/Mb and bTMB ≥12 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥20 mut/Mb, ≥16 mut/Mb and ≥12 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay.~tTMB ≥14 mut/Mb, tTMB ≥12 mut/Mb, tTMB ≥10 mut/Mb and tTMB ≥8 mut/Mb analysis sets included the subset of participants in FAS whose tTMB status was ≥14 mut/Mb, ≥12 mut/Mb, ≥10 mut/Mb and ≥8 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global cohort DCO date (approximately 44 months).

,
Interventionpercentage of participants (Number)
bTMB ≥20 mut/Mb analysis setbTMB ≥16 mut/Mb analysis setbTMB ≥12 mut/Mb analysis settTMB ≥14 mut/Mb analysis settTMB ≥12 mut/Mb analysis settTMB ≥10 mut/Mb analysis settTMB ≥8 mut/Mb analysis set
Global: Durvalumab + Tremelimumab27.531.228.761.342.637.736.7
Global: SoC Chemotherapy43.346.142.044.741.842.541.2

[back to top]

OS; China Cohort: China Programmed Cell Death Ligand 1 (PD-L1) Negative NSCLC Analysis Set

The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT02542293)
Timeframe: From baseline (Day 1, Week 0) until death due to any cause, assessed up to the China cohort DCO date (a maximum of approximately 44 months).

Interventionmonths (Median)
China: Durvalumab + Tremelimumab15.0
China: SoC Chemotherapy11.7

[back to top]

Serum Concentrations of Durvalumab

Blood samples were collected to determine the serum concentration of durvalumab. (NCT02542293)
Timeframe: Pre-dose and within 1 hour after end of infusion at Week 0 and 12; pre-dose on Week 24 and at follow-up Month 3

,
Interventionmicrogram per milliliter (µg/mL) (Geometric Mean)
Week 0: Pre-infusionWeek 0: End of infusionWeek 12: Pre-infusionWeek 12: End of infusionWeek 24: Pre-infusionFollow-up Month 3
China: Durvalumab + TremelimumabNA392.772.4448.985.65.4
Global: Durvalumab + TremelimumabNA418.677.5434.3108.88.8

[back to top]

Time From Randomization to Second Progression or Death (PFS2); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets

"The PFS2 was defined as the time from the date of randomization to the earliest of the progression events subsequent to that used for the primary variable PFS, or death (ie, date of PFS2 event or censoring - date of randomization + 1).~bTMB ≥20 mut/Mb, bTMB ≥16 mut/Mb and bTMB ≥12 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥20 mut/Mb, ≥16 mut/Mb and ≥12 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global cohort DCO date (approximately 44 months).

,
Interventionmonths (Median)
bTMB ≥20 mut/Mb analysis setbTMB ≥16 mut/Mb analysis setbTMB ≥12 mut/Mb analysis set
Global: Durvalumab + Tremelimumab10.610.99.9
Global: SoC Chemotherapy8.610.59.0

[back to top]

Serum Concentrations of Tremelimumab

Blood samples were collected to determine the serum concentration of tremelimumab. (NCT02542293)
Timeframe: Pre-dose and within 1 hour after end of infusion at Week 0 and 12, and at follow-up Month 3

,
Interventionµg/mL (Geometric Mean)
Week 0: Pre-infusionWeek 0: End of infusionWeek 12: Pre-infusionWeek 12: End of infusionFollow-up Month 3
China: Durvalumab + TremelimumabNA18.43.323.2NA
Global: Durvalumab + TremelimumabNA20.33.420.8NA

[back to top]

Overall Survival (OS); Global Cohort: Blood Tumor Mutational Burden (bTMB) ≥20 Mutations Per Megabase (Mut/Mb) Analysis Set

The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT02542293)
Timeframe: From baseline (Day 1, Week 0) until death due to any cause, assessed up to the Global cohort DCO date (a maximum of approximately 44 months).

Interventionmonths (Median)
Global: Durvalumab + Tremelimumab11.7
Global: SoC Chemotherapy9.1

[back to top]

Number of Participants With ADA Response to Tremelimumab

Blood samples were measured for the presence of ADAs and ADA-nAb for tremelimumab using validated assays. ADA prevalence is defined as the percentage of participants with positive ADA result at any time, baseline or post-baseline. Treatment-emergent ADA is defined as the sum of treatment-induced ADA and treatment-boosted ADA. ADA incidence is the percentage of participants who were treatment-emergent ADA-positive. Treatment-boosted ADA is defined as baseline positive ADA titer that was boosted to >=4 fold during the study period. Persistently positive is defined as having at least 2 post baseline ADA positive measurements with at least 16 weeks (112 days) between the first and last positive measurements, or an ADA positive result at the last available assessment. Transiently positive is defined as having at least 1 post baseline ADA positive measurement and not fulfilling the conditions for persistently positive. (NCT02542293)
Timeframe: At Weeks 0 and 12; 3 and 6 months after last dose of study treatment.

,
InterventionParticipants (Count of Participants)
ADA positive at any visit (ADA prevalence)Treatment-emergent ADA positive (ADA incidence)Treatment-boosted ADATreatment-induced ADA (positive post-baseline only)ADA positive post-baseline and positive at baselinePersistent positiveTransient positivenAb positive at any visitADA positive at baseline and not detected post-baseline
China: Durvalumab + Tremelimumab210110200
Global: Durvalumab + Tremelimumab493713642911339

[back to top]

Number of Participants With Anti-Drug Antibody (ADA) Response to Durvalumab

Blood samples were measured for the presence of ADAs and ADA-neutralizing antibodies (nAb) for durvalumab using validated assays. ADA prevalence is defined as the percentage of participants with positive ADA result at any time, baseline or post-baseline. Treatment-emergent ADA is defined as the sum of treatment-induced ADA and treatment-boosted ADA. ADA incidence is the percentage of participants who were treatment-emergent ADA-positive. Treatment-boosted ADA is defined as baseline positive ADA titer that was boosted to >=4 fold during the study period. Persistently positive is defined as having at least 2 post baseline ADA positive measurements with at least 16 weeks (112 days) between the first and last positive measurements, or an ADA positive result at the last available assessment. Transiently positive is defined as having at least 1 post baseline ADA positive measurement and not fulfilling the conditions for persistently positive. (NCT02542293)
Timeframe: At Weeks 0, 12, and 24; 3 and 6 months after last dose of study treatment.

,
InterventionParticipants (Count of Participants)
ADA positive at any visit (ADA prevalence)Treatment-emergent ADA positive (ADA incidence)Treatment-boosted ADATreatment-induced ADA (positive post-baseline only)ADA positive post-baseline and positive at baselinePersistent positiveTransient positivenAb positive at any visitADA positive at baseline and not detected post-baseline
China: Durvalumab + Tremelimumab110101000
Global: Durvalumab + Tremelimumab26120121112313

[back to top]

ORR; Global and China Cohorts: PD-L1-Negative NSCLC, FAS, PD-L1 TC ≥25%, and PD-L1 TC ≥50% Analysis Sets

"The ORR (per RECIST 1.1 using Investigator assessments) was defined as the percentage of participants with at least 1 visit response of CR or PR prior to PD.~PD-L1-negative analysis set included the subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by the Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~Global Cohort: The FAS included all randomized participants prior to the end of global recruitment.~China Cohort: The China FAS included all randomized participants in the China cohort and were used for all China only efficacy analyses.~PD-L1 TC ≥25% and PD-L1 TC ≥50% analysis sets included the subset of participants in the FAS whose PD-L1 status was TC ≥25% and TC ≥50% membrane expression in tumoral tissue, respectively at baseline as defined by the Ventana SP263 PD-L1 Assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global or China cohort DCO dates, as applicable (approximately 44 months) for each cohort.

,,,
Interventionpercentage of participants (Number)
PD-L1-negative NSCLC analysis setFASPD-L1 TC ≥25% analysis setPD-L1 TC ≥50% analysis set
China: Durvalumab + Tremelimumab23.135.954.860.0
China: SoC Chemotherapy41.439.040.646.4
Global: Durvalumab + Tremelimumab23.125.935.237.4
Global: SoC Chemotherapy38.841.743.944.0

[back to top]

OS at Months 12, 18 and 24; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets

"The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1).~PD-L1-negative analysis set included the subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by the Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~Global Cohort: The FAS included all randomized participants prior to the end of global recruitment.~China Cohort: The China FAS included all randomized participants in the China cohort and were used for all China only efficacy analyses." (NCT02542293)
Timeframe: Months 12, 18 and 24

,,,
Interventionpercentage of participants (Number)
Month 12: PD-L1-negative NSCLC analysis setMonth 12: FASMonth 18: PD-L1-negative NSCLC analysis setMonth 18: FASMonth 24: PD-L1-negative NSCLC analysis setMonth 24: FAS
China: Durvalumab + Tremelimumab68.072.844.054.636.044.2
China: SoC Chemotherapy46.453.139.341.817.930.4
Global: Durvalumab + Tremelimumab47.847.734.134.822.125.7
Global: SoC Chemotherapy52.850.034.534.622.323.4

[back to top]

OS at Months 12, 18 and 24; Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets

"The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1).~bTMB ≥20 mut/Mb, bTMB ≥16 mut/Mb and bTMB ≥12 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥20 mut/Mb, ≥16 mut/Mb and ≥12 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay." (NCT02542293)
Timeframe: Months 12, 18 and 24

,
Interventionpercentage of participants (Number)
Month 12: bTMB ≥20 mut/Mb analysis setMonth 12: bTMB ≥16 mut/Mb analysis setMonth 12: bTMB ≥12 mut/Mb analysis setMonth 18: bTMB ≥20 mut/Mb analysis setMonth 18: bTMB ≥16 mut/Mb analysis setMonth 18: bTMB ≥12 mut/Mb analysis setMonth 24: bTMB ≥20 mut/Mb analysis setMonth 24: bTMB ≥16 mut/Mb analysis setMonth 24: bTMB ≥12 mut/Mb analysis set
Global: Durvalumab + Tremelimumab49.350.546.936.235.529.426.124.021.3
Global: SoC Chemotherapy40.848.944.620.428.527.813.618.219.0

[back to top]

OS; Global and China Cohorts: FAS, PD-L1 Tumor Cell (TC) ≥25%, and PD-L1 TC ≥50% Analysis Sets

"The OS was defined as time from date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on last recorded date on which participant was known to be alive.~Global Cohort: The FAS included all randomized participants prior to the end of global recruitment. Any participants recruited in China, after global recruitment had ended, were not included in the FAS.~China Cohort: The China FAS included all randomized participants in the China cohort and were used for all China only efficacy analyses.~PD-L1 TC ≥25% and PD-L1 TC ≥50% analysis sets included the subset of participants in the FAS whose PD-L1 status was TC ≥25% and TC ≥50% membrane expression in tumoral tissue, respectively at baseline as defined by the Ventana SP263 PD-L1 Assay." (NCT02542293)
Timeframe: From baseline (Day 1, Week 0) until death due to any cause, assessed up to the Global or China cohort DCO dates, as applicable (a maximum of approximately 44 months) for each cohort.

,,,
Interventionmonths (Median)
FASPD-L1 TC ≥25% analysis setPD-L1 TC ≥50% analysis set
China: Durvalumab + Tremelimumab20.036.636.6
China: SoC Chemotherapy14.115.815.8
Global: Durvalumab + Tremelimumab10.912.214.1
Global: SoC Chemotherapy12.110.410.5

[back to top]

OS; Global Cohort: bTMB ≥16 Mut/Mb, bTMB ≥12 Mut/Mb, PD-L1-Negative NSCLC, bTMB <20 Mut/Mb, bTMB Non-Evaluable Population, tTMB ≥14 Mut/Mb, tTMB ≥12 Mut/Mb, tTMB ≥10 Mut/Mb, and tTMB ≥8 Mut/Mb Analysis Sets

"OS was defined as time from date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at time of analysis was censored based on last recorded date on which participant was known to be alive.~bTMB ≥16 mut/Mb, bTMB ≥12 mut/Mb and bTMB <20 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥16 mut/Mb, ≥12 mut/Mb and <20 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay.~PD-L1-negative analysis set included the subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by the Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~bTMB non-evaluable analysis set included the subset of participants in FAS whose bTMB status at baseline could not be determined by the GuardantOMNI CDx assay or whose sample was not available.~tTMB analysis sets are defined same as the bTMB analysis sets (NCT02542293)
Timeframe: From baseline (Day 1, Week 0) until death due to any cause, assessed up to the Global cohort DCO date (a maximum of approximately 44 months).

,
Interventionmonths (Median)
bTMB ≥16 mut/Mb analysis setbTMB ≥12 mut/Mb analysis setPD-L1 negative analysis setbTMB <20 mut/Mb analysis setbTMB non-evaluable analysis settTMB ≥14 mut/Mb analysis settTMB ≥12 mut/Mb analysis settTMB ≥10 mut/Mb analysis settTMB ≥8 mut/Mb analysis set
Global: Durvalumab + Tremelimumab12.110.911.19.99.317.511.111.111.0
Global: SoC Chemotherapy11.910.312.511.510.410.613.910.610.2

[back to top]

Alive and Progression-Free at 12 Months (APF12); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, and bTMB ≥12 Mut/Mb Analysis Sets

"The APF12 was defined as the percentage of participants who were alive and progression free at 12 months from randomization (ie, PFS rate at 12 months).~bTMB ≥20 mut/Mb, bTMB ≥16 mut/Mb and bTMB ≥12 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥20 mut/Mb, ≥16 mut/Mb and ≥12 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline then every 6 weeks up to 12 months.

,
Interventionpercentage of participants (Number)
bTMB ≥20 mut/Mb analysis setbTMB ≥16 mut/Mb analysis setbTMB ≥12 mut/Mb analysis set
Global: Durvalumab + Tremelimumab25.622.021.6
Global: SoC Chemotherapy7.012.313.8

[back to top]

PFS; Global and China Cohorts: PD-L1-Negative NSCLC, FAS, PD-L1 TC ≥25%, and PD-L1 TC ≥50% Analysis Sets

"PFS (per RECIST 1.1 using Investigator assessments) was defined as time from date of randomization until date of objective PD or death regardless of whether participant withdrew from randomized therapy or received another anticancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1).~PD-L1-negative analysis set included subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~Global Cohort: FAS included all randomized participants prior to end of global recruitment.~China Cohort: China FAS included all randomized participants in China cohort and were used for all China only efficacy analyses.~PD-L1 TC ≥25% and PD-L1 TC ≥50% analysis sets included subset of participants in FAS whose PD-L1 status was TC ≥25% and TC ≥50% membrane expression in tumoral tissue, respectively at baseline as defined by the Ventana SP263 PD-L1 Assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global or China cohort DCO dates, as applicable (approximately 44 months) for each cohort.

,,,
Interventionmonths (Median)
PD-L1-negative NSCLC analysis setFASPD-L1 TC ≥25% analysis setPD-L1 TC ≥50% analysis set
China: Durvalumab + Tremelimumab5.14.26.86.8
China: SoC Chemotherapy6.06.05.75.7
Global: Durvalumab + Tremelimumab4.14.04.24.6
Global: SoC Chemotherapy5.65.65.45.4

[back to top]

Progression-Free Survival (PFS); Global Cohort: bTMB ≥20 Mut/Mb, bTMB ≥16 Mut/Mb, bTMB ≥12 Mut/Mb, tTMB ≥14 Mut/Mb, tTMB ≥12 Mut/Mb, tTMB ≥10 Mut/Mb, and tTMB ≥8 Mut/Mb Analysis Sets

"The PFS (per Response Evaluation Criteria in Solid Tumors, version 1.1 [RECIST 1.1] using Investigator assessments) was defined as the time from the date of randomization until the date of objective PD or death (by any cause in the absence of progression) regardless of whether the participant withdrew from randomized therapy or received another anticancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1).~bTMB ≥20 mut/Mb, bTMB ≥16 mut/Mb and bTMB ≥12 mut/Mb analysis sets included the subset of participants in FAS whose bTMB status was ≥20 mut/Mb, ≥16 mut/Mb and ≥12 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay.~tTMB ≥14 mut/Mb, tTMB ≥12 mut/Mb, tTMB ≥10 mut/Mb and tTMB ≥8 mut/Mb analysis sets included the subset of participants in FAS whose tTMB status was ≥14 mut/Mb, ≥12 mut/Mb, ≥10 mut/Mb and ≥8 mut/Mb, respectively at baseline as defined by the GuardantOMNI CDx assay." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global cohort DCO date (approximately 44 months).

,
Interventionmonths (Median)
bTMB ≥20 mut/Mb analysis setbTMB ≥16 mut/Mb analysis setbTMB ≥12 mut/Mb analysis settTMB ≥14 mut/Mb analysis settTMB ≥12 mut/Mb analysis settTMB ≥10 mut/Mb analysis settTMB ≥8 mut/Mb analysis set
Global: Durvalumab + Tremelimumab4.24.23.98.75.24.34.4
Global: SoC Chemotherapy5.15.55.15.85.85.15.0

[back to top]

PFS2; Global and China Cohorts: PD-L1-Negative NSCLC and FAS Analysis Sets

"The PFS2 was defined as the time from the date of randomization to the earliest of the progression events subsequent to that used for the primary variable PFS, or death (ie, date of PFS2 event or censoring - date of randomization + 1).~PD-L1-negative analysis set included the subset of participants in FAS whose PD-L1 status was PD-L1-negative at baseline as defined by the Ventana SP263 PD-L1 Assay (ie, <1% PD-L1-membrane expression in tumoral tissue).~Global Cohort: The FAS included all randomized participants prior to the end of global recruitment.~China Cohort: The China FAS included all randomized participants in the China cohort and were used for all China only efficacy analyses." (NCT02542293)
Timeframe: Tumour scans performed at baseline and every 6 weeks up to 48 weeks relative to date of randomization, then every 8 weeks thereafter until confirmed PD/death. Up to Global or China cohort DCO dates, as applicable (approximately 44 months) for each cohort.

,,,
Interventionmonths (Median)
PD-L1-negative NSCLC analysis setFAS
China: Durvalumab + Tremelimumab13.815.5
China: SoC Chemotherapy10.312.9
Global: Durvalumab + Tremelimumab9.19.4
Global: SoC Chemotherapy12.410.4

[back to top] [back to top]

Overall Survival (OS), Groups A-C Only

Overall survival (OS) is defined as the time from randomization to the date of death. (NCT02574078)
Timeframe: up to approximately 60 months

InterventionMonths (Median)
Group A, Cohort A, Nivo20.0
Group A, Cohort A, Beva + Nivo30.8
Group A, Cohort A, Beva18.1
Group A, Cohort B, Nivo28.9
Group A, Cohort B, Peme + Nivo17.4
Group A, Cohort B, Peme18.4
Group B, NivoNA
Group B, BSC13.6
Group C, Nivo3.9
Group C, ICC15.8

[back to top]

Overall Survival (OS), Group D Only

Overall survival (OS) is defined as the time from randomization to the date of death. (NCT02574078)
Timeframe: up to approximately 60 months

Interventionmonths (Median)
Group D, Nivo + ErloNA
Group D, Erlo34.8

[back to top]

Objective Response Rate (ORR), Groups A-E

"Objective response rate (ORR) is defined as the number and percentage of participants with a best overall response (BOR) of confirmed complete response (CR) or partial response (PR). Best overall response (BOR) is defined as the best response designation, recorded between the date of first dose and the date of the initial objectively documented tumor progression per RECIST v1.1 or the date of subsequent therapy, whichever occurs first.~Confidence interval based on the Clopper and Pearson method." (NCT02574078)
Timeframe: up to approximately 48 months

InterventionPercentage of participants (Number)
Group A, Cohort A, Nivo23.1
Group A, Cohort A, Beva + Nivo16.7
Group A, Cohort A, Beva12.5
Group A, Cohort B, Nivo29.4
Group A, Cohort B, Peme + Nivo21.2
Group A, Cohort B, Peme3.1
Group B, Nivo18.8
Group B, BSC5.9
Group C, Nivo20.8
Group C, ICC15.4
Group D, Nivo + Erlo64.7
Group D, Erlo62.5
Group E23.1

[back to top]

Duration of Response (DOR), Groups A-D Only

"Duration of response (DOR) is defined as the time from first confirmed response (complete response (CR) or partial response (PR)) to the date of the initial objectively documented tumor progression as determined using RECIST 1.1 criteria or death due to any cause, whichever occurs first.~Median computed using Kaplan-Meier method." (NCT02574078)
Timeframe: up to approximately 48 months

Interventionmonths (Median)
Group A, Cohort A, Nivo12.780
Group A, Cohort A, Beva + NivoNA
Group A, Cohort A, Beva17.084
Group A, Cohort B, Nivo12.912
Group A, Cohort B, Peme + Nivo8.542
Group A, Cohort B, Peme14.982
Group B, NivoNA
Group B, BSCNA
Group C, Nivo3.877
Group C, ICC2.940
Group D, Nivo + Erlo8.805
Group D, Erlo10.152

[back to top]

Progression-Free Survival (PFS), Group E Only

Progression-free survival (PFS) is defined as the time from randomization to the date of the first documented tumor progression, as determined by investigators (per RECIST v1.1), or death due to any cause, whichever occurs first. (NCT02574078)
Timeframe: up to approximately 48 months

Interventionmonths (Median)
Group E9.63

[back to top]

Progression-Free Survival (PFS), Groups A-D Only

Progression-free survival (PFS) is defined as the time from randomization to the date of the first documented tumor progression, as determined by investigators (per RECIST v1.1), or death due to any cause, whichever occurs first. (NCT02574078)
Timeframe: up to approximately 48 months

InterventionMonths (Median)
Group A, Cohort A, Nivo15.0
Group A, Cohort A, Beva + Nivo6.7
Group A, Cohort A, Beva6.0
Group A, Cohort B, Nivo5.9
Group A, Cohort B, Peme + Nivo8.1
Group A, Cohort B, Peme5.0
Group B, Nivo9.6
Group B, BSC2.3
Group C, Nivo2.7
Group C, ICC6.7
Group D, Nivo + Erlo11.0
Group D, Erlo11.0

[back to top]

Overall Survival (OS) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)

OS is defined as the time from randomization to the date of death, regardless of the actual cause of the participant's death. The participants who were still alive at the time of data analysis or who were lost to follow-up OS time was censored at the last recorded date that the participant was known to be alive before the data cutoff date. OS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Weekly16.8
Chemotherapy13.0

[back to top]

Overall Survival (OS) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Full Analysis Set (FAS)

OS is defined as the time from randomization to the date of death, regardless of the actual cause of the participant's death. The participants who were still alive at the time of data analysis or who were lost to follow-up OS time was censored at the last recorded date that the participant was known to be alive before the data cutoff date. OS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Biweekly18.7
Chemotherapy13.3

[back to top]

Overall Survival (OS) in High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)

OS is defined as the time from randomization to the date of death, regardless of the actual cause of the participant's death. The participants who were still alive at the time of data analysis or who were lost to follow-up OS time was censored at the last recorded date that the participant was known to be alive before the data cutoff date. OS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Weekly19.3
Chemotherapy15.3

[back to top]

Overall Survival (OS) in High Programmed Death Ligand 1 (PD-L1) + Full Analysis Set (FAS)

OS is defined as the time from randomization to the date of death, regardless of the actual cause of the participant's death. The participants who were still alive at the time of data analysis or who were lost to follow-up OS time was censored at the last recorded date that the participant was known to be alive before the data cutoff date. OS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Biweekly20.1
Chemotherapy14.9

[back to top]

Overall Survival (OS) in Full Analysis Set (FAS)

OS is defined as the time from randomization to the date of death, regardless of the actual cause of the participant's death. The participants who were still alive at the time of data analysis or who were lost to follow-up OS time was censored at the last recorded date that the participant was known to be alive before the data cutoff date. OS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Biweekly15.0
Chemotherapy14.3

[back to top]

Duration of Response (DOR) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)

DOR was defined for participants with confirmed response, as the time from first documentation of objective response (Complete Response [CR] or Partial Response [PR]) to the date of first documentation of progression disease (PD) or death due to any cause, whichever occurred first. CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the SLD of all lesions. PD: At least a 20 percent (%) increase in the sum of the longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Weekly19.4
Chemotherapy8.4

[back to top]

Duration of Response (DOR) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1)+ Full Analysis Set (FAS)

DOR was defined for participants with confirmed response, as the time from first documentation of objective response (Complete Response [CR] or Partial Response [PR]) to the date of first documentation of progression disease (PD) or death due to any cause, whichever occurred first. CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the SLD of all lesions. PD: At least a 20 percent (%) increase in the sum of the longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Biweekly35.9
Chemotherapy8.4

[back to top]

Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer 13 (EORTC QLQ-LC13) at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ Modified HRQoL Analysis Set

EORTC QLQ-LC13 consisted of 13 questions relating to disease symptoms specific to lung cancer and treatment side effects typical of treatment with chemotherapy and radiotherapy. The EORTC QLQ-LC13 module generated one multiple-item score assessing dyspnea and a series of single item scores assessing coughing, hemoptysis, sore mouth, dysphagia, peripheral neuropathy, alopecia, pain in chest, pain in arms or shoulder and pain in other parts. Score range: 0 (no burden of symptom domain or single symptom item) to 100 (highest burden of symptoms for symptom domains and single items). (NCT02576574)
Timeframe: Baseline, End of treatment (up to Week 283.9)

,
Interventionscore on a scale (Mean)
DyspneaCoughingHemoptysisSore mouthDysphagiaPeripheral neuropathyAlopeciaPain in chestPain in arm or shoulderPain in other parts
Avelumab Weekly6.1-0.6-0.60.63.80.6-2.52.54.410.1
Chemotherapy4.9-5.20.01.9-0.59.915.0-0.51.41.4

[back to top]

Change From Baseline in European Quality Of Life 5-dimensions (EQ-5D-5L) Visual Analog Scale (VAS) in High Programmed Death Ligand 1 (PD-L1)+ Modified HRQoL Analysis Set

EQ-5D-5L is comprised of the following 5 participant-reported dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension has 5 levels: no problems, slight problems, moderate problems, severe problems, and extreme problems. The responses are used to derive overall score using a visual analog scale (VAS) that ranged from 0 to 100 millimeter (mm), where 0 is the worst health you can imagine and 100 is the best health you can imagine. (NCT02576574)
Timeframe: Baseline, End of treatment (Week 283.9)

Interventionmillimeter (Mean)
Avelumab Weekly-10.3
Chemotherapy-3.9

[back to top]

Number of Participants With At Least One Positive Anti-Drug Antibodies (ADAs) and Neutralizing Antibodies (NAbs) for Avelumab

Serum samples were analyzed by a validated electrochemiluminesce immunoassay to detect the presence of antidrug antibodies (ADA). Samples that screened positive were subsequently tested in a confirmatory assay were tested for neutralizing antibodies (nAb). Number of participants with ADA or nAb positive results for Avelumab were reported. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,
InterventionParticipants (Count of Participants)
ADAs to AvelumabNAbs to Avelumab
Avelumab Biweekly6643
Avelumab Weekly3818

[back to top]

Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Heart Rate Increase/Decrease

The number of participants with maximal on-treatment (TR) changes from baseline (BS) in Increase (Ic.)/Decrease (Dc.) heart rate (HR) (beats per minute [bpm]) were reported by using criteria: Ic./Dc. BS HR <100/>=100 bpm, on treatment change =<20 bpm, >20 - =<40 bpm, >40 bpm and missing; Ic./Dc. BS HR missing, on treatment change missing. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Ic. BS HR <100 bpm, on TR change =<20 bpmIc. BS HR <100 bpm, on TR change >20 - =<40 bpmIc. BS HR <100 bpm, on TR change >40 bpmIc. BS HR <100 bpm, on TR change missingIc. BS HR >= 100 bpm, on TR change =<20 bpmIc. BS HR >= 100 bpm, on TR change >20 - =<40 bpmIc. BS HR >= 100 bpm, on TR change >40 bpmIc. BS HR >= 100 bpm, on TR change missingIc. BS HR missing, on TR change missingDc. BS HR <100 bpm, on TR change =<20 bpmDc. BS HR <100 bpm, on TR change >20 - =<40 bpmDc. BS HR <100 bpm, on TR change >40 bpmDc. BS HR <100 bpm, on TR change missingDc. BS HR >= 100 bpm, on TR change =<20 bpmDc. BS HR >= 100 bpm, on TR change >20 - =<40 bpmDc. BS HR >= 100 bpm, on TR change >40 bpmDc. BS HR >= 100 bpm, on TR change missingDc. BS HR missing, on TR change missing
Avelumab Biweekly206862114310030267442141461130
Avelumab Weekly2026612530201022752151312710
Chemotherapy33285916463072385401162026372

[back to top]

Overall Survival (OS) in Modified Full Analysis Set (mFAS)

OS is defined as the time from randomization to the date of death, regardless of the actual cause of the participant's death. The participants who were still alive at the time of data analysis or who were lost to follow-up OS time was censored at the last recorded date that the participant was known to be alive before the data cutoff date. OS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Weekly15.4
Chemotherapy14.8

[back to top]

Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Body Temperature Increase

The number of participants with changes from baseline in increased Body Temperature (degree Celsius [°C]) were reported by using criteria: Baseline temperature (temp.) less than (<) 37°C, on treatment change <1°C, 1 - <2°C, 2 - <3°C, greater than or equal to (>=)3°C and missing; Baseline temp. 37 - <38°C, on treatment change <1°C, 1 - <2°C, 2 - <3°C, >=3°C and missing; Baseline temp. 38 - <39°C, on treatment change <1°C, 1 - <2°C, 2 - <3°C, >=3°C and missing; Baseline temp. 39-<40°C, on treatment change <1°C, 1 - <2°C, 2 - <3°C, >=3°C and missing; Baseline temp. >=40°C, on treatment change <1°C, 1 - <2°C, 2 - <3°C, >=3°C and missing; Baseline temp. missing, on treatment change missing. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Baseline temp. <37°C, on treatment change <1°CBaseline temp.<37°C, on treatment change 1 - <2°CBaseline temp. <37°C, on treatment change 2 - <3°CBaseline temp. <37°C, on treatment change >=3°CBaseline temp. <37°C, on treatment change missingBaseline temp. 37 - <38°C, on treatment change <1°CBaseline temp. 37 - <38°C, on treatment change 1 - <2°CBaseline temp. 37 - <38°C, on treatment change 2 - <3°CBaseline temp. 37 - <38°C, on treatment change >=3°CBaseline temp. 37 - <38°C, on treatment change missingBaseline temp. 38 - <39°C, on treatment change <1°CBaseline temp. 38 - <39°C, on treatment change 1 - <2°CBaseline temp. 38 - <39°C, on treatment change 2 - <3°CBaseline temp. 38 - <39°C, on treatment change >=3°CBaseline temp. 38 - <39°C, on treatment change missingBaseline temp. 39 - <40°C, on treatment change <1°CBaseline temp. 39 - <40°C, on treatment change 1 - <2°CBaseline temp. 39 - <40°C, on treatment change 2 - <3°CBaseline temp. 39 - <40°C, on treatment change >=3°CBaseline temp. 39 - <40°C, on treatment change missingBaseline temp. >=40°C, on treatment change <1°CBaseline temp. >=40°C, on treatment change 1 - <2°CBaseline temp. >=40°C, on treatment change 2 - <3°CBaseline temp. >=40°C, on treatment change >=3°CBaseline temp. >=40°C, on treatment change missingBaseline temp. missing, on treatment change missing
Avelumab Biweekly2714530162310010000010000000000
Avelumab Weekly256322161920000000000000000000
Chemotherapy4033300213710030000000000000002

[back to top]

Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance: Baseline Score Versus (Vs) Worst Post-baseline Score

ECOG performance status measured to assess participant's performance status on a scale of 0 to 5, where 0 = Fully active, able to carry on all pre-disease activities without restriction; 1 = Restricted in physically strenuous activity, ambulatory and able to carry out light or sedentary work; 2 = Ambulatory and capable of all selfcare but unable to carry out any work activities; 3 = Capable of only limited self-care, confined to bed/chair for more than 50 percent of waking hours; 4 = Completely disabled, cannot carry on any self-care, totally confined to bed/chair; 5 = dead. ECOG performance status was reported in terms of number of participants with baseline value vs worst post-baseline value (that is [i.e.] highest score). (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Baseline score 0, worst post-baseline score 0Baseline score 0, worst post-baseline score 1Baseline score 0, worst post-baseline score 2Baseline score 0, worst post-baseline score 3Baseline score 0, worst post-baseline score 4Baseline score 0, worst post-baseline score 5Baseline score 0, worst post-baseline score MissingBaseline score 1, worst post-baseline score 0Baseline score 1, worst post-baseline score 1Baseline score 1, worst post-baseline score 2Baseline score 1, worst post-baseline score 3Baseline score 1, worst post-baseline score 4Baseline score 1, worst post-baseline score 5Baseline score 1, worst post-baseline score MissingBaseline score >=2, worst post-baseline score 0Baseline score >=2, worst post-baseline score 1Baseline score >=2, worst post-baseline score 2Baseline score >=2, worst post-baseline score 3Baseline score >=2, worst post-baseline score 4Baseline score >=2, worst post-baseline score 5Baseline score >=2, worst post-baseline score missingBaseline score missing, worst post-baseline score 0Baseline score missing, worst post-baseline score 1Baseline score missing, worst post-baseline score 2Baseline score missing, worst post-baseline score 3Baseline score missing, worst post-baseline score 4Baseline score missing, worst post-baseline score 5Baseline score missing, worst post-baseline score missing
Avelumab Biweekly50568110611683516351000010000000000
Avelumab Weekly3549970042153321811600000100000000
Chemotherapy84831140144233466431600000000100000

[back to top]

Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer 13 (EORTC QLQ-LC13) at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ HRQoL Analysis Set

EORTC QLQ-LC13 consisted of 13 questions relating to disease symptoms specific to lung cancer and treatment side effects typical of treatment with chemotherapy and radiotherapy. The EORTC QLQ-LC13 module generated one multiple-item score assessing dyspnea and a series of single item scores assessing coughing, hemoptysis, sore mouth, dysphagia, peripheral neuropathy, alopecia, pain in chest, pain in arms or shoulder and pain in other parts. Score range: 0 (no burden of symptom domain or single symptom item) to 100 (highest burden of symptoms for symptom domains and single items). (NCT02576574)
Timeframe: Baseline, End of treatment (up to Week 283.9)

,
Interventionscore on a scale (Mean)
DyspneaCoughingHemoptysisSore mouthDysphagiaPeripheral neuropathyAlopeciaPain in chestPain in arm or shoulderPain in other parts
Avelumab Biweekly7.32.4-1.83.03.03.60.0-4.20.61.8
Chemotherapy5.2-4.31.51.9-0.610.814.2-1.51.91.5

[back to top]

Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Modified Full Analysis Set (mFAS)

PFS is defined as the time from date of randomization until date of the first documentation of progressive disease (PD) or death due to any cause in the absence of documented PD, whichever occurs first. PD is defined as at least a 20 percent (%) increase in the sum of the longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. PFS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Weekly5.6
Chemotherapy5.6

[back to top]

Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in Moderate and High Programmed Death Ligand 1 (PD-L1)+ Full Analysis Set (FAS)

PFS is defined as the time from date of randomization until date of the first documentation of progressive disease (PD) or death due to any cause in the absence of documented PD, whichever occurs first. PD is defined as at least a 20 percent (%) increase in the sum of the longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. PFS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Biweekly6.9
Chemotherapy5.6

[back to top]

Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1) + Modified Full Analysis Set (mFAS)

PFS is defined as the time from date of randomization until date of the first documentation of progressive disease (PD) or death due to any cause in the absence of documented PD, whichever occurs first. PD is defined as at least a 20 percent (%) increase in the sum of the longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. PFS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Weekly7.5
Chemotherapy5.6

[back to top]

Progression Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) Assessed by Independent Review Committee (IRC) in High Programmed Death Ligand 1 (PD-L1) + Full Analysis Set (FAS)

PFS is defined as the time from date of randomization until date of the first documentation of progressive disease (PD) or death due to any cause in the absence of documented PD, whichever occurs first. PD is defined as at least a 20 percent (%) increase in the sum of the longest diameter (SLD), taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. PFS was measured using Kaplan-Meier (KM) estimates. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionmonths (Median)
Avelumab Biweekly8.4
Chemotherapy5.6

[back to top]

Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in Moderate and High PD-L1+ Modified Full Analysis Set

Confirmed objective response was defined as the percentage of participants with a confirmed objective response of complete response (CR) or partial response (PR). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionpercentage of participants (Number)
Avelumab Weekly30.6
Chemotherapy30.6

[back to top]

Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Systolic Blood Pressure Increase/Decrease and Maximal Diastolic Blood Pressure Increase/Decrease

The number of participants with maximal on-treatment changes from baseline (BS) in Increase (Ic.)/Decrease (Dc.) Systolic Blood Pressure (SBP) and diastolic blood pressure (DBP) (millimeter of mercury [mmHg]) were reported by using criteria: Ic./Dc. BS SBP <140 mmHg and >=140 mmHg, on maximal treatment (TR) change =<20 mmHg, >20 - =<40 mmHg, >40 mmHg and missing; Ic./Dc. BS SBP missing, on maximal treatment (TR) change missing; Ic./Dc. BS DBP <90 mmHg and >= 90 mmHg, on maximal TR change =<20 mmHg, >20 - =<40 mmHg, >40 mmHg and missing; Ic./Dc. BS DBP missing on maximal TR change missing. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Ic. BS SBP <140 mmHg, on TR change =<20 mmHgIc. BS SBP <140 mmHg, on TR change >20 - =<40 mmHgIc. BS SBP <140 mmHg, on TR change >40 mmHgIc. BS SBP <140 mmHg, on TR change missingIc. BS SBP >=140 mmHg, on TR change =<20 mmHgIc. BS SBP >=140 mmHg, on TR change >20 - =<40 mmHgIc. BS SBP >=140 mmHg, on TR change >40 mmHgIc. BS SBP > = 140 mmHg, on TR change missingIc. BS SBP missing, on TR change missingDc. BS SBP <140 mmHg, on TR change =<20 mmHgDc. BS SBP <140 mmHg, on TR change >20 - =<40 mmHgDc. BS SBP <140 mmHg, on TR change >40 mmHgDc. BS SBP <140 mmHg, on TR change missingDc. BS SBP >=140 mmHg, on TR change =<20 mmHgDc. BS SBP >=140 mmHg, on TR change >20 - =<40 mmHgDc. BS SBP >=140 mmHg, on TR change >40 mmHgDc. BS SBP > = 140 mmHg, on TR change missingDc. BS SBP missing, on TR change missingIc. BS DBP <90 mmHg, on TR change =<20 mmHgIc. BS DBP <90 mmHg, on TR change >20 - =<40 mmHgIc. BS DBP <90 mmHg, on TR change >40 mmHgIc. BS DBP <90 mmHg, on TR change missingIc. BS DBP missing, on TR change missingIc. BS DBP >=90 mmHg, on TR change =<20 mmHgIc. BS DBP >=90 mmHg, on TR change >20 - =<40 mmHgIc. BS DBP >=90 mmHg, on TR change >40 mmHgIc. BS DBP >=90 mmHg, on TR change missingDc. BS DBP <90 mmHg, on TR change =<20 mmHgDc. BS DBP <90 mmHg, on TR change >20 - =<40 mmHgDc. BS DBP <90 mmHg, on TR change >40 mmHgDc. BS DBP <90 mmHg, on TR change missingDc. BS DBP >=90 mmHg, on TR change =<20 mmHgDc. BS DBP >=90 mmHg,on TR change >20 - =<40 mmHgDc. BS DBP >=90 mmHg, on TR change >40 mmHgDc. BS DBP >=90 mmHg, on TR change missingDc. BS DBP missing, on TR change missing
Avelumab Biweekly219611614417030220706141125123027248017024000279383171112100
Avelumab Weekly20956164265020204707461510202603715012111264331559010
Chemotherapy2781041021785121322691213234182140245123128000415321231810001

[back to top]

Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Weight Increase/Decrease

The number of participants with maximal on-treatment changes from baseline in Increase (Ic.)/Decrease (Dc.) in maximal weight were reported by using criteria: Ic./Dc. From baseline, on treatment (TR) change <10 percentage (%), >=10% and missing. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Ic. from baseline, on TR change <10%Ic. from baseline, on TR change >=10%Ic. from baseline, on TR change missingDc. from baseline, on TR change <10%Dc. from baseline, on TR change >=10%Dc. from baseline, on TR change missing
Avelumab Biweekly30238212964421
Avelumab Weekly28028102585010
Chemotherapy43639254235225

[back to top]

Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in Moderate and High PD-L1+ Full Analysis Set

Confirmed objective response was defined as the percentage of participants with a confirmed objective response of complete response (CR) or partial response (PR). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionpercentage of participants (Number)
Avelumab Biweekly33.5
Chemotherapy30.3

[back to top] [back to top]

Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life (EORTC QLQ-C30) Global Health Status at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ Modified HRQoL Analysis Set

EORTC QLQ-C30 was a 30-question tool used to assess the overall quality of life (QoL) in cancer participants. It consisted of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, role, cognitive, emotional, social), and 9 symptom scales/items (Fatigue, nausea and vomiting, pain, dyspnea, sleep disturbance, appetite loss, constipation, diarrhea, financial impact). The EORTC QLQ-C30 GHS/QoL score ranged from 0 to 100; High score indicated better GHS/QoL. Score 0 represents: very poor physical condition and QoL. Score 100 represents: excellent overall physical condition and QoL. (NCT02576574)
Timeframe: Baseline, End of treatment (Week 283.9)

Interventionscore on a scale (Mean)
Avelumab Weekly-12.9
Chemotherapy-4.5

[back to top]

Change From Baseline in European Organization for the Research and Treatment of Cancer Quality of Life (EORTC QLQ-C30) Global Health Status at End of Treatment (EOT) in High Programmed Death Ligand 1 (PD-L1)+ HRQoL Analysis Set

EORTC QLQ-C30 was a 30-question tool used to assess the overall quality of life (QoL) in cancer participants. It consisted of 15 domains: 1 global health status (GHS) scale, 5 functional scales (Physical, role, cognitive, emotional, social), and 9 symptom scales/items (Fatigue, nausea and vomiting, pain, dyspnea, sleep disturbance, appetite loss, constipation, diarrhea, financial impact). The EORTC QLQ-C30 GHS/QoL score ranged from 0 to 100; High score indicated better GHS/QoL. Score 0 represents: very poor physical condition and QoL. Score 100 represents: excellent overall physical condition and QoL. (NCT02576574)
Timeframe: Baseline, End of treatment (up to Week 283.9)

Interventionscore on a scale (Mean)
Avelumab Biweekly-0.3
Chemotherapy-6.1

[back to top]

Number of Participants With Potentially Clinically Significant Abnormalities (PCSA) in Electrocardiogram (ECG) Parameters

ECG parameters included heart rate, PR interval, QRS interval, corrected QT interval using Bazett's formula (QTcB) and corrected QT interval using Fridericia's formula (QTcF). PCSA criteria for abnormal value of ECG parameters: any heart rate <= 50 bpm and decrease from baseline >=20 bpm , any hear rate >= 120 bpm and increase from baseline >= 20 bpm; PR interval: >= 220 milliseconds (ms) and increase from baseline >= 20 ms; QRS interval >= 120 ms; QTcF > 450 ms, > 480 ms, > 500 ms, QTcF increase from baseline > 30 ms and QTcF increase from baseline > 60 ms; QTcB > 450 ms, > 480 ms, > 500 ms, QTcB increase from baseline > 30 ms and QTcB increase from baseline > 60 ms. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Heart Rate <= 50 bpm and decrease from baseline >= 20 bpmHeart Rate >= 120 bpm and decrease from baseline >= 20 bpmPR interval >= 220 ms and increase from baseline >= 20 msQRS interval >= 120 msQTcF > 450 msQTcF > 480 msQTcF > 500 msQTcF increase from baseline > 30 msQTcF increase from baseline > 60 msQTcB > 450 msQTcB > 480 msQTcB > 500 msQTcB increase from baseline > 30 msQTcB increase from baseline > 60 ms
Avelumab Biweekly1100181953206491363211
Avelumab Weekly16510134112131115257
Chemotherapy073152411539137024164919

[back to top]

Number of Participants With Shift From Baseline to Greater Than or Equal to (>=) Grade 3 in Laboratory Parameter Values Based on National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE), Version 4.03

Number of participants with shifts from Baseline values (Grade 0/1/2/3) to abnormal post-baseline values (shift to >= Grade 4) were reported as per NCI-CTCAE, v4.03 graded from Grade 1 to 5. Grade 1: Mild; Grade 2: Moderate; Grade 3: Severe; Grade 4: Life-threatening; Grade 5: Death. Shifts in laboratory parameter (anemia, lymphocyte count decreased, neutrophil count decreased, platelet count decreased, white blood cell count decreased, alanine aminotransferase increased, alkaline phosphatase increased, aspartate aminotransferase increased, blood bilirubin increased, creatine phosphokinase increased, creatinine increased and Hyperglycemia) were reported. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
Anemia: Grade 0 to Grade 3Anemia: Grade 1 to Grade 3Anemia: Grade 2 to Grade 3Lymphocyte count decreased: Grade 0 to Grade 3Lymphocyte count decreased: Grade 0 to Grade 4Lymphocyte count decreased: Grade 1 to Grade 3Lymphocyte count decreased: Grade 2 to Grade 3Lymphocyte count decreased: Grade 2 to Grade 4Lymphocyte count decreased: Grade 3 to Grade 4Neutrophil count decreased: Grade 0 to Grade 3Neutrophil count decreased: Grade 0 to Grade 4Neutrophil count decreased: Grade 1 to Grade 3Platelet count decreased: Grade 0 to Grade 3Platelet count decreased: Grade 0 to Grade 4Platelet count decreased: Grade 1 to Grade 3White blood cell count decreased: Grade 0 to Grade 3White blood cell count decreased: Grade 0 to Grade 4Alanine aminotransferase increased: Grade 0 to Grade 3Alanine aminotransferase increased: Grade 0 to Grade 4Alanine aminotransferase increased: Grade 1 to Grade 3Alkaline phosphatase increased: Grade 0 to Grade 3Alkaline phosphatase increased: Grade 1 to Grade 3Alkaline phosphatase increased: Grade 1 to Grade 4Alkaline phosphatase increased: Grade 2 to Grade 3Aspartate aminotransferase increased: Grade 0 to Grade 3Aspartate aminotransferase increased: Grade 0 to Grade 4Aspartate aminotransferase increased: Grade 1 to Grade 3Blood bilirubin increased: Grade 0 to Grade 3Blood bilirubin increased: Grade 0 to Grade 4Creatine phosphokinase increased: Grade 0 to Grade 3Creatine phosphokinase increased: Grade 0 to Grade 4Creatine phosphokinase increased: Grade 1 to Grade 4Creatine phosphokinase increased: Grade 2 to Grade 3Creatinine increased: Grade 0 to Grade 3Creatinine increased: Grade 1 to Grade 3Hyperglycemia: Grade 0 to Grade 3Hyperglycemia: Grade 0 to Grade 4
Avelumab Biweekly4441612700401000001211010071000650060210
Avelumab Weekly2561521060043013131810330142041501140200
Chemotherapy5830631312121165251182002411532111032130100041353

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and AEs of Special Interest (AESIs)

Adverse event (AE) was defined as any untoward medical occurrence in a participant, which does not necessarily have causal relationship with treatment. A serious AE was defined as an AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial or prolonged in participant hospitalization; congenital anomaly/birth defect or was otherwise considered medically important. The term TEAEs were those events with onset dates occurring during the on-treatment period or if the worsening of an event is during the on-treatment period TEAEs included both serious TEAEs and non-serious TEAEs. Any AE that was suspicious to be a potential Immune-related adverse event (irAE) including infusion related reactions were considered AESIs. Number of participants with TEAEs and AESIs were reported. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

,,
InterventionParticipants (Count of Participants)
TEAEsAESIs
Avelumab Biweekly346158
Avelumab Weekly308160
Chemotherapy484173

[back to top]

Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High PD-L1+ Modified Full Analysis Set

Confirmed objective response was defined as the percentage of participants with a confirmed objective response of complete response (CR) or partial response (PR). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionpercentage of participants (Median)
Avelumab Weekly34.6
Chemotherapy30.2

[back to top]

Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Independent Review Committee (IRC) in High PD-L1+ Full Analysis Set

Confirmed objective response was defined as the percentage of participants with a confirmed objective response of complete response (CR) or partial response (PR). CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to approximately 71.5 months)

Interventionpercentage of participants (Number)
Avelumab Biweekly37.7
Chemotherapy30.1

[back to top]

Number of Participants With Maximal On-Treatment Changes From Baseline in Vital Signs - Maximal Respiration Rate Increase/Decrease

The number of participants with maximal on-treatment (TR) changes from baseline (BS) in Increase (Ic.)/Decrease (Dc.) maximal Respiration Rate (RR) were reported by using criteria: Ic./Dc. BS RR <20 breaths per minute (breaths/min), on TR change =<5 breaths/min, >5 - =<10 breaths/min, >10 breaths/min and missing. Ic./Dc. BS RR missing, on TR change missing. Ic./Dc. BS RR >=20 breaths/min, on TR change =<5 breaths/min, >5 - =<10 breaths/min, >10 breaths/min and missing. (NCT02576574)
Timeframe: Time from date of randomization up to data cutoff (assessed up to 71.5 months)

,,
InterventionParticipants (Count of Participants)
Ic. BS RR <20 breaths/min, on TR change =<5 breaths/minIc.BS RR<20 breaths/min, on TR change >5 - = <10 breaths/minIc. BS RR <20 breaths/min, on TR change >10 breaths/minIc. BS RR <20 breaths/min, on TR change missingIc. BS RR >=20 breaths/min, on TR change =<5 breaths/minIc.BS RR >=20 breaths/min, on TR change >5 - =<10 breaths/minIc. BS RR >=20 breaths/min, on TR change >10 breaths/minIc. BS RR >=20 breaths/min, on TR change missingIc. BS RR missing, on TR change missingDc. BS RR <20 breaths/min, on TR change =<5 breaths/minDc. BS RR <20 breaths/min, on TR change >5 - =<10 breaths/minDc. BS RR <20 breaths/min, on TR change >10 breaths/minDc. BS RR <20 breaths/min, on TR change missingDc. BS RR >=20 breaths/min, on TR ch =<5 breaths/minDc.BS RR >=20 breaths/min, on TR change >5 - =<10 breaths/minDc. BS RR >=20 breaths/min, on TR change >10 breaths/minDc. BS RR >=20 breaths/min, on TR change missingDc. BS RR missing, on TR change missing
Avelumab Biweekly2211811189537623280117914476
Avelumab Weekly22413146841212362045813221
Chemotherapy3062621412420151132581141012231511

[back to top]

Number of Participants Who Experienced an Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a study participant administered study drug and which does not necessarily have to have a causal relationship with this study drug. For participants who switched from the Control group to receiving pembro, AEs that occurred after the first dose of pembro are excluded from this interim analysis, but will be included in the final analysis. The number of participants who experienced an AE is presented. (NCT02578680)
Timeframe: Through Database Cutoff Date of 08-Nov-2017 (Up to approximately 21 months); Serious AEs: Up to 90 days after last dose of study treatment, Other AEs: Up to 30 days after last dose of study treatment

InterventionParticipants (Count of Participants)
Pembrolizumab404
Control200

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 as Assessed by Blinded Central Imaging

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 RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. Note: The appearance of one or more new lesions was also considered PD. The PFS per RECIST 1.1 is presented. (NCT02578680)
Timeframe: Through Database Cutoff Date of 08-Nov-2017 (Up to approximately 21 months)

InterventionMonths (Median)
Pembrolizumab8.8
Control4.9

[back to top]

Number of Participants Who Discontinued Any Study Drug Due to an AE

The number of participants who discontinued any randomized study drug due to an AE is presented. (NCT02578680)
Timeframe: Through Database Cutoff Date of 08-Nov-2017 (Up to approximately 21 months)

InterventionParticipants (Count of Participants)
Pembrolizumab112
Control30

[back to top]

Duration of Response (DOR) Per RECIST 1.1 as Assessed by Blinded Central Imaging

For participants who demonstrated a confirmed CR (disappearance of all target lesions) or 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 a CR or PR until PD 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. Per RECIST 1.1, PD was defined as ≥20% increase in the sum of diameters of target lesions. In addition to the relative increase of 20%, the sum must also have demonstrated an absolute increase of ≥5 mm. Note: The appearance of one or more new lesions was also considered PD. DOR assessments were based on blinded central imaging review with confirmation. The DOR per RECIST 1.1 for all participants who experienced a confirmed CR or PR is presented. (NCT02578680)
Timeframe: From time of first documented evidence of CR or PR through database cutoff date of 08-Nov-2017 (Up to approximately 21 months)

InterventionMonths (Median)
Pembrolizumab11.2
Control7.8

[back to top]

Overall Response Rate (ORR) Per RECIST 1.1 as Assessed by Blinded Central Imaging

ORR was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The percentage of participants who experienced a CR or PR is presented. (NCT02578680)
Timeframe: Through Database Cutoff Date of 08-Nov-2017 (Up to approximately 21 months)

InterventionPercentage of Participants (Number)
Pembrolizumab47.6
Control18.9

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to death due to any cause. Participants without documented death at the time of the interim analysis were censored at the date of the last follow-up. The OS is presented. (NCT02578680)
Timeframe: Through Database Cutoff Date of 08-Nov-2017 (Up to approximately 21 months)

InterventionMonths (Median)
PembrolizumabNA
Control11.3

[back to top]

Overall Response Rate (ORR) Per RECIST 1.1

The primary objective of this randomized phase II trial to determine the overall response rate (ORR per RECIST 1.1) in Chemotherapy naive patients with stage IV NSCLC after the administration of standard platinum-based chemotherapy before MK-3475 (arm A) and administration of MK-3475 administered before standard platinum-based chemotherapy (arm B). Overall Response (OR) = CR + PR. (NCT02591615)
Timeframe: 18 Months

Interventionproportion of participants (Number)
Arm A (Both Squamous and Non-Squamous)0.395
Arm B (Both Squamous and Non-Squamous)0.404

[back to top]

Incidence of Treatment-Emergent Adverse Events (Safety and Tolerability)

To characterize the adverse events related to MK-3475 by frequency, type and grade in patients with Chemotherapy naive advanced NSCLC based on the sequence of administration with first-line chemotherapy. A count of participants experiencing an adverse event is summarized here, the detailed summary is in the adverse events section of this report. (NCT02591615)
Timeframe: 24 Months

Interventionparticipants (Number)
Arm A Squamous Carcinoma9
Arm A Non-squamous Carcinoma34
Arm B Squamous Carcinoma10
Arm B Non-squamous Carcinoma37

[back to top]

Compare Progression-Free Survival (PFS) Per RECIST 1.1

To compare the progression-free survival (PFS) per RECIST 1.1 in previously untreated patients with advanced NSCLC treated with first line carboplatin-based chemotherapy followed by MK-3475 to patients treated with MK-3475 prior to first-line carboplatin-based chemotherapy. (NCT02591615)
Timeframe: 24 Months

InterventionMonths (Median)
Arm A (Both Squamous and Non-Squamous)5.8
Arm B (Both Squamous and Non-Squamous)4.0

[back to top]

Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC30 Score for ITT Population

TTD in the overall population is defined as time from randomization to the earliest time with a ≥10-point increase from baseline for symptoms domains (or decrease for functioning domains from baseline for cough, dyspnea [single item and multi-item scales] chest pain [single item], pain in arm/shoulder and fatigue as measured by the EORTC QLQ-C30. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

,
Interventionmonths (Median)
Dyspnoea scoreFatigue score
Active Comparator: Premetrexed/Docetaxel8.31.2
Experimental: Alectinib13.35.6

[back to top]

Plasma Concentration of Alectinib

(NCT02604342)
Timeframe: Predose (2 hours) at Baseline, Week 3 and Week 6

Interventionnanogram/milliliter (ng/mL) (Geometric Mean)
Experimental: Alectinib559

[back to top]

Duration of Response for Lesions in the CNS (C-DOR) Using RECIST Version 1.1 as Assessed by IRC

DOR was defined as the time from when response (CR or PR) was first documented to first documented disease progression or death, whichever occurred first. C-DOR was defined in a similar way for lesions in the CNS, taking into account all lesions in the body. DOR was evaluated for participants who had a BOR of CR or PR. (NCT02604342)
Timeframe: From the first documented CR or PR to the first documented disease progression, death, or study end (up to 33 months)

Interventionmonths (Median)
Experimental: Alectinib13.9
Active Comparator: Premetrexed/DocetaxelNA

[back to top]

Overall Survival (OS)

Overall survival (OS) was defined as the time from randomization to death from any cause. OS was confounded by cross-over of participants to the alectinib arm. (NCT02604342)
Timeframe: Randomization to death from any cause, through study end (up to 33 months)

Interventionmonths (Median)
Experimental: Alectinib27.8
Active Comparator: Premetrexed/DocetaxelNA

[back to top]

Percentage of Participants With Adverse Events (AEs)

An adverse event (AE) is any untoward medical occurrence in a participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

InterventionPercentage of Participants (Number)
Experimental: Alectinib89.6
Active Comparator: Premetrexed/Docetaxel89.2

[back to top]

Percentage of Participants With CNS Objective Response Rate (ORR) With Measurable CNS Metastases at Baseline Using RECIST Version 1.1 as Assessed By IRC

Overall response rate in subjects with confirmed CNS response (C-ORR) was defined as the percentage of subjects who attained Complete Response (CR) or Partial Response (PR) for lesions in the CNS. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

Interventionpercentage of participants (Number)
Experimental: Alectinib66.7
Active Comparator: Premetrexed/Docetaxel0.0

[back to top]

Percentage of Participants With Disease Control in C-ITT Population Using RECIST Version 1.1 as Assessed by IRC

Disease Control Rate (DCR) was defined as the percentage of participants who attained CR, PR, or stable disease (SD) of at least 5 weeks. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters, SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum of diameters since the treatment started. (NCT02604342)
Timeframe: From first documented CR, PR, or SD lasting at least 5 weeks through study end (up to 33 months)

Interventionpercentage of participants (Number)
Experimental: Alectinib82.7
Active Comparator: Premetrexed/Docetaxel25.0

[back to top]

Percentage of Participants With ORR in C-ITT Population Using RECIST Version 1.1 as Assessed by IRC

ORR was defined as the percentage of participants who attained CR or PR. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

Interventionpercentage of participants (Number)
Experimental: Alectinib48.1
Active Comparator: Premetrexed/Docetaxel0.0

[back to top]

PFS Using RECIST Version 1.1 as Assessed by IRC

"PFS was defined as the time from randomization to the first documented disease progression, as determined using RECIST v1.1, or death from any cause, whichever occurred first. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 mm and the appearance of new lesions.~This outcome measure was assessed as part of the primary analysis and was not repeated during final analysis." (NCT02604342)
Timeframe: Approximately 15 months (Tumor assessments at baseline, every 6 weeks until progressive disease (PD), death or withdrawal from study prior to PD)

Interventionmonths (Median)
Experimental: Alectinib7.1
Active Comparator: Premetrexed/Docetaxel1.6

[back to top]

Plasma Concentration of Alectinib Metabolite

(NCT02604342)
Timeframe: Predose (2 hours) at Baseline, Week 3 and Week 6

Interventionng/mL (Geometric Mean)
Experimental: Alectinib240

[back to top]

Progression-Free Survival (PFS) Using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 as Assessed by Investigator

PFS was defined as the time from randomization to the first documented disease progression, as determined using RECIST v1.1, or death from any cause, whichever occurred first. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 millimeter (mm) and the appearance of new lesions. (NCT02604342)
Timeframe: Randomization to first documented disease progression, death from any cause, or study end (up to 33 months)

Interventionmonths (Median)
Experimental: Alectinib10.9
Active Comparator: Premetrexed/Docetaxel1.4

[back to top]

Time to CNS Progression in C-ITT Population Using RECIST Version 1.1 as Assessed by IRC

"Time to CNS progression was defined as the time from randomization until radiographic evidence of CNS progression. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 mm and the appearance of new lesions.~This outcome measure assessment was part of the primary analysis and was not repeated during final analysis." (NCT02604342)
Timeframe: Approximately 15 months (Tumor assessments at baseline, every 6 weeks until progressive disease (PD), death or withdrawal from study prior to PD)

Interventionmonths (Median)
Experimental: AlectinibNA
Active Comparator: Premetrexed/Docetaxel1.6

[back to top]

TTD in Composite of Three Symptoms (Cough, Dyspnea, and Chest Pain) Using EORTC QLQ-LC13 Score for C-ITT Population

TTD for a composite of three symptoms (cough, dyspnea, chest pain) in the overall population is defined as time from randomization to the earliest time with a ≥10-point increase from baseline for any component of the composite of the three following symptoms [cough, dyspnea [multi-item subscales QLQ-LC13] and chest pain]) as measured by the EORTC QLQ-LC13. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

Interventionmonths (Median)
Experimental: Alectinib2.8
Active Comparator: Premetrexed/Docetaxel1.4

[back to top]

TTD in Composite of Three Symptoms (Cough, Dyspnea, and Chest Pain) Using EORTC QLQ-LC13 Score for ITT Population

TTD for a composite of three symptoms (cough, dyspnea, chest pain) in the overall population is defined as time from randomization to the earliest time with a ≥10-point increase from baseline for any component of the composite of the three following symptoms [cough, dyspnea [multi-item subscales QLQ-LC13] and chest pain]) as measured by the EORTC QLQ-LC13. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

Interventionmonths (Median)
Experimental: Alectinib1.4
Active Comparator: Premetrexed/Docetaxel1.4

[back to top]

Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (EORTC QLQ-C30) Over Time

Percentage of participants who filled out an EORTC QLQ-C30 questionnaire at a visit. The EORTC QLQ-C30 questionnaire consisted of 30 questions generating five functional scores (physical, role, cognitive, emotional, and social); a global health status/global quality of life scale score; three symptom scale scores (fatigue, pain, and nausea and vomiting); and six stand alone one-item scores that capture additional symptoms (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea) and perceived financial burden. (NCT02604342)
Timeframe: Baseline through Week 138

Interventionpercentage of participants (Number)
BaselineTreatment - Week 3Treatment - Week 6Treatment - Week 12Treatment - Week 18Treatment - Week 24Treatment - Week 30Treatment - Week 36Treatment - Week 42Treatment - Week 48Treatment - Week 54Treatment - Week 60Treatment - Week 66Treatment - Week 72
Active Comparator: Premetrexed/Docetaxel85.083.360.080.050.010066.766.766.7100100100100100

[back to top]

Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core-30 (EORTC QLQ-C30) Over Time

Percentage of participants who filled out an EORTC QLQ-C30 questionnaire at a visit. The EORTC QLQ-C30 questionnaire consisted of 30 questions generating five functional scores (physical, role, cognitive, emotional, and social); a global health status/global quality of life scale score; three symptom scale scores (fatigue, pain, and nausea and vomiting); and six stand alone one-item scores that capture additional symptoms (dyspnea, appetite loss, sleep disturbance, constipation, and diarrhea) and perceived financial burden. (NCT02604342)
Timeframe: Baseline through Week 138

Interventionpercentage of participants (Number)
BaselineTreatment - Week 3Treatment - Week 6Treatment - Week 12Treatment - Week 18Treatment - Week 24Treatment - Week 30Treatment - Week 36Treatment - Week 42Treatment - Week 48Treatment - Week 54Treatment - Week 60Treatment - Week 66Treatment - Week 72Treatment - Week 78Treatment - Week 84Treatment - Week 90Treatment - Week 96Treatment - Week 102Treatment - Week 108Treatment - Week 114Treatment - Week 120Treatment - Week 126Treatment - Week 132Treatment - Week 138
Experimental: Alectinib92.496.197.295.588.591.196.289.895.310097.110089.788.984.678.388.993.884.610083.3100100100100

[back to top]

Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer-13 (EORTC QLQ-LC13) Over Time

Percentage of participants who filled out an EORTC QLQ-LC13 questionnaire at a visit. The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. (NCT02604342)
Timeframe: Baseline through Week 138

Interventionpercentage of participants (Number)
BaselineTreatment - Week 3Treatment - Week 6Treatment - Week 12Treatment - Week 18Treatment - Week 24Treatment - Week 30Treatment - Week 36Treatment - Week 42Treatment - Week 48Treatment - Week 54Treatment - Week 60Treatment - Week 66Treatment - Week 72
Active Comparator: Premetrexed/Docetaxel82.583.363.380.050.010066.766.766.7100100100100100

[back to top]

Compliance of European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Lung Cancer-13 (EORTC QLQ-LC13) Over Time

Percentage of participants who filled out an EORTC QLQ-LC13 questionnaire at a visit. The EORTC QLQ-LC13 module generated one multiple-item scale score assessing dyspnea and a series of single item scores assessing chest pain, arm/shoulder pain, pain in other parts, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. (NCT02604342)
Timeframe: Baseline through Week 138

Interventionpercentage of participants (Number)
BaselineTreatment - Week 3Treatment - Week 6Treatment - Week 12Treatment - Week 18Treatment - Week 24Treatment - Week 30Treatment - Week 36Treatment - Week 42Treatment - Week 48Treatment - Week 54Treatment - Week 60Treatment - Week 66Treatment - Week 72Treatment - Week 78Treatment - Week 84Treatment - Week 90Treatment - Week 96Treatment - Week 102Treatment - Week 108Treatment - Week 114Treatment - Week 120Treatment - Week 126Treatment - Week 132Treatment - Week 138
Experimental: Alectinib92.496.197.295.588.591.196.287.895.310097.110089.788.984.678.388.910084.610083.380.0100100100

[back to top]

Compliance of European Quality of Life (EuroQoL) 5 Dimension 5 Levels (EQ-5D-5L) Questionnaire Over Time

Percentage of participants who filled out an ED-5D-5L questionnaire at a visit. EQ-5D-5L: A generic preference-based health utility measure that provides a single index value for health status. The instrument consists of two parts. The first part, health-state classification, contains five dimensions of health: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. (NCT02604342)
Timeframe: Baseline through Week 60

Interventionpercentage of participants (Number)
Treatment - Week 0Treatment - Week 3Treatment - Week 6Treatment - Week 12Treatment - Week 18Treatment - Week 24Treatment - Week 30Treatment - Week 36Treatment - Week 42Treatment - Week 48
Active Comparator: Premetrexed/Docetaxel82.978.858.68066.710066.750500

[back to top]

Compliance of European Quality of Life (EuroQoL) 5 Dimension 5 Levels (EQ-5D-5L) Questionnaire Over Time

Percentage of participants who filled out an ED-5D-5L questionnaire at a visit. EQ-5D-5L: A generic preference-based health utility measure that provides a single index value for health status. The instrument consists of two parts. The first part, health-state classification, contains five dimensions of health: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. (NCT02604342)
Timeframe: Baseline through Week 60

Interventionpercentage of participants (Number)
Treatment - Week 0Treatment - Week 3Treatment - Week 6Treatment - Week 12Treatment - Week 18Treatment - Week 24Treatment - Week 30Treatment - Week 36Treatment - Week 42Treatment - Week 48Treatment - Week 54Treatment - Week 60
Experimental: Alectinib88.986.691.986.872.182.4808091.762.510050

[back to top]

Duration of Response (DOR) Using RECIST Version 1.1 as Assessed by Investigator and IRC

"DOR was defined as the time from when response (CR or PR) was first documented to first documented disease progression or death, whichever occurred first. DOR was evaluated for participants who had a best overall response (BOR) of CR or PR.~The IRC assessment was part of the primary analysis and was not repeated during final analysis." (NCT02604342)
Timeframe: From the first documented CR or PR to the first documented disease progression, death, or study end (up to 33 months)

,
Interventionmonths (Median)
Assessed by InvestigatorAssessed by IRC
Active Comparator: Premetrexed/Docetaxel2.7NA
Experimental: Alectinib12.09.7

[back to top]

Percentage of Participants With Disease Control Using RECIST Version 1.1 as Assessed by Investigator and IRC

"Disease control rate (DCR) was defined as the percentage of participants who attained CR, PR, or stable disease (SD) of at least 5 weeks. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters, SD: Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum of diameters since the treatment started.~The IRC assessment was part of the primary analysis and was not repeated during final analysis." (NCT02604342)
Timeframe: Approximately 15 months (Tumor assessments at baseline, every 6 weeks until progressive disease (PD), death or withdrawal from study prior to PD)

,
Interventionpercentage of participants (Number)
Assessed by InvestigatorAssessed by IRC
Active Comparator: Premetrexed/Docetaxel25.048.6
Experimental: Alectinib86.176.4

[back to top]

Percentage of Participants With Objective Response of CR or PR Using RECIST Version 1.1 as Assessed by Investigator and IRC

"ORR was defined as the percentage of participants who attained CR or PR. As per RECIST v1.1, CR: Disappearance of all target lesions and any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm, PR: At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters.~The IRC assessment was part of the primary analysis and was not repeated during final analysis." (NCT02604342)
Timeframe: Approximately 15 months (Tumor assessments at baseline, every 6 weeks until progressive disease (PD), death or withdrawal from study prior to PD)

,
Interventionpercentage of participants (Number)
Assessed by InvestigatorAssessed by IRC
Active Comparator: Premetrexed/Docetaxel2.511.4
Experimental: Alectinib50.636.1

[back to top]

PFS in C-ITT Population Using RECIST Version 1.1 as Assessed by Investigator and IRC

"PFS was defined as the time from randomization to the first documented disease progression, as determined using RECIST v1.1, or death from any cause, whichever occurred first. As per RECIST v1.1, disease progression is a 20% increase in the sum of the diameters of target lesions, an increase in size of measurable lesions by at least 5 mm and the appearance of new lesions.~This outcome measure assessment was part of the primary analysis and was not repeated during final analysis." (NCT02604342)
Timeframe: Approximately 15 months (Tumor assessments at baseline, every 6 weeks until progressive disease (PD), death or withdrawal from study prior to PD)

,
Interventionmonths (Median)
Assessed by InvestigatorAssessed by IRC
Active Comparator: Premetrexed/Docetaxel1.41.5
Experimental: Alectinib9.78.1

[back to top]

Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC13 Score for C-ITT Population

TTD in the overall population is defined as time from randomization to the earliest time with a ≥10-point increase from baseline for symptoms domains (or decrease for functioning domains from baseline for cough, dyspnea [single item and multi-item scales] chest pain [single item], pain in arm/shoulder and fatigue as measured by the EORTC QLQ-LC13. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

,
Interventionmonths (Median)
Coughing scoreDyspnoea scorePain in chest scorePain in arm or shoulder score
Active Comparator: Premetrexed/Docetaxel16.61.4NA1.7
Experimental: AlectinibNA9.7NA11.1

[back to top]

Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC13 Score for ITT Population

TTD in the overall population is defined as time from randomization to the earliest time with a ≥10-point increase from baseline for symptoms domains (or decrease for functioning domains from baseline for cough, dyspnea [single item and multi-item scales] chest pain [single item], pain in arm/shoulder and fatigue as measured by the EORTC QLQ-LC13. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

,
Interventionmonths (Median)
Coughing scoreDyspnoea scorePain in chest scorePain in arm or shoulder score
Active Comparator: Premetrexed/Docetaxel16.63.3NA1.9
Experimental: Alectinib18.14.1NA12.5

[back to top]

Time to Deterioration (TTD) in Lung Cancer Symptoms Using EORTC QLQ-LC30 Score for C-ITT Population

TTD in the overall population is defined as time from randomization to the earliest time with a ≥10-point increase from baseline for symptoms domains (or decrease for functioning domains from baseline for cough, dyspnea [single item and multi-item scales] chest pain [single item], pain in arm/shoulder and fatigue as measured by the EORTC QLQ-C30. (NCT02604342)
Timeframe: Baseline through study end (up to 33 months)

,
Interventionmonths (Median)
Dyspnoea scoreFatigue score
Active Comparator: Premetrexed/Docetaxel8.31.0
Experimental: Alectinib16.614.4

[back to top]

The 2-year Survival Rate After Initial Study Treatment.

The proportion of patients who are alive at 2 years following initiation of study treatment. (NCT02624700)
Timeframe: 2 years 3 months

InterventionParticipants (Count of Participants)
A: Pemetrexed + Sorafenib1
B: Pemetrexed + Sorafenib0

[back to top]

Number of Participants at Risk and Affected by Adverse Events (AEs)

To further characterize the safety and side effect profile of the combination. All participants' AEs will be listed and summary descriptive statistics will be calculated.The Adverse events (AEs) are reported using criteria in the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (CTCAE v4.0). (NCT02624700)
Timeframe: 2 years 3 months

,
InterventionParticipants (Count of Participants)
At RiskAffected
A: Pemetrexed + Sorafenib99
B: Pemetrexed + Sorafenib44

[back to top]

The Duration of Progression-free Survival (PFS).

Progression-free survival (PFS) defined as the time (in days) from initiation of study treatment until documented disease progression or death, whichever occurs first. (NCT02624700)
Timeframe: 2 years 3 months

,
InterventionDays (Number)
Participant AParticipant BParticipant CParticipant DParticipant EParticipant FParticipant GParticipant HParticipant IParticipant JParticipant KParticipant LParticipant M
A: Pemetrexed + Sorafenib141602812025959644956NANANANA
B: Pemetrexed + SorafenibNANANANANANANANANA2210016019

[back to top]

Changes Between Initial and Post Treatment Dimensions of Periapical Lesions

Changes in the dimensions of periapical lesions will be performed according to the analysis of initial and post treatment radiographs (baseline, 3, 6, 12, and 24-months subsequent to obturation) after being photographed using a digital camera Kodak EasyShare Max (Z990) with millimetre measurer in order to obtain interpretation of sizes of periapical lesions during conversion of pixels in mm2 by digital data processing in Adobe Photoshop CS software. Sucessful radiographic assessment will include decrease in size of the periapical lesion at the recall time of 24 months. (NCT02625298)
Timeframe: baseline, 3, 6, 12 and 24 months

,
Interventionsquare millimeters (Mean)
InitialBaseline3 months6 months12 months24 months
MTA+ Cercamed34.0417.5412.017.864.852.48
ProRoot MTA37.3416.2012.7411.598.382.99

[back to top]

Presence of Clinical Symptoms

Clinical examination will be used to assess the presence of spontaneous or provoked pain, discomfort during chewing, numbness or tenderness to percussion and/or palpation, altered tooth mobility, tooth crown discoloration or abscess and/or sinus tract. (NCT02625298)
Timeframe: baseline

,
Interventionparticipants (Number)
spontaneous or provoked paindiscomfort during chewingnumbnesstenderness to percussion and/or palpationaltered tooth mobilitytooth crown discolorationabscess and/or sinus tract
MTA+ Cercamed5612110
ProRoot MTA5832211

[back to top]

Percentage of Participants With Anti-Therapeutic Antibodies (ATAs) of Atezolizumab

Baseline prevalence and post-baseline incidence of anti-drug antibodies (ADA) to Atezolizumab in the Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed) (NCT02657434)
Timeframe: Prd (0 h) on D1 of Cy1,2,3,4,8,16 (Cy length=21 days) and thereafter on D1 of every 8th cycle, at treatment discontinuation & then every 30 days (up to 120 days) after last dose of atezolizumab (up to app 25 months)

InterventionPercentage of Participants (Number)
Baseline Evaluable ParticipantsPost-Baseline Evaluable Participants
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed1.835.4

[back to top]

Percentage of Participants With an Objective Response (Complete Response [CR] or Partial Response [PR]) Assessed by the Investigator Using RECIST V1.1

An objective response is defined as either an unconfirmed CR or a PR, as determined by the investigator using RECIST v1.1. Objective Response Rate is defined as the proportion of patients who had an objective response. (NCT02657434)
Timeframe: Randomization up to approximately 25 months

,
InterventionPercentage of Participants (Number)
RespondersNon-Responders
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed51.748.3
Arm B (Carboplatin or Cisplatin + Pemetrexed)37.462.6

[back to top]

Minimum Observed Serum Atezolizumab Concentration (Cmin)

Minimum observed serum atezolizumab concentration (Cmin) prior to infusion at selected cycles (Arm A) (NCT02657434)
Timeframe: Predose (Prd; 0 hour [h]) on D1 of Cy 2,3,4,8,16 (Cy length=21 days) and thereafter on D1 of every 8th cycle (up to approximately 25 months)

Interventionμg/mL (Mean)
Cy2D1Cy3D1Cy4D1Cy8D1Cy16D1Cy24D1Treatment Discontinuation VisitDay 120 Post Last Dose
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed69.811515122123425712913.4

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by EORTC Quality-of-Life Lung Cancer Module (QLQ-LC13) Symptom Score

The EORTC QLQ-LC13 module incorporates one multiple item scale to assess dyspnea and a series of single items assessing pain, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. The EORTC QLQ-LC13 is scored according to the EORTC scoring manual (Fayers et al. 2001). All EORTC scales and single-item measures are linearly transformed so that each score has a range of 0-100. A high score for a functional/global health status scale represents a high or healthy level of functioning/HRQoL (Health-Related Quality of Life); however, a high score for a symptom scale or item represents a high level of symptomatology or problems. A≥10-point change in the symptoms subscale score is perceived by patients as clinically significant (Osoba et al. 1998). (NCT02657434)
Timeframe: Baseline up to 3 and 6 months after disease progression or loss of clinical benefit (up to approximately 25 months)

InterventionUnits on a scale (Mean)
Coughing: Week 3Coughing: Week 6Coughing: Week 9Coughing: Week 12Coughing: Week 15Coughing: Week 18Coughing: Week 21Coughing: Week 24Coughing: Week 27Coughing: Week 30Coughing: Week 33Coughing: Week 36Coughing: Week 39Coughing: Week 42Coughing: Week 45Coughing: Week 48Coughing: Week 51Coughing: Week 54Coughing: Week 57Coughing: Week 60Coughing: Week 63Coughing: Week 66Coughing: Week 69Coughing: Week 72Coughing: Week 75Coughing: Week 78Coughing: Week 81Coughing: Week 84Coughing: Week 87Coughing: Week 90Coughing: Week 93Coughing: Time of First PdCoughing: Time of Last Tx DoseCoughing: Survival Follow-Up Week 12Coughing: Survival Follow-Up Week 24Dyspnoea: Week 3Dyspnoea: Week 6Dyspnoea: Week 9Dyspnoea: Week 12Dyspnoea: Week 15Dyspnoea: Week 18Dyspnoea: Week 21Dyspnoea: Week 24Dyspnoea: Week 27Dyspnoea: Week 30Dyspnoea: Week 33Dyspnoea: Week 36Dyspnoea: Week 39Dyspnoea: Week 42Dyspnoea: Week 45Dyspnoea: Week 48Dyspnoea: Week 51Dyspnoea: Week 54Dyspnoea: Week 57Dyspnoea: Week 60Dyspnoea: Week 63Dyspnoea: Week 66Dyspnoea: Week 69Dyspnoea: Week 72Dyspnoea: Week 75Dyspnoea: Week 78Dyspnoea: Week 81Dyspnoea: Week 84Dyspnoea: Week 87Dyspnoea: Week 90Dyspnoea: Week 93Dyspnoea: Time of First PDDyspnoea: Time of Last Tx DoseDyspnoea: Survival Follow-Up Week 12Dyspnoea: Survival Follow-Up Week 24Pain In Chest: Week 3Pain In Chest: Week 6Pain In Chest: Week 9Pain In Chest: Week 12Pain In Chest: Week 15Pain In Chest: Week 18Pain In Chest: Week 21Pain In Chest: Week 24Pain In Chest: Week 27Pain In Chest: Week 30Pain In Chest: Week 33Pain In Chest: Week 36Pain In Chest: Week 39Pain In Chest: Week 42Pain In Chest: Week 45Pain In Chest: Week 48Pain In Chest: Week 51Pain In Chest: Week 54Pain In Chest: Week 57Pain In Chest: Week 60Pain In Chest: Week 63Pain In Chest: Week 66Pain In Chest: Week 69Pain In Chest: Week 72Pain In Chest: Week 75Pain In Chest: Week 78Pain In Chest: Week 81Pain In Chest: Week 84Pain In Chest: Week 87Pain In Chest: Week 90Pain In Chest: Week 93Pain In Chest: Time of First PdPain In Chest: Time of Last Tx DosePain In Chest: Survival Follow-Up Week 12Pain In Chest: Survival Follow-Up Week 24Pain In Arm Or Shoulder: Week 3Pain In Arm Or Shoulder: Week 6Pain In Arm Or Shoulder: Week 9Pain In Arm Or Shoulder: Week 12Pain In Arm Or Shoulder: Week 15Pain In Arm Or Shoulder: Week 18Pain In Arm Or Shoulder: Week 21Pain In Arm Or Shoulder: Week 24Pain In Arm Or Shoulder: Week 27Pain In Arm Or Shoulder: Week 30Pain In Arm Or Shoulder: Week 33Pain In Arm Or Shoulder: Week 36Pain In Arm Or Shoulder: Week 39Pain In Arm Or Shoulder: Week 42Pain In Arm Or Shoulder: Week 45Pain In Arm Or Shoulder: Week 48Pain In Arm Or Shoulder: Week 51Pain In Arm Or Shoulder: Week 54Pain In Arm Or Shoulder: Week 57Pain In Arm Or Shoulder: Week 60Pain In Arm Or Shoulder: Week 63Pain In Arm Or Shoulder: Week 66Pain In Arm Or Shoulder: Week 69Pain In Arm Or Shoulder: Week 72Pain In Arm Or Shoulder: Week 75Pain In Arm Or Shoulder: Week 78Pain In Arm Or Shoulder: Week 81Pain In Arm Or Shoulder: Week 84Pain In Arm Or Shoulder: Week 87Pain In Arm Or Shoulder: Week 90Pain In Arm Or Shoulder: Week 93Pain In Arm Or Shoulder: Time of First PdPain In Arm Or Shoulder: Time of Last Tx DosePain In Arm Or Shoulder:Survival Follow-Up Week 12Pain In Arm Or Shoulder:Survival Follow-Up Week 24
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed-3.41-9.82-8.29-10.71-11.53-11.63-10.50-10.03-11.65-11.39-11.97-12.94-13.41-14.70-14.72-13.92-10.97-13.81-13.53-10.71-12.82-12.61-11.90-16.67-18.84-23.81-18.52-29.17-25.00-16.670.00-10.48-8.13-6.94-14.29-1.41-1.17-3.34-0.46-1.12-3.88-1.45-0.42-2.08-2.78-3.02-2.91-4.95-6.33-3.61-5.34-1.69-1.90-4.99-0.40-6.55-4.50-4.76-1.52-6.76-3.17-9.88-6.94-2.785.5622.22-0.160.4413.434.760.81-6.32-4.24-0.60-3.56-3.36-2.74-4.76-2.71-3.06-3.88-2.91-3.99-7.17-5.63-4.64-5.49-4.29-5.80-3.57-6.84-4.50-5.95-4.550.000.007.414.178.330.000.00-3.81-0.83-2.78-4.76-2.60-5.44-8.09-4.37-7.34-6.26-1.83-5.26-3.79-3.33-1.94-3.24-3.99-8.60-6.06-4.22-3.38-6.19-2.90-4.17-3.42-1.80-7.14-7.58-8.70-2.383.704.17-16.670.000.001.900.335.560.00

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality-of-Life Questionnaire-Core 30 (QLQ-C30) Symptom Score

EORTC QLQ-C30 is a validated and reliable self-report measure that consists of 30 questions that assess five aspects of patient functioning (physical, emotional, role, cognitive, and social), three symptom scales (fatigue, nausea and vomiting, pain), global health/quality of life, and six single items (dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties). EORTC QLQ-C30 is scored according to the EORTC scoring manual (Fayers et al. 2001). All EORTC scales and single-item measures are linearly transformed so that each score has a range of 0-100. A high score for a functional/global health status scale represents a high or healthy level of functioning/HRQoL (Health-Related Quality of Life); however a high score for a symptom scale or item represents a high level of symptomatology or problems. A ≥10-point change in the symptoms subscale score is perceived by patients as clinically significant (Osoba et al. 1998). (NCT02657434)
Timeframe: Baseline up to 3 and 6 months after disease progression or loss of clinical benefit (up to approximately 25 months)

InterventionUnits on a scale (Mean)
Dyspnoea: Week 3Dyspnoea: Week 6Dyspnoea: Week 9Dyspnoea: Week 12Dyspnoea: Week 15Dyspnoea: Week 18Dyspnoea: Week 21Dyspnoea: Week 24Dyspnoea: Week 27Dyspnoea: Week 30Dyspnoea: Week 33Dyspnoea: Week 36Dyspnoea: Week 39Dyspnoea: Week 42Dyspnoea: Week 45Dyspnoea: Week 48Dyspnoea: Week 51Dyspnoea: Week 54Dyspnoea: Week 57Dyspnoea: Week 60Dyspnoea: Week 63Dyspnoea: Week 66Dyspnoea: Week 69Dyspnoea: Week 72Dyspnoea: Week 75Dyspnoea: Week 78Dyspnoea: Week 81Dyspnoea: Week 84Dyspnoea: Week 87Dyspnoea: Week 90Dyspnoea: Survival Follow-Up Week 12Dyspnoea: Survival Follow-Up Week 24
Arm B (Carboplatin or Cisplatin + Pemetrexed)-1.95-1.230.20-0.91-1.67-2.12-5.67-2.75-2.53-1.25-1.83-3.83-1.67-3.27-6.25-5.13-14.81-9.68-7.69-11.11-7.69-9.52-16.67-4.17-6.670.000.000.000.000.009.205.26

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by European Organization for the Research and Treatment of Cancer (EORTC) Quality-of-Life Questionnaire-Core 30 (QLQ-C30) Symptom Score

EORTC QLQ-C30 is a validated and reliable self-report measure that consists of 30 questions that assess five aspects of patient functioning (physical, emotional, role, cognitive, and social), three symptom scales (fatigue, nausea and vomiting, pain), global health/quality of life, and six single items (dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties). EORTC QLQ-C30 is scored according to the EORTC scoring manual (Fayers et al. 2001). All EORTC scales and single-item measures are linearly transformed so that each score has a range of 0-100. A high score for a functional/global health status scale represents a high or healthy level of functioning/HRQoL (Health-Related Quality of Life); however a high score for a symptom scale or item represents a high level of symptomatology or problems. A ≥10-point change in the symptoms subscale score is perceived by patients as clinically significant (Osoba et al. 1998). (NCT02657434)
Timeframe: Baseline up to 3 and 6 months after disease progression or loss of clinical benefit (up to approximately 25 months)

InterventionUnits on a scale (Mean)
Dyspnoea: Week 3Dyspnoea: Week 6Dyspnoea: Week 9Dyspnoea: Week 12Dyspnoea: Week 15Dyspnoea: Week 18Dyspnoea: Week 21Dyspnoea: Week 24Dyspnoea: Week 27Dyspnoea: Week 30Dyspnoea: Week 33Dyspnoea: Week 36Dyspnoea: Week 39Dyspnoea: Week 42Dyspnoea: Week 45Dyspnoea: Week 48Dyspnoea: Week 51Dyspnoea: Week 54Dyspnoea: Week 57Dyspnoea: Week 60Dyspnoea: Week 63Dyspnoea: Week 66Dyspnoea: Week 69Dyspnoea: Week 72Dyspnoea: Week 75Dyspnoea: Week 78Dyspnoea: Week 81Dyspnoea: Week 84Dyspnoea: Week 87Dyspnoea: Week 90Dyspnoea: Week 93Dyspnoea: Survival Follow-Up Week 12Dyspnoea: Survival Follow-Up Week 24
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed-1.39-4.71-6.33-3.39-2.44-3.44-2.89-2.45-1.32-2.71-3.74-6.67-8.42-9.12-2.11-7.41-2.92-2.78-5.630.00-5.13-1.80-3.570.00-5.80-4.76-11.11-4.178.330.000.008.00-14.29

[back to top]

Duration of Response (DOR) as Determined by the Investigator Using RECIST v1.1

DOR is defined as the time interval from the date of the first occurrence of a CR or PR (whichever status is recorded first) until the first date that progressive disease or death is documented, whichever occurs first. (NCT02657434)
Timeframe: Randomization up to approximately 25 months

InterventionMonths (Number)
Arm B (Carboplatin or Cisplatin + Pemetrexed)6.4
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed9.5

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms as Reported Using the Symptoms in Lung Cancer (SILC) Scale Score

Change from baseline per SILC scale will be analyzed for each lung cancer symptoms scores. SILC questionnaire comprises 3 individual symptoms & are scored at individual symptom level, thus have a dyspnea score, chest pain score, & cough score. There are a total of 9 questions in SILC questionnaire, each question has a minimum value of 0 & maximum value of 4. Each individual symptom score is calculated as average of responses for symptom items. 'Chest pain' score is mean of question 1 & 2, 'Cough' score is mean of question 3 & 4 and 'Dyspnea' score is mean of question 5 to 9 in SILC questionnaire. An increase in score is suggestive of a worsening in symptomology. A score change of ≥0.3 points for dyspnea & cough symptom scores is considered to be clinically significant; whereas a score change of≥0.5 points for chest pain score is considered to be clinically significant. (NCT02657434)
Timeframe: Baseline up to 3 and 6 months after disease progression or loss of clinical benefit (up to approximately 25 months)

InterventionUnits on a scale (Mean)
Chest Pain: Week 1Chest Pain: Week 2Chest Pain: Week 3Chest Pain: Week 4Chest Pain: Week 5Chest Pain: Week 6Chest Pain: Week 7Chest Pain: Week 8Chest Pain: Week 9Chest Pain: Week 10Chest Pain: Week 11Chest Pain: Week 12Chest Pain: Week 13Chest Pain: Week 14Chest Pain: Week 15Chest Pain: Week 16Chest Pain: Week 17Chest Pain: Week 18Chest Pain: Week 19Chest Pain: Week 20Chest Pain: Week 21Chest Pain: Week 22Chest Pain: Week 23Chest Pain: Week 24Chest Pain: Week 25Chest Pain: Week 26Chest Pain: Week 27Chest Pain: Week 28Chest Pain: Week 29Chest Pain: Week 30Chest Pain: Week 31Chest Pain: Week 32Chest Pain: Week 33Chest Pain: Week 34Chest Pain: Week 35Chest Pain: Week 36Chest Pain: Week 37Chest Pain: Week 38Chest Pain: Week 39Chest Pain: Week 40Chest Pain: Week 41Chest Pain: Week 42Chest Pain: Week 43Chest Pain: Week 44Chest Pain: Week 45Chest Pain: Week 46Chest Pain: Week 47Chest Pain: Week 48Chest Pain: Week 49Chest Pain: Week 50Chest Pain: Week 51Chest Pain: Week 52Chest Pain: Week 53Chest Pain: Week 54Chest Pain: Week 55Chest Pain: Week 56Chest Pain: Week 57Chest Pain: Week 58Chest Pain: Week 59Chest Pain: Week 60Chest Pain: Week 61Chest Pain: Week 62Chest Pain: Week 63Chest Pain: Week 64Chest Pain: Week 65Chest Pain: Week 66Chest Pain: Week 67Chest Pain: Week 68Chest Pain: Week 69Chest Pain: Week 70Chest Pain: Week 71Chest Pain: Week 72Chest Pain: Week 73Chest Pain: Week 74Chest Pain: Week 75Chest Pain: Week 76Chest Pain: Week 77Chest Pain: Week 78Chest Pain: Week 79Chest Pain: Week 80Chest Pain: Week 81Chest Pain: Week 82Chest Pain: Week 83Chest Pain: Week 84Chest Pain: Week 85Chest Pain: Week 86Chest Pain: Week 87Chest Pain: Week 88Chest Pain: Week 89Chest Pain: Week 90Chest Pain: Week 91Chest Pain: Time of First PdChest Pain: Time of Last Tx DoseCough: Week 1Cough: Week 2Cough: Week 3Cough: Week 4Cough: Week 5Cough: Week 6Cough: Week 7Cough: Week 8Cough: Week 9Cough: Week 10Cough: Week 11Cough: Week 12Cough: Week 13Cough: Week 14Cough: Week 15Cough: Week 16Cough: Week 17Cough: Week 18Cough: Week 19Cough: Week 20Cough: Week 21Cough: Week 22Cough: Week 23Cough: Week 24Cough: Week 25Cough: Week 26Cough: Week 27Cough: Week 28Cough: Week 29Cough: Week 30Cough: Week 31Cough: Week 32Cough: Week 33Cough: Week 34Cough: Week 35Cough: Week 36Cough: Week 37Cough: Week 38Cough: Week 39Cough: Week 40Cough: Week 41Cough: Week 42Cough: Week 43Cough: Week 44Cough: Week 45Cough: Week 46Cough: Week 47Cough: Week 48Cough: Week 49Cough: Week 50Cough: Week 51Cough: Week 52Cough: Week 53Cough: Week 54Cough: Week 55Cough: Week 56Cough: Week 57Cough: Week 58Cough: Week 59Cough: Week 60Cough: Week 61Cough: Week 62Cough: Week 63Cough: Week 64Cough: Week 65Cough: Week 66Cough: Week 67Cough: Week 68Cough: Week 69Cough: Week 70Cough: Week 71Cough: Week 72Cough: Week 73Cough: Week 74Cough: Week 75Cough: Week 76Cough: Week 77Cough: Week 78Cough: Week 79Cough: Week 80Cough: Week 81Cough: Week 82Cough: Week 83Cough: Week 84Cough: Week 85Cough: Week 86Cough: Week 87Cough: Week 88Cough: Week 89Cough: Week 90Cough: Week 91Cough: Time of First PdCough: Time of Last Tx DoseDyspnoea: Week 1Dyspnoea: Week 2Dyspnoea: Week 3Dyspnoea: Week 4Dyspnoea: Week 5Dyspnoea: Week 6Dyspnoea: Week 7Dyspnoea: Week 8Dyspnoea: Week 9Dyspnoea: Week 10Dyspnoea: Week 11Dyspnoea: Week 12Dyspnoea: Week 13Dyspnoea: Week 14Dyspnoea: Week 15Dyspnoea: Week 16Dyspnoea: Week 17Dyspnoea: Week 18Dyspnoea: Week 19Dyspnoea: Week 20Dyspnoea: Week 21Dyspnoea: Week 22Dyspnoea: Week 23Dyspnoea: Week 24Dyspnoea: Week 25Dyspnoea: Week 26Dyspnoea: Week 27Dyspnoea: Week 28Dyspnoea: Week 29Dyspnoea: Week 30Dyspnoea: Week 31Dyspnoea: Week 32Dyspnoea: Week 33Dyspnoea: Week 34Dyspnoea: Week 35Dyspnoea: Week 36Dyspnoea: Week 37Dyspnoea: Week 38Dyspnoea: Week 39Dyspnoea: Week 40Dyspnoea: Week 41Dyspnoea: Week 42Dyspnoea: Week 43Dyspnoea: Week 44Dyspnoea: Week 45Dyspnoea: Week 46Dyspnoea: Week 47Dyspnoea: Week 48Dyspnoea: Week 49Dyspnoea: Week 50Dyspnoea: Week 51Dyspnoea: Week 52Dyspnoea: Week 53Dyspnoea: Week 54Dyspnoea: Week 55Dyspnoea: Week 56Dyspnoea: Week 57Dyspnoea: Week 58Dyspnoea: Week 59Dyspnoea: Week 60Dyspnoea: Week 61Dyspnoea: Week 62Dyspnoea: Week 63Dyspnoea: Week 64Dyspnoea: Week 65Dyspnoea: Week 66Dyspnoea: Week 67Dyspnoea: Week 68Dyspnoea: Week 69Dyspnoea: Week 70Dyspnoea: Week 71Dyspnoea: Week 72Dyspnoea: Week 73Dyspnoea: Week 74Dyspnoea: Week 75Dyspnoea: Week 76Dyspnoea: Week 77Dyspnoea: Week 78Dyspnoea: Week 79Dyspnoea: Week 80Dyspnoea: Week 81Dyspnoea: Week 82Dyspnoea: Week 83Dyspnoea: Week 84Dyspnoea: Week 85Dyspnoea: Week 86Dyspnoea: Week 87Dyspnoea: Week 88Dyspnoea: Week 89Dyspnoea: Week 90Dyspnoea: Week 91Dyspnoea: Time of First PdDyspnoea: Time of Last Tx Dose
Arm B (Carboplatin or Cisplatin + Pemetrexed)0.20-0.01-0.050.03-0.02-0.050.070.030.060.030.02-0.02-0.05-0.070.130.020.02-0.06-0.15-0.020.030.04-0.08-0.020.05-0.050.040.240.060.010.020.210.040.210.330.150.150.120.200.15-0.05-0.06-0.22-0.10-0.20-0.060.020.02-0.09-0.13-0.020.140.130.110.120.130.120.270.230.140.230.460.250.150.110.420.360.330.070.250.30-0.130.130.380.380.00-0.25-0.750.00-0.25-0.25-0.75-0.250.00-1.000.00-1.00-1.00-1.000.000.000.280.17-0.08-0.13-0.05-0.11-0.14-0.16-0.22-0.20-0.17-0.22-0.21-0.13-0.24-0.23-0.11-0.20-0.20-0.19-0.25-0.25-0.19-0.29-0.34-0.27-0.24-0.25-0.19-0.19-0.25-0.08-0.130.010.05-0.10-0.270.01-0.110.020.02-0.10-0.27-0.31-0.200.00-0.18-0.25-0.43-0.26-0.50-0.37-0.45-0.29-0.24-0.27-0.10-0.30-0.41-0.17-0.31-0.55-0.230.04-0.29-0.45-0.61-0.17-0.64-0.75-0.57-0.83-0.90-1.25-0.63-0.63-0.63-1.00-0.50-1.00-0.25-0.50-0.50-0.50-1.00-0.500.000.000.000.000.000.000.00-0.08-0.150.200.110.100.210.220.230.300.350.340.460.390.380.340.440.450.370.350.350.300.300.220.160.240.300.500.300.430.460.430.450.350.480.360.470.420.590.530.590.650.620.630.500.320.580.490.550.530.550.630.520.670.720.810.820.770.840.680.770.660.730.850.950.670.460.400.670.370.470.430.330.40-0.050.450.500.55-0.070.000.000.000.100.100.000.000.000.200.000.200.200.200.200.000.580.47

[back to top]

Maximum Observed Serum Atezolizumab Concentration (Cmax)

Maximum observed serum atezolizumab concentration (Cmax) after infusion (Arm A) (NCT02657434)
Timeframe: Day 1 of Cycle 1 (Cycle length=21 days)

Interventionμg/mL (Mean)
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed403

[back to top]

Overall Survival (OS)

OS is defined as time from randomization to death from any cause. (NCT02657434)
Timeframe: Randomization up to approximately 39 months

InterventionMonths (Median)
Arm B (Carboplatin or Cisplatin + Pemetrexed)13.6
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed17.5

[back to top]

Overall Survival Rate at Year 1

The Overall Survival Rate at the 1-year landmark time point is defined as the probabilities that participants are alive 1-year after randomization. (NCT02657434)
Timeframe: Year 1

InterventionPercentage (Number)
Arm B (Carboplatin or Cisplatin + Pemetrexed)55.04
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed59.72

[back to top]

Overall Survival Rate Year 2

The Overall Survival Rate at the 2-year landmark time point is defined as the probabilities that participants are alive 2-years after randomization. (NCT02657434)
Timeframe: Year 2

InterventionPercentage (Number)
Arm B (Carboplatin or Cisplatin + Pemetrexed)34.01
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed39.13

[back to top]

Progression Free Survival (PFS) as Assessed by the Investigator Using Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1)

PFS is defined as the time from randomization to the first occurrence of disease progression as determined by the investigator using RECIST v1.1 or death from any cause, whichever occurred first. (NCT02657434)
Timeframe: Randomization up to approximately 39 months

InterventionMonths (Median)
Arm B (Carboplatin or Cisplatin + Pemetrexed)5.2
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed7.7

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms as Assessed by EORTC Quality-of-Life Lung Cancer Module (QLQ-LC13) Symptom Score

The EORTC QLQ-LC13 module incorporates one multiple item scale to assess dyspnea and a series of single items assessing pain, coughing, sore mouth, dysphagia, peripheral neuropathy, alopecia, and hemoptysis. The EORTC QLQ-LC13 is scored according to the EORTC scoring manual (Fayers et al. 2001). All EORTC scales and single-item measures are linearly transformed so that each score has a range of 0-100. A high score for a functional/global health status scale represents a high or healthy level of functioning/HRQoL (Health-Related Quality of Life); however, a high score for a symptom scale or item represents a high level of symptomatology or problems. A≥10-point change in the symptoms subscale score is perceived by patients as clinically significant (Osoba et al. 1998). (NCT02657434)
Timeframe: Baseline up to 3 and 6 months after disease progression or loss of clinical benefit (up to approximately 25 months)

InterventionUnits on a scale (Mean)
Coughing: Week 3Coughing: Week 6Coughing: Week 9Coughing: Week 12Coughing: Week 15Coughing: Week 18Coughing: Week 21Coughing: Week 24Coughing: Week 27Coughing: Week 30Coughing: Week 33Coughing: Week 36Coughing: Week 39Coughing: Week 42Coughing: Week 45Coughing: Week 48Coughing: Week 51Coughing: Week 54Coughing: Week 57Coughing: Week 60Coughing: Week 63Coughing: Week 66Coughing: Week 69Coughing: Week 72Coughing: Week 75Coughing: Week 78Coughing: Week 81Coughing: Week 84Coughing: Week 87Coughing: Week 90Coughing: Time of First PdCoughing: Time of Last Tx DoseCoughing: Survival Follow-Up Week 12Coughing: Survival Follow-Up Week 24Dyspnoea: Week 3Dyspnoea: Week 6Dyspnoea: Week 9Dyspnoea: Week 12Dyspnoea: Week 15Dyspnoea: Week 18Dyspnoea: Week 21Dyspnoea: Week 24Dyspnoea: Week 27Dyspnoea: Week 30Dyspnoea: Week 33Dyspnoea: Week 36Dyspnoea: Week 39Dyspnoea: Week 42Dyspnoea: Week 45Dyspnoea: Week 48Dyspnoea: Week 51Dyspnoea: Week 54Dyspnoea: Week 57Dyspnoea: Week 60Dyspnoea: Week 63Dyspnoea: Week 66Dyspnoea: Week 69Dyspnoea: Week 72Dyspnoea: Week 75Dyspnoea: Week 78Dyspnoea: Week 81Dyspnoea: Week 84Dyspnoea: Week 87Dyspnoea: Week 90Dyspnoea: Time of First PDDyspnoea: Time of Last Tx DoseDyspnoea: Survival Follow-Up Week 12Dyspnoea: Survival Follow-Up Week 24Pain In Chest: Week 3Pain In Chest: Week 6Pain In Chest: Week 9Pain In Chest: Week 12Pain In Chest: Week 15Pain In Chest: Week 18Pain In Chest: Week 21Pain In Chest: Week 24Pain In Chest: Week 27Pain In Chest: Week 30Pain In Chest: Week 33Pain In Chest: Week 36Pain In Chest: Week 39Pain In Chest: Week 42Pain In Chest: Week 45Pain In Chest: Week 48Pain In Chest: Week 51Pain In Chest: Week 54Pain In Chest: Week 57Pain In Chest: Week 60Pain In Chest: Week 63Pain In Chest: Week 66Pain In Chest: Week 69Pain In Chest: Week 72Pain In Chest: Week 75Pain In Chest: Week 78Pain In Chest: Week 81Pain In Chest: Week 84Pain In Chest: Week 87Pain In Chest: Week 90Pain In Chest: Time of First PdPain In Chest: Time of Last Tx DosePain In Chest: Survival Follow-Up Week 12Pain In Chest: Survival Follow-Up Week 24Pain In Arm Or Shoulder: Week 3Pain In Arm Or Shoulder: Week 6Pain In Arm Or Shoulder: Week 9Pain In Arm Or Shoulder: Week 12Pain In Arm Or Shoulder: Week 15Pain In Arm Or Shoulder: Week 18Pain In Arm Or Shoulder: Week 21Pain In Arm Or Shoulder: Week 24Pain In Arm Or Shoulder: Week 27Pain In Arm Or Shoulder: Week 30Pain In Arm Or Shoulder: Week 33Pain In Arm Or Shoulder: Week 36Pain In Arm Or Shoulder: Week 39Pain In Arm Or Shoulder: Week 42Pain In Arm Or Shoulder: Week 45Pain In Arm Or Shoulder: Week 48Pain In Arm Or Shoulder: Week 51Pain In Arm Or Shoulder: Week 54Pain In Arm Or Shoulder: Week 57Pain In Arm Or Shoulder: Week 60Pain In Arm Or Shoulder: Week 63Pain In Arm Or Shoulder: Week 66Pain In Arm Or Shoulder: Week 69Pain In Arm Or Shoulder: Week 72Pain In Arm Or Shoulder: Week 75Pain In Arm Or Shoulder: Week 78Pain In Arm Or Shoulder: Week 81Pain In Arm Or Shoulder: Week 84Pain In Arm Or Shoulder: Week 87Pain In Arm Or Shoulder: Week 90Pain In Arm Or Shoulder: Time of First PdPain In Arm Or Shoulder: Time of Last Tx DosePain In Arm Or Shoulder:Survival Follow-Up Week 12Pain In Arm Or Shoulder:Survival Follow-Up Week 24
Arm B (Carboplatin or Cisplatin + Pemetrexed)-2.50-3.57-1.85-3.91-6.33-4.00-6.00-4.81-3.850.42-0.46-6.11-5.00-8.97-13.19-7.69-17.59-17.20-11.54-14.04-10.26-9.52-11.11-8.33-13.33-16.67-16.67-33.330.00-33.33-6.01-5.193.57-8.770.210.120.891.150.00-1.07-3.56-2.29-0.711.250.77-1.111.30-0.43-2.08-4.56-9.88-6.45-2.99-11.11-6.84-9.52-16.67-5.56-6.670.0011.110.000.000.002.373.969.924.68-1.250.710.82-2.07-1.46-3.47-3.33-2.41-5.13-4.17-0.46-1.67-3.89-7.69-6.25-7.69-11.11-7.53-1.280.00-5.13-4.76-16.67-8.33-6.670.000.000.00-33.33-33.336.561.345.951.75-4.69-4.46-2.88-6.90-3.89-5.07-6.00-2.41-5.561.25-5.56-1.67-5.000.00-0.69-1.71-2.78-1.08-1.28-5.26-5.134.760.00-8.33-6.670.000.000.000.000.002.19-0.5013.101.75

[back to top]

Plasma Concentrations for Pemetrexed in Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed)

(NCT02657434)
Timeframe: Prd (0 h), 5-10 mins before end of pemetrexed infusion (infusion duration=10 mins), 1 h post-infusion on D1 of Cy1,3 (Cy length=21 days)

Interventionng/mL (Mean)
Cy1D1 PredoseCy1D1 Before End of InfusionCy1D1 Post InfusionCy3D1 PredoseCy3D1 Before End of InufsionCy3D1 Post Infusion
Arm A (Atezolizumab + Carboplatin or Cisplatin + PemetrexedNA86500436001.837940050100

[back to top]

Change From Baseline in Patient-Reported Lung Cancer Symptoms as Reported Using the Symptoms in Lung Cancer (SILC) Scale Score

Change from baseline per SILC scale will be analyzed for each lung cancer symptoms scores. SILC questionnaire comprises 3 individual symptoms & are scored at individual symptom level, thus have a dyspnea score, chest pain score, & cough score. There are a total of 9 questions in SILC questionnaire, each question has a minimum value of 0 & maximum value of 4. Each individual symptom score is calculated as average of responses for symptom items. 'Chest pain' score is mean of question 1 & 2, 'Cough' score is mean of question 3 & 4 and 'Dyspnea' score is mean of question 5 to 9 in SILC questionnaire. An increase in score is suggestive of a worsening in symptomology. A score change of ≥0.3 points for dyspnea & cough symptom scores is considered to be clinically significant; whereas a score change of≥0.5 points for chest pain score is considered to be clinically significant. (NCT02657434)
Timeframe: Baseline up to 3 and 6 months after disease progression or loss of clinical benefit (up to approximately 25 months)

InterventionUnits on a scale (Mean)
Chest Pain: Week 1Chest Pain: Week 2Chest Pain: Week 3Chest Pain: Week 4Chest Pain: Week 5Chest Pain: Week 6Chest Pain: Week 7Chest Pain: Week 8Chest Pain: Week 9Chest Pain: Week 10Chest Pain: Week 11Chest Pain: Week 12Chest Pain: Week 13Chest Pain: Week 14Chest Pain: Week 15Chest Pain: Week 16Chest Pain: Week 17Chest Pain: Week 18Chest Pain: Week 19Chest Pain: Week 20Chest Pain: Week 21Chest Pain: Week 22Chest Pain: Week 23Chest Pain: Week 24Chest Pain: Week 25Chest Pain: Week 26Chest Pain: Week 27Chest Pain: Week 28Chest Pain: Week 29Chest Pain: Week 30Chest Pain: Week 31Chest Pain: Week 32Chest Pain: Week 33Chest Pain: Week 34Chest Pain: Week 35Chest Pain: Week 36Chest Pain: Week 37Chest Pain: Week 38Chest Pain: Week 39Chest Pain: Week 40Chest Pain: Week 41Chest Pain: Week 42Chest Pain: Week 43Chest Pain: Week 44Chest Pain: Week 45Chest Pain: Week 46Chest Pain: Week 47Chest Pain: Week 48Chest Pain: Week 49Chest Pain: Week 50Chest Pain: Week 51Chest Pain: Week 52Chest Pain: Week 53Chest Pain: Week 54Chest Pain: Week 55Chest Pain: Week 56Chest Pain: Week 57Chest Pain: Week 58Chest Pain: Week 59Chest Pain: Week 60Chest Pain: Week 61Chest Pain: Week 62Chest Pain: Week 63Chest Pain: Week 64Chest Pain: Week 65Chest Pain: Week 66Chest Pain: Week 67Chest Pain: Week 68Chest Pain: Week 69Chest Pain: Week 70Chest Pain: Week 71Chest Pain: Week 72Chest Pain: Week 73Chest Pain: Week 74Chest Pain: Week 75Chest Pain: Week 76Chest Pain: Week 77Chest Pain: Week 78Chest Pain: Week 79Chest Pain: Week 80Chest Pain: Week 81Chest Pain: Week 82Chest Pain: Week 83Chest Pain: Week 84Chest Pain: Week 85Chest Pain: Week 86Chest Pain: Week 87Chest Pain: Week 88Chest Pain: Week 89Chest Pain: Week 90Chest Pain: Week 91Chest Pain: Week 92Chest Pain: Week 93Chest Pain: Week 94Chest Pain: Week 95Chest Pain: Time of First PdChest Pain: Time of Last Tx DoseCough: Week 1Cough: Week 2Cough: Week 3Cough: Week 4Cough: Week 5Cough: Week 6Cough: Week 7Cough: Week 8Cough: Week 9Cough: Week 10Cough: Week 11Cough: Week 12Cough: Week 13Cough: Week 14Cough: Week 15Cough: Week 16Cough: Week 17Cough: Week 18Cough: Week 19Cough: Week 20Cough: Week 21Cough: Week 22Cough: Week 23Cough: Week 24Cough: Week 25Cough: Week 26Cough: Week 27Cough: Week 28Cough: Week 29Cough: Week 30Cough: Week 31Cough: Week 32Cough: Week 33Cough: Week 34Cough: Week 35Cough: Week 36Cough: Week 37Cough: Week 38Cough: Week 39Cough: Week 40Cough: Week 41Cough: Week 42Cough: Week 43Cough: Week 44Cough: Week 45Cough: Week 46Cough: Week 47Cough: Week 48Cough: Week 49Cough: Week 50Cough: Week 51Cough: Week 52Cough: Week 53Cough: Week 54Cough: Week 55Cough: Week 56Cough: Week 57Cough: Week 58Cough: Week 59Cough: Week 60Cough: Week 61Cough: Week 62Cough: Week 63Cough: Week 64Cough: Week 65Cough: Week 66Cough: Week 67Cough: Week 68Cough: Week 69Cough: Week 70Cough: Week 71Cough: Week 72Cough: Week 73Cough: Week 74Cough: Week 75Cough: Week 76Cough: Week 77Cough: Week 78Cough: Week 79Cough: Week 80Cough: Week 81Cough: Week 82Cough: Week 83Cough: Week 84Cough: Week 85Cough: Week 86Cough: Week 87Cough: Week 88Cough: Week 89Cough: Week 90Cough: Week 91Cough: Week 92Cough: Week 93Cough: Week 94Cough: Week 95Cough: Time of First PdCough: Time of Last Tx DoseDyspnoea: Week 1Dyspnoea: Week 2Dyspnoea: Week 3Dyspnoea: Week 4Dyspnoea: Week 5Dyspnoea: Week 6Dyspnoea: Week 7Dyspnoea: Week 8Dyspnoea: Week 9Dyspnoea: Week 10Dyspnoea: Week 11Dyspnoea: Week 12Dyspnoea: Week 13Dyspnoea: Week 14Dyspnoea: Week 15Dyspnoea: Week 16Dyspnoea: Week 17Dyspnoea: Week 18Dyspnoea: Week 19Dyspnoea: Week 20Dyspnoea: Week 21Dyspnoea: Week 22Dyspnoea: Week 23Dyspnoea: Week 24Dyspnoea: Week 25Dyspnoea: Week 26Dyspnoea: Week 27Dyspnoea: Week 28Dyspnoea: Week 29Dyspnoea: Week 30Dyspnoea: Week 31Dyspnoea: Week 32Dyspnoea: Week 33Dyspnoea: Week 34Dyspnoea: Week 35Dyspnoea: Week 36Dyspnoea: Week 37Dyspnoea: Week 38Dyspnoea: Week 39Dyspnoea: Week 40Dyspnoea: Week 41Dyspnoea: Week 42Dyspnoea: Week 43Dyspnoea: Week 44Dyspnoea: Week 45Dyspnoea: Week 46Dyspnoea: Week 47Dyspnoea: Week 48Dyspnoea: Week 49Dyspnoea: Week 50Dyspnoea: Week 51Dyspnoea: Week 52Dyspnoea: Week 53Dyspnoea: Week 54Dyspnoea: Week 55Dyspnoea: Week 56Dyspnoea: Week 57Dyspnoea: Week 58Dyspnoea: Week 59Dyspnoea: Week 60Dyspnoea: Week 61Dyspnoea: Week 62Dyspnoea: Week 63Dyspnoea: Week 64Dyspnoea: Week 65Dyspnoea: Week 66Dyspnoea: Week 67Dyspnoea: Week 68Dyspnoea: Week 69Dyspnoea: Week 70Dyspnoea: Week 71Dyspnoea: Week 72Dyspnoea: Week 73Dyspnoea: Week 74Dyspnoea: Week 75Dyspnoea: Week 76Dyspnoea: Week 77Dyspnoea: Week 78Dyspnoea: Week 79Dyspnoea: Week 80Dyspnoea: Week 81Dyspnoea: Week 82Dyspnoea: Week 83Dyspnoea: Week 84Dyspnoea: Week 85Dyspnoea: Week 86Dyspnoea: Week 87Dyspnoea: Week 88Dyspnoea: Week 89Dyspnoea: Week 90Dyspnoea: Week 91Dyspnoea: Week 92Dyspnoea: Week 93Dyspnoea: Week 94Dyspnoea: Week 95Dyspnoea: Time of First PdDyspnoea: Time of Last Tx Dose
Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed0.300.210.060.03-0.01-0.02-0.04-0.07-0.030.020.04-0.010.050.060.080.080.000.120.02-0.010.010.020.040.090.140.090.040.030.040.040.05-0.010.030.110.110.170.060.040.130.040.090.030.130.100.030.080.090.050.030.000.120.00-0.01-0.080.030.04-0.01-0.12-0.23-0.150.00-0.04-0.060.09-0.030.130.020.12-0.050.030.04-0.22-0.10-0.02-0.07-0.06-0.21-0.25-0.18-0.12-0.250.00-0.50-0.30-0.20-0.500.500.000.000.000.000.000.001.000.500.200.13-0.06-0.08-0.07-0.22-0.33-0.33-0.22-0.28-0.29-0.35-0.33-0.40-0.37-0.34-0.33-0.33-0.31-0.31-0.36-0.39-0.36-0.41-0.44-0.29-0.27-0.22-0.32-0.32-0.34-0.34-0.37-0.45-0.44-0.29-0.32-0.31-0.37-0.31-0.33-0.35-0.30-0.31-0.37-0.36-0.41-0.34-0.29-0.38-0.40-0.40-0.15-0.10-0.13-0.16-0.35-0.26-0.25-0.45-0.50-0.52-0.38-0.53-0.57-0.49-0.39-0.43-0.47-0.35-0.53-0.43-0.56-0.72-0.77-0.54-0.67-0.65-0.82-0.57-0.64-1.04-0.79-0.890.13-0.200.00-0.38-0.83-0.880.000.25-0.500.500.500.500.00-0.45-0.290.160.150.120.170.170.160.250.240.170.160.260.220.300.220.190.260.150.220.220.270.220.280.220.240.280.240.240.250.200.210.150.160.170.240.230.220.240.220.190.190.160.170.220.230.210.230.140.140.160.240.300.270.220.230.160.180.110.160.070.270.230.160.270.140.120.160.260.250.160.070.130.060.090.270.450.020.090.030.19-0.26-0.170.180.450.600.32-0.15-0.47-0.70-0.20-0.50-0.50-0.70-0.40-0.80-1.000.400.18

[back to top]

Plasma Concentrations for Cisplatin in Arm A (Atezolizumab + Carboplatin or Cisplatin + Pemetrexed)

(NCT02657434)
Timeframe: Prd (0 h), 5-10 mins before end of cisplatin infusion (infusion duration=30-60 mins), 1 h post-infusion on D1 of Cy1,3 (Cy length=21 days)

Interventionng/mL (Mean)
Cy1D1 PredoseCy1D1 Before End of InfusionCy1D1 Post InfusionCy3D1 PredoseCy3D1 Before End of InfusionCy3D1 Post Infusion
Arm A (Atezolizumab + Carboplatin or Cisplatin + PemetrexedNA3630240029030202740

[back to top]

Plasma Concentrations for Carboplatin in Arm A(Atezolizumab + Carboplatin or Cisplatin + Pemetrexed)

(NCT02657434)
Timeframe: Prd (0 h), 5-10 minutes (mins) before end of carboplatin infusion (infusion duration=1-2 h), 1 h post-infusion on D1 of Cy1,3 (Cy length=21 days)

Interventionng/mL (Mean)
Cy1D1 PredoseCy1D1 Before End of InfusionCy1D1 Post InfusionCy3D1 PredoseCy3D1 Before End of InfusionCy3D1 Post Infusion
Arm A (Atezolizumab + Carboplatin or Cisplatin + PemetrexedNA14900128002201790013900

[back to top]

Number of Participants With Laboratory Abnormalities in Hepatic Tests - Part 2

Number of participant with specific liver laboratory abnormalities graded by Common Terminology Criteria for Adverse Events (CTCAE v4.0) to determine the safety and tolerability of Nivolumab and Ipilimumab combined with chemotherapy. (NCT02659059)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Count of Participants)
ALT OR AST >3XULNALT OR AST >5XULNALT OR AST >10XULNALT OR AST >20XULNTOTAL BILIRUBIN >2XULNConcurrent ALT OR AST >3XULN & BILIRUBIN >2XULN within 1 dayConcurrent ALT OR AST >3XULN & BILIRUBIN >2XULN within 30 days
Nivolumab+Ipilimumab+Chemotherapy2211000

[back to top]

Progression Free Survival (PFS) - Part 1

Progression Free Survival (PFS) was defined as the time between the date of first dose and the first date of documented progression, as determined by blinded independent central review (BICR), or death due to any cause, whichever occurred first. Progressive Disease (PD): 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 also demonstrate an absolute increase of at least 5 mm. (NCT02659059)
Timeframe: From first dose to the first date of documented progression, or death due to any cause, whichever occurred first (Up to approximately 72 months)

InterventionMonths (Median)
Nivolumab+Ipilimumab5.19

[back to top]

Overall Survival (OS) - Part 2

Overall survival (OS) was defined as the time from date of first treatment to the date of death due to any cause. A participant who has not died will be censored at the last known date alive. (NCT02659059)
Timeframe: From the date of first treatment to the date of death due to any cause (Up to approximately 59 months)

InterventionMonths (Median)
Nivolumab+Ipilimumab+Chemotherapy19.35

[back to top]

Overall Survival (OS) - Part 1

Overall survival (OS) was defined as the time from date of first treatment to the date of death due to any cause. A participant who has not died will be censored at the last known date alive. (NCT02659059)
Timeframe: From the date of first treatment to the date of death due to any cause (Up to approximately 72 months)

InterventionMonths (Median)
Nivolumab+Ipilimumab20.83

[back to top]

Objective Response Rate (ORR) - Part 2

"Objective response rate (ORR) was defined as the percentage of treated participants with confirmed complete response (CR) or partial response (PR) per RECIST 1.1 based on investigator assessment.~CR = Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR = At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters." (NCT02659059)
Timeframe: From first dose up to approximately 59 months

InterventionPercentage of participants (Number)
Nivolumab+Ipilimumab+Chemotherapy47.2

[back to top]

Objective Response Rate (ORR) - Part 1

"Objective response rate (ORR) was defined as the percentage of treated participants with confirmed complete response (CR) or partial response (PR) per RECIST 1.1 based on Blinded Independent Central Review (BICR) assessment.~CR = Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR = At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters." (NCT02659059)
Timeframe: From first dose up to approximately 72 months

InterventionPercentage of participants (Number)
Nivolumab+Ipilimumab32.3

[back to top]

Number of Participants With Dose Limiting Toxicities (DLTs) - Part 2

"Dose limiting toxicities (DLTs) were defined as any of the items listed below.~Any Grade 2 drug-related uveitis or eye pain that does not respond to topical therapy and does not improve to Grade 1 severity within the re-treatment period OR requires systemic treatment.~Any Grade 2 drug-related pneumonitis or interstitial lung disease that does not resolve to dose delay and systemic steroids in 14 days.~Any Grade 3 non-skin drug-related adverse event with the exception of laboratory abnormalities that cannot be alleviated or controlled by appropriate care within 14 days.~Any Grade 4 drug-related adverse event including laboratory abnormalities except Grade 4 leukopenia or neutropenia lasting < 14 days and asymptomatic amylase/lipase elevation.~Drug-related hepatic function laboratory abnormalities." (NCT02659059)
Timeframe: 9 weeks after first dose

InterventionParticipants (Count of Participants)
Nivolumab+Ipilimumab+Chemotherapy1

[back to top]

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

Number of participants with adverse events (AEs) including serious adverse events (SAEs) and deaths graded by Common Terminology Criteria for Adverse Events (CTCAE v4.0) to determine the safety and tolerability of Nivolumab and Ipilimumab combined with chemotherapy. (NCT02659059)
Timeframe: Deaths are from first dose to database lock (Up to 24 months). AEs and SAEs are from first dose to 30 days post last dose

InterventionParticipants (Count of Participants)
Adverse Events (AEs)Serious Adverse Events (SAEs)Deaths due to Disease progressionDeaths due to Study drug toxicityDeaths due to unknown causesDeaths due to other causes
Nivolumab+Ipilimumab+Chemotherapy36269016

[back to top]

Progression Free Survival (PFS) by Tumor Mutation Burden (TMB) Levels - Part 1

"Progression Free Survival (PFS) by tumor mutational burden (TMB) using DNA derived from tumor specimens was defined as the time between the date of first dose and the first date of documented progression, as determined by blinded independent central review (BICR), or death due to any cause, whichever occurred first. Progressive Disease (PD): 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 also demonstrate an absolute increase of at least 5 mm.~High TMB = ≥ 10 mutations per megabase Low TMB = < 10 mutations per megabase" (NCT02659059)
Timeframe: From first dose to the first date of documented progression, or death due to any cause, whichever occurred first (Up to approximately 72 months)

InterventionMonths (Median)
High TMB (>=10 Mutations/MB)Low TMB (<10 Mutations/MB)
Nivolumab+Ipilimumab10.842.79

[back to top]

Progression Free Survival (PFS) - Part 2

Progression Free Survival (PFS) was defined as the time between the date of first dose and the first date of documented progression, as determined by investigator (per RECIST 1.1), or death due to any cause, whichever occurred first. Progressive Disease (PD): 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 also demonstrate an absolute increase of at least 5 mm. (NCT02659059)
Timeframe: From first dose to the first date of documented progression, or death due to any cause, whichever occurred first (Up to approximately 59 months)

InterventionMonths (Median)
Nivolumab+Ipilimumab+Chemotherapy10.81

[back to top]

Progression Free Survival (PFS) by PD-L1 Expression Levels - Part 1

"Progression Free Survival (PFS) by PD-L1 expression levels was defined as the time between the date of first dose and the first date of documented progression, as determined by blinded independent central review (BICR), or death due to any cause, whichever occurred first. Progressive Disease (PD): 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 also demonstrate an absolute increase of at least 5 mm.~PD-L1 ≥1% = PD-L1 positive (membranous staining in ≥ 1% tumor cells) PD-L1 <1% = PD-L1 negative (membranous staining in <1% tumor cells)" (NCT02659059)
Timeframe: From first dose to the first date of documented progression, or death due to any cause, whichever occurred first (Up to approximately 72 months)

InterventionMonths (Median)
PD-L1 ≥1%PD-L1 <1%
Nivolumab+Ipilimumab6.802.92

[back to top]

Overall Survival (OS) by Tumor Mutation Burden (TMB) Levels - Part 1

"Overall survival (OS) by tumor mutational burden (TMB) using DNA derived from tumor specimens was defined as the time from date of first treatment to the date of death due to any cause. A participant who has not died will be censored at the last known date alive.~High TMB = ≥ 10 mutations per megabase Low TMB = < 10 mutations per megabase" (NCT02659059)
Timeframe: From the date of first treatment to the date of death due to any cause (Up to approximately 72 months)

InterventionMonths (Median)
High TMB (>=10 Mutations/MB)Low TMB (<10 Mutations/MB)
Nivolumab+Ipilimumab47.3111.33

[back to top]

Overall Survival (OS) by PD-L1 Expression Levels - Part 1

"Overall survival (OS) by PD-L1 expression levels was defined as the time from date of first treatment to the date of death due to any cause. A participant who has not died will be censored at the last known date alive.~PD-L1 ≥1% = PD-L1 positive (membranous staining in ≥ 1% tumor cells) PD-L1 <1% = PD-L1 negative (membranous staining in <1% tumor cells)" (NCT02659059)
Timeframe: From the date of first treatment to the date of death due to any cause (Up to approximately 72 months)

InterventionMonths (Median)
PD-L1 ≥1%PD-L1 <1%
Nivolumab+Ipilimumab26.5113.70

[back to top]

Objective Response Rate (ORR) by Tumor Mutation Burden (TMB) Levels - Part 1

"Objective response rate (ORR) by tumor mutational burden (TMB) using DNA derived from tumor specimens was defined as the percentage of treated participants with confirmed complete response (CR) or partial response (PR) per RECIST 1.1 based on Blinded Independent Central Review (BICR) assessment.~CR = Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR = At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.~High TMB = ≥ 10 mutations per megabase Low TMB = < 10 mutations per megabase" (NCT02659059)
Timeframe: From first dose up to approximately 72 months

InterventionPercentage of participants (Number)
High TMB (>=10 Mutations/MB)Low TMB (<10 Mutations/MB)
Nivolumab+Ipilimumab52.116.0

[back to top]

Objective Response Rate (ORR) by PD-L1 Positive and Negative Levels - Part 1

Objective response rate (ORR) in PD-L1 positive (PD-L1 ≥1%) and PD-L1 negative (PD-L1 <1%) participants was defined as the percentage of treated participants with confirmed complete response (CR) or partial response (PR) per RECIST 1.1 based on Blinded Independent Central Review (BICR) assessment. (NCT02659059)
Timeframe: From first dose to database lock (Up to 18 months)

InterventionPercentage of participants (Number)
PD-L1 ≥1%PD-L1 <1%
Nivolumab+Ipilimumab41.314.9

[back to top]

Objective Response Rate (ORR) by PD-L1 Expression Levels-Part 1

"Objective response rate (ORR) by PD-L1 expression levels was defined as the percentage of treated participants with confirmed complete response (CR) or partial response (PR) per RECIST 1.1 based on Blinded Independent Central Review (BICR) assessment.~CR = Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to < 10 mm.~PR = At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.~PD-L1 ≥1% = PD-L1 positive (membranous staining in ≥ 1% tumor cells) PD-L1 <1% = PD-L1 negative (membranous staining in <1% tumor cells)" (NCT02659059)
Timeframe: From first dose up to approximately 72 months

InterventionPercentage of participants (Number)
PD-L1 ≥1%PD-L1 <1%
Nivolumab+Ipilimumab44.217.1

[back to top]

Number of Participants With Laboratory Abnormalities in Thyroid Tests - Part 2

Number of participants with specific thyroid laboratory abnormalities graded by Common Terminology Criteria for Adverse Events (CTCAE v4.0) to determine the safety and tolerability of Nivolumab and Ipilimumab combined with chemotherapy. (NCT02659059)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Count of Participants)
TSH > ULNTSH > ULN with TSH <= ULN at baselineTSH > ULN with at least one FT3/FT4 test value < LLNTSH > ULN with all other FT3/FT4 test values ≥ LLNTSH > ULN with FT3/FT4 test missingTSH < LLNTSH < LLN with TSH >= LLN at baselineTSH < LLN with at least one FT3/FT4 test value > ULNTSH < LLN with all other FT3/FT4 test values <= ULNTSH < LLN with FT3/Ft4 test missing
Nivolumab+Ipilimumab+Chemotherapy1078111312760

[back to top]

Overall Survival

"Overall survival is defined as the time from randomization until death. In the event that no death was documented prior to study termination or analysis cutoff, OS was censored at the last known date the subject was known to be alive, either through completion of on-study visits or through survival follow-up contact.~The treatment effect on OS was evaluated using the stratified log-rank test (stratified by tumor histology).~The Kaplan-Meier curves were also plotted. A Cox proportional hazard model with an adjustment for tumor histology (biphasic vs sarcomatoid) was used to compute the estimated hazard ratio and two-sided 95% CI. The treatment effect on OS was evaluated using the stratified log-rank test (stratified by tumor histology). The significance level to be used in the OS analysis at the final analysis was based on α = 0.04999 (two-sided)." (NCT02709512)
Timeframe: 18 months

Interventionmonths (Median)
Drug: ADI-PEG 20 Plus Pem Platinum9.30
Drug: Placebo Plus Pem Platinum7.66

[back to top]

Overall Survival Phase 3 Interim Analysis

"The primary analysis of OS Phase 3 was performed at the interim analysis. This was performed once 50% of the planned OS events for phase 3 have occurred (ie, 169 of the 338 planned OS events). This interim analysis will evaluate OS in the ITT population in an unblinded manner. The OS data at the second interim analysis will be analyzed to support the following decisions:~Futility stopping: Terminate the study due to futility at the interim analysis. Sample size re-estimation: Increase the target number of OS events after the second interim analysis.. The treatment effect on OS will be evaluated using the stratified log-rank test (stratified by tumor histology)." (NCT02709512)
Timeframe: Approximately 18 months

Interventionmonths (Median)
Drug: ADI-PEG 20 Plus Pem Platinum9.82
Drug: Placebo Plus Pem Platinum7.49

[back to top]

Progression Free Survival

The key secondary endpoint for the phase 3 portion is PFS, which will be analyzed only if the analysis of OS is statistically significant at the final analysis, with alpha level of 0.05 (two-sided) using the same statistical methodologies as applied to OS. (NCT02709512)
Timeframe: approximately 18 months

Interventionmonths (Median)
Drug: ADI-PEG 20 Plus Pem Platinum6.24
Drug: Placebo Plus Pem Platinum5.65

[back to top]

Response Rate

"Objective Response Rate is calculated as the proportion of subjects whose best tumor response from all post-baseline tumor assessments is complete response (CR) or partial response (PR). The best tumor response is the best response recorded from the start of the treatment until the end of treatment taking into account any requirement for confirmation.~To test Objective Response Rate significance, a Relative Risk Ratio (ADI-PEG 20 / Placebo) was calculated as the common relative risk of having a response (CR or PR) based on the Mantel-Haenszel estimator controlling for tumor histology (biphasic versus sarcomatoid)." (NCT02709512)
Timeframe: approximately 18 months

InterventionParticipants (Count of Participants)
Drug: ADI-PEG 20 Plus Pem Platinum12
Drug: Placebo Plus Pem Platinum12

[back to top]

Number of Subjects With NSCLC Who Achieved DCB

Objective response to study treatment will be assessed by RECIST 1.1 by a study radiologist. Partial and complete responses will be confirmed by a repeat imaging occurring at least 4 weeks after the initial identification of response; unconfirmed responses will be considered stable or progressive disease dependent on results of the second CT scan. Durable clinical benefit (DCB) will be defined as stable disease (Neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum diameters while on study), OR complete response (disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.), OR partial response (At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) lasting longer than 6 months. (NCT02710396)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Cohort 13
Cohort 20
Cohort 30

[back to top]

Objective Response Rate (ORR)

ORR is defined as participants with a partial (At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) or complete response (Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm) as assessed by RECIST 1.1 criteria. (NCT02710396)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Cohort 14
Cohort 21
Cohort 30

[back to top]

Progression Free Survival (PFS)

The length of time from treatment initiation to progression of disease (at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. The appearance of one or more new lesions is also considered progression) as assessed by RECIST 1.1. (NCT02710396)
Timeframe: Up to 2 years

Interventionmonths (Median)
Cohort 14.80
Cohort 23.94

[back to top]

Overall Survival (OS)

The length of time from the start of treatment to death. (NCT02710396)
Timeframe: Up to 28 months

InterventionMonths (Median)
Cohort 114.06
Cohort 223.49

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab in Part B Cycles 1, 2, 4, 6, and 8

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose for participants that received either dose regimen in Part B for Cohort 1, Cohort 2, Cohort 3, and Cohorts 1+2+3 combined. Per protocol, blood sampling for Ctrough was taken at pre-dose prior to the Cycle 1, 2, 4, 6, and 8 infusion. Cycle 1 length was 28 days (21-day pembrolizumab treatment cycle starting at Day 8 of Cycle 1). Cycle 2, 4, 6, and 8 length was 21 days. Ctrough is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Predose prior to the Cycles 1, 2, 4, 6, and 8 infusion

,,,
Interventionµg/mL (Geometric Mean)
Cycle 1Cycle 2Cycle 4Cycle 6Cycle 8
Part B Cohort 1: Pembrolizumab+Cisplatin+PemetrexedNA16.637.853.865.4
Part B Cohort 2: Pembrolizumab+Carboplatin+PemetrexedNA17.428.140.552.7
Part B Cohort 3: Pembrolizumab+Carboplatin+PaclitaxelNA8.6925.421.534.5
Part B Combined Cohorts 1, 2, and 3NA13.331.538.650.3

[back to top]

Maximum Concentration (Cmax) of Epacadostat in Part A

Cmax was the maximum observed concentration of epacadostat in plasma for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, Cmax for Cohort 1 was measured on Days 1, 5, and 12 and Cmax for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). Cmax is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
InterventionnM (Geometric Mean)
Day 5
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab852
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab371

[back to top]

Maximum Concentration (Cmax) of Pembrolizumab in Part B Cycle 1

Cmax was the maximum observed concentration of pembrolizumab in serum for participants that received either dose regimen in Part B for Cohort 1, Cohort 2, Cohort 3, and Cohorts 1+2+ 3 combined. Per protocol, the analysis for the Cmax of pembrolizumab was performed in Cycle 1 only. Blood samples were collected predose and postdose within 30 minutes post pembrolizumab infusion during Cycle 1 (21-day pembrolizumab treatment cycle starting at Day 8 of Cycle 1). Cmax is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (21-day pembrolizumab treatment cycle starting at Day 8 of Cycle 1): Day 1 predose and postdose within 30 minutes after the end of pembrolizumab infusion

Interventionµg/mL (Geometric Mean)
Part B Cohort 1: Pembrolizumab+Cisplatin+Pemetrexed73.8
Part B Cohort 2: Pembrolizumab+Carboplatin+Pemetrexed64.3
Part B Cohort 3: Pembrolizumab+Carboplatin+Paclitaxel65.3
Part B Combined Cohorts 1, 2, and 368.0

[back to top]

Trough Concentration (Ctrough) of Pembrolizumab in Part A Cycles 1, 2, 4, 6, and 8

Ctrough was the lowest concentration of pembrolizumab in serum just before the next dose for participants that received either dose regimen in Part A for Cohort 1 combined, Cohort 2 combined, and Cohorts 1+2 combined. Per protocol, blood sampling for Ctrough was taken at predose prior to the Cycle 1, 2, 4, 6, and 8 infusion. Cycle 1 length was 28 days (21-day pembrolizumab treatment cycle starting at Day 8 of Cycle 1). Cycle 2, 4, 6, and 8 length was 21 days. Ctrough is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Predose prior to the Cycles 1, 2, 4, 6, and 8 infusion

,,
Interventionµg/mL (Geometric Mean)
Cycle 1Cycle 2Cycle 4Cycle 6Cycle 8
Part A Combined Cohort 1NA17.324.528.538.2
Part A Combined Cohort 2NA17.941.851.047.9
Part A Combined Cohorts 1 And 2NA17.635.039.845.2

[back to top]

Trough Concentration (Ctrough) of Epacadostat in Part A

Ctrough was the lowest concentration of epacadostat in plasma just before the next dose for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, Ctrough for Cohort 1 was measured on Days 1, 5, and 12 and Ctrough for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). Ctrough is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose

,
InterventionnM (Geometric Mean)
Day 1Day 5Day 12
Part A Cohort 1: Epacadostat 100 mgNA87.795.4
Part A Cohort 1: Epacadostat 25 mgNANANA

[back to top]

Trough Concentration (Ctrough) of Epacadostat in Part A

Ctrough was the lowest concentration of epacadostat in plasma just before the next dose for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, Ctrough for Cohort 1 was measured on Days 1, 5, and 12 and Ctrough for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). Ctrough is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose

,
InterventionnM (Geometric Mean)
Day 5
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab106.0
Part A Cohort 2: Epacadostat 25 mg+PembrolizumabNA

[back to top]

Time to Maximum Concentration (Tmax) of Epacadostat in Part A

Tmax was the time required to reach the maximum concentration of epacadostat in plasma for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, Tmax for Cohort 1 was measured on Days 1, 5, and 12 and Tmax for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). Tmax is presented as a Median with a full range. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
Interventionhours (Median)
Day 1Day 5Day 12
Part A Cohort 1: Epacadostat 100 mg2.002.002.00
Part A Cohort 1: Epacadostat 25 mg2.002.002.00

[back to top]

Maximum Concentration (Cmax) of Pembrolizumab in Part A Cycle 1

Cmax was the maximum observed concentration of pembrolizumab in serum for participants that received either dose regimen in Part A for Cohort 1 combined, Cohort 2 combined, and Cohorts 1+2 combined. Per protocol, the analysis for the Cmax of pembrolizumab was performed in Cycle 1 only. Blood samples were collected predose and postdose within 30 minutes post pembrolizumab infusion during Cycle 1 (21-day pembrolizumab treatment cycle starting at Day 8 of Cycle 1). Cmax is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (21-day pembrolizumab treatment cycle starting at Day 8 of Cycle 1): Day 1 predose and postdose within 30 minutes after the end of pembrolizumab infusion

Interventionµg/mL (Geometric Mean)
Part A Combined Cohort 180.0
Part A Combined Cohort 273.8
Part A Combined Cohorts 1 and 276.2

[back to top]

Time to Maximum Concentration (Tmax) of Epacadostat in Part A

Tmax was the time required to reach the maximum concentration of epacadostat in plasma for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, Tmax for Cohort 1 was measured on Days 1, 5, and 12 and Tmax for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). Tmax is presented as a Median with a full range. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
Interventionhours (Median)
Day 5
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab2.00
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab2.00

[back to top]

Terminal Half-Life (t1/2) of Epacadostat in Part A

t1/2 was the time required to divide the epacadostat concentration by two after reaching pseudo-equilibrium. Plasma t1/2 was measured for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, t1/2 for Cohort 1 was measured on Days 1, 5, and 12 and t1/2 for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). t1/2 is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
Interventionhours (Geometric Mean)
Day 1Day 5Day 12
Part A Cohort 1: Epacadostat 100 mg2.552.822.43
Part A Cohort 1: Epacadostat 25 mg3.943.014.27

[back to top]

Terminal Half-Life (t1/2) of Epacadostat in Part A

t1/2 was the time required to divide the epacadostat concentration by two after reaching pseudo-equilibrium. Plasma t1/2 was measured for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, t1/2 for Cohort 1 was measured on Days 1, 5, and 12 and t1/2 for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). t1/2 is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
Interventionhours (Geometric Mean)
Day 5
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab3.77
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab2.62

[back to top]

Maximum Concentration (Cmax) of Epacadostat in Part A

Cmax was the maximum observed concentration of epacadostat in plasma for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, Cmax for Cohort 1 was measured on Days 1, 5, and 12 and Cmax for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). Cmax is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
InterventionnM (Geometric Mean)
Day 1Day 5Day 12
Part A Cohort 1: Epacadostat 100 mg106011001200
Part A Cohort 1: Epacadostat 25 mg327269294

[back to top]

Area Under the Concentration-Time Curve From Zero to the Time of the Last Measurable Concentration (AUC0-t) of Epacadostat in Part A

AUC0-t was defined as the AUC from zero to the time of the last measurable concentration of epacadostat in plasma for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, AUC0-t for Cohort 1 was measured on Days 1, 5, and 12 and AUC0-t for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). AUC0-t is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
InterventionnM•hour (Geometric Mean)
Day 1Day 5Day 12
Part A Cohort 1: Epacadostat 100 mg425046704710
Part A Cohort 1: Epacadostat 25 mg85510601020

[back to top]

Number of Participants Who Experienced At Least One Adverse Event (AE)

An adverse event is defined as any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An adverse event can therefore be any unfavourable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a preexisting condition that is temporally associated with the use of the Sponsor's product, is also an adverse event. The number of participants who experienced an AE was reported for each arm. (NCT02862457)
Timeframe: Up to approximately 39.7 months

InterventionParticipants (Count of Participants)
Part A Cohort 1: Epacadostat 25 mg3
Part A Cohort 1: Epacadostat 100 mg2
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab3
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab6
Part B Cohort 1: Pembrolizumab+Cisplatin+Pemetrexed7
Part B Cohort 2: Pembrolizumab+Carboplatin+Pemetrexed6
Part B Cohort 3: Pembrolizumab+Carboplatin+Paclitaxel6

[back to top]

Number of Participants Who Discontinued Study Treatment Due to An Adverse Event (AE)

An adverse event is defined as any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An adverse event can therefore be any unfavourable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a preexisting condition that is temporally associated with the use of the Sponsor's product, is also an adverse event. The number of participants who discontinued due to an AE was reported for each arm. (NCT02862457)
Timeframe: Up to approximately 38.5 months

InterventionParticipants (Count of Participants)
Part A Cohort 1: Epacadostat 25 mg0
Part A Cohort 1: Epacadostat 100 mg1
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab0
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab1
Part B Cohort 1: Pembrolizumab+Cisplatin+Pemetrexed0
Part B Cohort 2: Pembrolizumab+Carboplatin+Pemetrexed3
Part B Cohort 3: Pembrolizumab+Carboplatin+Paclitaxel3

[back to top]

Number of Participants Experiencing Dose-Limiting Toxicities (DLTs) According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0 (NCI-CTCAE v.4.0)

A DLT was defined as the occurrence of any treatment-emergent adverse event occurring up to and including Study Day 7 for Part A Cohort 1 or Day 21 for Part A Cohort 2 and Part B. The following criteria defined DLTs: Grade (G) 4 thrombocytopenia; G4 neutropenia (despite optimal supportive care in Part B) lasting >1 week; febrile neutropenia (only if considered clinically significant in Part B); G4 toxicity; G3 laboratory abnormality lasting >1 week: G3 toxicity excluding nausea or vomiting controlled within 72 hours, rash in the absence of desquamation, no mucosal involvement, does not require systemic steroids, and resolves to G1 by the next scheduled dose of pembrolizumab or 14 days; G2 or higher episcleritis, uveitis, or iritis; unable to receive 75% of epacadostat or 1 dose of pembrolizumab during the DLT observation period because of toxicity, even if the toxicity does not meet DLT criteria; or >2 week delay in initiating Cycle 2 due to toxicity. (NCT02862457)
Timeframe: Up to Day 7 for Part A Cohort 1; up to Day 21 for Part A Cohort 2 and Part B

InterventionParticipants (Count of Participants)
Part A Cohort 1: Epacadostat 25 mg0
Part A Cohort 1: Epacadostat 100 mg0
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab0
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab1
Part B Cohort 1: Pembrolizumab+Cisplatin+Pemetrexed1
Part B Cohort 2: Pembrolizumab+Carboplatin+Pemetrexed2
Part B Cohort 3: Pembrolizumab+Carboplatin+Paclitaxel2

[back to top]

Area Under the Concentration-Time Curve From Zero to the Time of the Last Measurable Concentration (AUC0-t) of Epacadostat in Part A

AUC0-t was defined as the AUC from zero to the time of the last measurable concentration of epacadostat in plasma for epacadostat administered alone (Cohort 1) and epacadostat administered with pembrolizumab (Cohort 2). Per protocol, AUC0-t for Cohort 1 was measured on Days 1, 5, and 12 and AUC0-t for Cohort 2 was measured on Day 5 only. Blood samples were collected pre-dose and post-dose at multiple time points up to 12 days during Cycle 1 (28-day cycle). AUC0-t is presented as a geometric mean with a percent geometric coefficient of variation. (NCT02862457)
Timeframe: Cycle 1 (28-day cycle): Days 1, 5, and 12 at predose and 0.5, 1, 2, 4, 6, 8 and 10 hours postdose

,
InterventionnM•hour (Geometric Mean)
Day 5
Part A Cohort 2: Epacadostat 100 mg+Pembrolizumab3950
Part A Cohort 2: Epacadostat 25 mg+Pembrolizumab1260

[back to top]

Overall Survival (OS)

"Overall Survival (OS) is defined as the time between the date of randomization and the date of death due to any cause. OS will be censored on the last date a participant was known to be alive.~Median based on Kaplan-Meier Estimates" (NCT02864251)
Timeframe: From randomization to the date of death due to any cause (up to approximately 67 months)

InterventionMonths (Median)
Arm A: Nivolumab Plus Platinum-doublet Chemotherapy19.35
Arm B: Nivolumab Plus Ipilimumab17.12
Arm C: Platinum Doublet Chemotherapy15.90

[back to top]

Objective Response Rate (ORR) by Blinded Independent Centralized Review (BICR)

"ORR is number of randomized participants who have confirmed best overall response (BOR) of complete response (CR) or partial response (PR) using RECIST v1.1 criteria by BICR assessment.~BOR is the best response designation, between randomization and objectively documented progression per RECIST v1.1 criteria by BICR or the date of subsequent anti-cancer therapy, whichever occurs first.~PR is at least a 30% decrease in the sum of diameters of target lesions, using the baseline sum diameters as reference. CR is disappearance of all target lesions and a reduction in the short axis of pathological lymph nodes to <10 mm (whether target or non-target).~Radiographic tumor response assessments from Week 7 (± 7 days), then every 6 weeks (± 7 days) until Week 49 and every 12 weeks (± 7 days) thereafter, until disease progression, treatment discontinued, or the start of subsequent anti-cancer therapy.~CR+PR, confidence interval based on the Clopper and Pearson method." (NCT02864251)
Timeframe: From randomization to the date of objectively documented progression, date of death, or the date of subsequent therapy (up to approximately 67 months)

InterventionPercent of Participants (Number)
Arm A: Nivolumab Plus Platinum-doublet Chemotherapy30.6
Arm B: Nivolumab Plus Ipilimumab13.7
Arm C: Platinum Doublet Chemotherapy26.7

[back to top]

Duration of Response (DOR) by Blinded Independent Centralized Review (BICR)

"DOR is the time between the date of first response (CR or PR) and the date of first documented disease progression as determined by Response Evaluation Criteria In Solid Tumors (RECIST 1.1) or death due to any cause (death occurring after re-treatment or randomization to new combination treatment was not included), whichever occurred first.~PR is at least a 30% decrease in the sum of diameters of target lesions, using the baseline sum diameters as reference. CR is disappearance of all target lesions and a reduction in the short axis of pathological lymph nodes to <10 mm (whether target or non-target).~Radiographic tumor response assessments from Week 7 (± 7 days), then every 6 weeks (± 7 days) until Week 49 and every 12 weeks (± 7 days) thereafter, until disease progression, treatment discontinued, or the start of subsequent anti-cancer therapy.~Participants who neither progress nor die were censored on the date of their last assessment.~Median computed using Kaplan-Meier method" (NCT02864251)
Timeframe: From randomization to the date of first documented disease progression or death due to any cause (approximately 67 months)

InterventionMonths (Median)
Arm A: Nivolumab Plus Platinum-doublet Chemotherapy6.67
Arm B: Nivolumab Plus Ipilimumab50.04
Arm C: Platinum Doublet Chemotherapy5.55

[back to top]

12 Month Progression Free Survival Rates (PFSR) by Blinded Independent Centralized Review (BICR)

The PFSR at 12 months is defined as the percent of treated participants remaining progression free and surviving at 12 months since the first dosing date. Progression is the appearance of one or more new lesions. Point estimates are derived from Kaplan-Meier analyses. (NCT02864251)
Timeframe: 12 Months after first treatment dose

InterventionPercent of Participants (Number)
Arm A: Nivolumab Plus Platinum-doublet Chemotherapy21.2
Arm B: Nivolumab Plus Ipilimumab12.2
Arm C: Platinum Doublet Chemotherapy15.9

[back to top]

9 Month Progression Free Survival Rates (PFSR) by Blinded Independent Centralized Review (BICR)

"The PFSR at 9 months is defined as the percent of treated participants remaining progression free and surviving at 9 months since the first dosing date. Progression is the appearance of one or more new lesions.~Point estimates are derived from Kaplan-Meier analyses." (NCT02864251)
Timeframe: 9 months after first treatment dose

InterventionPercent of Participants (Number)
Arm A: Nivolumab Plus Platinum-doublet Chemotherapy25.9
Arm B: Nivolumab Plus Ipilimumab12.2
Arm C: Platinum Doublet Chemotherapy19.8

[back to top]

Progression Free Survival (PFS) by Blinded Independent Centralized Review (BICR)

"PFS is defined as the time between the date of randomization and the date of first documented tumor progression, as determined by BICR (per RECIST v1.1 criteria), or death due to any cause, whichever occurs first.~Participants who died without reported progression will be considered to have progressed on the date of their death. Subsequent therapy was accounted for by censoring at the last evaluable tumor assessment on or prior to the date of subsequent therapy.~Progression is the appearance of one or more new lesions. RECIST - response evaluation criteria in solid tumors is a standard system to measure tumor response to treatment.~Based on Kaplan-Meier estimates" (NCT02864251)
Timeframe: From randomization to the date of first documented tumor progression or death (approximately 58 months)

InterventionMonths (Median)
Arm A: Nivolumab Plus Platinum-doublet Chemotherapy5.59
Arm B: Nivolumab Plus Ipilimumab1.54
Arm C: Platinum Doublet Chemotherapy5.45

[back to top]

Progression Free Survival (PFS)

Progression Free Survival is defined as the time between the date of randomization and the date of first documented tumor progression per Blinded Independent Central Review (BICR) assessments (using adapted m-RECIST and RECIST 1.1), or death due to any cause, whichever occurs first. Participants who received subsequent anticancer therapy prior to documented progression were censored at the date of the last evaluable tumor assessment conducted on or prior to the date of initiation of the subsequent anticancer therapy. (NCT02899299)
Timeframe: Up to 40 months

InterventionMonths (Median)
Treatment A6.77
Treatment B7.20

[back to top]

Objective Response Rate (ORR) According to PD-L1 Expression Level

PD-L1 Expression is defined as the percent of tumor cells membrane staining in a minimum of 100 evaluable tumor cells per validated Dako PD-L1 immunohistochemistry (IHC) assay. This is referred to as quantifiable PD-L1 expression and efficacy is determined by objective response rate (ORR) analysis. (NCT02899299)
Timeframe: Up to 40 months

,
InterventionPercentage of Participants (Number)
<1% PD-L1≥1% PD-L1
Treatment A21.143.5
Treatment B38.544.3

[back to top]

Disease Control Rate (DCR)

Disease Control Rate is defined as the percentage of all randomized participants whose Best Overall Response was Complete Response, Partial Response, Stable Disease or Non-CR/Non-PD per adapted m-RECIST and RECIST 1.1 as assessed by Blinded Independent Central Review (BICR). (NCT02899299)
Timeframe: Up to 40 months

InterventionPercentage of Participants (Number)
Treatment A76.6
Treatment B85.1

[back to top]

Objective Response Rate (ORR)

Objective Response Rate is defined as the percentage of randomized participants who achieve a best overall response of complete response or partial response per Blinded Independent Central Review (BICR) assessments (Per adapted m-RECIST for pleural mesothelioma and RECIST 1.1, confirmation of response required). (NCT02899299)
Timeframe: Up to 40 months

InterventionPercentage of Participants (Number)
Treatment A39.6
Treatment B42.7

[back to top]

Overall Survival (OS)

Overall Survival was defined as the time from randomization to the date of death due to any cause. A participant who has not died was censored at last known date alive. (NCT02899299)
Timeframe: From randomization to the date of death (Up to 40 Months)

InterventionMonths (Median)
Treatment A18.07
Treatment B14.09

[back to top]

Progression Free Survival (PFS) According to PD-L1 Expression Level

PD-L1 Expression is defined as the percent of tumor cells membrane staining in a minimum of 100 evaluable tumor cells per validated Dako PD-L1 immunohistochemistry (IHC) assay. This is referred to as quantifiable PD-L1 expression and efficacy is determined by progression free survival (PFS) analysis. (NCT02899299)
Timeframe: Up to 40 months

,
InterventionMonths (Median)
<1% PD-L1≥1% PD-L1
Treatment A4.146.97
Treatment B8.317.06

[back to top]

Overall Survival (OS) According to PD-L1 Expression Level

PD-L1 Expression is defined as the percent of tumor cells membrane staining in a minimum of 100 evaluable tumor cells per validated Dako PD-L1 immunohistochemistry (IHC) assay. This is referred to as quantifiable PD-L1 expression and efficacy is determined by overall survival (OS) analysis. (NCT02899299)
Timeframe: Up to 40 months

,
InterventionMonths (Median)
<1% PD-L1≥1% PD-L1
Treatment A17.2818.04
Treatment B16.4913.27

[back to top]

Area Under the Concentration-time Curve From Zero Time (Predose) to the Time of the Last Measurable Concentration (AUC0-t)

(NCT02937116)
Timeframe: Cycle 1: pre-dose, post-dose at 0, 1, and 6 hours, and Days 2, 3, 8, 15, and 22, and 29

Interventionh*ug/ml (Geometric Mean)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)4800
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)12300
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)39800
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)10800

[back to top]

The Half-life (t1/2) of IBI308 in Plasma After Single Dose Administration

(NCT02937116)
Timeframe: Cycle 1: pre-dose, post-dose at 0, 1, 6 hours, and Days 2, 3, 8, 15, and 22, and 29

InterventionDays (Geometric Mean)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)17
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)12.7
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)12.5
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)16.1

[back to top]

Time to Maximum Concentration (Tmax) of Sintilimab in Solid Tumor Participants

(NCT02937116)
Timeframe: Cycle 1: pre-dose, post-dose at 0, 1, 6 hours, and Days 2, 3, 8, 15, and 22, and 29

Interventionhours (Median)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)1.05
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)2.07
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)2.27
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)1.93

[back to top]

Clearance of IBI308 in Plasma After Single Dose Administration

(NCT02937116)
Timeframe: Cycle 1: pre-dose, post-dose at 0, 1, 6 hours, and Days 2, 3, 8, 15, and 22, and 29

Interventionml/h (Geometric Mean)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)8.53
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)11.7
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)13.7
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)12.9

[back to top]

DOR According to RECIST 1.1 as Assessed by Investigator

(NCT02937116)
Timeframe: Through out the study (up to 2 years)

InterventionDays (Median)
MEL: Sintilimab 200mg Q3W (Cohort A)NA
Malignant Tumor or Neuroendocrine Tumor: Sintilimab 200mg Q3W (Cohort B)NA
NSCLC: Sintilimab 200mg Q3W (Cohort C)368.0
nsNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort D)NA
scNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort E)170.5
Gastric or Gastroesophageal Junction Adenocarcinoma: Sintilimab 200mg Q3W + Chemotherapy (Cohort F)181.0
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort G)NA
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort H)NA

[back to top]

Maximum Concentration (Cmax) of Sintilimab in Solid Tumor Participants

(NCT02937116)
Timeframe: Cycle 1: pre-dose, post-dose at 0, 1, 6 hours, and Days 2, 3, 8, 15, and 22, and 29

Interventionug/ml (Geometric Mean)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)21.9
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)69.7
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)220
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)54.6

[back to top]

TTR According to RECIST 1.1 as Assessed by Investigator

(NCT02937116)
Timeframe: Through out the study (up to 2 years)

InterventionDays (Median)
MEL: Sintilimab 200mg Q3W (Cohort A)63.0
Malignant Tumor or Neuroendocrine Tumor: Sintilimab 200mg Q3W (Cohort B)64.0
NSCLC: Sintilimab 200mg Q3W (Cohort C)63.0
nsNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort D)63.0
scNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort E)62.0
Gastric or Gastroesophageal Junction Adenocarcinoma: Sintilimab 200mg Q3W + Chemotherapy (Cohort F)63.0
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort G)62.0
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort H)62.0

[back to top]

Number of Participants Experiencing Dose-limiting Toxicities (DLTs)

(NCT02937116)
Timeframe: Up to 28 days in Cycle 1

InterventionParticipants (Number)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)0
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)0
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)0
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)0

[back to top]

Volume of Distribution of IBI308 in Plasma After Single Dose Administration

(NCT02937116)
Timeframe: Cycle 1: pre-dose, post-dose at 0, 1, 6 hours, and Days 2, 3, 8, 15, and 22, and 29

InterventionL (Geometric Mean)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)5.02
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)5.14
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)5.95
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)7.2

[back to top]

PFS According to RECIST 1.1 as Assessed by Investigator

(NCT02937116)
Timeframe: Through out the study (up to 2 years)

InterventionDays (Median)
MEL: Sintilimab 200mg Q3W (Cohort A)62.0
Malignant Tumor or Neuroendocrine Tumor: Sintilimab 200mg Q3W (Cohort B)66.0
NSCLC: Sintilimab 200mg Q3W (Cohort C)84.0
nsNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort D)377.0
scNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort E)194.0
Gastric or Gastroesophageal Junction Adenocarcinoma: Sintilimab 200mg Q3W + Chemotherapy (Cohort F)230.0
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort G)NA
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort H)NA

[back to top]

OS for Participants

(NCT02937116)
Timeframe: Through out the study

InterventionDays (Median)
MEL: Sintilimab 200mg Q3W (Cohort A)518.0
Malignant Tumor or Neuroendocrine Tumor: Sintilimab 200mg Q3W (Cohort B)342.0
NSCLC: Sintilimab 200mg Q3W (Cohort C)431.0
nsNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort D)566.0
scNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort E)461.0
Gastric or Gastroesophageal Junction Adenocarcinoma: Sintilimab 200mg Q3W + Chemotherapy (Cohort F)NA
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort G)NA
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort H)NA

[back to top]

Objective Response Rate (ORR) According to RECIST 1.1 as Assessed by Independent Review Committee by Investigator

ORR was defined as the percentage of participants in the analysis population who had achieved BOR of CR or PR according to RECIST 1.1. (NCT02937116)
Timeframe: Through out the study (up to 2 years)

Interventionpercentage of participants (Number)
MEL: Sintilimab 200mg Q3W (Cohort A)4.5
Malignant Tumor or Neuroendocrine Tumor: Sintilimab 200mg Q3W (Cohort B)14.9
NSCLC: Sintilimab 200mg Q3W (Cohort C)13.5
nsNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort D)61.9
scNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort E)55.0
Gastric or Gastroesophageal Junction Adenocarcinoma: Sintilimab 200mg Q3W + Chemotherapy (Cohort F)85.0
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort G)42.9
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort H)14.3

[back to top]

Number of All Study Participants Who Demonstrate a Tumor Response

(NCT02937116)
Timeframe: Through out the study (up to 2 years)

InterventionParticipants (Number)
Solid Tumors: Sintilimab 1mg/kg Q2W (Part A1)0
Solid Tumors: Sintilimab 3mg/kg Q2W (Part A2)1
Solid Tumors: Sintilimab 10mg/kg Q2W (Part A3)0
Solid Tumors: Sintilimab 200mg/kg Q3W (Part A4)1
MEL: Sintilimab 200mg Q3W (Cohort A)1
Malignant Tumor or Neuroendocrine Tumor: Sintilimab 200mg Q3W (Cohort B)13
NSCLC: Sintilimab 200mg Q3W (Cohort C)5
nsNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort D)13
scNSCLC: Sintilimab 200mg Q3W + Chemotherapy (Cohort E)11
Gastric or Gastroesophageal Junction Adenocarcinoma: Sintilimab 200mg Q3W + Chemotherapy (Cohort F)17
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort G)3
Neuroendocrine Tumors: Sintilimab 200mg Q3W + Chemotherapy (Cohort H)1

[back to top]

Part 3 Schedule Finding: Incidence of Dose-limiting Toxicity (DLT) During the DLT Evaluation Window

Part 3 of the study was a schedule finding phase to establish the recommended phase 2 dosing schedules for Part 4 and assess the safety and tolerability for the NKTR-214/nivolumab/ipilimumab triplet combination. The results presented are for the DLT Population. (NCT02983045)
Timeframe: Dose-limiting toxicities (DLTs) were assessed during a 3-week (21-day) DLT evaluation period beginning with the first dose of ipilimumab.

,,
InterventionParticipants (Count of Participants)
Patients with at least one eventEndocrine disorders: Adrenal insufficiencyEndocrine disorders: HyperthyroidismMetabolism and nutrition disorders: Hyponatraemia
Schedule Finding (Schedule 1): NKTR-214 + Nivolumab + Ipilimumab1001
Schedule Finding (Schedule 2): NKTR-214 + Nivolumab + Ipilimumab1100
Schedule Finding (Schedule 3): NKTR-214 + Nivolumab + Ipilimumab1010

[back to top] [back to top]

Part 1 Dose Escalation: Incidence of Dose-limiting Toxicity (DLT) During the DLT Evaluation Window

Part 1of the study was a dose-escalation phase that evaluated the safety and tolerability and defined the maximum tolerated dose or recommended Phase 2 dose of the NKTR-214/nivolumab doublet across 5 dosage/schedule levels. The results presented are for the DLT Population. (NCT02983045)
Timeframe: Includes DLTs that occurred within the DLT window of at least 21 days after the first dose of study treatment (28 days for every 2 weeks dosing; 21 days for every 3 weeks dosing). Patients were counted only once under each preferred term.

,,,,
InterventionParticipants (Count of Participants)
At least 1 DLTMetabolism and Nutrition Disorders: AcidosisMetabolism and Nutrition Disorders: HyperglycaemiaVascular Disorders: Hypotension
Dose Escalation Cohort 1: NKTR-214 (0.006 mg/kg) q3w + Nivolumab (240 mg) q2w0000
Dose Escalation Cohort 2: NKTR-214 (0.006 mg/kg) q2w + Nivolumab (240 mg) q2w0000
Dose Escalation Cohort 3: NKTR-214 (0.003 mg/kg) q2w + Nivolumab (240 mg) q2w0000
Dose Escalation Cohort 4: NKTR-214 (0.006 mg/kg) q3w + Nivolumab (360 mg) q3w0000
Dose Escalation Cohort 5: NKTR-214 (0.009 mg/kg) q3w + Nivolumab (360 mg) q3w2111

[back to top]

Major Pathologic Response (MPR) Rate

Major pathologic response (MPR) rate is defined as number of randomized participants with NCT02998528)
Timeframe: From randomization up to a median of 30 months after randomization.

InterventionParticipants (Count of Participants)
Arm B: Platinum Doublet Chemo16
Arm C: Nivo 360 mg + Platinum Doublet Chemo66

[back to top]

Event-Free Survival (EFS)

Event-free survival (EFS) is defined as the length of time from randomization to any of the following events: any progression of disease precluding surgery, progression or recurrence disease based on blinded independent central review (BICR) assessment per response evaluation criteria in solid tumors (RECIST) 1.1 after surgery, or death due to any cause. Participants who don't undergo surgery for reason other than progression will be considered to have an event at progression or death. Progression is 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 also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progression). (NCT02998528)
Timeframe: From randomization to disease progression, reoccurrence, or death due to any cause. (Up to a median of 30 months)

InterventionMonths (Median)
Arm B: Platinum Doublet Chemo20.80
Arm C: Nivo 360 mg + Platinum Doublet Chemo31.57

[back to top]

Time to Death or Distant Metastases (TTDM)

TTDM is defined as the time between the date of randomization and the first date of distant metastasis or the date of death in the absence of distant metastasis. Distant metastasis is defined as any new lesion that is outside of the thorax using blinded independent central review (BICR) according to response evaluation criteria in solid tumors (RECIST) 1.1. Patients who have not developed distant metastasis or died at the time of analysis will be censored on the date of their last evaluable tumor assessment. (NCT02998528)
Timeframe: From randomization to the first date of distant metastasis or the date of death in the absence of distant metastasis (Up to a median of 30 months)

InterventionMonths (Median)
Arm B: Platinum Doublet Chemo26.71
Arm C: Nivo 360 mg + Platinum Doublet ChemoNA

[back to top]

Pathologic Complete Response (pCR) Rate

Pathologic complete response (pCR) rate is defined as the number of randomized participants with absence of residual tumor in lung and lymph nodes as evaluated by blinded independent pathological review (BIPR). (NCT02998528)
Timeframe: From randomization up to a median of 30 months after randomization.

InterventionParticipants (Count of Participants)
Arm B: Platinum Doublet Chemo4
Arm C: Nivo 360 mg + Platinum Doublet Chemo43

[back to top]

OS at 24 Months in PD-L1 TC >= 50% LREM Analysis Set

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 24 months were defined as the Kaplan-Meier estimate of OS at 24 months. (NCT03003962)
Timeframe: From date of randomization till 24 months

Interventionpercentage of participants (Number)
Durvalumab36.9
Platinum-based SoC32.6

[back to top]

Alive and Progression-Free at 12 Months (APF12)

The APF12 was defined as the Kaplan-Meier estimate of percentage of participants alive and progression free at 12 months based on PFS (per RECIST 1.1 as assessed using Investigator assessments) analysis. (NCT03003962)
Timeframe: From date of randomization until 12 months

Interventionpercentage of participants (Number)
Durvalumab25.5
Platinum-based SoC13.3

[back to top]

DoR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >=50% LREM Analysis Set

DoR (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (i.e. date of PFS event or censoring - date of first response + 1). DoR was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab12.2
Platinum-based SoC4.2

[back to top]

DoR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >=50% Analysis Set

DoR (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (i.e. date of PFS event or censoring - date of first response + 1). DoR was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab12.2
Platinum-based SoC4.2

[back to top]

DoR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >=25% LREM Analysis Set

DoR (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (i.e. date of PFS event or censoring - date of first response + 1). DoR was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab11.6
Platinum-based SoC4.2

[back to top]

APF12 in PD-L1 TC >= 50% LREM Analysis Set

The APF12 was defined as the Kaplan-Meier estimate of percentage of participants alive and progression free at 12 months based on PFS (per RECIST 1.1 as assessed using Investigator assessments) analysis. (NCT03003962)
Timeframe: From date of randomization until 12 months

Interventionpercentage of participants (Number)
Durvalumab26.5
Platinum-based SoC14.5

[back to top]

APF12 in PD-L1 TC >= 50% Analysis Set

The APF12 was defined as the Kaplan-Meier estimate of percentage of participants alive and progression free at 12 months based on PFS (per RECIST 1.1 as assessed using Investigator assessments) analysis. (NCT03003962)
Timeframe: From date of randomization until 12 months

Interventionpercentage of participants (Number)
Durvalumab26.1
Platinum-based SoC11.7

[back to top]

APF12 in PD-L1 TC >= 25% LREM Analysis Set

The APF12 was defined as the Kaplan-Meier estimate of percentage of participants alive and progression free at 12 months based on PFS (per RECIST 1.1 as assessed using Investigator assessments) analysis. (NCT03003962)
Timeframe: From date of randomization until 12 months

Interventionpercentage of participants (Number)
Durvalumab24.1
Platinum-based SoC16.4

[back to top]

Time to Deterioration of EORTC QLQ-LC13 in PD-L1 TC >= 25% LREM Analysis Set

Time to symptom deterioration was defined as the time from the date of randomization until the date if the first clinically meaningful symptom deterioration a decrease in the function scales or the global health status/ HRQoL from baseline of ≥10) that was confirmed at a subsequent visit or death (by any cause) in the absence of a clinically meaningful symptom deterioration, regardless of whether the participant withdraws from the study treatment or received another anticancer therapy prior to symptom deterioration. The QLQ-LC13 is a lung cancer specific module from the EORTC for lung cancer that comprised of 13 questions to assess lung cancer symptoms (cough, hemoptysis, dyspnea, and site-specific pain), treatment-related side-effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication. (NCT03003962)
Timeframe: From randomization until date of first symptom deterioration that is confirmed, assessed up to maximum of approximately 69 months (DCO 27 October 2022)

,
Interventionmonths (Median)
CoughDyspneaChest pain
Durvalumab9.23.69.8
Platinum-based SoC8.23.66.6

[back to top]

Time to Deterioration of EORTC QLQ-LC13

Time to symptom deterioration was defined as the time from the date of randomization until the date if the first clinically meaningful symptom deterioration a decrease in the function scales or the global health status/ HRQoL from baseline of ≥10) that was confirmed at a subsequent visit or death (by any cause) in the absence of a clinically meaningful symptom deterioration, regardless of whether the participant withdraws from the study treatment or received another anticancer therapy prior to symptom deterioration. The QLQ-LC13 is a lung cancer specific module from the EORTC for lung cancer that comprised of 13 questions to assess lung cancer symptoms (cough, hemoptysis, dyspnea, and site-specific pain), treatment-related side-effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication. (NCT03003962)
Timeframe: From randomization until date of first symptom deterioration that is confirmed, assessed up to maximum of approximately 69 months (DCO 27 October 2022)

,
Interventionmonths (Median)
CoughDyspneaChest pain
Durvalumab7.52.89.0
Platinum-based SoC6.63.66.4

[back to top]

Time to Deterioration of EORTC QLQ-C30 in PD-L1 TC >= 25% LREM Analysis Set

Time to symptom deterioration was defined as the time from the date of randomization until the date if the first clinically meaningful deterioration a decrease in the function scales or the global health status/ HRQoL from baseline of ≥10) that was confirmed at a subsequent visit or death (by any cause) in the absence of a clinically meaningful symptom deterioration, regardless of whether the participant withdraws from the study treatment or received another anticancer therapy prior to symptom deterioration. The EORTC QLQ-C30 consisted of 30 questions that were combined to produce 5 functional scales (physical, role, cognitive, emotional, and social), 3 symptom scales (fatigue, pain, and nausea/vomiting), 5 individual items (dyspnea, insomnia, appetite loss, constipation, and diarrhea), and a global measure of health status. The EORTC QLQ-C30 was scored according to the EORTC QLQ-C30 scoring manual. (NCT03003962)
Timeframe: From randomization until date of first symptom deterioration that is confirmed, assessed up to a maximum of approximately 69 months (DCO 27 October 2022)

,
Interventionmonths (Median)
Global health statusPhysical functioningAppetite lossFatigue
Durvalumab7.47.49.35.5
Platinum-based SoC5.54.73.71.8

[back to top]

Time to Deterioration of EORTC QLQ-C30

Time to symptom deterioration was defined as the time from the date of randomization until the date if the first clinically meaningful deterioration (a decrease in the function scales or the global health status/ health-related quality of life [HRQoL] from baseline of ≥10) that was confirmed at a subsequent visit or death in the absence of a clinically meaningful symptom deterioration, regardless of whether the participant withdraws from the study treatment or received another anticancer therapy prior to symptom deterioration. The EORTC QLQ-C30 consisted of 30 questions that were combined to produce 5 functional scales (physical, role, cognitive, emotional, and social), 3 symptom scales (fatigue, pain, and nausea/vomiting), 5 individual items (dyspnea, insomnia, appetite loss, constipation, and diarrhea), and a global measure of health status. The EORTC QLQ-C30 was scored according to the EORTC QLQ-C30 scoring manual. (NCT03003962)
Timeframe: From randomization until date of first symptom deterioration that is confirmed, assessed up to a maximum of approximately 69 months (DCO 27 October 2022)

,
Interventionmonths (Median)
Global health statusPhysical functioningAppetite lossFatigue
Durvalumab7.37.49.24.9
Platinum-based SoC3.83.83.61.8

[back to top]

Percentage of Participants With Antidrug Antibody (ADA) Response to Durvalumab

Treatment-emergent ADA positive was defined as either treatment-induced or treatment-boosted ADA. Treatment-boosted ADA was defined as a baseline positive ADA titer that was boosted to a 4-fold or higher-level following study drug administration. Persistently positive was defined as positive at least 2 post-baseline assessments with at least 16 weeks between the first and last positive assessment or positive at last post-baseline assessment. Transiently positive was defined as having at least 1 post-baseline ADA positive assessment and not fulfilling the conditions of persistently positive. (NCT03003962)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Treatment-emergent ADA positiveTreatment-boosted ADAPersistently positiveTransiently positive
Durvalumab0.80.400.8

[back to top]

Percentage of Participants With ADA Response to Durvalumab in LREM Analysis Set

Treatment-emergent ADA positive was defined as either treatment-induced or treatment-boosted ADA. Treatment-boosted ADA was defined as a baseline positive ADA titer that was boosted to a 4-fold or higher-level following study drug administration. Persistently positive was defined as positive at least 2 post-baseline assessments with at least 16 weeks between the first and last positive assessment or positive at last post-baseline assessment. Transiently positive was defined as having at least 1 post-baseline ADA positive assessment and not fulfilling the conditions of persistently positive. (NCT03003962)
Timeframe: Up to 24 weeks

Interventionpercentage of participants (Number)
Treatment-emergent ADA positiveTreatment-boosted ADAPersistently positiveTransiently positive
Durvalumab1.00.501.0

[back to top]

Number of Participants With ECOG Performance Status in PD-L1 TC >=25% LREM Analysis Set

ECOG performance status was assessed at the times specified based on the following and scored as 0: fully active: able to carry out all usual activities without restrictions. 1: Restricted in strenuous activity, but ambulatory and able to carry out any work activities; up and about more than 50% of waking hours. 3: Capable of only limited self-care; confined to bed or chair more than 50% of waking hours. 4: Completely disabled; unable to carry out any self-care and totally confined to bed or chair and 5: death. Data for only participants with restricted activity has been reported. (NCT03003962)
Timeframe: From Baseline and until follow-up period of 57 months

,
Interventionnumber of participants (Number)
Restricted activity, BaselineRestricted activity, Follow-up Month 57
Durvalumab2210
Platinum-based SoC2032

[back to top]

Number of Participants With Eastern Cooperative Oncology Group (ECOG) Performance Status

ECOG performance status was assessed at the times specified based on the following and scored as 0: fully active: able to carry out all usual activities without restrictions. 1: Restricted in strenuous activity, but ambulatory and able to carry out any work activities; up and about more than 50% of waking hours. 3: Capable of only limited self-care; confined to bed or chair more than 50% of waking hours. 4: Completely disabled; unable to carry out any self-care and totally confined to bed or chair and 5: death. Data for only participants with restricted activity has been reported. (NCT03003962)
Timeframe: From Baseline and until follow-up period of 57 months

,
Interventionnumber of participants (Number)
Restricted activity, BaselineRestricted activity, Follow-up Month 57
Durvalumab2670
Platinum-based SoC2552

[back to top]

Change From Baseline in European Organization for Research and Treatment of Cancer (EORTC) 30-Item Core Quality of Life Questionnaire Version 3 (QLQ-C30)

The EORTC QLQ-C30 consisted of 30 questions that were combined to produce 5 functional scales (physical, role, cognitive, emotional, and social), 3 symptom scales (fatigue, pain, and nausea/vomiting), 5 individual items (dyspnea, insomnia, appetite loss, constipation, and diarrhea), and a global measure of health status. The EORTC QLQ-C30 was scored according to the EORTC QLQ-C30 scoring manual. An outcome variable consisted of a score from 0 to 100. Higher scores on the global health status and functioning scales indicate better health status/function, but higher scores on symptom scales/items represent greater symptom severity. Baseline was defined as the last non-missing assessment prior to randomization. The mixed model repeated measures (MMRM) analysis of EORTC QLQ-C30 considered all data from baseline to PD or 12 months. (NCT03003962)
Timeframe: Baseline and 12 months

,
Interventionscores on a scale (Mean)
Global health statusPhysical functioningFatigueAppetite loss
Durvalumab-0.7-3.31.4-1.1
Platinum-based SoC-7.4-6.17.29.1

[back to top]

Change From Baseline in EORTC QLQ-LC13 in PD-L1 TC >= 25% LREM Analysis Set

The QLQ-LC13 is a lung cancer specific module from the EORTC for lung cancer that comprised of 13 questions to assess lung cancer symptoms (cough, hemoptysis, dyspnea, and site-specific pain), treatment-related side-effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication. An outcome variable consisted of a score from 0 to 100. Higher scores on symptom scales represent greater symptom severity. Baseline was defined as the last non-missing assessment prior to randomization. The MMRM analysis of EORTC QLQ-LC13 considered all data from baseline to PD or 12 months. (NCT03003962)
Timeframe: Baseline and 12 months

,
Interventionscores on a scale (Mean)
CoughDyspneaChest pain
Durvalumab-6.62.9-0.7
Platinum-based SoC-8.63.9-1.0

[back to top]

Change From Baseline in EORTC QLQ-C30 in PD-L1 TC >= 25% LREM Analysis Set

The EORTC QLQ-C30 consisted of 30 questions that were combined to produce 5 functional scales (physical, role, cognitive, emotional, and social), 3 symptom scales (fatigue, pain, and nausea/vomiting), 5 individual items (dyspnea, insomnia, appetite loss, constipation, and diarrhea), and a global measure of health status. The EORTC QLQ-C30 was scored according to the EORTC QLQ-C30 scoring manual. An outcome variable consisted of a score from 0 to 100. Higher scores on the global health status and functioning scales indicate better health status/function, but higher scores on symptom scales/items represent greater symptom severity. Baseline was defined as the last non-missing assessment prior to randomization. The MMRM analysis of EORTC QLQ-C30 considered all data from baseline to PD or 12 months. (NCT03003962)
Timeframe: Baseline and 12 months

,
Interventionscores on a scale (Mean)
Global health statusPhysical functioningFatigueAppetite loss
Durvalumab-1.4-3.92.2-0.2
Platinum-based SoC-7.3-6.07.79.8

[back to top]

Change From Baseline in EORTC 13-Item Lung Cancer Quality of Life Questionnaire (QLQ-LC13)

The QLQ-LC13 is a lung cancer specific module from the EORTC for lung cancer that comprised of 13 questions to assess lung cancer symptoms (cough, hemoptysis, dyspnea, and site-specific pain), treatment-related side-effects (sore mouth, dysphagia, peripheral neuropathy, and alopecia), and pain medication. An outcome variable consisted of a score from 0 to 100. Higher scores on symptom scales represent greater symptom severity. Baseline was defined as the last non-missing assessment prior to randomization. The MMRM analysis of EORTC QLQ-LC13 considered all data from baseline to PD or 12 months. (NCT03003962)
Timeframe: Baseline and 12 months

,
Interventionscores on a scale (Mean)
CoughDyspneaChest pain
Durvalumab-6.42.4-1.0
Platinum-based SoC-7.94.1-0.2

[back to top]

Time From Randomization to Second Progression (PFS2)

PFS2 was defined as the time from the date of randomization to the earliest of the progression events defined according to local clinical practice (subsequent to that used for the primary variable PFS) or death (i.e., date of PFS2 event or censoring - date of randomization + 1). PFS2 was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) until confirmed objective disease progression. Disease then assessed as per local practice until 2nd progression or death (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab11.3
Platinum-based SoC9.3

[back to top]

Progression Free Survival (PFS) Based on Investigator Assessment According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1)

The PFS (per RECIST 1.1) was defined as the time from the date of randomization until the date of objective disease progression or death regardless of whether the participants withdrew from randomized therapy or received another anticancer therapy prior to progression (i.e., date of PFS event or censoring - date of randomization +1). Progression of disease per RECIST 1.1, when either 1 of the criteria met: Target lesion (TL): at least a 20% increase in the sum of diameters of TLs, for reference the smallest sum on study. In addition, the sum must also demonstrate an absolute increase of at least 5 millimeters (mm). Non-target lesion (NTL): Unequivocal progression of existing NTLs. It may be due to an important progression in 1 lesion only or in several lesions. In all cases the progression must be clinically significant for the physician to consider changing (or stopping) therapy. New lesions: the presence of 1 or more new lesions was assessed as progression. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months DCO 27 October 2022)

Interventionmonths (Median)
Durvalumab5.4
Platinum-based SoC4.8

[back to top]

Duration of Response (DoR) as Per RECIST 1.1 Using Investigator Assessment

DoR (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (i.e. date of PFS event or censoring - date of first response + 1). DoR was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab11.9
Platinum-based SoC4.2

[back to top]

Objective Response Rate (ORR) as Per RECIST 1.1 Using Investigator Assessment

ORR (per RECIST 1.1 using Investigator assessments) was defined as the percentage of participants with an unconfirmed response of complete response (CR) or partial response (PR). CR was defined as disappearance of all target lesions (TLs). Any pathological lymph nodes selected as TLs had a reduction in short axis to < 10 millimeter (mm). PR was defined as at least a 30% decrease in the sum of diameters of TLs, with reference the baseline sum of diameters if criteria for PD are not met. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionpercentage of participants (Number)
Durvalumab37.6
Platinum-based SoC37.4

[back to top]

ORR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >= 25% LREM Analysis Set

ORR (per RECIST 1.1 using Investigator assessments)was defined as the percentage of participants with an unconfirmed response of CR or PR. CR was defined as disappearance of all TLs. Any pathological lymph nodes selected as TLs had a reduction in short axis to < 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of TLs, with reference the baseline sum of diameters if criteria for PD are not met. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionpercentage of participants (Number)
Durvalumab38.5
Platinum-based SoC40.2

[back to top]

ORR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >= 50% Analysis Set

ORR (per RECIST 1.1 using Investigator assessments) was defined as the percentage of participants with an unconfirmed response of CR or PR. CR was defined as disappearance of all TLs. Any pathological lymph nodes selected as TLs had a reduction in short axis to < 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of TLs, with reference the baseline sum of diameters if criteria for PD are not met. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionpercentage of participants (Number)
Durvalumab42.1
Platinum-based SoC40.7

[back to top]

ORR as Per RECIST 1.1 Using Investigator Assessment in PD-L1 TC >= 50% LREM Analysis Set

ORR (per RECIST 1.1 using Investigator assessments) was defined as the percentage of participants with an unconfirmed response of CR or PR. CR was defined as disappearance of all TLs. Any pathological lymph nodes selected as TLs had a reduction in short axis to < 10 mm. PR was defined as at least a 30% decrease in the sum of diameters of TLs, with reference the baseline sum of diameters if criteria for PD are not met. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionpercentage of participants (Number)
Durvalumab44.0
Platinum-based SoC43.7

[back to top]

OS at 18 Months

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 18 months were defined as the Kaplan-Meier estimate of OS at 18 months. (NCT03003962)
Timeframe: From date of randomization till 18 months.

Interventionpercentage of participants (Number)
Durvalumab42.5
Platinum-based SoC34.2

[back to top]

OS at 18 Months in PD-L1 TC > = 25% LREM Analysis Set

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 18 months were defined as the Kaplan-Meier estimate of OS at 18 months. (NCT03003962)
Timeframe: From date of randomization till 18 months

Interventionpercentage of participants (Number)
Durvalumab43.0
Platinum-based SoC41.4

[back to top]

OS at 18 Months in PD-L1 TC >= 50% Analysis Set

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 18 months were defined as the Kaplan-Meier estimate of OS at 18 months. (NCT03003962)
Timeframe: From date of randomization till 18 months

Interventionpercentage of participants (Number)
Durvalumab43.2
Platinum-based SoC34.9

[back to top]

OS at 18 Months in PD-L1 TC >= 50% LREM Analysis Set

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 18 months were defined as the Kaplan-Meier estimate of OS at 18 months. (NCT03003962)
Timeframe: From date of randomization till 18 months

Interventionpercentage of participants (Number)
Durvalumab44.3
Platinum-based SoC42.2

[back to top]

OS at 24 Months

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 24 months were defined as the Kaplan-Meier estimate of OS at 24 months. (NCT03003962)
Timeframe: From date of randomization till 24 months

Interventionpercentage of participants (Number)
Durvalumab34.6
Platinum-based SoC27.2

[back to top]

OS at 24 Months in PD-L1 TC > = 25% LREM Analysis Set

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 24 months were defined as the Kaplan-Meier estimate of OS at 24 months. (NCT03003962)
Timeframe: From date of randomization till 24 months

Interventionpercentage of participants (Number)
Durvalumab34.7
Platinum-based SoC32.8

[back to top]

OS at 24 Months in PD-L1 TC >= 50% Analysis Set

OS was defined as the time from the date of randomization until death due to any cause. Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. The percentage of participants alive at 24 months were defined as the Kaplan-Meier estimate of OS at 24 months. (NCT03003962)
Timeframe: From date of randomization till 24 months

Interventionpercentage of participants (Number)
Durvalumab37.0
Platinum-based SoC27.0

[back to top]

PFS2 in PD-L1 TC >= 50% LREM Analysis Set

PFS2 was defined as the time from the date of randomization to the earliest of the progression events defined according to local clinical practice (subsequent to that used for the primary variable PFS) or death (i.e., date of PFS2 event or censoring - date of randomization + 1). PFS2 was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) until confirmed objective disease progression. Disease then assessed as per local practice until 2nd progression or death (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab12.0
Platinum-based SoC10.9

[back to top]

PFS2 in PD-L1 TC >= 50% Analysis Set

PFS2 was defined as the time from the date of randomization to the earliest of the progression events defined according to local clinical practice (subsequent to that used for the primary variable PFS) or death (i.e., date of PFS2 event or censoring - date of randomization + 1). PFS2 was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) until confirmed objective disease progression. Disease then assessed as per local practice until 2nd progression or death (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab11.3
Platinum-based SoC8.8

[back to top]

PFS2 in PD-L1 TC >= 25% LREM Analysis Set

PFS2 was defined as the time from the date of randomization to the earliest of the progression events defined according to local clinical practice (subsequent to that used for the primary variable PFS) or death (i.e., date of PFS2 event or censoring - date of randomization + 1). PFS2 was calculated using the Kaplan-Meier technique. (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) until confirmed objective disease progression. Disease then assessed as per local practice until 2nd progression or death (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab11.8
Platinum-based SoC10.9

[back to top]

PFS Based on Investigator Assessment According to RECIST 1.1 in PD-L1 TC >= 50% LREM Analysis Set

The PFS (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of randomization until the date of objective disease progression or death regardless of whether the participants withdrew from randomized therapy or received another anticancer therapy prior to progression (i.e., date of PFS event or censoring - date of randomization +1). (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab5.7
Platinum-based SoC5.5

[back to top]

PFS Based on Investigator Assessment According to RECIST 1.1 in PD-L1 TC >= 50% Analysis Set

The PFS (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of randomization until the date of objective disease progression or death regardless of whether the participants withdrew from randomized therapy or received another anticancer therapy prior to progression (i.e., date of PFS event or censoring - date of randomization +1). (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab5.6
Platinum-based SoC4.5

[back to top]

PFS Based on Investigator Assessment According to RECIST 1.1 in LREM Analysis Set

The PFS (per RECIST 1.1 using Investigator assessments) was defined as the time from the date of randomization until the date of objective disease progression or death regardless of whether the participants withdrew from randomized therapy or received another anticancer therapy prior to progression (i.e., date of PFS event or censoring - date of randomization +1). (NCT03003962)
Timeframe: Tumor assessments (per RECIST 1.1) every 6 weeks for the first 48 weeks relative to the date of randomization and then every 8 weeks thereafter until confirmed objective disease progression (up to a maximum of approximately 69 months)

Interventionmonths (Median)
Durvalumab5.5
Platinum-based SoC5.6

[back to top]

Overall Survival (OS)

OS is defined as the time from the date of randomization until death due to any cause (date of death or censoring-date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. (NCT03003962)
Timeframe: From date of randomization until death due to any cause. Assessed up to a maximum of approximately 69 months [data cut-off (DCO) 27 October 2022]

Interventionmonths (Median)
Durvalumab14.6
Platinum-based SoC12.8

[back to top]

OS in PD-L1 TC >= 50% LREM Analysis Set

OS is defined as the time from the date of randomization until death due to ay cause (date of death or censoring-date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. (NCT03003962)
Timeframe: From date of randomization until death due to any cause. Assessed up to a maximum of approximately 69 months (DCO 27 October 2022)

Interventionmonths (Median)
Durvalumab14.9
Platinum-based SoC14.9

[back to top]

OS in PD-L1 TC >= 50% Analysis Set

OS is defined as the time from the date of randomization until death due to any cause (date of death or censoring-date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. (NCT03003962)
Timeframe: From date of randomization until death due to any cause. Assessed up to a maximum of approximately 69 months (DCO 27 October 2022)

Interventionmonths (Median)
Durvalumab14.6
Platinum-based SoC11.8

[back to top]

OS in Participants With LREM

OS is defined as the time from the date of randomization until death due to any cause (date of death or censoring-date of randomization + 1). Any participant not known to have died at the time of analysis was censored based on the last recorded date on which the participant was known to be alive. (NCT03003962)
Timeframe: From date of randomization until death due to any cause. Assessed up to a maximum of approximately 69 months (DCO 27 October 2022)

Interventionmonths (Median)
Durvalumab14.6
Platinum-based SoC15.0

[back to top]

Number of Participants With Grade 3 or Grade 4 Adverse Events

Assess toxicity of Nivolumab plus single agent chemotherapy compared with single agent chemotherapy alone. Number of grade 3 and 4 toxicities as defined by the NCI Common Terminology Criteria for Adverse Events (NCI CTCAE) v4. (NCT03041181)
Timeframe: 6 months

,
Interventionparticipants (Number)
GR4 Thromboembolic EventGR3 Adverse Events
Arm A00
Arm B10

[back to top]

Phases 1 and 2: Objective Response Rate (ORR)

ORR was defined as the percentage of participants having a complete response (CR) or partial response (PR) as determined by investigator assessment of radiographic disease per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. (NCT03085914)
Timeframe: Up to Week 18

,,,,,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial Response
Group A: Epa + Pembrolizumab + mFOLFOX605
Group B: Epa + Pembrolizumab + Nab-Paclitaxel and Gemcitabine12
Group C: Epa + Pembrolizumab + Paclitaxel and Carboplatin03
Group D: Epa + Pembrolizumab + Pemetrexed and Platinum Agent02
Group E: Epa + Pembrolizumab + Cyclophosphamide03
Group F: Epa + Pembrolizumab + Gemcitabine and Platinum Agent01
Group G: Epa + Pembrolizumab + 5-FU and Platinum Agent05

[back to top]

Phases 1 & 2: Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious TEAEs

A TEAE is any AE either reported for the first time or worsening of a pre-existing event after first dose of epacadostat, pembrolizumab, or chemotherapy. Serious adverse event is defined as an event that meets 1 of the following criteria: is fatal or life threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability, incapacity, or a substantial disruption of a person's ability to conduct normal life functions, constitutes a congenital anomaly or birth defect,is a medically important event that may jeopardize the participant or may require medical or surgical intervention to prevent 1 of the outcomes listed above. (NCT03085914)
Timeframe: Up to 21 months

,,,,,,
InterventionParticipants (Count of Participants)
TEAESerious TEAE
Group A: Epa + Pembrolizumab + mFOLFOX695
Group B: Epa + Pembrolizumab + Nab-Paclitaxel and Gemcitabine95
Group C: Epa + Pembrolizumab + Paclitaxel and Carboplatin113
Group D: Epa + Pembrolizumab + Pemetrexed and Platinum Agent96
Group E: Epa + Pembrolizumab + Cyclophosphamide134
Group F: Epa + Pembrolizumab + Gemcitabine and Platinum Agent86
Group G: Epa + Pembrolizumab + 5-FU and Platinum Agent115

[back to top]

Phases 1 and 2: Number of Participants With Dose Limiting Toxicities (DLTs)

A DLT was defined as the occurrence of any of the protocol-specified toxicities occurring up to and including Day 28 for the cohorts where mFOLFOX6 and nab-paclitaxel/gemcitabine are administered and Day 21 for all other chemotherapy regimens in Phase 1, except those with a clear alternative explanation (eg, disease progression) or transient (≤ 72 hours) abnormal laboratory values without associated clinically significant signs or symptoms based on investigator determination. (NCT03085914)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
Group A: Epa + Pembrolizumab + mFOLFOX62
Group B: Epa + Pembrolizumab + Nab-Paclitaxel and Gemcitabine0
Group C: Epa + Pembrolizumab + Paclitaxel and Carboplatin0
Group D: Epa + Pembrolizumab + Pemetrexed and Platinum Agent0
Group E: Epa + Pembrolizumab + Cyclophosphamide0
Group F: Epa + Pembrolizumab + Gemcitabine and Platinum Agent3
Group G: Epa + Pembrolizumab + 5-FU and Platinum Agent0

[back to top]

Objective Response Rate (ORR) Per Blinded Independent Central Review (BICR) by RECIST 1.1 of NKTR-214 Plus Pembrolizumab for Dose Expansion Cohorts 2 and 3.

"ORR per BICR by RECIST 1.1 for the Response Evaluable Population dose expansion Cohorts 2 and 3.~ORR is defined as the proportion of enrolled participants who achieved a Best Overall Response (BOR) of CR or PR. CR is defined as disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) had to have reduction in short axis to < 10 mm. PR is defined as at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. ORR is calculated as the sum of CR and PR.~The Response Evaluable Population was subjects who received at least 1 dose (or partial dose) of study drug, had measurable disease (per RECIST 1.1) at baseline, and had at least 1 post-baseline assessment of tumor response." (NCT03138889)
Timeframe: Until disease progression, death, unacceptable toxicity, symptomatic deterioration, Investigator's decision to discontinue treatment, patient withdrew consent or lost to follow-up, or study terminated by Sponsor; or until maximum of 2 years.

InterventionParticipants (Count of Participants)
Dose Expansion Cohort 2: NKTR-214 (0.006 mg/kg) + Pembro (200 mg)13
Dose Expansion Cohort 3: NKTR-214 (0.010 mg/kg) + Pembro (200 mg)2

[back to top]

Objective Response Rate (ORR) Per Investigator's Assessment by RECIST 1.1 of NKTR-214 at a Dose of 0.006 mg/kg With Pembrolizumab and Platinum-based Chemotherapy for Dose Expansion Cohorts 4+5.

"ORR per Investigator's Assessment* by RECIST 1.1 for the Response Evaluable Population dose expansion Cohorts 4 +5. The Response Evaluable Population was subjects who received at least 1 dose (or partial dose) of study drug, had measurable disease (per RECIST 1.1) at baseline, and had at least 1 post-baseline assessment of tumor response.~Objective response is the sum of confirmed complete response and confirmed partial response.~*Efficacy endpoint for Cohort 4 +5 is per Investigator's Assessment due to the early termination of the study and incompleteness of BICR data for these cohorts." (NCT03138889)
Timeframe: Until disease progression, death, unacceptable toxicity, symptomatic deterioration, Investigator's decision to discont. treatment, patient withdrew consent or lost to follow-up, or study terminated by Sponsor; or until maximum of 2 years.

InterventionParticipants (Count of Participants)
Dose Expansion Cohorts 4/52

[back to top]

Number of Participants Experiencing Dose-Limiting Toxicities in Dose Optimization Cohort 1a

"DLTs were assesses in the Dose Optimization Cohort 1 a, which had doses of NKTR-214 as 0.008 mg/kg, 0.010 mg/kg, and 0.012 m/kg, I combination with pembrolizumab at 200 mg.~A single DLT (hypotension) was reported in 1 patient in dose optimization Cohort 1a." (NCT03138889)
Timeframe: DLTs were assessed at 21 days from Cycle 1

,,
InterventionParticipants (Count of Participants)
At least 1 DLTVascular Disorders: Hypotension
Dose Optimization Cohort 1a: NKTR-214 (0.008 mg/kg) + Pembro (200 mg)00
Dose Optimization Cohort 1a: NKTR-214 (0.010 mg/kg) + Pembro (200 mg)11
Dose Optimization Cohort 1a: NKTR-214 (0.012 mg/kg) + Pembro (200 mg)00

[back to top]

Number of Participants With Treatment-Emergent Adverse Events [Safety and Tolerability] for Dose Optimization Cohort 1a.

Safety and Tolerability of NKTR-214 (starting at dose of 0.008 mg/kg) in combination with pembrolizumab (Keytruda®) as evaluated by incidence of drug-emergent Adverse Events (AEs), Serious Adverse Events (SAEs), AEs leading to drug discontinuation, and fatal AEs. (NCT03138889)
Timeframe: AEs reported starting immediately after first dose of study drug(s) until 100 days after the last dose of all study drugs, up to approximately 28 months.

,,,
InterventionParticipants (Count of Participants)
Subjects Reporting at Least One TEAESubjects Reporting at Least One Serious TEAESubjects Reporting at Least One TEAE Leading to DeathSubjects Reporting at Least One TEAE Leading to Drug Discontinuation
NKTR-214 (0.008 mg/kg) + Pembro (200 mg)4202
NKTR-214 (0.010 mg/kg) + Pembro (200 mg)7403
NKTR-214 (0.012 mg/kg) + Pembro (200 mg)7201
Total18806

[back to top]

PK of Tremelimumab; Peak and Trough Serum Concentrations

To evaluate PK, blood samples were collected at pre-specified timepoints and peak and trough serum concentrations of tremelimumab were determined. Peak concentration on Week 0 is the post-infusion concentration of Week 0 (collected within 10 minutes of the end of infusion). Trough concentrations on Weeks 3 and 12 are the pre-infusion concentrations of Weeks 3 and 12, respectively. (NCT03164616)
Timeframe: Samples were collected post-dose on Day 1 (Week 0), pre-dose on Weeks 3 and 12 and at follow-up (3 months after the last valid dose). Assessed at the global cohort DCO of 12 March 2021.

Interventionμg/mL (Geometric Mean)
Week 0Week 3Week 12Follow-up (3 months)
T + D + SoC23.174.167.820.86

[back to top]

Pharmacokinetics (PK) of Durvalumab; Peak and Trough Serum Concentrations

To evaluate PK, blood samples were collected at pre-specified timepoints and peak and trough serum concentrations of durvalumab were determined. Peak concentration on Week 0 is the post-infusion concentration of Week 0 (collected within 10 minutes of the end of infusion). Trough concentrations on Weeks 3 and 12 are the pre-infusion concentrations of Weeks 3 and 12, respectively. (NCT03164616)
Timeframe: Samples were collected post-dose on Day 1 (Week 0), pre-dose on Weeks 3 and 12 and at follow-up (3 months after the last valid dose). Assessed at the global cohort DCO of 12 March 2021.

,
Interventionmicrograms/milliliter (μg/mL) (Geometric Mean)
Week 0Week 3Week 12Follow-up (3 months)
D + SoC505.0191.53212.1116.06
T + D + SoC418.8082.08195.6213.42

[back to top]

Number of Patients With Anti-Drug Antibody (ADA) Response to Durvalumab

Blood samples were collected at pre-specified timepoints and number of patients who developed detectable ADAs against durvalumab was determined. ADA prevalence is defined as percentage of patients with positive ADA result at any time, baseline or post-baseline. Treatment-emergent ADA is defined as either treatment-induced ADA or treatment-boosted ADA. ADA incidence is percentage of patients who were treatment-emergent ADA-positive. Treatment-boosted ADA is defined as baseline positive ADA titer that was boosted by ≥4-fold during the study period. Persistently positive is defined as having ≥2 post-baseline ADA positive measurements with ≥16 weeks (112 days) between the first and last positive, or an ADA positive result at the last available assessment. Transiently positive is defined as having ≥1 post-baseline ADA positive measurement and not fulfilling the conditions for persistently positive. Presence of neutralizing antibody (nAb) was tested for all ADA positive samples. (NCT03164616)
Timeframe: Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, durvalumab).

,
InterventionParticipants (Count of Participants)
ADA positive at any visit (ADA prevalence)Treatment-emergent ADA positive (ADA incidence)Treatment-boosted ADATreatment-induced ADA (ADA positive post-baseline only)ADA positive at baseline onlyADA positive post-baseline and positive at baselinePersistently positiveTransiently positivenAb positive at any visit
D + SoC33191181327133
T + D + SoC4229227878263

[back to top]

Number of Patients With ADA Response to Tremelimumab

Blood samples were collected at pre-specified timepoints and number of patients who developed detectable ADAs against tremelimumab was determined. ADA prevalence is defined as percentage of patients with positive ADA result at any time, baseline or post-baseline. Treatment-emergent ADA is defined as either treatment-induced ADA or treatment-boosted ADA. ADA incidence is percentage of patients who were treatment-emergent ADA-positive. Treatment-boosted ADA is defined as baseline positive ADA titer that was boosted by ≥4-fold during the study period. Persistently positive is defined as having ≥2 post-baseline ADA positive measurements with ≥16 weeks (112 days) between the first and last positive, or an ADA positive result at the last available assessment. Transiently positive is defined as having ≥1 post-baseline ADA positive measurement and not fulfilling the conditions for persistently positive. Presence of nAb was tested for all ADA positive samples. (NCT03164616)
Timeframe: Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, tremelimumab).

InterventionParticipants (Count of Participants)
ADA positive at any visit (ADA prevalence)Treatment-emergent ADA positive (ADA incidence)Treatment-boosted ADATreatment-induced ADA (ADA positive post-baseline only)ADA positive at baseline onlyADA positive post-baseline and positive at baselinePersistently positiveTransiently positivenAb positive at any visit
T + D + SoC443833545221831

[back to top]

Time From Randomization to Second Progression (PFS2)

PFS2 was defined as the time from the date of randomization to the earliest of the progression event (subsequent to that used for the primary variable PFS) or death. The date of second progression was recorded by the Investigator and defined according to local standard clinical practice and could involve any of: objective radiological imaging, symptomatic progression or death. (NCT03164616)
Timeframe: Tumor scans performed at baseline, Week 6, Week 12 and then every 8 weeks relative to date of randomization until radiological progression. Assessed until global cohort DCO of 24 July 2019 (maximum of approximately 25 months).

Interventionmonths (Median)
T + D + SoC10.4
D + SoC10.2
SoC Alone9.4

[back to top]

Progression-Free Survival (PFS); D + SoC Compared With SoC Alone

PFS (per RECIST version 1.1 [RECIST 1.1] using Blinded Independent Central Review [BICR] assessments) was defined as time from date of randomization until date of objective disease progression or death (by any cause in the absence of progression), regardless of whether the patient withdrew from randomized therapy or received another anticancer therapy prior to progression. Median PFS was calculated using the Kaplan-Meier technique. The final analysis of PFS in the global cohort was pre-specified after approximately 497 BICR PFS events occurred across the D + SoC and SoC alone treatment arms (75% maturity). (NCT03164616)
Timeframe: Tumor scans performed at baseline, Week 6, Week 12 and then every 8 weeks relative to date of randomization until radiological progression. Assessed until global cohort DCO of 24 July 2019 (maximum of approximately 25 months).

Interventionmonths (Median)
D + SoC5.5
SoC Alone4.8

[back to top]

PFS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC

PFS (per RECIST 1.1 using BICR assessments) was defined as time from date of randomization until date of objective disease progression or death (by any cause in the absence of progression), regardless of whether the patient withdrew from randomized therapy or received another anticancer therapy prior to progression. Median PFS was calculated using the Kaplan-Meier technique. (NCT03164616)
Timeframe: Tumor scans performed at baseline, Week 6, Week 12 and then every 8 weeks relative to date of randomization until radiological progression. Assessed until global cohort DCO of 24 July 2019 (maximum of approximately 25 months.

Interventionmonths (Median)
T + D + SoC6.2
D + SoC5.5
SoC Alone4.8

[back to top]

Overall Survival (OS); D + SoC Compared With SoC Alone

OS was defined as the time from the date of randomization until death due to any cause. Any patient not known to have died at the time of analysis was censored based on the last recorded date on which the patient was known to be alive. Median OS was calculated using the Kaplan-Meier technique. The final analysis of OS in the global cohort was pre-specified after approximately 532 OS events occurred across the D + SoC and SoC alone treatment arms (80% maturity). (NCT03164616)
Timeframe: From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).

Interventionmonths (Median)
D + SoC13.3
SoC Alone11.7

[back to top]

OS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC

OS was defined as the time from the date of randomization until death due to any cause. Any patient not known to have died at the time of analysis was censored based on the last recorded date on which the patient was known to be alive. Median OS was calculated using the Kaplan-Meier technique. (NCT03164616)
Timeframe: From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).

Interventionmonths (Median)
T + D + SoC14.0
D + SoC13.3
SoC Alone11.7

[back to top]

Duration of Response (DoR)

DoR (per RECIST 1.1 using BICR assessments) was defined as the time from the date of first documented response until date of documented progression or death in the absence of disease progression. The end of response coincided with the date of progression or death from any cause used for the RECIST 1.1 PFS endpoint. The time of the initial response was defined as the latest of the dates contributing towards the first visit of PR or CR. Results are presented for the pre-specified DoR analysis using unconfirmed responses based on BICR. (NCT03164616)
Timeframe: Tumor scans performed at baseline, Week 6, Week 12 and then every 8 weeks relative to date of randomization until radiological progression. Assessed until global cohort DCO of 24 July 2019 (maximum of approximately 25 months).

Interventionmonths (Median)
T + D + SoC7.4
D + SoC6.0
SoC Alone4.2

[back to top]

Time to Deterioration of PRO Symptoms, Assessed Using EORTC QLQ-Lung Cancer Module 13 (QLQ-LC13)

The EORTC QLQ-LC13 is a disease-specific 13-item self-administered questionnaire for lung cancer, to be used in conjunction with the EORTC QLQ-C30. It comprises both multi-item and single-item measures of lung cancer-associated symptoms (ie, coughing, hemoptysis, dyspnea, and pain) and treatment-related symptoms from conventional chemotherapy and radiotherapy (ie, hair loss, neuropathy, sore mouth, and dysphagia). Scores from 0 to 100 were derived for each symptom item, with higher scores representing greater level of symptoms. Time to deterioration was defined as time from randomization until the date of first clinically meaningful deterioration that was confirmed at a subsequent visit or death (by any cause) in the absence of a clinically meaningful deterioration. (NCT03164616)
Timeframe: At baseline, Weeks 3, 6, 9, 12, 16 and 20, then Q4W until PD, on Day 28 and 2 months post-PD, then every 8 weeks until second progression/death (whichever came first). Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).

,,
Interventionmonths (Median)
QLQ-LC13 CoughQLQ-LC13 HemoptysisQLQ-LC13 DyspneaQLQ-LC13 Pain in ChestQLQ-LC13 Pain in Arm or ShoulderQLQ-LC13 Pain in Other Parts
D + SoC11.014.05.09.58.98.9
SoC Alone8.811.43.68.68.85.8
T + D + SoC9.717.85.410.08.99.7

[back to top] [back to top]

Objective Response Rate (ORR)

ORR (per RECIST 1.1 using BICR assessments) was defined as the percentage of patients with at least one visit response of complete response (CR) or partial response (PR). Results are presented for the pre-specified ORR analysis using unconfirmed responses based on BICR. (NCT03164616)
Timeframe: Tumor scans performed at baseline, Week 6, Week 12 and then every 8 weeks relative to date of randomization until radiological progression. Assessed until global cohort DCO of 24 July 2019 (maximum of approximately 25 months).

Interventionpercentage of patients (Number)
T + D + SoC46.3
D + SoC48.5
SoC Alone33.4

[back to top]

Objective Response Rate (ORR) by BICR by PD-LI Tumor Cell Expression

PD-L1 expression was defined as the percent of tumor cells with membrane staining in a minimum of 100 evaluable tumor cells per validated Dako PD-L1 IHC 28-8 pharmDx test. PD-L1 expression was classified as PD-L1 ≥1% (≥1% tumor cells with membrane staining in a minimum of a hundred evaluable tumor cells), PD-L1 < 1% and PD-L1 not quantifiable (without quantifiable PD-L1 expression), PD-L1 expression ≥ 50%, PD-L1 expression 1 to 49% (NCT03215706)
Timeframe: From date of randomization until date of documented tumor progression or subsequent anti-cancer therapy, whichever occurs first (assessed up to October 2019, approximately 23 months)

,
InterventionPercentage of Participants (Number)
BASELINE PD-L1 EXPRESSION ≥1%BASELINE PD-L1 EXPRESSION < 1%Non-Quantifiable PD-L1 ExpressionBASELINE PD-L1 EXPRESSION 1 - 49%BASELINE PD-L1 EXPRESSION >= 50%
Treatment A41.931.133.337.848.7
Treatment B27.620.928.024.530.9

[back to top]

Duration of Response (DoR)

DoR was defined as the time between the date of first confirmed documented response (CR or PR) to the date of the first documented BICR-assessed tumor progression (per RECIST 1.1), or death from any cause, whichever occurred first. Participants who started subsequent therapy (including palliative local therapy) without a prior reported progression were censored at the last evaluable tumor assessments prior to initiation of the subsequent anticancer therapy (including palliative local therapy). Participants who died without a reported prior progression were considered to have progressed on the date of their death. For subjects who neither progressed nor died, DoR was censored on the date of their last evaluable tumor assessment. DoR was evaluated for responders (confirmed CR or PR) only. (NCT03215706)
Timeframe: From date of first confirmed response to date of tumor progression (assessed up to October 2019, approximately 23 months)

InterventionMonths (Median)
Treatment A10.02
Treatment B5.09

[back to top]

Objective Response Rate (ORR) by BICR

ORR was defined as the number of randomized participants with a best overall response (BOR) of confirmed CR or PR based on BICR assessments (using RECIST v1.1 criteria), divided by the number of all randomized participants. BOR was recorded between the date of randomization and the date of objectively documented progression per RECIST 1.1 or the date of initiation of palliative local therapy or the date of initiation of subsequent anti-cancer therapy, whichever occurred first. For participants without documented progression or palliative local therapy or subsequent anti-cancer therapy, all available response designations contributed to the BOR determination. For participants who continued treatment beyond progression, the BOR was determined based on response designations recorded up to the time of the initial RECIST 1.1 defined progression. (NCT03215706)
Timeframe: From date of randomization until date of documented tumor progression or subsequent anti-cancer therapy, whichever occurs first (assessed up to October 2019, approximately 23 months)

InterventionPercentage of Participants (Number)
Treatment A37.7
Treatment B25.1

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to the date of death from any cause. OS was censored on the last date a subject was known to be alive. Survival follow-up was to be conducted every 3 months after participants's off-treatment date. (NCT03215706)
Timeframe: From date of randomization to date of death (assessed up to October 2019, approximately 23 months)

InterventionMonths (Median)
Treatment A14.13
Treatment B10.74

[back to top]

Progression Free Survival (PFS) by BICR

PFS (primary definition) was defined as the time from the randomization date to the date of the first documented tumor progression based on BICR assessment (per RECIST 1.1), or death from any cause, whichever occurred first. Participants who died without a reported prior progression were considered to have progressed on the date of their death. Participants who had not progressed or died were censored on the date of their last evaluable tumor assessment. Participants who did not have any on-study tumor assessments and did not die were censored on the randomization date. Participants who started any palliative local therapy or subsequent anticancer therapy without a prior reported progression were censored at the last evaluable tumor assessment prior to initiation of the palliative local therapy or subsequent anti-cancer therapy, whichever procedure occurred first. (NCT03215706)
Timeframe: From date of randomization until date of documented tumor progression or death due to any cause, whichever occurs first (assessed up to October 2019, approximately 23 months)

InterventionMonths (Median)
Treatment A6.83
Treatment B4.96

[back to top]

Time to Response (TTR)

TTR was defined as the time from randomization to the date of the first confirmed documented response (CR or PR), as assessed by the BICR. TTR was evaluated for responders (confirmed CR or PR) only. (NCT03215706)
Timeframe: From date of randomization to date of first confirmed documented response (assessed up to October 2019, approximately 23 months)

InterventionMonths (Median)
Treatment A2.51
Treatment B1.56

[back to top]

OS by PD-L1 Tumor Cell Expression

OS was defined as the time from randomization to the date of death from any cause. OS was censored on the last date a subject was known to be alive. Survival follow-up was to be conducted every 3 months after participants's off-treatment date. (NCT03215706)
Timeframe: From date of randomization to date of death (assessed up to October 2019, approximately 23 months)

,
InterventionMonths (Median)
< 1% PD-L1 Expression>= 1% PD-L1 Expressionwith 1-49% PD-L1 Expressionwith >= 50% PD-L1 ExpressionNon-Quantifiable PD-L1 Expression
Treatment A14.0314.2314.4614.1310.84
Treatment B9.9510.5810.2511.8617.05

[back to top]

PFS by BICR by PD-L1 Tumor Cell Expression

PFS (primary definition) was defined as the time from the randomization date to the date of the first documented tumor progression based on BICR assessment (per RECIST 1.1), or death from any cause, whichever occurred first. Participants who died without a reported prior progression were considered to have progressed on the date of their death. Participants who had not progressed or died were censored on the date of their last evaluable tumor assessment. Participants who did not have any on-study tumor assessments and did not die were censored on the randomization date. Participants who started any palliative local therapy or subsequent anticancer therapy without a prior reported progression were censored at the last evaluable tumor assessment prior to initiation of the palliative local therapy or subsequent anti-cancer therapy, whichever procedure occurred first. (NCT03215706)
Timeframe: From date of randomization until date of documented tumor progression or death due to any cause, whichever occurs first (assessed up to October 2019, approximately 23 months)

,
InterventionMonths (Median)
< 1% PD-L1 Expression>= 1% PD-L1 Expressionwith 1-49% PD-L1 Expressionwith >= 50% PD-L1 ExpressionNon-Quantifiable PD-L1 Expression
Treatment A5.827.036.748.286.14
Treatment B4.864.705.294.275.78

[back to top]

Duration Of Response

"Time from the first documentation of objective tumor response (CR or PR) to the first documentation of objective tumor progression or death due to any cause, whichever occurs first~Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.~Partial Response (PR): At least a 30% decrease in the sum of 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 on study(this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note:the appearance of one or more new lesions is also considered progression)." (NCT03256136)
Timeframe: From the first documented response until disease progression or death, up to approximately 2 years

Interventionmonths (Median)
Nivolumab Plus Ipilimumab EGFRNA
Nivolumab Plus Ipilimumab ALKNA
Nivolumab + Carboplatin + Pemetrexed With EGFR Chemo Naive0.9
Nivolumab + Carboplatin + Pemetrexed ALK Chemo Naive1.4

[back to top]

Progression Free Survival (PFS)

"Time from initiation of the study drugs to progression or death, whichever occurs first. Disease progression was assessed via RECIST 1.1~Progressive Disease (PD): At least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study(this includes the baseline sum if that is the smallest on study). 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).~Confidence Intervals (CIs) were calculated using the Kaplan Meier (KM) method" (NCT03256136)
Timeframe: From the start of treatment until disease progression or death due to any cause, up to approximately 2 years

Interventionmonths (Median)
Nivolumab Plus Ipilimumab EGFR1.3
Nivolumab Plus Ipilimumab ALK0.7
Nivolumab + Carboplatin + Pemetrexed With EGFR Chemo Naive4.65
Nivolumab + Carboplatin + Pemetrexed ALK Chemo Naive2.8

[back to top]

Disease Control Rate (DCR), Presented in Numbers of Participants

"The number of patients that achieved either complete response (CR), partial response (PR), or stable disease (SD) per RECIST version 1.1~Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.~Partial Response (PR): At least a 30% decrease in the sum of 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 diameters while on study." (NCT03256136)
Timeframe: Up to approximately 2 years

InterventionParticipants (Count of Participants)
Nivolumab Plus Ipilimumab EGFR0
Nivolumab Plus Ipilimumab ALK0
Nivolumab + Carboplatin + Pemetrexed With EGFR Chemo Naive3
Nivolumab + Carboplatin + Pemetrexed ALK Chemo Naive1

[back to top]

Overall Survival (OS)

Time from initiation of the study drugs to date of death due to any cause. CIs are the KM estimate CIs. (NCT03256136)
Timeframe: From the start of treatment until death due to any cause, up to approximately 2 years

Interventionmonths (Median)
Nivolumab Plus Ipilimumab EGFR22.3
Nivolumab Plus Ipilimumab ALK7.6
Nivolumab + Carboplatin + Pemetrexed With EGFR Chemo Naive7.75
Nivolumab + Carboplatin + Pemetrexed ALK Chemo Naive15.9

[back to top]

Objective Response Rate (ORR), Presented in Numbers of Participants

"Complete response (CR) or partial response (PR) per Response Evaluation Criteria in Solid Tumors (RECIST 1.1)~Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.~Partial Response (PR): At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters." (NCT03256136)
Timeframe: Up to approximately 2 years

InterventionParticipants (Count of Participants)
Nivolumab Plus Ipilimumab EGFR0
Nivolumab Plus Ipilimumab ALK0
Nivolumab + Carboplatin + Pemetrexed With EGFR Chemo Naive1
Nivolumab + Carboplatin + Pemetrexed ALK Chemo Naive1

[back to top]

Part A: Area Under the Plasma Concentration From Time Zero to Infinity (AUC[0-infinity]) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part A: TSR-042 and Niraparib 200 mg QDNA
Part A: TSR-042 and Niraparib 300 mg QDNA

[back to top]

Part A: AUC at Steady State (AUCss) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part A: TSR-042 and Niraparib 200 mg QD16.3481
Part A: TSR-042 and Niraparib 300 mg QD29.4312

[back to top]

Part A: AUC(0-infinity) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part A: TSR-042 and Niraparib 200 mg QDNA
Part A: TSR-042 and Niraparib 300 mg QDNA

[back to top]

Part A: Cmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part A: TSR-042 and Niraparib 200 mg QD158.6875
Part A: TSR-042 and Niraparib 300 mg QD138.9167

[back to top]

Part A: Ctau of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part A: TSR-042 and Niraparib 200 mg QD33.6000
Part A: TSR-042 and Niraparib 300 mg QD29.8000

[back to top]

Part A: Disease Control Rate

Disease Control Rate is defined as the percentage of participants who had achieved CR, PR, or stable disease (SD) per RECIST version 1.1. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionPercentage of participants (Number)
Part A: TSR-042 and Niraparib 200 mg QD43.8
Part A: TSR-042 and Niraparib 300 mg QD33.3

[back to top]

Part A: Duration of Response

Duration of response is defined as the time from first documentation of response (CR or PR) until the time of first documentation of disease progression by RECIST version 1.1 or death by any cause. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionMonths (Median)
Part A: TSR-042 and Niraparib 200 mg QD7.59
Part A: TSR-042 and Niraparib 300 mg QDNA

[back to top]

Part A: Maximum Observed Plasma (Cmax) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part A: TSR-042 and Niraparib 200 mg QD0.460600
Part A: TSR-042 and Niraparib 300 mg QD0.625667

[back to top]

Part A: Number of Participants With Dose-limiting Toxicity (DLT)

An event was considered a DLT if it occurred within the first 21 days of treatment, and met one of following DLT criteria:hematologic toxicity- Grade4 thrombocytopenia persists for >=7 days from the time of adverse event(AE) onset, Grade 4 neutropenia, Grade 4 or Grade 3 febrile neutropenia persists for >=7 days, Grade4 or Grade3 anemia requiring blood transfusion, Grade 3 thrombocytopenia associated with clinically significant bleeding, Grade 3 neutropenia associated with infection as described in Common Terminology Criteria for Adverse Events (CTCAE) version 4.0, drug related Grade 3 or 4 non-hematologic toxicity, drug related Grade 3 or 4 nonhematologic laboratory abnormality if any of the following also occur: abnormality leads to hospitalization and abnormality persists for >=7 days from the time of AE onset; drug related toxicity leading to prolonged delay (>2 weeks) in initiating Cycle 2. DLTs were collected during first cycle to establish recommended phase 2 dose (RP2D). (NCT03307785)
Timeframe: 21 days

InterventionParticipants (Count of Participants)
Part A: TSR-042 and Niraparib 200 mg QD2
Part A: TSR-042 and Niraparib 300 mg QD0

[back to top]

Part A: Number of Participants With Positive Anti-TSR-042 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-042. The presence of anti-TSR-042 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). Anti-drug Antibody Population consisted of all participants who received at least 1 dose of study treatment and had provided a pre-dose blood sample and at least 1 post-dose blood sample at or after 96 hours. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionParticipants (Count of Participants)
Part A: TSR-042 and Niraparib 200 mg QD0
Part A: TSR-042 and Niraparib 300 mg QD0

[back to top]

Part A: Objective Response Rate

Objective Response Rate is defined as the percentage of participants who achieved confirmed complete response (CR) or partial response (PR), evaluated using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 based on Investigator assessment; where CR=Disappearance of all target lesions. Any pathological lymph nodes must be <10 millimeters (mm) in the short axis. PR=At least a 30 percent (%) decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionPercentage of participants (Number)
Part A: TSR-042 and Niraparib 200 mg QD25.0
Part A: TSR-042 and Niraparib 300 mg QD0

[back to top]

Part A: Observed Concentration at the End of the Dosing Interval (Ctau) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part A: TSR-042 and Niraparib 200 mg QD0.191953
Part A: TSR-042 and Niraparib 300 mg QD0.342333

[back to top]

Part A: Progression-free Survival

Progression-free Survival is defined as the date of first dose to the date of disease progression or death due to any cause, whichever occurs earlier. Progressive Disease (PD) is defined as at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionMonths (Median)
Part A: TSR-042 and Niraparib 200 mg QD6.2
Part A: TSR-042 and Niraparib 300 mg QD2.8

[back to top]

Part A: Time to Reach Maximum Plasma Concentration (Tmax) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part A: TSR-042 and Niraparib 200 mg QD2.250
Part A: TSR-042 and Niraparib 300 mg QD4.083

[back to top]

Part A: Tmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part A: TSR-042 and Niraparib 200 mg QD0.817
Part A: TSR-042 and Niraparib 300 mg QD1.750

[back to top]

Part B: AUC(0-infinity) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part B: TSR-042 and Carboplatin-paclitaxelNA

[back to top]

Part B: Cmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part B: TSR-042 and Carboplatin-paclitaxel148.2000

[back to top]

Part B: Ctau of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part B: TSR-042 and Carboplatin-paclitaxel33.9778

[back to top]

Part B: Disease Control Rate

Disease Control Rate is defined as the percentage of participants who had achieved CR, PR, or SD per RECIST version 1.1. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionPercentage of participants (Number)
Part B: TSR-042 and Carboplatin-paclitaxel57.1

[back to top]

Part B: Number of Participants With DLT

An event was considered a DLT if it occurred within the first 21 days of treatment, and met one of following DLT criteria:hematologic toxicity- Grade4 thrombocytopenia persists for >=7 days from the time of AE onset, Grade 4 neutropenia, Grade 4 or Grade 3 febrile neutropenia persists for >=7 days, Grade4 or Grade3 anemia requiring blood transfusion, Grade 3 thrombocytopenia associated with clinically significant bleeding, Grade 3 neutropenia associated with infection as described in CTCAE version 4.0, drug related Grade 3 or 4 non-hematologic toxicity, drug related Grade 3 or 4 nonhematologic laboratory abnormality if any of the following also occur: abnormality leads to hospitalization and abnormality persists for >=7 days from the time of AE onset; drug related toxicity leading to prolonged delay (>2 weeks) in initiating Cycle 2. DLTs were collected during first cycle to establish RP2D. (NCT03307785)
Timeframe: 21 days

InterventionParticipants (Count of Participants)
Part B: TSR-042 and Carboplatin-paclitaxel1

[back to top]

Part B: Number of Participants With Positive Anti-TSR-042 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-042. The presence of anti-TSR-042 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). (NCT03307785)
Timeframe: Up to 28.5 months

InterventionParticipants (Count of Participants)
Part B: TSR-042 and Carboplatin-paclitaxel4

[back to top]

Part B: Objective Response Rate

Objective Response Rate is defined as the percentage of participants who achieved confirmed CR or PR, evaluated using RECIST version 1.1 based on Investigator assessment; where CR=Disappearance of all target lesions. Any pathological lymph nodes must be <10 mm in the short axis. PR=At least a 30% decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionPercentage of participants (Number)
Part B: TSR-042 and Carboplatin-paclitaxel42.9

[back to top]

Part F: Number of Participants With Non-serious TEAEs, STEAEs and AESIs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. An SAE is defined as any untoward medical occurrence that, at any dose; results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgement. TEAE is any event that was not present prior to the initiation of study treatment or any event already present that worsens in either intensity or frequency following exposure to study treatment. Number of participants with TEAEs, SAEs and AESIs are reported. (NCT03307785)
Timeframe: Up to 3.5 months

InterventionParticipants (Count of Participants)
Non-serious TEAEsSTEAEsAESIs
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexed100

[back to top]

Part E: Number of Participants With Non-serious TEAEs, STEAEs and AESIs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. An SAE is defined as any untoward medical occurrence that, at any dose; results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgement. TEAE is any event that was not present prior to the initiation of study treatment or any event already present that worsens in either intensity or frequency following exposure to study treatment. Number of participants with TEAEs, SAEs and AESIs are reported. (NCT03307785)
Timeframe: Up to 4.4 months

InterventionParticipants (Count of Participants)
Non-serious TEAEsSTEAEsAESIs
Part E: TSR-042 and Carboplatin-pemetrexed220

[back to top]

Part D: Vz of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=3
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab5.49556.15429.8356

[back to top]

Part D: Vss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=3
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab5.14935.63998.0614

[back to top]

Part D: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=4
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab0.5000.5500.500

[back to top]

Part D: Number of Participants With Non-serious TEAEs, STEAEs and AESIs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. An SAE is defined as any untoward medical occurrence that, at any dose; results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgement. TEAE is any event that was not present prior to the initiation of study treatment or any event already present that worsens in either intensity or frequency following exposure to study treatment. Number of participants with TEAEs, SAEs and AESIs are reported. (NCT03307785)
Timeframe: Up to 9.5 months

InterventionParticipants (Count of Participants)
Non-serious TEAEsSTEAEsAESIs
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab630

[back to top]

Part D: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=4
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab256.8000416.0000429.7500

[back to top]

Part D: CL of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=3
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab0.01020.00870.0104

[back to top]

Part D: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=3
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab51140.4410118845.392896800.1534

[back to top]

Part D: AUC0-t of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=6Cycle 4, n=5Cycle 5, n=5Cycle 11, n=4
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab32819.540152409.0255119847.099977794.6422

[back to top]

Part C: Vz/F of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

,
InterventionLiter (Mean)
Cycle 2, n=2,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab551.4003
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab559.3634

[back to top]

Part C: Vz of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionLiter (Mean)
Cycle 4, n=5,5Cycle 5, n=4,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab6.64626.16989.6347
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab5.89225.20668.5098

[back to top]

Part C: Vss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionLiter (Mean)
Cycle 4, n=5,5Cycle 5, n=4,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab6.45676.24058.9478
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab5.51774.80357.7750

[back to top]

Part C: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionHours (Median)
Cycle 4, n=5,7Cycle 5, n=5,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab1.5172.0832.000
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab0.6170.8252.417

[back to top]

Part C: Tmax,ss of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

,
InterventionHours (Median)
Cycle 2, n=4,4Cycle 5, n=4,3Cycle 11, n=3,1
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab4.9502.1582.000
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab3.9581.9672.083

[back to top]

Part C: Number of Participants With Non-serious TEAEs, STEAEs and AESIs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. An SAE is defined as any untoward medical occurrence that, at any dose; results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgement. TEAE is any event that was not present prior to the initiation of study treatment or any event already present that worsens in either intensity or frequency following exposure to study treatment. Number of participants with TEAEs, SAEs and AESIs are reported. (NCT03307785)
Timeframe: Up to 22.5 months

,
InterventionParticipants (Count of Participants)
Non-serious TEAEsSTEAEsAESIs
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab530
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab730

[back to top]

Part C: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionMicrogram per milliliter (Mean)
Cycle 4, n=4,6Cycle 5, n=5,6Cycle 11, n=3,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab70.425082.9800105.4667
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab64.266753.150060.9500

[back to top]

Part C: Ctau,ss of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

,
InterventionMicrogram per milliliter (Mean)
Cycle 2, n=5,4
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab0.484000
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab1.094250

[back to top]

Part C: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionMicrogram per milliliter (Mean)
Cycle 4, n=5,7Cycle 5, n=5,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab234.4000325.2000349.7500
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab227.1429417.3333321.0000

[back to top]

Part C: Cmax,ss of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

,
InterventionMicrogram per milliliter (Mean)
Cycle 2, n=4,4Cycle 5, n=4,3Cycle 11, n=3,1
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab0.8545000.7097500.468333
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab1.4047501.0736670.638000

[back to top]

Part C: CL/F of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

,
InterventionLiter per hour (Mean)
Cycle 2, n=2,4
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab10.3777
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab14.4947

[back to top]

Part C: CL of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionLiter per hour (Mean)
Cycle 4, n=5,5Cycle 5, n=5,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab0.01000.00820.0079
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab0.01040.00820.0086

[back to top]

Part C: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionHours*microgram per milliliter (Mean)
Cycle 4, n=5,5Cycle 5, n=5,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab55974.4268137399.5881147907.5379
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab50924.1510131288.0291119189.5905

[back to top]

Part C: AUC0-t of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

,
InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=6,7Cycle 4, n=5,7Cycle 5, n=5,6Cycle 11, n=4,2
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab29830.654254268.1379137777.3188143070.3093
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab26311.178949198.8237131281.9046119161.8795

[back to top]

Part C: AUC0-t of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

,
InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=6,7Cycle 2, n=4,4
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab3.451314.7473
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab11.360124.9691

[back to top]

Part B: Vz of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=10Cycle 5, n=8Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel5.36617.26278.0529

[back to top]

Part B: Vss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=10Cycle 5, n=8Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel5.18936.77017.4351

[back to top]

Part B: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Cycle 4, n=10Cycle 5, n=9Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel0.5751.5000.542

[back to top]

Part B: Number of Participants With Non-serious TEAEs, STEAEs and AESIs

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. An SAE is defined as any untoward medical occurrence that, at any dose; results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgement. TEAE is any event that was not present prior to the initiation of study treatment or any event already present that worsens in either intensity or frequency following exposure to study treatment. Number of participants with TEAEs, SAEs and AESIs are reported. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionParticipants (Count of Participants)
Non-Serious TEAEsSTEAEsAESIs
Part B: TSR-042 and Carboplatin-paclitaxel1490

[back to top]

Part B: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=9Cycle 5, n=8Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel67.244459.300057.4833

[back to top]

Part B: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=10Cycle 5, n=9Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel225.8000421.1111446.6667

[back to top]

Part B: CL of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Cycle 4, n=10Cycle 5, n=8Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel0.00980.00820.0092

[back to top]

Part B: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 4, n=10Cycle 5, n=8Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel55073.0926130694.9752117033.3612

[back to top]

Part B: AUC0-t of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=14Cycle 4, n=10Cycle 5, n=9Cycle 11, n=6
Part B: TSR-042 and Carboplatin-paclitaxel28097.984654730.0053124309.5455117030.4081

[back to top]

Part A: Volume of Distribution After Oral Administration (Vz/F) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 1, n=1,0Cycle 2, n=8,2
Part A: TSR-042 and Niraparib 200 mg QD380.1478716.1418

[back to top]

Part A: Volume of Distribution After Oral Administration (Vz/F) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 2, n=8,2
Part A: TSR-042 and Niraparib 300 mg QD487.8826

[back to top]

Part A: Volume of Distribution After Intravenous Administration (Vz) of of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=5,2Cycle 5, n=8,1Cycle 11, n=4,0
Part A: TSR-042 and Niraparib 200 mg QD5.69256.88117.4795

[back to top]

Part A: Volume of Distribution After Intravenous Administration (Vz) of of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=5,2Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD10.39846.3070

[back to top]

Part C: Ctau of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab37.2750
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab28.7500

[back to top]

Part C: Ctau of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab0.161233
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab0.478714

[back to top]

Part C: Cmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab138.6667
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab158.4857

[back to top]

Part C: Cmax of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab0.243667
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab0.891571

[back to top]

Part F: AUC0-t of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168 and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: CL of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: CL of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Cmax of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168 and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Cmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Cmax,ss of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part C: AUCss of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab21.4926
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab26.5006

[back to top]

Part C: AUC(0-infinity) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part C: TSR-042, Niraparib 200 mg QD and BevacizumabNA
Part C: TSR-042, Niraparib 300 mg QD and BevacizumabNA

[back to top]

Part C: AUC(0-infinity) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part C: TSR-042, Niraparib 200 mg QD and BevacizumabNA
Part C: TSR-042, Niraparib 300 mg QD and BevacizumabNA

[back to top]

Part B: Tmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part B: TSR-042 and Carboplatin-paclitaxel1.000

[back to top]

Part B: Progression-free Survival

Progression-free Survival is defined as the date of first dose to the date of disease progression or death due to any cause, whichever occurs earlier. PD 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. (NCT03307785)
Timeframe: Up to 28.5 months

InterventionMonths (Median)
Part B: TSR-042 and Carboplatin-paclitaxel17.6

[back to top]

Part A: Tmax at Steady State (Tmax,ss) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Cycle 2, n=10,3
Part A: TSR-042 and Niraparib 300 mg QD2.250

[back to top]

Part A: Tmax at Steady State (Tmax,ss) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Cycle 2, n=10,3Cycle 5, n=7,0Cycle 11, n=2,0
Part A: TSR-042 and Niraparib 200 mg QD4.0082.0001.817

[back to top]

Part A: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Cycle 4, n=9,4Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD1.3002.783

[back to top]

Part A: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Cycle 4, n=9,4Cycle 5, n=8,1Cycle 11, n=5,0
Part A: TSR-042 and Niraparib 200 mg QD1.5000.6250.567

[back to top]

Part A: Volume of Distribution After Intravenous Administration (Vss) of of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=5,2Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD9.86805.9659

[back to top]

Part A: Volume of Distribution After Intravenous Administration (Vss) of of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Cycle 4, n=5,2Cycle 5, n=8,1Cycle 11, n=4,0
Part A: TSR-042 and Niraparib 200 mg QD5.56476.65807.3107

[back to top]

Part F: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Ctau of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Ctau of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Ctau,ss of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: AUCss of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Disease Control Rate

Disease Control Rate is defined as the percentage of participants who had achieved CR, PR, or SD per RECIST version 1.1. (NCT03307785)
Timeframe: Up to 3.5 months

InterventionPercentage of participants (Number)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexed100

[back to top]

Part F: Duration of Response

Duration of response is defined as the time from first documentation of response (CR or PR) until the time of first documentation of disease progression by RECIST version 1.1 or death by any cause. (NCT03307785)
Timeframe: Up to 3.5 months

InterventionMonths (Median)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Number of Participants With DLT

An event was considered a DLT if it occurred within the first 21 days of treatment, and met one of following DLT criteria:hematologic toxicity- Grade4 thrombocytopenia persists for >=7 days from the time of AE onset, Grade 4 neutropenia, Grade 4 or Grade 3 febrile neutropenia persists for >=7 days, Grade4 or Grade3 anemia requiring blood transfusion, Grade 3 thrombocytopenia associated with clinically significant bleeding, Grade 3 neutropenia associated with infection as described in CTCAE version 4.0, drug related Grade 3 or 4 non-hematologic toxicity, drug related Grade 3 or 4 nonhematologic laboratory abnormality if any of the following also occur: abnormality leads to hospitalization and abnormality persists for >=7 days from the time of AE onset; drug related toxicity leading to prolonged delay (>2 weeks) in initiating Cycle 2. DLTs were collected during first cycle to establish RP2D. (NCT03307785)
Timeframe: 21 days

InterventionParticipants (Count of Participants)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexed0

[back to top]

Part F: Number of Participants With Positive Anti-TSR-022 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-022. The presence of anti-TSR-022 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). (NCT03307785)
Timeframe: Up to 3.5 months

InterventionParticipants (Count of Participants)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Number of Participants With Positive Anti-TSR-042 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-042. The presence of anti-TSR-042 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). (NCT03307785)
Timeframe: Up to 3.5 months

InterventionParticipants (Count of Participants)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Objective Response Rate

Objective Response Rate is defined as the percentage of participants who achieved confirmed CR or PR, evaluated using RECIST version 1.1 based on Investigator assessment; where CR=Disappearance of all target lesions. Any pathological lymph nodes must be <10 mm in the short axis. PR=At least a 30% decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters. (NCT03307785)
Timeframe: Up to 3.5 months

InterventionPercentage of participants (Number)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexed0

[back to top]

Part F: AUC0-t of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168 and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: AUC(0-infinity) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: AUC(0-infinity) of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168 and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168 and 336 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part E: Vz of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose (each cycle was 21 days)

InterventionLiter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Vss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose (each cycle was 21 days)

InterventionLiter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose (each cycle was 21 days)

InterventionHours (Median)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Tmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Progression-free Survival

Progression-free Survival is defined as the date of first dose to the date of disease progression or death due to any cause, whichever occurs earlier. PD 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. (NCT03307785)
Timeframe: Up to 4.4 months

InterventionMonths (Median)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Objective Response Rate

Objective Response Rate is defined as the percentage of participants who achieved confirmed CR or PR, evaluated using RECIST version 1.1 based on Investigator assessment; where CR=Disappearance of all target lesions. Any pathological lymph nodes must be <10 mm in the short axis. PR=At least a 30% decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters. (NCT03307785)
Timeframe: Up to 4.4 months

InterventionPercentage of participants (Number)
Part E: TSR-042 and Carboplatin-pemetrexed0

[back to top]

Part E: Number of Participants With Positive Anti-TSR-042 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-042. The presence of anti-TSR-042 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). (NCT03307785)
Timeframe: Up to 4.4 months

InterventionParticipants (Count of Participants)
Part E: TSR-042 and Carboplatin-pemetrexed0

[back to top]

Part E: Number of Participants With DLT

An event was considered a DLT if it occurred within the first 21 days of treatment, and met one of following DLT criteria:hematologic toxicity- Grade4 thrombocytopenia persists for >=7 days from the time of AE onset, Grade 4 neutropenia, Grade 4 or Grade 3 febrile neutropenia persists for >=7 days, Grade4 or Grade3 anemia requiring blood transfusion, Grade 3 thrombocytopenia associated with clinically significant bleeding, Grade 3 neutropenia associated with infection as described in CTCAE version 4.0, drug related Grade 3 or 4 non-hematologic toxicity, drug related Grade 3 or 4 nonhematologic laboratory abnormality if any of the following also occur: abnormality leads to hospitalization and abnormality persists for >=7 days from the time of AE onset; drug related toxicity leading to prolonged delay (>2 weeks) in initiating Cycle 2. DLTs were collected during first cycle to establish RP2D. (NCT03307785)
Timeframe: 21 days

InterventionParticipants (Count of Participants)
Part E: TSR-042 and Carboplatin-pemetrexed0

[back to top]

Part E: Duration of Response

Duration of response is defined as the time from first documentation of response (CR or PR) until the time of first documentation of disease progression by RECIST version 1.1 or death by any cause. (NCT03307785)
Timeframe: Up to 4.4 months

InterventionMonths (Median)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Disease Control Rate

Disease Control Rate is defined as the percentage of participants who had achieved CR, PR, or SD per RECIST version 1.1. (NCT03307785)
Timeframe: Up to 4.4 months

InterventionPercentage of participants (Number)
Part E: TSR-042 and Carboplatin-pemetrexed0

[back to top]

Part E: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Ctau of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: Cmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: CL of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: AUC0-t of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part E: AUC(0-infinity) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part E: TSR-042 and Carboplatin-pemetrexedNA

[back to top]

Part D: Tmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab1.250

[back to top]

Part F: Progression-free Survival

Progression-free Survival is defined as the date of first dose to the date of disease progression or death due to any cause, whichever occurs earlier. PD 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. (NCT03307785)
Timeframe: Up to 3.5 months

InterventionMonths (Median)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Tmax of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Tmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Tmax,ss of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Tmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Vss of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Vss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Vz of TSR-022

Blood samples were collected at indicated time points. PK parameters of TSR-022 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part F: Vz of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 1, 2, 3, 24, 48, 96, 168, and 336 hours post-dose; Cycle 2: Pre-dose; Cycle 5: Pre-dose, 0.5, 2, 24, 48, 96, 168, and 336 hours post-dose (each cycle was 21 days)

InterventionLiter (Mean)
Part F: TSR-042, TSR-022, and Carboplatin-pemetrexedNA

[back to top]

Part A: Area Under the Plasma Concentration From Time Zero to t (AUC[0-t]) of Niraparib

Blood samples were collected at indicated time points. Pharmacokinetic (PK) parameters of niraparib were calculated using non-compartmental methods. PK population consisted of all participants who received at least 1 dose of study treatment and had at least 1 PK sample. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

,
InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=15,6Cycle 2, n=10,3
Part A: TSR-042 and Niraparib 200 mg QD6.043014.9172
Part A: TSR-042 and Niraparib 300 mg QD7.834129.3024

[back to top]

Part A: AUC(0-t) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=16,6Cycle 4, n=9,4Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD29420.834729311.8182141328.9288

[back to top]

Part A: AUC(0-t) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 1, n=16,6Cycle 4, n=9,4Cycle 5, n=8,1Cycle 11, n=5,0
Part A: TSR-042 and Niraparib 200 mg QD26827.770355408.3992137108.2574139591.8834

[back to top]

Part A: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 4, n=7,2Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD46122.3924141306.3782

[back to top]

Part A: AUCss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Mean)
Cycle 4, n=7,2Cycle 5, n=8,1Cycle 11, n=4,0
Part A: TSR-042 and Niraparib 200 mg QD58598.5076135171.2753151575.3645

[back to top]

Part A: Clearance After Intravenous Administration (CL) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Cycle 4, n=7,2Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD0.01100.0071

[back to top]

Part A: Clearance After Intravenous Administration (CL) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Cycle 4, n=7,2Cycle 5, n=8,1Cycle 11, n=4,0
Part A: TSR-042 and Niraparib 200 mg QD0.00900.00810.0072

[back to top]

Part A: Clearance After Oral Administration (CL/F) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Cycle 2, n=8,2
Part A: TSR-042 and Niraparib 300 mg QD10.2833

[back to top]

Part A: Clearance After Oral Administration (CL/F) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionLiter per hour (Mean)
Cycle 1, n=1,0Cycle 2, n=8,2
Part A: TSR-042 and Niraparib 200 mg QD28.563816.6295

[back to top]

Part A: Cmax at Steady State (Cmax,ss) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 2, n=10,3
Part A: TSR-042 and Niraparib 300 mg QD1.706667

[back to top]

Part D: Progression-free Survival

Progression-free Survival is defined as the date of first dose to the date of disease progression or death due to any cause, whichever occurs earlier. PD 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. (NCT03307785)
Timeframe: Up to 9.5 months

InterventionMonths (Median)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab7.6

[back to top]

Part D: Objective Response Rate

Objective Response Rate is defined as the percentage of participants who achieved confirmed CR or PR, evaluated using RECIST version 1.1 based on Investigator assessment; where CR=Disappearance of all target lesions. Any pathological lymph nodes must be <10 mm in the short axis. PR=At least a 30% decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters. (NCT03307785)
Timeframe: Up to 9.5 months

InterventionPercentage of participants (Number)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab50.0

[back to top]

Part D: Number of Participants With Positive Anti-TSR-042 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-042. The presence of anti-TSR-042 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). (NCT03307785)
Timeframe: Up to 9.5 months

InterventionParticipants (Count of Participants)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab0

[back to top]

Part D: Number of Participants With DLT

An event was considered a DLT if it occurred within the first 21 days of treatment, and met one of following DLT criteria:hematologic toxicity- Grade4 thrombocytopenia persists for >=7 days from the time of AE onset, Grade 4 neutropenia, Grade 4 or Grade 3 febrile neutropenia persists for >=7 days, Grade4 or Grade3 anemia requiring blood transfusion, Grade 3 thrombocytopenia associated with clinically significant bleeding, Grade 3 neutropenia associated with infection as described in CTCAE version 4.0, drug related Grade 3 or 4 non-hematologic toxicity, drug related Grade 3 or 4 nonhematologic laboratory abnormality if any of the following also occur: abnormality leads to hospitalization and abnormality persists for >=7 days from the time of AE onset; drug related toxicity leading to prolonged delay (>2 weeks) in initiating Cycle 2. DLTs were collected during first cycle to establish RP2D. (NCT03307785)
Timeframe: 21 days

InterventionParticipants (Count of Participants)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab0

[back to top]

Part D: Disease Control Rate

Disease Control Rate is defined as the percentage of participants who had achieved CR, PR, or SD per RECIST version 1.1. (NCT03307785)
Timeframe: Up to 9.5 months

InterventionPercentage of participants (Number)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab83.3

[back to top]

Part D: Ctau of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab36.5250

[back to top]

Part D: Cmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab188.5000

[back to top]

Part D: AUC(0-infinity) of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycles 1 and 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionHours*microgram per milliliter (Geometric Mean)
Part D: TSR-042, Carboplatin-paclitaxel and BevacizumabNA

[back to top]

Part C: Tmax of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab1.200
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab0.583

[back to top]

Part A: Cmax at Steady State (Cmax,ss) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 2, n=10,3Cycle 5, n=7,0Cycle 11, n=2,0
Part A: TSR-042 and Niraparib 200 mg QD0.9259000.8390710.768000

[back to top]

Part A: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=9,4Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD178.5000319.0000

[back to top]

Part A: Cmax,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=9,4Cycle 5, n=8,1Cycle 11, n=5,0
Part A: TSR-042 and Niraparib 200 mg QD222.5556393.8750405.4000

[back to top]

Part A: Ctau at Steady State (Ctau,ss) of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 2: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose; Cycles 5 and 11: Pre-dose, 2 hours post-dose (each cycle was 21 days)

,
InterventionMicrogram per milliliter (Mean)
Cycle 2, n=11,6
Part A: TSR-042 and Niraparib 200 mg QD0.507900
Part A: TSR-042 and Niraparib 300 mg QD0.622000

[back to top]

Part A: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=8,1Cycle 5, n=8,1
Part A: TSR-042 and Niraparib 300 mg QD63.300071.2000

[back to top]

Part A: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=8,1Cycle 5, n=8,1Cycle 11, n=4,0
Part A: TSR-042 and Niraparib 200 mg QD77.362568.550093.8000

[back to top]

Part A: Number of Participants With Non-serious Treatment-emergent Adverse Events (TEAEs), Serious TEAEs (STEAEs) and Adverse Events of Special Interest (AESIs)

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of a study treatment, whether or not considered related to the study treatment. An SAE is defined as any untoward medical occurrence that, at any dose; results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or any other situation according to medical or scientific judgement. TEAE is any event that was not present prior to the initiation of study treatment or any event already present that worsens in either intensity or frequency following exposure to study treatment. Number of participants with TEAEs, SAEs and AESIs are reported. (NCT03307785)
Timeframe: Up to 28.5 months

,
InterventionParticipants (Count of Participants)
Non-serious TEAEsSTEAEsAESIs
Part A: TSR-042 and Niraparib 200 mg QD16111
Part A: TSR-042 and Niraparib 300 mg QD641

[back to top]

Part D: Ctau,ss of TSR-042

Blood samples were collected at indicated time points. PK parameters of TSR-042 were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 4: Pre-dose, 0.5, 2, 24, 96, 168, 336 and 504 hours post-dose; Cycles 5 and 11: Pre-dose, 0.5, 2, 24, 96, 168, 336, 504 and 1008 hours post-dose (each cycle was 21 days)

InterventionMicrogram per milliliter (Mean)
Cycle 4, n=5Cycle 5, n=5Cycle 11, n=3
Part D: TSR-042, Carboplatin-paclitaxel and Bevacizumab53.520048.080043.3000

[back to top]

Part C: Tmax of Niraparib

Blood samples were collected at indicated time points. PK parameters of niraparib were calculated using non-compartmental methods. (NCT03307785)
Timeframe: Cycle 1: Pre-dose, 1, 2, 4, 6, 8 and 24 hours post-dose (each cycle was 21 days)

InterventionHours (Median)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab3.975
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab4.050

[back to top]

Part C: Progression-free Survival

Progression-free Survival is defined as the date of first dose to the date of disease progression or death due to any cause, whichever occurs earlier. PD 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. (NCT03307785)
Timeframe: Up to 22.5 months

InterventionMonths (Median)
Part C: TSR-042, Niraparib 200 mg QD and BevacizumabNA
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab9.1

[back to top]

Part C: Objective Response Rate

Objective Response Rate is defined as the percentage of participants who achieved confirmed CR or PR, evaluated using RECIST version 1.1 based on Investigator assessment; where CR=Disappearance of all target lesions. Any pathological lymph nodes must be <10 mm in the short axis. PR=At least a 30% decrease in the sum of the diameters of target lesions, taking as a reference, the Baseline sum of the diameters. (NCT03307785)
Timeframe: Up to 22.5 months

InterventionPercentage of participants (Number)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab50.0
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab14.3

[back to top]

Part C: Number of Participants With Positive Anti-TSR-042 Antibodies

Serum samples were collected at indicated time points and tested for the presence of antibodies that bind to TSR-042. The presence of anti-TSR-042 antibodies were assessed using a tiered approach using electrochemiluminescence (that is, screening, confirmation, titer, and neutralizing antibody assay). (NCT03307785)
Timeframe: Up to 22.5 months

InterventionParticipants (Count of Participants)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab0
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab0

[back to top]

Part C: Number of Participants With DLT

An event was considered a DLT if it occurred within the first 21 days of treatment, and met one of following DLT criteria:hematologic toxicity- Grade4 thrombocytopenia persists for >=7 days from the time of AE onset, Grade 4 neutropenia, Grade 4 or Grade 3 febrile neutropenia persists for >=7 days, Grade4 or Grade3 anemia requiring blood transfusion, Grade 3 thrombocytopenia associated with clinically significant bleeding, Grade 3 neutropenia associated with infection as described in CTCAE version 4.0, drug related Grade 3 or 4 non-hematologic toxicity, drug related Grade 3 or 4 nonhematologic laboratory abnormality if any of the following also occur: abnormality leads to hospitalization and abnormality persists for >=7 days from the time of AE onset; drug related toxicity leading to prolonged delay (>2 weeks) in initiating Cycle 2. DLTs were collected during first cycle to establish RP2D. (NCT03307785)
Timeframe: 21 days

InterventionParticipants (Count of Participants)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab1
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab1

[back to top]

Part C: Disease Control Rate

Disease Control Rate is defined as the percentage of participants who had achieved CR, PR, or SD per RECIST version 1.1. (NCT03307785)
Timeframe: Up to 22.5 months

InterventionPercentage of participants (Number)
Part C: TSR-042, Niraparib 200 mg QD and Bevacizumab83.3
Part C: TSR-042, Niraparib 300 mg QD and Bevacizumab85.7

[back to top]

Absolute Value of Tumor Mutational Burden (TMB) in Tumor Tissue

Mutational load within tumor tissue was defined as number per megabase of the genome, coding, base substitution, and indel mutations present in the sample. Mutational load was determined in whole blood samples using next generation deoxyribonucleic acid (DNA) sequencing followed by computational analysis. (NCT03317496)
Timeframe: Pre-dose on Day 1 of Cycle 1

InterventionMutations per megabase (Mean)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin4.3
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin2.8
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin4.4
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin2.5

[back to top]

Duration of Response (DOR) as Per RECIST v 1.1 by Investigator Assessment

DOR was defined as time from first documentation of objective response (confirmed CR or PR) to the date of first PD documentation or death due to any cause, whichever occurs first. CR: disappearance of target and non-target lesions, with exception of nodal disease and normalization of tumor markers. All nodes, target and non-target must have short axis measures less than (<)10 millimeter(mm). PR: >=30% decrease in sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference baseline sum of diameters. Non-target lesions must be non-PD. PD: >=20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to relative increase of 20%, sum must also demonstrate an absolute increase of at least 5mm, appearance of one or more new lesions was considered PD. Median DOR was not derived for < 5 participants. (NCT03317496)
Timeframe: From date of first documented response to date of first documented PD or death due to any cause, whichever occurred first (maximum up to 5 years approximately)

InterventionMonths (Median)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/CarboplatinNA
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin9.6
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/CarboplatinNA
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/CisplatinNA

[back to top]

Number of Participants With Grade 3 or Higher TEAEs Based on National Cancer Institute Common Toxicity Criteria for Adverse Events (NCI CTCAE) v 4.03

AE was any untoward medical occurrence in a participant who received any study drug without regard to possibility of causal relationship. TEAEs were those events with onset dates occurring during the on-treatment period. On-treatment period was defined as time from first dose of any study treatment and up to 30 days after last dose or start day of new anti-cancer drug therapy minus 1 day, whichever occurred first. TEAEs were graded by the investigator using NCI CTCAE v 4.03 as Grade 1 = mild; Grade 2 = moderate; Grade 3 = severe; Grade 4 = life-threatening; Grade 5 = death. In this outcome measure, number of participants with grade 3 or higher TEAEs were reported. (NCT03317496)
Timeframe: From start of the treatment up to 30 days after last dose or start of new anticancer therapy minus 1 day, whichever occurred first (maximum up to 5 years approximately)

InterventionParticipants (Count of Participants)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin5
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin12
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin6
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin37

[back to top] [back to top]

Overall Survival (OS)

OS was defined as the time from the first dose of study treatment to the date of death due to any cause. Participants last known to be alive were censored at the date of last contact. The median duration of OS was not derived for less than (<) 10 participants. (NCT03317496)
Timeframe: From first dose of study treatment until death due to any cause (maximum up to 5 years approximately)

InterventionMonths (Median)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/CarboplatinNA
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin18.1
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/CarboplatinNA
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin15.1

[back to top]

Percentage of Participants With Confirmed Objective Response (OR) as Per Response Evaluation Criteria in Solid Tumors (RECIST) Version (v) 1.1 by Investigator Assessment

OR: complete response(CR) or partial response(PR)determined by investigator according to RECIST v1.1 from date of first dose of study treatment until date of first documentation of progressive disease(PD),confirmed by repeat assessments performed no less than 4 weeks after first response. CR: disappearance of target and non-target lesions, with exception of nodal disease and normalization of tumor markers. All nodes, target and non-target must have short axis measures less than (<)10 millimeter(mm). PR: >=30% decrease in sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference baseline sum of diameters. Non-target lesions must be non-PD. PD: >=20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to relative increase of 20%, sum must also demonstrate an absolute increase of at least 5mm, appearance of one or more new lesions was considered PD. (NCT03317496)
Timeframe: From start of the treatment until disease progression or death due to any cause, whichever occurred first (maximum up to 3.5 years approximately)

InterventionPercentage of Participants (Number)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin50.0
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin53.8
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin33.3
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin39.0

[back to top]

Phase 1b Lead-in: Number of Participants With Dose-Limiting Toxicities (DLT)

DLTs=occurrence of any AEs attributable to study treatment in first 2 treatment cycles:Hematologic: grade(G)4 neutropenia lasting >7days;febrile neutropenia with body temperature >=38 degree Celsius for >1hour; G>=3 neutropenic infection(absolute neutrophil count <1.0*10^9/L),G>=3 thrombocytopenia (platelet count<50.0-25.0*10^9/L)with bleeding;G4 thrombocytopenia(PC<25.0*10^9/L),G4 anemia(life-threatening).Non-hematologic: any G4 toxicities;G3 toxicities persisting for >3days despite medical treatment(nausea,vomiting,diarrhea)except endocrinopathies controlled with hormonal therapy;ALT/AST >3*upper limit of normal(ULN)if normal at baseline or 2*Baseline(>ULN at baseline)with total bilirubin >2*ULN and alkaline phosphatase <2*ULN;G3 QTcF prolongation after correction of any reversible cause(electrolyte abnormalities/hypoxia).Delay of >=3weeks in scheduled administration/failure to deliver 75% of doses due to toxicities attributable to any study treatment. DLT-evaluable analysis set. (NCT03317496)
Timeframe: Day 1 up to Week 6 (first 2 treatment cycles; 1 cycle = 21 days)

InterventionParticipants (Count of Participants)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin0
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin1
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin0
Phase 1b Lead-in: Avelumab 1200mg+Gemcitabine/Cisplatin1

[back to top]

Progression Free Survival (PFS) as Per RECIST v 1.1 by Investigator Assessment

PFS was defined as the time from the date of first dose of study treatment to the date of the first documentation of PD per RECIST v1.1 or death due to any cause, whichever occurred first. PD: >=20% increase in sum of diameters of target lesions, taking as reference the smallest sum on study. In addition to relative increase of 20%, sum must also demonstrate an absolute increase of at least 5mm, appearance of one or more new lesions was considered PD. The median duration of PFS was not derived for less than (<) 10 participants. (NCT03317496)
Timeframe: From start of treatment until disease progression or death due to any cause, whichever occurred first (maximum up to 5 years approximately)

InterventionMonths (Median)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/CarboplatinNA
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin9.8
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/CarboplatinNA
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin5.4

[back to top]

Time-to-Tumor Response (TTR) as Per RECIST v 1.1 by Investigator Assessment

TTR was defined as the time from the date of first dose of study treatment to the first documentation of objective response (CR or PR) as assessed by investigator according to RECIST v 1.1. CR: disappearance of target and non-target lesions, with exception of nodal disease and normalization of tumor markers. All nodes, target and non-target must have short axis measures less than (<)10 millimeter(mm). PR: >=30% decrease in sum of measures (longest diameter for tumor lesions and short axis measure for nodes) of target lesions, taking as reference baseline sum of diameters. Non-target lesions must be non-PD. (NCT03317496)
Timeframe: From first dose of study treatment until first documentation of CR or PR (maximum up to 5 years approximately)

InterventionMonths (Median)
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin2.8
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin1.4
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin1.3
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin1.5

[back to top]

Number of Participants With Grade 3 or Higher Laboratory Abnormalities by CTCAE Grade

Participants with laboratory abnormalities of any Grade as per NCI CTCAE toxicity grading v4.03 were summarized:hematology(anemia,hemoglobin increased,lymphocyte count decreased,lymphocyte count increased, neutrophil count decreased,platelet count decreased and white blood cell decreased)and clinical chemistry(alanine aminotransferase increased,alkaline phosphatase,increased,aspartate,aminotransferase increased,blood bilirubin increased,cholesterol high,creatinine phosphokinase[cpk] increased,creatinine increased,gamma-glutamyl transferase[ggt] increased,hypercalcemia,hyperglycemia,hyperkalemia, hypermagnesemia,hypernatremia,hypertriglyceridemia,hypoalbuminemia,hypocalcemia,hypoglycemia,hypokalemia,hypomagnesemia,hyponatremia, hypophosphatemia,serum amylase increased and lipase increased).As per NCI CTCAE toxicity grading v4.03, Grade1=mild;Grade2=moderate;Grade3=severe;Grade4=life-threatening;Grade 5=death.Parameters with at least 1 participant with abnormal value are reported. (NCT03317496)
Timeframe: From screening up to 90 days after last dose of study drug (maximum up to 5 years approximately)

,,,
InterventionParticipants (Count of Participants)
ALANINE AMINOTRANSFERASE INCREASEDALKALINE PHOSPHATASE INCREASEDASPARTATE AMINOTRANSFERASE INCREASEDBLOOD BILIRUBIN INCREASEDCPK INCREASEDCREATININE INCREASEDGGT INCREASEDHYPERGLYCEMIAHYPERKALEMIAHYPOKALEMIAHYPONATREMIALIPASE INCREASEDSERUM AMYLASE INCREASEDHYPOPHOSPHATEMIAHYPOMAGNESEMIAHYPOCALCEMIAHYPERTRIGLYCERIDEMIAHYPERNATREMIAHYPERMAGNESEMIAHYPERCALCEMIAANEMIALYMPHOCYTE COUNT DECREASEDLYMPHOCYTE COUNT INCREASEDNEUTROPHIL COUNT DECREASEDPLATELET COUNT DECREASEDWHITE BLOOD CELL DECREASED
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin00000010001101000000010303
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin10110122102220003000550827
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin10100102010102020000230332
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin31201044254443212111671211116

[back to top]

Number of Participants With Programmed Death-Ligand 1 (PD-L1) Expression

PD-L1 expression was determined using the Ventana PD-L1 SP263 IHC assay. PD-L1-positive status in UC cohorts was defined using an algorithm that combines assessments of PD-L1 staining on tumor and immune cells scored by pathologists and in NSCLC cohorts was defined as PD-L1 expression on >=1% of tumor cells. PD-L1 expression at baseline and on-treatment were reported in this outcome measure. (NCT03317496)
Timeframe: Baseline and Cycle 2 Day 8 (each cycle of 21 days)

,,,
InterventionParticipants (Count of Participants)
Baseline- Positive PD-L1Baseline- Negative PD-L1Baseline- Unknown PD-L1On-treatment (Cycle 2 Day 8)- Positive PD-L1On-treatment (Cycle 2 Day 8)- Negative PD-L1On-treatment (Cycle 2 Day 8)- Unknown PD-L1
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin141105
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin6702110
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin042024
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin281303632

[back to top]

Number of Participants With Treatment Emergent Adverse Events (TEAEs) and Serious TEAEs

Adverse event (AE) was any untoward medical occurrence in a participants who received any study drug without regard to possibility of causal relationship. Serious adverse event was any untoward medical occurrence that at any dose resulted in any of following outcomes/deemed significant for any other reason: death; initial /prolonged inpatient hospitalization; life-threatening experience (immediate risk of dying); persistent or significant disability/incapacity; congenital anomaly/birth defect. TEAEs were those events with onset dates occurring during the on-treatment period. On-treatment period was defined as time from first dose of any study treatment and up to 30 days after last dose or start day of new anti-cancer drug therapy minus 1 day, whichever occurred first. (NCT03317496)
Timeframe: From start of the treatment up to 30 days after last dose or start of new anticancer therapy minus 1 day, whichever occurred first (maximum up to 5 years approximately)

,,,
InterventionParticipants (Count of Participants)
TEAEsSerious TEAEs
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin65
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin139
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin63
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin4020

[back to top]

Serum Concentration of Avelumab

The lower limit of quantification (LLOQ) for avelumab was 0.2 micrograms per milliliter. Pharmacokinetic concentration analysis set was subset of safety analysis set and included participants who had at least one concentration measurement for avelumab or other study drugs which they were assigned to receive. Treatment groups with same dose and administration frequency were combined as pre-specified in reporting and analysis plan. (NCT03317496)
Timeframe: Pre-dose, 1 hour post-dose on Day 1 of Cycle 1, 2, 3, 6, 10, 14; 336 hours post-dose on Day 15 of Cycle 1, 2, 3 (each cycle of 21 days)

,,,
InterventionMicrograms per milliliter (Geometric Mean)
Cycle 1/Day 1- 1 hourCycle 1/Day 15- 336 hoursCycle 2/Day 1- pre-doseCycle 2/Day 1- 1 hourCycle 2/Day 15- 336 hoursCycle 3/Day 1- pre-doseCycle 3/Day 1- 1 hourCycle 3/Day 15- 336 hoursCycle 6/Day 1- pre-doseCycle 6/Day 1- 1 hourCycle 10/Day 1- pre-doseCycle 10/Day 1- 1 hourCycle 14/Day 1- pre-doseCycle 14/Day 1- 1 hour
Phase 1b Lead-in: Avelumab 1200 mg + Pemetrexed/Carboplatin284.116.514.328104.516.824.87893.8126.1710.86245.46.6206.04012.2429.05
Phase 1b Lead-in: Avelumab 800 mg + Gemcitabine/Cisplatin172.29.5703.754197.013.555.651204.017.159.518208.38.695222.013.37216.6
Phase 1b Lead-in: Avelumab 800 mg + Pemetrexed/Carboplatin173.312.324.780164.216.878.122147.019.4011.3892.539.523179.014.97215.3
Phase 1b Lead-in+Phase 2:Avelumab 1200mg+Gemcitabine/Cisplatin300.214.715.013311.918.666.771296.822.5510.39344.018.55242.916.49402.2

[back to top]

Time to Response (TTR) by IRRC Assessment

TTR was defined as the time of participants from the first treatment administration to the first incidence of a confirmed CR (disappearance of all lesions) or PR (at least a 30% decrease in the sum of diameters [SOD] of target lesions, taking as reference the baseline SOD) according to RECIST 1.1, which was modified for this study to follow a maximum of 10 target lesions and a maximum of 5 target lesions per organ. The time from first treatment administration to the first incidence of treatment response was reported as the TTR (NCT03607539)
Timeframe: Trough Database Cutoff Date of 15-Nov-2019 (Up to approximately 16 months)

InterventionMonths (Median)
Sintilimab Combination1.51
Placebo Combination2.63

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 as Assessed by Independent Radiographic Review Committee (IRRC)

PFS was defined as the time from randomization to the first documented PD or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥20% increase in the sum of diameters of target lesions. In addition to the relative increase of 20%, the sum must also have demonstrated an absolute increase of ≥5 mm. The appearance of one or more new lesions was also considered PD (NCT03607539)
Timeframe: Trough Database Cutoff Date of 15-Nov-2019 (Up to approximately 16 months)

InterventionMonths (Median)
Sintilimab Combination8.9
Placebo Combination5.0

[back to top]

Duration of Response (DOR) by IRRC Assessment

(NCT03607539)
Timeframe: From time of first documented evidence of CR or PR trough Database Cutoff Date of 15-Nov-2019 (Up to approximately 16 months)

InterventionMonths (Median)
Sintilimab CombinationNA
Placebo Combination5.52

[back to top]

Overall Survival (OS)

The analysis population consisted of all randomized participants. (NCT03607539)
Timeframe: Trough Database Cutoff Date of 15-Nov-2019 (Up to approximately 16 months)

InterventionMonths (Median)
Sintilimab CombinationNA
Placebo CombinationNA

[back to top]

Objective Response Rate (ORR) by IRRC Assessment

ORR was defined as the percentage of participants in the analysis population who had a confirmed CR (disappearance of all lesions) or PR (at least a 30% decrease in the sum of diameters [SOD] of target lesions, taking as reference the baseline SOD) according to RECIST 1.1, which was modified for this study to follow a maximum of 10 target lesions and a maximum of 5 target lesions per organ. The percentage of participants who experienced a confirmed CR or PR according to RECIST 1.1 as assessed by IRC was reported as the ORR (NCT03607539)
Timeframe: Trough Database Cutoff Date of 15-Nov-2019 (Up to approximately 16 months)

InterventionPercentage of Participants (Number)
Sintilimab Combination51.9
Placebo Combination29.8

[back to top]

Disease Control Rate (DCR) by IRRC Assessment

(NCT03607539)
Timeframe: Trough Database Cutoff Date of 15-Nov-2019 (Up to approximately 16 months)

InterventionPercentage of Participants (Number)
Sintilimab Combination86.8
Placebo Combination75.6

[back to top]

Number of Participants Who Discontinued From Study Treatment Due to an AE

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a study intervention. The number of participants who discontinued treatment due to an AE was assessed. (NCT03631784)
Timeframe: Up to approximately 1 year

InterventionParticipants (Count of Participants)
Pembrolizumab + cCRT + Paclitaxel + Carboplatin48
Pembrolizumab + cCRT + Pemetrexed + Cisplatin26

[back to top]

Percentage of Participants Who Developed Grade 3 or Higher Pneumonitis

Pneumonitis included the MedDRA preferred terms for radiation pneumonitis are acute interstitial pneumonitis, autoimmune lung disease, interstitial lung disease, pneumonitis, idiopathic pneumonia syndrome, organizing pneumonia, and immune-mediated pneumonitis. As per common terminology criteria for Adverse Events, version 4.0, pneumonitis was graded as follows: Grade (Gr) 1- asymptomatic, clinical or diagnostic observations only; intervention not indicated; Gr 2- symptomatic, medical intervention indicated, limiting instrumental activities of daily living (ADL); Gr 3- severe symptoms; limiting self-care activities of daily living (ADL), oxygen indicated; Gr 4- life-threatening respiratory compromise; urgent intervention indicated (e.g., tracheotomy or intubation); Gr 5- death. (NCT03631784)
Timeframe: Up to approximately 3 years

InterventionPercentage of Participants (Number)
Pembrolizumab + cCRT + Paclitaxel + Carboplatin8.0
Pembrolizumab + cCRT + Pemetrexed + Cisplatin6.9

[back to top]

Number of Participants Who Experienced an Adverse Event (AE)

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a study intervention. The number of participants with at least one AE was assessed. (NCT03631784)
Timeframe: Up to approximately 1 1/4 years

InterventionParticipants (Count of Participants)
Pembrolizumab + cCRT + Paclitaxel + Carboplatin108
Pembrolizumab + cCRT + Pemetrexed + Cisplatin101

[back to top]

Overall Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1)

ORR was defined as the percentage of participants who experienced 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) and was assessed using modified RECIST 1.1 by blinded independent central review (BICR). (NCT03631784)
Timeframe: Up to approximately 3 years

InterventionPercentage of Participants (Number)
Pembrolizumab + cCRT + Paclitaxel + Carboplatin71.4
Pembrolizumab + cCRT + Pemetrexed + Cisplatin75.5

[back to top]

Percentage of Participants Discontinuing Study Intervention Due to an AE.

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. The percentage of participants who discontinued the study intervention due to an AE is presented. (NCT03664024)
Timeframe: Up to ~28 months

InterventionPercentage of Participants (Number)
Pembrolizumab Plus Platinum-doublet Chemotherapy38.5

[back to top]

Percentage of Participants Who Experienced One or More Adverse Events (AEs)

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. The percentage of participants who experienced an AE is presented. (NCT03664024)
Timeframe: Up to ~31 months

InterventionPercentage of Participants (Number)
Pembrolizumab Plus Platinum-doublet Chemotherapy100

[back to top]

Overall Survival (OS)

OS is defined as the time from the start of treatment to death due to any cause. The Kaplan-Meier estimate of median PFS using the product-limit (Kaplan-Meier) method for censored data is presented. (NCT03664024)
Timeframe: Up to ~36 months

InterventionMonths (Median)
Pembrolizumab Plus Platinum-doublet Chemotherapy18.1

[back to top]

Objective Response Rate

Objective response rate is the proportion of participants who have a confirmed complete response (CR) or partial response (PR). Objective response rate is assessed by investigator review according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial Response (PR): At least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters. Participants with missing data are considered non-responders. The percentage of participants with an ORR is presented. (NCT03664024)
Timeframe: Up to ~25 months

InterventionPercentage of Participants (Number)
Pembrolizumab Plus Platinum-doublet Chemotherapy40.2

[back to top]

Progression Free Survival (PFS)

PFS is defined as the time from enrollment to the first documented disease progression or death due to any cause, whichever occurs first as assessed by investigator review according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1). The Kaplan-Meier estimate of median PFS using the product-limit (Kaplan-Meier) method for censored data is presented. (NCT03664024)
Timeframe: Up to ~36 months

InterventionMonths (Median)
Pembrolizumab Plus Platinum-doublet Chemotherapy7.2

[back to top]

Tumor Mutation Burden (TMB) in Cell-free Circulating Tumor Deoxyribonucleic Acid (ctDNA)

Cell-free ctDNA allows the exploration of tumor features from blood samples. TMB is a measure of mutational load in tumor cells and expressed as the number of somatic mutations per megabase (mut/MB) of DNA. The mean TMB in cell-free ctDNA of participants is presented. (NCT03664024)
Timeframe: Baseline (Day 1)

InterventionMut/MB (Mean)
Pembrolizumab Plus Platinum-doublet Chemotherapy Overall9
Pembrolizumab Plus Platinum-doublet Chemotherapy Responder8
Pembrolizumab Plus Platinum-doublet Chemotherapy Non-responder10

[back to top]

Number of Participants With Adverse Events

To characterize the toxicity of carboplatin plus pemetrexed plus atezolizumab plus bevacizumab in immunotherapy and chemotherapy-naïve patients with stage IV non-squamous non-small cell lung cancer as defined by the NCI Common Terminology Criteria for Adverse Events (NCI CTCAE) v5. (NCT03713944)
Timeframe: From C1D1 up to a maximum of 20 Months or until death

InterventionParticipants (Count of Participants)
Patient had at least one adverse event of any gradePatient had at least one grade 3 or greater adverse eventPatient had at least one grade 3 or greater treatment related adverse eventPatient had serious adverse event
Arm A30241912

[back to top]

Disease Control Rate

Disease control rate will include complete response (CR], partial response (PR), and stable disease (SD), as per RECIST v1.1 criteria. (NCT03713944)
Timeframe: From C1D1 until death or up to a maximum of 28 months

Interventionpercentage of participants (Number)
Arm A92.9

[back to top]

Overall Response Rate

"Overall Response rate will include confirmed complete response (CR) + confirmed partial response (PR), as determined as per RECIST v1.1 criteria and assessed by the local investigator or designee.~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." (NCT03713944)
Timeframe: From C1D1 until death or up to a maximum of 28 months.

Interventionpercentage of participants (Number)
Arm A42.9

[back to top]

Overall Survival

To estimate the overall survival (OS) of carboplatin plus pemetrexed plus atezolizumab plus bevacizumab in immunotherapy and chemotherapy-naïve patients with stage IV non-squamous non-small cell lung cancer. (NCT03713944)
Timeframe: From C1D1 up to a maximum of 28 Months or until death

Interventionmonths (Median)
Arm A22.4

[back to top]

Progression Free Survival

Progression free survival (PFS) defined as the time from the initiation of treatment to the time when the criteria for disease progression is met as defined by RECIST v1.1 or death of any cause.Disease 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 (NCT03713944)
Timeframe: From C1D1 until progression or death up to maximum of 20 Months

Interventionmonths (Median)
Arm A11.3

[back to top]

Objective Response Rate (ORR), Per Investigator Assessment Using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 Criteria

"ORR= number of subjects with best overall response (BOR) of complete or partial response (CR, PR) divided by number of evaluable subjects. BOR= best response recorded from start of treatment to first progression (PD)/new anticancer therapy, otherwise last follow-up.~CR: disappearance of target and non-target lesions; no new lesions; pathological lymph nodes < 10mm.~PR: 30% decrease in target lesions; non-target lesions not progressed or not evaluated; no new lesions.~PD: 20% increase of target lesions and/or new lesion(s).~ORR was to be compared using Fisher's exact test within each ALK inhibitor treatment arm (each mutation vs. no mutation) and also within the subset of subjects with no mutation comparing each experimental arm vs. pemetrexed. Due to early accrual closure (few subjects), only the number of subjects with BOR of CR or PR are provided, by mutation, no statistical testing. Analysis was to occur after each patient was potentially followed ≥ 24 weeks." (NCT03737994)
Timeframe: Baseline to 24 weeks

,
InterventionParticipants (Count of Participants)
Mutation = I1171: BOR of CR or PRMutation = None: BOR of CR or PR
Brigatinib01
Lorlatinib12

[back to top]

Progression-free Survival (PFS), Per Investigator Assessment Using RECIST v1.1 Criteria

Progression-free survival time is defined as the time from second step registration to the date of the first disease progression / progressive disease (PD), death, or last known follow-up (censored). Progressive disease is defined as a 20% increase of target lesions and/or new lesion(s). Percentage of participants progression-free at a given time (PFS) was to be estimated by the Kaplan-Meier method. Median PFS and PFS at specific time points was to be reported, with 95% confidence intervals, for each mutation (or no mutation)/regimen combination. Due to early accrual closure resulting in few participants, only the number of participants who progressed or died, by mutation, with no statistical testing. (NCT03737994)
Timeframe: Baseline to the date of the first disease progression / progressive disease, death, or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

InterventionParticipants (Count of Participants)
Mutation = I1107: progression or death
LDK378 (Ceritinib)0

[back to top]

Overall Survival (OS)

Survival time is defined as the time from second step registration to the date of death, or last known follow-up (censored). Percentage of participants alive at a given time (OS) was to be estimated by the Kaplan-Meier method. Median OS and OS at specific time points was to be reported, with 95% confidence intervals, for each mutation/regimen combination. Due to early accrual closure resulting in few patients, only the number of participants who died are provided, by mutation, and no statistical testing was done. (NCT03737994)
Timeframe: Baseline to the date of death or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

,
InterventionParticipants (Count of Participants)
Mutation = I1171: deathsMutation = None: deaths
Brigatinib01
Lorlatinib00

[back to top]

Progression-free Survival (PFS), Per Investigator Assessment Using RECIST v1.1 Criteria

Progression-free survival time is defined as the time from second step registration to the date of the first disease progression / progressive disease (PD), death, or last known follow-up (censored). Progressive disease is defined as a 20% increase of target lesions and/or new lesion(s). Percentage of participants progression-free at a given time (PFS) was to be estimated by the Kaplan-Meier method. Median PFS and PFS at specific time points was to be reported, with 95% confidence intervals, for each mutation (or no mutation)/regimen combination. Due to early accrual closure resulting in few participants, only the number of participants who progressed or died, by mutation, with no statistical testing. (NCT03737994)
Timeframe: Baseline to the date of the first disease progression / progressive disease, death, or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

InterventionParticipants (Count of Participants)
Mutation = none: progression or death
Ensartinib2

[back to top]

Objective Response Rate (ORR), Per Investigator Assessment Using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 Criteria

"ORR= number of subjects with best overall response (BOR) of complete or partial response (CR, PR) divided by number of evaluable subjects. BOR= best response recorded from start of treatment to first progression (PD)/new anticancer therapy, otherwise last follow-up.~CR: disappearance of target and non-target lesions; no new lesions; pathological lymph nodes < 10mm.~PR: 30% decrease in target lesions; non-target lesions not progressed or not evaluated; no new lesions.~PD: 20% increase of target lesions and/or new lesion(s).~ORR was to be compared using Fisher's exact test within each ALK inhibitor treatment arm (each mutation vs. no mutation) and also within the subset of subjects with no mutation comparing each experimental arm vs. pemetrexed. Due to early accrual closure (few subjects), only the number of subjects with BOR of CR or PR are provided, by mutation, no statistical testing. Analysis was to occur after each patient was potentially followed ≥ 24 weeks." (NCT03737994)
Timeframe: Baseline to 24 weeks

InterventionParticipants (Count of Participants)
Mutation = None: BOR of CR or PR
Ensartinib0

[back to top]

Progression-free Survival (PFS), Per Investigator Assessment Using RECIST v1.1 Criteria

Progression-free survival time is defined as the time from second step registration to the date of the first disease progression / progressive disease (PD), death, or last known follow-up (censored). Progressive disease is defined as a 20% increase of target lesions and/or new lesion(s). Percentage of participants progression-free at a given time (PFS) was to be estimated by the Kaplan-Meier method. Median PFS and PFS at specific time points was to be reported, with 95% confidence intervals, for each mutation (or no mutation)/regimen combination. Due to early accrual closure resulting in few participants, only the number of participants who progressed or died, by mutation, with no statistical testing. (NCT03737994)
Timeframe: Baseline to the date of the first disease progression / progressive disease, death, or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

,
InterventionParticipants (Count of Participants)
Mutation = I1107: progression or deathMutation = none: progression or death
Brigatinib11
Lorlatinib01

[back to top]

Objective Response Rate (ORR), Per Investigator Assessment Using the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 Criteria

"ORR= number of subjects with best overall response (BOR) of complete or partial response (CR, PR) divided by number of evaluable subjects. BOR= best response recorded from start of treatment to first progression (PD)/new anticancer therapy, otherwise last follow-up.~CR: disappearance of target and non-target lesions; no new lesions; pathological lymph nodes < 10mm.~PR: 30% decrease in target lesions; non-target lesions not progressed or not evaluated; no new lesions.~PD: 20% increase of target lesions and/or new lesion(s).~ORR was to be compared using Fisher's exact test within each ALK inhibitor treatment arm (each mutation vs. no mutation) and also within the subset of subjects with no mutation comparing each experimental arm vs. pemetrexed. Due to early accrual closure (few subjects), only the number of subjects with BOR of CR or PR are provided, by mutation, no statistical testing. Analysis was to occur after each patient was potentially followed ≥ 24 weeks." (NCT03737994)
Timeframe: Baseline to 24 weeks

InterventionParticipants (Count of Participants)
Mutation = I1171: BOR of CR or PR
LDK378 (Ceritinib)0

[back to top]

Duration of Overall Response, Per Investigator Assessment Using RECIST v1.1

"Duration of overall response (DOR) is defined as the time from the first occurrence of a documented BOR of CR or PR to the first date of recorded disease progression (PD) or death from any cause (whichever occurs first). BOR is the best response recorded from the start of treatment to first progression (PD)/new anticancer therapy, otherwise last follow-up.~CR: disappearance of target and non-target lesions; no new lesions; pathological lymph nodes < 10mm.~PR: 30% decrease in target lesions; non-target lesions not progressed or not evaluated; no new lesions.~PD: 20% increase of target lesions and/or new lesion(s).~Median DOR was to be estimated, with 95% confidence intervals, for each mutation/regimen combination using the Kaplan-Meier method. Due to early accrual closure resulting in few subjects, only the mean and range DOR are provided, by mutation, and no statistical testing was done." (NCT03737994)
Timeframe: Baseline to the date of the first disease progression / progressive disease, death, or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

Interventionmonths (Mean)
Mutation = I1171Mutation = None
Lorlatinib27.919.2

[back to top]

Duration of Overall Response, Per Investigator Assessment Using RECIST v1.1

"Duration of overall response (DOR) is defined as the time from the first occurrence of a documented BOR of CR or PR to the first date of recorded disease progression (PD) or death from any cause (whichever occurs first). BOR is the best response recorded from the start of treatment to first progression (PD)/new anticancer therapy, otherwise last follow-up.~CR: disappearance of target and non-target lesions; no new lesions; pathological lymph nodes < 10mm.~PR: 30% decrease in target lesions; non-target lesions not progressed or not evaluated; no new lesions.~PD: 20% increase of target lesions and/or new lesion(s).~Median DOR was to be estimated, with 95% confidence intervals, for each mutation/regimen combination using the Kaplan-Meier method. Due to early accrual closure resulting in few subjects, only the mean and range DOR are provided, by mutation, and no statistical testing was done." (NCT03737994)
Timeframe: Baseline to the date of the first disease progression / progressive disease, death, or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

Interventionmonths (Mean)
Mutation = None
Brigatinib3.5

[back to top]

Number of Participants by Highest Grade Adverse Event Reported

Common Terminology Criteria for Adverse Events (version 4.0) grades adverse event severity from 1=mild to 5=death. Summary data is provided in this outcome measure; see Adverse Events Module for specific adverse event data. (NCT03737994)
Timeframe: Baseline to the date of last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

InterventionParticipants (Count of Participants)Participants (Count of Participants)
Mutation = I110772512695Mutation = I110772512697Mutation = I110772512700Mutation = none72512697Mutation = none72512695Mutation = none72512698
Grade 1Grade 2Grade 3Grade 4Grade 5
Lorlatinib1
LDK378 (Ceritinib)1
LDK378 (Ceritinib)0
Brigatinib0
Lorlatinib0
Lorlatinib2
Ensartinib3
Brigatinib1
Ensartinib0

[back to top]

Overall Survival (OS)

Survival time is defined as the time from second step registration to the date of death, or last known follow-up (censored). Percentage of participants alive at a given time (OS) was to be estimated by the Kaplan-Meier method. Median OS and OS at specific time points was to be reported, with 95% confidence intervals, for each mutation/regimen combination. Due to early accrual closure resulting in few patients, only the number of participants who died are provided, by mutation, and no statistical testing was done. (NCT03737994)
Timeframe: Baseline to the date of death or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

InterventionParticipants (Count of Participants)
Mutation = None: deaths
Ensartinib2

[back to top]

Overall Survival (OS)

Survival time is defined as the time from second step registration to the date of death, or last known follow-up (censored). Percentage of participants alive at a given time (OS) was to be estimated by the Kaplan-Meier method. Median OS and OS at specific time points was to be reported, with 95% confidence intervals, for each mutation/regimen combination. Due to early accrual closure resulting in few patients, only the number of participants who died are provided, by mutation, and no statistical testing was done. (NCT03737994)
Timeframe: Baseline to the date of death or last known follow-up. Maximum follow-up time was 33.6 months, median 8.9 months.

InterventionParticipants (Count of Participants)
Mutation = I1171: deaths
LDK378 (Ceritinib)0

[back to top]

Progression-free Survival

"Progression-free survival (PFS) based on investigator assessments according to Response Evaluation Criteria in Solid Tumours version 1.1.~PFS is defined as time from date of randomization until the date of objective radiological disease progression using Response Evaluation Criteria in Solid Tumours version 1.1 (RECIST 1.1) or death (by any cause in the absence of progression)." (NCT03775486)
Timeframe: From randomization until date of objective radiological disease progression or death, or last evaluable assessment in the absence of progression, up to 18 months

,
InterventionParticipants (Count of Participants)
RECIST progression: Target LesionsRECIST progression: Non Target LesionsRECIST progression: New LesionsDeath in the absence of progressionCensored subjects: Censored RECIST progressionCensored subjects: Censored deathCensored subjects: Progression-free at time of analysisCensored subjects: Progression-free prior to lost to follow-upCensored subjects: Progression-free prior to withdrawal of consentCensored subjects: Progression-free prior to discontinuation due to other reasonCensored subjects: No post-baseline evaluable tumor assessment
Durvalumab/Olaparib Combination Therapy402844801440302
Durvalumab/Placebo Therapy633039900340202

[back to top]

Time to Deterioration in European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-Lung Cancer (LC)13

"Disease-related symptoms assessed by time to deterioration (for maintenance phase) in European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-Lung Cancer (LC)13.~Symptom deterioration is defined as an increase in the score from baseline of less than or equal to 10) that is confirmed at a subsequent assessment, or death (by any cause) in the absence of a clinically meaningful symptom deterioration.~NA is not applicable. The upper confidence limit was not calculable because of an insufficient number of participants with events." (NCT03775486)
Timeframe: From randomization until date of objective radiological disease progression or death, or last evaluable assessment in the absence of progression, up to 18 months

,
Interventionmonths (Median)
EORTC QLQ-LC13: DyspnoeaEORTC QLQ-LC13: CoughingEORTC QLQ-LC13: HaemoptysisEORTC QLQ-LC13: Pain In ChestEORTC QLQ-LC13: Pain In Arm Or ShoulderEORTC QLQ-LC13: Pain In Other Parts
Durvalumab/Olaparib Combination Therapy10.011.715.013.815.010.3
Durvalumab/Placebo Therapy9.710.612.611.59.710.6

[back to top]

Time to Deterioration in EORTC Quality of Life Questionnaire (QLQ) QLQ-C30

"Disease-related symptoms and health-related quality of life (HRQoL) assessed by time to deterioration (for maintenance phase) in EORTC QLQ-C30.~NA is not applicable. The upper confidence limit was not calculable because of an insufficient number of participants with events." (NCT03775486)
Timeframe: From randomization until date of first symptom deterioration that is confirmed, up to 18 months.

,
Interventiontime to deterioration (months) (Median)
EORTC QLQ-C30: FatigueEORTC QLQ-C30: Nausea And VomitingEORTC QLQ-C30: PainEORTC QLQ-C30: DyspnoeaEORTC QLQ-C30: InsomniaEORTC QLQ-C30: Appetite LossEORTC QLQ-C30: ConstipationEORTC QLQ-C30: DiarrhoeaPhysical FunctioningRole FunctioningEmotional FunctioningCognitive FunctioningSocial FunctioningGlobal Health Status/Quality of Life
Durvalumab/Olaparib Combination Therapy8.812.210.212.213.811.712.213.812.010.012.210.29.310.2
Durvalumab/Placebo Therapy1012.69.711.010.611.512.011.512.010.611.010.610.09.7

[back to top] [back to top]

Presence of Anti-drug Antibodies (ADAs) for Durvalumab

Presence of anti-drug antibodies (ADAs) for durvalumab, as assessed at 3, 6, 12, 16 and 20 weeks after start of treatment and every 12 weeks thereafter until 3 and 6 months after last dose of durvalumab (NCT03775486)
Timeframe: Assessed from start of initial therapy up to 2 years.

,
InterventionParticipants (Count of Participants)
ADA prevalence (any ADA positive, baseline or post-baseline)ADA incidence (treatment-induced or treatment-boosted)ADA positive post-baseline and positive at baselineADA positive post-baseline and not detected at baseline (treatment-induced)ADA not detected at post-baseline and positive at baselineTreatment-boosted ADAPersistent positiveTransient positiveNeutralizing anti-drug antibody positive at any visit
Durvalumab/Olaparib Combination Therapy1150560051
Durvalumab/Placebo Therapy941440321

[back to top]

Overall Survival

"Overall survival (OS) across the maintenance phase.~OS is defined as time from date of randomization until the date of death by any cause" (NCT03775486)
Timeframe: From randomization until the date of death due to any cause, up to 18 months.

,
InterventionParticipants (Count of Participants)
DeathCensored participants (still in survival at follow up or terminated study prior to death)
Durvalumab/Olaparib Combination Therapy4490
Durvalumab/Placebo Therapy4590

[back to top]

Change From Baseline in European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ)-Lung Cancer (LC)13

"Disease-related symptoms assessed by change from baseline (for maintenance phase) in EORTC QLQ-LC13. Average adjusted mean over first 11 cycles is presented.~The EORTC QLQ-LC13 was scored according to the published scoring manual. An outcome variable consisting of a score from 0 to 100 was derived for each of the symptom scales in the EORTC QLQ-LC13. Higher scores on symptom scales represent greater symptom severity." (NCT03775486)
Timeframe: From randomization until date of objective radiological disease progression or death, or last evaluable assessment in the absence of progression, up to 18 months

,
Interventionchange from baseline score (Mean)
EORTC QLQ-LC13: DyspnoeaEORTC QLQ-LC13: CoughingEORTC QLQ-LC13: Pain in chest
Durvalumab/Olaparib Combination Therapy-1.27-2.141.31
Durvalumab/Placebo Therapy-0.76-3.093.57

[back to top]

Duration of Response

"Duration of response (DoR) defined as time from the date of first documented response following randomization until the first date of documented progression or death in the absence of disease progression.~Percentage of participants remaining in response at 3, 6, 9 and 12 months estimated using the Kaplan-Meier method." (NCT03775486)
Timeframe: From date of first documented response until objective radiological disease progression or death, up to 18 months.

,
Interventionpercent (Number)
Percentage of participants remaining in response at 3 monthsPercentage of participants remaining in response at 6 monthsPercentage of participants remaining in response at 9 monthsPercentage of participants remaining in response at 12 months
Durvalumab/Olaparib Combination Therapy90.579.169.269.2
Durvalumab/Placebo Therapy85.165.765.765.7

[back to top]

Concentration of Durvalumab

Concentration (pharmacokinetics) of durvalumab (NCT03775486)
Timeframe: Assessed from start of initial therapy up to 2 years.

,
Interventionμg/mL (Geometric Mean)
Cycle 01 Day 01 (Initial Therapy Phase) Post doseCycle 02 Day 01 (Initial Therapy Phase) Pre doseCycle 04 Day 01 (Initial Therapy Phase) Pre doseCycle 01 (Maintenance Phase) Post doseCycle 02 (Maintenance Phase) Pre doseCycle 05 (Maintenance Phase) Pre doseCycle 08 (Maintenance Phase) Pre doseCycle 11 (Maintenance Phase) Pre doseCycle 14 (Maintenance Phase) Pre doseCycle 17 (Maintenance Phase) Pre doseCycle 20 (Maintenance Phase) Pre doseMonth 03 (Maintenance Phase) Pre dose
Durvalumab/Olaparib Combination Therapy417.15276.812155.461535.078159.157166.644198.932210.794276.612264.096528.04612.289
Durvalumab/Placebo Therapy453.72476.959154.947524.306160.315147.848154.057186.092182.042217.137112.27612.769

[back to top]

Change From Baseline in EORTC Quality of Life Questionnaire (QLQ) QLQ-C30

"Disease-related symptoms and health-related quality of life (HRQoL) assessed by change from baseline (for maintenance phase) in EORTC QLQ-C30. Average adjusted mean over first 11 cycles is presented.~The EORTC QLQ-C30 was scored according to the published scoring manual. An outcome variable consisting of a score from 0 to 100 was derived for each of the symptom scales, each of the function scales, and the global health status/QoL scale in the EORTC QLQ-C30. Higher scores on the global health status and function scales indicate better health status/function.~A high scale score represents a higher response level. Thus a high score for a functional scale represents a high / healthy level of functioning, a high score for the global health status / quality of life (QoL) represents a high QoL, but a high score for a symptom scale / item represents a high level of symptomatology / problems." (NCT03775486)
Timeframe: Includes all assessments occurring within the first 12 months of randomization or until disease progression, up to 18 months.

,
Interventionchange from baseline score (Mean)
EORTC QLQ-C30: FatigueEORTC QLQ-C30: Appetite loss
Durvalumab/Olaparib Combination Therapy0.15-0.13
Durvalumab/Placebo Therapy-1.49-3.35

[back to top] [back to top]

Number of Participants With Positive Antidrug Antibodies (ADA)

Serum samples were analyzed by a validated assay method to detect the presence of antidrug antibodies (ADA). Number of participants with positive ADA were reported. (NCT03840915)
Timeframe: Time from first treatment assessed up to approximately 26 months

InterventionParticipants (Count of Participants)
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed9
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxel3
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + Gemcitabine2
Cohort D: Bintrafusp Alfa + Docetaxel3

[back to top]

Number of Participants With Dose-Limiting Toxicities (DLTs)

DLT was defined as Adverse Events(AEs) with any of following toxicities: Grade 4 nonhematologic toxicity or hematologic toxicity lasting more than equal to(>=) 7 days despite medical intervention; Grade 3 nausea, vomiting, and diarrhea lasting >= 3 days despite supportive care; Any Grade 3 or Grade 4 nonhematologic lab value leading to hospitalization or persisting for >= 7 days; Grade 3 or Grade 4: grade 3 is defined as absolute neutrophil count (ANC) less than (<) 1,000/Cubic Millimeter(mm3) with a temperature of > 38.3 degree Celsius (°C); grade 4 is defined as ANC < 1,000/mm3 with a temperature of > 38.3°C, with life-threatening consequences; Thrombocytopenia < 25,000/mm3 associated with bleeding not resulting in hemodynamic instability or a life-threatening bleeding resulting in urgent intervention; Bleeding events >= Grade 3 occurring within 5 days of bintrafusp alfa treatment; Prolonged delay(> 3 weeks) in initiating Cycle 2 due to treatment-related toxicity; Grade 5 toxicity. (NCT03840915)
Timeframe: Day 1 Week 1 up to Week 3

InterventionParticipants (Count of Participants)
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed1
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxel1
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + Gemcitabine0
Cohort D: Bintrafusp Alfa + Docetaxel3

[back to top]

Duration of Response (DOR)

DOR was defined for participants with confirmed response, as the time from first documentation of confirmed objective response (Complete Response [CR] or Partial Response [PR]) according to RECIST 1.1 to the date of first documentation of progression disease (PD) or death due to any cause, whichever occurred first. CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the SLD of all lesions. PD: At least a 20 percent (%) increase in the SLD, taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. Results were calculated based on Kaplan-Meier estimates. (NCT03840915)
Timeframe: Time from first documentation of a confirmed objective response to PD or death due to any cause (assessed up to approximately 26 months)

Interventionmonths (Median)
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed9.6
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxelNA
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + Gemcitabine10.5
Cohort D: Bintrafusp Alfa + Docetaxel3.4

[back to top]

Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Serious TEAEs

An AE was defined as any untoward medical occurrence in a participant administered a pharmaceutical product and which did not necessarily have a causal relationship with this treatment. An AE could therefore be any unfavorable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether considered related to the medicinal product or protocol-specified procedure. Serious AE was defined AE that resulted in any of the following outcomes: death; life threatening; persistent/significant disability/incapacity; initial/prolonged inpatient hospitalization; congenital anomaly/birth defect. TEAE was defined as events with onset date or worsening during the on-treatment period. TEAEs included serious TEAEs and non-serious TEAEs. (NCT03840915)
Timeframe: Time from first treatment assessed up to approximately 26 months

,,,
InterventionParticipants (Count of Participants)
TEAEsSerious TEAEs
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed4031
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxel95
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + Gemcitabine96
Cohort D: Bintrafusp Alfa + Docetaxel129

[back to top]

Percentage of Participants With Confirmed Objective Response According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) as Assessed by Investigator (IRC)

Percentage of participants with confirmed objective response that is at least one overall assessment of complete response (CR) or partial response (PR) reported here. CR: Disappearance of all evidence of target and non-target lesions. PR: At least 30% reduction from baseline in the sum of the longest diameter (SLD) of all lesions. Confirmed CR = at least 2 determinations of CR at least 4 weeks apart and before progression. Confirmed PR = at least 2 determinations of PR at least 4 weeks apart and before progression (and not qualifying for a CR). Confirmed objective response was determined according to RECIST v1.1 and as adjudicated by Investigator. (NCT03840915)
Timeframe: Time from first treatment assessed up to approximately 26 months

InterventionPercentage of Participants (Number)
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed45.0
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxel66.7
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + Gemcitabine44.4
Cohort D: Bintrafusp Alfa + Docetaxel16.7

[back to top]

Overall Survival (OS)

OS was defined as the time from first administration of study intervention to the date of death due to any cause. The OS was analyzed by using the Kaplan-Meier method. (NCT03840915)
Timeframe: Time from first treatment assessed up to approximately 26 months

Interventionmonths (Median)
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed11.4
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxel11.8
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + GemcitabineNA
Cohort D: Bintrafusp Alfa + Docetaxel16.5

[back to top]

Progression-Free Survival (PFS) According to Response Evaluation Criteria in Solid Tumors (RECIST Version 1.1) Assessed by Investigator

PFS was defined as the time from first administration of study intervention until date of the first documentation of disease progression (PD) or death due to any cause, whichever occurred first. PD: At least a 20 percent (%) increase in the SLD, taking as reference the smallest SLD recorded from baseline or the appearance of 1 or more new lesions. Kaplan-Meier estimates was used to calculate PFS. (NCT03840915)
Timeframe: Time from first administration of study drug until the first documentation of PD or death, assessed up to approximately 26 months

Interventionmonths (Median)
Cohort A: Bintrafusp Alfa + Cisplatin/Carboplatin + Pemetrexed5.0
Cohort B: Bintrafusp Alfa + Carboplatin + Paclitaxel or Nab-paclitaxel4.1
Cohort C: Bintrafusp Alfa + Cisplatin/Carboplatin + Gemcitabine5.4
Cohort D: Bintrafusp Alfa + Docetaxel2.6

[back to top]

Number of Participants Who Discontinued Study Treatment Due to an Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a participant administered a study treatment and which does not necessarily have to have a causal relationship with this treatment. An AE could therefore be any unfavorable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the study treatment or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a pre-existing condition that was temporally associated with the use of study treatment, was also an AE. The number of participants who discontinued study treatment due to an AE is presented. (NCT03850444)
Timeframe: Up to 56.7 months

InterventionParticipants (Count of Participants)
Pembrolizumab26
Chemotherapy (SOC Treatment)22

[back to top]

Number of Participants Who Experienced At Least One Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a participant administered a study treatment and which does not necessarily have to have a causal relationship with this treatment. An AE could therefore be any unfavorable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the study treatment or protocol-specified procedure. Any worsening (i.e., any clinically significant adverse change in frequency and/or intensity) of a pre-existing condition that was temporally associated with the use of study treatment, was also an AE. The number of participants who experienced at least one AE is presented. (NCT03850444)
Timeframe: Up to 59.7 months

InterventionParticipants (Count of Participants)
Pembrolizumab126
Chemotherapy (SOC Treatment)124

[back to top]

Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%

ORR was determined for participants with a TPS of ≥1%. ORR was determined per RECIST 1.1 and was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target and non-target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The percentage of participants who had a TPS ≥1% and who experienced a CR or PR is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionPercentage of participants (Number)
Pembrolizumab32.8
Chemotherapy (SOC Treatment)24.6

[back to top]

Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%

ORR was determined for participants with a TPS of ≥20%. ORR was determined per RECIST 1.1 and was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target and non-target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The percentage of participants who had a TPS ≥20% and who experienced a CR or PR is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionPercentage of participants (Number)
Pembrolizumab35.6
Chemotherapy (SOC Treatment)24.3

[back to top]

Objective Response Rate (ORR) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%

ORR was determined for participants with a TPS of ≥50%. ORR was determined per RECIST 1.1 and was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target and non-target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The percentage of participants who had a TPS ≥50% and who experienced a CR or PR is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionPercentage of participants (Number)
Pembrolizumab41.7
Chemotherapy (SOC Treatment)24.3

[back to top]

Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥1%

OS was determined for participants with a TPS of ≥1% and was defined as the time from randomization until death due to any cause. Participants without documented death at the time of the analysis were censored at the date of the last follow-up. The OS was calculated using the product-limit (Kaplan-Meier) method for censored data. The OS for participants with a TPS ≥1% is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionMonths (Median)
Pembrolizumab20.0
Chemotherapy (SOC Treatment)13.7

[back to top]

Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥20%

OS was determined for participants with a TPS of ≥20% and was defined as the time from randomization until death due to any cause. Participants without documented death at the time of the analysis were censored at the date of the last follow-up. The OS was calculated using the product-limit (Kaplan-Meier) method for censored data. The OS for participants with a TPS ≥20% is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionMonths (Median)
Pembrolizumab20.0
Chemotherapy (SOC Treatment)13.7

[back to top]

Overall Survival (OS) in Participants With a Tumor Proportion Score (TPS) of ≥50%

OS was determined for participants with a TPS of ≥50% and was defined as the time from randomization until death due to any cause. Participants without documented death at the time of the analysis were censored at the date of the last follow-up. The OS was calculated using the product-limit (Kaplan-Meier) method for censored data. The OS for participants with a TPS ≥50% is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionMonths (Median)
Pembrolizumab20.0
Chemotherapy (SOC Treatment)14.0

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥1%

PFS was determined for participants with a TPS of ≥1% and was defined as the time from randomization until the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. The appearance of one or more new lesions or progression in non-target lesions was also considered PD. The PFS per RECIST 1.1 was calculated using the product-limit (Kaplan-Meier) method for censored data. The PFS for participants with a TPS ≥1% is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionMonths (Median)
Pembrolizumab6.3
Chemotherapy (SOC Treatment)6.4

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥20%

PFS was determined for participants with a TPS of ≥20% and was defined as the time from randomization until the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. The appearance of one or more new lesions or progression in non-target lesions was also considered PD. The PFS per RECIST 1.1 was calculated using the product-limit (Kaplan-Meier) method for censored data. The PFS for participants with a TPS ≥20% is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionMonths (Median)
Pembrolizumab6.3
Chemotherapy (SOC Treatment)6.5

[back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST 1.1) as Assessed by Blinded Independent Central Review (BICR) in Participants With a Tumor Proportion Score (TPS) of ≥50%

PFS was determined for participants with a TPS of ≥50% and was defined as the time from randomization until the first documented progressive disease (PD) or death due to any cause, whichever occurred first. Per RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. The appearance of one or more new lesions or progression in non-target lesions was also considered PD. The PFS per RECIST 1.1 was calculated using the product-limit (Kaplan-Meier) method for censored data. The PFS for participants with a TPS ≥50% is presented. (NCT03850444)
Timeframe: Up to 23.2 months

InterventionMonths (Median)
Pembrolizumab8.3
Chemotherapy (SOC Treatment)6.5

[back to top]

Overall Response Rate (ORR) Per RECIST 1.1 as Assessed by Blinded Central Imaging

ORR was defined as the percentage of participants in the analysis population who had a Complete Response (CR: Disappearance of all target lesions) or a Partial Response (PR: ≥30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters) per RECIST 1.1. The percentage of participants who experienced a CR or PR is presented. (NCT03950674)
Timeframe: Through Database Cutoff Date of 20-May-2019 (Up to approximately 31.2 months)

InterventionPercentage of Participants (Number)
Pembrolizumab56.0
Control33.3

[back to top]

Duration of Response (DOR) Per RECIST 1.1 as Assessed by Blinded Central Imaging

For participants who demonstrated a confirmed CR (disappearance of all target lesions) or 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 a CR or PR until PD or death. DOR for participants who had not progressed or died at the time of analysis was censored at the date of their last tumor assessment. Per RECIST 1.1, PD was defined as ≥20% increase in the sum of diameters of target lesions. In addition to the relative increase of 20%, the sum must also have demonstrated an absolute increase of ≥5 mm. Note: The appearance of one or more new lesions was also considered PD. DOR assessments were based on blinded central imaging review with confirmation. The DOR per RECIST 1.1 for all participants who experienced a confirmed CR or PR is presented. (NCT03950674)
Timeframe: From time of first documented evidence of CR or PR through database cutoff date of 20-May-2019 (Up to approximately 31.2 months)

InterventionMonths (Median)
Pembrolizumab13.6
Control9.7

[back to top]

Number of Participants Who Experienced an Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a study participant administered study drug and which does not necessarily have to have a causal relationship with this study drug. The number of participants who experienced an AE is presented. (NCT03950674)
Timeframe: Through Database Cutoff Date of 20-May-2019 (Up to approximately 31.2 months)

InterventionParticipants (Count of Participants)
Pembrolizumab25
Control15

[back to top]

Overall Survival (OS)

OS was defined as the time from randomization to death due to any cause. Participants without documented death at the time of the analysis were censored at the date of the last follow-up. The OS is presented. (NCT03950674)
Timeframe: Through Database Cutoff Date of 20-May-2019 (Up to approximately 31.2 months)

InterventionMonths (Median)
PembrolizumabNA
Control25.9

[back to top] [back to top]

Progression-Free Survival (PFS) Per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 as Assessed by Blinded Central Imaging

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 RECIST 1.1, PD was defined as ≥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 also demonstrate an absolute increase of ≥5 mm. Note: The appearance of one or more new lesions was also considered PD. The PFS per RECIST 1.1 is presented. (NCT03950674)
Timeframe: Through Database Cutoff Date of 20-May-2019 (Up to approximately 31.2 months)

InterventionMonths (Median)
Pembrolizumab16.5
Control7.1

[back to top]

Number of Participants Who Discontinued Any Study Drug Due to an AE

The number of participants who discontinued any randomized study drug due to an AE is presented. (NCT03950674)
Timeframe: Up to approximately 24 months

InterventionParticipants (Count of Participants)
Pembrolizumab9
Control3

[back to top]

Disease Control Rate (DCR)

"Percentage of participants with complete response (CR), partial response (PR), or stable disease as best response.~CR: Complete disappearance of all target and non-target lesions. No new lesions. No disease related symptoms. Any lymph nodes must have reduction in short axis to <1.0cm. All disease must be assessed using the same technique as baseline.~PR: Applies only to participants with at least one measurable lesion. At least 30% decrease under baseline of the sum of appropriate diameters of all target measurable lesions. No unequivocal progression of non-measurable disease. No new lesions. All target measurable lesions must be assessed using the same techniques as baseline.~Stable disease: Does not qualify for CR, PR, Progression or Symptomatic Deterioration. All target measurable lesions must be assessed using the same techniques as baseline." (NCT03971474)
Timeframe: From date of registration to a maximum of 3 years or death

InterventionPercentage of participants (Number)
Standard of Care (Inv. Choice)73
Ramucirumab + Pembrolizumab75

[back to top]

Duration of Response (DOR)

Time from date of first documentation of response (complete or partial) to date of first documentation of progression assessed by local review or symptomatic deterioration, or death due to any cause among participants who achieve a response. Participants last known to be alive without report of progression are censored at date of last disease assessment. For participants with a missing scan (or consecutive missing scans) whose subsequent scan determines progression, the expected date of the first missing scan (as defined by the disease assessment schedule) is used as the date of progression. (NCT03971474)
Timeframe: From date of registration to a maximum of 3 years or death

Interventionmonths (Median)
Standard of Care (Inv. Choice)5.6
Ramucirumab + Pembrolizumab12.9

[back to top]

Investigator Assessed-progression-free Survival (IA-PFS)

"Time from date of registration to date of first documentation of progression assessed by local review or symptomatic deterioration, or death due to any cause. Participants last known to be alive without report of progression are censored at date of last disease assessment. For participants with a missing scan (or consecutive missing scans) whose subsequent scan determines progression, the expected date of the first missing scan is used as the date of progression.~Symptomatic deterioration: Global deterioration of health status requiring discontinuation of treatment without objective evidence of progression. Efforts should be made to obtain objective evidence of progression after discontinuation." (NCT03971474)
Timeframe: From date of registration to a maximum of 3 years or death

Interventionmonths (Median)
Standard of Care (Inv. Choice)5.2
Ramucirumab + Pembrolizumab4.5

[back to top]

Overall Survival (OS), Subgroup Analysis by Stratification Factors

Number of participants who died due to any cause. Participants last known to be alive are censored at date of last contact. (NCT03971474)
Timeframe: From date of registration to a maximum of 3 years or death

,
InterventionParticipants (Number)
PD-L1: < 1%PD-L1: >= 1%Histology: SquamousHistology: Non-squamous
Ramucirumab + Pembrolizumab21191827
Standard of Care (Inv. Choice)21272427

[back to top] [back to top]

Investigator-Assessed Progression-Free Survival (IA-PFS), Subgroup Analysis by Stratification Factors

"Number of participants with progression assessed by local review or symptomatic deterioration, or death due to any cause. Participants last known to be alive without report of progression are censored at date of last disease assessment. For participants with a missing scan (or consecutive missing scans) whose subsequent scan determines progression, the expected date of the first missing scan is used as the date of progression.~Symptomatic deterioration: Global deterioration of health status requiring discontinuation of treatment without objective evidence of progression. Efforts should be made to obtain objective evidence of progression after discontinuation." (NCT03971474)
Timeframe: From date of registration to a maximum of 3 years or death

,
InterventionParticipants (Number)
Histology: Non-squamousHistology: SquamousPD-L1: < 1%PD-L1: >= 1%
Ramucirumab + Pembrolizumab34232724
Standard of Care (Inv. Choice)34282534

[back to top]

Response Rate (RR)

"Percentage of participants with a complete or partial, confirmed or unconfirmed response.~Complete Response (CR): Complete disappearance of all target and non-target lesions. No new lesions. No disease related symptoms. Any lymph nodes (whether target or non-target) must have reduction in short axis to < 1.0 cm. All disease must be assessed using the same technique as baseline.~Partial Response (PR): Applies only to participants with at least one measurable lesion. Greater than or equal to 30% decrease under baseline of the sum of appropriate diameters of all target measurable lesions. No unequivocal progression of non-measurable disease. No new lesions. All target measurable lesions must be assessed using the same techniques as baseline." (NCT03971474)
Timeframe: From date of registration to a maximum of 3 years or death

InterventionPercentage of participants (Number)
Standard of Care (Inv. Choice)28
Ramucirumab + Pembrolizumab22

[back to top]

Overall Survival (OS)

Time from date of registration to date of death due to any cause. Participants last known to be alive are censored at date of last contact. (NCT03971474)
Timeframe: From date of registration to a maximum of 2 years and 11 months or death

Interventionmonths (Median)
Standard of Care (Inv. Choice)11.6
Ramucirumab + Pembrolizumab14.5

[back to top]

the Maximum Tolerated Dose (MTD)

The primary endpoint was the MTD of anlotinib, at which less than 33% of patients experienced a DLT in the frst treatment cycle. A DLT involving hematological toxicity was defned as grade 4 and above, non-hematological toxicity as grade 3 and above, and liver and kidney function injury as grade 2 and above. (NCT04012619)
Timeframe: 1 month

Interventionmg (Number)
Anlotinib Hydrochloride Combined With AP10

[back to top]

Response Rate

Will be evaluated using the method of Kaplan-Meier. Confidence intervals for median times will be determined using the Brookmeyer-Crowley method. Confidence intervals around landmark times will be determined using Greenwood's formula for the variance and based on a log-log transformation applied on the survival function. Binary proportions will be calculated with associated confidence intervals for binary outcomes, such as response. Means and/or medians will be calculated for continuous outcomes. Confidence bounds will be provided for means and quartiles and ranges for median values. All confidence bounds will be presented as 95% bounds. (NCT04372927)
Timeframe: Through study completion (up to 4 months)

Interventionparticipants (Number)
Treatment (Chemotherapy, Durvalumab, Radiation Therapy)0

[back to top]

Frequency and Severity of Pneumonitis

The primary toxicity of interest is grade 3 or higher pneumonitis. The incidence of grade 3 or worse pneumonitis attributable to treatment will be evaluated and compared against the PACIFIC trial results. All toxicities of all grades will be monitored on study and reported. Binary proportions will be calculated with associated confidence intervals for binary outcomes, such as toxicity. (NCT04372927)
Timeframe: Through study completion (up to 4 months)

Interventionparticipants (Number)
Treatment (Chemotherapy, Durvalumab, Radiation Therapy)1

[back to top]

Frequency of Adverse Events

Frequency and severity of toxicities will be graded with Common Terminology Criteria for Adverse Events (CTCAE), version 5. Toxicities will be summarized as the proportion of patients with such toxicities, in addition to total number of toxicities (allowing for multiple toxicities within a patient) among all patients. All toxicities of all grades will be monitored on study and reported. Binary proportions will be calculated with associated confidence intervals for binary outcomes, such as toxicity. (NCT04372927)
Timeframe: Through study completion (up to 4 months)

Interventionparticipants (Number)
Treatment (Chemotherapy, Durvalumab, Radiation Therapy)1

[back to top]

Overall Survival (OS)

Will be evaluated using the method of Kaplan-Meier. Confidence intervals for median times will be determined using the Brookmeyer-Crowley method. Confidence intervals around landmark times will be determined using Greenwood's formula for the variance and based on a log-log transformation applied on the survival function. Binary proportions will be calculated with associated confidence intervals for binary outcomes, such as response. Means and/or medians will be calculated for continuous outcomes. Confidence bounds will be provided for means and quartiles and ranges for median values. All confidence bounds will be presented as 95% bounds. (NCT04372927)
Timeframe: From study registration to death due to any cause

Interventionmonths (Mean)
Treatment (Chemotherapy, Durvalumab, Radiation Therapy)4

[back to top]

Run-in Part: Number of Participants With Dose Limiting Toxicities (DLTs)

A DLT was defined as an adverse event (AE) or abnormal laboratory value assessed as unrelated to disease, progressive disease, inter-current illness, or concomitant medications, that despite optimal therapeutic intervention that occurred within the first 21 days of treatment with capmatinib in combination with osimertinib. (NCT04816214)
Timeframe: Up to 21 Days

InterventionParticipants (Count of Participants)
Run-in Part: Capmatinib + Osimertinib0

[back to top]

Run-in Part: Maximum Plasma Concentration (Cmax) of Osimertinib and Its Metabolites (AZ5104 and AZ7550)

Blood samples were collected. Cmax of osimertinib and its metabolites (AZ5104 and AZ7550) was calculated from plasma concentration-time data using non-compartmental methods and summarized using descriptive statistics. (NCT04816214)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 8 and 24 hours post-dose on Days 1 and 15 of Cycle 1 (Cycle = 21 days)

Interventionng/mL (Mean)
Osimertinib: Day 1Osimertinib: Day 15AZ5104: Day 1AZ5104: Day 15AZ7550: Day 1AZ7550: Day 15
Run-in Part: Capmatinib + Osimertinib99.52654.8622.03.6133.0

[back to top]

Run-in Part: Disease Control Rate (DCR) as Per Investigator Assessment

DCR was defined as the percentage of participants with a BOR of CR, PR, and stable disease (SD) as per investigator judgment and according to RECIST v1.1. Criteria for CR: Disappearance of all lesions and pathologic lymph nodes; PR: ≥30% decrease in SLD of the target lesions or no new lesions or no progression of non-target lesions; SD: Neither sufficient shrinkage to qualify for PR or CR nor and increase in lesions which would qualify for PD. PD: ≥20% increase in SLD compared to the smallest SLD in the study, or progression of non-target lesions or new lesions. (NCT04816214)
Timeframe: From randomization up to end of study, assessed up to 39 weeks

Interventionpercentage of participants (Number)
Run-in Part: Capmatinib + Osimertinib66.7

[back to top]

Run-in Part: Duration of Response (DOR) as Per Investigator Assessment

DOR was defined as the time from first documented response of CR or PR to the date of first documented PD or death due to any cause. Criteria for CR: Disappearance of all lesions and pathologic lymph nodes; PR: ≥30% decrease in the SLD of the target lesions or no new lesions or no progression of non-target lesions; PD: ≥20% increase in SLD compared to the smallest SLD in the study, or progression of non-target lesions or new lesions. (NCT04816214)
Timeframe: Up to disease progression or death or end of study, assessed up to 39 weeks

Interventionmonths (Median)
Run-in Part: Capmatinib + Osimertinib6.93

[back to top]

Run-in Part: Time to Response (TTR) as Per Investigator Assessment

TTR was defined as the duration of time between the date of fist dose of treatmwnr and the date of the first documented response of either CR or PR as per investigator judgment and according to RECIST v1.1 criteria. Criteria for CR: Disappearance of all lesions and pathologic lymph nodes; PR: ≥30% decrease in SLD of the target lesions or no new lesions or no progression of non-target lesions. (NCT04816214)
Timeframe: From first dose of treatment up to end of study, assessed up to 39 weeks

Interventionmonths (Median)
Run-in Part: Capmatinib + OsimertinibNA

[back to top]

Run-in Part: Median Duration of Exposure to Each Study Drug

Duration of exposure is defined as the time (in weeks) between the first and the last dose of study treatment. (NCT04816214)
Timeframe: From the first dose until last dose of study treatment, assessed up to 39 weeks

Interventionweeks (Median)
CapmatinibOsimertinib
Run-in Part: Capmatinib + Osimertinib23.524.0

[back to top]

Run-in Part: Number of Participants With at Least One Dose Interruption and Dose Reduction of Each Study Drug

Number of participants with at least one dose interruption and dose reduction were reported for each study drug. (NCT04816214)
Timeframe: From the first dose until last dose of study treatment, assessed up to 39 weeks

InterventionParticipants (Count of Participants)
Dose Interruption: CapmatinibDose Interruption: OsimertinibDose Reduction: CapmatinibDose Reduction: Osimertinib
Run-in Part: Capmatinib + Osimertinib5430

[back to top]

Run-in Part: Time to Maximum Plasma Concentration (Tmax) of Capmatinib

"Blood samples were collected. Tmax of capmatinib was calculated from plasma concentration-time data using non-compartmental methods and summarized using descriptive statistics.~Actual recorded sampling times were considered for the calculations" (NCT04816214)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, and 8 hours post-dose on Days 1 and 15 of Cycle 1 (Cycle = 21 days)

Interventionhours (Median)
Day 1Day 15
Run-in Part: Capmatinib + Osimertinib1.921.00

[back to top]

Run-in Part: Time to Maximum Plasma Concentration (Tmax) of Osimertinib and Its Metabolites (AZ5104 and AZ7550)

"Blood samples were collected. Tmax of osimertinib and its metabolites (AZ5104 and AZ7550) was calculated from plasma concentration-time data using non-compartmental methods and summarized using descriptive statistics.~Actual recorded sampling times were considered for the calculations" (NCT04816214)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 8 and 24 hours post-dose on Days 1 and 15 of Cycle 1 (Cycle = 21 days)

Interventionhours (Median)
Osimertinib: Day 1Osimertinib: Day 15AZ5104: Day 1AZ5104: Day 15AZ7550: Day 1AZ7550: Day 15
Run-in Part: Capmatinib + Osimertinib6.004.0023.14.005.682.25

[back to top]

Run-in Part: Maximum Plasma Concentration (Cmax) of Capmatinib

"Blood samples were collected. Cmax of capmatinib was calculated from plasma concentration-time data using non-compartmental methods and summarized using descriptive statistics.~Actual recorded sampling times were considered for the calculations" (NCT04816214)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, and 8 hours post-dose on Days 1 and 15 of Cycle 1 (Cycle = 21 days)

Interventionnanograms per milliliter (ng/mL) (Mean)
Day 1Day 15
Run-in Part: Capmatinib + Osimertinib55105750

[back to top]

Run-in Part: Progression-Free Survival (PFS) as Per Investigator Assessment

PFS was defined as the time (in months) from first dose of treatment to the date of the first documented progression or death due to any cause as per investigator judgment and according to RECIST v1.1. Progression was defined as a ≥20% increase in SLD compared to smallest SLD in the study, or progression of non-target lesions or new lesions. PFS was censored if no PFS event (progression or death) was observed. (NCT04816214)
Timeframe: From first dose of treatment until first documented progression or death or end of study, assessed up to 39 weeks

Interventionmonths (Median)
Run-in Part: Capmatinib + Osimertinib4.58

[back to top]

Run-in Part: Dose Intensity of Each Study Drug

Dose intensity was computed as the ratio of actual cumulative dose (milligrams) received and actual duration of exposure (weeks) to study drug. (NCT04816214)
Timeframe: From the first dose until last dose of study treatment, assessed up to 39 weeks

Interventionmilligrams per week (mg/week) (Mean)
CapmatinibOsimertinib
Run-in Part: Capmatinib + Osimertinib5042.8534.9

[back to top]

Run-in Part: Overall Response Rate (ORR) as Per Investigator Assessment

ORR was defined as the percentage of participants with a confirmed best overall response (BOR) of complete response (CR) or partial response (PR), as per investigator judgment and according to RECIST v1.1. Criteria for CR: Disappearance of all lesions and pathologic lymph nodes; PR: ≥30% decrease in the SLD of the target lesions or no new lesions or no progression of non-target lesions. (NCT04816214)
Timeframe: Up to end of study, assessed up to 39 weeks

Interventionpercentage of participants (Number)
Run-in Part: Capmatinib + Osimertinib50.0

[back to top]

Run-in Part: Area Under the Curve to the Last Measurable Concentration (AUClast) of Osimertinib and Its Metabolites (AZ5104 and AZ7550)

Blood samples were collected. AUClast of osimertinib and its metabolites (AZ5104 and AZ7550) was calculated from plasma concentration-time data using non-compartmental methods and summarized using descriptive statistics. (NCT04816214)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, 8 and 24 hours post-dose on Days 1 and 15 of Cycle 1 (Cycle = 21 days)

Interventionng*hr/mL (Mean)
Osimertinib: Day 1Osimertinib: Day 15AZ5104: Day 1AZ5104: Day 15AZ7550: Day 1AZ7550: Day 15
Run-in Part: Capmatinib + Osimertinib1790138082.913647.1112

[back to top]

Run-in Part: Area Under the Curve to the Last Measurable Concentration (AUClast) of Capmatinib

Blood samples were collected. AUClast of capmatinib was calculated from plasma concentration-time data using non-compartmental methods and summarized using descriptive statistics. (NCT04816214)
Timeframe: Pre-dose, 0.5, 1, 2, 3, 4, 6, and 8 hours post-dose on Days 1 and 15 of Cycle 1 (Cycle = 21 days)

Interventionnanograms*hours/milliliter (ng*hr/mL) (Mean)
Day 1Day 15
Run-in Part: Capmatinib + Osimertinib2010021300

[back to top]