Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT03164616
Other study ID # D419MC00004
Secondary ID 2017-000920-81
Status Active, not recruiting
Phase Phase 3
First received
Last updated
Start date June 1, 2017
Est. completion date May 28, 2025

Study information

Verified date February 2024
Source AstraZeneca
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This is a randomized, open-label, multi-center, global, Phase III study to determine the efficacy and safety of durvalumab + tremelimumab combination therapy + Standard of care (SoC) chemotherapy or durvalumab monotherapy + SoC chemotherapy versus SoC chemotherapy alone as first line treatment in patients with metastatic non small-cell lung cancer (NSCLC) with tumors that lack activating epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) fusions.


Description:

Adult patients with a histologically or cytologically documented metastatic NSCLC, with tumors that lack activating EGFR mutations and ALK fusions, are eligible for enrollment. Patients will be randomized in a 1:1:1 ratio to receive treatment with durvalumab + tremelimumab combination therapy + SoC chemotherapy, durvalumab monotherapy + SoC chemotherapy, or SoC chemotherapy alone. Tumor evaluation scans will be performed until objective disease progression as efficacy assessment. All patients will be followed for survival until the end of the study. An independent data monitoring committee (IDMC) composed of independent experts will be convened to confirm the safety and tolerability of the proposed dose and schedule.


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 1186
Est. completion date May 28, 2025
Est. primary completion date March 12, 2021
Accepts healthy volunteers No
Gender All
Age group 18 Years to 130 Years
Eligibility Inclusion Criteria: For inclusion in the study, patients should fulfill the following criteria: 1. Aged at least 18 years. 2. Histologically or cytologically documented Stage IV NSCLC. 3. Confirmed tumor PD-L1 status prior to randomization. 4. Patients must have tumors that lack activating EGFR mutations and ALK fusions. 5. No prior chemotherapy or any other systemic therapy for metastatic NSCLC. 6. World Health Organization (WHO)/Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. 7. No prior exposure to immunemediated therapy, excluding therapeutic anticancer vaccines. Exclusion Criteria: Patients should not enter the study if any of the following exclusion criteria are fulfilled: 1. Mixed small-cell lung cancer and NSCLC histology, sarcomatoid variant. 2. Active or prior documented autoimmune or inflammatory disorders. 3. Brain metastases or spinal cord compression unless the patient's condition is stable and off steroids. 4. Active infection including tuberculosis, hepatitis B, hepatitis C, or human immunodeficiency virus.

Study Design


Intervention

Drug:
Durvalumab
IV infusions every 3 weeks for 12 weeks (4 cycles) and every 4 weeks thereafter until disease progression or other discontinuation criteria
Tremelimumab
IV infusions every 3 weeks for 12 weeks (4 cycles). An additional dose of tremelimumab will be administered in the week 16.
Abraxane + carboplatin
Standard of care chemotherapy (squamous and non-squamous patients): Abraxane 100 mg/m2 on Days 1, 8, and 15 of each 21-day cycle. Carboplatin Area under the plasma drug concentration-time curve (AUC) 5 or 6 via IV infusion on Day 1 of each 21-day cycle for 4 to 6 cycles (ie, 4 cycles for Treatment Arms 1 and 2 and 4 to 6 cycles for Treatment Arm 3).
Gemcitabine + cisplatin
Standard of care chemotherapy (squamous patients only): Gemcitabine 1000 or 1250 mg/m2 via IV infusion on Days 1 and 8 of each 21-day cycle + cisplatin 75 mg/m2 via IV infusion on Day 1 of each 21-day cycle, for 4 to 6 cycles (ie, 4 cycles for Treatment Arms 1 and 2 and 4 to 6 cycles for Treatment Arm 3).
Gemcitabine + carboplatin
Standard of care chemotherapy (squamous patients only): Gemcitabine 1000 or 1250 mg/m2 via IV infusion on Days 1 and 8 of each 21-day cycle + carboplatin AUC 5 or 6 via IV infusion on Day 1 of each 21-day cycle for 4 to 6 cycles (ie, 4 cycles for Treatment Arms 1 and 2 and 4 to 6 cycles for Treatment Arm 3).
Pemetrexed + carboplatin
Standard of care chemotherapy (non-squamous patients only): Pemetrexed 500 mg/m2 and carboplatin AUC 5 or 6 via IV infusion on Day 1 of each 21-day cycle for 4 to 6 cycles (ie, 4 cycles for Treatment Arms 1 and 2 and 4 to 6 cycles for Treatment Arm 3); then continue pemetrexed 500 mg/m2 maintenance [i.e., q4w for Treatment Arms 1 and 2. For Treatment Arm 3, Pemetrexed maintenance therapy can be given either q3w or q4w (dependent on Investigator decision and local standards)] until objective disease progression.
Pemetrexed + cisplatin
Standard of care chemotherapy (non-squamous patients only): Pemetrexed 500 mg/m2 and cisplatin 75 mg/m2 via IV infusion on Day 1 of each 21-day cycle, for 4 to 6 cycles (ie, 4 cycles for Treatment Arms 1 and 2 and 4 to 6 cycles for Treatment Arm 3); then continue pemetrexed 500 mg/m2 maintenance [i.e., q4w for Treatment Arms 1 and 2. For Treatment Arm 3, Pemetrexed maintenance therapy can be given either q3w or q4w (dependent on Investigator decision and local standards)] until objective disease progression.

Locations

Country Name City State
Brazil Research Site Barretos
Brazil Research Site Belo Horizonte
Brazil Research Site Curitiba
Brazil Research Site Porto Alegre
Brazil Research Site Porto Alegre
Brazil Research Site Porto Alegre
Brazil Research Site Ribeirao Preto
Brazil Research Site Rio de Janeiro
Brazil Research Site Santo Andre
Brazil Research Site Santo André
Brazil Research Site São José do Rio Preto
Brazil Research Site Sao Paulo
Brazil Research Site São Paulo
Brazil Research Site São Paulo
Bulgaria Research Site Plovdiv
Bulgaria Research Site Plovdiv
Bulgaria Research Site Sofia
Bulgaria Research Site Sofia
Bulgaria Research Site Sofia
Bulgaria Research Site Sofia
Bulgaria Research Site Varna
China Research Site Beijing
China Research Site Beijing
China Research Site Changchun
China Research Site Changsha
China Research Site Fuzhou
China Research Site Guangzhou
China Research Site Hangzhou
China Research Site Harbin
China Research Site Hefei
China Research Site Kunming
China Research Site Linyi
China Research Site Liuzhou
China Research Site Nanjing
China Research Site Qingdao
China Research Site Shanghai
China Research Site Shanghai
China Research Site Shanghai
China Research Site Shantou
China Research Site Wuhan
China Research Site Wuhan
China Research Site Xining
China Research Site Yangzhou
China Research Site Zhanjiang
China Research Site Zhengzhou
China Research Site Zhengzhou
Germany Research Site Berlin
Germany Research Site Essen
Germany Research Site Freiburg
Germany Research Site Gauting
Germany Research Site Hamburg
Germany Research Site Hamburg
Germany Research Site Heidelberg
Germany Research Site Immenhausen
Germany Research Site Mainz
Germany Research Site Oldenburg
Germany Research Site Würzburg
Hong Kong Research Site Shatin
Hungary Research Site Budapest
Hungary Research Site Budapest
Hungary Research Site Kecskemét
Hungary Research Site Miskolc
Hungary Research Site Törökbálint
Japan Research Site Bunkyo-ku
Japan Research Site Chuo-ku
Japan Research Site Fukuoka-shi
Japan Research Site Hiroshima-shi
Japan Research Site Iwakuni-shi
Japan Research Site Kanazawa
Japan Research Site Kashiwa
Japan Research Site Koto-ku
Japan Research Site Kurume-shi
Japan Research Site Matsuyama-shi
Japan Research Site Okayama-shi
Japan Research Site Okayama-shi
Japan Research Site Osakasayama
Japan Research Site Sapporo-shi
Japan Research Site Sunto-gun
Japan Research Site Toyoake-shi
Japan Research Site Ube-shi
Japan Research Site Yokohama-shi
Japan Research Site Yokohama-shi
Korea, Republic of Research Site Busan
Korea, Republic of Research Site Chungcheongbuk-do
Korea, Republic of Research Site Daegu
Korea, Republic of Research Site Incheon
Korea, Republic of Research Site Seongnam-si
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Ulsan
Mexico Research Site Aguascalientes
Mexico Research Site Cuautitlan Izcalli
Mexico Research Site Guadalajara
Mexico Research Site Mexico
Mexico Research Site México
Mexico Research Site Mexico City
Mexico Research Site Monterrey
Mexico Research Site Monterrey
Mexico Research Site Tuxtla
Peru Research Site Arequipa
Peru Research Site Lima
Peru Research Site Lima
Peru Research Site Lima
Peru Research Site Lima
Peru Research Site San Isidro
Poland Research Site Olsztyn
Poland Research Site Tomaszów Mazowiecki
Poland Research Site Warszawa
Poland Research Site Wodzislaw Slaski
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Omsk
Russian Federation Research Site Saint Petersburg
Russian Federation Research Site Saint-Petersburg
Russian Federation Research Site Sankt-Peterburg
Russian Federation Research Site St. Petersburg
South Africa Research Site Durban
South Africa Research Site Johannesburg
South Africa Research Site Kraaifontein
South Africa Research Site Parktown
South Africa Research Site Pretoria
South Africa Research Site Rondebosch
South Africa Research Site Vereeniging
Taiwan Research Site Changhua City
Taiwan Research Site Kaohsiung
Taiwan Research Site Kaohsiung City
Taiwan Research Site Taichung
Taiwan Research Site Taichung
Taiwan Research Site Tainan
Taiwan Research Site Taipei
Taiwan Research Site Taipei
Taiwan Research Site Taipei
Taiwan Research Site Tao-Yuan
Thailand Research Site Bangkok
Thailand Research Site Bangkok
Thailand Research Site Bangkok
Thailand Research Site Muang
Thailand Research Site Songkhla
Ukraine Research Site Dnipro
Ukraine Research Site Ivano-Frankivsk
Ukraine Research Site Kirovohrad
Ukraine Research Site Kyiv
Ukraine Research Site Kyiv
Ukraine Research Site Lviv
Ukraine Research Site Odesa
Ukraine Research Site Sumy
Ukraine Research Site Vinnytsia
Ukraine Research Site Zaporizhzhia
United Kingdom Research Site Leicester
United Kingdom Research Site London
United Kingdom Research Site London
United Kingdom Research Site London
United Kingdom Research Site Manchester
United States Research Site Bakersfield California
United States Research Site Canton Ohio
United States Research Site Chattanooga Tennessee
United States Research Site Fort Myers Florida
United States Research Site Fort Worth Texas
United States Research Site Houston Texas
United States Research Site Jacksonville Florida
United States Research Site Kansas City Missouri
United States Research Site Louisville Kentucky
United States Research Site Nashville Tennessee
United States Research Site Phoenix Arizona
United States Research Site Pittsburgh Pennsylvania
United States Research Site Richmond Virginia
United States Research Site Saint Petersburg Florida
United States Research Site Santa Monica California
United States Research Site West Palm Beach Florida
Vietnam Research Site Hanoi
Vietnam Research Site Ho Chi Minh

Sponsors (1)

Lead Sponsor Collaborator
AstraZeneca

Countries where clinical trial is conducted

United States,  Vietnam,  Brazil,  Bulgaria,  China,  Germany,  Hong Kong,  Hungary,  Japan,  Korea, Republic of,  Mexico,  Peru,  Poland,  Russian Federation,  South Africa,  Taiwan,  Thailand,  Ukraine,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary 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). 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).
Primary 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). From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).
Secondary 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. 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.
Secondary 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. From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).
Secondary 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. 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).
Secondary Best Objective Response (BoR) The BoR was calculated based on the overall visit responses from each RECIST 1.1 assessment. BOR was defined as the best response a patient had following randomization, but prior to starting any subsequent cancer therapy and up to and including RECIST 1.1 progression or the last evaluable assessment in the absence of RECIST 1.1 progression, as determined by BICR. Categorization of BoR was based on RECIST using the following 'response' categories: CR and PR and the following 'non-response' categories: stable disease (SD) =6 weeks, progression (ie, PD) and not evaluable (NE). Results are presented for number (%) of patients in each specified category. 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).
Secondary 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. 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).
Secondary 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. 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).
Secondary 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. 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.
Secondary 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. 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.
Secondary 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. Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, durvalumab).
Secondary 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. Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, tremelimumab).
Secondary 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) The EORTC QLQ-Core 30 version 3 (QLQ-C30 v3) was included for assessing HRQoL. It assesses HRQoL/health status through 9 multi-item scales: 5 functional scales (physical, role, cognitive, emotional, and social), 3 symptom scales (fatigue, pain, and nausea and vomiting), and a global health and QoL scale. 6 single-item symptom measures are also included: dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial difficulties. Scores from 0 to 100 were derived for each of the 15 domains, with higher scores representing greater functioning, greater HRQoL, or 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. 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).
Secondary 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. 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).

External Links