Study of Durvalumab + Tremelimumab With Chemotherapy or Durvalumab With Chemotherapy or Chemotherapy Alone for Patients With Lung Cancer (POSEIDON).

Sponsor
AstraZeneca (Industry)
Overall Status
Recruiting
CT.gov ID
NCT03164616
Collaborator
(none)
1,193
182
3
95.9
6.6
0.1

Study Details

Study Description

Brief 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.

Condition or Disease Intervention/Treatment Phase
  • Drug: Durvalumab
  • Drug: Tremelimumab
  • Drug: Abraxane + carboplatin
  • Drug: Gemcitabine + cisplatin
  • Drug: Gemcitabine + carboplatin
  • Drug: Pemetrexed + carboplatin
  • Drug: Pemetrexed + cisplatin
Phase 3

Detailed 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.

Study Design

Study Type:
Interventional
Anticipated Enrollment :
1193 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
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 Small-Cell Lung Cancer (NSCLC) (POSEIDON)
Actual Study Start Date :
Jun 1, 2017
Actual Primary Completion Date :
Mar 12, 2021
Anticipated Study Completion Date :
May 28, 2025

Arms and Interventions

Arm Intervention/Treatment
Experimental: Treatment Arm 1

durvalumab + tremelimumab combination therapy + SoC chemotherapy

Drug: Durvalumab
IV infusions every 3 weeks for 12 weeks (4 cycles) and every 4 weeks thereafter until disease progression or other discontinuation criteria

Drug: Tremelimumab
IV infusions every 3 weeks for 12 weeks (4 cycles). An additional dose of tremelimumab will be administered in the week 16.

Drug: 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).

Drug: 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).

Drug: 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).

Drug: 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.

Drug: 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.

Experimental: Treatment Arm 2

durvalumab monotherapy + SoC chemotherapy

Drug: Durvalumab
IV infusions every 3 weeks for 12 weeks (4 cycles) and every 4 weeks thereafter until disease progression or other discontinuation criteria

Drug: 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).

Drug: 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).

Drug: 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).

Drug: 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.

Drug: 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.

Active Comparator: Treatment Arm 3

SoC chemotherapy alone

Drug: 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).

Drug: 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).

Drug: 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).

Drug: 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.

Drug: 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.

Outcome Measures

Primary Outcome Measures

  1. Progression-Free Survival (PFS); D + SoC Compared With SoC Alone [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).]

    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).

  2. Overall Survival (OS); D + SoC Compared With SoC Alone [From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).]

    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).

Secondary Outcome Measures

  1. PFS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC [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.]

    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.

  2. OS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC [From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).]

    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.

  3. Objective Response Rate (ORR) [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).]

    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.

  4. Best Objective Response (BoR) [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).]

    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.

  5. Duration of Response (DoR) [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).]

    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.

  6. Time From Randomization to Second Progression (PFS2) [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).]

    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.

  7. Pharmacokinetics (PK) of Durvalumab; Peak and Trough Serum Concentrations [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.]

    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.

  8. PK of Tremelimumab; Peak and Trough Serum Concentrations [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.]

    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.

  9. Number of Patients With Anti-Drug Antibody (ADA) Response to Durvalumab [Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, 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.

  10. Number of Patients With ADA Response to Tremelimumab [Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, 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.

  11. 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) [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).]

    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.

  12. Time to Deterioration of PRO Symptoms, Assessed Using EORTC QLQ-Lung Cancer Module 13 (QLQ-LC13) [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).]

    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.

Eligibility Criteria

Criteria

Ages Eligible for Study:
18 Years to 130 Years
Sexes Eligible for Study:
All
Accepts Healthy Volunteers:
No
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.

Contacts and Locations

Locations

Site City State Country Postal Code
1 Research Site Phoenix Arizona United States 85054
2 Research Site Bakersfield California United States 93309
3 Research Site Santa Monica California United States 90404
4 Research Site Fort Myers Florida United States 33901
5 Research Site Jacksonville Florida United States 32224
6 Research Site Saint Petersburg Florida United States 33705
7 Research Site West Palm Beach Florida United States 33401
8 Research Site Louisville Kentucky United States 40202
9 Research Site Kansas City Missouri United States 64132
10 Research Site Canton Ohio United States 44710
11 Research Site Pittsburgh Pennsylvania United States 15212
12 Research Site Chattanooga Tennessee United States 37404
13 Research Site Nashville Tennessee United States 37203
14 Research Site Fort Worth Texas United States 76104
15 Research Site Houston Texas United States 77090
16 Research Site Richmond Virginia United States 23298
17 Research Site Barretos Brazil 14784-400
18 Research Site Belo Horizonte Brazil 30380-472
19 Research Site Curitiba Brazil 81520-060
20 Research Site Porto Alegre Brazil 90035-003
21 Research Site Porto Alegre Brazil 90610-000
22 Research Site Porto Alegre Brazil 91350-200
23 Research Site Ribeirao Preto Brazil 14015-140
24 Research Site Rio de Janeiro Brazil 20231-050
25 Research Site Santo Andre Brazil 09080-110
26 Research Site Santo André Brazil 09060-650
27 Research Site Sao Paulo Brazil 01209-000
28 Research Site Sao Paulo Brazil 01246-000
29 Research Site São José do Rio Preto Brazil 15090-000
30 Research Site São Paulo Brazil 03102-002
31 Research Site Plovdiv Bulgaria 4000
32 Research Site Plovdiv Bulgaria 4004
33 Research Site Sofia Bulgaria 1330
34 Research Site Sofia Bulgaria 1431
35 Research Site Sofia Bulgaria 1618
36 Research Site Sofia Bulgaria 1784
37 Research Site Varna Bulgaria 9010
38 Research Site Beijing China 100021
39 Research Site Beijing China 100053
40 Research Site Beijing China 100142
41 Research Site Changchun China 130012
42 Research Site Changsha China 410013
43 Research Site Fuzhou China 350014
44 Research Site Guangzhou China 510080
45 Research Site Hangzhou China 310022
46 Research Site Harbin China 150081
47 Research Site He Fei China 230022
48 Research Site Hefei China 230001
49 Research Site Hefei China 230601
50 Research Site Jinan China 2501117
51 Research Site Kunming China 650118
52 Research Site Kunming China CN-650034
53 Research Site Linyi China 276000
54 Research Site Liuzhou China 545006
55 Research Site Nanjing China 210009
56 Research Site Qingdao China 110016
57 Research Site Shanghai China 200030
58 Research Site Shanghai China 200032
59 Research Site Shanghai China 200080
60 Research Site Shanghai China 200433
61 Research Site Shantou China 515041
62 Research Site Wenzhou China 325015
63 Research Site Wuhan China 430022
64 Research Site Wuhan China 430079
65 Research Site Xi'an China 710000
66 Research Site Xining China 810001
67 Research Site Yangzhou China 225001
68 Research Site Zhanjiang China 524001
69 Research Site Zhengzhou China 450008
70 Research Site Zhengzhou China 450052
71 Research Site Berlin Germany 13125
72 Research Site Essen Germany 45122
73 Research Site Freiburg Germany 79106
74 Research Site Gauting Germany 82131
75 Research Site Hamburg Germany 20251
76 Research Site Hamburg Germany 21075
77 Research Site Heidelberg Germany 69126
78 Research Site Immenhausen Germany 34376
79 Research Site Mainz Germany 55131
80 Research Site Oldenburg Germany 26121
81 Research Site Würzburg Germany 97067
82 Research Site Shatin Hong Kong 00000
83 Research Site Budapest Hungary 1083
84 Research Site Budapest Hungary 1121
85 Research Site Kecskemét Hungary 6000
86 Research Site Miskolc Hungary 3529
87 Research Site Törökbálint Hungary 2045
88 Research Site Bunkyo-ku Japan 113-8603
89 Research Site Chuo-ku Japan 104-0045
90 Research Site Fukuoka-shi Japan 812-8582
91 Research Site Hiroshima-shi Japan 730-0011
92 Research Site Iwakuni-shi Japan 740-8510
93 Research Site Kanazawa Japan 920-8641
94 Research Site Kashiwa Japan 227-8577
95 Research Site Koto-ku Japan 135-8550
96 Research Site Kurume-shi Japan 830-0011
97 Research Site Matsuyama-shi Japan 790-0007
98 Research Site Okayama-shi Japan 700-8558
99 Research Site Okayama-shi Japan 700-8607
100 Research Site Osakasayama Japan 589-8511
101 Research Site Sapporo-shi Japan 003-0804
102 Research Site Sunto-gun Japan 411-8777
103 Research Site Toyoake-shi Japan 470-1101
104 Research Site Ube-shi Japan 755-0241
105 Research Site Yokohama-shi Japan 236-0004
106 Research Site Yokohama-shi Japan 241-8515
107 Research Site Busan Korea, Republic of 47392
108 Research Site Chungcheongbuk-do Korea, Republic of 28644
109 Research Site Daegu Korea, Republic of 42415
110 Research Site Incheon Korea, Republic of 21565
111 Research Site Seongnam-si Korea, Republic of 13620
112 Research Site Seoul Korea, Republic of 05505
113 Research Site Seoul Korea, Republic of 06591
114 Research Site Seoul Korea, Republic of 120-752
115 Research Site Seoul Korea, Republic of 135-710
116 Research Site Ulsan Korea, Republic of 44033
117 Research Site Aguascalientes Mexico 20230
118 Research Site Cuautitlan Izcalli Mexico 54769
119 Research Site Guadalajara Mexico 44280
120 Research Site Mexico City Mexico 0 3100
121 Research Site Mexico Mexico 14080
122 Research Site Monterrey Mexico 64060
123 Research Site Monterrey Mexico 64460
124 Research Site México Mexico 04739
125 Research Site Tuxtla Mexico 29030
126 Research Site Arequipa Peru AREQUIPA01
127 Research Site Lima Peru L27
128 Research Site Lima Peru LIMA 29
129 Research Site Lima Peru LIMA 34
130 Research Site Lima Peru LIMA 41
131 Research Site San Isidro Peru 27
132 Research Site Olsztyn Poland 10-357
133 Research Site Tomaszów Mazowiecki Poland 97-200
134 Research Site Warszawa Poland 02-781
135 Research Site Wodzisław Śląski Poland 44-300
136 Research Site Moscow Russian Federation 105229
137 Research Site Moscow Russian Federation 115280
138 Research Site Moscow Russian Federation 115478
139 Research Site Moscow Russian Federation 125367
140 Research Site Omsk Russian Federation 644013
141 Research Site Saint Petersburg Russian Federation 195271
142 Research Site Saint-Petersburg Russian Federation 194291
143 Research Site St. Petersburg Russian Federation 197758
144 Research Site Durban South Africa 4091
145 Research Site Johannesburg South Africa 0001
146 Research Site Kraaifontein South Africa 7570
147 Research Site Parktown South Africa 2193
148 Research Site Pretoria South Africa 0001
149 Research Site Rondebosch South Africa 7700
150 Research Site Vereeniging South Africa 1930
151 Research Site Changhua City Taiwan 50006
152 Research Site Kaohsiung City Taiwan 83301
153 Research Site Kaohsiung Taiwan 82445
154 Research Site Taichung Taiwan 40447
155 Research Site Taichung Taiwan 40705
156 Research Site Tainan Taiwan 70403
157 Research Site Taipei Taiwan 10002
158 Research Site Taipei Taiwan 112
159 Research Site Taipei Taiwan 235
160 Research Site Tao-Yuan Taiwan 333
161 Research Site Bangkok Thailand 10210
162 Research Site Bangkok Thailand 10330
163 Research Site Bangkok Thailand 10400
164 Research Site Muang Thailand 50200
165 Research Site Songkhla Thailand 90110
166 Research Site Dnipro Ukraine 49102
167 Research Site Ivano-Frankivsk Ukraine 76018
168 Research Site Kirovohrad Ukraine 25006
169 Research Site Kyiv Ukraine 03022
170 Research Site Kyiv Ukraine 03115
171 Research Site Lviv Ukraine 79031
172 Research Site Odesa Ukraine 65055
173 Research Site Sumy Ukraine 40022
174 Research Site Vinnytsia Ukraine 21029
175 Research Site Zaporizhzhia Ukraine 69040
176 Research Site Leicester United Kingdom LE1 5WW
177 Research Site London United Kingdom EC1M 6BQ
178 Research Site London United Kingdom NW1 2PG
179 Research Site London United Kingdom W6 8RF
180 Research Site Manchester United Kingdom M20 4BX
181 Research Site Hanoi Vietnam 100000
182 Research Site Ho Chi Minh Vietnam 700000

Sponsors and Collaborators

  • AstraZeneca

Investigators

  • Study Director: Xiaojin Shi, M.D., MSc, One MedImmune Way, Gaithersburg, Maryland 20878, United States

Study Documents (Full-Text)

More Information

Additional Information:

Publications

None provided.
Responsible Party:
AstraZeneca
ClinicalTrials.gov Identifier:
NCT03164616
Other Study ID Numbers:
  • D419MC00004
First Posted:
May 23, 2017
Last Update Posted:
Aug 18, 2022
Last Verified:
Aug 1, 2022
Individual Participant Data (IPD) Sharing Statement:
Yes
Plan to Share IPD:
Yes
Studies a U.S. FDA-regulated Drug Product:
Yes
Studies a U.S. FDA-regulated Device Product:
No
Keywords provided by AstraZeneca
Additional relevant MeSH terms:

Study Results

Participant Flow

Recruitment Details Study was conducted in 142 study centers across 18 countries in North and Latin America, Europe, Asia Pacific and Africa. First patient randomized: 27 June 2017. Results are presented for the global cohort at final analysis data cut-offs (DCOs) of 24 July 2019 (Response Evaluation Criteria in Solid Tumors [RECIST]-based endpoints) and 12 March 2021 (all other data). Once global enrollment was complete, enrollment was started in mainland China. Results for the China cohort will be reported later.
Pre-assignment Detail Patients were randomized in a stratified manner as per programmed cell death ligand 1 (PD-L1) tumor expression status (PD-L1 tumor cells [TC] ≥50% versus [vs] <50%), disease stage (IVA vs IVB), and histology (non-squamous vs squamous) in a 1:1:1 ratio to receive treatment with tremelimumab + durvalumab combination therapy + standard of care (SoC) chemotherapy (also referred to as T+ D + SoC), durvalumab monotherapy + SoC chemotherapy (also referred to as D + SoC), or SoC chemotherapy alone.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 milligrams (mg) + durvalumab 1500 mg combination therapy + SoC chemotherapy via intravenous (IV) infusion every 3 weeks (Q3W) for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion every 4 weeks (Q4W) from Week 12 until progressive disease (PD), unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Period Title: Overall Study
STARTED 338 338 337
Received Treatment 331 335 331
COMPLETED 80 65 40
NOT COMPLETED 258 273 297

Baseline Characteristics

Arm/Group Title T + D + SoC D + SoC SoC Alone Total
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator. Total of all reporting groups
Overall Participants 338 338 337 1013
Age (years) [Mean (Standard Deviation) ]
Mean (Standard Deviation) [years]
62.6
(9.43)
63.5
(9.10)
63.1
(9.87)
63.1
(9.47)
Sex: Female, Male (Count of Participants)
Female
69
20.4%
85
25.1%
89
26.4%
243
24%
Male
269
79.6%
253
74.9%
248
73.6%
770
76%
Race/Ethnicity, Customized (Count of Participants)
White
205
60.7%
182
53.8%
179
53.1%
566
55.9%
Black or African American
8
2.4%
4
1.2%
8
2.4%
20
2%
Asian
99
29.3%
123
36.4%
128
38%
350
34.6%
Native Hawaiian or other Pacific Islander
2
0.6%
0
0%
0
0%
2
0.2%
American Indian or Alaska Native
12
3.6%
17
5%
9
2.7%
38
3.8%
Other
12
3.6%
12
3.6%
13
3.9%
37
3.7%
Race/Ethnicity, Customized (Count of Participants)
Hispanic or Latino
51
15.1%
54
16%
55
16.3%
160
15.8%
Not Hispanic or Latino
287
84.9%
284
84%
282
83.7%
853
84.2%
Age (Count of Participants)
≥18 to <50
29
8.6%
27
8%
30
8.9%
86
8.5%
≥50 to <65
162
47.9%
142
42%
146
43.3%
450
44.4%
≥65 to <75
112
33.1%
130
38.5%
121
35.9%
363
35.8%
≥75
35
10.4%
39
11.5%
40
11.9%
114
11.3%

Outcome Measures

1. Primary Outcome
Title Progression-Free Survival (PFS); D + SoC Compared With SoC Alone
Description 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).
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. Analysis of PFS was assessed as a primary outcome measure for comparison of the D + SoC vs SoC alone treatment arms only. The alternative treatment arm comparisons were assessed as a secondary outcome measure.
Arm/Group Title D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 338 337
Median (95% Confidence Interval) [months]
5.5
4.8
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection D + SoC, SoC Alone
Comments D + SoC vs SoC alone. A hazard ratio (HR) <1 favors D + SoC to be associated with a longer PFS than SoC alone.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value 0.00093
Comments Analysis was performed using a stratified log-rank test adjusting for PD-L1 tumor expression (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using the Breslow approach for handling ties.
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.74
Confidence Interval (2-Sided) 95%
0.620 to 0.885
Parameter Dispersion Type:
Value:
Estimation Comments The HR and confidence interval (CI) were estimated from a stratified Cox proportional hazards model adjusting for PD-L1 (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using Efron method to control for ties.
2. Primary Outcome
Title Overall Survival (OS); D + SoC Compared With SoC Alone
Description 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).
Time Frame From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. Analysis of OS was assessed as a primary outcome measure for comparison of the D + SoC vs SoC alone treatment arms only. The alternative treatment arm comparisons were assessed as a secondary outcome measure.
Arm/Group Title D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 338 337
Median (95% Confidence Interval) [months]
13.3
11.7
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection D + SoC, SoC Alone
Comments D + SoC vs SoC alone. An HR <1 favors D + SoC to be associated with a longer OS than SoC alone.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value 0.07581
Comments Analysis was performed using a stratified log-rank test adjusting for PD-L1 tumor expression (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using the Breslow approach for handling ties.
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.86
Confidence Interval (2-Sided) 95%
0.724 to 1.016
Parameter Dispersion Type:
Value:
Estimation Comments The HR and CI were estimated from a stratified Cox proportional hazards model adjusting for PD-L1 (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using Efron method to control for ties.
3. Secondary Outcome
Title PFS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC
Description 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.
Time Frame 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.

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 338 338 337
Median (95% Confidence Interval) [months]
6.2
5.5
4.8
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection D + SoC, SoC Alone
Comments T + D + SoC vs SoC alone. An HR <1 favors T + D + SoC to be associated with a longer PFS than SoC alone.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value 0.00031
Comments Analysis was performed using a stratified log-rank test adjusting for PD-L1 tumor expression (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using the Breslow approach for handling ties.
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.72
Confidence Interval (2-Sided) 95%
0.600 to 0.860
Parameter Dispersion Type:
Value:
Estimation Comments The HR and CI were estimated from a stratified Cox proportional hazards model adjusting for PD-L1 (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using Efron method to control for ties.
4. Secondary Outcome
Title OS; T + D + SoC Compared With SoC Alone and T + D + SoC Compared With D + SoC
Description 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.
Time Frame From baseline until death due to any cause. Assessed until global cohort DCO of 12 March 2021 (maximum of approximately 45 months).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 338 338 337
Median (95% Confidence Interval) [months]
14.0
13.3
11.7
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection D + SoC, SoC Alone
Comments T + D + SoC vs SoC alone. An HR <1 favors T + D + SoC to be associated with a longer OS than SoC alone.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value 0.00304
Comments Analysis was performed using a stratified log-rank test adjusting for PD-L1 tumor expression (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using the Breslow approach for handling ties.
Method Log Rank
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.77
Confidence Interval (2-Sided) 95%
0.650 to 0.916
Parameter Dispersion Type:
Value:
Estimation Comments The HR and CI were estimated from a stratified Cox proportional hazards model adjusting for PD-L1 (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using Efron method to control for ties.
5. Secondary Outcome
Title Objective Response Rate (ORR)
Description 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.
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. The denominator was a subset of the FAS who had measurable disease at baseline.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 335 330 332
Number [percentage of patients]
46.3
48.5
33.4
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection SoC Alone, SoC Alone
Comments D + SoC vs SoC alone. An odds ratio >1 favors D + SoC compared to SoC alone. Analysis was performed using logistic regression adjusting for PD-L1 tumor expression (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB), with the CI calculated using a profile likelihood approach.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value
Comments
Method
Comments
Method of Estimation Estimation Parameter Odds Ratio (OR)
Estimated Value 1.90
Confidence Interval (2-Sided) 95%
1.382 to 2.619
Parameter Dispersion Type:
Value:
Estimation Comments
Statistical Analysis 2
Statistical Analysis Overview Comparison Group Selection D + SoC, SoC Alone
Comments T + D + SoC vs SoC alone. An odds ratio >1 favors T + D + SoC compared to SoC alone. Analysis was performed using logistic regression adjusting for PD-L1 tumor expression (TC ≥50% vs TC <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB), with the CI calculated using a profile likelihood approach.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value
Comments
Method
Comments
Method of Estimation Estimation Parameter Odds Ratio (OR)
Estimated Value 1.72
Confidence Interval (2-Sided) 95%
1.260 to 2.367
Parameter Dispersion Type:
Value:
Estimation Comments
6. Secondary Outcome
Title Best Objective Response (BoR)
Description 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.
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. The denominator was a subset of the FAS who had measurable disease at baseline.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 335 330 332
CR
2
0.6%
3
0.9%
0
0%
PR
153
45.3%
157
46.4%
111
32.9%
SD ≥6 weeks
120
35.5%
107
31.7%
150
44.5%
PD
48
14.2%
60
17.8%
61
18.1%
NE
12
3.6%
3
0.9%
10
3%
7. Secondary Outcome
Title Duration of Response (DoR)
Description 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.
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. Only patients with an objective response were included in the analysis.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 155 160 111
Median (Inter-Quartile Range) [months]
7.4
6.0
4.2
8. Secondary Outcome
Title Time From Randomization to Second Progression (PFS2)
Description 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.
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 338 338 337
Median (95% Confidence Interval) [months]
10.4
10.2
9.4
Statistical Analysis 1
Statistical Analysis Overview Comparison Group Selection SoC Alone, SoC Alone
Comments D + SoC vs SoC alone. An HR <1 favors D + SoC to be associated with a longer PFS2 than SoC alone.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value
Comments
Method
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.79
Confidence Interval (2-Sided) 95%
0.666 to 0.928
Parameter Dispersion Type:
Value:
Estimation Comments The HR and CI were estimated from a stratified Cox proportional hazards model adjusting for PD-L1 (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using Efron method to control for ties.
Statistical Analysis 2
Statistical Analysis Overview Comparison Group Selection D + SoC, SoC Alone
Comments T + D + SoC vs SoC alone. An HR <1 favors T + D + SoC to be associated with a longer PFS2 than SoC alone.
Type of Statistical Test Superiority
Comments
Statistical Test of Hypothesis p-Value
Comments
Method
Comments
Method of Estimation Estimation Parameter Hazard Ratio (HR)
Estimated Value 0.75
Confidence Interval (2-Sided) 95%
0.632 to 0.883
Parameter Dispersion Type:
Value:
Estimation Comments The HR and CI were estimated from a stratified Cox proportional hazards model adjusting for PD-L1 (TC ≥50% vs <50%), histology (squamous vs non-squamous) and disease stage (IVA vs IVB) and using Efron method to control for ties.
9. Secondary Outcome
Title Pharmacokinetics (PK) of Durvalumab; Peak and Trough Serum Concentrations
Description 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.
Time Frame 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.

Outcome Measure Data

Analysis Population Description
Global cohort: The PK analysis set included all patients who received at least 1 dose of durvalumab or tremelimumab per the protocol for whom any post-dose PK data were available and without any protocol deviations that would significantly affect the PK analyses. PK for durvalumab was performed for the T + D + SoC and D + SoC treatment arms only.
Arm/Group Title T + D + SoC D + SoC
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 327 330
Week 0
418.80
(164.20)
505.01
(48.76)
Week 3
82.08
(84.38)
91.53
(100.58)
Week 12
195.62
(58.65)
212.11
(60.37)
Follow-up (3 months)
13.42
(166.36)
16.06
(249.88)
10. Secondary Outcome
Title PK of Tremelimumab; Peak and Trough Serum Concentrations
Description 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.
Time Frame 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.

Outcome Measure Data

Analysis Population Description
Global cohort: The PK analysis set included all patients who received at least 1 dose of durvalumab or tremelimumab per the protocol for whom any post-dose PK data were available and without any protocol deviations that would significantly affect the PK analyses. PK for tremelimumab was performed for the T + D + SoC treatment arm only.
Arm/Group Title T + D + SoC
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 327
Week 0
23.17
(65.62)
Week 3
4.16
(80.83)
Week 12
7.82
(75.68)
Follow-up (3 months)
0.86
(87.65)
11. Secondary Outcome
Title Number of Patients With Anti-Drug Antibody (ADA) Response to Durvalumab
Description 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.
Time Frame Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, durvalumab).

Outcome Measure Data

Analysis Population Description
Global cohort: The ADA evaluable patients included those in the safety analysis set with non-missing baseline ADA sample and at least 1 post-baseline ADA sample. ADA for durvalumab was performed for the T + D + SoC and D + SoC treatment arms only. The denominator was durvalumab ADA evaluable patients.
Arm/Group Title T + D + SoC D + SoC
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 286 285
ADA positive at any visit (ADA prevalence)
42
12.4%
33
9.8%
Treatment-emergent ADA positive (ADA incidence)
29
8.6%
19
5.6%
Treatment-boosted ADA
2
0.6%
1
0.3%
Treatment-induced ADA (ADA positive post-baseline only)
27
8%
18
5.3%
ADA positive at baseline only
8
2.4%
13
3.8%
ADA positive post-baseline and positive at baseline
7
2.1%
2
0.6%
Persistently positive
8
2.4%
7
2.1%
Transiently positive
26
7.7%
13
3.8%
nAb positive at any visit
3
0.9%
3
0.9%
12. Secondary Outcome
Title Number of Patients With ADA Response to Tremelimumab
Description 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.
Time Frame Samples were collected on Day 1 (Week 0), Week 12 and at 3 months after the last dose of study treatment (ie, tremelimumab).

Outcome Measure Data

Analysis Population Description
Global cohort: The ADA evaluable patients included those in the safety analysis set with non-missing baseline ADA sample and at least 1 post-baseline ADA sample. ADA for tremelimumab was performed for the T + D + SoC treatment arm only. The denominator was tremelimumab ADA evaluable patients.
Arm/Group Title T + D + SoC
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 278
ADA positive at any visit (ADA prevalence)
44
13%
Treatment-emergent ADA positive (ADA incidence)
38
11.2%
Treatment-boosted ADA
3
0.9%
Treatment-induced ADA (ADA positive post-baseline only)
35
10.4%
ADA positive at baseline only
4
1.2%
ADA positive post-baseline and positive at baseline
5
1.5%
Persistently positive
22
6.5%
Transiently positive
18
5.3%
nAb positive at any visit
31
9.2%
13. Secondary Outcome
Title 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)
Description 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.
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. For QLQ-C30 function scales and global health status/HRQoL, only patients with baseline scores ≥10 (and with data available) were included in the analysis. For QLQ-C30 symptom scales/items, only patients with baseline scores ≤90 (and with data available) were included in the analysis.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 325 326 321
QLQ-C30 Global Health Status / HRQoL
8.3
7.8
5.6
QLQ-C30 Physical Functioning
7.7
8.3
5.3
QLQ-C30 Role Functioning
6.6
7.4
4.8
QLQ-C30 Cognitive Functioning
7.6
7.4
5.8
QLQ-C30 Emotional Functioning
8.5
9.5
7.5
QLQ-C30 Social Functioning
6.4
7.1
5.7
QLQ-C30 Fatigue
3.7
3.8
2.8
QLQ-C30 Pain
8.9
6.5
5.7
QLQ-C30 Nausea / Vomiting
7.8
5.6
5.6
QLQ-C30 Dyspnea
7.9
6.9
6.7
QLQ-C30 Insomnia
8.3
7.4
5.8
QLQ-C30 Appetite Loss
7.2
7.2
7.0
QLQ-C30 Constipation
9.2
8.7
6.1
QLQ-C30 Diarrhea
11.0
9.8
10.8
14. Secondary Outcome
Title Time to Deterioration of PRO Symptoms, Assessed Using EORTC QLQ-Lung Cancer Module 13 (QLQ-LC13)
Description 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.
Time Frame 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).

Outcome Measure Data

Analysis Population Description
Global cohort: The FAS included all randomized patients. For QLQ-LC13 symptom scales/items, only patients with baseline scores ≤90 (and with data available) were included in the analysis.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
Measure Participants 325 326 321
QLQ-LC13 Cough
9.7
11.0
8.8
QLQ-LC13 Hemoptysis
17.8
14.0
11.4
QLQ-LC13 Dyspnea
5.4
5.0
3.6
QLQ-LC13 Pain in Chest
10.0
9.5
8.6
QLQ-LC13 Pain in Arm or Shoulder
8.9
8.9
8.8
QLQ-LC13 Pain in Other Parts
9.7
8.9
5.8

Adverse Events

Time Frame Adverse events: from first dose of study treatment until the earlier of 90 days after last dose or the date of first subsequent anticancer therapy. All-cause mortality (death due to any cause): from randomization up to global cohort final analysis DCO (12 March 2021). Maximum timeframe of approximately 45 months.
Adverse Event Reporting Description Safety analysis set included all patients who received at least 1 dose of study treatment (analyzed according to actual treatment received). 1 patient randomized to T + D + SoC arm and 1 patient randomized to D + SoC arm only received SoC and were included in SoC alone arm of the safety analysis set. All-cause mortality was determined for patients in the FAS (analyzed according to randomized treatment arm, regardless of the treatment received). Results are reported for the global cohort only.
Arm/Group Title T + D + SoC D + SoC SoC Alone
Arm/Group Description During chemotherapy (combination) stage: Patients received tremelimumab 75 mg + durvalumab 1500 mg combination therapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Patients also received an additional dose of tremelimumab 75 mg (in combination with durvalumab) at Week 16 post-chemotherapy. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received durvalumab 1500 mg monotherapy + SoC chemotherapy via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Patients then continued to receive durvalumab monotherapy 1500 mg IV infusion Q4W from Week 12 until PD, unless there was unacceptable toxicity, withdrawal of consent, or another discontinuation criterion was met. Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q4W from Week 12 until PD, unless contraindicated per the Investigator. During chemotherapy (combination) stage: Patients received SoC chemotherapy alone via IV infusion Q3W for 4 doses/cycles (Weeks 0, 3, 6 and 9). Patients could also receive SoC chemotherapy for an additional 2 cycles Q3W on Weeks 12 and 15 if clinically indicated and at the Investigator's discretion before the patient entered follow-up. The SoC chemotherapy was the Investigator's choice of one of the following regimens: abraxane + carboplatin (squamous and non-squamous patients), pemetrexed + cisplatin or carboplatin (non-squamous patients only), or gemcitabine + cisplatin or carboplatin (squamous patients only). Post-chemotherapy (maintenance) stage: Non-squamous patients who received pemetrexed + carboplatin/cisplatin during chemotherapy stage could receive pemetrexed maintenance therapy, given Q3W or Q4W (dependent on Investigator decision and local standards) from Week 12 until PD, unless contraindicated per the Investigator.
All Cause Mortality
T + D + SoC D + SoC SoC Alone
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 251/338 (74.3%) 265/338 (78.4%) 285/337 (84.6%)
Serious Adverse Events
T + D + SoC D + SoC SoC Alone
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 146/330 (44.2%) 134/334 (40.1%) 117/333 (35.1%)
Blood and lymphatic system disorders
Leukopenia 2/330 (0.6%) 3 1/334 (0.3%) 1 1/333 (0.3%) 1
Anaemia 18/330 (5.5%) 32 23/334 (6.9%) 30 21/333 (6.3%) 25
Neutropenia 4/330 (1.2%) 5 6/334 (1.8%) 8 3/333 (0.9%) 5
Pancytopenia 2/330 (0.6%) 2 5/334 (1.5%) 6 3/333 (0.9%) 4
Thrombocytopenia 8/330 (2.4%) 8 6/334 (1.8%) 7 3/333 (0.9%) 3
Disseminated intravascular coagulation 1/330 (0.3%) 1 0/334 (0%) 0 1/333 (0.3%) 1
Febrile neutropenia 7/330 (2.1%) 7 5/334 (1.5%) 5 4/333 (1.2%) 4
Cardiac disorders
Acute coronary syndrome 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Acute myocardial infarction 2/330 (0.6%) 2 3/334 (0.9%) 3 1/333 (0.3%) 1
Angina pectoris 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Angina unstable 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Atrial fibrillation 2/330 (0.6%) 2 1/334 (0.3%) 1 0/333 (0%) 0
Atrial tachycardia 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Autoimmune myocarditis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Bundle branch block left 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Cardiac arrest 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Cardiac failure 2/330 (0.6%) 2 0/334 (0%) 0 3/333 (0.9%) 3
Cardiac failure acute 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Cardiac failure congestive 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Cardiac tamponade 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Cardiopulmonary failure 2/330 (0.6%) 2 0/334 (0%) 0 1/333 (0.3%) 1
Coronary artery disease 1/330 (0.3%) 1 2/334 (0.6%) 2 0/333 (0%) 0
Myocardial infarction 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Pericardial effusion 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Right ventricular failure 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Supraventricular tachycardia 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Tachyarrhythmia 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Ventricular fibrillation 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Ear and labyrinth disorders
Deafness bilateral 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Deafness unilateral 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Endocrine disorders
Adrenal insufficiency 3/330 (0.9%) 3 2/334 (0.6%) 2 0/333 (0%) 0
Diabetes insipidus 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Hypopituitarism 2/330 (0.6%) 2 1/334 (0.3%) 1 0/333 (0%) 0
Eye disorders
Cataract 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 2
Gastrointestinal disorders
Abdominal discomfort 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Abdominal pain upper 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Autoimmune pancreatitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Colitis 5/330 (1.5%) 5 3/334 (0.9%) 3 0/333 (0%) 0
Diarrhoea 8/330 (2.4%) 8 3/334 (0.9%) 4 2/333 (0.6%) 2
Diverticular perforation 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Duodenal ulcer haemorrhage 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Dysphagia 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Enteritis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Enterocolitis 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Faecaloma 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Gastric ulcer 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Gastric ulcer haemorrhage 1/330 (0.3%) 1 0/334 (0%) 0 1/333 (0.3%) 1
Gastric ulcer perforation 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Gastritis 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Gastrointestinal haemorrhage 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Gastrointestinal inflammation 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Inguinal hernia 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Intestinal obstruction 0/330 (0%) 0 2/334 (0.6%) 2 0/333 (0%) 0
Mesenteric vein thrombosis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Nausea 2/330 (0.6%) 2 1/334 (0.3%) 2 2/333 (0.6%) 2
Oesophageal obstruction 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Oesophageal stenosis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Oesophagitis 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Pancreatitis 2/330 (0.6%) 2 0/334 (0%) 0 1/333 (0.3%) 1
Peptic ulcer 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Small intestinal obstruction 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Upper gastrointestinal haemorrhage 1/330 (0.3%) 1 1/334 (0.3%) 1 1/333 (0.3%) 1
Vomiting 2/330 (0.6%) 2 2/334 (0.6%) 2 0/333 (0%) 0
General disorders
Asthenia 1/330 (0.3%) 1 2/334 (0.6%) 2 2/333 (0.6%) 2
Cardiac death 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Death 3/330 (0.9%) 3 6/334 (1.8%) 6 1/333 (0.3%) 1
Face oedema 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Fatigue 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
General physical health deterioration 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Malaise 0/330 (0%) 0 1/334 (0.3%) 2 0/333 (0%) 0
Oedema peripheral 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Pyrexia 8/330 (2.4%) 8 1/334 (0.3%) 1 1/333 (0.3%) 1
Sudden death 1/330 (0.3%) 1 3/334 (0.9%) 3 0/333 (0%) 0
Hepatobiliary disorders
Autoimmune hepatitis 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Cholangitis 0/330 (0%) 0 2/334 (0.6%) 2 0/333 (0%) 0
Cholecystitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Drug-induced liver injury 3/330 (0.9%) 3 0/334 (0%) 0 0/333 (0%) 0
Hepatic failure 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Hepatitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Immune-mediated hepatitis 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Immune system disorders
Anaphylactic reaction 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Drug hypersensitivity 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Haemophagocytic lymphohistiocytosis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Hypersensitivity 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Infections and infestations
Abscess oral 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Acute hepatitis B 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Appendicitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Arthritis bacterial 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Bronchitis 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Bronchitis bacterial 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
COVID-19 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
COVID-19 pneumonia 1/330 (0.3%) 1 1/334 (0.3%) 1 1/333 (0.3%) 1
Candida pneumonia 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Cellulitis 1/330 (0.3%) 1 0/334 (0%) 0 3/333 (0.9%) 4
Chikungunya virus infection 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Cystitis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Cytomegalovirus colitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Device related infection 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Diarrhoea infectious 0/330 (0%) 0 2/334 (0.6%) 2 0/333 (0%) 0
Disseminated tuberculosis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Empyema 2/330 (0.6%) 2 1/334 (0.3%) 1 1/333 (0.3%) 1
Encephalitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Encephalitis viral 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Enterocolitis infectious 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Gastroenteritis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Gastroenteritis viral 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Herpes zoster 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Infection 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Infective exacerbation of chronic obstructive airways disease 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Influenza 0/330 (0%) 0 2/334 (0.6%) 2 0/333 (0%) 0
Lower respiratory tract infection 1/330 (0.3%) 1 2/334 (0.6%) 2 2/333 (0.6%) 2
Neutropenic sepsis 0/330 (0%) 0 0/334 (0%) 0 2/333 (0.6%) 3
Nosocomial infection 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Ophthalmic herpes zoster 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Oral candidiasis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Perirectal abscess 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Pharyngitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Pleural infection 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Pneumocystis jirovecii pneumonia 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Pneumonia 36/330 (10.9%) 42 21/334 (6.3%) 23 16/333 (4.8%) 17
Pneumonia bacterial 1/330 (0.3%) 2 0/334 (0%) 0 0/333 (0%) 0
Pneumonia klebsiella 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Postoperative wound infection 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Pulmonary sepsis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Pyelonephritis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Respiratory tract infection 3/330 (0.9%) 4 0/334 (0%) 0 0/333 (0%) 0
Sepsis 5/330 (1.5%) 5 2/334 (0.6%) 2 2/333 (0.6%) 2
Septic shock 1/330 (0.3%) 1 0/334 (0%) 0 1/333 (0.3%) 1
Staphylococcal infection 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Tracheitis 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Tracheobronchitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Upper respiratory tract infection 2/330 (0.6%) 2 0/334 (0%) 0 2/333 (0.6%) 2
Urinary tract infection 1/330 (0.3%) 1 0/334 (0%) 0 2/333 (0.6%) 2
Vascular device infection 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Viral infection 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Injury, poisoning and procedural complications
Femoral neck fracture 2/330 (0.6%) 2 1/334 (0.3%) 1 0/333 (0%) 0
Femur fracture 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Foot fracture 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Hip fracture 0/330 (0%) 0 2/334 (0.6%) 2 2/333 (0.6%) 2
Infusion related reaction 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Radiation oesophagitis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Road traffic accident 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Spinal compression fracture 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Subdural haematoma 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Subdural haemorrhage 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Toxicity to various agents 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Upper limb fracture 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Investigations
Alanine aminotransferase increased 2/330 (0.6%) 2 3/334 (0.9%) 3 0/333 (0%) 0
Aspartate aminotransferase increased 0/330 (0%) 0 3/334 (0.9%) 3 0/333 (0%) 0
Lipase increased 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Neutrophil count decreased 0/330 (0%) 0 0/334 (0%) 0 3/333 (0.9%) 6
Platelet count decreased 2/330 (0.6%) 2 2/334 (0.6%) 2 1/333 (0.3%) 1
Transaminases increased 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
White blood cell count decreased 0/330 (0%) 0 1/334 (0.3%) 1 1/333 (0.3%) 1
Metabolism and nutrition disorders
Cachexia 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Decreased appetite 2/330 (0.6%) 2 1/334 (0.3%) 1 1/333 (0.3%) 1
Dehydration 2/330 (0.6%) 2 1/334 (0.3%) 1 2/333 (0.6%) 2
Fluid overload 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Hypercalcaemia 1/330 (0.3%) 1 1/334 (0.3%) 1 1/333 (0.3%) 1
Hyperglycaemia 2/330 (0.6%) 2 3/334 (0.9%) 3 1/333 (0.3%) 1
Hyperkalaemia 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Hypokalaemia 2/330 (0.6%) 2 2/334 (0.6%) 2 0/333 (0%) 0
Hyponatraemia 3/330 (0.9%) 4 0/334 (0%) 0 1/333 (0.3%) 1
Type 1 diabetes mellitus 1/330 (0.3%) 2 1/334 (0.3%) 1 0/333 (0%) 0
Musculoskeletal and connective tissue disorders
Arthralgia 0/330 (0%) 0 1/334 (0.3%) 1 1/333 (0.3%) 1
Arthritis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Back pain 0/330 (0%) 0 2/334 (0.6%) 2 0/333 (0%) 0
Bone pain 0/330 (0%) 0 2/334 (0.6%) 2 0/333 (0%) 0
Intervertebral disc protrusion 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Mandibular mass 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Muscular weakness 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Musculoskeletal chest pain 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Musculoskeletal pain 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Myositis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Polymyositis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Neoplasms benign, malignant and unspecified (incl cysts and polyps)
Cancer fatigue 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Cancer pain 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Colon cancer 1/330 (0.3%) 1 2/334 (0.6%) 2 0/333 (0%) 0
Eyelid naevus 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Neuroendocrine carcinoma metastatic 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Rectal cancer 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Tumour associated fever 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Nervous system disorders
Brain oedema 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Cerebral infarction 3/330 (0.9%) 3 2/334 (0.6%) 2 3/333 (0.9%) 4
Cerebral ischaemia 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Cerebrovascular accident 4/330 (1.2%) 4 1/334 (0.3%) 1 1/333 (0.3%) 1
Coordination abnormal 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Dizziness 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Encephalitis autoimmune 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Haemorrhagic stroke 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Headache 0/330 (0%) 0 0/334 (0%) 0 2/333 (0.6%) 2
Hemiplegia 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Ischaemic stroke 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Leukoencephalopathy 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Nervous system disorder 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Polyneuropathy in malignant disease 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Seizure 2/330 (0.6%) 2 1/334 (0.3%) 1 1/333 (0.3%) 1
Spinal cord disorder 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Syncope 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Transient ischaemic attack 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Vocal cord paralysis 0/330 (0%) 0 0/334 (0%) 0 2/333 (0.6%) 2
Product Issues
Device occlusion 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Psychiatric disorders
Anxiety 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Completed suicide 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Suicide attempt 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Renal and urinary disorders
Acute kidney injury 6/330 (1.8%) 6 4/334 (1.2%) 4 1/333 (0.3%) 1
Autoimmune nephritis 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Bladder mass 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Calculus urinary 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Chronic kidney disease 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 2
Glomerulonephritis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Nephritis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Renal failure 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Renal impairment 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Reproductive system and breast disorders
Uterine haemorrhage 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Respiratory, thoracic and mediastinal disorders
Atelectasis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Chronic obstructive pulmonary disease 2/330 (0.6%) 3 2/334 (0.6%) 2 5/333 (1.5%) 8
Dyspnoea 3/330 (0.9%) 3 1/334 (0.3%) 1 2/333 (0.6%) 2
Epistaxis 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Haemoptysis 2/330 (0.6%) 2 7/334 (2.1%) 8 3/333 (0.9%) 3
Interstitial lung disease 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Lung infiltration 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Pleural effusion 0/330 (0%) 0 1/334 (0.3%) 1 2/333 (0.6%) 2
Pneumonia aspiration 0/330 (0%) 0 2/334 (0.6%) 2 1/333 (0.3%) 1
Pneumonitis 6/330 (1.8%) 6 5/334 (1.5%) 5 1/333 (0.3%) 1
Pneumothorax 2/330 (0.6%) 2 0/334 (0%) 0 0/333 (0%) 0
Pulmonary artery thrombosis 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Pulmonary embolism 5/330 (1.5%) 5 4/334 (1.2%) 4 9/333 (2.7%) 9
Pulmonary haemorrhage 0/330 (0%) 0 2/334 (0.6%) 3 3/333 (0.9%) 3
Respiratory failure 1/330 (0.3%) 1 1/334 (0.3%) 1 0/333 (0%) 0
Skin and subcutaneous tissue disorders
Drug eruption 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Pemphigoid 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Rash 1/330 (0.3%) 1 2/334 (0.6%) 2 0/333 (0%) 0
Rash maculo-papular 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Vascular disorders
Brachiocephalic vein thrombosis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Deep vein thrombosis 3/330 (0.9%) 3 0/334 (0%) 0 1/333 (0.3%) 1
Embolism 1/330 (0.3%) 1 2/334 (0.6%) 2 1/333 (0.3%) 1
Haematoma 0/330 (0%) 0 0/334 (0%) 0 1/333 (0.3%) 1
Hypertensive crisis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Hypotension 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Lymphoedema 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Peripheral artery occlusion 0/330 (0%) 0 1/334 (0.3%) 1 0/333 (0%) 0
Shock 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Shock haemorrhagic 0/330 (0%) 0 1/334 (0.3%) 1 1/333 (0.3%) 1
Superior vena cava syndrome 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Vasculitis 1/330 (0.3%) 1 0/334 (0%) 0 0/333 (0%) 0
Other (Not Including Serious) Adverse Events
T + D + SoC D + SoC SoC Alone
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 309/330 (93.6%) 302/334 (90.4%) 301/333 (90.4%)
Blood and lymphatic system disorders
Leukopenia 41/330 (12.4%) 80 32/334 (9.6%) 59 39/333 (11.7%) 80
Anaemia 150/330 (45.5%) 213 137/334 (41%) 188 146/333 (43.8%) 203
Neutropenia 98/330 (29.7%) 177 75/334 (22.5%) 135 77/333 (23.1%) 161
Thrombocytopenia 56/330 (17%) 83 38/334 (11.4%) 67 54/333 (16.2%) 77
Endocrine disorders
Hyperthyroidism 19/330 (5.8%) 20 20/334 (6%) 21 2/333 (0.6%) 3
Hypothyroidism 39/330 (11.8%) 42 21/334 (6.3%) 24 4/333 (1.2%) 4
Gastrointestinal disorders
Abdominal pain 19/330 (5.8%) 22 16/334 (4.8%) 19 10/333 (3%) 10
Constipation 63/330 (19.1%) 85 72/334 (21.6%) 98 79/333 (23.7%) 96
Diarrhoea 65/330 (19.7%) 98 57/334 (17.1%) 72 49/333 (14.7%) 66
Nausea 136/330 (41.2%) 281 120/334 (35.9%) 258 121/333 (36.3%) 243
Stomatitis 17/330 (5.2%) 18 20/334 (6%) 25 16/333 (4.8%) 17
Vomiting 59/330 (17.9%) 82 51/334 (15.3%) 74 45/333 (13.5%) 80
General disorders
Asthenia 56/330 (17%) 67 31/334 (9.3%) 42 39/333 (11.7%) 49
Fatigue 81/330 (24.5%) 116 80/334 (24%) 119 74/333 (22.2%) 93
Mucosal inflammation 17/330 (5.2%) 19 11/334 (3.3%) 15 6/333 (1.8%) 6
Oedema peripheral 27/330 (8.2%) 35 23/334 (6.9%) 29 30/333 (9%) 34
Pyrexia 48/330 (14.5%) 65 30/334 (9%) 38 22/333 (6.6%) 30
Infections and infestations
Upper respiratory tract infection 22/330 (6.7%) 32 12/334 (3.6%) 13 18/333 (5.4%) 27
Investigations
Alanine aminotransferase increased 45/330 (13.6%) 66 43/334 (12.9%) 63 44/333 (13.2%) 59
Amylase increased 28/330 (8.5%) 47 24/334 (7.2%) 41 16/333 (4.8%) 27
Aspartate aminotransferase increased 42/330 (12.7%) 67 36/334 (10.8%) 59 38/333 (11.4%) 51
Blood creatinine increased 21/330 (6.4%) 33 12/334 (3.6%) 16 12/333 (3.6%) 21
Gamma-glutamyltransferase increased 18/330 (5.5%) 18 17/334 (5.1%) 20 11/333 (3.3%) 11
Lipase increased 20/330 (6.1%) 34 12/334 (3.6%) 23 7/333 (2.1%) 12
Neutrophil count decreased 39/330 (11.8%) 66 46/334 (13.8%) 96 59/333 (17.7%) 110
Platelet count decreased 22/330 (6.7%) 28 19/334 (5.7%) 24 29/333 (8.7%) 35
Weight decreased 23/330 (7%) 23 29/334 (8.7%) 30 20/333 (6%) 24
White blood cell count decreased 24/330 (7.3%) 44 21/334 (6.3%) 50 27/333 (8.1%) 53
Metabolism and nutrition disorders
Decreased appetite 92/330 (27.9%) 115 71/334 (21.3%) 89 81/333 (24.3%) 117
Hyperglycaemia 18/330 (5.5%) 28 18/334 (5.4%) 25 12/333 (3.6%) 13
Hypokalaemia 25/330 (7.6%) 38 11/334 (3.3%) 19 14/333 (4.2%) 20
Hyponatraemia 9/330 (2.7%) 12 16/334 (4.8%) 19 18/333 (5.4%) 25
Musculoskeletal and connective tissue disorders
Arthralgia 41/330 (12.4%) 55 28/334 (8.4%) 31 20/333 (6%) 23
Back pain 25/330 (7.6%) 27 16/334 (4.8%) 21 15/333 (4.5%) 16
Musculoskeletal chest pain 18/330 (5.5%) 21 9/334 (2.7%) 9 20/333 (6%) 20
Pain in extremity 17/330 (5.2%) 20 8/334 (2.4%) 9 8/333 (2.4%) 8
Nervous system disorders
Dizziness 20/330 (6.1%) 22 22/334 (6.6%) 28 9/333 (2.7%) 15
Headache 37/330 (11.2%) 49 23/334 (6.9%) 29 24/333 (7.2%) 26
Psychiatric disorders
Insomnia 26/330 (7.9%) 30 40/334 (12%) 42 29/333 (8.7%) 36
Respiratory, thoracic and mediastinal disorders
Cough 33/330 (10%) 39 38/334 (11.4%) 42 22/333 (6.6%) 23
Dyspnoea 30/330 (9.1%) 30 29/334 (8.7%) 32 24/333 (7.2%) 25
Haemoptysis 12/330 (3.6%) 14 16/334 (4.8%) 22 17/333 (5.1%) 24
Productive cough 11/330 (3.3%) 12 19/334 (5.7%) 20 9/333 (2.7%) 10
Skin and subcutaneous tissue disorders
Alopecia 33/330 (10%) 34 36/334 (10.8%) 38 20/333 (6%) 20
Pruritus 36/330 (10.9%) 43 30/334 (9%) 41 15/333 (4.5%) 18
Rash 63/330 (19.1%) 81 46/334 (13.8%) 58 22/333 (6.6%) 23
Vascular disorders
Hypertension 20/330 (6.1%) 21 9/334 (2.7%) 13 6/333 (1.8%) 9

Limitations/Caveats

This study also incorporates a China tail, comprising additional patients randomized after the end of the global cohort recruitment. Efficacy and safety of patients randomized in China will be reported at a later date once this separate analysis has been completed.

More Information

Certain Agreements

Principal Investigators are NOT employed by the organization sponsoring the study.

There IS an agreement between Principal Investigators and the Sponsor (or its agents) that restricts the PI's rights to discuss or publish trial results after the trial is completed.

Results Point of Contact

Name/Title Global Clinical Lead
Organization AstraZeneca
Phone 1-877-240-9479
Email information.center@astrazeneca.com
Responsible Party:
AstraZeneca
ClinicalTrials.gov Identifier:
NCT03164616
Other Study ID Numbers:
  • D419MC00004
First Posted:
May 23, 2017
Last Update Posted:
Aug 18, 2022
Last Verified:
Aug 1, 2022