Clinical Trials Logo

Clinical Trial Details — Status: Active, not recruiting

Administrative data

NCT number NCT02453282
Other study ID # D419AC00001
Secondary ID 2015-001279-39
Status Active, not recruiting
Phase Phase 3
First received
Last updated
Start date July 21, 2015
Est. completion date December 31, 2024

Study information

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

Clinical Trial Summary

This is a randomized, open-label, multi-center, global, Phase III study to determine the efficacy and safety of MEDI4736 + tremelimumab combination therapy and MEDI4736 monotherapy versus platinum-based SoC chemotherapy in the first-line treatment of patients with epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) wild-type locally advanced or metastatic NSCLC


Description:

Patients will be randomized in a 1:1:1 ratio to receive treatment with MEDI4736 + tremelimumab combination therapy, MEDI4736 monotherapy, or Standard of Care (SoC) therapy.


Recruitment information / eligibility

Status Active, not recruiting
Enrollment 1118
Est. completion date December 31, 2024
Est. primary completion date October 4, 2018
Accepts healthy volunteers No
Gender All
Age group 18 Years to 130 Years
Eligibility Inclusion Criteria: For inclusion in the study, patients should fulfill the following criteria: - Aged at least 18 years - Documented evidence of Stage IV NSCLC - No sensitizing EGFR mutation or ALK rearrangement - No prior chemotherapy or any other systemic therapy for recurrent/metastatic NSCLC - World Health Organization (WHO) Performance Status of 0 or 1 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. Brain metastases or spinal cord compression unless asymptomatic, treated and stable (not requiring steroids) 3. Prior exposure to Immunomodulatory therapy (IMT), including, but not limited to, other anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), anti-programmed cell death1 (PD-1), anti-programmed cell death ligand 1 (PD-L1), or anti PD-L2 antibodies, excluding therapeutic anticancer vaccines 4. Active or prior documented autoimmune or inflammatory disorders (including inflammatory bowel disease [eg, colitis or Crohn's disease]

Study Design


Related Conditions & MeSH terms


Intervention

Biological:
MEDI4736 (Durvalumab)

MEDI4736 (Durvalumab)+Tremelimumab

Drug:
Paclitaxel + Carboplatin
Chemotherapy Agents
Gemcitabine + Cisplatin
Chemotherapy Agents
Gemcitabine + Carboplatin
Chemotherapy Agents
Pemetrexed + Cisplatin
Chemotherapy Agents
Pemetrexed + Carboplatin
Chemotherapy Agents
Biological:
Tremelimumab


Locations

Country Name City State
Australia Research Site Box Hill
Australia Research Site Gosford
Australia Research Site Kogarah
Australia Research Site Melbourne
Australia Research Site Port Macquarie
Australia Research Site Southport
Australia Research Site St Leonards
Belgium Research Site Brussels
Belgium Research Site Charleroi
Belgium Research Site Duffel
Belgium Research Site Leuven
Belgium Research Site Liège
Canada Research Site Kingston Ontario
Canada Research Site Newmarket Ontario
Canada Research Site Oshawa Ontario
Canada Research Site Regina Saskatchewan
Canada Research Site Saskatoon Saskatchewan
Canada Research Site Sault Ste. Marie Ontario
Canada Research Site St. Catharines Ontario
Canada Research Site Sudbury Ontario
Canada Research Site Toronto Ontario
France Research Site Bordeaux Cedex
France Research Site Brest Cedex
France Research Site Creteil
France Research Site Lille
France Research Site Lyon Cedex 08
France Research Site Marseille cedex
Germany Research Site Aachen
Germany Research Site Bad Berka
Germany Research Site Berlin
Germany Research Site Berlin
Germany Research Site Freiburg
Germany Research Site Gauting
Germany Research Site Hamburg
Germany Research Site Heidelberg
Germany Research Site Hemer
Germany Research Site Homburg/Saar
Germany Research Site Immenhausen
Germany Research Site Löwenstein
Germany Research Site Lubeck
Germany Research Site Mainz
Germany Research Site Münster
Germany Research Site Oldenburg
Germany Research Site Ulm
Germany Research Site Velbert
Germany Research Site Würzburg
Hungary Research Site Budapest
Hungary Research Site Deszk
Hungary Research Site Edelény
Hungary Research Site Kaposvár
Hungary Research Site Kecskemét
Hungary Research Site Miskolc
Hungary Research Site Nyíregyháza
Hungary Research Site Pécs
Hungary Research Site Székesfehérvár
Italy Research Site Genova
Italy Research Site Meldola
Italy Research Site Milano
Italy Research Site Milano
Italy Research Site Milano
Italy Research Site San Giovanni Rotondo
Italy Research Site Siena
Japan Research Site Fukushima-shi
Japan Research Site Himeji-shi
Japan Research Site Hirakata-shi
Japan Research Site Hirosaki-shi
Japan Research Site Iizuka-shi
Japan Research Site Iwakuni-shi
Japan Research Site Izumi-shi
Japan Research Site Kanazawa
Japan Research Site Kishiwada-shi
Japan Research Site Kobe-shi
Japan Research Site Koga-shi
Japan Research Site Kyoto-shi
Japan Research Site Kyoto-shi
Japan Research Site Mitaka-shi
Japan Research Site Nagaoka-shi
Japan Research Site Nagoya-shi
Japan Research Site Nagoya-shi
Japan Research Site Okayama-shi
Japan Research Site Osaka-shi
Japan Research Site Osaka-shi
Japan Research Site Osakasayama
Japan Research Site Saga-shi
Japan Research Site Saitama-shi
Japan Research Site Sakai-shi
Japan Research Site Sendai-shi
Japan Research Site Shinjuku-ku
Japan Research Site Tokushima-shi
Japan Research Site Ube-shi
Japan Research Site Yokohama-shi
Japan Research Site Yokohama-shi
Japan Research Site Yokosuka-shi
Korea, Republic of Research Site Busan
Korea, Republic of Research Site Changwon-si
Korea, Republic of Research Site Cheongju-si
Korea, Republic of Research Site Daegu
Korea, Republic of Research Site Daegu
Korea, Republic of Research Site Daejeon
Korea, Republic of Research Site Goyang-si
Korea, Republic of Research Site Incheon
Korea, Republic of Research Site Jinju-si
Korea, Republic of Research Site Seongnam-si
Korea, Republic of Research Site Seongnam-Si
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Seoul
Korea, Republic of Research Site Suwon-si
Korea, Republic of Research Site Ulsan
Netherlands Research Site Amsterdam
Netherlands Research Site Arnhem
Netherlands Research Site Breda
Netherlands Research Site Den Bosch
Netherlands Research Site Groningen
Netherlands Research Site Maastricht
Netherlands Research Site Rotterdam
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Moscow
Russian Federation Research Site Obninsk
Russian Federation Research Site Omsk
Russian Federation Research Site Saint Petersburg
Russian Federation Research Site Saint-Petersburg
Russian Federation Research Site St. Petersburg
Russian Federation Research Site St. Petersburg
Spain Research Site A Coruña
Spain Research Site Alicante
Spain Research Site Barcelona
Spain Research Site Barcelona
Spain Research Site Gerona
Spain Research Site Jaén
Spain Research Site León
Spain Research Site Madrid
Spain Research Site Madrid
Spain Research Site Madrid
Spain Research Site Majadahonda
Spain Research Site Málaga
Spain Research Site Sevilla
Spain Research Site Sevilla
Spain Research Site Valencia
Switzerland Research Site Bellinzona
Switzerland Research Site Lausanne
Taiwan Research Site Kaohsiung
Taiwan Research Site Kaohsiung
Taiwan Research Site Taichung
Taiwan Research Site Tainan City
Taiwan Research Site Taipei
Taiwan Research Site Taipei
Taiwan Research Site Tao-Yuan
Thailand Research Site Bangkok
Thailand Research Site Chiang Mai
Thailand Research Site Hat Yai
Thailand Research Site Khon Kaen
United States Research Site Athens Georgia
United States Research Site Bakersfield California
United States Research Site Baltimore Maryland
United States Research Site Charlotte North Carolina
United States Research Site Cleveland Ohio
United States Research Site Fullerton California
United States Research Site Honolulu Hawaii
United States Research Site Jacksonville Florida
United States Research Site La Jolla California
United States Research Site Los Angeles California
United States Research Site Los Angeles California
United States Research Site Madison Wisconsin
United States Research Site Mineola New York
United States Research Site Minneapolis Minnesota
United States Research Site Nashville Tennessee
United States Research Site Nashville Tennessee
United States Research Site New Haven Connecticut
United States Research Site New York New York
United States Research Site New York New York
United States Research Site New York New York
United States Research Site North Charleston South Carolina
United States Research Site Omaha Nebraska
United States Research Site Pembroke Pines Florida
United States Research Site Redondo Beach California
United States Research Site Richmond Virginia
United States Research Site Sacramento California
United States Research Site Saint Louis Missouri
United States Research Site San Luis Obispo California
United States Research Site Santa Maria California
United States Research Site Scottsdale Arizona
United States Research Site Summit New Jersey
United States Research Site Tampa Florida
United States Research Site Tucson Arizona
United States Research Site West Hollywood California
United States Research Site Yuma Arizona
Vietnam Research Site Hanoi
Vietnam Research Site Hanoi
Vietnam Research Site Hanoi City
Vietnam Research Site Ho Chi Minh
Vietnam Research Site Ho Chi Minh
Vietnam Research Site Ho Chi Minh city

Sponsors (1)

Lead Sponsor Collaborator
AstraZeneca

Countries where clinical trial is conducted

United States,  Vietnam,  Australia,  Belgium,  Canada,  France,  Germany,  Hungary,  Italy,  Japan,  Korea, Republic of,  Netherlands,  Russian Federation,  Spain,  Switzerland,  Taiwan,  Thailand, 

Outcome

Type Measure Description Time frame Safety issue
Primary Overall Survival (OS); PD-L1 (TC >=25%) Analysis Set Population, Durvalumab Monotherapy Vs SoC Chemotherapy and Durvalumab + Tremelimumab Vs SoC Chemotherapy The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. From baseline (Day 1, Week 0) until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).
Primary Progression-Free Survival (PFS); PD-L1 (TC >=25%) Analysis Set Population, Durvalumab + Tremelimumab Vs SoC Chemotherapy The PFS per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST 1.1) using blinded independent central review (BICR) assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). Progressive disease (PD) was defined as at least a 20% increase in the sum of diameters of target lesions (TLs) and an absolute increase of at least 5 millimeter (mm), taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary OS; PD-L1 (TC >=25%) Analysis Set Population, Durvalumab + Tremelimumab Vs Durvalumab Monotherapy The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. From baseline until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary OS; PD-L1 (TC >=1%) Analysis Set Population The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. From baseline until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary OS; FAS Population The OS was defined as the time from the date of randomization until death due to any cause (ie, date of death or censoring - date of randomization + 1). Any participant not known to have died at the time of analysis were censored based on the last recorded date on which the participant was known to be alive. Median OS was calculated using the Kaplan-Meier technique. From baseline until death due to any cause, assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary PFS; PD-L1 (TC >=25%) Analysis Set Population, Durvalumab Monotherapy Vs SoC Chemotherapy and Durvalumab + Tremelimumab Vs Durvalumab Monotherapy The PFS per RECIST 1.1 using BICR assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). The PD was defined as at least a 20% increase in the sum of diameters of TLs and an absolute increase of at least 5 mm, taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary PFS; PD-L1 (TC >=1%) Analysis Set Population The PFS per RECIST 1.1 using BICR assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). The PD was defined as at least a 20% increase in the sum of diameters of TLs and an absolute increase of at least 5 mm, taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary PFS; FAS Population The PFS per RECIST 1.1 using BICR assessments was defined as the time from the date of randomization until the date of objective disease progression or death (by any cause in the absence of progression) regardless of whether the participant withdraw from randomized therapy or received another anti-cancer therapy prior to progression (ie, date of PFS event or censoring - date of randomization + 1). The PD was defined as at least a 20% increase in the sum of diameters of TLs and an absolute increase of at least 5 mm, taking as reference the smallest sum of diameters since treatment started including the baseline sum of diameters. Median PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary Objective Response Rate (ORR); PD-L1 (TC >=25%) Analysis Set Population The ORR per RECIST 1.1 using BICR assessments was defined as the percentage of participants with at least 1 visit response of Complete Response (CR) or Partial Response (PR). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary ORR; PD-L1 (TC >=1%) Analysis Set Population The ORR per RECIST 1.1 using BICR assessments was defined as the percentage of participants with at least 1 visit response of CR or PR. The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary ORR; FAS Population The ORR per RECIST 1.1 using BICR assessments was defined as the percentage of participants with at least 1 visit response of CR or PR. The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary Duration of Response (DoR); PD-L1 (TC >=25%) Analysis Set Population The DoR per RECIST 1.1 using BICR assessments was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (ie, date of PFS event or censoring - date of first response + 1). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary DoR; PD-L1 (TC >=1%) Analysis Set Population The DoR per RECIST 1.1 using BICR assessments was defined as the time from the date of first documented response (CR or PR) until the first date of documented progression or death in the absence of disease progression (ie, date of PFS event or censoring - date of first response + 1). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary DoR; FAS Population The DoR per RECIST 1.1 using BICR assessments was defined as the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (ie, date of PFS event or censoring - date of first response + 1). The CR was defined as disappearance of all TLs (any pathological lymph nodes selected as TLs must have a reduction in short axis to <10 mm) and PR was defined as at least a 30% decrease in the sum of diameters of TLs (taking as reference the baseline sum of diameters as long as criteria for PD are not met). Tumour scans performed at baseline then every 6 weeks up to 48 weeks relative to the date of randomization, then every 8 weeks thereafter until confirmed disease progression. Assessed up to the data cut-off date (a maximum of approximately 3 years).
Secondary Percentage of Participants Alive and Progression Free at 12 Months (APF12); PD-L1 (TC >=25%) Analysis Set Population The APF12 was defined as the percentage of participants who were alive and progression free per RECIST v1.1 using BICR assessments at 12 months after randomization. The PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 12 months.
Secondary Percentage of Participants APF12; PD-L1 (TC >=1%) Analysis Set Population The APF12 was defined as the percentage of participants who were alive and progression free per RECIST v1.1 using BICR assessments at 12 months after randomization. The PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 12 months.
Secondary Percentage of Participants APF12; FAS Population The APF12 was defined as the percentage of participants who were alive and progression free per RECIST v1.1 using BICR assessments at 12 months after randomization. The PFS was calculated using the Kaplan-Meier technique. Tumour scans performed at baseline then every 6 weeks up to 12 months.
Secondary Time From Randomization to Second Progression (PFS2); PD-L1 (TC >=25%) Analysis Set Population The PFS2 was defined as the time from the date of randomization to the earliest of the progression events (subsequent to that used for the primary variable PFS and excluding any confirmation of progression scans performed for first progression) or death (ie, date of PFS2 event or censoring - date of randomization + 1). The second progression event was determined by local standard clinical practice which may have included any of the following: objective radiological imaging, symptomatic progression, or death. Tumour scans performed at baseline then every 6 weeks up to Week 48, then every 8 weeks thereafter until 1st progression. Disease then assessed per local practice until 2nd progression. Assessed up to data cut-off date (maximum of approximately 3 years).
Secondary PFS2; PD-L1 (TC >=1%) Analysis Set Population The PFS2 was defined as the time from the date of randomization to the earliest of the progression events (subsequent to that used for the primary variable PFS and excluding any confirmation of progression scans performed for first progression) or death (ie, date of PFS2 event or censoring - date of randomization + 1). The second progression event was determined by local standard clinical practice which may have included any of the following: objective radiological imaging, symptomatic progression, or death. Tumour scans performed at baseline then every 6 weeks up to Week 48, then every 8 weeks thereafter until 1st progression. Disease then assessed per local practice until 2nd progression. Assessed up to data cut-off date (maximum of approximately 3 years).
Secondary PFS2; FAS Population The PFS2 was defined as the time from the date of randomization to the earliest of the progression events (subsequent to that used for the primary variable PFS and excluding any confirmation of progression scans performed for first progression) or death (ie, date of PFS2 event or censoring - date of randomization + 1). The second progression event was determined by local standard clinical practice which may have included any of the following: objective radiological imaging, symptomatic progression, or death. Tumour scans performed at baseline then every 6 weeks up to Week 48, then every 8 weeks thereafter until 1st progression. Disease then assessed per local practice until 2nd progression. Assessed up to data cut-off date (maximum of approximately 3 years).
Secondary Change From Baseline in Disease-Related Symptoms as Assessed by European Organisation for Research and Treatment of Cancer Quality of Life Questionnaires (EORTC QLQ) at 12 Months Patient reported outcomes for 5 disease related symptoms was assessed using the EORTC QLQ-Core 30 (C30) items questionnaire (fatigue and appetite loss) and the EORTC QLQ-Lung Cancer module 13 (LC13) (dysponea, cough and chest pain). An outcome variable consisting of a score from 0 to 100 was derived for each of the symptom scales/symptom items with higher scores representing greater symptom severity. An improvement in symptoms were indicated by a negative change in score from baseline. A positive change in score from baseline indicated a deterioration of symptoms. A minimum clinically meaningful change is defined as an absolute change in the score from baseline of >=10. At baseline then every 4 weeks for the first 8 weeks relative to the date of randomization, then every 8 weeks until second progression/death, whichever comes first. Assessed up to 12 months.
Secondary Serum Concentrations of Durvalumab Blood samples were collected to determine the serum concentration of durvalumab. Pre-dose and within 1 hour after end of infusion at Week 0, 12 and 24, and at follow-up Month 3.
Secondary Serum Concentrations of Tremelimumab Blood samples were collected to determine the serum concentration of tremelimumab. Pre-dose and within 1 hour after end of infusion at Week 0 and 12, and at follow-up Month 3.
Secondary Maximum Serum Concentration at Steady State (Cmax_ss) of Durvalumab Blood samples were collected to determine the Cmax_ss of durvalumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. Within 1 hour after end of infusion on infusion day at Week 12.
Secondary Cmax_ss of Tremelimumab Blood samples were collected to determine the Cmax_ss of tremelimumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. Within 1 hour after end of infusion on infusion day at Week 12.
Secondary Trough Serum Concentration at Steady State (Ctrough_ss) of Durvalumab Blood samples were collected to determine the Ctrough_ss of durvalumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. Pre-dose at Week 12.
Secondary Ctrough_ss of Tremelimumab Blood samples were collected to determine the Ctrough_ss of tremelimumab. Steady state was defined as Cycle 4 (Week 12). PK parameters were determined using standard non-compartmental methods. Pre-dose at Week 12.
Secondary Number of Participants With Anti-Drug Antibody (ADA) Response to Durvalumab Blood samples were measured for the presence of ADAs and ADA-neutralizing antibodies (nAb) for durvalumab using validated assays. Tiered analysis was performed to include screening, confirmatory, and titer assay components, and positive-negative cut points previously statistically determined from drug-naïve validation samples were employed. Immunogenicity results were analyzed by summarizing the number of participants who developed detectable ADAs against durvalumab. Persistently positive is defined as having at least 2 post-baseline ADA positive measurements with at least 16 weeks (112 days) between the first and last positive measurements, or an ADA positive result at the last available assessment. Transiently positive is defined as having at least one post-baseline ADA positive measurement and not fulfilling the conditions for persistently positive. At Weeks 0, 12, and 24; 3 and 6 months after last dose of study treatment.
Secondary Number of Participants With ADA Response to Tremelimumab Blood samples were measured for the presence of ADAs and ADA-nAb for tremelimumab using validated assays. Tiered analysis was performed to include screening, confirmatory, and titer assay components, and positive-negative cut points previously statistically determined from drug-naïve validation samples were employed. Immunogenicity results were analyzed by summarizing the number of participants who developed detectable ADAs against tremelimumab. Persistently positive is defined as having at least 2 post-baseline ADA positive measurements with at least 16 weeks (112 days) between the first and last positive measurements, or an ADA positive result at the last available assessment. Transiently positive is defined as having at least one post-baseline ADA positive measurement and not fulfilling the conditions for persistently positive. At Weeks 0 and 12; 3 and 6 months after last dose of study treatment.

External Links