Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT02420821
Other study ID # WO29637
Secondary ID 2014-004684-20
Status Completed
Phase Phase 3
First received
Last updated
Start date May 20, 2015
Est. completion date December 13, 2021

Study information

Verified date January 2023
Source Hoffmann-La Roche
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This multi-center, randomized, open-label study will evaluate the efficacy and safety of atezolizumab plus bevacizumab versus sunitinib in participants with inoperable, locally advanced, or metastatic RCC who have not received prior systemic active or experimental therapy, either in the adjuvant or metastatic setting.


Recruitment information / eligibility

Status Completed
Enrollment 915
Est. completion date December 13, 2021
Est. primary completion date February 14, 2020
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - Definitive diagnosis of unresectable locally advanced or metastatic RCC with clear-cell histology and/or a component of sarcomatoid carcinoma, with no prior treatment in the metastatic setting - Evaluable Memorial Sloan Kettering Cancer Center risk score - Measurable disease, as defined by RECIST v1.1 - Karnofsky performance status greater than or equal to 70% - Adequate hematologic and end-organ function prior to randomization Exclusion Criteria: Disease-Specific Exclusions: - Radiotherapy for RCC within 14 days prior to treatment - Active central nervous system disease - Uncontrolled pleural effusion, pericardial effusion, or ascites - Uncontrolled hypercalcemia - Any other malignancies within 5 years except for low-risk prostate cancer or those with negligible risk of metastasis or death General Medical Exclusions: - Life expectancy less than 12 weeks - Participation in another experimental drug study within 4 weeks prior to treatment - Pregnant or lactating women - Known hypersensitivity to any component of atezolizumab or other study medication - History of autoimmune disease except controlled, treated hypothyroidism or type I diabetes mellitus - History of idiopathic pulmonary fibrosis, organizing pneumonia, drug-induced pneumonitis, or idiopathic pneumonitis - Positive human immunodeficiency virus test - Active or chronic hepatitis B or C - Severe infections within 4 weeks prior to treatment - Exposure to oral or IV antibiotics within 2 weeks prior to treatment - Live attenuated vaccines within 4 weeks prior to treatment (for influenza vaccination participants must agree not to receive live, attenuated influenza vaccine within 4 weeks prior to treatment, during treatment or within 5 months following the last dose) - Significant cardiovascular disease - Prior allogeneic stem cell or solid organ transplantation Exclusion Criteria Related to Medications: - Prior treatment with cluster of differentiation 137 agonists, anti-cytotoxic T-lymphocyte associated protein-4, anti-programmed death (PD)-1, or anti-PD-L1 therapeutic antibody or pathway-targeting agents - Treatment with immunostimulatory agents for non-malignant conditions within 6 weeks or immunosuppressive agents within 2 weeks prior to treatment Bevacizumab- and Sunitinib-Specific Exclusions: - History of hypertensive crisis or hypertensive encephalopathy - Baseline electrocardiogram showing corrected QT interval greater than 460 milliseconds

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Atezolizumab (MPDL3280A), an engineered anti-PD-L1 antibody
Atezolizumab will be administered at a fixed dose of 1200 milligrams (mg) via intravenous (IV) infusion on Days 1 and 22 of each 42-day cycle.
Bevacizumab
Bevacizumab will be administered at a dose of 15 milligrams per kilogram (mg/kg) via IV infusion on Days 1 and 22 of each 42-day cycle.
Sunitinib
Sunitinib will be administered at a dose of 50 mg once daily, orally via capsule, on Day 1 through Day 28 of each 42-day cycle.

Locations

Country Name City State
Australia Lifehouse Camperdown New South Wales
Australia Austin Hospital; Medical Oncology Heidelberg Victoria
Australia Ashford Cancer Center Research Kurralta Park South Australia
Australia Macquarie University Hospital Macquarie Park New South Wales
Australia St John of God Hospital Murdoch Western Australia
Australia Icon Cancer Foundation South Brisbane Queensland
Australia Calvary Mater Newcastle; Medical Oncology Waratah New South Wales
Bosnia and Herzegovina University Clinical Centre of the Republic of Srpska Banja Luka
Brazil Hospital de Caridade de Ijui; Oncologia Ijui RS
Brazil Hospital Sao Lucas - PUCRS Porto Alegre RS
Brazil Santa Casa de Misericordia de Porto Alegre Porto Alegre RS
Brazil Instituto do Cancer do Estado de Sao Paulo - ICESP Sao Paulo SP
Canada Royal Victoria Hospital Barrie Ontario
Canada Queen Elizabeth II Health Sciences Centre; Oncology Halifax Nova Scotia
Canada Hamilton Health Sciences - Juravinski Cancer Centre Hamilton Ontario
Canada London Regional Cancer Centre London Ontario
Canada Jewish General Hospital Montreal Quebec
Canada Lakeridge Health Oshawa; Oncology Oshawa Ontario
Canada The Ottawa Hospital Cancer Centre; Oncology Ottawa Ontario
Canada CHU de Quebec Hotel-Dieu de Quebec Quebec City Quebec
Canada Princess Margaret Cancer Center Toronto Ontario
Canada Sunnybrook Odette Cancer Centre Toronto Ontario
Czechia Masarykuv onkologicky ustav Brno
Czechia Fakultni nemocnice Olomouc; Onkologicka klinika Olomouc
Czechia Fakultni Poliklinika Vseobecne Fakultni Niemocnice; Onkologicka Klinika Praha 2
Czechia Thomayerova nemocnice Praha 4 - Krc
Denmark Aarhus Universitetshospital; Kræftafdelingen Aarhus N
Denmark Herlev Hospital; Afdeling for Kræftbehandling Herlev
Denmark Odense Universitetshospital, Onkologisk Afdeling R Odense C
France ICO Paul Papin; Oncologie Medicale. Angers
France Hopital Saint Andre; Oncologie 2 Bordeaux
France Centre Francois Baclesse; Urologie Gynecologie Caen
France Centre Oscar Lambret Lille
France Centre Léon Bérard Lyon
France Institut Paoli Calmettes; Oncologie Medicale Marseille
France Centre D'Oncologie de Gentilly; Oncology Nancy
France APHP - Hospital Saint Louis Paris
France Hopital Europeen Georges Pompidou; Service D'Oncologie Medicale Paris
France ICO - Site René Gauducheau Saint Herblain
France Institut Gustave Roussy; Departement Oncologie Medicale Villejuif
Germany Universitätsklinikum "Carl Gustav Carus"; Klinik und Poliklinik für Urologie Dresden
Germany Universitätsklinikum Essen; Innere Klinik und Poliklinik für Tumorforschung Essen
Germany Nationales Centrum für Tumorerkrankungen Heidelberg (NCT); Thoraxklinik Heidelberg Heidelberg
Germany Klinikum d.Universität München Campus Großhadern München
Germany Universitätsklinikum Tübingen; Klinik für Urologie Tübingen
Italy Azienda USL8 Arezzo-Presidio Ospedaliero 1 San Donato;U.O.C. Oncologia Arezzo Toscana
Italy IRST Istituto Scientifico Romagnolo Per Lo Studio E Cura Dei Tumori, Sede Meldola; Oncologia Medica Meldola Emilia-Romagna
Italy Asst Grande Ospedale Metropolitano Niguarda; Dipartimento Di Ematologia Ed Oncologia Milano Lombardia
Italy Irccs Ospedale San Raffaele;Oncologia Medica Milano Lombardia
Italy A.O. Universitaria Policlinico Di Modena; Oncologia Modena Emilia-Romagna
Italy Az. Osp. Cardarelli; Divisione Di Oncologia Napoli Campania
Italy Fondazione IRCCS Policlinico San Matteo, Oncologia Pavia Lombardia
Italy Azienda Ospedaliera San Camillo Forlanini; Oncologia Medica Roma Lazio
Japan Nagoya University Hospital; Urology Aichi
Japan Chiba Cancer Center Chiba
Japan Kyushu University Hospital Fukuoka
Japan Gunma University Hospital Gunma
Japan Hokkaido University Hospital Hokkaido
Japan University of Tsukuba Hospital; Urology Ibaraki
Japan Iwate Medical University Hospital Iwate
Japan Kitasato University Hospital Kanagawa
Japan Yokohama City University Hospital Kanagawa
Japan Kumamoto University Hospital Kumamoto
Japan Niigata University Medical & Dental Hospital Niigata
Japan Okayama University Hospital Okayama
Japan Kindai University Hospital Osaka
Japan Osaka International Cancer Institute; Urology Osaka
Japan Osaka Metropolitan University Hospital Osaka
Japan Osaka University Hospital Osaka
Japan Tokushima University Hospital Tokushima
Japan Keio University Hospital Tokyo
Japan Nippon Medical School Hospital Tokyo
Japan The Cancer Institute Hospital, JFCR; Urology Tokyo
Japan Tokyo Medical and Dental University Hospital Tokyo
Japan Tokyo Women's Medical University Tokyo
Japan Toranomon Hospital Tokyo
Korea, Republic of Chungnam National University Hospital Daejeon
Korea, Republic of National Cancer Center Goyang-si
Korea, Republic of Seoul National University Bundang Hospital Seongnam-si
Korea, Republic of Asan Medical Center Seoul
Korea, Republic of Samsung Medical Center Seoul
Korea, Republic of Seoul National University Hospital Seoul
Korea, Republic of Severance Hospital, Yonsei University Health System Seoul
Mexico Cancerología Queretaro
Mexico Centro Oncologico Estatal ISSEMYM Toluca
Poland Centrum Onkologii Ziemi Lubelskiej im. sw. Jana z Dukli Lublin
Poland Szpital Kliniczny; Przemienienia Panskiego;Uniwersytetu Medyczny im.; Karola Marcinkowskiego w Pozna Poznan
Poland MAGODENT Sp. z o.o. Warsaw
Poland Saint Elizabeth's Hospital Warsaw
Russian Federation ALTAI REGIONAL ONCOLOGICAL CENTER; "Nadezhda" Clinic Barnaul Altaj
Russian Federation City Clinical Oncology Hospital Moscow
Russian Federation P.A. Herzen Oncological Inst. ; Oncology Moscow
Russian Federation GBUZ Nizhegorodskay Region: Clinical Diagnostic Center Nizhni Novgorod Niznij Novgorod
Singapore National Cancer Centre; Medical Oncology Singapore
Singapore National University Hospital Singapore
Spain Hospital Clínic i Provincial; Servicio de Oncología Barcelona
Spain Hospital Duran i Reynals; Oncologia Barcelona
Spain Hospital Univ Vall d'Hebron; Servicio de Oncologia Barcelona
Spain Hospital Universitario Reina Sofia; Servicio de Oncologia Córdoba Cordoba
Spain Hospital General Universitario Gregorio Marañon; Servicio de Oncologia Madrid
Spain Hospital Ramon y Cajal; Servicio de Oncologia Madrid
Spain Hospital Universitario 12 de Octubre; Servicio de Oncologia Madrid
Spain Corporacio Sanitaria Parc Tauli; Servicio de Oncologia Sabadell Barcelona
Spain Hospital Universitario Virgen del Rocio; Servicio de Oncologia Sevilla
Taiwan Taichung Veterans General Hospital; Division of Urology Taichung
Taiwan National Taiwan Uni Hospital; Dept of Oncology Taipei
Taiwan Chang Gung Medical Foundation-Linkou, Urinary Oncology Taoyuan
Thailand Chulalongkorn Hospital; Medical Oncology Bangkok
Thailand Faculty of Med. Siriraj Hosp.; Med.-Div. of Med. Oncology Bangkok
Thailand Ramathibodi Hospital; Dept of Med.-Div. of Med. Onc Bangkok
Thailand Maharaj Nakorn Chiangmai Hospital; Department of Surgery/ Urology unit Chiangmai
Thailand Songklanagarind Hospital; Department of Oncology Songkhla
Turkey Hacettepe University Medical Faculty Ankara
Turkey Trakya University Medical Faculty Research And Practice Hospital Medical Oncology Department Edirne
Turkey Istanbul Uni Cerrahpasa Medical Faculty Hospital; Medical Oncology Istanbul
United Kingdom Clatterbridge Cancer Centre Bebington
United Kingdom Queen Elizabeth Hospital Birmingham
United Kingdom Royal Blackburn Hospital Blackburn
United Kingdom Addenbrookes Nhs Trust; Oncology Clinical Trials Unit Cambridge
United Kingdom Barts Health NHS Trust - St Bartholomew's Hospital London
United Kingdom Royal Free Hospital; Dept of Oncology London
United Kingdom Christie Hospital Nhs Trust; Medical Oncology Manchester
United Kingdom Churchill Hospital; Oxford Cancer and Haematology Centre Oxford
United Kingdom Southampton General Hospital; Medical Oncology Southampton
United Kingdom Royal Marsden Hospital; Dept of Medical Oncology Sutton
United Kingdom Singleton Hospital; Pharmacy Department Swansea
United States New York Oncology Hematology,P.C.-Albany Albany New York
United States Piedmont Cancer Institute, PC Atlanta Georgia
United States University of Colorado; Anschutz Cancer Pavilion Aurora Colorado
United States Lynn Cancer Institute/Boca Raton Regional Hospital Boca Raton Florida
United States Beth Israel Deaconess Medical Center Boston Massachusetts
United States Dana Farber Cancer Inst. Boston Massachusetts
United States Massachusetts General Hospital Boston Massachusetts
United States Rocky Mountain Cancer Center; Medical Oncology Boulder Colorado
United States SCRI Tennessee Oncology Chattanooga Chattanooga Tennessee
United States The University of Chicago Chicago Illinois
United States Oncology Hematology Care Inc Cincinnati Ohio
United States Cleveland Clinic Foundation; Taussig Cancer Center Cleveland Ohio
United States Texas Oncology-Baylor Sammons Cancer Center Dallas Texas
United States Hackensack University Medical Center Hackensack New Jersey
United States Comprehensive Cancer Centers of Nevada - Eastern Avenue Las Vegas Nevada
United States Norton Cancer Institute Louisville Kentucky
United States Sarah Cannon Cancer Center and Research Institute Nashville Tennessee
United States Vanderbilt Univ Medical Ctr Nashville Tennessee
United States Memorial Sloan-Kettering Cancer Center New York New York
United States University of California at Irvine Medical Center; Department of Oncology Orange California
United States Florida Cancer Specialists - Port Charlotte Port Charlotte Florida
United States Oncology and Hematology Associates of SW Virginia-Raonoke Roanoke Virginia
United States Florida Cancer Specialist, North Region Saint Petersburg Florida
United States University of California San Francisco California
United States Seattle Cancer Care Alliance Seattle Washington
United States Northwest Cancer Specialists, P.C. Tigard Oregon
United States University of Arizona Cancer Center Tucson Arizona
United States Georgetown U; Lombardi Comp Can Washington District of Columbia

Sponsors (1)

Lead Sponsor Collaborator
Hoffmann-La Roche

Countries where clinical trial is conducted

United States,  Australia,  Bosnia and Herzegovina,  Brazil,  Canada,  Czechia,  Denmark,  France,  Germany,  Italy,  Japan,  Korea, Republic of,  Mexico,  Poland,  Russian Federation,  Singapore,  Spain,  Taiwan,  Thailand,  Turkey,  United Kingdom, 

Outcome

Type Measure Description Time frame Safety issue
Primary Percentage of Participants With Disease Progression as Determined by the Investigator According to Response Evaluation Criteria in Solid Tumors Version 1.1 (RECIST v1.1) or Death From Any Cause in Programmed Death-Ligand 1 (PD-L1)-Selected Population Tumor response was assessed by the investigator according to RECIST v1.1. Disease Progression (PD) was defined as greater than or equal to (>/=) 20 percent (%) relative increase in the sum of diameters (SoD) of all target lesions (TLs), taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 millimeters (mm); >/=1 new lesion(s); and/or unequivocal progression of existing non-TLs. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Primary Progression-Free Survival (PFS) as Determined by the Investigator According to RECIST v1.1 in PD-L1-Selected Population PFS was defined as the time from randomization to PD, as determined by the investigator per RECIST v1.1, or death from any cause, whichever occurred first. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PFS event were censored at the last tumor assessment date. Participants with no post-baseline tumor assessments were censored at the randomization date + 1 day. Participants with a PFS event who missed >/=2 scheduled assessments immediately prior to the PFS event were censored at the last tumor assessment prior to the missed visits. Median PFS was estimated by Kaplan-Meier method and 95% confidence interval (CI) was assessed using the method of Brookmeyer and Crowley. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Primary Percentage of Participants Who Died of Any Cause in ITT Population Percentage of participants who died of any cause was reported. Baseline until death from any cause (until data cut-off date 14 February 2020, up to approximately 57 months)
Primary Overall Survival (OS) in ITT Population OS was defined as the time from randomization to death due to any cause. Participants who were not reported as having died at the date of analysis were censored at the date when they were last known to be alive. Participants who did not have post-baseline information were censored at the date of randomization + 1 day. Median OS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until death from any cause (until data cut-off date 14 February 2020, up to approximately 57 months)
Secondary Percentage of Participants Who Died of Any Cause in PD-L1-Selected Population Percentage of participants who died of any cause was reported. Baseline until death from any cause (until data cut-off date 14 February 2020, up to approximately 57 months)
Secondary OS in PD-L1-Selected Population OS was defined as the time from randomization to death due to any cause. Participants who were not reported as having died at the date of analysis were censored at the date when they were last known to be alive. Participants who did not have post-baseline information were censored at the date of randomization + 1 day. Median OS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until death from any cause (until data cut-off date 14 February 2020, up to approximately 57 months)
Secondary Percentage of Participants With PD as Determined by an Independent Review Committee (IRC) According to RECIST v1.1 or Death From Any Cause in ITT Population Tumor response was assessed by an IRC according to RECIST v1.1. PD was defined as >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary PFS as Determined by an IRC According to RECIST v1.1 in ITT Population PFS was defined as the time from randomization to PD, as determined by an IRC per RECIST v1.1, or death from any cause, whichever occurred first. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PFS event were censored at the last tumor assessment date. Participants with no post-baseline tumor assessments were censored at the randomization date + 1 day. Median PFS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With PD as Determined by an IRC According to RECIST v1.1 or Death From Any Cause in PD-L1-Selected Population Tumor response was assessed by an IRC according to RECIST v1.1. PD was defined as >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary PFS as Determined by an IRC According to RECIST v1.1 in PD-L1-Selected Population PFS was defined as the time from randomization to PD, as determined by an IRC per RECIST v1.1, or death from any cause, whichever occurred first. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PFS event were censored at the last tumor assessment date. Participants with no post-baseline tumor assessments were censored at the randomization date + 1 day. Median PFS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With an Objective Response of Complete Response (CR) or Partial Response (PR) as Determined by the Investigator According to RECIST v1.1 in Objective Response Rate (ORR)-Evaluable Population Tumor response was assessed by the investigator according to RECIST v1.1. Objective response was defined as percentage of participants with a documented CR or PR. CR was defined as disappearance of all TLs/non-TLs and (if applicable) normalization of tumor marker level or reduction in short axis of any pathological lymph nodes to less than (<) 10 mm. PR was defined as >/=30% decrease in the SoD of TLs (taking as reference the baseline SoD) or persistence of >/=1 non-TL(s) and/or (if applicable) maintenance of tumor marker level above the normal limits. The 95% CI was computed using Clopper-Pearson approach. Participants without any post-baseline tumor assessments were considered non-responders. Baseline until documented CR/PR, PD, or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Duration of Response (DOR) as Determined by the Investigator According to RECIST v1.1 in DOR-Evaluable Population DOR was defined as the time from the first occurrence of CR/PR to PD as determined by the investigator per RECIST v1.1, or death from any cause, whichever occurred first. CR: disappearance of TLs/non-TLs and normalization of tumor marker level or reduction in short axis of any pathological lymph nodes to <10 mm. PR: >/=30% decrease in the SoD of TLs (taking as reference the baseline SoD) or persistence of >/=1 non-TL(s) and/or maintenance of tumor marker level above the normal limits. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PD or death after a CR/PR were censored at last tumor assessment. Participants without tumor assessments after a CR/PR were censored at first CR/PR + 1 day. Median DOR was estimated by Kaplan-Meier method and 95% CI by the method of Brookmeyer and Crowley. Baseline until documented CR/PR, PD, or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With an Objective Response of CR or PR as Determined by an IRC According to RECIST v1.1 in ORR-Evaluable Population Tumor response was assessed by an IRC according to RECIST v1.1. Objective response was defined as percentage of participants with a documented CR or PR. CR was defined as disappearance of all TLs/non-TLs and (if applicable) normalization of tumor marker level or reduction in short axis of any pathological lymph nodes to <10 mm. PR was defined as >/=30% decrease in the SoD of TLs (taking as reference the baseline SoD) or persistence of >/=1 non-TL(s) and/or (if applicable) maintenance of tumor marker level above the normal limits. The 95% CI was computed using Clopper-Pearson approach. Participants without any post-baseline tumor assessments were considered non-responders. Baseline until documented CR/PR, PD, or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary DOR as Determined by an IRC According to RECIST v1.1 in DOR-Evaluable Population DOR was defined as the time from the first occurrence of CR/PR to PD as determined by an IRC per RECIST v1.1, or death from any cause, whichever occurred first. CR: disappearance of TLs/non-TLs and normalization of tumor marker level or reduction in short axis of any pathological lymph nodes to <10 mm. PR: >/=30% decrease in the SoD of TLs (taking as reference the baseline SoD) or persistence of >/=1 non-TL(s) and/or maintenance of tumor marker level above the normal limits. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PD or death after a CR/PR were censored at last tumor assessment. Participants without tumor assessments after a CR/PR were censored at first CR/PR + 1 day. Median DOR was estimated by Kaplan-Meier method and 95% CI by the method of Brookmeyer and Crowley. Baseline until documented CR/PR, PD, or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With PD as Determined by the Investigator According to Immune-Modified RECIST or Death From Any Cause in ITT Population Tumor response was assessed by the investigator according to immune-modified RECIST. PD was defined as >/=20% relative increase in the SoD of all TLs and all new measurable lesions, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary PFS as Determined by the Investigator According to Immune-Modified RECIST in ITT Population PFS was defined as the time from randomization to PD, as determined by the investigator per immune-modified RECIST or death from any cause, whichever occurred first. PD: >/=20% relative increase in the SoD of all TLs and all new measurable lesions, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm. Participants without PFS event were censored at the last tumor assessment date. Participants with no post-baseline tumor assessments were censored at the randomization date + 1 day. Median PFS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With an Objective Response of CR or PR as Determined by the Investigator According to Immune-Modified RECIST in ORR-Evaluable Population Tumor response was assessed by the investigator according to immune-modified RECIST. Objective response was defined as percentage of participants with a documented CR or PR. CR was defined as disappearance of all TLs/non-TLs or reduction in short axis of any pathological lymph nodes to <10 mm. PR was defined as >/=30% decrease in the SoD of TLs and all new measurable lesions (taking as reference the baseline SoD), in the absence of CR. The 95% CI was computed using Clopper-Pearson approach. Participants without any post-baseline tumor assessments were considered non-responders. Baseline until documented CR/PR, PD, or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary DOR as Determined by the Investigator According to Immune-Modified RECIST in DOR-Evaluable Population DOR was defined as the time from the first occurrence of CR/PR to PD as determined by the investigator per immune-modified RECIST or death from any cause, whichever occurred first. CR: disappearance of TLs/non-TLs or reduction in short axis of any pathological lymph nodes to <10 mm. PR: >/=30% decrease in the SoD of TLs and all new measurable lesions (taking as reference the baseline SoD), in the absence of CR. PD: >/=20% relative increase in the SoD of all TLs and all new measurable lesions, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm. Participants without PD or death after a CR/PR were censored at last tumor assessment. Participants without tumor assessments after a CR/PR were censored at first CR/PR + 1 day. Median DOR was estimated by Kaplan-Meier method and 95% CI by the method of Brookmeyer and Crowley. Baseline until documented CR/PR, PD, or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With PD as Determined by the Investigator According to RECIST v1.1 or Death From Any Cause in ITT Population Tumor response was assessed by the investigator according to RECIST v1.1. PD was defined as >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary PFS as Determined by the Investigator According to RECIST v1.1 in ITT Population PFS was defined as the time from randomization to PD, as determined by the investigator per RECIST v1.1 or death from any cause, whichever occurred first. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PFS event were censored at the last tumor assessment date. Participants with no post-baseline tumor assessments were censored at the randomization date + 1 day. Participants with a PFS event who missed >/=2 scheduled assessments immediately prior to the PFS event were censored at the last tumor assessment prior to the missed visits. Median PFS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants With PD as Determined by the Investigator According to RECIST v1.1 or Death From Any Cause in Participants With Sarcomatoid Histology Tumor response was assessed by the investigator according to RECIST v1.1. PD was defined as >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary PFS as Determined by the Investigator According to RECIST v1.1 in Participants With Sarcomatoid Histology PFS was defined as the time from randomization to PD, as determined by the investigator per RECIST v1.1 or death from any cause, whichever occurred first. PD: >/=20% relative increase in the SoD of all TLs, taking as reference the smallest SoD on study, including baseline, and an absolute increase of >/=5 mm; >/=1 new lesion(s); and/or unequivocal progression of non-TLs. Participants without PFS event were censored at the last tumor assessment date. Participants with no post-baseline tumor assessments were censored at the randomization date + 1 day. Median PFS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until documented PD or death, whichever occurred first (until data cut-off date 29 September 2017, up to approximately 24 months)
Secondary Percentage of Participants Who Died of Any Cause in Participants With Sarcomatoid Histology Percentage of participants with sarcomatoid histology who died of any cause was reported. Baseline until death from any cause (until data cut-off date 14 February 2020, up to approximately 57 months)
Secondary OS in Participants With Sarcomatoid Histology OS was defined as the time from randomization to death due to any cause. Participants who were not reported as having died at the date of analysis were censored at the date when they were last known to be alive. Participants who did not have post-baseline information were censored at the date of randomization + 1 day. Median OS was estimated by Kaplan-Meier method and 95% CI was assessed using the method of Brookmeyer and Crowley. Baseline until death from any cause (until data cut-off date 14 February 2020, up to approximately 57 months)
Secondary Change From Baseline in Symptom Interference as Determined by M.D. Anderson Symptom Inventory (MDASI) Part II Score The MDASI is a cancer-related, multi-symptom, valid, and reliable self-report questionnaire that comprises of 23 items and two subscales: symptom severity (17 items) and symptom interference (6 items). In Part II, participants were asked to rate how much the symptoms have interfered with 6 areas of function (general activity, walking, work, mood, relations with other people, and enjoyment of life) in the last 24 hours. Each item was rated on a scale of 0 (does not interfere) to 10 (interfered completely) and total Part II score was calculated as an average of 6-item scores. Repeated measures model-estimated least-squares (LS) mean score for changes from baseline is reported at each timepoint, where a negative value indicates improvement. Here, 'Number Analyzed' = number of participants evaluable at specified time point. Baseline (Day 1 Cycle 1); Day 22 Cycle 1; Day 1 and 22 of every cycle from Cycle 2 up to Cycle 19; Cycle length = 42 days
Secondary Change From Baseline in Symptom Severity as Determined by MDASI Part I Score The MDASI is a cancer-related, multi-symptom, valid, and reliable self-report questionnaire that comprises of 23 items and two subscales: symptom severity (17 items) and symptom interference (6 items). In Part I, participants were asked to rate how severe the symptoms (pain, fatigue, nausea, disturbed sleep, feeling of being distressed, shortness of breath, remembering things, lack of appetite, drowsy, dry mouth, feeling sad, vomiting, numbness or tingling, rash/skin changes, headache, mouth/throat sores, and diarrhea) were when "at their worst" in the last 24 hours. Each item was rated on a scale of 0 (not present) to 10 (as bad as you can imagine). Mixed-effects model-estimated LS mean score for change from baseline at the end-of treatment is reported for each item, where a negative value indicates improvement. Baseline; End of Treatment (EoT) visit (up to approximately 27 months)
Secondary Change From Baseline in Symptom Severity as Determined by Brief Fatigue Inventory (BFI) Interference Scale Score The BFI is a valid and reliable self-report questionnaire used to assess the severity and impact of cancer-related fatigue. BFI interference subscale (6 items) assessed the impact of fatigue on global domains (general activity, mood, walking ability, normal work, relations with other people, and enjoyment of life) in the last 24 hours. Each item was rated on a scale of 0 (does not interfere) to 10 (interfered completely). Change from baseline in the mean score of all 6 items at each timepoint is reported, where a negative value indicates improvement. Baseline (Day 1 Cycle 1); every week for first 12 weeks, Days 1 and 22 of each cycle (Cycle 3 up to 19), within 30 days of PD (up to 27 months), at EoT (up to 27 months) and at 6, 12, 24, and 36 weeks after EoT (overall up to 27 months); 1 cycle=42 days
Secondary Change From Baseline in Symptom Severity as Determined by BFI Worst Fatigue Item The BFI is a valid and reliable self-report questionnaire used to assess the severity and impact of cancer-related fatigue. BFI worst fatigue item assessed the severity of fatigue at its worst in the last 24 hours. The item was rated on a scale of 0 (not present) to 10 (as bad as you can imagine). Change from baseline in the score at each time point is reported, where a negative value indicates improvement. Baseline (Day 1 Cycle 1); every week for first 12 weeks, Days 1 and 22 of each cycle (Cycle 3 up to 19), within 30 days of PD (up to 27 months), at EoT (up to 27 months) and at 6, 12, 24, and 36 weeks after EoT (overall up to 27 months); 1 cycle=42 days
Secondary Change From Baseline in Treatment Side Effects Burden as Determined by Functional Assessment of Cancer Therapy Kidney Symptom Index (FKSI-19) General Population 5 (GP5) Item Score The FKSI-19 is a 19-item tool designed to assess the most important symptoms and concerns related to treatment effectiveness in advanced kidney cancer. The FKSI-19 GP5 item (bothered by the side effect of treatment) assessed side effects burden in the past 7 days on a 5-point scale (0=not at all, 1=a little bit, 2=somewhat, 3=quite a bit, 4=very much). Repeated measures model-estimated LS mean score for changes from baseline is reported at each timepoint, where a negative value indicates improvement. Day 1 and 22 of every cycle (Baseline = Day 1 Cycle 1) up to Cycle 19; Cycle length = 42 days
Secondary Number of Participants With Anti-Therapeutic Antibodies (ATAs) Against Atezolizumab The number of participants with "Treatment-induced ATAs" and "Treatment-enhanced ATA" against atezolizumab at any time during or after treatment was reported. Treatment-induced ATA = a participant with negative or missing Baseline ATA result(s) and at least one positive post-Baseline ATA result. Treatment-enhanced ATA = a participant with positive ATA result at Baseline who has one or more post Baseline titer results that are at least 0.60 titer unit greater than the Baseline titer result. Here, 'Overall Number of Participants Analyzed' = number of participants with a non-missing baseline ATA sample; 'Number Analyzed' = number of participants with a non-missing ATA sample at indicated timepoint. Baseline (Predose [Hour 0] at Day 1 Cycle 1); Post-Baseline (Predose at Cycles 2, 4, and 8, and every eight cycles thereafter up to EoT [up to approximately 27 months] and 120 days after EoT [up to approximately 27 months]) (Cycle length=42 days)
Secondary Number of Participants With ATAs Against Bevacizumab The number of participants with "Treatment-induced ATAs" and "Treatment-enhanced ATA" against bevacizumab at any time during or after treatment was reported. Treatment-induced ATA = a participant with negative or missing Baseline ATA result(s) and at least one positive post-Baseline ATA result. Treatment-enhanced ATA = a participant with positive ATA result at Baseline who has one or more post Baseline titer results that are at least 0.60 titer unit greater than the Baseline titer result. Baseline (Predose [Hour 0] at Day 1 Cycle 1); Post-Baseline (Predose at Cycle 3, at EoT [up to approximately 27 months] and at 120 days after EoT [up to approximately 27 months]) (Cycle length=42 days)
Secondary Maximum Observed Serum Concentration (Cmax) for Atezolizumab Cmax for atezolizumab was estimated from plasma concentration versus time data. 30 minutes after the end of bevacizumab infusion (atezolizumab infusion duration: 30-60 min; bevacizumab infusion duration: 30-90 minutes) on Day 1 of Cycle 1 (Cycle length = 42 days)
Secondary Minimum Observed Serum Concentration (Cmin) for Atezolizumab Cmin for atezolizumab was estimated from plasma concentration versus time data. Predose (Hour 0) on Day 22 of Cycle 1; predose (Hour 0) on Day 1 of Cycles 2; Cycle length = 42 days
Secondary Cmax for Bevacizumab Cmax for bevacizumab was estimated from plasma concentration versus time data. 30 minutes after the end of bevacizumab infusion (atezolizumab infusion duration: 30-60 min; bevacizumab infusion duration: 30-90 minutes) on Day 1 of Cycle 1 (Cycle length = 42 days)
Secondary Cmin for Bevacizumab Cmin for bevacizumab was estimated from plasma concentration versus time data. Pre-dose (Hour 0) on Day 1 of Cycle 3 (Cycle length = 42 days)
See also
  Status Clinical Trial Phase
Active, not recruiting NCT04987203 - Study to Compare Tivozanib in Combination With Nivolumab to Tivozanib Monotherapy in Subjects With Renal Cell Carcinoma Phase 3
Recruiting NCT06391879 - Establishment of a Multidimensional Prediction Model for the Natural Course of VHL Disease-related Renal Cell Carcinoma
Completed NCT02526017 - Study of Cabiralizumab in Combination With Nivolumab in Patients With Selected Advanced Cancers Phase 1
Recruiting NCT05059444 - ORACLE: Observation of ResiduAl Cancer With Liquid Biopsy Evaluation
Terminated NCT03655613 - APL-501 or Nivolumab in Combination With APL-101 in Locally Advanced or Metastatic HCC and RCC Phase 1/Phase 2
Active, not recruiting NCT03170960 - Study of Cabozantinib in Combination With Atezolizumab to Subjects With Locally Advanced or Metastatic Solid Tumors Phase 1/Phase 2
Withdrawn NCT05418387 - A Social Support Intervention to Improve Treatment Among Hispanic Kidney and Liver Cancer Patients in Arizona N/A
Recruiting NCT04623502 - An Investigation of Kidney and Urothelial Tumor Metabolism in Patients Undergoing Surgical Resection and/or Biopsy N/A
Completed NCT02853344 - Study of Pembrolizumab (MK-3475) Monotherapy in Locally Advanced/Metastatic Renal Cell Carcinoma (MK-3475-427/KEYNOTE-427) Phase 2
Terminated NCT04088500 - A Study of Combination Nivolumab and Ipilimumab Retreatment in Patients With Advanced Renal Cell Carcinoma Phase 2
Completed NCT05070637 - Circulating Tumor Cell Reducing No-touch Nephrectomy N/A
Active, not recruiting NCT03634540 - A Trial of Belzutifan (PT2977, MK-6482) in Combination With Cabozantinib in Patients With Clear Cell Renal Cell Carcinoma (ccRCC) (MK-6482-003) Phase 2
Not yet recruiting NCT06049030 - A Study of HS-10516 in Patients With Advanced Clear Cell Renal Cell Carcinoma Phase 1
Completed NCT03652077 - A Safety and Tolerability Study of INCAGN02390 in Select Advanced Malignancies Phase 1
Completed NCT01358721 - Phase I Biomarker Study (BMS-936558) Phase 1
Active, not recruiting NCT04503148 - Anesthesia and Cancer Study: Renal Cell Carcinoma N/A
Completed NCT02386826 - INC280 Combined With Bevacizumab in Patients With Glioblastoma Multiforme Phase 1
Not yet recruiting NCT05808608 - A Study of AK104 Plus Axitinib in Advanced/Metastatic Special Pathological Subtypes of Renal Cell Carcinoma Phase 1/Phase 2
Withdrawn NCT03323710 - Study of Propranolol Plus Sunitinib in First-line Treatment of Metastatic Renal Cell Carcinoma Phase 2
Not yet recruiting NCT02787915 - DC1s-CTL Cellular Therapy for Renal Cell Carcinoma Phase 1/Phase 2