45
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma : The CheckMate 143 Phase 3 Randomized Clinical Trial

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Key Points

          Question

          Does programmed cell death 1 immune checkpoint inhibition with nivolumab improve overall survival compared with bevacizumab treatment for patients with recurrent glioblastoma?

          Findings

          In this randomized phase 3 clinical trial of 369 patients diagnosed with recurrent glioblastoma treated with nivolumab, an improved survival benefit was not observed in patients who received nivolumab compared with bevacizumab-treated control patients.

          Meaning

          Additional research is needed; nivolumab monotherapy did not improve overall survival compared with bevacizumab in the treatment of recurrent glioblastoma. A study of nivolumab in combination with radiotherapy and temozolomide in patients with newly diagnosed glioblastoma with methylated MGMT promoter is ongoing.

          Abstract

          This randomized clinical trial examines the effect of programmed cell death 1 immune checkpoint inhibition with nivolumab on overall survival for patients with recurrent glioblastoma.

          Abstract

          Importance

          Clinical outcomes for glioblastoma remain poor. Treatment with immune checkpoint blockade has shown benefits in many cancer types. To our knowledge, data from a randomized phase 3 clinical trial evaluating a programmed death-1 (PD-1) inhibitor therapy for glioblastoma have not been reported.

          Objective

          To determine whether single-agent PD-1 blockade with nivolumab improves survival in patients with recurrent glioblastoma compared with bevacizumab.

          Design, Setting, and Participants

          In this open-label, randomized, phase 3 clinical trial, 439 patients with glioblastoma at first recurrence following standard radiation and temozolomide therapy were enrolled, and 369 were randomized. Patients were enrolled between September 2014 and May 2015. The median follow-up was 9.5 months at data cutoff of January 20, 2017. The study included 57 multicenter, multinational clinical sites.

          Interventions

          Patients were randomized 1:1 to nivolumab 3 mg/kg or bevacizumab 10 mg/kg every 2 weeks until confirmed disease progression, unacceptable toxic effects, or death.

          Main Outcomes and Measures

          The primary end point was overall survival (OS).

          Results

          A total of 369 patients were randomized to nivolumab (n = 184) or bevacizumab (n = 185). The MGMT promoter was methylated in 23.4% (43/184; nivolumab) and 22.7% (42/185; bevacizumab), unmethylated in 32.1% (59/184; nivolumab) and 36.2% (67/185; bevacizumab), and not reported in remaining patients. At median follow-up of 9.5 months, median OS (mOS) was comparable between groups: nivolumab, 9.8 months (95% CI, 8.2-11.8); bevacizumab, 10.0 months (95% CI, 9.0-11.8); HR, 1.04 (95% CI, 0.83-1.30); P = .76. The 12-month OS was 42% in both groups. The objective response rate was higher with bevacizumab (23.1%; 95% CI, 16.7%-30.5%) vs nivolumab (7.8%; 95% CI, 4.1%-13.3%). Grade 3/4 treatment-related adverse events (TRAEs) were similar between groups (nivolumab, 33/182 [18.1%]; bevacizumab, 25/165 [15.2%]), with no unexpected neurological TRAEs or deaths due to TRAEs.

          Conclusions and Relevance

          Although the primary end point was not met in this randomized clinical trial, mOS was comparable between nivolumab and bevacizumab in the overall patient population with recurrent glioblastoma. The safety profile of nivolumab in patients with glioblastoma was consistent with that in other tumor types.

          Trial Registration

          ClinicalTrials.gov Identifier: NCT02017717

          Related collections

          Most cited references14

          • Record: found
          • Abstract: found
          • Article: not found

          Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck

          Background Patients with recurrent or metastatic squamous-cell carcinoma of the head and neck after platinum chemotherapy have a very poor prognosis and limited therapeutic options. Nivolumab, an anti-programmed death 1 (PD-1) monoclonal antibody, was assessed as treatment for this condition. Methods In this randomized, open-label, phase 3 trial, we assigned, in a 2:1 ratio, 361 patients with recurrent squamous-cell carcinoma of the head and neck whose disease had progressed within 6 months after platinum-based chemotherapy to receive nivolumab (at a dose of 3 mg per kilogram of body weight) every 2 weeks or standard, single-agent systemic therapy (methotrexate, docetaxel, or cetuximab). The primary end point was overall survival. Additional end points included progression-free survival, rate of objective response, safety, and patient-reported quality of life. Results The median overall survival was 7.5 months (95% confidence interval [CI], 5.5 to 9.1) in the nivolumab group versus 5.1 months (95% CI, 4.0 to 6.0) in the group that received standard therapy. Overall survival was significantly longer with nivolumab than with standard therapy (hazard ratio for death, 0.70; 97.73% CI, 0.51 to 0.96; P=0.01), and the estimates of the 1-year survival rate were approximately 19 percentage points higher with nivolumab than with standard therapy (36.0% vs. 16.6%). The median progression-free survival was 2.0 months (95% CI, 1.9 to 2.1) with nivolumab versus 2.3 months (95% CI, 1.9 to 3.1) with standard therapy (hazard ratio for disease progression or death, 0.89; 95% CI, 0.70 to 1.13; P=0.32). The rate of progression-free survival at 6 months was 19.7% with nivolumab versus 9.9% with standard therapy. The response rate was 13.3% in the nivolumab group versus 5.8% in the standard-therapy group. Treatment-related adverse events of grade 3 or 4 occurred in 13.1% of the patients in the nivolumab group versus 35.1% of those in the standard-therapy group. Physical, role, and social functioning was stable in the nivolumab group, whereas it was meaningfully worse in the standard-therapy group. Conclusions Among patients with platinum-refractory, recurrent squamous-cell carcinoma of the head and neck, treatment with nivolumab resulted in longer overall survival than treatment with standard, single-agent therapy. (Funded by Bristol-Myers Squibb; CheckMate 141 ClinicalTrials.gov number, NCT02105636 .).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial.

            In 2004, a randomised phase III trial by the European Organisation for Research and Treatment of Cancer (EORTC) and National Cancer Institute of Canada Clinical Trials Group (NCIC) reported improved median and 2-year survival for patients with glioblastoma treated with concomitant and adjuvant temozolomide and radiotherapy. We report the final results with a median follow-up of more than 5 years. Adult patients with newly diagnosed glioblastoma were randomly assigned to receive either standard radiotherapy or identical radiotherapy with concomitant temozolomide followed by up to six cycles of adjuvant temozolomide. The methylation status of the methyl-guanine methyl transferase gene, MGMT, was determined retrospectively from the tumour tissue of 206 patients. The primary endpoint was overall survival. Analyses were by intention to treat. This trial is registered with Clinicaltrials.gov, number NCT00006353. Between Aug 17, 2000, and March 22, 2002, 573 patients were assigned to treatment. 278 (97%) of 286 patients in the radiotherapy alone group and 254 (89%) of 287 in the combined-treatment group died during 5 years of follow-up. Overall survival was 27.2% (95% CI 22.2-32.5) at 2 years, 16.0% (12.0-20.6) at 3 years, 12.1% (8.5-16.4) at 4 years, and 9.8% (6.4-14.0) at 5 years with temozolomide, versus 10.9% (7.6-14.8), 4.4% (2.4-7.2), 3.0% (1.4-5.7), and 1.9% (0.6-4.4) with radiotherapy alone (hazard ratio 0.6, 95% CI 0.5-0.7; p<0.0001). A benefit of combined therapy was recorded in all clinical prognostic subgroups, including patients aged 60-70 years. Methylation of the MGMT promoter was the strongest predictor for outcome and benefit from temozolomide chemotherapy. Benefits of adjuvant temozolomide with radiotherapy lasted throughout 5 years of follow-up. A few patients in favourable prognostic categories survive longer than 5 years. MGMT methylation status identifies patients most likely to benefit from the addition of temozolomide. EORTC, NCIC, Nélia and Amadeo Barletta Foundation, Schering-Plough.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              MGMT gene silencing and benefit from temozolomide in glioblastoma.

              Epigenetic silencing of the MGMT (O6-methylguanine-DNA methyltransferase) DNA-repair gene by promoter methylation compromises DNA repair and has been associated with longer survival in patients with glioblastoma who receive alkylating agents. We tested the relationship between MGMT silencing in the tumor and the survival of patients who were enrolled in a randomized trial comparing radiotherapy alone with radiotherapy combined with concomitant and adjuvant treatment with temozolomide. The methylation status of the MGMT promoter was determined by methylation-specific polymerase-chain-reaction analysis. The MGMT promoter was methylated in 45 percent of 206 assessable cases. Irrespective of treatment, MGMT promoter methylation was an independent favorable prognostic factor (P<0.001 by the log-rank test; hazard ratio, 0.45; 95 percent confidence interval, 0.32 to 0.61). Among patients whose tumor contained a methylated MGMT promoter, a survival benefit was observed in patients treated with temozolomide and radiotherapy; their median survival was 21.7 months (95 percent confidence interval, 17.4 to 30.4), as compared with 15.3 months (95 percent confidence interval, 13.0 to 20.9) among those who were assigned to only radiotherapy (P=0.007 by the log-rank test). In the absence of methylation of the MGMT promoter, there was a smaller and statistically insignificant difference in survival between the treatment groups. Patients with glioblastoma containing a methylated MGMT promoter benefited from temozolomide, whereas those who did not have a methylated MGMT promoter did not have such a benefit. Copyright 2005 Massachusetts Medical Society.
                Bookmark

                Author and article information

                Journal
                JAMA Oncol
                JAMA Oncol
                JAMA Oncology
                American Medical Association
                2374-2437
                2374-2445
                July 2020
                21 May 2020
                21 May 2020
                : 6
                : 7
                : 1-8
                Affiliations
                [1 ]Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
                [2 ]AUSL–IRCCS Institute of Neurological Sciences, Bologna, Italy
                [3 ]Department of Neurology and Yale Cancer Center, Yale University, New Haven, Connecticut
                [4 ]University College London Hospitals, London, United Kingdom
                [5 ]The Johns Hopkins Hospital, Baltimore, Maryland
                [6 ]Neurology Clinic, University of Heidelberg, National Center for Tumor Diseases, Heidelberg, Germany
                [7 ]Yale School of Medicine, New Haven, Connecticut
                [8 ]Cleveland Clinic, Cleveland, Ohio
                [9 ]Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
                [10 ]Dr Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt, Germany
                [11 ]Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
                [12 ]Hospital Universitario 12 de Octubre, Madrid, Spain
                [13 ]University of Wollongong School of Medicine, Wollongong, NSW, Australia
                [14 ]Moffitt Cancer Center, University of South Florida, Tampa, Florida
                [15 ]Bristol Myers Squibb, Princeton, New Jersey
                [16 ]Duke University Hospital, Durham, North Carolina
                Author notes
                Article Information
                Accepted for Publication: March 4, 2020.
                Corresponding Author: David A. Reardon, MD, Dana-Farber/Harvard Cancer Center, 450 Brookline Ave, D2134, Boston, MA 02215-5450 ( david_reardon@ 123456dfci.harvard.edu ).
                Published Online: May 21, 2020. doi:10.1001/jamaoncol.2020.1024
                Open Access: This is an open access article distributed under the terms of the CC-BY-NC-ND License. © 2020 Reardon DA et al. JAMA Oncology.
                Author Contributions: Dr Reardon had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Drs Reardon and Brandes were co-lead authors.
                Concept and design: Reardon, Brandes, Omuro, Lim, Baehring, Sahebjam, Tatsuoka, Taitt, Zwirtes, Sampson.
                Acquisition, analysis, or interpretation of data: Reardon, Brandes, Omuro, Mulholland, Lim, Wick, Baehring, Ahluwalia, Roth, Bähr, Phuphanich, Sepulveda, De Souza, Sahebjam, Carleton, Taitt, Zwirtes, Weller.
                Drafting of the manuscript: Reardon, Brandes, Omuro, Baehring, De Souza, Tatsuoka, Taitt, Zwirtes, Weller.
                Critical revision of the manuscript for important intellectual content: Brandes, Omuro, Mulholland, Lim, Wick, Baehring, Ahluwalia, Roth, Bähr, Phuphanich, Sepulveda, De Souza, Sahebjam, Carleton, Tatsuoka, Taitt, Zwirtes, Sampson, Weller.
                Statistical analysis: Tatsuoka.
                Administrative, technical, or material support: Omuro, Wick, Baehring, Roth, Bähr, Phuphanich, De Souza, Carleton, Taitt, Zwirtes, Weller.
                Supervision: Reardon, Brandes, Omuro, Mulholland, Roth, Phuphanich, Sepulveda, Carleton, Tatsuoka, Taitt, Zwirtes, Sampson, Weller.
                Conflict of Interest Disclosures: Dr Reardon has received grant funding from Acerta Pharmaceuticals, Incyte, Midatech, Omniox, and Tragara; grant funding and personal fees from Agenus, Celldex, EMD Serono, and Inovio; and personal fees from Advantagene, Genentech/Roche, Merck, Merck KGaA, Monteris, Novocure, Oncorus, Oxigene, Regeneron, Stemline Therapeutics, and Taiho Oncology. Dr Brandes has received travel grants from Roche and Celgene. Dr Omuro has received personal fees from Bristol Myers Squibb, BTG, AstraZeneca, Inovio, Merck, Stemline, Novocure, and Alexion. Dr Mulholland has received grant funding, travel support, and nonfinancial support (to forward plan immuno-oncology use at Mount Vernon) from Bristol Myers Squibb. Dr Lim has received grant funding from Bristol Myers Squibb, Kryin-Kwoya, Biohaven, Accuary, and Arbor; personal fees for an advisory board from SQZ Biotechnologies, VBI Technologies, and Tocagen; consultancy fees from Stryker and Baxter; and grant funding for laboratory research from DNATrix; and has a patent combining local chemotherapy with immunotherapy pending to Arbor and a patent combining stereotactic radiosurgery with immunotherapy that has been issued. Dr Baehring has served as a consultant to Bristol Myers Squibb. Dr Ahluwalia has received research funding from Bristol Myers Squibb; grants and consultancy fees from Incyte, Bristol Myers Squibb, AstraZeneca, Novocure, and AbbVie; grant funding from Tracon, Novartis, Pharmacyclics, Merck, and Bayer; consultancy fees from Monteris Medical, Caris Life Sciences, MRI Solutions, CBT Pharmaceuticals, Flatiron, Karyopharm, Tocagen, Bayer, and Varian Medical Systems; personal fees from Kadman and VBI vaccines; stock options from MimiVax and Doctible; and personal fees from Forma Therapeutics outside the submitted work. Dr Roth has received personal fees from Bristol Myers Squibb, Covagen, Medac, Novocure, Roche, Debiopharm, and Virometix; grants from MSD outside the submitted work. Dr Bähr has received research funding and personal fees from Bristol Myers Squibb and personal fees from Novocure and Medac. Dr Phuphanich reported grants from Bristol Myers during the conduct of the study. Dr Sepulveda has received personal fees from Bayer, AbbVie, Novartis, GW Pharma, Celgene, and Pierre Fabre; and grant funding and personal fees from Pfizer and Catalysis Pharma. Dr De Souza reported other from BioSceptre outside the submitted work. Dr Sahebjam has received grant funding from Merck and Bristol Myers Squibb and funding from Bristol Myers Squibb, Merck, Brooklyn ImmunoTherapeutics, Lilly Pharmaceuticals, Cortice Bioscience, Merck, and Bristol Myers Squibb outside the submitted work. Drs Carleton, Tatsuoka, and Taitt are employed by Bristol Myers Squibb. Dr Zwirtes is employed by and owns stock in Bristol Myers Squibb. Dr Sampson has served as a consultant/advisory board member for Bristol Myers Squibb and Brainlab; has received grant funding and personal fees from and is a patent holder for Celldex Therapeutics; has received grant funding and personal fees from, owns equity/stock in, and is a patent holder for Annias Immunotherapeutics; and owns stock in Istari. Dr Weller has received fees for patient enrollment per study contract from Bristol Myers Squibb; grant funding and personal fees from AbbVie, MSD, Novocure, Merck (EMD Serono), and Roche; grant funding from Actelion, Acceleron, Bayer, Tragara, OGD Pharma, Piqur, and Dracen; and personal fees from Basilea, Celgene, Celldex, Progenics, Tocagen, and Orbus.
                Funding Support: This study was supported by Bristol Myers Squibb.
                Role of the Funder/Sponsor: Bristol Myers Squibb participated in study design, monitoring, and data analysis. The sponsor and authors were involved in data collection and development of the report and approved the decision to submit the final report for publication.
                Data Sharing Statement : See Supplement 4. Bristol Myers Squibb’s policy on data sharing may be found at https://www.bms.com/researchers-and-partners/independent-research/data-sharing-request-process.html.
                Additional Contributions: We thank the patients and their families; investigators, and research staff at all study sites; Ono Pharmaceutical, Osaka, Japan; and the staff of Dako, an Agilent Technologies, Inc, company, for collaborative development of the PD-L1 IHC 28-8 pharmDx assay. Editorial assistance was provided by Bridget Sackey-Aboagye, PhD, of Chrysalis Medical Communications, Inc.
                Article
                coi200016
                10.1001/jamaoncol.2020.1024
                7243167
                32437507
                701426b7-b6f4-4a43-af26-eb521c4b3d2b
                Copyright 2020 Reardon DA et al. JAMA Oncology.

                This is an open access article distributed under the terms of the CC-BY-NC-ND License.

                History
                : 26 November 2019
                : 4 March 2020
                Categories
                Research
                Research
                Original Investigation
                Featured
                Online First
                Comments

                Comments

                Comment on this article