23
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Long-term Clinical Outcomes and Biomarker Analyses of Atezolizumab Therapy for Patients With Metastatic Triple-Negative Breast Cancer : A Phase 1 Study

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          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

          Is single-agent atezolizumab therapy safe and does it provide clinical benefit in patients with metastatic triple-negative breast cancer (mTNBC)?

          Findings

          In this phase 1 study of 116 patients with mTNBC, the safety profile was consistent with that of atezolizumab in other tumor types. With a median follow-up of longer than 2 years, patients with an objective response to atezolizumab had a durable clinical response, and patients with higher tumor immune cell infiltration had better clinical outcomes.

          Meaning

          Single-agent atezolizumab was well tolerated and showed durable clinical activity in patients with mTNBC.

          Abstract

          This phase 1 study evaluates the safety, clinical activity, and biomarkers associated with the use of single-agent atezolizumab in women with metastatic triple-negative breast cancer.

          Abstract

          Importance

          Atezolizumab (anti–programmed cell death ligand 1 [PD-L1]) is well tolerated and clinically active in multiple cancer types. Its safety and clinical activity in metastatic triple-negative breast cancer (mTNBC) has not been reported.

          Objective

          To evaluate the safety, clinical activity, and biomarkers associated with the use of single-agent atezolizumab in patients with mTNBC.

          Design, Setting, and Participants

          Women with mTNBC (defined by investigator assessment) were enrolled between January 2013 and February 2016 in a multicohort open-label, phase 1 study at US and European academic medical centers. Median follow-up was 25.3 months (range, 0.4-45.6 months). Eligible patients regardless of line of therapy had measurable disease by Response Evaluation Criteria in Solid Tumors, version 1.1; Eastern Cooperative Oncology Group performance status of 0 to 1; and a representative tumor sample for assessment of immune cell (IC) PD-L1 expression.

          Interventions

          Atezolizumab was given intravenously every 3 weeks until unacceptable toxic effects or loss of clinical benefit.

          Main Outcomes and Measures

          Primary outcome was safety and tolerability. Activity and exploratory outcomes included objective response rate (ORR), duration of response, progression-free survival (PFS), and overall survival (OS). Outcomes were assessed in all patients and in key patient subgroups.

          Results

          Among 116 evaluable patients (median age, 53 years [range, 29-82 years]), treatment-related adverse events occurred in 73 (63%); 58 (79%) were grade 1 to 2. Most adverse events occurred within the first treatment year. The ORRs were numerically higher in first-line (5 of 21 [24%]) than in second-line or greater patients (6 of 94 [6%]). Median duration of response was 21 months (range, 3 to ≥38 months). Median PFS was 1.4 (95% CI, 1.3-1.6) months by RECIST and 1.9 (95% CI, 1.4-2.5) months by irRC. In first-line patients, median OS was 17.6 months (95% CI, 10.2 months to not estimable). Patients with PD-L1 expression of at least 1% tumor-infiltrating ICs had higher ORRs and longer OS (12% [11 of 91]; 10.1 [95% CI, 7.0-13.8] months, respectively) than those with less than 1% ICs (0 of 21; 6.0 [95% CI, 2.6-12.6] months, respectively). High levels of ICs (>10%) were independently associated with higher ORRs and longer OS.

          Conclusions and Relevance

          Single-agent atezolizumab was well tolerated and provided durable clinical benefit in patients with mTNBC with stable or responding disease and in earlier lines of treatment.

          Trial Registration

          ClinicalTrials.gov identifier: NCT01375842

          Related collections

          Most cited references8

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

          Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199.

          Recent studies suggest that tumor-infiltrating lymphocytes (TILs) are associated with disease-free (DFS) and overall survival (OS) in operable triple-negative breast cancer (TNBC). We seek to validate the prognostic impact of TILs in primary TNBCs in two adjuvant phase III trials conducted by the Eastern Cooperative Oncology Group (ECOG). Full-face hematoxylin and eosin–stained sections of 506 tumors from ECOG trials E2197 and E1199 were evaluated for density of TILs in intraepithelial (iTILs) and stromal compartments (sTILs). Patient cases of TNBC from E2197 and E1199 were randomly selected based on availability of sections. For the primary end point of DFS, association with TIL scores was determined by fitting proportional hazards models stratified on study. Secondary end points were OS and distant recurrence–free interval (DRFI). Reporting recommendations for tumor marker prognostic studies criteria were followed, and all analyses were prespecified. The majority of 481 evaluable cancers had TILs (sTILs, 80%; iTILs, 15%). With a median follow-up of 10.6 years, higher sTIL scores were associated with better prognosis; for every 10% increase in sTILs, a 14% reduction of risk of recurrence or death (P = .02), 18% reduction of risk of distant recurrence (P = .04), and 19% reduction of risk of death (P = .01) were observed. Multivariable analysis confirmed sTILs to be an independent prognostic marker of DFS, DRFI, and OS. In two national randomized clinical trials using contemporary adjuvant chemotherapy, we confirm that stromal lymphocytic infiltration constitutes a robust prognostic factor in TNBCs. Studies assessing outcomes and therapeutic efficacies should consider stratification for this parameter.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers.

            Modulation of immunologic interactions in cancer tissue is a promising therapeutic strategy. To investigate the immunogenicity of human epidermal growth factor receptor 2 (HER2) -positive and triple-negative (TN) breast cancers (BCs), we evaluated tumor-infiltrating lymphocytes (TILs) and immunologically relevant genes in the neoadjuvant GeparSixto trial.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer.

              Recent studies in multiple epithelial cancers have shown that the inhibitory receptor programmed cell death 1 (PD-1) is expressed on tumor-infiltrating lymphocytes and/or programmed death ligand 1 (PD-L1) is expressed on tumor cells, suggesting that antitumor immunity may be modulated by the PD-1/PD-L1 signaling pathway. In addition, phase 1 clinical trials with monoclonal antibodies targeting PD-1 or PD-L1 have shown promising results in several human cancers. The purpose of this study was to investigate the impact of PD-L1 expression in human breast cancer specimens. We conducted an immunohistochemistry study using a tissue microarray encompassing 650 evaluable formalin-fixed breast cancer cases with detailed clinical annotation and outcomes data. PD-L1 was expressed in 152 (23.4 %) of the 650 breast cancer specimens. Expression was significantly associated with age, tumor size, AJCC primary tumor classification, tumor grade, lymph node status, absence of ER expression, and high Ki-67 expression. In univariate analysis, PD-L1 expression was associated with a significantly worse OS. In multivariate analysis, PD-L1 expression remained an independent negative prognostic factor for OS. In subset analyses, expression of PD-L1 was associated with significantly worse OS in the luminal B HER2(-) subtype, the luminal B HER2(+) subtype, the HER2 subtype, and the basal-like subtype. This is the first study to demonstrate that PD-L1 expression is an independent negative prognostic factor in human breast cancer. This finding has important implications for the application of antibody therapies targeting the PD-1/PD-L1 signaling pathway in this disease.
                Bookmark

                Author and article information

                Journal
                JAMA Oncol
                JAMA Oncol
                JAMA Oncol
                JAMA Oncology
                American Medical Association
                2374-2437
                2374-2445
                13 September 2018
                January 2019
                13 September 2019
                : 5
                : 1
                : 74-82
                Affiliations
                [1 ]Bloomberg-Kimmel Institute for Cancer Immunotherapy, Department of Oncology, Johns Hopkins University, Baltimore, Maryland
                [2 ]now with Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh
                [3 ]Medical Oncology Department, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, Spain
                [4 ]Department of Medicine (Oncology), Yale Cancer Center, New Haven, Connecticut
                [5 ]Comprehensive Cancer Centers of Nevada, University of Nevada Las Vegas School of Medicine, Las Vegas
                [6 ]The Angeles Clinic and Research Institute, Los Angeles, California
                [7 ]Department of Medicine,Dana-Farber Cancer Institute, Boston, Massachusetts
                [8 ]Department of Medicine, Hematology/Oncology, Massachusetts General Hospital, Boston
                [9 ]Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
                [10 ]Department of Medical Oncology, Centre Léon-Bérard, Lyon, France
                [11 ]Department of Medical Oncology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
                [12 ]HonorHealth Research Institute, Scottsdale, Arizona
                [13 ]Ventana Medical Systems, Inc, Tucson, Arizona
                [14 ]Genentech, Inc, South San Francisco, California
                [15 ]now with Bellicum Pharmaceuticals, Inc, South San Francisco, California
                [16 ]Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, England
                Author notes
                Article Information
                Accepted for Publication: July 17, 2018.
                Corresponding Author: Leisha A. Emens, MD, PhD, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Ave, Pittsburgh, PA 15213 ( emensla@ 123456upmc.edu ).
                Published Online: September 13, 2018. doi:10.1001/jamaoncol.2018.4224
                Author Contributions: Dr Emens had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.
                Study concept and design: Gordon, Grossman, O’Hear, Fassò, Molinero, Schmid.
                Acquisition, analysis, or interpretation of data: All authors.
                Drafting of the manuscript: Emens, Gordon, ElGabry, Grossman, O’Hear, Fassò, Molinero, Schmid.
                Critical revision of the manuscript for important intellectual content: Emens, Cruz, Eder, Braiteh, Chung, Tolaney, Kuter, Nanda, Cassier, Delord, Gordon, Chang, Sarkar, Grossman, O’Hear, Fassò, Molinero, Schmid.
                Statistical analysis: Chang, Sarkar.
                Obtained funding: Delord.
                Administrative, technical, or material support: Chung, Nanda, Gordon, ElGabry, Molinero.
                Study supervision: Braiteh, Tolaney, Nanda, Gordon, Grossman, Molinero.
                Conflict of Interest Disclosures: Dr Emens has received research support from Roche/Genentech, Corvus, AstraZeneca, and EMD Serono; research grants from Aduro Biotech, Merck, Maxcyte, and the Breast Cancer Research Foundation; advisory board honoraria from Medimmune, AstraZeneca, Celgene, Vaccinex, Peregrine, Bayer, Gritstone, Abbvie, Replimune, Roche-Genentech, Bristol-Myers Squibb, Syndax, and Amgen; other financial support for advisory boards from eTHeRNA and Molecuvax; royalties from Aduro Biotech; and stock options from Molecuvax. Dr Emens was also a member of the US Food and Drug Administration Advisory Committee on Tissue, Cell, and Gene Therapies; and is currently on the Board of Directors for the Society of Immunotherapy of Cancer, and chair of a Data and Safety Monitoring Board for Syndax. Dr Braiteh has received speaking and consulting fees from Amgen, AstraZeneca/Medimmune, Biotheranostics, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Clovis, Eli Lilly, Incyte, Ipsen, Insys, Merck, Merrimack, Pfizer, and Roche/Genentech; advisory board honoraria from Amgen, AstraZeneca/Medimmune, Bayer, Biotheranostics, Boehringer Ingelheim, Clovis, Eli Lilly, Heron Therapeutics, Incyte, Ipsen, Insys, Lexicon, Merck, Merrimack, Pfizer, and Roche/Genentech; and travel support from Amgen, AstraZeneca/Medimmune, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Eli Lilly, Halozyme Therapeutics, Heron Therapeutics, Incyte, Ipsen, Insys, Merck, Merrimack, Pfizer, and Roche/Genentech. Dr Tolaney has received research funding from Roche/Genentech; research funding/consulting fees from Eli Lilly, Novartis, AstraZeneca, Merck, Pfizer, Nektar, Nanostring, and Eisai; and grant support from Exelixis. Dr Nanda has received research funding from Celgene, Corcept, and Merck; and advisory board honoraria from AstraZeneca, Celgene, Roche/Genentech, Merck, Novartis, Pfizer, Syndax, and Puma. She also reports DSMB participation for G1 Therapeutics. Dr Cassier has received grants from Novartis, AstraZeneca, Bristol-Myers Squib, and MSD; personal fees from Novartis, Amgen, and AstraZeneca; and nonfinancial support from Plexxikon, AstraZeneca, and MSD. Dr Gordon has received research funding from AbbVie, Amgen, Array BioPharma, Calithera Biosciences, Celldex, Deciphera, Endocyte, ESSA Pharma, Gilead Sciences, GlaxoSmithKline, Incyte, Lilly, Lilly/ImClone, MedImmune, Merck Serono, Millennium, OncoMed, Pfizer, Plexxikon, Roche/Genentech, Seattle Genetics, Tokai Pharmaceuticals, and TRACON Pharma; and consulting or advisory honoraria from Castle Biosciences, Deciphera, and RedHill Biopharma. Dr ElGabry is a Roche employee. Dr Chang, Ms Sarkar, and Drs Grossman, O’Hear, Fassò, and Molinero are Genentech employees. Drs O’Hear and Molinero are holders of Roche stock. Dr Schmid’s spouse is an employee of Roche/Genentech. Dr Schmid has received honoraria from AstraZeneca, Bayer, Boehringer Ingelheim, Celgene, Eisai, Novartis, Pfizer, Puma, and Roche/Genentech. His institution has received research funding or grants from Astellas, AstraZeneca, Medivation, Novartis, Oncogenex, and Roche/Genentech. No other disclosures are reported.
                Funding/Support: This study was sponsored by F. Hoffmann-La Roche Ltd.
                Role of the Funder/Sponsor: The protocol was developed by the sponsor (F. Hoffmann-La Roche Ltd) and advisors. Data were collected collaboratively by the sponsor and clinical investigators. Statisticians employed by the sponsor analyzed the data. The sponsor participated in the preparation, review, and approval of the manuscript; and decision to submit the manuscript for publication.
                Previous Presentation: This study was presented in part, based on earlier data, at the following meetings: San Antonio Breast Cancer Symposium; December 10, 2014; San Antonio, Texas; American Association for Cancer Research Annual Meeting; April 20, 2015; Philadelphia, Pennsylvania; and American Association for Cancer Research Annual Meeting; April 3, 2017; Washington, DC.
                Additional Contributions: We thank the patients, their families, and the clinical study site investigators and staff. Daniel S. Chen, MD, PhD, Gregg Fine, MD, and Priti S. Hegde, PhD, all employees of Genentech, contributed to the design and conduct of the study (D.S.C., G.F.) and to the study biomarker plan (P.S.H.). Medical writing assistance was provided by Ernestine Chung, PhD, and Jonathan Lee, PhD, of Health Interactions and funded by F. Hoffmann-La Roche, Ltd.
                Article
                PMC6439773 PMC6439773 6439773 coi180079
                10.1001/jamaoncol.2018.4224
                6439773
                30242306
                56591f39-4147-4129-b55c-8acc3d7dc32e
                Copyright 2018 American Medical Association. All Rights Reserved.
                History
                : 13 April 2018
                : 10 July 2018
                : 17 July 2018
                Funding
                Funded by: F. Hoffmann-La Roche Ltd
                Categories
                Research
                Research
                Original Investigation
                Online First

                Comments

                Comment on this article